WO2022247885A1 - 三并杂环类化合物的结晶和盐及其应用 - Google Patents

三并杂环类化合物的结晶和盐及其应用 Download PDF

Info

Publication number
WO2022247885A1
WO2022247885A1 PCT/CN2022/095126 CN2022095126W WO2022247885A1 WO 2022247885 A1 WO2022247885 A1 WO 2022247885A1 CN 2022095126 W CN2022095126 W CN 2022095126W WO 2022247885 A1 WO2022247885 A1 WO 2022247885A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
angles
following
alternatively
Prior art date
Application number
PCT/CN2022/095126
Other languages
English (en)
French (fr)
Other versions
WO2022247885A9 (zh
Inventor
毛魏魏
樊莉莉
钱文远
韦昌青
陈曙辉
Original Assignee
正大天晴药业集团股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 正大天晴药业集团股份有限公司 filed Critical 正大天晴药业集团股份有限公司
Priority to CN202280033492.7A priority Critical patent/CN117295739A/zh
Publication of WO2022247885A1 publication Critical patent/WO2022247885A1/zh
Publication of WO2022247885A9 publication Critical patent/WO2022247885A9/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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • This application relates to the crystal of the compound of formula (I), its salt and the crystal of the salt, and its application in the preparation of medicines for treating diseases related to JAKs.
  • Janus kinases are cytoplasmic tyrosine kinases that transmit cytokine signals, from membrane receptors to STAT transcription factors.
  • the JAK family consists of four members: JAK1, JAK2, JAK3 and TYK2.
  • the JAK-STAT pathway transduces extracellular signals from a variety of cytokines, growth factors and hormones to the nucleus and is responsible for the expression of thousands of protein-coding genes.
  • JAK-STAT intracellular signal transduction serves interferons, most interleukins, and various cytokines and endocrine factors, such as EPO, TPO, GH, OSM, LIF, CNTF, GM-CSF, and PRL (Vainchenker W. et al .(2008).
  • JAK-1, JAK-2 and TYK-2 are expressed in various tissue cells of the human body, and JAK-3 is mainly expressed in various hematopoietic tissue cells, mainly in bone marrow cells, thymocytes, NK cells and activated B lymphocytes , T lymphocytes.
  • JAK1 has emerged as a novel target in disease areas such as immunity, inflammation and cancer.
  • a base mutation JAK2V617F on the JAK2 gene in humans is associated with polycythemia vera (PV), essential thrombocythemia (ET), idiopathic myelofibrosis (IMF), chronic It is closely related to the occurrence of myeloid leukemia (CML).
  • JAK3 Mutations in either JAK3 or ⁇ c can cause severe combined immunodeficiency.
  • the abnormal activity of JAK3 is manifested by a large reduction of T cells and NK cells and loss of B cell function, which seriously affects the normal biological functions of the immune system.
  • JAK3 Based on its functional characteristics and special tissue distribution, JAK3 has become an attractive drug target for immune system-related diseases.
  • the loss of TYK2 function will cause defects in the signaling pathways of various cytokine receptors, which will lead to viral infection, decreased antibacterial immune function, and increased the possibility of lung infection (John J.O'Shea, 2004 , Nature Reviews Drug Discovery 3, 555-564).
  • JAK inhibitors selectively bind to different cytokine receptors, endow signal transduction specificity, and thus play different physiological roles.
  • This selective mode of action makes JAK inhibitors relatively specific for diseases treat.
  • IL-2 or IL-4 receptors bind JAK1 and JAK3 along with a common gamma chain, whereas type I receptors with the same beta chain bind JAK2.
  • Type I receptors using gp130 (glycoprotein 130) and activated by heterodimeric cytokines preferentially bind JAK1/2 and TYK2.
  • Type I receptors activated by hormone-like cytokines bind and activate the JAK2 kinase.
  • Type II receptors for interferons bind JAK1 and TYK2, whereas receptors of the IL-10 cytokine family bind JAK1/2 and TYK2.
  • Various specific combinations of the cytokines and their receptors with JAK family members trigger different physiological effects and provide the possibility for the treatment of different diseases. Heterodimerization of JAK1 with other JAKs to transduce cytokine-driven pro-inflammatory signaling. Therefore, inhibition of JAK1 and/or other JAKs is expected to be of therapeutic benefit for a range of inflammatory disorders and other diseases driven by JAK-mediated signaling (Daniella M. Schwartz, 2017, Nature Reviews Drug Discovery 16, 843-862. )
  • the application provides a compound of formula (I) or a salt thereof
  • the salt of the compound of formula (I) is selected from phosphate or oxalate.
  • the present application provides crystals of the compound of formula (I) or a salt thereof.
  • the application provides crystals of the phosphate salt of the compound of formula (I) or crystals of the oxalate salt of the compound of formula (I).
  • the application provides the type A crystal of the compound of formula (I), which has diffraction peaks at the following 2 ⁇ angles in the X-ray powder diffraction pattern using Cu K ⁇ radiation: 5.74 ⁇ 0.20°, 7.23 ⁇ 0.20° and 12.28 ⁇ 0.2 °;
  • the X-ray powder diffraction pattern of the type A crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.26 ⁇ 0.20°, 5.74 ⁇ 0.20°, 7.23 ⁇ 0.20°, 9.25 ⁇ 0.20°, 12.28 ⁇ 0.20° and 14.46 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the type A crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.26 ⁇ 0.20°, 5.74 ⁇ 0.20°, 7.23 ⁇ 0.20°, 9.25 ⁇ 0.20°, 12.28 ⁇ 0.2°, 14.46 ⁇ 0.2°, 14.89 ⁇ 0.2°, and 21.06 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the type A crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.26 ⁇ 0.20°, 5.74 ⁇ 0.20°, 7.23 ⁇ 0.20°, 9.25 ⁇ 0.20°, 12.28 ⁇ 0.20°, 14.46 ⁇ 0.20°, 14.89 ⁇ 0.20°, 15.75 ⁇ 0.20°, 16.82 ⁇ 0.20°, 19.37 ⁇ 0.20°, and 21.06 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the type A crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.26 ⁇ 0.20°, 5.74 ⁇ 0.20°, 7.23 ⁇ 0.20°, 8.80 ⁇ 0.20°, 9.25 ⁇ 0.20°, 12.28 ⁇ 0.20°, 14.23 ⁇ 0.20°, 14.46 ⁇ 0.20°, 14.89 ⁇ 0.20°, 15.75 ⁇ 0.20°, 16.82 ⁇ 0.20°, 17.32 ⁇ 0.20°, 17.84 ⁇ 0.20°, 19.37 ⁇ 0.20°, 21.06 ⁇ 0.20°, 22.76 ⁇ 0.20°, 24.11 ⁇ 0.20°, 26.16 ⁇ 0.20°, 26.76 ⁇ 0.20°, and 27.16 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the type A crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.26 ⁇ 0.20°, 5.74 ⁇ 0.20°, 7.23 ⁇ 0.20°, 9.25 ⁇ 0.20°, 9.64 ⁇ 0.20°, 10.50 ⁇ 0.20°, 12.28 ⁇ 0.20°, and 14.46 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the type A crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.26 ⁇ 0.20°, 5.74 ⁇ 0.20°, 7.23 ⁇ 0.20°, 9.25 ⁇ 0.20°, 9.64 ⁇ 0.20°, 10.50 ⁇ 0.20°, 12.28 ⁇ 0.2°, 14.46 ⁇ 0.2°, 14.89 ⁇ 0.2°, and 21.06 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the type A crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.26 ⁇ 0.20°, 5.74 ⁇ 0.20°, and 21.06 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the type A crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.26 ⁇ 0.20°, 5.74 ⁇ 0.20°, 7.23 ⁇ 0.20°, 8.80 ⁇ 0.20°, 9.25 ⁇ 0.20°, 9.64 ⁇ 0.20°, 10.50 ⁇ 0.20°, 12.28 ⁇ 0.20°, 14.23 ⁇ 0.20°, 14.46 ⁇ 0.20°, 14.89 ⁇ 0.20°, 15.75 ⁇ 0.20°, 16.82 ⁇ 0.20°, 17.32 ⁇ 0.20°, 17.84 ⁇ 0.20°, 19.37 ⁇ 0.20°, 21.06 ⁇ 0.20°, 22.76 ⁇ 0.20°, 24.11 ⁇ 0.20°, 26.16 ⁇ 0.20°, 26.76 ⁇ 0.20°, and 27.16 ⁇ 0.20°.
  • the application provides the type A crystal of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, including 3, 4, 5, 6, 7, 8, 9, 10 or 11 diffraction peaks: 5.26 ⁇ 0.20°, 5.74 ⁇ 0.20°, 7.23 ⁇ 0.20°, 9.25 ⁇ 0.20°, 12.28 ⁇ 0.20°, 14.46 ⁇ 0.20°, 14.89 ⁇ 0.20°, 15.75 ⁇ 0.20° , 16.82 ⁇ 0.20°, 19.37 ⁇ 0.20° and 21.06 ⁇ 0.20°.
  • the application provides the type A crystal of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, comprising 3, 4, 5, 6, 7 or Eight diffraction peaks: 5.26 ⁇ 0.20°, 5.74 ⁇ 0.20°, 7.23 ⁇ 0.20°, 9.25 ⁇ 0.20°, 12.28 ⁇ 0.2°, 14.46 ⁇ 0.2°, 14.89 ⁇ 0.2° and 21.06 ⁇ 0.2°.
  • the XRPD pattern of type A crystal of the compound of formula (I) is shown in FIG. 1 .
  • the differential scanning calorimetry (DSC) spectrum of Form A crystal of the compound of formula (I) has an onset point of an exothermic peak at 188.8 ⁇ 3°C.
  • the DSC spectrum of type A crystal of the compound of formula (I) is shown in FIG. 2 .
  • thermogravimetric analysis (TGA) pattern of the Form A crystal of the compound of formula (I) has a weight loss of 3.13% at 200 ⁇ 3°C.
  • the TGA spectrum of type A crystal of the compound of formula (I) is shown in FIG. 3 .
  • the present application provides a method for preparing type A crystals of the compound of formula (I), the method comprising the following steps: dissolving the compound of formula (I) in ethanol to precipitate a solid.
  • the compound of formula (I) is mixed with ethanol, and the solution of the compound of formula (I) is clarified by heating and stirring.
  • the resulting solution is cooled to precipitate a solid.
  • the above method further comprises the step of isolating the solid and then drying the solid; optionally, drying under vacuum.
  • the application also provides type B crystals of the compound of formula (I), which has diffraction peaks at the following 2 ⁇ angles in the X-ray powder diffraction pattern using Cu K ⁇ radiation: 5.08 ⁇ 0.20°, 9.78 ⁇ 0.20° and 13.76 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the type B crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.08 ⁇ 0.20°, 9.78 ⁇ 0.20°, 13.76 ⁇ 0.20°, 18.96 ⁇ 0.20°, and 24.34 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the type B crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.08 ⁇ 0.20°, 9.78 ⁇ 0.20°, 13.76 ⁇ 0.20°, 15.80 ⁇ 0.20°, 18.96 ⁇ 0.20°, 19.61 ⁇ 0.20°, and 24.34 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the type B crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.08 ⁇ 0.20°, 6.31 ⁇ 0.20°, 9.78 ⁇ 0.20°, 13.76 ⁇ 0.20°, 15.80 ⁇ 0.20°, 18.96 ⁇ 0.20°, 19.61 ⁇ 0.20°, and 24.34 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the type B crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 5.08 ⁇ 0.20°, 6.31 ⁇ 0.20°, 9.78 ⁇ 0.20°, 11.75 ⁇ 0.20°, 13.76 ⁇ 0.20°, 14.17 ⁇ 0.20°, 15.80 ⁇ 0.20°, 18.96 ⁇ 0.20°, 19.61 ⁇ 0.20°, 23.87 ⁇ 0.20°, and 24.34 ⁇ 0.20°.
  • the application provides the type B crystal of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, comprising 3, 4, 5, 6, 7, 8, 9, 10 or 11 diffraction peaks: 5.08 ⁇ 0.20°, 6.31 ⁇ 0.20°, 9.78 ⁇ 0.20°, 11.75 ⁇ 0.20°, 13.76 ⁇ 0.20°, 14.17 ⁇ 0.20°, 15.80 ⁇ 0.20°, 18.96 ⁇ 0.20° , 19.61 ⁇ 0.20°, 23.87 ⁇ 0.20° and 24.34 ⁇ 0.20°.
  • the application provides the type B crystal of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, comprising 3, 4, 5, 6, 7 or Eight diffraction peaks: 5.08 ⁇ 0.20°, 6.31 ⁇ 0.20°, 9.78 ⁇ 0.20°, 13.76 ⁇ 0.20°, 15.80 ⁇ 0.20°, 18.96 ⁇ 0.20°, 19.61 ⁇ 0.20° and 24.34 ⁇ 0.20°.
  • the XRPD spectrum of the type B crystal of the compound of formula (I) is shown in FIG. 4 .
  • the differential scanning calorimetry (DSC) spectrum of the Form B crystal of the compound of formula (I) has an onset point of an endothermic peak at 172.9 ⁇ 3°C.
  • the DSC spectrum of the type B crystal of the compound of formula (I) is shown in FIG. 5 .
  • thermogravimetric analysis (TGA) spectrum of the crystal type B of the compound of formula (I) has a weight loss of 5.18% at 150 ⁇ 3°C.
  • the TGA spectrum of the type B crystal of the compound of formula (I) is shown in FIG. 6 .
  • the present application provides a method for preparing type B crystals of the compound of formula (I), the method comprising the following steps: the compound of formula (I) is mixed with DMF and MEK, precipitated and separated to obtain a solid.
  • the above precipitation process is carried out at -20° C. to room temperature.
  • the above method further comprises the step of drying the isolated solid; optionally, drying at room temperature.
  • the application also provides type C crystals of the compound of formula (I), which has diffraction peaks at the following 2 ⁇ angles in the X-ray powder diffraction pattern using Cu K ⁇ radiation: 6.21 ⁇ 0.20°, 9.04 ⁇ 0.20° and 12.38 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the type C crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 6.21 ⁇ 0.20°, 9.04 ⁇ 0.20°, 11.54 ⁇ 0.20°, 12.38 ⁇ 0.20°, and 23.29 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the type C crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 6.21 ⁇ 0.20°, 9.04 ⁇ 0.20°, 11.54 ⁇ 0.20°, 12.38 ⁇ 0.20°, 16.27 ⁇ 0.20°, 23.29 ⁇ 0.20°, and 25.62 ⁇ 0.20°.
  • the application provides the type C crystal of the compound of formula (I), which contains 3, 4, 5, 6 or 7 crystals selected from the following 2 ⁇ angles in the X-ray powder diffraction pattern using Cu K ⁇ radiation. Diffraction peaks: 6.21 ⁇ 0.20°, 9.04 ⁇ 0.20°, 11.54 ⁇ 0.20°, 12.38 ⁇ 0.20°, 16.27 ⁇ 0.20°, 23.29 ⁇ 0.20° and 25.62 ⁇ 0.20°.
  • the XRPD pattern of the type C crystal of the compound of formula (I) is shown in FIG. 7 .
  • the differential scanning calorimetry (DSC) spectrum of the type C crystal of the compound of formula (I) has an onset point of an endothermic peak at 140.2 ⁇ 3°C.
  • the DSC spectrum of the type C crystal of the compound of formula (I) is shown in FIG. 8 .
  • thermogravimetric analysis (TGA) spectrum of the type C crystal of the compound of formula (I) has a weight loss of 3.10% at 130 ⁇ 3°C.
  • the TGA spectrum of the type C crystal of the compound of formula (I) is shown in FIG. 9 .
  • the present application provides a method for preparing type C crystals of the compound of formula (I), the method comprising the following steps: dissolving the compound of formula (I) in ACN, and cooling down to precipitate a solid.
  • a filtration operation is also included; optionally, the filtration is performed using a 0.45 micron PTFE filter head.
  • the above-mentioned cooling refers to cooling from 50° C. to 5° C. at a rate of 0.1° C./minute.
  • the above method further includes the step of isolating the obtained solid and then drying the solid; optionally, drying at room temperature.
  • the application also provides the D-type crystal of the compound of formula (I), which has diffraction peaks at the following 2 ⁇ angles in the X-ray powder diffraction pattern using Cu K ⁇ radiation: 7.13 ⁇ 0.20°, 18.22 ⁇ 0.20° and 21.45 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the D-type crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 7.13 ⁇ 0.20°, 10.04 ⁇ 0.20°, 11.24 ⁇ 0.20°, 15.97 ⁇ 0.20°, 18.22 ⁇ 0.20°, and 21.45 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the D-type crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 7.13 ⁇ 0.20°, 10.04 ⁇ 0.20°, 11.24 ⁇ 0.20°, 15.97 ⁇ 0.20°, 16.94 ⁇ 0.20°, 18.22 ⁇ 0.20°, 21.45 ⁇ 0.20°, and 22.71 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the D-type crystal of the compound of formula (I) using Cu K ⁇ radiation has diffraction peaks at the following 2 ⁇ angles: 7.13 ⁇ 0.20°, 10.04 ⁇ 0.20°, 11.24 ⁇ 0.20°, 15.97 ⁇ 0.20°, 16.94 ⁇ 0.20°, 18.22 ⁇ 0.20°, 20.28 ⁇ 0.20°, 21.45 ⁇ 0.20°, 22.71 ⁇ 0.20°, and 26.21 ⁇ 0.20°.
  • the application provides the type D crystal of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, including 3, 4, 5, 6, 7, 8, 9 or 10 diffraction peaks: 7.13 ⁇ 0.20°, 10.04 ⁇ 0.20°, 11.24 ⁇ 0.20°, 15.97 ⁇ 0.20°, 16.94 ⁇ 0.20°, 18.22 ⁇ 0.20°, 20.28 ⁇ 0.20°, 21.45 ⁇ 0.20°, 22.71 ⁇ 0.20° and 26.21 ⁇ 0.20°.
  • the application provides the D-type crystal of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, comprising 3, 4, 5, 6, 7 or Eight diffraction peaks: 7.13 ⁇ 0.20°, 10.04 ⁇ 0.20°, 11.24 ⁇ 0.20°, 15.97 ⁇ 0.20°, 16.94 ⁇ 0.20°, 18.22 ⁇ 0.20°, 21.45 ⁇ 0.20° and 22.71 ⁇ 0.20°.
  • the XRPD spectrum of the type D crystal of the compound of formula (I) is shown in FIG. 10 .
  • the peak position and relative intensity of the diffraction peaks of the X-ray powder diffraction pattern using Cu K ⁇ radiation are shown in Table 4:
  • thermogravimetric analysis (TGA) spectrum of the type D crystal of the compound of formula (I) has a weight loss of 1.83% at 140 ⁇ 3°C.
  • the TGA spectrum of the D-type crystal of the compound of formula (I) is shown in FIG. 11 .
  • the present application provides a method for preparing type D crystals of the compound of formula (I), the method comprising the following steps: mixing the compound of formula (I) with maleic acid and acetone, and separating the solid.
  • the above method further includes a step of drying the separated solid; optionally, drying at room temperature under vacuum conditions.
  • the present application also provides the phosphate salt of the compound of formula (I).
  • the present application also provides crystals of the phosphate salt of the compound of formula (I).
  • the ratio of the number of molecules of the compound of formula (I) to phosphoric acid is 1: (1-1.5); in some embodiments, the formula ( 1) The ratio of the number of molecules of the compound to phosphoric acid is 1:1.
  • the phosphate salt of the compound of formula (I) is a compound of formula (II),
  • the phosphate of the compound of formula (I) is a crystal of the phosphate of the compound of formula (I).
  • the application also provides crystallization of the phosphate salt of the compound of formula (I), which has diffraction peaks at the following 2 ⁇ angles in the X-ray powder diffraction pattern using Cu K ⁇ radiation: 7.87 ⁇ 0.20°, 16.64 ⁇ 0.20° and 20.04 ⁇ 0.20°.
