CN116120323A - Solid form aza-condensed ring amide compound and use thereof - Google Patents

Solid form aza-condensed ring amide compound and use thereof Download PDF

Info

Publication number
CN116120323A
CN116120323A CN202211427370.4A CN202211427370A CN116120323A CN 116120323 A CN116120323 A CN 116120323A CN 202211427370 A CN202211427370 A CN 202211427370A CN 116120323 A CN116120323 A CN 116120323A
Authority
CN
China
Prior art keywords
ray powder
radiation
powder diffraction
diffraction pattern
alternatively
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202211427370.4A
Other languages
Chinese (zh)
Inventor
王洁
王叶挺
龚登凰
米国瑞
马二倩
马玉秀
范丽雪
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
CSPC Zhongqi Pharmaceutical Technology Shijiazhuang Co Ltd
Original Assignee
CSPC Zhongqi Pharmaceutical Technology Shijiazhuang Co Ltd
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 CSPC Zhongqi Pharmaceutical Technology Shijiazhuang Co Ltd filed Critical CSPC Zhongqi Pharmaceutical Technology Shijiazhuang Co Ltd
Publication of CN116120323A publication Critical patent/CN116120323A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • 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
    • C07D487/02Heterocyclic 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 two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • 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

Landscapes

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

Abstract

The invention provides a compound shown in a solid form, a compound shown in a crystal form shown in the formula (A), a specific crystal form, a pharmaceutical composition containing the compound and application thereof, wherein the compound shown in the crystal form shown in the formula (A) and the specific crystal form have good crystallinity and stability, and the in-vitro and in-vivo efficacy shows that the compound shown in the formula (A) has good crystallinity and stability on wild type and mutant kinase and fine particle sizeThe cell and the in-vivo tumor have good inhibition effect and good potential of patent medicine.
Figure DDA0003942748500000011

Description

Solid form aza-condensed ring amide compound and use thereof
The present application claims priority from a prior application filed by the applicant in 2021 at 11/15 to the national intellectual property agency of China, having patent application number 202111345026.6, entitled "solid form aza-fused ring amide-type compounds and uses thereof". The entirety of this prior application is incorporated by reference into this application.
Technical Field
The application relates to the field of medicines, in particular to an aza-condensed ring amide compound in a solid form, an aza-condensed ring amide compound in a crystal form and a specific crystal form thereof, a pharmaceutical composition containing the aza-condensed ring amide compound and application of the aza-condensed ring amide compound in preparation of medicines for preventing and/or treating one or more of TRK, ROS1 and ALK mediated diseases.
Background
Tropomyosin-related kinase or Tropomyosin Receptor Kinase (TRK) is a class of nerve growth factor receptors whose family consists of three subtypes, TRKA, TRKB and TRKC, of high homology, encoded by the neurotrophic receptor tyrosine kinase 1 (NTRK 1), NTRK2 and NTRK3 genes, respectively. When the TRK receptor protein binds to the corresponding ligand, different physiological functions can be achieved by activating downstream signaling pathways, such as the RAS/MAPK pathway, plcγ pathway, and PI3K pathway. TRK family proteins are normally expressed primarily in neural tissues, involved in neural cell differentiation and survival, and in axon and dendrite formation, and play an important role in embryonic development and maintenance of normal function of the nervous system.
TRK kinase is activated in malignant tumors by a variety of mechanisms, mainly structural rearrangements and changes in expression. For example, the rearrangement of the gene NTRK encoding the TRK kinase with other genes results in fusion oncogenes, which result in structural and expression changes in the TRK kinase that are no longer regulated and controlled by nerve growth factor ligands, constitutive activation occurs, and neoplastic development is promoted. In addition, the results of gene sequencing also show that TRK kinase has a close relationship with the occurrence, metastasis and exacerbation of various tumors, and is expressed in various tumors, such as non-small cell lung cancer, colorectal cancer, melanoma, gall bladder cancer, thyroid cancer, glioblastoma and the like.
Currently, the first generation TRK inhibitors Larotrectinib (LOXO-101) and Entrectrinib (RXDX-101) were approved for sale by the U.S. Food and Drug Administration (FDA) in 2018 and 2019, respectively. Larotrectinib is a potent, oral, selective tropomyosin receptor kinase inhibitor whose efficacy data was published as early as the ASCO conference at 2017, month 6, and in phase I and phase II clinical trials 55 subjects were enrolled, of which 46 had an estimated Overall Response Rate (ORR) of 78%. Entrectrinib is a potent inhibitor of TRK, ROS1 and ALK proteins, and can pass the blood brain barrier, with 79% of the ORRs in 24 evaluable patients in phase I clinical trials.
Like other targeted drugs, TRK inhibitors also face drug resistance issues. Mutations in the NTRK kinase domain cause conformational changes in the TRK family of protein kinase domains or changes in binding affinity to ATP, thereby affecting binding of the TRK inhibitor to the target, types of mutations are G595R, G639R, G667C, etc. To solve the problem of drug resistance of the first generation TRK inhibitors, the second generation TRK inhibitors such as LOXO-195, TPX-005, etc. have been studied.
Figure BDA0003942748480000021
Disclosure of Invention
In one aspect, the present application provides a compound of formula (a) in solid form.
Figure BDA0003942748480000022
In some embodiments of the present application, the solid form of the compound of formula (A) described above may be subjected to infrared spectrometry using a tabletting method, and the infrared spectrum includes characteristic peaks (. + -. 4 cm) at the following positions -1 ):3429,1643,1488,1453,1232,1027。
In some aspects of the present application, the compound of formula (a) in solid form described above has an infrared spectrum substantially as shown in figure 12.
In some embodiments of the present application, the compound of formula (A) in solid form described above may be tested using X-ray powder diffraction.
In some embodiments of the present application, the compound of formula (A) in solid form has a chemical purity of 95% or more; preferably, it has a chemical purity of ≡97%; further preferably, it has a chemical purity of ≡98%.
In some aspects of the present application, the compound of formula (a) in solid form described above is amorphous and uses Cu-ka radiation having an infrared spectrum substantially as shown in figure 12.
In another aspect, the present application provides a compound of formula (a) in crystalline form.
In another aspect, the present application provides crystalline form I of a compound of formula (a), having an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 9.2, 17.9, 18.5.
In some aspects of the present application, form I above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 8.9,9.2, 17.9, 18.5, 23.8.
In some aspects of the present application, form I above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 8.9,9.2, 10.1, 17.9, 18.5, 23.8, 28.0.
In some aspects of the present application, form I above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 8.9,9.2, 10.1, 16.1, 17.9, 18.5, 23.8, 27.0, 28.0.
In some aspects of the present application, form I above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 8.9,9.2, 10.1, 16.1, 16.6, 17.9, 18.5, 23.8, 27.0, 28.0.
In some aspects of the present application, form I above, using Cu-ka radiation, has an X-ray powder diffraction pattern substantially as shown in figure 1.
In another aspect, the present application provides crystalline form II of a compound of formula (a), having an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 9.1, 18.3, 20.6.
In some aspects of the present application, form II above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.4,9.1, 18.3, 20.6.
In some aspects of the present application, form II above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.4,9.1, 18.3, 20.6, 27.7.
In some aspects of the present application, form II above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.4,9.1, 18.3, 18.9, 20.6, 27.7.
In some aspects of the present application, the above-described form II, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.4,9.1, 18.3, 18.9, 20.6, 22.4, 23.9, 27.7.
In some aspects of the present application, form II above, using Cu-ka radiation, has an X-ray powder diffraction pattern substantially as shown in figure 2.
In some aspects of the present application, the differential scanning calorimetry curve of the above-mentioned form II has an endothermic peak at 155.27 ±5 ℃.
In some aspects of the present application, the aforementioned form II has a differential scanning calorimetry curve with endothermic peaks at 58.39 ±5 ℃ and 155.27 ±5 ℃.
In some aspects of the present application, the above-described form II has a DSC profile substantially as shown in figure 3.
In some aspects of the present application, the thermogravimetric analysis of form II has a weight loss of 0.1910% ± 0.2% between room temperature and 75±5 ℃.
In some aspects of the present application, form II above has a TGA profile substantially as shown in figure 3.
In another aspect, the present application provides crystalline form III of formula (a), having an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°) at the following 2θ angles using Cu-ka radiation: 5.6, 13.3, 15.8, 18.4.
In some aspects of the present application, the above-described crystalline form III, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 5.6, 13.3, 15.8, 18.4, 23.7.
In some aspects of the present application, the above-described crystalline form III, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 5.6, 10.9, 13.3, 15.8, 18.4, 19.0, 23.7.
In some aspects of the present application, the above-described crystalline form III, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 5.6, 10.9, 11.3, 13.3, 15.8, 17.1, 18.4, 19.0, 23.7.
In some aspects of the present application, the above-described crystalline form III, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 5.6, 10.9, 11.3, 13.3, 15.8, 17.1, 18.4, 19.0, 19.9, 23.7.
In some aspects of the present application, the above-described crystalline form III, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 5.6,7.4, 10.9, 11.3, 13.3, 15.8, 17.1, 18.4, 19.0, 19.9, 23.7.
In some aspects of the present application, form III above, using Cu-ka radiation, has an X-ray powder diffraction pattern substantially as shown in fig. 4.
In some aspects of the present application, the aforementioned form III has a differential scanning calorimetry curve with an endothermic peak at 182.29 ±5 ℃.
In some aspects of the present application, form III above has a DSC profile substantially as shown in figure 5.
In another aspect, the present application provides crystalline form IV of a compound of formula (a), having an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 8.5, 10.0, 17.2, 17.9.
In some aspects of the present application, form IV above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 8.5, 10.0, 13.1, 17.2, 17.9, 25.0.
In some aspects of the present application, form IV above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 8.5, 10.0, 13.1, 17.2, 17.9, 20.0, 25.0.
In some aspects of the present application, form IV above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 8.5, 10.0, 13.1, 15.7, 17.2, 17.9, 20.0, 25.0, 25.9.
In some aspects of the present application, form IV above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 8.5, 10.0, 13.1, 15.7, 17.2, 17.9, 19.5, 20.0, 25.0, 25.9.
In some aspects of the present application, form IV above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 5.5,8.5, 10.0, 13.1, 14.0, 15.7, 17.2, 17.9, 19.5, 20.0, 25.0, 25.9.
In some aspects of the present application, form IV above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 5.5,8.5, 10.0, 13.1, 14.0, 15.7, 17.2, 17.9, 19.0, 19.5, 20.0, 25.0, 25.9, 26.4.
In some aspects of the present application, form IV above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 5.5,8.5, 10.0, 13.1, 14.0, 15.7, 16.5, 17.2, 17.9, 19.0, 19.5, 20.0, 22.9, 25.0, 25.9, 26.4.
In some aspects of the present application, form IV above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 5.5,8.5, 10.0, 13.1, 14.0, 15.7, 16.5, 17.2, 17.9, 19.0, 19.5, 20.0, 22.9, 23.4, 23.7, 25.0, 25.9, 26.4.
In some aspects of the present application, form IV above, using Cu-ka radiation, has an X-ray powder diffraction pattern substantially as shown in fig. 6 or 8.
In some aspects of the present application, the differential scanning calorimetry curve of form IV has an endothermic peak at 192.07 ±5 ℃.
In some aspects of the present application, the aforementioned form IV has a differential scanning calorimetry curve with endothermic peaks at 64.82±5 ℃ and 192.07 ±5 ℃.
In some aspects of the present application, form IV above has a DSC profile substantially as shown in figure 7.
In some aspects of the present application, the thermogravimetric analysis of form IV above has a weight loss of 1.1526% ± 0.2% between room temperature and 75±5 ℃.
In some aspects of the present application, form IV above has a TGA profile substantially as shown in figure 7.
In another aspect, the present application provides crystalline form V of a compound of formula (a), having an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.0, 12.7, 18.2.
In some aspects of the present application, form V above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.0, 12.7, 18.2, 23.4.
In some aspects of the present application, form V above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.0, 12.7, 16.7, 18.2, 23.4.
In some aspects of the present application, form V above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.0, 12.7, 16.7, 18.2, 23.4, 25.4, 28.1.
In some aspects of the present application, form V above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.0,7.0, 12.7, 14.2, 16.7, 18.2, 23.4, 25.4, 28.1.
In some aspects of the present application, form V above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.0,7.0, 12.7, 14.2, 16.7, 18.2, 23.4, 24.5, 25.4, 28.1.
In some aspects of the present application, form V above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.0,7.0, 12.1, 12.7, 14.2, 16.7, 17.6, 18.2, 23.4, 24.5, 25.4, 28.1.
In some aspects of the present application, form V above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.0,7.0, 12.1, 12.7, 14.2, 16.7, 17.6, 18.2, 20.2, 23.4, 24.5, 25.4, 28.1, 28.8.
In some aspects of the present application, form V above uses Cu-ka radiation having an X-ray powder diffraction pattern substantially as shown in fig. 9.
In another aspect, the present application provides crystalline form VI of a compound of formula (a), having an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.1, 16.9, 22.3.
In some aspects of the present application, form VI above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.1, 16.9, 18.6, 22.3.
In some aspects of the present application, form VI above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.1,7.4, 11.5, 16.9, 18.6, 22.3.
In some aspects of the present application, form VI above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.1,7.4, 11.5, 16.9, 18.6, 19.4, 22.3, 25.3.
In some aspects of the present application, form VI above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.1,7.4, 11.5, 16.9, 18.6, 19.4, 20.1, 22.3, 25.3, 28.6.
In some aspects of the present application, form VI above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.1,7.4, 11.5, 16.9, 18.6, 19.4, 20.1, 22.3, 22.8, 23.2, 25.3, 28.6.
In some aspects of the present application, form VI above, using Cu-ka radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.1,7.4, 11.5, 16.9, 18.6, 19.4, 20.1, 22.3, 22.8, 23.2, 25.3, 26.7, 28.6.
In some aspects of the present application, form VI above, using Cu-ka radiation, has an X-ray powder diffraction pattern substantially as shown in fig. 10.
In some aspects of the present application, the differential scanning calorimetry curve of form VI has an endothermic peak at 184.47 ±5 ℃.
In some aspects of the present application, form VI above has a DSC profile substantially as shown in figure 11.
In some aspects of the present application, the thermogravimetric analysis of form VI has a weight loss of 0.253% ± 0.2% between room temperature and 200 ℃ ± 5 ℃.
In some aspects of the present application, form VI above has a TGA profile substantially as shown in figure 11.
In another aspect, the present application provides a crystalline composition comprising one or more of form I, form II, form III, form IV, form V, form VI of a compound of formula (a).
In some aspects of the present application, the crystalline form II comprises greater than 50%, greater than 60%, greater than 70%, greater than 80%, greater than 90%, or greater than 95% by weight of the crystalline composition.
In some aspects of the present application, the crystalline form III comprises greater than 50%, greater than 60%, greater than 70%, greater than 80%, greater than 90%, or greater than 95% by weight of the crystalline composition.
In some aspects of the present application, the form IV comprises greater than 50%, greater than 60%, greater than 70%, greater than 80%, greater than 90%, or greater than 95% by weight of the crystalline composition.
In some aspects of the present application, form VI comprises greater than 50%, greater than 60%, greater than 70%, greater than 80%, greater than 90%, or greater than 95% by weight of the crystalline composition.
In another aspect, the present application provides a pharmaceutical composition comprising a compound of formula (a) in solid form, a compound of formula (a) in crystalline form, or a crystalline composition as described above.
In some embodiments of the present application, the pharmaceutical composition comprises one or more of form I, form II, form III, form IV, form V, and form VI of the compound of formula (a).
In another aspect, the present application also provides a pharmaceutical composition comprising a compound of formula (a) in solid form, a compound of formula (a) in crystalline form, or a crystalline composition as described above, and a pharmaceutically acceptable carrier.
In some aspects of the present application, the pharmaceutical composition comprises one or more of form I, form II, form III, form IV, form V, form VI of the compound of formula (a), and a pharmaceutically acceptable carrier.
In another aspect, the present application provides the use of a compound of formula (a) in solid form, a compound of formula (a) in crystalline form, a crystalline composition as described above, a crystalline form I, a crystalline form II, a crystalline form III, a crystalline form IV, a crystalline form V or a crystalline form VI of a compound of formula (a) as described above, or a pharmaceutical composition as described above, as a medicament or in the preparation of a medicament.
In some aspects of the present application, the medicament is for treating a pain disorder, a cell proliferative disorder, an inflammatory disorder, a neurodegenerative disorder, or an infectious disorder.
In some aspects of the present application, the medicament is for preventing and/or treating a disease mediated by one or more of TRK, ROS1, or ALK.
In some aspects of the present application, the medicament is for preventing and/or treating NTRK gene rearrangement/fusion and/or drug resistant mutation positive tumors, or ROS1 gene rearrangement/fusion and/or drug resistant mutation positive tumors; preferably, the tumor is a solid tumor or a hematological tumor; further preferably, the tumor is a solid tumor.
In some aspects of the present application, the NTRK resistance mutation is NTRK1-G595R, NTRK1-G667C, NTRK-G623R or NTRK3-G696A; preferably, the NTRK resistance mutation is NTRK1-G595R, NTRK3-G623R or NTRK1-G667C; further preferably, the NTRK resistant mutation is NTRK1-G595R or NTRK1-G667C.
In another aspect, the present application also provides a compound of formula (a) in solid form, a compound of formula (a) in crystalline form, a crystalline composition as described above, a crystalline form I, a crystalline form II, a crystalline form III, a crystalline form IV, a crystalline form V or a crystalline form VI of a compound of formula (a) as described above, or a pharmaceutical composition as described above, for use in the prevention and/or treatment of a disease mediated by one or more of TRK, ROS1 or ALK.
In another aspect, the present application also provides a method of preventing and/or treating a disease mediated by one or more of TRK, ROS1, or ALK, comprising: administering to the patient a therapeutically effective amount of a compound of formula (a) in solid form as described above, a compound of formula (a) in crystalline form, a crystalline composition as described above, form I, form II, form III, form IV, form V or form VI of a compound of formula (a) or a pharmaceutical composition as described above.
In some aspects of the present application, the disease is a NTRK gene rearrangement/fusion and/or drug resistance mutation positive tumor, or a ROS1 gene rearrangement/fusion and/or drug resistance mutation positive tumor; preferably, the tumor is a solid tumor or a hematological tumor; further preferably, the tumor is a solid tumor.
In some aspects of the present application, the NTRK resistance mutation is NTRK1-G595R, NTRK1-G667C, NTRK-G623R or NTRK3-G696A; preferably, the NTRK resistance mutation is NTRK1-G595R, NTRK3-G623R or NTRK1-G667C; further preferably, the NTRK resistant mutation is NTRK1-G595R or NTRK1-G667C.
In some aspects of the present application, the disease is selected from pain, cell proliferative, inflammatory, neurodegenerative, or infectious diseases.
In one embodiment, the TRK mediated disease described above is selected from a disease mediated through one, two, or three of TRKA, TRKB, or TRKC.
In one embodiment, the above-described diseases involve deregulation of the NTRK gene, the TRK protein, or their expression, activity or level; preferably, it involves NTRK gene fusion, amplification, rearrangement, mutation or high expression; further preferred are NTRK gene rearrangements/fusions or mutations.
In some aspects of the present application, the NTRK is mutated to NTRK1-G595R, NTRK1-G667C, NTRK3-G623R or NTRK3-G696A; preferably, the NTRK resistance mutation is NTRK1-G595R, NTRK3-G623R or NTRK1-G667C; further preferably, the NTRK resistant mutation is NTRK1-G595R or NTRK1-G667C.
In one embodiment, the above-described diseases involve deregulation of the ROS1 gene, ROS1 protein, or their expression, activity, or level; preferably, ROS1 gene fusion, amplification, rearrangement, mutation or high expression is involved; further preferred are ROS1 gene rearrangements/fusions or mutations.
In one embodiment, the disease described above involves a deregulation of the expression, activity or level of one or more genes, proteins, or thereof in TRK, ALK, ROS 1; preferably involving fusion, amplification, rearrangement, mutation or high expression of one or more of NTRK, ALK, ROS genes; further preferred are gene fusions or mutations involving one or more of NTRK, ALK, ROS 1.
In one embodiment, the cell proliferative disorder described above is a tumor or cancer.
In one embodiment, the tumor or cancer is a solid tumor or a hematological tumor; preferably a solid tumor; further preferred are solid tumors positive for NTRK gene rearrangement/fusion and/or resistance mutation, or solid tumors positive for ROS1 gene rearrangement/fusion and/or resistance mutation.
In some aspects of the present application, the NTRK resistance mutation is NTRK1-G595R, NTRK1-G667C, NTRK-G623R or NTRK3-G696A; preferably, the NTRK resistance mutation is NTRK1-G595R, NTRK3-G623R or NTRK1-G667C; further preferably, the NTRK resistant mutation is NTRK1-G595R or NTRK1-G667C.
In one embodiment, the tumor or cancer is a hematological malignancy, lung cancer, breast cancer, ovarian cancer, prostate cancer, pancreatic cancer, glioma, colorectal cancer, melanoma, cancer of the head and neck, gall bladder cancer, thyroid cancer, glioblastoma, gastric cancer, neuroblastoma, or salivary gland cancer; preferably, the lung cancer is non-small cell lung cancer.
Definition and description
The following terms and phrases used herein are intended to have the following meanings unless otherwise indicated. A particular phrase or terminology, unless otherwise specifically defined, should not be construed as being ambiguous or otherwise clear, but rather should be construed in a generic sense. The "compound represented by formula (a) in solid form" referred to herein means a compound represented by formula (a) in solid form.
The "compound represented by formula (a) in crystalline form" referred to herein means a compound represented by formula (a) in crystalline form, and includes anhydrous and solvent-free forms, hydrate forms, and solvate forms of the compound represented by formula (a).
The term "solvate" or "solvate" refers to an association of a stoichiometric or non-stoichiometric ratio of a solvent molecule with a compound of formula (a) herein, and includes an association containing both a water molecule and one or more other solvent molecules, as well as an association containing only one or more other solvent molecules.
The term "hydrate" refers to an association of water molecules in stoichiometric or non-stoichiometric proportions with a compound of formula (a) of the present application.
The "anhydrous and solvent-free form" means that no water molecules or solvent molecules are contained, or that water molecules or solvent molecules coexist with the compound represented by formula (a) in a non-intermolecular force-bonded manner, for example, in an adsorption manner.
In the context of the present application, the characteristic diffraction peaks, diffraction peaks and/or the 2 theta angle values in the X-ray powder diffraction pattern are all in degrees (°).
The term "crystalline composition" refers to a solid form comprising one, two or more of form I, form II, form III, form IV, form V, or form VI mentioned herein. Furthermore, the crystalline composition may optionally contain, in addition to the crystalline forms of the present application, other crystalline forms or other amorphous forms of the compound represented by formula (a) or a salt thereof, or impurities other than these substances. It will be appreciated by those skilled in the art that the sum of the contents of the individual components in the crystalline composition should be 100%.
The "room temperature" is room temperature in the conventional sense in the art, typically 10 to 30℃and preferably 25.+ -. 5 ℃.
In an X-ray powder diffraction pattern, the term "substantially" or "substantially as shown" refers to a substantially pure crystalline form in which 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%, or at least 97%, or at least 98%, or at least 99% of the peaks in the powder X-ray diffraction pattern appear in the given pattern. Further, as the content of a certain crystal form in a product gradually decreases, some diffraction peaks ascribed to the crystal form in the X-ray powder diffraction pattern thereof may be reduced due to factors of the detection sensitivity of the instrument. Furthermore, there may be slight errors in the position of the peaks for any given crystal form, which is also well known in the crystallography arts. For example, the position of the peak may be shifted due to a change in temperature at the time of analyzing the sample, a shift in the sample, calibration of the instrument, or the like, and a measurement error of the 2θ value is sometimes about ±0.3°, typically about ±0.2°. Thus, this error should be taken into account when determining each crystal structure, and the term "substantially" or "substantially as shown in the drawings" is also intended to cover such differences in diffraction peak positions, meaning ± 0.3 °, preferably ± 0.2 °.
In a DSC profile or TGA profile, the term "substantially" or "substantially as shown" means that for an isomorphous form of an isomorphous compound, the errors in thermal transition onset temperature, endothermic peak temperature, exothermic peak temperature, melting point, weight loss onset temperature, or weight loss end point temperature, etc., are typically within about 5 ℃, usually within about 3 ℃, in a continuous analysis. When describing a compound as having a given thermal transition onset temperature, endothermic peak temperature, exothermic peak temperature, melting point, weight loss onset temperature, or weight loss end temperature, etc., this temperature is referred to as + -5 deg.c.
The term "cell proliferative disorder" as used herein refers to a condition in which the growth rate of a population of cells is lower or higher than the expected rate for a given physiological state and condition.
The term "tumor" encompasses benign tumors, malignant tumors, and borderline tumors, wherein malignant tumors are also collectively referred to as cancers.
The term "preventing" as used herein refers to a compound or drug that, when used in a disease or disorder (e.g., cancer), reduces the frequency of symptoms of or delays the onset of a medical disorder in a subject as compared to a subject to whom the compound or drug (e.g., a combination product as claimed herein) was not administered.
The term "treating" as used herein refers to alleviating, alleviating or ameliorating a symptom of a disease or disorder, ameliorating a symptom of underlying metabolism, inhibiting a disease or symptom, e.g., preventing the development of a disease or disorder, alleviating a disease or disorder, causing regression of a disease or disorder, alleviating a condition caused by a disease or disorder, or preventing a symptom of a disease or disorder.
The term "pharmaceutically acceptable carrier" or "pharmaceutically acceptable adjuvant" refers to those carriers or adjuvants which do not have a significant irritating effect on the organism and which do not impair the biological activity and properties of the active compound.
Intermediate compounds of the present application may be prepared by a variety of synthetic methods known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combining them with other chemical synthetic methods, and equivalent alternatives known to those skilled in the art, preferred embodiments including but not limited to the examples of the present application.
The chemical reactions of the embodiments of the present application are accomplished in a suitable solvent that is suitable for the chemical changes of the present application and the reagents and materials needed. In order to obtain the compounds of the present application, modifications or choices of synthesis steps or reaction schemes based on the existing embodiments are sometimes required by those skilled in the art.
The present application will be specifically described by examples, which are not meant to be limiting in any way.
All solvents used in this application are commercially available and can be used without further purification.
Technical effects
The compound shown in the formula (A) in a solid form, the compound shown in the formula (A) in a crystal form and a specific crystal form have one or more of the following beneficial effects:
(1) The compound shown in the formula (A) in a solid form has good properties and is convenient to weigh, transfer, separate, purify and store.
(2) The compound shown in the formula (A) in a crystal form and a specific crystal form have good crystallinity;
(3) The compound shown in the formula (A) in a crystal form and a specific crystal form are easy to purify, filter and separate, and particularly the crystal form IV is simple and convenient to prepare and high in yield;
(4) The preferred crystal form has good physical stability and chemical stability and good medicinal prospect;
(5) In vitro kinase activity inhibition assays showed: the compounds of formula (a) exhibit excellent inhibitory activity against various kinases (e.g., TRK, ALK, ROS 1) and mutants thereof, particularly against TRK and mutant forms thereof;
(5) In vitro cytostatic activity assays showed: the compound shown in the formula (A) has strong inhibition effect on a plurality of NTRK mutant cells and IC on inhibition activity of 6 cells 50 Below 10nM, preferably below 5nM, more preferably below 1 nM;
(6) The in vivo tumor inhibition test results show that: compared with a control compound, the compound shown in the formula (A) has better in-vivo anti-tumor effect, better tolerance and higher possibility of drug forming;
(7) The in vivo action mechanism research experiment shows that: the compound shown in the formula (A) can inhibit TRK phosphorylation in tumor tissues, and further effectively inhibit the phosphorylation of PLC gamma 1 and AKT, and inhibit the growth of the tumor tissues.
Drawings
Fig. 1: x-ray powder diffraction pattern of form I of example 1.
Fig. 2: x-ray powder diffraction pattern of form II of example 2.
Fig. 3: DSC-TGA spectrum of form II of example 2.
Fig. 4: x-ray powder diffraction pattern of form III of example 3.
Fig. 5: DSC-TGA spectrum of form III of example 3.
Fig. 6: x-ray powder diffraction pattern of form IV of example 4.
Fig. 7: DSC-TGA spectrum of form IV of example 4.
Fig. 8: x-ray powder diffraction pattern of form IV of example 5.
Fig. 9: x-ray powder diffraction pattern of form V of example 7.
Fig. 10: x-ray powder diffraction pattern of form VI of example 8.
Fig. 11: DSC-TGA spectrum of form VI of example 8.
Fig. 12: IR spectrum of the solid obtained in example 0.
Fig. 13: test example 4 results.
Detailed Description
1. X-ray powder diffraction (X-ray powder diffractometer, XRPD)
(1) Instrument model: bruker D8 advanced X-ray powder diffractometer
The testing method comprises the following steps: about 5-20 mg of sample (examples 1-4, examples 6-8) for XRPD detection
The detailed XRPD parameters are as follows:
x-ray generator: cu, K alpha,
Figure BDA0003942748480000101
light pipe voltage: 40kV, light pipe current: 40mA
Scanning range: 3-40 ° (2 theta)
Scanning step length: 0.02 degree
Sample tray: zero background sample tray.
(2) Instrument model: d2 PHASER desk-top X ray diffractometer
The testing method comprises the following steps: about 100-200 mg sample (example 0, example 5) for XRPD detection
The detailed XRPD parameters are as follows:
x-ray generator: cu, K alpha,
Figure BDA0003942748480000111
light pipe voltage: 30kV, light pipe current: 10mA
Scanning range: 3 ° -60 ° (2θ)
Scanning step length: 0.02 degree
Sample tray: zero background sample tray.
2. Differential scanning calorimetric analysis (Differential Scanning Calorimeter, DSC)
Instrument model: TA Discovery 250 differential scanning calorimeter
The testing method comprises the following steps: the sample was placed in a perforated aluminum crucible and heated to the final temperature at a ramp rate of 10 ℃/min after equilibration at 25 ℃.
Sample amount: 1-3 mg
Type of air flow: nitrogen gas
Flow rate: 50mL/min
Heating initiation temperature: 25 ℃.
3. Thermogravimetric analysis (Thermal Gravimetric Analyzer, TGA)
Instrument model: TA Discovery 55 thermogravimetric analyzer (TA, US)
The testing method comprises the following steps: the sample was placed in an aluminum crucible peeled in advance, and after the sample mass was automatically weighed in a TGA furnace, the sample was heated to the final temperature at a rate of 10 ℃/min.
Sample amount: 1-5 mg
Type of air flow: nitrogen gas
Sample cell airflow rate: 60mL/min
Heating initiation temperature: room temperature
Termination temperature: 250/300 ℃.
4. Dynamic moisture desorption analysis (DVS)
Instrument model: DVS Intrinsic dynamic steam adsorber (SMS)
The testing method comprises the following steps: a sufficient amount of sample (10-20 mg) was placed in the sample chamber previously peeled and automatically weighed. The sample was dried at 40℃until dm/dt was less than 0.002% (only for anhydrate, starting at 25℃for hydrate). Cool to 25 ℃, and start the test using the operating parameters in the table below.
Stage time 60 minutes
Drying/test temperature 40℃/25℃
Circulation Whole cycle
Each RH equilibration time 1 hour
Data storage rate 5 seconds
Total air flow 200sccm
Total air flow rate after the experiment 200sccm
5. High Performance Liquid Chromatography (HPLC)
Instrument model: agilent 1260 services (Waters, US)
Chromatographic column:
Figure BDA0003942748480000121
Express C18 4.6x 100mm,2.7μm
test conditions: wavelength 248nm; column temperature 40 DEG C
Flow rate: 1.0mL/min
Sample injection volume: 5. Mu.L.
6. Nuclear magnetic resonance spectrum (Nuclear Magnetic Resonance Spectroscopy NMRS)
Instrument model: bruker AVANCE III 400 (Bruker, GER)
Content and test solvent: 1 H-NMR, test solvent was DMSO-d6.
7. Infrared spectrum (Infrared Spectroscopy, IR)
Detection instrument: perkinElmer Spectrum 100FT-IR infrared spectrum analyzer
The testing method comprises the following steps: 3mg of the sample was weighed, diluted with KBr and tabletted, and examined at room temperatureThe specific parameters are as follows: detection range: 4000-400cm -1 Wavenumber, resolution: 4cm -1
Abbreviations: DCM: dichloromethane; DIPEA: diisopropylethylamine; DMF: n, N-dimethylformamide; EA: ethyl acetate; PE: petroleum ether; DMSO: dimethyl sulfoxide; TBTU: O-benzotriazol-N, N, N ', N' -tetramethylurea tetrafluoroboric acid; BOP: benzotriazol-1-yloxy tris (dimethylamino) phosphonium hexafluorophosphate; ATP: adenosine 5' -triphosphate; DTT:1, 4-dithiothreitol; MTT:3- (4, 5-dimethyl-2-thiazole) -2, 5-diphenyl tetrazolium bromide thiazole blue.
For a better understanding of the present application, reference is made to the following specific examples, which are not intended to limit the scope of the present application. The following preparation examples, comparative examples, test examples or test examples were selected according to conventional methods and conditions or according to the specifications of the commercial products without specifying the specific conditions.
Preparation example 1: preparation of Compounds of formula (A)
Figure BDA0003942748480000122
Step a: (2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidine (5.8236 g,28.958 mmol), 5-chloropyrazolo [1,5-a ]]A mixed solution of pyrimidine-3-carboxylic acid ethyl ester (6.534 g,28.958 mmol), n-butanol (50 mL) and diisopropylamine (8.780 g,86.874 mmol) was reacted at 100deg.C for 4h and concentrated under reduced pressure to give 5- ((2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester crude (B1). No purification was carried out and used directly in the next reaction, (ES, m/z): 391.05[ M+H ]] +
Step b: 5- ((2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) pyrazolo [1,5-a]The crude pyrimidine-3-carboxylic acid ethyl ester was dissolved in absolute ethanol (50 mL), stirred at 75deg.C until the system was clear and transparent, then LiOH (4.86 g,115.832 mmol) aqueous solution (50 mL) was added and stirred at 75deg.C for 5h. After cooling to room temperature, the mixture was concentrated under reduced pressure to remove absolute ethanol. Slowly dripping 1N HCl aqueous solution to adjust the pH to 3 to the upper limit4, a large amount of white solid is separated out, the mixture is stirred at room temperature for 30min and then is filtered, and a filter cake is washed by a small amount of purified water. The filter cake is collected, dried and weighed to obtain white powdery solid 5- ((2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) pyrazolo [1, 5-a) ]Pyrimidine-3-carboxylic acid (9.9 g). The filtrate was extracted with EA (2X 50 mL), the organic phases were combined, washed with water (2X 50 mL) and saturated aqueous NaCl solution (50 mL), and dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated under reduced pressure. Column chromatography purification (PE: ea=4:1-2:1, v/v), collection of the product points, concentration under reduced pressure, yield 5- ((2 r,4 s) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) pyrazolo [1,5-a as a white powdery solid]Pyrimidine-3-carboxylic acid (386 mg). Co-yield 5- ((2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid purification (B2, 10.284 g, 98%), (ES, m/z): 363.04[ M+H ]] +
Step c: 1-Boc-4- (4-aminophenyl) piperazine (918 mg,3.312 mmol) was added to a solution containing 5- ((2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (intermediate B2, 1000mg,2.76 mmol) was reacted with TBTU (1063 mg,3.312 mmol) in anhydrous DMF (10 mL) followed by dropwise addition of DIPEA (1284 mg,9.936 mmol) at 0deg.C overnight at room temperature. The reaction solution was stirred with water (50 mL) to precipitate a solid, which was filtered under reduced pressure to give a cake, which was dried in a vacuum oven to give tert-butyl 4- (4- (5- ((2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) pyrazolo [1, 5-a) ]Pyrimidine-3-carboxamido) phenylpiperazine-1-carboxylate (B3, 1320mg, 77%). (ES, m/z): 622.09[ M+H ]] +
Step d: to tert-butyl 4- (4- (5- ((2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide) phenylpiperazine-1-carboxylate (1.320 g,2.125 mmol) was added with DCM and CF 3 COOH (12 mL,3/1, v/v), stirring at room temperature for 4 hours, concentrating the reaction solution under reduced pressure, adding water (80 mL) and EA (10 mL) to the residue, adjusting the base (pH=9) with ammonia water, stirring to precipitate a solid, obtaining a filter cake by vacuum filtration, rinsing the filter cake with a small amount of water, air-drying, and weighing to obtain 5- ((2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) -N- (4- (piperazin-1-yl) phenyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (B4,227 mg, 82%),(ES,m/z):522.09[M+H] +
Step e: glycolic acid (306 mg,4.026 mmol) was added to a solution containing 5- ((2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) -N- (4- (piperazin-4-yl) phenyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (intermediate B4, 700mg, 1.348 mmol) was reacted with BOP (710 mg,1.610 mmol) in anhydrous DMF (10 mL) followed by dropwise addition of DIPEA (520 mg,4.026 mmol) at 0deg.C with stirring at room temperature for 4h. The reaction mixture was mixed with water (80 mL), the mixture was extracted with EA (55 mL. Times.2), and the mixture was extracted with H 2 O (80 mL), brine (80 mL), drying over anhydrous sodium sulfate, filtering, concentrating the filtrate under reduced pressure, eluting with a column chromatography silica gel column, eluting with 1% (v/v) MeOH-DCM followed by 2% (v/v) MeOH-DCM, collecting the product points and concentrating to give 5- ((2R, 4S) -2- (2, 5-difluorophenyl) -4-fluoropyrrolidin-1-yl) -N- (4- (4- (2-hydroxyacetyl) piperazin-1-yl) phenyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (A, 666mg, 86%), (ES, m/z): 580.14[ M+H ]] +1 H NMR(600MHz,DMSO-d 6 )δ9.810(s,1H),8.906-8.723(m,1H),8.283-8.229(m,1H),7.623(s,1H),7.343(s,1H),7.210(s,2H),7.061-6.842(m,4H),5.711-5.495(m,2H),4.631(t,J=5.4Hz,1H),4.556-4.548(m,1H),4.318-4.225(m,1H),4.150(d,J=5.4Hz,2H),3.637(s,2H),3.513(s,2H),3.124-3.106(m,4H),2.957-2.912(m,1H)。
Control preparation examples 1-5:
another: compounds D1 to D5 were prepared by reference to the preparation routes and procedures described in the patent documents WO2019029629A1 and WO2012034095A 1.
Figure BDA0003942748480000131
Figure BDA0003942748480000141
/>
Example 0: preparation of the Compound of formula (A) in solid form
A sample of preparation 1 (about 50 mg) was taken and pumped under heating (50 ℃ C.) using an oil pump for 3 hours to give an off-white solid with a purity of 98.6%. IR (KBr, cm-1): 3429.38, 1643.78, 1487.95, 1453.15, 1232.01, 1026.79. See fig. 12. The sample was subjected to X-ray powder diffraction and shown to be amorphous.
Example 1: preparation of Compound of formula (A) Crystal form I
The sample of example 0 (about 20 mg) was weighed into a sample bottle at room temperature, tetrahydrofuran (0.2 mL) was added to obtain a clear solution, methanol (1.4 mL) was gradually added dropwise thereto, stirred at room temperature for 1 day, and filtered to obtain a solid. The sample was taken for X-ray powder diffraction and shown to be a crystalline solid (form I) with good crystallinity, the spectrum is shown in figure 1 and the XRPD diffraction peak data is shown in table 1.
Table 1 example 1 XRPD diffraction peak data table for form I
Figure BDA0003942748480000142
Figure BDA0003942748480000151
Note that: peaks with a relative peak intensity >4.0% were selected and listed in the table.
Example 2: preparation of Compound of formula (A) Crystal form II
An appropriate amount of form I obtained in example 1 was weighed and dried at 50 ℃ for 3 hours to give a solid. The sample was taken for X-ray powder diffraction and showed a crystalline solid (form II) with good crystallinity, the spectrum is shown in figure 2 and the XRPD diffraction peak data is shown in table 2. Samples were taken for DSC-TGA testing, the DSC plot showed an endothermic peak at 58.39 ℃and 155.27 ℃each, and the TGA plot showed 0.1910% weight loss of the samples between room temperature and 75℃as shown in FIG. 3.
TABLE 2 XRPD diffraction peak data for example 2 form II
Figure BDA0003942748480000152
Example 3: preparation of crystalline form III of the Compound of formula (A)
The sample of example 0 (about 30 mg) was weighed into a sample bottle at room temperature, acetonitrile (0.3 mL) was added to prepare a solution, stirred at room temperature for 3 days, filtered to obtain a solid, and dried at 50℃for 3 hours. The sample was taken for X-ray powder diffraction and showed a crystalline solid (form III) with good crystallinity, the spectrum is shown in fig. 4 and the XRPD diffraction peak data is shown in table 3. Samples were taken for DSC-TGA testing and DSC plots showed an endothermic peak at 182.29C, see FIG. 5.
TABLE 3 XRPD diffraction peak data for example 3 form III
Peak position (2θ) ° Relative strength% Peak position (2θ) ° Relative strength% Peak position (2θ) ° Relative strength%
5.618 100.0 15.802 47.2 19.901 6.7
7.431 5.5 16.272 5.6 23.688 9.9
10.937 8.8 17.053 5.9 23.845 5.4
11.319 8.4 18.428 13.2
13.284 25.5 18.983 9.2
Note that: peaks with a relative peak intensity >5.0% were selected and listed in the table.
Example 4: preparation of Compound of formula (A) form IV
The sample of example 0 (about 20 mg) was weighed into a sample bottle at room temperature, tetrahydrofuran (0.2 mL) was added to each to prepare a clear solution, isopropanol (1.3 mL) was gradually added dropwise thereto, stirred at room temperature for 1 day, filtered to obtain a solid, and dried at 50℃for 3 hours. The sample was taken for X-ray powder diffraction and showed a crystalline solid (form IV) with good crystallinity, the spectrum is shown in fig. 6 and the XRPD diffraction peak data is shown in table 4. Samples were taken for DSC-TGA testing, with DSC plots showing two endotherm peaks at 64.82℃and 192.07 ℃and TGA plots showing 1.1526% weight loss of samples between room temperature and 75℃as shown in FIG. 7.
TABLE 4 XRPD diffraction peak data for example 4 form IV
Peak position (2θ) ° Relative strength% Peak position (2θ) ° Relative strength% Peak position (2θ) ° Relative strength%
5.390 11.4 17.184 100.0 23.371 15.2
8.527 97.1 17.897 45.7 24.172 8.7
10.046 59.4 18.994 11.8 25.017 27.2
13.117 16.6 19.463 14.1 25.893 24.3
14.091 3.8 19.958 19.0 26.453 15.6
15.258 7.0 21.259 9.7 29.194 5.4
15.731 20.1 22.052 6.8
16.505 9.0 22.958 10.7
Note that: peaks with a relative peak intensity >3.5% were selected and listed in the table.
Examples 5 to 6: preparation of Compound of formula (A) form IV
Using the samples obtained in example 0, form IV was also obtained under the conditions as listed in table 5. Wherein example 5 refers to the preparation method of example 4.
TABLE 5 Experimental conditions and results for examples 5-6
Figure BDA0003942748480000161
TABLE 6 XRPD diffraction peak data for example 5 form IV
Peak position (2θ) ° Relative strength% Peak position (2θ) ° Relative strength% Peak position (2θ) ° Relative strength%
5.479 9.6 17.202 74.6 22.932 11.6
8.573 100.0 17.468 18.6 23.369 13.6
10.066 55.4 17.928 32.2 23.737 14.3
11.527 5.8 18.612 5.3 24.219 8.2
13.129 23.9 19.015 11.3 25.023 34.6
13.999 10.0 19.469 15.4 25.358 20.6
14.229 6.5 19.982 24.8 25.841 22.6
15.144 5.1 21.321 5.5 26.421 11.6
15.692 19.8 22.032 7.0
16.457 8.4 22.493 5.7
Note that: peaks with a relative peak intensity >5% were selected and listed in the table.
Example 7: preparation of Compound of formula (A) form V
The sample of example 0 (about 34 mg) was weighed into a sample bottle, dissolved in acetone (0.5 mL), stirred at 5℃for 1 day, and filtered to give a solid at room temperature. The sample was taken for X-ray powder diffraction and showed a crystalline solid (form V) with good crystallinity, the spectrum is shown in fig. 9 and the XRPD diffraction peak data is shown in table 7.
TABLE 7 XRPD diffraction peak data for example 7 form V
Peak position (2θ) ° Relative strength% Peak position (2θ) ° Relative strength% Peak position (2θ) ° Relative strength%
6.003 100.0 17.582 8.1 23.448 52.5
7.048 12.4 18.190 45.0 23.986 4.1
10.203 4.1 18.526 4.3 24.451 9.9
11.571 4.3 20.247 8.7 25.395 18.4
12.113 7.2 20.578 6.6 26.628 4.3
12.729 42.2 20.783 8.9 28.120 19.5
14.225 9.6 20.954 6.1 28.793 7.1
16.727 27.7 23.228 18.3
Note that: peaks with a relative peak intensity >4.0% were selected and listed in the table.
Example 8: preparation of Compound of formula (A) Crystal form VI
An appropriate amount of form V obtained in example 7 was weighed and dried at 50 ℃ for 3 hours to give a solid. The sample was taken for X-ray powder diffraction and showed a crystalline solid (form VI) with good crystallinity, the spectrum is shown in fig. 10 and the XRPD diffraction peak data is shown in table 8. Samples were taken for DSC-TGA testing, the DSC plot showed an endothermic peak at 184.47 ℃and the TGA plot showed a weight loss of 0.253% for samples between room temperature and 200℃as seen in FIG. 11.
TABLE 8 XRPD diffraction peak data for example 8 form VI
Figure BDA0003942748480000171
Note that: peaks with a relative peak intensity >3.0% were selected and listed in the table.
Comparative examples 1 to 2
An appropriate amount of the sample of example 0 was weighed, dissolved in tetrahydrofuran to prepare a clear solution of 10mg/0.3mL, the resulting solution was equally divided in centrifuge tubes, 0.3mL each tube, then 0.1mL of solvent 2 shown in the following table was added to each tube, covered with a film and the wells were pricked, and the solvent was evaporated at room temperature.
Table 9 Experimental conditions and results for comparative examples 1-2
Comparative example Solvent 2 Results
Comparative example 1 Methanol Adhesive article
Comparative example 2 Acetone (acetone) Adhesive article
Test example 1: solid stability experiments of different Crystal forms of the Compound of formula (A)
An appropriate amount of form III (example 3) of the compound of formula (A) was weighed into a vial, and placed under conditions of high temperature (60 ℃, sealing) and acceleration (40 ℃ C./75% RH, open) for 7 days, and samples were subjected to purity detection and X-ray powder diffraction, respectively, to examine the stability of form III (example 3) of the compound of formula (A) under different conditions, and the results are shown in Table 10.
A sample of form IV (example 4) was weighed and placed in a vial in appropriate amounts and placed under conditions of high temperature (60 ℃, sealed), high humidity (25 ℃/92.5% rh, open) and acceleration (40 ℃/75% rh, open) for 7 days, respectively, and the samples were subjected to purity detection and X-ray powder diffraction, respectively, and the stability of form IV (example 4) of the compound of formula (a) under different conditions was examined, and the results are shown in table 11.
Table 10 results of solid stability experiments for form III
Figure BDA0003942748480000181
TABLE 11 solid stability test results for form IV
Figure BDA0003942748480000182
The data indicate that: form III of example 3 remained chemically stable and form stable under both high temperature and accelerated conditions; form IV of example 4 remains chemically stable and form stable under high temperature, high humidity and acceleration conditions.
Test example 2: DVS test of different crystalline forms of the compound of formula (a)
The samples of the crystal form III and the crystal form IV of the example 3 and the example 4 are respectively placed in a DVS sample chamber for testing. The sample after DVS test was taken and subjected to X-ray powder diffraction, and the results are shown in table 12.
TABLE 12 DVS test results for different crystal forms
Examples and initial crystalline forms DVS post-crystal form
EXAMPLE 3 form III The crystal form is unchanged
EXAMPLE 4 form IV The crystal form is unchanged
The data indicate that: form III and form IV remained unchanged after DVS testing.
Test example 1 TRK kinase inhibition test
1. The operation steps are as follows:
1.1 kinase reaction:
adding a certain concentration into the compound plate in turnGradient test compound, enzyme solution (negative control wells added to kinase buffer (1 Xkinase buffer (Cisbio, cat#62EZBFDD), pH 7.5, 5mM MgCl 2 1mM DTT)), and centrifuged at 1000rpm for 30 seconds. Plates were sealed and incubated in a constant temperature incubator at 25℃for 30 minutes. A substrate solution of TK-Sub-biotin (Cisbio, cat#61 TKOBL) and ATP (Sigma, cat#R0441) was prepared, and the substrate mixed solution was added to a 384-well plate and centrifuged at 1000rpm for 30 seconds. Plates were sealed and incubated in a constant temperature incubator at 25℃for 60 minutes.
Figure BDA0003942748480000183
Figure BDA0003942748480000191
1.2 kinase assay:
TK antibody and XL665 were diluted, mixed and added to assay plates and centrifuged at 1000rpm for 30 seconds. Plates were sealed and incubated in a constant temperature incubator at 25℃for 60 minutes. The assay plate was placed on an Envision machine to read. (HTRF 665/615 ratio: 665nm signal value/615 nm signal value)
Inhibition ratio = (ratio) Negative control well -ratio of Compound pore ) Ratio (ratio) Negative control well -ratio of Enzyme-free control wells )×100%
1.3 data analysis and Curve fitting
Fitting data in XLFit excel plug-in version 5.4.0.8 to obtain IC 50 Values.
1.4QC parameters
The reference compound is contained in each plate and its IC 50 Each time within 3 times.
2. Test results: as shown in Table 13
TABLE 13 inhibitory Activity of different Compounds against TRK kinase
Figure BDA0003942748480000192
Remarks: the RXDX-101, LOXO-195 and LOXO-101 are all disclosed compounds and can be commercially obtained into marketed products (pharmaceutical or chemical grade products); a compound of formula (a): preparation example 1 sample.
The results show that: the compounds of formula (A) exhibit higher kinase inhibitory activity in various kinases, better than or equal to RXDX-101, LOXO-195 and LOXO-101 in TRKA, TRKB, TRKC and TRKC-G696A, while the inhibitory activity in various mutant drug-resistant kinases (G595R, G667C, G623R) is significantly better than RXDX-101, LOXO-195 and LOXO-101.
Test example 2, ALK and ROS1 kinase inhibition assay
1. The operation steps are as follows:
1.1 kinase reaction:
the compound was diluted to a concentration in DMSO and diluted 4-fold gradient. Respectively adding a compound with a certain concentration into a 384-well plate, and incubating for 10min at room temperature with an enzyme solution and DMSO; adding fluorescein labeled peptide, and incubating at 28deg.C for a certain time with ATP (sigma, cat. No.: A7699-1G, lot. No.: 987-65-5); adding a stop solution. And (5) reading.
Inhibition ratio formula corresponding to single concentration: inhibition ratio= (OD Negative control well -OD Compound pore )/(OD Negative control well -OD Enzyme-free control wells )×100%
Figure BDA0003942748480000201
1.2 data analysis and Curve fitting
Fitting data in XLFit excel plug-in version 4.3.1 to obtain IC 50 Values, results are shown in table 14.
TABLE 14 ALK and ROS1 kinase inhibitory Activity of different Compounds
Figure BDA0003942748480000202
Note that: a compound of formula (a): preparation example 1 sample.
The results show that: the compound shown in the formula (A) has stronger inhibition activity in ROS1 kinase, and is obviously superior to RXDX-101 and LOXO-101 and is superior to LOXO-195; the ALK kinase inhibitor also has good inhibition activity, and is obviously superior to LOXO-101 and LOXO-195.
Test example 3 in vitro cytostatic test
1. Cell lines
6 sources of cell lines for the test: kang Yuanbo Biotechnology (Beijing) Co., ltd
Cell type: murine B cells
Culture medium: RPMI-1640+10% FBS
2. Test method
Cells in the logarithmic growth phase were harvested and counted using a platelet counter. Uniformly inoculating a cell suspension with a certain density into a 96-well plate by blowing, vibrating and uniformly dispersing the cell suspension into each hole by 100 mu L of each hole; adding 100 mu L of a drug solution with a certain concentration gradient into each hole, and setting three compound holes for each drug concentration; CO at 37 DEG C 2 Culturing in an incubator for 72 hours; MTT working solution (5 mg/mL) was added, 20. Mu.L per well; acting at 37 ℃ for 4 hours; the plate centrifuge is centrifuged at 1000rpm/min for 5min, 180 mu L of culture medium is sucked and removed, 150 mu L of DMSO is added, a micropore oscillator is used for shaking and mixing uniformly, the bottom of the plate is wiped clean, and an Optical Density (OD) value is detected at 550nm of an enzyme-labeling instrument.
3. Data analysis
Inhibition ratio = (control well OD-test well OD)/(control well OD-blank well OD) ×100% and half inhibition concentration IC was calculated from each concentration inhibition ratio using SPSS software 50 Values.
4. Test results: the results are shown in Table 15:
TABLE 15 inhibitory Activity of different Compounds against different cell lines
Figure BDA0003942748480000203
Figure BDA0003942748480000211
Note that: a compound of formula (a): preparation example 1 sample.
TABLE 16 inhibitory Activity of control compounds against different cell lines
Figure BDA0003942748480000212
The results show that: the compound shown in the formula (A) shows better in-vitro cell activity in various wild type and mutant drug-resistant cell strains, and is obviously superior to RXDX-101, LOXO-195, LOXO-101 and compounds D1-D5 in the prior art.
Test example 4: research on in vivo mechanism of Compounds of formula (A)
1. Test method
1.1 model preparation:
taking mutant drug-resistant cells Ba/F3 LMNA-NTRK1-G595R in logarithmic growth phase, collecting, and re-suspending in serum-free culture medium to make cell concentration 6×10 7 -10×10 7 Each mL, and an equal volume of Matrigel (Matrigel) was added to the cell suspension to give a final cell concentration of 3X 10 7 -5×10 7 And each mL. Nunununu mice (4-6 weeks, females) were inoculated subcutaneously with 0.1mL of tumor cell suspension in the anterior axilla with an inoculum size of 3×10 6 -5×10 6 Animal models were prepared.
1.2 test groups:
the maximum tumor diameter and the minimum tumor diameter of the nude mice transplanted tumor are measured by a vernier caliper, and the tumor volume is calculated: the calculation formula of Tumor Volume (TV) is: v=1/2×a×b 2 Wherein a and b represent the maximum and minimum diameters, respectively, of the tumor mass. Nude mice with proper tumor volume are selected, and animals are equally divided into 7 groups (200-300 mm) according to tumor volume by adopting a random digital method 3 ) Each group of 3.
1.3 administration of drugs
The compound represented by the formula (A) was administered by gavage according to the body weight of the animal in a volume of 10mL/kg, and the compound represented by the formula (A) was formulated to have a desired administration concentration using "3% DMSO+96% HP-. Beta. -CD (0.5 g/mL) +1% HCl".
Three control groups are adopted, and tumor tissues are frozen and stored 4 hours after the solvent is added. The other groups were given 100mg/kg of the compound represented by the formula (A), and tumor tissues were frozen at 0.25h, 1h, 4h, 8h, 12h and 24h, respectively.
1.4 protein extraction and quantification
Tumor tissue of a certain mass was taken and added to a corresponding volume of protein lysate (RIPA lysate (Thermo Fisher, cat. No. 89900) of protease inhibitor (clomplete, mini, EDTA-free, EASYpack; roche, cat. No. 04693159001) of phosphatase inhibitor (photosstop, EASYpack; roche, cat. No. 04906837001) =8:1:1), homogenized and lysed in ice bath for 30min. The supernatant was subjected to low-temperature high-speed centrifugation, and BCA protein was quantified (according to BCA protein quantification kit (Tiangen, cat# PA 115-01). Finally, the protein concentration is regulated to uniform concentration by using a lysate, and then loading buffer solution (loading buffer) is added, and the mixture is boiled for 10min at 100 ℃.
1.5Western-blot
4-20% of 10-hole preformed adhesive is adopted; the loading amount is 100 mug; 140V electrophoresis for 1-1.5h; wet converting 300mA for 1.5-2 h; blocking with 5% BSA for 2-3h; incubation at 4deg.C overnight (Trk 1:5000, p-Trk, PLCγ1, p-PLCγ1, AKT, p-AKT, actin 1:1000); 4×5min0.1% TBST wash; the secondary antibody was incubated for 2h (1:5000) at room temperature, ECL luminescence, exposure.
Figure BDA0003942748480000221
2. Test results: as shown in fig. 13.
From the test results, it can be seen that: as time goes on, TRK, p-TRK, p-PLC gamma 1 and p-AKT in FIG. 13 are all obviously reduced, and the compounds shown in the formula (A) can obviously reduce the protein levels of TRK and p-TRK, so that phosphorylation of p-PLC gamma 1/PLC gamma 1 and p-AKT/AKT is effectively inhibited, and cell growth and proliferation are regulated and controlled.
Test example 5: in vivo efficacy experiment of compound on NTRK mutant drug-resistant tumor model
Test method
1.1 model preparation
Taking the logarithmic phase of growthCells were collected, resuspended in serum-free medium to a cell concentration of 6X 10 7 -10×10 7 Each mL, and an equal volume of Matrigel (Matrigel) was added to the cell suspension to give a final cell concentration of 3X 10 7 -5×10 7 And each mL. Nunununu mice (4-6 weeks, females) were inoculated subcutaneously with 0.1mL of tumor cell suspension in the anterior axilla with an inoculum size of 3×10 6 -5×10 6 Animal models were prepared.
1.2 test group
The maximum tumor diameter and the minimum tumor diameter of the nude mice transplanted tumor are measured by a vernier caliper, and the tumor volume is calculated: the calculation formula of Tumor Volume (TV) is: v=1/2×a×b 2 Wherein a and b represent the maximum and minimum diameters, respectively, of the tumor mass. Nude mice with proper tumor volume are selected, and animals are equally divided into 7 groups (100-200 mm) according to tumor volume by adopting a random digital method 3 ) Each group had 6.
1.3 observations index
The animals were dosed by stomach irrigation at the same day as the day of the group, the dosing volume was 10mL/kg, LOXO-195 was formulated as the desired dosing solution using 0.5% CMC-Na, and the compound of formula (A) was formulated as the desired dosing solution using "3% DMSO+96% HP-beta-CD (0.5 g/mL) +1% HCl". Tumor diameters were measured twice weekly and tumor volumes were calculated. The specific indexes are as follows:
animal body weight: animals were weighed daily prior to the morning dosing, and a weight loss of greater than 20% was defined as a toxic response to the drug (the next day of last dosing was observed);
tumor volume (Tumor volume, TV) =v=1/2×a×b 2 Wherein a and b represent the maximum and minimum diameters, respectively, of the tumor mass (the next day of last administration was observed);
relative tumor proliferation rate T/C (%): T/C (%) =trtv/crtv×100% (TRTV: dosing group RTV, CRTV: control group RTV);
tumor Growth Inhibition (TGI) = [1- (Ti-T0)/(Vi-V0) ]x100%. (wherein Ti represents the average tumor volume of the administration group on a certain day; T0 represents the average tumor volume of the administration group at the beginning of administration; vi represents the average tumor volume of the vehicle control group on a certain day (same day as Ti; V0 represents the average tumor volume of the vehicle control group at the beginning of administration);
Tumor inhibition rate: at the end of the experiment, animals were sacrificed by cervical removal, tumor masses were removed and weighed, photographed, and tumor inhibition was calculated as tumor inhibition = (average tumor weight of control group-average tumor weight of dosing group)/average tumor weight of control group x 100%.
Test results
2.1Ba/F3 LMNA-NTRK1-G667C model
2.1.1 Effect of drug on body weight of tumor-bearing mice
The body weight of each dose group of each compound had an ascending trend, and the ascending trend was more pronounced than that of the control group. The weight of each dose group of each compound is obviously increased, which is possibly related to the compound, and the condition of mice is better and the weight is obviously increased due to the inhibition of tumor growth. The results are shown in Table 17.
2.1.2 effects of drugs on tumor weight and tumor inhibition Rate in tumor-bearing mice
The data results show that: at the same administration dose (100 mg/kg), the compound shown in the formula (A) has more remarkable inhibition on tumor growth compared with LOXO-195; further, the compound represented by the formula (A) (100 mg/kg) also exhibited a better tumor suppressing effect than the LOXO-195 group (200 mg/kg) at a higher administration dose. The results are shown in Table 17.
TABLE 17 in vivo results of Ba/F3 LMNA-NTRK1-G667C model
Figure BDA0003942748480000231
/>
Figure BDA0003942748480000241
2.2Ba/F3 LMNA-NTRK1-G595R model
2.2.1 Effect of drug on body weight of tumor-bearing mice
The body weight of each dose group of each compound had an ascending trend, and the ascending trend was more pronounced than that of the control group. The weight of each dose group of each compound is obviously increased, which is possibly related to the compound, and the condition of mice is better and the weight is obviously increased due to the inhibition of tumor growth. The results are shown in Table 17.
2.2.2 effects of drugs on tumor weight and tumor inhibition Rate in tumor-bearing mice
The data results show that: compared with LOXO-195 (100 mg/kg), the compound shown in the formula (A) can realize remarkable inhibition of tumor tissue weight at a lower administration dosage (50 mg/kg), and the tumor weight inhibition rate is more than 90%. The results are shown in Table 18.
TABLE 18 in vivo results for Ba/F3 LMNA-NTRK1-G595R model
Figure BDA0003942748480000242
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this application that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Claims (11)

1. A compound of formula (A) in solid form,
Figure FDA0003942748470000011
preferably, the compound represented by the formula (A) in solid form is characterized by comprising characteristic peaks (+ -4 cm) at the following positions by infrared spectrometry using a tabletting method -1 ):3429,1643,1488,1453,1232,1027;
Further preferred, the compound of formula (a) in solid form has an infrared spectrum substantially as shown in fig. 12.
2. A compound of formula (a) in crystalline form.
3. The compound of formula (a) in crystalline form according to claim 2, characterized in that it is in crystalline form I of the compound of formula (a), using Cu-ka radiation, having an X-ray powder diffraction pattern with characteristic diffraction peaks at the following 2Θ angles: (±0.2°): 9.2 17.9, 18.5; alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 8.9,9.2, 17.9, 18.5, 23.8;
Alternatively, its X-ray powder diffraction pattern using Cu-ka radiation has characteristic diffraction peaks (±0.2°) at the following 2θ angles: 8.9,9.2, 10.1, 17.9, 18.5, 23.8, 28.0;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 8.9,9.2, 10.1, 16.1, 17.9, 18.5, 23.8, 27.0, 28.0;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 8.9,9.2, 10.1, 16.1, 16.6, 17.9, 18.5, 23.8, 27.0, 28.0;
alternatively, it has an X-ray powder diffraction pattern substantially as shown in figure 1.
4. The compound of formula (a) in crystalline form according to claim 2, characterized in that it is form II of the compound of formula (a), using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 9.1 18.3, 20.6;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.4,9.1, 18.3, 20.6;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ angles: (±0.2°): 6.4,9.1, 18.3, 20.6, 27.7;
Alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.4,9.1, 18.3, 18.9, 20.6, 27.7;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.4,9.1, 18.3, 18.9, 20.6, 22.4, 23.9, 27.7;
alternatively, it has an X-ray powder diffraction pattern substantially as shown in figure 2;
or, the differential scanning calorimetric curve has an endothermic peak at 155.27 +/-5 ℃;
alternatively, it has a DSC profile substantially as shown in figure 3.
5. The compound of formula (a) in crystalline form according to claim 2, characterized in that it is form III of the compound of formula (a), using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 5.6 13.3, 15.8, 18.4;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 5.6 13.3, 15.8, 18.4, 23.7;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 5.6 10.9, 13.3, 15.8, 18.4, 19.0, 23.7;
Alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 5.6 10.9, 11.3, 13.3, 15.8, 17.1, 18.4, 19.0, 23.7;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 5.6,7.4, 10.9, 11.3, 13.3, 15.8, 17.1, 18.4, 19.0, 19.9, 23.7;
alternatively, it has an X-ray powder diffraction pattern substantially as shown in figure 4;
or, the differential scanning calorimetric curve has an endothermic peak at 182.29 +/-5 ℃;
alternatively, it has a DSC profile substantially as shown in figure 5.
6. The compound of formula (a) in crystalline form according to claim 2, characterized in that it is form IV of the compound of formula (a), using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 8.5 10.0, 17.2, 17.9;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ angles: 8.5 10.0, 13.1, 17.2, 17.9, 25.0;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 8.5 10.0, 13.1, 17.2, 17.9, 20.0, 25.0;
Alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 8.5 10.0, 13.1, 15.7, 17.2, 17.9, 20.0, 25.0, 25.9;
alternatively, it has an X-ray powder diffraction pattern substantially as shown in figure 6 or figure 8.
Or, the differential scanning calorimetric curve has an endothermic peak at 192.07 +/-5 ℃;
alternatively, it has a DSC profile substantially as shown in figure 7.
7. The compound of formula (a) in crystalline form according to claim 2, characterized in that it is form V of the compound of formula (a), using Cu-ka radiation, having an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.0 12.7, 18.2;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.0 12.7, 18.2, 23.4;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.0 12.7, 16.7, 18.2, 23.4;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.0 12.7, 16.7, 18.2, 23.4, 25.4, 28.1;
Alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.0,7.0, 12.7, 14.2, 16.7, 18.2, 23.4, 24.5, 25.4, 28.1;
alternatively, it has an X-ray powder diffraction pattern substantially as shown in figure 9.
8. The compound of formula (a) in crystalline form according to claim 2, characterized in that it is form VI of the compound of formula (a), using Cu-ka radiation, having an X-ray powder diffraction pattern with characteristic diffraction peaks (±0.2°): 6.1 16.9, 22.3;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.1 16.9, 18.6, 22.3;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.1,7.4, 11.5, 16.9, 18.6, 22.3;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.1,7.4, 11.5, 16.9, 18.6, 19.4, 22.3, 25.3;
alternatively, using Cu-ka radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks (±0.2°): 6.1,7.4, 11.5, 16.9, 18.6, 19.4, 20.1, 22.3, 25.3, 28.6;
Alternatively, it has an X-ray powder diffraction pattern substantially as shown in figure 10.
9. A crystalline composition comprising one or more of form I of claim 3, form II of claim 4, form III of claim 5, form IV of claim 6, form V of claim 7, form VI of claim 8; preferably, the form II comprises more than 50% by weight of the crystalline composition; preferably 60% or more; more preferably 70% or more; more preferably 80% or more; more preferably 90% or more; more preferably 95% or more;
or, the crystalline form III comprises more than 50% by weight of the crystalline composition; preferably 60% or more; more preferably 70% or more; more preferably 80% or more; more preferably 90% or more; more preferably 95% or more;
or, the form IV comprises more than 50% by weight of the crystalline composition; preferably 60% or more; more preferably 70% or more; more preferably 80% or more; more preferably 90% or more; more preferably 95% or more;
or the crystalline form VI comprises more than 50% by weight of the crystalline composition; preferably 60% or more; more preferably 70% or more; more preferably 80% or more; more preferably 90% or more; more preferably 95% or more.
10. A pharmaceutical composition comprising a compound of formula (a) according to claim 1 in solid form, or a compound of formula (a) according to claim 2 in crystalline form, or a crystalline composition according to claim 9;
preferably, the pharmaceutical composition comprises one or more of form I of claim 3, form II of claim 4, form III of claim 5, form IV of claim 6, form V of claim 7, form VI of claim 8.
11. Use of a compound of formula (a) according to claim 1 in solid form, a compound of formula (a) according to any one of claims 2-8 in crystalline form, a crystalline composition according to claim 9, or a pharmaceutical composition according to claim 10 as a medicament or in the preparation of a medicament; preferably, the medicament is for the prevention and/or treatment of one or more of TRK, ROS or ALK mediated diseases; further preferably, the disease is selected from pain diseases, cell proliferative diseases, inflammatory diseases, neurodegenerative diseases or infectious diseases; further preferably, the cell proliferative disease is a tumor or cancer; further preferred, the tumor or cancer is a solid tumor or hematological tumor; further preferred, the tumor or cancer is hematological malignancy, lung cancer, breast cancer, ovarian cancer, prostate cancer, pancreatic cancer, glioma, colorectal cancer, melanoma, cancer of the head and neck, gall bladder cancer, thyroid cancer, glioblastoma, gastric cancer, neuroblastoma, or salivary gland cancer; preferably, the lung cancer is non-small cell lung cancer.
CN202211427370.4A 2021-11-15 2022-11-14 Solid form aza-condensed ring amide compound and use thereof Pending CN116120323A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111345026 2021-11-15
CN2021113450266 2021-11-15

Publications (1)

Publication Number Publication Date
CN116120323A true CN116120323A (en) 2023-05-16

Family

ID=86310695

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211427370.4A Pending CN116120323A (en) 2021-11-15 2022-11-14 Solid form aza-condensed ring amide compound and use thereof

Country Status (2)

Country Link
CN (1) CN116120323A (en)
WO (1) WO2023083356A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116120322A (en) * 2021-11-15 2023-05-16 石药集团中奇制药技术(石家庄)有限公司 Salt of aza-condensed ring amide compound, crystal form and use thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012034095A1 (en) * 2010-09-09 2012-03-15 Irm Llc Compounds and compositions as trk inhibitors
CA2858958C (en) * 2011-12-12 2016-10-04 Dr. Reddy's Laboratories Ltd. Substituted pyrazolo[1,5-a]pyridine as tropomyosin receptor kinase (trk) inhibitors
CN111542522B (en) * 2017-08-11 2021-10-15 苏州韬略生物科技有限公司 Substituted pyrazolopyrimidines useful as kinase inhibitors
CN111039946A (en) * 2018-10-15 2020-04-21 上海轶诺药业有限公司 Preparation and application of imidazo aromatic ring compounds
US20230227460A1 (en) * 2020-05-15 2023-07-20 Cspc Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd Fused aza-heterocyclic amide compound and use thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116120322A (en) * 2021-11-15 2023-05-16 石药集团中奇制药技术(石家庄)有限公司 Salt of aza-condensed ring amide compound, crystal form and use thereof

Also Published As

Publication number Publication date
WO2023083356A1 (en) 2023-05-19

Similar Documents

Publication Publication Date Title
KR20090107013A (en) Crystalline forms of thiazolidinedione derivative and its manufacturing method
JP7100625B2 (en) Crystal form, salt type and method for producing the substituted 2-H-pyrazole derivative
JP2023525380A (en) Aza-fused cyclic amide compound and use thereof
CN116120323A (en) Solid form aza-condensed ring amide compound and use thereof
CN113861191B (en) Crystal forms of CDK4/6 active compound and application thereof
CN117295742A (en) New forms of compound I and their use
CN112174982A (en) Lopitinib crystal form and preparation method thereof
CN115466266B (en) mTOR protein degradation targeting chimeric body, preparation method and application thereof
WO2020007219A1 (en) Crystalline form of egfr inhibitor and preparation method therefor
WO2022063229A1 (en) Salt of arylaminoquinazoline-containing compound, and preparation method therefor and use thereof
CN116768856A (en) Salt of substituted amino six-membered nitrogen heterocyclic compound, crystal form, preparation method and application thereof
WO2023036252A1 (en) Pyrrolopyrimidine or pyrrolopyridine derivative and medical use thereof
CN116120322A (en) Salt of aza-condensed ring amide compound, crystal form and use thereof
CN114437077A (en) Compounds useful as kinase inhibitors and uses thereof
AU2018278283A1 (en) Pyridoquinazoline derivatives useful as protein kinase inhibitors
CN112020357B (en) Salt of indazolyl-containing tricyclic derivative and crystal form thereof
WO2023104107A1 (en) Salt of 3,4-dihydroisoquinoline compound and use thereof
CN108299419B (en) Novel crystal forms of novel EGFR kinase inhibitor and preparation method thereof
CN116115618A (en) Medicine for treating tumor
WO2022083733A1 (en) Bruton tyrosine kinase inhibitor compound in solid form and use thereof
CN109422739B (en) Deuterated indoleamine 2, 3-dioxygenase inhibitor and application thereof
EA043251B1 (en) CRYSTAL FORM OF THE COMPOUND FOR INHIBITION OF CDK4/6 ACTIVITY AND ITS APPLICATION
TW202311267A (en) Polymorph of imidazolidinone compound, preparation method therefor and use thereof
CN117247382A (en) Crystal forms of pyridopyrimidinone compounds
WO2022033552A1 (en) Cdk kinase inhibitor, preparation method therefor, pharmaceutical composition, and application

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination