WO2020253836A1 - 喹唑啉类化合物的晶型、盐型及其制备方法 - Google Patents

喹唑啉类化合物的晶型、盐型及其制备方法 Download PDF

Info

Publication number
WO2020253836A1
WO2020253836A1 PCT/CN2020/097158 CN2020097158W WO2020253836A1 WO 2020253836 A1 WO2020253836 A1 WO 2020253836A1 CN 2020097158 W CN2020097158 W CN 2020097158W WO 2020253836 A1 WO2020253836 A1 WO 2020253836A1
Authority
WO
WIPO (PCT)
Prior art keywords
crystal form
compound
formula
angles
ray powder
Prior art date
Application number
PCT/CN2020/097158
Other languages
English (en)
French (fr)
Inventor
陈新海
张丽
周凯
于衍新
胡伯羽
章晖宇
Original Assignee
南京明德新药研发有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 南京明德新药研发有限公司 filed Critical 南京明德新药研发有限公司
Priority to CN202080044785.6A priority Critical patent/CN114144418B/zh
Priority to EP20827468.8A priority patent/EP3988553A4/en
Priority to US17/620,464 priority patent/US20220242851A1/en
Priority to JP2021575887A priority patent/JP7350374B2/ja
Publication of WO2020253836A1 publication Critical patent/WO2020253836A1/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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 invention relates to a crystal form, a salt form of a compound as a Pan-HER tyrosine kinase inhibitor and a preparation method thereof, and to its application in the preparation of a medicine for treating solid tumors.
  • HER Human epidermal growth factor receptor
  • EGFR Human epidermal growth factor receptor
  • HER is overexpressed or abnormal in a variety of tumor cells such as breast cancer, non-small cell lung cancer, gastric cancer, pancreatic cancer, ovarian cancer, colorectal cancer, head and neck squamous cell carcinoma, malignant glioma and prostate cancer
  • the phenomenon of activation has been shown that the overexpression or abnormal activation of HER is closely related to the degree of differentiation, malignancy and prognosis of tumors (Baselga. J., Oncologist 2002, 7, 2-8). Therefore, inhibiting HER has become a hot spot in anti-tumor drug research.
  • Pan-HER tyrosine kinase irreversible inhibitor inhibits HER1, HER2 and HER4 at the same time. Studies have shown that this irreversible inhibition of HER family receptors can not only increase the activity of the drug, but also reduce the generation of drug resistance. Some resistant tumor cell lines, such as the H1975 cell line resistant to erlotinib, have a significant inhibitory effect. The only irreversible pan-HER tyrosine kinase inhibitors that have been approved for marketing are Afatinib and Neratinib. Many inhibitors are in clinical research, such as Poziotinib, Dacomitinib, and Canertinib. There is still unmet market demand.
  • Poziotinib (control compound 1) is a pan-HER inhibitor developed by WO2008150118, and its structure is as follows.
  • Patent WO2015043515 discloses the control compound 2, which has EGFR and HER2 inhibitory effects, and its structure is as follows.
  • the present invention provides crystal form A of the compound represented by formula (I), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 17.4 ⁇ 0.2°, 20.8 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 14.6 ⁇ 0.2°, 15.2 ⁇ 0.2°, 17.4 ⁇ 0.2°, 19.0 ⁇ 0.2°, 20.8 ⁇ 0.2°, 22.1 ⁇ 0.2°, 24.1 ⁇ 0.2°, 29.5 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 12.0 ⁇ 0.2°, 12.7 ⁇ 0.2°, 14.6 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.9 ⁇ 0.2°, 17.4 ⁇ 0.2°, 19.0 ⁇ 0.2°, 20.8 ⁇ 0.2°, 22.1 ⁇ 0.2°, 24.1 ⁇ 0.2°, 29.5 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3°, 3.7°, 6.9°, 11.2°, 12.0°, 12.7°, 14.6°, 15.2 °, 15.9°, 16.9°, 17.4°, 17.9°, 18.5°, 19.0°, 19.4°, 20.1°, 20.4°, 20.8°, 22.1°, 22.4°, 23.5°, 24.1°, 24.4°, 25.4°, 25.8°, 26.6°, 27.1°, 28.3°, 29.0°, 29.5°.
  • the XRPD pattern of the above crystal form A is shown in FIG. 1.
  • the XRPD pattern analysis data of the above-mentioned crystal form A is shown in Table 1:
  • the present invention provides a compound represented by formula (II).
  • the present invention provides the B crystal form of the compound represented by formula (II), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 8.0 ⁇ 0.2°, 11.4 ⁇ 0.2°, 18.7 ⁇ 0.2°.
  • the present invention provides crystal form B of the compound represented by formula (II), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 12.7 ⁇ 0.2°, 18.7 ⁇ 0.2°, 24.8 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 8.0 ⁇ 0.2°, 11.4 ⁇ 0.2°, 12.7 ⁇ 0.2°, 15.0 ⁇ 0.2°, 16.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.7 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 11.4 ⁇ 0.2°, 12.7 ⁇ 0.2°, 16.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 20.0 ⁇ 0.2°, 22.4 ⁇ 0.2°, 23.1 ⁇ 0.2°, 24.8 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 8.0 ⁇ 0.2°, 11.4 ⁇ 0.2°, 12.7 ⁇ 0.2°, 15.0 ⁇ 0.2°, 16.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.7 ⁇ 0.2°, 22.4 ⁇ 0.2°, 23.1 ⁇ 0.2°, 24.8 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 6.825°, 8.04°, 11.412°, 12.72°, 13.454°, 15.007°, 16.067°, 16.817 °, 17.402°, 18.009°, 18.691°, 20.012°, 20.748°, 21.265°, 22.03°, 22.383°, 22.779°, 23.075°, 23.648°, 24.475°, 24.77°, 25.593°, 25.914°, 26.248°, 27.076°, 27.527°, 28.557°, 28.987°.
  • the XRPD pattern of the above-mentioned crystal form B is shown in FIG. 2.
  • the XRPD pattern analysis data of the above-mentioned crystal form B is shown in Table 2:
  • the differential scanning calorimetry curve of the above-mentioned crystal form B has an endothermic peak at 207.4 ⁇ 3.0°C.
  • the DSC spectrum of the above-mentioned crystal form B is shown in FIG. 3.
  • the present invention provides a compound represented by formula (III).
  • the present invention provides crystal form C of the compound represented by formula (III), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 7.6 ⁇ 0.2°, 8.5 ⁇ 0.2°.
  • the present invention provides crystal form C of the compound represented by formula (III), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 13.8 ⁇ 0.2°, 17.7 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form C has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 7.6 ⁇ 0.2°, 8.5 ⁇ 0.2°, 11.3 ⁇ 0.2°, 13.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 17.7 ⁇ 0.2°, 18.3 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form C has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 7.6 ⁇ 0.2°, 11.3 ⁇ 0.2°, 13.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 17.7 ⁇ 0.2°, 23.7 ⁇ 0.2°, 24.9 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form C has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 7.6 ⁇ 0.2°, 8.5 ⁇ 0.2°, 11.3 ⁇ 0.2°, 13.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 17.7 ⁇ 0.2°, 18.3 ⁇ 0.2°, 23.7 ⁇ 0.2°, 24.1 ⁇ 0.2°, 24.9 ⁇ 0.2°, 26.6 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form C has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 7.6 ⁇ 0.2°, 8.5 ⁇ 0.2°, 11.3 ⁇ 0.2°, 13.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 17.7 ⁇ 0.2°, 18.3 ⁇ 0.2°, 22.6 ⁇ 0.2°, 23.7 ⁇ 0.2°, 24.9 ⁇ 0.2°, 26.6 ⁇ 0.2°. .
  • the X-ray powder diffraction pattern of the above-mentioned crystal form C has characteristic diffraction peaks at the following 2 ⁇ angles: 3.315°, 3.621°, 7.651°, 8.528°, 10.898°, 11.3°, 13.081°, 13.472 °, 13.767°, 15.247°, 15.93°, 16.47°, 17.103°, 17.733°, 18.322°, 18.759°, 19.173°, 19.369°, 20.123°, 20.532°, 21.146°, 21.914°, 22.629°, 22.841°, 23.709°, 24.127°, 24.938°, 25.581°, 26.221°, 26.588°, 27.005°, 27.658°, 28.109°, 28.508°, 29°, 29.289°.
  • the XRPD pattern of the above-mentioned crystal form C is shown in FIG. 4.
  • the XRPD pattern analysis data of the above-mentioned crystal form C is shown in Table 3:
  • the present invention provides the D crystal form of the compound represented by the formula (III), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 3.5 ⁇ 0.2°, 9.5 ⁇ 0.2°, 10.5 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form D has characteristic diffraction peaks at the following 2 ⁇ angles: 3.5 ⁇ 0.2°, 9.5 ⁇ 0.2°, 10.5 ⁇ 0.2°, 14.0 ⁇ 0.2°, 16.3 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.0 ⁇ 0.2°, 23.4 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form D has characteristic diffraction peaks at the following 2 ⁇ angles: 3.5 ⁇ 0.2°, 9.5 ⁇ 0.2°, 10.5 ⁇ 0.2°, 14.0 ⁇ 0.2°, 16.3 ⁇ 0.2°, 18.7 ⁇ 0.2°, 23.4 ⁇ 0.2°, 25.6 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form D has characteristic diffraction peaks at the following 2 ⁇ angles: 3.502°, 9.505°, 10.509°, 12.166°, 12.446°, 14.008°, 15.385°, 15.684 °, 16.275°, 17.495°, 18.681°, 19.016°, 19.404°, 20.356°, 20.954°, 21.802°, 22.489°, 23.019°, 23.457°, 24.245°, 24.977°, 25.583°, 26.253°, 26.688°, 27.359°, 28.425°, 28.762°, 29.821°, 31.581°, 32.411°, 32.907°, 33.155°, 34.579°, 36.315°.
  • the XRPD pattern of the above-mentioned crystal form D is shown in FIG. 5.
  • the XRPD pattern analysis data of the above-mentioned crystal form D is shown in Table 4:
  • the present invention provides a compound represented by formula (IV).
  • the present invention provides the E crystal form of the compound represented by formula (IV), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 12.8 ⁇ 0.2°, 13.2 ⁇ 0.2°.
  • the present invention provides the E crystal form of the compound represented by formula (IV), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 16.2 ⁇ 0.2°, 23.2 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form E has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 12.8 ⁇ 0.2°, 13.2 ⁇ 0.2°, 14.9 ⁇ 0.2°, 16.2 ⁇ 0.2°, 18.7 ⁇ 0.2°, 23.2 ⁇ 0.2°, 24.8 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form E has characteristic diffraction peaks at the following 2 ⁇ angles: 3.3 ⁇ 0.2°, 8.1 ⁇ 0.2°, 13.2 ⁇ 0.2°, 14.9 ⁇ 0.2°, 16.2 ⁇ 0.2°, 18.7 ⁇ 0.2°, 23.2 ⁇ 0.2°, 24.8 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form E has characteristic diffraction peaks at the following 2 ⁇ angles: 3.305°, 3.601°, 8.088°, 11.28°, 12.762°, 13.177°, 14.206°, 14.875 °, 16.256°, 17.264°, 18.661°, 19.993°, 21.397°, 21.934°, 23.196°, 24.755°, 25.209°, 27.381°, 28.76°, 30.802°, 32.96°, 33.325°.
  • the XRPD pattern of the above-mentioned crystal form E is shown in FIG. 6.
  • the present invention provides the F crystal form of the compound represented by formula (IV), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 3.6 ⁇ 0.2°, 7.5 ⁇ 0.2°, 16.6 ⁇ 0.2°.
  • the present invention provides the F crystal form of the compound represented by formula (IV), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 3.6 ⁇ 0.2°, 12.2 ⁇ 0.2°, 16.6 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form F has characteristic diffraction peaks at the following 2 ⁇ angles: 3.6 ⁇ 0.2°, 7.5 ⁇ 0.2°, 12.2 ⁇ 0.2°, 15.1 ⁇ 0.2°, 16.6 ⁇ 0.2°, 23.6 ⁇ 0.2°, 24.6 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form F has characteristic diffraction peaks at the following 2 ⁇ angles: 3.62°, 6.981°, 7.501°, 10.896°, 11.18°, 12.198°, 13.734°, 14.789 °, 15.111°, 16.611°, 18.146°, 19.586°, 20.104°, 20.375°, 20.924°, 21.084°, 21.537°, 22.111°, 22.614°, 23.638°, 24.614°, 25.077°.
  • the XRPD pattern of the above crystal form F is shown in FIG. 7.
  • the XRPD pattern analysis data of the above-mentioned crystal form F is shown in Table 6:
  • the present invention provides a method for preparing the crystal form of compound A represented by formula (I), including:
  • the solvent is selected from methanol, ethanol and ethyl acetate.
  • the present invention provides a method for preparing the crystal form of compound B represented by formula (II), including:
  • the solvent is selected from tetrahydrofuran.
  • the stirring temperature is 35°C to 45°C.
  • the stirring time is 24 hours to 48 hours.
  • the weight-volume ratio of the compound to the solvent is 1 g:10-50 mL.
  • the present invention provides another preparation method of compound B crystalline form represented by formula (II), including:
  • the solvent is selected from ethanol and ethyl acetate.
  • the stirring temperature is 65°C to 75°C.
  • the stirring time is 16 hours to 24 hours.
  • the weight-volume ratio of the compound to the solvent is 1 g: 6-10 mL.
  • the present invention provides a method for preparing the crystal form of compound C represented by formula (III), including:
  • the solvent is selected from acetonitrile.
  • the stirring temperature is 35°C to 45°C.
  • the stirring time is 24 hours to 48 hours.
  • the weight-volume ratio of the compound to the solvent is 1 g:7-10 mL.
  • the present invention provides a method for preparing the crystal form of compound D represented by formula (III), including:
  • the method for preparing the above-mentioned crystal form D wherein the solvent is selected from ethyl acetate, acetonitrile-water (1:1) and methanol-water (1:1).
  • the stirring temperature is 35°C to 45°C.
  • the stirring time is 24 hours to 48 hours.
  • the weight-volume ratio of the compound to the solvent is 1 g:7-10 mL.
  • the present invention also provides the application of the above crystal form in the preparation of drugs for treating Pan-HER tyrosine kinase inhibitor related diseases.
  • the present invention also provides the application of the above-mentioned compound or crystal form in the preparation of medicines for treating Pan-HER tyrosine kinase inhibitor-related diseases.
  • the above application is characterized in that the Pan-HER tyrosine kinase inhibitor-related drugs are drugs for tumors.
  • 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 combining them with other chemical synthesis methods, and those skilled in the art.
  • Well-known equivalent alternatives, preferred implementations include but are not limited to the embodiments of the present invention.
  • the solvent used in the present invention is commercially available.
  • the present invention uses the following abbreviations: DCM stands for dichloromethane; DMF stands for N,N-dimethylformamide; DMSO stands for dimethyl sulfoxide; EtOH stands for ethanol; MeOH stands for methanol; TFA stands for trifluoroacetic acid; ATP stands for Adenosine triphosphate; HEPES stands for 4-hydroxyethylpiperazine ethanesulfonic acid; MgCl 2 stands for magnesium dichloride.
  • the compound of the present invention has good crystal form stability and is easy to prepare medicine; the compound of the present invention has obvious inhibitory activity on HER1, HER2 and HER4, and has obvious inhibitory activity on the proliferation of NCI-N87, OE21 and OE19 cells.
  • the compound of the present invention has excellent pharmacokinetic properties; the compound of the present invention has a significant effect of inhibiting tumor growth and has no significant effect on the weight of mice at the effective dose, and the safety is better.
  • Test method Approximately 10-20mg sample is used for XRPD detection.
  • Tube voltage 40kV
  • tube current 40mA
  • Test method Take a sample (0.5 ⁇ 1mg) and place it in a DSC aluminum pan for testing. Under the condition of 25mL/min N 2 and at a heating rate of 10°C/min, heat the sample from 30°C to 300°C.
  • Figure 1 is an XRPD spectrum of Cu-K ⁇ radiation of the crystal form of compound A of formula (I).
  • Figure 2 is an XRPD spectrum of Cu-K ⁇ radiation of the B crystal form of compound of formula (II).
  • Figure 3 is a DSC spectrum of the crystal form of formula (II) B.
  • Fig. 4 is an XRPD spectrum of Cu-K ⁇ radiation of the crystal form C of compound of formula (III).
  • Figure 5 is an XRPD spectrum of Cu-K ⁇ radiation of the D crystal form of compound of formula (III).
  • Fig. 6 is an XRPD spectrum of Cu-K ⁇ radiation of the crystal form E of compound of formula (IV).
  • Figure 7 is an XRPD spectrum of Cu-K ⁇ radiation of the F crystal of the compound of formula (IV).
  • the purpose of this test is to detect the in vitro inhibitory activity of the compound against HER1 (ErbB1), HER2 (ErbB2), and HER4 (ErbB4).
  • the enzymes used in this experiment are human ErbB1, ErbB2 and ErbB4.
  • Eurofins Pharma Discovery Service provides an activity detection method.
  • the results of the inhibitory activity of the test compounds against HER1, HER2, and HER4 are shown in Table 11.
  • test compound buffer 5 ⁇ L
  • peptide substrate poly(Glu, Tyr) 4:1) (2.5 ⁇ L)
  • ErbB 4-20ng, 2.5 ⁇ L
  • MnCl 2 50mM, 1.25 ⁇ L
  • DH 2 O 3.75 ⁇ L
  • [ ⁇ - 33 P]ATP 10 ⁇ L
  • the meter detects the intensity of the emitted photons, and compares the cpm (times per minute) of the enzyme sample with the cpm of the internal control sample.
  • the level of the photon intensity reflects the strength of the tyrosine kinase activity.
  • the compound of formula (I) has obvious inhibitory activity on HER1, HER2 and HER4.
  • Experimental purpose to detect the inhibitory activity of the test compound on cell proliferation.
  • Luciferase in Cell-Titer-Glo reagent uses luciferin, oxygen and ATP as reaction substrates to produce oxyluciferin and release energy in the form of light. Since the luciferase reaction requires ATP, the total amount of light produced by the reaction is directly proportional to the total amount of ATP that reflects cell viability.
  • Cell line NCI-N87 cell line (ATCC-CRL-5822), BT-474 cell line (ATCC-HTB-20), OE21 (ECACC-96062201)
  • Cell culture medium (RPMI 1640 medium (Invitrogen#22400-105) ; 10% serum Invitrogen#10090148; L-glutamine 1 ⁇ , Gibco#25030-081; Double antibody Hyclone#SV30010)
  • the compound stock solution is 10 mM, and the compound is diluted with DMSO to make the initial concentration 4 mM. Add the compound to the compound mother liquid plate, 9 ⁇ L per well.
  • Test result The test result is shown in Table 12.
  • the mother liquor of the compound to be tested was diluted 3-fold with DMSO to a total of 10 concentrations.
  • the compound to be tested was further diluted with the cell culture medium into 10 ⁇ compound solution (maximum concentration 100 ⁇ M containing 1% DMSO).
  • 10 ⁇ compound solution maximal concentration 100 ⁇ M containing 1% DMSO.
  • the final concentration of the model standard control Staurosporine (staurosporine) and all tested compounds are all starting at 10 ⁇ M, with a gradient of 3 times dilution, and a total of 10 test concentration points.
  • Inhibition percentage% (ZPE-sample detection value)/(ZPE-HPE) ⁇ 100%
  • the processed data will be analyzed with GraphPad Prism 6 analysis software for nonlinear regression analysis to obtain a dose-response curve, and calculate the half inhibitory concentration (IC 50 ) of the test compound on OE19 cells.
  • IC 50 half inhibitory concentration
  • the compound of formula (I) has obvious inhibitory activity on the proliferation of NCI-N87, OE21 and OE19 cells.
  • mice Female BALB/c nude mice, 6-8 weeks old, weighing 18-22 grams; Supplier: Shanghai Lingchang Biotechnology Co., Ltd. provides experimental methods and procedures:
  • Human gastric cancer NCI-N87 cells cultured in a monolayer in vitro, culture conditions are RPMI-1640 medium with 10% fetal bovine serum, 100U/mL penicillin, 100U/mL0 streptomycin and 2mM glutamine, 37°C, 5% CO 2 culture. Use pancreatin-EDTA for routine digestion and passage twice a week. When the cell saturation is 80%-90%, the cells are collected, counted, and seeded.
  • NCI-N87 cells PBS+Matrigel, 1:1 subcutaneously on the right back of each mouse, and start grouping when the average tumor volume reaches 158mm 3
  • test compound was formulated into clear solutions of 0.04mg/mL and 0.08mg/mL, the solvent was 10% NMP (N-methylpyrrolidone) + 10% ethylene glycol stearate + 80% water
  • the experimental index is to investigate whether the tumor growth is inhibited, delayed or cured.
  • the tumor diameter was measured with vernier calipers twice a week.
  • TGI (%) reflects the tumor growth inhibition rate.
  • TGI(%) [1-(Average tumor volume at the end of a certain treatment group-average tumor volume at the beginning of the treatment group)/(Average tumor volume at the end of treatment in the solvent control group- The average tumor volume at the start of treatment in the solvent control group)] ⁇ 100%.
  • T/C (%) average tumor volume at the end of a certain treatment group/average tumor volume at the end of the solvent control group ⁇ 100%.
  • the body weight of experimental animals is used as a reference index for indirect determination of drug toxicity.
  • the body weight of the mice in the treatment group of this model showed a downward trend, and there was no other morbidity or death.
  • Table 13 Evaluation of anti-tumor efficacy of compounds of formula (I) on human gastric cancer NCI-N87 cell subcutaneous xenograft tumor model
  • c.p value is calculated based on tumor volume.
  • the in vivo efficacy of the compound of formula (I) in human gastric cancer NCI-N87 cell subcutaneous xenograft tumor model was evaluated.
  • the treatment group formula (I) compound @0.8mg/kg is equivalent to the reference compound Poziotinib@0.8mg/kg, and both show significant tumor inhibition; the low-dose group formula (I) compound
  • the reference compound Poziotinib has equivalent or better efficacy.
  • mice Female BALB/c nude mice, 6-8 weeks old, weighing 18-22 grams; Supplier: Shanghai Xipuer-Bikai Laboratory Animal Co., Ltd.
  • Human gastric cancer NCI-N87 cells were cultured in a monolayer in vitro.
  • the culture conditions were RPMI-1640 medium with 10% fetal bovine serum, 100U/mL penicillin, 100 ⁇ g/mL streptomycin, 37°C, 5% CO 2 culture.
  • test compound was prepared as a clear solution of 0.05 mg/mL, and the solvent was 10% NMP (N-methylpyrrolidone) + 10% ethylene glycol stearate + 80% water
  • the experimental index is to investigate whether the tumor growth is inhibited, delayed or cured.
  • the tumor diameter was measured with vernier calipers twice a week.
  • TGI (%) reflects the tumor growth inhibition rate.
  • TGI(%) [1-(Average tumor volume at the end of a certain treatment group-average tumor volume at the beginning of the treatment group)/(Average tumor volume at the end of treatment in the solvent control group- The average tumor volume at the start of treatment in the solvent control group)] ⁇ 100%.
  • T/C (%) average tumor volume at the end of a certain treatment group/average tumor volume at the end of the solvent control group ⁇ 100%.
  • the body weight of experimental animals is used as a reference index for indirect determination of drug toxicity.
  • the body weight of the mice in the treatment group of this model showed a downward trend, and there was no other morbidity or death.
  • Table 14 Evaluation of antitumor efficacy of compounds of formula (I) on human gastric cancer NCI-N87 cell subcutaneous xenograft tumor model (calculated based on tumor volume on the 21st day after administration)
  • c.p value is calculated based on tumor volume.
  • mice Female BALB/c nude mice, 6-8 weeks old, weighing 17-23 grams; Supplier: Shanghai Xipuer-Bikai Laboratory Animal Co., Ltd.
  • Human esophageal cancer OE21 cells are cultured in a monolayer in vitro, and the culture conditions are RPMI-1640 medium with 10% fetal bovine serum, 100U/mL penicillin, 100U/mL 0 streptomycin and 2mM glutamine, and culture at 37°C with 5% CO 2 .
  • OE21 cells PBS
  • test compound is formulated into a clear or suspension solution of 0.15mg/mL ⁇ 0.3mg/mL, the solvent is 10% NMP + 10% ethylene glycol stearate + 80% water
  • the experimental index is to investigate whether the tumor growth is inhibited, delayed or cured.
  • the tumor diameter was measured with vernier calipers twice a week.
  • TGI (%) reflects the tumor growth inhibition rate.
  • TGI(%) [(1-(Average tumor volume at the end of a certain treatment group-average tumor volume at the beginning of the treatment group))/(Average tumor at the end of treatment in the solvent control group Volume-the average tumor volume at the start of treatment in the solvent control group)] ⁇ 100%.
  • T/C(%) Average tumor volume at the end of a certain treatment group/average tumor volume at the end of the solvent control group ⁇ 100%.
  • mice in the Poziotinib@2/1.5mg/kg and formula (I) compound@2/1.5mg/kg administration groups all showed varying degrees of weight loss and dander; Poziotinib@2/1.5 mg/kg.
  • 1 mouse and 3 mice lost more than 15% of their body weight and were treated with drug withdrawal, and the whole group of mice had a dark yellow urine toxicity prompt;
  • the dose of Poziotinib@2mg/kg was reduced to 1.5mg/kg
  • the dose of compound of formula (I)@2mg/kg was reduced to 1.5mg/kg. After the dose adjustment, the weight of each group recovered significantly.
  • Table 15 Evaluation of antitumor efficacy of compounds of formula (I) on OE21 xenograft tumor model (calculated based on tumor volume on the 21st day after administration)
  • Tumor growth inhibition is calculated by T/C and TGI, refer to Experimental Example 4.
  • the p value is calculated based on the tumor volume on the 21st day after administration, and the statistical method of One-Way ANOVA is used to compare with the Vehicle group.
  • the efficacy of the compound of formula (I) at a low dose of 0.5 mpk is equivalent to that of the reference compound Poziotnib and the compound of formula (I) at a high dose of 1.5 mpk.
  • the compound of formula (I) has a significant anti-tumor effect, and its efficacy is equivalent to that of the reference compound Poziotinib, but the weight of the mice in the high-dose group of the reference compound decreased significantly, and the weight of 3 mice lost more than 15%. The drug was stopped, and there were mice in the whole group. Dark yellow urine indicates toxicity.
  • the compound of formula (I) has little effect on the weight of mice, no drug withdrawal, no dark yellow urine toxicity prompt, and the tolerability is significantly better than the reference compound.
  • the compound of formula (I) has a lower effective dose, better tolerability, and a better safety window than the reference compound Poziotinib.
  • This experiment aims to investigate the plasma pharmacokinetics of female BALB/c mice after a single intravenous injection and intragastric administration of the examples of this patent and the reference compound.
  • mice Female BALB/c nude mice, 7-9 weeks old, weighing 17-23 grams; Supplier: Shanghai Xipuer-Bikai Laboratory Animal Co., Ltd.
  • Sample collection The experimental animals collect 0.03 mL of blood samples from saphenous vein puncture at each time point, and record the actual blood collection time. All blood samples were put into a commercial EDTA-K2 anticoagulation tube with a specification of 1.5 mL (the supplier is Jiangsu Kangjian Medical Products Co., Ltd.). After the blood sample was collected, within half an hour, the supernatant plasma was collected by centrifugation at 4°C and 3000g for 10 minutes, quickly placed in dry ice, and stored in a refrigerator at -80°C for LC-MS/MS analysis.
  • C max peak concentration
  • T max peak time
  • T 1/2 time required to clear half of the compound
  • AUC 0-last 0- the concentration integral area within the last sampling time.
  • mice The results of pharmacokinetic studies in mice showed that the half-life of the compound of formula (I) was longer than that of the reference compound, and the oral plasma exposure was significantly better than that of the reference compound Poziotinib and the control compound 2.
  • the results of the CYP inhibitory activity of the compound of the present invention in human liver microsomes show that the compound of formula (I) and Poziotinib have no inhibitory activity on CYP1A2 and CYP3A4.
  • the inhibitory activity of the compound of formula (I) on CYP2C19 is equivalent to that of the reference compound Poziotinib, and the activity on CYP2C9 and CYP2D6 is improved, which is 2 and 6 times better than the reference compound, respectively.
  • the compound of formula (I) has improved CYP activity and is better than the reference compound, so the risk of drug-drug interaction is lower.

Landscapes

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

Abstract

一种作为Pan-HER酪氨酸激酶抑制剂的化合物的晶型、盐型及其制备方法,并涉及其在制备治疗实体瘤的药物中的应用。

Description

喹唑啉类化合物的晶型、盐型及其制备方法
本申请主张如下优先权:
CN201910533240.0,申请日2019-06-19。
技术领域
本发明涉及一种作为Pan-HER酪氨酸激酶抑制剂的化合物的晶型、盐型及其制备方法,并涉及其在制备治疗实体瘤的药物中的应用。
背景技术
人类表皮生长因子受体(HER,EGFR)是蛋白酪氨酸激酶家族的一员,广泛分布于人体各组织细胞膜上,可以调节细胞的增殖,生长,转移和凋亡。其结构由三部分组成:胞外的配体结合区、跨膜区以及胞内的酪氨酸激酶区。根据受体的结构差异,可以将HER区分为四种亚型,分别为HER1(EGFR,ErbB-1)、HER2(ErbB-2)、HER3(ErbB-3)及HER4(ErbB-4)。研究发现,HER在乳腺癌、非小细胞肺癌、胃癌、胰腺癌、卵巢癌、结直肠癌、头颈部鳞癌、恶性胶质瘤以及前列腺癌等多种肿瘤细胞中均存在过表达或异常激活的现象。另外,研究表明,HER的过表达或异常激活与肿瘤的分化程度、恶性程度及预后等密切相关(Baselga.J.,Oncologist 2002,7,2-8)。因此,抑制HER成为了抗肿瘤药物研究的热点。
目前,已经上市的靶向性的HER抑制剂包括吉非替尼(Gefitinib)、埃罗替尼(Erlotinib)、拉帕替尼(Lapatinib)等。然而,由于这些上市药物有效应答率不高,容易产生耐药性以及存在一些毒副作用,因此迫切需要研发其他具有优良抗肿瘤效果,同时能够克服耐药性,并且耐受性良好的抗肿瘤药物。
Pan-HER酪氨酸激酶不可逆抑制剂同时抑制HER1、HER2以及HER4,研究表明,这种对HER家族受体的不可逆抑制除了能提高药物的活性外,还能减少耐药性的产生,同时对一些产生耐药的肿瘤细胞系,如对埃罗替尼耐药的H1975细胞系,具有显著的抑制效果。目前已经被批准上市的pan-HER酪氨酸激酶不可逆抑制剂只有Afatinib和Neratinib,多个抑制剂处于临床研究,如:Poziotinib,Dacomitinib,Canertinib,仍有未满足的市场需求。
因此,有必要进一步开发Pan-HER酪氨酸激酶不可逆抑制剂,用于癌症的治疗。
Poziotinib(对照化合物1)是WO2008150118开发的pan-HER抑制剂,其结构如下。
Figure PCTCN2020097158-appb-000001
专利WO2015043515公开了对照化合物2,具有EGFR和HER2抑制作用,其结构如下。
Figure PCTCN2020097158-appb-000002
发明内容
本发明提供了式(I)所示化合物的A晶型,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:3.3±0.2°,17.4±0.2°,20.8±0.2°。
Figure PCTCN2020097158-appb-000003
本发明的一些方案中,上述A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,14.6±0.2°,15.2±0.2°,17.4±0.2°,19.0±0.2°,20.8±0.2°,22.1±0.2°,24.1±0.2°,29.5±0.2°。
本发明的一些方案中,上述A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,12.0±0.2°,12.7±0.2°,14.6±0.2°,15.2±0.2°,15.9±0.2°,17.4±0.2°,19.0±0.2°,20.8±0.2°,22.1±0.2°,24.1±0.2°,29.5±0.2°。
本发明的一些方案中,上述A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3°,3.7°,6.9°,11.2°,12.0°,12.7°,14.6°,15.2°,15.9°,16.9°,17.4°,17.9°,18.5°,19.0°,19.4°,20.1°,20.4°,20.8°,22.1°,22.4°,23.5°,24.1°,24.4°,25.4°,25.8°,26.6°,27.1°,28.3°,29.0°,29.5°。
本发明的一些方案中,上述A晶型,其XRPD图谱如图1所示。
本发明的一些方案中,上述A晶型的XRPD图谱解析数据如表1所示:
表1式(I)化合物A晶型的XRPD解析数据
Figure PCTCN2020097158-appb-000004
Figure PCTCN2020097158-appb-000005
本发明提供了式(Ⅱ)所示化合物。
Figure PCTCN2020097158-appb-000006
本发明提供了式(Ⅱ)所示化合物的B晶型,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:8.0±0.2°,11.4±0.2°,18.7±0.2°。
本发明提供了式(Ⅱ)所示化合物的B晶型,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:12.7±0.2°,18.7±0.2°,24.8±0.2°。
本发明的一些方案中,上述B晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:8.0±0.2°,11.4±0.2°,12.7±0.2°,15.0±0.2°,16.1±0.2°,18.7±0.2°,20.0±0.2°,20.7±0.2°。
本发明的一些方案中,上述B晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:11.4±0.2°,12.7±0.2°,16.1±0.2°,18.7±0.2°,20.0±0.2°,22.4±0.2°,23.1±0.2°,24.8±0.2°。
本发明的一些方案中,上述B晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:8.0±0.2°,11.4±0.2°,12.7±0.2°,15.0±0.2°,16.1±0.2°,18.7±0.2°,20.0±0.2°,20.7±0.2°,22.4±0.2°,23.1±0.2°,24.8±0.2°。
本发明的一些方案中,上述B晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:6.825°,8.041°,11.412°,12.72°,13.454°,15.007°,16.067°,16.817°,17.402°,18.009°,18.691°,20.012°,20.748°,21.265°,22.03°,22.383°,22.779°,23.075°,23.648°,24.475°,24.77°,25.593°,25.914°,26.248°,27.076°,27.527°,28.557°,28.987°。
本发明的一些方案中,上述B晶型,其XRPD图谱如图2所示。
本发明的一些方案中,上述B晶型的XRPD图谱解析数据如表2所示:
表2式(II)化合物B晶型的XRPD解析数据
Figure PCTCN2020097158-appb-000007
本发明的一些方案中,上述B晶型,其差示扫描量热曲线在207.4±3.0℃有一个吸热峰的起始点。
本发明的一些方案中,上述B晶型,其DSC图谱如图3所示。
本发明提供了式(Ⅲ)所示化合物。
Figure PCTCN2020097158-appb-000008
本发明提供了式(III)所示化合物的C晶型,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:3.3±0.2°,7.6±0.2°,8.5±0.2°。
本发明提供了式(III)所示化合物的C晶型,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:3.3±0.2°,13.8±0.2°,17.7±0.2°。
本发明的一些方案中,上述C晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,7.6±0.2°,8.5±0.2°,11.3±0.2°,13.8±0.2°,15.2±0.2°,17.7±0.2°,18.3±0.2°。
本发明的一些方案中,上述C晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,7.6±0.2°,11.3±0.2°,13.8±0.2°,15.2±0.2°,17.7±0.2°,23.7±0.2°,24.9±0.2°。
本发明的一些方案中,上述C晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,7.6±0.2°,8.5±0.2°,11.3±0.2°,13.8±0.2°,15.2±0.2°,17.7±0.2°,18.3±0.2°,23.7±0.2°,24.1±0.2°,24.9±0.2°,26.6±0.2°。
本发明的一些方案中,上述C晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,7.6±0.2°,8.5±0.2°,11.3±0.2°,13.8±0.2°,15.2±0.2°,17.7±0.2°,18.3±0.2°,22.6±0.2°,23.7±0.2°,24.9±0.2°,26.6±0.2°。。
本发明的一些方案中,上述C晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.315°,3.621°,7.651°,8.528°,10.898°,11.3°,13.081°,13.472°,13.767°,15.247°,15.93°,16.47°,17.103°,17.733°,18.322°,18.759°,19.173°,19.369°,20.123°,20.532°,21.146°,21.914°,22.629°,22.841°,23.709°,24.127°,24.938°,25.581°,26.221°,26.588°,27.005°,27.658°,28.109°,28.508°,29°,29.289°。
本发明的一些方案中,上述C晶型,其XRPD图谱如图4所示。
本发明的一些方案中,上述C晶型的XRPD图谱解析数据如表3所示:
表3式(III)化合物C晶型的XRPD解析数据
Figure PCTCN2020097158-appb-000009
Figure PCTCN2020097158-appb-000010
本发明提供了式(III)所示化合物的D晶型,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:3.5±0.2°,9.5±0.2°,10.5±0.2°。
本发明的一些方案中,上述D晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.5±0.2°,9.5±0.2°,10.5±0.2°,14.0±0.2°,16.3±0.2°,18.7±0.2°,19.0±0.2°,23.4±0.2°。
本发明的一些方案中,上述D晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.5±0.2°,9.5±0.2°,10.5±0.2°,14.0±0.2°,16.3±0.2°,18.7±0.2°,23.4±0.2°,25.6±0.2°。
本发明的一些方案中,上述D晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.502°,9.505°,10.509°,12.166°,12.446°,14.008°,15.385°,15.684°,16.275°,17.495°,18.681°,19.016°,19.404°,20.356°,20.954°,21.802°,22.489°,23.019°,23.457°,24.245°,24.977°,25.583°,26.253°,26.688°,27.359°,28.425°,28.762°,29.821°,31.581°,32.411°,32.907°,33.155°,34.579°,36.315°。
本发明的一些方案中,上述D晶型,其XRPD图谱如图5所示。
本发明的一些方案中,上述D晶型的XRPD图谱解析数据如表4所示:
表4式(III)化合物D晶型的XRPD解析数据
Figure PCTCN2020097158-appb-000011
Figure PCTCN2020097158-appb-000012
本发明提供了式(IV)所示化合物。
Figure PCTCN2020097158-appb-000013
本发明提供了式(IV)所示化合物的E晶型,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:3.3±0.2°,12.8±0.2°,13.2±0.2°。
本发明提供了式(IV)所示化合物的E晶型,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:3.3±0.2°,16.2±0.2°,23.2±0.2°。
本发明的一些方案中,上述E晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,12.8±0.2°,13.2±0.2°,14.9±0.2°,16.2±0.2°,18.7±0.2°,23.2±0.2°,24.8±0.2°。
本发明的一些方案中,上述E晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,8.1±0.2°,13.2±0.2°,14.9±0.2°,16.2±0.2°,18.7±0.2°,23.2±0.2°,24.8±0.2°。
本发明的一些方案中,上述E晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.305°,3.601°,8.088°,11.28°,12.762°,13.177°,14.206°,14.875°,16.256°,17.264°,18.661°,19.993°,21.397°,21.934°,23.196°,24.755°,25.209°,27.381°,28.76°,30.802°,32.96°,33.325°。
本发明的一些方案中,上述E晶型,其XRPD图谱如图6所示。
本发明的一些方案中,上述E晶型的XRPD图谱解析数据如表5所示:
表5式(IV)化合物E晶型的XRPD解析数据
Figure PCTCN2020097158-appb-000014
本发明提供了式(IV)所示化合物的F晶型,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:3.6±0.2°,7.5±0.2°,16.6±0.2°。
本发明提供了式(IV)所示化合物的F晶型,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:3.6±0.2°,12.2±0.2°,16.6±0.2°。
本发明的一些方案中,上述F晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.6±0.2°,7.5±0.2°,12.2±0.2°,15.1±0.2°,16.6±0.2°,23.6±0.2°,24.6±0.2°。
本发明的一些方案中,上述F晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.62°,6.981°,7.501°,10.896°,11.18°,12.198°,13.734°,14.789°,15.111°,16.611°,18.146°,19.586°,20.104°,20.375°,20.924°,21.084°,21.537°,22.111°,22.614°,23.638°,24.614°,25.077°。
本发明的一些方案中,上述F晶型,其XRPD图谱如图7所示。
本发明的一些方案中,上述F晶型的XRPD图谱解析数据如表6所示:
表6式(IV)化合物F晶型的XRPD解析数据
Figure PCTCN2020097158-appb-000015
本发明提供了式(I)所示化合物A晶型的制备方法,包括:
(1)将式(I)所示化合物加入溶剂中,使其溶解;
(2)加热搅拌,冷却,过滤,干燥,得到式(I)化合物的A晶型。
本发明的一些方案中,上述A晶型的制备方法,其中,所述溶剂选自甲醇、乙醇和乙酸乙酯。
本发明提供了式(II)所示化合物B晶型的制备方法,包括:
(1)将式(I)所示化合物加入溶剂中,使其溶解;
(2)加入马来酸,搅拌,过滤,干燥,得到式(II)化合物的B晶型。
本发明的一些方案中,上述B晶型的制备方法,其中,所述溶剂选自四氢呋喃。
本发明的一些方案中,上述B晶型的制备方法,其中,搅拌温度为35℃~45℃。
本发明的一些方案中,上述B晶型的制备方法,其中,搅拌时间为24小时~48小时。
本发明的一些方案中,上述B晶型的制备方法,其中,化合物与溶剂的重量-体积比为1g:10~50mL。
本发明提供了式(II)所示化合物B晶型的另一种制备方法,包括:
(1)将式(I)所示化合物加入溶剂中,使其溶解;
(2)加热,加入马来酸,搅拌,析出固体,冷却,过滤,干燥,得到式(II)化合物的B晶型。
本发明的一些方案中,上述B晶型的制备方法,其中,所述溶剂选自乙醇、乙酸乙酯。
本发明的一些方案中,上述B晶型的制备方法,其中,搅拌温度为65℃~75℃。
本发明的一些方案中,上述B晶型的制备方法,其中,搅拌时间为16小时~24小时。
本发明的一些方案中,上述B晶型的制备方法,其中,化合物与溶剂的重量-体积比为1g:6~10mL。
本发明提供了式(III)所示化合物C晶型的制备方法,包括:
(1)将式(I)所示化合物和盐酸盐加入溶剂中,使其成混悬液;
(2)加热,搅拌,离心,干燥,得到式(III)化合物的C晶型。
本发明的一些方案中,上述C晶型的制备方法,其中,所述溶剂选自乙腈。
本发明的一些方案中,上述C晶型的制备方法,其中,搅拌温度为35℃~45℃。
本发明的一些方案中,上述C晶型的制备方法,其中,搅拌时间为24小时~48小时。
本发明的一些方案中,上述C晶型的制备方法,其中,化合物与溶剂的重量-体积比为1g:7~10mL。
本发明提供了式(III)所示化合物D晶型的制备方法,包括:
(1)将式(I)所示化合物和盐酸盐加入溶剂中,使其成混悬液;
(2)加热,搅拌,离心,干燥,得到式(III)化合物的D晶型。
本发明的一些方案中,上述D晶型的制备方法,其中,所述溶剂选自乙酸乙酯、乙腈-水(1:1)和甲醇-水(1:1)。
本发明的一些方案中,上述D晶型的制备方法,其中,搅拌温度为35℃~45℃。
本发明的一些方案中,上述D晶型的制备方法,其中,搅拌时间为24小时~48小时。
本发明的一些方案中,上述D晶型的制备方法,其中,化合物与溶剂的重量-体积比为1g:7~10mL。
本发明还提供了上述晶型在制备治疗Pan-HER酪氨酸激酶抑制剂相关病症的药物上的应用。
本发明还提供了上述化合物或晶型在制备治疗Pan-HER酪氨酸激酶抑制剂相关病症的药物上的应用。
本发明的一些方案中,上述应用,其特征在于,所述Pan-HER酪氨酸激酶抑制剂相关药物是用于肿瘤的药物。
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。
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。
本发明所使用的溶剂可经市售获得。本发明采用下述缩略词:DCM代表二氯甲烷;DMF代表N,N-二甲基甲酰胺;DMSO代表二甲亚砜;EtOH代表乙醇;MeOH代表甲醇;TFA代表三氟乙酸;ATP代表三磷酸腺苷;HEPES代表4-羟乙基哌嗪乙磺酸;MgCl 2代表二氯化镁。
技术效果
本发明化合物的晶型稳定性好,易于成药;本发明化合物对HER1、HER2和HER4抑制活性明显,并且对NCI-N87、OE21和OE19细胞增殖抑制活性明显。本发明化合物具有优异的药代动力学性质;本发明化合物有显著抑制肿瘤生长的效果且起效剂量下对小鼠体重无显著影响,安全性更好。
1.1 X-射线粉末衍射(X-ray powder diffractometer,XRPD)
仪器型号:Bruker D8 advance X-射线衍射仪
测试方法:大约10~20mg样品用于XRPD检测。
详细的XRPD参数如下:
光管:Cu,Cu:K-Alpha
Figure PCTCN2020097158-appb-000016
管电压:40kV,管电流:40mA.
发射狭缝:0.60mm
探测器狭缝:10.50mm
防散射狭缝:7.10mm
扫描范围(2θ角):3或4-40deg
扫描速率:10deg/min
样品盘转速:15rpm/0rpm
1.2差热分析(Differential Scanning Calorimeter,DSC)
仪器型号:DSC Q2000差示扫描量热仪
测试方法:取样品(0.5~1mg)置于DSC铝锅内进行测试,在25mL/min N 2条件下,以10℃/min的升温速率,加热样品从30℃到300℃。
1.3高效液相色谱分析方法
配样浓度:0.5mg/mL
固体稳定性试验HPLC方法色谱条件参见下表7:
表7
Figure PCTCN2020097158-appb-000017
附图说明
图1为式(I)化合物A晶型的Cu-Kα辐射的XRPD谱图。
图2为式(II)化合物B晶型的Cu-Kα辐射的XRPD谱图。
图3为式(II)B晶型的DSC谱图。
图4为式(III)化合物C晶型的Cu-Kα辐射的XRPD谱图。
图5为式(III)化合物D晶型的Cu-Kα辐射的XRPD谱图。
图6为式(IV)化合物E晶型的Cu-Kα辐射的XRPD谱图。
图7为式(IV)化合物F晶型的Cu-Kα辐射的XRPD谱图。
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。
实施例1:式(I)化合物的制备
Figure PCTCN2020097158-appb-000018
合成路线:
Figure PCTCN2020097158-appb-000019
第一步
将化合物1-1(900.0mg,3.93mmol)溶于N,N-二甲基甲酰胺(20mL),加入碳酸钾(1.09g,7.85mmol)和化合物Q-1(1.60g,7.07mmol)。反应液在75℃搅拌16小时。反应液冷却到20℃,加水(30mL)稀释,用乙酸乙酯(30mL x 2)萃取。合并的有机相,依次用水(40mL x 2),饱和食盐水(40mL x 2)洗涤,用无水硫酸钠干燥。过滤,滤液减压浓缩。剩余物经硅胶柱层析法分离纯化(乙酸乙酯:石油醚=1:3),得到化合物1-2。 1H NMR(400MHz,CDCl 3)δ7.42(s,1H),6.45(s,1H),4.75(d,J=8.4Hz,1H),4.10-4.40(m,2H),3.87(s,3H),3.86(s,3H),3.75-3.80(m,1H),1.90-2.10(m,4H),1.50-1.70(m,4H),1.41(s,9H)。LC-MS:m/z=458.1[M+Na] +
第二步
将化合物1-2(1.50g,3.44mmol)溶于甲醇(30mL)中,加入湿钯碳(10%,50.0mg),反应液在20℃,氢气氛围下搅拌0.5小时。过滤反应液,滤液减压浓缩,干燥得到化合物1-3。 1H NMR(400MHz,CDCl 3)δ7.09(s,1H),6.10(s,1H),5.36(brs,2H),4.15-4.40(m,2H),3.85(s,3H),3.82(s,3H),3.65-3.80(m,1H),1.95-2.15(m,4H),1.90-1.85(m,2H),1.47(s,9H),1.30-1.45(m,2H)。LC-MS:m/z=406.2[M+H] +
第三步
将化合物1-3(1.30g,3.21mmol)和醋酸铵(2.47g,32.06mmol)加入原甲酸三甲酯(31.29g,294.89mmol)中,反应液在70℃搅拌10小时。降温至20℃,反应液减压浓缩,剩余物加入水(10mL)打浆,过滤,固体干燥后得到化合物1-4。 1H NMR(400MHz,DMSO-d 6)δ7.83(s,1H),7.08(s,1H),7.00(s,1H),5.17(d,J=8.4Hz,1H),4.10-4.20(m,2H),3.95-4.00(m,1H),3.92(s,3H),1.90-2.00(m,4H),1.75-1.90(m,2H),1.50-1.60(m,2H),1.43(s,9H)。LC-MS:m/z=401.1[M+H] +
第四步
将化合物1-4(1.20g,3.00mmol)加入氯化氢的二氧六环溶液(4N,10mL),反应液在0℃搅拌0.4小时。反应液减压浓缩,得到化合物1-5的盐酸盐。LC-MS:m/z=301.0[M+H] +
第五步
在0℃,将1-5的盐酸盐(1.10g,3.27mmol),三乙胺(4.6mL,32.66mmol)溶入二氯甲烷(30mL)中,滴加三氟乙酸酐(1.37g,6.53mmol),反应液在20℃搅拌1小时。减压浓缩,剩余物加水(40mL)打浆,过滤,固体干燥得到化合物1-6。 1H NMR(400MHz,DMSO-d 6)δ11.90(s,1H),7.80-7.90(m,1H),7.13(m,1H),7.00(m,1H),5.28(d,J=9.2Hz,1H),4.60-4.70(m,1H),4.40-4.50(m,1H),4.00-4.10(m,1H),3.91(s,3H),1.90-2.20(m,6H),1.55-1.75(m,2H)。LC-MS:m/z=397.0[M+H] +
第六步
将化合物1-6(500.0mg,1.26mmol)溶于三氯氧磷(24.1mL,260.22mmol)中,加入N,N二甲基甲酰胺(92.2mg,1.26mmol),反应液在100℃搅拌5小时。减压浓缩,剩余物加入3,4-二氯-2-氟苯胺(340.6mg,1.89mmol)的异丙醇(5mL)溶液。反应液在90℃搅拌1小时。反应液冷却到20℃,加入饱和碳酸氢钠水溶液(10mL)淬灭,用乙酸乙酯(10mL x 2)萃取。合并有机相,用无水硫酸钠干燥。过滤,滤液减压浓缩,剩余物用硅胶柱层析法分离纯化(乙酸乙酯:石油醚=2:1),得到化合物1-7。 1H NMR(400MHz,CDCl 3)δ8.50-8.65(m,2H),7.40-7.50(m,1H),7.15-7.25(m,2H),6.59(s,1H),4.75-4.85(m,1H),4.55-4.65(m,2H),4.05-4.10(m,1H),3.98(s,3H),2.10-2.40(m,4H),1.90-2.00(m,2H),1.60-1.75(m,2H)。LC-MS:m/z=557.9[M+H] +
第七步
将化合物1-7(305.0mg,547.2μmol)溶于甲醇(15mL)中,加入碳酸钾(378.2mg,2.74mmol)。反应液在50℃搅拌14个小时。反应液减压浓缩,剩余物用硅胶制备板分离纯化(二氯甲烷:甲醇=10:1),得到化合 物1-8。 1H NMR(400MHz,DMSO-d 6)δ9.45(s,1H),8.28(s,1H),7.67(t,J=8.0Hz,1H),7.58(d,J=10.0Hz,1H),7.20(s,1H),7.14(s,1H),5.17(d,J=8.4Hz,1H),4.05-4.10(m,2H),3.98-4.05(m,1H),3.93(s,3H),2.15-2.30(m,4H),1.95-2.10(m,2H),1.70-1.80(m,2H)。LC-MS:m/z=462.0[M+H] +
第八步
在0℃下,将化合物1-8(220.0mg,475.8μmol)溶于四氢呋喃(6mL)和水(6mL)的混合溶剂,加入碳酸氢钠(119.9mg,1.43mmol),缓慢滴入丙烯酰氯(38.8mg,428.3μmol)。反应液在0℃继续搅拌0.5小时。加入MeOH(1mL)淬灭,减压浓缩,剩余物用制备薄层层析板分离纯化(二氯甲烷:甲醇=10:1),得到式(I)化合物。 1H NMR(400MHz,DMSO-d 6)δ9.35(s,1H),8.24(s,1H),7.66(t,J=8.0Hz,1H),7.57(d,J=8.8Hz,1H),7.14(s,1H),7.06(s,1H),6.72(dd,J 1=16.8Hz,J 2=10.4Hz,1H),6.18(dd,J 1=16.8Hz,J 2=1.6Hz,1H),5.68(dd,J 1=10.4Hz,J 2=1.6Hz,1H),5.35(d,J=9.2Hz,1H),4.50-4.60(m,2H),4.05-4.15(m,1H),3.92(s,3H),1.85-2.20(m,6H),1.60-1.70(m,1H),1.50-1.60(m,1H)。LC-MS:m/z=538.0[M+Na] +
实施例2:
式(I)化合物A晶型的制备
将式(I)所示化合物(50g)加入甲醇和乙酸乙酯(v:v=50mL:250mL)混合溶液中,得到的混悬液在80℃-90℃加热搅拌,慢慢冷却,过滤,固体减压干燥,得到式(I)化合物的A晶型。
式(II)化合物B晶型的制备
将式(I)化合物(40.9g)溶于乙醇中(320mL),混合物加热至70℃,搅拌下加入顺丁烯二酸(9.4g)的乙醇溶液(80mL)。反应液转为澄清后又析出固体,混合物在70℃搅拌0.5小时,加入乙酸乙酯(400mL),混合物在70℃继续搅拌16小时,冷却至室温,过滤,收集滤饼,减压干燥,得到式(II)化合物B晶型。
称取大约500mg式(I)化合物加入到40mL玻璃瓶中,加入25mL的四氢呋喃使其溶解,加入磁子后,将上述样品置于磁力加热搅拌器上(40℃,890rpm)进行反应。然后缓慢加入马来酸与溶剂混合物(马来酸:丙酮=200mg:1500μL),继续于磁力加热搅拌器上(40℃,890rpm)进行反应,将混悬状样品(40℃,890rpm条件下被磁力搅拌24小时后)于3000rpm下离心5分钟后,所得固体样品于30℃真空干燥箱中进行干燥,得到式(II)化合物B晶型。
称取适量式(I)化合物,大约50mg的马来酸盐加入到2.0mL玻璃小瓶中,加入适量溶剂(见下表8),使其成为混悬液;加入磁子后,将上述样品置于磁力加热搅拌器上,40℃条件下振荡2天后,离心,将所得的固体样品置于35℃真空干燥箱中干燥过夜,得到式(II)化合物B晶型。
表8式(II)化合物B晶型的制备
Figure PCTCN2020097158-appb-000020
式(III)化合物C晶型和D晶型的制备
称取适量式(I)化合物,大约20-30mg的盐酸盐加入到2.0mL玻璃小瓶中,加入适量溶剂(见下表9),使其成为混悬液;加入磁子后,将上述样品置于磁力加热搅拌器上,40℃条件下振荡2天后,离心,将所得的固体样品置于35℃真空干燥箱中干燥过夜,得到式(III)化合物C晶型或D晶型。
表9式(III)化合物C晶型和D晶型的制备
Figure PCTCN2020097158-appb-000021
式(IV)化合物E晶型的制备
称取25mg式(I)化合物,大约20-30mg的硫酸盐加入到2.0mL玻璃小瓶中,加入200μL甲醇,使其成为混悬液;加入磁子后,将上述样品置于磁力加热搅拌器上,40℃条件下振荡2天后,离心,将所得的固体样品置于35℃真空干燥箱中干燥过夜,得到式(IV)化合物E晶型。
式(IV)化合物F晶型的制备
称取50.284mg式(I)化合物,大约50mg的硫酸盐加入到2.0mL玻璃小瓶中,加入200μL乙醇,使其成为混悬液;加入磁子后,将上述样品置于磁力加热搅拌器上,40℃条件下振荡2天后,离心,将所得的固体样品置于35℃真空干燥箱中干燥过夜,得到式(IV)化合物F晶型。
实施例3:式(I)化合物B晶型的固体稳定性试验
根据影响因素和加速试验条件,准确称重式(I)化合物B晶型约5mg置于干燥洁净的玻璃瓶中,一式两份,摊成薄薄一层,作为正式供试样品,放置于影响因素试验条件下(60℃,92.5%RH)和加速条件下(40℃/75%RH和60℃/75%RH),其样品为完全暴露放样,用铝箔纸盖上,扎上小孔。另分别取少量样品放置在40mL玻璃样品瓶同样条件下待测定晶型状态。光照(1.2×10 6Lux·hr/近紫外200w·hr/m 2)条件下放置的样品为室温完全暴露放样。试验结果见表10。
表10式(I)化合物B晶型的固体稳定性试验结果
Figure PCTCN2020097158-appb-000022
结论:式(I)化合物B晶型具有良好的稳定性。
生物化学检测:体外评价
实验例1酶活性评价
本试验目的是检测化合物对HER1(ErbB1),HER2(ErbB2),HER4(ErbB4)的体外抑制活性。本试验采用的酶为人源ErbB1,ErbB2和ErbB4,Eurofins Pharma Discovery Service提供活性检测方法,测试化合物对HER1,HER2,HER4抑制活性结果如表11所示。
实验步骤和方法(96孔板):
加入5倍稀释的测试化合物缓冲液(5μL),多肽底物poly(Glu,Tyr)(4:1)(2.5μL),ErbB(4-20ng,2.5μL),MnCl 2(50mM,1.25μL),dH 2O(3.75μL),[γ- 33P]ATP(10μL),在30℃孵化10分钟。加入3%磷酸终止反应,取10μL标本转移至Filtermate A,用75mM磷酸清洗滤片3次,用甲醇清洗1次,滤片转移到密封塑料口袋,加入闪烁液混合物(4mL),在闪烁发光计数仪检测发出的光子强度,将酶样本的cpm(次数/分钟)与内控样品的cpm进行比较,光子强度的高低反映了酪氨酸激酶活性的强弱。
表11:式(I)化合物体外酶活性筛选试验结果
化合物 HER1 IC 50(nM) HER2 IC 50(nM) HER4 IC 50(nM)
式(I)化合物 8 5 3
结论:式(I)化合物对HER1,HER2和HER4抑制活性明显。
实验例2细胞增殖抑制活性评价:
实验目的:检测待测化合物对细胞增殖抑制活性。
实验原理:Cell-Titer-Glo试剂中的荧光素酶利用荧光素、氧和ATP作为反应底物,产生氧化荧光素,并以光的形式释放能量。由于荧光素酶反应需要ATP,因而反应产生的光的总量和反应细胞活力的ATP数总量成正比。
实验材料:
细胞系:NCI-N87细胞系(ATCC-CRL-5822),BT-474细胞系(ATCC-HTB-20),OE21(ECACC-96062201)细胞培养基:(RPMI 1640培养基(Invitrogen#22400-105;10%血清Invitrogen#10090148;左旋谷酰胺1×,Gibco#25030-081;双抗Hyclone#SV30010)
Cell
Figure PCTCN2020097158-appb-000023
发光法细胞活力检测试剂盒(Promega#G7573)
384孔细胞培养板(Greiner#781090)
化合物板(LABCYTE#LP-0200)
CO 2培养箱(Thermo#371)
Vi-cell细胞计数仪(Beckman Coulter)
移液器(Eppendorf)
移液管(Greiner)
移液枪(Eppendorf)
多功能酶标仪(Envision Reader)
ECHO Liquid-handling workstation(Labcyte-ECHO555)
实验步骤和方法:
2.1第1天:
按照细胞种板示意图在384或96孔板中分别按每孔1000个细胞,25μL每孔的密度种板,边缘孔不种 细胞补25μL PBS。
2.2第0天:
(1)化合物母液为10mM,用DMSO稀释化合物使其初始浓度为4mM。在化合物母液板上加入化合物,每孔9μL。
(2)用ECHO液体工作站做化合物稀释并向细胞板每孔加入125nL化合物,第2列和23列细胞孔每孔加125nL DMSO,第1列和24列PBS孔每孔加125nL DMSO。
(3)细胞板每孔补加25μL培养基,最终细胞板每孔为50μL,化合物浓度为1μM,3倍稀释,10个浓度,左右复孔,DMSO终浓度为0.25%。
2.3加好化合物后,1000rpm离心1min,将细胞板放置于37℃、5%CO 2培养箱中培养3天。
2.4第3天:
从培养箱中取出细胞板,在室温下平衡30分钟。向每孔加入25μL Cell-Titer-Glo试剂,振摇一分钟使它被充分混匀,1000rpm离心1分钟。10分钟后,在PerkinElmer Envision上读板,设置荧光读取时间0.2秒。
试验结果:试验结果如表12。
OE19细胞CTG实验
用胰酶消化饱和度已达到80%-90%的OE19细胞,离心重悬计数,调整细胞浓度,90ml/孔,加入96孔细胞培养板,使OE19细胞每孔个数为8000个。在含5%CO 2的细胞培养箱中37℃培养过夜。
用DMSO将待测化合物母液3倍梯度稀释,共10个浓度。在实验开始前,在无菌条件下将梯度稀释好的待测化合物用细胞培养液进一步稀释为10×化合物溶液(最高浓度100μM含1%DMSO)。将配制好的10×化合物溶液加入到细胞培养板中,每孔加入10μL。这样,模型标准对照Staurosporine(星形孢菌素)及所有受试化合物的终浓度均以10μM为起始浓度,梯度3倍稀释,共10个测试浓度点。
细胞培养72小时后,取出96孔细胞培养板,加入Cell Titer Glo试剂,50ml/孔,混匀离心,室温避光孵育10分钟。将细胞板放入Envision进行读数。
根据原始数据计算抑制率,抑制率计算公式为:
抑制百分比%=(ZPE-样本检测值)/(ZPE-HPE)×100%
以DAY3(以加化合物记为实验第一天,DAY3为读数当天)的含10mM staurosporine的孔作为HPE(100%抑制对照)孔,以DAY3的含0.1%的DMSO孔为ZPE(0%抑制对照)孔。受试化合物每个浓度点设两个复孔。
处理后的数据将用GraphPad Prism 6分析软件来做非线性回归分析,得到剂量效应曲线,并计算出待测化合物对OE19细胞的半抑制浓度(IC 50)。试验结果如表12.
表12:式(I)化合物体外细胞增殖抑制活性筛选试验结果
Figure PCTCN2020097158-appb-000024
结论:式(I)化合物对NCI-N87,OE21和OE19细胞增殖抑制活性明显。
实验例3人胃癌NCI-N87细胞皮下异种移植肿瘤BALB/c裸小鼠模型的体内药效学研究:
实验目的:研究本专利待测化合物对人胃癌NCI-N87细胞皮下异种移植瘤在BALB/c裸小鼠模型体内药效进行评估
实验动物:雌性BALB/c裸小鼠,6-8周龄,体重18-22克;供应商:上海灵畅生物科技有限公司提供实验方法与步骤:
3.1细胞培养
人胃癌NCI-N87细胞,体外单层培养,培养条件为RPMI-1640培养基中加10%胎牛血清,100U/mL青霉素,100U/mL0链霉素和2mM谷氨酰胺,37℃,5%CO 2培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞饱和度为80%-90%时,收取细胞,计数,接种。
3.2肿瘤细胞接种(肿瘤接种)
将0.2ml(10×106)NCI-N87细胞(PBS+Matrigel,1:1)皮下接种于每只小鼠的右后背,肿瘤平均体积达到158mm 3时开始分组给药
3.3受试物的配制:
受试化合物配制成0.04mg/mL和0.08mg/mL的澄清溶液,溶媒为10%NMP(N-甲基吡咯烷酮)+10%乙二醇硬脂酸酯+80%水
3.4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。每周两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%。
肿瘤增殖率T/C(%):计算公式如下:T/C(%)=某处理组给药结束时平均瘤体积/溶剂对照组治疗结束时平均瘤体积×100%。
3.5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)(具体数据见表13)。治疗组在试验结束时给药后第28天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。两组间比较用T-test进行分析,三组或多组间比较用one-way ANOVA进行分析,经检验,F值有显著 性差异,应用Games-Howell法进行检验。用SPSS 17.0进行所有数据分析。p<0.05认为有显著性差异。
3.6试验结果
3.6.1死亡率、发病率及体重变化情况
实验动物的体重作为间接测定药物毒性的参考指标。在此模型治疗组小鼠体重有下降趋势,无其他发病或死亡现象。
3.6.2抗肿瘤药效评价指标
表13式(I)化合物对人胃癌NCI-N87细胞皮下异种移植瘤模型的抑瘤药效评价
(基于给药后第28天肿瘤体积计算得出)
Figure PCTCN2020097158-appb-000025
注:
a.平均值±SEM。
b.肿瘤生长抑制由T/C和TGI(TGI(%)=[1-(T28-T0)/(V28-V0)]×100)计算。
c.p值根据肿瘤体积计算。
3.7试验结论和讨论
在本实验评价了式(I)化合物在人胃癌NCI-N87细胞皮下异种移植瘤模型中的体内药效。与溶剂对照组相比,治疗组式(I)化合物@0.8mg/kg与参照化合物Poziotinib@0.8mg/kg药效相当,都展现了显著的抑瘤作用;低剂量组式(I)化合物与参照化合物Poziotinib药效相当或者更优。
实验例4人胃癌NCI-N87细胞皮下异种移植肿瘤BALB/c裸小鼠模型的体内药效学研究:
实验目的:研究本专利待测化合物对人胃癌NCI-N87细胞皮下异种移植瘤在BALB/c裸小鼠模型体内药效进行评估
实验动物:雌性BALB/c裸小鼠,6-8周龄,体重18-22克;供应商:上海西普尔-必凯实验动物有限公司
实验方法与步骤:
4.1细胞培养
人胃癌NCI-N87细胞,体外单层培养,培养条件为RPMI-1640培养基中加10%胎牛血清,100U/mL青霉素,100μg/mL链霉素,37℃,5%CO 2培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞饱和度为80%-90%时,收取细胞,计数,接种。
4.2肿瘤细胞接种(肿瘤接种)
将0.2ml(10×10 6)NCI-N87细胞(PBS+Matrigel,1:1)皮下接种于每只小鼠的右后背,肿瘤平均体积达到156mm 3时开始分组给药
4.3受试物的配制:
受试化合物配制成0.05mg/mL的澄清溶液,溶媒为10%NMP(N-甲基吡咯烷酮)+10%乙二醇硬脂酸酯+80%水
4.4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。每周两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%。
肿瘤增殖率T/C(%):计算公式如下:T/C(%)=某处理组给药结束时平均瘤体积/溶剂对照组治疗结束时平均瘤体积×100%。
4.5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)(具体数据见表14)。治疗组在试验结束时给药后第28天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。两组间比较用T-test进行分析,三组或多组间比较用one-way ANOVA进行分析,经检验,F值有显著性差异,应用Games-Howell法进行检验。用SPSS 17.0进行所有数据分析。p<0.05认为有显著性差异。
4.6试验结果
4.6.1死亡率、发病率及体重变化情况
实验动物的体重作为间接测定药物毒性的参考指标。在此模型治疗组小鼠体重有下降趋势,无其他发病或死亡现象。
4.6.2抗肿瘤药效评价指标
表14式(I)化合物对人胃癌NCI-N87细胞皮下异种移植瘤模型的抑瘤药效评价(基于给药后第21天肿瘤体积计算得出)
Figure PCTCN2020097158-appb-000026
注:
a.平均值±SEM。
b.肿瘤生长抑制由T/C和TGI(TGI(%)=[1-(T21-T0)/(V21-V0)]×100)计算。
c.p值根据肿瘤体积计算。
4.7试验结论和讨论
在本实验评价了式(I)化合物和对照化合物2在人胃癌NCI-N87细胞皮下异种移植瘤模型中的体内药效。与溶剂对照组相比,治疗组式(I)化合物@0.5mg/kg展现了显著的抑瘤作用,抑瘤率为T/C=10.69%,TGI=119.41%,p=0.007;同剂量下对照化合物2药效不明显T/C=89.35%,TGI=18.83%;式(I)化合物药效显著优于对照化合物2。
实验例5人食管癌OE21细胞皮下异种移植肿瘤BALB/c裸小鼠模型的体内药效学研究:
实验目的:研究待测化合物对人食管癌OE21细胞皮下异种移植肿瘤BALB/c裸小鼠模型体内药效进行评估。
实验动物:雌性BALB/c裸小鼠,6-8周龄,体重17-23克;供应商:上海西普尔-必凯实验动物有限公司
实验方法与步骤:
5.1细胞培养
人食管癌OE21细胞体外单层培养,培养条件为RPMI-1640培养基中加10%胎牛血清,100U/mL青霉素,100U/mL0链霉素和2mM谷氨酰胺,37℃5%CO 2培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞饱和度为80%-90%时,收取细胞,计数,接种。
5.2肿瘤细胞接种(肿瘤接种)
将0.1ml(20.1)OE21细胞(PBS)皮下接种于每只小鼠的右后背,肿瘤平均体积达到112mm 3时开始分组给药。
5.3受试物的配制:
受试化合物配制成0.15mg/mL~0.3mg/mL的澄清或混悬液溶液,溶媒为10%NMP+10%乙二醇硬脂酸酯 +80%水
5.4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。每周两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(%)=某处理组给药结束时平均瘤体积/溶剂对照组治疗结束时平均瘤体积×100%。
5.5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)。三组或多组间比较用one-way ANOVA。如果F值有显著性差异,应在ANOVA分析之后再进行多重比较。用SPSS 17.0进行所有数据分析。p<0.05认为有显著性差异。
5.6试验结果
5.6.1死亡率、发病率及体重变化情况
在此模型中Poziotinib@2/1.5mg/kg和式(I)化合物@2/1.5mg/kg给药组的动物均显示有不同程度的体重下降和出现皮屑的现象;Poziotinib@2/1.5mg/kg在分组给药后第6天和第7天开始依次有1只和3只小鼠体重下降超过15%,作停药处理,且有整组小鼠尿液深黄色的毒性提示;给药第3天开始Poziotinib@2mg/kg降低剂量到1.5mg/kg,式(I)化合物@2mg/kg降低剂量到1.5mg/kg。剂量调整后,各组体重明显恢复。
5.6.2抗肿瘤药效评价指标
表15式(I)化合物对OE21异种移植瘤模型的抑瘤药效评价(基于给药后第21天肿瘤体积计算得出)
Figure PCTCN2020097158-appb-000027
Figure PCTCN2020097158-appb-000028
注:
a.平均值±SEM。
b.肿瘤生长抑制由T/C和TGI,计算,参考实验例4。
c.p值根据给药后第21天的肿瘤体积计算,采用One-Way ANOVA的统计方法,分别和Vehicle组比较得出。
5.7试验结论和讨论
在本实验评价了式(I)化合物和参照化合物Poziotinib对OE21异种移植瘤模型中的体内药效。开始给药21天后,溶剂对照组荷瘤鼠的瘤体积达到948mm 3,受试物Poziotinib@2/1.5mg/kg和式(I)化合物@2/1.5mg/kg组与溶剂对照组比较有显著的抑瘤作用,抑瘤率分别为Poziotinib@2/1.5mg/kg组:T/C=6.05%,TGI=106.65%,p=0.008;式(I)化合物@2/1.5mg/kg组:T/C=5.82%,TGI=106.65%,p=0.008。式(I)化合物低剂量0.5mpk药效与参照化合物Poziotnib和式(I)化合物高剂量1.5mpk药效相当。式(I)化合物抑瘤效果显著,药效与参考化合物Poziotinib相当,但是参考化合物高剂量组小鼠体重下降显著,3只小鼠体重下降超过15%,予以停药,并且全组有小鼠尿液深黄色毒性提示。式(I)化合物对小鼠体重影响小,无停药,无尿液深黄色毒性提示,耐受性显著优于参考化合物。式(I)化合物起效剂量更低,耐受性更好,安全窗口更优于参考化合物Poziotinib。
实验例6小鼠药代动力学研究试验
实验目的:本实验旨在考察本专利实施例与参照化合物单次静脉注射和灌胃给药后雌性BALB/c小鼠体内血浆药代动力学。
实验动物:雌性BALB/c裸小鼠,7-9周龄,体重17-23克;供应商:上海西普尔-必凯实验动物有限公司
样品采集:实验动物每个时间点从隐静脉穿刺采集血液样本0.03mL,记录实际采血时间。所有血样均加入规格为1.5mL的商品化EDTA-K2抗凝管中(供应商为江苏康健医疗用品有限公司)。血样采集后,在半小时内,于4℃、3000g离心10分钟吸取上清血浆,迅速至于干冰中,于-80℃冰箱保存,用于LC-MS/MS分析。
数据分析:采用WinNonlin TMVersion6.3(Pharsight,MountainView,CA)药动学软件的非房室模型处理血浆浓度,使用线性对数梯形法方法计算药动学参数C max,T max,T 1/2,AUC 0-last
表16式(I)化合物与参考化合物PK结果对比
Figure PCTCN2020097158-appb-000029
Figure PCTCN2020097158-appb-000030
C max:达峰浓度;T max:达峰时间;T 1/2:清除一半化合物所需时长;AUC 0-last:0-末次取样时间内的浓度积分面积。
结论:在小鼠药代动力学研究结果显示,式(I)化合物半衰期比参考化合物更长,口服血浆暴露量显著优于参照化合物Poziotinib和对照化合物2。
实验例7 CYP酶抑制活性评价
本发明化合物和参考化合物Poziotinib在人肝微粒体中CYP酶活性测试结果如下表所示(表17)
表17式(I)化合物与参考化合物CYP酶抑制结果对比
Figure PCTCN2020097158-appb-000031
本发明化合物在人肝微粒体CYP抑制活性结果显示,式(I)化合物和Poziotinib对CYP1A2和CYP3A4无抑制活性。式(I)化合物对CYP2C19抑制活性与参考化合物Poziotinib相当,对CYP2C9和CYP2D6的活性有改善,分别优于参考化合物2倍和6倍。综合比较,式(I)化合物对CYP活性有改善并优于参考化合物,因此药物-药物相互作用风险更低。

Claims (34)

  1. 式(I)所示化合物的A晶型,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:3.3±0.2°,17.4±0.2°,20.8±0.2°。
    Figure PCTCN2020097158-appb-100001
  2. 根据权利要求1所述的A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,14.6±0.2°,15.2±0.2°,17.4±0.2°,19.0±0.2°,20.8±0.2°,22.1±0.2°,24.1±0.2°,29.5±0.2°。
  3. 根据权利要求2所述的A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,12.0±0.2°,12.7±0.2°,14.6±0.2°,15.2±0.2°,15.9±0.2°,17.4±0.2°,19.0±0.2°,20.8±0.2°,22.1±0.2°,24.1±0.2°,29.5±0.2°。
  4. 根据权利要求3所述的A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3°,3.7°,6.9°,11.2°,12.0°,12.7°,14.6°,15.2°,15.9°,16.9°,17.4°,17.9°,18.5°,19.0°,19.4°,20.1°,20.4°,20.8°,22.1°,22.4°,23.5°,24.1°,24.4°,25.4°,25.8°,26.6°,27.1°,28.3°,29.0°,29.5°。
  5. 根据权利要求4所述的A晶型,其XRPD图谱如图1所示。
  6. 式(Ⅱ)所示化合物。
    Figure PCTCN2020097158-appb-100002
  7. 式(Ⅱ)所示化合物的B晶型,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:8.0±0.2°,11.4±0.2°,18.7±0.2°。
  8. 根据权利要求7所述的B晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:8.0±0.2°,11.4±0.2°,12.7±0.2°,15.0±0.2°,16.1±0.2°,18.7±0.2°,20.0±0.2°,20.7±0.2°。
  9. 根据权利要求8所述的B晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:8.0±0.2°,11.4±0.2°,12.7±0.2°,15.0±0.2°,16.1±0.2°,18.7±0.2°,20.0±0.2°,20.7±0.2°,22.4±0.2°,23.1±0.2°,24.8±0.2°。
  10. 根据权利要求9所述的B晶型,其XRPD图谱如图2所示。
  11. 根据权利要求7~10任意一项所述的B晶型,其差示扫描量热曲线在207.4±3.0℃有一个吸热峰的起始点。
  12. 根据权利要求11所述的B晶型,其DSC图谱如图3所示。
  13. 式(Ⅲ)所示化合物。
    Figure PCTCN2020097158-appb-100003
  14. 式(Ⅲ)所示化合物的C晶型,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:3.3±0.2°,7.6±0.2°,8.5±0.2°。
  15. 根据权利要求14所述的C晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,7.6±0.2°,8.5±0.2°,11.3±0.2°,13.8±0.2°,15.2±0.2°,17.7±0.2°,18.3±0.2°。
  16. 根据权利要求15所述的C晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,7.6±0.2°,8.5±0.2°,11.3±0.2°,13.8±0.2°,15.2±0.2°,17.7±0.2°,18.3±0.2°,22.6±0.2°,23.7±0.2°,24.9±0.2°,26.6±0.2°。
  17. 根据权利要求16所述的C晶型,其XRPD图谱如图4所示。
  18. 式(Ⅲ)所示化合物的D晶型,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:3.5±0.2°,9.5±0.2°,10.5±0.2°。
  19. 根据权利要求18所述的D晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.5±0.2°,9.5±0.2°,10.5±0.2°,14.0±0.2°,16.3±0.2°,18.7±0.2°,23.4±0.2°,25.6±0.2°。
  20. 根据权利要求19所述的D晶型,其XRPD图谱如图5所示。
  21. 式(Ⅳ)所示化合物。
    Figure PCTCN2020097158-appb-100004
  22. 式(Ⅳ)所示化合物的E晶型,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:3.3±0.2°,12.8±0.2°,13.2±0.2°。
  23. 根据权利要求22所述的E晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.3±0.2°,8.1±0.2°,13.2±0.2°,14.9±0.2°,16.2±0.2°,18.7±0.2°,23.2±0.2°,24.8±0.2°。
  24. 根据权利要求23所述的E晶型,其XRPD图谱如图6所示。
  25. 式(Ⅳ)所示化合物的F晶型,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:3.6±0.2°,7.5±0.2°,16.6±0.2°。
  26. 根据权利要求25所述的F晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:3.6±0.2°,7.5±0.2°,12.2±0.2°,15.1±0.2°,16.6±0.2°,23.6±0.2°,24.6±0.2°。
  27. 根据权利要求26所述的F晶型,其XRPD图谱如图7所示。
  28. 式(I)所示化合物A晶型的制备方法,包括:
    (1)将式(I)所示化合物加入溶剂中,使其溶解;
    (2)加热搅拌,冷却,过滤,干燥,得到式(I)化合物的A晶型。
  29. 式(II)所示化合物B晶型的制备方法,包括:
    (1)将式(I)所示化合物加入溶剂中,使其溶解;
    (2)加热,加入马来酸,搅拌,析出固体,冷却,过滤,干燥,得到式(II)化合物的B晶型。
  30. 根据权利要求28所述的制备方法,其中,所述溶剂选自四氢呋喃。
  31. 根据权利要求29所述的制备方法,其中,所述溶剂选自乙醇、乙酸乙酯。
  32. 根据权利要求28或29所述的制备方法,其中,化合物与溶剂的重量-体积比为1g:7~50mL。
  33. 根据权利要求6、13或21任意一项所述的化合物或根据权利要求1~5、7~12、14~20或22~27任意一项所述的晶型或根据权利要求28~32任意一项所述的制备方法得到的晶型在制备治疗Pan-HER酪氨酸激酶抑制剂相关病症的药物上的应用。
  34. 根据权利要求33所述的应用,其特征在于,所述Pan-HER酪氨酸激酶抑制剂相关药物是用于肿瘤的药物。
PCT/CN2020/097158 2019-06-19 2020-06-19 喹唑啉类化合物的晶型、盐型及其制备方法 WO2020253836A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CN202080044785.6A CN114144418B (zh) 2019-06-19 2020-06-19 喹唑啉类化合物的晶型、盐型及其制备方法
EP20827468.8A EP3988553A4 (en) 2019-06-19 2020-06-19 CRYSTALLINE AND SALT FORM OF A QUINAZOLINE COMPOUND, AND METHOD FOR THE PREPARATION THEREOF
US17/620,464 US20220242851A1 (en) 2019-06-19 2020-06-19 Crystal form and salt of quinazoline compound and preparation method thereof
JP2021575887A JP7350374B2 (ja) 2019-06-19 2020-06-19 キナゾリン系化合物の結晶体、塩およびその調製方法

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910533240.0 2019-06-19
CN201910533240 2019-06-19

Publications (1)

Publication Number Publication Date
WO2020253836A1 true WO2020253836A1 (zh) 2020-12-24

Family

ID=74040641

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/097158 WO2020253836A1 (zh) 2019-06-19 2020-06-19 喹唑啉类化合物的晶型、盐型及其制备方法

Country Status (5)

Country Link
US (1) US20220242851A1 (zh)
EP (1) EP3988553A4 (zh)
JP (1) JP7350374B2 (zh)
CN (1) CN114144418B (zh)
WO (1) WO2020253836A1 (zh)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117567466B (zh) * 2024-01-16 2024-04-16 成都金瑞基业生物科技有限公司 一种喹唑啉衍生物的制备方法

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008150118A2 (en) 2007-06-05 2008-12-11 Hanmi Pharm. Co., Ltd. Novel amide derivative for inhibiting the growth of cancer cells
WO2015043515A1 (zh) 2013-09-28 2015-04-02 正大天晴药业集团股份有限公司 喹唑啉衍生物及其制备方法
CN105294717A (zh) * 2015-11-18 2016-02-03 乳源瑶族自治县大众药品贸易有限公司 一种egfr抑制剂的盐、晶型及其用途
CN105461729A (zh) * 2015-11-18 2016-04-06 乳源瑶族自治县大众药品贸易有限公司 一种egfr抑制剂的盐、晶型及其用途
CN106588943A (zh) * 2015-10-19 2017-04-26 广东东阳光药业有限公司 一种egfr抑制剂的盐、晶型及其用途
WO2019120213A1 (zh) * 2017-12-19 2019-06-27 南京明德新药研发股份有限公司 喹唑啉衍生物及其应用

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6704422B2 (ja) * 2015-03-20 2020-06-03 チア タイ ティエンチン ファーマシューティカル グループ カンパニー リミテッドChia Tai Tianqing Pharmaceutical Group Co.,Ltd. キナゾリン誘導体の塩およびその製造方法

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008150118A2 (en) 2007-06-05 2008-12-11 Hanmi Pharm. Co., Ltd. Novel amide derivative for inhibiting the growth of cancer cells
CN101679384A (zh) * 2007-06-05 2010-03-24 韩美药品株式会社 用于抑制癌细胞生长的新酰胺衍生物
WO2015043515A1 (zh) 2013-09-28 2015-04-02 正大天晴药业集团股份有限公司 喹唑啉衍生物及其制备方法
CN105555782A (zh) * 2013-09-28 2016-05-04 正大天晴药业集团股份有限公司 喹唑啉衍生物及其制备方法
CN106588943A (zh) * 2015-10-19 2017-04-26 广东东阳光药业有限公司 一种egfr抑制剂的盐、晶型及其用途
CN105294717A (zh) * 2015-11-18 2016-02-03 乳源瑶族自治县大众药品贸易有限公司 一种egfr抑制剂的盐、晶型及其用途
CN105461729A (zh) * 2015-11-18 2016-04-06 乳源瑶族自治县大众药品贸易有限公司 一种egfr抑制剂的盐、晶型及其用途
WO2019120213A1 (zh) * 2017-12-19 2019-06-27 南京明德新药研发股份有限公司 喹唑啉衍生物及其应用

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BASELGA. J., ONCOLOGIST, vol. 7, 2002, pages 2 - 8
ROSKOSKI ROBERT JR.: "ErbB/HER Protein-tyrosine kinases: Structures and Small Molecule Inhibitors", PHARMACOLOGICAL RESEARCH, vol. 87, 30 September 2014 (2014-09-30), XP055328149, DOI: 20200805161227A *

Also Published As

Publication number Publication date
EP3988553A1 (en) 2022-04-27
EP3988553A4 (en) 2023-06-14
JP2022537758A (ja) 2022-08-29
JP7350374B2 (ja) 2023-09-26
US20220242851A1 (en) 2022-08-04
CN114144418A (zh) 2022-03-04
CN114144418B (zh) 2023-10-10

Similar Documents

Publication Publication Date Title
CN106928150B (zh) 丙烯酰胺苯胺衍生物及其药学上的应用
EP2612860B1 (en) Quinazoline derivatives substituted by aniline, preparation method and use thereof
EP2716633B1 (en) Quinazoline derivative as tyrosine-kinase inhibitor, preparation method therefor and application thereof
KR20170031778A (ko) 피리딘 아미도피리미딘 유도체, 그의 제조 방법 및 용도
KR20140145157A (ko) 상피 성장 인자 수용체 키나제 억제제의 고체 형태
US20200361908A1 (en) Crystals of aniline pyrimidine compound serving as egfr inhibitor
Fan et al. Discovery of Dioxino [2, 3-f] quinazoline derivative VEGFR-2 inhibitors exerting significant antipro-liferative activity in HUVECs and mice
WO2022266015A1 (en) Fused heteroaryl compounds useful as anticancer agents
EP3102571A1 (en) Substituted pyrimidines useful as egfr-t790m kinase inhibitors
BR112021005750A2 (pt) inibidor de fgfr4 e uso do mesmo
WO2020253836A1 (zh) 喹唑啉类化合物的晶型、盐型及其制备方法
CN110857292A (zh) 一种egfr激酶抑制剂及其制备方法和应用
CN112351971B (zh) 喹啉或喹唑啉类化合物及其应用
WO2020007219A1 (zh) 一种egfr抑制剂的晶型及其制备方法
WO2022257965A1 (zh) 固体形式的周期蛋白依赖性激酶9抑制剂及其用途
JP2019513730A (ja) 癌を治療するためのATM(毛細血管拡張性運動失調症変異)キナーゼモジュレータとしてのN,N−ジメチル−3−[[5−(3−メチル−2−オキソ−1−テトラヒドロピラン−4−イル−イミダゾ[4,5−c]キノリン−8−イル)−2−ピリジル]オキシ]プロパン−1−アミンオキシド
WO2021088987A1 (zh) 作为选择性her2抑制剂的盐型、晶型及其应用
CN111362924B (zh) 氘代的嘧啶衍生物及其用途
KR20220008313A (ko) 퀴나졸리논계 화합물의 결정형 및 이의 제조방법
CN113087671A (zh) 一种氰基取代吡啶及氰基取代嘧啶类化合物、制备方法及其应用
WO2020224585A1 (zh) 一种mTORC1/2双激酶活性抑制剂的盐型、晶型及其制备方法
CN108299419B (zh) 一种新型egfr激酶抑制剂的几种新晶型及其制备方法
CN116003406B (zh) 杂芳环氮氧化合物及其制备方法和应用
WO2022242688A1 (zh) 一种含氰基取代的大环类化合物的晶型及其制备方法
CN108047132A (zh) 二苯胺基吡啶类抗肿瘤化合物及其制备方法与用途

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: 20827468

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021575887

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020827468

Country of ref document: EP

Effective date: 20220119