  • the X-ray powder diffraction pattern using Cu K ⁇ radiation of the phosphate salt of the compound of formula (I) has diffraction peaks at the following 2 ⁇ angles: 6.69 ⁇ 0.20 °, 7.87 ⁇ 0.20 ° , 14.15 ⁇ 0.20°, 16.64 ⁇ 0.20° and 20.04 ⁇ 0.20°.
  • the X-ray powder diffraction pattern using Cu K ⁇ radiation of the phosphate salt of the compound of formula (I) has diffraction peaks at the following 2 ⁇ angles: 6.69 ⁇ 0.20 °, 7.87 ⁇ 0.20 ° , 14.15 ⁇ 0.20°, 16.64 ⁇ 0.20°, 20.04 ⁇ 0.20°, 21.73 ⁇ 0.20°, 26.17 ⁇ 0.20° and 28.11 ⁇ 0.20°.
  • the X-ray powder diffraction pattern using Cu K ⁇ radiation of the phosphate salt of the compound of formula (I) has diffraction peaks at the following 2 ⁇ angles: 6.69 ⁇ 0.20 °, 7.87 ⁇ 0.20 ° , 14.15 ⁇ 0.20°, 15.21 ⁇ 0.20°, 16.64 ⁇ 0.20°, 20.04 ⁇ 0.20°, 21.73 ⁇ 0.20°, 22.21 ⁇ 0.20°, 25.07 ⁇ 0.20°, 26.17 ⁇ 0.20°, 26.81 ⁇ 0.20° and 28.11 ⁇ 0.20° .
  • the X-ray powder diffraction pattern using Cu K ⁇ radiation of the phosphate salt of the compound of formula (I) has diffraction peaks at the following 2 ⁇ angles: 6.69 ⁇ 0.20 °, 7.87 ⁇ 0.20 ° , 14.15 ⁇ 0.20°, 15.21 ⁇ 0.20°, 16.64 ⁇ 0.20°, 20.04 ⁇ 0.20°, 21.73 ⁇ 0.20°, 22.21 ⁇ 0.20°, 23.66 ⁇ 0.20°, 25.07 ⁇ 0.20°, 26.17 ⁇ 0.20°, 26.81 ⁇ 0.20° , 27.50 ⁇ 0.20° and 28.11 ⁇ 0.20°.
  • the X-ray powder diffraction pattern using Cu K ⁇ radiation of the phosphate salt of the compound of formula (I) has diffraction peaks at the following 2 ⁇ angles: 6.69 ⁇ 0.20 °, 7.87 ⁇ 0.20 ° , 14.15 ⁇ 0.20°, 15.21 ⁇ 0.20°, 16.64 ⁇ 0.20°, 20.04 ⁇ 0.20°, 21.73 ⁇ 0.20°, 22.21 ⁇ 0.20°, 23.66 ⁇ 0.20°, 25.07 ⁇ 0.20°, 25.94 ⁇ 0.20°, 26.17 ⁇ 0.20° , 26.81 ⁇ 0.20°, 27.50 ⁇ 0.20°, 28.11 ⁇ 0.20°, 29.06 ⁇ 0.20° and 29.85 ⁇ 0.20°.
  • the application provides the crystallization of the phosphate salt of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, comprising 3, 4, 5, 6, 7 selected from the following 2 ⁇ angles. , 8, 9, 10, 11, 12, 13 or 14 diffraction peaks: 6.69 ⁇ 0.20°, 7.87 ⁇ 0.20°, 14.15 ⁇ 0.20°, 15.21 ⁇ 0.20°, 16.64 ⁇ 0.20°, 20.04 ⁇ 0.20°, 21.73 ⁇ 0.20°, 22.21 ⁇ 0.20°, 23.66 ⁇ 0.20°, 25.07 ⁇ 0.20°, 26.17 ⁇ 0.20°, 26.81 ⁇ 0.20°, 27.50 ⁇ 0.20°, and 28.11 ⁇ 0.20°.
  • the application provides the crystallization of the phosphate salt of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, comprising 3, 4, 5, 6, 7 selected from the following 2 ⁇ angles. Or 8 diffraction peaks: 6.69 ⁇ 0.20°, 7.87 ⁇ 0.20°, 14.15 ⁇ 0.20°, 16.64 ⁇ 0.20°, 20.04 ⁇ 0.20°, 21.73 ⁇ 0.20°, 26.17 ⁇ 0.20° and 28.11 ⁇ 0.20°.
  • the XRPD pattern of the crystallization of the phosphate salt of the compound of formula (I) is shown in FIG. 12 .
  • the peak positions and relative strengths of the diffraction peaks of the X-ray powder diffraction spectrum using Cu K ⁇ radiation are as shown in Table 5 Show:
  • the differential scanning calorimetry (DSC) spectrum of the phosphate salt of the compound of formula (I) has an onset point of an exothermic peak at 141.9 ⁇ 3.0°C.
  • the DSC spectrum of the crystallization of the phosphate salt of the compound of formula (I) is shown in FIG. 13 .
  • the crystalline thermogravimetric analysis (TGA) spectrum of the phosphate salt of the compound of formula (I) has a weight loss of 5.50% at 130 ⁇ 3°C.
  • the TGA spectrum of the crystallization of the phosphate salt of the compound of formula (I) is shown in FIG. 14 .
  • the present application provides a method for preparing phosphate crystals of the compound of formula (I), said method comprising the following steps: mixing the compound of formula (I) with phosphoric acid, EtOH and H 2 O, and separating to obtain a solid.
  • the above preparation method includes stirring under heating after the mixing; optionally, stirring under heating to 40-80°C; optionally, heating to Stirring was carried out at 50°C.
  • the above method further includes a step of drying the isolated solid; optionally drying at room temperature under vacuum.
  • the present application also provides the oxalate salt of the compound of formula (I).
  • the ratio of the number of molecules of the compound of formula (I) to oxalic acid is 1:(1-1.5); in some embodiments, the The ratio of the number of molecules of the compound of formula (I) to oxalic acid is 1:1.1.
  • the oxalate salt of the compound of formula (I) is a compound of formula (III),
  • the oxalate of the compound of formula (I) is a crystal of the oxalate of the compound of formula (I), such as type I crystal of the oxalate of the compound of formula (I) or the formula ( I) Form II crystal of the oxalate salt of the compound.
  • the application also provides type I crystals of the oxalate salt of the compound of formula (I), which has diffraction peaks at the following 2 ⁇ angles in the X-ray powder diffraction pattern using Cu K ⁇ radiation: 5.06 ⁇ 0.20°, 12.69 ⁇ 0.20° and 15.21 ⁇ 0.20°.
  • the X-ray powder diffraction pattern using Cu K ⁇ radiation of the type I crystal of the oxalate salt of the compound of formula (I) has diffraction peaks at the following 2 ⁇ angles: 5.06 ⁇ 0.20°, 12.69 ⁇ 0.20°, 15.21 ⁇ 0.20°, 17.92 ⁇ 0.20°, 20.77 ⁇ 0.20°, and 27.32 ⁇ 0.20°.
  • the X-ray powder diffraction pattern using Cu K ⁇ radiation of the type I crystal of the oxalate salt of the compound of formula (I) has diffraction peaks at the following 2 ⁇ angles: 5.06 ⁇ 0.20°, 11.28 ⁇ 0.20°, 12.69 ⁇ 0.20°, 15.21 ⁇ 0.20°, 16.48 ⁇ 0.20°, 17.92 ⁇ 0.20°, 19.52 ⁇ 0.20°, 20.77 ⁇ 0.20°, 22.90 ⁇ 0.20°, and 27.32 ⁇ 0.20°.
  • the application provides the type I crystal of the oxalate salt of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, comprising 3, 4, 5 or Six diffraction peaks: 5.06 ⁇ 0.20°, 12.69 ⁇ 0.20°, 15.21 ⁇ 0.20°, 17.92 ⁇ 0.20°, 20.77 ⁇ 0.20° and 27.32 ⁇ 0.20°.
  • the XRPD pattern of type I crystal of the oxalate salt of the compound of formula (I) is shown in FIG. 15 .
  • the peak positions and relative intensities of the diffraction peaks of the X-ray powder diffraction spectrum using Cu K ⁇ radiation are as follows: Table 6 shows:
  • the DSC spectrum of the type I crystal of the oxalate salt of the compound of formula (I) is shown in FIG. 16 .
  • thermogravimetric analysis (TGA) pattern of the type I crystal of the oxalate salt of the compound of formula (I) has a weight loss of 11.44% at 150 ⁇ 3°C.
  • the TGA spectrum of type I crystal of the oxalate salt of the compound of formula (I) is shown in FIG. 17 .
  • the present application provides a method for preparing type I crystals of the oxalate salt of the compound of formula (I), the method comprising the following steps: mixing the compound of formula (I) with oxalic acid and acetone, and separating the solid.
  • the above preparation method includes stirring under heating after the mixing; optionally, the heating temperature is 40-80°C; optionally, the heating temperature is 50°C.
  • the above method further includes a step of drying the separated solid; optionally, drying at room temperature under vacuum conditions.
  • the application also provides type II crystals of the oxalate salt of the compound of formula (I), which has diffraction peaks at the following 2 ⁇ angles in the X-ray powder diffraction pattern using Cu K ⁇ radiation: 5.44 ⁇ 0.20°, 11.75 ⁇ 0.20° and 13.77 ⁇ 0.20°.
  • the X-ray powder diffraction pattern using Cu K ⁇ radiation of the type II crystal of the oxalate salt of the compound of formula (I) has diffraction peaks at the following 2 ⁇ angles: 5.44 ⁇ 0.20°, 10.85 ⁇ 0.20°, 11.75 ⁇ 0.20°, 13.77 ⁇ 0.20°, 16.27 ⁇ 0.20°, and 27.42 ⁇ 0.20°.
  • the X-ray powder diffraction pattern using Cu K ⁇ radiation of the type II crystal of the oxalate salt of the compound of formula (I) has diffraction peaks at the following 2 ⁇ angles: 5.44 ⁇ 0.20°, 10.85 ⁇ 0.20°, 11.75 ⁇ 0.20°, 13.77 ⁇ 0.20°, 16.01 ⁇ 0.20°, 16.27 ⁇ 0.20°, 23.39 ⁇ 0.20°, and 27.42 ⁇ 0.20°.
  • the X-ray powder diffraction pattern using Cu K ⁇ radiation of the type II crystal of the oxalate salt of the compound of formula (I) has diffraction peaks at the following 2 ⁇ angles: 5.44 ⁇ 0.20°, 10.85 ⁇ 0.20°, 11.75 ⁇ 0.20°, 13.77 ⁇ 0.20°, 16.01 ⁇ 0.20°, 16.27 ⁇ 0.20°, 17.85 ⁇ 0.20°, 18.39 ⁇ 0.20°, 20.83 ⁇ 0.20°, 21.75 ⁇ 0.20°, 23.39 ⁇ 0.20°, 25.13 ⁇ 0.20° and 27.42 ⁇ 0.20°.
  • the application provides the type II crystal of the oxalate salt of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, comprising 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or 13 diffraction peaks: 5.44 ⁇ 0.20°, 10.85 ⁇ 0.20°, 11.75 ⁇ 0.20°, 13.77 ⁇ 0.20°, 16.01 ⁇ 0.20°, 16.27 ⁇ 0.20°, 17.85 ⁇ 0.20°, 18.39 ⁇ 0.20°, 20.83 ⁇ 0.20°, 21.75 ⁇ 0.20°, 23.39 ⁇ 0.20°, 25.13 ⁇ 0.20°, and 27.42 ⁇ 0.20°.
  • the application provides the type II crystal of the oxalate salt of the compound of formula (I), which uses Cu K ⁇ radiation in the X-ray powder diffraction pattern, comprising 3, 4, 5, 6, 7 or 8 diffraction peaks: 5.44 ⁇ 0.20°, 10.85 ⁇ 0.20°, 11.75 ⁇ 0.20°, 13.77 ⁇ 0.20°, 16.01 ⁇ 0.20°, 16.27 ⁇ 0.20°, 23.39 ⁇ 0.20° and 27.42 ⁇ 0.20°.
  • the XRPD spectrum of the type II crystal of the oxalate salt of the compound of formula (I) is shown in FIG. 18 .
  • the peak positions and relative intensities of the diffraction peaks of the X-ray powder diffraction spectrum using Cu K ⁇ radiation are as follows: Table 7 shows:
  • Table 7 XRPD data of Form II crystals of the oxalate salt of the compound of formula (I)
  • the DSC spectrum of the type II crystal of the oxalate salt of the compound of formula (I) is shown in FIG. 19 .
  • thermogravimetric analysis (TGA) pattern of the type II crystal of the oxalate salt of the compound of formula (I) has a weight loss of 7.78% at 150 ⁇ 3°C.
  • the TGA spectrum of the type II crystal of the oxalate salt of the compound of formula (I) is shown in FIG. 20 .
  • the present application provides a method for preparing type II crystals of the oxalate salt of the compound of formula (I), the method comprising the following steps: mixing the compound of formula (I) with oxalic acid, EtOH and H 2 O, separating A solid was obtained.
  • the above preparation method includes stirring under heating after the mixing; optionally, stirring at 40-80°C; optionally, stirring at 50°C .
  • the above preparation method further includes a drying step; optionally, drying is performed at room temperature under vacuum conditions.
  • the present application provides a crystal form composition comprising the crystals described in the present application, wherein the crystals account for more than 50% of the weight of the crystal form composition, preferably more than 80%, more preferably more than 90%, Preferably more than 95%.
  • the present application provides a pharmaceutical composition comprising a therapeutically effective amount of the crystal of the compound of formula (I), the salt of the compound of formula (I) or its crystal, or its crystal form combination.
  • the pharmaceutical composition of the present application may or may not contain pharmaceutically acceptable auxiliary materials.
  • the pharmaceutical composition of the present application may further include one or more other therapeutic agents.
  • the present application provides a solid pharmaceutical composition, the pharmaceutical composition comprising a therapeutically effective amount of the crystals of the compound of formula (I) described herein, the salt of the compound of formula (I) or its crystals, or its crystalline composition.
  • the present application also provides the crystals of the compound of formula (I), the salt of the compound of formula (I) or its crystals, its crystal form composition, or its pharmaceutical composition in the preparation for treatment or Use in drugs for the prevention of JAK1 and/or JAK2 related diseases.
  • the application also provides the crystals of the compound of formula (I), the salt of the compound of formula (I) or its crystals, its crystal form composition, or its pharmaceutical composition in the treatment or prevention of JAK1 and /or applications in JAK2-associated diseases.
  • the present application also provides the crystals of the compound of formula (I) described in the present application, the salt of the compound of formula (I) or its crystals, and its combination of crystal forms for the treatment or prevention of JAK1 and/or JAK2-related diseases substance, or its pharmaceutical composition.
  • the present application also provides a method for treating or preventing JAK1 and/or JAK2-related diseases, comprising administering a therapeutically effective amount of the compound of formula (I) described in the present application to a mammal (preferably a human) in need of the treatment Crystals, salts of compounds of formula (I) or crystals thereof, crystal composition thereof, or pharmaceutical compositions thereof.
  • the JAK1 and/or JAK2-related diseases are selected from inflammatory disorders (such as arthritis) and the like.
  • the compounds of the present application, their crystals, their salts and the crystals of their salts exhibited good selective inhibition of JAK1 and/or JAK2 in the in vitro activity test of the four Janus kinase subtypes JAK1, JAK2, JAk3 and TYK2;
  • the animal to be tested may have good oral bioavailability, high exposure, good pharmacokinetic properties, and good drug efficacy in vivo.
  • the crystals of the compound of formula (I) of the present application, its salts and the crystals of their salts can exhibit advantages such as stable physical and chemical properties, little influence by light, heat and humidity, good solubility, and less likely to undergo crystal transformation, which is beneficial to drug production.
  • the position of the peak or the relative intensity of the peak may vary due to factors such as measuring instruments, measuring methods/conditions, and the like.
  • the measurement error of the 2 ⁇ value may be ⁇ 0.2°. Therefore, when determining each crystal form, this error should be taken into consideration, and values within the error range also belong to the scope of the present application.
  • the position of the endothermic peak in DSC may vary due to factors such as measuring instruments, measuring methods/conditions, and the like.
  • the error may be ⁇ 5°C, or ⁇ 3°C. Therefore, when determining each crystal form, this error should be taken into consideration, and values within the error range also belong to the scope of the present application.
  • “Pharmaceutically acceptable excipients” refer to the inert substances that are administered together with the active ingredients and are beneficial to the administration of the active ingredients, including but not limited to acceptable drugs for humans or animals approved by the State Food and Drug Administration (such as livestock) any glidants, sweeteners, diluents, preservatives, dyes/colorants, flavor enhancers, surfactants, wetting agents, dispersants, disintegrants, suspending agents, stabilizers, Isotonic agent, solvent or emulsifier.
  • Non-limiting examples of such excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • composition refers to a mixture of one or more compounds of the present application or their salts and pharmaceutically acceptable auxiliary materials.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound of the present application to an organism.
  • the pharmaceutical composition of the present application can be prepared by combining the compound of the present application with suitable pharmaceutically acceptable auxiliary materials, for example, it can be formulated into solid, semi-solid, liquid or gaseous preparations, such as tablets, pills, capsules, powders , granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres and aerosols, etc.
  • Typical routes of administration of the crystals described herein or pharmaceutical compositions thereof include, but are not limited to, oral, rectal, topical, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous Internal administration.
  • the pharmaceutical composition of the present application can be produced by methods well known in the art, such as conventional mixing methods, dissolving methods, granulating methods, dragee-making methods, pulverizing methods, emulsifying methods, freeze-drying methods and the like.
  • the pharmaceutical composition is in oral form.
  • the pharmaceutical compositions can be formulated by mixing the active compounds with pharmaceutically acceptable excipients well known in the art. These excipients enable the compounds of the present application to be formulated into tablets, pills, lozenges, dragees, capsules, liquids, gels, slurries, suspensions, etc. for oral administration to patients.
  • Therapeutic dosages of the compounds of the present application may depend, for example, on the particular use for the treatment, the mode of administration of the compound, the health and state of the patient, and the judgment of the prescribing physician.
  • the ratio or concentration of the compounds of the present application in the pharmaceutical composition may vary, depending on various factors, including dosage, chemical properties (eg, hydrophobicity) and route of administration.
  • treating means administering a compound or formulation described herein to improve or eliminate a disease or one or more symptoms associated with the disease, and includes:
  • prevention means administering a compound or formulation described herein to prevent a disease or one or more symptoms associated with the disease, and includes: preventing a disease or disease state from occurring in a mammal, especially when Such mammals are susceptible to the disease state, but have not been diagnosed as having the disease state.
  • the term "therapeutically effective amount” refers to a non-toxic but sufficient amount of the drug or agent to achieve the desired effect.
  • the determination of the effective amount varies from person to person, depending on the age and general condition of the recipient, and also depends on the specific active substance. The appropriate effective amount in each case can be determined by those skilled in the art according to routine experiments.
  • a therapeutically effective amount of the crystals described herein is from about 0.0001 to 20 mg/Kg body weight/day, such as from 0.001 to 10 mg/Kg body weight/day.
  • the dosage frequency of the crystals described herein is determined by the needs of the individual patient, for example, once or twice a day, or more times a day. Dosing can be intermittent, eg, wherein the patient receives a daily dose of crystals for a period of several days, followed by a period of several or more days in which the patient receives no daily dose of crystals.
  • parameter values should be understood as being modified by the term "about”.
  • the term “about” indicates an error value exists, for example, it means a variation within ⁇ 5%, such as ⁇ 1% or ⁇ 0.1%, of a certain value.
  • DCM dichloromethane
  • DMF N,N-dimethylformamide
  • DMSO dimethylsulfoxide
  • EtOH stands for ethanol
  • MeOH stands for methanol
  • TsOH stands for p-toluenesulfonic acid
  • ATP Represents adenosine triphosphate
  • MEK represents methyl ethyl ketone
  • ACN represents acetonitrile
  • Boc represents tert-butoxycarbonyl
  • TBS represents tert-butyldimethylsilyl
  • Et represents ethyl
  • Ts represents p-toluenesulfonyl
  • PE represents petroleum ether
  • EA or EtOAc represents ethyl acetate
  • ADDP represents azodicarbonyl dipiperidine
  • TBAF represents tetrabutylammonium fluoride
  • DCM dichloromethane
  • THF represents tetrahydrofuran
  • Test method About 10-20 mg of sample is used for XRPD detection.
  • Phototube voltage 40kV
  • phototube current 40mA
  • Test method Take a sample ( ⁇ 1mg) and place it in a DSC aluminum pot for testing. Under the condition of 50mL/min N 2 , heat the sample from 25°C to 300°C at a heating rate of 10°C/min.
  • Test method Take a sample (2-5mg) and place it in a TGA platinum pot for testing. Under the condition of 25mL/min N 2 , heat the sample from room temperature to a weight loss of 20% at a heating rate of 10°C/min.
  • Fig. 1 is the Cu-K ⁇ radiation XRPD spectrum of type A crystal of the compound of formula (I).
  • Fig. 2 is the DSC spectrum of type A crystal of the compound of formula (I).
  • Fig. 3 is the TGA spectrum of type A crystal of the compound of formula (I).
  • Fig. 4 is the Cu-K ⁇ radiation XRPD spectrum of type B crystal of the compound of formula (I).
  • Fig. 5 is a DSC spectrum of type B crystal of the compound of formula (I).
  • Fig. 6 is the TGA spectrum of type B crystal of the compound of formula (I).
  • Fig. 7 is the Cu-K ⁇ radiation XRPD spectrum of type C crystal of the compound of formula (I).
  • Fig. 8 is a DSC spectrum of type C crystal of the compound of formula (I).
  • Fig. 9 is a TGA spectrum of type C crystal of the compound of formula (I).
  • Fig. 10 is the Cu-K ⁇ radiation XRPD spectrum of the type D crystal of the compound of formula (I).
  • Fig. 11 is the TGA spectrum of the D-type crystal of the compound of formula (I).
  • Fig. 12 is the Cu-K ⁇ radiation XRPD spectrum of the crystallization of the phosphate salt of the compound of formula (I).
  • Fig. 13 is a DSC spectrum of the crystallization of the phosphate salt of the compound of formula (I).
  • Fig. 14 is a TGA spectrum of the crystallization of the phosphate salt of the compound of formula (I).
  • Fig. 15 is the Cu-K ⁇ radiation XRPD spectrum of type I crystals of the oxalate salt of the compound of formula (I).
  • Fig. 16 is a DSC spectrum of the type I crystal of the oxalate salt of the compound of formula (I).
  • Fig. 17 is a TGA spectrum of the type I crystal of the oxalate salt of the compound of formula (I).
  • Fig. 18 is the Cu-K ⁇ radiation XRPD spectrum of the oxalate salt type II crystal of the compound of formula (I).
  • Fig. 19 is a DSC spectrum of the type II crystal of the oxalate salt of the compound of formula (I).
  • Fig. 20 is a TGA spectrum of the type II crystal of the oxalate salt of the compound of formula (I).
  • Embodiment 1 the preparation of formula (I) compound
  • Step 1 Under nitrogen protection at -78°C, add n-butyllithium in n-hexyl dropwise to a solution of tert-butyldimethyl(2-propynyloxy)silane (200g, 1174.24mmol) in tetrahydrofuran (2L) alkane solution (2.5M, 427.54mL), and the reaction solution was stirred at -78°C for 30 minutes. Then, a solution of compound 1-1 (250 g, 971.7 mmol) in tetrahydrofuran (2 L) was added dropwise to the reaction solution at -78°C. The reaction solution was reacted at -78°C for 3 hours.
  • Step 2 Add hydrazine hydrate (34.71 g, 1.03 mol, 98%) to a solution of compound 1-2 (400 g, 935.44 mmol) in DMF (3 L) under ice bath. The reaction was carried out at 25°C for 2 hours. LC-MS showed that the reaction was complete, the reaction solution was diluted with water (10L), extracted with EA (2L*2), the combined reaction solution was washed with saturated brine (2L), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain Compounds 1-3. MS (ESI) 442 [M+H] + .
  • Step 3 NaBH 4 (77.71 g, 2.05 mol) was added in portions to a solution of compound 1-3 (432 g, 978.18 mmol) in THF (3 L) under ice-bath conditions. Afterwards, methanol (0.6 L) was slowly added dropwise, and the reaction solution was stirred at 25° C. for 12 hours. LC-MS showed that the reaction was complete.
  • Step 4 To a solution of compound 1-4 (336 g, 840.84 mmol) in tetrahydrofuran (4 L) was added tributylphosphine (340.24 g, 1.68 mol) under ice-bath conditions. The reaction solution was stirred for 30 minutes under ice-bath conditions, and ADDP (424.31 g, 1.68 mol) was added to the reaction solution. The reaction solution was stirred and reacted at 20° C. for 12 hours. LC-MS showed the reaction was complete.
  • Step 5 To a solution of compound 1-5 (390 g, 1.02 mol) in THF (1 L) was added TBAF (1 M, 1.02 L, 1.02 mol) at room temperature, and the reaction was carried out at 20° C. for 1.5 hours. LC-MS showed the reaction was complete.
  • MS (ESI) 268 [M+H] + MS (ESI) 268 [M+H] + .
  • Step 6 Add manganese dioxide (37.40 g, 430.19 mmol) to a solution of compound 1-6 (11.5 g, 43.02 mmol) in DCM (150 mL) and MeOH (15 mL), replace with nitrogen for 3 times, and then Stir at °C for 12 hours. LC-MS showed the reaction was complete. The reaction solution was filtered and concentrated to obtain compound 1-7. MS (ESI) 266 [M+H] + .
  • Step 8 To a solution of compound 1-8 (11 g, 41.94 mmol) in DCM (150 mL) was added TMSI (10.91 g, 54.52 mmol, 7.42 mL) at 0°C. The reaction solution was stirred at 0°C for 1 hour. TLC showed disappearance of starting material and formation of new spots. The reaction solution was concentrated under reduced pressure to obtain the hydroiodide salt of compound 1-9. MS (ESI) 163 [M+H] + .
  • Step 9 To a solution of compound 1-10 (20g, 131.08mmol, 1eq) in DCM (50mL) was added p-toluenesulfonyl chloride (27.49g, 144.19mmol, 1.1eq), DMAP (1.6g, 13.11 mmol, 0.1eq) and triethylamine (19.9g, 196, 62mmol, 27.37mL), the resulting solution was stirred at 25°C for 16 hours, the reaction was complete, the solvent was removed under reduced pressure, and saturated NaHCO 3 solution (50mL) was added, filtered, The filter cake was washed with water and dried to obtain product 1-11. MS (ESI) 307 [M+H] + .
  • Step 10 To a solution of compound 1-11 (10 g, 32.60 mmol, 1 eq) in DCM (50 mL) was added dropwise tetrabutylammonium nitrate (29.78 g, 97.79 mmol, 3 eq) in dichloromethane ( 50mL) solution, and then slowly dropwise added trifluoroacetic anhydride (20.54g, 97.79mmol, 13.60mL, 3eq).
  • Step 11 Add DIEA (26.73g, 206.82mmol) to a solution of compound 1-9 (12g, 41.36mmol, hydroiodide) and compound 1-12 (11.64g, 33.09mmol) in isopropanol (200mL) ,36.0mL). The reaction liquid was replaced with nitrogen three times, and stirred at 90° C. for 12 hours. LC-MS showed the reaction was complete. The reaction solution was cooled, added H 2 O (200 mL), filtered and dried to obtain compound 1-13. MS (ESI) 478 [M+H] + .
  • Step 12 Add Fe (9.36 g, 167.54 mmol) and NH 4 Cl (12.55 g, 234.56 mmol) to a solution of compound 1-13 (16 g, 33.51 mmol) in THF (200 mL) and H 2 O (50 mL) , replaced with nitrogen three times, and stirred at 100° C. for 1 hour. LCMS showed the reaction was complete. The reaction solution was filtered, and the filtrate was diluted with H 2 O (100 mL), and then extracted with ethyl acetate (150 mL*2). The filter cake was washed with DCM:MeOH (20:1, 100 mL*3). The extract and the washing solution of the filter cake were combined, dried over sodium sulfate, filtered, and concentrated to obtain compound 1-14. MS (ESI) 448 [M+H] + .
  • Step 13 To a solution of compound 1-14 (150 mg, 335.19 ⁇ mol) and TsOH (5.8 mg, 33.52 ⁇ mol) in AcOH (5 mL) was added tetramethylorthocarbonate (456.4 mg, 3.35 mmol). The reaction liquid was replaced with nitrogen three times, and stirred at 50° C. for 2 hours. LCMS showed the reaction was complete. The reaction solution was concentrated to remove the solvent, and H 2 O (5 mL) was added to the obtained concentrate to dilute, and extracted with dichloromethane (5 mL*3). The organic phases were combined, washed with saturated brine, dried over sodium sulfate, filtered and concentrated to obtain compound 1-15. MS (ESI) 488 [M+H] + .
  • Step 14 Compound 1-15 (180 mg, 369.21 ⁇ mol) was dissolved in THF (10 mL), then TBAF (1M, 738.4 ⁇ L) was added. The reaction liquid was replaced with nitrogen three times, and stirred at 70° C. for 12 hours. LC-MS showed the reaction was complete. The reaction solution was concentrated, and the resulting concentrate was diluted with aqueous NaHCO 3 (15 mL), extracted with DCM (15 mL*3), and the organic phases were combined, dried over sodium sulfate, filtered, and concentrated to obtain a crude product.
  • JAK2, JAK3 and TYK2 were diluted in a solution prepared as follows: 20 mM 3-(N-morpholine) propanesulfonic acid (MOPS), 1 mM EDTA, 0.01% Brij-35.5% glycerol, 0.1% ⁇ -mercaptoethanol, 1 mg/ mL BSA.
  • JAK1 was diluted in a solution prepared as follows: 20 mM TRIS, 0.2 mM EDTA, 0.1% ⁇ -mercaptoethanol, 0.01% Brij-35.5% glycerol.
  • the compound of formula (I) in Example 1 was prepared as a solution in 100% DMSO at a concentration 50 times that of the subsequent assay.
  • the tested compound of formula (I) was serially diluted 3 times to obtain a total of 9 concentrations ranging from 10 ⁇ M to 0.001 ⁇ M, and the content of DMSO in the detection reaction system was 2%.
  • a working stock solution of this compound was added as the first component of the reaction to the corresponding assay well, followed by the addition of the remaining components following the assay protocol detailed below.
  • JAK1(h) was incubated with 20 mM Tris/HCl pH 7.5, 0.2 mM EDTA, 500 ⁇ M MGEEPLYWSFPAKKK (SEQ ID NO: 1), 10 mM magnesium acetate and [ ⁇ - 33P ]-ATP (activity and concentration as required).
  • the reaction was started by adding Mg/ATP mixture and after 40 min incubation at room temperature, the reaction was stopped by adding phosphoric acid at a concentration of 0.5%. Then 10 ⁇ L of the reactant was spotted on a P30 filter pad and washed three times with 0.425% phosphoric acid and once with methanol within 4 minutes, dried and counted by scintillation.
  • JAK2(h) was incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 100 ⁇ M KTFCGTPEYLAPEVRREPRILSEEEQEM FRDFDYIADWC (SEQ ID NO: 2), 10 mM magnesium acetate and [ ⁇ - 33P ]-ATP (activity and concentration as required).
  • the reaction was started by adding Mg/ATP mixture and after 40 min incubation at room temperature, the reaction was stopped by adding phosphoric acid at a concentration of 0.5%. Then 10 ⁇ L of the reactant was spotted on a P30 filter pad and washed three times with 0.425% phosphoric acid and once with methanol within 4 minutes, dried and counted by scintillation.
  • JAK3(h) was incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 500 ⁇ M GGEEEEYFELVKKKK (SEQ ID NO: 3), 10 mM magnesium acetate and [ ⁇ - 33P ]-ATP (activity and concentration as required).
  • the reaction was started by adding Mg/ATP mixture and after 40 min incubation at room temperature, the reaction was stopped by adding phosphoric acid at a concentration of 0.5%. Then 10 ⁇ L of the reactant was spotted on a P30 filter pad and washed three times with 0.425% phosphoric acid and once with methanol within 4 minutes, dried and counted by scintillation.
  • TYK2(h) were incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 250 ⁇ M GGMEDIYFEFMGGKKK (SEQ ID NO: 4), 10 mM magnesium acetate and [ ⁇ - 33P ]-ATP (activity and concentration as required).
  • the reaction was started by adding Mg/ATP mixture and after 40 min incubation at room temperature, the reaction was stopped by adding phosphoric acid at a concentration of 0.5%. Then 10 ⁇ L of the reactant was spotted on a P30 filter pad and washed three times with 0.425% phosphoric acid and once with methanol within 4 minutes, dried and counted by scintillation.
  • IC 50 results were analyzed using XLFIT5 (formula 205) from IDBS, see Table 8 for details.
  • Example 1 After the compound of formula (I) of Example 1 was dissolved in 5% DMSO and 95% (v:v) of SBE- ⁇ -CD with a mass percent concentration of 12%, the resulting clear solution was injected through the tail vein respectively And intragastrically administered to male SD rats (fasted overnight before administration, 7-8 weeks old). After giving the test compound, at 0.117, 0.333, 1, 2, 4, 7 and 24 hours for intravenous injection group (1mg/kg) rats, for intragastric administration (5mg/kg) rats at 0.25, 0.5, 1 , 2, 4, 8 and 24 hours, blood was collected from the mandibular vein of each rat and centrifuged to obtain plasma.
  • the blood drug concentration was determined by LC-MS/MS method, and the relevant pharmacokinetic parameters were calculated by the non-compartmental model linear logarithmic trapezoidal method using WinNonlin TM Version 6.3 pharmacokinetic software.
  • the test results are as follows:
  • AUC 0-inf Area under the plasma concentration-time curve from time 0 to time extrapolated to infinity.
  • the rat collagen-induced arthritis model was used to verify the effect of the compound of formula (I) on treating arthritis.
  • Lewis rats were immunized, and the day of the first immunization was recorded as day 0, and the subsequent days were marked sequentially.
  • 50 microliters of the prepared collagen emulsion (containing 200 micrograms of CII) was subcutaneously injected into the tail (2-3 centimeters from the base of the tail).
  • the same volume of collagen emulsion was subcutaneously injected into the tail by the same method.
  • Lewis rats in the normal group do not need to be immunized.
  • Example 1 On the 27th day of modeling, the animals after modeling were divided into groups, and the corresponding compound of formula (I) in Example 1 was given, and the rats were given different doses (see Table 11 for specific doses) of 0.5% dissolved in water.
  • the body weight of the rats in the treatment group also recovered in a dose-dependent manner, and when the administration ended on the 14th day, the body weight of the rats in the 3mg/kg BID group returned to the normal group. rat level.
  • Mobile phase A 10mmol/L ammonium formate solution (adjust the pH value to 3.5 with formic acid)
  • each sample was set out for complete exposure, and in addition, two (1.2g each) tape Pack the samples, put each sample into a double-layer medicinal low-density polyethylene bag, each layer of medicinal low-density polyethylene bag is buckled and sealed separately, and then put the medicinal low-density polyethylene bag into a package of dry Put it in an aluminum foil bag of the agent and heat seal it.
  • control samples Under the condition of light (total illuminance 1.2 ⁇ 10 6 Lux ⁇ hr/near ultraviolet 200w ⁇ hr/m 2 ), add 2 copies of control samples.
  • the packaging method of the control samples is the same as that of the light samples, but the surface glass is covered with aluminum film.
  • the above samples were sampled and analyzed at 5 days, 10 days and 30 days respectively.
  • the samples placed under the conditions of light (visible light 1200000Lux, ultraviolet 200W) are fully exposed at room temperature.
  • Each of the crystals described in this application can exhibit the following favorable properties under the conditions of light, high temperature, high humidity, and accelerated test: stable physical and chemical properties, stable content of each single impurity and total impurities, basically unchanged moisture content, and no crystallization. Type transformation, and the hygroscopicity of the crystal form is small.

Abstract

涉及式(Ⅰ)化合物的结晶、其盐及其盐的结晶,以及它们在制备用于治疗或预防与JAKs相关疾病的药物中的应用。

Description

三并杂环类化合物的结晶和盐及其应用
相关申请的引用
本申请要求于2021年05月26日向中华人民共和国国家知识产权局提交的第202110578454.7号中国专利申请的优先权,在此将其全部内容以援引的方式整体并入本文中。
技术领域
本申请涉及式(Ⅰ)化合物的结晶、其盐及其盐的结晶,以及其在制备治疗与JAKs相关疾病的药物中的应用。
背景技术
Janus激酶(JAKs)是一种胞质酪氨酸激酶,可传递细胞因子信号,从膜受体到STAT转录因子。JAK家庭包含四个成员:JAK1、JAK2、JAK3和TYK2。JAK-STAT通路将来自多种细胞因子、生长因子和激素的细胞外信号传导到细胞核,并且负责数千个蛋白质编码基因的表达。JAK-STAT细胞内信号转导服务于干扰素、大多数白介素、以及多种细胞因子和内分泌因子,如EPO、TPO、GH、OSM、LIF、CNTF、GM-CSF和PRL(Vainchenker W.et al.(2008)。
JAK-1、JAK-2和TYK-2在人体各组织细胞中均有表达,JAK-3主要表达于各造血组织细胞中,主要存在于骨髓细胞、胸腺细胞、NK细胞及活化的B淋巴细胞、T淋巴细胞中。JAK1已成为免疫、炎症和癌症等疾病领域的新型靶点。人体中的JAK2基因上的一个碱基突变JAK2V617F,其与骨髓增生性疾病中的真性红细胞增多症(PV)、特发性血小板增多症(ET)、特发性骨髓纤维化(IMF)、慢性粒细胞白血病(CML)等的发生密切相关。JAK3或γc突变都可导致重症联合免疫缺陷。JAK3活性异常表现为T细胞和NK细胞大量减少、B细胞功能丧失,严重影响免疫***等的正常生物学功能。基于其功能特点和特殊的组织分布,JAK3已成为针对免疫***相关疾病极具吸引力的药物靶点。在小鼠中,TYK2功能缺失会引起多种细胞因子受体的信号通路发生缺陷,进而导致病毒感染、抗菌免疫功能下降并增加了肺部感染的可能性等(John J.O’Shea,2004,Nature Reviews Drug Discovery 3,555-564)。不同的JAK家族成员选择性地结合在不同的细胞因子受体上,赋予信号传导特异性,从而发挥不同的生理学作用,这种选择性的作用方式使得JAK抑制剂可以相对特异性地应用于疾病治疗。如IL-2或IL-4受体连同共同的γ链与JAK1和JAK3结合,而具有相同β链的I型受体与JAK2结合。使用gp130(糖蛋白130)的I型受体和由杂二聚体细胞因子激活的I型受体优先结合JAK1/2和TYK2。由激素样细胞因子激活的I型受体结合并激活JAK2激酶。干扰素的II型受体结合JAK1和TYK2,而IL-10细胞因子家族的受体与JAK1/2和TYK2结合。所述细胞因子及其受体与JAK家族成员的各种特异结合引发不同的生理学作用,为不同疾病的治疗提供可能。将JAK1与其它JAKs杂二聚化以转导细胞因子驱动的促炎信号传导。因此,预期抑制JAK1和/或其它JAK对于一系列炎性病症和其它由JAK介导的信号转导驱动的疾病是具有治疗益处的(Daniella M.Schwartz,2017,Nature Reviews Drug Discovery 16,843-862。)
发明内容
一方面,本申请提供式(Ⅰ)化合物或其盐
Figure PCTCN2022095126-appb-000001
在一些实施方案中,所述式(Ⅰ)化合物的盐选自磷酸盐或草酸盐。
另一方面,本申请提供式(Ⅰ)化合物或其盐的结晶。
在一些实施方案中,本申请提供式(Ⅰ)化合物的磷酸盐的结晶或式(Ⅰ)化合物的草酸盐的结晶。
一方面,本申请提供了式(Ⅰ)化合物的A型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.74±0.20°、7.23±0.20°和12.28±0.2°;
Figure PCTCN2022095126-appb-000002
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°、12.28±0.20°和14.46±0.2°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°、12.28±0.2°、14.46±0.2°、14.89±0.2°和21.06±0.2°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°、12.28±0.20°、14.46±0.20°、14.89±0.20°、15.75±0.20°、16.82±0.20°、19.37±0.20°和21.06±0.20°。
本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、8.80±0.20°、9.25±0.20°、12.28±0.20°、14.23±0.20°、14.46±0.20°、14.89±0.20°、15.75±0.20°、16.82±0.20°、17.32±0.20°、17.84±0.20°、19.37±0.20°、21.06±0.20°、22.76±0.20°、24.11±0.20°、26.16±0.20°、26.76±0.20°和27.16±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°、9.64±0.20°、10.50±0.20°、12.28±0.20°和14.46±0.2°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°、9.64±0.20°、10.50±0.20°、12.28±0.2°、14.46±0.2°、14.89±0.2°和21.06±0.2°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°、9.64±0.20°、10.50±0.20°、12.28±0.20°、14.46±0.20°、14.89±0.20°、15.75±0.20°、16.82±0.20°、19.37±0.20°和21.06±0.20°。
本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、8.80±0.20°、9.25±0.20°、9.64±0.20°、10.50±0.20°、12.28±0.20°、14.23±0.20°、14.46±0.20°、14.89±0.20°、15.75±0.20°、16.82±0.20°、17.32±0.20°、17.84±0.20°、19.37±0.20°、21.06±0.20°、22.76±0.20°、24.11±0.20°、26.16±0.20°、26.76±0.20°和27.16±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的A型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7、8、9、10或11个衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°、12.28±0.20°、14.46±0.20°、14.89±0.20°、15.75±0.20°、16.82±0.20°、19.37±0.20°和21.06±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的A型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7或8个衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°,12.28±0.2°、14.46±0.2°、14.89±0.2°和21.06±0.2°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的XRPD图谱如图1所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的XRPD图谱中,其使用Cu Kα辐射的X射线粉末衍射图谱的衍射峰峰位置及相对强度如表1所示:
表1:式(Ⅰ)化合物的A型结晶的XRPD数据
编号 2θ[±0.20°] 相对强度[%] 编号 2θ[±0.20°] 相对强度[%]
1 5.26 19.49 12 16.82 10.67
2 5.74 37.39 13 17.32 6.18
3 7.23 100.00 14 17.84 9.24
4 8.80 7.43 15 19.37 11.38
5 9.25 25.32 16 21.06 16.32
6 10.50 5.16 17 22.76 8.73
7 12.28 47.04 18 24.11 5.60
8 14.23 17.55 19 26.16 9.76
9 14.46 39.79 20 26.76 6.91
10 14.89 11.33 21 27.16 7.08
11 15.75 10.70      
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的差示扫描量热(DSC)图谱在188.8±3℃处有一个放热峰的起始点。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的DSC图谱如图2所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的热重分析(TGA)图谱在200±3℃处失重达3.13%。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的A型结晶的TGA图谱如图3所示。
又一方面,本申请提供一种式(Ⅰ)化合物的A型结晶的制备方法,所述方法包括如下步骤:式(Ⅰ)化合物溶于乙醇中,析出固体。在一些实施方案中,式(Ⅰ)化合物与乙醇混合,通过加热搅拌,使式(Ⅰ)化合物溶液澄清。在一些实施方案中,式(Ⅰ)化合物溶于乙醇中之后,将所得溶液进行冷却,以析出固体。在一些实施方案中,上述方法还包括分离得到固体并随后将固体进行干燥的步骤;任选地,在真空条件下进行干燥。
另一方面,本申请还提供了式(Ⅰ)化合物的B型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.08±0.20°、9.78±0.20°和13.76±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的B型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.08±0.20°、9.78±0.20°、13.76±0.20°、18.96±0.20°和24.34±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的B型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.08±0.20°、9.78±0.20°、13.76±0.20°、15.80±0.20°、18.96±0.20°、19.61±0.20°和24.34±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的B型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.08±0.20°、6.31±0.20°、9.78±0.20°、13.76±0.20°、15.80±0.20°、18.96±0.20°、19.61±0.20°和24.34±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的B型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.08±0.20°、6.31±0.20°、9.78±0.20°、11.75±0.20°、13.76±0.20°、14.17±0.20°、15.80±0.20°、18.96±0.20°、19.61±0.20°、23.87±0.20°和24.34±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的B型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7、8、9、10或11个衍射峰:5.08±0.20°、6.31±0.20°、9.78±0.20°、11.75±0.20°、13.76±0.20°、14.17±0.20°、15.80±0.20°、18.96±0.20°、19.61±0.20°、23.87±0.20°和24.34±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的B型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7或8个衍射峰:5.08±0.20°、6.31±0.20°、9.78±0.20°、13.76±0.20°、15.80±0.20°、18.96±0.20°、19.61±0.20°和24.34±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的B型结晶的XRPD图谱如图4所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的B型结晶的XRPD图谱中,其使用Cu Kα辐射的X射线粉末衍射图谱的衍射峰的峰位置及相对强度如表2所示:
表2:式(Ⅰ)化合物的B型结晶的XRPD数据
编号 2θ[±0.20°] 相对强度[%] 编号 2θ[±0.20°] 相对强度[%]
1 5.08 100.00 7 15.80 5.51
2 6.31 4.73 8 18.96 7.60
3 9.78 46.35 9 19.61 6.83
4 11.75 3.95 10 23.87 5.59
5 13.76 53.39 11 24.34 9.98
6 14.17 9.88      
在本申请的一些实施方案中,所述式(Ⅰ)化合物的B型结晶的差示扫描量热(DSC)图谱在172.9±3℃处有一个吸热峰的起始点。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的B型结晶的DSC图谱如图5所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的B型结晶的热重分析(TGA)图谱在150±3℃处失重达5.18%。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的B型结晶的TGA图谱如图6所示。
又一方面,本申请提供一种式(Ⅰ)化合物的B型结晶的制备方法,所述方法包括如下步骤:式(Ⅰ)化合物与DMF及MEK混合,析出并分离得到固体。
在一些实施方案中,上述析出过程在-20℃~室温条件下进行。
在一些实施方案中,上述方法还包括将分离得到的固体进行干燥的步骤;任选地,在室温条件下干燥。
另一方面,本申请还提供了式(Ⅰ)化合物的C型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:6.21±0.20°、9.04±0.20°和12.38±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的C型结晶的使用Cu Kα辐射的X射线粉末衍射图 谱在下列2θ角处具有衍射峰:6.21±0.20°、9.04±0.20°、11.54±0.20°、12.38±0.20°和23.29±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的C型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:6.21±0.20°、9.04±0.20°、11.54±0.20°、12.38±0.20°、16.27±0.20°、23.29±0.20°和25.62±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的C型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6或7个衍射峰:6.21±0.20°、9.04±0.20°、11.54±0.20°、12.38±0.20°、16.27±0.20°、23.29±0.20°和25.62±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的C型结晶的XRPD图谱如图7所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的C型结晶的XRPD图谱中,其使用Cu Kα辐射的X射线粉末衍射图谱的衍射峰的峰位置及相对强度如表3所示:
表3:式(Ⅰ)化合物的C型结晶的XRPD图谱解析数据
编号 2θ[±0.20°] 相对强度[%] 编号 2θ[±0.20°] 相对强度[%]
1 6.21 100.00 5 16.27 3.54
2 9.04 29.07 6 23.29 4.79
3 11.54 6.28 7 25.62 4.26
4 12.38 18.89      
在本申请的一些实施方案中,所述式(Ⅰ)化合物的C型结晶的差示扫描量热(DSC)图谱在140.2±3℃处有一个吸热峰的起始点。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的C型结晶的DSC图谱如图8所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的C型结晶的热重分析(TGA)图谱在130±3℃处失重达3.10%。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的C型结晶的TGA图谱如图9所示。
又一方面,本申请提供一种式(Ⅰ)化合物的C型结晶的制备方法,所述方法包括如下步骤:将式(Ⅰ)化合物溶于ACN中,降温析出固体。
在本申请的一些实施方案中,上述制备方法中,将式(Ⅰ)化合物溶于ACN中之后,还包括过滤操作;任选地,采用0.45微米PTFE滤头进行所述过滤。
在本申请的一些实施方案中,上述降温指从50℃按照0.1℃/分钟降温至5℃。
在本申请的一些实施方案中,上述方法还包括分离得到固体并随后将固体进行干燥的步骤;任选地,在室温条件下干燥。
另一方面,本申请还提供了式(Ⅰ)化合物的D型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:7.13±0.20°、18.22±0.20°和21.45±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的D型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:7.13±0.20°、10.04±0.20°、11.24±0.20°、15.97±0.20°、18.22±0.20°和21.45±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的D型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:7.13±0.20°、10.04±0.20°、11.24±0.20°、15.97±0.20°、16.94±0.20°、18.22±0.20°、21.45±0.20°和22.71±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的D型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:7.13±0.20°、10.04±0.20°、11.24±0.20°、15.97±0.20°、16.94±0.20°、18.22±0.20°、20.28±0.20°、21.45±0.20°、22.71±0.20°和26.21±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的D型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7、8、9或10个衍射峰:7.13±0.20°、10.04±0.20°、11.24±0.20°、15.97±0.20°、16.94±0.20°、18.22±0.20°、20.28±0.20°、21.45±0.20°、22.71±0.20°和26.21±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的D型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7或8个衍射峰:7.13±0.20°、10.04±0.20°、11.24±0.20°、15.97±0.20°、16.94±0.20°、18.22±0.20°、21.45±0.20°和22.71±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的D型结晶的XRPD图谱如图10所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的D型结晶的XRPD中,其使用Cu Kα辐射的X射线粉末衍射图谱的衍射峰的峰位置及相对强度如表4所示:
表4:式(Ⅰ)化合物的D型结晶的XRPD图谱解析数据
编号 2θ[±0.20°] 相对强度[%] 编号 2θ[±0.20°] 相对强度[%]
1 7.13 100.00 6 18.22 26.78
2 10.04 8.44 7 20.28 7.27
3 11.24 13.76 8 21.45 25.17
4 15.97 12.42 9 22.71 10.84
5 16.94 11.92 10 26.21 8.07
在本申请的一些实施方案中,所述式(Ⅰ)化合物的D型结晶的热重分析(TGA)图谱在140±3℃处失重达1.83%。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的D型结晶的TGA图谱如图11所示。
又一方面,本申请提供一种式(Ⅰ)化合物的D型结晶的制备方法,所述方法包括如下步骤:将式(Ⅰ)化合物与马来酸、丙酮混合,分离固体。
在本申请的一些实施方案中,上述方法还包括将分离得到的固体进行干燥的步骤;任选地,在室温、真空条件下进行干燥。
另一方面,本申请还提供了式(Ⅰ)化合物的磷酸盐。
另一方面,本申请还提供了式(Ⅰ)化合物的磷酸盐的结晶。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐中,式(Ⅰ)化合物与磷酸的分子个数比例为1:(1-1.5);在一些实施方案中,式(Ⅰ)化合物与磷酸的分子个数比例为1:1。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐为式(Ⅱ)化合物,
Figure PCTCN2022095126-appb-000003
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐为式(Ⅰ)化合物的磷酸盐的结晶。
另一方面,本申请还提供了式(Ⅰ)化合物的磷酸盐的结晶,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:7.87±0.20°、16.64±0.20°和20.04±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、16.64±0.20°和20.04±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、16.64±0.20°、20.04±0.20°、21.73±0.20°、26.17±0.20°和28.11±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、15.21±0.20°、16.64±0.20°、20.04±0.20°、21.73±0.20°、22.21±0.20°、25.07±0.20°、26.17±0.20°、26.81±0.20°和28.11±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、15.21±0.20°、16.64±0.20°、20.04±0.20°、21.73±0.20°、22.21±0.20°、23.66±0.20°、25.07±0.20°、26.17±0.20°、26.81±0.20°、27.50±0.20°和28.11±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、15.21±0.20°、16.64±0.20°、20.04±0.20°、21.73±0.20°、22.21±0.20°、23.66±0.20°、25.07±0.20°、25.94±0.20°、26.17±0.20°、26.81±0.20°、27.50±0.20°、28.11±0.20°、29.06±0.20°和29.85±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的磷酸盐的结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7、8、9、10、11、12、13或14个衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、15.21±0.20°、16.64±0.20°、20.04±0.20°、21.73±0.20°、22.21±0.20°、23.66±0.20°、25.07±0.20°、26.17±0.20°、26.81±0.20°、27.50±0.20°和28.11±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的磷酸盐的结晶,其使用Cu Kα辐射的X射线粉末衍射图谱 中,包含选自下述2θ角处的3、4、5、6、7或8个衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、16.64±0.20°、20.04±0.20°、21.73±0.20°、26.17±0.20°和28.11±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的XRPD图谱如图12所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的XRPD图谱中,其使用Cu Kα辐射的X射线粉末衍射图谱的衍射峰的峰位置及相对强度如表5所示:
表5:式(Ⅰ)化合物的磷酸盐的结晶的XRPD数据
编号 2θ[±0.20°] 相对强度[%] 编号 2θ[±0.20°] 相对强度[%]
1 6.69 50.68 11 23.66 10.28
2 7.87 100.00 12 25.07 16.28
3 14.15 43.11 13 25.94 13.52
4 15.21 16.29 14 26.17 22.03
5 16.64 80.67 15 26.81 18.85
6 18.28 4.91 16 27.50 12.14
7 20.04 59.77 17 28.11 44.87
8 21.30 6.15 18 29.06 3.59
9 21.73 20.72 19 29.85 3.19
10 22.21 12.94      
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的差示扫描量热(DSC)图谱在141.9±3.0℃处有一个放热峰的起始点。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的DSC图谱如图13所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的热重分析(TGA)图谱在130±3℃处失重达5.50%。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的磷酸盐的结晶的TGA图谱如图14所示。
又一方面,本申请提供一种式(Ⅰ)化合物的磷酸盐的结晶的制备方法,所述方法包括如下步骤:将式(Ⅰ)化合物与磷酸、EtOH及H 2O混合,分离得到固体。
在本申请的一些实施方案中,上述制备方法包括在所述混合后,在加热条件下进行搅拌;任选地,在加热至40~80℃的条件下进行搅拌;任选地,在加热至50℃的条件下进行搅拌。
在本申请的一些实施方案中,上述方法还包括将分离得到的固体进行干燥的步骤;任选地在室温、真空条件下干燥。
另一方面,本申请还提供了式(Ⅰ)化合物的草酸盐。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐中,式(Ⅰ)化合物与草酸的分子个数比例为1:(1-1.5);在一些实施方案中,所述式(Ⅰ)化合物与草酸的分子个数比例为1:1.1。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐为式(Ⅲ)化合物,
Figure PCTCN2022095126-appb-000004
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐为式(Ⅰ)化合物的草酸盐的结晶,例如式(Ⅰ)化合物的草酸盐的I型结晶或式(Ⅰ)化合物的草酸盐的II型结晶。
另一方面,本申请还提供了式(Ⅰ)化合物的草酸盐的I型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.06±0.20°、12.69±0.20°和15.21±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的I型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.06±0.20°、12.69±0.20°、15.21±0.20°、17.92±0.20°、20.77±0.20°和27.32±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的I型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.06±0.20°、11.28±0.20°、12.69±0.20°、15.21±0.20°、16.48±0.20°、17.92±0.20°、19.52±0.20°、20.77±0.20°、22.90±0.20°和27.32±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的草酸盐的I型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5或6个衍射峰:5.06±0.20°、12.69±0.20°、15.21±0.20°、17.92±0.20°、20.77±0.20°和27.32±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的I型结晶的XRPD图谱如图15所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的I型结晶的XRPD图谱中,其使用Cu Kα辐射的X射线粉末衍射图谱的衍射峰的峰位置及相对强度如表6所示:
表6:式(Ⅰ)化合物的草酸盐的I型结晶的XRPD数据
编号 2θ[±0.20°] 相对强度[%] 编号 2θ[±0.20°] 相对强度[%]
1 5.06 100.00 7 17.92 9.17
2 10.03 2.21 8 19.52 3.45
3 11.28 3.74 9 20.77 6.05
4 12.69 16.97 10 22.90 4.48
5 15.21 40.90 11 27.32 8.78
6 16.48 4.02      
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的I型结晶的DSC图谱如图16所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的I型结晶的热重分析(TGA)图谱在150±3℃处失重达11.44%。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的I型结晶的TGA图谱如图17所示。
又一方面,本申请提供一种式(Ⅰ)化合物的草酸盐的I型结晶的制备方法,所述方法包括如下步骤:将式(Ⅰ)化合物与草酸、丙酮混合,分离固体。
在本申请的一些实施方案中,上述制备方法包括在所述混合后,在加热下进行搅拌;任选地,加热温度为40~80℃;任选地,加热温度为50℃。
在本申请的一些实施方案中,上述方法还包括将分离得到的固体进行干燥的步骤;任选地,在室温、真空条件下进行干燥。
另一方面,本申请还提供了式(Ⅰ)化合物的草酸盐的II型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.44±0.20°、11.75±0.20°和13.77±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的II型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.44±0.20°、10.85±0.20°、11.75±0.20°、13.77±0.20°、16.27±0.20°和27.42±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的II型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.44±0.20°、10.85±0.20°、11.75±0.20°、13.77±0.20°、16.01±0.20°、16.27±0.20°、23.39±0.20°和27.42±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的II型结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.44±0.20°、10.85±0.20°、11.75±0.20°、13.77±0.20°、16.01±0.20°、16.27±0.20°、17.85±0.20°、18.39±0.20°、20.83±0.20°、21.75±0.20°、23.39±0.20°、25.13±0.20°和27.42±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的草酸盐的II型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7、8、9、10、11、12或13个衍射峰:5.44±0.20°、10.85±0.20°、11.75±0.20°、13.77±0.20°、16.01±0.20°、16.27±0.20°、17.85±0.20°、18.39±0.20°、20.83±0.20°、21.75±0.20°、23.39±0.20°、25.13±0.20°和27.42±0.20°。
