WO2024083183A1 - Salt and crystal form of phosphonyl derivative and use thereof in medicine - Google Patents

Salt and crystal form of phosphonyl derivative and use thereof in medicine Download PDF

Info

Publication number
WO2024083183A1
WO2024083183A1 PCT/CN2023/125385 CN2023125385W WO2024083183A1 WO 2024083183 A1 WO2024083183 A1 WO 2024083183A1 CN 2023125385 W CN2023125385 W CN 2023125385W WO 2024083183 A1 WO2024083183 A1 WO 2024083183A1
Authority
WO
WIPO (PCT)
Prior art keywords
salt
formula
compound represented
compound
ray powder
Prior art date
Application number
PCT/CN2023/125385
Other languages
French (fr)
Chinese (zh)
Inventor
宫正
蒋西
蒋琦
胡健涌
范江
窦赢
Original Assignee
西藏海思科制药有限公司
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 西藏海思科制药有限公司 filed Critical 西藏海思科制药有限公司
Publication of WO2024083183A1 publication Critical patent/WO2024083183A1/en

Links

Definitions

  • the present invention relates to the field of medicine, and in particular to a salt and a crystal form of a compound represented by formula (I) and a preparation method thereof, as well as a pharmaceutical composition and application thereof in medicine.
  • Epidermal growth factor receptor is a transmembrane protein tyrosine kinase that can act as a receptor for EGF family members to trigger the EGFR signaling pathway in human epithelial cells, thereby regulating cell proliferation, invasion, metastasis, apoptosis and angiogenesis (Nat. Rev. Cancer, 2007, 7, 169-181; Expert Opin. Ther. Targets, 2012, 16, 15-31.).
  • PROTAC proteolysis targeting chimera
  • PROTAC technology By introducing ligands that can bind to different target proteins into PROTAC molecules, PROTAC technology can be used to treat various diseases. This technology has also received widespread attention in recent years (ACS Chem. Biol. 2017, 12, 892-898; Drug Discovery Today Technol. 2019, 31, 15-27.).
  • PCT/CN2022/090243 describes a compound represented by formula (I), which is a Protacs small molecule with good EGFR inhibition and degradation activity.
  • the purpose of the present invention is to provide a pharmaceutically acceptable salt of a compound represented by formula (I), a crystal form thereof, and a preparation method thereof, a pharmaceutical composition thereof, and use thereof in preparing drugs for EGFR-related diseases such as cancer diseases.
  • the advantages of the compound represented by formula (I) or the pharmaceutically acceptable salt of its stereoisomer and its solvate and the crystalline or amorphous form of the compound represented by formula (I) include but are not limited to easy processing and crystallization, convenient handling, easy purification, easy industrialization, good fluidity, easy micronization, high solubility, good pharmacokinetic properties and good stability, and are suitable for preparing pharmaceutical preparations.
  • the present invention provides a compound represented by formula (I) or a pharmaceutically acceptable salt of its stereoisomer and a solvate thereof.
  • the pharmaceutically acceptable salt is selected from maleate, 2-naphthalenesulfonate, 1,5-naphthalene disulfonate, fumarate, hydrohalide (preferably hydrobromide and hydrochloride), sulfate, phosphate, L-tartrate, citrate, L-malate, hippurate, D-glucuronate, glycolate, mucate, succinate, lactate, orotate, pamoate, glycinate, alanine, arginine, cinnamate, benzoate, benzenesulfonate, p-toluenesulfonate, acetate, propionate, valerate, triphenylacetate, L-proline, ferulate, 2-hydroxyethanesulfonate, mandelate, nitrate, methanesulfonate, malonate, gentisate, salicylate, oxalate, or glutarate;
  • hydrohalide preferably hydrobromide and hydro
  • the pharmaceutically acceptable salt is selected from benzenesulfonate, L-malate, phosphate, sulfate, p-toluenesulfonate, hydrochloride, maleate, 2-naphthalenesulfonate, hydrobromide, methanesulfonate, citrate, mandelate, lactobionate, succinate, salicylate, 1,5-naphthalenedisulfonate, fumarate, nicotinate, hippurate, and oxalate;
  • the pharmaceutically acceptable salt is selected from the group consisting of methanesulfonate, maleate, 2-naphthalenesulfonate, oxalate;
  • the pharmaceutically acceptable salt is selected from the group consisting of mesylate;
  • the molar ratio of the compound represented by formula (I): the pharmaceutically acceptable salt is 1:0.5 to 1:3.5;
  • the molar ratio of the compound represented by formula (I): the pharmaceutically acceptable salt is 1:1, 1:2, 1:3;
  • the pharmaceutically acceptable salt is selected from a methanesulfonate salt, and the molar ratio of the compound represented by formula (I): methanesulfonic acid is 1:1, 1:2, 1:3;
  • the pharmaceutically acceptable salt is selected from a methanesulfonate salt, and the molar ratio of the compound represented by formula (I): methanesulfonic acid is 1:2;
  • the pharmaceutically acceptable salt is selected from maleate, and the molar ratio of the compound represented by formula (I): maleic acid is 1:2 or 1:1;
  • the pharmaceutically acceptable salt is selected from 2-naphthalenesulfonate, and the molar ratio of the compound represented by formula (I): 2-naphthalenesulfonic acid is 1:1, 1:2;
  • the pharmaceutically acceptable salt is selected from oxalates, and the molar ratio of the compound represented by formula (I): oxalic acid is 1:1;
  • the pharmaceutically acceptable salt is selected from benzenesulfonate, and the molar ratio of the compound represented by formula (I): benzenesulfonic acid is 1:1, 1:2 or 1:3;
  • the pharmaceutically acceptable salt is selected from L-malate, and the molar ratio of the compound represented by formula (I): L-malic acid is 1:2;
  • the pharmaceutically acceptable salt is selected from phosphates, and the molar ratio of the compound represented by formula (I): phosphoric acid is 1:1, 1:2;
  • the pharmaceutically acceptable salt is selected from sulfates, and the molar ratio of the compound represented by formula (I): sulfuric acid is 1:1, 1:2 or 1:3;
  • the pharmaceutically acceptable salt is selected from p-toluenesulfonate, and the molar ratio of the compound represented by formula (I): p-toluenesulfonic acid is 1:1, 1:2;
  • the pharmaceutically acceptable salt is selected from hydrochloride, and the molar ratio of the compound represented by formula (I): hydrochloric acid is 1:1, 1:2 or 1:3;
  • the pharmaceutically acceptable salt is selected from maleate salts, and the molar ratio of the compound represented by formula (I): maleic acid is 1:1, 1:2;
  • the pharmaceutically acceptable salt is selected from hydrobromide, and the molar ratio of the compound represented by formula (I): hydrobromic acid is 1:1, 1:2 or 1:3;
  • the pharmaceutically acceptable salt is selected from a methanesulfonate salt, and the molar ratio of the compound represented by formula (I): methanesulfonic acid is 1:1, 1:2 or 1:3;
  • the pharmaceutically acceptable salt is selected from citrate, and the molar ratio of the compound represented by formula (I): citric acid is 1:1, 1:2;
  • the pharmaceutically acceptable salt is selected from mandelate, and the molar ratio of the compound represented by formula (I): mandelic acid is 1:1, 1:2;
  • the pharmaceutically acceptable salt is selected from succinate, and the molar ratio of the compound represented by formula (I): succinic acid is 1:1, 1:2 or 1:3;
  • the pharmaceutically acceptable salt is selected from fumarate, and the molar ratio of the compound represented by formula (I): fumaric acid is 1:0.5, 1:2;
  • the pharmaceutically acceptable salt is selected from hippurate, and the molar ratio of the compound represented by formula (I): hippuric acid is 1:1;
  • the pharmaceutically acceptable salt is selected from salicylates, and the molar ratio of the compound represented by formula (I): salicylic acid is 1:1;
  • the pharmaceutically acceptable salt is selected from 1,5-dinaphthylidenesulfonate, and the molar ratio of the compound represented by formula (I): 1,5-dinaphthylidenesulfonic acid is 1:1, 1:2;
  • the pharmaceutically acceptable salt is selected from nicotinate, and the molar ratio of the compound represented by formula (I): nicotinic acid is 1:3;
  • the pharmaceutically acceptable salts described above are amorphous.
  • the present invention relates to a maleate crystalline form 1 of a compound represented by formula (I); in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2 ⁇ positions: 14.83° ⁇ 0.2°, 16.53° ⁇ 0.2°, 20.34° ⁇ 0.2°, 22.87° ⁇ 0.2°, and 23.92° ⁇ 0.2°; in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2 ⁇ positions: 4.77° ⁇ 0.2°, 6.75° ⁇ 0.2°, 8.80° ⁇ 0.2°, 14.83° ⁇ 0.2°, 16.53° ⁇ 0.2°, 20.34° ⁇ 0.2°, 22.87° ⁇ 0.2°, and 23.92° ⁇ 0.2°.
  • its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ positions: 4.77° ⁇ 0.2°, 6.75° ⁇ 0.2°, 8.80° ⁇ 0.2°, 10.97° ⁇ 0.2°, 14.83° ⁇ 0.2°, 16.53° ⁇ 0.2°, 16.97° ⁇ 0.2°, 18.68° ⁇ 0.2°, 20.34° ⁇ 0.2°, 21.08° ⁇ 0.2°, 22.87° ⁇ 0.2°, 23.92° ⁇ 0.2°, 24.61° ⁇ 0.2°; in some embodiments, Cu-K ⁇ radiation is used, and its X-ray powder diffraction pattern is shown in Figure 1.
  • the present invention relates to a dimaleate crystalline form 1 of a compound represented by formula (I); in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2 ⁇ positions: 4.18° ⁇ 0.2°, 8.24° ⁇ 0.2°, 18.40° ⁇ 0.2°, 20.48° ⁇ 0.2°, 21.96° ⁇ 0.2°; in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2 ⁇ positions: 4.18° ⁇ 0.2°, 8.24° ⁇ 0.2°, 18.40° ⁇ 0.2°, 18.80° ⁇ 0.2°, 20.48° ⁇ 0.2°, 21.96° ⁇ 0.2°, 23.66° ⁇ 0.2°, 24.34° ⁇ 0.2°; In some embodiments, Cu-K ⁇ radiation is used, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ positions: 4.18° ⁇ 0.2°, 8.24° ⁇ 0.2°, 12.30
  • the present invention relates to a 2-naphthalenesulfonate salt crystal form 1 of a compound represented by formula (I); in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2 ⁇ positions: 5.82° ⁇ 0.2°, 17.52° ⁇ 0.2°, 20.13° ⁇ 0.2°, 21.16° ⁇ 0.2°, and 26.80° ⁇ 0.2°; in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2 ⁇ positions: 5.82° ⁇ 0.2°, 12.58° ⁇ 0.2°, 14.92° ⁇ 0.2°, 17.52° ⁇ 0.2°, 20.13° ⁇ 0.2°, 21.16° ⁇ 0.2°, 22.95° ⁇ 0.2°, 26.80 ° ⁇ 0.2°; in some embodiments, Cu-K ⁇ radiation is used, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ positions: 5.82° ⁇ 0.2
  • the present invention relates to an oxalate crystal form 1 of a compound represented by formula (I); in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ positions: 19.69° ⁇ 0.2°, 20.07° ⁇ 0.2°, 23.75° ⁇ 0.2°, 24.45° ⁇ 0.2°, 26.82° ⁇ 0.2°; in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ positions: 13.79° ⁇ 0.2°, 17.86° ⁇ 0.2°, 19.69° ⁇ 0.2°, 20.07° ⁇ 0.2°, 23.75° ⁇ 0.2°, 24.45° ⁇ 0.2°, 24.82° ⁇ 0.2°, 26.82° ⁇ 0.2°, 27.07° ⁇ 0.2°; in some embodiments, Cu-K ⁇ radiation is used, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ positions: 6.91° ⁇
  • the present invention relates to an amorphous dimethanesulfonate of a compound represented by formula (I), and its X-ray powder diffraction pattern is shown in FIG36 .
  • the present invention relates to a crystalline form 1 of a compound represented by formula (I); in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ positions: 5.03° ⁇ 0.2°, 15.35° ⁇ 0.2°, 19.43° ⁇ 0.2°, 19.88° ⁇ 0.2°; in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ positions: 2°, 5.03° ⁇ 0.2°, 8.03° ⁇ 0.2°, 13.25° ⁇ 0.2°, 14.57° ⁇ 0.2°, 14.88° ⁇ 0.2°, 15.35° ⁇ 0.2°, 19.43° ⁇ 0.2°, 19.88° ⁇ 0.2°, 23.83 ⁇ 0.2°, 24.71° ⁇ 0.2°, 26.44° ⁇ 0.2°, 29.93° ⁇ 0.2°; in some embodiments, using Cu-K ⁇ radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ positions:
  • the present invention relates to a method for preparing a pharmaceutically acceptable salt of a compound represented by formula (I), wherein the method comprises: a step of forming a salt using the compound represented by formula (I) and an acid; in some embodiments, the solvent used is selected from one or more of C1-6 halogenated alkane solvents, C2-6 ester solvents, C2-6 ether solvents, C1-6 alcohol solvents or water; in some embodiments, the solvent used is selected from one or more of dichloromethane, 1,2-dichloroethane, ethyl acetate, methanol, ethanol, isopropanol, propanol, ether, tetrahydrofuran and water.
  • the present invention relates to a pharmaceutical composition, wherein the pharmaceutical composition contains a therapeutically effective amount of a crystalline or amorphous pharmaceutically acceptable salt of any one of the compounds represented by formula (I) above, and a pharmaceutically acceptable carrier or excipient.
  • the present invention relates to the use of a crystalline or amorphous pharmaceutically acceptable salt of any one of the compounds represented by formula (I) or the pharmaceutical composition described above in the preparation of a drug for treating diseases (preferably cancer) related to the inhibition or degradation of EGFR.
  • the present invention relates to the use of a crystalline or amorphous pharmaceutically acceptable salt of any one of the compounds represented by formula (I) or the pharmaceutical composition described above in the preparation of a drug for treating diseases (preferably cancer) related to the inhibition or degradation of EGFR.
  • the pharmaceutical composition of the present invention may be in the form of a unit preparation (the amount of the main drug in the unit preparation is also referred to as "preparation strength").
  • Effective amount or “therapeutically effective amount” as used herein refers to administering a sufficient amount of a compound disclosed herein that will alleviate one or more symptoms of the disease or condition (e.g., cancer) being treated to some extent.
  • the result is a reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired change in a biological system.
  • an "effective amount” for therapeutic use is the amount of a compound disclosed herein required to provide a clinically significant reduction in disease symptoms.
  • therapeutically effective amounts include, but are not limited to, 1-800 mg, 1-700 mg, 1-600 mg, 2-600 mg, 3-600 mg, 4-600 mg, 5-600 mg, 6-600 mg, 10-600 mg, 20-600 mg, 25-600 mg, 30-600 mg, 40-600 mg, 50-600 mg, 60-600 mg, 70-600 mg, 75-600 mg, 80-600 mg, 90-600 mg, 100-600 mg, 200-600 mg, 1-500 mg, 2-500 mg, 3-500 mg, 4-500 mg, 5-500 mg, 6-500 mg, 10-500 mg g, 20-500mg, 25-500mg, 30-500mg, 40-500mg, 50-500mg, 60-500mg, 70-500mg, 75-500mg, 80-500mg, 90-500mg, 100-500mg, 125-500mg, 150-500mg, 200-500mg, 250-500mg, 300-500mg, 400-500mg, 5-400mg,
  • examples of therapeutically effective amounts include, but are not limited to, 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 125 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, 210 mg, 220 mg, 230 mg, 240 mg, 250 mg, 300 mg.
  • a method for treating a disease in a mammal comprising administering to a subject a therapeutically effective amount of a pharmaceutically acceptable salt or cocrystal of the compound of the present invention, preferably 1-800 mg, wherein the disease is preferably a disease related to the inhibition or degradation of EGFR (preferably cancer).
  • a method for treating a disease in a mammal comprising administering a pharmaceutically acceptable salt or co-crystal of a compound of the present invention to a subject at a daily dose of 1-1000 mg/day
  • the daily dose may be a single dose or divided doses, in some embodiments, the daily dose includes but is not limited to 10-1500 mg/day, 10-1000 mg/day, 10-800 mg/day, 25-800 mg/day, 50-800 mg/day, 100-800 mg/day, 200-800 mg/day, 25-400 mg/day, 50- 400 mg/day, 100-400 mg/day, 200-400 mg/day, in some embodiments, daily doses include but are not limited to 10 mg/day, 20 mg/day, 25 mg/day, 50 mg/day, 80 mg/day, 100 mg/day, 125 mg/day, 150 mg/day, 160 mg/day, 200 mg/day, 300 mg/day, 320 mg/day, 400 mg/day, 480 mg/day, 600 mg/day, 640 mg/day
  • the present invention relates to a kit, which may include a composition in a single-dose or multi-dose form, and the kit contains a pharmaceutically acceptable salt or co-crystal of the compound of the present invention, and the amount of the pharmaceutically acceptable salt or co-crystal of the compound of the present invention is the same as that in the above-mentioned pharmaceutical composition.
  • the crystal form of the compound shown in formula (I) of the present invention has excellent physical properties, including but not limited to solubility, dissolution rate, light resistance, low hygroscopicity, high temperature resistance, and high humidity resistance.
  • the crystal form of the present invention can significantly reduce the filtration time, shorten the production cycle, and save costs during the preparation process.
  • the crystal form of the present invention also has good light stability, thermal stability, and wet stability, which can ensure the reliability of the crystal form during storage and transportation, thereby ensuring the safety of the preparation, and the crystal form does not need to be specially packaged to prevent the influence of light, temperature, and humidity, thereby reducing costs.
  • the crystal form will not be degraded due to the influence of light, high temperature, and high humidity, thereby improving the safety of the preparation and the effectiveness after long-term storage. Patients taking the crystal form will not worry about the photosensitivity reaction of the preparation due to exposure to sunlight.
  • the crystalline form of the compound represented by formula (I) of the present invention degrades very little or less when stored or transported at ambient temperature, has good thermal stability, can be stably maintained for a long time, and is suitable for standard preparation production processes.
  • the crystal form of the compound represented by formula (I) described in the present invention is suitable and convenient for mass preparation.
  • the preparation prepared using the aforementioned crystal form can reduce irritation and improve absorption, so that the problem of metabolic rate can be solved, toxicity can be significantly reduced, safety can be improved, and the quality and efficacy of the preparation can be effectively guaranteed.
  • the crystalline structure of the present invention can be analyzed using various analytical techniques known to those skilled in the art, including but not limited to, X-ray powder diffraction (XRD), ion chromatography (IC), differential scanning calorimetry (DSC) and/or thermogravimetric analysis (TGA), also known as thermogravimetry (TG).
  • XRD X-ray powder diffraction
  • IC ion chromatography
  • DSC differential scanning calorimetry
  • TGA thermogravimetric analysis
  • TG thermogravimetric analysis
  • crystal forms of the present invention are not limited to characteristic spectra that are completely identical to the characteristic spectra described in the accompanying drawings disclosed in the present invention, such as XRD, DSC, TGA, and any crystal forms having characteristic spectra that are substantially the same or essentially the same as those described in the accompanying drawings fall within the scope of the present invention.
  • the melting peak height of a DSC curve depends on many factors related to sample preparation and instrument geometry, while the peak position is relatively insensitive to experimental details. Therefore, in some embodiments, the crystalline compound of the present invention is characterized by a DSC pattern with a characteristic peak position, having substantially the same properties as the DSC pattern provided in the accompanying drawings of the present invention, with an error tolerance of ⁇ 3 ° C.
  • the terms "about” and “approximately” used herein generally refer to the numerical value of the variable and all numerical values of the variable within experimental error (e.g., within a 95% confidence interval for the mean value) or within ⁇ 10% of the specified numerical value, or a wider range.
  • amorphous refers to any solid material that is not ordered in three dimensions.
  • amorphous solids can be characterized by known techniques, including XRPD crystal diffraction analysis, differential scanning calorimetry (DSC), solid-state nuclear magnetic resonance (ssNMR) spectroscopy, or a combination of these techniques. As described below, the XRPD pattern produced by an amorphous solid has no obvious diffraction characteristic peaks.
  • crystalline form or “crystal” refers to any solid material that exhibits a three-dimensional ordering, in contrast to an amorphous solid material, which produces a characteristic XRPD pattern with well-defined peaks.
  • seed crystals refer to the formation of crystal nuclei by adding insoluble additives in the crystallization method, which accelerates or promotes the growth of enantiomer crystals with the same crystal form or stereo configuration.
  • composition refers to a mixture of one or more compounds described herein or their physiologically/pharmaceutically acceptable salts and other components, wherein the other components include physiologically/pharmaceutically acceptable carriers and excipients.
  • carrier refers to a carrier or diluent that does not cause significant irritation to an organism and does not eliminate the biological activity and properties of the administered compound.
  • excipient refers to an inert substance added to a pharmaceutical composition to further enhance the administration of a compound.
  • the “IC 50" in the present invention refers to the half inhibitory concentration, which refers to the concentration at which half of the maximum inhibitory effect is achieved.
  • ether solvent refers to a chain compound or a cyclic compound containing an ether bond -O- and having 1 to 10 carbon atoms. Specific examples include but are not limited to: tetrahydrofuran, ethyl ether, propylene glycol methyl ether, methyl tert-butyl ether, isopropyl ether or 1,4-dioxane.
  • the "alcohol solvent” described in the present invention refers to a group derived from one or more "hydroxyl groups” replacing one or more hydrogen atoms on a "C 1-6 alkyl group", wherein the "hydroxyl group” and "C 1-6 alkyl group” are as defined above, and specific examples include but are not limited to: methanol, ethanol, isopropanol, n-propanol, isopentanol or trifluoroethanol.
  • ester solvent refers to a combination of a low-level organic acid containing 1 to 4 carbon atoms and a low-level alcohol containing 1 to 6 carbon atoms. Specific examples include but are not limited to: ethyl acetate, isopropyl acetate or butyl acetate.
  • keton solvent refers to a compound in which a carbonyl group (-C(O)-) is connected to two hydrocarbon groups.
  • ketones can be divided into aliphatic ketones, alicyclic ketones, aromatic ketones, saturated ketones and unsaturated ketones. Specific examples include but are not limited to: acetone, acetophenone, 4-methyl-2-pentanone.
  • nitrile solvent refers to a group derived from one or more "cyano” replacing one or more hydrogen atoms on a "C 1-6 alkyl".
  • the "cyano” and “C 1-6 alkyl” are as defined above, and specific examples include but are not limited to: acetonitrile or propionitrile.
  • halogenated hydrocarbon solvent refers to a group derived from one or more "halogen atoms” replacing one or more hydrogen atoms on a "C 1-6 alkyl group", wherein the "halogen atom” and "C 1-6 alkyl group” are as defined above, and specific examples include but are not limited to: dichloromethane, 1,2-dichloroethane, chloroform or carbon tetrachloride.
  • crystal of the present invention can be used interchangeably.
  • room temperature generally refers to 4-30°C, preferably refers to 20 ⁇ 5°C.
  • the drying temperature of the present invention is generally 20 to 100° C., preferably 25 to 70° C., and can be either normal pressure drying or reduced pressure drying (vacuum drying). Preferably, the drying is carried out under reduced pressure.
  • X-ray powder diffraction pattern refers to an experimentally observed diffraction pattern or a parameter, data or value derived therefrom.
  • An XRPD pattern is usually characterized by peak position (abscissa) and/or peak intensity (ordinate).
  • the "2 ⁇ or 2 ⁇ angle" described in the present invention refers to the diffraction angle
  • is the Bragg angle, which is based on the peak position expressed in degrees (°) set in the X-ray diffraction experiment, and is usually the horizontal coordinate unit in the diffraction spectrum. If the incident beam forms an angle ⁇ with a certain lattice plane and the reflection is diffracted, the experimental setting needs to record the reflected beam at an angle of 2 ⁇ .
  • substantially the same means that representative peak positions and intensity variations are taken into account. For example, one skilled in the art will appreciate that peak positions (2 ⁇ ) will show some variation, typically up to 0.1 to 0.2 degrees, and that the instrument used to measure diffraction will also cause some variation. In addition, one skilled in the art will appreciate that relative peak intensities will vary due to instrumental differences as well as degree of crystallinity, preferred orientation, prepared sample surface, and other factors known to one skilled in the art, and should be considered as only qualitative measurements.
  • the “differential scanning calorimetry or DSC” mentioned in the present invention refers to measuring the temperature difference and heat flow difference between a sample and a reference object during the process of heating or maintaining a constant temperature of the sample, so as to characterize all physical and chemical changes related to thermal effects and obtain the phase change information of the sample.
  • Hygroscopic weight gain due to moisture absorption is less than 15% but not less than 2%;
  • weight gain due to moisture absorption is less than 2% but not less than 0.2%;
  • moisture gain is less than 0.2%.
  • the crystal form disclosed in the present invention can be prepared by the following common methods for preparing crystal forms:
  • the volatilization experiment is to evaporate the clear solution of the sample at different temperatures until the solvent is dry.
  • the slurry experiment is to stir the supersaturated solution of the sample (with insoluble solids) at a certain temperature in different solvent systems.
  • the anti-solvent test is to dissolve the sample in a good solvent, add an anti-solvent, stir the precipitated solid for a short time and then filter it immediately.
  • the cooling crystallization experiment is to dissolve a certain amount of sample into the corresponding solvent at high temperature, and then stir and crystallize directly at room temperature or low temperature.
  • the polymer template experiment is to add different types of polymer materials to the sample clear solution and leave it open at room temperature to evaporate until the solvent is dry.
  • the thermal method experiment is to treat the sample under certain thermal method crystallization conditions and cool it to room temperature.
  • the water vapor diffusion experiment is to place the sample in a certain humidity environment at room temperature.
  • FIG1 is an X-ray powder diffraction pattern of maleate salt form 1 of the compound represented by formula (I).
  • FIG2 is a thermogravimetric analysis spectrum of the maleate salt form 1 of the compound represented by formula (I).
  • FIG3 is a differential scanning calorimetry curve of maleate salt form 1 of the compound represented by formula (I).
  • FIG4 is an isothermal adsorption curve of maleate salt form 1 of the compound represented by formula (I).
  • FIG5 is a DVS spectrum of the maleate salt form 1 of the compound represented by formula (I).
  • FIG6 is an X-ray powder diffraction pattern of the dimaleate crystalline form 1 of the compound represented by formula (I).
  • FIG. 7 is a thermogravimetric analysis spectrum of the dimaleate crystal form 1 of the compound represented by formula (I).
  • FIG8 is a differential scanning calorimetry curve of the dimaleate crystal form 1 of the compound represented by formula (I).
  • FIG9 is an isothermal adsorption curve of the dimaleate crystal form 1 of the compound represented by formula (I).
  • FIG10 is a DVS spectrum of the dimaleate crystal form 1 of the compound represented by formula (I).
  • FIG11 is an X-ray powder diffraction pattern of 2-naphthalenesulfonic acid form 1 of the compound represented by formula (I).
  • FIG12 is a thermogravimetric analysis spectrum of 2-naphthalenesulfonic acid form 1 of the compound represented by formula (I).
  • FIG13 is a differential scanning calorimetry curve of 2-naphthalenesulfonic acid form 1 of the compound represented by formula (I).
  • FIG14 is an isothermal adsorption curve of 2-naphthalenesulfonic acid form 1 of the compound represented by formula (I).
  • FIG15 is a DVS spectrum of 2-naphthalenesulfonic acid form 1 of the compound represented by formula (I).
  • FIG16 is an X-ray powder diffraction pattern of oxalate crystal form 1 of the compound represented by formula (I).
  • FIG. 17 is a thermogravimetric analysis spectrum of the oxalate salt form 1 of the compound represented by formula (I).
  • FIG18 is a differential scanning calorimetry curve of the oxalate crystal form 1 of the compound represented by formula (I).
  • FIG19 is an X-ray powder diffraction pattern of the amorphous benzenesulfonate salt of the compound represented by formula (I).
  • FIG. 20 is an X-ray powder diffraction pattern of the amorphous triphenylsulfonate salt of the compound represented by formula (I).
  • FIG21 is an X-ray powder diffraction pattern of the amorphous di-L-malate salt of the compound represented by formula (I).
  • FIG. 22 is an X-ray powder diffraction pattern of the amorphous phosphate of the compound represented by formula (I).
  • FIG. 23 is an X-ray powder diffraction pattern of the amorphous diphosphate of the compound represented by formula (I).
  • FIG. 24 is an X-ray powder diffraction pattern of the amorphous sulfate salt of the compound represented by formula (I).
  • FIG25 is an X-ray powder diffraction pattern of the amorphous disulfate salt of the compound represented by formula (I).
  • FIG26 is an X-ray powder diffraction pattern of the amorphous trisulfate salt of the compound represented by formula (I).
  • FIG. 27 is an X-ray powder diffraction pattern of the amorphous di-p-toluenesulfonate salt of the compound represented by formula (I).
  • FIG28 is an X-ray powder diffraction pattern of the amorphous hydrochloride salt of the compound represented by formula (I).
  • FIG29 is an X-ray powder diffraction pattern of the amorphous dihydrochloride salt of the compound represented by formula (I).
  • FIG30 is an X-ray powder diffraction pattern of the amorphous trihydrochloride salt of the compound represented by formula (I).
  • FIG31 is an X-ray powder diffraction pattern of the amorphous di-2-naphthalenesulfonate salt of the compound represented by formula (I).
  • FIG32 is an X-ray powder diffraction pattern of the amorphous hydrobromide salt of the compound represented by formula (I).
  • FIG33 is an X-ray powder diffraction pattern of the amorphous dihydrobromide salt of the compound represented by formula (I).
  • FIG34 is an X-ray powder diffraction pattern of the amorphous trihydrobromide salt of the compound represented by formula (I).
  • FIG35 is an X-ray powder diffraction pattern of the amorphous methanesulfonate salt of the compound represented by formula (I).
  • FIG36 is an X-ray powder diffraction pattern of the amorphous dimesylate salt of the compound represented by formula (I).
  • FIG37 is an X-ray powder diffraction pattern of the amorphous trimesylate salt of the compound represented by formula (I).
  • FIG38 is an X-ray powder diffraction pattern of the amorphous mandelate salt of the compound represented by formula (I).
  • FIG39 is an X-ray powder diffraction pattern of the amorphous bimandelate salt of the compound represented by formula (I).
  • FIG40 is an X-ray powder diffraction pattern of the amorphous succinate salt of the compound represented by formula (I).
  • FIG41 is an X-ray powder diffraction pattern of the amorphous salicylate of the compound represented by formula (I).
  • FIG42 is an X-ray powder diffraction pattern of amorphous 1,5-naphthalene disulfonate of the compound represented by formula (I).
  • FIG43 is an X-ray powder diffraction pattern of amorphous di-1,5-naphthalene disulfonate of the compound represented by formula (I).
  • FIG44 is an X-ray powder diffraction pattern of the amorphous hemi-fumarate salt of the compound represented by formula (I).
  • Figure 45 is an X-ray powder diffraction pattern of the amorphous difumarate salt of the compound represented by formula (I).
  • FIG46 is an X-ray powder diffraction pattern of the amorphous trinicotinate salt of the compound represented by formula (I).
  • FIG47 is an X-ray powder diffraction pattern of the amorphous hippurate salt of the compound represented by formula (I).
  • Figure 48 is an X-ray powder diffraction pattern of Form 1 of the compound represented by formula (I).
  • FIG49 is a thermogravimetric analysis spectrum of Form 1 of the compound represented by formula (I).
  • Figure 50 is a differential scanning calorimetry curve of Form 1 of the compound represented by formula (I).
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • HPLC determination was performed using an Agilent 1260DAD high pressure liquid chromatograph (Eclipse Plus C18, 150 ⁇ 4.6mm).
  • the known starting materials of the present invention can be synthesized by methods known in the art, or can be purchased from companies such as Titan Technology, Anage Chemical, Shanghai Demo, Chengdu Kelon Chemical, Shaoyuan Chemical Technology, and Bailingwei Technology.
  • 1G (130 g, 0.19 mmol) was dissolved in a mixture of THF (1.3 L) and water (400 mL), and ammonium chloride (51 g, 0.95 mmol) and zinc powder (62 g, 0.95 mmol) were added. The temperature was slowly raised to 40-60 °C for reaction for 1-2 hours. The reaction solution was cooled to room temperature and filtered. The filter cake was washed with 1 L of DCM. The organic phases were combined, and the organic phases were washed with 0.5 L of ammonia water and 0.5 L of saturated brine in sequence. After drying over anhydrous sodium sulfate, the mixture was concentrated under reduced pressure to obtain 1H as a yellow solid.
  • Test Example 1 Proliferation Inhibitory Activity of NCI-H1975 (EGFR-L858R-T790M) and A431 (EGFR-WT) Cells
  • NCI-H1975 (EGFR-L858R-T790M) and A431 (EGFR-WT) cells were purchased from ATCC, and the culture medium was RPMI1640 + 10% FBS and DMEM + 10% FBS, respectively, and cultured in a 37 ° C, 5% CO 2 incubator. On the first day, NCI-H1975 (EGFR-L858R-T790M) and A431 (EGFR-WT) cells in the exponential growth phase were collected, and live cells were counted using an automatic cell analyzer (countstar).
  • the cell suspension was adjusted with culture medium and plated on a 96-well cell culture plate, with 1000 NCI-H1975 (EGFR-L858R-T790M) cells per well and 3000 A431 cells per well.
  • the culture medium was aspirated, and 90 ⁇ L of fresh culture medium and 10 ⁇ L of different concentrations of compounds were added to each well, with a final DMSO concentration of 0.1% per well.
  • the cells were cultured in an incubator at 37°C and 5% CO 2 for 72 hours.
  • CTG solution promega, G7572
  • 50 ⁇ L of CTG solution pre-melted and equilibrated to room temperature was added to each well, mixed with a microplate shaker for 2 minutes, and placed at room temperature for 10 minutes before measuring the fluorescence signal value with a microplate reader (PHERAstar FSX).
  • V sample is the reading of the drug treatment group
  • V vehicle control is The values are the average values of the solvent control group.
  • origin9.2 software nonlinear regression model was used to draw the S-shaped dose-survival rate curve and calculate the IC 50 value.
  • the compound has good proliferation inhibitory activity against NCI-H1975 (EGFR-L858R-T790M) cells, but poor proliferation inhibitory activity against A431 (EGFR-WT) cells, and has good selectivity.
  • Test Example 2 Proliferation Inhibitory Activity on Cells NCI-H1975 EGFR-L858R-T790M-C797S
  • Cells NCI-H1975 EGFR-L858R-T790M-C797S were cultured in a 37°C, 5% CO 2 incubator in RPMI1640+10% FBS+100 ⁇ g/mL hygromycin. Cells in the exponential growth phase were collected, and the cell suspension was adjusted to an appropriate concentration with a medium without hygromycin and plated on a 96-well plate with a density of 1500 cells/well and a volume of 90 ⁇ L. 10 ⁇ L of compounds of different concentrations were added, and a solvent control group of cells plus DMSO was set up, and the concentration of DMSO was 0.1%. The cell culture plate was placed in a 37°C, 5% CO 2 incubator for 72 hours.
  • the compound has good proliferation inhibitory activity on NCI-H1975 EGFR-L858R-T790M-C797S cells.
  • mice Male SD rats, about 220 g, 6-8 weeks old, 3 rats/compound, purchased from Chengdu Dashuo Experimental Animal Co., Ltd.
  • Intragastric administration solvent 0.5% MC (MC: methylcellulose)
  • Test Example 4 Beagle dog pharmacokinetic test
  • mice Male beagle dogs, about 8-11 kg, 3 per compound, purchased from Beijing Mas Biotechnology Co., Ltd.
  • test method On the day of the test, beagle dogs were randomly divided into groups according to body weight. They were fasted but not watered for 12-14 hours one day before administration and were fed 4 hours after administration.

Abstract

Provided in the present invention are a pharmaceutically acceptable salt of a compound as represented by formula (I) or a stereoisomer thereof, a solvate and/or a crystal form thereof, a preparation method therefor, a pharmaceutical composition thereof, and the use thereof in medicine.

Description

一种膦酰衍生物的盐及晶型和在医药上的用途A salt and crystal form of a phosphono derivative and its use in medicine 技术领域Technical Field
本发明涉及医药领域,具体的说,是涉及一种式(I)所示的化合物的盐和晶型及其制备方法,以及其用于药物组合物和在医药上的应用。The present invention relates to the field of medicine, and in particular to a salt and a crystal form of a compound represented by formula (I) and a preparation method thereof, as well as a pharmaceutical composition and application thereof in medicine.
背景技术Background technique
表皮生长因子受体(EGFR)是一种跨膜蛋白酪氨酸激酶,可作为EGF家族成员触发人类上皮细胞中EGFR信号通路的受体,从而调节细胞增殖,侵袭,转移,凋亡和血管生成(Nat.Rev.Cancer,2007,7,169-181;Expert Opin.Ther.Targets,2012,16,15-31.)。人体内EGFR基因的过度表达、突变或扩增致使EGFR活性异常增加,会导致许多恶性肿瘤如食道癌、胶质母细胞瘤、肛门癌、头颈部上皮癌、乳腺癌、肺癌、特别是非小细胞肺癌(NSCLC)的产生(Cells,2019,8,350-361.)。PROTAC(proteolysis targeting chimera)分子是一类能够同时结合靶向蛋白和E3泛素连接酶的双功能化合物,此类化合物能够被细胞的蛋白酶体识别,引起靶向蛋白的降解,能够有效地降低靶向蛋白在细胞中的含量。通过在PROTAC分子引入能结合不同靶向蛋白的配体,使PROTAC技术应用于各种疾病的治疗成为可能,该技术近年来同时得到了广泛的关注(ACS Chem.Biol.2017,12,892-898;Drug Discovery Today Technol.2019,31,15-27.)。Epidermal growth factor receptor (EGFR) is a transmembrane protein tyrosine kinase that can act as a receptor for EGF family members to trigger the EGFR signaling pathway in human epithelial cells, thereby regulating cell proliferation, invasion, metastasis, apoptosis and angiogenesis (Nat. Rev. Cancer, 2007, 7, 169-181; Expert Opin. Ther. Targets, 2012, 16, 15-31.). Overexpression, mutation or amplification of the EGFR gene in the human body leads to abnormal increase in EGFR activity, which can lead to the occurrence of many malignant tumors such as esophageal cancer, glioblastoma, anal cancer, head and neck epithelial cancer, breast cancer, lung cancer, especially non-small cell lung cancer (NSCLC) (Cells, 2019, 8, 350-361.). PROTAC (proteolysis targeting chimera) molecules are a class of bifunctional compounds that can simultaneously bind to target proteins and E3 ubiquitin ligases. Such compounds can be recognized by the proteasome of the cell, causing the degradation of the target protein, and can effectively reduce the content of the target protein in the cell. By introducing ligands that can bind to different target proteins into PROTAC molecules, PROTAC technology can be used to treat various diseases. This technology has also received widespread attention in recent years (ACS Chem. Biol. 2017, 12, 892-898; Drug Discovery Today Technol. 2019, 31, 15-27.).
PCT/CN2022/090243记载了式(I)所示的化合物,该化合物是一种具有良好的EGFR抑制和降解活性的Protacs小分子。
PCT/CN2022/090243 describes a compound represented by formula (I), which is a Protacs small molecule with good EGFR inhibition and degradation activity.
发明内容Summary of the invention
本发明的目的是提供一种式(I)所示化合物的可药用盐及其晶型和制备方法,其药物组合物以及其在制备EGFR相关疾病如癌症疾病的药物中的用途。The purpose of the present invention is to provide a pharmaceutically acceptable salt of a compound represented by formula (I), a crystal form thereof, and a preparation method thereof, a pharmaceutical composition thereof, and use thereof in preparing drugs for EGFR-related diseases such as cancer diseases.
式(I)所示化合物或其立体异构体的可药用盐及其溶剂化物与式(I)所示化合物的晶型或无定型其优势包括但不限于易于加工和结晶、方便处理、易于纯化,易于工业化,流动性好、易于微粉化、较高的溶解度、较好的药代动力学特性和良好的稳定性,适合制备药物制剂。The advantages of the compound represented by formula (I) or the pharmaceutically acceptable salt of its stereoisomer and its solvate and the crystalline or amorphous form of the compound represented by formula (I) include but are not limited to easy processing and crystallization, convenient handling, easy purification, easy industrialization, good fluidity, easy micronization, high solubility, good pharmacokinetic properties and good stability, and are suitable for preparing pharmaceutical preparations.
本发明提供一种式(I)所示化合物或其立体异构体的可药用盐及其溶剂化物,
The present invention provides a compound represented by formula (I) or a pharmaceutically acceptable salt of its stereoisomer and a solvate thereof.
在一些实施方案中,可药用盐选自马来酸盐、2-萘磺酸盐、1,5-萘二磺酸盐、富马酸盐、氢卤酸盐(优选为氢溴酸盐和盐酸盐)、硫酸盐、磷酸盐、L-酒石酸盐、柠檬酸盐、L-苹果酸盐、马尿酸盐、 D-葡萄糖醛酸盐、乙醇酸盐、粘酸盐、琥珀酸盐、乳酸盐、乳清酸盐、帕莫酸盐、甘氨酸盐、丙氨酸盐、精氨酸盐、肉桂酸盐、苯甲酸盐、苯磺酸盐、对甲苯磺酸盐、乙酸盐、丙酸盐、戊酸盐、三苯基乙酸盐、L-脯氨酸盐、阿魏酸盐、2-羟基乙磺酸盐、扁桃酸盐、硝酸盐、甲磺酸盐、丙二酸盐、龙胆酸盐、水杨酸盐、草酸盐或戊二酸盐;In some embodiments, the pharmaceutically acceptable salt is selected from maleate, 2-naphthalenesulfonate, 1,5-naphthalene disulfonate, fumarate, hydrohalide (preferably hydrobromide and hydrochloride), sulfate, phosphate, L-tartrate, citrate, L-malate, hippurate, D-glucuronate, glycolate, mucate, succinate, lactate, orotate, pamoate, glycinate, alanine, arginine, cinnamate, benzoate, benzenesulfonate, p-toluenesulfonate, acetate, propionate, valerate, triphenylacetate, L-proline, ferulate, 2-hydroxyethanesulfonate, mandelate, nitrate, methanesulfonate, malonate, gentisate, salicylate, oxalate, or glutarate;
在一些实施方案中,可药用盐选自苯磺酸盐、L-苹果酸盐、磷酸盐、硫酸盐、对甲苯磺酸盐、盐酸盐、马来酸盐、2-萘磺酸盐、氢溴酸盐、甲磺酸盐、柠檬酸盐、扁桃酸盐、乳糖酸盐、琥珀酸盐、水杨酸盐、1,5-萘二磺酸盐、富马酸盐、烟酸盐、马尿酸盐和草酸盐;In some embodiments, the pharmaceutically acceptable salt is selected from benzenesulfonate, L-malate, phosphate, sulfate, p-toluenesulfonate, hydrochloride, maleate, 2-naphthalenesulfonate, hydrobromide, methanesulfonate, citrate, mandelate, lactobionate, succinate, salicylate, 1,5-naphthalenedisulfonate, fumarate, nicotinate, hippurate, and oxalate;
在一些实施方案中,可药用盐选自甲磺酸盐、马来酸盐、2-萘磺酸盐、草酸盐;In some embodiments, the pharmaceutically acceptable salt is selected from the group consisting of methanesulfonate, maleate, 2-naphthalenesulfonate, oxalate;
在一些实施方案中,可药用盐选自甲磺酸盐;In some embodiments, the pharmaceutically acceptable salt is selected from the group consisting of mesylate;
在一些实施方案中,式(I)所示化合物:可药用盐的摩尔比为1:0.5~1:3.5;In some embodiments, the molar ratio of the compound represented by formula (I): the pharmaceutically acceptable salt is 1:0.5 to 1:3.5;
在一些实施方案中,式(I)所示化合物:可药用盐的摩尔比为1:1、1:2、1:3;In some embodiments, the molar ratio of the compound represented by formula (I): the pharmaceutically acceptable salt is 1:1, 1:2, 1:3;
在一些实施方案中,可药用盐选自甲磺酸盐,式(I)所示化合物:甲磺酸的摩尔比为1:1、1:2、1:3;In some embodiments, the pharmaceutically acceptable salt is selected from a methanesulfonate salt, and the molar ratio of the compound represented by formula (I): methanesulfonic acid is 1:1, 1:2, 1:3;
在一些实施方案中,可药用盐选自甲磺酸盐,式(I)所示化合物:甲磺酸的摩尔比为1:2;In some embodiments, the pharmaceutically acceptable salt is selected from a methanesulfonate salt, and the molar ratio of the compound represented by formula (I): methanesulfonic acid is 1:2;
在一些实施方案中,可药用盐选自马来酸盐,式(I)所示化合物:马来酸的摩尔比为1:2或1:1;In some embodiments, the pharmaceutically acceptable salt is selected from maleate, and the molar ratio of the compound represented by formula (I): maleic acid is 1:2 or 1:1;
在一些实施方案中,可药用盐选自2-萘磺酸盐,式(I)所示化合物:2-萘磺酸的摩尔比为1:1、1:2;In some embodiments, the pharmaceutically acceptable salt is selected from 2-naphthalenesulfonate, and the molar ratio of the compound represented by formula (I): 2-naphthalenesulfonic acid is 1:1, 1:2;
在一些实施方案中,可药用盐选自草酸盐,式(I)所示化合物:草酸的摩尔比为1:1;In some embodiments, the pharmaceutically acceptable salt is selected from oxalates, and the molar ratio of the compound represented by formula (I): oxalic acid is 1:1;
在一些实施方案中,可药用盐选自苯磺酸盐,式(I)所示化合物:苯磺酸的摩尔比为1:1、1:2或1:3;In some embodiments, the pharmaceutically acceptable salt is selected from benzenesulfonate, and the molar ratio of the compound represented by formula (I): benzenesulfonic acid is 1:1, 1:2 or 1:3;
在一些实施方案中,可药用盐选L-苹果酸盐,式(I)所示化合物:L-苹果酸的摩尔比为1:2;In some embodiments, the pharmaceutically acceptable salt is selected from L-malate, and the molar ratio of the compound represented by formula (I): L-malic acid is 1:2;
在一些实施方案中,可药用盐选自磷酸盐,式(I)所示化合物:磷酸的摩尔比为1:1、1:2;In some embodiments, the pharmaceutically acceptable salt is selected from phosphates, and the molar ratio of the compound represented by formula (I): phosphoric acid is 1:1, 1:2;
在一些实施方案中,可药用盐选自硫酸盐,式(I)所示化合物:硫酸的摩尔比为1:1、1:2或1:3;In some embodiments, the pharmaceutically acceptable salt is selected from sulfates, and the molar ratio of the compound represented by formula (I): sulfuric acid is 1:1, 1:2 or 1:3;
在一些实施方案中,可药用盐选自对甲苯磺酸盐,式(I)所示化合物:对甲苯磺酸的摩尔比为1:1、1:2;In some embodiments, the pharmaceutically acceptable salt is selected from p-toluenesulfonate, and the molar ratio of the compound represented by formula (I): p-toluenesulfonic acid is 1:1, 1:2;
在一些实施方案中,可药用盐选自盐酸盐,式(I)所示化合物:盐酸的摩尔比为1:1、1:2或1:3;In some embodiments, the pharmaceutically acceptable salt is selected from hydrochloride, and the molar ratio of the compound represented by formula (I): hydrochloric acid is 1:1, 1:2 or 1:3;
在一些实施方案中,可药用盐选自马来酸盐,式(I)所示化合物:马来酸的摩尔比为1:1、1:2;In some embodiments, the pharmaceutically acceptable salt is selected from maleate salts, and the molar ratio of the compound represented by formula (I): maleic acid is 1:1, 1:2;
在一些实施方案中,可药用盐选自氢溴酸盐,式(I)所示化合物:氢溴酸的摩尔比为1:1、1:2或1:3;In some embodiments, the pharmaceutically acceptable salt is selected from hydrobromide, and the molar ratio of the compound represented by formula (I): hydrobromic acid is 1:1, 1:2 or 1:3;
在一些实施方案中,可药用盐选自甲磺酸盐,式(I)所示化合物:甲磺酸的摩尔比为1:1、1:2或1:3;In some embodiments, the pharmaceutically acceptable salt is selected from a methanesulfonate salt, and the molar ratio of the compound represented by formula (I): methanesulfonic acid is 1:1, 1:2 or 1:3;
在一些实施方案中,可药用盐选自柠檬酸盐,式(I)所示化合物:柠檬酸的摩尔比为1:1、1:2;In some embodiments, the pharmaceutically acceptable salt is selected from citrate, and the molar ratio of the compound represented by formula (I): citric acid is 1:1, 1:2;
在一些实施方案中,可药用盐选自扁桃酸盐,式(I)所示化合物:扁桃酸的摩尔比为1:1、1:2;In some embodiments, the pharmaceutically acceptable salt is selected from mandelate, and the molar ratio of the compound represented by formula (I): mandelic acid is 1:1, 1:2;
在一些实施方案中,可药用盐选自琥珀酸盐,式(I)所示化合物:琥珀酸的摩尔比为1:1、1:2或1:3;In some embodiments, the pharmaceutically acceptable salt is selected from succinate, and the molar ratio of the compound represented by formula (I): succinic acid is 1:1, 1:2 or 1:3;
在一些实施方案中,可药用盐选自富马酸盐,式(I)所示化合物:富马酸的摩尔比为1:0.5、1:2;In some embodiments, the pharmaceutically acceptable salt is selected from fumarate, and the molar ratio of the compound represented by formula (I): fumaric acid is 1:0.5, 1:2;
在一些实施方案中,可药用盐选自马尿酸盐,式(I)所示化合物:马尿酸的摩尔比为1:1;In some embodiments, the pharmaceutically acceptable salt is selected from hippurate, and the molar ratio of the compound represented by formula (I): hippuric acid is 1:1;
在一些实施方案中,可药用盐选自水杨酸盐,式(I)所示化合物:水杨酸的摩尔比为1:1;In some embodiments, the pharmaceutically acceptable salt is selected from salicylates, and the molar ratio of the compound represented by formula (I): salicylic acid is 1:1;
在一些实施方案中,可药用盐选自1,5-二萘磺酸盐,式(I)所示化合物:1,5-二萘磺酸的摩尔比为1:1、1:2; In some embodiments, the pharmaceutically acceptable salt is selected from 1,5-dinaphthylidenesulfonate, and the molar ratio of the compound represented by formula (I): 1,5-dinaphthylidenesulfonic acid is 1:1, 1:2;
在一些实施方案中,可药用盐选自烟酸盐,式(I)所示化合物:烟酸的摩尔比为1:3;In some embodiments, the pharmaceutically acceptable salt is selected from nicotinate, and the molar ratio of the compound represented by formula (I): nicotinic acid is 1:3;
在一些实施方案中,上述可药用盐为无定型。In some embodiments, the pharmaceutically acceptable salts described above are amorphous.
本发明涉及一种式(I)所示化合物的马来酸盐晶型1;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:14.83°±0.2°、16.53°±0.2°、20.34°±0.2°、22.87°±0.2°、23.92°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:4.77°±0.2°、6.75°±0.2°、8.80°±0.2°、14.83°±0.2°、16.53°±0.2°、20.34°±0.2°、22.87°±0.2°、23.92°±0.2°;在一些实施方案中,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:4.77°±0.2°、6.75°±0.2°、8.80°±0.2°、10.97°±0.2°、14.83°±0.2°、16.53°±0.2°、16.97°±0.2°、18.68°±0.2°、20.34°±0.2°、21.08°±0.2°、22.87°±0.2°、23.92°±0.2°、24.61°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图如图1所示。The present invention relates to a maleate crystalline form 1 of a compound represented by formula (I); in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2θ positions: 14.83°±0.2°, 16.53°±0.2°, 20.34°±0.2°, 22.87°±0.2°, and 23.92°±0.2°; in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2θ positions: 4.77°±0.2°, 6.75°±0.2°, 8.80°±0.2°, 14.83°±0.2°, 16.53°±0.2°, 20.34°±0.2°, 22.87°±0.2°, and 23.92°±0.2°. 2.87°±0.2°, 23.92°±0.2°; in some embodiments, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ positions: 4.77°±0.2°, 6.75°±0.2°, 8.80°±0.2°, 10.97°±0.2°, 14.83°±0.2°, 16.53°±0.2°, 16.97°±0.2°, 18.68°±0.2°, 20.34°±0.2°, 21.08°±0.2°, 22.87°±0.2°, 23.92°±0.2°, 24.61°±0.2°; in some embodiments, Cu-Kα radiation is used, and its X-ray powder diffraction pattern is shown in Figure 1.
本发明涉及一种式(I)所示化合物的二马来酸盐晶型1;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:4.18°±0.2°、8.24°±0.2°、18.40°±0.2°、20.48°±0.2°、21.96°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:4.18°±0.2°、8.24°±0.2°、18.40°±0.2°、18.80°±0.2°、20.48°±0.2°、21.96°±0.2°、23.66°±0.2°、24.34°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:4.18°±0.2°、8.24°±0.2°、12.30±0.2°、13.21°±0.2°、16.34°±0.2°、16.57°±0.2°、18.40°±0.2°、18.80°±0.2°、20.00°±0.2°、20.48°±0.2°、20.81°±0.2°、21.96°±0.2°、23.66°±0.2°、24.34°±0.2°、25.73°±0.2°、27.98°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图如图6所示。The present invention relates to a dimaleate crystalline form 1 of a compound represented by formula (I); in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2θ positions: 4.18°±0.2°, 8.24°±0.2°, 18.40°±0.2°, 20.48°±0.2°, 21.96°±0.2°; in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2θ positions: 4.18°±0.2°, 8.24°±0.2°, 18.40°±0.2°, 18.80°±0.2°, 20.48°±0.2°, 21.96°±0.2°, 23.66°±0.2°, 24.34°±0.2°; In some embodiments, Cu-Kα radiation is used, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ positions: 4.18°±0.2°, 8.24°±0.2°, 12.30±0.2°, 13.21°±0.2°, 16.34°±0.2°, 16.57°±0.2°, 18.40°±0.2°, 18.80°±0.2°, 20.00°±0.2°, 20.48°±0.2°, 20.81°±0.2°, 21.96°±0.2°, 23.66°±0.2°, 24.34°±0.2°, 25.73°±0.2°, and 27.98°±0.2°; in some embodiments, Cu-Kα radiation is used, and its X-ray powder diffraction pattern is shown in Figure 6.
本发明涉及一种式(I)所示化合物的2-萘磺酸盐晶型1;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:5.82°±0.2°、17.52°±0.2°、20.13°±0.2°、21.16°±0.2°、26.80°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:5.82°±0.2°、12.58°±0.2°、14.92°±0.2°、17.52°±0.2°、20.13°±0.2°、21.16°±0.2°、22.95°±0.2°、26.80°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:5.82°±0.2°、7.10°±0.2°、8.78°±0.2°、11.59°±0.2°、12.58°±0.2°、14.92°±0.2°、17.03°±0.2°、17.52°±0.2°、18.80°±0.2°、20.13°±0.2°、21.16°±0.2°、21.72°±0.2°、22.22°±0.2°、22.95°±0.2°、26.80°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图如图11所示。The present invention relates to a 2-naphthalenesulfonate salt crystal form 1 of a compound represented by formula (I); in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2θ positions: 5.82°±0.2°, 17.52°±0.2°, 20.13°±0.2°, 21.16°±0.2°, and 26.80°±0.2°; in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction spectrum has characteristic diffraction peaks at the following 2θ positions: 5.82°±0.2°, 12.58°±0.2°, 14.92°±0.2°, 17.52°±0.2°, 20.13°±0.2°, 21.16°±0.2°, 22.95°±0.2°, 26.80 °±0.2°; in some embodiments, Cu-Kα radiation is used, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ positions: 5.82°±0.2°, 7.10°±0.2°, 8.78°±0.2°, 11.59°±0.2°, 12.58°±0.2°, 14.92°±0.2°, 17.03°±0.2°, 17.52°±0.2°, 18.80°±0.2°, 20.13°±0.2°, 21.16°±0.2°, 21.72°±0.2°, 22.22°±0.2°, 22.95°±0.2°, 26.80°±0.2°; in some embodiments, Cu-Kα radiation is used, and its X-ray powder diffraction pattern is shown in Figure 11.
本发明涉及一种式(I)所示化合物的草酸盐晶型1;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:19.69°±0.2°、20.07°±0.2°、23.75°±0.2°、24.45°±0.2°、26.82°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:13.79°±0.2°、17.86°±0.2°、19.69°±0.2°、20.07°±0.2°、23.75°±0.2°、24.45°±0.2°、24.82°±0.2°、26.82°±0.2°、27.07°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:6.91°±0.2°、7.63°±0.2°、13.52°±0.2°、13.79°±0.2°、17.86°±0.2°、18.87°±0.2°、19.69°±0.2°、20.07°±0.2°、20.71°±0.2°、23.75°±0.2°、24.45°±0.2°、24.82°±0.2°、26.82°±0.2°、27.07°±0.2°、29.44°±0.2°、31.28°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图如图16所示。The present invention relates to an oxalate crystal form 1 of a compound represented by formula (I); in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ positions: 19.69°±0.2°, 20.07°±0.2°, 23.75°±0.2°, 24.45°±0.2°, 26.82°±0.2°; in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ positions: 13.79°±0.2°, 17.86°±0.2°, 19.69°±0.2°, 20.07°±0.2°, 23.75°±0.2°, 24.45°±0.2°, 24.82°±0.2°, 26.82°±0.2°, 27.07° ±0.2°; in some embodiments, Cu-Kα radiation is used, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ positions: 6.91°±0.2°, 7.63°±0.2°, 13.52°±0.2°, 13.79°±0.2°, 17.86°±0.2°, 18.87°±0.2°, 19.69°±0.2°, 20.07°±0.2°, 20.71°±0.2°, 23.75°±0.2°, 24.45°±0.2°, 24.82°±0.2°, 26.82°±0.2°, 27.07°±0.2°, 29.44°±0.2°, 31.28°±0.2°; in some embodiments, Cu-Kα radiation is used, and its X-ray powder diffraction pattern is shown in Figure 16.
本发明涉及一种无定型的式(I)所示的化合物的二甲磺酸盐,其X-射线粉末衍射图如图36所示。The present invention relates to an amorphous dimethanesulfonate of a compound represented by formula (I), and its X-ray powder diffraction pattern is shown in FIG36 .
本发明涉及一种式(I)所示的化合物的晶型1;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:5.03°±0.2°、15.35°±0.2°、19.43°±0.2°、19.88°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰: 5.03°±0.2°、8.03°±0.2°、13.25°±0.2°、14.57°±0.2°、14.88°±0.2°、15.35°±0.2°、19.43°±0.2°、19.88°±0.2°、23.83±0.2°、24.71°±0.2°、26.44°±0.2°、29.93°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:5.03°±0.2°、8.03°±0.2°、8.99°±0.2°、13.25°±0.2°、14.57°±0.2°、14.88°±0.2°、15.35°±0.2°、19.43°±0.2°、19.88°±0.2°、20.17±0.2°、22.32±0.2°、22.55±0.2°、23.83±0.2°、24.71°±0.2°、26.17±0.2°、26.44°±0.2°、29.49±0.2°、29.93°±0.2°;在一些实施方案中,使用Cu-Kα辐射,其X-射线粉末衍射图如图48所示。The present invention relates to a crystalline form 1 of a compound represented by formula (I); in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ positions: 5.03°±0.2°, 15.35°±0.2°, 19.43°±0.2°, 19.88°±0.2°; in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ positions: 2°, 5.03°±0.2°, 8.03°±0.2°, 13.25°±0.2°, 14.57°±0.2°, 14.88°±0.2°, 15.35°±0.2°, 19.43°±0.2°, 19.88°±0.2°, 23.83±0.2°, 24.71°±0.2°, 26.44°±0.2°, 29.93°±0.2°; in some embodiments, using Cu-Kα radiation, its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ positions: 5.03°±0.2°, 8.03°±0.2°, 8.99°±0.2°, 48°±0.2°; in some embodiments, Cu-Kα radiation is used, and the X-ray powder diffraction pattern thereof is shown in FIG48.
本发明涉及一种式(I)所示化合物的可药用盐的制备方法,其中,所述方法包括:以式(I)所示化合物和酸成盐的步骤;在一些实施方案中,所用溶剂选自C1-6卤代烷烃类溶剂、C2-6酯类溶剂、C2-6醚类溶剂、C1-6醇类溶剂或水中的一种或多种;在一些实施方案中,所用溶剂选自二氯甲烷、1,2-二氯乙烷、乙酸乙酯、甲醇、乙醇、异丙醇、丙醇、乙醚、四氢呋喃和水中的一种或多种。The present invention relates to a method for preparing a pharmaceutically acceptable salt of a compound represented by formula (I), wherein the method comprises: a step of forming a salt using the compound represented by formula (I) and an acid; in some embodiments, the solvent used is selected from one or more of C1-6 halogenated alkane solvents, C2-6 ester solvents, C2-6 ether solvents, C1-6 alcohol solvents or water; in some embodiments, the solvent used is selected from one or more of dichloromethane, 1,2-dichloroethane, ethyl acetate, methanol, ethanol, isopropanol, propanol, ether, tetrahydrofuran and water.
本发明涉及一种药物组合物,其中,所述药物组合物含有治疗有效量的前述任意一种式(I)所示化合物的可药用盐的晶型或无定型、及药学上可接受的载体或赋形剂。The present invention relates to a pharmaceutical composition, wherein the pharmaceutical composition contains a therapeutically effective amount of a crystalline or amorphous pharmaceutically acceptable salt of any one of the compounds represented by formula (I) above, and a pharmaceutically acceptable carrier or excipient.
本发明涉及前述任意一种式(I)所示化合物的可药用盐的晶型或无定型或者上述的药物组合物在制备用于治疗与抑制或降解EGFR相关疾病(优选癌症)的药物中的应用。The present invention relates to the use of a crystalline or amorphous pharmaceutically acceptable salt of any one of the compounds represented by formula (I) or the pharmaceutical composition described above in the preparation of a drug for treating diseases (preferably cancer) related to the inhibition or degradation of EGFR.
本发明涉及前述任意一种式(I)所示化合物的可药用盐的晶型或无定型或者上述的药物组合物在制备用于治疗与抑制或降解EGFR相关疾病(优选癌症)的药物中的应用。The present invention relates to the use of a crystalline or amorphous pharmaceutically acceptable salt of any one of the compounds represented by formula (I) or the pharmaceutical composition described above in the preparation of a drug for treating diseases (preferably cancer) related to the inhibition or degradation of EGFR.
在一些实施方案中,本发明的药物组合物可以为单位制剂形式(单位制剂中主药的量也被称为“制剂规格”)。In some embodiments, the pharmaceutical composition of the present invention may be in the form of a unit preparation (the amount of the main drug in the unit preparation is also referred to as "preparation strength").
本申请中所述“有效量”或“治疗有效量”是指给予足够量的本申请公开的化合物,其将在某种程度上缓解所治疗的疾病或病症(例如癌症)的一种或多种症状。在一些实施方案中,结果是减少和/或缓和疾病的体征、症状或原因,或生物系统的任何其它希望改变。例如,针对治疗用途的“有效量”是提供临床上显著的疾病症状降低所需的包含本申请公开的化合物的量。治疗有效量的实例(以游离碱形式计算)包括但不限于1-800mg、1-700mg、1-600mg、2-600mg、3-600mg、4-600mg、5-600mg、6-600mg、10-600mg、20-600mg、25-600mg、30-600mg、40-600mg、50-600mg、60-600mg、70-600mg、75-600mg、80-600mg、90-600mg、100-600mg、200-600mg、1-500mg、2-500mg、3-500mg、4-500mg、5-500mg、6-500mg、10-500mg、20-500mg、25-500mg、30-500mg、40-500mg、50-500mg、60-500mg、70-500mg、75-500mg、80-500mg、90-500mg、100-500mg、125-500mg、150-500mg、200-500mg、250-500mg、300-500mg、400-500mg、5-400mg、10-400mg、20-400mg、25-400mg、30-400mg、40-400mg、50-400mg、60-400mg、70-400mg、75-400mg、80-400mg、90-400mg、100-400mg、125-400mg、150-400mg、200-400mg、250-400mg、300-400mg、1-300mg、2-300mg、5-300mg、10-300mg、20-300mg、25-300mg、30-300mg、40-300mg、50-300mg、60-300mg、70-300mg、75-300mg、80-300mg、90-300mg、100-300mg、125-300mg、150-300mg、200-300mg、250-300mg、1-200mg、2-200mg、5-200mg、10-200mg、20-200mg、25-200mg、30-200mg、40-200mg、50-200mg、60-200mg、70-200mg、75-200mg、80-200mg、90-200mg、100-200mg、125-200mg、150-200mg、1-100mg、2-100mg、5-100mg、10-100mg、15-100mg、20-100mg、25-100mg、30-100mg、40-100mg、50-100mg、60-100mg、70-100mg、75-100mg、80-100mg、90-100mg。"Effective amount" or "therapeutically effective amount" as used herein refers to administering a sufficient amount of a compound disclosed herein that will alleviate one or more symptoms of the disease or condition (e.g., cancer) being treated to some extent. In some embodiments, the result is a reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired change in a biological system. For example, an "effective amount" for therapeutic use is the amount of a compound disclosed herein required to provide a clinically significant reduction in disease symptoms. Examples of therapeutically effective amounts (calculated as the free base) include, but are not limited to, 1-800 mg, 1-700 mg, 1-600 mg, 2-600 mg, 3-600 mg, 4-600 mg, 5-600 mg, 6-600 mg, 10-600 mg, 20-600 mg, 25-600 mg, 30-600 mg, 40-600 mg, 50-600 mg, 60-600 mg, 70-600 mg, 75-600 mg, 80-600 mg, 90-600 mg, 100-600 mg, 200-600 mg, 1-500 mg, 2-500 mg, 3-500 mg, 4-500 mg, 5-500 mg, 6-500 mg, 10-500 mg g, 20-500mg, 25-500mg, 30-500mg, 40-500mg, 50-500mg, 60-500mg, 70-500mg, 75-500mg, 80-500mg, 90-500mg, 100-500mg, 125-500mg, 150-500mg, 200-500mg, 250-500mg, 300-500mg, 400-500mg, 5-400mg, 10-400mg, 20-400mg, 25-400mg, 30-400mg, 40-400mg, 50-400mg, 60-400mg, 70-400mg, 75-400mg, 80-400mg, 90 -400mg, 100-400mg, 125-400mg, 150-400mg, 200-400mg, 250-400mg, 300-400mg, 1-300mg, 2-300mg, 5-300mg, 10-300mg, 20-300mg, 25-300mg, 30-300mg, 40-300mg, 50-300mg, 60-300mg, 70-300mg, 75-300mg, 80-300mg, 90-300mg, 100-300mg, 125-300mg, 150-300mg, 200-300mg, 250-300mg, 1-200mg, 2-200mg, 5-200mg g, 10-200mg, 20-200mg, 25-200mg, 30-200mg, 40-200mg, 50-200mg, 60-200mg, 70-200mg, 75-200mg, 80-200mg, 90-200mg, 100-200mg, 125-200mg, 150-200mg, 1-100mg, 2-100mg, 5-100mg, 10-100mg, 15-100mg, 20-100mg, 25-100mg, 30-100mg, 40-100mg, 50-100mg, 60-100mg, 70-100mg, 75-100mg, 80-100mg, 90-100mg.
在一些实施方案中,治疗有效量的实例包括但不限于1mg、5mg、10mg、15mg、20mg、25mg、30mg、35mg、40mg、45mg、50mg、55mg、65mg、70mg、75mg、80mg、85mg、90mg、95mg、100mg、110mg、120mg、125mg、130mg、140mg、150mg、160mg、170mg、180mg、190mg、200mg、210mg、220mg、230mg、240mg、250mg、300mg。 In some embodiments, examples of therapeutically effective amounts include, but are not limited to, 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 125 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, 210 mg, 220 mg, 230 mg, 240 mg, 250 mg, 300 mg.
一种用于治疗哺乳动物的疾病的方法,所述方法包括给予受试者治疗有效量的本发明化合物可药用盐或共晶,治疗有效量优选1-800mg,所述的疾病优选抑制或降解EGFR相关疾病(优选癌症)。A method for treating a disease in a mammal, comprising administering to a subject a therapeutically effective amount of a pharmaceutically acceptable salt or cocrystal of the compound of the present invention, preferably 1-800 mg, wherein the disease is preferably a disease related to the inhibition or degradation of EGFR (preferably cancer).
一种用于治疗哺乳动物的疾病的方法,所述方法包括,将药物本发明化合物可药用盐或共晶以1-1000mg/天的日剂量给予受试者,所述日剂量可以为单剂量或分剂量,在一些实施方案中,日剂量包括但不限于10-1500mg/天、10-1000mg/天、10-800mg/天、25-800mg/天、50-800mg/天、100-800mg/天、200-800mg/天、25-400mg/天、50-400mg/天、100-400mg/天、200-400mg/天,在一些实施方案中,日剂量包括但不限于10mg/天、20mg/天、25mg/天、50mg/天、80mg/天、100mg/天、125mg/天、150mg/天、160mg/天、200mg/天、300mg/天、320mg/天、400mg/天、480mg/天、600mg/天、640mg/天、800mg/天、1000mg/天。A method for treating a disease in a mammal, the method comprising administering a pharmaceutically acceptable salt or co-crystal of a compound of the present invention to a subject at a daily dose of 1-1000 mg/day, the daily dose may be a single dose or divided doses, in some embodiments, the daily dose includes but is not limited to 10-1500 mg/day, 10-1000 mg/day, 10-800 mg/day, 25-800 mg/day, 50-800 mg/day, 100-800 mg/day, 200-800 mg/day, 25-400 mg/day, 50- 400 mg/day, 100-400 mg/day, 200-400 mg/day, in some embodiments, daily doses include but are not limited to 10 mg/day, 20 mg/day, 25 mg/day, 50 mg/day, 80 mg/day, 100 mg/day, 125 mg/day, 150 mg/day, 160 mg/day, 200 mg/day, 300 mg/day, 320 mg/day, 400 mg/day, 480 mg/day, 600 mg/day, 640 mg/day, 800 mg/day, 1000 mg/day.
本发明涉及一种试剂盒,该试剂盒可以包括单剂量或多剂量形式的组合物,该试剂盒包含本发明化合物可药用盐或共晶,本发明化合物可药用盐或共晶的量与上述药物组合物中其量相同。The present invention relates to a kit, which may include a composition in a single-dose or multi-dose form, and the kit contains a pharmaceutically acceptable salt or co-crystal of the compound of the present invention, and the amount of the pharmaceutically acceptable salt or co-crystal of the compound of the present invention is the same as that in the above-mentioned pharmaceutical composition.
本发明所述的式(I)所示化合物的晶型具有优良的物理性质,其包括但不限于溶解度、溶出率、耐光照性、低吸湿性、耐高温性、耐高湿性。例如,本发明所述的晶型在制剂过程中可明显降低过滤时间,缩短生产周期,节约成本。本发明所述的晶型还具有良好的光稳定性、热稳定性和湿稳定性,可保证所述晶型在储存和运输时的可靠性,从而保证制剂的安全性,并且所述晶型不需要为防止受光照、温度和湿度的影响而采取特殊包装处理,从而降低了成本。所述晶型不会因光照、高温和高湿影响产生降解,提高了制剂的安全性和长期贮藏后的有效性。服用所述晶型的患者不会担忧制剂因暴露于日光下产生光敏反应。The crystal form of the compound shown in formula (I) of the present invention has excellent physical properties, including but not limited to solubility, dissolution rate, light resistance, low hygroscopicity, high temperature resistance, and high humidity resistance. For example, the crystal form of the present invention can significantly reduce the filtration time, shorten the production cycle, and save costs during the preparation process. The crystal form of the present invention also has good light stability, thermal stability, and wet stability, which can ensure the reliability of the crystal form during storage and transportation, thereby ensuring the safety of the preparation, and the crystal form does not need to be specially packaged to prevent the influence of light, temperature, and humidity, thereby reducing costs. The crystal form will not be degraded due to the influence of light, high temperature, and high humidity, thereby improving the safety of the preparation and the effectiveness after long-term storage. Patients taking the crystal form will not worry about the photosensitivity reaction of the preparation due to exposure to sunlight.
本发明所述的式(I)所示化合物的晶型在环境温度下储存或运输时极少或较少降解,具有较好的热稳定性,可长时间稳定保持,且适用于标准的制剂生产过程。The crystalline form of the compound represented by formula (I) of the present invention degrades very little or less when stored or transported at ambient temperature, has good thermal stability, can be stably maintained for a long time, and is suitable for standard preparation production processes.
本发明所述的式(I)所示化合物的晶型适合和便于大量制备,用前述晶型制备得到的制剂可减少刺激性并提高吸收,使得代谢速度方面的问题得以解决,毒性得以显著降低,安全性得以提高,有效地保证了制剂的质量和效能。The crystal form of the compound represented by formula (I) described in the present invention is suitable and convenient for mass preparation. The preparation prepared using the aforementioned crystal form can reduce irritation and improve absorption, so that the problem of metabolic rate can be solved, toxicity can be significantly reduced, safety can be improved, and the quality and efficacy of the preparation can be effectively guaranteed.
其中可以理解的是,本发明所述的“优选地,……,进一步在以下2θ位置具有特征衍射峰”,或者“更优选地,……,更进一步在以下2θ位置具有特征衍射峰”等等诸如此类的表达,是指在前面所述2θ位置具有特征衍射峰的基础上,进一步还在所述的“以下2θ位置”具有特征衍射峰。It can be understood that the expressions such as “preferably, ..., further having a characteristic diffraction peak at the following 2θ position” or “more preferably, ..., further having a characteristic diffraction peak at the following 2θ position” described in the present invention mean that on the basis of having a characteristic diffraction peak at the aforementioned 2θ position, there is further a characteristic diffraction peak at the “following 2θ position”.
可以理解的是,本发明描述的和保护的数值为近似值。数值内的变化可能归因于设备的校准、设备误差、晶体的纯度、晶体大小、样本大小以及其他因素。It is understood that the numerical values described and protected by the present invention are approximate values. The variation in the numerical values may be due to the calibration of the equipment, equipment error, purity of the crystal, crystal size, sample size and other factors.
本发明晶型结构可以使用本领域普通技术人员已知的各种分析技术分析,包括但不限于,X-射线粉末衍射(XRD)、离子色谱(IC)、差示扫描量热法(DSC)和/或热重分析(Thermogravimetric Analysis,TGA),又叫热重法(Thermogravimetry,TG)。The crystalline structure of the present invention can be analyzed using various analytical techniques known to those skilled in the art, including but not limited to, X-ray powder diffraction (XRD), ion chromatography (IC), differential scanning calorimetry (DSC) and/or thermogravimetric analysis (TGA), also known as thermogravimetry (TG).
可以理解的是,本发明的晶型不限于与本发明公开的附图中描述的特征图谱完全相同的特征图谱,比如XRD、DSC、TGA,具有与附图中描述的哪些图谱基本上相同或本质上相同的特征图谱的任何晶型均落入本发明的范围内。It is to be understood that the crystal forms of the present invention are not limited to characteristic spectra that are completely identical to the characteristic spectra described in the accompanying drawings disclosed in the present invention, such as XRD, DSC, TGA, and any crystal forms having characteristic spectra that are substantially the same or essentially the same as those described in the accompanying drawings fall within the scope of the present invention.
可以理解的是,差示扫描量热(DSC)领域中所熟知的,DSC曲线的熔融峰高取决于与样品制备和仪器几何形状有关的许多因素,而峰位置对实验细节相对不敏感。因此,在一些实施方案中,本发明的结晶化合物的特征在于具有特征峰位置的DSC图,具有与本发明附图中提供的DSC图实质上相同的性质,误差容限为±3℃。It is understood that, as is well known in the field of differential scanning calorimetry (DSC), the melting peak height of a DSC curve depends on many factors related to sample preparation and instrument geometry, while the peak position is relatively insensitive to experimental details. Therefore, in some embodiments, the crystalline compound of the present invention is characterized by a DSC pattern with a characteristic peak position, having substantially the same properties as the DSC pattern provided in the accompanying drawings of the present invention, with an error tolerance of ± 3 ° C.
除非另有说明,本文使用的所述技术和科学术语具有与本发明所属领域技术人员通常所理解的 相同的含义。若存在矛盾,则以本申请提供的定义为准。当以范围、优选范围、或者优选的数值上限以及优选的数值下限的形式表述某个量、浓度或其他值或参数的时候,应当理解相当于具体揭示了通过将任意一对范围上限或优选数值与任意范围下限或优选数值结合起来的任何范围,而不考虑该范围是否具体揭示。除非另有说明,本文所列出的数值范围旨在包括范围的端点和该范围内的所有整数和分数(小数)。Unless otherwise defined, the technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. The same meaning. If there is a contradiction, the definition provided in the present application shall prevail. When a certain amount, concentration or other value or parameter is expressed in the form of a range, a preferred range, or a preferred upper numerical limit and a preferred lower numerical limit, it should be understood that it is equivalent to specifically revealing any range by combining any pair of upper range limits or preferred values with any lower range limit or preferred values, regardless of whether the range is specifically disclosed. Unless otherwise specified, the numerical ranges listed herein are intended to include the endpoints of the range and all integers and fractions (decimals) within the range.
除非有相反的陈述,在说明书和权利要求书中使用的术语具有下述含义。Unless stated otherwise, the terms used in the specification and claims have the following meanings.
本发明所述的“任选”或“任选地”是指随后所描述的事件或环境可以但不必须发生,该说明包括该事件或环境发生或不发生的场合。The term "optional" or "optionally" used herein means that the subsequently described event or circumstance may but need not occur, and the description includes instances where the event or circumstance occurs or does not occur.
本发明所述的“约”、“大约”当与数值变量并用时,通常指该变量的数值和该变量的所有数值在实验误差内(例如对于平均值95%的置信区间内)或在指定数值的±10%内,或更宽范围内。When used with a numerical variable, the terms "about" and "approximately" used herein generally refer to the numerical value of the variable and all numerical values of the variable within experimental error (e.g., within a 95% confidence interval for the mean value) or within ±10% of the specified numerical value, or a wider range.
除非另有说明,本文的百分比、份数等都按重量计。Unless otherwise specified, percentages, parts, etc. herein are all by weight.
本发明所述的“无定型”是指三维上无排序的任意固体物质。在一些情况中,无定形固体可通过已知技术表征,所述技术包括XRPD晶体衍射分析、差示扫描量热(DSC)、固态核磁共振(ssNMR)波谱分析或这些技术的组合。如以下所说明,无定形固体产生的XRPD图谱无明显的衍射特征峰。"Amorphous" as used herein refers to any solid material that is not ordered in three dimensions. In some cases, amorphous solids can be characterized by known techniques, including XRPD crystal diffraction analysis, differential scanning calorimetry (DSC), solid-state nuclear magnetic resonance (ssNMR) spectroscopy, or a combination of these techniques. As described below, the XRPD pattern produced by an amorphous solid has no obvious diffraction characteristic peaks.
本发明所述的“晶型”或“晶体”是指呈现三维排序的任意固体物质,与无定型固体物质相反,其产生具有边界清楚的峰的特征性XRPD图谱。As used herein, "crystalline form" or "crystal" refers to any solid material that exhibits a three-dimensional ordering, in contrast to an amorphous solid material, which produces a characteristic XRPD pattern with well-defined peaks.
本发明所述的“晶种”是指在结晶法中,通过加入不溶的添加物,形成晶核,加快或促进与之晶型或立体构型相同的对映异构体结晶的生长。The "seed crystals" mentioned in the present invention refer to the formation of crystal nuclei by adding insoluble additives in the crystallization method, which accelerates or promotes the growth of enantiomer crystals with the same crystal form or stereo configuration.
本发明所述的“药物组合物”表示一种或多种文本所述化合物或其生理学/药学上可接受的盐与其他组成成分的混合物,其中其它组分包含生理学/药学上可接受的载体和赋形剂。The "pharmaceutical composition" described in the present invention refers to a mixture of one or more compounds described herein or their physiologically/pharmaceutically acceptable salts and other components, wherein the other components include physiologically/pharmaceutically acceptable carriers and excipients.
本发明所述的“载体”指的是不会对生物体产生明显刺激且不会消除所给予化合物的生物活性和特性的载体或稀释剂。The "carrier" mentioned in the present invention refers to a carrier or diluent that does not cause significant irritation to an organism and does not eliminate the biological activity and properties of the administered compound.
本发明所述的“赋形剂”指的是加入到药物组合物中以进一步依赖于化合物给药的惰性物质。The term "excipient" as used herein refers to an inert substance added to a pharmaceutical composition to further enhance the administration of a compound.
本发明所述的“IC50”指半数抑制浓度,指达到最大抑制效果一半时的浓度。The "IC 50 " in the present invention refers to the half inhibitory concentration, which refers to the concentration at which half of the maximum inhibitory effect is achieved.
本发明所述的“醚类溶剂”是指含有醚键-O-且碳原子数为1至10个的链状化合物或环状化合物,具体实例包括但不限于:四氢呋喃、乙醚、丙二醇甲醚、甲基叔丁基醚、异丙醚或1,4-二氧六环。The "ether solvent" described in the present invention refers to a chain compound or a cyclic compound containing an ether bond -O- and having 1 to 10 carbon atoms. Specific examples include but are not limited to: tetrahydrofuran, ethyl ether, propylene glycol methyl ether, methyl tert-butyl ether, isopropyl ether or 1,4-dioxane.
本发明所述的“醇类溶剂”是指一个或多个“羟基”取代“C1-6烷基”上的一个或多个氢原子所衍生的基团,所述“羟基”和“C1-6烷基”如前文所定义,具体实例包括但不限于:甲醇、乙醇、异丙醇、正丙醇、异戊醇或三氟乙醇。The "alcohol solvent" described in the present invention refers to a group derived from one or more "hydroxyl groups" replacing one or more hydrogen atoms on a "C 1-6 alkyl group", wherein the "hydroxyl group" and "C 1-6 alkyl group" are as defined above, and specific examples include but are not limited to: methanol, ethanol, isopropanol, n-propanol, isopentanol or trifluoroethanol.
本发明所述的“酯类溶剂”是指含碳原子数为1至4个的低级有机酸与含碳原子数为1至6个的低级醇的结合物,具体实例包括但不限于:乙酸乙酯、乙酸异丙酯或乙酸丁酯。The "ester solvent" described in the present invention refers to a combination of a low-level organic acid containing 1 to 4 carbon atoms and a low-level alcohol containing 1 to 6 carbon atoms. Specific examples include but are not limited to: ethyl acetate, isopropyl acetate or butyl acetate.
本发明所述的“酮类溶剂”是指羰基(-C(O)-)与两个烃基相连的化合物,根据分子中烃基的不同,酮可分为脂肪酮、脂环酮、芳香酮、饱和酮和不饱和酮,具体实例包括但不限于:丙酮、苯乙酮、4-甲基-2-戊酮。The "ketone solvent" described in the present invention refers to a compound in which a carbonyl group (-C(O)-) is connected to two hydrocarbon groups. Depending on the different hydrocarbon groups in the molecule, ketones can be divided into aliphatic ketones, alicyclic ketones, aromatic ketones, saturated ketones and unsaturated ketones. Specific examples include but are not limited to: acetone, acetophenone, 4-methyl-2-pentanone.
本发明所述的“腈类溶剂”是指一个或多个“氰基”取代“C1-6烷基”上的一个或多个氢原子所衍生的基团,所述“氰基”和“C1-6烷基”如前文所定义,具体实例包括但不限于:乙腈或丙腈。The "nitrile solvent" described in the present invention refers to a group derived from one or more "cyano" replacing one or more hydrogen atoms on a "C 1-6 alkyl". The "cyano" and "C 1-6 alkyl" are as defined above, and specific examples include but are not limited to: acetonitrile or propionitrile.
本发明所述的“卤代烃类溶剂”是指一个或多个“卤素原子”取代“C1-6烷基”上的一个或多个氢原子所衍生的基团,所述“卤素原子”和“C1-6烷基”如前文所定义,具体实例包括但不限于:二氯甲烷、1,2-二氯乙烷、氯仿或四氯化碳。 The "halogenated hydrocarbon solvent" described in the present invention refers to a group derived from one or more "halogen atoms" replacing one or more hydrogen atoms on a "C 1-6 alkyl group", wherein the "halogen atom" and "C 1-6 alkyl group" are as defined above, and specific examples include but are not limited to: dichloromethane, 1,2-dichloroethane, chloroform or carbon tetrachloride.
本发明所述的“本发明的晶体”、“本发明的晶型”、“本发明的多晶型物”等可互换使用。The "crystal of the present invention", "crystal form of the present invention", "polymorph of the present invention" and the like described in the present invention can be used interchangeably.
本发明所述“室温”一般指4~30℃,优选地指20±5℃。The "room temperature" mentioned in the present invention generally refers to 4-30°C, preferably refers to 20±5°C.
本发明所述干燥温度一般为20~100℃,优选25~70℃,可以为,可以常压干燥,也可以减压干燥(真空干燥)。优选的,干燥在减压下干燥。The drying temperature of the present invention is generally 20 to 100° C., preferably 25 to 70° C., and can be either normal pressure drying or reduced pressure drying (vacuum drying). Preferably, the drying is carried out under reduced pressure.
本发明所述的“X射线粉末衍射图谱(XRPD图谱)”是指实验观察的衍射图或源于其的参数、数据或值。XRPD图谱通常由峰位(横坐标)和/或峰强度(纵坐标)表征。The "X-ray powder diffraction pattern (XRPD pattern)" of the present invention refers to an experimentally observed diffraction pattern or a parameter, data or value derived therefrom. An XRPD pattern is usually characterized by peak position (abscissa) and/or peak intensity (ordinate).
本发明所述的“2θ或2θ角度”是指指衍射角,θ为布拉格角,是基于X射线衍射实验中设置的以度数(°)表示的峰位,并且通常是在衍射图谱中的横坐标单位。如果入射束与某晶格面形成θ角时反射被衍射,则实验设置需要以2θ角记录反射束。应当理解,在本文中提到的特定晶型的特定2θ值意图表示使用本文所述的X射线衍射实验条件所测量的2θ值(以度数表示),所述2θ的误差范围为±0.3,可以是±0.3、±0.2或±0.1。The "2θ or 2θ angle" described in the present invention refers to the diffraction angle, θ is the Bragg angle, which is based on the peak position expressed in degrees (°) set in the X-ray diffraction experiment, and is usually the horizontal coordinate unit in the diffraction spectrum. If the incident beam forms an angle θ with a certain lattice plane and the reflection is diffracted, the experimental setting needs to record the reflected beam at an angle of 2θ. It should be understood that the specific 2θ value of a specific crystal form mentioned in this article is intended to represent the 2θ value (expressed in degrees) measured using the X-ray diffraction experimental conditions described herein, and the error range of 2θ is ±0.3, which can be ±0.3, ±0.2 or ±0.1.
本发明所述的“基本上相同”意指将代表性峰位和强度变化考虑在内。例如,本领域技术人员会理解峰位(2θ)会显示一些变化,通常多达0.1~0.2度,并且用于测量衍射的仪器也会导致一些变化。另外,本领域技术人员会理解相对峰强度会因仪器间的差异以及结晶性程度、择优取向、制备的样品表面及本领域技术人员已知的其它因素而出现变化,并应将其看作仅为定性测量。"Substantially the same" as used herein means that representative peak positions and intensity variations are taken into account. For example, one skilled in the art will appreciate that peak positions (2θ) will show some variation, typically up to 0.1 to 0.2 degrees, and that the instrument used to measure diffraction will also cause some variation. In addition, one skilled in the art will appreciate that relative peak intensities will vary due to instrumental differences as well as degree of crystallinity, preferred orientation, prepared sample surface, and other factors known to one skilled in the art, and should be considered as only qualitative measurements.
本发明所述的“差示扫描量热法或DSC”是指在样品升温或恒温过程中,测量样品与参考物之间的温度差、热流差,以表征所有与热效应有关的物理变化和化学变化,得到样品的相变信息。The "differential scanning calorimetry or DSC" mentioned in the present invention refers to measuring the temperature difference and heat flow difference between a sample and a reference object during the process of heating or maintaining a constant temperature of the sample, so as to characterize all physical and chemical changes related to thermal effects and obtain the phase change information of the sample.
依据《中国药典》2020年版四部中“9103药物引湿性指导原则”中引湿性特征描述与引湿性增重的界定,According to the description of hygroscopic characteristics and the definition of hygroscopic weight gain in the "9103 Drug Hygroscopicity Guidance" in Part IV of the 2020 edition of the Chinese Pharmacopoeia,
潮解:吸收足量水分形成液体;Deliquesce: Absorbs enough water to form a liquid;
极具引湿性:引湿增重不小于15%;Highly hygroscopic: weight gain due to moisture absorption is not less than 15%;
有引湿性:引湿增重小于15%但不小于2%;Hygroscopic: weight gain due to moisture absorption is less than 15% but not less than 2%;
略有引湿性:引湿增重小于2%但不小于0.2%;Slightly hygroscopic: weight gain due to moisture absorption is less than 2% but not less than 0.2%;
无或几乎无引湿性:引湿增重小于0.2%。No or almost no hygroscopicity: moisture gain is less than 0.2%.
本发明公开的晶型可以经如下的常见的制备晶型的方法制备:The crystal form disclosed in the present invention can be prepared by the following common methods for preparing crystal forms:
1、挥发实验是将样品澄清溶液在不同温度下敞口挥发至溶剂干。1. The volatilization experiment is to evaporate the clear solution of the sample at different temperatures until the solvent is dry.
2、晶浆实验是将样品的过饱和溶液(有不溶固体存在)在不同溶剂体系中某个温度下进行搅拌。2. The slurry experiment is to stir the supersaturated solution of the sample (with insoluble solids) at a certain temperature in different solvent systems.
3、抗溶剂实验是取样品溶解在良溶剂中,加入抗溶剂,析出固体短时搅拌后立即过滤处理。3. The anti-solvent test is to dissolve the sample in a good solvent, add an anti-solvent, stir the precipitated solid for a short time and then filter it immediately.
4、冷却结晶实验是在高温下将一定量的样品溶解到相应溶剂中,然后直接在室温或低温搅拌析晶。4. The cooling crystallization experiment is to dissolve a certain amount of sample into the corresponding solvent at high temperature, and then stir and crystallize directly at room temperature or low temperature.
5、高分子模板实验是在样品澄清溶液中加入不同种类的高分子材料,置于室温下敞口挥发至溶剂干。5. The polymer template experiment is to add different types of polymer materials to the sample clear solution and leave it open at room temperature to evaporate until the solvent is dry.
6、热方法实验是将样品按一定热方法结晶条件处理并冷却至室温。6. The thermal method experiment is to treat the sample under certain thermal method crystallization conditions and cool it to room temperature.
7、水汽扩散实验是将样品在室温下一定湿度环境中放置。7. The water vapor diffusion experiment is to place the sample in a certain humidity environment at room temperature.
附图说明BRIEF DESCRIPTION OF THE DRAWINGS
图1为式(I)所示化合物的马来酸盐晶型1的X-射线粉末衍射图谱。FIG1 is an X-ray powder diffraction pattern of maleate salt form 1 of the compound represented by formula (I).
图2为式(I)所示化合物的马来酸盐晶型1的热重分析图谱。FIG2 is a thermogravimetric analysis spectrum of the maleate salt form 1 of the compound represented by formula (I).
图3为式(I)所示化合物的马来酸盐晶型1的差示扫描量热分析曲线。 FIG3 is a differential scanning calorimetry curve of maleate salt form 1 of the compound represented by formula (I).
图4为式(I)所示化合物的马来酸盐晶型1的等温吸附曲线。FIG4 is an isothermal adsorption curve of maleate salt form 1 of the compound represented by formula (I).
图5为式(I)所示化合物的马来酸盐晶型1的DVS图谱。FIG5 is a DVS spectrum of the maleate salt form 1 of the compound represented by formula (I).
图6为式(I)所示化合物的二马来酸盐晶型1的X-射线粉末衍射图谱。FIG6 is an X-ray powder diffraction pattern of the dimaleate crystalline form 1 of the compound represented by formula (I).
图7为式(I)所示化合物的二马来酸盐晶型1的热重分析图谱。FIG. 7 is a thermogravimetric analysis spectrum of the dimaleate crystal form 1 of the compound represented by formula (I).
图8为式(I)所示化合物的二马来酸盐晶型1的差示扫描量热分析曲线。FIG8 is a differential scanning calorimetry curve of the dimaleate crystal form 1 of the compound represented by formula (I).
图9为式(I)所示化合物的二马来酸盐晶型1的等温吸附曲线。FIG9 is an isothermal adsorption curve of the dimaleate crystal form 1 of the compound represented by formula (I).
图10为式(I)所示化合物的二马来酸盐晶型1的DVS图谱。FIG10 is a DVS spectrum of the dimaleate crystal form 1 of the compound represented by formula (I).
图11为式(I)所示化合物的2-萘磺酸晶型1的X-射线粉末衍射图谱。FIG11 is an X-ray powder diffraction pattern of 2-naphthalenesulfonic acid form 1 of the compound represented by formula (I).
图12为式(I)所示化合物的2-萘磺酸晶型1的热重分析图谱。FIG12 is a thermogravimetric analysis spectrum of 2-naphthalenesulfonic acid form 1 of the compound represented by formula (I).
图13为式(I)所示化合物的2-萘磺酸晶型1的差示扫描量热分析曲线。FIG13 is a differential scanning calorimetry curve of 2-naphthalenesulfonic acid form 1 of the compound represented by formula (I).
图14为式(I)所示化合物的2-萘磺酸晶型1的等温吸附曲线。FIG14 is an isothermal adsorption curve of 2-naphthalenesulfonic acid form 1 of the compound represented by formula (I).
图15为式(I)所示化合物的2-萘磺酸晶型1的DVS图谱。FIG15 is a DVS spectrum of 2-naphthalenesulfonic acid form 1 of the compound represented by formula (I).
图16为式(I)所示化合物的草酸盐晶型1的X-射线粉末衍射图谱。FIG16 is an X-ray powder diffraction pattern of oxalate crystal form 1 of the compound represented by formula (I).
图17为式(I)所示化合物的草酸盐晶型1的热重分析图谱。FIG. 17 is a thermogravimetric analysis spectrum of the oxalate salt form 1 of the compound represented by formula (I).
图18为式(I)所示化合物的草酸盐晶型1的差示扫描量热分析曲线。FIG18 is a differential scanning calorimetry curve of the oxalate crystal form 1 of the compound represented by formula (I).
图19为式(I)所示化合物的苯磺酸盐无定型的X-射线粉末衍射图谱。FIG19 is an X-ray powder diffraction pattern of the amorphous benzenesulfonate salt of the compound represented by formula (I).
图20为式(I)所示化合物的三苯磺酸盐无定型的X-射线粉末衍射图谱。FIG. 20 is an X-ray powder diffraction pattern of the amorphous triphenylsulfonate salt of the compound represented by formula (I).
图21为式(I)所示化合物的二L-苹果酸盐无定型的X-射线粉末衍射图谱。FIG21 is an X-ray powder diffraction pattern of the amorphous di-L-malate salt of the compound represented by formula (I).
图22为式(I)所示化合物的磷酸盐无定型的X-射线粉末衍射图谱。FIG. 22 is an X-ray powder diffraction pattern of the amorphous phosphate of the compound represented by formula (I).
图23为式(I)所示化合物的二磷酸盐无定型的X-射线粉末衍射图谱。FIG. 23 is an X-ray powder diffraction pattern of the amorphous diphosphate of the compound represented by formula (I).
图24为式(I)所示化合物的硫酸盐无定型的X-射线粉末衍射图谱。FIG. 24 is an X-ray powder diffraction pattern of the amorphous sulfate salt of the compound represented by formula (I).
图25为式(I)所示化合物的二硫酸盐无定型的X-射线粉末衍射图谱。FIG25 is an X-ray powder diffraction pattern of the amorphous disulfate salt of the compound represented by formula (I).
图26为式(I)所示化合物的三硫酸盐无定型的X-射线粉末衍射图谱。FIG26 is an X-ray powder diffraction pattern of the amorphous trisulfate salt of the compound represented by formula (I).
图27为式(I)所示化合物的二对甲苯磺酸盐无定型的X-射线粉末衍射图谱。FIG. 27 is an X-ray powder diffraction pattern of the amorphous di-p-toluenesulfonate salt of the compound represented by formula (I).
图28为式(I)所示化合物的盐酸盐无定型的X-射线粉末衍射图谱。FIG28 is an X-ray powder diffraction pattern of the amorphous hydrochloride salt of the compound represented by formula (I).
图29为式(I)所示化合物的二盐酸盐无定型的X-射线粉末衍射图谱。FIG29 is an X-ray powder diffraction pattern of the amorphous dihydrochloride salt of the compound represented by formula (I).
图30为式(I)所示化合物的三盐酸盐无定型的X-射线粉末衍射图谱。FIG30 is an X-ray powder diffraction pattern of the amorphous trihydrochloride salt of the compound represented by formula (I).
图31为式(I)所示化合物的二-2-萘磺酸盐无定型的X-射线粉末衍射图谱。FIG31 is an X-ray powder diffraction pattern of the amorphous di-2-naphthalenesulfonate salt of the compound represented by formula (I).
图32为式(I)所示化合物的氢溴酸盐无定型的X-射线粉末衍射图谱。FIG32 is an X-ray powder diffraction pattern of the amorphous hydrobromide salt of the compound represented by formula (I).
图33为式(I)所示化合物的二氢溴酸盐无定型的X-射线粉末衍射图谱。FIG33 is an X-ray powder diffraction pattern of the amorphous dihydrobromide salt of the compound represented by formula (I).
图34为式(I)所示化合物的三氢溴酸盐无定型的X-射线粉末衍射图谱。FIG34 is an X-ray powder diffraction pattern of the amorphous trihydrobromide salt of the compound represented by formula (I).
图35为式(I)所示化合物的甲磺酸盐无定型的X-射线粉末衍射图谱。FIG35 is an X-ray powder diffraction pattern of the amorphous methanesulfonate salt of the compound represented by formula (I).
图36为式(I)所示化合物的二甲磺酸盐无定型的X-射线粉末衍射图谱。FIG36 is an X-ray powder diffraction pattern of the amorphous dimesylate salt of the compound represented by formula (I).
图37为式(I)所示化合物的三甲磺酸盐无定型的X-射线粉末衍射图谱。FIG37 is an X-ray powder diffraction pattern of the amorphous trimesylate salt of the compound represented by formula (I).
图38为式(I)所示化合物的扁桃酸盐无定型的X-射线粉末衍射图谱。FIG38 is an X-ray powder diffraction pattern of the amorphous mandelate salt of the compound represented by formula (I).
图39为式(I)所示化合物的二扁桃酸盐无定型的X-射线粉末衍射图谱。FIG39 is an X-ray powder diffraction pattern of the amorphous bimandelate salt of the compound represented by formula (I).
图40为式(I)所示化合物的琥珀酸盐无定型的X-射线粉末衍射图谱。FIG40 is an X-ray powder diffraction pattern of the amorphous succinate salt of the compound represented by formula (I).
图41为式(I)所示化合物的水杨酸盐无定型的X-射线粉末衍射图谱。FIG41 is an X-ray powder diffraction pattern of the amorphous salicylate of the compound represented by formula (I).
图42为式(I)所示化合物的1,5-萘二磺酸盐无定型的X-射线粉末衍射图谱。 FIG42 is an X-ray powder diffraction pattern of amorphous 1,5-naphthalene disulfonate of the compound represented by formula (I).
图43为式(I)所示化合物的二-1,5-萘二磺酸盐无定型的X-射线粉末衍射图谱。FIG43 is an X-ray powder diffraction pattern of amorphous di-1,5-naphthalene disulfonate of the compound represented by formula (I).
图44为式(I)所示化合物的半富马酸盐无定型的X-射线粉末衍射图谱。FIG44 is an X-ray powder diffraction pattern of the amorphous hemi-fumarate salt of the compound represented by formula (I).
图45为式(I)所示化合物的二富马酸盐无定型的X-射线粉末衍射图谱。Figure 45 is an X-ray powder diffraction pattern of the amorphous difumarate salt of the compound represented by formula (I).
图46为式(I)所示化合物的三烟酸盐无定型的X-射线粉末衍射图谱。FIG46 is an X-ray powder diffraction pattern of the amorphous trinicotinate salt of the compound represented by formula (I).
图47为式(I)所示化合物的马尿酸盐无定型的X-射线粉末衍射图谱。FIG47 is an X-ray powder diffraction pattern of the amorphous hippurate salt of the compound represented by formula (I).
图48为式(I)所示化合物的晶型1的X-射线粉末衍射图谱。Figure 48 is an X-ray powder diffraction pattern of Form 1 of the compound represented by formula (I).
图49为式(I)所示化合物的晶型1的热重分析图谱。FIG49 is a thermogravimetric analysis spectrum of Form 1 of the compound represented by formula (I).
图50为式(I)所示化合物的晶型1的差示扫描量热分析曲线。Figure 50 is a differential scanning calorimetry curve of Form 1 of the compound represented by formula (I).
具体实施方式Detailed ways
化合物的结构是通过核磁共振(NMR)或(和)质谱(MS)来确定的。NMR位移(δ)以10-6(ppm)的单位给出。NMR的测定是用(Bruker Avance III 400和Bruker Avance 300)核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代氯仿(CDCl3),氘代甲醇(CD3OD),内标为四甲基硅烷(TMS)。The structures of the compounds were determined by nuclear magnetic resonance (NMR) or (and) mass spectrometry (MS). NMR shifts (δ) are given in units of 10 -6 (ppm). NMR measurements were performed using (Bruker Avance III 400 and Bruker Avance 300) NMR spectrometers, with deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD) as the solvent, and tetramethylsilane (TMS) as the internal standard.
MS的测定用(Agilent 6120B(ESI)和Agilent 6120B(APCI))。MS was used for determination (Agilent 6120B (ESI) and Agilent 6120B (APCI)).
HPLC的测定使用安捷伦1260DAD高压液相色谱仪(Eclipse Plus C18,150×4.6mm)。HPLC determination was performed using an Agilent 1260DAD high pressure liquid chromatograph (Eclipse Plus C18, 150×4.6mm).
本发明的己知的起始原料可以采用或按照本领域已知的方法来合成,或可购买于泰坦科技、安耐吉化学、上海德默、成都科龙化工、韶远化学科技、百灵威科技等公司。The known starting materials of the present invention can be synthesized by methods known in the art, or can be purchased from companies such as Titan Technology, Anage Chemical, Shanghai Demo, Chengdu Kelon Chemical, Shaoyuan Chemical Technology, and Bailingwei Technology.
以下通过具体实施例详细说明本发明的实施过程和产生的有益效果,旨在帮助阅读者更好地理解本发明的实质和特点,不作为对本案可实施范围的限定。The following describes in detail the implementation process of the present invention and the beneficial effects produced by the specific embodiments, which is intended to help readers better understand the essence and characteristics of the present invention, and is not intended to limit the scope of implementation of the present invention.
实施例1:化合物1的制备
Example 1: Preparation of Compound 1
第一步:1C的制备Step 1: Preparation of 1C
将1A(16g,56.45mmol)和1B(14.8g,59.27mmol)溶于DMSO(200mL)中,加入碳酸钾(23.5g,0.17mol),120℃搅拌反应4小时,反应液冷却至室温,加入300mL水,有黄色固体析出,抽滤,滤饼水洗3次,二氯甲烷将滤饼复溶,无水硫酸钠干燥,减压浓缩,柱层析纯化(流动相:二氯甲烷/甲醇(V/V)=50/1-15/1)得到1C(22g,收率:76%)。1A (16 g, 56.45 mmol) and 1B (14.8 g, 59.27 mmol) were dissolved in DMSO (200 mL), potassium carbonate (23.5 g, 0.17 mol) was added, and the mixture was stirred at 120°C for 4 hours. The reaction solution was cooled to room temperature, 300 mL of water was added, and a yellow solid was precipitated. The solid was filtered and the filter cake was washed with water 3 times. The filter cake was redissolved in dichloromethane, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified by column chromatography (mobile phase: dichloromethane/methanol (V/V) = 50/1-15/1) to obtain 1C (22 g, yield: 76%).
第四步:1E的制备 Step 4: Preparation of 1E
将1C(400g,0.78mol)、1D(158g,1.24mol)溶于1,4-二氧六环(1.6L)和水(0.4L)混合溶剂,加入碳酸钾(216g,1.56mol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(38g,0.047mol),氮气置换3次,90℃搅拌反应过夜,反应完全后,减压浓缩后加入500mL乙酸乙酯稀释反应液,水洗3次,饱和氯化钠洗涤1次,有机相经无水硫酸钠干燥后减压浓缩得到1E粗品,加入PE/EA=3/1混合液(1600mL)打浆30分钟,抽滤,滤饼减压浓缩得到1E(377g,收率:94%)。1C (400 g, 0.78 mol) and 1D (158 g, 1.24 mol) were dissolved in a mixed solvent of 1,4-dioxane (1.6 L) and water (0.4 L), potassium carbonate (216 g, 1.56 mol) and [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium dichloromethane complex (38 g, 0.047 mol) were added, and nitrogen was replaced three times. The reaction was stirred at 90°C overnight. After the reaction was complete, the reaction solution was concentrated under reduced pressure and then 500 mL of ethyl acetate was added to dilute the reaction solution. The reaction solution was washed with water three times and saturated sodium chloride once. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain a crude product of 1E. A PE/EA=3/1 mixed solution (1600 mL) was added and slurried for 30 minutes. The mixture was filtered and the filter cake was concentrated under reduced pressure to obtain 1E (377 g, yield: 94%).
LCMS m/z=515.3[M+H]+ LCMS m/z=515.3[M+H] +
第五步:1F的制备Step 5: Preparation of 1F
将1E(377g,0.73mol)溶于甲醇(600mL)中,加入氯化氢-二氧六环溶液(4mol/L,2.5L),室温下搅拌反应60min,减压浓缩得到粗品,粗品中加入水2.5L和浓盐酸500mL,乙酸乙酯(1.5L*2)萃取,水相用氢氧化钠溶液调节pH至13,二氯甲烷(1.5L*2)萃取,合并有机相并用无水硫酸钠干燥,抽滤,滤液减压浓缩得后加入PE/MTBE混合溶液(v/v=1/1,1.2L)打浆,抽滤,滤饼减压浓缩干得1F(256g,收率:85%)。1E (377 g, 0.73 mol) was dissolved in methanol (600 mL), and hydrogen chloride-dioxane solution (4 mol/L, 2.5 L) was added. The reaction was stirred at room temperature for 60 min, and the crude product was concentrated under reduced pressure. 2.5 L of water and 500 mL of concentrated hydrochloric acid were added to the crude product, and the product was extracted with ethyl acetate (1.5 L*2). The pH of the aqueous phase was adjusted to 13 with sodium hydroxide solution, and the product was extracted with dichloromethane (1.5 L*2). The organic phases were combined and dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a PE/MTBE mixed solution (v/v=1/1, 1.2 L) for slurrying, filtered, and the filter cake was concentrated under reduced pressure to obtain 1F (256 g, yield: 85%).
第六步:1G的制备Step 6: Preparation of 1G
将1F(120g,0.29mol)溶于DMSO(600mL)中,依次加入2-(2,6-二氧代哌啶-3-基)-5-氟异吲哚啉-1,3-二酮(80g,0.29mmol)和二异丙基乙胺(112g,0.87mmol),100℃反应过夜。加入2.0L水,析出固体,抽滤,滤饼用200mL二氯甲烷溶解、萃取,有机相用无水硫酸钠干燥后,减压浓缩,用硅胶柱层析纯化(流动相:二氯甲烷/甲醇(V/V)=100/1-10/1)得到1G(186g,收率:96%)。1F (120 g, 0.29 mol) was dissolved in DMSO (600 mL), and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione (80 g, 0.29 mmol) and diisopropylethylamine (112 g, 0.87 mmol) were added in sequence, and the mixture was reacted at 100°C overnight. 2.0 L of water was added, and the solid was precipitated and filtered. The filter cake was dissolved and extracted with 200 mL of dichloromethane. The organic phase was dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified by silica gel column chromatography (mobile phase: dichloromethane/methanol (V/V) = 100/1-10/1) to obtain 1G (186 g, yield: 96%).
第七步:1H的制备Step 7: Preparation of 1H
将1G(130g,0.19mmol)溶于THF(1.3L)和水(400mL)的混合液中,加入氯化铵(51g,0.95mmol)和锌粉(62g,0.95mmol),缓慢升温至40-60℃反应1-2小时,反应液冷却至室温,过滤,滤饼用1L的DCM洗涤,合并有机相,有机相用依次用0.5L氨水和0.5L饱和食盐水洗涤,无水硫酸钠干燥后减压浓缩,得到1H,黄色固体。1G (130 g, 0.19 mmol) was dissolved in a mixture of THF (1.3 L) and water (400 mL), and ammonium chloride (51 g, 0.95 mmol) and zinc powder (62 g, 0.95 mmol) were added. The temperature was slowly raised to 40-60 °C for reaction for 1-2 hours. The reaction solution was cooled to room temperature and filtered. The filter cake was washed with 1 L of DCM. The organic phases were combined, and the organic phases were washed with 0.5 L of ammonia water and 0.5 L of saturated brine in sequence. After drying over anhydrous sodium sulfate, the mixture was concentrated under reduced pressure to obtain 1H as a yellow solid.
LCMS m/z=641.3[M+H]+ LCMS m/z=641.3[M+H] +
第八步:化合物1的制备Step 8: Preparation of Compound 1
将1H(80g,126mmol)和1M(51g,126mmol)溶于DMF(500mL)中,加入对甲苯磺酸一水合物(48g,252mmol),100℃反应过夜。冷却至室温,缓慢加入800mL的饱和碳酸氢钠水溶液,抽滤,滤饼用二氯甲烷溶解、萃取,有机相用无水硫酸钠干燥后,减压浓缩,用硅胶柱层析纯化(二氯甲烷/甲醇(V/V)=100/1-100/4),柱层析得到64g化合物1(64g,收率:50%)。1H (80 g, 126 mmol) and 1M (51 g, 126 mmol) were dissolved in DMF (500 mL), p-toluenesulfonic acid monohydrate (48 g, 252 mmol) was added, and the mixture was reacted at 100°C overnight. After cooling to room temperature, 800 mL of saturated sodium bicarbonate aqueous solution was slowly added, and the mixture was filtered, and the filter cake was dissolved and extracted with dichloromethane. The organic phase was dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified by silica gel column chromatography (dichloromethane/methanol (V/V) = 100/1-100/4). 64 g of compound 1 (64 g, yield: 50%) was obtained by column chromatography.
LCMS m/z=502.7[(M+2H)/2]+LCMS m/z=502.7[(M+2H)/2] + .
1H NMR(400MHz,D2O/CF3COOD(v/v=1:1))δ8.23(s,1H),7.96-7.75(m,5H),7.46(s,1H),7.35(s,1H),7.31-7.15(m,2H),6.91(d,1H),5.12(dd,1H),4.22-4.07(m,5H),4.03(s,3H),3.93-3.70(m,6H),3.62-3.48(m,2H),3.39-3.18(m,4H),2.95-2.85(m,2H),2.82-2.67(m,1H),2.63-2.44(m,1H),2.37-2.16(m,3H),2.12-1.88(m,9H),1.19-1.08(m,2H),0.73-0.65(m,2H)。 1 H NMR (400 MHz, D 2 O/CF 3 COOD (v/v=1:1))δ8.23(s,1H),7.96-7.75(m,5H),7.46(s,1H),7.35( s, 1H), 7.31-7.15 (m, 2H), 6.91 (d, 1H), 5.12 (dd, 1H), 4.22-4.07 (m, 5H), 4.03 (s, 3H), 3.93-3.70 (m, 6H) ,3.62-3.48(m,2H),3.39-3.18(m,4H),2.95-2.85(m,2H),2.82-2.67(m,1H),2.63-2.44(m,1H),2.37-2.16( m, 3H), 2.12-1.88 (m, 9H), 1.19-1.08 (m, 2H), 0.73-0.65 (m, 2H).
实施例2:化合物1的马来酸盐晶型1的制备Example 2: Preparation of Maleate Salt Form 1 of Compound 1
取化合物1约100mg,加入2.0ml四氢呋喃,加入0.2ml混有13mg马来酸的四氢呋喃溶液。在室温下搅拌过夜,离心,所得固体在室温下真空干燥过夜,得到化合物1的马来酸盐晶型1,其XRD、DSC、TGA、等温吸附曲线和DVS依次为图1-5。Take about 100 mg of compound 1, add 2.0 ml of tetrahydrofuran, add 0.2 ml of tetrahydrofuran solution mixed with 13 mg of maleic acid, stir overnight at room temperature, centrifuge, and vacuum dry the obtained solid at room temperature overnight to obtain maleate crystal form 1 of compound 1, whose XRD, DSC, TGA, isothermal adsorption curve and DVS are shown in Figures 1-5 respectively.
实施例3:化合物1的二马来酸晶型1的制备 Example 3: Preparation of dimaleic acid crystal form 1 of compound 1
取化合物1约100mg,加入2.0ml四氢呋喃,加入0.4ml混有26mg马来酸的四氢呋喃溶液,析出。在室温下搅拌2天,离心,所得固体在室温下真空干燥过夜,得到化合物1的二马来酸盐晶型1,其XRD、DSC、TGA、等温吸附曲线和DVS依次为图6-10。
Take about 100 mg of compound 1, add 2.0 ml of tetrahydrofuran, add 0.4 ml of tetrahydrofuran solution mixed with 26 mg of maleic acid, and precipitate. Stir at room temperature for 2 days, centrifuge, and vacuum dry the obtained solid at room temperature overnight to obtain dimaleate crystal form 1 of compound 1, whose XRD, DSC, TGA, isothermal adsorption curve and DVS are shown in Figures 6-10 respectively.
通过1H NMR(400MHz,DMSO-d6)的峰位移解析,化合物1的化学位移5.12(dd,J=12.9,5.4Hz,1H)为38号位置的-CH峰,6.15(s,4H)的峰为马来酸的-CH峰,其比例为1:4,故可解析得到化合物1与马来酸的比例为1:2。Through the peak shift analysis of 1H NMR (400MHz, DMSO-d 6 ), the chemical shift 5.12 (dd, J=12.9, 5.4Hz, 1H) of compound 1 is the -CH peak at position 38, and the peak at 6.15 (s, 4H) is the -CH peak of maleic acid. The ratio is 1:4. Therefore, it can be analyzed that the ratio of compound 1 to maleic acid is 1:2.
实施例4:化合物1的2-萘磺酸盐晶型1的制备Example 4: Preparation of 2-naphthalenesulfonate salt form 1 of compound 1
取化合物1约100mg,加入2.0ml氯仿,加入0.2ml混有23mg 2-萘磺酸的乙醇溶液。在室温下搅拌2天,离心,所得固体在室温下真空干燥过夜,得到化合物1的2-萘磺酸盐晶型1,其XRD、DSC、TGA、等温吸附曲线和DVS依次为图11-15。Take about 100 mg of compound 1, add 2.0 ml of chloroform, and add 0.2 ml of ethanol solution mixed with 23 mg of 2-naphthalenesulfonic acid. Stir at room temperature for 2 days, centrifuge, and vacuum dry the obtained solid overnight at room temperature to obtain 2-naphthalenesulfonate salt form 1 of compound 1, whose XRD, DSC, TGA, isothermal adsorption curves and DVS are shown in Figures 11-15 respectively.
通过1H NMR(400MHz,DMSO-d6)的峰位移解析,化合物1的化学位移5.12(dd,J=12.9,5.4Hz,1H)为38号位置的-CH峰,8.31-8.14(m,4H)的峰为2-萘磺酸的峰,其比例为1:4,故可解析得到化合物1与2-萘磺酸的比例为1:1。Through the peak shift analysis of 1H NMR (400MHz, DMSO-d 6 ), the chemical shift 5.12 (dd, J=12.9,5.4Hz,1H) of compound 1 is the -CH peak at position 38, and the peaks of 8.31-8.14 (m,4H) are the peaks of 2-naphthalenesulfonic acid, and the ratio is 1:4. Therefore, it can be analyzed that the ratio of compound 1 to 2-naphthalenesulfonic acid is 1:1.
实施例5:化合物1的草酸盐晶型1的制备Example 5: Preparation of Oxalate Form 1 of Compound 1
取化合物1约50mg,加入2.0ml二氯甲烷,加入0.2ml混有15mg草酸的乙醇溶液。在室温下混匀后在4℃下搅拌过夜,离心,所得固体在室温下真空干燥过夜,得到化合物1的草酸盐晶型1,其XRD、DSC、TGA依次为图16-18。Take about 50 mg of compound 1, add 2.0 ml of dichloromethane, add 0.2 ml of ethanol solution mixed with 15 mg of oxalic acid, mix well at room temperature, stir overnight at 4°C, centrifuge, and dry the obtained solid in vacuum at room temperature overnight to obtain oxalate crystal form 1 of compound 1, whose XRD, DSC, and TGA are shown in Figures 16-18 respectively.
离子色谱(IC)检测结果显示成盐比为1:1。
Ion chromatography (IC) test results showed that the salt ratio was 1:1.
实施例6:化合物1的苯磺酸盐无定型的制备Example 6: Preparation of amorphous benzenesulfonate salt of compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.1ml混有10mg苯磺酸的甲醇溶液。在室温下搅拌过夜,加入正庚烷2.0mL和2.0mL甲基叔丁基醚,搅拌6h,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的苯磺酸盐无定型。其XRD如图19所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, add 0.1 ml of methanol solution mixed with 10 mg of benzenesulfonic acid. Stir overnight at room temperature, add 2.0 ml of n-heptane and 2.0 ml of methyl tert-butyl ether, stir for 6 hours, precipitate solid, centrifuge, and dry the obtained solid under vacuum at room temperature overnight to obtain amorphous benzenesulfonate of compound 1. Its XRD is shown in Figure 19.
实施例7:化合物1的三苯磺酸盐无定型的制备Example 7: Preparation of amorphous triphenylsulfonate salt of compound 1
取化合物1约50mg,加入1.0ml四氢呋喃,加入0.3ml混有29mg苯磺酸的四氢呋喃溶液。在室温下搅拌过夜,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的三苯磺酸盐,其XRD如图20所示。Take about 50 mg of compound 1, add 1.0 ml of tetrahydrofuran, add 0.3 ml of tetrahydrofuran solution mixed with 29 mg of benzenesulfonic acid, stir at room temperature overnight, precipitate solid, centrifuge, and dry the obtained solid under vacuum at room temperature overnight to obtain tribenzenesulfonate of compound 1, whose XRD is shown in Figure 20.
实施例8:化合物1的二-L-苹果酸盐无定型的制备 Example 8: Preparation of amorphous di-L-malate salt of compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.2ml混有15mg L-苹果酸的甲醇溶液。在室温下搅拌过夜,加入2.0mL甲基叔丁基醚,搅拌6h,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的二-L-苹果酸盐无定型。其XRD如图21所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, and add 0.2 ml of methanol solution mixed with 15 mg of L-malic acid. Stir overnight at room temperature, add 2.0 mL of methyl tert-butyl ether, stir for 6 hours, precipitate solid, centrifuge, and vacuum dry the obtained solid at room temperature overnight to obtain the amorphous di-L-malate of compound 1. Its XRD is shown in Figure 21.
实施例9:化合物1的磷酸盐无定型的制备Example 9: Preparation of amorphous phosphate of compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.1ml混有6mg磷酸的乙醇溶液,析出固体。在室温下搅拌过夜,离心,所得固体在室温下真空干燥过夜,得到化合物1的磷酸盐无定型,其XRD如图22所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, add 0.1 ml of ethanol solution mixed with 6 mg of phosphoric acid, and precipitate solid. Stir overnight at room temperature, centrifuge, and dry the obtained solid in vacuum at room temperature overnight to obtain amorphous phosphate of compound 1, whose XRD is shown in Figure 22.
实施例10:化合物1的二磷酸盐无定型的制备Example 10: Preparation of the amorphous diphosphate salt of compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有13mg磷酸的乙醇溶液,析出固体。在室温下搅拌过夜,离心,所得固体在室温下真空干燥过夜,得到化合物1的二磷酸盐无定型,其XRD如图23所示。Take about 50 mg of compound 1, add 1.0 ml of chloroform, add 0.2 ml of ethanol solution mixed with 13 mg of phosphoric acid, and precipitate solid. Stir overnight at room temperature, centrifuge, and dry the obtained solid in vacuum at room temperature overnight to obtain the amorphous diphosphate of compound 1, whose XRD is shown in Figure 23.
实施例11:化合物1的硫酸盐无定型的制备Example 11: Preparation of the Sulfate Amorphous Form of Compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.1ml混有5.5mg硫酸的甲醇溶液。在室温下搅拌过夜,加入2.0mL甲基叔丁基醚,搅拌6h,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的硫酸盐无定型。其XRD如图24所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, add 0.1 ml of methanol solution mixed with 5.5 mg of sulfuric acid. Stir overnight at room temperature, add 2.0 ml of methyl tert-butyl ether, stir for 6 hours, precipitate solid, centrifuge, and vacuum dry the obtained solid at room temperature overnight to obtain the amorphous sulfate of compound 1. Its XRD is shown in Figure 24.
实施例12:化合物1的二硫酸盐无定型的制备Example 12: Preparation of the disulfate amorphous form of compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.2ml混有11mg硫酸的甲醇溶液。在室温下搅拌过夜,加入2.0mL甲基叔丁基醚,搅拌6h,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的二硫酸盐无定型,其XRD如图25所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, add 0.2 ml of methanol solution mixed with 11 mg of sulfuric acid, stir at room temperature overnight, add 2.0 ml of methyl tert-butyl ether, stir for 6 hours, precipitate solid, centrifuge, and dry the obtained solid under vacuum at room temperature overnight to obtain the amorphous disulfate of compound 1, whose XRD is shown in Figure 25.
实施例13:化合物1的三硫酸盐无定型的制备Example 13: Preparation of the trisulfate amorphous form of compound 1
取化合物1约50mg,加入1.0ml四氢呋喃,加入0.3ml混有16.5mg硫酸的四氢呋喃溶液。在室温下搅拌过夜,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的三硫酸盐无定型,其XRD如图26所示。Take about 50 mg of compound 1, add 1.0 ml of tetrahydrofuran, add 0.3 ml of tetrahydrofuran solution mixed with 16.5 mg of sulfuric acid, stir at room temperature overnight, precipitate solid, centrifuge, and dry the obtained solid in vacuum at room temperature overnight to obtain the trisulfate amorphous form of compound 1, whose XRD is shown in Figure 26.
实施例14:化合物1的二对甲苯磺酸盐无定型的制备Example 14: Preparation of amorphous di-p-toluenesulfonate salt of compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.2ml混有19mg对甲苯磺酸的甲醇溶液。在室温下搅拌过夜,加入2.0mL甲基叔丁基醚,析出固体,搅拌6h,离心,所得固体在室温下真空干燥过夜,得到化合物1的二对甲苯磺酸盐无定型,其XRD如图27所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, add 0.2 ml of methanol solution mixed with 19 mg of p-toluenesulfonic acid, stir at room temperature overnight, add 2.0 mL of methyl tert-butyl ether, precipitate solid, stir for 6 hours, centrifuge, and dry the obtained solid under vacuum at room temperature overnight to obtain amorphous di-p-toluenesulfonate of compound 1, whose XRD is shown in Figure 27.
实施例15:化合物1的盐酸盐无定型的制备Example 15: Preparation of amorphous hydrochloride of compound 1
取式(I)所示化合物约50mg,加入1.0ml二氯甲烷,加入0.1ml混有6mg盐酸的甲醇溶液。在室温下搅拌过夜,加入2.0mL甲基叔丁基醚,析出固体,搅拌6h,离心,所得固体在室温下真空干燥过夜,得到化合物1的盐酸盐无定型,其XRD如图28所示。Take about 50 mg of the compound represented by formula (I), add 1.0 ml of dichloromethane, add 0.1 ml of methanol solution mixed with 6 mg of hydrochloric acid, stir at room temperature overnight, add 2.0 ml of methyl tert-butyl ether, precipitate solid, stir for 6 hours, centrifuge, and dry the obtained solid under vacuum at room temperature overnight to obtain the amorphous hydrochloride of compound 1, whose XRD is shown in Figure 28.
实施例16:化合物1的二盐酸盐无定型的制备Example 16: Preparation of amorphous dihydrochloride salt of compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.2ml混有11mg盐酸的甲醇溶液。在室温下搅拌过夜,加入2.0mL甲基叔丁基醚,搅拌6h,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的二盐酸盐无定型,其XRD如图29所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, add 0.2 ml of methanol solution mixed with 11 mg of hydrochloric acid, stir at room temperature overnight, add 2.0 ml of methyl tert-butyl ether, stir for 6 hours, precipitate solid, centrifuge, and dry the obtained solid under vacuum at room temperature overnight to obtain the amorphous dihydrochloride of compound 1, whose XRD is shown in Figure 29.
实施例17:化合物1的三盐酸盐无定型的制备 Example 17: Preparation of amorphous trihydrochloride salt of compound 1
取化合物1约50mg,加入1.0ml四氢呋喃,加入0.3ml混有17mg盐酸的四氢呋喃溶液,析出固体。在室温下搅拌过夜,离心,所得固体在室温下真空干燥过夜,得到化合物1的三盐酸盐无定型,其XRD如图30所示。Take about 50 mg of compound 1, add 1.0 ml of tetrahydrofuran, add 0.3 ml of tetrahydrofuran solution mixed with 17 mg of hydrochloric acid, and precipitate solid. Stir overnight at room temperature, centrifuge, and dry the obtained solid in vacuum at room temperature overnight to obtain the trihydrochloride amorphous form of compound 1, whose XRD is shown in Figure 30.
实施例18:化合物1的二-2-萘磺酸盐无定型的制备Example 18: Preparation of amorphous di-2-naphthalenesulfonate of compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有23mg 2-萘磺酸的乙醇溶液。在室温下搅拌过夜,加入2.0mL正庚烷,搅拌6h,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的二-2-萘磺酸盐无定型,其XRD如图31所示。Take about 50 mg of compound 1, add 1.0 ml of chloroform, and add 0.2 ml of ethanol solution mixed with 23 mg of 2-naphthalenesulfonic acid. Stir overnight at room temperature, add 2.0 mL of n-heptane, stir for 6 hours, precipitate solid, centrifuge, and vacuum dry the solid at room temperature overnight to obtain the amorphous di-2-naphthalenesulfonate of compound 1, whose XRD is shown in Figure 31.
实施例19:化合物1的氢溴酸盐无定型的制备Example 19: Preparation of the Amorphous Hydrobromide Salt of Compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.1ml混有11mg氢溴酸的甲醇溶液。在室温下搅拌过夜,加入2.0mL甲基叔丁基醚,搅拌6h,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的氢溴酸盐无定型,其XRD如图32所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, add 0.1 ml of methanol solution mixed with 11 mg of hydrobromic acid, stir at room temperature overnight, add 2.0 ml of methyl tert-butyl ether, stir for 6 hours, precipitate solid, centrifuge, and dry the obtained solid under vacuum at room temperature overnight to obtain the amorphous hydrobromide salt of compound 1, whose XRD is shown in Figure 32.
实施例20:化合物1的二氢溴酸盐无定型的制备Example 20: Preparation of amorphous dihydrobromide salt of compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.2ml混有22mg氢溴酸的甲醇溶液。在室温下搅拌过夜,加入2.0mL甲基叔丁基醚,搅拌6h,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的二氢溴酸盐无定型,其XRD如图33所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, add 0.2 ml of methanol solution mixed with 22 mg of hydrobromic acid, stir at room temperature overnight, add 2.0 ml of methyl tert-butyl ether, stir for 6 hours, precipitate solid, centrifuge, and dry the obtained solid under vacuum at room temperature overnight to obtain the amorphous dihydrobromide salt of compound 1, whose XRD is shown in Figure 33.
实施例21:化合物1的三氢溴酸盐无定型的制备Example 21: Preparation of amorphous trihydrobromide salt of compound 1
取化合物1约50mg,加入1.0ml四氢呋喃,加入0.3ml混有33mg氢溴酸的四氢呋喃溶液,析出固体。在室温下搅拌过夜,离心,所得固体在室温下真空干燥过夜,得到化合物1的三氢溴酸盐无定型,其XRD如图34所示。Take about 50 mg of compound 1, add 1.0 ml of tetrahydrofuran, add 0.3 ml of tetrahydrofuran solution mixed with 33 mg of hydrobromic acid, and precipitate solid. Stir overnight at room temperature, centrifuge, and dry the obtained solid in vacuum at room temperature overnight to obtain the amorphous trihydrobromide salt of compound 1, whose XRD is shown in Figure 34.
实施例22:化合物1的甲磺酸盐无定型的制备Example 22: Preparation of amorphous mesylate salt of compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.1ml混有5mg甲磺酸的甲醇溶液。在室温下搅拌过夜,加入2.0mL甲基叔丁基醚,搅拌6h,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的甲磺酸盐无定型,其XRD如图35所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, add 0.1 ml of methanol solution mixed with 5 mg of methanesulfonic acid, stir at room temperature overnight, add 2.0 ml of methyl tert-butyl ether, stir for 6 hours, precipitate solid, centrifuge, and dry the obtained solid under vacuum at room temperature overnight to obtain amorphous methanesulfonate of compound 1, whose XRD is shown in Figure 35.
实施例23:化合物1的二甲磺酸盐无定型的制备Example 23: Preparation of the amorphous dimesylate salt of compound 1
取化合物1约50mg,加入1.0ml二氯甲烷,加入0.2ml混有11mg甲磺酸的甲醇溶液。在室温下搅拌过夜,加入2.0mL甲基叔丁基醚,搅拌6h,析出固体,离心,所得固体在室温下真空干燥过夜,得到化合物1的二甲磺酸盐无定型,其XRD如图36所示。Take about 50 mg of compound 1, add 1.0 ml of dichloromethane, add 0.2 ml of methanol solution mixed with 11 mg of methanesulfonic acid, stir at room temperature overnight, add 2.0 ml of methyl tert-butyl ether, stir for 6 hours, precipitate solid, centrifuge, and dry the obtained solid under vacuum at room temperature overnight to obtain the dimethanesulfonate amorphous form of compound 1, whose XRD is shown in Figure 36.
通过1H NMR(400MHz,DMSO-d6)的峰位移解析,化合物1的化学位移5.12(dd,J=12.9,5.4Hz,1H)为38号位置的-CH峰,2.70(s,6H)的峰为甲磺酸的峰,其比例为1:6,故可解析得到化合物1与甲磺酸的比例为1:2。Through the peak shift analysis of 1H NMR (400MHz, DMSO-d6), the chemical shift 5.12 (dd, J=12.9, 5.4Hz, 1H) of compound 1 is the -CH peak at position 38, and the peak at 2.70 (s, 6H) is the peak of methanesulfonic acid, and the ratio is 1:6. Therefore, it can be analyzed that the ratio of compound 1 to methanesulfonic acid is 1:2.
实施例24:化合物1的三甲磺酸盐无定型的制备Example 24: Preparation of the amorphous trimesylate salt of compound 1
取化合物1约50mg,加入1.0ml四氢呋喃,加入0.3ml混有16mg甲磺酸的四氢呋喃溶液,析出固体。在室温下搅拌过夜,离心,所得固体在室温下真空干燥过夜,得到化合物1的三甲磺酸盐无定型,其XRD如图37所示。About 50 mg of compound 1 was added to 1.0 ml of tetrahydrofuran, and 0.3 ml of tetrahydrofuran solution mixed with 16 mg of methanesulfonic acid was added to precipitate a solid. The mixture was stirred overnight at room temperature, centrifuged, and the obtained solid was dried in vacuum at room temperature overnight to obtain an amorphous trimesylate salt of compound 1, whose XRD is shown in FIG37 .
实施例25:化合物1的扁桃酸盐无定型的制备Example 25: Preparation of amorphous mandelate salt of compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有9mg扁桃酸的乙醇溶液,将溶液在 4℃下搅拌过夜,未析出固体,加入约4mL正庚烷得到混悬液,在4℃下搅拌过夜。离心,所得固体在室温下真空干燥过夜,得到化合物1的扁桃酸盐无定型,其XRD如图38所示。Take about 50 mg of compound 1, add 1.0 ml of chloroform, add 0.2 ml of ethanol solution mixed with 9 mg of mandelic acid, and stir the solution in After stirring at 4°C overnight, no solid was precipitated, and about 4 mL of n-heptane was added to obtain a suspension, which was stirred at 4°C overnight. After centrifugation, the obtained solid was dried in vacuo at room temperature overnight to obtain the amorphous mandelate salt of compound 1, whose XRD is shown in FIG38 .
实施例26:化合物1的二扁桃酸盐无定型的制备Example 26: Preparation of the Amorphous Dimandelate Salt of Compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有18mg扁桃酸的乙醇溶液,将溶液在4℃下搅拌过夜,未析出固体,加入约4mL正庚烷得到混悬液,在4℃下搅拌过夜。离心,所得固体在室温下真空干燥过夜,得到化合物1的二扁桃酸盐无定型,其XRD如图39所示。About 50 mg of compound 1 was taken, 1.0 ml of chloroform was added, 0.2 ml of ethanol solution mixed with 18 mg of mandelic acid was added, and the solution was stirred at 4°C overnight. No solid was precipitated, and about 4 mL of n-heptane was added to obtain a suspension, which was stirred at 4°C overnight. Centrifugation was performed, and the obtained solid was vacuum dried at room temperature overnight to obtain the amorphous dimandelate salt of compound 1, whose XRD is shown in Figure 39.
实施例27:化合物1的琥珀酸盐无定型的制备Example 27: Preparation of amorphous succinate of compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有7mg琥珀酸的乙醇溶液,将溶液在4℃下搅拌过夜,未析出固体,加入约4mL正庚烷得到混悬液,在4℃下搅拌过夜。离心,所得固体在室温下真空干燥过夜,得到化合物1的琥珀酸盐无定型,其XRD如图40所示。About 50 mg of compound 1 was taken, 1.0 ml of chloroform was added, 0.2 ml of ethanol solution mixed with 7 mg of succinic acid was added, and the solution was stirred at 4°C overnight. No solid was precipitated, and about 4 mL of n-heptane was added to obtain a suspension, which was stirred at 4°C overnight. Centrifugation was performed, and the obtained solid was vacuum dried at room temperature overnight to obtain amorphous succinate of compound 1, whose XRD is shown in Figure 40.
实施例28:化合物1的水杨酸盐无定型的制备Example 28: Preparation of amorphous salicylate of compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有8mg水杨酸的乙醇溶液,将溶液在4℃下搅拌过夜,未析出固体,加入约4mL正庚烷得到混悬液,在4℃下搅拌过夜。离心,所得固体在室温下真空干燥过夜,得到化合物1的琥珀酸盐无定型,其XRD、DSC如图41所示。About 50 mg of compound 1 was taken, 1.0 ml of chloroform was added, 0.2 ml of ethanol solution mixed with 8 mg of salicylic acid was added, and the solution was stirred at 4°C overnight. No solid was precipitated, and about 4 mL of n-heptane was added to obtain a suspension, which was stirred at 4°C overnight. Centrifugation was performed, and the obtained solid was vacuum dried at room temperature overnight to obtain the amorphous succinate of compound 1, whose XRD and DSC are shown in Figure 41.
实施例29:化合物1的1,5-萘二磺酸盐无定型的制备Example 29: Preparation of amorphous 1,5-naphthalene disulfonate salt of compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有22mg 1,5-萘二磺酸的乙醇溶液,将溶液在4℃下搅拌过夜,析出固体。离心,所得固体在室温下真空干燥过夜,得到化合物1的1,5-萘二磺酸盐无定型,其XRD如图42所示。Take about 50 mg of compound 1, add 1.0 ml of chloroform, add 0.2 ml of ethanol solution mixed with 22 mg of 1,5-naphthalene disulfonic acid, stir the solution at 4°C overnight, and precipitate solid. Centrifuge, and vacuum dry the obtained solid at room temperature overnight to obtain the amorphous 1,5-naphthalene disulfonic acid salt of compound 1, whose XRD is shown in Figure 42.
实施例30:化合物1的二-1,5-萘二磺酸盐无定型的制备Example 30: Preparation of amorphous di-1,5-naphthalene disulfonate of compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有44mg 1,5-萘二磺酸的乙醇溶液,将溶液在4℃下搅拌过夜,析出固体。离心,所得固体在室温下真空干燥过夜,得到化合物1的二-1,5-萘二磺酸盐无定型,其XRD如图43所示。Take about 50 mg of compound 1, add 1.0 ml of chloroform, add 0.2 ml of ethanol solution mixed with 44 mg of 1,5-naphthalenedisulfonic acid, stir the solution at 4°C overnight, and precipitate solid. Centrifuge, and vacuum dry the obtained solid at room temperature overnight to obtain the amorphous di-1,5-naphthalenedisulfonate of compound 1, whose XRD is shown in Figure 43.
实施例31:化合物1的半富马酸盐无定型的制备Example 31: Preparation of amorphous hemifumarate of compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有7mg富马酸的乙醇溶液,将溶液在4℃下搅拌过夜,析出固体。离心,所得固体在室温下真空干燥过夜,得到化合物1的半富马酸盐无定型,其XRD如图44所示。Take about 50 mg of compound 1, add 1.0 ml of chloroform, add 0.2 ml of ethanol solution mixed with 7 mg of fumaric acid, stir the solution at 4°C overnight, and precipitate solid. Centrifuge, and vacuum dry the obtained solid at room temperature overnight to obtain amorphous hemifumarate of compound 1, whose XRD is shown in Figure 44.
实施例32:化合物1的二富马酸盐无定型的制备Example 32: Preparation of amorphous difumarate of compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有14mg富马酸的乙醇溶液,将溶液在4℃下搅拌过夜,析出固体,加入约4mL正庚烷得到混悬液,在4℃下搅拌过夜。离心,所得固体在室温下真空干燥过夜,得到化合物1的二富马酸盐无定型,其XRD如图45所示。Take about 50 mg of compound 1, add 1.0 ml of chloroform, add 0.2 ml of ethanol solution mixed with 14 mg of fumaric acid, stir the solution at 4°C overnight, precipitate solid, add about 4 mL of n-heptane to obtain a suspension, stir overnight at 4°C. Centrifuge, and dry the obtained solid in vacuum at room temperature overnight to obtain amorphous difumarate of compound 1, whose XRD is shown in Figure 45.
实施例33:化合物1的三烟酸盐无定型的制备Example 33: Preparation of the Amorphous Trinicotinate of Compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有8mg烟酸的乙醇溶液,将溶液在4℃下搅拌过夜,未析出固体,加入约4mL正庚烷得到混悬液,在4℃下搅拌过夜。离心,所得固体在室温下真空干燥过夜,得到化合物1的三烟酸酸盐无定型,其XRD如图46所示。About 50 mg of compound 1 was taken, 1.0 ml of chloroform was added, 0.2 ml of ethanol solution mixed with 8 mg of nicotinic acid was added, and the solution was stirred at 4°C overnight. No solid was precipitated, and about 4 mL of n-heptane was added to obtain a suspension, which was stirred at 4°C overnight. Centrifugation was performed, and the obtained solid was vacuum dried at room temperature overnight to obtain an amorphous trinicotinate of compound 1, whose XRD is shown in Figure 46.
实施例34:化合物1的马尿酸盐无定型的制备Example 34: Preparation of the Amorphous Hippurate Salt of Compound 1
取化合物1约50mg,加入1.0ml氯仿,加入0.2ml混有11mg马尿酸的乙醇溶液,将溶液 在4℃下搅拌过夜,未析出固体,加入约4mL正庚烷得到混悬液,在4℃下搅拌过夜。离心,所得固体在室温下真空干燥过夜,得到化合物1的马尿酸盐无定型,其XRD如图47所示。Take about 50 mg of compound 1, add 1.0 ml of chloroform, add 0.2 ml of ethanol solution mixed with 11 mg of hippuric acid, and mix the solution. After stirring at 4°C overnight, no solid was precipitated, and about 4 mL of n-heptane was added to obtain a suspension, which was stirred at 4°C overnight. After centrifugation, the obtained solid was dried under vacuum at room temperature overnight to obtain the hippurate amorphous form of compound 1, whose XRD is shown in FIG47 .
实施例35:化合物1的晶型1的制备Example 35: Preparation of Form 1 of Compound 1
取化合物1约50mg,加入1.0ml四氢呋喃,将溶液在室温下搅拌过夜,离心,所得固体在室温下真空干燥过夜,得到化合物1的晶型1,其XRD、TGA、DSC如图48-50所示。About 50 mg of compound 1 was taken, 1.0 ml of tetrahydrofuran was added, the solution was stirred at room temperature overnight, centrifuged, and the obtained solid was dried under vacuum at room temperature overnight to obtain Form 1 of compound 1, whose XRD, TGA and DSC are shown in Figures 48-50.
X-射线粉末衍射仪(XRD)/DSC/TGA/DVS/IC测试X-ray powder diffractometer (XRD)/DSC/TGA/DVS/IC test
XRD/DSC/TGA/DVS测试参数详见表1.XRD/DSC/TGA/DVS test parameters are shown in Table 1.
表1 XRD/DSC/TGA/DVS测试仪器和参数


Table 1 XRD/DSC/TGA/DVS test instruments and parameters


离子色谱(IC)
Ion Chromatography (IC)
表2:化合物1的马来酸盐晶型1的XRD峰列表

Table 2: XRD peak list of maleate salt form 1 of compound 1

表3:化合物1的二马来酸盐晶型1的XRD峰列表

Table 3: XRD peak list of dimaleate salt form 1 of compound 1

表4:化合物1的2-萘磺酸盐晶型1的XRD峰列表

Table 4: XRD peak list of 2-naphthalenesulfonate salt form 1 of compound 1

表5:化合物1的草酸盐晶型1的XRD峰列表
Table 5: XRD peak list of oxalate salt form 1 of compound 1
表6:化合物1的晶型1的XRD峰列表


Table 6: XRD peak list of Form 1 of Compound 1


表7.化合物1的可药用盐无定型水中溶解度(mg/mL)
Table 7. Solubility of amorphous form of pharmaceutically acceptable salt of compound 1 in water (mg/mL)
表8.化合物1的可药用盐的晶型引湿性
Table 8. Crystalline Hygroscopicity of Pharmaceutically Acceptable Salts of Compound 1
生物活性测试Biological activity test
测试例1:NCI-H1975(EGFR-L858R-T790M)和A431(EGFR-WT)细胞的增殖抑制活性Test Example 1: Proliferation Inhibitory Activity of NCI-H1975 (EGFR-L858R-T790M) and A431 (EGFR-WT) Cells
NCI-H1975(EGFR-L858R-T790M)和A431(EGFR-WT)细胞购自于ATCC,培养基分别为RPMI1640+10%FBS和DMEM+10%FBS,于37℃,5%CO2孵箱中培养。第一天,收集处于指数生长期的NCI-H1975(EGFR-L858R-T790M)和A431(EGFR-WT)细胞,用自动细胞分析仪(countstar)进行活细胞计数。用培养基将细胞悬液调整后铺板96孔细胞培养板,NCI-H1975(EGFR-L858R-T790M)细胞每孔1000个,A431细胞每孔3000个。第二天,吸去培养基,每孔加入90μL新鲜培养基和10μL不同浓度化合物,每孔DMSO终浓度为0.1%。于37℃,5%CO2孵箱中培养72小时。药物处理72小时后,每孔加入50μL预先融化并平衡到室温的CTG溶液(promega,G7572),用微孔板震荡器混匀2min,于室温放置10min后用酶标仪(PHERAstar FSX)测定荧光信号值。NCI-H1975 (EGFR-L858R-T790M) and A431 (EGFR-WT) cells were purchased from ATCC, and the culture medium was RPMI1640 + 10% FBS and DMEM + 10% FBS, respectively, and cultured in a 37 ° C, 5% CO 2 incubator. On the first day, NCI-H1975 (EGFR-L858R-T790M) and A431 (EGFR-WT) cells in the exponential growth phase were collected, and live cells were counted using an automatic cell analyzer (countstar). The cell suspension was adjusted with culture medium and plated on a 96-well cell culture plate, with 1000 NCI-H1975 (EGFR-L858R-T790M) cells per well and 3000 A431 cells per well. On the second day, the culture medium was aspirated, and 90 μL of fresh culture medium and 10 μL of different concentrations of compounds were added to each well, with a final DMSO concentration of 0.1% per well. The cells were cultured in an incubator at 37°C and 5% CO 2 for 72 hours. After 72 hours of drug treatment, 50 μL of CTG solution (promega, G7572) pre-melted and equilibrated to room temperature was added to each well, mixed with a microplate shaker for 2 minutes, and placed at room temperature for 10 minutes before measuring the fluorescence signal value with a microplate reader (PHERAstar FSX).
细胞存活率用公式Vsample/Vvehicle controlx100%计算。其中Vsample为药物处理组的读数,Vvehicle control 为溶剂对照组的平均值。应用origin9.2软件,使用非线性回归模型绘制S型剂量-存活率曲线并计算IC50值。The cell viability was calculated using the formula V sample /V vehicle control x 100%. V sample is the reading of the drug treatment group, V vehicle control is The values are the average values of the solvent control group. Using origin9.2 software, nonlinear regression model was used to draw the S-shaped dose-survival rate curve and calculate the IC 50 value.
表9对NCI-H1975(EGFR-L858R-T790M)与A431(EGFR-WT)细胞的增殖抑制活性结果
Table 9 Results of proliferation inhibition activity on NCI-H1975 (EGFR-L858R-T790M) and A431 (EGFR-WT) cells
结论:化合物对NCI-H1975(EGFR-L858R-T790M)细胞具有良好的增殖抑制活性;对A431(EGFR-WT)细胞增殖抑制活性差,具有良好的选择性。Conclusion: The compound has good proliferation inhibitory activity against NCI-H1975 (EGFR-L858R-T790M) cells, but poor proliferation inhibitory activity against A431 (EGFR-WT) cells, and has good selectivity.
测试例2:对细胞NCI-H1975 EGFR-L858R-T790M-C797S的增殖抑制活性Test Example 2: Proliferation Inhibitory Activity on Cells NCI-H1975 EGFR-L858R-T790M-C797S
细胞NCI-H1975 EGFR-L858R-T790M-C797S培养于37℃,5%CO2孵箱中,培养基为RPMI1640+10%FBS+100μg/mL潮霉素。收集处于指数生长期的细胞,用不含潮霉素的培养基将细胞悬液调整到适当浓度后铺板96孔板,铺板密度为1500个/孔,体积90μL。加入10μL不同浓度的化合物,并设置细胞加DMSO的溶媒对照组,DMSO的浓度均为0.1%。细胞培养板置于37℃,5%CO2孵箱中培养72小时。培养结束后,按照CellTiter-Glo试剂盒(Promega,G7572)操作说明,每孔加入50μL预先融化并平衡到室温的CTG溶液,用微孔板震荡器混匀2min,于室温放置10min后用酶标仪(Envision2104)测定荧光信号值。细胞存活率(Surviving cells%)数据采用式(2)处理,并使用GraphPad Prism 5.0软件,使用非线性回归模型绘制S型剂量-存活率曲线并计算IC50值。其中Vsample为药物处理组的读数,Vvehicle control为对照组的读数。Cells NCI-H1975 EGFR-L858R-T790M-C797S were cultured in a 37°C, 5% CO 2 incubator in RPMI1640+10% FBS+100μg/mL hygromycin. Cells in the exponential growth phase were collected, and the cell suspension was adjusted to an appropriate concentration with a medium without hygromycin and plated on a 96-well plate with a density of 1500 cells/well and a volume of 90μL. 10μL of compounds of different concentrations were added, and a solvent control group of cells plus DMSO was set up, and the concentration of DMSO was 0.1%. The cell culture plate was placed in a 37°C, 5% CO 2 incubator for 72 hours. After the culture, according to the instructions of the CellTiter-Glo kit (Promega, G7572), 50 μL of CTG solution pre-melted and equilibrated to room temperature was added to each well, mixed with a microplate shaker for 2 minutes, and placed at room temperature for 10 minutes before measuring the fluorescence signal value with an ELISA reader (Envision2104). The cell survival rate (Surviving cells%) data were processed using formula (2), and GraphPad Prism 5.0 software was used to draw an S-shaped dose-survival rate curve using a nonlinear regression model and calculate the IC 50 value. Where V sample is the reading of the drug treatment group, and V vehicle control is the reading of the control group.
Surviving cells%=Vsample/Vvehicle controlx100%  (式2)Surviving cells%=V sample /V vehicle control x100% (Formula 2)
表10对细胞NCI-H1975 EGFR-L858R-T790M-C797S的增殖抑制活性
Table 10 Proliferation inhibitory activity on cells NCI-H1975 EGFR-L858R-T790M-C797S
结论:化合物对细胞NCI-H1975 EGFR-L858R-T790M-C797S具有良好的增殖抑制活性。Conclusion: The compound has good proliferation inhibitory activity on NCI-H1975 EGFR-L858R-T790M-C797S cells.
测试例3:大鼠药代动力学测试Test Example 3: Pharmacokinetic Test in Rats
1.1试验动物:雄性SD大鼠,220g左右,6~8周龄,3只/化合物。购于成都达硕实验动物有限公司。1.1 Experimental animals: Male SD rats, about 220 g, 6-8 weeks old, 3 rats/compound, purchased from Chengdu Dashuo Experimental Animal Co., Ltd.
1.2试验设计:试验当天,SD大鼠按体重随机分组。给药前1天禁食不禁水12~14h,给药后4h给食。1.2 Experimental design: On the day of the experiment, SD rats were randomly divided into groups according to body weight. They were fasted but not watered for 12-14 hours one day before administration and fed 4 hours after administration.
给药信息
Dosing Information
注:灌胃给药溶媒:0.5%MC(MC:甲基纤维素)Note: Intragastric administration solvent: 0.5% MC (MC: methylcellulose)
于给药前及给药后异氟烷麻醉经眼眶取血0.15ml,置于EDTAK2离心管中,5000rpm,4℃离心10min,收集血浆。静脉组和灌胃组采血时间点均为:0,15,30min,1,2,4,6,8,24h。分析检测前,所有样品存于-80℃,用LC-MS/MS对样品进行定量分析。Before and after drug administration, 0.15 ml of blood was collected from the eye socket under isoflurane anesthesia, placed in an EDTAK2 centrifuge tube, and centrifuged at 5000 rpm and 4°C for 10 min to collect plasma. The blood collection time points for the intravenous group and the gavage group were: 0, 15, 30 min, 1, 2, 4, 6, 8, 24 h. Before analysis and testing, all samples were stored at -80°C and quantitatively analyzed by LC-MS/MS.
表11大鼠药代动力学参数
Table 11 Pharmacokinetic parameters in rats
由表11可知,化合物1的不同盐具有良好的口服性能。It can be seen from Table 11 that different salts of Compound 1 have good oral properties.
测试例4:比格犬药代动力学测试Test Example 4: Beagle dog pharmacokinetic test
试验动物:雄性比格犬,8~11kg左右,3只/化合物,购于北京玛斯生物技术有限公司。Experimental animals: Male beagle dogs, about 8-11 kg, 3 per compound, purchased from Beijing Mas Biotechnology Co., Ltd.
试验方法:试验当天,比格犬按体重随机分组。给药前1天禁食不禁水12~14h,给药后4h给食。
Test method: On the day of the test, beagle dogs were randomly divided into groups according to body weight. They were fasted but not watered for 12-14 hours one day before administration and were fed 4 hours after administration.
于给药前及给药后通过颈静脉或四肢静脉取血1ml,置于EDTAK2离心管中。5000rpm,4℃离心10min,收集血浆。灌胃组采血时间点均为:0,15,30min,1,2,4,6,8,10,12,24,48h。分析检测前,所有样品存于-80℃,用LC-MS/MS对样品进行定量分析Before and after administration, 1 ml of blood was collected from the jugular vein or limb vein and placed in an EDTAK2 centrifuge tube. The blood was centrifuged at 5000 rpm and 4°C for 10 min to collect plasma. The blood collection time points for the gavage group were: 0, 15, 30 min, 1, 2, 4, 6, 8, 10, 12, 24, 48 h. Before analysis and testing, all samples were stored at -80°C and quantitatively analyzed by LC-MS/MS.
表12犬药代动力学参数
Table 12 Pharmacokinetic parameters in dogs
由表12可知,化合物1的不同盐具有良好的口服性能。 It can be seen from Table 12 that different salts of Compound 1 have good oral properties.

Claims (18)

  1. 式(I)所示化合物或其立体异构体的可药用盐及其溶剂化物,
    A compound represented by formula (I) or a pharmaceutically acceptable salt of its stereoisomer and a solvate thereof,
    所述可药用盐选自马来酸盐、2-萘磺酸、1,5-萘二磺酸盐、富马酸盐、氢卤酸盐(优选为氢溴酸盐和盐酸盐)、硫酸盐、磷酸盐、L-酒石酸盐、柠檬酸盐、L-苹果酸盐、马尿酸盐、D-葡萄糖醛酸盐、乙醇酸盐、粘酸盐、琥珀酸盐、乳酸盐、乳清酸盐、帕莫酸盐、甘氨酸盐、丙氨酸盐、精氨酸盐、肉桂酸盐、苯甲酸盐、苯磺酸盐、对甲苯磺酸盐、乙酸盐、丙酸盐、戊酸盐、三苯基乙酸盐、L-脯氨酸盐、阿魏酸盐、2-羟基乙磺酸盐、扁桃酸盐、硝酸盐、甲磺酸盐、丙二酸盐、龙胆酸盐、水杨酸盐、草酸盐或戊二酸盐;优选自苯磺酸盐、L-苹果酸盐、磷酸盐、硫酸盐、对甲苯磺酸盐、盐酸盐、马来酸盐、2-萘磺酸盐、氢溴酸盐、甲磺酸盐、柠檬酸盐、扁桃酸盐、乳糖酸盐、琥珀酸盐、水杨酸盐、1,5-萘二磺酸盐、富马酸盐、烟酸盐、马尿酸盐和草酸盐;更优选自甲磺酸盐。The pharmaceutically acceptable salt is selected from maleate, 2-naphthalenesulfonic acid, 1,5-naphthalenedisulfonate, fumarate, hydrohalide (preferably hydrobromide and hydrochloride), sulfate, phosphate, L-tartrate, citrate, L-malate, hippurate, D-glucuronate, glycolate, mucate, succinate, lactate, orotate, pamoate, glycinate, alanine, arginine, cinnamate, benzoate, benzenesulfonate, p-toluenesulfonate, acetate, propionate, valerate, triphenylacetate, L-proline salt, ferulate salt, 2-hydroxyethanesulfonate salt, mandelate salt, nitrate salt, methanesulfonate salt, malonate salt, gentisate salt, salicylate salt, oxalate salt or glutarate salt; preferably selected from benzenesulfonate, L-malate, phosphate, sulfate, p-toluenesulfonate, hydrochloride, maleate, 2-naphthalenesulfonate, hydrobromide, methanesulfonate, citrate, mandelate salt, lactobionate, succinate, salicylate, 1,5-naphthalenedisulfonate, fumarate, nicotinate, hippurate and oxalate salt; more preferably selected from methanesulfonate salt.
  2. 根据权利要求1所述的可药用盐及其溶剂化物,式(I)所示化合物:可药用盐的摩尔比为1:0.5~1:3.5;The pharmaceutically acceptable salt and solvate thereof according to claim 1, wherein the molar ratio of the compound represented by formula (I): the pharmaceutically acceptable salt is 1:0.5 to 1:3.5;
    优选地,式(I)所示化合物:可药用盐的摩尔比为1:1、1:2、1:3。Preferably, the molar ratio of the compound represented by formula (I): the pharmaceutically acceptable salt is 1:1, 1:2, or 1:3.
  3. 一种式(I)所示化合物的马来酸盐晶型1,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:14.83°±0.2°、16.53°±0.2°、20.34°±0.2°、22.87°±0.2°、23.92°±0.2°;优选地,进一步在以下2θ位置具有特征衍射峰:4.77°±0.2°、6.75°±0.2°、8.80°±0.2°;更优选地,更进一步在以下2θ位置具有特征衍射峰:10.97°±0.2°、16.97°±0.2°、18.68°±0.2°、21.08°±0.2°、24.61°±0.2°。A maleate crystalline form 1 of a compound represented by formula (I), using Cu-Kα radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks at the following 2θ positions: 14.83°±0.2°, 16.53°±0.2°, 20.34°±0.2°, 22.87°±0.2°, 23.92°±0.2°; preferably, further has characteristic diffraction peaks at the following 2θ positions: 4.77°±0.2°, 6.75°±0.2°, 8.80°±0.2°; more preferably, further has characteristic diffraction peaks at the following 2θ positions: 10.97°±0.2°, 16.97°±0.2°, 18.68°±0.2°, 21.08°±0.2°, 24.61°±0.2°.
  4. 根据权利要求3所述的晶型1,使用Cu-Kα辐射,其X-射线粉末衍射图如图1所示。According to the crystalline form 1 of claim 3, using Cu-Kα radiation, its X-ray powder diffraction pattern is shown in Figure 1.
  5. 一种式(I)所示化合物的二马来酸盐晶型1,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:4.18°±0.2°、8.24°±0.2°、18.40°±0.2°、20.48°±0.2°、21.96°±0.2°;优选地,进一步在以下2θ位置具有特征衍射峰:18.80°±0.2°、23.66°±0.2°、24.34°±0.2°;更优选地,更进一步在以下2θ位置具有特征衍射峰:12.30±0.2°、13.21°±0.2°、16.34°±0.2°、16.57°±0.2°、20.00°±0.2°、20.81°±0.2°、25.73°±0.2°、27.98°±0.2°。A dimaleate crystalline form 1 of a compound represented by formula (I), using Cu-Kα radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks at the following 2θ positions: 4.18°±0.2°, 8.24°±0.2°, 18.40°±0.2°, 20.48°±0.2°, 21.96°±0.2°; preferably, further has characteristic diffraction peaks at the following 2θ positions: 18.80°±0. 2°, 23.66°±0.2°, 24.34°±0.2°; more preferably, it further has characteristic diffraction peaks at the following 2θ positions: 12.30±0.2°, 13.21°±0.2°, 16.34°±0.2°, 16.57°±0.2°, 20.00°±0.2°, 20.81°±0.2°, 25.73°±0.2°, 27.98°±0.2°.
  6. 根据权利要求5所述的晶型1,使用Cu-Kα辐射,其X-射线粉末衍射图如图6所示。According to the crystalline form 1 of claim 5, using Cu-Kα radiation, its X-ray powder diffraction pattern is shown in Figure 6.
  7. 一种式(I)所示的化合物的2-萘磺酸盐晶型1,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:5.82°±0.2°、17.52°±0.2°、20.13°±0.2°、21.16°±0.2°、26.80°±0.2°;优选地,进一步在以下2θ位置具有特征衍射峰:12.58°±0.2°、14.92°±0.2°、22.95°±0.2°;更优选地,更进一步在以下2θ位置具有特征衍射峰:7.10°±0.2°、8.78°±0.2°、11.59°±0.2°、17.03°±0.2°、18.80°±0.2°、21.72°±0.2°、22.22°±0.2°。A 2-naphthalenesulfonate salt form 1 of a compound represented by formula (I), using Cu-Kα radiation, has an X-ray powder diffraction pattern having characteristic diffraction peaks at the following 2θ positions: 5.82°±0.2°, 17.52°±0.2°, 20.13°±0.2°, 21.16°±0.2°, 26.80°±0.2°; preferably, further having characteristic diffraction peaks at the following 2θ positions: 1 2.58°±0.2°, 14.92°±0.2°, 22.95°±0.2°; more preferably, further having characteristic diffraction peaks at the following 2θ positions: 7.10°±0.2°, 8.78°±0.2°, 11.59°±0.2°, 17.03°±0.2°, 18.80°±0.2°, 21.72°±0.2°, 22.22°±0.2°.
  8. 根据权利要求7所述的晶型1,使用Cu-Kα辐射,其X-射线粉末衍射图如图11所示。 According to the crystalline form 1 of claim 7, using Cu-Kα radiation, its X-ray powder diffraction pattern is shown in Figure 11.
  9. 一种式(I)所示化合物的草酸盐晶型1,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:19.69°±0.2°、20.07°±0.2°、23.75°±0.2°、24.45°±0.2°、26.82°±0.2°;更优选地,进一步在以下2θ位置具有特征衍射峰:13.79°±0.2°、17.86°±0.2°、24.82°±0.2°、27.07°±0.2°;更进一步在以下2θ位置具有特征衍射峰:6.91°±0.2°、7.63°±0.2°、13.52°±0.2°、18.87°±0.2°、20.71°±0.2°、29.44°±0.2°、31.28°±0.2°。An oxalate crystalline form 1 of the compound represented by formula (I), using Cu-Kα radiation, has an X-ray powder diffraction pattern with characteristic diffraction peaks at the following 2θ positions: 19.69°±0.2°, 20.07°±0.2°, 23.75°±0.2°, 24.45°±0.2°, 26.82°±0.2°; more preferably, further has characteristic diffraction peaks at the following 2θ positions: 13.79 °±0.2°, 17.86°±0.2°, 24.82°±0.2°, 27.07°±0.2°; further, characteristic diffraction peaks are present at the following 2θ positions: 6.91°±0.2°, 7.63°±0.2°, 13.52°±0.2°, 18.87°±0.2°, 20.71°±0.2°, 29.44°±0.2°, 31.28°±0.2°.
  10. 根据权利要求9所述的晶型1,使用Cu-Kα辐射,其X-射线粉末衍射图如图16所示。According to the crystalline form 1 of claim 9, using Cu-Kα radiation, its X-ray powder diffraction pattern is shown in Figure 16.
  11. 根据权利要求1所述的式(I)所示的化合物的可药用盐,所述的可药用盐为无定型固体形式。The pharmaceutically acceptable salt of the compound represented by formula (I) according to claim 1, wherein the pharmaceutically acceptable salt is in the form of an amorphous solid.
  12. 一种无定型的式(I)所示的化合物的二甲磺酸盐。An amorphous dimethanesulfonate of the compound represented by formula (I).
  13. 一种式(I)所示化合物的晶型1,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:5.03°±0.2°、15.35°±0.2°、19.43°±0.2°、19.88°±0.2°;优选地,进一步在以下2θ位置具有特征衍射峰:8.03°±0.2°、13.25°±0.2°、14.57°±0.2°、14.88°±0.2°、23.83±0.2°、24.71°±0.2°、26.44°±0.2°、29.93°±0.2°;更优选地,更进一步在以下2θ位置具有特征衍射峰:8.99°±0.2°、20.17±0.2°、22.32±0.2°、22.55±0.2°、26.17±0.2°、29.49±0.2°。A crystalline form 1 of a compound represented by formula (I), using Cu-Kα radiation, has an X-ray powder diffraction pattern having characteristic diffraction peaks at the following 2θ positions: 5.03°±0.2°, 15.35°±0.2°, 19.43°±0.2°, 19.88°±0.2°; preferably, further having characteristic diffraction peaks at the following 2θ positions: 8.03°±0.2°, 13.25°±0.2°, 14.57°±0 .2°, 14.88°±0.2°, 23.83±0.2°, 24.71°±0.2°, 26.44°±0.2°, 29.93°±0.2°; more preferably, it further has characteristic diffraction peaks at the following 2θ positions: 8.99°±0.2°, 20.17±0.2°, 22.32±0.2°, 22.55±0.2°, 26.17±0.2°, 29.49±0.2°.
  14. 根据权利要求13所述的晶型1,使用Cu-Kα辐射,其X-射线粉末衍射图如图48所示。According to the crystalline form 1 of claim 13, using Cu-Kα radiation, its X-ray powder diffraction pattern is shown in Figure 48.
  15. 一种式(I)所示化合物的可药用盐的制备方法,其中,所述方法包括:以式(I)所示化合物和酸成盐的步骤。A method for preparing a pharmaceutically acceptable salt of a compound represented by formula (I), wherein the method comprises: a step of forming a salt with the compound represented by formula (I) and an acid.
  16. 根据权利要求15所述的制备方法,其中,所用溶剂选自C1-6卤代烷烃类溶剂、C2-6酯类溶剂、C2-6醚类溶剂、C1-6醇类溶剂或水中的一种或多种,优选地所用溶剂选自二氯甲烷、1,2-二氯乙烷、乙酸乙酯、甲醇、乙醇、异丙醇、丙醇、乙醚、四氢呋喃和水中的一种或多种。The preparation method according to claim 15, wherein the solvent used is selected from one or more of C1-6 halogenated alkane solvents, C2-6 ester solvents, C2-6 ether solvents, C1-6 alcohol solvents or water, and preferably the solvent used is selected from one or more of dichloromethane, 1,2-dichloroethane, ethyl acetate, methanol, ethanol, isopropanol, propanol, ether, tetrahydrofuran and water.
  17. 一种药物组合物,其中,所述药物组合物含有治疗有效量的权利要求1~15任意一项所述的晶型或无定型、及药学上可接受的载体或赋形剂。A pharmaceutical composition, wherein the pharmaceutical composition contains a therapeutically effective amount of the crystalline form or amorphous form according to any one of claims 1 to 15, and a pharmaceutically acceptable carrier or excipient.
  18. 权利要求1或2所述的可药用盐及其溶剂化物、权利要求3~14任意一项所述的晶型或无定型或者权利要求17所述的药物组合物在制备用于治疗与抑制或降解EGFR相关疾病的药物中的应用,优选地所述的疾病选自癌症。 Use of the pharmaceutically acceptable salt and solvate thereof according to claim 1 or 2, the crystalline form or amorphous form according to any one of claims 3 to 14, or the pharmaceutical composition according to claim 17 in the preparation of a medicament for treating a disease associated with the inhibition or degradation of EGFR, wherein the disease is preferably selected from cancer.
PCT/CN2023/125385 2022-10-20 2023-10-19 Salt and crystal form of phosphonyl derivative and use thereof in medicine WO2024083183A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211284088.5 2022-10-20
CN202211284088 2022-10-20

Publications (1)

Publication Number Publication Date
WO2024083183A1 true WO2024083183A1 (en) 2024-04-25

Family

ID=90736990

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/125385 WO2024083183A1 (en) 2022-10-20 2023-10-19 Salt and crystal form of phosphonyl derivative and use thereof in medicine

Country Status (1)

Country Link
WO (1) WO2024083183A1 (en)

Similar Documents

Publication Publication Date Title
US9012464B2 (en) Salts and polymorphic forms of Afatinib
CN106822127B (en) Lenalinib maleate and crystalline forms
US8642588B2 (en) Short-acting benzodiazepine salts and their polymorphic forms
JP6880023B2 (en) Pharmaceutically acceptable salts of β-guanidinopropionic acid with improved properties and their use
CN114890943A (en) Solid forms of { [5- (3-chlorophenyl) -3-hydroxypyridine-2-carbonyl ] amino } acetic acid, compositions and uses
ES2764139T3 (en) Cyclin-dependent protein kinase inhibitor hydroxyethyl sulfonate, crystal form thereof and method of preparation thereof
CN112010839B (en) Crystalline forms of a targeted silk/threonine kinase inhibitor
US10208065B2 (en) Crystalline free bases of C-Met inhibitor or crystalline acid salts thereof, and preparation methods and uses thereof
US10323035B2 (en) Co-crystal of a CDK inhibitor and an MEK inhibitor and process of preparation thereof
JP6676626B2 (en) Crystal of mesylate salt of acylthiourea compound and method for producing the same
BR112020015759A2 (en) PHARMACEUTICAL COMPOUND, SALTS OF THE SAME, FORMULATIONS OF THE SAME AND METHODS TO PRODUCE AND USE THE SAME
KR20180111797A (en) Crystalline forms of quinolone analogues and salts thereof
WO2024083183A1 (en) Salt and crystal form of phosphonyl derivative and use thereof in medicine
KR101928987B1 (en) A New and Highly Pure Crystalline monohydrate of mirabegron, Method for Preparing or Use Thereof
US11111234B2 (en) Salt of a quinazoline derivative-like tyrosine kinase inhibitor and crystal form thereof
WO2024041605A1 (en) Pharmaceutically acceptable salt of heteroaryl derivative parp inhibitor and use thereof
US11440908B2 (en) Crystalline forms of dasatinib
CN111344280B (en) (E) -N- (3-cyano-7-ethoxy-4- ((4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide maleate and crystalline forms
EP4169915A1 (en) Crystalline form of compound
WO2024041608A1 (en) Crystal form of heteroaryl derivative parp inhibitor and use thereof
US20230121346A1 (en) The salts of a compound and the crystalline forms thereof
US20210380586A1 (en) 2-indolyl imidazo[4,5-d]phenanthroline polymorphs and compositions regarding the same
WO2015107545A1 (en) Water soluble salts of dasatinib hydrate
RU2807142C1 (en) Maleate salts of (e)-n-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide and their crystalline forms
WO2016050016A1 (en) Substituted heterocyclic compound as kinase inhibitor, and preparation method therefor and uses thereof