WO2022083733A1 - 固体形式的布鲁顿酪氨酸激酶抑制剂化合物及其用途 - Google Patents

固体形式的布鲁顿酪氨酸激酶抑制剂化合物及其用途 Download PDF

Info

Publication number
WO2022083733A1
WO2022083733A1 PCT/CN2021/125689 CN2021125689W WO2022083733A1 WO 2022083733 A1 WO2022083733 A1 WO 2022083733A1 CN 2021125689 W CN2021125689 W CN 2021125689W WO 2022083733 A1 WO2022083733 A1 WO 2022083733A1
Authority
WO
WIPO (PCT)
Prior art keywords
ray powder
formula
powder diffraction
diffraction pattern
angles
Prior art date
Application number
PCT/CN2021/125689
Other languages
English (en)
French (fr)
Inventor
曲凡志
张翱
丁健
门利玲
杨汉煜
孙静
Original Assignee
上海润石医药科技有限公司
中国科学院上海药物研究所
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 上海润石医药科技有限公司, 中国科学院上海药物研究所 filed Critical 上海润石医药科技有限公司
Publication of WO2022083733A1 publication Critical patent/WO2022083733A1/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the present invention relates to the compound represented by formula (A), its solvate or hydrate solid form, crystal form, specific crystal form and pharmaceutical composition as small molecule Bruton's tyrosine kinase inhibitor, and also relates to the compound in the The application in the preparation of medicines for treating Bruton's tyrosine kinase-related disorders.
  • Bruton's tyrosine kinase is a non-receptor tyrosine kinase belonging to the TEC tyrosine kinase family.
  • TEC family members include Tec, Bmx, BTK, Itk and Txk.
  • BTK is the most widely studied member of the TEC family and is a key regulator of the B cell receptor (BCR) signaling pathway. It is widely expressed in various types of malignant hematological tumors and is involved in the proliferation, differentiation and apoptosis of B cells. Therefore, BTK has become an important molecular target for the drug treatment of malignant hematological tumors.
  • Ibrutinib (trade name Imbruvica) is the first BTK small molecule inhibitor to be marketed and belongs to the first generation of BTK inhibitors. It was approved by the US FDA in 2013 for Clinical treatment of Mantle Cell Lymphoma (MCL) and Chronic Lymphocytic Leukemia (CLL), etc. Since then, ibrutinib has continued to expand its indications.
  • MCL Mantle Cell Lymphoma
  • CLL Chronic Lymphocytic Leukemia
  • the currently approved indications also include 17p deletion chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/Small Lymphocytic Lymphoma, SLL), Waldenstrom Macroglobulinemia (WM), marginal zone lymphoma (Marginal Zone Lymphoma, MZL), chronic graft-resistant Host disease (chronic Graft-Versus-Host Disease, cGVHD).
  • Ibrutinib can form covalent binding with cysteine 481 (Cys481) in the ATP-binding domain of BTK kinase, irreversibly inhibit BTK activation, block BTK signaling pathway, thereby inhibiting the proliferation and survival of B lymphoma cells. achieve the purpose of tumor treatment.
  • Ibrutinib has achieved substantial efficacy in clinical treatment. However, due to the poor target selectivity of ibrutinib, there are certain toxic and side effects in clinical practice.
  • Acalabrutinib (ACP-196, trade name Calquence) was approved for the treatment of MCL and CLL in 2017 and belongs to the second generation of BTK targeted drugs. Compared with ibrutinib, acaltinib is more selective for BTK and has lower off-target toxicity.
  • Zanubrutinib Zanubrutinib (Zabrutinib, BGB-3111), developed by BeiGene Biotechnology Co., Ltd., was approved by the FDA in November 2019 for the treatment of adult MCL patients, becoming the first Chinese local antibody to receive FDA breakthrough therapy designation.
  • the first-generation inhibitors have high inhibitory activity against BTK, but the target selection and bioavailability are poor; the second-generation inhibitors have good selectivity, but the inhibition rate of BTK is lower than that of the first-generation inhibitors.
  • the core skeleton of the compounds currently on the market or under research is mainly a bicyclic system.
  • Compound A has excellent BTK inhibitory activity and maintains good BTK inhibitory selectivity, excellent in vivo antitumor activity, good oral administration performance and good metabolic stability, and has the potential to be developed as a BTK selective inhibitor .
  • the present invention provides a compound of formula (A), a solvate or a hydrate thereof in solid form.
  • the present invention provides a compound of formula (A), a solvate or a hydrate thereof in crystalline form.
  • the above-mentioned crystalline form is characterized in that the crystalline form is a solvent-free and anhydrous crystalline form or a hydrated crystalline form, preferably a solvent-free and anhydrous crystalline form.
  • the present invention provides the compound represented by formula (A), its solvate or hydrate crystal form I, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 13.6 ⁇ 0.2°.
  • the above-mentioned crystalline form I its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 13.6 ⁇ 0.2°, 23.4 ⁇ 0.2°.
  • the above-mentioned crystal form I its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 13.6 ⁇ 0.2°, 18.7 ⁇ 0.2°, 23.4 ⁇ 0.2°. .
  • the above-mentioned crystal form I its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 13.6 ⁇ 0.2°, 16.2 ⁇ 0.2°, 18.7 ⁇ 0.2°, 23.4 ⁇ 0.2°.
  • the above-mentioned crystal form I its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 13.6 ⁇ 0.2°, 16.2 ⁇ 0.2°, 18.7 ⁇ 0.2°, 23.4 ⁇ 0.2°, 26.1 ⁇ 0.2°.
  • the above-mentioned crystal form I its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 13.6 ⁇ 0.2°, 16.2 ⁇ 0.2°, 18.7 ⁇ 0.2°, 23.4 ⁇ 0.2°, 24.2 ⁇ 0.2°, 24.8 ⁇ 0.2°, 26.1 ⁇ 0.2°.
  • the above-mentioned crystal form I its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 13.6 ⁇ 0.2°, 16.2 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.4 ⁇ 0.2°, 21.7 ⁇ 0.2°, 23.4 ⁇ 0.2°, 24.2 ⁇ 0.2°, 24.8 ⁇ 0.2°, 26.1 ⁇ 0.2°.
  • the above-mentioned crystal form I its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 13.6 ⁇ 0.2°, 16.2 ⁇ 0.2°, 16.7 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.4 ⁇ 0.2°, 21.7 ⁇ 0.2°, 23.4 ⁇ 0.2°, 24.2 ⁇ 0.2°, 24.8 ⁇ 0.2°, 26.1 ⁇ 0.2°, 27.6 ⁇ 0.2°.
  • the above-mentioned crystal form I its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 13.6 ⁇ 0.2°, 16.2 ⁇ 0.2°, 16.7 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.4 ⁇ 0.2°, 20.8 ⁇ 0.2°, 21.7 ⁇ 0.2°, 23.4 ⁇ 0.2°, 24.2 ⁇ 0.2°, 24.8 ⁇ 0.2°, 26.1 ⁇ 0.2°, 27.6 ⁇ 0.2°.
  • the above-mentioned crystal form I its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 13.6 ⁇ 0.2°, 16.2 ⁇ 0.2°, 16.7 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.4 ⁇ 0.2°, 20.8 ⁇ 0.2°, 21.7 ⁇ 0.2°, 22.5 ⁇ 0.2°, 23.4 ⁇ 0.2°, 24.2 ⁇ 0.2°, 24.8 ⁇ 0.2°, 26.1 ⁇ 0.2°, 27.6 ⁇ 0.2°, 30.4 ⁇ 0.2°.
  • the above-mentioned crystal form I its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystal form I its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystalline form I has an X-ray powder diffraction pattern substantially as shown in FIG. 3 .
  • the above-mentioned crystal form I the differential scanning calorimetry curve has an exothermic peak onset at 272.89 ⁇ 3°C.
  • the above-mentioned crystal form I has an exothermic peak at 274.74 ⁇ 3° C. in the differential scanning calorimetry curve.
  • the above-mentioned crystalline form I has a DSC pattern substantially as shown in FIG. 4 .
  • the present invention provides the compound represented by formula (A), its solvate or hydrate crystal form II, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 8.5 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.0 ⁇ 0.2°.
  • the above-mentioned crystal form II its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 7.0 ⁇ 0.2°, 8.5 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.0 ⁇ 0.2°, 22.1 ⁇ 0.2°.
  • the above-mentioned crystal form II its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 7.0 ⁇ 0.2°, 7.9 ⁇ 0.2°, 8.5 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.0 ⁇ 0.2°, 22.1 ⁇ 0.2°, 25.3 ⁇ 0.2°.
  • the above-mentioned crystal form II its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 7.0 ⁇ 0.2°, 7.9 ⁇ 0.2°, 8.5 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.0 ⁇ 0.2°, 18.0 ⁇ 0.2°, 22.1 ⁇ 0.2°, 25.3 ⁇ 0.2°, 26.3 ⁇ 0.2°.
  • the above-mentioned crystal form II its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 7.0 ⁇ 0.2°, 7.9 ⁇ 0.2°, 8.5 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.0 ⁇ 0.2°, 17.1 ⁇ 0.2°, 18.0 ⁇ 0.2°, 19.1 ⁇ 0.2°, 22.1 ⁇ 0.2°, 25.3 ⁇ 0.2°, 26.3 ⁇ 0.2°.
  • the above-mentioned crystal form II its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 7.0 ⁇ 0.2°, 7.9 ⁇ 0.2°, 8.5 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.0 ⁇ 0.2°, 17.1 ⁇ 0.2°, 18.0 ⁇ 0.2°, 19.1 ⁇ 0.2°, 22.1 ⁇ 0.2°, 25.3 ⁇ 0.2°, 26.3 ⁇ 0.2°.
  • the above-mentioned crystal form II its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 7.0 ⁇ 0.2°, 7.9 ⁇ 0.2°, 8.5 ⁇ 0.2°, 10.6 ⁇ 0.2°, 12.8 ⁇ 0.2°, 15.0 ⁇ 0.2°, 17.1 ⁇ 0.2°, 18.0 ⁇ 0.2°, 19.1 ⁇ 0.2°, 21.3 ⁇ 0.2°, 22.1 ⁇ 0.2°, 25.3 ⁇ 0.2°, 26.3 ⁇ 0.2°.
  • the above-mentioned crystal form II its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 7.0 ⁇ 0.2°, 7.9 ⁇ 0.2°, 8.5 ⁇ 0.2°, 10.6 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 14.1 ⁇ 0.2°, 15.0 ⁇ 0.2°, 17.1 ⁇ 0.2°, 18.0 ⁇ 0.2°, 19.1 ⁇ 0.2°, 21.3 ⁇ 0.2°, 22.1 ⁇ 0.2°, 25.3 ⁇ 0.2°, 26.3 ⁇ 0.2°.
  • the above-mentioned crystal form II its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystal form II its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystalline form II has an X-ray powder diffraction pattern substantially as shown in FIG. 5 .
  • the differential scanning calorimetry curve of the differential scanning calorimetry curve has three onset points of endothermic peaks at 49.26 ⁇ 3°C, 84.59 ⁇ 3°C and 168.35 ⁇ 3°C, respectively.
  • the above-mentioned crystal form II has three endothermic peaks at 69.36 ⁇ 3°C, 98.45 ⁇ 3°C and 176.96 ⁇ 3°C respectively in the differential scanning calorimetry curve.
  • the above-mentioned crystalline form II has a DSC pattern substantially as shown in FIG. 6 .
  • the above-mentioned crystal form II has a weight loss of 3.417% ⁇ 0.2% in the thermogravimetric analysis curve between room temperature and 100 ⁇ 3°C.
  • the above-mentioned crystalline form II has a TGA pattern substantially as shown in FIG. 6 .
  • the present invention provides the compound represented by formula (A), its solvate or hydrate crystal form III, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.1 ⁇ 0.2°.
  • the above-mentioned crystal form III its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.1 ⁇ 0.2°, 25.5 ⁇ 0.2°.
  • the above-mentioned crystal form III its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.0 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.1 ⁇ 0.2°, 22.3 ⁇ 0.2°, 25.5 ⁇ 0.2°.
  • the present invention provides the compound represented by formula (A), its solvate or hydrate crystal form III, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.1 ⁇ 0.2°, 16.2 ⁇ 0.2°.
  • the above-mentioned crystal form III its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.1 ⁇ 0.2°, 16.2 ⁇ 0.2°, 25.5 ⁇ 0.2°.
  • the above-mentioned crystal form III its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.0 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.1 ⁇ 0.2°, 16.2 ⁇ 0.2°, 22.3 ⁇ 0.2°, 25.5 ⁇ 0.2°.
  • the above-mentioned crystal form III its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.0 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.1 ⁇ 0.2°, 16.2 ⁇ 0.2°, 19.3 ⁇ 0.2°, 22.3 ⁇ 0.2°, 25.5 ⁇ 0.2°.
  • the above-mentioned crystal form III its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.0 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.1 ⁇ 0.2°, 16.2 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.1 ⁇ 0.2°, 19.3 ⁇ 0.2°, 22.3 ⁇ 0.2°, 25.5 ⁇ 0.2°.
  • the above-mentioned crystal form III its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.3 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.0 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.6 ⁇ 0.2°, 15.1 ⁇ 0.2°, 16.2 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.1 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.4 ⁇ 0.2°, 22.3 ⁇ 0.2°, 25.5 ⁇ 0.2°.
  • the above-mentioned crystal form III its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystal form III its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystalline form III has an X-ray powder diffraction pattern substantially as shown in FIG. 7 .
  • the differential scanning calorimetry curve of the differential scanning calorimetry curve has three onset points of endothermic peaks at 38.13 ⁇ 3°C, 75.07 ⁇ 3°C and 167.19 ⁇ 3°C, respectively.
  • the above-mentioned crystalline form III has three endothermic peaks at 57.40 ⁇ 3°C, 96.32 ⁇ 3°C and 177.55 ⁇ 3°C in the differential scanning calorimetry curve, respectively.
  • the above-mentioned crystalline form III has a DSC pattern substantially as shown in 8.
  • thermogravimetric analysis curve has a weight loss of 3.075% ⁇ 0.2% before 90 ⁇ 3°C.
  • the above-mentioned crystalline form III has a TGA pattern substantially as shown in FIG. 8 .
  • the present invention provides the compound represented by formula (A), its solvate or hydrate crystal form IV, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 6.3 ⁇ 0.2°, 9.7 ⁇ 0.2°, 12.8 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form IV has characteristic diffraction peaks at the following 2 ⁇ angles: 5.1 ⁇ 0.2°, 6.3 ⁇ 0.2°, 9.7 ⁇ 0.2°, 12.8 ⁇ 0.2°, 14.1 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form IV has characteristic diffraction peaks at the following 2 ⁇ angles: 5.1 ⁇ 0.2°, 6.3 ⁇ 0.2°, 9.7 ⁇ 0.2°, 12.8 ⁇ 0.2°, 14.1 ⁇ 0.2°, 19.0 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form IV has characteristic diffraction peaks at the following 2 ⁇ angles: 5.1 ⁇ 0.2°, 6.3 ⁇ 0.2°, 9.7 ⁇ 0.2°, 12.8 ⁇ 0.2°, 14.1 ⁇ 0.2°, 15.6 ⁇ 0.2°, 19.0 ⁇ 0.2°, 21.9 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form IV has characteristic diffraction peaks at the following 2 ⁇ angles: 5.1 ⁇ 0.2°, 6.3 ⁇ 0.2°, 9.7 ⁇ 0.2°, 12.8 ⁇ 0.2°, 14.1 ⁇ 0.2°, 19.0 ⁇ 0.2°, 20.4 ⁇ 0.2°, 23.2 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form IV has characteristic diffraction peaks at the following 2 ⁇ angles: 5.1 ⁇ 0.2°, 6.3 ⁇ 0.2°, 9.7 ⁇ 0.2°, 12.8 ⁇ 0.2°, 14.1 ⁇ 0.2°, 19.0 ⁇ 0.2°, 20.4 ⁇ 0.2°, 21.9 ⁇ 0.2°, 23.2 ⁇ 0.2°, 26.3 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form IV has characteristic diffraction peaks at the following 2 ⁇ angles: 5.1 ⁇ 0.2°, 6.3 ⁇ 0.2°, 9.7 ⁇ 0.2°, 12.8 ⁇ 0.2°, 14.1 ⁇ 0.2°, 15.6 ⁇ 0.2°, 19.0 ⁇ 0.2°, 20.4 ⁇ 0.2°, 21.9 ⁇ 0.2°, 23.2 ⁇ 0.2°, 26.3 ⁇ 0.2°.
  • the above-mentioned crystal form IV its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystal form IV its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystalline form IV has an X-ray powder diffraction pattern substantially as shown in FIG. 9 .
  • the differential scanning calorimetry curve of the differential scanning calorimetry curve has two onset points of endothermic peaks at 45.75 ⁇ 3°C and 159.89 ⁇ 3°C, respectively.
  • the above-mentioned crystal form IV has two endothermic peaks at 70.65 ⁇ 3°C and 166.96 ⁇ 3°C in the differential scanning calorimetry curve, respectively.
  • the above-mentioned crystalline form IV has a DSC pattern substantially as shown in FIG. 10 .
  • the above-mentioned crystal form IV has a weight loss of 7.863% ⁇ 0.2% in the thermogravimetric analysis curve between room temperature and 200 ⁇ 3°C.
  • the above-mentioned crystalline form IV has a TGA pattern substantially as shown in FIG. 10 .
  • the present invention provides the compound represented by formula (A), its solvate or hydrate crystal form V, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 5.6 ⁇ 0.2°, 7.1 ⁇ 0.2°, 14.3 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form V has characteristic diffraction peaks at the following 2 ⁇ angles: 5.6 ⁇ 0.2°, 7.1 ⁇ 0.2°, 11.5 ⁇ 0.2°, 13.8 ⁇ 0.2°, 14.3 ⁇ 0.2°.
  • the above-mentioned crystal form V its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 5.6 ⁇ 0.2°, 7.1 ⁇ 0.2°, 11.5 ⁇ 0.2°, 13.8 ⁇ 0.2°, 14.3 ⁇ 0.2°, 17.0 ⁇ 0.2°, 21.9 ⁇ 0.2°.
  • the above-mentioned crystalline form V its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 5.6 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.9 ⁇ 0.2°, 11.5 ⁇ 0.2°, 13.8 ⁇ 0.2°, 14.3 ⁇ 0.2°, 17.0 ⁇ 0.2°, 19.1 ⁇ 0.2°, 21.9 ⁇ 0.2°.
  • the above-mentioned crystalline form V its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 5.6 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.9 ⁇ 0.2°, 11.5 ⁇ 0.2°, 13.8 ⁇ 0.2°, 14.3 ⁇ 0.2°, 17.0 ⁇ 0.2°, 18.3 ⁇ 0.2°, 19.1 ⁇ 0.2°, 20.7 ⁇ 0.2°, 21.9 ⁇ 0.2°.
  • the above-mentioned crystalline form V its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 5.6 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.9 ⁇ 0.2°, 11.5 ⁇ 0.2°, 13.8 ⁇ 0.2°, 14.3 ⁇ 0.2°, 17.0 ⁇ 0.2°, 18.3 ⁇ 0.2°, 19.1 ⁇ 0.2°, 20.7 ⁇ 0.2°, 21.9 ⁇ 0.2°, 25.2 ⁇ 0.2°.
  • the above-mentioned crystalline form V its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 5.6 ⁇ 0.2°, 7.1 ⁇ 0.2°, 8.9 ⁇ 0.2°, 11.5 ⁇ 0.2°, 13.8 ⁇ 0.2°, 14.3 ⁇ 0.2°, 17.0 ⁇ 0.2°, 18.3 ⁇ 0.2°, 19.1 ⁇ 0.2°, 20.7 ⁇ 0.2°, 21.9 ⁇ 0.2°, 25.2 ⁇ 0.2°, 27.2 ⁇ 0.2°.
  • the above-mentioned crystal form V its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystal form V its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystalline form V has an X-ray powder diffraction pattern substantially as shown in FIG. 11 .
  • the above-mentioned crystal form V has an endothermic peak at 174.53 ⁇ 3°C in its differential scanning calorimetry curve.
