[go: up one dir, main page]
More Web Proxy on the site http://driver.im/

WO2023093859A1 - Salt of axl kinase inhibitor, preparation method therefor and use thereof - Google Patents

Salt of axl kinase inhibitor, preparation method therefor and use thereof Download PDF

Info

Publication number
WO2023093859A1
WO2023093859A1 PCT/CN2022/134408 CN2022134408W WO2023093859A1 WO 2023093859 A1 WO2023093859 A1 WO 2023093859A1 CN 2022134408 W CN2022134408 W CN 2022134408W WO 2023093859 A1 WO2023093859 A1 WO 2023093859A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
salt
pharmaceutically acceptable
crystal form
Prior art date
Application number
PCT/CN2022/134408
Other languages
French (fr)
Chinese (zh)
Inventor
张林林
马昌友
吴有智
裴俊杰
吴舰
徐丹
朱春霞
田舟山
Original Assignee
南京正大天晴制药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 南京正大天晴制药有限公司 filed Critical 南京正大天晴制药有限公司
Priority to CN202280074029.7A priority Critical patent/CN118251400A/en
Publication of WO2023093859A1 publication Critical patent/WO2023093859A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C303/00Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides
    • C07C303/32Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides of salts of sulfonic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C303/00Preparation of esters or amides of sulfuric acids; Preparation of sulfonic acids or of their esters, halides, anhydrides or amides
    • C07C303/42Separation; Purification; Stabilisation; Use of additives
    • C07C303/44Separation; Purification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C309/00Sulfonic acids; Halides, esters, or anhydrides thereof
    • C07C309/01Sulfonic acids
    • C07C309/02Sulfonic acids having sulfo groups bound to acyclic carbon atoms
    • C07C309/03Sulfonic acids having sulfo groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton
    • C07C309/04Sulfonic acids having sulfo groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton containing only one sulfo group
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the invention belongs to the technical field of medicine, and the compound is an AXL kinase inhibitor, and specifically relates to a salt of the AXL inhibitor, a preparation method and a medical application thereof.
  • RTKs Receptor tyrosine kinases
  • Ligand-receptor binding induces receptor dimerization and activation of its intracellular kinase domain, which in turn leads to the recruitment, phosphorylation and activation of multiple downstream signaling cascades (Robinson, D.R. et al., Oncogene, 19:5548-5557, 2000).
  • RTKs have been identified in the human genome that regulate a variety of cellular processes, including cell survival, growth, differentiation, proliferation, adhesion, and motility (Segaliny, A.I. et al., J. Bone Oncol, 4:1 -12, 2015).
  • AXL (also known as UFO, ARK, and Tyro7) belongs to the TAM family of receptor tyrosine kinases, which also includes Mer and Tyro3. Among them, AXL and Tyro3 have the most similar gene structure, while AXL and Mer have the most similar amino acid sequence of tyrosine kinase domain. Like other receptor tyrosine kinases (RTKs), the structure of the TAM family consists of an extracellular domain, a transmembrane domain, and a conserved intracellular kinase domain. The extracellular domain of AXL has a unique structure that juxtaposes immunoglobulin and type III fibronectin repeat units and is reminiscent of a neutrophil adhesion molecule.
  • TAM family members have a common ligand—growth arrest specific protein 6 (Gas6), which can bind to all TAM receptor tyrosine kinases. After AXL binds to Gas6, it will lead to receptor dimerization and AXL autophosphorylation, thereby activating multiple downstream signal transduction pathways and participating in multiple processes of tumorigenesis (Linger, R.M et al., Ther.Targets, 14(10 ), 1073-1090, 2010; Rescigno, J. et al., Oncogene, 6(10), 1909-1913, 1991).
  • Gas6 growth arrest specific protein 6
  • AXL is widely expressed in normal tissues of the human body, such as monocytes, macrophages, platelets, endothelial cells, cerebellum, heart, skeletal muscle, liver, and kidney, among which the expression is highest in cardiac muscle and skeletal muscle, and bone marrow CD34+ cells and stromal cells also have a higher expression High expression, very low expression in normal lymphoid tissue (Wu YM, Robinson DR, Kung HJ, Cancer Res, 64(20), 7311-7320, 2004; hung BI et al., DNA Cell Biol, 22(8), 533-540 , 2003).
  • AXL gene was overexpressed or ectopically expressed in hematopoietic cells, mesenchymal cells and endothelial cells.
  • the overexpression of AXL kinase is particularly prominent.
  • Inhibition of AXL receptor tyrosine kinase can reduce the pro-survival signals of tumor cells, block the invasion ability of tumors, and increase the sensitivity of targeted drug therapy and chemotherapy. Therefore, finding effective AXL inhibitors is an important direction for the development of tumor-targeted drugs.
  • the present invention provides a pharmaceutically acceptable salt of the compound of formula I, wherein the salt is selected from organic acid salts or inorganic acid salts, wherein the organic acid salts are selected from methanesulfonate, benzenesulfonate, oxalate
  • the inorganic acid salt is selected from one of hydrochloride, hydrobromide, sulfate or phosphate, the compound of formula I
  • the structure is as follows:
  • the organic acid salt is mesylate.
  • the mesylate salt is a hydrate of the mesylate salt.
  • the mesylate salt is the dihydrate of the mesylate salt.
  • the molar ratio of the compound of formula I to the organic acid in the organic acid salt is 1:1.
  • the molar ratio of the compound of formula I to the inorganic acid in the inorganic acid salt is 1:1 or 1:2.
  • the salt of an inorganic acid is a hydrochloride.
  • the molar ratio of the compound of formula I to hydrogen chloride in the hydrochloride salt is 1:1 or 1:2.
  • the molar ratio of the compound of formula I to hydrogen chloride in the hydrochloride salt is 1:2.
  • the molar ratio of the compound of formula I to sulfuric acid in the sulfate salt is 1:1.
  • the molar ratio of the compound of formula I to hydrobromic acid in the hydrobromide salt is 1:1.
  • the molar ratio of the compound of formula I to phosphoric acid in the phosphate salt is 1:1.
  • the salt in the present invention is obtained by a salt-forming reaction between the compound of formula I and the corresponding acid.
  • the compound of formula I is converted into a cation and combined with the acid radical of the corresponding acid to form the salt. Therefore, the molar ratio of the compound of formula I to the acid in the present invention can be understood as the molar ratio of the cation of the compound of formula I to the acid group of the corresponding acid in the salt.
  • the present invention provides the mesylate salt of the compound of formula I, wherein the molar ratio of the compound of formula I to methanesulfonic acid is 1:1, or the molar ratio of the cation of the compound of formula I to the acid group of methanesulfonic acid The ratio is 1:1.
  • the present invention provides a crystal form of a pharmaceutically acceptable salt of the compound of formula I, wherein the salt is selected from organic acid salts or inorganic acid salts, wherein the organic acid salts are selected from methanesulfonate, benzenesulfonate , oxalate, fumarate, citrate and hippurate, the inorganic acid salt is selected from hydrochloride, hydrobromide or phosphate.
  • the present invention provides a crystalline form of the mesylate salt of the compound of Formula I.
  • the 2 ⁇ of the X-ray powder diffraction pattern of the mesylate salt crystal is detailed in Table 1 below:
  • the present invention provides a crystalline form of the mesylate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 1 .
  • the present invention provides a crystalline form of the monohydrochloride salt of a compound of formula I.
  • the 2 ⁇ of the X-ray powder diffraction pattern of the monohydrochloride crystal is detailed in Table 2 below:
  • the present invention provides a crystalline form of the monohydrochloride salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 4 . In some embodiments, the present invention provides a crystalline form of the dihydrochloride salt of the compound of formula I.
  • the dihydrochloride salt is crystalline, and the 2 ⁇ of its X-ray powder diffraction pattern is detailed in Table 3 below:
  • the present invention provides a crystal form of the dihydrochloride salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 5 .
  • the present invention provides a crystalline form of the phosphate salt of a compound of formula I.
  • the 2 ⁇ of the X-ray powder diffraction pattern of the phosphate crystal is detailed in Table 4 below:
  • the present invention provides a crystalline form of the phosphate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 6 .
  • the present invention provides a crystalline form of a hippurate salt of a compound of Formula I.
  • the 2 ⁇ of the X-ray powder diffraction pattern of the hippurate crystal is detailed in Table 5 below:
  • the present invention provides a crystalline form of hippurate of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 7 .
  • the present invention provides an amorphous form of the sulfate salt of the compound of formula I.
  • the present invention provides a crystalline form of the hydrobromide salt of a compound of formula I.
  • the 2 ⁇ of the hydrobromide crystal is shown in Table 6 below in detail in its X-ray powder diffraction pattern:
  • the present invention provides a crystal form of the hydrobromide salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 9 .
  • the present invention provides a crystalline form of the besylate salt of the compound of formula I.
  • the benzenesulfonate salt is crystalline, and the 2 ⁇ of its X-ray powder diffraction pattern is detailed in Table 7 below:
  • the present invention provides a crystalline form of the besylate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 10 .
  • the present invention provides a crystalline form of the oxalate salt of the compound of formula I.
  • the oxalate salt crystals, the 2 ⁇ of its X-ray powder diffraction pattern are detailed in Table 8 below:
  • the present invention provides the crystalline form of the oxalate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 11 .
  • the present invention provides a crystalline form of the fumarate salt of the compound of formula I.
  • the 2 ⁇ of the X-ray powder diffraction pattern of the fumarate crystals is detailed in Table 9 below:
  • the present invention provides a crystalline form of the fumarate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 12 .
  • the present invention provides a crystalline form of the citrate salt of the compound of formula I.
  • the 2 ⁇ of the X-ray powder diffraction pattern of the citrate crystals is detailed in Table 10 below:
  • the present invention provides a crystalline form of the citrate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 13 .
  • the present invention provides a crystal form A of the compound of formula I, whose X-ray powder diffraction pattern is 7.6° ⁇ 0.2°, 10.2° ⁇ 0.2°, 17.6° ⁇ 0.2°, 20.3° ⁇ 0.2° at 2 ⁇ There are diffraction peaks at 20.9° ⁇ 0.2°.
  • the crystal form A has an X-ray powder diffraction pattern at 2 ⁇ of 4.1° ⁇ 0.2°, 7.6° ⁇ 0.2°, 10.2° ⁇ 0.2°, 12.6° ⁇ 0.2°, 13.0° ⁇ 0.2°, 17.6° There are diffraction peaks at ⁇ 0.2°, 19.7° ⁇ 0.2°, 20.3° ⁇ 0.2°, 20.9° ⁇ 0.2° and 22.2° ⁇ 0.2°.
  • the crystal form A has an X-ray powder diffraction pattern at 2 ⁇ of 4.1° ⁇ 0.2°, 5.6° ⁇ 0.2°, 7.6° ⁇ 0.2°, 10.2° ⁇ 0.2°, 10.9° ⁇ 0.2°, 12.6° ⁇ 0.2°, 13.0° ⁇ 0.2°, 15.2° ⁇ 0.2°, 17.6° ⁇ 0.2°, 19.7° ⁇ 0.2°, 20.3° ⁇ 0.2°, 20.9° ⁇ 0.2°, 22.2° ⁇ 0.2°, 23.2° ⁇ 0.2 °, 24.6° ⁇ 0.2°, 27.0° ⁇ 0.2°, 28.8° ⁇ 0.2°, 37.0° ⁇ 0.2° and 37.7° ⁇ 0.2° have diffraction peaks.
  • the 2 ⁇ of the X-ray powder diffraction pattern of the crystal form A is detailed in the table below:
  • the X-ray powder diffraction of the crystal form A in 2 ⁇ angle has a pattern as shown in FIG. 14 .
  • the present invention provides a method for preparing a pharmaceutically acceptable salt of the compound of formula I or a crystal form of the pharmaceutically acceptable salt thereof, which comprises the step of forming a salt of the compound of formula I with a corresponding acid.
  • the reaction solvent of the preparation method is selected from mixed solvents of alcohol solvents and alkane solvents, mixed solvents of ketone solvents and alkane solvents, mixed solvents of ester solvents and alkane solvents, nitrile solvents - A mixed solvent of an aqueous solvent and an alkane solvent, a mixed solvent of an alkylbenzene solvent and an alkane solvent, or a mixed solvent of a halogenated hydrocarbon solvent and an alkane solvent.
  • the alcohol solvent is selected from methanol, ethanol or isopropanol; the ketone solvent is selected from acetone or butanone; preferably acetone; the ester solvent is selected from ethyl acetate or butyl acetate ; Preferred ethyl acetate;
  • the nitriles-water solvent is selected from the mixed solution of nitriles-water, and the alkane solvent is selected from n-heptane.
  • the crystal form of the pharmaceutically acceptable salt is a mesylate salt crystal form
  • the mesylate salt crystal form is prepared by a method comprising the following steps:
  • step S2 stirring and reacting the compound of formula (I), methanesulfonic acid and ethyl acetate, adding the crystal form obtained in step S1 and continuing to stir;
  • the present invention also provides a pharmaceutical composition comprising a pharmaceutically acceptable salt of the compound of formula I.
  • the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
  • the pharmaceutical composition is a solid pharmaceutical preparation suitable for oral administration, preferably a tablet or a capsule.
  • the present invention also provides a pharmaceutical composition comprising the crystal form of the pharmaceutically acceptable salt of the compound of formula I.
  • the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
  • the pharmaceutical composition is a solid pharmaceutical preparation suitable for oral administration, preferably a tablet or a capsule.
  • the present invention provides a crystal form composition, wherein the pharmaceutically acceptable salt of the compound of formula I above, and the crystal form of the pharmaceutically acceptable salt of the compound of formula I account for the weight of the crystal form composition above 50.
  • the present invention also provides a pharmaceutical composition comprising the above crystal composition; the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
  • the present invention also provides a pharmaceutically acceptable salt of the compound of formula I or its crystal form composition or its pharmaceutical composition for use as a medicine.
  • the present invention also provides a pharmaceutically acceptable salt crystal form of the compound of formula I or its crystal form composition or pharmaceutical composition thereof for use as a medicine.
  • the present invention also provides a method for preventing and/or treating AXL kinase-mediated diseases or disease states, which comprises administering the salt of the compound of formula I of the present invention, or its crystal form, to individuals in need Composition or its pharmaceutical composition.
  • the present invention also provides a method for preventing and/or treating AXL kinase-mediated diseases or disease states, which comprises administering the crystalline form of the salt of the compound of formula I of the present invention, Its crystal form composition or its pharmaceutical composition.
  • the present invention also provides the salt of the compound of formula I of the present invention, its crystal form composition or its pharmaceutical composition for preventing and/or treating AXL kinase-mediated diseases or disease states.
  • the present invention also provides the crystalline form of the salt of the compound of formula I of the present invention, its crystalline form composition or its pharmaceutical composition for preventing and/or treating AXL kinase-mediated diseases or disease states .
  • the AXL kinase-mediated disease or condition is cancer.
  • the cancer is a disease associated with hematological and solid tumors.
  • the pharmaceutically acceptable salts of the present invention also include their hydrated forms.
  • pharmaceutically acceptable carrier refers to those carriers that have no obvious stimulating effect on the body and will not impair the biological activity and performance of the active compound. Including but not limited to any diluents, disintegrants, binders, glidants, and wetting agents approved by the State Food and Drug Administration for human or animal use.
  • fumaric acid refers to fumaric acid, which has the structure:
  • alcohol solvent refers to a substance derived from one or more hydroxyl groups (OH) replacing one or more hydrogen atoms on a C1-C6 alkane
  • C1-C6 alkane refers to a substance containing 1-6 carbon atoms
  • Specific examples of linear or branched alkanes and alcohol solvents include, but are not limited to: methanol, ethanol, isopropanol or n-propanol.
  • alkane solvent refers to straight chain or branched chain or cyclic alkanes containing 5-7 carbon atoms, specific examples include but not limited to n-hexane, cyclohexane, n-heptane.
  • ester solvent refers to a chain compound containing an ester group -COOR and a carbon number of 3-10, wherein R is a C1-C6 alkyl group, and the C1-C6 alkyl group refers to a chain compound containing 1-6
  • R is a C1-C6 alkyl group
  • C1-C6 alkyl group refers to a chain compound containing 1-6
  • Specific examples of straight-chain or branched-chain alkanes with carbon atoms and ester solvents include but are not limited to methyl acetate, ethyl acetate, and propyl acetate.
  • halogenated hydrocarbon solvent refers to a substance derived from one or more halogen atoms replacing one or more hydrogen atoms on a C1-C6 alkane
  • the C1-C6 alkane refers to a substance containing 1-6 carbon atoms
  • the halogen atoms refer to fluorine, chlorine, bromine, and iodine.
  • Specific examples of halogenated hydrocarbon solvents include but are not limited to dichloromethane or chloroform.
  • ketone solvent refers to a chain or cyclic compound containing carbonyl -CO- and having 3-10 carbon atoms, specific examples include but not limited to acetone, methyl ethyl ketone or cyclohexanone.
  • benzene-based solvent means a solvent containing a phenyl group, and specific examples include toluene, xylene, cumene, or chlorobenzene.
  • equivalent refers to the equivalent amount of other raw materials required in accordance with the equivalent relationship of chemical reactions, taking the basic raw materials used in each step as 1 equivalent.
  • the "X-ray powder diffraction pattern" in the present invention is obtained by using Cu-K ⁇ radiation measurement.
  • the diffraction pattern obtained from a crystalline compound is often characteristic for a particular crystal, where the relative intensity of the bands (especially at low angles) may vary due to The effect of dominant orientation due to differences in crystallization conditions, particle size and other measurement conditions varies. Therefore, the relative intensities of the diffraction peaks are not characteristic of the targeted crystals.
  • XRPD X-ray powder diffraction
  • the position of the peak can move, and the measurement error of the 2 ⁇ value is sometimes about ⁇ 0.2°. Therefore, this error should be taken into account when determining each crystal structure.
  • the peak positions of their XRPD spectra are similar on the whole, and the relative intensity error may be large.
  • DSC Differential Scanning Calorimetry
  • Thermogravimetric analysis refers to a thermal analysis technique that measures the relationship between the mass of the sample to be tested and the temperature change at a programmed temperature.
  • TGA Thermogravimetric analysis
  • IMDM (Iscove's Modified Dulbecco's Medium): Iscove (person's name) modified Dulbecco (person's name) medium.
  • Fig. 1 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound mesylate in embodiment 5;
  • Fig. 2 shows the differential scanning calorimetry (DSC) figure of formula I compound mesylate in embodiment 5;
  • Fig. 3 shows the thermogravimetric (TGA) figure of formula I compound mesylate in embodiment 5;
  • Fig. 4 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound monohydrochloride in embodiment 3;
  • Fig. 5 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound dihydrochloride in embodiment 3;
  • Fig. 6 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound phosphate in embodiment 3;
  • Fig. 7 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound hippurate in embodiment 3;
  • Fig. 8 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound sulfate in embodiment 3;
  • Fig. 9 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound hydrobromide in embodiment 3;
  • Fig. 10 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound besylate in embodiment 3;
  • Fig. 11 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound oxalate in embodiment 3;
  • Fig. 12 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound fumarate in embodiment 3;
  • Fig. 13 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound citrate in embodiment 3;
  • Figure 14 shows the X-ray powder diffraction (XRPD) spectrum of the crystal form A of the compound of formula I in Example 6.
  • test conditions of each instrument are as follows:
  • Temperature rise range room temperature - 300°C
  • Method Put the sample in an aluminum pan, then place the aluminum pan in a platinum pan, expose it in a nitrogen atmosphere, and raise the temperature from room temperature to the set temperature at a rate of 10°C/min.
  • Heating range 20-300°C
  • the sample was placed in an aluminum pan, and after capping, the temperature was raised from 20°C to the set temperature at a rate of 10°C/min in a nitrogen atmosphere.
  • the crude product was purified by reverse-phase high-performance liquid chromatography (column is YMC Actus Triart C18, 30*150mm, particle diameter 5 ⁇ m, mobile phase A: water (10mmol/L ammonium bicarbonate), mobile phase B: acetonitrile, flow rate: 60mL/ min, gradient: 20%B to 50%B, 8min, wavelength: 220nm, retention time: 6.83min, column temperature: 25°C), the title product (20.2mg) was obtained.
  • Example 1 Related compounds prepared in Example 1 carry out related enzyme activities, cells, and related activities in vivo
  • the specific structure of the positive drug 1 (BGB324) used in the activity test is as follows:
  • 1 ⁇ enzyme buffer 200 ⁇ L of Enzymatic buffer kinase 5X, 10 ⁇ L of 500 mM MgCl 2 , 10 ⁇ L of 100 mM DTT, 6.26 ⁇
  • DMSO dilute the compounds and positive drugs prepared in the examples from 10 mM to 100 ⁇ M, and titrate with a compound titrator (Tecan, D300e), and the titrator will automatically spray the required concentration into each well, the first step A concentration of 1 ⁇ M, 1/2 log gradient dilution, a total of 8 concentrations. Centrifuge at 2500rpm for 30s and incubate at room temperature for 15min.
  • ATP (Sigma, A7699) was diluted with 1 ⁇ enzyme buffer, from 10 mM to 75 ⁇ M (5 ⁇ ), and the final concentration was 15 ⁇ M; substrate TK Substrate 3-biotin (Cisbio, 61TK0BLC) was diluted with 1 ⁇ enzyme buffer solution from 500 ⁇ M to 5 ⁇ M (5 ⁇ ), and the final concentration was 1 ⁇ M; ATP was mixed with the substrate in equal volume, and 4 ⁇ L was added to each well using a BioTek automatic liquid dispenser; centrifuged at 2500 rpm for 30 s, at 25 ° C React for 45 minutes.
  • Streptavidin-XL665 (Cisbio, 610SAXLG) was diluted from 16.67 ⁇ M to 250nM (4 ⁇ ) with HTRF KinEASE detection buffer (cisbio), and the final concentration was 62.5nM;
  • TK Antibody-Cryptate (Cisbio) was diluted with HTRF KinEASE detection buffer (cisbio) was diluted from 100 ⁇ to 5 ⁇ , and the final concentration was 1 ⁇ ;
  • XL665 was mixed with Antibody in equal volume, 10 ⁇ L was added to each well using a BioTek automatic dispenser, centrifuged at 2500 rpm for 30 seconds, and reacted at 25°C for 1 hour. After the reaction, the multifunctional plate reader HTRF was used for detection.
  • MV-4-11 human myelomonocytic leukemia cell line, medium: IMDM+10% fetal bovine serum
  • IMDM+10% fetal bovine serum was purchased from Nanjing Kebai Biotechnology Co., Ltd., and placed in an incubator at 37°C and 5% CO 2 nourish.
  • Cells in the logarithmic growth phase were plated in 96-well plates at cell densities of 8000/well, 6000/well, 5000/well, 4000/well and 3000/well, and a blank control group was set at the same time.
  • Signal value of the test substance the mean value of the fluorescent signal of the cell + medium + compound group
  • Signal value of the blank group the average value of the fluorescence signal of the culture medium group (containing 0.5% DMSO);
  • Signal value of negative control group mean value of fluorescence signal of cell+medium group (containing 0.5% DMSO).
  • the IC 50 (MV4-11, nM) of the antiproliferative activity of the compound of Example 1 on MV4-11 cells was 6.97.
  • test compound The inhibitory effect of the test compound and the positive drug on the growth of human acute monocytic leukemia cell MV-4-11 xenografted tumor model in nude mice in vivo.
  • MV-4-11 cells in the logarithmic growth phase were collected, counted and resuspended, and the cell concentration was adjusted to 7.0 ⁇ 10 7 cells/mL; injected subcutaneously into the right axilla of nude mice, each animal was inoculated with 200 L (14 ⁇ 10 6 cells/monkey), the MV-4-11 xenograft tumor model was established. When the tumor volume reaches 100-300 mm 3 , tumor-bearing mice with good health and similar tumor volume are selected.
  • Solvent control group PEG400&citric acid buffer (20:80, v:v).
  • tumor volume (mm 3 ) l ⁇ w 2 /2
  • RTV relative tumor volume
  • TV initial is the tumor volume measured during group administration
  • TV t is the tumor volume at each measurement during administration.
  • TV t (T) represents the tumor volume measured each time in the treatment group
  • TV initial (T) represents the tumor volume of the treatment group when administered in groups
  • TV t (C) represents the tumor volume measured each time in the solvent control group
  • TV initial (C) represents the tumor volume of the solvent control group at the time of group administration.
  • RTV T represents the RTV of the treatment group
  • RTV C represents the RTV of the solvent control group.
  • the experimental data in the table is the relevant data obtained when the experiment ends (the end of the experiment is defined as: after 21 days or when the tumor volume of the solvent control group reaches 2000 mm 3 and the experiment ends (whichever is reached earlier)).
  • Each compound was prepared as a 10 mg/mL stock solution in DMSO.
  • mice Male ICR mice aged 6-10 weeks (source of mice: Weitong Lihua Experimental Animal Technology Co., Ltd.), 6 in each group, were fasted overnight and fed 4 hours after administration. On the day of the experiment, the mice were given 10 mg kg- 1 compound test solution by intragastric administration. At 0, 5min, 15min, 30min, 1h, 2h, 4h, 8h, and 24h after administration, about 100 ⁇ L of blood was collected from the orbit of the mice and placed in an EDTA-K 2 anticoagulant tube. Whole blood samples were centrifuged at 1500-1600 g for 10 min, and the separated plasma was stored in a -40-20°C refrigerator for biological sample analysis. LC-MS/MS method was used to determine the plasma concentration.
  • Fig. 4-13 is respectively formula I compound hydrochloride, dihydrochloride, phosphate, hippurate, sulfate, hydrobromide, benzenesulfonate, oxalate, fumarate, citric acid X-ray powder diffraction (XRPD) pattern of the salt.
  • XRPD X-ray powder diffraction
  • Embodiment 6 (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6,7,8,9-tetrahydro-5H-benzo[7] wheel Preparation of Form A of En-2-yl)amino)pyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethylphosphine oxide (compound of formula I)

Landscapes

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

Abstract

The present invention provides a salt of a compound of formula (I) and a crystal form thereof, a preparation method therefor and use thereof. The salt is selected from the group consisting of mesylate, benzene sulfonate, oxalate, fumarate, citrate, equine urate, hydrochloride, hydrobromate, sulfate, or phosphate.

Description

AXL激酶抑制剂的盐、其制备方法和用途Salts of AXL kinase inhibitors, their preparation methods and uses 技术领域technical field
本发明属于医药技术领域,所述化合物是AXL激酶抑制剂,具体涉及AXL抑制剂的盐、其制备方法以及医药用途。The invention belongs to the technical field of medicine, and the compound is an AXL kinase inhibitor, and specifically relates to a salt of the AXL inhibitor, a preparation method and a medical application thereof.
背景技术Background technique
受体酪氨酸激酶(RTK)是多域跨膜蛋白,可作为细胞外配体的传感器。配体受体结合诱导受体二聚化并激活其胞内激酶结构域,继而导致多个下游信号级联反应的募集、磷酸化和激活(Robinson,D.R.等,Oncogene,19:5548-5557,2000)。迄今为止,已在人类基因组中鉴定出58个RTK,它们可调节多种细胞过程,包括细胞存活、生长、分化、增殖、粘附和运动(Segaliny,A.I.等,J.Bone Oncol,4:1-12,2015)。Receptor tyrosine kinases (RTKs) are multidomain transmembrane proteins that serve as sensors of extracellular ligands. Ligand-receptor binding induces receptor dimerization and activation of its intracellular kinase domain, which in turn leads to the recruitment, phosphorylation and activation of multiple downstream signaling cascades (Robinson, D.R. et al., Oncogene, 19:5548-5557, 2000). To date, 58 RTKs have been identified in the human genome that regulate a variety of cellular processes, including cell survival, growth, differentiation, proliferation, adhesion, and motility (Segaliny, A.I. et al., J. Bone Oncol, 4:1 -12, 2015).
AXL(又称为UFO、ARK和Tyro7)属于受体酪氨酸激酶TAM家族,该家族成员还包括Mer和Tyro3。其中,AXL和Tyro3具有最为相似的基因结构,而AXL和Mer具有最为相似的酪氨酸激酶域氨基酸序列。与其他受体酪氨酸激酶(RTKs)一样,TAM家族的结构包含胞外域、跨膜域和保守的胞内激酶域。AXL的细胞外结构域具有独特的使免疫球蛋白和III型纤维连接蛋白重复单元并置的结构并且使人联想到中性细胞粘附分子的结构。TAM家族成员有1个共同配体—生长抑制特异性蛋白6(Gas6),该配体能够与所有TAM受体酪氨酸激酶结合。AXL与Gas6结合后,会导致受体二聚化和AXL自磷酸化,从而激活下游多条信号转导通路,并参与肿瘤发生的多个过程(Linger,R.M等,Ther.Targets,14(10),1073-1090,2010;Rescigno,J.等,Oncogene,6(10),1909-1913,1991)。AXL (also known as UFO, ARK, and Tyro7) belongs to the TAM family of receptor tyrosine kinases, which also includes Mer and Tyro3. Among them, AXL and Tyro3 have the most similar gene structure, while AXL and Mer have the most similar amino acid sequence of tyrosine kinase domain. Like other receptor tyrosine kinases (RTKs), the structure of the TAM family consists of an extracellular domain, a transmembrane domain, and a conserved intracellular kinase domain. The extracellular domain of AXL has a unique structure that juxtaposes immunoglobulin and type III fibronectin repeat units and is reminiscent of a neutrophil adhesion molecule. TAM family members have a common ligand—growth arrest specific protein 6 (Gas6), which can bind to all TAM receptor tyrosine kinases. After AXL binds to Gas6, it will lead to receptor dimerization and AXL autophosphorylation, thereby activating multiple downstream signal transduction pathways and participating in multiple processes of tumorigenesis (Linger, R.M et al., Ther.Targets, 14(10 ), 1073-1090, 2010; Rescigno, J. et al., Oncogene, 6(10), 1909-1913, 1991).
AXL广泛表达于人体正常组织,如单核细胞、巨噬细胞、血小板、内皮细胞、小脑、心脏、骨骼肌、肝脏和肾脏等,其中心肌和骨骼肌表达最高,骨髓CD34+细胞和基质细胞也有较高的表达,正常淋巴组织表达很低(Wu YM,Robinson DR,Kung HJ,Cancer Res,64(20),7311-7320,2004;hung BI等,DNA Cell Biol,22(8),533-540,2003)。在对许多癌细胞的研究中发现,在造血细胞、间质细胞和内皮细胞中,AXL基因都存在着超表达或异位表达。在各类白血病和多数的实体瘤中,AXL激酶的超表达现象尤为突出。通过抑制AXL受体酪氨酸激酶可以降低肿瘤细胞的促存活信号、阻滞肿瘤的侵袭能力,增加靶向药物治疗和化疗敏感度。因此寻找有效的AXL抑制剂是当前肿瘤靶向药物研发的重要方向。AXL is widely expressed in normal tissues of the human body, such as monocytes, macrophages, platelets, endothelial cells, cerebellum, heart, skeletal muscle, liver, and kidney, among which the expression is highest in cardiac muscle and skeletal muscle, and bone marrow CD34+ cells and stromal cells also have a higher expression High expression, very low expression in normal lymphoid tissue (Wu YM, Robinson DR, Kung HJ, Cancer Res, 64(20), 7311-7320, 2004; hung BI et al., DNA Cell Biol, 22(8), 533-540 , 2003). In the study of many cancer cells, it was found that AXL gene was overexpressed or ectopically expressed in hematopoietic cells, mesenchymal cells and endothelial cells. In various leukemias and most solid tumors, the overexpression of AXL kinase is particularly prominent. Inhibition of AXL receptor tyrosine kinase can reduce the pro-survival signals of tumor cells, block the invasion ability of tumors, and increase the sensitivity of targeted drug therapy and chemotherapy. Therefore, finding effective AXL inhibitors is an important direction for the development of tumor-targeted drugs.
发明内容Contents of the invention
一方面,本发明提供了式I化合物的药学上可接受的盐,所述盐选自有机酸盐或无机酸盐,其中所述有机酸盐选自甲磺酸盐、苯磺酸盐、草酸盐、富马酸盐、柠檬酸盐和马尿酸盐中的一种,所述无机酸盐选自盐酸盐、氢溴酸盐、硫酸盐或磷酸盐中的一种,式I化合物结构如下:In one aspect, the present invention provides a pharmaceutically acceptable salt of the compound of formula I, wherein the salt is selected from organic acid salts or inorganic acid salts, wherein the organic acid salts are selected from methanesulfonate, benzenesulfonate, oxalate One of salt, fumarate, citrate and hippurate, the inorganic acid salt is selected from one of hydrochloride, hydrobromide, sulfate or phosphate, the compound of formula I The structure is as follows:
Figure PCTCN2022134408-appb-000001
Figure PCTCN2022134408-appb-000001
在一些实施方案中,所述有机酸盐为甲磺酸盐。In some embodiments, the organic acid salt is mesylate.
在一些实施方案中,所述甲磺酸盐为甲磺酸盐的水合物。In some embodiments, the mesylate salt is a hydrate of the mesylate salt.
在一些实施方案中,所述甲磺酸盐为甲磺酸盐的二水合物。In some embodiments, the mesylate salt is the dihydrate of the mesylate salt.
在一些实施方案中,所述有机酸盐中式I化合物与有机酸的摩尔比为1:1。In some embodiments, the molar ratio of the compound of formula I to the organic acid in the organic acid salt is 1:1.
在一些实施方案中,所述无机酸盐中式I化合物与无机酸的摩尔比为1:1或1:2。In some embodiments, the molar ratio of the compound of formula I to the inorganic acid in the inorganic acid salt is 1:1 or 1:2.
在一些实施方案中,所述无机酸盐为盐酸盐。In some embodiments, the salt of an inorganic acid is a hydrochloride.
在一些实施方案中,所述盐酸盐中式I化合物与氯化氢的摩尔比为1:1或1:2。In some embodiments, the molar ratio of the compound of formula I to hydrogen chloride in the hydrochloride salt is 1:1 or 1:2.
在一些实施方案中,所述盐酸盐中式I化合物与氯化氢的摩尔比为1:2。In some embodiments, the molar ratio of the compound of formula I to hydrogen chloride in the hydrochloride salt is 1:2.
在一些实施方案中,所述硫酸盐中式I化合物与硫酸的摩尔比为1:1。In some embodiments, the molar ratio of the compound of formula I to sulfuric acid in the sulfate salt is 1:1.
在一些实施方案中,所述氢溴酸盐中式I化合物与氢溴酸的摩尔比为1:1。In some embodiments, the molar ratio of the compound of formula I to hydrobromic acid in the hydrobromide salt is 1:1.
在一些实施方案中,所述磷酸盐中式I化合物与磷酸的摩尔比为1:1。In some embodiments, the molar ratio of the compound of formula I to phosphoric acid in the phosphate salt is 1:1.
可以理解为,本发明所说的盐是式I化合物与相应的酸通过成盐反应获得,在反应中,式I化合物转化为阳离子,与相应的酸的酸根结合,形成所述盐。因此,本发明中式I化合物与酸的摩尔比可以理解为盐中式I化合物的阳离子与相应酸的酸根的摩尔比。It can be understood that the salt in the present invention is obtained by a salt-forming reaction between the compound of formula I and the corresponding acid. In the reaction, the compound of formula I is converted into a cation and combined with the acid radical of the corresponding acid to form the salt. Therefore, the molar ratio of the compound of formula I to the acid in the present invention can be understood as the molar ratio of the cation of the compound of formula I to the acid group of the corresponding acid in the salt.
在一些典型实施方案中,本发明提供了式I化合物的甲磺酸盐,其中式I化合物与甲磺酸的摩尔比为1:1,或式I化合物的阳离子与甲磺酸的酸根的摩尔比为1:1。In some typical embodiments, the present invention provides the mesylate salt of the compound of formula I, wherein the molar ratio of the compound of formula I to methanesulfonic acid is 1:1, or the molar ratio of the cation of the compound of formula I to the acid group of methanesulfonic acid The ratio is 1:1.
另一方面,本发明提供了式I化合物的药学上可接受的盐的晶型,所述盐选自有机酸盐或无机酸盐,其中有机酸盐选自甲磺酸盐、苯磺酸盐、草酸盐、富马酸盐、柠檬酸盐和马尿酸盐,所述无机酸盐选自盐酸盐、氢溴酸盐或磷酸盐。In another aspect, the present invention provides a crystal form of a pharmaceutically acceptable salt of the compound of formula I, wherein the salt is selected from organic acid salts or inorganic acid salts, wherein the organic acid salts are selected from methanesulfonate, benzenesulfonate , oxalate, fumarate, citrate and hippurate, the inorganic acid salt is selected from hydrochloride, hydrobromide or phosphate.
在一些实施方案中,本发明提供了式I化合物的甲磺酸盐的结晶形式。In some embodiments, the present invention provides a crystalline form of the mesylate salt of the compound of Formula I.
在一些实施方案中,所述甲磺酸盐结晶,其X射线粉末衍射图的2θ详见下表1:In some embodiments, the 2θ of the X-ray powder diffraction pattern of the mesylate salt crystal is detailed in Table 1 below:
表1式I化合物的甲磺酸盐晶型X射线粉末衍射图数据Table 1 The mesylate salt crystal form X-ray powder diffraction pattern data of formula I compound
2θ(°)2θ(°) 高度(counts)height (counts) I/I 0(%) I/I 0 (%)
5.3005.300 5454 17.117.1
7.7937.793 4646 14.514.5
9.1749.174 102102 32.032.0
10.47910.479 6060 18.918.9
13.27113.271 8383 26.226.2
15.41515.415 318318 100.0100.0
16.06716.067 9090 28.428.4
17.16417.164 8989 28.128.1
17.69317.693 8080 25.025.0
18.20118.201 4141 12.812.8
18.95918.959 8383 26.126.1
20.33220.332 212212 66.766.7
20.85320.853 3838 11.811.8
21.88321.883 9999 31.031.0
22.51922.519 2626 8.28.2
23.10123.101 117117 36.936.9
23.48423.484 5151 16.016.0
24.94924.949 3434 10.610.6
25.79225.792 3737 11.511.5
26.33126.331 2828 8.78.7
29.54429.544 8282 25.925.9
33.00633.006 2626 8.28.2
39.46339.463 7171 22.322.3
.
在一些实施方案中,本发明提供了式I化合物的甲磺酸盐的晶型,其X射线粉末衍射图如图1所示。In some embodiments, the present invention provides a crystalline form of the mesylate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 1 .
在一些实施方案中,本发明提供了式I化合物的单盐酸盐的结晶形式。In some embodiments, the present invention provides a crystalline form of the monohydrochloride salt of a compound of formula I.
在一些实施方案中,所述单盐酸盐结晶,其X射线粉末衍射图的2θ详见下表2:In some embodiments, the 2θ of the X-ray powder diffraction pattern of the monohydrochloride crystal is detailed in Table 2 below:
表2式I化合物的单盐酸盐晶型X射线粉末衍射图数据Table 2 The monohydrochloride crystal form X-ray powder diffraction pattern data of the compound of formula I
Figure PCTCN2022134408-appb-000002
Figure PCTCN2022134408-appb-000002
Figure PCTCN2022134408-appb-000003
Figure PCTCN2022134408-appb-000003
在一些实施方案中,本发明提供了式I化合物的单盐酸盐的晶型,其X射线粉末衍射图如图4所示。在一些实施方案中,本发明提供了式I化合物的二盐酸盐的结晶形式。In some embodiments, the present invention provides a crystalline form of the monohydrochloride salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 4 . In some embodiments, the present invention provides a crystalline form of the dihydrochloride salt of the compound of formula I.
在一些实施方案中,所述二盐酸盐结晶,其X射线粉末衍射图的2θ详见下表3:In some embodiments, the dihydrochloride salt is crystalline, and the 2θ of its X-ray powder diffraction pattern is detailed in Table 3 below:
表3式I化合物的二盐酸盐晶型X射线粉末衍射图数据Table 3 The dihydrochloride salt crystal form X-ray powder diffraction pattern data of the compound of formula I
Figure PCTCN2022134408-appb-000004
Figure PCTCN2022134408-appb-000004
Figure PCTCN2022134408-appb-000005
Figure PCTCN2022134408-appb-000005
在一些实施方案中,本发明提供了式I化合物的二盐酸盐的晶型,其X射线粉末衍射图如图5所示。In some embodiments, the present invention provides a crystal form of the dihydrochloride salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 5 .
在一些实施方案中,本发明提供了式I化合物的磷酸盐的结晶形式。In some embodiments, the present invention provides a crystalline form of the phosphate salt of a compound of formula I.
在一些实施方案中,所述磷酸盐结晶,其X射线粉末衍射图的2θ详见下表4:In some embodiments, the 2θ of the X-ray powder diffraction pattern of the phosphate crystal is detailed in Table 4 below:
表4式I化合物的磷酸盐晶型X射线粉末衍射图数据Phosphate crystal form X-ray powder diffraction pattern data of the compound of formula I in table 4
2θ(°)2θ(°) 高度(counts)height (counts) I/I 0(%) I/I 0 (%)
4.9214.921 3434 22.322.3
6.5956.595 77 4.84.8
9.7679.767 153153 100.0100.0
10.99410.994 23twenty three 15.415.4
11.73211.732 24twenty four 15.615.6
13.38813.388 4545 29.829.8
14.56914.569 4949 32.432.4
15.32215.322 2020 12.912.9
16.03016.030 1313 8.78.7
18.43918.439 5555 36.036.0
19.04319.043 6666 43.343.3
20.48420.484 1717 11.311.3
21.34321.343 22twenty two 14.214.2
22.58422.584 4646 29.929.9
24.36624.366 8282 53.853.8
24.89024.890 6060 39.039.0
25.99425.994 6060 39.139.1
29.30529.305 7272 47.347.3
32.09632.096 77 4.94.9
33.98833.988 1313 8.38.3
35.57335.573 99 5.95.9
37.02237.022 1212 7.97.9
39.39139.391 99 5.85.8
.
在一些实施方案中,本发明提供了式I化合物的磷酸盐的晶型,其X射线粉末衍射图如图6所示。In some embodiments, the present invention provides a crystalline form of the phosphate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 6 .
在一些实施方案中,本发明提供了式I化合物的马尿酸盐的结晶形式。In some embodiments, the present invention provides a crystalline form of a hippurate salt of a compound of Formula I.
在一些实施方案中,所述马尿酸盐结晶,其X射线粉末衍射图的2θ详见下表5:In some embodiments, the 2θ of the X-ray powder diffraction pattern of the hippurate crystal is detailed in Table 5 below:
表5式I化合物的马尿酸盐晶型X射线粉末衍射图数据Table 5 The hippurate crystal form X-ray powder diffraction pattern data of the compound of formula I
Figure PCTCN2022134408-appb-000006
Figure PCTCN2022134408-appb-000006
Figure PCTCN2022134408-appb-000007
Figure PCTCN2022134408-appb-000007
在一些实施方案中,本发明提供了式I化合物的马尿酸盐的晶型,其X射线粉末衍射图如图7所示。In some embodiments, the present invention provides a crystalline form of hippurate of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 7 .
在一些实施方案中,本发明提供了式I化合物的硫酸盐的无定型。In some embodiments, the present invention provides an amorphous form of the sulfate salt of the compound of formula I.
在一些实施方案中,本发明提供了式I化合物的氢溴酸盐的结晶形式。In some embodiments, the present invention provides a crystalline form of the hydrobromide salt of a compound of formula I.
在一些实施方案中,所述氢溴酸盐结晶,其X射线粉末衍射图的2θ详见下表6:In some embodiments, the 2θ of the hydrobromide crystal is shown in Table 6 below in detail in its X-ray powder diffraction pattern:
表6式I化合物的氢溴酸盐晶型X射线粉末衍射图数据The hydrobromide salt crystal form X-ray powder diffraction pattern data of table 6 formula I compound
2θ(°)2θ(°) 高度(counts)height (counts) I/I 0(%) I/I 0 (%)
5.6715.671 1515 6.66.6
8.4578.457 5656 24.224.2
11.31811.318 145145 62.562.5
12.02912.029 6969 29.929.9
12.92012.920 205205 88.288.2
13.21913.219 110110 47.447.4
13.75313.753 181181 78.278.2
14.79114.791 4242 17.917.9
16.77916.779 143143 61.561.5
18.05718.057 112112 48.348.3
21.01021.010 141141 61.061.0
21.48421.484 121121 52.152.1
22.05622.056 6262 26.626.6
22.61422.614 9898 42.242.2
23.27023.270 232232 100.0100.0
24.12924.129 181181 77.977.9
24.90724.907 9494 40.440.4
25.63625.636 148148 63.763.7
27.15527.155 100100 43.043.0
28.29628.296 5757 24.724.7
29.46429.464 3232 13.713.7
33.01933.019 3232 13.613.6
34.69934.699 6767 28.828.8
.
在一些实施方案中,本发明提供了式I化合物的氢溴酸盐的晶型,其X射线粉末衍射图如图9所示。In some embodiments, the present invention provides a crystal form of the hydrobromide salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 9 .
在一些实施方案中,本发明提供了式I化合物的苯磺酸盐的结晶形式。In some embodiments, the present invention provides a crystalline form of the besylate salt of the compound of formula I.
在一些实施方案中,所述苯磺酸盐结晶,其X射线粉末衍射图的2θ详见下表7:In some embodiments, the benzenesulfonate salt is crystalline, and the 2θ of its X-ray powder diffraction pattern is detailed in Table 7 below:
表7苯磺酸盐晶型X射线粉末衍射图数据Table 7 Besylate crystal form X-ray powder diffraction pattern data
2θ(°)2θ(°) 高度(counts)height (counts) I/I 0(%) I/I 0 (%)
10.98010.980 2525 12.312.3
12.14412.144 9090 43.743.7
13.60113.601 6060 29.529.5
15.20015.200 125125 60.860.8
17.06617.066 116116 56.656.6
18.53618.536 205205 100.0100.0
19.38419.384 9999 48.548.5
20.16020.160 111111 54.054.0
21.25021.250 7373 35.735.7
21.97521.975 203203 99.299.2
24.50524.505 154154 75.075.0
26.75926.759 7878 38.038.0
27.89727.897 4747 23.123.1
30.58030.580 3636 17.517.5
32.11532.115 1818 8.68.6
.
在一些实施方案中,本发明提供了式I化合物的苯磺酸盐的晶型,其X射线粉末衍射图如图10所示。In some embodiments, the present invention provides a crystalline form of the besylate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 10 .
在一些实施方案中,本发明提供了式I化合物的草酸盐的结晶形式。In some embodiments, the present invention provides a crystalline form of the oxalate salt of the compound of formula I.
在一些实施方案中,所述草酸盐结晶,其其X射线粉末衍射图的2θ详见下表8:In some embodiments, the oxalate salt crystals, the 2θ of its X-ray powder diffraction pattern are detailed in Table 8 below:
表8式I化合物的草酸盐晶型X射线粉末衍射图数据Table 8 X-ray powder diffraction pattern data of the oxalate salt crystal form of the compound of formula I
2θ(°)2θ(°) 高度(counts)height (counts) I/I 0(%) I/I 0 (%)
5.9125.912 9494 14.614.6
8.8348.834 174174 26.926.9
10.27010.270 114114 17.717.7
11.76011.760 7878 12.112.1
14.71014.710 646646 100.0100.0
17.65617.656 485485 75.075.0
18.22918.229 7272 11.211.2
19.31819.318 5454 8.48.4
19.72019.720 6060 9.49.4
21.56421.564 194194 30.030.0
25.44125.441 103103 16.016.0
26.59026.590 121121 18.718.7
27.01627.016 100100 15.515.5
32.37532.375 77 1.01.0
34.95734.957 2929 4.54.5
.
在一些实施方案中,本发明提供了式I化合物的草酸盐的晶型,其X射线粉末衍射图如图11所示。In some embodiments, the present invention provides the crystalline form of the oxalate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 11 .
在一些实施方案中,本发明提供了式I化合物的富马酸盐的结晶形式。In some embodiments, the present invention provides a crystalline form of the fumarate salt of the compound of formula I.
在一些实施方案中,所述富马酸盐结晶,其X射线粉末衍射图的2θ详见下表9:In some embodiments, the 2θ of the X-ray powder diffraction pattern of the fumarate crystals is detailed in Table 9 below:
表9式I化合物的富马酸盐晶型X射线粉末衍射图数据Table 9 Fumarate salt crystal form X-ray powder diffraction pattern data of the compound of formula I
Figure PCTCN2022134408-appb-000008
Figure PCTCN2022134408-appb-000008
Figure PCTCN2022134408-appb-000009
Figure PCTCN2022134408-appb-000009
在一些实施方案中,本发明提供了式I化合物的富马酸盐的晶型,其X射线粉末衍射图如图12所示。In some embodiments, the present invention provides a crystalline form of the fumarate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 12 .
在一些实施方案中,本发明提供了式I化合物的柠檬酸盐的结晶形式。In some embodiments, the present invention provides a crystalline form of the citrate salt of the compound of formula I.
在一些实施方案中,所述柠檬酸盐结晶,其X射线粉末衍射图的2θ详见下表10:In some embodiments, the 2θ of the X-ray powder diffraction pattern of the citrate crystals is detailed in Table 10 below:
表10式I化合物的柠檬酸盐晶型X射线粉末衍射图数据The citrate crystal form X-ray powder diffraction pattern data of the compound of formula I in table 10
2θ(°)2θ(°) 高度(counts)height (counts) I/I 0(%) I/I 0 (%)
18.13818.138 9090 100.0100.0
19.46719.467 6363 70.070.0
26.07226.072 5555 61.561.5
31.10131.101 1414 15.415.4
.
在一些实施方案中,本发明提供了式I化合物的柠檬酸盐的晶型,其X射线粉末衍射图如图13所示。In some embodiments, the present invention provides a crystalline form of the citrate salt of the compound of formula I, the X-ray powder diffraction pattern of which is shown in FIG. 13 .
另一方面,本发明提供了一种式I化合物的晶型A,其X射线粉末衍射图在2θ为7.6°±0.2°、10.2°±0.2°、17.6°±0.2°、20.3°±0.2°和20.9°±0.2°处具有衍射峰。In another aspect, the present invention provides a crystal form A of the compound of formula I, whose X-ray powder diffraction pattern is 7.6°±0.2°, 10.2°±0.2°, 17.6°±0.2°, 20.3°±0.2° at 2θ There are diffraction peaks at 20.9°±0.2°.
进一步地,所述晶型A,其X射线粉末衍射图在2θ为4.1°±0.2°、7.6°±0.2°、10.2°±0.2°、12.6°±0.2°、13.0°±0.2°、17.6°±0.2°、19.7°±0.2°、20.3°±0.2°、20.9°±0.2°和22.2°±0.2°处具有衍射峰。Further, the crystal form A has an X-ray powder diffraction pattern at 2θ of 4.1°±0.2°, 7.6°±0.2°, 10.2°±0.2°, 12.6°±0.2°, 13.0°±0.2°, 17.6° There are diffraction peaks at ±0.2°, 19.7°±0.2°, 20.3°±0.2°, 20.9°±0.2° and 22.2°±0.2°.
进一步地,所述晶型A,其X射线粉末衍射图在2θ为4.1°±0.2°、5.6°±0.2°、7.6°±0.2°、10.2°±0.2°、10.9°±0.2°、12.6°±0.2°、13.0°±0.2°、15.2°±0.2°、17.6°±0.2°、19.7°±0.2°、20.3°±0.2°、20.9°±0.2°、22.2°±0.2°、23.2°±0.2°、24.6°±0.2°、27.0°±0.2°、28.8°±0.2°、37.0°±0.2°和37.7°±0.2°处具有衍射峰。Further, the crystal form A has an X-ray powder diffraction pattern at 2θ of 4.1°±0.2°, 5.6°±0.2°, 7.6°±0.2°, 10.2°±0.2°, 10.9°±0.2°, 12.6° ±0.2°, 13.0°±0.2°, 15.2°±0.2°, 17.6°±0.2°, 19.7°±0.2°, 20.3°±0.2°, 20.9°±0.2°, 22.2°±0.2°, 23.2°±0.2 °, 24.6°±0.2°, 27.0°±0.2°, 28.8°±0.2°, 37.0°±0.2° and 37.7°±0.2° have diffraction peaks.
在一些实施方案中,所述晶型A,其X射线粉末衍射图的2θ详见下表:In some embodiments, the 2θ of the X-ray powder diffraction pattern of the crystal form A is detailed in the table below:
表11晶型A的X射线粉末衍射图数据Table 11 X-ray powder diffraction pattern data of crystal form A
Figure PCTCN2022134408-appb-000010
Figure PCTCN2022134408-appb-000010
Figure PCTCN2022134408-appb-000011
Figure PCTCN2022134408-appb-000011
在一些实施方案中,所述晶型A以2θ角度表示的X射线粉末衍射具有如图14所示的图谱。In some embodiments, the X-ray powder diffraction of the crystal form A in 2θ angle has a pattern as shown in FIG. 14 .
另一方面,本发明提供了式I化合物的药学上可接受的盐或其药学上可接受的盐的晶型的制备方法,其包括将式I化合物与相应的酸成盐的步骤。In another aspect, the present invention provides a method for preparing a pharmaceutically acceptable salt of the compound of formula I or a crystal form of the pharmaceutically acceptable salt thereof, which comprises the step of forming a salt of the compound of formula I with a corresponding acid.
在一些实施方案中,所述制备方法的反应溶剂选自醇类溶剂与烷烃类溶剂的混合溶剂、酮类溶剂与烷烃类溶剂的混合溶剂、酯类溶剂与烷烃类溶剂的混合溶剂、腈类-水类溶剂与烷烃类溶剂的混合溶剂、烷苯类溶剂与烷烃类溶剂的混合溶剂或卤代烃类溶剂与烷烃类溶剂的混合溶剂。In some embodiments, the reaction solvent of the preparation method is selected from mixed solvents of alcohol solvents and alkane solvents, mixed solvents of ketone solvents and alkane solvents, mixed solvents of ester solvents and alkane solvents, nitrile solvents - A mixed solvent of an aqueous solvent and an alkane solvent, a mixed solvent of an alkylbenzene solvent and an alkane solvent, or a mixed solvent of a halogenated hydrocarbon solvent and an alkane solvent.
在一些实施方案中,所述醇类溶剂选自甲醇、乙醇或异丙醇;所述酮类溶剂选自丙酮或丁酮;优选丙酮;所述酯类溶剂选自乙酸乙酯或乙酸丁酯;优选乙酸乙酯;所述腈类-水类溶剂选自腈类-水的混合溶液,所述烷烃类溶剂选自正庚烷。In some embodiments, the alcohol solvent is selected from methanol, ethanol or isopropanol; the ketone solvent is selected from acetone or butanone; preferably acetone; the ester solvent is selected from ethyl acetate or butyl acetate ; Preferred ethyl acetate; The nitriles-water solvent is selected from the mixed solution of nitriles-water, and the alkane solvent is selected from n-heptane.
在一些实施方案中,所述药学上可接受的盐的晶型为甲磺酸盐晶型,所述甲磺酸盐晶型通过包括以下步骤的方法制备得到:In some embodiments, the crystal form of the pharmaceutically acceptable salt is a mesylate salt crystal form, and the mesylate salt crystal form is prepared by a method comprising the following steps:
S1:将式I化合物、甲磺酸与甲苯搅拌得到式I化合物的甲磺酸盐晶型的晶种;S1: stirring the compound of formula I, methanesulfonic acid and toluene to obtain a seed crystal of the mesylate salt crystal form of the compound of formula I;
S2:将式(I)化合物、甲磺酸与乙酸乙酯搅拌反应后加入步骤S1得到的晶型继续搅拌;S2: stirring and reacting the compound of formula (I), methanesulfonic acid and ethyl acetate, adding the crystal form obtained in step S1 and continuing to stir;
S3:加入乙酸乙酯、丙酮和权利要求8所述的式I化合物的晶型,搅拌析晶得到所述甲磺酸盐晶型(或称甲磺酸盐晶型II)。S3: adding ethyl acetate, acetone and the crystal form of the compound of formula I according to claim 8, stirring and crystallizing to obtain the mesylate salt crystal form (or called mesylate salt crystal form II).
另一方面,本发明还提供了包含所述式I化合物的药学上可接受的盐的药物组合物。In another aspect, the present invention also provides a pharmaceutical composition comprising a pharmaceutically acceptable salt of the compound of formula I.
在一些实施方案中,所述药物组合物进一步包含一种或多种药学上可接受的载体。In some embodiments, the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
在一些实施方案中,所述药物组合物为适于口服的固体药物制剂,优选片剂或胶囊。In some embodiments, the pharmaceutical composition is a solid pharmaceutical preparation suitable for oral administration, preferably a tablet or a capsule.
另一方面,本发明还提供了包含所述式I化合物的药学上可接受的盐的晶型的药物组合物。In another aspect, the present invention also provides a pharmaceutical composition comprising the crystal form of the pharmaceutically acceptable salt of the compound of formula I.
在一些实施方案中,所述药物组合物进一步包含一种或多种药学上可接受的载体。In some embodiments, the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
在一些实施方案中,所述药物组合物为适于口服的固体药物制剂,优选片剂或胶囊。In some embodiments, the pharmaceutical composition is a solid pharmaceutical preparation suitable for oral administration, preferably a tablet or a capsule.
另一方面,本发明提供一种晶型组合物,其中上述式I化合物的药学上可接受的盐、上述式I化合物的药学上可接受的盐的晶型占所述晶型组合物重量的50%以上。In another aspect, the present invention provides a crystal form composition, wherein the pharmaceutically acceptable salt of the compound of formula I above, and the crystal form of the pharmaceutically acceptable salt of the compound of formula I account for the weight of the crystal form composition above 50.
进一步占所述晶型组合物重量的80%以上;Further accounting for more than 80% of the weight of the crystal composition;
进一步占所述晶型组合物重量的90%以上;Further accounting for more than 90% of the weight of the crystal composition;
进一步占所述晶型组合物重量的95%以上。It further accounts for more than 95% of the weight of the crystal composition.
另一方面,本发明还提供了包含上述晶型组合物的药物组合物;所述药物组合物进一步包含一种或多种药学上可接受的载体。On the other hand, the present invention also provides a pharmaceutical composition comprising the above crystal composition; the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
另一方面,本发明还提供了用作药物的式I化合物的药学上可接受的盐或其晶型组合物或其药物组合物。On the other hand, the present invention also provides a pharmaceutically acceptable salt of the compound of formula I or its crystal form composition or its pharmaceutical composition for use as a medicine.
另一方面,本发明还提供了用作药物的式I化合物的药学上可接受的盐的晶型或其晶型组合物或其药物组合物。On the other hand, the present invention also provides a pharmaceutically acceptable salt crystal form of the compound of formula I or its crystal form composition or pharmaceutical composition thereof for use as a medicine.
另一方面,本发明还提供了用于预防和/或治疗AXL激酶介导的疾病或疾病状态的方法,其包括向有需要的个体给予本发明的所述式I化合物的盐、其晶型组合物或其药物组合物。In another aspect, the present invention also provides a method for preventing and/or treating AXL kinase-mediated diseases or disease states, which comprises administering the salt of the compound of formula I of the present invention, or its crystal form, to individuals in need Composition or its pharmaceutical composition.
另一方面,本发明还提供了用于预防和/或治疗AXL激酶介导的疾病或疾病状态的方法,其包括向有需要的个体给予本发明的所述式I化合物的盐的晶型、其晶型组合物或其药物组合物。On the other hand, the present invention also provides a method for preventing and/or treating AXL kinase-mediated diseases or disease states, which comprises administering the crystalline form of the salt of the compound of formula I of the present invention, Its crystal form composition or its pharmaceutical composition.
另一方面,本发明还提供了用于预防和/或治疗AXL激酶介导的疾病或疾病状态的本发明的所述式I化合物的 盐、其晶型组合物或其药物组合物。On the other hand, the present invention also provides the salt of the compound of formula I of the present invention, its crystal form composition or its pharmaceutical composition for preventing and/or treating AXL kinase-mediated diseases or disease states.
另一方面,本发明还提供了用于预防和/或治疗AXL激酶介导的疾病或疾病状态的本发明的所述式I化合物的盐的晶型、其晶型组合物或其药物组合物。In another aspect, the present invention also provides the crystalline form of the salt of the compound of formula I of the present invention, its crystalline form composition or its pharmaceutical composition for preventing and/or treating AXL kinase-mediated diseases or disease states .
在一些实施方案中,所述AXL激酶介导的疾病或疾病状态为癌症。In some embodiments, the AXL kinase-mediated disease or condition is cancer.
在一些典型的实施方案中,所述癌症为与血液肿瘤和实体瘤相关的疾病。In some exemplary embodiments, the cancer is a disease associated with hematological and solid tumors.
相关定义related definition
除非有特定说明,下列用在说明书和权利要求书中的术语具有下述含义:Unless otherwise specified, the following terms used in the specification and claims have the following meanings:
本发明的药学上可接受的盐还包括它们的水合物形式。The pharmaceutically acceptable salts of the present invention also include their hydrated forms.
术语“药学上可接受的载体”是指对机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些载体。包括但不限于国家食品药品监督管理局许可的可用于人或动物的任何稀释剂、崩解剂、粘合剂、助流剂、润湿剂。The term "pharmaceutically acceptable carrier" refers to those carriers that have no obvious stimulating effect on the body and will not impair the biological activity and performance of the active compound. Including but not limited to any diluents, disintegrants, binders, glidants, and wetting agents approved by the State Food and Drug Administration for human or animal use.
术语“富马酸”指反丁烯二酸,具有结构:
Figure PCTCN2022134408-appb-000012
The term "fumaric acid" refers to fumaric acid, which has the structure:
Figure PCTCN2022134408-appb-000012
术语“醇类溶剂”是指一个或多个羟基(OH)取代C1-C6烷烃上的一个或多个氢原子所衍生的物质,所述C1-C6烷烃是指含有1-6个碳原子的直链或支链的烷烃,醇类溶剂的具体实例包括但不限于:甲醇、乙醇、异丙醇或正丙醇。The term "alcohol solvent" refers to a substance derived from one or more hydroxyl groups (OH) replacing one or more hydrogen atoms on a C1-C6 alkane, and the C1-C6 alkane refers to a substance containing 1-6 carbon atoms Specific examples of linear or branched alkanes and alcohol solvents include, but are not limited to: methanol, ethanol, isopropanol or n-propanol.
术语“烷烃类溶剂”是指含有5-7个碳原子的直链或支链或环状的烷烃,具体实例包括但不限于正己烷、环己烷、正庚烷。The term "alkane solvent" refers to straight chain or branched chain or cyclic alkanes containing 5-7 carbon atoms, specific examples include but not limited to n-hexane, cyclohexane, n-heptane.
术语“酯类溶剂”是指含有酯基-COOR且碳原子数为3-10个的链状化合物,其中R为C1-C6烷基,所述C1-C6烷基是指含有1-6个碳原子的直链或支链烷烃,酯类溶剂的具体实例包括但不限于乙酸甲酯、乙酸乙酯、乙酸丙酯。The term "ester solvent" refers to a chain compound containing an ester group -COOR and a carbon number of 3-10, wherein R is a C1-C6 alkyl group, and the C1-C6 alkyl group refers to a chain compound containing 1-6 Specific examples of straight-chain or branched-chain alkanes with carbon atoms and ester solvents include but are not limited to methyl acetate, ethyl acetate, and propyl acetate.
术语“卤代烃类溶剂”是指一个或多个卤素原子取代C1-C6烷烃上的一个或多个氢原子所衍生的物质,所述C1-C6烷烃是指含有1-6个碳原子的直链或支链的烷烃,所述卤素原子是指氟、氯、溴、碘,卤代烃类溶剂的具体实例包括但不限于二氯甲烷或氯仿。The term "halogenated hydrocarbon solvent" refers to a substance derived from one or more halogen atoms replacing one or more hydrogen atoms on a C1-C6 alkane, and the C1-C6 alkane refers to a substance containing 1-6 carbon atoms For linear or branched alkanes, the halogen atoms refer to fluorine, chlorine, bromine, and iodine. Specific examples of halogenated hydrocarbon solvents include but are not limited to dichloromethane or chloroform.
术语“酮类溶剂”是指含有羰基-CO-且碳原子数为3-10个的链状或环状化合物,具体实例包括但不限于丙酮、丁酮或环己酮。The term "ketone solvent" refers to a chain or cyclic compound containing carbonyl -CO- and having 3-10 carbon atoms, specific examples include but not limited to acetone, methyl ethyl ketone or cyclohexanone.
术语“苯类溶剂”是指含有苯基的溶剂,具体实例包括甲苯、二甲苯、异丙苯或氯苯。The term "benzene-based solvent" means a solvent containing a phenyl group, and specific examples include toluene, xylene, cumene, or chlorobenzene.
术语“当量”是指按照化学反应的当量关系,以每步骤中所用基本原料为1当量,所需要的其他原料的当量用量。The term "equivalent" refers to the equivalent amount of other raw materials required in accordance with the equivalent relationship of chemical reactions, taking the basic raw materials used in each step as 1 equivalent.
本发明中的“X射线粉末衍射图谱”为使用Cu-Kα辐射测量得到。The "X-ray powder diffraction pattern" in the present invention is obtained by using Cu-Kα radiation measurement.
需要说明的是,在X射线粉末衍射光谱(XRPD)中,由结晶化合物得到的衍射谱图对于特定的结晶往往是特征性的,其中谱带(尤其是在低角度)的相对强度可能会因为结晶条件、粒径和其它测定条件的差异而产生的优势取向效果而变化。因此,衍射峰的相对强度对所针对的结晶并非是特征性的。判断是否与已知的结晶相同时,更应该注意的是峰的相对位置而不是它们的相对强度。此外,对任何给定的结晶而言,峰的位置可能存在轻微误差,这在结晶学领域中也是公知的。例如,由于分析样品时温度的变化、样品移动、或仪器的标定等,峰的位置可以移动,2θ值的测定误差有时约为±0.2°。因此,在确定每种结晶结构时,应该将此误差考虑在内。在XRPD图谱中通常用2θ角或晶面距d表示峰位置,两者之间具有简单的换算关系:d=λ/2sinθ,其中d代表晶面距,λ代表入射X射线的波长,θ为衍射角。对于同种化合物的同种结晶,其XRPD谱的峰位置在整体上具有相似性,相对强度误差可能较大。还应指出的是,在混合物的鉴定中,由于含量下降等因素会造成部分衍射线的缺失,此时,无需依赖高纯试样中观察到的全部谱带,甚至一条谱带也可能对给定的结晶是特征性的。It should be noted that in X-ray powder diffraction (XRPD), the diffraction pattern obtained from a crystalline compound is often characteristic for a particular crystal, where the relative intensity of the bands (especially at low angles) may vary due to The effect of dominant orientation due to differences in crystallization conditions, particle size and other measurement conditions varies. Therefore, the relative intensities of the diffraction peaks are not characteristic of the targeted crystals. When judging whether it is identical to a known crystal, more attention should be paid to the relative positions of the peaks rather than their relative intensities. Furthermore, for any given crystal there may be slight errors in the position of the peaks, as is well known in the art of crystallography. For example, due to temperature changes, sample movement, or instrument calibration when analyzing samples, the position of the peak can move, and the measurement error of the 2θ value is sometimes about ±0.2°. Therefore, this error should be taken into account when determining each crystal structure. In the XRPD spectrum, the 2θ angle or the crystal plane distance d is usually used to represent the peak position, and there is a simple conversion relationship between the two: d=λ/2sinθ, where d represents the crystal plane distance, λ represents the wavelength of the incident X-ray, and θ is Diffraction angle. For the same crystal of the same compound, the peak positions of their XRPD spectra are similar on the whole, and the relative intensity error may be large. It should also be pointed out that in the identification of mixtures, due to factors such as content decline, some diffraction lines will be missing. At this time, it is not necessary to rely on all the bands observed in the high-purity sample, and even one band may affect the given Certain crystallization is characteristic.
差示扫描量热法(DSC)测定当晶体由于其晶体结构发生变化或晶体熔融而吸收或释放热时的转变温度。对于同种化合物的同种晶型,在连续的分析中,热转变温度和熔点误差典型的在约5℃之内,通常在约3℃之内。当描述某个化合物具有某一给定的DSC峰或熔点时,指的是该DSC峰或熔点±5℃。DSC提供了一种辨别不同晶型的辅助方法。不同的晶体形态可根据其不同的转变温度特征而加以识别。需要指出的是对于混合物而言,其DSC峰或熔点可能会在更大的范围内波动。此外,由于在物质熔化的过程中伴有分解,因此熔化温度与升温速率相关。Differential Scanning Calorimetry (DSC) measures the transition temperature when a crystal absorbs or releases heat due to a change in its crystal structure or melting of the crystal. For the same crystal form of the same compound, thermal transition temperatures and melting points are typically within about 5°C, usually within about 3°C, in successive analyses. When it is described that a compound has a given DSC peak or melting point, it is meant that the DSC peak or melting point ± 5°C. DSC provides an auxiliary method to distinguish different crystal forms. Different crystal forms can be identified by their different transition temperature characteristics. It should be pointed out that for the mixture, its DSC peak or melting point may fluctuate in a larger range. In addition, since the melting process of the substance is accompanied by decomposition, the melting temperature is related to the heating rate.
热重分析(TGA)指的是在程序控制温度下测量待测样品的质量与温度变化之间关系的一种热分析技术。当被测物质在加热过程中有升华或汽化现象时,其分解出了气体或失去了结晶水时,引起被测物质量发生变化。这时, 热重曲线就不是直线而是有所下降。通过分析热重曲线,即可知道被测物质在什么温度下产生变化,并且根据所失重量,可计算失去了多少物质量。Thermogravimetric analysis (TGA) refers to a thermal analysis technique that measures the relationship between the mass of the sample to be tested and the temperature change at a programmed temperature. When the measured substance sublimes or vaporizes during the heating process, it decomposes into gas or loses crystal water, causing the quantity of the measured substance to change. At this time, the thermogravimetric curve is not a straight line but a decline. By analyzing the thermogravimetric curve, you can know at what temperature the measured substance changes, and according to the lost weight, you can calculate how much the substance is lost.
在提到例如XRPD图谱、DSC图谱或TGA图谱时,术语“如……所示”包括与本文描绘的那些不一定相同,但在被本领域技术人员考虑时落入实验误差的限度内的图谱。When referring to, for example, an XRPD pattern, DSC pattern or TGA pattern, the term "as shown" includes patterns that are not necessarily identical to those depicted herein, but which fall within the limits of experimental error when considered by those skilled in the art .
如无特殊说明,本发明的简称具有如下含义:Unless otherwise specified, the abbreviation of the present invention has the following meanings:
M:mol/LM: mol/L
mM:mmol/LmM: mmol/L
nM:nmol/LnM: nmol/L
Boc:叔丁氧羰基Boc: tert-butoxycarbonyl
1H NMR:核磁共振氢谱 1 H NMR: Proton Magnetic Resonance Spectroscopy
MS(ESI+):质谱MS(ESI+): mass spectrum
DMSO-d 6:氘代二甲基亚砜 DMSO-d 6 : deuterated dimethyl sulfoxide
CDCl 3:氘代氯仿 CDCl 3 : deuterated chloroform
DTT:二硫苏糖醇DTT: Dithiothreitol
SEB:Supplemented Enzymatic Buffer(补充酶缓冲液)SEB: Supplemented Enzymatic Buffer (Supplemented Enzymatic Buffer)
IMDM(Iscove's Modified Dulbecco's Medium):Iscove(人名)改良的Dulbecco(人名)培养基。IMDM (Iscove's Modified Dulbecco's Medium): Iscove (person's name) modified Dulbecco (person's name) medium.
室温:25℃。Room temperature: 25°C.
附图说明Description of drawings
为了更清楚地说明本发明实施例和现有技术的技术方案,下面对实施例和现有技术中所需要使用的附图作简单地介绍,显而易见地,下面描述中的附图仅仅是本发明的一些实施例,对于本领域普通技术人员来讲,还可以根据这些附图获得其他的附图。In order to more clearly illustrate the embodiments of the present invention and the technical solutions of the prior art, the following briefly introduces the accompanying drawings that need to be used in the embodiments and the prior art. Obviously, the accompanying drawings in the following description are only examples For some embodiments of the invention, those skilled in the art can also obtain other drawings according to these drawings.
图1显示实施例5中式I化合物甲磺酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 1 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound mesylate in embodiment 5;
图2显示实施例5中式I化合物甲磺酸盐的差示扫描量热(DSC)图;Fig. 2 shows the differential scanning calorimetry (DSC) figure of formula I compound mesylate in embodiment 5;
图3显示实施例5中式I化合物甲磺酸盐的热重(TGA)图;Fig. 3 shows the thermogravimetric (TGA) figure of formula I compound mesylate in embodiment 5;
图4显示实施例3中式I化合物单盐酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 4 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound monohydrochloride in embodiment 3;
图5显示实施例3中式I化合物二盐酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 5 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound dihydrochloride in embodiment 3;
图6显示实施例3中式I化合物磷酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 6 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound phosphate in embodiment 3;
图7显示实施例3中式I化合物马尿酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 7 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound hippurate in embodiment 3;
图8显示实施例3中式I化合物硫酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 8 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound sulfate in embodiment 3;
图9显示实施例3中式I化合物氢溴酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 9 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound hydrobromide in embodiment 3;
图10显示实施例3中式I化合物苯磺酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 10 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound besylate in embodiment 3;
图11显示实施例3中式I化合物草酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 11 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound oxalate in embodiment 3;
图12显示实施例3中式I化合物富马酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 12 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound fumarate in embodiment 3;
图13显示实施例3中式I化合物柠檬酸盐的X-射线粉末衍射(XRPD)谱图;Fig. 13 shows the X-ray powder diffraction (XRPD) spectrogram of formula I compound citrate in embodiment 3;
图14显示实施例6中式I化合物的晶型A的X-射线粉末衍射(XRPD)谱图。Figure 14 shows the X-ray powder diffraction (XRPD) spectrum of the crystal form A of the compound of formula I in Example 6.
具体实施方式Detailed ways
下面通过实施例更详细地描述本发明。但这些具体描述仅用于说明本发明的技术方案,不对本发明构成任何限制。The present invention is described in more detail below by way of examples. However, these specific descriptions are only used to illustrate the technical solution of the present invention, and do not constitute any limitation to the present invention.
各仪器测试条件如下:The test conditions of each instrument are as follows:
(1)X-射线粉末衍射仪(X-ray Powder Diffraction,XRPD)(1) X-ray powder diffractometer (X-ray Powder Diffraction, XRPD)
仪器型号:Bruker D2 Phaser 2 nd Instrument model: Bruker D2 Phaser 2 nd
Figure PCTCN2022134408-appb-000013
Figure PCTCN2022134408-appb-000013
Figure PCTCN2022134408-appb-000014
Figure PCTCN2022134408-appb-000014
(2)热重分析仪(Thermogravimetric,TGA)(2) Thermogravimetric Analyzer (Thermogravimetric, TGA)
仪器型号:TA Instruments TGA25Instrument model: TA Instruments TGA25
吹扫气:氮气Purge gas: Nitrogen
升温速率:10℃/minHeating rate: 10°C/min
升温范围:室温-300℃Temperature rise range: room temperature - 300°C
方法:将样品置于铝盘中,再将铝盘置于铂盘中,敞口在氮气氛围中以10℃/min的速度从室温升温至设定的温度。Method: Put the sample in an aluminum pan, then place the aluminum pan in a platinum pan, expose it in a nitrogen atmosphere, and raise the temperature from room temperature to the set temperature at a rate of 10°C/min.
(3)差示扫描量热仪(Differential Scanning Calorimeter,DSC)(3) Differential Scanning Calorimeter (DSC)
仪器型号:TA Instruments DSC25Instrument model: TA Instruments DSC25
吹扫气:氮气Purge gas: Nitrogen
升温速率:10℃/minHeating rate: 10°C/min
升温范围:20-300℃Heating range: 20-300°C
方法:样品置于铝盘中,压盖后在氮气氛围中以10℃/min的速度从20℃升温至设定的温度。Method: The sample was placed in an aluminum pan, and after capping, the temperature was raised from 20°C to the set temperature at a rate of 10°C/min in a nitrogen atmosphere.
(4)动态水分吸附(DVS)(4) Dynamic moisture adsorption (DVS)
仪器型号:Surface Measurement System(SMS)-DVS IntrinsicInstrument model: Surface Measurement System (SMS)-DVS Intrinsic
Figure PCTCN2022134408-appb-000015
Figure PCTCN2022134408-appb-000015
具体的仪器设定参数如下:The specific instrument setting parameters are as follows:
实施例1(S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基)嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦的制备Example 1 (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6,7,8,9-tetrahydro-5H-benzo[7] wheel Preparation of En-2-yl)amino)pyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethylphosphine oxide
Figure PCTCN2022134408-appb-000016
Figure PCTCN2022134408-appb-000016
a)2-碘-4-(甲氧基甲基)苯胺a) 2-iodo-4-(methoxymethyl)aniline
在二氯甲烷(261mL)/水(135mL)的溶液中加入4-(甲氧基甲基)苯胺(9g)、碘(16.65g)和碳酸氢钠(16.53g),22℃下搅拌16h。反应液用饱和硫代硫酸钠(10ml)在室温下下猝灭。所得混合物用二氯甲烷(3x 100mL)萃取,接着用饱和氯化钠水溶液(1x100mL)洗涤合并的有机层,有机层再用无水硫酸钠干燥。过滤后,滤液减压浓缩。残渣经硅胶柱层析纯化(石油醚/乙酸乙酯=1/1v/v),得到标题产物(16g)。MS(ESI+):264.0(M+H).4-(Methoxymethyl)aniline (9g), iodine (16.65g) and sodium bicarbonate (16.53g) were added to a solution of dichloromethane (261mL)/water (135mL), and stirred at 22°C for 16h. The reaction was quenched with saturated sodium thiosulfate (10 ml) at room temperature. The resulting mixture was extracted with dichloromethane (3 x 100 mL), then the combined organic layers were washed with saturated aqueous sodium chloride (1 x 100 mL), and the organic layer was dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1 v/v) to obtain the title product (16 g). MS(ESI+):264.0(M+H).
b)(2-氨基-5-(甲氧基甲基)苯基)二甲基氧化膦b) (2-Amino-5-(methoxymethyl)phenyl)dimethylphosphine oxide
在氮气气氛下,向N,N-二甲基甲酰胺(224mL)中加入2-碘-4-(甲氧基甲基)苯胺(16g,60.82mmol,1.00当量)、磷酸钾(14.20g)、醋酸钯(0.68g)和4,5-双二苯基膦-9,9-二甲基氧杂蒽(1.76g)的搅拌溶液中添加二甲基氧化膦(5.22g),于120℃下搅拌反应2小时。将混合物冷却至室温。过滤所得混合物,用N,N-二甲基甲酰胺(3x5mL)洗涤滤饼。滤液减压浓缩。用硅胶柱层析(二氯甲烷/甲醇=20/1v/v)纯化残余物得到标题产物(12.9g)。MS(ESI+):214.1(M+H).To N,N-dimethylformamide (224 mL) was added 2-iodo-4-(methoxymethyl)aniline (16 g, 60.82 mmol, 1.00 equiv), potassium phosphate (14.20 g) under nitrogen atmosphere , palladium acetate (0.68g) and 4,5-bisdiphenylphosphine-9,9-dimethylxanthene (1.76g) in a stirred solution was added dimethylphosphine oxide (5.22g), at 120 ° C The reaction was stirred for 2 hours. The mixture was cooled to room temperature. The resulting mixture was filtered and the filter cake was washed with N,N-dimethylformamide (3x5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane/methanol=20/1 v/v) to obtain the title product (12.9 g). MS(ESI+):214.1(M+H).
c)(2-((2,5-二氯嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦c) (2-((2,5-dichloropyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethylphosphine oxide
在室温下向N,N-二甲基甲酰胺(22mL)中加入(2-氨基-5-(甲氧基甲基)苯基)二甲基氧化膦(1.10g)、2,4,5-三氯嘧啶(1.23g)和N,N-二异丙基乙胺(2.00g)搅拌3h。所得混合物用二氯甲烷(30mL)稀释。在0℃下加水(10ml)使反应猝灭。所得混合物用二氯甲烷(3x 50mL)萃取。合并的有机层用饱和氯化钠(1x50mL)洗涤并用无水硫酸钠干燥。过滤后,滤液减压浓缩。用硅胶柱层析(二氯甲烷/甲醇=20/1v/v)纯化残余物得到标题产物(1.28g)。To N,N-dimethylformamide (22 mL) was added (2-amino-5-(methoxymethyl)phenyl)dimethylphosphine oxide (1.10 g), 2,4,5 - Trichloropyrimidine (1.23g) and N,N-diisopropylethylamine (2.00g) were stirred for 3h. The resulting mixture was diluted with dichloromethane (30 mL). The reaction was quenched by adding water (10ml) at 0°C. The resulting mixture was extracted with dichloromethane (3 x 50 mL). The combined organic layers were washed with saturated sodium chloride (1x50 mL) and dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane/methanol=20/1 v/v) to obtain the title product (1.28 g).
MS(ESI+):360.0(M+H).MS(ESI+):360.0(M+H).
d)(S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基)嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦d) (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6,7,8,9-tetrahydro-5H-benzo[7]annulene -2-yl)amino)pyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethylphosphine oxide
向异丙醇(2mL)中加入(2-((2,5-二氯嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦(50.00mg)和(S)-7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]环烯-2-胺(31.98mg),然后加入氯化氢的1,4-二氧六环溶液(10滴,4M),130℃下用微波辐射3.5小时。然后混合物冷却到室温,减压浓缩。粗品经反相高效液相色谱法纯化(柱为YMC Actus Triart C18,30*150mm,粒径5μm,流动相A:水(10mmol/L碳酸氢铵),流动相B:乙腈,流速:60mL/min,梯度:20%B至50%B,8min,波长:220nm,保留时间:6.83min,柱温:25℃),得标题产物(20.2mg)。To isopropanol (2 mL) was added (2-((2,5-dichloropyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethylphosphine oxide (50.00 mg) and (S)-7-(pyrrolidin-1-yl)-6,7,8,9-tetrahydro-5H-benzo[7]cycloalken-2-amine (31.98mg), then added 1 of hydrogen chloride , 4-dioxane solution (10 drops, 4M), irradiated with microwave at 130°C for 3.5 hours. The mixture was then cooled to room temperature and concentrated under reduced pressure. The crude product was purified by reverse-phase high-performance liquid chromatography (column is YMC Actus Triart C18, 30*150mm, particle diameter 5 μm, mobile phase A: water (10mmol/L ammonium bicarbonate), mobile phase B: acetonitrile, flow rate: 60mL/ min, gradient: 20%B to 50%B, 8min, wavelength: 220nm, retention time: 6.83min, column temperature: 25°C), the title product (20.2mg) was obtained.
1H NMR(400MHz,DMSO-d 6,ppm):δ11.07(s,1H),9.26(s,1H),8.52(d,J=4.6Hz,1H),8.17(s,1H),7.53(dd,J=14.0,2.0Hz,1H),7.44(q,J=3.1Hz,2H),7.26(dd,J=8.1,2.3Hz,1H),6.97(d,J=8.1Hz,1H),4.42(s,2H),3.31(s,3H),3.01–2.75(m,2H),2.55(s,5H),2.50(s,2H),1.84(s,2H),1.81(s,3H),1.77(s,3H),1.70(q,J=3.6,3.2Hz,4H),1.54(s,2H).MS(ESI+):554.2(M+H). 1 H NMR (400MHz, DMSO-d 6 , ppm): δ11.07(s, 1H), 9.26(s, 1H), 8.52(d, J=4.6Hz, 1H), 8.17(s, 1H), 7.53 (dd, J=14.0,2.0Hz,1H),7.44(q,J=3.1Hz,2H),7.26(dd,J=8.1,2.3Hz,1H),6.97(d,J=8.1Hz,1H) ,4.42(s,2H),3.31(s,3H),3.01–2.75(m,2H),2.55(s,5H),2.50(s,2H),1.84(s,2H),1.81(s,3H ),1.77(s,3H),1.70(q,J=3.6,3.2Hz,4H),1.54(s,2H).MS(ESI+):554.2(M+H).
实施例2 活性测定Example 2 Activity Determination
实施例1制备的相关化合物进行相关的酶活、细胞、体内相关的活性Related compounds prepared in Example 1 carry out related enzyme activities, cells, and related activities in vivo
活性测试中所使用的阳性药1(BGB324)具体结构如下:The specific structure of the positive drug 1 (BGB324) used in the activity test is as follows:
Figure PCTCN2022134408-appb-000017
Figure PCTCN2022134408-appb-000017
阳性药2(TP0903)具体结构如下:The specific structure of positive drug 2 (TP0903) is as follows:
Figure PCTCN2022134408-appb-000018
Figure PCTCN2022134408-appb-000018
以上化合物均从上海升泓生物科技有限公司购买。All the above compounds were purchased from Shanghai Shenghong Biotechnology Co., Ltd.
(1)AXL激酶抑制活性(1) AXL kinase inhibitory activity
1.实验流程1. Experimental process
a)AXL酶(Carna,08-107)配置及加入:用1×酶缓冲液(将200μL的Enzymatic buffer kinase 5X,10μL的500mM的MgCl 2,10μL的100mM的DTT,6.26μL的2500nM的SEB,加入773.75μL的H 2O,配置成1ml的1×酶缓冲液)将33.33ng/uL的AXL酶稀释到0.027ng/μL(1.67×,final conc.=0.016ng/μL),使用BioTek(MultiFlo FX)自动分液仪,化合物孔和阳性对照孔分别加6μL的1.67倍终浓度的酶溶液;在阴性对照孔中加6μL的1×Enzymatic buffer。 a) AXL enzyme (Carna, 08-107) configuration and addition: use 1× enzyme buffer (200 μL of Enzymatic buffer kinase 5X, 10 μL of 500 mM MgCl 2 , 10 μL of 100 mM DTT, 6.26 μL of 2500 nM SEB, Add 773.75 μL of H 2 O to make 1ml of 1×enzyme buffer) Dilute 33.33ng/uL of AXL enzyme to 0.027ng/μL (1.67×, final conc.=0.016ng/μL), use BioTek (MultiFlo FX) Automatic dispenser, add 6 μL of 1.67-fold final concentration of enzyme solution to compound wells and positive control wells; add 6 μL of 1×Enzymatic buffer to negative control wells.
b)化合物配制及加入:使用DMSO将实施例中制备的化合物及阳性药从10mM稀释到100μM,用化合物滴定仪(Tecan,D300e)进行滴定,滴定仪自动喷入每孔所需浓度,第1个浓度为1μM,1/2log梯度稀释,共8个浓度。2500rpm离心30s,室温孵育15min。b) Compound preparation and addition: use DMSO to dilute the compounds and positive drugs prepared in the examples from 10 mM to 100 μM, and titrate with a compound titrator (Tecan, D300e), and the titrator will automatically spray the required concentration into each well, the first step A concentration of 1 μM, 1/2 log gradient dilution, a total of 8 concentrations. Centrifuge at 2500rpm for 30s and incubate at room temperature for 15min.
c)ATP、底物配制及加入:ATP(Sigma,A7699)用1×酶缓冲液进行稀释,从10mM稀释到75μM(5×),终浓度为15μM;底物TK Substrate 3-biotin(Cisbio,61TK0BLC)用1×酶缓冲液,从500μM稀释到5μM(5×),终浓度为1μM,;ATP同底物等体积混合,使用BioTek自动分液仪4μL加入每孔;2500rpm离心30s,25℃反应45min。c) ATP, substrate preparation and addition: ATP (Sigma, A7699) was diluted with 1× enzyme buffer, from 10 mM to 75 μM (5×), and the final concentration was 15 μM; substrate TK Substrate 3-biotin (Cisbio, 61TK0BLC) was diluted with 1× enzyme buffer solution from 500 μM to 5 μM (5×), and the final concentration was 1 μM; ATP was mixed with the substrate in equal volume, and 4 μL was added to each well using a BioTek automatic liquid dispenser; centrifuged at 2500 rpm for 30 s, at 25 ° C React for 45 minutes.
d)检测试剂配制及加入:Streptavidin-XL665(Cisbio,610SAXLG)用HTRF KinEASE detection buffer(cisbio)从16.67μM稀释到250nM(4×),终浓度为62.5nM;TK Antibody-Cryptate(Cisbio)用HTRF KinEASE detection buffer(cisbio)从100×稀释到5×,终浓度为1×;XL665同Antibody等体积混合,使用BioTek自动分液仪10μL加入每孔,2500rpm离心30s,25℃反应1小时。反应结束后,用多功能读板仪HTRF进行检测。d) Detection reagent preparation and addition: Streptavidin-XL665 (Cisbio, 610SAXLG) was diluted from 16.67μM to 250nM (4×) with HTRF KinEASE detection buffer (cisbio), and the final concentration was 62.5nM; TK Antibody-Cryptate (Cisbio) was diluted with HTRF KinEASE detection buffer (cisbio) was diluted from 100× to 5×, and the final concentration was 1×; XL665 was mixed with Antibody in equal volume, 10 μL was added to each well using a BioTek automatic dispenser, centrifuged at 2500 rpm for 30 seconds, and reacted at 25°C for 1 hour. After the reaction, the multifunctional plate reader HTRF was used for detection.
2.数据分析2. Data analysis
使用GraphPad Prism 5软件log(inhibitor)vs.response-Variable slope拟合量效曲线,得到化合物对AXL激酶抑制的IC 50值。 Use GraphPad Prism 5 software log(inhibitor) vs.response-Variable slope to fit the dose-effect curve, and obtain the IC 50 value of the compound on AXL kinase inhibition.
抑制率计算公式如下:The formula for calculating the inhibition rate is as follows:
Figure PCTCN2022134408-appb-000019
Figure PCTCN2022134408-appb-000019
3.实验结果详见下表3. The experimental results are detailed in the table below
表12化合物AXL抑制活性IC 50数据 Table 12 Compound AXL inhibitory activity IC 50 data
Figure PCTCN2022134408-appb-000020
Figure PCTCN2022134408-appb-000020
Figure PCTCN2022134408-appb-000021
Figure PCTCN2022134408-appb-000021
(2)化合物对细胞增殖抑制检测(2) Detection of compound's inhibition of cell proliferation
1.实验流程1. Experimental process
MV-4-11(人髓性单核细胞白血病细胞株,培养基:IMDM+10%胎牛血清)购自南京科佰生物科技有限公司,置于37℃,5%CO 2的培养箱中培养。取对数生长期的细胞分别以8000个/孔、6000个/孔、5000个/孔、4000个/孔和3000个/孔的细胞密度铺在96孔板中,并同时设置空白对照组。 MV-4-11 (human myelomonocytic leukemia cell line, medium: IMDM+10% fetal bovine serum) was purchased from Nanjing Kebai Biotechnology Co., Ltd., and placed in an incubator at 37°C and 5% CO 2 nourish. Cells in the logarithmic growth phase were plated in 96-well plates at cell densities of 8000/well, 6000/well, 5000/well, 4000/well and 3000/well, and a blank control group was set at the same time.
将待测化合物以及阳性药溶解在二甲基亚砜中以制备10mM的储液,并置于-80℃冰箱中长期保存。细胞铺板24h后,用二甲基亚砜稀释10mM的化合物储液得到200倍浓度的工作液(最高浓度200或2000μM,3倍梯度,共10个浓度),每个浓度各取3μL加入到197μL的完全培养基中,稀释得到3倍浓度的工作液,然后取50μL加入到100μL的细胞培养液中(二甲基亚砜终浓度为0.5%,v/v),每个浓度设置两个复孔。加药处理72h后,每孔加入50μl的
Figure PCTCN2022134408-appb-000022
(购自Promega),按照说明书的操作流程在Envision(PerkinElmer)上测定荧光信号,使用GraphPad Prism 5软件log(inhibitor)vs.response-Variable slope拟合量效曲线,得到化合物对细胞增殖抑制的IC 50值。抑制率计算公式:
Dissolve the test compound and the positive drug in dimethyl sulfoxide to prepare a 10 mM stock solution, and store it in a -80°C refrigerator for long-term storage. 24 hours after cell plating, dilute the 10 mM compound stock solution with dimethyl sulfoxide to obtain a 200-fold working solution (maximum concentration 200 or 2000 μM, 3-fold gradient, 10 concentrations in total), and add 3 μL of each concentration to 197 μL In the complete culture medium, dilute to obtain a 3-fold working solution, then take 50 μL and add it to 100 μL cell culture medium (the final concentration of dimethyl sulfoxide is 0.5%, v/v), and set two replicates for each concentration. hole. After 72 hours of drug treatment, add 50 μl of
Figure PCTCN2022134408-appb-000022
(purchased from Promega), the fluorescent signal was measured on Envision (PerkinElmer) according to the operating procedures of the instructions, and the dose-effect curve was fitted using GraphPad Prism 5 software log (inhibitor) vs. response-Variable slope to obtain the IC of the compound on cell proliferation inhibition 50 value. Inhibition rate calculation formula:
Figure PCTCN2022134408-appb-000023
Figure PCTCN2022134408-appb-000023
其中:in:
受试物信号值:细胞+培养基+化合物组荧光信号均值;Signal value of the test substance: the mean value of the fluorescent signal of the cell + medium + compound group;
空白组信号值:培养基组(含0.5%DMSO)荧光信号均值;Signal value of the blank group: the average value of the fluorescence signal of the culture medium group (containing 0.5% DMSO);
阴性对照组信号值:细胞+培养基组(含0.5%DMSO)荧光信号均值。Signal value of negative control group: mean value of fluorescence signal of cell+medium group (containing 0.5% DMSO).
2.实验结果2. Experimental results
实施例1的化合物MV4-11细胞的抗增殖活性的IC 50(MV4-11,nM)为6.97。 The IC 50 (MV4-11, nM) of the antiproliferative activity of the compound of Example 1 on MV4-11 cells was 6.97.
(3)化合物的MV4-11体内药效(3) MV4-11 in vivo efficacy of the compound
测试化合物以及阳性药对人急性单核细胞白血病细胞MV-4-11裸鼠移植瘤模型肿瘤体内生长的抑制作用。The inhibitory effect of the test compound and the positive drug on the growth of human acute monocytic leukemia cell MV-4-11 xenografted tumor model in nude mice in vivo.
1.小鼠模型的构建1. Construction of mouse model
收取对数生长期MV-4-11细胞,细胞计数后重悬后,调整细胞浓度至7.0×10 7细胞/mL;注射到裸鼠前右侧腋窝皮下,每只动物接种200为L(14×10 6细胞/只),建立MV-4-11移植瘤模型。待瘤体积达到100~300mm 3,挑选健康状况良好、肿瘤体积相近的荷瘤鼠。 MV-4-11 cells in the logarithmic growth phase were collected, counted and resuspended, and the cell concentration was adjusted to 7.0×10 7 cells/mL; injected subcutaneously into the right axilla of nude mice, each animal was inoculated with 200 L (14 ×10 6 cells/monkey), the MV-4-11 xenograft tumor model was established. When the tumor volume reaches 100-300 mm 3 , tumor-bearing mice with good health and similar tumor volume are selected.
2.化合物的配置2. Compound Configuration
将化合物以及阳性药,用适当的溶剂涡旋振荡后超声使化合物完全溶解后缓慢加入适量体积柠檬酸缓冲液,涡旋振荡,使液体混合均匀,得到浓度为0.1、0.5、1mg mL -1的给药制剂。 Vortex the compound and the positive drug with an appropriate solvent, then ultrasonically dissolve the compound completely, then slowly add an appropriate volume of citrate buffer, vortex, and mix the liquid evenly to obtain the concentration of 0.1, 0.5, 1 mg mL -1 Dosing preparations.
溶剂对照组:PEG400&柠檬酸缓冲液(20:80,v:v)。Solvent control group: PEG400&citric acid buffer (20:80, v:v).
3.动物分组及给药3. Grouping and administration of animals
将建模的小鼠随机分组(n=6),于分组当天开始给予相关化合物和阳性药,21天后或溶剂对照组肿瘤体积达到2000mm 3结束实验(以先达到指标为准),给药体积均为10mL·kg -1。化合物以及阳性药均采取灌胃方式给予,每天给予一次。实验开始后每周测量2次瘤径和动物体重,计算肿瘤体积。 The modeled mice were randomly grouped (n=6), and related compounds and positive drugs were administered on the day of grouping, and the experiment was ended after 21 days or when the tumor volume of the solvent control group reached 2000 mm (whichever was reached first), and the administration volume Both are 10 mL·kg -1 . Both the compound and the positive drug were administered by intragastric administration, once a day. After the experiment started, the tumor diameter and animal body weight were measured twice a week, and the tumor volume was calculated.
4.数据分析4. Data Analysis
肿瘤体积(TV)计算公式为:肿瘤体积(mm 3)=l×w 2/2, The formula for calculating tumor volume (TV) is: tumor volume (mm 3 )=l×w 2 /2,
其中,l表示肿瘤长径(mm);w表示肿瘤短径(mm)。Among them, l represents the long diameter of the tumor (mm); w represents the short diameter of the tumor (mm).
相对肿瘤体积(RTV)的计算公式为:RTV=TV t/TV initial The formula for calculating the relative tumor volume (RTV) is: RTV=TV t /TV initial
其中,TV initial为分组给药时测量到的肿瘤体积;TV t为给药期间每一次测量时的肿瘤体积。 Among them, TV initial is the tumor volume measured during group administration; TV t is the tumor volume at each measurement during administration.
肿瘤生长抑制率TGI(%)的计算公式为:TGI=100%×[1-(TV t(T)-TV initial(T))/(TV t(C)-TV initial(C))] The calculation formula of tumor growth inhibition rate TGI(%) is: TGI=100%×[1-(TV t(T) -TV initial(T) )/(TV t(C) -TV initial(C) )]
其中,TV t(T)表示治疗组每次测量的肿瘤体积;TV initial(T)表示分组给药时治疗组的肿瘤体积;TV t(C)表示溶剂对照组每次测量的肿瘤体积;TV initial(C)表示分组给药时溶剂对照组的肿瘤体积。 Among them, TV t (T) represents the tumor volume measured each time in the treatment group; TV initial (T) represents the tumor volume of the treatment group when administered in groups; TV t (C) represents the tumor volume measured each time in the solvent control group; TV initial (C) represents the tumor volume of the solvent control group at the time of group administration.
相对肿瘤增殖率(%T/C)的计算公式为:%T/C=100%×(RTV T/RTV C) The formula for calculating the relative tumor proliferation rate (%T/C) is: %T/C=100%×(RTV T /RTV C )
其中,RTV T表示治疗组RTV;RTV C表示溶剂对照组RTV。 Among them, RTV T represents the RTV of the treatment group; RTV C represents the RTV of the solvent control group.
试验数据用Microsoft Office Excel 2007软件进行计算和相关统计学处理。The experimental data were calculated and related statistical processing with Microsoft Office Excel 2007 software.
5.实验结果如下详见下表:5. The experimental results are shown in the table below:
表13化合物的体内药效In vivo efficacy of the compound of table 13
Figure PCTCN2022134408-appb-000024
Figure PCTCN2022134408-appb-000024
备注:表中的实验数据为实验结束(实验结束定义为:21天后或溶剂对照组肿瘤体积达到2000mm 3结束实验(以先达到指标为准))时,获得的相关数据。 Remarks: The experimental data in the table is the relevant data obtained when the experiment ends (the end of the experiment is defined as: after 21 days or when the tumor volume of the solvent control group reaches 2000 mm 3 and the experiment ends (whichever is reached earlier)).
(4)化合物的ICR小鼠药代动力学研究(4) Pharmacokinetic study of compounds in ICR mice
1.化合物的灌胃处方配置1. Prescription configuration for gavage of compounds
将各化合物用DMSO配制成10mg/mL的储备液。Each compound was prepared as a 10 mg/mL stock solution in DMSO.
混合溶媒配制:Tween 80:PEG400:Water=1:9:90(v/v/v)Mixed solvent preparation: Tween 80:PEG400:Water=1:9:90(v/v/v)
分别准确吸取浓度为10mg/mL的化合物DMSO储备液450μl至玻璃瓶,加入适当体积的DMSO和混合溶媒,最终制剂中溶媒的比例为DMSO:混合溶媒(v/v)=10:90,涡旋(或超声),分散均匀,分别得浓度为1mg/mL的4.5mL给药试液。Accurately draw 450 μl of the compound DMSO stock solution with a concentration of 10 mg/mL into a glass bottle, add an appropriate volume of DMSO and a mixed solvent, the ratio of the solvent in the final preparation is DMSO:mixed solvent (v/v)=10:90, vortex (or ultrasound), and disperse evenly to obtain 4.5mL administration test solutions with a concentration of 1mg/mL respectively.
2.试验方案2. Test plan
取雄性6~10周龄ICR小鼠(小鼠来源:维通利华实验动物技术有限公司),每组6只,小鼠禁食过夜,给药后4小时喂食。实验当天,小鼠分别灌胃给予10mg kg- 1化合物试液。给药后小鼠在0、5min、15min、30min、1h、2h、4h、8h、24h,由眼眶采血约100μL,置于EDTA-K 2抗凝管中。将全血样品于1500~1600g离心10min,将分离得到的血浆保存于-40~-20℃冰箱中,用于生物样品分析。LC-MS/MS方法测定血药浓度。 Male ICR mice aged 6-10 weeks (source of mice: Weitong Lihua Experimental Animal Technology Co., Ltd.), 6 in each group, were fasted overnight and fed 4 hours after administration. On the day of the experiment, the mice were given 10 mg kg- 1 compound test solution by intragastric administration. At 0, 5min, 15min, 30min, 1h, 2h, 4h, 8h, and 24h after administration, about 100 μL of blood was collected from the orbit of the mice and placed in an EDTA-K 2 anticoagulant tube. Whole blood samples were centrifuged at 1500-1600 g for 10 min, and the separated plasma was stored in a -40-20°C refrigerator for biological sample analysis. LC-MS/MS method was used to determine the plasma concentration.
3.数据分析及结果3. Data analysis and results
采用Pharsight Phoenix 7.0中的非房室模型计算药代动力学参数,具体结果详见下表。The pharmacokinetic parameters were calculated using the non-compartmental model in Pharsight Phoenix 7.0, and the specific results are shown in the table below.
表14化合物的小鼠药代动力学结果The mouse pharmacokinetic result of table 14 compound
化合物compound Cmax(ng/mL)Cmax(ng/mL) Tmax(h)Tmax(h) AUC0-24(ng.h/mL)AUC0-24(ng.h/mL) T 1/2(h) T 1/2 (h)
实施例1Example 1 324324 2.172.17 13001300 1.351.35
阳性药2(TP-0903)Positive drug 2 (TP-0903) 26.826.8 0.250.25 52.252.2 1.201.20
实施例3(S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基)嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦(式I化合物)的盐以及晶型的制备Example 3 (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6,7,8,9-tetrahydro-5H-benzo[7] wheel En-2-yl)amino)pyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethylphosphine oxide (compound of formula I) salt and crystal form
分别取约50mg式I化合物,即式I化合物,和1.05当量的酸(盐酸同时设置酸与式I化合物摩尔比为2.10的情况),加入1mL溶剂并在室温下搅拌2天。所得澄清液通过5℃搅拌和缓慢挥发的方法尝试结晶,固体通过离心分离,在40℃下,鼓风干燥或减压干燥2-5小时后用于XRPD表征。Take about 50mg of the compound of formula I, that is, the compound of formula I, and 1.05 equivalents of acid (hydrochloric acid while setting the molar ratio of acid to compound of formula I to 2.10), add 1mL of solvent and stir at room temperature for 2 days. The resulting clear liquid was crystallized by stirring at 5°C and slowly volatilized, and the solid was separated by centrifugation, and then dried by air or under reduced pressure at 40°C for 2-5 hours and then used for XRPD characterization.
表15式I化合物成盐结果Table 15 formula I compound salt formation result
Figure PCTCN2022134408-appb-000025
Figure PCTCN2022134408-appb-000025
Figure PCTCN2022134408-appb-000026
Figure PCTCN2022134408-appb-000026
Figure PCTCN2022134408-appb-000027
Figure PCTCN2022134408-appb-000027
图4-13分别为式I化合物盐酸盐、二盐酸盐、磷酸盐、马尿酸盐、硫酸盐、氢溴酸盐、苯磺酸盐、草酸盐、富马酸盐、柠檬酸盐的X-射线粉末衍射(XRPD)谱图。Fig. 4-13 is respectively formula I compound hydrochloride, dihydrochloride, phosphate, hippurate, sulfate, hydrobromide, benzenesulfonate, oxalate, fumarate, citric acid X-ray powder diffraction (XRPD) pattern of the salt.
实施例4(S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基)嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦(式I化合物)甲磺酸盐的制备方法Example 4 (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6,7,8,9-tetrahydro-5H-benzo[7] wheel En-2-yl) amino) pyrimidin-4-yl) amino) -5- (methoxymethyl) phenyl) dimethyl phosphine oxide (formula I compound) methanesulfonate
向20-mL玻璃小瓶中先后添加(S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基))嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦(50mg)、甲苯(1mL)和甲磺酸(10mg),在室温下搅拌反应2小时,得到混悬液状态的(S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基))嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦甲磺酸盐晶型。Add (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6,7,8,9-tetrahydro-5H- Benzo[7]annulen-2-yl)amino))pyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethylphosphine oxide (50 mg), toluene (1 mL) and Methanesulfonic acid (10 mg), stirred and reacted at room temperature for 2 hours to obtain (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6 ,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)amino))pyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethyl Phosphine oxide mesylate crystal form.
实施例5(S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基)嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化磷(式I化合物)甲磺酸盐的制备方法Example 5 (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6,7,8,9-tetrahydro-5H-benzo[7] wheel En-2-yl) amino) pyrimidin-4-yl) amino) -5-(methoxymethyl) phenyl) dimethyl phosphorus oxide (formula I compound) methanesulfonate
向20-mL玻璃小瓶中先后添加(S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基))嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦(1g)、乙酸乙酯(20mL)和甲磺酸(172.8mg),在室温下搅拌反应10分钟后加入实施例4制备的甲磺酸盐晶型为晶种(5mg)搅拌30分钟。向玻璃小瓶中先后加入乙酸乙酯(20mL)、丙酮(10mL)和实施例4制备的甲磺酸盐晶型晶种(100.mg)搅拌析晶20分钟。抽滤,湿滤饼在真空下于40℃下干燥20小时,得到淡黄色固体粉末状态的(S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基))嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦甲磺酸盐晶型。Add (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6,7,8,9-tetrahydro-5H- Benzo[7]annulen-2-yl)amino))pyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethylphosphine oxide (1g), ethyl acetate (20mL ) and methanesulfonic acid (172.8 mg), stirred and reacted at room temperature for 10 minutes, and then added the mesylate salt crystal form prepared in Example 4 as a seed crystal (5 mg) and stirred for 30 minutes. Ethyl acetate (20 mL), acetone (10 mL) and the mesylate crystal seed crystal (100. mg) prepared in Example 4 were successively added to the glass vial and stirred for 20 minutes for crystallization. Suction filtration, the wet cake was dried under vacuum at 40°C for 20 hours to obtain (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl) )-6,7,8,9-tetrahydro-5H-benzo[7]annulen-2-yl)amino))pyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl ) Dimethylphosphine oxide mesylate crystal form.
1H NMR(400MHz,DMSO-d6):11.11(s,1H);9.46(br,1H);9.36(s,1H);8.58-8.50(m,1H);8.19(s,1H);7.60-7.43,(m,3H);7.37(dd,J=8.2,2.2Hz,1H);7.04(d,J=8.2Hz,1H);4.44(s,2H);3.53-3.46(m,3H);3.33(s,3H);3.15(s,2H);2.82-2.62(m,4H);2.32-2.29(m,5H);1.99(s,2H);1.89-1.75(m,8H);1.40(q,J=12.3Hz,2H). 1 H NMR (400MHz, DMSO-d6): 11.11(s,1H); 9.46(br,1H); 9.36(s,1H); 8.58-8.50(m,1H); 8.19(s,1H); 7.43, (m, 3H); 7.37 (dd, J = 8.2, 2.2Hz, 1H); 7.04 (d, J = 8.2Hz, 1H); 4.44 (s, 2H); 3.53-3.46 (m, 3H); 3.33(s,3H); 3.15(s,2H); 2.82-2.62(m,4H); 2.32-2.29(m,5H); 1.99(s,2H); 1.89-1.75(m,8H); 1.40( q,J=12.3Hz,2H).
其XRPD图谱参见附图1,DSC图谱参见附图2,TGA图参见图3。See accompanying drawing 1 for its XRPD pattern, refer to accompanying drawing 2 for its DSC pattern, and refer to Fig. 3 for its TGA pattern.
实施例6(S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基)嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦(式I化合物)的晶型A的制备Embodiment 6 (S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6,7,8,9-tetrahydro-5H-benzo[7] wheel Preparation of Form A of En-2-yl)amino)pyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethylphosphine oxide (compound of formula I)
于3mL玻璃瓶中先后加入200mg按照实施例1方法制备的S)-(2-((5-氯-2-((7-(吡咯烷-1-基)-6,7,8,9-四氢-5H-苯并[7]轮烯-2-基)氨基)嘧啶-4-基)氨基)-5-(甲氧基甲基)苯基)二甲基氧化膦和2mL纯化水,常温磁力搅拌6小时后,将样品离心,取湿样置于40℃减压干燥21小时后得176mg的晶型A,收率88.0%,其XRPD图谱详见图14,X射线粉末衍射图数据详见表16。Add 200 mg of S)-(2-((5-chloro-2-((7-(pyrrolidin-1-yl)-6,7,8,9- Tetrahydro-5H-benzo[7]annulen-2-yl)amino)pyrimidin-4-yl)amino)-5-(methoxymethyl)phenyl)dimethylphosphine oxide and 2 mL of purified water, After 6 hours of magnetic stirring at room temperature, the sample was centrifuged, and the wet sample was dried under reduced pressure at 40°C for 21 hours to obtain 176 mg of Form A, with a yield of 88.0%. The XRPD pattern is shown in Figure 14, X-ray powder diffraction pattern data See Table 16 for details.
表16游离碱晶型A的X射线粉末衍射图数据Table 16 X-ray powder diffraction pattern data of free base crystal form A
2θ(°)2θ(°) 高度(counts)height (counts) I/I 0(%) I/I 0 (%)
4.14.1 4545 15.415.4
5.65.6 4242 14.314.3
7.67.6 109109 36.936.9
10.210.2 107107 36.436.4
10.910.9 1313 4.44.4
12.612.6 5959 20.120.1
13.013.0 4444 14.814.8
15.215.2 1818 6.26.2
17.617.6 246246 83.483.4
19.719.7 8585 28.928.9
20.320.3 184184 62.562.5
20.920.9 294294 100.0100.0
22.222.2 6060 20.520.5
23.223.2 22twenty two 7.57.5
24.624.6 1919 6.66.6
27.027.0 4040 13.613.6
28.828.8 1414 4.64.6
37.037.0 66 2.12.1
37.737.7 1212 4.14.1

Claims (12)

  1. 一种式I化合物的药学上可接受的盐,所述盐选自有机酸盐或无机酸盐,其中所述有机酸盐选自甲磺酸盐、苯磺酸盐、草酸盐、富马酸盐、柠檬酸盐和马尿酸盐中的一种,所述无机酸盐选自盐酸盐、氢溴酸盐、硫酸盐或磷酸盐中的一种,式I化合物结构如下:A pharmaceutically acceptable salt of a compound of formula I, the salt is selected from organic acid salts or inorganic acid salts, wherein the organic acid salts are selected from methanesulfonate, benzenesulfonate, oxalate, fumarate A kind of in acid salt, citrate and hippurate, described inorganic acid salt is selected from a kind of in hydrochloride, hydrobromide, sulfate or phosphate, formula I compound structure is as follows:
    Figure PCTCN2022134408-appb-100001
    Figure PCTCN2022134408-appb-100001
  2. 根据权利要求1所述的式I化合物的药学上可接受的盐,所述有机酸盐为甲磺酸盐。According to the pharmaceutically acceptable salt of the compound of formula I according to claim 1, the organic acid salt is mesylate.
  3. 根据权利要求2所述的式I化合物的药学上可接受的盐,所述甲磺酸盐为水合物形式,进一步为二水合物形式。The pharmaceutically acceptable salt of the compound of formula I according to claim 2, the mesylate salt is in the form of hydrate, further in the form of dihydrate.
  4. 根据权利要求1-3任一项所述的式I化合物的药学上可接受的盐,所述有机酸盐中式I化合物与有机酸的摩尔比为1:1。The pharmaceutically acceptable salt of the compound of formula I according to any one of claims 1-3, wherein the molar ratio of the compound of formula I to the organic acid in the organic acid salt is 1:1.
  5. 根据权利要求1所述的式I化合物的药学上可接受的盐,所述无机酸盐中式I化合物与无机酸的摩尔比为1:1或1:2;进一步地,所述无机酸盐为盐酸盐,所述盐酸盐中式I化合物与氯化氢的摩尔比为1:1或1:2。The pharmaceutically acceptable salt of the compound of formula I according to claim 1, wherein the molar ratio of the compound of formula I to the inorganic acid in the inorganic acid salt is 1:1 or 1:2; further, the inorganic acid salt is Hydrochloride, in which the molar ratio of the compound of formula I to hydrogen chloride is 1:1 or 1:2.
  6. 一种式I化合物的药学上可接受的盐的晶型,所述盐选自有机酸盐或无机酸盐,其中所述有机酸盐选自甲磺酸盐、苯磺酸盐、草酸盐、富马酸盐、柠檬酸盐和马尿酸盐,所述无机酸盐选自盐酸盐、氢溴酸盐或磷酸盐。A crystal form of a pharmaceutically acceptable salt of a compound of formula I, the salt is selected from organic acid salts or inorganic acid salts, wherein the organic acid salts are selected from methanesulfonate, benzenesulfonate, oxalate , fumarate, citrate and hippurate, the inorganic acid salt being selected from hydrochloride, hydrobromide or phosphate.
    Figure PCTCN2022134408-appb-100002
    Figure PCTCN2022134408-appb-100002
  7. 根据权利要求6所述的式I化合物的药学上可接受的盐的晶型,所述盐为甲磺酸盐,进一步地,所述式I化合物的甲磺酸盐晶型的X射线粉末衍射图如图1所示。The crystal form of the pharmaceutically acceptable salt of the compound of formula I according to claim 6, the salt is a mesylate salt, further, the X-ray powder diffraction of the mesylate salt crystal form of the compound of formula I The picture is shown in Figure 1.
  8. 一种式I化合物的晶型,其X射线粉末衍射图在2θ为7.6°±0.2°、10.2°±0.2°、17.6°±0.2°、20.3°±0.2°和20.9°±0.2°处具有衍射峰;A crystal form of a compound of formula I, the X-ray powder diffraction pattern of which has diffraction at 2θ of 7.6°±0.2°, 10.2°±0.2°, 17.6°±0.2°, 20.3°±0.2° and 20.9°±0.2° peak;
    进一步地,X射线粉末衍射图在2θ为4.1°±0.2°、7.6°±0.2°、10.2°±0.2°、12.6°±0.2°、13.0°±0.2°、17.6°±0.2°、19.7°±0.2°、20.3°±0.2°、20.9°±0.2°和22.2°±0.2°处具有衍射峰;Further, the X-ray powder diffraction pattern is 4.1°±0.2°, 7.6°±0.2°, 10.2°±0.2°, 12.6°±0.2°, 13.0°±0.2°, 17.6°±0.2°, 19.7°±0.2° at 2θ There are diffraction peaks at 0.2°, 20.3°±0.2°, 20.9°±0.2° and 22.2°±0.2°;
    进一步地,其X射线粉末衍射图在2θ为4.1°±0.2°、5.6°±0.2°、7.6°±0.2°、10.2°±0.2°、10.9°±0.2°、12.6°±0.2°、13.0°±0.2°、15.2°±0.2°、17.6°±0.2°、19.7°±0.2°、20.3°±0.2°、20.9°±0.2°、22.2°±0.2°、23.2°±0.2°、24.6°±0.2°、27.0°±0.2°、28.8°±0.2°、37.0°±0.2°和37.7°±0.2°处具有衍射峰;Further, its X-ray powder diffraction pattern at 2θ is 4.1°±0.2°, 5.6°±0.2°, 7.6°±0.2°, 10.2°±0.2°, 10.9°±0.2°, 12.6°±0.2°, 13.0° ±0.2°, 15.2°±0.2°, 17.6°±0.2°, 19.7°±0.2°, 20.3°±0.2°, 20.9°±0.2°, 22.2°±0.2°, 23.2°±0.2°, 24.6°±0.2 °, 27.0°±0.2°, 28.8°±0.2°, 37.0°±0.2° and 37.7°±0.2° have diffraction peaks;
    进一步地,以2θ角度表示的X射线粉末衍射具有如图14所示的图谱。Further, X-ray powder diffraction represented by 2θ angle has a spectrum as shown in FIG. 14 .
  9. 权利要求1-5任一项所述的式I化合物的药学上可接受的盐或权利要求6-7任一项所述的式I化合物的药学上可接受的盐的晶型的制备方法,其包括将式I化合物与相应的酸成盐的步骤。The preparation method of the pharmaceutically acceptable salt of the compound of formula I described in any one of claims 1-5 or the crystal form of the pharmaceutically acceptable salt of the compound of formula I described in any one of claims 6-7, It comprises the step of salt-forming a compound of formula I with a corresponding acid.
  10. 根据权利要求9所述的制备方法,所述药学上可接受的盐的晶型为甲磺酸盐晶型,所述甲磺酸盐晶型通过包括以下步骤的方法制备得到:According to the preparation method according to claim 9, the crystal form of the pharmaceutically acceptable salt is a mesylate salt crystal form, and the mesylate salt crystal form is prepared by a method comprising the following steps:
    S1:将式I化合物、甲磺酸与甲苯搅拌得到式I化合物的甲磺酸盐晶型的晶种;S1: stirring the compound of formula I, methanesulfonic acid and toluene to obtain a seed crystal of the mesylate salt crystal form of the compound of formula I;
    S2:将式(I)化合物、甲磺酸与乙酸乙酯搅拌反应后加入步骤S1得到的晶型继续搅拌;S2: stirring and reacting the compound of formula (I), methanesulfonic acid and ethyl acetate, adding the crystal form obtained in step S1 and continuing to stir;
    S3:加入乙酸乙酯、丙酮和权利要求8所述的式I化合物的晶型,搅拌析晶得到所述甲磺酸盐晶型。S3: adding ethyl acetate, acetone and the crystal form of the compound of formula I according to claim 8, stirring and crystallizing to obtain the mesylate salt crystal form.
  11. 药物组合物,其包括权利要求1-5任一项所述的式I化合物的药学上可接受的盐、权利要求6-7任一项所述的式I化合物的药学上可接受的盐的晶型、权利要求8所述的式I化合物的晶型或权利要求9-10任一项所述的方法制备得到的药学上可接受的盐或药学上可接受的盐的晶型与药学上可接受的载体。A pharmaceutical composition comprising the pharmaceutically acceptable salt of the compound of formula I described in any one of claims 1-5, the pharmaceutically acceptable salt of the compound of formula I described in any one of claims 6-7 Crystal form, the crystal form of the compound of formula I described in claim 8 or the pharmaceutically acceptable salt prepared by the method described in any one of claims 9-10 or the crystal form of the pharmaceutically acceptable salt and the pharmaceutically acceptable salt acceptable carrier.
  12. 权利要求1-5任一项所述的式I化合物的药学上可接受的盐、权利要求6-7任一项所述的式I化合物的药学上可接受的盐的晶型、权利要求8所述的式I化合物的晶型或权利要求9-10任一项所述的方法制备得到的药学上可接受的盐或药学上可接受的盐的晶型在制备用于预防和/或治疗AXL激酶介导的疾病或疾病状态的药物中的用途。The pharmaceutically acceptable salt of the compound of formula I described in any one of claims 1-5, the crystal form of the pharmaceutically acceptable salt of the compound of formula I described in any one of claims 6-7, claim 8 The crystalline form of the compound of formula I or the pharmaceutically acceptable salt or the crystalline form of the pharmaceutically acceptable salt prepared by the method described in any one of claims 9-10 is used for prevention and/or treatment Use in medicine for an AXL kinase mediated disease or condition.
PCT/CN2022/134408 2021-11-26 2022-11-25 Salt of axl kinase inhibitor, preparation method therefor and use thereof WO2023093859A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280074029.7A CN118251400A (en) 2021-11-26 2022-11-25 Salts of AXL kinase inhibitors, methods of preparation and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111422552.8 2021-11-26
CN202111422552.8A CN116178433A (en) 2021-11-26 2021-11-26 Salts of AXL kinase inhibitors, methods of preparation and use thereof

Publications (1)

Publication Number Publication Date
WO2023093859A1 true WO2023093859A1 (en) 2023-06-01

Family

ID=86442748

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/134408 WO2023093859A1 (en) 2021-11-26 2022-11-25 Salt of axl kinase inhibitor, preparation method therefor and use thereof

Country Status (2)

Country Link
CN (2) CN116178433A (en)
WO (1) WO2023093859A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024199102A1 (en) * 2023-03-24 2024-10-03 南京正大天晴制药有限公司 Method for preparing axl inhibitor

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101796046A (en) * 2007-07-16 2010-08-04 阿斯利康(瑞典)有限公司 Pyrimidine derivatives 934
CN102356075A (en) * 2009-01-23 2012-02-15 里格尔药品股份有限公司 Compositions and methods for inhibition of the jak pathway
WO2015038868A1 (en) * 2013-09-13 2015-03-19 Cephalon, Inc. Fused bicyclic 2,4-diaminopyrimidine derivatives
CN106458914A (en) * 2014-03-28 2017-02-22 常州捷凯医药科技有限公司 Heterocyclic compounds as AXL inhibitors
WO2018102366A1 (en) * 2016-11-30 2018-06-07 Ariad Pharmaceuticals, Inc. Anilinopyrimidines as haematopoietic progenitor kinase 1 (hpk1) inhibitors
WO2020253860A1 (en) * 2019-06-21 2020-12-24 江苏豪森药业集团有限公司 Aryl phosphorus oxide derivative inhibitor, preparation method therefor and use thereof
WO2021125803A1 (en) * 2019-12-16 2021-06-24 한국화학연구원 Novel pyrimidin derivative and use thereof
WO2021239133A1 (en) * 2020-05-29 2021-12-02 南京正大天晴制药有限公司 Pyrimidine compound as axl inhibitor

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101796046A (en) * 2007-07-16 2010-08-04 阿斯利康(瑞典)有限公司 Pyrimidine derivatives 934
CN102356075A (en) * 2009-01-23 2012-02-15 里格尔药品股份有限公司 Compositions and methods for inhibition of the jak pathway
WO2015038868A1 (en) * 2013-09-13 2015-03-19 Cephalon, Inc. Fused bicyclic 2,4-diaminopyrimidine derivatives
CN106458914A (en) * 2014-03-28 2017-02-22 常州捷凯医药科技有限公司 Heterocyclic compounds as AXL inhibitors
WO2018102366A1 (en) * 2016-11-30 2018-06-07 Ariad Pharmaceuticals, Inc. Anilinopyrimidines as haematopoietic progenitor kinase 1 (hpk1) inhibitors
WO2020253860A1 (en) * 2019-06-21 2020-12-24 江苏豪森药业集团有限公司 Aryl phosphorus oxide derivative inhibitor, preparation method therefor and use thereof
WO2021125803A1 (en) * 2019-12-16 2021-06-24 한국화학연구원 Novel pyrimidin derivative and use thereof
WO2021239133A1 (en) * 2020-05-29 2021-12-02 南京正大天晴制药有限公司 Pyrimidine compound as axl inhibitor

Also Published As

Publication number Publication date
CN118251400A (en) 2024-06-25
CN116178433A (en) 2023-05-30

Similar Documents

Publication Publication Date Title
JP7383652B2 (en) B-RAF Kinase Maleate Salt, Crystal Form, Preparation Method, and Use thereof
TWI675839B (en) Form crystal of bisulfate of janus kinase (jak) inhibitor and preparation method thereof
TW201236684A (en) Pharmaceutically acceptable salts of (E)-N-[4-[[3-chloro-4-(2-pyridylmethoxy)phenyl]amino]-3-cyano-7-ethoxy-6-quinolyl]-3-[(2R)-1-methylpyrrolidin-2-yl]prop-2-enamide, preparation process and pharmaceutical use there of
CN109970745A (en) Substituted pyrrolo-triazine class compound and its medical composition and its use
JP6916562B2 (en) Compounds, pharmaceutically acceptable salts thereof, solvates, stereoisomers and tautomers, and drug compositions, hyperproliferative disorder therapeutic agents, hyperproliferative disorder prophylaxis agents, drugs, cancer therapeutic agents, cancer Prophylactic agents and kinase signaling regulators
WO2023093861A1 (en) Mono-p-toluenesulfonate of axl kinase inhibitor and crystal form thereof
JP2019526605A (en) Crystal form and salt form of substituted 2-H-pyrazole derivative and method for producing the same
WO2023093859A1 (en) Salt of axl kinase inhibitor, preparation method therefor and use thereof
CN112424202B (en) Crystal forms of active compounds inhibiting CDK4/6 and uses thereof
WO2023174400A1 (en) Salt of substituted amino six-membered nitric heterocyclic compound, crystal form thereof, method for preparing same, and use thereof
WO2019228330A1 (en) Substituted benzo[d]imidazole compound and pharmaceutical composition thereof
WO2023045816A1 (en) Benzocycloheptane compound as axl inhibitor
EP3750894B1 (en) Urea-substituted aromatic ring-linked dioxazoline compound, preparation method therefor, and uses thereof
JP2023543281A (en) Salts of arylaminoquinazoline-containing compounds, and their preparation and use
WO2018099451A1 (en) Crystal form of compound
CN115785107B (en) Substituted 8, 9-dihydropyrimido [5,4-b ] indolizine compound, pharmaceutical composition and application thereof
WO2024027825A1 (en) Cdk inhibitor and polymorph of phosphate thereof
WO2023109761A1 (en) Crystal of pyrazolopyrimidinone compound and salt thereof
CN108299419B (en) Novel crystal forms of novel EGFR kinase inhibitor and preparation method thereof
WO2023083356A1 (en) Nitrogen fused-heterocyclic amide compound in solid form and use thereof
CN113227073B (en) Salts of selective inhibitors of EGFR tyrosine kinase and crystalline forms thereof
CN107663207B (en) Mesylate crystal of EGFR kinase inhibitor and preparation method thereof
CN117794913A (en) Polymorphs of pyrimidine derivatives and pharmaceutically acceptable salts thereof and uses thereof
EA043251B1 (en) CRYSTAL FORM OF THE COMPOUND FOR INHIBITION OF CDK4/6 ACTIVITY AND ITS APPLICATION
KR20220088712A (en) Salts of compounds and crystalline forms 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: 22897941

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280074029.7

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE