WO2020140934A1 - Salt of selective inhibitor of egfr tyrosine kinase and crystal form thereof - Google Patents

Salt of selective inhibitor of egfr tyrosine kinase and crystal form thereof Download PDF

Info

Publication number
WO2020140934A1
WO2020140934A1 PCT/CN2020/070062 CN2020070062W WO2020140934A1 WO 2020140934 A1 WO2020140934 A1 WO 2020140934A1 CN 2020070062 W CN2020070062 W CN 2020070062W WO 2020140934 A1 WO2020140934 A1 WO 2020140934A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound
water
tartrate
egfr
Prior art date
Application number
PCT/CN2020/070062
Other languages
French (fr)
Chinese (zh)
Inventor
林建东
王小建
Original Assignee
山东轩竹医药科技有限公司
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 山东轩竹医药科技有限公司 filed Critical 山东轩竹医药科技有限公司
Priority to CN202080007085.XA priority Critical patent/CN113227073B/en
Publication of WO2020140934A1 publication Critical patent/WO2020140934A1/en

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
    • C07D403/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond

Definitions

  • the invention relates to a selective inhibitor salt of EGFR tyrosine kinase and its mutant, its polymorphic form, preparation method, pharmaceutical composition containing the inhibitor salt and its crystalline form, and its preparation Use in medicine for treating and/or preventing abnormal cell proliferation diseases.
  • Protein kinase is a very important signaling entity in intracellular communication, where it modifies many proteins by catalyzing the transport of phosphate groups from ATP, which acts as a phosphorus donor, to the phenolic hydroxyl group on the protein tyrosine side chain.
  • ATP phosphate groups
  • tyrosine kinase is incorporated into the intracellular domain of a very large transmembrane protein with a homologous ligand binding domain in the extracellular domain, whereby ligand binding activates tyrosine kinase within the cell.
  • RTK receptor tyrosine kinases
  • EGFR epidermal growth factor RTK
  • NSCLC non-small cell lung cancer
  • EGFR is a member of the erbB (type I) subfamily of RTK, along with erbB-2, erbB-3, and erbB-4.
  • EGFR-EGFR homodimers are commonly used for signaling, the more common process in this family is that ligands induce heterodimerization, making signaling entities It will be, for example, EGFR:erbB-2 or erb-B2:erbB-3 and appropriate ligands.
  • the easiest way to reactivate the system is to increase the expression of one of the other erbBs, and even before treatment, this is often seen, and can help explain why many tumors that overexpress wt EGFR do not respond to EGFR inhibition.
  • HGFR RTK HGFR
  • erbB-3 tumorigenic heterodimers with erbB family members
  • HGFR inhibitors A common mechanism of resistance to EGFR inhibitors.
  • dm-EGFR double mutant receptor
  • NSCLCs with double mutants such as L858R/T790M are common among initial responders who subsequently develop resistance to EGFR inhibitors. Although it is not known whether such subclones always exist or only appear after treatment, it seems most likely that mutations already exist in short-term responders and may appear as remutations in long-term responders who develop resistance later.
  • 2-((2-(dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide can selectively modulate protein kinases that are resistant to current EGFR-based inhibitor therapy , Especially type I receptor tyrosine kinase (RTK) family or erbB family, and more particularly active novel compounds of certain mutant forms of EGFR receptor, thereby effectively inhibiting cell proliferation and cell invasion, inhibiting metastasis, and inducing cells Apoptosis or inhibit angiogenesis.
  • RTK type I receptor tyrosine kinase
  • crystal form plays an important role in the process of drug development. Different forms of compounds have different bioavailability, solubility, stability and other physical and chemical properties. In order to meet the requirements of preparations, production, transportation, etc., we I) The crystal form of the compound was studied in order to find a crystal form with good properties.
  • the present invention provides tartrates of compounds of formula (I), specifically L-(+)-tartrates of compounds of formula (I).
  • the present invention also provides an EGFR inhibitor N-(5-((4-(7-cyano-1,3-dimethyl-1H-indol-5-yl)pyrimidine-2 represented by formula (I) -Yl)amino)-2-((2-(dimethylamino)ethyl)methylamino)-4-methoxyphenyl)acrylamide L-(+)-tartrate crystal form, including crystal Form I and Form II.
  • the present invention also provides L-(+)-tartrate crystal form I (hereinafter referred to as crystal form I) of the compound of formula (I) and L-(+)-tartrate crystal form II (hereinafter referred to as crystal form I) of the compound of formula (I)
  • the preparation method of crystal form II) for short includes the pharmaceutical composition of the crystal form, and the application of the crystal form in the preparation of a medicine for preventing and/or treating abnormal cell proliferation diseases.
  • the present invention provides tartrate of the compound represented by formula (I),
  • the molar ratio of the compound of formula (I) to tartaric acid in the tartrate salt of the compound of formula (I) is 1:3-1:1, preferably 1:1.
  • the tartaric acid in the tartaric acid salt of the compound of formula (I) is L-(+)-tartaric acid.
  • the present invention also provides L-(+)-tartrate crystal form I of the compound represented by formula (I),
  • the molar ratio of the compound of formula (I) to L-(+)-tartaric acid is 1:1, and Cu-K ⁇ radiation is used to express the X-ray powder diffraction pattern at an angle of 2 ⁇ , at 8.9 ⁇ 0.2°, 11.2 ⁇ There are characteristic peaks at 0.2°, 12.0 ⁇ 0.2°, 12.5 ⁇ 0.2°, 13.1 ⁇ 0.2°, 14.7 ⁇ 0.2°, 17.5 ⁇ 0.2°, 19.1 ⁇ 0.2°.
  • the crystalline form I in the X-ray powder diffraction pattern expressed at a 2 ⁇ angle using Cu-K ⁇ radiation, is also 16.3 ⁇ 0.2°, 16.6 ⁇ 0.2 There are characteristic peaks at °, 20.0 ⁇ 0.2°, 20.6 ⁇ 0.2°, 21.1 ⁇ 0.2°, 21.6 ⁇ 0.2°.
  • the crystalline form I has substantially the same X-ray powder diffraction pattern as in FIG. 1.
  • the Form I differential scanning calorimetry has an endothermic peak in the range of 190°C to 230°C. In some embodiments, the DSC pattern of Form I has an endothermic peak in the range of 200°C to 225°C. In some embodiments, the maximum endothermic transition temperature of the DSC pattern of Form I is 215.5 ⁇ 5°C. In some embodiments, the crystalline form I has a differential scanning calorimetry curve substantially as shown in FIG. 2.
  • the TGA pattern of Form I shows no significant weight loss at 0°C to 200°C.
  • the crystalline form I has a TGA pattern substantially as shown in FIG. 3.
  • the present invention also provides a method for preparing L-(+)-tartrate crystal form I of the compound of formula (I), the steps of which include: combining the compound of formula (I) with 1 equivalent of L-( +)-Tartaric acid is added to an organic solvent, water or N-methylpyrrolidone is added under stirring conditions, filtered after the reaction is completed, and dried to obtain the L-(+)-tartrate crystal form I of the compound of formula (I); the formula ( I) Compound N-(5-((4-(7-cyano-1,3-dimethyl-1H-indol-5-yl)pyrimidin-2-yl)amino)-2-((2- (Dimethylamino)ethyl)methylamino)-4-methoxyphenyl)acrylamide can be obtained by the preparation method in the embodiment of the present invention.
  • the organic solvent is selected from one or more combinations of one or more solvents selected from lower nitriles, lower alcohols containing more than 2 carbon atoms, and oxygen-containing heterocyclics .
  • the organic solvent is selected from one or both of acetonitrile, ethanol, propanol, isopropanol, butanol, sec-butanol, tert-butanol and 1,4-dioxane Any combination of the above solvents.
  • the ratio of organic solvent to water or the ratio of organic solvent to N-methylpyrrolidone in the above preparation method is 1:1-20:1, preferably 1:1-10:1, further preferably 2:1-10:1.
  • the present invention also provides another method for preparing L-(+)-tartrate crystal form I of the compound of formula (I).
  • the steps include: adding the compound of formula (I) to a single solvent or a mixed solvent and heating to a certain level After the temperature, add a certain amount of L-(+)-tartaric acid, maintain the temperature to continue the reaction until the end of the reaction, lower the temperature and crystallize, separate, and dry to obtain L-(+)-tartrate crystal form I of the compound of formula (I).
  • the single solvent described in the above preparation method is selected from lower alcohols, lower ketones, lower esters, oxygenated heterocyclics, lower nitriles, lower alkanes or lower halogenated alkanes;
  • the mixed solvent is selected from a mixed solvent of alcohols and water, a mixed solvent of nitriles and water, a mixed solvent of ketones and water, or a mixed solvent of oxygen-containing heterocyclics and water.
  • the single solvent in the above preparation method is selected from methanol, ethanol, propanol, isopropanol, n-butanol, isobutanol, t-butanol, acetone, butanone, methyl ethyl Ketone, acetonitrile, ethyl formate, ethyl acetate, methyl acetate, tetrahydrofuran, 1,4-dioxane, n-pentane, isopentane, n-hexane, isohexane, cyclohexane, cycloheptane, toluene Or dichloromethane; the mixed solvent is selected from ethanol/water, propanol/water, isopropanol/water, butanol/water, sec-butanol/water, tert-butanol/water, acetonitrile/water, 1,4 -Dioxan
  • the ratio of organic solvent to water in the mixed solvent, is selected from 1:1-20:1, further preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 1: 1-10:1, more preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 2:1-10:1.
  • the ratio of organic solvent to water is selected from 1:1, 2:1, 3:1, 3:2, 4:1, 5:1, 5:2, 5:3, 6:1, 8:1, 8:3, 8:5, 10:1 or 10:3;
  • the certain temperature described in the above preparation method is selected from 40°C-80°C, preferably 50°C-70°C.
  • certain equivalents described in the above preparation method are selected from 0.9-1.2 equivalents, preferably 1.0-1.1 equivalents, more preferably 1.0 equivalents.
  • the present invention also provides a third method for preparing L-(+)-tartrate crystal form I of the compound of formula (I).
  • the steps include: adding the compound of formula (I) to a single solvent or a mixed solvent, and slurry washing at room temperature Next, add a certain equivalent of L-(+)-tartaric acid, maintain the temperature and continue the reaction until the end of the reaction. Place the crystal at room temperature, separate, and dry to obtain L-(+)-tartrate crystal form I of the compound of formula (I).
  • the single solvent described in the above preparation method is selected from lower alcohols, lower ketones, lower esters, oxygenated heterocyclics, lower nitriles, lower alkanes or lower halogenated alkanes;
  • the mixed solvent is selected from a mixed solvent of alcohols and water, a mixed solvent of nitriles and water, a mixed solvent of ketones and water, or a mixed solvent of oxygen-containing heterocyclics and water.
  • the single solvent in the above preparation method is selected from methanol, ethanol, propanol, isopropanol, n-butanol, isobutanol, t-butanol, acetone, butanone, methyl ethyl Ketone, acetonitrile, ethyl formate, ethyl acetate, methyl acetate, tetrahydrofuran, 1,4-dioxane, n-pentane, isopentane, n-hexane, isohexane, cyclohexane, cycloheptane, toluene Or dichloromethane; the mixed solvent is selected from ethanol/water, propanol/water, isopropanol/water, butanol/water, sec-butanol/water, tert-butanol/water, acetonitrile/water, 1,4 -Dioxan
  • the ratio of organic solvent to water in the mixed solvent, is selected from 1:1-20:1, further preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 1: 1-10:1, more preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 2:1-10:1.
  • the ratio of organic solvent to water is selected from 1:1, 2:1, 3:1, 3:2, 4:1, 5:1, 5:2, 5:3, 6:1, 8:1, 8:3, 8:5, 10:1 or 10:3;
  • certain equivalents described in the above preparation method are selected from 0.9-1.2 equivalents, preferably 1.0-1.1 equivalents, more preferably 1.0 equivalents.
  • the present invention also provides L-(+)-tartrate crystal form II of the compound represented by formula (I),
  • the molar ratio of the compound of formula (I) to tartaric acid is 1:1, and the Cu-K ⁇ radiation is used to express the X-ray powder diffraction pattern at an angle of 2 ⁇ , at 9.0 ⁇ 0.2°, 11.3 ⁇ 0.2°, 12.0 ⁇ 0.2 There are characteristic peaks at °, 12.7 ⁇ 0.2°, 13.9 ⁇ 0.2°, 17.9 ⁇ 0.2°, 19.1 ⁇ 0.2°, 20.3 ⁇ 0.2°, 21.2 ⁇ 0.2°.
  • the crystalline form II in the X-ray powder diffraction pattern expressed at a 2 ⁇ angle using Cu-K ⁇ radiation, is also 16.3 ⁇ 0.2°, 17.3 ⁇ 0.2 °, 23.5 ⁇ 0.2°, 24.4 ⁇ 0.2°, 25.5 ⁇ 0.2°, 25.9 ⁇ 0.2° have characteristic peaks; preferably, the X-ray powder diffraction pattern of the crystalline form II is basically shown in FIG. 4.
  • the invention also provides a method for preparing crystalline form II.
  • the steps include: adding the compound of formula (I) and 1 equivalent of L-(+)-tartaric acid to an organic solvent, adding water under normal temperature stirring conditions, and continuing to stir at normal temperature At the end of the reaction, filter and dry to obtain Form II.
  • the organic solvent described in the above preparation method is selected from methanol.
  • the present invention also provides a pharmaceutical composition of L-(+)-tartaric acid crystal form I or crystal form II of the compound of formula (I) and one or more pharmaceutically acceptable carriers and/or diluents
  • the pharmaceutical composition may be It is prepared into any pharmaceutically acceptable dosage form and administered to patients in need thereof by oral, parenteral, rectal, or pulmonary administration.
  • oral administration it can be made into conventional solid preparations, such as tablets, capsules, pills, granules, etc.; it can also be made into oral liquid preparations, such as oral solutions, oral suspensions, syrups, etc.
  • a suitable filler, binder, disintegrant, lubricant, etc. may be added.
  • injections When used for parenteral administration, it can be made into injections, including injections, sterile powders for injection and concentrated solutions for injection. When it is prepared as an injection, it can be produced by a conventional method in the existing pharmaceutical field. When preparing an injection, an additional agent may not be added, or an appropriate additional agent may be added according to the nature of the drug. When used for rectal administration, it can be made into suppositories. When used for pulmonary administration, it can be made into inhalants or sprays.
  • the present invention also provides the use of L-(+)-tartrate crystal form I or crystal form II of the compound of formula (I) in the preparation of a medicament for the treatment and/or prevention of EGFR-mediated hyperproliferative diseases.
  • the present invention also provides a method for treating and/or preventing hyperproliferative diseases mediated by EGFR, which comprises administering to a subject in need thereof an effective amount of the compound of formula (I) L-( +)-Step of tartrate form I or form II.
  • the hyperproliferative disease described above is selected from cancer.
  • the EGFR described above includes wild-type EGFR and EGFR mutants.
  • the above-mentioned EGFR mutants include one or any combination of one or more of EGFR 19 exon mutation, EGFR 20 exon mutation and EGFR 21 exon mutation .
  • the aforementioned exon 20 mutation of EGFR is selected from NPG, ASV or T790M.
  • the cancer described above is selected from lung cancer, non-small cell lung cancer, colorectal cancer, pancreatic cancer, head and neck cancer, breast cancer, ovarian cancer, uterine cancer, liver cancer, gastric cancer, prostate cancer, glioblastoma Tumor and epithelial cell carcinoma.
  • the cancer described above is caused by mutations in exon 20 of EGFR.
  • the above-mentioned cancer is caused by the T790M mutation in EGFR 20 exon mutation, and there is also an EGFR 19 exon insertion mutation or EGFR 21 exon point mutation.
  • the present disclosure also provides the use of the L-(+)-tartrate crystalline form I or crystalline form II of the compound of formula (I) in the preparation of reagents that can be used to inhibit the level of EGFR enzymes in cells.
  • the agent is used in an in vivo or in vitro method.
  • the present disclosure also provides a method of inhibiting the level of EGFR in a cell, which includes the step of administering to the cell an effective amount of L-(+)-tartrate salt form I or form II of the compound of formula (I).
  • the method is performed in vivo or in vitro.
  • lower alcohols refers to straight-chain or branched-chain fatty alcohols containing 1 to 6 carbon atoms, or cyclic fatty alcohols, examples of which include but are not limited to: ethanol, propanol, isopropanol, Butanol, sec-butanol, tert-butanol and cyclopentanol.
  • lower nitrile contains 1-6 carbon atoms, and examples thereof include, but are not limited to, acetonitrile, propionitrile, and adiponitrile.
  • lower ketones contains fatty ketone compounds with 1-6 carbon atoms, and examples thereof include, but are not limited to: methyl ethyl ketone, methyl isopropyl ketone, acetone, methyl butanone, and methyl isobutyl ketone.
  • lower esters contains fatty ester compounds of 1-6 carbon atoms, examples of which include but are not limited to: methyl formate, ethyl formate, propyl formate, methyl acetate, ethyl acetate, Propyl acetate, isopropyl acetate, methyl propionate, ethyl propionate, propyl propionate and isopropyl propionate.
  • lower alkanes contains straight-chain or branched-chain aliphatic alkanes with 1-7 carbon atoms, or cyclic aliphatic alkanes, or aromatic alkanes, examples of which include but are not limited to: n-pentane, isopentane , N-hexane, isohexane, cyclopentane, cyclohexane and toluene.
  • lower halogenated alkane contains a linear or branched aliphatic alkane or cyclic aliphatic alkane containing 1 to 6 carbon atoms and substituted with at least one halogen atom.
  • Examples include, but are not limited to: methylene chloride, chloroform, carbon tetrachloride, 1,1-dichloroethane, 1,2-dichloroethane, and 1,1,1-trichloroethane.
  • the term "separation" is used to separate solids by conventional methods, such as filtration, centrifugation and discarding the supernatant.
  • filtration includes but is not limited to atmospheric pressure filtration, reduced pressure filtration and the like.
  • room temperature and "normal temperature” refer to indoor ambient temperature, usually 10-30°C, such as 20-25°C.
  • drying includes but is not limited to natural drying at room temperature, infrared lamp drying, oven drying, dryer drying, and drying under vacuum conditions.
  • the drying temperature is room temperature or 30-60°C, preferably 40-50°C.
  • cooling crystallization refers to cooling the reaction system to 0-35°C, preferably to 10-30°C, and more preferably 15-°C by cooling in an ice-water bath, cooling by autothermal cooling, or cooling by refrigeration equipment. At 25°C, it is more preferable to lower the temperature until most crystals precipitate.
  • Compounds can exist in two or more crystalline states, molecules with the same structure, crystallized into different solid forms, called polymorphs or polymorphs. When referring to a specific crystalline form, it is often called “crystal”, which is the term “crystal form” used herein.
  • the position of the absorption peak in the X-ray powder diffraction pattern of each crystal form may be in the range of ⁇ 0.2° of the specific value of the above invention, for example, in the range of ⁇ 0.1°, the absorption measured by differential scanning calorimetry
  • the thermal transition temperature may be within the range of the above specific numerical value ⁇ 5.0°C (for example, ⁇ 3.0°C or ⁇ 2.0°C).
  • the present disclosure also uses thermal weight loss analysis (TGA) to analyze the relationship between the degree of crystal form decomposition or sublimation and evaporation (loss of weight) and temperature. It should be understood that the same crystal form is affected by sample purity, particle size, different types of equipment, different test methods, etc., and there are certain errors in the obtained values.
  • TGA thermal weight loss analysis
  • the temperature at which the crystal form decomposes, sublimates, or evaporates may be within the range of the specific values disclosed above ⁇ 3.0°C, for example, ⁇ 2.0°C.
  • the term "subject" refers to an animal or a human, preferably a mammal or a human.
  • the term "effective amount” refers to an amount sufficient to achieve a desired therapeutic or preventive effect, for example, an amount that achieves relief of symptoms related to the disease to be treated.
  • the term "treatment” aims to reduce or eliminate the targeted disease state or disorder. If a subject receives a therapeutic amount of the crystalline form or its pharmaceutical composition according to the methods described herein, the subject's one or more indications and symptoms exhibit observable and/or detectable Decrease or improve, the subject is successfully "treated”. It should also be understood that the treatment of the disease state or disorder includes not only complete treatment but also incomplete treatment, but has achieved some biological or medical related results.
  • the compound of formula (I) of the present invention, its tartrate and its L-(+)-tartrate crystal form have excellent EGFR inhibitory activity, especially with a higher selection of certain mutant forms of EGFR receptor Sexuality and inhibitory activity can effectively inhibit cell proliferation and cell invasion, inhibit metastasis, induce apoptosis or inhibit angiogenesis;
  • the L-(+)-tartrate salt crystal form of the compound of formula (I) of the present invention has good stability, and has good properties, fluidity and compressibility, which is convenient for detection, preparation, transportation and storage;
  • the L-(+)-tartrate salt crystal form of the compound of formula (I) of the present invention has high purity, less residual solvent, higher solubility, good dissolution, good stability and easy quality control.
  • FIG. 1 is an X-ray powder diffraction pattern of L-(+)-tartrate salt crystal form I of the compound of formula (I).
  • the ordinate represents the diffraction intensity, and the abscissa represents the diffraction angle (2 ⁇ );
  • Fig. 2 is a differential scanning calorimetry (DSC) thermal analysis diagram of L-(+)-tartrate salt crystal form I of the compound of formula (I), the ordinate represents heat flow (W/g), and the abscissa represents temperature (°C) ;
  • Fig. 3 is a thermogravimetric analysis (TGA) curve of L-(+)-tartrate crystal form I of the compound of formula (I).
  • the ordinate represents the mass percentage (%), and the abscissa represents the temperature (°C);
  • Fig. 5 is an X-ray powder diffraction pattern of L-(+)-tartrate crystalline form II of the compound of formula (I).
  • the ordinate indicates the diffraction intensity, and the abscissa indicates the diffraction angle (2 ⁇ ).
  • the crystal structure of the present invention is not limited to providing a crystal structure of an X-ray powder diffraction pattern exactly the same as the X-ray powder diffraction pattern drawn in the drawings disclosed in the present application, and X-ray powder diffraction patterns substantially the same as those disclosed in the drawings Any crystal structure of the ray powder diffraction pattern is included in the scope of the present invention.
  • Conditions for X-ray powder diffraction measurement Cu palladium, K ⁇ 1 1.54060, step size 0.0203, 0.3 seconds per step.
  • the X-ray powder diffracted in a crystal form expressed at a 2 ⁇ angle (°) has a characteristic peak.
  • the X-ray powder diffraction pattern of the L-(+)-tartrate salt form I of the compound of formula (I) is shown in FIG. 1, and this form I has a peak at the following diffraction angle 2 ⁇ (°):
  • Measurement conditions Purge with nitrogen at 50 ml/min, collect data at a heating rate of 10°C/min between room temperature and 225°C, and plot with the endothermic peak facing down.
  • the actual measured start temperature and maximum temperature have a certain degree of variability based on the measurement parameters and heating rate.
  • Test conditions Purge with nitrogen at 60 ml/min and collect data at a heating rate of 10°C/min between room temperature and 300°C.
  • Conditions for X-ray powder diffraction measurement Cu palladium, K ⁇ 1 1.54060, step size 0.0203, 0.3 seconds per step.
  • the X-ray powder diffracted in a crystal form expressed at a 2 ⁇ angle (°) has a characteristic peak.
  • the X-ray powder diffraction pattern of the L-(+)-tartrate salt form II of the compound of formula (I) is shown in FIG. 5, which has a peak at the following diffraction angle 2 ⁇ (°):
  • kits and services are used to measure the inhibition of various kinases including (but not limited to): ALK, ABL, AXL, Aur B and C, BLK, erbB-2, erbB-4.EGFR, mutant EGFR, HPK, IRAK1, RON, ROS1, SLK, STK10, TIE2, TRK, c-Met, Lck, Lyn, Src, Fyn, Syk, Zap-70, Itk, Tec, Btk, EGFR, ErbB2, Kdr, Flt-1, Flt- 3.
  • kits and services are used to measure the inhibition of various kinases including (but not limited to): ALK, ABL, AXL, Aur B and C, BLK, erbB-2, erbB-4.EGFR, mutant EGFR, HPK, IRAK1, RON, ROS1, SLK, STK10, TIE2, TRK, c-Met, Lck, Lyn, Src, Fyn,
  • kinase inhibitory activity of compounds of formula (I-VIII) is measured by means of the following assay.
  • Human EGF receptor tyrosine kinase was isolated from A431 human squamous cell carcinoma cells overexpressing EGF receptor by the following method. Cells were grown in 50% Dulbecco's modified Eagle's and 50% HAM F-12 nutrient medium (Gibco) containing 10% fetal bovine serum in roller bottles.
  • the supernatant was equilibrated with 10 mL of wheat pre-equilibrated with 50 mM Hepes, 10% glycerol, 0.1% Triton X-100 and 150 mM NaCl pH 7.5 (equilibration buffer) at 40°C.
  • the embryo agglutination agarose was equilibrated for 2 hours.
  • the contaminated protein was washed from the resin with an equilibrium buffer containing 1M NaCl, and the enzyme was eluted with an equilibrium buffer containing 0.5M N-acetyl-1-D-glucosamine, followed by 1 mM urea. The enzyme was eluted with 0.1 mg/ml EGF.
  • the receptor appears to be homogeneous.
  • various mutant forms of epidermal growth factor receptor can be isolated from appropriate cell lines containing epidermal growth factor receptor.
  • the EGFRdel746-750 mutant protein can be extracted from PC-9 cells, and the L858R/T790M double mutant EGFR protein can be isolated from H1975 cells.
  • the enzyme assay used to determine IC 50 was performed in a total volume of 25 ⁇ L. All compounds were diluted to a 500 ⁇ M stock solution in 100% DMSO and serially diluted 4 times to achieve 10 doses. The "Max” and “Min” controls contain 100% DMSO. “Max” represents the DMSO control without enzyme, and “Min” represents the low control without compound. 10 ⁇ l of the compound was transferred to 90 ⁇ l of 1x kinase base buffer for intermediate dilution.
  • the enzyme assay used to determine IC 50 was performed in a total volume of 25 ⁇ L. All compounds were diluted to a 500 ⁇ M stock solution in 100% DMSO and serially diluted 4 times to achieve 10 doses. The "Max” and “Min” controls contain 100% DMSO. “Max” represents the DMSO control without enzyme, and “Min” represents the low control without compound. 10 ⁇ l of the compound was transferred to 90 ⁇ l of 1x kinase base buffer for intermediate dilution.
  • the enzyme assay used to determine IC 50 was performed in a total volume of 25 ⁇ L. All compounds were diluted to a 500 ⁇ M stock solution in 100% DMSO and serially diluted 4 times to achieve 10 doses. The "Max” and “Min” controls contain 100% DMSO. “Max” represents the DMSO control without enzyme, and “Min” represents the low control without compound. 10 ⁇ l of the compound was transferred to 90 ⁇ l of 1x kinase base buffer for intermediate dilution.
  • the compound of formula (I) is prepared according to the method in the examples; the structure of compound AZD9291 is as follows and prepared according to the prior art method.
  • the H1975 cells were cryopreserved in liquid nitrogen. Before thawing the cells, 15 mL of cell culture medium (RPMI 1640 medium supplied with 10% fetal bovine serum and 1% penicillin/streptomycin) was placed in a T75 flask and incubated in a humidified 37°C/5% CO 2 incubator Incubate the flask in advance for 15 minutes to equilibrate the medium to the proper pH and temperature. The vial was removed from the liquid nitrogen and quickly thawed by placing in a water bath at 37°C for 1-2 minutes with gentle stirring, then decontaminated by wiping with 70% ethanol, and then opened in a Class II biosafety cabinet.
  • RPMI 1640 medium supplied with 10% fetal bovine serum and 1% penicillin/streptomycin
  • the contents of the vial were transferred dropwise to 10 mL of cell culture medium in a sterile 15 mL conical tube. The tube was then centrifuged at 200 ⁇ g for 5 minutes, and the supernatant was aspirated. The cell pellet was resuspended with 1 mL of fresh cell culture medium, and transferred to a T75 flask containing cell culture medium.
  • test compound was dissolved in DMSO at 30 mM.
  • 45 ⁇ L of compound was transferred to a 384-well compound source plate (LABCYTE catalog number P-05525) and serially diluted at a ratio of 1:3, resulting in a 13-point dilution.
  • a 384-well compound source plate (LABCYTE catalog number P-05525)
  • serially diluted at a ratio of 1:3, resulting in a 13-point dilution.
  • 20 nL of these compound DMSO dilutions (10 points, from 1.11 mM to 0.056 ⁇ M) were dispensed into new 384-well assay plates by Echo 550.
  • the cells were harvested from the flask to the cell culture medium as described above and the cell count was calculated using an automated cell counter (Thermo Fisher Scientific, Countess TM ). Dilute the cells with culture medium to 25,000 cells/ml and add 40 ⁇ L of cell suspension to each well of the 384-well cell culture plate as indicated. The final concentration is 1,000 cells/well. Add medium alone as a low control. The plate was covered with a lid and placed in a 37°C/5% CO 2 incubator for 72 hours.
  • the luminescence reading is converted to% inhibition by applying the following equation: (Lum HC -Lum Cpd )/(Lum HC -Lum LC ). Logarithmic curve, then IC 50 was calculated by four-parameter fit to the XLFit.
  • PC-9 cells The inhibition of PC-9 cells was measured in the same manner as described for H1975 cells.
  • the medium for A431 cells was Dulbecco's modified Eagle's medium supplied with 10% fetal bovine serum and 1% penicillin/streptomycin.
  • the DMSO dilution used for A431 determination is 10 points from 30nM to 1.52uM. The rest of the procedure was performed in the same manner as described for H1975 cells.
  • PC9 cells contain EGFR d746-750 single mutant (SM).
  • H1975 cells contain EGFR L858R/T790M double mutant (DM).
  • A431 cells contain unmutated EGFR wild type (WT).
  • the compound of formula (I) of the present invention has excellent EGFR inhibitory activity and anti-tumor proliferation activity, especially high inhibitory activity and selectivity for EGFR mutants.
  • the compound of formula (I) is prepared according to the method in the examples; the compound AZD9291 is prepared according to the prior art method.
  • the H1975 cells were cryopreserved in liquid nitrogen. Before thawing the cells, 15 mL of cell culture medium (RPMI 1640 medium supplied with 10% fetal bovine serum and 1% penicillin/streptomycin) was placed in a T75 flask and incubated in a humidified 37°C/5% CO 2 incubator Incubate the flask in advance for 15 minutes to equilibrate the medium to the proper pH and temperature. The vial was removed from the liquid nitrogen and quickly thawed by placing in a water bath at 37°C for 1-2 minutes with gentle stirring, then decontaminated by wiping with 70% ethanol, and then opened in a Class II biosafety cabinet.
  • RPMI 1640 medium supplied with 10% fetal bovine serum and 1% penicillin/streptomycin
  • the contents of the vial were transferred dropwise to 10 mL of cell culture medium in a sterile 15 mL conical tube. The tube was then centrifuged at 200 ⁇ g for 5 minutes, and the supernatant was aspirated. The cell pellet was resuspended with 1 mL of fresh cell culture medium, and transferred to a T75 flask containing cell culture medium.
  • the cells were harvested from the flask to the cell culture medium and the number of cells was calculated using an automated cell counter (ThermoFisher Scientific, Countess TM ).
  • the cells were diluted with culture medium to 250,000 cells/ml, and 40 ⁇ L of cell suspension was added to each well of the 384-well cell culture plate as indicated. The final concentration is 10,000 cells/well.
  • the plate was covered with a lid and placed in a 37°C/5% CO2 incubator overnight to adhere the cells.
  • test compound was dissolved in DMSO at 10 mM.
  • 45uL of compound was transferred to a 384-well compound source plate (LABCYTE catalog number P-05525) and serially diluted at a ratio of 1:3, resulting in a 13-point dilution.
  • a 384-well compound source plate (LABCYTE catalog number P-05525)
  • serially diluted at a ratio of 1:3, resulting in a 13-point dilution.
  • 40 nL of these compound DMSO dilutions 11 points, from 1.11 mM to 0.019 uM were dispensed into H1975 cell plates by Echo 550.
  • the medium for A431 cells was Dulbecco's modified Eagle's medium supplied with 10% fetal bovine serum and 1% penicillin/streptomycin.
  • the DMSO dilution used for the A431 measurement is 11 points from 10 mM to 0.17 uM.
  • EGF EGF (1 ⁇ g/mL) was added to each well and stimulated for 10 minutes. The rest of the procedure was performed in the same manner as described for H1975 cells.
  • the human lung cell line PC9 (Exon 19 lacks EGFR) was obtained from the American Type Culture Collection. PC 9 cells were maintained in RPMI1640 containing 10% fetal bovine serum and 2 mM glutamine. The cells were grown in a humidified incubator at 37°C and 5% CO 2 .
  • the assay for measuring cell phosphorylation of endogenous p-EGFR in cell lysates was performed according to the protocol described in the R&D Systems DuoSet IC Human Phosphorylation-EGF R ELISA (R&D Systems catalog number #DYCI095). 40 ⁇ L of cells were seeded (10,000 cells/well) in growth medium in a Coming black transparent bottom 384-well plate, and incubated at 37° C. and 5% CO 2 overnight.
  • the cells were acoustically administered using Echo 555, where the compound was serially diluted in 100% DMSO.
  • the plate was incubated for another 2 hours, then after aspirating the medium, 40 ⁇ L x lysis buffer was added to each well.
  • Greiner black high-binding 384-well plates were coated with capture gas and then blocked with 3% BSA. After removing the block, 15 ⁇ L of lysate was transferred to Greiner black high binding 384-well plate and incubated for 2 hours. After aspirating and washing the plate with PBS, 20 ⁇ L of detection antibody was added and incubated for 2 hours. After aspirating and washing the plate with PBS, 20 ⁇ L of QuantaBlu fluorescent peroxidase substrate (Thermo Fisher Scientific Cat.
  • the compound of formula (I) of the present invention can effectively inhibit the autophosphorylation of EGFR, and can effectively inhibit the autophosphorylation of EGFR mutants, thereby inhibiting the overexpression of EGFR and its mutants, and thereby inhibiting tumor proliferation.
  • the compound L-(+)-tartrate crystal form I of formula (I) is prepared according to the method in the examples.
  • Investigation condition 1 Put the test products under the conditions of 60°C-opening, 40°C-opening, 25°CRH75% opening, ultraviolet light-opening, respectively, on the 5th and 10th day and satisfy the total illuminance ⁇ 1.2 ⁇ 10 6 Lux ⁇ h, near ultraviolet energy ⁇ 200w ⁇ h/m 2 samples were taken to determine the content of related substances, XRD inspection was added on the 10th day, and compared with the 0 day sample.
  • Crystal form I of the compound of formula (I) when placed under the above high-temperature and high-humidity conditions for 10 days and under conditions satisfying ultraviolet illuminance, the related substances and crystalline form XRD patterns of the samples are not obvious Variety. Crystal form I has good stability, which is beneficial to the preparation, transportation and storage of medicines, and basically meets the needs of medicines.
  • Test article L-(+)-tartrate crystal form I of the compound of formula (I).
  • the L-(+)-tartrate crystal form I of the compound of formula (I) of the present invention has almost no hygroscopicity and good properties, which is convenient for the production of medicines, preparation, transportation and storage of preparations, which is conducive to ensuring the stability and safety of medicines .
  • Example 7 In vivo pharmacokinetic experiment of SD female rats of the compound of the present invention
  • Test article L-(+)-tartrate crystal form I of the compound of formula (I).
  • Test animals female SD rats, 6/administration route, body weight 204-233 g/animal.
  • Compound PO administration 2% HPC + 0.1% Tween 80;
  • Preparation method of 28% HP- ⁇ -CD solution Weigh about 28g of HP- ⁇ -CD, add sterile water for injection to a final volume of 100mL, and stir to dissolve.
  • Preparation of 2% HPC + 0.1% Tween 80 Weigh about 2 g of HPC (hydroxypropyl cellulose), add sterile water for injection to a final volume of 100 mL, then add 100 ⁇ L of Tween 80, and mix.
  • HPC hydroxypropyl cellulose
  • test drug solution is administered according to the method in Table 5 below:
  • Blood sample collection Animals collect whole blood (about 0.3 mL) in the jugular vein for pharmacokinetic analysis.
  • Time point of blood collection for IV administration animals before administration, 2min, 10min, 30min, 1h, 2h, 4h, 8h, 24h after administration.
  • Time point of blood collection for PO-administered animals before administration, 15min, 30min, 1h, 2h, 4h, 8h, 24h, 48h, 72h after administration.
  • the centrifuge tube was coated with K2-EDTA and stored in an ice-water bath; after the blood sample was collected, it was transferred to the above-mentioned centrifuge tube, mixed manually, and the centrifugation was completed within 1 hour.
  • the whole blood sample was stored in an ice-water bath before centrifugation. 10min. After centrifugation, the collected plasma was divided into 2 tubes and stored below -70°C.
  • the plasma samples of the compounds were analyzed by protein precipitation method, and the validated LC-MS/MS method was used for detection and analysis.
  • AUC last represents the area under the drug-time curve 0 ⁇ t; AUC inf represents the area under the drug-time curve 0 ⁇ ; CL represents the clearance rate; V ss represents the steady-state apparent volume of distribution; T max represents the time when the blood drug concentration reaches the peak; C max represents the peak concentration of the blood drug concentration; t 1/2 represents the half-life; MRT represents the average residence time; F% represents the absolute bioavailability

Abstract

The present invention relates to a pharmaceutically acceptable salt of N-(5-((4-(7-cyano-1,3-dimethyl-1H-indol-5-yl)pyrimidin-2-yl)amino)-2-((2-(dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide as shown in formula (I), specifically, a tartrate and crystal forms I and II of a tartrate of the compound of formula (I), a preparation method therefor, a pharmaceutical preparation containing the above-mentioned salt and crystal forms, and the use thereof in the preparation of a drug for treating and/or preventing diseases mediated by EGFR.

Description

EGFR酪氨酸激酶的选择性抑制剂的盐及其晶型Salts and crystal forms of selective inhibitors of EGFR tyrosine kinase
本申请要求于2019年01月05日提交中国专利局、申请号为201910009657.7、发明名称为“EGFR酪氨酸激酶的选择性抑制剂的盐及其晶型”的中国专利申请的优先权,其全部内容通过引用结合在本申请中。This application claims the priority of the Chinese patent application filed on January 05, 2019 in the Chinese Patent Office with the application number 201910009657.7 and the invention titled "Salts and Crystal Forms of Selective Inhibitors of EGFR Tyrosine Kinases". The entire contents are incorporated by reference in this application.
技术领域Technical field
本发明涉及一种EGFR酪氨酸激酶及其突变体的选择性抑制剂的盐、其多晶型、制备方法、含有所述抑制剂盐及其晶型的药物组合物,以及其在制备用于治疗和/或预防异常细胞增殖疾病的药物中的用途。The invention relates to a selective inhibitor salt of EGFR tyrosine kinase and its mutant, its polymorphic form, preparation method, pharmaceutical composition containing the inhibitor salt and its crystalline form, and its preparation Use in medicine for treating and/or preventing abnormal cell proliferation diseases.
背景技术Background technique
蛋白激酶(PK)是细胞内通信中非常重要的信号传导实体,其中其通过催化磷酸基从充当磷供体的ATP输送至蛋白质酪氨酸侧链上的酚羟基来修饰许多蛋白质。有时,酪氨酸激酶并入在胞外域中具有同源配体结合域的极大跨膜蛋白质的细胞内结构域中,由此配体结合在细胞内活化酪氨酸激酶。此类分子为受体酪氨酸激酶(RTK)。Protein kinase (PK) is a very important signaling entity in intracellular communication, where it modifies many proteins by catalyzing the transport of phosphate groups from ATP, which acts as a phosphorus donor, to the phenolic hydroxyl group on the protein tyrosine side chain. Sometimes, tyrosine kinase is incorporated into the intracellular domain of a very large transmembrane protein with a homologous ligand binding domain in the extracellular domain, whereby ligand binding activates tyrosine kinase within the cell. Such molecules are receptor tyrosine kinases (RTK).
十几年前,表皮生长因子RTK(EGFR、erbB-1)的两种4-苯胺基喹唑啉抑制剂吉非替尼(gefitinib)和埃罗替尼(erlotinib)被批准用于肺癌。EGFR为实体瘤中最常见的失调激酶之一,其中经常在包括非小细胞肺癌(NSCLC)的肿瘤类型的50%或更多中看到过度表达或突变。More than a decade ago, two 4-anilinoquinazoline inhibitors, gefitinib and erlotinib, of the epidermal growth factor RTK (EGFR, erbB-1) were approved for lung cancer. EGFR is one of the most common dysregulated kinases in solid tumors, where overexpression or mutations are often seen in 50% or more of tumor types including non-small cell lung cancer (NSCLC).
当追踪吉非替尼和埃罗替尼的反应者时,发现抗性的开始可能与若干不同的遗传变化有关。虽然在极少情况下,肿瘤似乎挑选完全不同的信号传导系统来驱动肿瘤,但通常抗性涉及原始系统的扭曲。EGFR是RTK的erbB(I型)子族的一员,与erbB-2、erbB-3和erbB-4一起。这些受体通过诱发其二聚的配体而活化,且虽然EGFR-EGFR同源二聚体常用于信号传导,但此家族中的更常见过程是配体诱发异源二聚,使得信号传导实体将为例如EGFR:erbB-2或erb-B2:erbB-3和适当配体。使系统再次活化的最简单方法是增加其它erbB之一的表达,且甚至在治疗前,也时常看到此,并可以帮助解释许多过度表达wt EGFR的肿瘤未对EGFR抑制起 反应的原因。一种有点相关的机制涉及RTK HGFR,其虽然并非erbB家族成员,但已经展示在过度表达时与erbB家族成员、尤其是erbB-3形成致瘤的异源二聚物,且HGFR的过度表达是对EGFR抑制剂产生抗性的常见机制。至少在实验室背景下,HGFR抑制剂添加至这些细胞将使对EGFR抑制剂的敏感性恢复。第三种和最常见的抗性模式是EGFR的进一步突变,产生双重突变受体(dm-EGFR),所述受体降低其对EGFR抑制剂的敏感性。这些中最常见的是所谓的“看门”突变T790M,且具有例如L858R/T790M的双重突变体的NSCLC在随后发展对EGFR抑制剂的抗性的初始反应者中是常见的。虽然不知道这类亚克隆是始终存在还是仅仅在治疗后出现,但似乎最可能的是,突变已存在于短期反应者中,并在后面发展抗性的长期反应者中可能作为重新突变出现。When following the responders of gefitinib and erlotinib, it was found that the onset of resistance may be related to several different genetic changes. Although in rare cases, tumors seem to pick completely different signaling systems to drive tumors, resistance usually involves distortion of the original system. EGFR is a member of the erbB (type I) subfamily of RTK, along with erbB-2, erbB-3, and erbB-4. These receptors are activated by ligands that induce their dimerization, and although EGFR-EGFR homodimers are commonly used for signaling, the more common process in this family is that ligands induce heterodimerization, making signaling entities It will be, for example, EGFR:erbB-2 or erb-B2:erbB-3 and appropriate ligands. The easiest way to reactivate the system is to increase the expression of one of the other erbBs, and even before treatment, this is often seen, and can help explain why many tumors that overexpress wt EGFR do not respond to EGFR inhibition. A somewhat related mechanism involves RTK HGFR, which, although not a member of the erbB family, has been shown to form tumorigenic heterodimers with erbB family members, especially erbB-3, when overexpressed, and the overexpression of HGFR is A common mechanism of resistance to EGFR inhibitors. At least in the laboratory setting, the addition of HGFR inhibitors to these cells will restore sensitivity to EGFR inhibitors. The third and most common pattern of resistance is the further mutation of EGFR, resulting in a double mutant receptor (dm-EGFR), which reduces its sensitivity to EGFR inhibitors. The most common of these is the so-called "gatekeeper" mutation T790M, and NSCLCs with double mutants such as L858R/T790M are common among initial responders who subsequently develop resistance to EGFR inhibitors. Although it is not known whether such subclones always exist or only appear after treatment, it seems most likely that mutations already exist in short-term responders and may appear as remutations in long-term responders who develop resistance later.
下述式(I)所述的N-(5-((4-(7-氰基-1,3-二甲基-1H-吲哚-5-基)嘧啶-2-基)氨基)-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺可以选择性地调节对当前基于EGFR的抑制疗法具有抗性的蛋白激酶,尤其是I型受体酪氨酸激酶(RTK)家族或erbB家族,更尤其是EGFR受体的某些突变形式的活性的新颖化合物,从而有效抑制细胞增殖和细胞侵入、抑制转移、诱发细胞凋亡或抑制血管生成。N-(5-((4-(7-cyano-1,3-dimethyl-1H-indol-5-yl)pyrimidin-2-yl)amino) described in the following formula (I) 2-((2-(dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide can selectively modulate protein kinases that are resistant to current EGFR-based inhibitor therapy , Especially type I receptor tyrosine kinase (RTK) family or erbB family, and more particularly active novel compounds of certain mutant forms of EGFR receptor, thereby effectively inhibiting cell proliferation and cell invasion, inhibiting metastasis, and inducing cells Apoptosis or inhibit angiogenesis.
Figure PCTCN2020070062-appb-000001
Figure PCTCN2020070062-appb-000001
晶型的研究在药物研发过程中发挥着重要的作用,不同形态的化合物具有不同的生物利用度、溶解度、稳定性等理化性质,为了满足制剂、生产、运输等情况的要求,我们对式(I)化合物的晶型进行了研究,以期发现具有良好性质的晶型。The study of crystal form plays an important role in the process of drug development. Different forms of compounds have different bioavailability, solubility, stability and other physical and chemical properties. In order to meet the requirements of preparations, production, transportation, etc., we I) The crystal form of the compound was studied in order to find a crystal form with good properties.
发明内容Summary of the invention
本发明提供了式(I)化合物的酒石酸盐,具体的为式(I)化合物的L-(+)-酒石酸盐。The present invention provides tartrates of compounds of formula (I), specifically L-(+)-tartrates of compounds of formula (I).
本发明还提供了式(I)所示的EGFR抑制剂N-(5-((4-(7-氰基-1,3-二甲基-1H-吲哚-5-基)嘧啶-2-基)氨基)-2-((2-(二甲基氨基)乙基)甲基氨基)-4-甲氧基苯基)丙烯酰胺的L-(+)-酒石酸盐晶型,包括晶型I和晶型II。The present invention also provides an EGFR inhibitor N-(5-((4-(7-cyano-1,3-dimethyl-1H-indol-5-yl)pyrimidine-2 represented by formula (I) -Yl)amino)-2-((2-(dimethylamino)ethyl)methylamino)-4-methoxyphenyl)acrylamide L-(+)-tartrate crystal form, including crystal Form I and Form II.
本发明还提供了上述式(I)化合物的L-(+)-酒石酸盐晶型I(以下简称晶型I)和式(I)化合物的L-(+)-酒石酸盐晶型II(以下简称晶型II)的制备方法,包含该晶型的药物组合物,以及该晶型在制备预防和/或治疗异常细胞增殖疾病的药物中的应用。The present invention also provides L-(+)-tartrate crystal form I (hereinafter referred to as crystal form I) of the compound of formula (I) and L-(+)-tartrate crystal form II (hereinafter referred to as crystal form I) of the compound of formula (I) The preparation method of crystal form II) for short, includes the pharmaceutical composition of the crystal form, and the application of the crystal form in the preparation of a medicine for preventing and/or treating abnormal cell proliferation diseases.
本发明提供了式(I)所示化合物的酒石酸盐,The present invention provides tartrate of the compound represented by formula (I),
Figure PCTCN2020070062-appb-000002
Figure PCTCN2020070062-appb-000002
在一些实施方案中,所述的式(I)化合物的酒石酸盐中式(I)化合物与酒石酸的摩尔比为1:3-1:1,优选1:1。In some embodiments, the molar ratio of the compound of formula (I) to tartaric acid in the tartrate salt of the compound of formula (I) is 1:3-1:1, preferably 1:1.
在一些实施方案中,所述的式(I)化合物的酒石酸盐中的酒石酸为L-(+)-酒石酸。In some embodiments, the tartaric acid in the tartaric acid salt of the compound of formula (I) is L-(+)-tartaric acid.
本发明还提供了一种式(I)所示化合物的L-(+)-酒石酸盐晶型I,The present invention also provides L-(+)-tartrate crystal form I of the compound represented by formula (I),
Figure PCTCN2020070062-appb-000003
Figure PCTCN2020070062-appb-000003
其中,式(I)化合物与L-(+)-酒石酸的摩尔比为1:1,并且使用Cu-Kα辐射,以2θ角度表示的X-射线粉末衍射图谱,在8.9±0.2°、11.2±0.2°、 12.0±0.2°、12.5±0.2°、13.1±0.2°、14.7±0.2°、17.5±0.2°、19.1±0.2°处有特征峰。Among them, the molar ratio of the compound of formula (I) to L-(+)-tartaric acid is 1:1, and Cu-Kα radiation is used to express the X-ray powder diffraction pattern at an angle of 2θ, at 8.9±0.2°, 11.2± There are characteristic peaks at 0.2°, 12.0±0.2°, 12.5±0.2°, 13.1±0.2°, 14.7±0.2°, 17.5±0.2°, 19.1±0.2°.
在一些实施方案中,在使用Cu-Kα辐射,以2θ角度表示的X-射线粉末衍射图谱中,所述晶型I在包含上述特征峰的基础上,还在16.3±0.2°、16.6±0.2°、20.0±0.2°、20.6±0.2°、21.1±0.2°、21.6±0.2°处有特征峰。In some embodiments, in the X-ray powder diffraction pattern expressed at a 2θ angle using Cu-Kα radiation, the crystalline form I, on the basis of including the above characteristic peaks, is also 16.3±0.2°, 16.6±0.2 There are characteristic peaks at °, 20.0±0.2°, 20.6±0.2°, 21.1±0.2°, 21.6±0.2°.
在一些实施方案中,所述晶型I具有与图1基本相同的X-射线粉末衍射图谱。In some embodiments, the crystalline form I has substantially the same X-ray powder diffraction pattern as in FIG. 1.
在一些实施方案中,所述晶型I的差示扫描量热分析图(DSC图谱)在190℃至230℃范围内具有吸热峰。在一些实施方案中,所述晶型I的DSC图谱在200℃至225℃范围内具有吸热峰。在一些实施方案中,所述晶型I的DSC图谱的最大吸热转变温度为215.5±5℃。在一些实施方案中,所述晶型I具有基本上如图2所示的差示扫描量热曲线。In some embodiments, the Form I differential scanning calorimetry (DSC profile) has an endothermic peak in the range of 190°C to 230°C. In some embodiments, the DSC pattern of Form I has an endothermic peak in the range of 200°C to 225°C. In some embodiments, the maximum endothermic transition temperature of the DSC pattern of Form I is 215.5±5°C. In some embodiments, the crystalline form I has a differential scanning calorimetry curve substantially as shown in FIG. 2.
在一些实施方案中,所述晶型I的TGA图谱显示,在0℃~200℃处无明显失重现象。在一些实施方案中,所述晶型I具有基本上如图3所示的TGA图谱。In some embodiments, the TGA pattern of Form I shows no significant weight loss at 0°C to 200°C. In some embodiments, the crystalline form I has a TGA pattern substantially as shown in FIG. 3.
在另一方面,本发明还提供了一种式(I)化合物的L-(+)-酒石酸盐晶型I的制备方法,其步骤包括,将式(I)化合物与1当量的L-(+)-酒石酸加入有机溶剂中,搅拌条件下加入水或N-甲基吡咯烷酮,反应结束后过滤,干燥得式(I)化合物的L-(+)-酒石酸盐晶型I;所述式(I)化合物N-(5-((4-(7-氰基-1,3-二甲基-1H-吲哚-5-基)嘧啶-2-基)氨基)-2-((2-(二甲基氨基)乙基)甲基氨基)-4-甲氧基苯基)丙烯酰胺可通过本发明实施例中的制备方法得到。In another aspect, the present invention also provides a method for preparing L-(+)-tartrate crystal form I of the compound of formula (I), the steps of which include: combining the compound of formula (I) with 1 equivalent of L-( +)-Tartaric acid is added to an organic solvent, water or N-methylpyrrolidone is added under stirring conditions, filtered after the reaction is completed, and dried to obtain the L-(+)-tartrate crystal form I of the compound of formula (I); the formula ( I) Compound N-(5-((4-(7-cyano-1,3-dimethyl-1H-indol-5-yl)pyrimidin-2-yl)amino)-2-((2- (Dimethylamino)ethyl)methylamino)-4-methoxyphenyl)acrylamide can be obtained by the preparation method in the embodiment of the present invention.
在一些优选的实施方案中,所述有机溶剂选自低级腈类、含2个以上碳原子的低级醇类、和含氧杂环类中的一种或两种以上的溶剂之间的任意组合。In some preferred embodiments, the organic solvent is selected from one or more combinations of one or more solvents selected from lower nitriles, lower alcohols containing more than 2 carbon atoms, and oxygen-containing heterocyclics .
在一些优选的实施方案中,所述有机溶剂选自乙腈、乙醇、丙醇、异丙醇、丁醇、仲丁醇、叔丁醇和1,4-二氧六环中的一种或两种以上的溶剂之间的任意组合。In some preferred embodiments, the organic solvent is selected from one or both of acetonitrile, ethanol, propanol, isopropanol, butanol, sec-butanol, tert-butanol and 1,4-dioxane Any combination of the above solvents.
在一些优选的实施方案中,上述制备方法中的有机溶剂与水的比例或 有机溶剂与N-甲基吡咯烷酮的比例为1:1-20:1,优选1:1-10:1,进一步优选2:1-10:1。In some preferred embodiments, the ratio of organic solvent to water or the ratio of organic solvent to N-methylpyrrolidone in the above preparation method is 1:1-20:1, preferably 1:1-10:1, further preferably 2:1-10:1.
本发明还提供了式(I)化合物的L-(+)-酒石酸盐晶型I的另一种制备方法,其步骤包括,将式(I)化合物加入单一溶剂或混合溶剂中,加热至一定温度后,加入一定当量的L-(+)-酒石酸,维持温度继续反应至反应结束,降温析晶,分离,干燥得到式(I)化合物的L-(+)-酒石酸盐晶型I。The present invention also provides another method for preparing L-(+)-tartrate crystal form I of the compound of formula (I). The steps include: adding the compound of formula (I) to a single solvent or a mixed solvent and heating to a certain level After the temperature, add a certain amount of L-(+)-tartaric acid, maintain the temperature to continue the reaction until the end of the reaction, lower the temperature and crystallize, separate, and dry to obtain L-(+)-tartrate crystal form I of the compound of formula (I).
在一些优选的实施方案中,上述制备方法中所述的单一溶剂选自低级醇类、低级酮类、低级酯类、含氧杂环类、低级腈类、低级烷烃类或低级卤代烷烃类;所述的混合溶剂选自醇类与水混合溶剂、腈类与水混合溶剂、酮类与水混合溶剂或含氧杂环类与水混合溶剂。In some preferred embodiments, the single solvent described in the above preparation method is selected from lower alcohols, lower ketones, lower esters, oxygenated heterocyclics, lower nitriles, lower alkanes or lower halogenated alkanes; The mixed solvent is selected from a mixed solvent of alcohols and water, a mixed solvent of nitriles and water, a mixed solvent of ketones and water, or a mixed solvent of oxygen-containing heterocyclics and water.
在一些优选的实施方案中,上述制备方法中所述单一溶剂选自甲醇、乙醇、丙醇、异丙醇、正丁醇、异丁醇、叔丁醇、丙酮、丁酮、甲基乙基酮、乙腈、甲酸乙酯、乙酸乙酯、乙酸甲酯、四氢呋喃、1,4-二氧六环、正戊烷、异戊烷、正己烷、异己烷、环己烷、环庚烷、甲苯或二氯甲烷;所述混合溶剂选自乙醇/水、丙醇/水、异丙醇/水、丁醇/水、仲丁醇/水、叔丁醇/水、乙腈/水、1,4-二氧六环/水或丙酮/水。In some preferred embodiments, the single solvent in the above preparation method is selected from methanol, ethanol, propanol, isopropanol, n-butanol, isobutanol, t-butanol, acetone, butanone, methyl ethyl Ketone, acetonitrile, ethyl formate, ethyl acetate, methyl acetate, tetrahydrofuran, 1,4-dioxane, n-pentane, isopentane, n-hexane, isohexane, cyclohexane, cycloheptane, toluene Or dichloromethane; the mixed solvent is selected from ethanol/water, propanol/water, isopropanol/water, butanol/water, sec-butanol/water, tert-butanol/water, acetonitrile/water, 1,4 -Dioxane/water or acetone/water.
在一些优选的实施方案中,所述混合溶剂中,有机溶剂与水的比例选自1:1-20:1,进一步优选的,所述混合溶剂中,有机溶剂与水的比例选自1:1-10:1,更优选的,所述混合溶剂中,有机溶剂与水的比例选自2:1-10:1。In some preferred embodiments, in the mixed solvent, the ratio of organic solvent to water is selected from 1:1-20:1, further preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 1: 1-10:1, more preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 2:1-10:1.
在一些优选的实施方案中,所述混合溶剂中,有机溶剂与水的比例选自1:1、2:1、3:1、3:2、4:1、5:1、5:2、5:3、6:1、8:1、8:3、8:5、10:1或10:3;In some preferred embodiments, in the mixed solvent, the ratio of organic solvent to water is selected from 1:1, 2:1, 3:1, 3:2, 4:1, 5:1, 5:2, 5:3, 6:1, 8:1, 8:3, 8:5, 10:1 or 10:3;
在一些优选的实施方案中,上述制备方法中所述的一定温度选自40℃-80℃,优选50℃-70℃。In some preferred embodiments, the certain temperature described in the above preparation method is selected from 40°C-80°C, preferably 50°C-70°C.
在一些优选的实施方案中,上述制备方法中所述的一定当量选自0.9-1.2当量,优选1.0-1.1当量,更优选1.0当量。In some preferred embodiments, certain equivalents described in the above preparation method are selected from 0.9-1.2 equivalents, preferably 1.0-1.1 equivalents, more preferably 1.0 equivalents.
本发明还提供了式(I)化合物的L-(+)-酒石酸盐晶型I的第三种制备方法,其步骤包括,将式(I)化合物加入单一溶剂或混合溶剂中,常温浆洗条件下,加入一定当量的L-(+)-酒石酸,维持温度继续反应至反应结 束,常温放置析晶,分离,干燥得到式(I)化合物的L-(+)-酒石酸盐晶型I。The present invention also provides a third method for preparing L-(+)-tartrate crystal form I of the compound of formula (I). The steps include: adding the compound of formula (I) to a single solvent or a mixed solvent, and slurry washing at room temperature Next, add a certain equivalent of L-(+)-tartaric acid, maintain the temperature and continue the reaction until the end of the reaction. Place the crystal at room temperature, separate, and dry to obtain L-(+)-tartrate crystal form I of the compound of formula (I).
在一些优选的实施方案中,上述制备方法中所述的单一溶剂选自低级醇类、低级酮类、低级酯类、含氧杂环类、低级腈类、低级烷烃类或低级卤代烷烃类;所述的混合溶剂选自醇类与水混合溶剂、腈类与水混合溶剂、酮类与水混合溶剂或含氧杂环类与水混合溶剂。In some preferred embodiments, the single solvent described in the above preparation method is selected from lower alcohols, lower ketones, lower esters, oxygenated heterocyclics, lower nitriles, lower alkanes or lower halogenated alkanes; The mixed solvent is selected from a mixed solvent of alcohols and water, a mixed solvent of nitriles and water, a mixed solvent of ketones and water, or a mixed solvent of oxygen-containing heterocyclics and water.
在一些优选的实施方案中,上述制备方法中所述单一溶剂选自甲醇、乙醇、丙醇、异丙醇、正丁醇、异丁醇、叔丁醇、丙酮、丁酮、甲基乙基酮、乙腈、甲酸乙酯、乙酸乙酯、乙酸甲酯、四氢呋喃、1,4-二氧六环、正戊烷、异戊烷、正己烷、异己烷、环己烷、环庚烷、甲苯或二氯甲烷;所述混合溶剂选自乙醇/水、丙醇/水、异丙醇/水、丁醇/水、仲丁醇/水、叔丁醇/水、乙腈/水、1,4-二氧六环/水或丙酮/水。In some preferred embodiments, the single solvent in the above preparation method is selected from methanol, ethanol, propanol, isopropanol, n-butanol, isobutanol, t-butanol, acetone, butanone, methyl ethyl Ketone, acetonitrile, ethyl formate, ethyl acetate, methyl acetate, tetrahydrofuran, 1,4-dioxane, n-pentane, isopentane, n-hexane, isohexane, cyclohexane, cycloheptane, toluene Or dichloromethane; the mixed solvent is selected from ethanol/water, propanol/water, isopropanol/water, butanol/water, sec-butanol/water, tert-butanol/water, acetonitrile/water, 1,4 -Dioxane/water or acetone/water.
在一些优选的实施方案中,所述混合溶剂中,有机溶剂与水的比例选自1:1-20:1,进一步优选的,所述混合溶剂中,有机溶剂与水的比例选自1:1-10:1,更优选的,所述混合溶剂中,有机溶剂与水的比例选自2:1-10:1。In some preferred embodiments, in the mixed solvent, the ratio of organic solvent to water is selected from 1:1-20:1, further preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 1: 1-10:1, more preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 2:1-10:1.
在一些优选的实施方案中,所述混合溶剂中,有机溶剂与水的比例选自1:1、2:1、3:1、3:2、4:1、5:1、5:2、5:3、6:1、8:1、8:3、8:5、10:1或10:3;In some preferred embodiments, in the mixed solvent, the ratio of organic solvent to water is selected from 1:1, 2:1, 3:1, 3:2, 4:1, 5:1, 5:2, 5:3, 6:1, 8:1, 8:3, 8:5, 10:1 or 10:3;
在一些优选的实施方案中,上述制备方法中所述的一定当量选自0.9-1.2当量,优选1.0-1.1当量,更优选1.0当量。In some preferred embodiments, certain equivalents described in the above preparation method are selected from 0.9-1.2 equivalents, preferably 1.0-1.1 equivalents, more preferably 1.0 equivalents.
在另一方面,本发明还提供了一种式(I)所示化合物的L-(+)-酒石酸盐晶型II,In another aspect, the present invention also provides L-(+)-tartrate crystal form II of the compound represented by formula (I),
Figure PCTCN2020070062-appb-000004
Figure PCTCN2020070062-appb-000004
其中,式(I)化合物与酒石酸的摩尔比为1:1,并且使用Cu-Kα辐 射,以2θ角度表示的X-射线粉末衍射图谱,在9.0±0.2°、11.3±0.2°、12.0±0.2°、12.7±0.2°、13.9±0.2°、17.9±0.2°、19.1±0.2°、20.3±0.2°、21.2±0.2°处有特征峰。Wherein, the molar ratio of the compound of formula (I) to tartaric acid is 1:1, and the Cu-Kα radiation is used to express the X-ray powder diffraction pattern at an angle of 2θ, at 9.0±0.2°, 11.3±0.2°, 12.0±0.2 There are characteristic peaks at °, 12.7±0.2°, 13.9±0.2°, 17.9±0.2°, 19.1±0.2°, 20.3±0.2°, 21.2±0.2°.
在一些实施方案中,在使用Cu-Kα辐射,以2θ角度表示的X-射线粉末衍射图谱中,所述晶型II在包含上述特征峰的基础上,还在16.3±0.2°、17.3±0.2°、23.5±0.2°、24.4±0.2°、25.5±0.2°、25.9±0.2°处有特征峰;优选的,所述晶型II的X-射线粉末衍射图谱基本如图4所示。In some embodiments, in the X-ray powder diffraction pattern expressed at a 2θ angle using Cu-Kα radiation, the crystalline form II, on the basis of including the above characteristic peaks, is also 16.3±0.2°, 17.3±0.2 °, 23.5±0.2°, 24.4±0.2°, 25.5±0.2°, 25.9±0.2° have characteristic peaks; preferably, the X-ray powder diffraction pattern of the crystalline form II is basically shown in FIG. 4.
本发明还提供了一种晶型II的制备方法,其步骤包括,将式(I)化合物和1当量L-(+)-酒石酸加入有机溶剂中,常温搅拌条件下,加入水,继续常温搅拌至反应结束,过滤,干燥,得晶型II。The invention also provides a method for preparing crystalline form II. The steps include: adding the compound of formula (I) and 1 equivalent of L-(+)-tartaric acid to an organic solvent, adding water under normal temperature stirring conditions, and continuing to stir at normal temperature At the end of the reaction, filter and dry to obtain Form II.
在一些优选的实施方案中,上述制备方法中所述的有机溶剂选自甲醇。In some preferred embodiments, the organic solvent described in the above preparation method is selected from methanol.
本发明还提供了式(I)化合物的L-(+)-酒石酸晶型I或晶型II与一种或多种药用载体和/或稀释剂的药物组合物,所述药物组合物可制备成药学上可接受的任一剂型,以口服、肠胃外、直肠或经肺给药等方式施用于需要其的患者。用于口服给药时,可制成常规的固体制剂,如片剂、胶囊剂、丸剂、颗粒剂等;也可制成口服液体制剂,如口服溶液剂、口服混悬剂、糖浆剂等。制成口服制剂时,可以加入适宜的填充剂、粘合剂、崩解剂、润滑剂等。用于肠胃外给药时,可制成注射剂,包括注射液、注射用无菌粉末与注射用浓溶液。制成注射剂时,可采用现有制药领域中的常规方法生产,配制注射剂时,可以不加入附加剂,也可根据药物的性质加入适宜的附加剂。用于直肠给药时,可制成栓剂等。用于经肺给药时,可制成吸入剂或喷雾剂等。The present invention also provides a pharmaceutical composition of L-(+)-tartaric acid crystal form I or crystal form II of the compound of formula (I) and one or more pharmaceutically acceptable carriers and/or diluents, the pharmaceutical composition may be It is prepared into any pharmaceutically acceptable dosage form and administered to patients in need thereof by oral, parenteral, rectal, or pulmonary administration. When used for oral administration, it can be made into conventional solid preparations, such as tablets, capsules, pills, granules, etc.; it can also be made into oral liquid preparations, such as oral solutions, oral suspensions, syrups, etc. When making an oral preparation, a suitable filler, binder, disintegrant, lubricant, etc. may be added. When used for parenteral administration, it can be made into injections, including injections, sterile powders for injection and concentrated solutions for injection. When it is prepared as an injection, it can be produced by a conventional method in the existing pharmaceutical field. When preparing an injection, an additional agent may not be added, or an appropriate additional agent may be added according to the nature of the drug. When used for rectal administration, it can be made into suppositories. When used for pulmonary administration, it can be made into inhalants or sprays.
本发明还提供了式(I)化合物的L-(+)-酒石酸盐晶型I或晶型II在制备治疗和/或预防EGFR所介导的过度增殖性疾病的药物中的用途。The present invention also provides the use of L-(+)-tartrate crystal form I or crystal form II of the compound of formula (I) in the preparation of a medicament for the treatment and/or prevention of EGFR-mediated hyperproliferative diseases.
本发明还提供了一种治疗和/或预防由EGFR所介导的过度增殖性疾病的方法,其包括向有此需要的受试者施用有效量的所述式(I)化合物的L-(+)-酒石酸盐晶型I或晶型II的步骤。The present invention also provides a method for treating and/or preventing hyperproliferative diseases mediated by EGFR, which comprises administering to a subject in need thereof an effective amount of the compound of formula (I) L-( +)-Step of tartrate form I or form II.
在一些优选的实施方案中,上述所述的过度增殖性疾病选自癌症。In some preferred embodiments, the hyperproliferative disease described above is selected from cancer.
在一些优选的实施方案中,上述所述的EGFR包括野生型EGFR和 EGFR突变体。In some preferred embodiments, the EGFR described above includes wild-type EGFR and EGFR mutants.
在一些优选的实施方案中,上述所述的EGFR突变体包括EGFR 19位外显子突变、EGFR 20位外显子突变和EGFR 21位外显子突变中的一种或两种以上的任意组合。In some preferred embodiments, the above-mentioned EGFR mutants include one or any combination of one or more of EGFR 19 exon mutation, EGFR 20 exon mutation and EGFR 21 exon mutation .
在一些优选的实施方案中,上述所述的EGFR 20位外显子突变选自NPG、ASV或T790M。In some preferred embodiments, the aforementioned exon 20 mutation of EGFR is selected from NPG, ASV or T790M.
在一些实施方案中,上述所述的癌症选自肺癌、非小细胞肺癌、结直肠癌、胰腺癌、头颈癌、乳腺癌、卵巢癌、子宫癌、肝癌、胃癌、前列腺癌、胶质母细胞瘤和上皮细胞癌。In some embodiments, the cancer described above is selected from lung cancer, non-small cell lung cancer, colorectal cancer, pancreatic cancer, head and neck cancer, breast cancer, ovarian cancer, uterine cancer, liver cancer, gastric cancer, prostate cancer, glioblastoma Tumor and epithelial cell carcinoma.
在一些优选的实施方案中,上述所述癌症由EGFR 20位外显子突变引起。In some preferred embodiments, the cancer described above is caused by mutations in exon 20 of EGFR.
在一些优选的实施方案中,上述所述癌症由EGFR 20位外显子突变中的T790M突变引起,同时存在EGFR 19位外显子插入突变或EGFR 21位外显子点突变。In some preferred embodiments, the above-mentioned cancer is caused by the T790M mutation in EGFR 20 exon mutation, and there is also an EGFR 19 exon insertion mutation or EGFR 21 exon point mutation.
本公开还提供所述的式(I)化合物的L-(+)-酒石酸盐晶型I或晶型II在制备试剂中的用途,所述试剂可用于抑制细胞中EGFR酶的水平。在一些优选的实施方案中,所述试剂用于体内或体外方法中。The present disclosure also provides the use of the L-(+)-tartrate crystalline form I or crystalline form II of the compound of formula (I) in the preparation of reagents that can be used to inhibit the level of EGFR enzymes in cells. In some preferred embodiments, the agent is used in an in vivo or in vitro method.
本公开还提供一种抑制细胞中EGFR水平的方法,其包括向细胞施用有效量的式(I)化合物的L-(+)-酒石酸盐晶型I或晶型II的步骤。在一些优选的实施方案中,所述方法在体内或体外进行。The present disclosure also provides a method of inhibiting the level of EGFR in a cell, which includes the step of administering to the cell an effective amount of L-(+)-tartrate salt form I or form II of the compound of formula (I). In some preferred embodiments, the method is performed in vivo or in vitro.
在发明中除非另有说明,否则本文中使用的科学和技术名词具有本领域技术人员所通常理解的含义。同时,为了更好地理解本发明,下面提供部分术语的定义和解释。Unless otherwise stated in the invention, the scientific and technical terms used herein have the meaning commonly understood by those skilled in the art. Meanwhile, in order to better understand the present invention, definitions and explanations of some terms are provided below.
本发明中,术语“低级醇类”是指含有1-6个碳原子的直链或支链脂肪醇,或环状脂肪醇,其实例包括但不限于:乙醇、丙醇、异丙醇、丁醇、仲丁醇、叔丁醇和环戊醇。In the present invention, the term "lower alcohols" refers to straight-chain or branched-chain fatty alcohols containing 1 to 6 carbon atoms, or cyclic fatty alcohols, examples of which include but are not limited to: ethanol, propanol, isopropanol, Butanol, sec-butanol, tert-butanol and cyclopentanol.
本发明中,术语“低级腈类”含有1-6个碳原子的腈类化合物,其实例包括但不限于:乙腈、丙腈和己二腈。In the present invention, the term "lower nitrile" contains 1-6 carbon atoms, and examples thereof include, but are not limited to, acetonitrile, propionitrile, and adiponitrile.
本发明中,术语“低级酮类”含有1-6个碳原子的脂肪酮类化合物,其 实例包括但不限于:甲乙酮、甲基异丙基酮、丙酮、甲基丁酮和甲基异丁酮。In the present invention, the term "lower ketones" contains fatty ketone compounds with 1-6 carbon atoms, and examples thereof include, but are not limited to: methyl ethyl ketone, methyl isopropyl ketone, acetone, methyl butanone, and methyl isobutyl ketone.
本发明中,术语“低级酯类”含有1-6个碳原子的脂肪酯类化合物,其实例包括但不限于:甲酸甲酯、甲酸乙酯、甲酸丙酯、乙酸甲酯、乙酸乙酯、乙酸丙酯、乙酸异丙酯、丙酸甲酯、丙酸乙酯、丙酸丙酯和丙酸异丙酯。In the present invention, the term "lower esters" contains fatty ester compounds of 1-6 carbon atoms, examples of which include but are not limited to: methyl formate, ethyl formate, propyl formate, methyl acetate, ethyl acetate, Propyl acetate, isopropyl acetate, methyl propionate, ethyl propionate, propyl propionate and isopropyl propionate.
本发明中,术语“低级烷烃类”含有1-7个碳原子的直链或支链脂肪烷烃,或环状脂肪烷烃,或芳香烷烃,其实例包括但不限于:正戊烷、异戊烷、正己烷、异己烷、环戊烷、环己烷和甲苯。In the present invention, the term "lower alkanes" contains straight-chain or branched-chain aliphatic alkanes with 1-7 carbon atoms, or cyclic aliphatic alkanes, or aromatic alkanes, examples of which include but are not limited to: n-pentane, isopentane , N-hexane, isohexane, cyclopentane, cyclohexane and toluene.
本发明中,术语“低级卤代烷烃类”含有1-6个碳原子的且被至少一个卤原子取代的直链或支链脂肪烷烃,或环状脂肪烷烃。其实例包括但不限于:二氯甲烷、三氯甲烷、四氯化碳、1,1-二氯乙烷、1,2-二氯乙烷和1,1,1-三氯乙烷。In the present invention, the term "lower halogenated alkane" contains a linear or branched aliphatic alkane or cyclic aliphatic alkane containing 1 to 6 carbon atoms and substituted with at least one halogen atom. Examples include, but are not limited to: methylene chloride, chloroform, carbon tetrachloride, 1,1-dichloroethane, 1,2-dichloroethane, and 1,1,1-trichloroethane.
本发明中,术语“分离”通过常规的方法,例如过滤、离心弃上清等,来分离固体。In the present invention, the term "separation" is used to separate solids by conventional methods, such as filtration, centrifugation and discarding the supernatant.
本发明中,术语“过滤”包括但不限于常压过滤、减压过滤等。In the present invention, the term "filtration" includes but is not limited to atmospheric pressure filtration, reduced pressure filtration and the like.
如本文中所使用的,术语“室温”、“常温”是指室内环境温度,通常指10-30℃,例如20-25℃。As used herein, the terms "room temperature" and "normal temperature" refer to indoor ambient temperature, usually 10-30°C, such as 20-25°C.
本发明中,术语“干燥”包括但不限于室温下自然晾干、红外灯干燥、烘箱烘干、干燥器干燥、真空条件下干燥。所述的干燥温度为室温或30-60℃,优选40-50℃。In the present invention, the term "drying" includes but is not limited to natural drying at room temperature, infrared lamp drying, oven drying, dryer drying, and drying under vacuum conditions. The drying temperature is room temperature or 30-60°C, preferably 40-50°C.
本发明中,术语“降温析晶”是指通过冰水浴降温、自热冷却降温、使用制冷设备降温等方式使反应体系降温至0-35℃,优选降温至10-30℃,进一步优选15-25℃,更优选降温至大部分晶体析出。化合物可以两种或多种结晶状态存在,结构相同的分子,结晶成不同的固体形式,称为多晶型物或多晶形等。当涉及具体结晶形式时,常称“crystal form”,即为本文中使用的术语“晶型”。In the present invention, the term "cooling crystallization" refers to cooling the reaction system to 0-35°C, preferably to 10-30°C, and more preferably 15-°C by cooling in an ice-water bath, cooling by autothermal cooling, or cooling by refrigeration equipment. At 25°C, it is more preferable to lower the temperature until most crystals precipitate. Compounds can exist in two or more crystalline states, molecules with the same structure, crystallized into different solid forms, called polymorphs or polymorphs. When referring to a specific crystalline form, it is often called "crystal", which is the term "crystal form" used herein.
在本公开中,各晶型X射线粉末衍射图谱中吸收峰的位置可以在上述发明的具体数值±0.2°的范围内,例如在±0.1°的范围内,差示扫描量热 法测定的吸热转变温度可以在上述具体数值±5.0℃(例如±3.0℃或±2.0℃)的范围内。In the present disclosure, the position of the absorption peak in the X-ray powder diffraction pattern of each crystal form may be in the range of ±0.2° of the specific value of the above invention, for example, in the range of ±0.1°, the absorption measured by differential scanning calorimetry The thermal transition temperature may be within the range of the above specific numerical value ±5.0°C (for example, ±3.0°C or ±2.0°C).
应当理解用不同类型设备或用不同的测试条件可能给出稍微不同的XRPD的图谱和峰值。不同晶型的图谱、峰值和各衍射峰的相对强度将受化合物纯度、样品的前处理、扫描速度、粒径和测试设备的校验和维修的影响。所提供的数值不能作为绝对值。It should be understood that using different types of equipment or using different test conditions may give slightly different XRPD patterns and peaks. The patterns, peaks and relative intensity of different diffraction peaks of different crystal forms will be affected by the purity of the compound, sample preparation, scanning speed, particle size, and calibration and maintenance of the test equipment. The value provided cannot be used as an absolute value.
应当理解用不同类型设备或用不同的测试条件可能给出稍微不同的DSC图谱和吸热转变温度读数。该数值将受化合物纯度、样品重量、加热速度、粒径和测试设备的校验和维修的影响。晶型的最大吸热转变温度可以在上述公开的具体数值±5.0℃的范围内。It should be understood that using different types of equipment or using different test conditions may give slightly different DSC patterns and endothermic transition temperature readings. This value will be affected by compound purity, sample weight, heating rate, particle size, and calibration and maintenance of test equipment. The maximum endothermic transition temperature of the crystal form may be within the range of the specific values disclosed above ±5.0°C.
本公开还采用热失重分析(TGA)对晶型发生分解或升华、蒸发的程度(失去重量)与温度的关系进行了分析。应当理解同种晶型受样品纯度、粒径、不同类型设备、不同的测试方法等的影响,所得到的数值存在一定误差。晶型发生分解或升华、蒸发时的温度可以在上述公开的具体数值±3.0℃的范围内,例如±2.0℃的范围内。The present disclosure also uses thermal weight loss analysis (TGA) to analyze the relationship between the degree of crystal form decomposition or sublimation and evaporation (loss of weight) and temperature. It should be understood that the same crystal form is affected by sample purity, particle size, different types of equipment, different test methods, etc., and there are certain errors in the obtained values. The temperature at which the crystal form decomposes, sublimates, or evaporates may be within the range of the specific values disclosed above ±3.0°C, for example, ±2.0°C.
本发明中,术语“受试者”是指动物或人,优选哺乳动物或人。In the present invention, the term "subject" refers to an animal or a human, preferably a mammal or a human.
本发明中,术语“有效量”是指足以实现所需治疗或预防效果的量,例如,实现减轻与待治疗疾病相关的症状的量。In the present invention, the term "effective amount" refers to an amount sufficient to achieve a desired therapeutic or preventive effect, for example, an amount that achieves relief of symptoms related to the disease to be treated.
本发明中,术语“治疗”目的是减轻或消除所针对的疾病状态或病症。如果受试者按照本文所述方法接受了治疗量的所述晶型或其药物组合物,该受试者一种或多种指征和症状表现出可观察到的和/或可检测出的降低或改善,则受试者被成功地“治疗”了。还应当理解,所述的疾病状态或病症的治疗不仅包括完全地治疗,还包括未达到完全地治疗,但实现了一些生物学或医学相关的结果。In the present invention, the term "treatment" aims to reduce or eliminate the targeted disease state or disorder. If a subject receives a therapeutic amount of the crystalline form or its pharmaceutical composition according to the methods described herein, the subject's one or more indications and symptoms exhibit observable and/or detectable Decrease or improve, the subject is successfully "treated". It should also be understood that the treatment of the disease state or disorder includes not only complete treatment but also incomplete treatment, but has achieved some biological or medical related results.
本发明式(I)化合物、其酒石酸盐以及其L-(+)-酒石酸盐晶型的主要优点包括:The main advantages of the compound of formula (I) of the present invention, its tartrate and its L-(+)-tartrate crystal form include:
(1)本发明式(I)化合物、其酒石酸盐以及其L-(+)-酒石酸盐晶型具有优异的EGFR的抑制活性,尤其是对EGFR受体的某些突变形式具有较高的选择性和抑制活性,可有效抑制细胞增殖和细胞侵入、抑制转移、 诱发细胞凋亡或抑制血管生成;(1) The compound of formula (I) of the present invention, its tartrate and its L-(+)-tartrate crystal form have excellent EGFR inhibitory activity, especially with a higher selection of certain mutant forms of EGFR receptor Sexuality and inhibitory activity can effectively inhibit cell proliferation and cell invasion, inhibit metastasis, induce apoptosis or inhibit angiogenesis;
(2)本发明式(I)化合物、其酒石酸盐以及其L-(+)-酒石酸盐晶型的毒副作用较小,尤其是在糖尿病、心脏毒性等方面副作用较小;(2) The compound of formula (I) of the present invention, its tartrate and its L-(+)-tartrate crystal form have less toxic and side effects, especially in the aspects of diabetes and cardiotoxicity;
(3)本发明式(I)化合物、其酒石酸盐以及其L-(+)-酒石酸盐晶型具有良好的药代动力学性质,具有优异的半衰期、较高的暴露量等性质,成药性良好。(3) The compound of formula (I) of the present invention, its tartrate and its L-(+)-tartrate crystal form have good pharmacokinetic properties, excellent half-life, high exposure, etc. good.
(4)本发明式(I)化合物的L-(+)-酒石酸盐晶型稳定性良好,且具有良好的性状、流动性和可压性,便于检测、制剂、运输和储藏;(4) The L-(+)-tartrate salt crystal form of the compound of formula (I) of the present invention has good stability, and has good properties, fluidity and compressibility, which is convenient for detection, preparation, transportation and storage;
(5)本发明式(I)化合物的L-(+)-酒石酸盐晶型纯度高,残留溶剂少,溶解度较高,溶出度好,稳定性好,质量易控。(5) The L-(+)-tartrate salt crystal form of the compound of formula (I) of the present invention has high purity, less residual solvent, higher solubility, good dissolution, good stability and easy quality control.
附图说明BRIEF DESCRIPTION
图1是式(I)化合物的L-(+)-酒石酸盐晶型I的X-射线粉末衍射图谱,纵坐标表示衍射强度(intensity),横坐标表示衍射角度(2θ);FIG. 1 is an X-ray powder diffraction pattern of L-(+)-tartrate salt crystal form I of the compound of formula (I). The ordinate represents the diffraction intensity, and the abscissa represents the diffraction angle (2θ);
图2是式(I)化合物的L-(+)-酒石酸盐晶型I的差示扫描量热(DSC)热分析图,纵坐标表示热流(W/g),横坐标表示温度(℃);Fig. 2 is a differential scanning calorimetry (DSC) thermal analysis diagram of L-(+)-tartrate salt crystal form I of the compound of formula (I), the ordinate represents heat flow (W/g), and the abscissa represents temperature (°C) ;
图3是式(I)化合物的L-(+)-酒石酸盐晶型I的热重分析(TGA)曲线,纵坐标表示质量百分比(%),横坐标表示温度(℃);Fig. 3 is a thermogravimetric analysis (TGA) curve of L-(+)-tartrate crystal form I of the compound of formula (I). The ordinate represents the mass percentage (%), and the abscissa represents the temperature (°C);
图4是式(I)化合物的L-(+)-酒石酸盐晶型I的氢谱;4 is a hydrogen spectrum of L-(+)-tartrate crystal form I of the compound of formula (I);
图5是式(I)化合物的L-(+)-酒石酸盐晶型II的X-射线粉末衍射图谱,纵坐标表示衍射强度(intensity),横坐标表示衍射角度(2θ)。Fig. 5 is an X-ray powder diffraction pattern of L-(+)-tartrate crystalline form II of the compound of formula (I). The ordinate indicates the diffraction intensity, and the abscissa indicates the diffraction angle (2θ).
具体实施方式detailed description
以下通过实施例形式的具体实施方式,对本发明的上述内容作进一步的详细说明。但不应理解为本发明上述主题的范围仅限于以下实施例。凡基于本发明上述内容所实现的技术均属于本发明的范围。The above content of the present invention will be further described in detail below through specific implementations in the form of examples. However, it should not be understood that the scope of the above subject matter of the present invention is limited to the following embodiments. All technologies implemented based on the above contents of the present invention belong to the scope of the present invention.
实施例1:式(I)化合物的制备Example 1: Preparation of compound of formula (I)
(1)5-溴-1,3-二甲基-1H-吲哚-7-甲腈的制备(1) Preparation of 5-bromo-1,3-dimethyl-1H-indole-7-carbonitrile
Figure PCTCN2020070062-appb-000005
Figure PCTCN2020070062-appb-000005
将5-溴-3-甲基-1H-吲哚-7-甲腈(1.17g,5mmol)溶于四氢呋喃(20mL)中,冷却至0℃,分批加入NaH(260mg,6.5mmol)。加毕,0℃搅拌30分钟,然后缓慢滴加MeI(781mg,5.5mmol)。滴毕,自然升至室温并搅拌反应两小时,小心加水淬灭反应,乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥,过滤,滤液浓缩得粗品,柱层析纯化得目标化合物(694mg,收率:56%)。5-Bromo-3-methyl-1H-indole-7-carbonitrile (1.17 g, 5 mmol) was dissolved in tetrahydrofuran (20 mL), cooled to 0°C, and NaH (260 mg, 6.5 mmol) was added in portions. After the addition, the mixture was stirred at 0°C for 30 minutes, and then MeI (781 mg, 5.5 mmol) was slowly added dropwise. After the completion of the drop, the mixture was naturally raised to room temperature and stirred for two hours. The reaction was quenched carefully with water, extracted with ethyl acetate (20mL×3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to obtain a crude product. 694 mg, yield: 56%).
1HNMR(300MHz,CDCl 3):7.87(d,J=1.5Hz,1H),7.60(d,J=1.5Hz,1H),6.88(s,1H),4.06(s,3H),2.28(s,3H). 1 HNMR (300 MHz, CDCl 3 ): 7.87 (d, J = 1.5 Hz, 1H), 7.60 (d, J = 1.5 Hz, 1H), 6.88 (s, 1H), 4.06 (s, 3H), 2.28 (s ,3H).
(2)1,3-二甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吲哚-7-甲腈的制备(2) 1,3-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)-1H-indole- Preparation of 7-carbonitrile
Figure PCTCN2020070062-appb-000006
Figure PCTCN2020070062-appb-000006
将化合物5-溴-1,3-二甲基-1H-吲哚-7-甲腈(523mg,2.1mmol,1.0eq)溶于二氧六环(5mL)中,加入双联硼酸频哪醇酯(592mg,2.3mmol,1.1eq),乙酸钾(125mg,6.3mol,3eq),Pd(dppf)Cl 2(124mg,0.168mmol,0.08eq)置换氮气3次,然后加热至85℃,反应6小时。TLC,LCMS检测,反应完全后,抽滤,母液浓缩,柱层析纯化得目标化合物(350mg,收率:56%)。 The compound 5-bromo-1,3-dimethyl-1H-indole-7-carbonitrile (523mg, 2.1mmol, 1.0eq) was dissolved in dioxane (5mL), and bisboronic acid pinacol was added Ester (592 mg, 2.3 mmol, 1.1 eq), potassium acetate (125 mg, 6.3 mol, 3 eq), Pd (dppf) Cl 2 (124 mg, 0.168 mmol, 0.08 eq) replaced nitrogen 3 times, then heated to 85° C., reaction 6 hour. TLC and LCMS detection. After the reaction was completed, suction filtration, mother liquor concentration, and column chromatography purification to obtain the target compound (350 mg, yield: 56%).
1HNMR(300MHz,CDCl 3):□8.23(s,1H),7.99(s,1H),6.85(s,1H),4.09(s,3H),2.33(s,3H),1.39(s,12H). 1 HNMR (300MHz, CDCl 3 ): 8.23 (s, 1H), 7.99 (s, 1H), 6.85 (s, 1H), 4.09 (s, 3H), 2.33 (s, 3H), 1.39 (s, 12H ).
(3)5-(2-氯嘧啶-4-基)-1,3-二甲基-1H-吲哚-7-甲腈的制备(3) Preparation of 5-(2-chloropyrimidin-4-yl)-1,3-dimethyl-1H-indole-7-carbonitrile
Figure PCTCN2020070062-appb-000007
Figure PCTCN2020070062-appb-000007
氮气保护下,将化合物1,3-二甲基-5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吲哚-7-甲腈(296mg,1mmol,1.0eq)溶解在二氧六环和水(5/1mL)中,分别加入2,4-二氯嘧啶(162mg,1.1mmol,1.1eq),Pd(PPh 3) 4(115mg,0.1mmol,0.1eq),碳酸钾(411mg,3mmol,3eq)加毕升温至100℃反应3小时。TLC,LCMS检测,反应完全后,抽滤,滤液浓缩柱层析提纯,得目标化合物(164mg,收率:58%)。 Under nitrogen, the compound 1,3-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)-1H- Indole-7-carbonitrile (296mg, 1mmol, 1.0eq) was dissolved in dioxane and water (5/1mL), 2,4-dichloropyrimidine (162mg, 1.1mmol, 1.1eq), Pd were added respectively (PPh 3 ) 4 (115 mg, 0.1 mmol, 0.1 eq), potassium carbonate (411 mg, 3 mmol, 3 eq) was added and the temperature was raised to 100° C. for 3 hours. TLC and LCMS detection. After the reaction was completed, suction filtration was carried out, and the filtrate was concentrated and purified by column chromatography to obtain the target compound (164 mg, yield: 58%).
1HNMR(300MHz,DMSO-d 6):□8.65(d,J=5.1Hz,1H),8.55(s,1H),8.29(s,1H),7.70(d,J=5.1Hz,1H),6.95(s,1H),4.13(s,3H),2.35(s,3H). 1 HNMR (300 MHz, DMSO-d 6 ): 8.65 (d, J = 5.1 Hz, 1H), 8.55 (s, 1H), 8.29 (s, 1H), 7.70 (d, J = 5.1 Hz, 1H), 6.95(s, 1H), 4.13(s, 3H), 2.35(s, 3H).
(4)式(I)化合物的制备(4) Preparation of compound of formula (I)
Figure PCTCN2020070062-appb-000008
Figure PCTCN2020070062-appb-000008
将化合物5-(2-氯嘧啶-4-基)-1,3-二甲基-1H-吲哚-7-甲腈(164mg,0.58mmol,1.0eq)和化合物N-(5-氨基-2-{[2-(二甲基氨基)乙基](甲基)氨基}-4-甲氧基苯基)丙烯酰胺(170mg,0.58mmol,1.0eq)溶于2-戊醇(4mL)中,室温下分批加入一水对甲苯磺酸(123mg,0.64mmol,1.1eq),加完加热至120℃反应5小时,TLC,LCMS检测,反应完全后,降至室温,加入水(10mL),二氯甲烷/甲醇=10:1(10mL×3)萃取,有机相用盐水(10mL)洗,干燥,浓缩,柱层析提纯得式(I)化合物(48mg,收率:15%)。Compound 5-(2-chloropyrimidin-4-yl)-1,3-dimethyl-1H-indole-7-carbonitrile (164mg, 0.58mmol, 1.0eq) and compound N-(5-amino- 2-{[2-(Dimethylamino)ethyl](methyl)amino}-4-methoxyphenyl)acrylamide (170mg, 0.58mmol, 1.0eq) dissolved in 2-pentanol (4mL) Medium, p-toluenesulfonic acid monohydrate (123mg, 0.64mmol, 1.1eq) was added in batches at room temperature. After the addition, the reaction was heated to 120°C for 5 hours. TLC, LCMS detection. After the reaction was completed, it was cooled to room temperature and water (10mL) was added. ), dichloromethane/methanol=10:1 (10 mL×3) extraction, the organic phase was washed with brine (10 mL), dried, concentrated, and purified by column chromatography to obtain the compound of formula (I) (48 mg, yield: 15%) .
1HNMR(300MHz,DMSO-d 6):□10.19(br,1H),9.07(s,1H),8.71(s,1H),8.51-8.49(m,2H),8.20(s,1H),7.59-7.57(m,1H),7.32(s,1H),7.04(s,1H),6.40-6.34(m,1H),6.27-6.21(m,1H),5.75-5.72(m,1H),4.04(s,3H),3.85(s,3H),2.89-2.87(m,2H),2.71(s,3H),2.34-2.32(m,2H),2.23(s,3H),2.17(s,6H).LCMS:(M+H) +:538.8.HPLC:94.8%. 1 HNMR (300MHz, DMSO-d 6 ): □10.19 (br, 1H), 9.07 (s, 1H), 8.71 (s, 1H), 8.51-8.49 (m, 2H), 8.20 (s, 1H), 7.59 -7.57(m,1H),7.32(s,1H),7.04(s,1H),6.40-6.34(m,1H),6.27-6.21(m,1H),5.75-5.72(m,1H),4.04 (s,3H),3.85(s,3H),2.89-2.87(m,2H),2.71(s,3H),2.34-2.32(m,2H),2.23(s,3H),2.17(s,6H ). LCMS: (M + H) + : 538.8. HPLC: 94.8%.
实施例2:式(I)化合物的L-(+)-酒石酸盐晶型I的制备Example 2: Preparation of L-(+)-tartrate crystal form I of the compound of formula (I)
制备方法一:Preparation method one:
取式(I)化合物(50mg,0.093mmol)置于10mL离心管中,加入1当量L-(+)-酒石酸(13.9mg,0.093mmol),向离心管中加入乙醇(2mL),常温搅拌0.5h后,加入水(0.5mL)继续搅拌。反应结束后,过滤,干燥。所得固体经XRPD测试为晶型I。Take the compound of formula (I) (50 mg, 0.093 mmol) in a 10 mL centrifuge tube, add 1 equivalent of L-(+)-tartaric acid (13.9 mg, 0.093 mmol), add ethanol (2 mL) to the centrifuge tube, and stir at room temperature for 0.5 After h, add water (0.5 mL) and continue stirring. After the reaction, it was filtered and dried. The obtained solid was tested to form I by XRPD.
制备方法二Preparation method two
取式(I)化合物(3.0g,5.57mmol)置于100mL圆底烧瓶中,加入刚配置好的丙酮与水的混合溶剂(丙酮与水的体积比为2:1),搅拌,升温至50℃,加入L-(+)-酒石酸(836mg,5.57mmol),50℃下搅拌至反应结束,自然降温,过滤,50℃真空干燥。所得固体(3.25g,收率84.7%)经XRPD测试为晶型I。Take the compound of formula (I) (3.0g, 5.57mmol) in a 100mL round bottom flask, add the mixed solvent of acetone and water (the volume ratio of acetone and water is 2:1), stir and heat to 50 At ℃, add L-(+)-tartaric acid (836mg, 5.57mmol), stir at 50℃ until the end of the reaction, naturally cool down, filter, and vacuum dry at 50℃. The obtained solid (3.25g, yield 84.7%) was tested to form I by XRPD.
制备方法三Preparation method three
取丙酮(3mL),加入水(0.5mL)配置成体积比为6:1的丙酮/水混合溶剂,取式(I)化合物(0.1g,0.19mmol)置于10mL圆底烧瓶中,加入上述丙酮/水混合溶剂(2mL),搅拌,升温至50℃,加入L-(+)-酒石酸(27.9mg,0.19mmol),维持温度搅拌至反应结束,降温,过滤,50℃真空干燥。所得固体(93.8mg,收率73.3%)经XRPD测试为晶型I。Take acetone (3mL), add water (0.5mL) to configure an acetone/water mixed solvent with a volume ratio of 6:1, take the compound of formula (I) (0.1g, 0.19mmol) in a 10mL round bottom flask, add the above Acetone/water mixed solvent (2mL) was stirred, and the temperature was raised to 50°C. L-(+)-tartaric acid (27.9mg, 0.19mmol) was added. The temperature was maintained and stirred until the reaction was completed. The obtained solid (93.8 mg, yield 73.3%) was tested as Form I by XRPD.
制备方法四Preparation method four
取式(I)化合物的L-(+)-酒石酸盐晶型I(50mg,0.072mmol)置于10mL圆底离心管中,向离心管中加入丙酮(5mL)与水(0.5mL)的混合溶剂,50℃恒温浆洗,反应结束后,搅拌条件下降至室温,离心(4000rpm,10min),舍弃上清液,下层固体于50℃真空干燥,经XRPD测试为晶型I。Take L-(+)-tartrate salt form I (50mg, 0.072mmol) of the compound of formula (I) in a 10mL round bottom centrifuge tube, add a mixture of acetone (5mL) and water (0.5mL) to the centrifuge tube The solvent was washed at a constant temperature of 50°C. After the reaction was completed, the stirring condition was lowered to room temperature, centrifugation (4000rpm, 10min), the supernatant was discarded, and the lower solid was dried at 50°C under vacuum, and the crystal form I was tested by XRPD.
制备方法五Preparation method five
取式(I)化合物的L-(+)-酒石酸盐晶型I(50×5mg,0.072mmol)4份,分别置于10mL离心管中,4个离心管中分别加入乙腈(2mL)、乙酸乙酯(2mL)、1,4-二氧六环(2mL)和正庚烷(2mL),依次编号1-4,并分别置于20℃恒温浆洗,反应结束后,离心(4000rpm,10min),舍弃上清液,下层固体50℃真空干燥,经XRPD测试为晶型I。Take 4 parts of L-(+)-tartrate crystal form I (50×5mg, 0.072mmol) of the compound of formula (I), put them into 10mL centrifuge tubes, and add acetonitrile (2mL) and acetic acid to the 4 centrifuge tubes respectively Ethyl ester (2mL), 1,4-dioxane (2mL) and n-heptane (2mL), sequentially numbered 1-4, and placed in a constant temperature slurry wash at 20 ℃, after the reaction is completed, centrifuged (4000rpm, 10min), The supernatant was discarded, and the lower solid was dried in vacuum at 50°C, and tested to form I by XRPD.
对通过上述方法制得的晶型I,进行测定:For Form I prepared by the above method, determine:
X-射线粉末衍射测定X-ray powder diffraction measurement
本发明的晶体结构不限于提供与本申请公开的附图中所绘的X-射线粉末衍射图完全相同的X-射线粉末衍射图的晶体结构,与附图中公开的那些基本相同的X-射线粉末衍射图的任何晶体结构都包含在本发明的范围内。The crystal structure of the present invention is not limited to providing a crystal structure of an X-ray powder diffraction pattern exactly the same as the X-ray powder diffraction pattern drawn in the drawings disclosed in the present application, and X-ray powder diffraction patterns substantially the same as those disclosed in the drawings Any crystal structure of the ray powder diffraction pattern is included in the scope of the present invention.
X-射线粉末衍射测定的条件:Cu钯,Kα1
Figure PCTCN2020070062-appb-000009
1.54060,步长0.0203,每步0.3秒。
Conditions for X-ray powder diffraction measurement: Cu palladium, Kα1
Figure PCTCN2020070062-appb-000009
1.54060, step size 0.0203, 0.3 seconds per step.
使用Cu-Kα辐射,以2θ角度(°)表示的X-射线粉末衍射的晶型处有特征峰。Using Cu-Kα radiation, the X-ray powder diffracted in a crystal form expressed at a 2θ angle (°) has a characteristic peak.
式(I)化合物的L-(+)-酒石酸盐晶型I的X-射线粉末衍射图谱示于图1中,该晶型I在以下衍射角度2θ(°)处有峰:The X-ray powder diffraction pattern of the L-(+)-tartrate salt form I of the compound of formula (I) is shown in FIG. 1, and this form I has a peak at the following diffraction angle 2θ (°):
8.9±0.2°、11.2±0.2°、12.0±0.2°、12.5±0.2°、13.1±0.2°、14.7±0.2°、17.5±0.2°、19.1±0.2°、16.3±0.2°、16.6±0.2°、20.0±0.2°、20.6±0.2°、21.1±0.2°、21.6±0.2°。8.9±0.2°, 11.2±0.2°, 12.0±0.2°, 12.5±0.2°, 13.1±0.2°, 14.7±0.2°, 17.5±0.2°, 19.1±0.2°, 16.3±0.2°, 16.6±0.2°, 20.0±0.2°, 20.6±0.2°, 21.1±0.2°, 21.6±0.2°.
差示扫描量热法Differential Scanning Calorimetry
通过差示扫描量热法(DSC)研究式(I)化合物的L-(+)-酒石酸盐晶型I的固态热性能。晶型I的DSC曲线显示于图2中。The solid-state thermal properties of L-(+)-tartrate crystal form I of the compound of formula (I) were studied by differential scanning calorimetry (DSC). The DSC curve of Form I is shown in FIG. 2.
测定条件:用氮气以50毫升/分钟吹扫,在室温至225℃之间以10℃/分钟加热速率收集数据,在吸热峰朝下的情况下绘图。Measurement conditions: Purge with nitrogen at 50 ml/min, collect data at a heating rate of 10°C/min between room temperature and 225°C, and plot with the endothermic peak facing down.
在DSC测定中,根据测量参数及加热速率,实际测得的开始温度和最高温度具有一定程度的可变性。In DSC measurement, the actual measured start temperature and maximum temperature have a certain degree of variability based on the measurement parameters and heating rate.
热重分析Thermogravimetric analysis
测试条件:用氮气以60毫升/分钟吹扫,在室温至300℃之间以10℃/分钟加热速率收集数据。Test conditions: Purge with nitrogen at 60 ml/min and collect data at a heating rate of 10°C/min between room temperature and 300°C.
式(I)化合物的L-(+)-酒石酸盐晶型I的TGA曲线显示于图3中。The TGA curve of L-(+)-tartrate salt form I of the compound of formula (I) is shown in FIG. 3.
核磁分析NMR analysis
1HNMR(400MHz,DMSO-d6):δ9.84(s,1H),8.94(s,1H),8.67(d,1H),8.47(s,1H),8.46(s,1H),8.15(s,1H),7.56(d,1H),7.29(s,1H),7.00(s,1H),6.55(m,1H),6.27(m,1H),5.74(m,1H),4.08(s,2H),4.02(s,3H),3.86(s,3H),3.06(t,2H),2.73(t,2H),2.64(s,3H),2.48(s,3H),2.47(s,3H),2.27(s,3H). 1 HNMR (400MHz, DMSO-d6): δ 9.84 (s, 1H), 8.94 (s, 1H), 8.67 (d, 1H), 8.47 (s, 1H), 8.46 (s, 1H), 8.15 (s , 1H), 7.56 (d, 1H), 7.29 (s, 1H), 7.00 (s, 1H), 6.55 (m, 1H), 6.27 (m, 1H), 5.74 (m, 1H), 4.08 (s, 2H), 4.02(s, 3H), 3.86(s, 3H), 3.06(t, 2H), 2.73(t, 2H), 2.64(s, 3H), 2.48(s, 3H), 2.47(s, 3H ), 2.27(s, 3H).
式(I)化合物的L-(+)-酒石酸盐晶型I的氢谱显示于图4中。The hydrogen spectrum of L-(+)-tartrate salt form I of the compound of formula (I) is shown in FIG. 4.
实施例3:式(I)化合物L-(+)-酒石酸盐晶型II的制备Example 3: Preparation of the compound of formula (I) L-(+)-tartrate crystal form II
取式(I)化合物(50mg,0.093mmol),置于10mL圆底烧瓶中,加入L-(+)-酒石酸(13.9mg,0.093mmol),然后加入甲醇(2mL),常温搅拌1h后,加入水(0.5mL),继续搅拌23h后过滤,50℃真空干燥,固体经XRPD检测,结果显示为晶型II。Take the compound of formula (I) (50 mg, 0.093 mmol), place it in a 10 mL round bottom flask, add L-(+)-tartaric acid (13.9 mg, 0.093 mmol), and then add methanol (2 mL). After stirring at room temperature for 1 h, add Water (0.5 mL), continue stirring for 23 h, then filter, dry at 50° C. in vacuo, and the solid was detected by XRPD. The result showed that it was Form II.
X-射线粉末衍射测定X-ray powder diffraction measurement
X-射线粉末衍射测定的条件:Cu钯,Kα1
Figure PCTCN2020070062-appb-000010
1.54060,步长0.0203,每步0.3秒。
Conditions for X-ray powder diffraction measurement: Cu palladium, Kα1
Figure PCTCN2020070062-appb-000010
1.54060, step size 0.0203, 0.3 seconds per step.
使用Cu-Kα辐射,以2θ角度(°)表示的X-射线粉末衍射的晶型处有特征峰。Using Cu-Kα radiation, the X-ray powder diffracted in a crystal form expressed at a 2θ angle (°) has a characteristic peak.
式(I)化合物的L-(+)-酒石酸盐晶型II的X-射线粉末衍射图谱示于图5中,该晶型II在以下衍射角度2θ(°)处有峰:The X-ray powder diffraction pattern of the L-(+)-tartrate salt form II of the compound of formula (I) is shown in FIG. 5, which has a peak at the following diffraction angle 2θ (°):
9.0±0.2°、11.3±0.2°、12.0±0.2°、12.7±0.2°、13.9±0.2°、17.9±0.2°、19.1±0.2°、20.3±0.2°、21.2±0.2°、16.3±0.2°、17.3±0.2°、23.5±0.2°、24.4±0.2°、25.5±0.2°、25.9±0.2°。9.0±0.2°, 11.3±0.2°, 12.0±0.2°, 12.7±0.2°, 13.9±0.2°, 17.9±0.2°, 19.1±0.2°, 20.3±0.2°, 21.2±0.2°, 16.3±0.2°, 17.3±0.2°, 23.5±0.2°, 24.4±0.2°, 25.5±0.2°, 25.9±0.2°.
实施例4:式(I)化合物的EGFR及肿瘤抑制活性考察Example 4: Investigation of EGFR and tumor inhibitory activity of the compound of formula (I)
缩写:abbreviation:
DMSO 二甲亚砜DMSO
DTT 二硫苏糖醇DTT dithiothreitol
ATP 三磷酸腺苷ATP adenosine triphosphate
EDTA 乙二胺四乙酸EDTA ethylenediaminetetraacetic acid
Ki 酶抑制常数Ki enzyme inhibition constant
DMEM 达尔伯克氏改良伊格尔氏培养基DMEM Dulbecco's Modified Eagle's Medium
NCS 新生小牛血清NCS Newborn Calf Serum
PBS 磷酸盐缓冲盐水PBS phosphate buffered saline
PMSF 苯基甲烷磺酰氟PMSF phenylmethanesulfonyl fluoride
ELISA 酶联免疫吸附测定ELISA
IgG 免疫球蛋白GIgG immunoglobulin G
FBS 胎牛血清FBS fetal bovine serum
BDNF 脑衍生神经营养因子BDNF brain-derived neurotrophic factor
4.1、EGFR激酶抑制测定试验4.1. EGFR kinase inhibition test
使用所属领域的技术人员众所周知的市售测定试剂盒和服务测量本发明化合物对激酶的抑制。这些试剂盒和服务用于测量包括(但不限于)以下的多种激酶的抑制:ALK、ABL、AXL、Aur B和C、BLK、erbB-2、erbB-4.EGFR、突变EGFR、HPK、IRAK1、RON、ROS1、SLK、STK10、TIE2、TRK、c-Met、Lck、Lyn、Src、Fyn、Syk、Zap-70、Itk、Tec、Btk、EGFR、ErbB2、Kdr、Flt-1、Flt-3、Tek、c-Met、InsR和Atk。这些测定试剂盒和服务的商业供应者包括Promega公司和Reaction Biology公司、EMD Millipore和CEREP。除市售测定试剂盒和服务外,式(I-VIII)化合物的激酶抑制活性借助于下述测定测量。The inhibition of kinases by compounds of the invention is measured using commercially available assay kits and services well known to those skilled in the art. These kits and services are used to measure the inhibition of various kinases including (but not limited to): ALK, ABL, AXL, Aur B and C, BLK, erbB-2, erbB-4.EGFR, mutant EGFR, HPK, IRAK1, RON, ROS1, SLK, STK10, TIE2, TRK, c-Met, Lck, Lyn, Src, Fyn, Syk, Zap-70, Itk, Tec, Btk, EGFR, ErbB2, Kdr, Flt-1, Flt- 3. Tek, c-Met, InsR and Atk. Commercial suppliers of these assay kits and services include Promega and Reaction Biology, EMD Millipore and CEREP. In addition to commercially available assay kits and services, the kinase inhibitory activity of compounds of formula (I-VIII) is measured by means of the following assay.
4.1.1、表皮生长因子受体酪氨酸激酶的纯化4.1.1. Purification of epidermal growth factor receptor tyrosine kinase
通过以下方法从过度表达EGF受体的A431人类鳞状细胞癌细胞分离人类EGF受体酪氨酸激酶。使细胞在滚瓶中含有10%胎牛血清的50%达尔伯克氏改良伊格尔氏和50%HAM F-12营养培养基(Gibco)中生长。将大约109个细胞在两体积含有以下的缓冲液中溶解:20mM 2-(4N-[2-羟乙基]哌嗪-1-基)乙磺酸(hepes)pH 7.4、5mM乙二醇双(2-氨基乙醚)N,N,N',N'-四乙酸、1%Triton X-lOO、10%丙三醇、0.1mM原钒酸钠、5mM 氟化钠、4mM焦磷酸盐、4mM苯甲酰胺、1mM二硫苏糖醇、80μg/mL抑肽酶、40μg/mL亮抑酶肽和1mM苯甲基磺酰氟。在25,000×g下离心10分钟后,使上层清液在40℃下用10mL预先经50mM Hepes、10%丙三醇、0.1%Triton X-100和150mM NaCl pH 7.5(平衡缓冲液)平衡的小麦胚凝集索琼脂糖平衡2小时。将污染蛋白质用含1M NaCl的平衡缓冲液从树脂洗涤,并用含0.5M N-乙酰基-1-D-葡糖胺的平衡缓冲液、接着1mM尿素洗脱该酶。该酶用0.1mg/ml EGF洗脱。如通过考马斯蓝染色的聚丙烯酰胺电泳凝胶评定,受体似乎为均质的。Human EGF receptor tyrosine kinase was isolated from A431 human squamous cell carcinoma cells overexpressing EGF receptor by the following method. Cells were grown in 50% Dulbecco's modified Eagle's and 50% HAM F-12 nutrient medium (Gibco) containing 10% fetal bovine serum in roller bottles. Approximately 109 cells were dissolved in two volumes of buffer containing the following: 20 mM 2-(4N-[2-hydroxyethyl] piperazin-1-yl)ethanesulfonic acid (hepes) pH 7.4, 5 mM ethylene glycol bis (2-aminoethyl ether) N,N,N',N'-tetraacetic acid, 1% Triton X-100, 10% glycerin, 0.1mM sodium orthovanadate, 5mM sodium fluoride, 4mM pyrophosphate, 4mM Benzamide, 1 mM dithiothreitol, 80 μg/mL aprotinin, 40 μg/mL leupeptin, and 1 mM benzylsulfonyl fluoride. After centrifugation at 25,000×g for 10 minutes, the supernatant was equilibrated with 10 mL of wheat pre-equilibrated with 50 mM Hepes, 10% glycerol, 0.1% Triton X-100 and 150 mM NaCl pH 7.5 (equilibration buffer) at 40°C. The embryo agglutination agarose was equilibrated for 2 hours. The contaminated protein was washed from the resin with an equilibrium buffer containing 1M NaCl, and the enzyme was eluted with an equilibrium buffer containing 0.5M N-acetyl-1-D-glucosamine, followed by 1 mM urea. The enzyme was eluted with 0.1 mg/ml EGF. As assessed by Coomassie blue stained polyacrylamide electrophoresis gel, the receptor appears to be homogeneous.
使用与前一段中所述相同的技术,可以从含表皮生长因子受体的适当细胞系分离表皮生长因子受体的各种突变形式。举例来说,EGFRdel746-750突变蛋白质可以从PC-9细胞提取,且L858R/T790M双重突变EGFR蛋白质可以从H1975细胞分离。Using the same technique as described in the previous paragraph, various mutant forms of epidermal growth factor receptor can be isolated from appropriate cell lines containing epidermal growth factor receptor. For example, the EGFRdel746-750 mutant protein can be extracted from PC-9 cells, and the L858R/T790M double mutant EGFR protein can be isolated from H1975 cells.
4.1.2、单突变体EGFR_d746-750的IC 50值的测定 4.1.2. Determination of IC 50 value of single mutant EGFR_d746-750
用于测定IC 50的酶测定在25μL总体积中进行。在100%DMSO中稀释所有化合物至500μM储备溶液,并连续稀释4倍,达成10个剂量。“Max”和“Min”对照含有100%DMSO。“Max”代表无酶的DMSO对照,“Min”代表无化合物的低对照。将10μl化合物转移至90μl 1x激酶基础缓冲液,进行中间稀释。将5μl中间稀释化合物转移至384孔测定板,接着将10μl含有(12.5nM EGFR_d746-750、5mMDTT、1x激酶基础缓冲液)的2.5x酶缓冲液添加至测定板。在室温下孵育10分钟且添加10μl含有(7.5μM肽、35μM ATP、25mM MgCl 2、1x激酶基础缓冲液)的2.5x底物缓冲液,以起始反应。在室温下孵育1小时且添加25μl终止缓冲液来结束反应。从Caliper收集转化数据,并从Caliper程序收集转化数据。在XLfit中拟合数据以获得IC 50值。 The enzyme assay used to determine IC 50 was performed in a total volume of 25 μL. All compounds were diluted to a 500 μM stock solution in 100% DMSO and serially diluted 4 times to achieve 10 doses. The "Max" and "Min" controls contain 100% DMSO. "Max" represents the DMSO control without enzyme, and "Min" represents the low control without compound. 10 μl of the compound was transferred to 90 μl of 1x kinase base buffer for intermediate dilution. 5 μl of the intermediate dilution compound was transferred to a 384-well assay plate, and then 10 μl of 2.5x enzyme buffer containing (12.5nM EGFR_d746-750, 5mMDTT, 1x kinase base buffer) was added to the assay plate. Incubate for 10 minutes at room temperature and add 10 μl of 2.5x substrate buffer containing (7.5 μM peptide, 35 μM ATP, 25 mM MgCl 2 , 1x kinase base buffer) to start the reaction. Incubate for 1 hour at room temperature and add 25 μl stop buffer to end the reaction. Collect conversion data from Caliper, and collect conversion data from the Caliper program. Data fitting to obtain IC50 values XLfit IC.
4.1.3、双重突变EGFR(EGFR_T790M/L858R)的IC 50值的测定 4.1.3. Determination of IC 50 value of double mutant EGFR (EGFR_T790M/L858R)
用于测定IC 50的酶测定在25μL总体积中进行。在100%DMSO中稀释所有化合物至500μM储备溶液,且连续稀释4倍,达成10个剂量。“Max”和“Min”对照含有100%DMSO。“Max”代表无酶的DMSO对照,“Min”代表无化合物的低对照。将10μl化合物转移至90μl 1x激酶基础 缓冲液,进行中间稀释。将5μl中间稀释化合物转移至384孔测定板,接着将10μl含有(25nM EGFR_T790M/L858R、5mM DTT、1x激酶基础缓冲液)的2.5x酶缓冲液添加至测定板。在室温下孵育10分钟且添加10μl含有(7.5μM肽、47.5μM ATP、25mMMgCl 2、1x激酶基础缓冲液)的2.5x底物缓冲液,以起始反应。在室温下孵育1小时且添加25μl终止缓冲液来结束反应。从Caliper收集转化数据,并从Caliper程序收集转化数据。在XLfit中拟合数据以获得IC 50值。 The enzyme assay used to determine IC 50 was performed in a total volume of 25 μL. All compounds were diluted to a 500 μM stock solution in 100% DMSO and serially diluted 4 times to achieve 10 doses. The "Max" and "Min" controls contain 100% DMSO. "Max" represents the DMSO control without enzyme, and "Min" represents the low control without compound. 10 μl of the compound was transferred to 90 μl of 1x kinase base buffer for intermediate dilution. 5 μl of the intermediate dilution compound was transferred to a 384-well assay plate, and then 10 μl of 2.5x enzyme buffer containing (25 nM EGFR_T790M/L858R, 5 mM DTT, 1x kinase base buffer) was added to the assay plate. Incubate at room temperature for 10 minutes and add 10 μl of 2.5x substrate buffer containing (7.5 μM peptide, 47.5 μM ATP, 25 mMMgCl 2 , 1x kinase base buffer) to start the reaction. Incubate for 1 hour at room temperature and add 25 μl stop buffer to end the reaction. Collect conversion data from Caliper, and collect conversion data from the Caliper program. Data fitting to obtain IC50 values XLfit IC.
4.1.4、wt EGFR的IC 50值的测定 4.1.4. Determination of IC 50 value of wt EGFR
用于测定IC 50的酶测定在25μL总体积中进行。在100%DMSO中稀释所有化合物至500μM储备溶液,且连续稀释4倍,达成10个剂量。“Max”和“Min”对照含有100%DMSO。“Max”代表无酶的DMSO对照,“Min”代表无化合物的低对照。将10μl化合物转移至90μl 1x激酶基础缓冲液,进行中间稀释。将5μl中间稀释化合物转移至384孔测定板,接着将10μl含有(20nM EGFR、5mM DTT、1x激酶基础缓冲液)的2.5x酶缓冲液添加至测定板。在室温下孵育10分钟且添加10μl含有(7.5μM肽、5.75μM ATP、25mM MgCl2、25mMMnCl 2、1x激酶基础缓冲液)的2.5x底物缓冲液,以起始反应。在室温下孵育1小时且添加25μl终止缓冲液来结束反应。从Caliper收集转化数据,并从Caliper程序收集转化数据。在XLfit中拟合数据以获得IC 50值。 The enzyme assay used to determine IC 50 was performed in a total volume of 25 μL. All compounds were diluted to a 500 μM stock solution in 100% DMSO and serially diluted 4 times to achieve 10 doses. The "Max" and "Min" controls contain 100% DMSO. "Max" represents the DMSO control without enzyme, and "Min" represents the low control without compound. 10 μl of the compound was transferred to 90 μl of 1x kinase base buffer for intermediate dilution. 5 μl of the intermediate dilution compound was transferred to a 384-well assay plate, and then 10 μl of 2.5x enzyme buffer containing (20 nM EGFR, 5 mM DTT, 1x kinase base buffer) was added to the assay plate. Incubate at room temperature for 10 minutes and add 10 μl of 2.5x substrate buffer containing (7.5 μM peptide, 5.75 μM ATP, 25 mM MgCl 2 , 25 mM MnCl 2 , 1x kinase base buffer) to start the reaction. Incubate for 1 hour at room temperature and add 25 μl of stop buffer to end the reaction. Conversion data is collected from Caliper, and conversion data is collected from the Caliper program. Data fitting to obtain IC50 values XLfit IC.
4.2、EGFR细胞抗增殖试验4.2 Anti-proliferation test of EGFR cells
供试品testing sample
式(I)化合物,按照实施例中的方法制备;化合物AZD9291结构如下,按照现有技术方法制备。The compound of formula (I) is prepared according to the method in the examples; the structure of compound AZD9291 is as follows and prepared according to the prior art method.
Figure PCTCN2020070062-appb-000011
Figure PCTCN2020070062-appb-000011
试验步骤experiment procedure
4.2.1、H1975抑制测定(细胞增殖)4.2.1, H1975 inhibition assay (cell proliferation)
将H1975细胞低温保存在液氮中。在细胞解冻前,将15mL细胞培养基(供有10%胎牛血清和1%青霉素/链霉素的RPMI 1640培养基)置于T75烧瓶中且在湿润的37℃/5%CO 2孵育箱中预先孵育烧瓶15分钟以使培养基平衡至适当pH值和温度。从液氮移出小瓶,并通过在缓和搅拌下置放于水浴中37℃下1-2分钟而快速解冻,然后通过用70%乙醇擦拭来去污,接着在II级生物安全柜中打开。将小瓶内含物逐滴转移至无菌15mL锥形管中的10mL细胞培养基中。接着将管在200×g下离心5分钟,且吸出上层清液。将细胞小球用1mL新鲜细胞培养基再次悬浮,且将其转移至含有细胞培养基的T75烧瓶中。 The H1975 cells were cryopreserved in liquid nitrogen. Before thawing the cells, 15 mL of cell culture medium (RPMI 1640 medium supplied with 10% fetal bovine serum and 1% penicillin/streptomycin) was placed in a T75 flask and incubated in a humidified 37°C/5% CO 2 incubator Incubate the flask in advance for 15 minutes to equilibrate the medium to the proper pH and temperature. The vial was removed from the liquid nitrogen and quickly thawed by placing in a water bath at 37°C for 1-2 minutes with gentle stirring, then decontaminated by wiping with 70% ethanol, and then opened in a Class II biosafety cabinet. The contents of the vial were transferred dropwise to 10 mL of cell culture medium in a sterile 15 mL conical tube. The tube was then centrifuged at 200×g for 5 minutes, and the supernatant was aspirated. The cell pellet was resuspended with 1 mL of fresh cell culture medium, and transferred to a T75 flask containing cell culture medium.
为使H1975细胞传代,首先,将粘附细胞以胰蛋白酶/EDTA清洗。接着添加胰蛋白酶/EDTA(T75烧瓶3mL)至烧瓶且旋动以保证细胞均匀涂布胰蛋白酶。接着在37℃下孵育烧瓶,直到细胞脱离。添加相等体积的细胞培养基来终止反应。收集脱离的细胞并在200×g下离心5分钟,接着再次悬浮在新鲜培养基中。接着,将细胞转移至含有细胞培养基的新T75烧瓶中。细胞每周在培养基中以1:2或1:4的比率传代培养三次。To passage H1975 cells, first, adherent cells were washed with trypsin/EDTA. Then trypsin/EDTA (3 mL in T75 flask) was added to the flask and swirled to ensure that the cells were evenly coated with trypsin. The flask was then incubated at 37°C until the cells detached. Add an equal volume of cell culture medium to stop the reaction. The detached cells were collected and centrifuged at 200×g for 5 minutes, and then resuspended in fresh medium. Next, the cells were transferred to a new T75 flask containing cell culture medium. The cells were subcultured in the medium at a ratio of 1:2 or 1:4 three times a week.
将测试化合物以30mM溶解于DMSO中。将45μL化合物转移至384孔化合物来源板(LABCYTE目录号P-05525)中且以1:3比率连续稀释,产生13点稀释液。采取相同体积的DMSO作为高对照。20nL这些化合物DMSO稀释液(10点,从1.11mM至0.056μM)通过Echo 550分配至新的384孔测定板中。The test compound was dissolved in DMSO at 30 mM. 45 μL of compound was transferred to a 384-well compound source plate (LABCYTE catalog number P-05525) and serially diluted at a ratio of 1:3, resulting in a 13-point dilution. Take the same volume of DMSO as a high control. 20 nL of these compound DMSO dilutions (10 points, from 1.11 mM to 0.056 μM) were dispensed into new 384-well assay plates by Echo 550.
从烧瓶收获细胞至如上所述的细胞培养基并使用自动化细胞计数器(Thermo Fisher Scientific,Countess TM)计算细胞数。用培养基稀释细胞成25,000个细胞/毫升,并添加40μL细胞悬浮液至如所指明的384孔细胞培养板的每个孔中。最终浓度为1,000个细胞/孔。添加仅仅培养基作为低对照。该板被盖覆盖且置于37℃/5%CO 2孵育箱中72小时。 The cells were harvested from the flask to the cell culture medium as described above and the cell count was calculated using an automated cell counter (Thermo Fisher Scientific, Countess ). Dilute the cells with culture medium to 25,000 cells/ml and add 40 μL of cell suspension to each well of the 384-well cell culture plate as indicated. The final concentration is 1,000 cells/well. Add medium alone as a low control. The plate was covered with a lid and placed in a 37°C/5% CO 2 incubator for 72 hours.
孵育72小时后,从孵育箱移出板且在室温下平衡15分钟。在实验前在37℃下孵育CellTiter Glo试剂(Promega,G9243)。使缓冲液平衡至 室温且用于溶解基质。为测定细胞存活力,添加40μLCellTiter-Glo试剂至待检测的每个孔(与培养基呈1:1)。接着板置于室温下30分钟,接着在EnSpire(PerkinElmer)上读取。After 72 hours of incubation, the plate was removed from the incubator and equilibrated at room temperature for 15 minutes. CellTiter Glo reagent (Promega, G9243) was incubated at 37°C before the experiment. The buffer was equilibrated to room temperature and used to dissolve the matrix. To determine cell viability, add 40 μL of CellTiter-Glo reagent to each well to be tested (1:1 with culture medium). The plate was then placed at room temperature for 30 minutes, and then read on EnSpire (PerkinElmer).
为估计IC 50,发光读数通过应用以下方程式转变成抑制%∶(Lum HC-Lum Cpd)/(Lum HC-Lum LC)。接着通过XLFit中拟合成四参数对数曲线,来计算IC 50To estimate the IC 50 , the luminescence reading is converted to% inhibition by applying the following equation: (Lum HC -Lum Cpd )/(Lum HC -Lum LC ). Logarithmic curve, then IC 50 was calculated by four-parameter fit to the XLFit.
4.2.2、PC-9生长抑制测定(细胞增殖)4.2.2, PC-9 growth inhibition assay (cell proliferation)
PC-9细胞的抑制测定以与针对H1975细胞所述相同的方式进行。The inhibition of PC-9 cells was measured in the same manner as described for H1975 cells.
4.2.3、A431抑制测定(细胞增殖).4.2.3, A431 inhibition assay (cell proliferation).
A431细胞的培养基为供有10%胎牛血清和1%青霉素/链霉素的达尔伯克氏改良伊格尔氏培养基。用于A431测定的DMSO稀释液为从30nM至1.52uM的10点。程序其余部分以与针对H1975细胞所述相同的方式进行。The medium for A431 cells was Dulbecco's modified Eagle's medium supplied with 10% fetal bovine serum and 1% penicillin/streptomycin. The DMSO dilution used for A431 determination is 10 points from 30nM to 1.52uM. The rest of the procedure was performed in the same manner as described for H1975 cells.
试验结果test results
表1细胞增值试验结果Table 1 Cell proliferation test results
Figure PCTCN2020070062-appb-000012
Figure PCTCN2020070062-appb-000012
PC9细胞含有EGFR d746-750单突变体(SM)。H1975细胞含有EGFR L858R/T790M双重突变体(DM)。A431细胞含有未突变的EGFR野生型(WT)。PC9 cells contain EGFR d746-750 single mutant (SM). H1975 cells contain EGFR L858R/T790M double mutant (DM). A431 cells contain unmutated EGFR wild type (WT).
试验结论Test Conclusions
本发明式(I)化合物具有优异的EGFR抑制活性和抗肿瘤增殖活性,尤其是对EGFR突变体具有较高的抑制活性和选择性。The compound of formula (I) of the present invention has excellent EGFR inhibitory activity and anti-tumor proliferation activity, especially high inhibitory activity and selectivity for EGFR mutants.
4.3、细胞EGFR自身磷酸化测定试验4.3. Cell EGFR autophosphorylation test
供试品testing sample
式(I)化合物,按照实施例中的方法制备;化合物AZD9291,按照现有技术方法制备。The compound of formula (I) is prepared according to the method in the examples; the compound AZD9291 is prepared according to the prior art method.
试验步骤experiment procedure
L858R/T790M双重突变体H1975自身磷酸化抑制测定(ELISA)L858R/T790M double mutant H1975 autophosphorylation inhibition assay (ELISA)
将H1975细胞低温保存在液氮中。在细胞解冻前,将15mL细胞培养基(供有10%胎牛血清和1%青霉素/链霉素的RPMI 1640培养基)置于T75烧瓶中且在湿润的37℃/5%CO 2孵育箱中预先孵育烧瓶15分钟以使培养基平衡至适当pH值和温度。从液氮移出小瓶,并通过在缓和搅拌下置放于水浴中37℃下1-2分钟而快速解冻,然后通过用70%乙醇擦拭来去污,接着在II级生物安全柜中打开。将小瓶内含物逐滴转移至无菌15mL锥形管中的10mL细胞培养基中。接着将管在200×g下离心5分钟,且吸出上层清液。将细胞小球用1mL新鲜细胞培养基再次悬浮,且将其转移至含有细胞培养基的T75烧瓶中。 The H1975 cells were cryopreserved in liquid nitrogen. Before thawing the cells, 15 mL of cell culture medium (RPMI 1640 medium supplied with 10% fetal bovine serum and 1% penicillin/streptomycin) was placed in a T75 flask and incubated in a humidified 37°C/5% CO 2 incubator Incubate the flask in advance for 15 minutes to equilibrate the medium to the proper pH and temperature. The vial was removed from the liquid nitrogen and quickly thawed by placing in a water bath at 37°C for 1-2 minutes with gentle stirring, then decontaminated by wiping with 70% ethanol, and then opened in a Class II biosafety cabinet. The contents of the vial were transferred dropwise to 10 mL of cell culture medium in a sterile 15 mL conical tube. The tube was then centrifuged at 200×g for 5 minutes, and the supernatant was aspirated. The cell pellet was resuspended with 1 mL of fresh cell culture medium, and transferred to a T75 flask containing cell culture medium.
为使H1975细胞传代,首先,将粘附细胞以胰蛋白酶/EDTA清洗。接着添加胰蛋白酶/EDTA(T75烧瓶3mL)至烧瓶且旋动以保证细胞均匀涂布胰蛋白酶。接着在37℃下孵育烧瓶,直到细胞脱离。添加相等体积的细胞培养基来终止反应。收集脱离的细胞且在200×g下离心5分钟,接着再次悬浮在新鲜培养基中。接着,将细胞转移至含有细胞培养基的新T75烧瓶中。细胞每周在培养基中以1:4的比率传代培养三次。To passage H1975 cells, first, adherent cells were washed with trypsin/EDTA. Then trypsin/EDTA (3 mL in T75 flask) was added to the flask and swirled to ensure that the cells were evenly coated with trypsin. The flask was then incubated at 37°C until the cells detached. Add an equal volume of cell culture medium to stop the reaction. The detached cells were collected and centrifuged at 200×g for 5 minutes, and then resuspended in fresh medium. Next, the cells were transferred to a new T75 flask containing cell culture medium. Cells were subcultured in the medium at a ratio of 1:4 three times a week.
从烧瓶收获细胞至细胞培养基且使用自动化细胞计数器(ThermoFisher Scientific,Countess TM)计算细胞数。用培养基稀释细胞成250,000个细胞/毫升,且添加40μL细胞悬浮液至如所指明的384孔细胞培养板的每个孔中。最终浓度为10,000个细胞/孔。板被盖覆盖且置于37℃/5%CO2孵育箱中过夜以粘附细胞。 The cells were harvested from the flask to the cell culture medium and the number of cells was calculated using an automated cell counter (ThermoFisher Scientific, Countess ). The cells were diluted with culture medium to 250,000 cells/ml, and 40 μL of cell suspension was added to each well of the 384-well cell culture plate as indicated. The final concentration is 10,000 cells/well. The plate was covered with a lid and placed in a 37°C/5% CO2 incubator overnight to adhere the cells.
第二天,将测试化合物以10mM溶解于DMSO中。将45uL化合物转移至384孔化合物来源板(LABCYTE目录号P-05525)中且以1:3比率连续稀释,产生13点稀释液。采取相同体积的DMSO作为高对照。40nL这些化合物DMSO稀释液(11点,从1.11mM至0.019uM)通过Echo 550分配至H1975细胞板中。The next day, the test compound was dissolved in DMSO at 10 mM. 45uL of compound was transferred to a 384-well compound source plate (LABCYTE catalog number P-05525) and serially diluted at a ratio of 1:3, resulting in a 13-point dilution. Take the same volume of DMSO as a high control. 40 nL of these compound DMSO dilutions (11 points, from 1.11 mM to 0.019 uM) were dispensed into H1975 cell plates by Echo 550.
将孵育板放回37℃/5%CO 2孵育箱2小时。用冰冷HBSS替换每个孔的培养基。接着去除HBSS,添加30μL细胞溶解缓冲液至每个孔并在板式震荡器上震荡板30分钟。在1,000rpm下离心5分钟以去除气泡且 转移25uL溶解产物上层清液以通过使用商业ELISA(R&D,DYC1095B-5)进行p-EGFR测定。 Return the incubation plate to the 37°C/5% CO 2 incubator for 2 hours. Replace the medium in each well with ice cold HBSS. Next, HBSS was removed, 30 μL of cell lysis buffer was added to each well and the plate was shaken on a plate shaker for 30 minutes. Centrifuge at 1,000 rpm for 5 minutes to remove air bubbles and transfer 25 uL of lysate supernatant to perform p-EGFR assay by using a commercial ELISA (R&D, DYC1095B-5).
为估计IC50,吸收读数通过应用以下方程式转变成相对活性%∶抑制%=(Abs HC-Abs cpd)/Abs HC。接着通过XLFit(IDBS,Guildford,Surrey)中拟合成四参数对数曲线,来计算IC 50To estimate the IC50, the absorption reading is converted to relative activity% by applying the following equation:% inhibition = (Abs HC- Abs cpd )/Abs HC . Logarithmic curve, then IC 50 was calculated by four-parameter fit to XLFit (IDBS, Guildford, Surrey) are.
4.3.1、野生型EGFR A431自身磷酸化抑制测定(ELISA)4.3.1. Wild type EGFR A431 autophosphorylation inhibition assay (ELISA)
A431细胞的培养基为供有10%胎牛血清和1%青霉素/链霉素的达尔伯克氏改良伊格尔氏培养基。用于A431测定的DMSO稀释液为从10mM至0.17uM的11点。用测试化合物处理2小时后,添加4.5μL EGF(1μg/mL)至每个孔并刺激10分钟。程序其余部分以与针对H1975细胞所述相同的方式进行。The medium for A431 cells was Dulbecco's modified Eagle's medium supplied with 10% fetal bovine serum and 1% penicillin/streptomycin. The DMSO dilution used for the A431 measurement is 11 points from 10 mM to 0.17 uM. After 2 hours of treatment with the test compound, 4.5 μL of EGF (1 μg/mL) was added to each well and stimulated for 10 minutes. The rest of the procedure was performed in the same manner as described for H1975 cells.
4.3.2、Exonl9缺失EGFR(活化单突变体)PC-9细胞自身磷酸化测定4.3.2 Exonl9 deletion EGFR (activated single mutant) PC-9 cell autophosphorylation assay
人类肺细胞系PC9(Exon 19缺失EGFR)是从美国典型菌种保藏中心获得。将PC 9细胞维持在含有10%胎牛血清和2mM谷氨酰胺的RPMI1640中。使细胞在湿润孵育箱中在37℃与5%CO 2下生长。测量细胞溶解产物中内源性p-EGFR的细胞磷酸化的测定是根据R&D Systems DuoSet IC人类磷酸化-EGF R ELISA(R&D Systems目录号#DYCI095)中所述的方案进行。将40μL细胞接种(10000个细胞/孔)于Coming黑色透明底384孔板中的生长培养基中,且在37℃与5%CO 2下孵育过夜。使用Echo 555声学上给与细胞,其中化合物在100%DMSO中连续稀释。板再孵育2小时,接着在吸出培养基后,将40μL x溶解缓冲液添加至每个孔。Greiner黑色高结合384孔板用捕捉气体涂布,然后用3%BSA阻断。在去除阻断后,将15μL溶解产物转移至Greiner黑色高结合384孔板且孵育2小时。在吸出和用PBS洗涤板后,添加20μL检测抗体并孵育2小时。在吸出和用PBS洗涤板后,添加20μL QuantaBlu荧光过氧化物酶底物(Thermo FisherScientific目录号15169)并孵育1小时。将20μL QuantaBlu终止溶液添加至板且在Envision板式读数器上使用激发352nm波长和发射460nm波长读取荧光。将由每种化合物得到的数据输入适合软件包(例如Origin)中以执行曲线拟合分析。由此数据,通过计 算得到50%作用所需的化合物浓度,确定IC 50值。 The human lung cell line PC9 (Exon 19 lacks EGFR) was obtained from the American Type Culture Collection. PC 9 cells were maintained in RPMI1640 containing 10% fetal bovine serum and 2 mM glutamine. The cells were grown in a humidified incubator at 37°C and 5% CO 2 . The assay for measuring cell phosphorylation of endogenous p-EGFR in cell lysates was performed according to the protocol described in the R&D Systems DuoSet IC Human Phosphorylation-EGF R ELISA (R&D Systems catalog number #DYCI095). 40 μL of cells were seeded (10,000 cells/well) in growth medium in a Coming black transparent bottom 384-well plate, and incubated at 37° C. and 5% CO 2 overnight. The cells were acoustically administered using Echo 555, where the compound was serially diluted in 100% DMSO. The plate was incubated for another 2 hours, then after aspirating the medium, 40 μL x lysis buffer was added to each well. Greiner black high-binding 384-well plates were coated with capture gas and then blocked with 3% BSA. After removing the block, 15 μL of lysate was transferred to Greiner black high binding 384-well plate and incubated for 2 hours. After aspirating and washing the plate with PBS, 20 μL of detection antibody was added and incubated for 2 hours. After aspirating and washing the plate with PBS, 20 μL of QuantaBlu fluorescent peroxidase substrate (Thermo Fisher Scientific Cat. No. 15169) was added and incubated for 1 hour. 20 μL of QuantaBlu stop solution was added to the plate and fluorescence was read on the Envision plate reader using excitation wavelength of 352 nm and emission wavelength of 460 nm. The data obtained from each compound is input into a suitable software package (eg Origin) to perform curve fitting analysis. From this data, the concentration of the compound required for 50% action was calculated to determine the IC 50 value.
试验结果test results
表2细胞EGFR自身磷酸化测定试验结果Table 2 Cell EGFR autophosphorylation test results
Figure PCTCN2020070062-appb-000013
Figure PCTCN2020070062-appb-000013
实验结论Experimental results
本发明式(I)化合物可有效抑制EGFR的自身磷酸化,且能够有效抑制EGFR突变体的自身磷酸化,从而抑制EGFR及其突变体的过表达,进而抑制肿瘤增殖。The compound of formula (I) of the present invention can effectively inhibit the autophosphorylation of EGFR, and can effectively inhibit the autophosphorylation of EGFR mutants, thereby inhibiting the overexpression of EGFR and its mutants, and thereby inhibiting tumor proliferation.
实施例5:式(I)化合物L-(+)-酒石酸盐晶型I的稳定性考察Example 5: Investigation of the stability of the crystalline form I of compound L-(+)-tartrate of formula (I)
供试品:testing sample:
式(I)化合物L-(+)-酒石酸盐晶型I,按照实施例中的方法制备。The compound L-(+)-tartrate crystal form I of formula (I) is prepared according to the method in the examples.
考察条件:Inspection conditions:
考察条件一:将供试品分别在60℃-开口、40℃-开口、25℃ RH75%开口、紫外光照-开口条件下放置,分别于第5、10天及满足总照度≥1.2×10 6Lux·h,近紫外能量≥200w·h/m 2条件取样,测定有关物质含量,于第10天增加XRD考察,与0天的样品进行比较。 Investigation condition 1: Put the test products under the conditions of 60℃-opening, 40℃-opening, 25℃RH75% opening, ultraviolet light-opening, respectively, on the 5th and 10th day and satisfy the total illuminance ≥1.2×10 6 Lux·h, near ultraviolet energy ≥200w·h/m 2 samples were taken to determine the content of related substances, XRD inspection was added on the 10th day, and compared with the 0 day sample.
有关物质测定:按照《中国药典》2015年版四部通则0512高效液相色谱法测定。Determination of related substances: According to the "Chinese Pharmacopoeia" 2015 edition of the four general rules 0512 high performance liquid chromatography.
XRD测定:按照《中国药典》2015年版四部通则0981第二法X射线粉末衍射法测定。XRD measurement: According to the "Chinese Pharmacopoeia" 2015 edition of four general principles 0981 second method X-ray powder diffraction method.
试验结果:test results:
表3晶型I的稳定性考察结果Table 3 Results of the investigation of the stability of Form I
Figure PCTCN2020070062-appb-000014
Figure PCTCN2020070062-appb-000014
Figure PCTCN2020070062-appb-000015
Figure PCTCN2020070062-appb-000015
*:总照度≥1.2×10 6Lux·h,近紫外能量≥200w·h/m 2 *: Total illuminance ≥1.2×10 6 Lux·h, near ultraviolet energy ≥200w·h/m 2
试验结论:Test Conclusions:
式(I)化合物的L-(+)-酒石酸盐晶型I,在上述高温高湿条件下放置10天以及在满足紫外光照度的条件下放置,样品的有关物质及晶型XRD图谱均没有明显变化。晶型I稳定性较好,利于药品的制备、运输和储藏,基本满足成药性的需求。The L-(+)-tartrate crystalline form I of the compound of formula (I), when placed under the above high-temperature and high-humidity conditions for 10 days and under conditions satisfying ultraviolet illuminance, the related substances and crystalline form XRD patterns of the samples are not obvious Variety. Crystal form I has good stability, which is beneficial to the preparation, transportation and storage of medicines, and basically meets the needs of medicines.
实施例6:晶型I的引湿性考察Example 6: Investigation of the hygroscopicity of Form I
供试品:式(I)化合物的L-(+)-酒石酸盐晶型I。Test article: L-(+)-tartrate crystal form I of the compound of formula (I).
实验方法:experimental method:
1、取干燥的具塞玻璃称量瓶,将称量瓶敞口,与瓶盖同置于25℃±1℃的恒温干燥器(下部放置氯化铵饱和溶液,相对湿度为80%±2%)内,放置24小时,盖好称量瓶盖,精密称定重量(m 1)。 1. Take a dry glass weighing bottle with a stopper, open the weighing bottle, and place it in the thermostat drier at 25°C±1°C with the cap (the lower part is a saturated ammonium chloride solution, the relative humidity is 80%±2 %), place for 24 hours, close the weighing bottle cap, and accurately weigh the weight (m 1 ).
2.取本品适量,平铺于上述称量瓶中,加盖精密称定重量(m 2)。 2. Take an appropriate amount of this product, lay it flat in the above weighing bottle, and cover with a precision weighing weight (m 2 ).
3.将称量瓶敞口,与瓶盖同置于上述恒温恒湿(25℃±1℃,RH80%±2%)条件下24小时。3. Open the weighing bottle and place it under the constant temperature and constant humidity (25℃±1℃, RH80%±2%) condition for 24 hours with the bottle cap.
4.盖好称量瓶盖,精密称定重量(m 3)。 4. Close the weighing bottle cap and accurately weigh the weight (m 3 ).
实验结果:Experimental results:
表4晶型I的引湿性考察结果Table 4 Results of the investigation of the hygroscopicity of Form I
供试品testing sample 放置条件Placement conditions 增重百分率(%) * Weight gain percentage (%) *
晶型IForm I 25℃±1℃、RH80%±2%放置1天25℃±1℃, RH80%±2%, leave for 1 day 0.110.11
*:
Figure PCTCN2020070062-appb-000016
*:
Figure PCTCN2020070062-appb-000016
实验结论:Experimental results:
本发明式(I)化合物的L-(+)-酒石酸盐晶型I几乎无引湿性,性状良好,便于药品的生产、制剂的制备、运输和储藏,有利于保证药品的稳定性和安全性。The L-(+)-tartrate crystal form I of the compound of formula (I) of the present invention has almost no hygroscopicity and good properties, which is convenient for the production of medicines, preparation, transportation and storage of preparations, which is conducive to ensuring the stability and safety of medicines .
实施例7:本发明化合物的SD雌性大鼠体内药代动力学实验Example 7: In vivo pharmacokinetic experiment of SD female rats of the compound of the present invention
供试品:式(I)化合物的L-(+)-酒石酸盐晶型I。Test article: L-(+)-tartrate crystal form I of the compound of formula (I).
受试动物:雌性SD大鼠,6只/给药途径,体重204-233g/只。Test animals: female SD rats, 6/administration route, body weight 204-233 g/animal.
供试品溶液制备:Preparation of test solution:
溶解方案:Dissolution scheme:
化合物IV给药:5%DMSO+10%PEG400+85%(28%HP-β-CD);Compound IV administration: 5% DMSO + 10% PEG400 + 85% (28% HP-β-CD);
化合物PO给药:2%HPC+0.1%吐温80;Compound PO administration: 2% HPC + 0.1% Tween 80;
空白溶媒1的配制(可根据实际需要调整配制体积):Preparation of blank solvent 1 (the preparation volume can be adjusted according to actual needs):
28%HP-β-CD溶液配制方法:称取HP-β-CD约28g,加入灭菌注射用水至终体积100mL,搅拌溶解。Preparation method of 28% HP-β-CD solution: Weigh about 28g of HP-β-CD, add sterile water for injection to a final volume of 100mL, and stir to dissolve.
空白溶媒2的配制(可根据实际需要调整配制体积):Preparation of blank solvent 2 (the preparation volume can be adjusted according to actual needs):
2%HPC+0.1%吐温80的配制:称取HPC(羟丙基纤维素)约2g,加入灭菌注射用水至终体积100mL,再加入100μL吐温80,混匀。Preparation of 2% HPC + 0.1% Tween 80: Weigh about 2 g of HPC (hydroxypropyl cellulose), add sterile water for injection to a final volume of 100 mL, then add 100 μL of Tween 80, and mix.
具体配制方法:Specific preparation method:
①称取适量供试品(供试品实际称样量=折算系数×供试品理论需要量),加入适量的2%HPC+0.1%吐温80,利用分散乳化机将其分散均匀,定容,作为SD雌性大鼠灌胃给药药液。① Weigh an appropriate amount of test sample (the actual sample size of the test sample = conversion factor × theoretical requirement of the test sample), add an appropriate amount of 2% HPC + 0.1% Tween 80, use a dispersing emulsifier to disperse it uniformly, set Rong, as an SD female rat by intragastric administration of the drug solution.
②称取适量供试品(供试品实际称样量=折算系数×供试品理论需要量),加入适量的DMSO和PEG400,涡旋,再加入适量的28%HP-β-CD定容至终体积,使终体积中DMSO:PEG400:28%HP-β-CD的体积比为5:10:85,50℃水浴保温30min,用0.22μm PTFE滤器过滤,作为SD雌性大鼠静脉给药药液。② Weigh an appropriate amount of test sample (the actual sample size of the test sample = conversion factor × theoretical requirement of the test sample), add the appropriate amount of DMSO and PEG400, vortex, and then add the appropriate amount of 28% HP-β-CD To the final volume, the volume ratio of DMSO: PEG400: 28% HP-β-CD in the final volume was 5:10:85, and the temperature was kept in a 50°C water bath for 30 minutes, filtered with a 0.22 μm PTFE filter, and administered intravenously as SD female rats Liquid medicine.
实验方法experimental method
给药:Administration:
将供试品药液按照下表5方法给药:The test drug solution is administered according to the method in Table 5 below:
表5table 5
Figure PCTCN2020070062-appb-000017
Figure PCTCN2020070062-appb-000017
血样采集:动物于颈静脉采集全血(约0.3mL)用于药代分析。Blood sample collection: Animals collect whole blood (about 0.3 mL) in the jugular vein for pharmacokinetic analysis.
IV给药动物采血时间点:给药前、给药后2min、10min、30min、1h、2h、4h、8h、24h。Time point of blood collection for IV administration animals: before administration, 2min, 10min, 30min, 1h, 2h, 4h, 8h, 24h after administration.
PO给药动物采血时间点:给药前、给药后15min、30min、1h、2h、4h、8h、24h、48h、72h。Time point of blood collection for PO-administered animals: before administration, 15min, 30min, 1h, 2h, 4h, 8h, 24h, 48h, 72h after administration.
离心管用K2-EDTA包被,冰水浴保存;血样采集后转移至上述离心管中,手动混匀,1h内完成离心,全血样品在离心前冰水浴保存,离心条件为:3000g,4℃,10min。离心后,收集血浆分装成2管,-70℃以下保存。The centrifuge tube was coated with K2-EDTA and stored in an ice-water bath; after the blood sample was collected, it was transferred to the above-mentioned centrifuge tube, mixed manually, and the centrifugation was completed within 1 hour. The whole blood sample was stored in an ice-water bath before centrifugation. 10min. After centrifugation, the collected plasma was divided into 2 tubes and stored below -70℃.
血浆样品分析Plasma sample analysis
化合物的血浆样品分析均采用蛋白沉淀法,采用经验证的LC-MS/MS方法进行检测分析。The plasma samples of the compounds were analyzed by protein precipitation method, and the validated LC-MS/MS method was used for detection and analysis.
表6雌性SD大鼠PK评价结果Table 6 PK evaluation results of female SD rats
Figure PCTCN2020070062-appb-000018
Figure PCTCN2020070062-appb-000018
AUC last代表药时曲线下面积0→t;AUC inf代表药时曲线下面积0→∞;CL代表清除率;V ss表示稳态表观分布容积;T max代表血药浓度达峰时间;C max代表血药浓度达峰浓度;t 1/2代表半衰期;MRT代表平均驻留时间;F%代表绝对生物利用度 AUC last represents the area under the drug-time curve 0→t; AUC inf represents the area under the drug-time curve 0→∞; CL represents the clearance rate; V ss represents the steady-state apparent volume of distribution; T max represents the time when the blood drug concentration reaches the peak; C max represents the peak concentration of the blood drug concentration; t 1/2 represents the half-life; MRT represents the average residence time; F% represents the absolute bioavailability
由表6的实验结果可知,本发明化合物的药代动力学性质良好,经5mg/kg静脉和10mg/kg灌胃给药后t 1/2分别为4.21和9.09h;暴露量分别为7038.37和8202.03h*ng/mL,口服生物利用度为58.27%。 According to the experimental results in Table 6, the pharmacokinetic properties of the compound of the present invention are good. After administration by intragastric administration of 5 mg/kg intravenously and 10 mg/kg, t 1/2 is 4.21 and 9.09 h, respectively; the exposure is 7038.37 and 8202.03h*ng/mL, oral bioavailability is 58.27%.
以上所述仅是本发明的优选实施方式,应当指出,对于本技术领域的普通技术人员来说,在不脱离本发明原理的前提下,还可以做出若干改进和润饰,这些改进和润饰也应视为本发明的保护范围。The above is only a preferred embodiment of the present invention. It should be pointed out that for those of ordinary skill in the art, without departing from the principles of the present invention, several improvements and retouches can be made. These improvements and retouches also It should be regarded as the protection scope of the present invention.

Claims (17)

  1. 式(I)所示化合物的酒石酸盐,The tartrate of the compound represented by formula (I),
    Figure PCTCN2020070062-appb-100001
    Figure PCTCN2020070062-appb-100001
  2. 如权利要求1所述化合物的酒石酸盐,其特征在于,式(I)化合物与酒石酸的摩尔比为1:3-1:1,优选1:1。The tartrate salt of the compound according to claim 1, wherein the molar ratio of the compound of formula (I) to tartaric acid is 1:3 to 1:1, preferably 1:1.
  3. 如权利要求1或2所述的酒石酸盐,其中,所述的酒石酸为L-(+)-酒石酸。The tartrate according to claim 1 or 2, wherein the tartaric acid is L-(+)-tartaric acid.
  4. 一种式(I)所示化合物的L-(+)-酒石酸盐晶型I,L-(+)-tartrate crystal form I of a compound of formula (I),
    Figure PCTCN2020070062-appb-100002
    Figure PCTCN2020070062-appb-100002
    其特征在于,式(I)化合物与L-(+)-酒石酸的摩尔比为1:1,并且使用Cu-Kα辐射,以2θ角度表示的X-射线粉末衍射图谱,在8.9±0.2°、11.2±0.2°、12.0±0.2°、12.5±0.2°、13.1±0.2°、14.7±0.2°、17.5±0.2°、19.1±0.2°处有特征峰。It is characterized in that the molar ratio of the compound of formula (I) to L-(+)-tartaric acid is 1:1, and using Cu-Kα radiation, the X-ray powder diffraction pattern expressed at a 2θ angle, at 8.9±0.2°, 11.2±0.2°, 12.0±0.2°, 12.5±0.2°, 13.1±0.2°, 14.7±0.2°, 17.5±0.2°, 19.1±0.2° have characteristic peaks.
  5. 如权利要求4所述的酒石酸盐晶型I,其特征在于,使用Cu-Kα辐射,以2θ角度表示的X-射线粉末衍射图谱,还在16.3±0.2°、16.6±0.2°、20.0±0.2°、20.6±0.2°、21.1±0.2°、21.6±0.2°处有特征峰。The crystalline form I of tartrate according to claim 4, characterized in that, using Cu-Kα radiation, the X-ray powder diffraction pattern expressed at a 2θ angle is also 16.3±0.2°, 16.6±0.2°, 20.0±0.2 There are characteristic peaks at °, 20.6±0.2°, 21.1±0.2°, and 21.6±0.2°.
  6. 如权利要求5所述的酒石酸盐晶型I,其特征在于,其X-射线粉末衍射图谱基本如图1所示。The tartrate crystalline form I according to claim 5, characterized in that its X-ray powder diffraction pattern is substantially as shown in FIG.
  7. 如权利要求4所述的酒石酸盐晶型I,其特征在于,其差示扫描量热分析图在190℃至230℃范围内具有吸热峰;优选的,最大吸热转变温度为215.5±5℃;更优选的,所述晶型A具有基本上如图2所示的差示扫描量热曲线。The crystalline form I of tartrate according to claim 4, wherein the differential scanning calorimetry analysis chart has an endothermic peak in the range of 190°C to 230°C; preferably, the maximum endothermic transition temperature is 215.5±5 °C; more preferably, the crystalline form A has a differential scanning calorimetry curve substantially as shown in FIG. 2.
  8. 如权利要求4所述的酒石酸盐晶型I的制备方法,其特征在于,将式(I)化合物与1当量的L-(+)-酒石酸加入有机溶剂中,搅拌条件下加入水或N-甲基吡咯烷酮,反应结束后过滤,干燥得式(I)化合物的L-(+)-酒石酸盐晶型I;The method for preparing crystalline form I of tartrate according to claim 4, characterized in that the compound of formula (I) and 1 equivalent of L-(+)-tartaric acid are added to an organic solvent, and water or N- is added under stirring Methylpyrrolidone, filtered after the reaction is completed, and dried to obtain L-(+)-tartrate crystal form I of the compound of formula (I);
    优选的,所述有机溶剂选自低级腈类、含2个以上碳原子的低级醇类、含氧杂环类或者上述有机溶剂的任意混合溶剂;Preferably, the organic solvent is selected from lower nitriles, lower alcohols containing 2 or more carbon atoms, oxygen-containing heterocyclics, or any mixed solvent of the above organic solvents;
    更优选的,所述有机溶剂选自乙腈、乙醇、丙醇、异丙醇、丁醇、仲丁醇、叔丁醇、1,4-二氧六环或上述有机溶剂的任意混合溶剂。More preferably, the organic solvent is selected from acetonitrile, ethanol, propanol, isopropanol, butanol, sec-butanol, tert-butanol, 1,4-dioxane, or any mixed solvent of the above organic solvents.
  9. 如权利要求4所述的酒石酸盐晶型I的制备方法,其特征在于,将式(I)化合物加入单一溶剂或混合溶剂中,加热至一定温度后或常温浆洗条件下,加入一定当量的L-(+)-酒石酸,维持温度继续反应至反应结束,降温析晶或常温放置析晶,分离,干燥得到式(I)化合物的L-(+)-酒石酸盐晶型I;The method for preparing crystalline form I of tartrate according to claim 4, characterized in that the compound of formula (I) is added to a single solvent or a mixed solvent, after heating to a certain temperature or under normal temperature slurry washing conditions, a certain equivalent of L is added -(+)-tartaric acid, maintain the temperature and continue the reaction to the end of the reaction, lower the temperature or crystallize at room temperature, separate, and dry to obtain L-(+)-tartrate crystal form I of the compound of formula (I);
    优选的,所述的单一溶剂选自低级醇类、低级酮类、低级酯类、含氧杂环类、低级腈类、低级烷烃类或低级卤代烷烃类,所述的混合溶剂选自醇类与水混合溶剂、腈类与水混合溶剂、酮类与水混合溶剂或含氧杂环类与水混合溶剂。Preferably, the single solvent is selected from lower alcohols, lower ketones, lower esters, oxygenated heterocyclics, lower nitriles, lower alkanes or lower halogenated alkanes, and the mixed solvent is selected from alcohols Mixed solvent with water, mixed solvent with nitriles and water, mixed solvent with ketones and water, or mixed solvent with oxygenated heterocyclics and water.
  10. 如权利要求9所述的制备方法,其特征在于,所述单一溶剂选自甲醇、乙醇、丙醇、异丙醇、正丁醇、异丁醇、叔丁醇、丙酮、丁酮、甲基乙基酮、乙腈、甲酸乙酯、乙酸乙酯、乙酸甲酯、四氢呋喃、1,4-二氧六环、正戊烷、异戊烷、正己烷、异己烷、环己烷、环庚烷、甲苯或二氯甲烷;所述混合溶剂选自乙醇/水、丙醇/水、异丙醇/水、丁醇/水、仲丁醇/水、叔丁醇/水、乙腈/水、1,4-二氧六环/水或丙酮/水;The preparation method according to claim 9, wherein the single solvent is selected from methanol, ethanol, propanol, isopropanol, n-butanol, isobutanol, tert-butanol, acetone, butanone, methyl Ethyl ketone, acetonitrile, ethyl formate, ethyl acetate, methyl acetate, tetrahydrofuran, 1,4-dioxane, n-pentane, isopentane, n-hexane, isohexane, cyclohexane, cycloheptane , Toluene or methylene chloride; the mixed solvent is selected from ethanol/water, propanol/water, isopropanol/water, butanol/water, sec-butanol/water, tert-butanol/water, acetonitrile/water, 1 ,4-dioxane/water or acetone/water;
    优选的,所述混合溶剂中,有机溶剂与水的比例选自1:1-20:1,进一步优选的,所述混合溶剂中,有机溶剂与水的比例选自1:1-10:1,更优选 的,所述混合溶剂中,有机溶剂与水的比例选自2:1-10:1;所述的一定温度选自40℃-80℃,优选50℃-70℃。Preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 1:1-20:1, further preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 1:1-10:1 More preferably, in the mixed solvent, the ratio of organic solvent to water is selected from 2:1-10:1; the certain temperature is selected from 40°C-80°C, preferably 50°C-70°C.
  11. 如权利要求9所述的制备方法,其特征在于,所述的一定当量选自0.9-1.2当量,优选1.0-1.1当量,更优选1.0当量。The preparation method according to claim 9, wherein the certain equivalent is selected from 0.9-1.2 equivalent, preferably 1.0-1.1 equivalent, more preferably 1.0 equivalent.
  12. 一种式(I)所示化合物的L-(+)-酒石酸盐晶型II,L-(+)-tartrate crystal form II of a compound of formula (I),
    Figure PCTCN2020070062-appb-100003
    Figure PCTCN2020070062-appb-100003
    其特征在于,式(I)化合物与酒石酸的摩尔比为1:1,并且使用Cu-Kα辐射,以2θ角度表示的X-射线粉末衍射图谱,在9.0±0.2°、11.3±0.2°、12.0±0.2°、12.7±0.2°、13.9±0.2°、17.9±0.2°、19.1±0.2°、20.3±0.2°、21.2±0.2°处有特征峰。It is characterized in that the molar ratio of the compound of formula (I) to tartaric acid is 1:1, and the Cu-Kα radiation is used to express the X-ray powder diffraction pattern at an angle of 2θ at 9.0±0.2°, 11.3±0.2°, 12.0 There are characteristic peaks at ±0.2°, 12.7±0.2°, 13.9±0.2°, 17.9±0.2°, 19.1±0.2°, 20.3±0.2°, 21.2±0.2°.
  13. 如权利要求12所述的晶型II,其特征在于,使用Cu-Kα辐射,以2θ角度表示的X-射线粉末衍射图谱,还在16.3±0.2°、17.3±0.2°、23.5±0.2°、24.4±0.2°、25.5±0.2°、25.9±0.2°处有特征峰;优选的,所述晶型II的X-射线粉末衍射图谱基本如图5所示。The crystalline form II according to claim 12, wherein the X-ray powder diffraction pattern expressed at an angle of 2θ using Cu-Kα radiation is also 16.3±0.2°, 17.3±0.2°, 23.5±0.2°, There are characteristic peaks at 24.4±0.2°, 25.5±0.2°, and 25.9±0.2°; preferably, the X-ray powder diffraction pattern of the crystal form II is basically shown in FIG. 5.
  14. 药物组合物,其含有如权利要求1-3中任一项所述的式(I)所示化合物的酒石酸盐、权利要求4-7中任一项所述的式(I)所示化合物的酒石酸盐晶型I或权利要求12-13中任一项所述的式(I)所示化合物的酒石酸盐晶型II以及一种或多种药用载体,所述药物组合物可以制备成药学上可接受的任一剂型。A pharmaceutical composition containing a tartrate salt of a compound represented by formula (I) according to any one of claims 1-3, and a compound represented by formula (I) according to any one of claims 4-7 The tartrate crystal form I or the tartrate crystal form II of the compound represented by formula (I) according to any one of claims 12-13 and one or more pharmaceutically acceptable carriers, the pharmaceutical composition can be prepared into a pharmaceutical Any dosage form that is acceptable.
  15. 权利要求1-3中任一项所述的式(I)所示化合物的酒石酸盐、权利要求4-7中任一项所述的式(I)所示化合物的酒石酸盐晶型I或权利要求12-13中任一项所述的式(I)所示化合物的酒石酸盐晶型II在制备用于治疗和/或预防由EGFR介导的过度增殖性疾病的药物中的应用,所述的过度增殖性疾病选自癌症。A tartrate salt of the compound represented by formula (I) according to any one of claims 1-3, or a tartrate crystal form I of the compound represented by formula (I) according to any one of claims 4-7, or the right The use of the tartrate crystal form II of the compound represented by formula (I) according to any one of claims 12 to 13 in the preparation of a medicament for the treatment and/or prevention of EGFR-mediated hyperproliferative diseases, said The hyperproliferative diseases are selected from cancer.
  16. 如权利要求15所述的用途,其中所述的癌症选自:肺癌、非小细胞肺癌、结直肠癌、胰腺癌、头颈癌、乳腺癌、卵巢癌、子宫癌、肝癌、胃癌前列腺癌、胶质母细胞瘤和上皮细胞癌。The use according to claim 15, wherein the cancer is selected from the group consisting of lung cancer, non-small cell lung cancer, colorectal cancer, pancreatic cancer, head and neck cancer, breast cancer, ovarian cancer, uterine cancer, liver cancer, gastric cancer, prostate cancer, gum Glioblastoma and epithelial cell carcinoma.
  17. 如权利要求15或16所述的用途,其中所述的EGFR选自野生型EGFR和突变型EGFR,优选的,突变型EGFR包括EGFR 19位外显子突变、EGFR 20位外显子突变和EGFR 21位外显子突变中的一种或两种以上突变,更优选的,所述EGFR 20位外显子突变选自NPG、ASV或T790M,所述的EGFR 19位外显子突变选自插入突变,所述的EGFR 21位外显子突变选自点突变。The use according to claim 15 or 16, wherein the EGFR is selected from wild type EGFR and mutant EGFR, preferably, mutant EGFR includes EGFR 19 exon mutation, EGFR 20 exon mutation and EGFR One or two or more mutations among the 21 exon mutations, more preferably, the EGFR 20 exon mutation is selected from NPG, ASV, or T790M, and the EGFR 19 exon mutation is selected from insertion Mutation, the EGFR 21 exon mutation is selected from point mutations.
PCT/CN2020/070062 2019-01-05 2020-01-02 Salt of selective inhibitor of egfr tyrosine kinase and crystal form thereof WO2020140934A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080007085.XA CN113227073B (en) 2019-01-05 2020-01-02 Salts of selective inhibitors of EGFR tyrosine kinase and crystalline forms thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910009657 2019-01-05
CN201910009657.7 2019-01-05

Publications (1)

Publication Number Publication Date
WO2020140934A1 true WO2020140934A1 (en) 2020-07-09

Family

ID=71407147

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/070062 WO2020140934A1 (en) 2019-01-05 2020-01-02 Salt of selective inhibitor of egfr tyrosine kinase and crystal form thereof

Country Status (2)

Country Link
CN (1) CN113227073B (en)
WO (1) WO2020140934A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103702990A (en) * 2011-07-27 2014-04-02 阿斯利康(瑞典)有限公司 2-(2,4,5-substituted -anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
WO2019101295A1 (en) * 2017-11-21 2019-05-31 Huawei Technologies Co., Ltd. Image and video processing apparatuses and methods

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104860941B (en) * 2014-02-25 2017-03-22 上海海雁医药科技有限公司 2,4-disubstituted phenyl-1,5-diamine derivatives and use thereof, and pharmaceutical composition and medicinal composition prepared from 2,4-disubstituted phenyl-1,5-diamine derivative
TWI726968B (en) * 2016-01-07 2021-05-11 開曼群島商Cs醫藥技術公司 Selective inhibitors of clinically important mutants of the egfr tyrosine kinase

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103702990A (en) * 2011-07-27 2014-04-02 阿斯利康(瑞典)有限公司 2-(2,4,5-substituted -anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
WO2019101295A1 (en) * 2017-11-21 2019-05-31 Huawei Technologies Co., Ltd. Image and video processing apparatuses and methods

Also Published As

Publication number Publication date
CN113227073A (en) 2021-08-06
CN113227073B (en) 2022-09-16

Similar Documents

Publication Publication Date Title
JP7414756B2 (en) Acrylanilide derivatives, their production methods and their pharmaceutical applications
TW201512176A (en) Protein tyrosine kinase modulators and methods of use
TW201638092A (en) Pyrimidine pyrrole compound, preparation method therefor, pharmaceutical composition, and uses thereof
US20200361908A1 (en) Crystals of aniline pyrimidine compound serving as egfr inhibitor
US20150152088A1 (en) Alkynyl heteroaromatic compound and use thereof
US20210101881A1 (en) Pyrimidine compound, preparation method thereof and medical use thereof
TW201922709A (en) Epidermal growth factor receptor inhibitors
CN111410651B (en) Salts of tyrosine kinase inhibitors and crystalline forms thereof
WO2020001351A1 (en) Egfr inhibitor, method for preparing the same, and uses thereof
WO2020140934A1 (en) Salt of selective inhibitor of egfr tyrosine kinase and crystal form thereof
WO2022257965A1 (en) Cyclin-dependent kinase 9 inhibitor in solid form and use thereof
WO2019228330A1 (en) Substituted benzo[d]imidazole compound and pharmaceutical composition thereof
WO2019223777A1 (en) Pyrrolopyrimidine compound containing arylamine substitution, preparation method and application thereof
WO2022063229A1 (en) Salt of arylaminoquinazoline-containing compound, and preparation method therefor and use thereof
CN111362924B (en) Deuterated pyrimidine derivatives and uses thereof
WO2021147996A1 (en) Salt of arylaminopurine derivative, preparation method therefor and use thereof
WO2020147838A1 (en) Salt of egfr inhibitor, crystal form, and preparation method therefor
CN111233774B (en) Amino pyrimidine compound
WO2022007841A1 (en) Egfr inhibitor, preparation method therefor, and pharmaceutical application thereof
TWI820414B (en) Quinazoline compounds, preparation method and use thereof
CN115701429B (en) 4- (1H-indol-1-yl) pyrimidine-2-amino derivative, and preparation method and application thereof
CN110642838B (en) EGFR inhibitor and preparation and application thereof
CN110642836B (en) EGFR inhibitor and preparation and application thereof
CN107663207B (en) Mesylate crystal of EGFR kinase inhibitor and preparation method thereof
WO2023093859A1 (en) Salt of axl kinase inhibitor, preparation method therefor and use thereof

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: 20735872

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20735872

Country of ref document: EP

Kind code of ref document: A1