  • the above-mentioned crystalline form V has a DSC pattern substantially as shown in FIG. 15 .
  • the present invention provides the compound represented by formula (A), its solvate or hydrate crystal form VI, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 6.2 ⁇ 0.2°, 9.4 ⁇ 0.2°, 12.5 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form VI has characteristic diffraction peaks at the following 2 ⁇ angles: 6.2 ⁇ 0.2°, 9.4 ⁇ 0.2°, 12.5 ⁇ 0.2°, 15.2 ⁇ 0.2°, 21.4 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form VI has characteristic diffraction peaks at the following 2 ⁇ angles: 5.0 ⁇ 0.2°, 6.2 ⁇ 0.2°, 9.4 ⁇ 0.2°, 12.5 ⁇ 0.2°, 15.2 ⁇ 0.2°, 21.4 ⁇ 0.2°, 24.7 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form VI has characteristic diffraction peaks at the following 2 ⁇ angles: 5.0 ⁇ 0.2°, 6.2 ⁇ 0.2°, 9.4 ⁇ 0.2°, 12.5 ⁇ 0.2°, 15.2 ⁇ 0.2°, 21.4 ⁇ 0.2°, 23.5 ⁇ 0.2°, 24.7 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form VI has characteristic diffraction peaks at the following 2 ⁇ angles: 5.0 ⁇ 0.2°, 6.2 ⁇ 0.2°, 9.4 ⁇ 0.2°, 12.5 ⁇ 0.2°, 15.2 ⁇ 0.2°, 18.8 ⁇ 0.2°, 21.4 ⁇ 0.2°, 23.5 ⁇ 0.2°, 24.7 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form VI has characteristic diffraction peaks at the following 2 ⁇ angles: 5.0 ⁇ 0.2°, 6.2 ⁇ 0.2°, 9.4 ⁇ 0.2°, 12.5 ⁇ 0.2°, 14.0 ⁇ 0.2°, 15.2 ⁇ 0.2°, 18.8 ⁇ 0.2°, 21.4 ⁇ 0.2°, 23.5 ⁇ 0.2°, 24.7 ⁇ 0.2°.
  • the above-mentioned crystal form VI its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystal form VI its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystalline form VI has an X-ray powder diffraction pattern substantially as shown in FIG. 12 .
  • the differential scanning calorimetry curve of the differential scanning calorimetry curve has two onset points of endothermic peaks at 45.54 ⁇ 3°C and 163.80 ⁇ 3°C, respectively.
  • the above-mentioned crystal form VI has two endothermic peaks at 86.78 ⁇ 3°C and 165.61 ⁇ 3°C in the differential scanning calorimetry curve, respectively.
  • the above-mentioned crystalline form VI has a DSC pattern substantially as shown in FIG. 13 .
  • the above-mentioned crystal form VI its thermogravimetric analysis curve has a weight loss of 3.153% ⁇ 0.2% between room temperature and 120 ⁇ 3°C, and has a weight loss of 1.500% between 120 and 180°C ( ⁇ 3°C). ⁇ 0.2% weight loss.
  • the above-mentioned crystalline form VI has a TGA pattern substantially as shown in FIG. 13 .
  • the present invention provides the compound represented by formula (A), its solvate or hydrate crystal form VII, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 5.7 ⁇ 0.2°, 10.8 ⁇ 0.2°, 21.8 ⁇ 0.2°.
  • the above-mentioned crystal form VII its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 5.7 ⁇ 0.2°, 7.5 ⁇ 0.2°, 10.8 ⁇ 0.2°, 12.5 ⁇ 0.2°, 21.8 ⁇ 0.2°.
  • the above-mentioned crystal form VII its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 5.7 ⁇ 0.2°, 7.5 ⁇ 0.2°, 10.8 ⁇ 0.2°, 12.5 ⁇ 0.2°, 17.4 ⁇ 0.2°, 21.8 ⁇ 0.2°, 25.1 ⁇ 0.2°.
  • the above-mentioned crystal form VII its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 5.7 ⁇ 0.2°, 7.5 ⁇ 0.2°, 10.8 ⁇ 0.2°, 11.6 ⁇ 0.2°, 12.5 ⁇ 0.2°, 17.4 ⁇ 0.2°, 18.0 ⁇ 0.2°, 21.8 ⁇ 0.2°, 25.1 ⁇ 0.2°.
  • the above-mentioned crystal form VII its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystal form VII its X-ray powder diffraction pattern has characteristic diffraction peaks ( ⁇ 0.2°) at the following 2 ⁇ angles:
  • the above-mentioned crystalline form VII has an X-ray powder diffraction pattern substantially as shown in FIG. 1 .
  • the differential scanning calorimetry curve of the differential scanning calorimetry curve has two onset points of endothermic peaks at 47.52 ⁇ 3°C and 151.56 ⁇ 3°C, respectively.
  • the above-mentioned crystalline form VII has two endothermic peaks at 74.24 ⁇ 3°C and 156.63 ⁇ 3°C in its differential scanning calorimetry curve, respectively.
  • the above-mentioned crystal form VII has a DSC pattern substantially as shown in FIG. 2 .
  • the above-mentioned crystal form VII has a weight loss of 8.053% ⁇ 0.2% in the thermogravimetric analysis curve between room temperature and 150 ⁇ 3°C.
  • the above-mentioned crystal form VII has a TGA pattern substantially as shown in FIG. 2 .
  • the present invention provides the preparation method of the compound represented by formula (A) in crystalline form, its solvate or hydrate, comprising:
  • a drying step is further included to obtain the compound represented by formula (A), its solvate or hydrate in crystalline form.
  • the solvent is selected from a single solvent or a mixed solvent, preferably, the solvent is selected from n-heptane, toluene, ethyl acetate, methanol, ethylene glycol or water one or both.
  • the suspension in the above preparation method, can be prepared by adding a poor solvent to the solution.
  • the stirring temperature is -20°C ⁇ 70°C, preferably -10 ⁇ 60°C, more preferably 0 ⁇ 50°C, still more preferably 15 ⁇ 40°C.
  • the stirring time is 0.1 hour to 75 hours, preferably 0.5 hour to 72 hours.
  • the weight-volume ratio of the compound to the solvent is 1 g: 1-90 mL, preferably 1 g: 5-50 mL, more preferably 1 g: 10-25 mL.
  • the volume ratio of the two solvents in the mixed solvent is 1:1-100, preferably 1:1-50, more preferably 1:1-20, further preferably 1:1 to 5.
  • the one with the less amount is regarded as 1, and the volume ratio is calculated.
  • the drying time is 0.1 hour to 75 hours, preferably 1 hour to 48 hours, more preferably 5 hours to 24 hours, and further preferably 5 hours to 12 hours. hours, more preferably 5 hours to 10 hours; the drying is preferably vacuum drying.
  • the present invention provides the preparation method of the crystal form I of the compound represented by formula (A), its solvate or hydrate, comprising:
  • the solvent is selected from a single solvent or a mixed solvent, preferably, the single solvent can be selected from ethyl acetate, and the mixed solvent can be selected from toluene and n-heptane.
  • the suspension in the above preparation method, can be prepared by adding a poor solvent to the solution.
  • the stirring temperature is -20°C to 70°C, preferably -10 to 60°C, more preferably 0 to 50°C, and even more preferably 15 to 40°C.
  • the stirring time is 0.1 hour to 75 hours, preferably 0.5 hour to 72 hours.
  • the weight-volume ratio of the compound to the solvent is 1 g: 1-90 mL, preferably 1 g: 5-50 mL, more preferably 1 g: 10-25 mL.
  • the volume ratio of the two solvents in the mixed solvent is 1:1-100, preferably 1:1-50, more preferably 1:1-20, further preferably 1:1 to 5, more preferably 1:1.
  • the present invention provides the preparation method of the crystal form II of the compound represented by formula (A), its solvate or hydrate, comprising:
  • the present invention provides a method for preparing crystal form II of the compound represented by formula (A), its solvate or hydrate, comprising: adding water to crystal form VII, stirring or beating, crystallization, and separating solids.
  • the solvent is water.
  • the stirring or beating temperature is -20°C to 70°C, preferably -10 to 60°C, more preferably 0 to 50°C, and even more preferably 15 to 40°C , more preferably room temperature.
  • the stirring or beating time is 0.1 hour to 75 hours, preferably 0.5 hour to 72 hours.
  • the weight-volume ratio of the compound to the solvent is 1 g: 1-90 mL, preferably 1 g: 5-50 mL, more preferably 1 g: 10-25 mL.
  • the present invention provides a preparation method of the compound represented by formula (A), its solvate or hydrate crystal form III, comprising:
  • the temperature of the vacuum drying is 30-70°C, preferably 40-60°C, more preferably 40-50°C.
  • the vacuum drying time is 0.1 hour to 75 hours, preferably 1 hour to 48 hours, more preferably 5 hours to 24 hours, and further preferably 5 hours to 5 hours. 12 hours, more preferably 5 hours to 10 hours.
  • the present invention provides the preparation method of the compound represented by formula (A), its solvate or hydrate crystal form IV, comprising:
  • the solvent is methanol.
  • the stirring temperature is -20°C ⁇ 70°C, preferably -10 ⁇ 60°C, more preferably 0 ⁇ 50°C, still more preferably 15 ⁇ 40°C, more preferably More preferably, it is room temperature.
  • the stirring time is 0.1 hour to 75 hours, preferably 0.5 hour to 72 hours, more preferably 1 hour to 48 hours, and further preferably 2 hours to 24 hours.
  • the above-mentioned preparation method wherein, the weight-volume ratio of the compound to the solvent is 1g:1 ⁇ 90mL, preferably 1g:5 ⁇ 50mL, more preferably 1g:10 ⁇ 25mL, still more preferably 1g: 10 ⁇ 15mL.
  • the present invention provides a preparation method of the compound represented by formula (A), its solvate or hydrate crystal form V, comprising:
  • the temperature of the vacuum drying is 30-70°C, preferably 40-60°C, more preferably 40-50°C.
  • the vacuum drying time is 0.1 hour to 75 hours, preferably 1 hour to 48 hours, more preferably 5 hours to 24 hours, and further preferably 5 hours to 5 hours. 12 hours, more preferably 5 hours to 10 hours.
  • the present invention provides the preparation method of the crystal form VI of the compound represented by formula (A), its solvate or hydrate, comprising:
  • the solvent is ethylene glycol.
  • the stirring temperature is -20°C ⁇ 70°C, preferably -10 ⁇ 60°C, more preferably 0 ⁇ 50°C, still more preferably 15 ⁇ 40°C, more preferably More preferably, it is room temperature.
  • the stirring time is 0.1 hour to 75 hours, preferably 0.5 hour to 72 hours, and more preferably 1 hour to 48 hours.
  • the weight-volume ratio of the compound to the solvent is 1 g: 1-90 mL, preferably 1 g: 5-50 mL, more preferably 1 g: 10-25 mL.
  • the present invention provides the preparation method of the crystal form VII of the compound represented by formula (A), its solvate or hydrate, comprising:
  • the solvent is dichloromethane.
  • the stirring temperature is -20°C ⁇ 70°C, preferably -10 ⁇ 60°C, more preferably 0 ⁇ 50°C, still more preferably 15 ⁇ 40°C, more preferably More preferably, it is room temperature.
  • the stirring time is 0.1 hour to 75 hours, preferably 0.5 hour to 72 hours, and more preferably 1 hour to 48 hours.
  • the weight-volume ratio of the compound to the solvent is 1 g: 1-90 mL, preferably 1 g: 5-50 mL, more preferably 1 g: 10-25 mL.
  • Another object of the present invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising the compound of formula (A) in solid form, its solvate or hydrate, the compound of formula (A) in crystalline form, its solvate or hydrate or its crystalline mixture.
  • the above-mentioned pharmaceutical composition comprises the aforementioned compound of formula (A), its solvate or hydrate crystal form I, crystal form II, crystal form III, crystal form IV, crystal form V, crystal form A mixture of one or more of VI.
  • the pharmaceutical composition comprises the aforementioned compound of formula (A) in solid form, a solvate or hydrate thereof, a compound of formula (A) in crystalline form, a solvate or hydrate thereof, or a crystal thereof mixtures, and containing one or more pharmaceutically acceptable carriers.
  • the pharmaceutical composition comprises the aforementioned compound of formula (A), its solvate or hydrate crystal form I, crystal form II, crystal form III, crystal form IV, crystal form V, crystal form A mixture of one or more of VI, and one or more pharmaceutically acceptable carriers.
  • the present invention also provides the compound represented by formula (A) in solid form, its solvate or hydrate, and the compound represented by formula (A) in crystalline form, its solvate or hydrate, and formula (A) Compound, its solvate or hydrate crystal form I, crystal form II, crystal form III, crystal form IV, crystal form V, crystal form VI, crystal form VII or the above pharmaceutical composition in the preparation of medicine for the treatment of BTK-related disorders application on.
  • the present invention also relates to the compound represented by the formula (A) in the solid form, its solvate or hydrate, the compound represented by the formula (A) in the crystalline form, its solvate or hydrate, the formula (A) ), its solvate or hydrate crystal form I, crystal form II, crystal form III, crystal form IV, crystal form V, crystal form VI, crystal form VII or the above pharmaceutical composition, for the treatment of BTK related conditions.
  • the present application also provides the compound of formula (A) in the aforementioned solid form for the treatment of BTK-related disorders, its solvate or hydrate, the compound of formula (A) in crystalline form, and its solvate Form I, Form II, Form III, Form IV, Form V, Form VI, Form VII or the foregoing pharmaceutical composition.
  • the aforementioned BTK-related disorders involve dysregulation of BTK protein expression, level or activity.
  • the aforementioned BTK-related disorders include tumor diseases or autoimmune diseases; preferably, the tumor diseases are hematological tumors; more preferably, leukemia or lymphoma; more preferably, B-cell lymphoma; For mantle cell lymphoma, chronic lymphocytic leukemia, small lymphocytic lymphoma, marginal zone lymphoma, follicular lymphoma, Waldenstrom's macroglobulinemia, or diffuse large B-cell lymphoma.
  • the compound represented by the formula (A), its solvate or hydrate, the compound represented by the formula (A), its solvate or hydrate, the formula (A) in crystalline form are also provided.
  • the compound, its solvate or hydrate crystal form I, crystal form II, crystal form III, crystal form IV, crystal form V, crystal form VI, crystal form VII or the above-mentioned pharmaceutical composition are prepared for the treatment of tumor diseases or The use of medicines in autoimmune diseases.
  • the tumor disease is hematological tumor; more preferably, leukemia or lymphoma; more preferably, B-cell lymphoma; still more preferably, mantle cell lymphoma, chronic lymphocytic leukemia, small lymphocytic lymphoma , marginal zone lymphoma, follicular lymphoma, Waldenstrom's macroglobulinemia, or diffuse large B-cell lymphoma.
  • the BTK-related disorder involves dysregulation of BTK protein expression, level or activity.
  • the present invention also relates to a method for treating a patient's condition, by administering to the patient the compound shown in the formula (A) of the above-mentioned solid form, a solvate or a hydrate thereof, a crystalline form of the formula (
  • the compound represented by A), its solvate or hydrate, the compound represented by formula (A), its solvate or hydrate crystal form I, crystal form II, crystal form III, crystal form IV, crystal form V, Form VI, form VII or the above-mentioned pharmaceutical composition the condition of the patient is a BTK-related condition.
  • the above-mentioned method for treating a patient's disorder wherein the BTK-related disorder includes a tumor disease or an autoimmune disease; preferably, the tumor disease is a hematological tumor; more preferably, a leukemia or lymphoma more preferably B cell lymphoma; more preferably mantle cell lymphoma, chronic lymphocytic leukemia, small lymphocytic lymphoma, marginal zone lymphoma, follicular lymphoma, Waldenström macroglobulinemia or diffuse lymphoma B-cell lymphoma.
  • the tumor disease is a hematological tumor; more preferably, a leukemia or lymphoma more preferably B cell lymphoma; more preferably mantle cell lymphoma, chronic lymphocytic leukemia, small lymphocytic lymphoma, marginal zone lymphoma, follicular lymphoma, Waldenström macroglobulinemia or diffuse lymphoma B-cell lymphoma.
  • solvate refers to an association formed by one or more solvent molecules and the compound represented by formula 2 of the present application, including those containing both water molecules and one or more other solvent molecules. associates.
  • hydrate refers to an association of one or more water molecules with a compound of formula 2 of the present application.
  • substantially as shown means at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or at least 95%, or at least 96% of the X-ray powder diffraction pattern. %, or at least 97%, or at least 98%, or at least 99% of the peaks are shown in its graph; or in the DSC graph, the error range of the onset or peak temperature of each endothermic or exothermic peak is ⁇ 10°C Within the range, preferably within the range of 5°C, more preferably within the range of 3°C; or in the TGA spectrum, the error range of the starting point or end point of the weight loss temperature is within the range of ⁇ 10°C, preferably within the range of 5°C, more preferably within the range of 3°C, the weight loss is within the range of ⁇ 10°C.
  • the percentages are within ⁇ 0.5%, preferably within ⁇ 0.3%, within ⁇ 0.2%. Further, when the content of a certain crystal form in the product is gradually reduced, some diffraction peaks in the powder X-ray diffraction pattern that belong to the crystal form may be reduced due to the detection sensitivity of the instrument.
  • characteristic diffraction peak refers to a diffraction peak in an X-ray powder diffraction pattern that can be used to represent the crystal form, which is related to the peak position, peak shape and relative peak intensity of the diffraction peak, for example, small angle peaks, peaks
  • the shape is sharp and the relative peak intensity is at least 2.9% or more, or at least 3% or more, or at least 5% or more, or at least 10% or more, or at least 20% or more, or at least 30% or more, or at least 40% or more, or at least 50% or more, or at least 60% or more, or at least 70% or more, or at least 75% or more of the diffraction peaks.
  • tumor includes benign tumors, malignant tumors, and borderline tumors, wherein malignant tumors are also collectively referred to as cancers.
  • treating generally refers to obtaining a desired pharmacological and/or physiological effect, including partial or complete stabilization or cure of a disease and/or effects resulting from a disease.
  • Treatment encompasses any treatment of a disease in a patient, including: (a) inhibiting the symptoms of the disease, ie, preventing its progression; or (b) alleviating the symptoms of the disease, ie, causing regression of the disease or symptoms.
  • effective amount or “therapeutically effective amount” means an amount of a compound of the present application that (i) treats a particular disease, or (ii) reduces, ameliorates, or eliminates one or more symptoms of a particular disease.
  • terapéuticaally effective amount means an amount of a compound sufficient to effect treatment of a disease when administered to a patient for the treatment of the disease.
  • the amount of a compound of the present application that constitutes a “therapeutically effective amount” will vary depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by those skilled in the art according to its own knowledge and the present disclosure.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable adjuvant” refers to those carriers or adjuvants that are not significantly irritating to the organism and that do not impair the biological activity and properties of the active compound.
  • the "heating temperature”, “cooling temperature” or “crystallization temperature” described in this application is in °C or °C, and the error range can be ⁇ 10, ⁇ 5, ⁇ 4, ⁇ 3, ⁇ 2 or ⁇ 1°C.
  • the intermediate compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by their combination with other chemical synthesis methods, and those skilled in the art.
  • Well-known equivalents, preferred embodiments include, but are not limited to, the examples of the present invention.
  • DCM dichloromethane
  • DIAD diisopropyl azodicarboxylate
  • DIPEA diisopropylethylamine
  • DMF N,N-dimethylformamide
  • EA ethyl acetate
  • HATU 2-(7 -benzotriazole oxide)-N,N,N',N'-tetramethylurea hexafluorophosphate
  • NBS N-bromosuccinimide
  • NIS N-iodosuccinimide Amine
  • Pd(dppf)Cl 2 [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride
  • Pd(PPh 3 ) 4 tetrakis(triphenylphosphine)palladium
  • PdCl 2 palladium dichloride
  • Pd(OAc) 2 palladium acetate
  • Pd(PPh 3 ) 2 Cl 2 bistriphenyl
  • the crystal form of the compound represented by the formula (A) mentioned in the present invention has good crystallinity, preferably the crystal form has good stability, is easy to prepare medicine, and has high bioavailability; the compound (A) of the present invention has excellent BTK inhibitory activity , and maintains good BTK inhibition selectivity, excellent in vivo antitumor activity, good oral administration performance, and can provide more effective treatment for diseases caused by abnormal expression of BTK, and in different species of liver microsomes ( Under the action of human liver microsomes, rat liver microsomes and mouse liver microsomes), the types and proportions of metabolites are small, and the prototype drugs are basically the main ones (60min: 84%-98%), and the metabolic stability is better.
  • Test method about 1 ⁇ 2mg sample is used for XRPD detection
  • Light tube voltage 45kV
  • light tube current 40mA
  • Test method Take a sample (1-5 mg) and place it in a DSC sample pan, cover the sample pan and poke holes, and heat the sample at a heating rate of 10°C/min to the final temperature after equilibrating at 25°C.
  • Termination temperature 300°C.
  • Test method The sample is placed in a peeled open aluminum sample pan. After the sample mass is automatically weighed in a TGA heating furnace, the sample is heated to the final temperature at a rate of 10°C/min.
  • Heating start temperature 25 ⁇ 30°C
  • Termination temperature 300°C.
  • Test method Add a sufficient amount of sample to the instrument to simulate dynamic water vapor adsorption, and record the change in weight at 25°C under different humidity balances.
  • the hygroscopicity of the sample is classified:
  • Hygroscopic hygroscopic weight gain is less than 15% but not less than 2%
  • hygroscopic weight gain is less than 2% but not less than 0.2%
  • hygroscopic weight gain is less than 0.2%.
  • FIG. 1 X-ray powder diffraction pattern of Form VII of Example 1.
  • Figure 3 X-ray powder diffraction pattern of Form I of Example 2.
  • FIG. 5 X-ray powder diffraction pattern of Form II of Example 4.
  • FIG. 6 DSC-TGA spectrum of Form II of Example 4.
  • FIG. 7 X-ray powder diffraction pattern of Form III of Example 5.
  • FIG. 8 DSC-TGA spectrum of Form III of Example 5.
  • FIG. 9 X-ray powder diffraction pattern of Form IV of Example 6.
  • FIG. 10 DSC-TGA spectrum of Form IV of Example 6.
  • FIG. 11 X-ray powder diffraction pattern of Form V of Example 7.
  • FIG. 12 X-ray powder diffraction pattern of Form VI of Example 8.
  • FIG. 14 DVS graph of Form I of Example 2.
  • FIG. 15 DSC-TGA spectrum of Form V of Example 7.
  • Figure 16 Schematic diagram of experimental results of the REC-1 xenograft tumor model.
  • Figure 17 Schematic diagram of experimental results of the TMD8 xenograft tumor model.
  • Figure 18 XRPD overlays of Form I before and after grinding.
  • the endothermic peak onset temperatures were 47.52 °C and 151.56 °C, respectively, and the peak temperatures were 74.24 °C and 156.63 °C, respectively. There is a weight loss of 8.053% between °C, see Figure 2.
  • Crystal form VII was weighed into a glass bottle of suitable volume, water (0.5 mL) was added, it was sealed with a parafilm, slurried at room temperature for 3 days, and filtered to obtain a solid. A sample was taken for X-ray powder diffraction, and it was shown to be a crystalline solid (crystal form II) with good crystallinity. The sample was taken for DSC-TGA test. The DSC chart showed that the sample had three endothermic peaks.
  • the endothermic peak starting point temperatures were 49.26 °C, 84.59 °C, and 168.35 °C, respectively, and the peak temperatures were 69.36 °C, 98.45 °C, and 176.96 °C, respectively.
  • TGA chart It is shown that the sample has a weight loss of 3.417% between room temperature and 100°C, see Figure 6 .
  • the crystal form II was taken and dried in a vacuum drying oven at 40°C for 12 hours to obtain a solid.
  • a sample was taken for X-ray powder diffraction, and it was shown to be a crystalline solid (crystal form III, anhydrous crystal form) with good crystallinity.
  • the crystal form III was heated to 160°C and then subjected to X-ray powder diffraction, and its XRPD spectrum did not change.
  • the DSC chart shows that the sample has three endothermic peaks, the endothermic peak onset temperatures are 38.13 °C, 75.07 °C and 167.19 °C, and the peak temperatures are 57.40 °C, 96.32 °C and 177.55 °C, respectively, TGA spectrum A period of 3.075% weight loss is shown before 90°C, see Figure 8.
  • the crystal form IV was dried in a vacuum drying oven at 40°C for 12 hours to obtain a solid.
  • the sample is shown as a crystalline solid (crystal form V, anhydrous crystal form), and the crystallinity is good, the spectrum is shown in Figure 11, the sample is taken for DSC-TGA test, the DSC chart shows that the sample has two There are endothermic peaks, the starting point temperatures of the endothermic peaks are 36.14°C and 166.61°C, and the peak temperatures are 65.23°C and 174.53°C, respectively.
  • the TGA diagram shows that the sample has a weight loss of 0.874% between room temperature and 280°C, see Figure 15 . Its XRPD diffraction peak data are shown in Table 6.
  • the crystalline form I (5 mg) and the crystalline form II (5 mg) of the compound of formula (A) were respectively weighed, placed at the bottom of the sample vial, and spread out into a thin layer. The samples were sampled and tested on the 7th day, and the test results were compared with the initial test results on the 0th day. The test results are shown in Table 8 below:
  • Example 2 crystal form I (about 97 mg) and place it in the DVS sample chamber for testing. The samples after DVS were taken for X-ray powder diffraction.
  • the DVS spectrum of the crystalline form I of the compound of formula (A) is shown in FIG. 14 .
  • the hygroscopic weight gain of the crystal form I of the compound of formula (A) at 25°C and 80% RH is 0.9%, and the hygroscopic weight gain at 25°C and 90% RH is 1.0%. transformation occurs.
  • reaction buffer 50 mM HEPES pH 7.4, 50 mM MgCl 2 , 0.5 mM MnCl 2 , 0.2 mM Na 3 VO 4 , 1 mM DTT
  • reaction buffer 50 mM HEPES pH 7.4, 50 mM MgCl 2 , 0.5 mM MnCl 2 , 0.2 mM Na 3 VO 4 , 1 mM DTT
  • final concentration 5 ⁇ M 50 mM HEPES pH 7.4, 50 mM MgCl 2 , 0.5 mM MnCl 2 , 0.2 mM Na 3 VO 4 , 1 mM DTT
  • the above reaction system was placed in a shaker at 37°C (100 rpm) for 1 hour, then the plate was washed three times with T-PBS, and the primary antibody PY99 100 ⁇ L/well (Santa Cruz) was added, and the reaction was shaken at 37°C for 0.5 hour. After washing the plate with T-PBS, 100 ⁇ L/well of horseradish peroxidase-labeled goat anti-mouse secondary antibody diluent was added, and the reaction was shaken at 37°C for 0.5 hours. After washing the plate with T-PBS, add 100 ⁇ L/well of 2 mg/mL OPD chromogenic solution, and react at 25°C for 1-10 minutes in the dark. Then, 50 ⁇ L/well of 2M H 2 SO 4 was added to stop the reaction, and the reaction was read with a tunable wavelength microplate reader SPECTRA MAX Plus384 with a wavelength of 490 nm.
  • Compounds S1, S10, ibrutinib, acalatinib, S18s, S19s and S20s are used as positive control compounds, wherein compounds S1, S10, S18s, S19s and S20s adopt the methods disclosed in the prior art (eg CN108101905A) or similar methods, ibrutinib and acalatinib were purchased from Selleck Company.
  • the IC 50 value was obtained by four-parameter regression using the software attached to the microplate reader. The results are listed in Table 9 below.
  • the cell suspension (Ramos: 10,000 cells/well; TMD8: 12,000 cells/well) was inoculated in a 96-well plate, and left for 2 hours in a 37°C incubator to wait for the cells to stabilize, and then add different concentrations of test compounds to each well. (3 duplicate wells for each concentration), and blank control (well containing only culture medium, no cells), negative control (well with cells only, no compound added) and positive compound control at the same time. After dosing for 72 h, 20 ⁇ L of MTT (5 mg/mL) was added to each well and incubated at 37°C for 4 h, followed by 100 ⁇ L of triple solution (10% SDS, 5% isobutanol, 0.01M HCl), and placed at 37°C overnight. The OD value was measured with a wavelength-tunable microplate reader SPECTRAmax Plus384 at a wavelength of 570 nm.
  • the inhibition rate of the compound was calculated by the following formula:
  • the IC 50 value was obtained by four-parameter regression using the software attached to the microplate reader. The experiment was repeated 3 times independently and the results are listed in Table 10 below.
  • the compounds S1, S10, ibrutinib, acalatinib, S18s, S19s and S20s described above were also used as positive control compounds.
  • Cell culture Human lymphoma TMD8 cells were cultured in vitro in suspension, and the culture conditions were RPMI 1640 medium (supplier: gibco; product number: 22400-089; production batch number: 4868546) with 10% fetal bovine serum, 100U/mL penicillin and 100 ⁇ g /mL streptomycin, incubate at 37°C with 5% CO 2 . Routine treatment passaging was performed twice a week. When the cell saturation was 80%-90%, cells were harvested, counted, and seeded.
  • Human mantle cell lymphoma REC-1 cells were cultured in vitro in suspension in RPMI 1640 medium (supplier: gibco; product number: 22400-089; production batch number: 1868795) with 10% fetal bovine serum, 100U/mL penicillin and 100 ⁇ g/mL streptomycin, 37 °C 5% CO2 culture. Routine treatment passaging was performed twice a week. When the cell saturation was 80%-90%, cells were harvested, counted, and seeded.
  • Tumor Measurements and Experimental Indicators examine whether tumor growth is inhibited, delayed or cured. Tumor diameters were measured with vernier calipers three times a week.
  • the formula for calculating tumor volume is:
  • V 0.5a ⁇ b 2 .
  • a and b represent the long and short diameters of the tumor, respectively.
  • TGI % or relative tumor proliferation rate T/C (%).
  • TGI % reflecting tumor growth inhibition rate.
  • TGI(%) [1-(average tumor volume at the end of administration of a certain treatment group-average tumor volume at the beginning of administration of this treatment group)/(average tumor volume at the end of treatment in the solvent control group-average at the beginning of treatment in the solvent control group Tumor volume)] ⁇ 100%.
  • T/C% T RTV /C RTV ⁇ 100% (T RTV : relative tumor volume of the treatment group; C RTV : relative tumor volume of the negative control group).
  • Statistical analysis including mean and standard error (SEM) of tumor volume for each time point for each group.
  • the treatment group showed the best therapeutic effect on the 15th day (REC-1 xenograft tumor model) and 17th day (TMD8 xenograft tumor model) after administration, so a statistical analysis was performed based on this data to evaluate between groups difference.
  • One-way ANOVA was used for comparison among three or more groups, and if there was a significant difference in F value, the Games-Howell method was used to test. All data analyses were performed with SPSS 17.0. p ⁇ 0.05 was considered a significant difference.
  • the in vivo efficacy of compound A in the human mantle cell lymphoma REC-1 xenograft tumor model is shown in Table 13 and FIG. 16 .
  • a. Mean ⁇ SEM; b. Tumor growth inhibition was calculated by T/C and TGI (TGI(%) [1-(T 15 -T 0 )/(V 15 -V 0 )] ⁇ 100); The cp value is calculated according to the tumor volume; d. Dosing mode: once a day; e. Detection using the sample of Preparation Example 1.
  • a. Mean ⁇ SEM; b. Tumor growth inhibition was calculated by T/C and TGI (TGI(%) [1-(T 17 -T 0 )/(V 17 -V 0 )] ⁇ 100); The cp value is calculated according to the tumor volume; d. Dosing mode: once a day; e. Detection using the sample of Preparation Example 1.
  • Mode of administration once a day.
  • Oral administration 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24 hours after administration;
  • Intravenous administration 5 minutes, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24 hours after administration;
  • venous blood was collected from the retroocular venous plexus of the rat, placed in a heparinized test tube, centrifuged at 11,000 rpm for 5 minutes, and the plasma was separated and frozen in a -20°C refrigerator.
  • the concentration of Compound A in rat plasma was determined by LC/MS/MS method.
  • the peak concentration Cmax and the peak time Tmax are measured values
  • AUC 0-t value of the area under the drug-time curve calculated by trapezoidal method
  • AUC 0- ⁇ AUC 0-t +C t / ke ,
  • C t is the blood drug concentration at the last measurable time point
  • ke is the elimination rate constant
  • Mean residence time MRT AUMC/AUC.
  • Absolute bioavailability F (AUC gavage ⁇ D vein )/(AUC vein ⁇ D gavage ) ⁇ 100%
  • SD rats purchased from Shanghai Sipple-Bike Laboratory Animal Co., Ltd., production license number SCXK (Shanghai) 2018-000612, half male and half male, weighing 170-250g, were randomly divided into 2 groups, 6 rats in each group, respectively Test compounds were administered by gavage and intravenously, as detailed in Table 18 below.
  • Oral administration 0.25, 0.5, 1.0, 2.0, 3.0, 5.0, 7.0, 9.0 and 24 hours after administration;
  • Intravenous administration 5 minutes, 0.25, 0.5, 1.0, 2.0, 3.0, 5.0, 7.0 and 24 hours after administration;
  • 0.2 mL of venous blood was collected from the retroocular venous plexus of rats, placed in an EDTA-K2 test tube, centrifuged at 11,000 rpm for 5 minutes, and the plasma was separated within 2 hours, and stored at -70°C for testing.
  • the concentration of Compound A in rat plasma was determined by LC/MS/MS method.
  • the calculation software and parameters are the same as those described in the corresponding parts in Experimental Example 7.
  • the test results are shown in Table 19 below:
  • Compound A is a novel, orally available, highly selective and highly active BTK inhibitor. Its in vitro and in vivo activity is significantly better than that of currently marketed BTK inhibitors. At the same dose, it inhibits tumor growth. The activity is significantly better than that of the positive control drug ibrutinib; further, the crystal form of the compound represented by the formula (A) mentioned in the present invention has good crystallinity, preferably the crystal form has good stability, is easy to formulate medicine, and is biologically stable. High utilization. It shows that compound A and its specific crystal form have great development value.

Landscapes

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

Abstract

提供了一种化合物A、其溶剂合物或水合物的固体形式、结晶形式、晶型及制备方法和用途,所得化合物A的结晶形式具有结晶度好、稳定性好,化合物A具有良好的布鲁顿酪氨酸激酶抑制活性、细胞抑制活性、体内抗肿瘤活性、药代性质和代谢稳定性。

Description

固体形式的布鲁顿酪氨酸激酶抑制剂化合物及其用途 技术领域
本发明涉及作为小分子布鲁顿酪氨酸激酶抑制剂的式(A)所示化合物、其溶剂合物或水合物的固体形式、结晶形式、具体晶型及药物组合物,并涉及其在制备治疗布鲁顿酪氨酸激酶相关病症药物中的应用。
背景技术
布鲁顿酪氨酸激酶(Bruton’s tyrosine kinase,BTK)是隶属于TEC酪氨酸激酶家族的一种非受体酪氨酸激酶。TEC家族成员包括Tec、Bmx、BTK、Itk和Txk。BTK是TEC家族中研究最广泛的成员,是B细胞受体(BCR)信号通路中的关键调节因子,在多种类型恶性血液肿瘤中广泛表达,参与B细胞的增殖、分化与凋亡。因此,BTK已成为恶性血液肿瘤药物治疗的重要分子靶点。
目前,已有4个BTK抑制剂获批上市,依鲁替尼(Ibrutinib,商品名Imbruvica)是首个上市的BTK小分子抑制剂,属于一代BTK抑制剂,于2013年被美国FDA批准用于套细胞淋巴瘤(Mantle Cell Lymphoma,MCL)和慢性淋巴细胞白血病(Chronic Lymphocytic Leukemia,CLL)等的临床治疗,其后依鲁替尼不断扩充适应症,目前获批的适应症还包括携带17p删除的慢性淋巴细胞白血病/小淋巴细胞淋巴瘤(CLL/Small Lymphocytic Lymphoma,SLL)、华氏巨球蛋白血症(Waldenstrom Macroglobulinemia,WM)、边缘区淋巴瘤(Marginal Zone Lymphoma,MZL)、慢性移植物抗宿主病(chronic Graft-Versus-Host Disease,cGVHD)。依鲁替尼可以与BTK激酶ATP结合域中第481位半胱氨酸(Cys481)形成共价结合,不可逆性抑制BTK活化,阻断BTK信号通路,从而抑制B淋巴瘤细胞的增殖和存活,达到肿瘤治疗的目的。依鲁替尼在临床治疗中取得了实质性的疗效。但由于依鲁替尼靶点选择性较差,临床存在一定毒副作用。阿卡替尼(Acalabrutinib,ACP-196,商品名Calquence)于2017年被批准用于MCL和CLL的治疗,属于第二代BTK靶向药。相对于依鲁替尼,阿卡替尼对BTK的选择性更高,脱靶毒性较低。另外,百济神州生物科技有限公司开发的Zanubrutinib(泽布替尼,BGB-3111)于2019年11月经FDA批准用于经治的成年MCL患者,成为首获FDA突破性疗法认定的中国本土抗癌药。日本小野制药有限公司研发的Tirabrutinib(ONO-4059)于2020年3月经日本医药品医疗器械综合机构PMDA)批准上市,用于复发或难治性的原发性中枢神经系统淋巴瘤(PCNSL)和淋巴浆细胞性淋巴瘤(LPL)的治疗。总的来讲,第一代抑制剂对BTK的抑制活性高,但靶点选择和生物利用率差;第二代抑制剂选择性好,但对BTK的抑制率低于第一代抑制剂。结构上,目前上市或在研的化合物的核心骨架主要是双环体系。
为了获得兼具第一代抑制剂的高活性和第二代抑制剂的良好选择性的新一代BTK抑制剂,中国科学院上海药物研究所在CN108101905A专利中公开了一类结构独特的三环化合物。其中,嘧啶并吲嗪类化合物S1和S10及嘧啶并吡呤类化合物S18、S19和S20显示了较高的BTK抑制活性,在进一步的工作中,还合成了S18、S19和S20的S构型的化合物(即S18s、S19s和S20s)[Yu Xue,et al.Discovery of 4,7-Diamino-5-(4-phenoxyphenyl)-6-methylenepyrimido[5,4-b]pyrrollizines as Novel Bruton’s Tyrosine Kinase Inhibitors.J.Med.Chem.,2018,61,4608-4627.]。但进一步研究发现S1、S10、S18s和S19s这些化合物代谢过程中不稳定,末端苯基的4-位易于氧化;而化合物S20s的口服生物利用度不理想。
Figure PCTCN2021125689-appb-000001
发明内容
化合物A具有优良的BTK抑制活性、且保持较好的BTK抑制选择性、具有优良的体内抗肿瘤活性、良好的口服给药性能和良好的代谢稳定性,具有开发为BTK选择性抑制剂的潜力。
Figure PCTCN2021125689-appb-000002
一方面,本发明提供了固体形式的式(A)所示化合物、其溶剂合物或水合物。
另一方面,本发明提供了结晶形式的式(A)所示化合物、其溶剂合物或水合物。
本发明的一些方案中,上述结晶形式,其特征在于,所述结晶形式为无溶剂且无水结晶形式或水合物结晶形式,优选为无溶剂且无水结晶形式。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型I,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°。
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,23.4±0.2°。
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,18.7±0.2°,23.4±0.2°。。
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,16.2±0.2°,18.7±0.2°,23.4±0.2°。
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,16.2±0.2°,18.7±0.2°,23.4±0.2°,26.1±0.2°。
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,16.2±0.2°,18.7±0.2°,23.4±0.2°,24.2±0.2°,24.8±0.2°,26.1±0.2°。
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,16.2±0.2°,18.7±0.2°,19.4±0.2°,21.7±0.2°,23.4±0.2°,24.2±0.2°,24.8±0.2°,26.1±0.2°。
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,16.2±0.2°,16.7±0.2°,18.7±0.2°,19.4±0.2°,21.7±0.2°,23.4±0.2°,24.2±0.2°,24.8±0.2°,26.1±0.2°,27.6±0.2°。
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,16.2±0.2°,16.7±0.2°,18.7±0.2°,19.4±0.2°,20.8±0.2°,21.7±0.2°,23.4±0.2°,24.2±0.2°,24.8±0.2°,26.1±0.2°,27.6±0.2°。
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,16.2±0.2°,16.7±0.2°,18.7±0.2°,19.4±0.2°,20.8±0.2°,21.7±0.2°,22.5±0.2°,23.4±0.2°,24.2±0.2°,24.8±0.2°,26.1±0.2°,27.6±0.2°,30.4±0.2°。
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 峰位置(2θ) 峰位置(2θ)
4.8 19.4 23.7
11.6 20.8 24.2
13.6 21.7 24.8
16.2 22.5 26.1
16.7 22.7 29.2
18.7 23.4 30.4
本发明的一些方案中,上述晶型I,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
4.8 45.9 19.4 10.9 23.7 9.1
11.6 100.0 20.8 9.9 24.2 14.9
13.6 56.0 21.7 11.2 24.8 16.4
16.2 25.9 22.5 9.0 26.1 20.7
16.7 8.6 22.7 8.4 29.2 6.0
18.7 34.1 23.4 60.4 30.4 8.4
本发明的一些方案中,上述晶型I,其具有基本上如图3所示的X-射线粉末衍射图谱。
本发明的一些方案中,上述晶型I,其差示扫描量热曲线在272.89±3℃有一个放热峰的起始点。
本发明的一些方案中,上述晶型I,其差示扫描量热曲线在274.74±3℃有一个放热峰。
本发明的一些方案中,上述晶型I,其具有基本上如图4所示的DSC图谱。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型II,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:8.5±0.2°,10.6±0.2°,15.0±0.2°。
本发明的一些方案中,上述晶型II,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0±0.2°,8.5±0.2°,10.6±0.2°,15.0±0.2°,22.1±0.2°。
本发明的一些方案中,上述晶型II,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0±0.2°,7.9±0.2°,8.5±0.2°,10.6±0.2°,15.0±0.2°,22.1±0.2°,25.3±0.2°。
本发明的一些方案中,上述晶型II,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0±0.2°,7.9±0.2°,8.5±0.2°,10.6±0.2°,15.0±0.2°,18.0±0.2°,22.1±0.2°,25.3±0.2°,26.3±0.2°。
本发明的一些方案中,上述晶型II,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0±0.2°,7.9±0.2°,8.5±0.2°,10.6±0.2°,15.0±0.2°,17.1±0.2°,18.0±0.2°,19.1±0.2°,22.1±0.2°,25.3±0.2°,26.3±0.2°。
本发明的一些方案中,上述晶型II,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.0±0.2°,7.9±0.2°,8.5±0.2°,10.6±0.2°,15.0±0.2°,17.1±0.2°,18.0±0.2°,19.1±0.2°,22.1±0.2°,25.3±0.2°,26.3±0.2°。
本发明的一些方案中,上述晶型II,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.0±0.2°,7.9±0.2°,8.5±0.2°,10.6±0.2°,12.8±0.2°,15.0±0.2°,17.1±0.2°,18.0±0.2°,19.1±0.2°,21.3±0.2°,22.1±0.2°,25.3±0.2°,26.3±0.2°。
本发明的一些方案中,上述晶型II,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.0±0.2°,7.9±0.2°,8.5±0.2°,10.6±0.2°,11.5±0.2°,12.8±0.2°,14.1±0.2°,15.0±0.2°,17.1±0.2°,18.0±0.2°,19.1±0.2°,21.3±0.2°,22.1±0.2°,25.3±0.2°,26.3±0.2°。
本发明的一些方案中,上述晶型II,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 峰位置(2θ) 峰位置(2θ)
4.3 14.1 21.3
7.0 15.0 22.1
7.9 17.1 25.1
8.5 18.0 25.3
10.6 18.2 26.3
11.5 19.1 30.7
12.8    
本发明的一些方案中,上述晶型II,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
4.3 18.8 14.1 9.4 21.3 16.1
7.0 29.6 15.0 100.0 22.1 49.3
7.9 27.3 17.1 28.0 25.1 13.9
8.5 96.2 18.0 28.3 25.3 49.7
10.6 83.8 18.2 18.2 26.3 33.7
11.5 14.4 19.1 14.3 30.7 11.2
12.8 11.5        
本发明的一些方案中,上述晶型II,其具有基本上如图5所示的X-射线粉末衍射图谱。
本发明的一些方案中,上述晶型II,其差示扫描量热曲线在49.26±3℃、84.59±3℃和168.35±3℃分别有三个吸热峰的起始点。
本发明的一些方案中,上述晶型II,其差示扫描量热曲线在69.36±3℃、98.45±3℃和176.96±3℃分别有三个吸热峰。
本发明的一些方案中,上述晶型II,其具有基本上如图6所示的DSC图谱。
本发明的一些方案中,上述晶型II,其热重分析曲线在室温~100±3℃之间有3.417%±0.2%的失重。
本发明的一些方案中,上述晶型II,其具有基本上如图6所示的TGA图谱。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型III,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°。
本发明的一些方案中,上述晶型III,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.1±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,25.5±0.2°。
本发明的一些方案中,上述晶型III,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.1±0.2°,8.0±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,22.3±0.2°,25.5±0.2°。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型III,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,16.2±0.2°。
本发明的一些方案中,上述晶型III,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.1±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,16.2±0.2°,25.5±0.2°。
本发明的一些方案中,上述晶型III,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.1±0.2°,8.0±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,16.2±0.2°,22.3±0.2°,25.5±0.2°。
本发明的一些方案中,上述晶型III,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.1±0.2°,8.0±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,16.2±0.2°,19.3±0.2°,22.3±0.2°,25.5±0.2°。
本发明的一些方案中,上述晶型III,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.1±0.2°,8.0±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,16.2±0.2°,17.2±0.2°,18.1±0.2°,19.3±0.2°,22.3±0.2°,25.5±0.2°。
本发明的一些方案中,上述晶型III,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.1±0.2°,8.0±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,16.2±0.2°,17.2±0.2°,18.1±0.2°,19.3±0.2°,21.4±0.2°,22.3±0.2°,25.5±0.2°。
本发明的一些方案中,上述晶型III,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 峰位置(2θ) 峰位置(2θ)
4.3 11.6 19.3
7.1 14.6 21.4
8.0 15.1 22.3
8.6 16.2 25.5
9.0 17.2 26.1
10.4 18.1 26.5
10.6 18.4 31.1
本发明的一些方案中,上述晶型III,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
4.3 68.2 11.6 11.1 19.3 22.1
7.1 45.2 14.6 20.4 21.4 17.9
8.0 47.6 15.1 93.3 22.3 45.6
8.6 72.5 16.2 28 25.5 67.7
9.0 15.0 17.2 22.1 26.1 24.7
10.4 48.7 18.1 24.4 26.5 26.9
10.6 100.0 18.4 20.8 31.1 10.0
本发明的一些方案中,上述晶型III,其具有基本上如图7所示的X-射线粉末衍射图谱。
本发明的一些方案中,上述晶型III,其差示扫描量热曲线在38.13±3℃、75.07±3℃和167.19±3℃ 分别有三个吸热峰的起始点。
本发明的一些方案中,上述晶型III,其差示扫描量热曲线在57.40±3℃、96.32±3℃和177.55±3℃分别有三个吸热峰。
本发明的一些方案中,上述晶型III,其具有基本上如8所示的DSC图谱。
本发明的一些方案中,上述晶型III,其热重分析曲线在90±3℃之前有3.075%±0.2%的失重。
本发明的一些方案中,上述晶型III,其具有基本上如图8所示的TGA图谱。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型IV,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:6.3±0.2°,9.7±0.2°,12.8±0.2°。
本发明的一些方案中,上述晶型IV,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.1±0.2°,6.3±0.2°,9.7±0.2°,12.8±0.2°,14.1±0.2°。
本发明的一些方案中,上述晶型IV,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.1±0.2°,6.3±0.2°,9.7±0.2°,12.8±0.2°,14.1±0.2°,19.0±0.2°。
本发明的一些方案中,上述晶型IV,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.1±0.2°,6.3±0.2°,9.7±0.2°,12.8±0.2°,14.1±0.2°,15.6±0.2°,19.0±0.2°,21.9±0.2°。
本发明的一些方案中,上述晶型IV,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.1±0.2°,6.3±0.2°,9.7±0.2°,12.8±0.2°,14.1±0.2°,19.0±0.2°,20.4±0.2°,23.2±0.2°。
本发明的一些方案中,上述晶型IV,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.1±0.2°,6.3±0.2°,9.7±0.2°,12.8±0.2°,14.1±0.2°,19.0±0.2°,20.4±0.2°,21.9±0.2°,23.2±0.2°,26.3±0.2°。
本发明的一些方案中,上述晶型IV,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.1±0.2°,6.3±0.2°,9.7±0.2°,12.8±0.2°,14.1±0.2°,15.6±0.2°,19.0±0.2°,20.4±0.2°,21.9±0.2°,23.2±0.2°,26.3±0.2°。
本发明的一些方案中,上述晶型IV,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 峰位置(2θ) 峰位置(2θ)
5.1 15.6 23.8
6.3 17.8 24.1
9.7 19.0 25.1
11.6 20.4 25.8
12.8 21.9 26.3
14.1 23.2  
本发明的一些方案中,上述晶型IV,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
5.1 14.1 15.6 9.5 23.8 6.9
6.3 44.7 17.8 5.6 24.1 5.1
9.7 52.2 19.0 16.4 25.1 5.6
11.6 4.4 20.4 12.7 25.8 4.5
12.8 100 21.9 9.8 26.3 11.7
14.1 17.5 23.2 15.9    
本发明的一些方案中,上述晶型IV,其具有基本上如图9所示的X-射线粉末衍射图谱。
本发明的一些方案中,上述晶型IV,其差示扫描量热曲线在45.75±3℃和159.89±3℃分别有两个吸热峰的起始点。
本发明的一些方案中,上述晶型IV,其差示扫描量热曲线在70.65±3℃和166.96±3℃分别有两个吸热峰。
本发明的一些方案中,上述晶型IV,其具有基本上如图10所示的DSC图谱。
本发明的一些方案中,上述晶型IV,其热重分析曲线在室温~200±3℃之间有7.863%±0.2%的失重。
本发明的一些方案中,上述晶型IV,其具有基本上如图10所示的TGA图谱。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型V,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.6±0.2°,7.1±0.2°,14.3±0.2°。
本发明的一些方案中,上述晶型V,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.6±0.2°,7.1±0.2°,11.5±0.2°,13.8±0.2°,14.3±0.2°。
本发明的一些方案中,上述晶型V,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.6±0.2°,7.1±0.2°,11.5±0.2°,13.8±0.2°,14.3±0.2°,17.0±0.2°,21.9±0.2°。
本发明的一些方案中,上述晶型V,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.6±0.2°,7.1±0.2°,8.9±0.2°,11.5±0.2°,13.8±0.2°,14.3±0.2°,17.0±0.2°,19.1±0.2°,21.9±0.2°。
本发明的一些方案中,上述晶型V,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.6±0.2°,7.1±0.2°,8.9±0.2°,11.5±0.2°,13.8±0.2°,14.3±0.2°,17.0±0.2°,18.3±0.2°,19.1±0.2°,20.7±0.2°,21.9±0.2°。
本发明的一些方案中,上述晶型V,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.6±0.2°,7.1±0.2°,8.9±0.2°,11.5±0.2°,13.8±0.2°,14.3±0.2°,17.0±0.2°,18.3±0.2°,19.1±0.2°,20.7±0.2°,21.9±0.2°,25.2±0.2°。
本发明的一些方案中,上述晶型V,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.6±0.2°,7.1±0.2°,8.9±0.2°,11.5±0.2°,13.8±0.2°,14.3±0.2°,17.0±0.2°,18.3±0.2°,19.1±0.2°,20.7±0.2°,21.9±0.2°,25.2±0.2°,27.2±0.2°。
本发明的一些方案中,上述晶型V,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 峰位置(2θ) 峰位置(2θ)
4.4 12.7 20.7
5.1 13.4 21.9
5.6 13.8 22.4
7.1 14.3 23.2
8.9 17.0 23.4
9.6 17.9 25.2
11.3 18.3 25.8
11.5 19.1 27.2
本发明的一些方案中,上述晶型V,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
4.4 6.7 12.7 6.7 20.7 11.6
5.1 7.1 13.4 4.1 21.9 19.0
5.6 86.7 13.8 40.6 22.4 6.6
7.1 37.6 14.3 100.0 23.2 5.2
8.9 10.9 17.0 12.6 23.4 8.8
9.6 7.0 17.9 8.3 25.2 8.3
11.3 20.2 18.3 11.8 25.8 5.3
11.5 36.9 19.1 11.4 27.2 8.9
本发明的一些方案中,上述晶型V,其具有基本上如图11所示的X-射线粉末衍射图谱。
本发明的一些方案中,上述晶型V,其差示扫描量热曲线在174.53±3℃有一个吸热峰。
本发明的一些方案中,上述晶型V,其具有基本上如图15所示的DSC图谱。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型VI,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:6.2±0.2°,9.4±0.2°,12.5±0.2°。
本发明的一些方案中,上述晶型VI,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:6.2±0.2°,9.4±0.2°,12.5±0.2°,15.2±0.2°,21.4±0.2°。
本发明的一些方案中,上述晶型VI,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.0±0.2°,6.2±0.2°,9.4±0.2°,12.5±0.2°,15.2±0.2°,21.4±0.2°,24.7±0.2°。
本发明的一些方案中,上述晶型VI,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.0±0.2°,6.2±0.2°,9.4±0.2°,12.5±0.2°,15.2±0.2°,21.4±0.2°,23.5±0.2°,24.7±0.2°。
本发明的一些方案中,上述晶型VI,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.0±0.2°,6.2±0.2°,9.4±0.2°,12.5±0.2°,15.2±0.2°,18.8±0.2°,21.4±0.2°,23.5±0.2°,24.7±0.2°。
本发明的一些方案中,上述晶型VI,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.0±0.2°,6.2±0.2°,9.4±0.2°,12.5±0.2°,14.0±0.2°,15.2±0.2°,18.8±0.2°,21.4±0.2°,23.5±0.2°,24.7±0.2°。
本发明的一些方案中,上述晶型VI,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 峰位置(2θ) 峰位置(2θ)
5.0 15.2 23.5
6.2 17.6 24.7
9.4 18.5 25.4
10.1 18.8 26.0
12.5 21.4 30.8
14.0 23.2  
本发明的一些方案中,上述晶型VI,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
5.0 11.5 15.2 22.3 23.5 10.1
6.2 35.8 17.6 2.9 24.7 10
9.4 100 18.5 6.6 25.4 4.1
10.1 3.2 18.8 6.6 26.0 4.8
12.5 65.6 21.4 23.9 30.8 3.2
14.0 5.2 23.2 7.3    
本发明的一些方案中,上述晶型VI,其具有基本上如图12所示的X-射线粉末衍射图谱。
本发明的一些方案中,上述晶型VI,其差示扫描量热曲线在45.54±3℃和163.80±3℃分别有两个吸热峰的起始点。
本发明的一些方案中,上述晶型VI,其差示扫描量热曲线在86.78±3℃和165.61±3℃分别有两个吸热峰。
本发明的一些方案中,上述晶型VI,其具有基本上如图13所示的DSC图谱。
本发明的一些方案中,上述晶型VI,其热重分析曲线在室温~120±3℃之间有3.153%±0.2%的失重,在120~180℃(±3℃)之间有1.500%±0.2%的失重。
本发明的一些方案中,上述晶型VI,其具有基本上如图13所示的TGA图谱。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型VII,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.7±0.2°,10.8±0.2°,21.8±0.2°。
本发明的一些方案中,上述晶型VII,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.7±0.2°,7.5±0.2°,10.8±0.2°,12.5±0.2°,21.8±0.2°。
本发明的一些方案中,上述晶型VII,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.7±0.2°,7.5±0.2°,10.8±0.2°,12.5±0.2°,17.4±0.2°,21.8±0.2°,25.1±0.2°。
本发明的一些方案中,上述晶型VII,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.7±0.2°,7.5±0.2°,10.8±0.2°,11.6±0.2°,12.5±0.2°,17.4±0.2°,18.0±0.2°,21.8±0.2°,25.1±0.2°。
本发明的一些方案中,上述晶型VII,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 峰位置(2θ) 峰位置(2θ)
5.7 14.7 18.7
7.5 15.1 20.0
10.8 16.2 21.8
11.6 17.4 23.3
12.5 18.0 25.1
本发明的一些方案中,上述晶型VII,其X-射线粉末衍射图谱在以下2θ角处具有特征衍射峰(±0.2°):
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
5.7 100.0 14.7 5.4 18.7 3.2
7.5 7.2 15.1 5.4 20.0 3.0
10.8 40.5 16.2 3.0 21.8 11.1
11.6 7.0 17.4 10.1 23.3 4.0
12.5 9.2 18.0 5.9 25.1 5.7
本发明的一些方案中,上述晶型VII,其具有基本上如图1所示的X-射线粉末衍射图谱。
本发明的一些方案中,上述晶型VII,其差示扫描量热曲线在47.52±3℃和151.56±3℃分别有两个吸热峰的起始点。
本发明的一些方案中,上述晶型VII,其差示扫描量热曲线在74.24±3℃和156.63±3℃分别有两个吸热峰。
本发明的一些方案中,上述晶型VII,其具有基本上如图2所示的DSC图谱。
本发明的一些方案中,上述晶型VII,其热重分析曲线在室温~150±3℃之间有8.053%±0.2%的失重。
本发明的一些方案中,上述晶型VII,其具有基本上如图2所示的TGA图谱。
另一方面,本发明提供了结晶形式的式(A)所示化合物、其溶剂合物或水合物的制备方法,包括:
(1)将式(A)所示化合物加入溶剂中,使其成混悬液或溶液;
(2)搅拌上述混悬液或溶液,析晶,分离固体;
任选地,还进一步包括干燥步骤,得到结晶形式的式(A)所示化合物、其溶剂合物或水合物。
本发明的一些方案中,上述的制备方法,其中,所述溶剂选自单一溶剂或混合溶剂,优选地,所述溶剂选自正庚烷、甲苯、乙酸乙酯、甲醇、乙二醇或水中的一种或两种。
本发明的一些方案中,上述制备方法,其中,所述混悬液可以通过向溶液中加入不良溶剂制备得到。
本发明的一些方案中,上述的制备方法,其中,搅拌温度为-20℃~70℃,优选为-10~60℃,进一步优选为0~50℃,更进一步优选为15~40℃。
本发明的一些方案中,上述的制备方法,其中,搅拌时间为0.1小时~75小时,优选为0.5小时~72小时。
本发明的一些方案中,上述的制备方法,其中,化合物与溶剂的重量-体积比为1g:1~90mL,优选为1g:5~50mL,进一步优选为1g:10~25mL。
本发明的一些方案中,上述的制备方法,其中,混合溶剂中两种溶剂的体积比为1:1~100,优选为1:1~50,进一步优选为1:1~20,进一步优选为1:1~5。两种溶剂中,以用量少者计为1,计算体积比。
本发明的一些方案中,上述的制备方法,其中,所述干燥的时间为0.1小时~75小时,优选为1小时~48小时,进一步优选为5小时~24小时,进一步优选为5小时~12小时,更进一步优选为5小时~10小时;所述干燥优选为真空干燥。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型I的制备方法,包括:
(1)将式(A)所示化合物加入溶剂中,使其成混悬液或溶液;
(2)搅拌上述混悬液或溶液,析晶,分离固体,得到式(A)所示化合物、其溶剂合物或水合物的晶型I。
本发明的一些方案中,上述的制备方法,其中,所述溶剂选自单一溶剂或混合溶剂,优选地,单一溶剂可以选自乙酸乙酯,混合溶剂可选自甲苯和正庚烷。
本发明的一些方案中,上述制备方法,其中,所述混悬液可以通过向溶液中加入不良溶剂制备得到。
本发明的一些方案中,上述的制备方法,其中,搅拌温度为-20℃~70℃,优选为-10~60℃进一步优选为0~50℃,更进一步优选为15~40℃。
本发明的一些方案中,上述的制备方法,其中,搅拌时间为0.1小时~75小时,优选为0.5小时~72小时。
本发明的一些方案中,上述的制备方法,其中,化合物与溶剂的重量-体积比为1g:1~90mL,优选为1g:5~50mL,进一步优选为1g:10~25mL。
本发明的一些方案中,上述的制备方法,其中,混合溶剂中两种溶剂的体积比为1:1~100,优选为1:1~50,进一步优选为1:1~20,进一步优选为1:1~5,进一步优选为1:1。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型II的制备方法,包括:
(1)将式(A)所示化合物加入溶剂中,使其成混悬液或溶液;
(2)搅拌上述混悬液或溶液,析晶,分离固体,得到式(A)所示化合物、其溶剂合物或水合物的晶型II。
或者,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型II的制备方法,包括:向晶型VII中加入水,搅拌或打浆,析晶,分离固体。
本发明的一些方案中,上述的制备方法,其中,所述溶剂为水。
本发明的一些方案中,上述的制备方法,其中,搅拌或打浆温度为-20℃~70℃,优选为-10~60℃,进一步优选为0~50℃,更进一步优选为15~40℃,更进一步优选为室温。
本发明的一些方案中,上述的制备方法,其中,搅拌或打浆时间为0.1小时~75小时,优选为0.5小时~72小时。
本发明的一些方案中,上述的制备方法,其中,化合物与溶剂的重量-体积比为1g:1~90mL,优选为1g:5~50mL,进一步优选为1g:10~25mL。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型III的制备方法,包括:
(1)将上述晶型II置于真空干燥箱中进行干燥,得到式(A)所示化合物、其溶剂合物或水合物的晶型III。
本发明的一些方案中,上述的制备方法,其中,所述真空干燥的温度为30~70℃,优选为40~60℃,进一步优选为40~50℃。
本发明的一些方案中,上述的制备方法,其中,所述真空干燥的时间为0.1小时~75小时,优选为1小时~48小时,进一步优选为5小时~24小时,进一步优选为5小时~12小时,更进一步优选为5小时~10小时。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型IV的制备方法,包括:
(1)将式(A)所示化合物加入溶剂中,使其成混悬液或溶液;
(2)搅拌上述混悬液或溶液,析晶,分离固体,得到式(A)所示化合物、其溶剂合物或水合物的晶型IV。
本发明的一些方案中,上述的制备方法,其中,所述溶剂为甲醇。
本发明的一些方案中,上述的制备方法,其中,搅拌温度为-20℃~70℃,优选为-10~60℃,进一步优选为0~50℃,更进一步优选为15~40℃,更进一步优选为室温。
本发明的一些方案中,上述的制备方法,其中,搅拌时间为0.1小时~75小时,优选为0.5小时~72小时,进一步优选为1小时~48小时,进一步优选为2小时~24小时。
本发明的一些方案中,上述的制备方法,其中,化合物与溶剂的重量-体积比为1g:1~90mL,优选为1g:5~50mL,进一步优选为1g:10~25mL,更进一步优选为1g:10~15mL。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型V的制备方法,包括:
(1)将上述晶型IV置于真空干燥箱中进行干燥,得到式(A)所示化合物、其溶剂合物或水合物的晶型V。
本发明的一些方案中,上述的制备方法,其中,所述真空干燥的温度为30~70℃,优选为40~60℃,进一步优选为40~50℃。
本发明的一些方案中,上述的制备方法,其中,所述真空干燥的时间为0.1小时~75小时,优选为1小时~48小时,进一步优选为5小时~24小时,进一步优选为5小时~12小时,更进一步优选为5小时~10小时。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型VI的制备方法,包括:
(1)将式(A)所示化合物加入溶剂中,使其成混悬液或溶液;
(2)搅拌上述混悬液或溶液,析晶,分离固体,得到式(A)所示化合物、其溶剂合物或水合物的晶型VI。
本发明的一些方案中,上述的制备方法,其中,所述溶剂为乙二醇。
本发明的一些方案中,上述的制备方法,其中,搅拌温度为-20℃~70℃,优选为-10~60℃,进一步优选为0~50℃,更进一步优选为15~40℃,更进一步优选为室温。
本发明的一些方案中,上述的制备方法,其中,搅拌时间为0.1小时~75小时,优选为0.5小时~72小时,进一步优选为1小时~48小时。
本发明的一些方案中,上述的制备方法,其中,化合物与溶剂的重量-体积比为1g:1~90mL,优选为1g:5~50mL,进一步优选为1g:10~25mL。
另一方面,本发明提供了式(A)所示化合物、其溶剂合物或水合物的晶型VII的制备方法,包括:
(1)将式(A)所示化合物加入溶剂中,使其成混悬液或溶液;
(2)搅拌上述混悬液或溶液,析晶,分离固体,得到式(A)所示化合物、其溶剂合物或水合物的晶型VII。
本发明的一些方案中,上述的制备方法,其中,所述溶剂为二氯甲烷。
本发明的一些方案中,上述的制备方法,其中,搅拌温度为-20℃~70℃,优选为-10~60℃,进一步优选为0~50℃,更进一步优选为15~40℃,更进一步优选为室温。
本发明的一些方案中,上述的制备方法,其中,搅拌时间为0.1小时~75小时,优选为0.5小时~72小时,进一步优选为1小时~48小时。
本发明的一些方案中,上述的制备方法,其中,化合物与溶剂的重量-体积比为1g:1~90mL,优选为1g:5~50mL,进一步优选为1g:10~25mL。
本发明的另一个目的在于提供了一种药物组合物,包含前述固体形式的式(A)化合物、其溶剂合物或水合物、结晶形式的式(A)化合物、其溶剂合物或水合物或其结晶混合物。
本发明的一些方案中,上述药物组合物,包含前述式(A)化合物、其溶剂合物或水合物的晶型I、晶型II、晶型III、晶型IV、晶型V、晶型VI中的一种或几种的混合物。
本发明的一些方案中,所述药物组合物包含前述固体形式的式(A)化合物、其溶剂合物或水合物、结晶形式的式(A)化合物、其溶剂合物或水合物或其结晶混合物,以及含有一种或多种可药用载体。
本发明的一些方案中,所述药物组合物包含前述式(A)化合物、其溶剂合物或水合物的晶型I、晶型II、晶型III、晶型IV、晶型V、晶型VI中的一种或几种的混合物,以及含有一种或多种可药用载体。
本发明还提供了上述固体形式的式(A)所示化合物、其溶剂合物或水合物、结晶形式的式(A)所示化合物、其溶剂合物或水合物、式(A)所示化合物、其溶剂合物或水合物的晶型I、晶型II、晶型III、晶型IV、晶型V、晶型VI、晶型VII或上述药物组合物在制备治疗BTK相关病症的药物上的应用。
另一方面,本发明还涉及上述固体形式的式(A)所示化合物、其溶剂合物或水合物、结晶形式的式(A)所示化合物、其溶剂合物或水合物、式(A)所示化合物、其溶剂合物或水合物的晶型I、晶型II、晶型III、晶型IV、晶型V、晶型VI、晶型VII或上述药物组合物,用于治疗BTK相关病症的应用。
又一方面,本申请还提供了用于治疗BTK相关病症的前述固体形式的式(A)所示化合物、其溶剂合物或水合物、结晶形式的式(A)所示化合物、其溶剂合物或水合物、式(A)所示化合物、其溶剂合物或水合物的晶型I、晶型II、晶型III、晶型IV、晶型V、晶型VI、晶型VII或前述药物组合物。
本发明的一些方案中,前述BTK相关病症涉及BTK蛋白表达、水平或活性的失调。
本发明的一些方案中,前述BTK相关病症包括肿瘤疾病或自身免疫性疾病;优选地,所述肿瘤疾病为血液瘤;进一步优选为白血病或淋巴瘤;进一步优选为B细胞淋巴瘤;更进一步优选为套细胞淋巴瘤、慢性淋巴细胞白血病、小淋巴细胞淋巴瘤、边缘区淋巴瘤、滤泡性淋巴瘤、华氏巨球蛋白血症或弥漫性大B细胞淋巴瘤。
另一方面,还提供了上述固体形式的式(A)所示化合物、其溶剂合物或水合物、结晶形式的式(A)所示化合物、其溶剂合物或水合物、式(A)所示化合物、其溶剂合物或水合物的晶型I、晶型II、晶型III、晶型IV、晶型V、晶型VI、晶型VII或上述药物组合物在制备治疗肿瘤疾病或自身免疫性疾病的药物中的应用。
本发明的一些方案中,所述肿瘤疾病为血液瘤;进一步优选为白血病或淋巴瘤;进一步优选为B细胞淋巴瘤;更进一步优选为套细胞淋巴瘤、慢性淋巴细胞白血病、小淋巴细胞淋巴瘤、边缘区淋巴瘤、滤泡性淋巴瘤、华氏巨球蛋白血症或弥漫性大B细胞淋巴瘤。
本发明的一些方案中,所述BTK相关病症涉及BTK蛋白表达、水平或活性的失调。
另一方面,本发明还涉及一种治疗患者的病症的方法,通过向患者施用治疗有效量的上述固体形式的式(A)所示化合物、其溶剂合物或水合物、结晶形式的式(A)所示化合物、其溶剂合物或水合物、式(A)所示化合物、其溶剂合物或水合物的晶型I、晶型II、晶型III、晶型IV、晶型V、晶型VI、晶型VII或上述药物组合物,所述患者的病症为BTK相关病症。
本发明的一些方案中,上述的治疗患者的病症的方法,其中,所述BTK相关病症包括肿瘤疾病或自身免疫性疾病;优选地,所述肿瘤疾病为血液瘤;进一步优选为白血病或淋巴瘤;进一步优选为B细胞淋巴瘤;更进一步优选为套细胞淋巴瘤、慢性淋巴细胞白血病、小淋巴细胞淋巴瘤、边缘区淋巴瘤、滤泡性淋巴瘤、华氏巨球蛋白血症或弥漫性大B细胞淋巴瘤。
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。
术语“溶剂化物”或“溶剂合物”是指一个或多个溶剂分子与本申请的式2所示化合物所形成的缔合物,包括同时含有水分子和一种或多种其它溶剂分子的缔合物。
术语“水合物”是指一个或多个水分子与本申请的式2所示化合物所形成的缔合物。
术语“基本上如图所示”是指X-射线粉末衍射图谱中至少50%,或至少60%,或至少70%,或至少80%,或至少90%,或至少95%,或至少96%,或至少97%,或至少98%,或至少99%的峰显示在其图中;或者DSC图谱中,各吸热峰或放热峰的起始点或峰值温度的误差范围在±10℃范围内,优选5℃范围内,进一步优选3℃范围内;或者TGA图谱中,失重温度起点或终点的误差范围在±10℃范围内,优选5℃范围内,进一步优选3℃范围内,失重百分比在±0.5%范围内,优选±0.3%范围内,±0.2%范围内。进一步地,当产品中某种晶型的含量逐渐降低时,其粉末X-射线衍射图谱中的一些归属于该晶型的衍射峰可能会由于仪器的检测灵敏度的因素而变少。
术语“特征衍射峰”是指在X-射线粉末衍射图谱中,可用于代表该晶型的衍射峰,其与衍射峰的峰位置、峰形和相对峰强度有关,例如,小角度峰,峰形尖锐,且相对峰强度至少2.9%以上,或至少3%以上,或至少5%以上,或至少10%以上,或至少20%以上,或至少30%以上,或至少40%以上,或至少50%以上,或至少60%以上,或至少70%以上,或至少75%以上的衍射峰。
术语“肿瘤”包含良性肿瘤、恶性肿瘤和交界性肿瘤,其中恶性肿瘤又统称为癌症。
术语“治疗”一般是指获得需要的药理和/或生理效应,包括部分或完全稳定或治愈疾病和/或由于疾病产生的作用。
本文使用的“治疗”涵盖了对患者疾病的任何治疗,包括:(a)抑制疾病的症状,即阻止其发展;或(b)缓解疾病的症状,即,导致疾病或症状退化。术语“有效量”或“治疗有效量”意指(i)治疗特定疾病,或(ii)减轻、改善或消除特定疾病的一种或多种症状的本申请化合物的用量。
术语“治疗有效量”意指化合物被给予患者用于治疗疾病时,足以实现对该疾病的治疗的量。构成“治疗有效量”的本申请化合物的量取决于该化合物、疾病状态及其严重性、给药方式以及待被治疗的哺乳动物的年龄而改变,但可例行性地由本领域技术人员根据其自身的知识及本公开内容而确定。
术语“可药用载体”或“药学上可接受的辅料”是指对有机体无明显刺激作用,而且不会损害活性化合物的生物活性及性能的那些载体或辅料。
如无特别说明,本申请所述的“加热温度”、“降温温度”或“析晶温度”,单位为℃或摄氏度,误差范围可以为±10、±5、±4、±3、±2或±1℃。
本发明的中间体化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明具体实施方式的化学反应是在合适的溶剂中完成的,所述的溶剂须适合于本发明的化学变化及其所需的试剂和物料。为了获得本发明的化合物,有时需要本领域技术人员在已有实施方式的基础上对合成步骤或者反应流程进行修改或选择。
下面会通过实施例具体描述本发明,这些实施例并不意味着对本发明的任何限制。
本发明所使用的所有溶剂是市售的,无需进一步纯化即可使用。
在该制备方法以及本发明中,使用的术语如下所述:
DCM:二氯甲烷;DIAD:偶氮二甲酸二异丙酯;DIPEA:二异丙基乙胺;DMF:N,N-二甲基甲酰胺;EA:乙酸乙酯;HATU:2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯;NBS:N-溴代丁二酰亚胺;NIS:N-碘代丁二酰亚胺;Pd(dppf)Cl 2:[1,1'-双(二苯基膦基)二茂铁]二氯化钯;Pd(PPh 3) 4:四(三苯基膦)钯;PdCl 2:二氯化钯;Pd(OAc) 2:醋酸钯;Pd(PPh 3) 2Cl 2:双三苯基磷二氯化钯;PE:石油醚;THF:四氢呋喃;DMSO:二甲基亚砜。
技术效果
本发明提及的式(A)所示化合物的晶型具有良好的结晶度,优选晶型具备良好的稳定性,易于成药,生物利用度高;本发明化合物(A)具有优良的BTK抑制活性、且保持较好的BTK抑制选择性、具有优良的体内抗肿瘤活性、良好的口服给药性能,对由BTK异常表达引起的疾病可以提供更有效的治疗,且在不同种属肝微粒体(人肝微粒体、大鼠肝微粒体和小鼠肝微粒体)作用下,代谢产物种类和占比少,基本以原型药物为主(60min:84%-98%),代谢稳定性更好。
1.1 X-射线粉末衍射(X-ray powder diffractometer,XRPD)
仪器型号:PANalytical EMPYREAN X-射线衍射仪
测试方法:大约1~2mg样品用于XRPD检测
详细的XRPD参数如下:
X射线发生器:Cu,Kα,
Figure PCTCN2021125689-appb-000003
光管电压:45kV,光管电流:40mA
扫描范围:3°-40°(2θ)
扫描步长:0.013°
扫描时间:20.4秒/步
光管类型:Empyrean Cu LFF HR(94300337310x)DK 420877
旋转时间:1s
样品盘:零背景样品盘
采集软件:HighScore Data Collector
分析软件:Jade 6。
1.2差示扫描量热分析(Differential Scanning Calorimeter,DSC)
仪器型号:Discovery DSC 250差示扫描量热仪
测试方法:取样品(1~5mg)置于DSC样品盘中,样品盘加盖并扎孔,将样品在25℃平衡后以10℃/min的升温速率加热至最终温度。
样品量:1~5mg
气流种类:氮气
流速:50mL/min
加热起始温度:25℃
终止温度:300℃。
1.3热重分析(Thermal Gravimetric Analyzer,TGA)
仪器型号:Discovery TGA 55热重分析仪
测试方法:将样品置于已去皮的开口铝制样品盘中,样品质量在TGA加热炉内自动称量后,将样品以10℃/min的速率加热至最终温度。
样品量:1~5mg
气流种类:氮气
流速:60mL/min
加热起始温度:25~30℃
终止温度:300℃。
1.4动态水分吸脱附分析(DVS)
仪器型号:Vsorp-Enhanced动态水蒸气吸附仪
测试方法:将足量的样品加入到仪器中模拟动态水蒸气吸附,并且记录25℃时不同湿度平衡时重量的变化。
据样品在吸附过程中达到80%RH时的增重,对样品吸湿性大小进行分类:
(1)潮解:吸收足量水分形成液体
(2)极具吸湿性:吸湿增重不小于15%
(3)有吸湿性:吸湿增重小于15%但不小于2%
(4)略有吸湿性:吸湿增重小于2%但不小于0.2%
(5)无吸湿性:吸湿增重小于0.2%。
1.4.1晶型I的测试参数:
样品重量:                        96.821mg
样品温度:                        25℃
循环时间:                        10min
1.4.2晶型II的测试参数:
样品重量:                        117.086mg
样品温度:                        25℃
循环时间:                        10min
1.5高效液相色谱(HPLC)
仪器型号:Agilent HPLC 1260高效液相色谱仪
色谱柱:CORTECS C18,4.6*150mm,2.7μm
测试条件:波长230nm;柱温30℃
1.6核磁共振波谱(Nuclear Magnetic Resonance Spectroscopy,NMRS)
仪器型号:Bruker AVANCE III HD 300/400
内容及测试溶剂: 1H-NMR,测试溶剂为DMSO-d6。
附图说明
图1:实施例1的晶型VII的X-射线粉末衍射图谱。
图2:实施例1的晶型VII的DSC-TGA图谱。
图3:实施例2的晶型I的X-射线粉末衍射图谱。
图4:实施例2的晶型I的DSC-TGA图谱。
图5:实施例4的晶型II的X-射线粉末衍射图谱。
图6:实施例4的晶型II的DSC-TGA图谱。
图7:实施例5的晶型III的X-射线粉末衍射图谱。
图8:实施例5的晶型III的DSC-TGA图谱。
图9:实施例6的晶型IV的X-射线粉末衍射图谱。
图10:实施例6的晶型IV的DSC-TGA图谱。
图11:实施例7的晶型V的X-射线粉末衍射图谱。
图12:实施例8的晶型VI的X-射线粉末衍射图谱。
图13:实施例8的晶型VI的DSC-TGA图谱。
图14:实施例2的晶型I的DVS图。
图15:实施例7的晶型V的DSC-TGA图谱。
图16:REC-1异种移植瘤模型的实验结果示意图。
图17:TMD8异种移植瘤模型的实验结果示意图。
图18:晶型I研磨前后XRPD叠合图。
具体实施方式
为了更好的理解本发明的内容,下面结合具体实施例来做进一步的说明,但具体的实施方式并不是对本发明的内容所做的限制。
制备例1:式(A)化合物的制备
1、中间体3的合成
Figure PCTCN2021125689-appb-000004
在250mL圆底烧瓶中加入4-氯-5-碘-7H-吡咯并[2,3-d]嘧啶(原料2,17.28g,1eq)和无水碳酸钾(2eq),真空干燥除水。加入干燥的DMF作为溶剂,粉碎的(S)-甲磺酸2-((叔丁氧基羰基)氨基)-丁-3-烯-1-基酯(原料1,24.6g,1.5eq),置换氮气。于55℃下加热搅拌12小时,时间可适当延长以确保反应完全。
反应完成后,加入水和乙酸乙酯萃取三次,合并酯层,用水反萃一次,用饱和食盐水洗。无水硫酸钠干燥。干法过柱(洗脱液:CHCl 3:MeOH=100:1,v/v,下同)得产品(S)-(1-(4-氯-5-碘-7H-吡咯并[2,3-d]嘧啶-7-基)丁-3-烯-2-基)氨基甲酸叔丁酯(中间体3,19.28g),产率为69.5%。
1H NMR(300MHz,CDCl 3)δ8.60(s,1H),7.39(s,1H),5.82(ddd,J=17.1,10.5,5.5Hz,1H),5.33-5.14(m,2H),4.80(s,1H),4.63-4.51(m,1H),4.51-4.42(m,1H),4.35(s,1H),1.33(s,9H)。ee>99.5%。
2、中间体4的合成
Figure PCTCN2021125689-appb-000005
在350mL耐压管中加入(S)-(1-(4-氯-5-碘-7H-吡咯并[2,3-d]嘧啶-7-基)丁-3-烯-2-基)氨基甲酸叔丁酯(中间体3,9.2g),加入1,4-二氧六环(40mL)作为溶剂,加入氨水(40mL)。于120℃下密封反应2.5小时。
反应完成后,冷却至室温,加入水和乙酸乙酯萃取,合并酯层,用饱和食盐水洗。无水硫酸钠干燥。干法过柱(洗脱液:CHCl 3:MeOH=30:1)得产品(S)-(1-(4-氨基-5-碘-7H-吡咯并[2,3-d]嘧啶-7-基)丁-3-烯-2-基)氨基甲酸叔丁酯(中间体4,6.86g),产率为78.0%。
1H NMR(300MHz,CDCl 3)δ8.25(s,1H),7.05(s,1H),5.87-5.74(m,1H),5.72(s,2H),5.34-5.13(m,3H),4.56-4.43(m,1H),4.34(dd,J=14.8,4.9Hz,1H),4.30-4.15(m,1H),1.35(s,9H)。ee>99.5%。
3、中间体6的合成
Figure PCTCN2021125689-appb-000006
在1L圆底烧瓶中加入(S)-(1-(4-氨基-5-碘-7H-吡咯并[2,3-d]嘧啶-7-基)丁-3-烯-2-基)氨基甲酸叔丁酯(中间体4,32.9g,1eq),N-(吡啶-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯甲酰胺(原料5,34.8g,1.4eq)和四(三苯基膦)钯(17.7g,0.2eq)。加入1,4-二氧六环(383mL)作为溶剂,并置换N 2。在搅拌下加入2M碳酸钠溶液(76.6mL)。于90℃下回流搅拌5小时。
加入水与乙酸乙酯萃取,合并酯层,用饱和食盐水洗。无水硫酸钠干燥。干法过柱,先用EA作为洗脱液除去大部分杂质,然后用CHCl 3:MeOH=30:1的混合物作为洗脱液。产品可能含有少量杂质,可用PE重结晶析出纯品。得(S)-(1-(4-氨基-5-(4-(吡啶-2-基氨基甲酰基)苯基)-7H-吡咯并[2,3-d]嘧啶-7-基)丁-3-烯-2-基)氨基甲酸叔丁酯(中间体6,28.4g),产率为74.3%。
4、中间体7的合成
Figure PCTCN2021125689-appb-000007
在1L圆底烧瓶中加入(S)-(1-(4-氨基-5-(4-(吡啶-2-基氨基甲酰基)苯基)-7H-吡咯并[2,3-d]嘧啶-7-基)丁-3-烯-2-基)氨基甲酸叔丁酯(中间体6,32.7g,1eq),加入600mL DMF作为溶剂。在搅拌下缓慢加入N-溴代丁二酰亚胺(12.8g,1.1eq),于室温下搅拌过夜。
反应完成后,加入水与乙酸乙酯萃取,合并酯层,用水反萃一次,饱和食盐水洗。无水硫酸钠干燥。干法过柱,首先用CHCl 3:MeOH=50:1的混合物作为洗脱液,然后换CHCl 3:MeOH=30:1的混合物作为洗脱液。得产品(S)-(1-(4-氨基-6-溴-5-(4-(吡啶-2-基氨基甲酰基)苯基)-7H-吡咯并[2,3-d]嘧啶-7-基)丁-3-烯-2-基)氨基甲酸叔丁酯(中间体7,25.8g),产率为68.2%。
5、中间体8的合成
Figure PCTCN2021125689-appb-000008
在250mL圆底烧瓶中加入(S)-(1-(4-氨基-6-溴-5-(4-(吡啶-2-基氨基甲酰基)苯基)-7H-吡咯并[2,3-d]嘧啶-7-基)丁-3-烯-2-基)氨基甲酸叔丁酯(中间体7,11.9g,1eq)和[1,1’-双(二苯基膦基)二茂铁]二氯化钯(1.66g,0.11eq),加入51mL THF作为溶剂,并置换氮气,多置换几次以确保完全。在搅拌下加入4M氢氧化钠溶液(8.2mL)。于85℃下回流搅拌15小时。
反应完全后,加入水与乙酸乙酯萃取,合并酯层,用饱和食盐水洗。无水硫酸钠干燥。干法过柱(洗脱液:CHCl 3:MeOH=30:1)得产品(S)-(4-氨基-6-亚甲基-5-(4-(吡啶-2-基氨基甲酰基)苯基)-7,8-二氢-6H-嘧啶并[5,4-b]吡呤-7-基)氨基甲酸叔丁酯(中间体8,8.79g),产率为85.9%。
6、化合物A的合成
Figure PCTCN2021125689-appb-000009
在250mL圆底烧瓶中加入(S)-(4-氨基-6-亚甲基-5-(4-(吡啶-2-基氨基甲酰基)苯基)-7,8-二氢-6H-嘧啶并[5,4-b]吡呤-7-基)氨基甲酸叔丁酯(中间体8,2.75g,1eq),加入110mL DCM作为溶剂。于搅拌下逐滴加入三氟乙酸(10.5mL)。于室温下搅拌3小时。反应完成后,直接将反应液旋干,多用甲醇带几次将三氟乙酸带出,旋干后得到氨基脱Boc保护粗品,直接投下一步。
将上一步产物移至250mL圆底烧瓶中,再加入三乙胺(1eq),搅拌五分钟后再加入2-丁炔酸(0.511g,1.1eq)和2-(7-氧化苯并三氮唑)-N,N,N’,N’-四甲基脲六氟磷酸酯(2.31g,1.1eq),加入100mL DCM作为溶剂。冰水浴降温至0℃,逐滴加入三乙胺(1.54mL+0.77mL)。逐渐 升温至室温,于室温下搅拌1.5小时。反应液呈微黄色。加入水和DCM萃取,合并有机相,用饱和食盐水洗。无水硫酸钠干燥,柱层析(CHCl 3:MeOH=30:1)后得终产物,即化合物A(1.88g),产率为73.3%。
1H NMR(400MHz,CDCl 3)δ8.98(s,1H),8.43(dt,J=8.3,1.0Hz,1H),8.34(ddd,J=5.0,1.9,0.9Hz,1H),8.22(s,1H),8.09-8.03(m,2H),7.81(ddd,J=8.4,7.4,1.9Hz,1H),7.69-7.63(m,2H),7.13(ddd,J=7.4,4.9,1.0Hz,1H),6.55(d,J=8.2Hz,1H),5.67(m,J=8.1,5.7,2.6Hz,1H),5.56(d,J=2.3Hz,1H),5.40(s,2H),5.27(d,J=2.3Hz,1H),4.70(dd,J=11.7,8.1Hz,1H),4.09-3.99(m,1H),1.99(s,3H)。
实施例1:式(A)化合物晶型VII的制备
室温,称取约20mg制备例1样品加入到适宜容积的玻璃瓶中,加入二氯甲烷(0.5mL),用封口膜将其密封,搅拌3天,过滤得固体。取样品进行X-射线粉末衍射,显示为结晶状固体(晶型VII),谱图见附图1,其XRPD衍射峰数据见表1。取样品进行1H-NMR测试,显示样品中含有约10%的二氯甲烷。取样品进行DSC-TGA测试,DSC图显示有两个吸热峰,吸热峰起始点温度分别为47.52℃和151.56℃,峰值温度分别为74.24℃和156.63℃,TGA图显示样品在室温~150℃之间有8.053%的失重,见附图2。
表1 实施例1晶型VII的XRPD衍射峰数据表
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
5.705 100 15.146 5.4 21.829 11.1
7.517 7.2 16.247 3.0 22.472 2.2
9.894 2.2 17.417 10.1 23.300 4.0
10.787 40.5 18.033 5.9 25.059 5.7
11.587 7.0 18.729 3.2 26.859 1.9
12.481 9.2 19.674 2.7 29.222 2.0
12.875 2.9 20.037 3.0 29.642 2.6
13.477 1.6 20.463 2.5 30.913 1.0
14.449 2.9 20.905 1.6    
14.699 5.4 21.316 2.9    
实施例2:式(A)化合物晶型I的制备
室温,称取20mg制备例1样品加入到适宜容积的玻璃瓶中,加入总体积为0.8mL的正庚烷/甲苯混合溶剂(V/V=1/1)中,用封口膜将其密封,搅拌3天,过滤得固体。取样品进行X-射线粉末衍射,显示为结晶状固体(晶型I,无水晶型),且结晶度良好,谱图见附图3,其XRPD衍射峰数据见表2。取样品进行DSC-TGA测试,DSC图显示在272.89℃有一个放热峰的起始点,峰值温度为274.74℃,见附图4。
表2 实施例2晶型I的XRPD衍射峰数据表
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
4.799 45.9 20.844 9.9 29.220 6.0
6.820 1.2 21.658 11.2 29.841 1.4
9.654 2.9 22.499 9.0 30.193 4.6
11.561 100.0 22.722 8.4 30.377 8.4
12.470 1.1 23.404 60.4 31.349 2.4
13.649 56.0 23.679 9.1 33.224 1.7
14.490 3.2 24.205 14.9 33.884 1.6
16.157 25.9 24.771 16.4 34.646 1.4
16.695 8.6 25.151 5.2 35.208 1.6
17.588 2.4 26.095 20.7 35.762 1.1
17.955 1.1 26.542 1.1 36.888 1.1
18.742 34.1 26.946 1.3 38.100 0.7
19.374 10.9 27.212 1.9 38.650 1.6
19.821 4.3 27.579 8.4 39.397 1.6
20.094 3.8 28.198 3.7    
20.529 5.2 28.549 5.1    
实施例3:式(A)化合物晶型I的制备
室温,称取20mg制备例1样品加入到适宜容积的玻璃瓶中,加入乙酸乙酯(0.5mL)中,用封口膜将其密封,搅拌3天。过滤得固体。取样品进行X-射线粉末衍射,显示为晶型I。
实施例4:式(A)化合物晶型II的制备
称取约20mg晶型VII加入到适宜容积的玻璃瓶中,加入水(0.5mL),用封口膜将其密封,室温打浆3天,过滤得固体。取样品进行X-射线粉末衍射,显示为结晶状固体(晶型II),且结晶度良好,谱图见附图5,其XRPD衍射峰数据见表3。取样品进行DSC-TGA测试,DSC图显示样品有三个吸热峰,吸热峰起始点温度分别为49.26℃、84.59℃和168.35℃,峰值温度分别为69.36℃、98.45℃和176.96℃,TGA图显示样品在室温~100℃之间有3.417%的失重,见附图6。
表3 实施例4晶型II的XRPD衍射峰数据表
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
4.260 18.8 18.010 28.3 26.320 33.7
5.201 4.2 18.245 18.2 28.026 6.4
5.201 4.2 19.136 14.3 28.396 2.5
6.965 29.6 20.724 4.4 29.115 4.7
7.858 27.3 20.871 7.7 30.680 11.2
8.514 96.2 21.264 16.1 30.953 5.9
10.589 83.8 22.117 49.3 31.854 3.1
11.508 14.4 23.235 6.7 32.436 2.5
12.847 11.5 23.525 5.1 32.885 2.9
14.133 9.4 23.807 3.8 34.800 2.7
15.041 100.0 25.110 13.9 35.720 4.3
15.843 3.6 25.349 49.7 37.546 3.4
17.101 28 25.833 9.5 39.565 4.2
实施例5:式(A)化合物晶型III的制备
取晶型II置于40℃真空干燥箱中干燥12h,得固体。取样品进行X-射线粉末衍射,显示为结晶状固体(晶型III,无水晶型),且结晶度良好,谱图见附图7,其XRPD衍射峰数据见表4。将晶型III加热到160℃再进行X-射线粉末衍射,其XRPD谱图未发生改变。取样品进行DSC-TGA测试,DSC图显示样品有三个吸热峰,吸热峰起始点温度分别为38.13℃、75.07℃和167.19℃,峰值温度分别为57.40℃、96.32℃和177.55℃,TGA图谱显示90℃之前有一段3.075%的失重,见附图8。
表4 实施例5晶型III的XRPD衍射峰数据表
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
4.301 68.2 13.580 3.5 22.275 45.6
7.071 45.2 14.569 20.4 25.518 67.7
7.950 47.6 15.133 93.3 26.083 24.7
8.645 72.5 16.156 28.0 26.503 26.9
8.999 15.0 17.246 22.1 28.185 4.9
10.353 48.7 18.089 24.4 29.262 6.6
10.642 100.0 18.428 20.8 30.904 8.8
11.639 11.1 19.255 22.1 31.060 10.0
13.046 6.1 21.381 17.9 32.029 3.1
实施例6:式(A)化合物晶型IV的制备
称取约20mg制备例1样品加入到适宜容积的玻璃瓶中,加入甲醇(0.3mL),用封口膜将其密封,室温搅拌,待析出大量固体后过滤。取样品进行X-射线粉末衍射,显示为结晶状固体(晶型IV),且结晶度良好,谱图见附图9,其XRPD衍射峰数据见表5。取样品进行DSC-TGA测试,DSC图显示样品有两个吸热峰,吸热峰起始点温度分别为45.75℃和159.89℃,峰值温度分别为70.65℃和166.96℃,TGA图显示样品在室温~200℃之间有7.863%的失重,见附图10。
表5 实施例6晶型IV的XRPD衍射峰数据表
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
5.061 14.1 19.005 16.4 26.319 11.7
6.322 44.7 20.357 12.7 26.742 2.7
9.657 52.2 21.934 9.8 27.473 3.8
11.649 4.4 23.234 15.9 29.263 3.9
12.769 100.0 23.785 6.9 32.439 2.9
14.095 17.5 24.127 5.1 37.220 1.7
15.631 9.5 25.059 5.6    
17.797 5.6 25.780 4.5    
实施例7:式(A)化合物晶型V的制备
取晶型IV置于40℃真空干燥箱中干燥12h,得固体。取样品进行X-射线粉末衍射,显示为结晶状固体(晶型V,无水晶型),且结晶度良好,谱图见附图11,取样品进行DSC-TGA测试,DSC图显示样品有两个吸热峰,吸热峰起始点温度分别为36.14℃和166.61℃,峰值温度分别为65.23℃和174.53℃,TGA图显示样品在室温~280℃之间有0.874%的失重,见附图15。其XRPD衍射峰数据见表6。
表6 实施例7晶型V的XRPD衍射峰数据表
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
4.432 6.7 13.396 4.1 22.406 6.6
5.114 7.1 13.781 40.6 23.232 5.2
5.613 86.7 14.279 100.0 23.431 8.8
6.335 3.0 16.997 12.6 25.191 8.3
7.109 37.6 17.339 2.8 25.833 5.3
8.501 1.7 17.902 8.3 26.581 2.6
8.895 10.9 18.323 11.8 27.172 8.9
9.564 7.0 19.125 11.4 28.104 2.0
10.670 2.9 19.466 2.4 28.459 1.9
11.299 20.2 19.871 1.6 29.335 1.7
11.508 36.9 20.713 11.6 32.343 1.5
12.732 6.7 21.908 19    
实施例8:式(A)化合物晶型VI的制备
称取约20mg制备例1样品加入到适宜容积的玻璃瓶中,加入乙二醇(0.5mL),用封口膜将其密封,室温打浆3天,过滤得固体。取样品进行X-射线粉末衍射,显示为结晶状固体(晶型VI),且结晶度良好,谱图见附图12,其XRPD衍射峰数据见表7。取样品进行DSC-TGA测试,DSC图显示样品有两个吸热峰,吸热峰起始点温度分别为45.54℃和163.80℃,峰值温度分别为86.78℃和165.61℃,TGA图显示样品在室温~120℃之间有3.153%的失重,在120~180℃之间有1.500% 的失重,见附图13。
表7 实施例8晶型VI的XRPD衍射峰数据表
峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度% 峰位置(2θ) 相对强度%
4.984 11.5 20.385 2.1 29.560 1.1
6.217 35.8 21.409 23.9 30.849 3.2
9.407 100.0 23.247 7.3 31.882 0.9
10.075 3.2 23.509 10.1 32.377 0.6
11.615 1.2 24.745 10 34.078 0.7
12.547 65.6 25.360 4.1 34.485 1.9
13.766 1.6 25.992 4.8 35.744 0.5
14.041 5.2 26.516 1.0 36.757 1.0
15.212 22.3 27.121 1.1 37.180 1.2
17.601 2.9 27.856 1.7 37.534 1.2
18.547 6.6 28.330 0.7 38.568 0.9
18.796 6.6 28.840 1.3    
19.175 0.6 29.288 0.7    
实验例1:式(A)化合物I晶型和II晶型的固体稳定性实验
考察式(A)化合物晶型I和晶型II在高湿(40℃/75%RH,敞口)条件下放置7天的稳定性。
分别称取式(A)化合物晶型I(5mg)和晶型II(5mg),置于样品瓶的底部,摊成薄薄一层。样品于第7天取样检测,检测结果与0天的初始检测结果进行比较,试验结果见下表8所示:
表8 式(A)化合物晶型I和晶型II的固体稳定性试验结果
Figure PCTCN2021125689-appb-000010
“RH”:相对湿度。
结论:式(A)化合物晶型I和晶型II均具有良好的稳定性。
实验例2:式(A)化合物晶型I的吸湿性研究
取式(A)化合物实施例2晶型I(约97mg)置于DVS样品室内进行测试。取DVS后的样品进行X-射线粉末衍射。
实验结果:
式(A)化合物晶型I的DVS谱图如图14所示。
式(A)化合物晶型I在25℃和80%RH下的吸湿增重为0.9%,在25℃和90%RH下的吸湿增重为1.0%,略有吸湿性,DVS后晶型未发生转变。
实验例3:研磨试验
取适量的晶型I(实施例2,约10mg)样品置于研钵中进行研磨,约5分钟,收集固体进行X-射线粉末衍射。晶型I研磨前后XRPD叠合图参见图18。
结论:研磨后晶型未发生改变,说明晶型I在机械加工过程中可保持稳定。
实验例4:布鲁顿激酶(BTK)分子水平酶活抑制活性评价
将酶反应底物Poly(Glu,Tyr) 4:1用无钾离子的PBS(10mM磷酸钠缓冲液,150mM NaCl,pH 7.2-7.4)稀释成20μg/mL包被酶标板,在37℃下反应12-16小时后,用200μL/孔的T-PBS(含0.1%Tween-20的PBS)洗板三次,于37℃烘箱中干燥酶标板1-2小时。在以上包被底物的酶标板中,首先加入用反应缓冲液(50mM HEPES pH 7.4,50mM MgCl 2,0.5mM MnCl 2,0.2mM Na 3VO 4,1mM DTT)稀释的ATP溶液49μL/孔(终浓度为5μM)。每孔中加入1μL待测试化合物(化合物孔)或含相应浓度的DMSO(阴性对照孔),每次实验需设无酶对照孔。再加入50μL以反应缓冲液稀释的BTK酪氨酸激酶蛋白启动反应。
将上述反应体系置于37℃摇床(100rpm)中1小时,然后T-PBS洗板三次,加入一抗PY99100μL/孔(Santa Cruz),37℃摇床反应0.5小时。T-PBS洗板后,加入辣根过氧化物酶标记的羊抗鼠二抗稀释液100μL/孔,37℃摇床反应0.5小时。T-PBS洗板后,加入2mg/mL的OPD显色液100μL/孔,25℃避光反应1-10分钟。然后加入2M H 2SO 4 50μL/孔中止反应,用可调波长式微孔板酶标仪SPECTRA MAX Plus384读数,波长为490nm。
以化合物S1、S10、依鲁替尼、阿卡替尼、S18s、S19s和S20s作为阳性对照化合物,其中,化合物S1、S10、S18s、S19s和S20s采用现有技术(例如CN108101905A)中公开的方法或类似方法制备,依鲁替尼和阿卡替尼购自Selleck公司。
各化合物的抑制率通过下列公式求得:
Figure PCTCN2021125689-appb-000011
IC 50值采用酶标仪随机附带软件以四参数法回归求得。结果列于下表9中。
表9 不同化合物对BTK的抑制作用
化合物 IC 50(nM)
S1 ~1
S10 <10
依鲁替尼 ~1
阿卡替尼 ~10
S18s ~1
S19s ~1
S20s ~1
化合物A 0.5
注:使用制备例1样品检测。
以上结果表明,化合物A对BTK的抑制活性优于前期化合物S1、S10、S18s、S19s和S20s,也优于目前已上市的第一代BTK抑制剂依鲁替尼和第二代BTK抑制剂阿卡替尼。
实验例5:化合物对人B淋巴瘤细胞Ramos(Burkitt淋巴瘤)和人弥漫大B淋巴瘤细胞TMD8的体外增殖抑制活性检测
将细胞悬液(Ramos:10000细胞/孔;TMD8:12000细胞/孔)接种于96孔板中,于37℃培养箱静置2小时待细胞状态稳定后,每孔加入不同浓度的受试化合物(每个浓度设3个复孔),并同时设置空白对照(仅包含培养液,不含细胞的孔)、阴性对照(仅加细胞,不加化合物的孔)及阳性化合物对照。加药处理72h后,每孔加入20μL MTT(5mg/mL)于37℃孵育4h,加入100μL三联液(10%SDS,5%异丁醇,0.01M HCl),37℃放置过夜。用可调波长式微孔板酶标仪SPECTRAmax Plus384在570nm波长条件下测定OD值。
化合物的抑制率通过下列公式求得:
Figure PCTCN2021125689-appb-000012
IC 50值采用酶标仪随机附带软件以四参数法回归求得。实验独立重复3次,其结果列于下表10中。
同样以上文所述化合物S1、S10、依鲁替尼、阿卡替尼、S18s、S19s和S20s作为阳性对照化合物。
表10 不同化合物对Ramos细胞和TMD8细胞的增殖抑制活性
化合物 Ramos细胞IC 50 TMD8细胞IC 50
S1 94.73μM 0.006μM
S10 8.72μM 0.030μM
依鲁替尼 12.91μM 0.005μM
阿卡替尼 38.16μM 0.023μM
化合物A 3.15μM 0.003μM
S18s 5.04μM 0.016μM
S19s 0.017μM
S20s 14.3μM 0.004μM
以上结果表明,在细胞水平,化合物A对B细胞淋巴瘤的增殖抑制能力优于前期化合物S1、S10、S18s、S19s和S20s,也优于目前已上市的第一代BTK抑制剂依鲁替尼和第二代BTK抑制剂阿卡替尼。进一步需要说明的是,与其他化合物相比,本发明的化合物A对于Ramos细胞具有较高的增殖抑制活性,且对于TMD8细胞具有更高的增殖抑制活性。
实验例6:体内抗肿瘤活性评价
实验动物:
TMD8异种移植瘤模型
1)种属:小鼠
2)品系:CB-17SCID
3)周龄及体重:6-8周;18-22g
4)性别:雌性
5)供应商:北京维通利华实验动物技术有限公司
REC-1异种移植瘤模型
1)种属:小鼠
2)品系:BALB/c裸小鼠
3)周龄及体重:6-8周;17-20g
4)性别:雌性
5)供应商:上海灵畅生物科技有限公司
细胞培养:人淋巴癌TMD8细胞体外悬浮培养,培养条件为RPMI 1640培养基(供应商:gibco;货号:22400-089;生产批号:4868546)中加10%胎牛血清,100U/mL青霉素和100μg/mL链霉素,37℃ 5%CO 2培养。一周两次进行常规处理传代。当细胞饱和度为80%-90%时,收取细胞,计数,接种。
人套细胞淋巴癌REC-1细胞体外悬浮培养,培养条件为RPMI 1640培养基(供应商:gibco;货号:22400-089;生产批号:1868795)中加10%胎牛血清,100U/mL青霉素和100μg/mL链霉素,37℃ 5%CO2培养。一周两次进行常规处理传代。当细胞饱和度为80%-90%时,收取细胞,计数,接种。
肿瘤细胞接种:将0.2mL 10×10 6个人淋巴癌TMD8细胞皮下接种于每只裸小鼠的右后背(PBS:Matrigel=1:1)。肿瘤平均体积达到104mm 3时开始分组给药。根据动物肿瘤体积通过一个基于Excel随机分组软件进行分组,每组6只小鼠。
将0.2mL 5×10 6个REC-1细胞皮下接种于每只裸小鼠的右后背(PBS:Matrigel=1:1)。肿瘤平均体积达到100mm 3时开始分组给药。根据动物肿瘤体积通过一个基于Excel随机分组软件进行分组,每组6只小鼠。
受试物的配制:
受试物配制方法参见下表11和表12:
表11 TMD8异种移植瘤模型受试物配制方法
Figure PCTCN2021125689-appb-000013
注:①样品现配现用,配置好置于4℃条件下存储,在给动物给药前需要轻轻将药物充分混匀;给药方式:灌胃;给药体积10μL/g;②使用制备例1样品检测。
表12 REC-1异种移植瘤模型受试物配制方法
Figure PCTCN2021125689-appb-000014
注:①样品现配现用,配置好置于4℃条件下存储,在给动物给药前需要轻轻将药物充分混匀;给药方式:灌胃;给药体积10μL/g;②使用制备例1样品检测。
实验动物日常观察:本实验方案的拟定及任何修改均通过了苏州药明康德新药开发股份有限公司实验动物管理与使用委员会(IACUC)的评估核准。实验动物的使用及福利遵照国际实验动物评估和认可委员会(AAALAC)的规定执行。每天监测动物的健康状况及死亡情况,例行检查包括观察肿瘤生长和药物治疗对动物日常行为表现的影响如行为活动,摄食摄水量(仅目测),体重变化(每周测量三次体重),外观体征或其它不正常情况。基于各组动物数量记录了组内动物死亡数和副作用。
肿瘤测量和实验指标:实验指标是考察肿瘤生长是否被抑制、延缓或治愈。每周三次用游标卡尺测量肿瘤直径。
肿瘤体积的计算公式为:
V=0.5a×b 2
a和b分别表示肿瘤的长径和短径。
化合物的抑瘤疗效用TGI(%)或相对肿瘤增殖率T/C(%)评价。TGI(%),反映肿瘤生长抑制率。
TGI(%)的计算:
TGI(%)=【1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)】×100%。
相对肿瘤增殖率T/C(%):计算公式如下:
T/C%=T RTV/C RTV×100%(T RTV:治疗组相对肿瘤体积;C RTV:阴性对照组相对肿瘤体积)。根据肿瘤测量的结果计算出相对肿瘤体积(RTV),计算公式为RTV=V t/V 0,其中V 0是分组给药时(即d 0)测量所得平均肿瘤体积,V t为某一次测量时的平均肿瘤体积,T RTV与C RTV取同一天数据。
统计分析:统计分析,包括每个组的每个时间点的肿瘤体积的平均值和标准误(SEM)。治疗组在给药后分别在第15天(REC-1异种移植瘤模型)和第17天(TMD8异种移植瘤模型)表现出最好的治疗效果,因此基于此数据进行统计学分析评估组间差异。三组或多组间比较用one-way ANOVA进行分析,如果F值有显著性差异,应用Games-Howell法进行检验。用SPSS17.0进行所有数据分析。p<0.05认为有显著性差异。
化合物A在人套细胞淋巴癌REC-1异种移植瘤模型中的体内药效如表13和图16所示。开始给药后15天,溶剂对照组荷瘤鼠的瘤体积达到3501mm 3,依鲁替尼25mg/kg组与溶剂对照组相比具有显著的抑瘤作用(T/C=38%,TGI=64%,p=0.008),瘤体积为1323mm 3。化合物A 15mg/kg和30mg/kg组的瘤体积分别为1034和680mm 3,与溶剂对照组相比具有显著的抑瘤作用(T/C值分别为30%和19%,TGI值分别为73%和83%,p=0.004和0.003)。
表13 化合物A对REC-1异种移植瘤模型的抑瘤药效评价(基于给药后第15天肿瘤体积计算得出)
Figure PCTCN2021125689-appb-000015
注:a.平均值±SEM;b.肿瘤生长抑制由T/C和TGI(TGI(%)=[1-(T 15-T 0)/(V 15-V 0)]×100)计算;c.p值根据肿瘤体积计算;d.给药方式:每天一次;e.使用制备例1样品检测。
化合物A在人淋巴癌TMD8异种移植瘤模型中的体内药效如表14和图17所示。开始给药后17天,溶剂对照组荷瘤鼠的瘤体积达到1852mm 3,依鲁替尼25mg/kg组与溶剂对照组相比具有显著的抑瘤作用(T/C=35.68%,TGI=68.18%,p<0.001),瘤体积为661mm 3。化合物A的5mg/kg和10mg/kg组的瘤体积分别为912和553mm 3,与溶剂对照组相比具有显著的抑瘤作用(T/C值分别为49.27%和29.85%,TGI值分别为53.78%和74.35%,p=0.003和<0.001)。
表14 化合物A对TMD8异种移植瘤模型的抑瘤药效评价(基于给药后第17天肿瘤体积计算得出)
Figure PCTCN2021125689-appb-000016
注:a.平均值±SEM;b.肿瘤生长抑制由T/C和TGI(TGI(%)=[1-(T 17-T 0)/(V 17-V 0)]×100)计算;c.p值根据肿瘤体积计算;d.给药方式:每天一次;e.使用制备例1样品检测。
结果表明,在两种BTK敏感的小鼠移植瘤模型中,化合物A具有显著的肿瘤生长抑制活性,明显优于目前已上市的第一代BTK抑制剂依鲁替尼。
另外,使用化合物S18s,重复上述在人淋巴癌TMD8异种移植瘤模型中的实验,T/C(%)结果列于下表中。在下表中,还列出了化合物A的T/C(%)结果作为对比。
表15 化合物A及S18对TMD8异种移植瘤模型的抑瘤效果
Figure PCTCN2021125689-appb-000017
注:给药方式:每天一次。
由上述数据可以看出,在更低剂量下(10mg/kg),化合物A表现出较化合物S18s(15mg/kg)更好的肿瘤生长抑制效果。
实验例7:大鼠药代动力学性质评价
SD大鼠14只,雄性,体重200-220g,随机分成4组,每组4/3只,分别灌胃和静脉给予受试化合物,具体安排见下表16
表16 受试化合物给药方法
组别 动物数 化合物 给药途径 给药剂量(mg/kg)
1 4 化合物A 灌胃(po) 3
2 3 化合物A 静脉(iv) 1
3 4 依鲁替尼 灌胃(po) 3
4 3 依鲁替尼 静脉(iv) 1
注:使用制备例1样品检测。灌胃给药以含1%吐温80的0.5%羧甲基纤维素钠(CMC-Na)配制,配置药物浓度为0.3mg/mL;静脉给药以5%DMSO/5%吐温80/90%生理盐水配制成溶液,配 置给药浓度为0.2mg/mL。
试验前禁食12小时,自由饮水。给药后2小时统一进食。
采血时间点及样品处理:
灌胃给药:给药后0.25,0.5,1.0,2.0,4.0,6.0,8.0和24小时;
静脉给药:给药后5分钟,0.25,0.5,1.0,2.0,4.0,6.0,8.0和24小时;
在以上设定时间点经大鼠眼球后静脉丛取静脉血0.3mL,置肝素化试管中,11000rpm离心5分钟,分离血浆,于-20℃冰箱中冷冻。
样品测试和数据分析
采用LC/MS/MS法测定大鼠血浆中化合物A的浓度。
采用WinNonlin 5.3软件(美国Pharsight公司)的非房室模型计算给药后的药代动力学参数。
达峰浓度C max和达峰时间T max为实测值;
药时曲线下面积AUC 0-t值:采用梯形法计算;
AUC 0-∞=AUC 0-t+C t/k e
C t为最后一个可测得时间点的血药浓度,
k e为消除速率常数;
消除半衰期t 1/2=0.693/k e
平均滞留时间MRT=AUMC/AUC。
清除率CL=D/AUC 0-∞;稳态分布容积Vss=CL×MRT
绝对生物利用度F=(AUC 灌胃×D 静脉)/(AUC 静脉×D 灌胃)×100%
试验结果见下表17:
表17 化合物A及依鲁替尼的药代动力学试验结果
Figure PCTCN2021125689-appb-000018
以上结果表明,化合物A在大鼠体内清除率显著低于依鲁替尼(20倍),血浆中药物暴露量也比依鲁替尼高达70倍,即同等剂量下化合物A的口服吸收更好,且化合物A具有更好的口服生物利用度。
实验例8:大鼠药代动力学性质评价
SD大鼠,购于上海西普尔-必凯实验动物有限公司,生产许可证号SCXK(沪)2018-000612只,雌雄各半,体重170-250g,随机分成2组,每组6只,分别灌胃和静脉给予受试化合物,具体安排见下表18。
表18 受试化合物给药方法
组别 动物数 化合物 给药途径 给药剂量(mg/kg)
1 6 化合物A(实施例2的晶型I) 灌胃(po) 15
2 6 化合物A(实施例2的晶型I) 静脉(iv) 7.5
注:使用实施例2的晶型I样品检测。灌胃给药以10%SDS水溶液:
Figure PCTCN2021125689-appb-000019
EL:0.5%MC生理盐水溶液=1:2:97(v:v:v)配制,配置药物浓度为1.5mg/mL;静脉给药以N,N-二甲基甲酰胺:生理盐水(40:60,v/v)配制成溶液,配置给药浓度为3mg/mL。
试验前禁食12小时,自由饮水。给药后4小时统一进食。
采血时间点及样品处理:
灌胃给药:给药后0.25,0.5,1.0,2.0,3.0,5.0,7.0,9.0和24小时;
静脉给药:给药后5分钟,0.25,0.5,1.0,2.0,3.0,5.0,7.0和24小时;
在以上设定时间点经大鼠眼球后静脉丛取静脉血0.2mL,置EDTA-K2化试管中,11000rpm离心5分钟,2h内分离血浆,-70℃保存待测。
样品测试和数据分析
采用LC/MS/MS法测定大鼠血浆中化合物A的浓度。计算软件和参数同实验例7中对应部 分的描述。试验结果见下表19:
表19 化合物A(实施例2的晶型I)的药代动力学试验结果
Figure PCTCN2021125689-appb-000020
以上结果表明,化合物A(实施例2的晶型I)在大鼠体内清除率较低,血浆中药物暴露量较高,且化合物A(实施例2的晶型I)具有良好的口服生物利用度。
因此,化合物A是一个结构新颖的、可口服的、高选择性的、高活性的BTK抑制剂,体内外活性明显优于目前国外已上市的BTK抑制剂,在同等剂量下,对肿瘤生长抑制活性显著优于阳性对照药依鲁替尼;进一步地,本发明提及的式(A)所示化合物的晶型具有良好的结晶度,优选晶型具备良好的稳定性,易于成药,且生物利用度高。说明化合物A及其具体晶型极具开发价值。
以上实施方式本质上仅为辅助说明,且并不欲用以限制申请目标的实施例或这些实施例的应用或用途。在本文中,用语“例示性”代表“作为一个实例、范例或说明”。本文中任一种例示性的实施形态并不必然可解读为相对于其他实施形态而言为优选或较有利者。
此外,尽管已于前述实施方式中提出至少一例示性实施例或比较例,但应了解本发明仍可存在大量的变化。同样应了解的是,本文所述的实施例并不欲用以通过任何方式限制所请求的申请目标的范围、用途或组态。相反的,前述实施方式将可提供本领域具有普通知识人员一种简便的指引以实施所述的一种或多种实施例。再者,可对要素的功能与排列进行各种变化而不脱离申请专利范围所界定的范围,且申请专利范围包含已知的均等物及在本专利申请案提出申请时的所有可预见均等物。

Claims (12)

  1. 固体形式的式(A)所示化合物、其溶剂合物或水合物,
    Figure PCTCN2021125689-appb-100001
  2. 结晶形式的式(A)所示化合物、其溶剂合物或水合物。
  3. 根据权利要求2所述的结晶形式的式(A)所示化合物、其溶剂合物或水合物,其特征在于,所述结晶形式为无溶剂且无水结晶形式或水合物结晶形式,优选为无溶剂且无水结晶形式。
  4. 根据权利要求2所述的结晶形式的式(A)所示化合物、其溶剂合物或水合物,其为式(A)所示化合物、其溶剂合物或水合物的晶型I,其特征在于,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,23.4±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,18.7±0.2°,23.4±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,16.2±0.2°,18.7±0.2°,23.4±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.8±0.2°,11.6±0.2°,13.6±0.2°,16.2±0.2°,18.7±0.2°,23.4±0.2°,26.1±0.2°;
    或者,其具有基本上如图3所示的X-射线粉末衍射图谱。
  5. 根据权利要求4所述的晶型I,其差示扫描量热曲线在在274.74±3℃有一个放热峰;
    或者,其具有基本上如图4所示的DSC图谱。
  6. 根据权利要求2所述的结晶形式的式(A)所示化合物、其溶剂合物或水合物,其为式(A)所示化合物、其溶剂合物或水合物的晶型II,其特征在于,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:8.5±0.2°,10.6±0.2°,15.0±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0±0.2°,8.5±0.2°,10.6±0.2°,15.0±0.2°,22.1±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0±0.2°,7.9±0.2°,8.5±0.2°,10.6±0.2°,15.0±0.2°,22.1±0.2°,25.3±0.2°;
    或者,其具有基本上如图5所示的X-射线粉末衍射图谱。
  7. 根据权利要求2所述的结晶形式的式(A)所示化合物、其溶剂合物或水合物,其为式(A)所示化合物、其溶剂合物或水合物的晶型III,其特征在于,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,16.2±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.1±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,16.2±0.2°,25.5±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.3±0.2°,7.1±0.2°,8.0±0.2°,8.6±0.2°,10.6±0.2°,15.1±0.2°,16.2±0.2°,22.3±0.2°,25.5±0.2°;
    或者,其具有基本上如图7所示的X-射线粉末衍射图谱。
  8. 根据权利要求2所述的结晶形式的式(A)所示化合物、其溶剂合物或水合物,其为式(A)所示化合物、其溶剂合物或水合物的晶型IV,其特征在于,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:6.3±0.2°,9.7±0.2°,12.8±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.1±0.2°,6.3±0.2°,9.7±0.2°,12.8±0.2°,14.1±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.1±0.2°,6.3±0.2°,9.7±0.2°,12.8±0.2°,14.1±0.2°,19.0±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.1±0.2°,6.3±0.2°,9.7±0.2°,12.8±0.2°,14.1±0.2°,15.6±0.2°,19.0±0.2°,21.9±0.2°;
    或者,其具有基本上如图9所示的X-射线粉末衍射图谱。
  9. 根据权利要求2所述的结晶形式的式(A)所示化合物、其溶剂合物或水合物,其为式(A)所示化合物、其溶剂合物或水合物的晶型V,其特征在于,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.6±0.2°,7.1±0.2°,14.3±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.6±0.2°,7.1±0.2°,11.5±0.2°,13.8±0.2°,14.3±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.6±0.2°,7.1±0.2°,11.5±0.2°,13.8±0.2°,14.3±0.2°,17.0±0.2°,21.9±0.2°;
    或者,其具有基本上如图11所示的X-射线粉末衍射图谱。
  10. 根据权利要求2所述的结晶形式的式(A)所示化合物、其溶剂合物或水合物,其为式(A)所示化合物、其溶剂合物或水合物的晶型VI,其特征在于,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:6.2±0.2°,9.4±0.2°,12.5±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:6.2±0.2°,9.4±0.2°,12.5±0.2°,15.2±0.2°,21.4±0.2°;
    或者,其X-射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.0±0.2°,6.2±0.2°,9.4±0.2°,12.5±0.2°,15.2±0.2°,21.4±0.2°,24.7±0.2°;
    或者,其具有基本上如图12所示的X-射线粉末衍射图谱。
  11. 一种药物组合物,包含权利要求1所述的固体形式的式(A)化合物、其溶剂合物或水合物、或权利要求2或3所述的结晶形式的式(A)化合物、其溶剂合物、水合物或其结晶组合物;
    优选地,所述药物组合物包含权利要求4或5所述式(A)所示化合物的晶型I、权利要求6所述式(A)所示化合物的晶型II、权利要求7所述式(A)所示化合物的晶型III、权利要求8所述式(A)所示化合物的晶型IV、权利要求9所述式(A)所示化合物的晶型V、权利要求10所述式(A)所示化合物的晶型VI中的一种或多种。
  12. 根据权利要求1所述的固体形式的式(A)所示化合物、其溶剂合物或水合物、权利要求2或3所述的结晶形式的式(A)所示化合物、其溶剂合物或水合物、权利要求4~10任意一项所述式(A)所示化合物、其溶剂合物或水合物的晶型、或权利要求11所述的药物组合物在制备治疗BTK相关病症的药物中的应用;
    优选地,所述BTK相关病症包括肿瘤疾病或自身免疫性疾病;优选地,所述肿瘤疾病为血液瘤;进一步优选为白血病或淋巴瘤;进一步优选为B细胞淋巴瘤;更进一步优选为套细胞淋巴瘤、慢性淋巴细胞白血病、小淋巴细胞淋巴瘤、边缘区淋巴瘤、滤泡性淋巴瘤、华氏巨球蛋白血症或弥漫性大B细胞淋巴瘤。
PCT/CN2021/125689 2020-10-23 2021-10-22 固体形式的布鲁顿酪氨酸激酶抑制剂化合物及其用途 WO2022083733A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011145818.4 2020-10-23
CN202011145818 2020-10-23

Publications (1)

Publication Number Publication Date
WO2022083733A1 true WO2022083733A1 (zh) 2022-04-28

Family

ID=81291647

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/125689 WO2022083733A1 (zh) 2020-10-23 2021-10-22 固体形式的布鲁顿酪氨酸激酶抑制剂化合物及其用途

Country Status (2)

Country Link
CN (1) CN114478547A (zh)
WO (1) WO2022083733A1 (zh)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101175755A (zh) * 2005-03-17 2008-05-07 诺瓦提斯公司 作为酪氨酸/苏氨酸激酶抑制剂特别是b-raf激酶的n-[3-(1-氨基-5,6,7,8-四氢-2,4,4b-三氮杂芴-9-基)-苯基]苯甲酰胺
CN108101905A (zh) * 2016-11-24 2018-06-01 中国科学院上海药物研究所 嘧啶并[5,4-b]吲嗪或嘧啶并[5,4-b]吡呤化合物、其制备方法及用途
CN111848634A (zh) * 2019-04-24 2020-10-30 中国科学院上海药物研究所 嘧啶并[5,4-b]吡呤化合物、其光学异构体、制备方法及用途
WO2021037188A1 (zh) * 2019-08-29 2021-03-04 中国科学院上海药物研究所 嘧啶并[5,4-b]吡呤化合物的制药用途

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101175755A (zh) * 2005-03-17 2008-05-07 诺瓦提斯公司 作为酪氨酸/苏氨酸激酶抑制剂特别是b-raf激酶的n-[3-(1-氨基-5,6,7,8-四氢-2,4,4b-三氮杂芴-9-基)-苯基]苯甲酰胺
CN108101905A (zh) * 2016-11-24 2018-06-01 中国科学院上海药物研究所 嘧啶并[5,4-b]吲嗪或嘧啶并[5,4-b]吡呤化合物、其制备方法及用途
CN111848634A (zh) * 2019-04-24 2020-10-30 中国科学院上海药物研究所 嘧啶并[5,4-b]吡呤化合物、其光学异构体、制备方法及用途
WO2021037188A1 (zh) * 2019-08-29 2021-03-04 中国科学院上海药物研究所 嘧啶并[5,4-b]吡呤化合物的制药用途

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LIU, YUTING ET AL.: ""A novel tricyclic BTK inhibitor suppresses B cell responses and osteoclastic bone erosion in rheumatoid arthritis"", 《ACTA PHARMACOLOGICA SINICA》, vol. 42, no. 10, 13 January 2021 (2021-01-13), XP037570969, DOI: 10.1038/s41401-020-00578-0 *
XUE, YU ET AL.: ""Discovery of 4, 7-Diamino-5-(4-phenoxyphenyl)-6-methylene-pyrimido[5, 4-b]pyrrolizines as Novel Bruton's Tyrosine Kinase Inhibitors"", JOURNAL OF MEDICINAL CHEMISTRY, vol. 61, no. 10, 1 May 2018 (2018-05-01), XP055745992, DOI: 10.1021/acs.jmedchem.8b00441 *

Also Published As

Publication number Publication date
CN114478547A (zh) 2022-05-13

Similar Documents

Publication Publication Date Title
ES2837431T3 (es) Moduladores del regulador de la conductancia transmembrana de fibrosis quística, composiciones farmacéuticas, métodos de tratamiento, y procesos para elaborar el modulador
EP3740206B1 (en) Inhibitors of cyclin-dependent kinase 7 (cdk7)
JP6948659B1 (ja) ピリダジニルチアアゾールカルボキシアミド化合物
KR20060054300A (ko) 선택적인 사이클린 의존성 키나제 4 억제제로서의이세싸이오네이트 염
RU2746045C2 (ru) Кристалл пирролопиримидина для получения jak-ингибитора
CN110382499A (zh) Fgfr抑制剂及其应用
WO2021228248A1 (zh) 氮杂稠环酰胺类化合物及其用途
WO2017088746A1 (zh) 新的表皮生长因子受体抑制剂及其应用
WO2015188681A1 (zh) 一种新型杂环化合物及其制备方法和作为激酶抑制剂的用途
JP7397452B2 (ja) ヘテロアリールカルボキシアミド化合物
TW202311259A (zh) 化合物i的新形式及其應用
WO2020216343A1 (zh) 嘧啶并[5,4-b]吡呤化合物、其光学异构体、制备方法及用途
WO2021249319A1 (zh) 三环化合物、药物组合物及其应用
WO2021129841A1 (zh) 用作ret激酶抑制剂的化合物及其应用
WO2020098716A1 (zh) 布鲁顿酪氨酸激酶的抑制剂
WO2023036252A1 (zh) 吡咯并嘧啶类或吡咯并吡啶类衍生物及其医药用途
WO2020221209A1 (zh) 一种cd73抑制剂,其制备方法和应用
WO2023083356A1 (zh) 固体形式的氮杂稠环酰胺类化合物及其用途
WO2022083733A1 (zh) 固体形式的布鲁顿酪氨酸激酶抑制剂化合物及其用途
WO2022171118A1 (zh) 一种具有抗肿瘤活性的化合物及其用途
EP3632912B1 (en) Pyridoquinazoline derivatives useful as protein kinase inhibitors
WO2022083745A1 (zh) 一种布鲁顿酪氨酸激酶抑制剂的用途
WO2023246858A1 (zh) 一种硼酸酯衍生物的可药用盐、其结晶形式及用途
WO2023246854A1 (zh) 一种硼酸酯衍生物的结晶、其制备方法及用途
WO2016188481A1 (zh) 苯并吡唑联吡啶类化合物、包含此类化合物的药物组合物及其用途

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: 21882145

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21882145

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 21882145

Country of ref document: EP

Kind code of ref document: A1

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 16/10/2023)