另一方面,本申请提供了式(Ⅰ)化合物的草酸盐的II型结晶,其使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7或8个衍射峰:5.44±0.20°、10.85±0.20°、11.75±0.20°、13.77±0.20°、16.01±0.20°、16.27±0.20°、23.39±0.20°和27.42±0.20°。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的II型结晶的XRPD图谱如图18所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的II型结晶的XRPD图谱中,其使用Cu Kα辐射的X射线粉末衍射图谱的衍射峰的峰位置及相对强度如表7所示:
表7:式(Ⅰ)化合物的草酸盐的II型结晶的XRPD数据
编号 2θ[±0.20°] 相对强度[%] 编号 2θ[±0.20°] 相对强度[%]
1 5.44 100.00 9 18.39 6.50
2 10.85 15.27 10 20.83 12.17
3 11.75 23.69 11 21.75 12.75
编号 2θ[±0.20°] 相对强度[%] 编号 2θ[±0.20°] 相对强度[%]
4 13.77 33.21 12 23.39 13.01
5 16.01 23.53 13 25.13 8.47
6 16.27 28.41 14 25.61 6.83
7 17.21 5.99 15 27.42 16.16
8 17.85 11.76      
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的II型结晶的DSC图谱如图19所示。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的II型结晶的热重分析(TGA)图谱在150±3℃处失重达7.78%。
在本申请的一些实施方案中,所述式(Ⅰ)化合物的草酸盐的II型结晶的TGA图谱如图20所示。
又一方面,本申请提供一种式(Ⅰ)化合物的草酸盐的II型结晶的制备方法,所述方法包括如下步骤:将式(Ⅰ)化合物与草酸、EtOH及H 2O混合,分离得到固体。
在本申请的一些实施方案中,上述制备方法包括在所述混合后,在加热下进行搅拌;任选地,在40~80℃条件下进行搅拌;任选地,在50℃条件下进行搅拌。
在本申请的一些实施方案中,上述制备方法还包括干燥步骤;任选地,在室温、真空条件下进行干燥。
又一方面,本申请提供了包含本申请所述结晶的晶型组合物,其中,所述结晶占晶型组合物重量的50%以上,较好为80%以上,更好是90%以上,最好是95%以上。
又一方面,本申请提供一种药物组合物,该药物组合物包含治疗有效量的本申请所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶、或其晶型组合物。本申请的药物组合物中可含有或不含有药学上可接受的辅料。此外,本申请的药物组合物可进一步包括一种或多种其它治疗剂。在一些实施方式中,本申请提供一种固态的药物组合物,该药物组合物包含治疗有效量的本申请所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶、或其晶型组合物。
又一方面,本申请还提供了本申请所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶、其晶型组合物、或者其药物组合物在制备用于治疗或预防JAK1和/或JAK2相关疾病的药物中的应用。
又一方面,本申请还提供了本申请所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶、其晶型组合物、或者其药物组合物在治疗或预防JAK1和/或JAK2相关疾病中的应用。
又一方面,本申请还提供了用于治疗或预防JAK1和/或JAK2相关疾病的本申请所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶、其晶型组合物、或者其药物组合物。
又一方面,本申请还提供了治疗或预防JAK1和/或JAK2相关疾病的方法,包括对需要该治疗的哺乳动物(优选人)给予治疗有效量的本申请所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶、其晶型组合物、或者其药物组合物。
本申请中,所述JAK1和/或JAK2相关疾病选自炎性病症(例如关节炎)等。
技术效果
本申请的化合物、其结晶、其盐及其盐的结晶在Janus激酶4个亚型JAK1、JAK2、JAk3和TYK2的体外活性测试中展现了对JAK1和/或JAK2的良好的选择性抑制;在待测动物中可具有良好的口服生物利用度,较高的暴露量,可具有良好的药代动力学性质,及良好的体内药效。且本申请的式(Ⅰ)化合物的结晶、其盐及其盐的结晶可表现出物理化学性质稳定、受光热湿度影响小、溶解性好、不易发生转晶等优势,利于成药。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在含有下列含义。一个特定的短语或术语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文出现商品名时,旨在指代其对应的商品或其活性成分。
需要说明的是,在粉末X-射线衍射光谱中,峰的位置或峰的相对强度可能会因为测定仪器、测定方法/条件等因素而产生差异。对任何特定的晶型,峰的位置可能存在误差,2θ值的测定误差可以为±0.2°。因此,在确定每种晶型时,应该将此误差考虑在内,在误差范围内的值也属于本申请的范围。
需要说明的是,对于同种晶型,DSC的吸热峰出现位置可能会因为测定仪器、测定方法/条件等因素而产生差异。对任何特定的晶型,吸热峰的位置可能存在误差,误差可以为±5℃,可以为±3℃。因此,在确定每种晶型时,应该将此误差考虑在内,在误差范围内的值也属于本申请的范围。
所述词语“包括(comprise)”或“包含(comprise)”及其英文变体例如comprises或comprising应 理解为开放的、非排他性的意义,即“包括但不限于”。
“药学上可接受的辅料”是指与活性成分一同给药的、有利于活性成分给药的惰性物质,包括但不限于国家食品药品监督管理局许可的可接受的用于人或动物(例如家畜)的任何助流剂、增甜剂、稀释剂、防腐剂、染料/着色剂、矫味增强剂、表面活性剂、润湿剂、分散剂、崩解剂、助悬剂、稳定剂、等渗剂、溶剂或乳化剂。所述辅料的非限制性实例包括碳酸钙、磷酸钙、各种糖和各类淀粉、纤维素衍生物、明胶、植物油和聚乙二醇。
术语“药物组合物”是指一种或多种本申请的化合物或其盐与药学上可接受的辅料组成的混合物。药物组合物的目的是有利于对有机体给予本申请的化合物。
本申请的药物组合物可通过将本申请的化合物与适宜的药学上可接受的辅料组合而制备,例如可配制成固态、半固态、液态或气态制剂,如片剂、丸剂、胶囊剂、粉剂、颗粒剂、膏剂、乳剂、悬浮剂、栓剂、注射剂、吸入剂、凝胶剂、微球及气溶胶等。
给予本申请所述结晶或其药物组合物的典型途径包括但不限于口服、直肠、局部、吸入、肠胃外、舌下、***内、鼻内、眼内、腹膜内、肌内、皮下、静脉内给药。
本申请的药物组合物可以采用本领域众所周知的方法制造,如常规的混合法、溶解法、制粒法、制糖衣药丸法、磨细法、乳化法、冷冻干燥法等。
在一些实施方案中,药物组合物是口服形式。对于口服给药,可以通过将活性化合物与本领域熟知的药学上可接受的辅料混合,来配制该药物组合物。这些辅料能使本申请的化合物被配制成片剂、丸剂、锭剂、糖衣剂、胶囊剂、液体、凝胶剂、浆剂、悬浮剂等,用于对患者的口服给药。
本申请化合物的治疗剂量可根据例如以下而定:治疗的具体用途、给予化合物的方式、患者的健康和状态,以及签处方医师的判断。本申请化合物在药用组合物中的比例或浓度可不固定,取决于多种因素,它们包括剂量、化学特性(例如疏水性)和给药途径。术语“治疗”意为将本申请所述化合物或制剂进行给药以改善或消除疾病或与所述疾病相关的一个或多个症状,且包括:
(i)抑制疾病或疾病状态,即遏制其发展;
(ii)缓解疾病或疾病状态,即使该疾病或疾病状态消退。
术语“预防”意为将本申请所述化合物或制剂进行给药以预防疾病或与所述疾病相关的一个或多个症状,且包括:预防疾病或疾病状态在哺乳动物中出现,特别是当这类哺乳动物易患有该疾病状态,但尚未被诊断为已患有该疾病状态时。
针对药物或药理学活性剂而言,术语“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。有效量的确定因人而异,取决于受体的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
本申请所述结晶的治疗有效量为从约0.0001到20mg/Kg体重/天,例如从0.001到10mg/Kg体重/天。
本申请所述结晶的剂量频率由患者个体的需求决定,例如,每天1次或2次,或每天更多次。给药可以是间歇性的,例如,其中在若干天的期间内,患者接受结晶的每日剂量,接着在若干天或更多天的期间,患者不接受结晶的每日剂量。
在本文中,除非上下文另有明确规定,否则单数术语涵盖复数指代物,反之亦然。类似地,除非上下文另有明确指示,词语“或”意在包括“和”。
除非另有说明,在本文中,参数值应当被理解为由术语“约”修饰。当用术语“约”描述本申请的参数时,术语“约”表示存在的误差值,例如表示在某一特定值的±5%、例如±1%或±0.1%的范围内变化。
为了描述和公开的目的,以引用的方式将所有的专利、专利申请和其它已确定的出版物在此明确地并入本文。这些出版物仅因为它们的公开早于本申请的申请日而提供。所有关于这些文件的日期的声明或这些文件的内容的表述是基于申请者可得的信息,并且不构成任何关于这些文件的日期或这些文件的内容的正确性的承认。而且,在任何国家,在本文中对这些出版物的任何引用并不构成关于该出版物成为本领域的公知常识的一部分的认可。
下面会通过实施例具体描述本申请,这些实施例并不意味着对本申请的任何限制。
本申请所使用的所有溶剂是市售的,无需进一步纯化即可使用。
本申请采用下述缩略词:DCM代表二氯甲烷;DMF代表N,N-二甲基甲酰胺;DMSO代表二甲亚砜;EtOH代表乙醇;MeOH代表甲醇;TsOH代表对甲苯磺酸;ATP代表三磷酸腺苷;MEK代表甲基乙基酮;ACN代表乙腈;Boc代表叔丁氧羰基;TBS代表叔丁基二甲基硅基;Et代表乙基;Ts代表对甲苯磺酰基;PE代表石油醚;EA或EtOAc代表乙酸乙酯;ADDP代表偶氮二甲酰二哌啶;TBAF代表四丁基氟 化铵;DCM代表二氯甲烷;THF代表四氢呋喃;TMSI代表三甲基碘硅烷;AcOH代表乙酸。
X-射线粉末衍射(X-ray powder diffraction,XRPD)
仪器型号:布鲁克D8 advance X-射线衍射仪
测试方法:大约10~20mg样品用于XRPD检测。
详细的XRPD参数如下:
光管:Cu,kα,
Figure PCTCN2022095126-appb-000005
光管电压:40kV,光管电流:40mA
发散狭缝:0.60mm
探测器狭缝:10.50mm
防散射狭缝:7.10mm
扫描范围:4-40deg(或3-40deg)
步径:0.02deg
步长:0.12秒
样品盘转速:15rpm
差示扫描量热(Differential Scanning Calorimetry,DSC)
仪器型号:TA Q2000差示扫描量热仪
测试方法:取样品(~1mg)置于DSC铝锅内进行测试,在50mL/min N 2条件下,以10℃/min的升温速率,将样品从25℃加热到300℃。
热重分析(Thermal Gravimetric Analysis,TGA)
仪器型号:TA Q5000IR热重分析仪
测试方法:取样品(2~5mg)置于TGA铂金锅内进行测试,在25mL/min N 2条件下,以10℃/min的升温速率,将样品从室温加热到失重20%。
附图说明
图1为式(Ⅰ)化合物的A型结晶的Cu-Kα辐射的XRPD谱图。
图2为式(Ⅰ)化合物的A型结晶的DSC谱图。
图3为式(Ⅰ)化合物的A型结晶的TGA谱图。
图4为式(Ⅰ)化合物的B型结晶的Cu-Kα辐射的XRPD谱图。
图5为式(Ⅰ)化合物的B型结晶的DSC谱图。
图6为式(Ⅰ)化合物的B型结晶的TGA谱图。
图7为式(Ⅰ)化合物的C型结晶的Cu-Kα辐射的XRPD谱图。
图8为式(Ⅰ)化合物的C型结晶的DSC谱图。
图9为式(Ⅰ)化合物的C型结晶的TGA谱图。
图10为式(Ⅰ)化合物的D型结晶的Cu-Kα辐射的XRPD谱图。
图11为式(Ⅰ)化合物的D型结晶的TGA谱图。
图12为式(Ⅰ)化合物的磷酸盐的结晶的Cu-Kα辐射的XRPD谱图。
图13为式(Ⅰ)化合物的磷酸盐的结晶的DSC谱图。
图14为式(Ⅰ)化合物的磷酸盐的结晶的TGA谱图。
图15为式(Ⅰ)化合物的草酸盐的I型结晶的Cu-Kα辐射的XRPD谱图。
图16为式(Ⅰ)化合物的草酸盐的I型结晶的DSC谱图。
图17为式(Ⅰ)化合物的草酸盐的I型结晶的TGA谱图。
图18为式(Ⅰ)化合物的草酸盐的II型结晶的Cu-Kα辐射的XRPD谱图。
图19为式(Ⅰ)化合物的草酸盐的II型结晶的DSC谱图。
图20为式(Ⅰ)化合物的草酸盐的II型结晶的TGA谱图。
具体实施方式
为了更好的理解本申请的内容,下面结合具体实施例来做进一步的说明,但具体的实施方式并不是对本申请的内容所做的限制。
实施例1:式(Ⅰ)化合物的制备
Figure PCTCN2022095126-appb-000006
步骤1:在-78℃氮气保护下,向溶有叔丁基二甲基(2-丙炔氧基)硅烷(200g,1174.24mmol)的四氢呋喃(2L)溶液中滴加正丁基锂的正己烷溶液(2.5M,427.54mL),反应液在-78℃下搅拌30分钟。然后向-78℃的反应液滴加化合物1-1(250g,971.7mmol)的四氢呋喃(2L)溶液。反应液在-78℃反应3小时。TLC(PE:EA=3:1,体积比)显示反应完全,向反应液中加入饱和氯化铵水溶液(2L)和水(1L)淬灭反应,EA(2L*3)萃取,合并的反应液用饱和食盐水洗涤(2L),无水硫酸钠干燥,过滤并减压浓缩,得到化合物1-2。 1H NMR(400MHz,CDCl 3)δ=5.11(br d,J=7.3Hz,1H),4.48(s,2H),4.35-4.27(m,1H),4.21(q,J=7.2Hz,2H),2.80-2.59(m,2H),2.30-2.13(m,1H),1.98(br dd,J=6.4,14.2Hz,1H),1.55-1.42(s,9H),1.36-1.27(m,3H),0.93(s,9H),0.19-0.07(s,6H)。
步骤2:在冰浴下,向溶有化合物1-2(400g,935.44mmol)的DMF(3L)溶液中加入水合肼(34.71g,1.03mol,98%)。该反应在25℃下反应2小时。LC-MS显示反应完全,该反应液用水(10L)稀释,EA(2L*2)萃取,合并的反应液用饱和食盐水洗涤(2L),无水硫酸钠干燥,过滤并减压浓缩,得到化合物1-3。MS(ESI)442[M+H] +
步骤3:在冰浴条件下,向化合物1-3(432g,978.18mmol)的THF(3L)溶液中分批加入NaBH 4(77.71g,2.05mol)。之后缓慢滴加甲醇(0.6L),反应液在25℃下搅拌反应12小时,LC-MS显示反应完全。在冰浴条件下,向反应液中加入饱和氯化铵水溶液(300mL)淬灭反应,然后用水(2L)稀释,EA(2L*2)萃取,合并的反应液用饱和食盐水洗涤(2L),无水硫酸钠干燥,过滤并减压浓缩,通过硅胶柱层析(DCM:MeOH=50:1~20:1)得到化合物1-4。MS(ESI)400[M+H] +
步骤4:在冰浴条件下,向化合物1-4(336g,840.84mmol)的四氢呋喃(4L)溶液中,加入三丁基膦(340.24g,1.68mol)。该反应液在冰浴条件下搅拌30分钟,向该反应液加入ADDP(424.31g,1.68mol)。反应液在20℃下搅拌反应12小时。LC-MS显示反应完全。反应液用水(2L)稀释,EA(2L*2)萃取,合并的反应液用 饱和食盐水洗涤(1.5L),无水硫酸钠干燥,过滤并减压浓缩,通过硅胶柱层析(PE:EA=20:1 to 2:1,体积比)得到化合物1-5。MS(ESI)382[M+H] +
步骤5:在室温条件下,向溶有化合物1-5(390g,1.02mol)的四氢呋喃(1L)溶液中加入TBAF(1M,1.02L,1.02mol),该反应在20℃下反应1.5小时。LC-MS显示反应完全。反应液用水(1L)稀释,用饱和NaHCO 3水溶液调至pH=8,EA(1L*3)萃取,合并的反应液用饱和食盐水洗涤(1L),无水硫酸钠干燥,过滤并减压浓缩,然后浓缩液用乙酸乙酯(1L)溶解,向该溶液慢慢滴加HCl/EtOAc(4M,200mL),搅拌1小时,有固体生成,过滤,得到化合物1-6。MS(ESI)268[M+H] +
步骤6:向溶有化合物1-6(11.5g,43.02mmol)的DCM(150mL)和MeOH(15mL)溶液中加入二氧化锰(37.40g,430.19mmol),用氮气置换3次,然后在65℃下搅拌12小时。LC-MS显示反应完全。将反应液过滤,浓缩得到化合物1-7。MS(ESI)266[M+H] +
步骤7:向溶有化合物1-7(11g,41.46mmol)的THF(150mL)溶液中加入NH 3·H 2O(51.89g,414.61mmol,57.03mL,28%purity)和I 2(31.57g,124.38mmol),将反应液用氮气置换3次,然后在25℃下搅拌12小时。LC-MS显示反应完全。向反应液中加入饱和亚硫酸钠水溶液淬灭反应,加入20mL水稀释,然后用乙酸乙酯(50mL*2)萃取。合并有机相,经饱和食盐水(30mL)洗涤,硫酸钠干燥,过滤,浓缩得到粗品。粗品经硅胶柱色谱分离(PE:EA=3:1)纯化得到化合物1-8。MS(ESI)263[M+H] +
步骤8:在0℃下,向溶有化合物1-8(11g,41.94mmol)的DCM(150mL)溶液加入TMSI(10.91g,54.52mmol,7.42mL)。该反应液在0℃下搅拌1小时。TLC显示原料消失,有新点生成。将反应液减压浓缩得到化合物1-9的氢碘酸盐。MS(ESI)163[M+H] +
步骤9:在25℃下向化合物1-10(20g,131.08mmol,1eq)的DCM(50mL)溶液中分别加入对甲苯磺酰氯(27.49g,144.19mmol,1.1eq)、DMAP(1.6g,13.11mmol,0.1eq)及三乙胺(19.9g,196,62mmol,27.37mL),所得溶液在25℃下搅拌16小时,反应完全,减压除去溶剂,加入饱和NaHCO 3溶液(50mL),过滤,滤饼水洗,干燥,得产物1-11。MS(ESI)307[M+H] +
步骤10:在-5℃下,向化合物1-11(10g,32.60mmol,1eq)的DCM(50mL)溶液中滴加四丁基硝酸铵(29.78g,97.79mmol,3eq)的二氯甲烷(50mL)溶液,然后缓慢滴加三氟乙酸酐(20.54g,97.79mmol,13.60mL,3eq)。所得溶液在-5℃搅拌30min,随后搅拌16h,反应完全,乙酸乙酯(500mL*3)萃取,合并的有机相依次用水(200mL*2)和食盐水洗(200mL*1),无水硫酸钠干燥、过滤、并减压浓缩,所得浓缩物在二氯甲烷中重结晶得产物1-12。MS(ESI)352[M+H] +
步骤11:向溶有化合物1-9(12g,41.36mmol,氢碘酸盐)、化合物1-12(11.64g,33.09mmol)的异丙醇(200mL)溶液中加入DIEA(26.73g,206.82mmol,36.0mL)。将反应液用氮气置换3次,然后在90℃下搅拌12小时。LC-MS显示反应完全。将反应液冷却,加入H 2O(200mL),过滤,干燥得到化合物1-13。MS(ESI)478[M+H] +
步骤12:向溶有化合物1-13(16g,33.51mmol)的THF(200mL)和H 2O(50mL)溶液中加入Fe(9.36g,167.54mmol)和NH 4Cl(12.55g,234.56mmol),用氮气置换3次,在100℃下搅拌1小时。LCMS显示反应完全。将反应液过滤,滤液用H 2O(100mL)稀释,然后用乙酸乙酯萃取(150mL*2)。滤饼用DCM:MeOH(20:1,100mL*3)洗涤。合并萃取液和洗涤滤饼的洗涤液,经硫酸钠干燥,过滤,浓缩得到化合物1-14。MS(ESI)448[M+H] +
步骤13:向溶有化合物1-14(150mg,335.19μmol)和TsOH(5.8mg,33.52μmol)的AcOH(5mL)溶液中加入原碳酸四甲酯(456.4mg,3.35mmol)。将反应液用氮气置换3次,在50℃下搅拌2小时。LCMS显示反应完全。将反应液浓缩除去溶剂,向所得浓缩物中加入H 2O(5mL)稀释,用二氯甲烷(5mL*3)萃取。合并有机相,经饱和食盐水洗涤,硫酸钠干燥,过滤,浓缩得到化合物1-15。MS(ESI)488[M+H] +
步骤14:将化合物1-15(180mg,369.21μmol)溶于THF(10mL),然后加入TBAF(1M,738.4μL)。将反应液用氮气置换3次,在70℃下搅拌12小时。LC-MS显示反应完全。将反应液浓缩,然后将所得浓缩物用NaHCO 3水溶液(15mL)稀释,用DCM(15mL*3)萃取,合并有机相,经硫酸钠干燥,过滤,浓缩得到粗品。粗品经制备型HPLC(Phenomenex Gemini NX 80*30mm*3μm;流动相:A相为水(10mM NH 4HCO 3)-ACN;B相(ACN)%:20%-50%,9min)纯化,分离得到式(Ⅰ)化合物。MS(ESI)334[M+H] +1H NMR(400MHz,DMSO-d 6)δ=11.72(br s,1H),8.38(br s,1H),7.42(br s,1H),6.85(br s,1H),6.71(br s,1H),5.41(br s,1H),4.69(br s,2H),4.09(br s,3H),3.05(br s,2H),2.68-2.55(m,1H),2.28(br s,1H)。
实施例2
取1克式(I)化合物置于100毫升的玻璃小瓶中,加入40毫升乙醇,加热至回流,在78℃下,搅拌至溶液澄清,冷却至室温,搅拌过夜。过滤得到固体,将固体真空干燥8小时,得式(I)化合物的A型结晶。
实施例3
取19.9毫克式(I)化合物置于5.0毫升的玻璃小瓶中,加入0.2毫升DMF以溶解固体。将该澄清溶液在室温条件下边磁力搅拌(转速约为750转每分钟)边滴加3毫升MEK,将得到的澄清溶液转至室温条件下磁力搅拌过夜后仍澄清,继续转至5℃条件下磁力搅拌过夜后仍澄清,接着转至-20℃条件下磁力搅拌过夜后,得到浑浊样品,离心分离得到固体,将固体转至室温敞口干燥三天,得式(I)化合物的B型结晶。
实施例4
取19.8毫克式(I)化合物置于5.0毫升的玻璃小瓶中,加入2.5毫升ACN,在50℃条件下放置过夜,使用0.45微米PTFE滤头过滤得到澄清溶液。将该澄清溶液从50℃按照0.1℃/分钟降温至5℃后,在5℃下磁力搅拌过夜,得到浑浊样品。离心分离得到固体,将固体转至室温敞口干燥过夜,得式(I)化合物C型结晶。
实施例5
取20.1毫克式(I)化合物置于HPLC玻璃小瓶中,加入8.5毫克马来酸、0.5毫升丙酮得到浑浊样品。将该样品在50℃条件下磁力搅拌两天,样品仍浑浊。离心分离得到固体,将固体转至室温真空干燥约四小时,得式(I)化合物D型结晶。
实施例6
取20.3毫克式(I)化合物置于HPLC玻璃小瓶中,加入3.5微升磷酸、0.5毫升EtOH/H 2O(19:1,v/v)得到浑浊样品。将该样品在50℃条件下磁力搅拌两天,样品仍浑浊。离心分离得到固体,将固体转至室温真空干燥约四小时,得式(I)化合物的磷酸盐的结晶。
实施例7
取20.3毫克式(I)化合物置于HPLC玻璃小瓶中,加入9.3毫克草酸、0.5毫升丙酮得到浑浊样品。将该样品在50℃条件下磁力搅拌两天,样品仍浑浊。离心分离得到固体,将固体转至室温真空干燥约四小时,得式(I)化合物的草酸盐的I型结晶。
实施例8
取20.1毫克式(I)化合物置于HPLC玻璃小瓶中,加入9.6毫克草酸、0.5毫升EtOH/H 2O(19:1,v/v)得到浑浊样品。将该样品在50℃条件下磁力搅拌两天,样品仍浑浊。离心分离得到固体,将固体转至室温真空干燥约四小时,得式(I)化合物的草酸盐的II型结晶。
生物活性测试
实验例1:JAK1、JAK2、JAK3、TYK2激酶体外活性测试
实验材料
重组人源JAK1、JAK2、JAK3、TYK2蛋白酶、主要仪器及试剂均由英国的Eurofins公司提供。
实验方法
JAK2、JAK3和TYK2在由如下配制的溶液中稀释:20mM 3-(N-吗啉)丙磺酸(MOPS),1mM EDTA,0.01%Brij-35.5%甘油,0.1%β-巯基乙醇,1mg/mL BSA。JAK1在由如下配制的溶液中稀释:20mM TRIS,0.2mM EDTA,0.1%β-巯基乙醇,0.01%Brij-35.5%甘油。将实施例1的式(Ⅰ)化合物制备成处于100%DMSO中的溶液并且其浓度达到后续测定时的浓度的50倍。将测试的式(Ⅰ)化合物进行3倍浓度梯度稀释,得到终浓度为10μM到0.001μM共9个浓度,DMSO在检测反应体系中的含量为2%。将该化合物的工作储备液作为反应的第一组分添加到相应的测定孔中,然后按照下面详述的测定方案加入其余组分。
JAK1(h)酶反应
将JAK1(h)与20mM Tris/HCl pH 7.5、0.2mM EDTA、500μM MGEEPLYWSFPAKKK(SEQ ID NO:1)、10mM乙酸镁和[γ- 33P]-ATP(根据需要制定活性和浓度)一起孵育。添加Mg/ATP混合物开始反应,在室温下孵育40分钟后,加入0.5%浓度的磷酸终止反应。然后取10μL反应物点在P30滤垫上并于4分钟内用0.425%磷酸洗涤三次和甲醇洗涤一次,干燥、闪烁计数。
JAK2(h)酶反应
将JAK2(h)与8mM MOPS pH 7.0、0.2mM EDTA、100μM KTFCGTPEYLAPEVRREPRILSEEEQEM FRDFDYIADWC(SEQ ID NO:2)、10mM乙酸镁和[γ- 33P]-ATP(根据需要制定活性和浓度)一起孵育。添加Mg/ATP混合物开始反应,在室温下孵育40分钟后,加入0.5%浓度的磷酸终止反应。然后取10μL反应物点在P30滤垫上并于4分钟内用0.425%磷酸洗涤三次和甲醇洗涤一次,干燥、闪烁计数。
JAK3(h)酶反应
将JAK3(h)与8mM MOPS pH 7.0、0.2mM EDTA、500μM GGEEEEYFELVKKKK(SEQ ID NO:3)、10mM乙酸镁和[γ- 33P]-ATP(根据需要制定活性和浓度)一起孵育。添加Mg/ATP混合物开始反应,在室温下孵育40分钟后,加入0.5%浓度的磷酸终止反应。然后取10μL反应物点在P30滤垫上并于4分钟内用0.425%磷酸洗涤三次和甲醇洗涤一次,干燥、闪烁计数。
TYK2(h)酶反应
将TYK2(h)与8mM MOPS pH 7.0、0.2mM EDTA、250μM GGMEDIYFEFMGGKKK(SEQ ID NO:4)、10mM乙酸镁和[γ- 33P]-ATP(根据需要制定活性和浓度)一起孵育。添加Mg/ATP混合物开始反应,在室温下孵育40分钟后,加入0.5%浓度的磷酸终止反应。然后取10μL反应物点在P30滤垫上并于4分钟内用0.425%磷酸洗涤三次和甲醇洗涤一次,干燥、闪烁计数。
数据分析
IC 50结果使用IDBS公司的XLFIT5(205公式)进行分析得到,具体见表8。
表8.式(Ⅰ)化合物体外筛选试验结果
Figure PCTCN2022095126-appb-000007
实验例2:药代动力学(PK)试验
将实施例1的式(Ⅰ)化合物溶解于5%DMSO和95%(v:v)的质量百分浓度为12%的SBE-β-CD中之后,将得到的澄清溶液分别经尾静脉注射和灌胃给予雄性SD大鼠体内(给予前过夜禁食,7~8周龄)。给予受试化合物后,对于静脉注射组(1mg/kg)大鼠在0.117、0.333、1、2、4、7和24小时,对于灌胃组(5mg/kg)大鼠在0.25、0.5、1、2、4、8和24小时,分别从各大鼠的下颌静脉采血并离心后获得血浆。采用LC-MS/MS法测定血药浓度,使用WinNonlin TM Version 6.3药动学软件,以非房室模型线性对数梯形法计算相关药代动力学参数。测试结果如下:
表9式(Ⅰ)化合物在大鼠中的PK测试结果
PK参数 式(Ⅰ)化合物
T 1/2(hr) 3.65
C max(nM) 10152
AUC 0-inf(nM.hr) 67786
生物利用度(%) 119%
注:T 1/2:半衰期;C max:达峰浓度;
AUC 0-inf:从0时间到外推至无穷大时的血浆浓度-时间曲线下面积。
实验例3:大鼠胶原诱导的关节炎(CIA)的体内药效研究
实验过程:
用大鼠胶原诱导的关节炎模型验证式(Ⅰ)化合物的治疗关节炎的作用。对Lewis大鼠进行免疫,第一次免疫当天记为第0天,随后的天数依序标注。将Lewis大鼠经异氟烷麻醉后,在尾部皮下(距尾根部2-3厘米)注射50微升的制备好的胶原乳剂(包含200微克CII)。第21天,采用同法在尾部皮下注射相同体积的胶原乳剂。正常组的Lewis大鼠无需免疫。在造模第27天将造模后的动物分组,并给予相应的实施例1的式(Ⅰ)化合物,对大鼠分别给予不同剂量(具体剂量见表11)的溶解在处于水中的0.5%HPMC E5、0.5%PVP K30和0.2%SLS的混合溶媒中的式(Ⅰ)化合物,并每天2次、口服给予(每个剂量组的受试动物数为8)。连续给药14天,期间观察大鼠状态,记录足体积肿胀情况并评分,评分标准见表10。
表10.关节炎临床评分标准
分值 临床症状
0 无红斑和红肿
1 跗骨附近或踝关节或跖骨处出现红斑或轻度红肿或一个脚趾处有红斑和红肿
2 踝关节和跖骨处轻微红斑和肿胀,有两个以上脚趾处有红肿和红斑
3 踝、腕关节和跖骨处中度红斑和肿胀
4 踝、腕关节、跖骨和脚趾全部严重红肿
实验结果:
式(Ⅰ)化合物在1和3mg/kg剂量下以BID给予时,以剂量依赖性趋势降低关节炎大鼠的临床评分,与溶媒对照组相比具有显著性差异,到给药第14天结束时,3mg/kg BID组中的大鼠的临床评分降到零(表11)。同时足体积肿胀程度也呈剂量依赖性趋势地降低,到第14天给药结束时,与溶媒对照组相比具有显著性差异,3mg/kg BID组中的大鼠的足体积肿胀降至1.28μL。治疗组(式(Ⅰ)化合物给予组)中的大鼠体重也呈剂量依赖性趋势地恢复,到第14天给药结束时,3mg/kg BID组中的大鼠体重恢复至正常组中的大鼠的水平。
表11大鼠中的CIA的体内药效研究主要参数*
Figure PCTCN2022095126-appb-000008
*注:与溶媒对照组相比,各组P<0.001(双因素方差分析)。
实验例4:稳定性测试
按照如下描述的各方法,对本申请所述的化合物的各结晶进行相关的测定。
有关物质测定HPLC方法色谱条件:
色谱柱:Waters XBridge C18,4.6mm×150mm,3.5μm
流动相A:10mmol/L甲酸铵溶液(用甲酸调节pH值至3.5)
流动相B:100%ACN
流速:0.8ml/min
柱温:40℃
检测波长:230nm。
含量测定HPLC方法色谱条件:
色谱柱:Waters XBridge C18,4.6mm×150mm,3.5μm
流动相:10mmol/L甲酸铵溶液(用甲酸调节pH值至3.5):乙腈(78:22)
流速:0.8ml/min
柱温:40℃
检测波长:220nm。
对映异构体测定HPLC方法色谱条件:
色谱柱:Chiral CD-Ph,4.6mm×250mm,5μm
流动相:0.1%三氟乙酸溶液:乙腈(65:35)
流速:1.5ml/min
柱温:40℃
检测波长:220nm。
样品配制:采用乙腈与水的混合溶剂(乙腈:水=50:50(v/v))溶解样品。
水分测定方法:
采用卡尔费休容量法测定,溶剂:甲醇。
固体稳定性放样方法
考察本申请所述的化合物的各结晶在以下条件放置的稳定性,并在不同的时间点取样检测有关物质、含量、对映异构体和晶型。
准确称重不同结晶置于敞口的扁形称量瓶中,一式3份。
摊成薄薄一层,作为正式供试样品,放置于影响因素试验条件下[40℃、60℃、(25℃,相对湿度75%)、(25℃,相对湿度92.5%)、光照(总照度1.2×10 6Lux·hr/近紫外200w·hr/m 2)],各样品为完全暴露放样,另外,“25℃,相对湿度92.5%”条件下增加两份(每份1.2g)带包装样品,每份样品分别装入双层药用低密度聚乙烯袋,每层药用低密度聚乙烯袋分别扎扣密封,再将药用低密度聚乙烯袋袋子放入已经装有一包干燥剂的铝箔袋中并热封。光照(总照度1.2×10 6Lux·hr/近紫外200w·hr/m 2)条件下增加2份对照样品,对照样品的包装方式与光照样品一致,但在表面皿外面覆盖铝膜。上述样品分别在5天、10天、30天进行取样分析。光照(可见光1200000Lux,紫外200W)条件下放置的样品为室温完全暴露放样。
另取该化合物的不同结晶装入双层药用低密度聚乙烯袋,每层药用低密度聚乙烯袋分别扎扣密封,再将药用低密度聚乙烯袋袋子放入已经装有一包干燥剂的铝箔袋中并热封,作为正式供试样品,放置于加速条件(40℃,相对湿度75%)下、长期试验条件(25℃,相对湿度60%)下和5℃的条件下,分别于1、3、6、9、12、18、24月取样分析,其中加速试验取样点为1、3、6月,长期试验取样点为3、6、9、12、18、24月。并利用动态蒸汽吸附仪考察考察上述晶型的吸湿性数据。
本申请所述的各结晶在光照、高温、高湿、加速试验条件下可表现出如下有利的性质:物理化学性质稳定,各单杂质及总杂质含量稳定,水分含量基本不变,不发生晶型转变,并且晶型的吸湿性小。
本领域技术人员将认识到,本公开的范围并不限于上文描述的各种具体实施方式和实施例,而是能够在不脱离本公开的精神和构思的情况下,进行各种修改、替换、或重新组合,这都落入了本公开的保护范围内。

Claims (15)

  1. 式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(I)化合物的盐为磷酸盐或草酸盐,
    Figure PCTCN2022095126-appb-100001
  2. 根据权利要求1所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(I)化合物的结晶的使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7或8个衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°、12.28±0.2°、14.46±0.2°、14.89±0.2°和21.06±0.2°;
    或者,所述式(I)化合物的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.74±0.20°、7.23±0.20°和12.28±0.2°;
    或者,在下列2θ角处具有衍射峰:5.74±0.20°、5.26±0.20°、7.23±0.20°、9.25±0.20°、12.28±0.20°和14.46±0.2°;
    或者,在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°、12.28±0.2°、14.46±0.2°、14.89±0.2°和21.06±0.2°;
    或者,在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、9.25±0.20°、12.28±0.20°、14.46±0.20°、14.89±0.20°、15.75±0.20°、16.82±0.20°、19.37±0.20°和21.06±0.20°;
    或者,在下列2θ角处具有衍射峰:5.26±0.20°、5.74±0.20°、7.23±0.20°、8.80±0.20°、9.25±0.20°、10.50±0.20°、12.28±0.20°、14.23±0.20°、14.46±0.20°、14.89±0.20°、15.75±0.20°、16.82±0.20°、17.32±0.20°、17.84±0.20°、19.37±0.20°、21.06±0.20°、22.76±0.20°、24.11±0.20°、26.16±0.20°、26.76±0.20°和27.16±0.20°;
    或者,所述式(I)化合物的结晶的XRPD图谱如图1所示。
  3. 根据权利要求1或2所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(I)化合物的结晶的差示扫描量热曲线在188.8±3℃处有一个放热峰的起始点;
    或者,所述式(I)化合物的结晶的DSC图谱如图2所示。
  4. 根据权利要求1所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(I)化合物的结晶的使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7或8个衍射峰:5.08±0.20°、6.31±0.20°、9.78±0.20°、13.76±0.20°、15.80±0.20°、18.96±0.20°、19.61±0.20°和24.34±0.20°;
    或者,所述式(I)化合物的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.08±0.20°、9.78±0.20°、13.76±0.20°;
    或者,在下列2θ角处具有衍射峰:5.08±0.20°、9.78±0.20°、13.76±0.20°、18.96±0.20°和24.34±0.20°;
    或者,在下列2θ角处具有衍射峰:5.08±0.20°、6.31±0.20°、9.78±0.20°、13.76±0.20°、15.80±0.20°、18.96±0.20°、19.61±0.20°、24.34±0.20°;
    或者,在下列2θ角处具有衍射峰:5.08±0.20°、6.31±0.20°、9.78±0.20°、11.75±0.20°、13.76±0.20°、14.17±0.20°、15.80±0.20°、18.96±0.20°、19.61±0.20°、23.87±0.20°和24.34±0.20°;
    或者,所述式(I)化合物的结晶的XRPD图谱如图4所示。
  5. 根据权利要求1或4所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(I)化合物的结晶的差示扫描量热曲线在172.9±3℃处有一个吸热峰的起始点;
    或者,所述式(I)化合物的结晶的DSC图谱如图5所示。
  6. 根据权利要求1所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(I)化合物的结晶的使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6或7个衍射峰:6.21±0.20°、9.04±0.20°、11.54±0.20°、12.38±0.20°、16.27±0.20°、23.29±0.20°和25.62±0.20°;
    或者,所述式(I)化合物的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:6.21±0.20°、9.04±0.20°和12.38±0.20°;
    或者,在下列2θ角处具有衍射峰:6.21±0.20°、9.04±0.20°、11.54±0.20°、12.38±0.20°和23.29±0.20°;
    或者,在下列2θ角处具有衍射峰:6.21±0.20°、9.04±0.20°、11.54±0.20°、12.38±0.20°、16.27±0.20°、23.29±0.20°和25.62±0.20°;
    或者,所述式(I)化合物的结晶的XRPD图谱如图7所示。
  7. 根据权利要求1或6所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(I)化合物的结晶的差示扫描量热曲线在140.2±3℃处有一个吸热峰的起始点;
    或者,所述式(I)化合物的结晶的DSC图谱如图8所示。
  8. 根据权利要求1所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(I)化合物的结晶的使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7或8个衍射峰:7.13±0.20°、10.04±0.20°、11.24±0.20°、15.97±0.20°、16.94±0.20°、18.22±0.20°、21.45±0.20°和22.71±0.20°;或者,所述式(I)化合物的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:7.13±0.20°、18.22±0.20°和21.45±0.20°;
    或者,在下列2θ角处具有衍射峰:7.13±0.20°、10.04±0.20°、11.24±0.20°、15.97±0.20°、18.22±0.20°和21.45±0.20°;
    或者,在下列2θ角处具有衍射峰:7.13±0.20°、10.04±0.20°、11.24±0.20°、15.97±0.20°、16.94±0.20°、18.22±0.20°、21.45±0.20°和22.71±0.20°;
    或者,在下列2θ角处具有衍射峰:7.13±0.20°、10.04±0.20°、11.24±0.20°、15.97±0.20°、16.94±0.20°、18.22±0.20°、20.28±0.20°、21.45±0.20°、22.71±0.20°和26.21±0.20°;
    或者,所述式(I)化合物的结晶的XRPD图谱如图10所示。
  9. 根据权利要求1所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(Ⅰ)化合物的盐为磷酸盐,所述式(Ⅰ)化合物与磷酸的分子个数比例为1:(1-1.5),或者所述式(Ⅰ)化合物与磷酸的分子个数比例为1:1。
  10. 根据权利要求1或9所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(Ⅰ)化合物的盐的结晶为式(Ⅰ)化合物的磷酸盐的结晶,所述磷酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7或8个衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、16.64±0.20°、20.04±0.20°、21.73±0.20°、26.17±0.20°和28.11±0.20°;
    或者,所述磷酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:7.87±0.20°、16.64±0.20°和20.04±0.20°;
    或者,在下列2θ角处具有衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、16.64±0.20°和20.04±0.20°;
    或者,在下列2θ角处具有衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、16.64±0.20°、20.04±0.20°、21.73±0.20°、26.17±0.20°和28.11±0.20°;
    或者,在下列2θ角处具有衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、15.21±0.20°、16.64±0.20°、20.04±0.20°、21.73±0.20°、22.21±0.20°、25.07±0.20°、26.17±0.20°、26.81±0.20°和28.11±0.20°;
    或者,在下列2θ角处具有衍射峰:6.69±0.20°、7.87±0.20°、14.15±0.20°、15.21±0.20°、16.64±0.20°、20.04±0.20°、21.73±0.20°、22.21±0.20°、23.66±0.20°、25.07±0.20°、26.17±0.20°、26.81±0.20°、27.50±0.20°和28.11±0.20°;
    或者,所述磷酸盐的结晶的XRPD图谱如图12所示。
  11. 根据权利要求1或10所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述磷酸盐的结晶的差示扫描量热曲线在141.9±3.0℃处有一个放热峰的起始点;
    或者,所述磷酸盐的结晶的DSC图谱如图13所示。
  12. 根据权利要求1所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(Ⅰ)化合物的盐为草酸盐,所述式(Ⅰ)化合物与草酸的分子个数比例为1:(1-1.5);或者,所述式(Ⅰ)化合物与草酸的分子个数比例为1:1.1。
  13. 根据权利要求1或12所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶,其中,所述式(Ⅰ)化合物的盐的结晶为式(Ⅰ)化合物的草酸盐,所述草酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5或6个衍射峰:5.06±0.20°、12.69±0.20°、15.21±0.20°、17.92±0.20°、20.77±0.20°和27.32±0.20°;
    或者,所述草酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.06±0.20°、12.69±0.20°和15.21±0.20°;
    或者,在下列2θ角处具有衍射峰:5.06±0.20°、12.69±0.20°、15.21±0.20°、17.92±0.20°、20.77±0.20°和27.32±0.20°;
    或者,在下列2θ角处具有衍射峰:5.06±0.20°、11.28±0.20°、12.69±0.20°、15.21±0.20°、16.48±0.20°、17.92±0.20°、19.52±0.20°、20.77±0.20°、22.90±0.20°和27.32±0.20°;
    或者,所述草酸盐的结晶的XRPD图谱如图15所示;
    或者其中,所述式(Ⅰ)化合物的盐的结晶为式(Ⅰ)化合物的草酸盐,所述草酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱中,包含选自下述2θ角处的3、4、5、6、7或8个衍射峰:5.44±0.20°、10.85±0.20°、11.75±0.20°、13.77±0.20°、16.01±0.20°、16.27±0.20°、23.39±0.20°和27.42±0.20°;
    或者,所述草酸盐的结晶的使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有衍射峰:5.44±0.20°、11.75±0.20°和13.77±0.20°;
    或者,在下列2θ角处具有衍射峰:5.44±0.20°、10.85±0.20°、11.75±0.20°、13.77±0.20°、16.27±0.20°和27.42±0.20°;
    或者,在下列2θ角处具有衍射峰:5.44±0.20°、10.85±0.20°、11.75±0.20°、13.77±0.20°、16.01±0.20°、16.27±0.20°、23.39±0.20°和27.42±0.20°;
    或者,在下列2θ角处具有衍射峰:5.44±0.20°、10.85±0.20°、11.75±0.20°、13.77±0.20°、16.01±0.20°、16.27±0.20°、17.85±0.20°、18.39±0.20°、20.83±0.20°、21.75±0.20°、23.39±0.20°、25.13±0.20°和27.42±0.20°;或者,所述草酸盐的结晶的XRPD图谱如图18所示。
  14. 一种药物组合物,所述药物组合物包含治疗有效量的权利要求1-13中任一项所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶。
  15. 用于治疗或预防JAK1和/或JAK2相关疾病的权利要求1-13中任一项所述的式(I)化合物的结晶、式(I)化合物的盐或其结晶、或者权利要求14所述的药物组合物。
PCT/CN2022/095126 2021-05-26 2022-05-26 三并杂环类化合物的结晶和盐及其应用 WO2022247885A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280033492.7A CN117295739A (zh) 2021-05-26 2022-05-26 三并杂环类化合物的结晶和盐及其应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110578454 2021-05-26
CN202110578454.7 2021-05-26

Publications (2)

Publication Number Publication Date
WO2022247885A1 true WO2022247885A1 (zh) 2022-12-01
WO2022247885A9 WO2022247885A9 (zh) 2023-11-02

Family

ID=84228441

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/095126 WO2022247885A1 (zh) 2021-05-26 2022-05-26 三并杂环类化合物的结晶和盐及其应用

Country Status (2)

Country Link
CN (1) CN117295739A (zh)
WO (1) WO2022247885A1 (zh)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102712640A (zh) * 2010-01-12 2012-10-03 弗·哈夫曼-拉罗切有限公司 三环杂环化合物、其组合物和应用方法
WO2013007765A1 (en) * 2011-07-13 2013-01-17 F. Hoffmann-La Roche Ag Fused tricyclic compounds for use as inhibitors of janus kinases
WO2020244614A1 (zh) * 2019-06-05 2020-12-10 南京明德新药研发有限公司 吡咯并嘧啶类化合物及其应用
WO2021104488A1 (zh) * 2019-11-27 2021-06-03 正大天晴药业集团股份有限公司 作为jak抑制剂的三并杂环类化合物及其应用

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102712640A (zh) * 2010-01-12 2012-10-03 弗·哈夫曼-拉罗切有限公司 三环杂环化合物、其组合物和应用方法
WO2013007765A1 (en) * 2011-07-13 2013-01-17 F. Hoffmann-La Roche Ag Fused tricyclic compounds for use as inhibitors of janus kinases
WO2020244614A1 (zh) * 2019-06-05 2020-12-10 南京明德新药研发有限公司 吡咯并嘧啶类化合物及其应用
WO2021104488A1 (zh) * 2019-11-27 2021-06-03 正大天晴药业集团股份有限公司 作为jak抑制剂的三并杂环类化合物及其应用

Also Published As

Publication number Publication date
WO2022247885A9 (zh) 2023-11-02
CN117295739A (zh) 2023-12-26

Similar Documents

Publication Publication Date Title
AU2019262144B2 (en) RIP1 inhibitory compounds and methods for making and using the same
CN107667108B (zh) 作为jak激酶抑制剂的萘啶化合物
US9809572B2 (en) Deuterated diaminopyrimidine compounds and pharmaceutical compositions comprising such compounds
RU2633694C2 (ru) Дейтерированный фениламинопиримидин и фармацевтическая композиция, содержащая такое соединение
KR102090453B1 (ko) 상피 성장 인자 수용체 키나제 억제제의 염
KR20170139073A (ko) B-raf 키나아제 억제제의 말레에이트 염, 결정질 형태, 제조방법, 및 이의 용도
US10550101B2 (en) Crystalline forms of mesylate salt of pyridinyl amino pyrimidine derivative, preparation methods therefor, and applications thereof
CN109689641B (zh) 一种取代的2-氢-吡唑衍生物的晶型、盐型及其制备方法
WO2017092413A1 (zh) 一种二氨基嘧啶化合物及包含该化合物的组合物
CN108349974A (zh) 一种取代的吡啶酰胺类化合物及其应用
WO2021104488A1 (zh) 作为jak抑制剂的三并杂环类化合物及其应用
CN115197208A (zh) 杂芳基类化合物、其制备方法及其在医药上的应用
WO2021143843A1 (zh) 一种pde3/pde4双重抑制剂的结晶及其应用
JP7464613B2 (ja) ジアリールチオヒダントイン化合物結晶
WO2022247885A1 (zh) 三并杂环类化合物的结晶和盐及其应用
WO2023193563A1 (zh) 一种噻吩并吡啶化合物的晶型a、制备方法及其药物组合物
CN108456214B (zh) 含噁唑或咪唑结构的喹唑啉类化合物及其应用
CN115038701B (zh) Jak抑制剂的晶型及其应用
CN108137614B (zh) 一种稠合嘧啶化合物及包含该化合物的组合物及其用途
WO2020147838A1 (zh) 一种egfr抑制剂的盐、晶型及其制备方法
CN117247382A (zh) 吡啶并嘧啶酮化合物的晶型
WO2022237808A1 (zh) 吡咯并嘧啶类化合物的晶型及其制备方法
CN108299419B (zh) 一种新型egfr激酶抑制剂的几种新晶型及其制备方法
TW202313014A (zh) 作為pd-l1相互作用的免疫調節劑的雜環化合物
WO2022135555A1 (zh) 嘌呤酮类化合物、其制备方法及其在医药上的应用

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: 22810609

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE