WO2013135187A1 - Solid dispersion of improved adsorption performance and preparation thereof - Google Patents

Solid dispersion of improved adsorption performance and preparation thereof Download PDF

Info

Publication number
WO2013135187A1
WO2013135187A1 PCT/CN2013/072643 CN2013072643W WO2013135187A1 WO 2013135187 A1 WO2013135187 A1 WO 2013135187A1 CN 2013072643 W CN2013072643 W CN 2013072643W WO 2013135187 A1 WO2013135187 A1 WO 2013135187A1
Authority
WO
WIPO (PCT)
Prior art keywords
solid dispersion
active ingredient
povidone
compound
formula
Prior art date
Application number
PCT/CN2013/072643
Other languages
French (fr)
Chinese (zh)
Inventor
易必慧
尚晓芳
陆惠萍
盛泽林
Original Assignee
苏州泽璟生物制药有限公司
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 苏州泽璟生物制药有限公司 filed Critical 苏州泽璟生物制药有限公司
Publication of WO2013135187A1 publication Critical patent/WO2013135187A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/146Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention is in the field of medicine, and relates to a solid dispersion and its preparation, and in particular to a solid dispersion for improving absorption properties and preparation thereof. Background technique
  • the compound of formula II is a novel multi-targeted oral drug for the treatment of tumors. Its primary development goal is to treat gastrointestinal stromal tumors and metastatic renal cell carcinoma that are not responsive or intolerant to standard therapies.
  • the compound shown by formula ⁇ is extremely poor in water solubility and is a poorly soluble drug. It has a low solubility and dissolution rate in physiological body fluids, and a low dissolution rate, which ultimately leads to low bioavailability and seriously affects the absorption of active ingredients by the human body. It is not conducive to giving full play to the therapeutic effects of the compound.
  • Solid dispersions can increase the bioavailability of poorly soluble drugs, but existing solid dispersions can only increase the area under the curve of the drug (AUC) by a factor of 2-6.
  • AUC area under the curve of the drug
  • One object of the present invention is to provide a solid dispersion of a compound of the formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, having a significant increase in bioavailability as an active ingredient.
  • Another object of the present invention is to provide a process for the preparation of the solid dispersion of the present invention.
  • a further object of the invention is to provide a pharmaceutical composition of the solid dispersion.
  • a first aspect of the invention provides a solid dispersion comprising:
  • R is CD 3 or CH 3 ;
  • weight ratio of (A) and (B) is 1: 1-1: 40.
  • the hydrophilic polymer carrier is povidone or a povidone-containing polymer carrier.
  • the hydrophilic polymer carrier is povidone K30 (PVP K30) or a povidone K30-containing polymer carrier, wherein the povidone K30 is contained in an amount of 10 to 100% by weight. Based on the total weight of the polymeric carrier.
  • the active ingredient is a compound of formula II, or a pharmaceutically acceptable salt, hydrate or solvate thereof
  • the active ingredient is a p-toluenesulfonate of a compound of formula II.
  • the povidone-containing K30-containing polymeric carrier further comprises other water-soluble carriers selected from the group consisting of polyethylene glycols, povidones, celluloses, surfactants, Or a combination thereof;
  • polyethylene glycol is selected from the group consisting of polyethylene glycol 4000, polyethylene glycol 6000, polyethylene glycol 8000 or a combination thereof;
  • the povidone is selected from the group consisting of povidone K25, povidone oxime 90, povidone S630, or a combination thereof;
  • the cellulose is selected from the group consisting of hypromellose E5, hyprothenol Cellulose E15, hydroxypropyl cellulose L, or a combination thereof;
  • the surfactant class is selected from the group consisting of poloxamer 407, poloxamer 188, or a combination thereof.
  • the povidone-containing K30-containing polymeric carrier comprises a povidone-based carrier selected from the group consisting of povidone K25, povidone 90, povidone S630, or a combination thereof.
  • the weight ratio of the povidone-containing K30-containing polymer carrier to the other water-soluble carrier is 1:4-4:1; preferably 1:2-4:1; more preferably For 1: 1-4: 1.
  • the weight ratio of the (A) and (B) is 1: 1-1: 20.
  • the weight ratio of the (A) and (B) is 1: 2-1: 5.
  • the X-ray powder diffraction pattern of the solid dispersion does not have an X-ray diffraction characteristic peak of the active component of the solid dispersion.
  • the area under the drug-time curve of the solid dispersion is 6-20 times, preferably 7-15 times, the area under the drug-time curve when the active ingredient of the solid dispersion is administered alone;
  • the solid dispersion is cumulatively dissolved in a sodium acetate buffered aqueous solution at 37 ° C, pH 4.5 for 10 minutes. Degree greater than 70%, preferably 75-95%;
  • the cumulative dissolution of the solid dispersion at 37 ° C, pH 4.5 in an aqueous solution of sodium acetate buffer for 120 minutes is 10-40 times, preferably 15-15, of the cumulative dissolution of the active ingredient of the solid dispersion. 30 times.
  • the area under the drug time curve is measured in the following animals: rat, mouse or canine.
  • a second aspect of the present invention provides a method for preparing a solid dispersion according to the first aspect of the present invention, comprising the method of:
  • step (b2) mixing the molten carrier obtained in the step (bl) with the active ingredient and melting the mixture to obtain a molten mixture
  • step (b3) cooling the molten mixture obtained in the step (b2) to obtain the solid dispersion according to the first aspect of the invention
  • the active ingredient is a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof,
  • R is CD 3 or CH 3
  • the hydrophilic polymer carrier is a hydrophilic polymer carrier containing povidone K30.
  • the solvent is selected from the group consisting of methanol, ethanol, isopropanol, acetone, chloroform, tetrahydrofuran, or Combination.
  • step (c2) includes the steps of:
  • the solvent in the mixture obtained in the step (cl) is removed by a rotary evaporator; or
  • the solvent in the mixture obtained in the step (cl) was removed by drying with a spray dryer.
  • the drying process parameters are: inlet air temperature 110 to 120 ° C, fan power 55 % to 65 %, peristaltic pump power 50 % to 55 %.
  • the solid dispersion is further treated with at least one additional processing step of grinding, sieving, rolling, milling, screening, mixing, or a combination thereof.
  • a third aspect of the invention provides a pharmaceutical composition, the composition comprising:
  • the excipient comprises: a filler, a diluent, a sweetener, a flavoring agent, an antioxidant, a coloring agent, a preservative, a lubricant, a binder, a disintegrating agent, or combination.
  • the composition is a capsule, a tablet, a pill, a powder, and a granule.
  • the pharmaceutical composition is for use in a pharmaceutical composition for inhibiting a phosphokinase such as raft chymase.
  • the pharmaceutical composition is for the treatment and prevention of cancer.
  • a process for the preparation of a pharmaceutical composition according to the third aspect of the present invention wherein the solid dispersion according to the first aspect of the present invention is mixed with a pharmaceutically acceptable excipient, thereby producing a Said pharmaceutical composition.
  • a fifth aspect of the invention provides the use of the solid dispersion of the first aspect of the invention for the preparation of a medicament for inhibiting a phosphokinase (e.g., raf ⁇ enzyme); or for the preparation of a medicament for treating a tumor.
  • a phosphokinase e.g., raf ⁇ enzyme
  • a sixth aspect of the invention provides a method of treating (i) the solid dispersion of the first aspect of the invention or (ii) the pharmaceutical composition of the third aspect of the invention for a patient in need of treatment.
  • Figure 1 shows an XRPD pattern of Solid Dispersion 1.
  • Figure 2 shows an XRPD pattern of Solid Dispersion 2.
  • Figure 3 shows an XRPD pattern of Solid Dispersion 3.
  • Figure 4 shows an XRPD pattern of Solid Dispersion 4.
  • Figure 5 shows an XRPD pattern of solid dispersion 5.
  • Figure 6 shows an XRPD pattern of solid dispersion 6.
  • Figure 7 shows an XRPD pattern of solid dispersion 7.
  • Figure 8 shows an XRPD pattern of solid dispersion 8.
  • Figure 9 shows an XRPD pattern of Solid Dispersion 9.
  • Figure 10 shows an XRPD pattern of solid dispersion 10.
  • Figure 1 1 shows an XRPD pattern of the solid dispersion 1 1 .
  • Figure 12 shows the physical mixture of the compound of the formula II, the compound of the formula II and the PVP K30 and the dissolution profile of the solid dispersion 1.
  • Figure 13 shows an XRPD pattern of solid dispersion 12.
  • Figure 14 shows a comparison of XRPD of the solid dispersion 4 after 1, 2, and 3 months accelerated stability test. The results are shown in Figure 14: From top to bottom, the compound of formula II is p-toluenesulfonate, new The prepared solid dispersion 4, the XRPD pattern when the solid dispersion 4 was left for one month, the solid dispersion 4 was left for two months, and the solid dispersion 4 was left for three months.
  • Figure 15 shows a comparison of XRPD of the solid dispersion 5 after 1, 2, and 3 months accelerated stability test. The results are shown in Figure 15: From top to bottom, the compound of formula II is p-toluenesulfonate, new The prepared solid dispersion 5, the XRPD pattern when the solid dispersion 5 was left for one month, the solid dispersion 5 was left for two months, and the solid dispersion 5 was left for three months.
  • Figure 16 shows a comparison of XRPD of the solid dispersion 1 after 1, 2, and 3 months accelerated stability test, and the results are shown in Figure 16: From the top to the bottom, the compound of the formula II, the newly prepared solid dispersion 1, The XRPD pattern of the solid dispersion 1 when it was left for one month, when the solid dispersion 1 was left for two months, and when the solid dispersion 1 was left for three months.
  • Figure 17 shows a comparison of XRPD of the solid dispersion 3 after 1, 2, and 3 months accelerated stability test. The results are shown in Figure 17: From the top to the bottom, the compound of the formula II, the newly prepared solid dispersion 3, The XRPD pattern when the solid dispersion 3 was left for one month, when the solid dispersion 3 was left for two months, and when the solid dispersion 3 was left for three months.
  • Solid dispersion Solid dispersion refers to a dispersion system in which the active ingredient is highly dispersed in a (solid) support to form a solid form. Active ingredient
  • active ingredient refers to a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt thereof, as an active ingredient
  • the term also encompasses various crystalline forms, hydrates or solvates of a compound of formula A or a derivative thereof, preferably:
  • pharmaceutically acceptable salt refers to a salt of the compound of the present invention which is formed with an acid or a base and which is suitable for use as a medicament.
  • Pharmaceutically acceptable salts include inorganic and organic salts.
  • a preferred class of salts are the salts of the compounds of the invention with acids.
  • Suitable acids for forming salts include, but are not limited to: mineral acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, Organic acids such as maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, benzoic acid, benzenesulfonic acid; and acidic amino acids such as aspartic acid and glutamic acid.
  • mineral acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid,
  • Organic acids such as maleic acid, lactic acid, malic acid, tartaric acid, cit
  • the pharmaceutically acceptable salt is p-toluenesulfonate.
  • the carrier used for the solid dispersion is a hydrophilic polymer carrier, preferably povidone or povidone-containing.
  • the povidone K30-containing polymer carrier used in the present invention refers to the weight of povidone K30.
  • hydrophilic polymeric carrier may also contain other water soluble carriers such as, but not limited to, polyethylene glycols, povidones, cellulosics, surfactants, or combinations thereof.
  • the polyethylene glycols are selected from the group consisting of polyethylene glycol 4000 (PEG 4000), polyethylene glycol 6000 (PEG)
  • the povidone is selected from the group consisting of povidone K90 (PVP K90), povidone K25 (PVP K25), povidone S630, or a combination thereof;
  • the cellulose is selected from the group consisting of hypromellose E5 (HPMC E5), hypromellose E15 (HPMC E15), hydroxypropylcellulose L (HPC-L), or a combination thereof;
  • the surfactant class is selected from the group consisting of poloxamer 407 (Poloxamer 407), Poloxamer 188 (Poloxamer 188), or a combination thereof.
  • the solid dispersions provided by the present invention include:
  • weight ratio of (A) and (B) is 1: 1-1: 40.
  • the weight ratio of the (A) and (B) is 1: 20-1: 1, preferably, the weight ratio of the (A) and (B) is 1: 5-1: 2.
  • the solid dispersion of the present invention can be obtained by the following method, however, the conditions of the method, such as the carrier, the solvent, the amounts of the respective components, the preparation temperature, the time required for preparation, and the like are not limited to the following explanations.
  • the solid dispersions of the present invention may also be conveniently prepared by combining various synthetic methods described in the specification or known in the art, such combinations being readily made by those skilled in the art to which the present invention pertains. .
  • the preparation method may be a melting method, a solvent method, a solvent-melting method, a spray drying method, or a surface dispersion method.
  • the preparation method is a melt method or a solvent method. Melting method
  • the (A) active ingredient and (B) the hydrophilic polymer carrier are uniformly mixed, heated to melt in a water bath or an oil bath, and then cooled to a solid, and pulverized to obtain the active substance in a powder form.
  • a solid dispersion of the ingredients are usually used.
  • the (B) hydrophilic polymer carrier may be heated and melted, and then (A) the active ingredient may be added, and then The molten mixture is rapidly cooled to a solid under vigorous stirring, and after pulverization, a solid dispersion of the active ingredient in a powder form is obtained.
  • Solvent method
  • the solvent method is a method in which (A) an active ingredient and (B) a hydrophilic polymer carrier are co-dissolved in a solvent, and after the mixture is homogeneous, the solvent in the solution is removed to obtain an active ingredient and a hydrophilic polymer.
  • the carrier is simultaneously precipitated, and after pulverization, a solid dispersion of the active ingredient in a powder form is obtained.
  • the solvent used in the solvent method includes: methanol, ethanol, isopropanol, acetone, tetrahydrofuran, or a combination thereof.
  • Structure identification methods for solid dispersions include differential scanning calorimetry, X-ray powder diffraction, and the like.
  • the solid dispersion prepared by the active ingredient, the carrier and the method of the present invention were subjected to X-ray powder diffraction (XRPD) detection, and the XRPD spectra of the three were compared. Characteristic peak of active ingredient
  • the X-ray diffraction characteristic peak of the active ingredient disappears substantially or completely.
  • the X-ray powder diffraction pattern of the solid dispersion of the present invention does not have an X-ray diffraction characteristic peak of the active component of the solid dispersion.
  • the typical diffraction peak (characteristic peak) of the active ingredient when measured by XRPD is as follows;
  • a compound of formula I which is at 14.58 ⁇ 0 ⁇ , 18 ⁇ 71 ⁇ 0 ⁇ , 21 ⁇ 08 ⁇ 0 ⁇ 1. , 23 ⁇ 35 ⁇ 0 ⁇ , 29.44 ⁇ 0. Strong diffraction peaks.
  • a compound of formula II which is at 12.49 ⁇ 0.1. , 22 ⁇ 40 ⁇ 0 ⁇ , 22 ⁇ 83 ⁇ 0 ⁇ , 24.69 ⁇ 0.1. 25.11 ⁇ 0.1° has strong diffraction peaks.
  • Some carriers have strong diffraction peaks.
  • PEG4000 and PEG6000 have strong diffraction peaks at 19.19 ⁇ 0. and 23.30 ⁇ 0.1°. ? 01 0 & 111 ⁇ 2 407 has strong diffraction peaks at 19.16 ⁇ 0.1 ° and 23.32 ⁇ 0.1 °.
  • Some carriers do not have strong diffraction peaks.
  • strong diffraction peaks are not detected in PVP K30, HPMC E5, and HPC-L. Characteristic peak of solid dispersion
  • the strong diffraction peak of the active ingredient disappears in the XRPD spectrum of the solid dispersion prepared by the present invention, thereby confirming the successful preparation of the solid dispersion of the present invention, which is not a simple physical mixing of the active ingredient and the carrier. But in an amorphous or molecular form.
  • compound of the invention refers to a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt thereof.
  • solid dispersion of the invention means a solid dispersion of the active ingredient which is a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt thereof.
  • the compound of the formula A (or a derivative thereof, or a salt thereof) has excellent inhibitory activity against phosphokinase (Kinase M column such as raft ⁇ enzyme, the solid dispersion of the present invention and the pharmaceutical composition containing the solid dispersion of the present invention as a main component
  • the compounds are useful for the treatment, prevention, and alleviation of diseases mediated by phosphokinase (Kinase M, such as raft ⁇ enzyme.
  • the compounds of the present invention are useful for treating diseases such as cancer, cardiovascular disease, obesity, diabetes, etc. .
  • compositions of the present invention comprise a solid dispersion of the present invention or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient in a safe and effective amount.
  • safe and effective amount is meant: The amount of solid dispersion is sufficient to significantly improve the condition without causing serious side effects.
  • the pharmaceutical composition contains from 1 to 2000 mg of the solid dispersion/agent of the invention, more preferably from 10 to 200 mg of the solid dispersion/agent of the invention.
  • the "one dose" is a capsule or a tablet.
  • “Pharmaceutically acceptable excipient” means: one or more compatible solid or liquid fillers or gel materials which are suitable for human use and which must be of sufficient purity and of sufficiently low toxicity.
  • “compatibility” it is meant herein that the components of the composition are compatible with the solid dispersions of the present invention and between them without significantly reducing the efficacy of the solid dispersion.
  • Examples of pharmaceutically acceptable excipients are cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), starches, monosaccharides or polysaccharides, gelatin , talc, solid lubricants (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.) , emulsifiers (such as Tween®), wetting agents (such as sodium decyl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • cellulose and its derivatives such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.
  • starches such as stearic acid, magnesium
  • the mode of administration of the solid dispersion or pharmaceutical composition of the present invention is not particularly limited, and representative modes of administration include, but are not limited to, oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical Dosing.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the solid dispersion is admixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or mixed with: (a) a filler or compatibilizer, for example , starch, lactose, sucrose, glucose, microcrystalline cellulose, mannitol and silicic acid; (b) binders, for example, hydroxypropylmethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and (a) humectants, for example, glycerin; (d) disintegrants, for example, agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, croscarmellose Sodium and sodium carbonate; (e) a slow solvent such as paraffin; (f) aspiration Accelerator, for example, a
  • Solid dosage forms such as tablets, troches, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other materials known in the art. They may contain opacifying agents and the release of the solid dispersion in such compositions may be released in a portion of the digestive tract in a delayed manner. Examples of embedding components that can be employed are polymeric and waxy materials. The solid dispersion may also form a microcapsule form with one or more of the above excipients as necessary.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, lozenges or elixirs.
  • the liquid dosage form may comprise an inert diluent conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or a mixture of these substances.
  • an inert diluent conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, di
  • compositions may contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfumes.
  • the suspension may contain a suspending agent, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan ester, microcrystalline cellulose, aluminum methoxide and agar or a mixture of these and the like.
  • a suspending agent for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan ester, microcrystalline cellulose, aluminum methoxide and agar or a mixture of these and the like.
  • compositions for parenteral injection may comprise a physiologically acceptable sterile aqueous or nonaqueous solution, dispersion, suspension or emulsion, and sterile powder for reconstitution into a sterile injectable solution or dispersion.
  • Suitable aqueous and nonaqueous vehicles, diluents, solvents or vehicles include water, ethanol, polyol, and suitable mixtures thereof.
  • Dosage forms for solid dispersions of the invention for topical administration include ointments, powders, patches, propellants and inhalants.
  • the solid dispersions of the present invention are mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
  • the solid dispersion of the present invention can be administered alone or in combination with other pharmaceutically acceptable compounds.
  • a pharmaceutical composition e.g., a human
  • a safe and effective amount of the solid dispersion of the present invention is suitably applied to a mammal (e.g., a human) in need of treatment wherein the dosage is a pharmaceutically effective effective dosage for administration to a 60 kg body weight.
  • the daily dose is usually from 1 to 2000 mg, preferably from 20 to 500 mg.
  • specific doses should also consider factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
  • the main advantages of the invention are:
  • Providing a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate thereof or The solvate is a solid dispersion of the active ingredient which significantly improves the in vitro dissolution of the active ingredient and is highly advantageous for mammals to absorb the active ingredient. Moreover, the solid dispersion has significantly superior in vivo pharmacokinetic parameters (especially the area under the curve of the drug) relative to the active ingredient compound itself, and is very stable.
  • API refers to the active ingredient
  • Dissolution medium 0.1% sodium decyl sulfate pH 4.5 acetic acid-sodium acetate buffer 900 mL/dissolution cup; temperature: 37 ⁇ 0.5 ° C; rotation speed: 100 rpm.
  • the samples were divided into three groups, each of which was configured with 3 portions.
  • the first group of samples is an active ingredient;
  • the second group of samples is a sample obtained by simply mixing the active ingredient with a carrier by a simple physical method such as stirring.
  • the third set of samples was a solid dispersion obtained according to the preparation method in each of the examples. The details are as follows.
  • Example 3 Solid dispersion 3
  • Example 4 Solid dispersion 4
  • the p-toluenesulfonate of the compound of the formula II and the PVP K30 are in a weight ratio of 1:4, and the solvent (methanol) method is the same as in the first embodiment except that the p-toluenesulfonate of the compound of the formula II is used instead. a compound of formula II.
  • the p-toluenesulfonate of the compound of the formula II and the PVP K30 are in a weight ratio of 1:3, and the solvent (methanol) method is the same as in the example 4 except that the p-toluenesulfonate of the compound of the formula II is used.
  • PVP solvent (methanol) method
  • the weight ratio of K30 is 1:3.
  • the preparation method was the same as in Example 1, except that the solvent was absolute ethanol.
  • Example 7 Solid Dispersion 7
  • the compound of formula II has p-toluenesulfonate and PVP K30 in a weight ratio of 1:4, solvent (anhydrous ethanol) method
  • the preparation method is the same as that of Example 1, except that the p-toluenesulfonate of the compound of the formula II is used instead of the compound of the formula II, and the solvent is anhydrous ethanol.
  • the compound of formula II has p-toluenesulfonate and PVP K30 in a weight ratio of 1:4, solvent (isopropyl alcohol) method
  • the preparation method is the same as in Example 1, except that the p-toluenesulfonate of the compound of the formula II is used instead of the compound of the formula II, and the solvent is isopropanol.
  • the compound of formula II has p-toluenesulfonate and PVP K30 in a weight ratio of 1:4, solvent (anhydrous ethanol) method
  • the lysed solution was spray dried with a Swiss-made Buchi Model 209 spray dryer.
  • the process parameters are as follows: inlet air temperature 1 10 ° C ⁇ 120 ° C, fan power 55% ⁇ 65%, peristaltic pump power 50% ⁇ 55%.
  • a white fine powder was obtained.
  • the obtained powder was vacuum dried in a vacuum oven at 50 ° C for 24 hours to obtain a solid dispersion 9 powder.
  • the compound of formula II has p-toluenesulfonate and PVP K30 in a weight ratio of 1:4, solvent (isopropyl alcohol) method
  • the preparation method was the same as in Example 9, except that the solvent was isopropyl alcohol.
  • the compound of formula II has p-toluenesulfonate and PVP K30 in a weight ratio of 1:2, solvent (isopropyl alcohol) method
  • the preparation method is the same as that of Example 9, except that p-toluenesulfonate and PVP of the compound of formula II are used.
  • the weight ratio of K30 is 1:2, and the solvent is isopropyl alcohol.
  • the physical mixture of the compound of the formula II, the compound of the formula II and the PVP K30 (the weight of the compound of the formula II and the PVP K30 are 50 mg and 200 mg, respectively) and the solid dispersion 1 (250 mg) are respectively added, and the paddle speed is 100 rpm, respectively.
  • 5 mL was sampled at 2 min, 5 min, 10 min, 15 min, 20 min, 30 min, 45 min, 60 min, 120 min, filtered through a 0.45 ⁇ microporous membrane filter, and the clarified filtrate was diluted to determine the drug concentration, and the dissolution of the three was calculated.
  • the solid dispersion 1 was selected for in vivo pharmacoassay with the corresponding API (i.e., the compound of formula II).
  • 0.2 ml of venous blood was taken from the posterior venous plexus of rats, placed in heparinized tubes, centrifuged at 3500 rpm for 10 min, and plasma was separated. Sample, stored at -20 °C.
  • Plasma samples were quantified for API content using a validated LC-MS/MS analytical method.
  • the pharmacokinetic parameters will be calculated based on the plasma concentration of each rat at different time points. The parameters are shown in Table 1.
  • the preparation method was the same as in Example 1, except that the compound of the formula I was used instead of the compound of the formula II, and the solvent was anhydrous ethanol.
  • Example 16 Solid Dispersion 14
  • Example 17 Comparative Test of Dissolution of Solid Dispersion 12
  • Example 18 In vivo pharmacokinetic test of solid dispersion 12
  • the solid dispersion 12 was selected for in vivo pharmacoassay with the corresponding API (i.e., the compound of formula I) in the same manner as in Example 13.
  • the solid dispersion 4 was selected for accelerated stability test (40 ° C, 75% H): taken at 1, 2, and 3 months for X-ray powder diffraction.
  • Solid dispersion 5 was selected for accelerated stability test (40 ° C, 75% H): 1, 2, 3 months The X-ray powder diffraction was taken out.
  • the solid dispersion 1 was selected for accelerated stability test (40 ° C, 75% H): 1 , 2, 3 months were taken out for X-ray powder diffraction.
  • Fig. 16 The XRPD results are shown in Fig. 16, which indicates that the solid dispersion 1 is very stable after the 1, 2, and 3 months accelerated stability test (no X-ray diffraction characteristic peak of the API, and the curves are basically the same), and no crystal form is formed. (ie, the original amorphous or molecular state does not change), still a solid dispersion that exists in an amorphous or molecular state.
  • the solid dispersion 3 was selected for accelerated stability test (40 ° C, 75% H): taken at 1, 2, and 3 months for X-ray powder diffraction.
  • API is a compound of formula II
  • the carrier is a mixed carrier of PVP K30 and other water-soluble carriers (see Table 3 for mixed carriers).
  • the preparation method was the same as that in Example 1, except that the mixed carrier was subjected to the conditions shown in Table 3, and the solvent was Anhydrous ethanol.
  • the XRPD test results show that the diffraction peaks (characteristic peaks) of the compound of formula II disappear completely, and exist in an amorphous or molecular form.
  • the solid dispersion comprising the active ingredient and the PVP K30-containing carrier prepared according to the present invention is very stable.
  • the solid dispersion prepared by the invention has significantly improved dissolution and good in vivo pharmacokinetic parameters, especially the area under the drug-time curve of the solid dispersion of the invention is its active ingredient alone.
  • the area under the curve of the drug at the time of administration is 6-20 times, preferably 7-15 times.
  • the solids dispersion of the compound of formula II has a more pronounced improvement in area under the drug time curve than the solid dispersion of the compound of formula I.
  • the solids dispersion of the solid dispersion of the compound of formula II is 13.9 times the area of the active ingredient of the compound of formula II, and the solid dispersion of the solid dispersion of the compound of formula I is 7.3 times the area of the active ingredient of the compound of formula I.

Abstract

A solid dispersion of improved adsorption performance and preparation thereof. Specifically, provided in the present invention is the solid dispersion. The solid dispersion comprises (A) serving as an active ingredient and represented by formula A, a compound or a derivative thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof, and, (B) a hydrophilic macromolecule carrier, where the weight ratio between (A) and (B) is between 1:1 and 1:40. The solid dispersion provided in the present invention significantly increases in vitro dissolution of active ingredients, and greatly increases adsorption of the active ingredients for mammals, thus effectively reducing dosage.

Description

种改善吸收性能的固体分散体及其制备  Solid dispersion for improving absorption performance and preparation thereof
技术领域  Technical field
本发明属于医药领域, 涉及一种固体分散体及其制备, 具体地涉及一种改善 吸收性能的固体分散体及其制备。 背景技术  The present invention is in the field of medicine, and relates to a solid dispersion and its preparation, and in particular to a solid dispersion for improving absorption properties and preparation thereof. Background technique
式 II所示的化合物是一种新型多靶向性的治疗肿瘤的口服药物。 其首要开 发目标为, 用于治疗对标准疗法没有响应或不能耐受之胃肠道基质肿瘤和转移 性肾细胞癌。
Figure imgf000002_0001
The compound of formula II is a novel multi-targeted oral drug for the treatment of tumors. Its primary development goal is to treat gastrointestinal stromal tumors and metastatic renal cell carcinoma that are not responsive or intolerant to standard therapies.
Figure imgf000002_0001
式 Π所示的化合物水溶性极差, 是难溶性药物, 在生理体液中的溶解度和溶 解速度较低, 溶出度也较低, 最终导致生物利用度较低, 严重影响人体对活性成 分的吸收, 不利于充分发挥该化合物的疾病治疗效果。  The compound shown by formula 极 is extremely poor in water solubility and is a poorly soluble drug. It has a low solubility and dissolution rate in physiological body fluids, and a low dissolution rate, which ultimately leads to low bioavailability and seriously affects the absorption of active ingredients by the human body. It is not conducive to giving full play to the therapeutic effects of the compound.
固体分散体可以提高难溶性药物的生物利用度, 但是现有的固体分散体, 最高只能将药物的药时曲线下面积 (AUC)提高 2-6倍。 例如, CN101632630A中, 较普罗布考而言, 普罗布考和 PVP K30的固体分散体, 其犬相对生物利用度也 仅仅提高了 5.8倍。  Solid dispersions can increase the bioavailability of poorly soluble drugs, but existing solid dispersions can only increase the area under the curve of the drug (AUC) by a factor of 2-6. For example, in CN101632630A, the relative bioavailability of probucol and PVP K30 solids is only 5.8 times higher than that of probucol.
因此, 开发一种生物利用度显著提高的式 II化合物的固体分散体, 对解决哺 乳动物对其活性成分吸收差的问题是非常有必要的。 发明内容  Therefore, the development of a solid dispersion of a compound of formula II with significantly improved bioavailability is highly important in addressing the problem of poor absorption of active ingredients by mammals. Summary of the invention
本发明的一个目的是提供一种生物利用度显著提高的以式 A化合物或其衍 生物、 或其药学上可接受的盐、 水合物或溶剂合物为活性成分的固体分散体。  SUMMARY OF THE INVENTION One object of the present invention is to provide a solid dispersion of a compound of the formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, having a significant increase in bioavailability as an active ingredient.
本发明的另一目的是提供本发明固体分散体的制备方法。  Another object of the present invention is to provide a process for the preparation of the solid dispersion of the present invention.
本发明的再一目的是提供所述固体分散体的药物组合物。  A further object of the invention is to provide a pharmaceutical composition of the solid dispersion.
本发明第一方面提供了一种固体分散体, 包括:  A first aspect of the invention provides a solid dispersion comprising:
(A) 作为活性成分的式 A化合物或其衍生物、 或其药学上可接受的盐、 水合物 或溶剂合物,  (A) a compound of the formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, as an active ingredient,
Figure imgf000002_0002
式中, R为 CD3或 CH3 ; 以及
Figure imgf000002_0002
Wherein R is CD 3 or CH 3 ;
(B) 亲水性高分子载体;  (B) a hydrophilic polymer carrier;
其中, (A)和 (B)的重量比为 1 : 1-1: 40。  Wherein the weight ratio of (A) and (B) is 1: 1-1: 40.
在另一优选例中, 所述的亲水性高分子载体是聚维酮或含聚维酮的高分子载 体。  In another preferred embodiment, the hydrophilic polymer carrier is povidone or a povidone-containing polymer carrier.
在另一优选例中, 所述的亲水性高分子载体是聚维酮 K30(PVP K30)或含聚维 酮 K30的高分子载体, 其中, 聚维酮 K30的含量为 10-100wt%, 按高分子载体的总 重量计。  In another preferred embodiment, the hydrophilic polymer carrier is povidone K30 (PVP K30) or a povidone K30-containing polymer carrier, wherein the povidone K30 is contained in an amount of 10 to 100% by weight. Based on the total weight of the polymeric carrier.
在另一优选例中,所述活性成分为式 II所示的化合物,或其药学上可接受的盐、 水合物或溶剂合
Figure imgf000003_0001
In another preferred embodiment, the active ingredient is a compound of formula II, or a pharmaceutically acceptable salt, hydrate or solvate thereof
Figure imgf000003_0001
在另一优选例中, 所述的活性成分是式 II化合物的对甲苯磺酸盐。  In another preferred embodiment, the active ingredient is a p-toluenesulfonate of a compound of formula II.
在另一优选例中, 所述含聚维酮 K30的高分子载体还包含选自下组的其他水溶 性载体: 聚乙二醇类、 聚维酮类、 纤维素类、 表面活性剂类、 或其组合;  In another preferred embodiment, the povidone-containing K30-containing polymeric carrier further comprises other water-soluble carriers selected from the group consisting of polyethylene glycols, povidones, celluloses, surfactants, Or a combination thereof;
其中,所述聚乙二醇类选自下组:聚乙二醇 4000、聚乙二醇 6000、聚乙二醇 8000 或其组合;  Wherein the polyethylene glycol is selected from the group consisting of polyethylene glycol 4000, polyethylene glycol 6000, polyethylene glycol 8000 or a combination thereof;
所述聚维酮类选自下组:聚维酮 K25、聚维酮 Κ90、聚维酮 S630、或其组合; 所述纤维素类选自下组: 羟丙甲纤维素 E5、 羟丙甲纤维素 E15、 羟丙纤维素 L、 或其组合;  The povidone is selected from the group consisting of povidone K25, povidone oxime 90, povidone S630, or a combination thereof; the cellulose is selected from the group consisting of hypromellose E5, hyprothenol Cellulose E15, hydroxypropyl cellulose L, or a combination thereof;
所述表面活性剂类选自下组: 泊洛沙姆 407、 泊洛沙姆 188、 或其组合。 在另一优选例中,所述含聚维酮 K30的高分子载体包含选自聚维酮 K25、聚维 酮 Κ90、 聚维酮 S630、 或其组合的聚维酮类载体。  The surfactant class is selected from the group consisting of poloxamer 407, poloxamer 188, or a combination thereof. In another preferred embodiment, the povidone-containing K30-containing polymeric carrier comprises a povidone-based carrier selected from the group consisting of povidone K25, povidone 90, povidone S630, or a combination thereof.
在另一优选例中, 所述含聚维酮 K30的高分子载体和其他水溶性载体的重量比 例为 1 : 4-4: 1; 较佳地为 1 : 2-4: 1; 更佳地为 1 : 1-4: 1。  In another preferred embodiment, the weight ratio of the povidone-containing K30-containing polymer carrier to the other water-soluble carrier is 1:4-4:1; preferably 1:2-4:1; more preferably For 1: 1-4: 1.
在另一优选例中, 所述 (A)和 (B)的重量比为 1 : 1-1: 20。  In another preferred embodiment, the weight ratio of the (A) and (B) is 1: 1-1: 20.
在另一优选例中, 所述 (A)和 (B)的重量比为 1 : 2-1: 5。  In another preferred embodiment, the weight ratio of the (A) and (B) is 1: 2-1: 5.
在另一优选例中, 所述固体分散体的 X-射线粉末衍射图中, 不具有所述固体 分散体的活性成分的 X-射线衍射特征峰。  In another preferred embodiment, the X-ray powder diffraction pattern of the solid dispersion does not have an X-ray diffraction characteristic peak of the active component of the solid dispersion.
在另一优选例中, 具有以下一个或多个特征:  In another preferred embodiment, there are one or more of the following features:
① 所述固体分散体的药时曲线下面积是所述固体分散体的活性成分单独给药 时的药时曲线下面积的 6-20倍, 较佳地 7-15倍;  1 The area under the drug-time curve of the solid dispersion is 6-20 times, preferably 7-15 times, the area under the drug-time curve when the active ingredient of the solid dispersion is administered alone;
② 所述固体分散体在 37°C, pH4.5的醋酸钠缓冲水溶液中 10分钟时累积溶出 度大于 70%, 较佳地为 75-95%; 2 The solid dispersion is cumulatively dissolved in a sodium acetate buffered aqueous solution at 37 ° C, pH 4.5 for 10 minutes. Degree greater than 70%, preferably 75-95%;
③ 所述固体分散体在 37°C, pH4.5的醋酸钠缓冲水溶液中 120分钟时累积溶 出度是所述固体分散体的活性成分累积溶出度的 10-40倍, 较佳地为 15-30倍。  3 The cumulative dissolution of the solid dispersion at 37 ° C, pH 4.5 in an aqueous solution of sodium acetate buffer for 120 minutes is 10-40 times, preferably 15-15, of the cumulative dissolution of the active ingredient of the solid dispersion. 30 times.
在另一优选例中, 所述的药时曲线下面积是在以下动物中测得的: 大鼠、 小鼠 或犬。  In another preferred embodiment, the area under the drug time curve is measured in the following animals: rat, mouse or canine.
本发明第二方面提供了一种本发明第一方面所述固体分散体的制备方法,包括 方法:  A second aspect of the present invention provides a method for preparing a solid dispersion according to the first aspect of the present invention, comprising the method of:
(a) 熔融法 1, 包括步骤:  (a) Melting method 1, including steps:
(al) 混合活性成分与亲水性高分子载体, 从而得到一含活性成分的混合物; (a2) 加热熔融步骤 (al)得到的含活性成分的混合物, 从而得到熔融后的混合 物; 和  (al) mixing the active ingredient with a hydrophilic polymer carrier to obtain a mixture containing the active ingredient; (a2) heating the molten component (al) to obtain the mixture containing the active ingredient, thereby obtaining a molten mixture;
(a3) 冷却步骤 (a2)得到的熔融后的混合物, 从而得到本发明第一方面所述的固 体分散体;  (a3) cooling the molten mixture obtained in the step (a2) to obtain the solid dispersion according to the first aspect of the invention;
 Or
(b)熔融法 2, 包括步骤:  (b) Melting method 2, including steps:
(bl) 加热熔融亲水性高分子载体, 从而得到熔融后的载体;  (bl) heating and melting the hydrophilic polymer carrier to obtain a molten carrier;
(b2) 将步骤 (bl)得到的熔融后的载体和活性成分混合后熔融,从而得到熔融后 的混合物; 和  (b2) mixing the molten carrier obtained in the step (bl) with the active ingredient and melting the mixture to obtain a molten mixture;
(b3) 冷却步骤 (b2)得到的熔融后的混合物,从而得到本发明第一方面所述的固 体分散体;  (b3) cooling the molten mixture obtained in the step (b2) to obtain the solid dispersion according to the first aspect of the invention;
 Or
(c) 溶剂法, 包括步骤:  (c) Solvent method, including steps:
(cl) 将活性成分与亲水性高分子载体共同溶解于溶剂中, 从而得到一混合物; 禾口  (cl) co-dissolving the active ingredient and the hydrophilic polymer carrier in a solvent to obtain a mixture;
(c2) 除去步骤 (cl)得到的混合物中的溶剂, 从而得到本发明第一方面所述的固 体分散体;  (c2) removing the solvent in the mixture obtained in the step (cl), thereby obtaining the solid dispersion according to the first aspect of the invention;
其中, 所述活性成分为式 A化合物或其衍生物、 或其药学上可接受的盐、 水合 物或溶剂合物,  Wherein the active ingredient is a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof,
Figure imgf000004_0001
Figure imgf000004_0001
式中, R为 CD3或 CH3Where R is CD 3 or CH 3 ,
所述亲水性高分子载体为含聚维酮 K30的亲水性高分子载体,  The hydrophilic polymer carrier is a hydrophilic polymer carrier containing povidone K30.
所述溶剂选自下组: 甲醇、 乙醇、 异丙醇、 丙酮、 二氯甲垸、 四氢呋喃, 或其 组合。 The solvent is selected from the group consisting of methanol, ethanol, isopropanol, acetone, chloroform, tetrahydrofuran, or Combination.
在另一优选例中, 所述步骤 (c2)包括步骤:  In another preferred embodiment, the step (c2) includes the steps of:
用旋转蒸发仪除去步骤 (cl)得到的混合物中的溶剂; 或者  The solvent in the mixture obtained in the step (cl) is removed by a rotary evaporator; or
用喷雾干燥器干燥除去步骤 (cl)得到的混合物中的溶剂。  The solvent in the mixture obtained in the step (cl) was removed by drying with a spray dryer.
在另一优选例中, 所述干燥的工艺参数为: 进风温度 110 〜 120°C, 风机功 率 55 %〜 65 %, 蠕动泵功率 50 %〜 55 %。  In another preferred embodiment, the drying process parameters are: inlet air temperature 110 to 120 ° C, fan power 55 % to 65 %, peristaltic pump power 50 % to 55 %.
在另一优选例中,所述固体分散体进一步用至少一种另外的加工步骤处理,所 述加工步骤为研磨、 筛分、 碾压、 碾磨、 筛选、 混合或其组合。  In another preferred embodiment, the solid dispersion is further treated with at least one additional processing step of grinding, sieving, rolling, milling, screening, mixing, or a combination thereof.
本发明第三方面提供了一种药物组合物, 所述组合物包括:  A third aspect of the invention provides a pharmaceutical composition, the composition comprising:
(1)本发明第一方面所述的固体分散体; 和  (1) The solid dispersion of the first aspect of the invention; and
(2)药学上可接受的赋形剂。  (2) A pharmaceutically acceptable excipient.
在另一优选例中, 所述赋形剂包括: 填充剂、 稀释剂、 甜味剂、 矫味剂、 抗氧 化剂、 着色剂、 防腐剂、 润滑剂、 粘合剂、 崩解剂、 或其组合。  In another preferred embodiment, the excipient comprises: a filler, a diluent, a sweetener, a flavoring agent, an antioxidant, a coloring agent, a preservative, a lubricant, a binder, a disintegrating agent, or combination.
在另一优选例中, 所述组合物为胶囊剂、 片剂、 丸剂、 散剂和颗粒剂。  In another preferred embodiment, the composition is a capsule, a tablet, a pill, a powder, and a granule.
在另一优选例中, 所述药物组合物, 用于抑制磷酸激酶 (如 raft敫酶)的药物 组合物。  In another preferred embodiment, the pharmaceutical composition is for use in a pharmaceutical composition for inhibiting a phosphokinase such as raft chymase.
在另一优选例中, 所述的药物组合物用于治疗和预防癌症。  In another preferred embodiment, the pharmaceutical composition is for the treatment and prevention of cancer.
本发明第四方面提供了一种本发明第三方面所述药物组合物的制备方法,将本 发明第一方面所述的固体分散体和药学上可接受的赋形剂混合,从而制得所述的药 物组合物。  According to a fourth aspect of the present invention, there is provided a process for the preparation of a pharmaceutical composition according to the third aspect of the present invention, wherein the solid dispersion according to the first aspect of the present invention is mixed with a pharmaceutically acceptable excipient, thereby producing a Said pharmaceutical composition.
本发明第五方面提供了一种本发明第一方面所述的固体分散体的用途,用于制 备抑制磷酸激酶 (如 raf^酶)的药物; 或用于制备治疗肿瘤的药物。  A fifth aspect of the invention provides the use of the solid dispersion of the first aspect of the invention for the preparation of a medicament for inhibiting a phosphokinase (e.g., raf^ enzyme); or for the preparation of a medicament for treating a tumor.
本发明第六方面提供了一种治疗方法, 将 (i)本发明第一方面所述的固体分散 体或 (ii)本发明第三方面所述的药物组合物, 施用于需要治疗的患者。  A sixth aspect of the invention provides a method of treating (i) the solid dispersion of the first aspect of the invention or (ii) the pharmaceutical composition of the third aspect of the invention for a patient in need of treatment.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文 (如实施例) 中具体描述的各技术特征之间都可以互相组合, 从而构成新的或优选的技术方 案。 限于篇幅, 在此不再一一累述。 附图说明  It is to be understood that within the scope of the present invention, the various technical features of the present invention and the technical features specifically described hereinafter (e.g., the embodiments) can be combined with each other to constitute a new or preferred technical solution. Due to space limitations, we will not repeat them here. DRAWINGS
图 1显示了固体分散体 1的 XRPD图。  Figure 1 shows an XRPD pattern of Solid Dispersion 1.
图 2显示了固体分散体 2的 XRPD图。  Figure 2 shows an XRPD pattern of Solid Dispersion 2.
图 3显示了固体分散体 3的 XRPD图。  Figure 3 shows an XRPD pattern of Solid Dispersion 3.
图 4显示了固体分散体 4的 XRPD图。  Figure 4 shows an XRPD pattern of Solid Dispersion 4.
图 5显示了固体分散体 5的 XRPD图。  Figure 5 shows an XRPD pattern of solid dispersion 5.
图 6显示了固体分散体 6的 XRPD图。 图 7显示了固体分散体 7的 XRPD图。 Figure 6 shows an XRPD pattern of solid dispersion 6. Figure 7 shows an XRPD pattern of solid dispersion 7.
图 8显示了固体分散体 8的 XRPD图。  Figure 8 shows an XRPD pattern of solid dispersion 8.
图 9显示了固体分散体 9的 XRPD图。  Figure 9 shows an XRPD pattern of Solid Dispersion 9.
图 10显示了固体分散体 10的 XRPD图。  Figure 10 shows an XRPD pattern of solid dispersion 10.
图 1 1显示了固体分散体 1 1的 XRPD图。  Figure 1 1 shows an XRPD pattern of the solid dispersion 1 1 .
图 12显示了式 II化合物、式 II化合物和 PVP K30的物理混合物以及固体分散 体 1的溶出曲线。  Figure 12 shows the physical mixture of the compound of the formula II, the compound of the formula II and the PVP K30 and the dissolution profile of the solid dispersion 1.
图 13显示了固体分散体 12的 XRPD图。  Figure 13 shows an XRPD pattern of solid dispersion 12.
图 14显示了固体分散体 4经 1、 2、 3个月加速稳定性试验后 XRPD对比图, 结 果如图 14所示: 从上往下依次是式 II化合物对甲基苯磺酸盐、 新制备的固体分 散体 4、 固体分散体 4放置一个月时、 固体分散体 4放置两个月时、 固体分散体 4 放置三个月时的 XRPD图。  Figure 14 shows a comparison of XRPD of the solid dispersion 4 after 1, 2, and 3 months accelerated stability test. The results are shown in Figure 14: From top to bottom, the compound of formula II is p-toluenesulfonate, new The prepared solid dispersion 4, the XRPD pattern when the solid dispersion 4 was left for one month, the solid dispersion 4 was left for two months, and the solid dispersion 4 was left for three months.
图 15显示了固体分散体 5经 1、 2、 3个月加速稳定性试验后 XRPD对比图, 结 果如图 15所示: 从上往下依次是式 II化合物对甲基苯磺酸盐、 新制备的固体分 散体 5、 固体分散体 5放置一个月时、 固体分散体 5放置两个月时、 固体分散体 5 放置三个月时的 XRPD图。  Figure 15 shows a comparison of XRPD of the solid dispersion 5 after 1, 2, and 3 months accelerated stability test. The results are shown in Figure 15: From top to bottom, the compound of formula II is p-toluenesulfonate, new The prepared solid dispersion 5, the XRPD pattern when the solid dispersion 5 was left for one month, the solid dispersion 5 was left for two months, and the solid dispersion 5 was left for three months.
图 16显示了固体分散体 1经 1、 2、 3个月加速稳定性试验后 XRPD对比图, 结 果如图 16所示: 从上往下依次是式 II化合物、 新制备的固体分散体 1、 固体分散 体 1放置一个月时、 固体分散体 1放置两个月时、 固体分散体 1放置三个月时的 XRPD图。  Figure 16 shows a comparison of XRPD of the solid dispersion 1 after 1, 2, and 3 months accelerated stability test, and the results are shown in Figure 16: From the top to the bottom, the compound of the formula II, the newly prepared solid dispersion 1, The XRPD pattern of the solid dispersion 1 when it was left for one month, when the solid dispersion 1 was left for two months, and when the solid dispersion 1 was left for three months.
图 17显示了固体分散体 3经 1、 2、 3个月加速稳定性试验后 XRPD对比图, 结 果如图 17所示: 从上往下依次是式 II化合物、 新制备的固体分散体 3、 固体分散 体 3放置一个月时、 固体分散体 3放置两个月时、 固体分散体 3放置三个月时的 XRPD图。 具体实施方式  Figure 17 shows a comparison of XRPD of the solid dispersion 3 after 1, 2, and 3 months accelerated stability test. The results are shown in Figure 17: From the top to the bottom, the compound of the formula II, the newly prepared solid dispersion 3, The XRPD pattern when the solid dispersion 3 was left for one month, when the solid dispersion 3 was left for two months, and when the solid dispersion 3 was left for three months. detailed description
本发明人通过长期而深入的研究,意外地发现一种包含 (A)作为活性成分的 式 A化合物或其衍生物、 或其药学上可接受的盐、 水合物或溶剂合物和 (B)亲水性 高分子载体 (尤其是含聚维酮 K30的高分子载体)的固体分散体,相对于活性成分 而言, 所述固体分散体的药时曲线下面积约为活性成分的 6-20倍; 所述固体分 散体明显提高其活性成分的体外溶出度,显著改善哺乳动物对活性成分的吸收, 从而有效地减小其使用剂量。 此外, 所述固体分散体还具有非常好的稳定性, 非常适用于制备相关疾病的药物组合物。 在此基础上, 发明人完成了本发明。 固体分散体 固体分散体 (solid dispersion)是指将活性成分高度分散于 (固体)载体中形成 的一种以固体形式存在的分散系统。 活性成分 The present inventors have unexpectedly discovered, by long-term and intensive research, a compound of the formula A or a derivative thereof comprising (A) as an active ingredient, or a pharmaceutically acceptable salt, hydrate or solvate thereof and (B) a solid dispersion of a hydrophilic polymer carrier (especially a polymer carrier containing povidone K30), wherein the area under the drug-time curve of the solid dispersion is about 6-20 of the active ingredient relative to the active ingredient The solid dispersion significantly increases the in vitro dissolution of the active ingredient, and significantly improves the absorption of the active ingredient by the mammal, thereby effectively reducing the dosage thereof. In addition, the solid dispersion also has very good stability and is very suitable for the preparation of pharmaceutical compositions for related diseases. On this basis, the inventors have completed the present invention. Solid dispersion Solid dispersion refers to a dispersion system in which the active ingredient is highly dispersed in a (solid) support to form a solid form. Active ingredient
如本文所用, 术语"活性成分"指作为活性成分的式 A化合物或其衍生物、 或 其药学上可接受的盐,  The term "active ingredient" as used herein refers to a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt thereof, as an active ingredient,
Figure imgf000007_0001
Figure imgf000007_0001
该术语还包括及式 A化合物或其衍生物的各种晶型形式、 水合物或溶剂合 优选地, 所 物:  The term also encompasses various crystalline forms, hydrates or solvates of a compound of formula A or a derivative thereof, preferably:
Figure imgf000007_0002
Figure imgf000007_0002
如本文所用, 术语"药学上可接受的盐"指本发明化合物与酸或碱所形成的 适合用作药物的盐。 药学上可接受的盐包括无机盐和有机盐。 一类优选的盐是 本发明化合物与酸形成的盐。适合形成盐的酸包括但并不限于: 盐酸、氢溴酸、 氢氟酸、 硫酸、 硝酸、 磷酸等无机酸, 甲酸、 乙酸、 丙酸、 草酸、 丙二酸、 琥 珀酸、 富马酸、 马来酸、 乳酸、 苹果酸、 酒石酸、 柠檬酸、 苦味酸、 甲磺酸、 苯甲磺酸, 苯磺酸等有机酸; 以及天冬氨酸、 谷氨酸等酸性氨基酸。  The term "pharmaceutically acceptable salt" as used herein refers to a salt of the compound of the present invention which is formed with an acid or a base and which is suitable for use as a medicament. Pharmaceutically acceptable salts include inorganic and organic salts. A preferred class of salts are the salts of the compounds of the invention with acids. Suitable acids for forming salts include, but are not limited to: mineral acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, Organic acids such as maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, benzoic acid, benzenesulfonic acid; and acidic amino acids such as aspartic acid and glutamic acid.
优选地, 所述药学上可接受的盐为对甲基苯磺酸盐。 载体  Preferably, the pharmaceutically acceptable salt is p-toluenesulfonate. Carrier
固体分散体所用的载体, 最常用的有水溶性载体、 难溶性载体和肠溶性载 本发明所述的 "载体 "为亲水性高分子载体,优选为聚维酮或含聚维酮的高分 子载体, 或者聚维酮 K30或含聚维酮 K30的高分子载体, 其中, 聚维酮 K30的含量 为 10-100wt%, 按高分子载体的总重量计。  The carrier used for the solid dispersion, the most commonly used water-soluble carrier, poorly soluble carrier and enteric carrier. The "carrier" of the present invention is a hydrophilic polymer carrier, preferably povidone or povidone-containing. A molecular carrier, or povidone K30 or a polymeric carrier containing povidone K30, wherein the povidone K30 is contained in an amount of 10 to 100% by weight based on the total weight of the polymer carrier.
更优选地, 本发明所用的含聚维酮 K30的高分子载体是指聚维酮 K30的重量含 量超过载体总重量 20%的载体; 或聚维酮 K30的重量含量超过载体总重量 40%的载 体; 或聚维酮 K30的重量含量超过载体总重量 60%的载体。 More preferably, the povidone K30-containing polymer carrier used in the present invention refers to the weight of povidone K30. A carrier having a weight exceeding 20% by weight of the carrier; or a carrier having a weight content of povidone K30 of more than 40% by weight based on the total weight of the carrier; or a carrier having a weight content of povidone K30 of more than 60% by weight based on the total weight of the carrier.
当然, 所述亲水性高分子载体还可以含有其他水溶性载体, 例如包括 (但不限 于): 聚乙二醇类、 聚维酮类、 纤维素类、 表面活性剂类、 或其组合。  Of course, the hydrophilic polymeric carrier may also contain other water soluble carriers such as, but not limited to, polyethylene glycols, povidones, cellulosics, surfactants, or combinations thereof.
所述聚乙二醇类选自下组: 聚乙二醇 4000(PEG 4000)、 聚乙二醇 6000(PEG The polyethylene glycols are selected from the group consisting of polyethylene glycol 4000 (PEG 4000), polyethylene glycol 6000 (PEG)
6000)、 聚乙二醇 8000(PEG 8000)或其组合; 6000), polyethylene glycol 8000 (PEG 8000) or a combination thereof;
所述聚维酮类选自下组: 聚维酮 K90(PVP K90)、 聚维酮 K25(PVP K25)、 聚 维酮 S630、 或其组合;  The povidone is selected from the group consisting of povidone K90 (PVP K90), povidone K25 (PVP K25), povidone S630, or a combination thereof;
所述纤维素类选自下组: 羟丙甲纤维素 E5(HPMC E5)、 羟丙甲纤维素 E15(HPMC E15)、 羟丙纤维素 L(HPC-L)、 或其组合;  The cellulose is selected from the group consisting of hypromellose E5 (HPMC E5), hypromellose E15 (HPMC E15), hydroxypropylcellulose L (HPC-L), or a combination thereof;
所述表面活性剂类选自下组: 泊洛沙姆 407(Poloxamer 407)、 泊洛沙姆 188(Poloxamer 188)、 或其组合。 组成  The surfactant class is selected from the group consisting of poloxamer 407 (Poloxamer 407), Poloxamer 188 (Poloxamer 188), or a combination thereof. Composition
本发明提供的固体分散体包括:  The solid dispersions provided by the present invention include:
(A) 作为活性成分的式 A化合物或其衍生物、 或其药学上可接受的盐、 水合物 或溶剂合物, 其中, 式 A化合物同上文中所述。 和  (A) A compound of the formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, as the active ingredient, wherein the compound of the formula A is as described above. with
(B) 亲水性高分子载体;  (B) a hydrophilic polymer carrier;
其中, (A)和 (B)的重量比为 1 : 1-1: 40。  Wherein the weight ratio of (A) and (B) is 1: 1-1: 40.
优选地, 所述 (A)和 (B)的重量比为 1 : 20-1: 1, 较佳地, 所述 (A)和 (B)的重量 比为 1 : 5-1: 2。  Preferably, the weight ratio of the (A) and (B) is 1: 20-1: 1, preferably, the weight ratio of the (A) and (B) is 1: 5-1: 2.
制备方法  Preparation
本发明所述固体分散体可通过如下的方法制得,然而该方法的条件,例如载体、 溶剂、 各成分的量、 制备温度、 制备所需时间等不限于下面的解释。 本发明所述固 体分散体还可以任选将在本说明书中描述的或本领域已知的各种合成方法组合起 来而方便的制得, 这样的组合可由本发明所属领域的技术人员容易的进行。  The solid dispersion of the present invention can be obtained by the following method, however, the conditions of the method, such as the carrier, the solvent, the amounts of the respective components, the preparation temperature, the time required for preparation, and the like are not limited to the following explanations. The solid dispersions of the present invention may also be conveniently prepared by combining various synthetic methods described in the specification or known in the art, such combinations being readily made by those skilled in the art to which the present invention pertains. .
具体地, 所述制备方法可以是熔融法、 溶剂法、 溶剂-熔融法、 喷雾干燥法、 或表面分散法。 优选地, 所述制备方法是熔融法或溶剂法。 熔融法  Specifically, the preparation method may be a melting method, a solvent method, a solvent-melting method, a spray drying method, or a surface dispersion method. Preferably, the preparation method is a melt method or a solvent method. Melting method
通常的, 所述熔融法是将 (A)活性成分和 (B)亲水性高分子载体混合均匀, 用水 浴或油浴加热至熔融,然后冷却成固体,粉碎后得到粉末状的所述活性成分的固体 分散体。  Usually, in the melting method, the (A) active ingredient and (B) the hydrophilic polymer carrier are uniformly mixed, heated to melt in a water bath or an oil bath, and then cooled to a solid, and pulverized to obtain the active substance in a powder form. A solid dispersion of the ingredients.
或者, 可将 (B) 亲水性高分子载体加热熔融后, 再加入 (A)活性成分, 然后将 熔融的混合物在剧烈搅拌下,迅速冷却成固体,粉碎后得到粉末状的所述活性成分 的固体分散体。 溶剂法 Alternatively, the (B) hydrophilic polymer carrier may be heated and melted, and then (A) the active ingredient may be added, and then The molten mixture is rapidly cooled to a solid under vigorous stirring, and after pulverization, a solid dispersion of the active ingredient in a powder form is obtained. Solvent method
通常的, 所述溶剂法是将 (A)活性成分和 (B)亲水性高分子载体共同溶解于溶剂 中, 混合物均匀后, 除去溶液中的溶剂, 可使活性成分和亲水性高分子载体同时析 出, 粉碎后即可得到粉末状的所述活性成分的固体分散体。  In general, the solvent method is a method in which (A) an active ingredient and (B) a hydrophilic polymer carrier are co-dissolved in a solvent, and after the mixture is homogeneous, the solvent in the solution is removed to obtain an active ingredient and a hydrophilic polymer. The carrier is simultaneously precipitated, and after pulverization, a solid dispersion of the active ingredient in a powder form is obtained.
所述溶剂法所使用的溶剂包括: 甲醇、 乙醇、 异丙醇、 丙酮、 四氢呋喃、 或其 组合。 结构鉴定  The solvent used in the solvent method includes: methanol, ethanol, isopropanol, acetone, tetrahydrofuran, or a combination thereof. Structure Identification
固体分散体的结构鉴定方法包括差示扫描量热分析、 X-射线粉末衍射等。 分 别对活性成分、 载体和本发明所述方法制备的固体分散体, 进行了 X-射线粉末衍 射 (XRPD)检测, 比较三者的 XRPD谱图。 活性成分的特征峰  Structure identification methods for solid dispersions include differential scanning calorimetry, X-ray powder diffraction, and the like. The solid dispersion prepared by the active ingredient, the carrier and the method of the present invention were subjected to X-ray powder diffraction (XRPD) detection, and the XRPD spectra of the three were compared. Characteristic peak of active ingredient
当制成本发明的固体分散体后, 活性成分的 X-射线衍射特征峰基本上或完全 消失。 换言之, 本发明固体分散体的 X-射线粉末衍射图中, 不具有所述固体分散 体的活性成分的 X-射线衍射特征峰。  When the solid dispersion of the present invention is formed, the X-ray diffraction characteristic peak of the active ingredient disappears substantially or completely. In other words, the X-ray powder diffraction pattern of the solid dispersion of the present invention does not have an X-ray diffraction characteristic peak of the active component of the solid dispersion.
用 XRPD测定时, 活性成分的典型衍射峰 (特征峰)如下;  The typical diffraction peak (characteristic peak) of the active ingredient when measured by XRPD is as follows;
式 I化合物, 其在 14·58±0· 、 18·71±0· 、 21·08±0·1。、 23·35±0· 、 29.44±0. 有强衍射峰。  A compound of formula I, which is at 14.58±0·, 18·71±0·, 21·08±0·1. , 23·35±0·, 29.44±0. Strong diffraction peaks.
式 II化合物, 其在 12·49±0·1。、 22·40±0· 、 22·83±0· 、 24.69±0.1。、 25.11±0.1° 有强衍射峰。  A compound of formula II which is at 12.49 ± 0.1. , 22·40±0·, 22·83±0·, 24.69±0.1. 25.11±0.1° has strong diffraction peaks.
式 II化合物的对甲基苯磺酸盐, 其在 4.40±0. 、 13.18±0. 、 14.74±0.1°、 a p-toluenesulfonate of the compound of formula II, which is at 4.40 ± 0., 13.18 ± 0., 14.74 ± 0.1 °,
16·64±0·1ο、 17·82±0·1ο、 20·01±0·1ο、 20·41±0·1ο、 20·78±0·1ο、 21·47±0·1ο、 22·83±0·1ο 有强衍射峰。 载体的特征峰 16·64±0·1 ο , 17·82±0·1 ο , 20·01±0·1 ο , 20·41±0·1 ο , 20·78±0·1 ο , 21·47±0 ·1 ο , 22·83±0·1 ο Strong diffraction peaks. Characteristic peak of the carrier
有些载体有强衍射峰, 例如, PEG4000和PEG6000在19.19±0. 、 23.30±0.1° 有强衍射峰。?010 &11½ 407在19.16±0.1°、 23.32±0.1°有强衍射峰。 有些载体没有 强衍射峰, 例如, PVP K30、 HPMC E5和 HPC-L等均没有检测到强衍射峰。 固体分散体的特征峰 Some carriers have strong diffraction peaks. For example, PEG4000 and PEG6000 have strong diffraction peaks at 19.19±0. and 23.30±0.1°. ? 01 0 & 111⁄2 407 has strong diffraction peaks at 19.16 ± 0.1 ° and 23.32 ± 0.1 °. Some carriers do not have strong diffraction peaks. For example, strong diffraction peaks are not detected in PVP K30, HPMC E5, and HPC-L. Characteristic peak of solid dispersion
本发明制备的固体分散体的 XRPD谱图中活性成分的强衍射峰消失, 从而确定 本发明所述的固体分散体制备成功, 其中不是活性成分和载体的简单的物理混合 物, 而是以无定型或分子形态存在。 药物组合物 The strong diffraction peak of the active ingredient disappears in the XRPD spectrum of the solid dispersion prepared by the present invention, thereby confirming the successful preparation of the solid dispersion of the present invention, which is not a simple physical mixing of the active ingredient and the carrier. But in an amorphous or molecular form. Pharmaceutical composition
术语 "本发明化合物"指式 A化合物或其衍生物、 或其药学上可接受的盐。 术语"本发明固体分散体"指活性成分为式 A化合物或其衍生物、 或其药学 上可接受的盐的固态分散体。  The term "compound of the invention" refers to a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt thereof. The term "solid dispersion of the invention" means a solid dispersion of the active ingredient which is a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt thereof.
由于式 A化合物 (或其衍生物、 或其盐)具有优异的对磷酸激酶 (KinaseM列如 raft敫酶的抑制活性, 因此本发明固体分散体以及含有本发明固体分散体为主要 成分的药物组合物可用于治疗、 预防以及缓解由对磷酸激酶 (KinaseM列如 raft敫 酶介导的疾病。 根据现有技术, 本发明化合物可用于治疗以下疾病: 癌症、 心 血管疾病、 肥胖病、 糖尿病等等。  Since the compound of the formula A (or a derivative thereof, or a salt thereof) has excellent inhibitory activity against phosphokinase (Kinase M column such as raft敫 enzyme, the solid dispersion of the present invention and the pharmaceutical composition containing the solid dispersion of the present invention as a main component The compounds are useful for the treatment, prevention, and alleviation of diseases mediated by phosphokinase (Kinase M, such as raft敫 enzyme. According to the prior art, the compounds of the present invention are useful for treating diseases such as cancer, cardiovascular disease, obesity, diabetes, etc. .
本发明的药物组合物包含安全有效量范围内的本发明固体分散体或其药 理上可接受的盐及药理上可以接受的赋形剂。 其中"安全有效量"指的是: 固体 分散体的量足以明显改善病情, 而不至于产生严重的副作用。 通常, 药物组合 物含有 l-2000mg本发明固体分散体 /剂, 更佳地, 含有 10-200mg本发明固体分 散体 /剂。 较佳地, 所述的"一剂"为一个胶囊或药片。  The pharmaceutical compositions of the present invention comprise a solid dispersion of the present invention or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient in a safe and effective amount. By "safe and effective amount" is meant: The amount of solid dispersion is sufficient to significantly improve the condition without causing serious side effects. In general, the pharmaceutical composition contains from 1 to 2000 mg of the solid dispersion/agent of the invention, more preferably from 10 to 200 mg of the solid dispersion/agent of the invention. Preferably, the "one dose" is a capsule or a tablet.
"药学上可以接受的赋形剂 "指的是: 一种或多种相容性固体或液体填料或 凝胶物质, 它们适合于人使用, 而且必须有足够的纯度和足够低的毒性。 "相容 性"在此指的是组合物中各组份能和本发明的固体分散体以及它们之间相互惨 和, 而不明显降低固体分散体的药效。 药学上可以接受的赋形剂部分例子有纤 维素及其衍生物 (如羧甲基纤维素钠、 乙基纤维素钠、 纤维素乙酸酯等)、 淀粉 类、 单元糖或多元糖、 明胶、 滑石、 固体润滑剂 (如硬脂酸、 硬脂酸镁)、 硫酸 钙、 植物油 (如豆油、 芝麻油、 花生油、 橄榄油等)、 多元醇 (如丙二醇、 甘油、 甘露醇、 山梨醇等)、 乳化剂 (如吐温 ®)、 润湿剂 (如十二垸基硫酸钠)、 着色剂、 调味剂、 稳定剂、 抗氧化剂、 防腐剂、 无热原水等。  "Pharmaceutically acceptable excipient" means: one or more compatible solid or liquid fillers or gel materials which are suitable for human use and which must be of sufficient purity and of sufficiently low toxicity. By "compatibility" it is meant herein that the components of the composition are compatible with the solid dispersions of the present invention and between them without significantly reducing the efficacy of the solid dispersion. Examples of pharmaceutically acceptable excipients are cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), starches, monosaccharides or polysaccharides, gelatin , talc, solid lubricants (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.) , emulsifiers (such as Tween®), wetting agents (such as sodium decyl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
本发明固体分散体或药物组合物的施用方式没有特别限制, 代表性的施用 方式包括 (但并不限于): 口服、 瘤内、 直肠、 肠胃外 (静脉内、 肌肉内或皮下)、 和局部给药。  The mode of administration of the solid dispersion or pharmaceutical composition of the present invention is not particularly limited, and representative modes of administration include, but are not limited to, oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical Dosing.
用于口服给药的固体剂型包括胶囊剂、 片剂、 丸剂、 散剂和颗粒剂。 在这 些固体剂型中, 固体分散体与至少一种常规惰性赋形剂 (或载体)混合, 如柠檬 酸钠或磷酸二钙, 或与下述成分混合: (a) 填料或增容剂, 例如, 淀粉、 乳糖、 蔗糖、 葡萄糖、 微晶纤维素、 甘露醇和硅酸; (b) 粘合剂, 例如, 羟丙甲基纤 维素、 藻酸盐、 明胶、 聚乙烯基吡咯垸酮、 蔗糖和阿拉伯胶; (c) 保湿剂, 例 如, 甘油; (d) 崩解剂, 例如, 琼脂、 碳酸钙、 马铃薯淀粉或木薯淀粉、 藻酸、 某些复合硅酸盐、交联羧甲基纤维素钠和碳酸钠; (e) 缓溶剂,例如石蜡;(f) 吸 收加速剂, 例如, 季胺化合物; (g) 润湿剂, 例如鲸蜡醇、 十二垸基硫酸钠、 单硬脂酸甘油酯; (h) 吸附剂, 例如, 高岭土; 和 (i) 润滑剂, 例如, 滑石、 硬 脂酸钙、 硬脂酸镁、 固体聚乙二醇、 十二垸基硫酸钠, 或其混合物。 胶囊剂、 片剂和丸剂中, 剂型也可包含缓冲剂。 Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In these solid dosage forms, the solid dispersion is admixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or mixed with: (a) a filler or compatibilizer, for example , starch, lactose, sucrose, glucose, microcrystalline cellulose, mannitol and silicic acid; (b) binders, for example, hydroxypropylmethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and (a) humectants, for example, glycerin; (d) disintegrants, for example, agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, croscarmellose Sodium and sodium carbonate; (e) a slow solvent such as paraffin; (f) aspiration Accelerator, for example, a quaternary amine compound; (g) a wetting agent such as cetyl alcohol, sodium dodecyl sulfate, glyceryl monostearate; (h) an adsorbent, for example, kaolin; and (i) Lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium dodecyl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage form may also contain a buffer.
固体剂型如片剂、 糖丸、 胶囊剂、 丸剂和颗粒剂可采用包衣和壳材制备, 如肠衣和其它本领域公知的材料。 它们可包含不透明剂, 并且, 这种组合物中 固体分散体的释放可以延迟的方式在消化道内的某一部分中释放。 可采用的包 埋组分的实例是聚合物质和蜡类物质。 必要时, 固体分散体也可与上述赋形剂 中的一种或多种形成微胶囊形式。  Solid dosage forms such as tablets, troches, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other materials known in the art. They may contain opacifying agents and the release of the solid dispersion in such compositions may be released in a portion of the digestive tract in a delayed manner. Examples of embedding components that can be employed are polymeric and waxy materials. The solid dispersion may also form a microcapsule form with one or more of the above excipients as necessary.
用于口服给药的液体剂型包括药学上可接受的乳液、 溶液、 悬浮液、 糖桨 或酊剂。除了固体分散体外,液体剂型可包含本领域中常规采用的惰性稀释剂, 如水或其它溶剂, 增溶剂和乳化剂, 例知, 乙醇、 异丙醇、 碳酸乙酯、 乙酸乙 酯、 丙二醇、 1,3-丁二醇、 二甲基甲酰胺以及油, 特别是棉籽油、 花生油、 玉 米胚油、 橄榄油、 蓖麻油和芝麻油或这些物质的混合物等。  Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, lozenges or elixirs. In addition to the solid dispersion, the liquid dosage form may comprise an inert diluent conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or a mixture of these substances.
除了这些惰性稀释剂外, 组合物也可包含助剂, 如润湿剂、 乳化剂和悬浮 剂、 甜味剂、 矫味剂和香料。  In addition to these inert diluents, the compositions may contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfumes.
除了固体分散体外, 悬浮液可包含悬浮剂, 例如, 乙氧基化异十八垸醇、 聚氧乙烯山梨醇和脱水山梨醇酯、 微晶纤维素、 甲醇铝和琼脂或这些物质的混 合物等。  In addition to the solid dispersion, the suspension may contain a suspending agent, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan ester, microcrystalline cellulose, aluminum methoxide and agar or a mixture of these and the like.
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、 分 散液、 悬浮液或乳液, 和用于重新溶解成无菌的可注射溶液或分散液的无菌粉 末。 适宜的含水和非水载体、 稀释剂、 溶剂或赋形剂包括水、 乙醇、 多元醇及 其适宜的混合物。  Compositions for parenteral injection may comprise a physiologically acceptable sterile aqueous or nonaqueous solution, dispersion, suspension or emulsion, and sterile powder for reconstitution into a sterile injectable solution or dispersion. Suitable aqueous and nonaqueous vehicles, diluents, solvents or vehicles include water, ethanol, polyol, and suitable mixtures thereof.
用于局部给药的本发明固体分散体的剂型包括软膏剂、 散剂、 贴剂、 喷射 剂和吸入剂。 本发明固体分散体在无菌条件下与生理上可接受的载体及任何防 腐剂、 缓冲剂, 或必要时可能需要的推进剂一起混合。  Dosage forms for solid dispersions of the invention for topical administration include ointments, powders, patches, propellants and inhalants. The solid dispersions of the present invention are mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
本发明固体分散体可以单独给药, 或者与其他药学上可接受的化合物联合 给药。 使用药物组合物时, 是将安全有效量的本发明固体分散体适用于需要治 疗的哺乳动物 (如人),其中施用时剂量为药学上认为的有效给药剂量,对于 60kg 体重的人而言, 日给药剂量通常为 l〜2000mg, 优选 20〜500mg。 当然, 具体 剂量还应考虑给药途径、 病人健康状况等因素, 这些都是熟练医师技能范围之 内的。 本发明的主要优点是:  The solid dispersion of the present invention can be administered alone or in combination with other pharmaceutically acceptable compounds. When a pharmaceutical composition is used, a safe and effective amount of the solid dispersion of the present invention is suitably applied to a mammal (e.g., a human) in need of treatment wherein the dosage is a pharmaceutically effective effective dosage for administration to a 60 kg body weight. The daily dose is usually from 1 to 2000 mg, preferably from 20 to 500 mg. Of course, specific doses should also consider factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled physician. The main advantages of the invention are:
(1) 提供了一种以式 A化合物或其衍生物、或其药学上可接受的盐、水合物或 溶剂合物为活性成分固体分散体,所述固体分散体显著改善了活性成分的体外溶 出度, 非常有利于哺乳动物对该活性成分的吸收。 而且相对于其活性成分化合 物本身而言,所述固体分散体具有显著优异的体内药代动力学参数 (尤其是药时 曲线下面积), 且非常稳定。 (1) Providing a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate thereof or The solvate is a solid dispersion of the active ingredient which significantly improves the in vitro dissolution of the active ingredient and is highly advantageous for mammals to absorb the active ingredient. Moreover, the solid dispersion has significantly superior in vivo pharmacokinetic parameters (especially the area under the curve of the drug) relative to the active ingredient compound itself, and is very stable.
(2) 提供了一种所述固体分散体的制备方法。 下面结合具体实施, 进一步阐述本发明。 应理解, 这些实施例仅用于说明 本发明而不用于限制本发明的范围。 下列实施例中未注明具体条件的实验方 法, 通常按照常规条件, 或按照制造厂商所建议的条件。 除非另外说明, 否则 百分比和份数按重量计算。  (2) A method of preparing the solid dispersion is provided. The invention will be further elucidated below in conjunction with specific implementations. It is to be understood that the examples are merely illustrative of the invention and are not intended to limit the scope of the invention. The experimental methods in which the specific conditions are not specified in the following examples are usually carried out according to conventional conditions or according to the conditions recommended by the manufacturer. Percentages and parts are by weight unless otherwise stated.
如本文所用 "API" 是指活性成分。 实施例  As used herein, "API" refers to the active ingredient. Example
固体分散体的溶出度测试方法  Dissolution test method for solid dispersion
按照《中华人民共和国药典》 2010年版二部附录 XC溶出度测定法第二法 (桨 法)进行检测。 条件如下: 溶出介质: 0.1%十二垸基硫酸钠 pH 4.5醋酸-醋酸钠 缓冲液 900mL/溶出杯; 温度: 37±0.5°C ; 转速: 100 转 /分钟。  According to the "People's Republic of China Pharmacopoeia" 2010 edition of the second appendix XC dissolution method of the second method (paddle method) for testing. The conditions are as follows: Dissolution medium: 0.1% sodium decyl sulfate pH 4.5 acetic acid-sodium acetate buffer 900 mL/dissolution cup; temperature: 37 ± 0.5 ° C; rotation speed: 100 rpm.
具体地, 将样品分为三组, 每组样品分别配置 3份。 其中, 第 1组样品是活 性成分; 第 2组样品是只是将活性成分与载体通过简单的物理方法 (如搅拌)混合 后的样品。 第三组样品是按照各个实施例中制备方法得到的固体分散体。 具体 如下表。  Specifically, the samples were divided into three groups, each of which was configured with 3 portions. Among them, the first group of samples is an active ingredient; the second group of samples is a sample obtained by simply mixing the active ingredient with a carrier by a simple physical method such as stirring. The third set of samples was a solid dispersion obtained according to the preparation method in each of the examples. The details are as follows.
Figure imgf000012_0001
Figure imgf000012_0001
使用所述溶出介质, 按照常规方法分别检测以上 3组样品的溶出度, 每组 检测 3份样品, 取其平均值。 比较 3组的测试结果。 实施例 1 固体分散体 1  Using the dissolution medium, the dissolution rates of the above three groups of samples were respectively measured according to a conventional method, and each sample was tested for 3 samples, and the average value thereof was taken. Compare the test results of the three groups. Example 1 Solid dispersion 1
式 II化合物与 PVP K30, 重量比例为 1 : 4, 溶剂(甲醇)法  Formula II compound and PVP K30, weight ratio of 1: 4, solvent (methanol) method
称取 1重量份的式 II化合物和 4重量份的 PVP K30置同一烧瓶中,烧瓶置 80°C油 浴中, 加入甲醇至烧瓶中, 密塞在冷凝水回流条件下搅拌, 直至化合物和 PVP K30 均溶清,再搅拌 30分钟。将溶清的液体用旋转蒸发器在 80°C时快速蒸干,得到固体。 将固体在 50°C真空干燥箱内干燥 24小时, 粉碎或研细, 即得固体分散体 1。 其 XRPD测试结果如图 1所示, 固体分散体 1中, 式 II化合物的衍射峰 (特征峰) 全部消失, 以无定型或分子形态存在。 实施例 2 固体分散体 2 Weigh 1 part by weight of the compound of formula II and 4 parts by weight of PVP K30 in the same flask, place the flask in an oil bath at 80 ° C, add methanol to the flask, and stir the plug under reflux of the condensed water until the compound and PVP K30 was dissolved and stirred for another 30 minutes. The dissolved liquid was quickly evaporated to dryness at 80 ° C using a rotary evaporator to give a solid. The solid was dried in a vacuum oven at 50 ° C for 24 hours, pulverized or ground to obtain a solid dispersion 1. The XRPD test results are shown in Fig. 1. In the solid dispersion 1, the diffraction peaks (characteristic peaks) of the compound of the formula II disappeared completely, and existed in an amorphous or molecular form. Example 2 Solid Dispersion 2
式 II化合物与 PVP K30, 重量比例为 1 : 3, 溶剂(甲醇)法  Formula II compound and PVP K30, weight ratio of 1: 3, solvent (methanol) method
制备方法同实施例 1, 不同点在于式 II化合物与 PVP K30的重量比例为 1 : 3。 其 XRPD测试结果如图 2所示, 固体分散体 2中, 式 II化合物的衍射峰 (特征峰) 全部消失, 以无定型或分子形态存在。 实施例 3 固体分散体 3  The preparation method was the same as in Example 1, except that the weight ratio of the compound of the formula II to the PVP K30 was 1:3. The XRPD test results are shown in Fig. 2. In the solid dispersion 2, the diffraction peaks (characteristic peaks) of the compound of the formula II disappeared completely, and existed in an amorphous or molecular form. Example 3 Solid dispersion 3
式 II化合物与 PVP K30, 重量比例为 1 : 2, 溶剂(甲醇)法  Formula II compound and PVP K30, weight ratio of 1: 2, solvent (methanol) method
制备方法同实施例 1, 不同点在于式 II化合物与 PVP K30的重量比例为 1 : 2。 其 XRPD测试结果如图 3所示, 固体分散体 3中, 式 II化合物的衍射峰 (特征峰) 全部消失, 以无定型或分子形态存在。 实施例 4 固体分散体 4  The preparation method was the same as in Example 1, except that the weight ratio of the compound of the formula II to the PVP K30 was 1:2. The XRPD test results are shown in Fig. 3. In the solid dispersion 3, the diffraction peaks (characteristic peaks) of the compound of the formula II disappeared completely, and existed in an amorphous or molecular form. Example 4 Solid dispersion 4
式 II化合物的对甲基苯磺酸盐与 PVP K30, 重量比例为 1 : 4, 溶剂(甲醇)法 制备方法同实施例 1, 不同点在于采用式 II化合物的对甲基苯磺酸盐代替式 II 化合物。  The p-toluenesulfonate of the compound of the formula II and the PVP K30 are in a weight ratio of 1:4, and the solvent (methanol) method is the same as in the first embodiment except that the p-toluenesulfonate of the compound of the formula II is used instead. a compound of formula II.
其 XRPD测试结果如图 4所示, 固体分散体 4中, 式 II化合物的对甲基苯磺酸 盐的衍射峰 (特征峰)全部消失, 以无定型或分子形态存在。 实施例 5 固体分散体 5  The XRPD test results are shown in Fig. 4. In the solid dispersion 4, the diffraction peaks (characteristic peaks) of the p-toluenesulfonate of the compound of the formula II disappeared, and existed in an amorphous or molecular form. Example 5 Solid dispersion 5
式 II化合物的对甲基苯磺酸盐与 PVP K30, 重量比例为 1 : 3, 溶剂(甲醇)法 制备方法同实施例 4, 不同点在于采用式 II化合物的对甲基苯磺酸盐和 PVP The p-toluenesulfonate of the compound of the formula II and the PVP K30 are in a weight ratio of 1:3, and the solvent (methanol) method is the same as in the example 4 except that the p-toluenesulfonate of the compound of the formula II is used. PVP
K30的重量比例为 1 : 3。 The weight ratio of K30 is 1:3.
其 XRPD测试结果如图 5所示, 固体分散体 5中, 式 II化合物的对甲基苯磺酸 盐的衍射峰 (特征峰)全部消失, 以无定型或分子形态存在。 实施例 6 固体分散体 6  The XRPD test results are shown in Fig. 5. In the solid dispersion 5, the diffraction peaks (characteristic peaks) of the p-toluenesulfonate of the compound of the formula II disappeared, and existed in an amorphous or molecular form. Example 6 Solid dispersion 6
式 II化合物与 PVP K30, 重量比例为 1 : 4, 溶剂 (无水乙醇)法  Formula II compound and PVP K30, weight ratio of 1: 4, solvent (anhydrous ethanol) method
制备方法同实施例 1, 不同点在于溶剂为无水乙醇。  The preparation method was the same as in Example 1, except that the solvent was absolute ethanol.
其 XRPD测试结果如图 6所示, 固体分散体 6中, 式 II化合物的衍射峰 (特征峰) 全部消失, 以无定型或分子形态存在。 实施例 7 固体分散体 7 式 II化合物的对甲基苯磺酸盐与 PVP K30, 重量比例为 1 : 4, 溶剂 (无水乙醇) 法 The XRPD test results are shown in Fig. 6. In the solid dispersion 6, the diffraction peaks (characteristic peaks) of the compound of the formula II disappeared completely, and existed in an amorphous or molecular form. Example 7 Solid Dispersion 7 The compound of formula II has p-toluenesulfonate and PVP K30 in a weight ratio of 1:4, solvent (anhydrous ethanol) method
制备方法同实施例 1, 不同点在于采用式 II化合物的对甲基苯磺酸盐代替式 II 化合物, 溶剂为无水乙醇。  The preparation method is the same as that of Example 1, except that the p-toluenesulfonate of the compound of the formula II is used instead of the compound of the formula II, and the solvent is anhydrous ethanol.
其 XRPD测试结果如图 7所示, 固体分散体 7中, 式 II化合物的对甲基苯磺酸 盐的衍射峰 (特征峰)全部消失, 以无定型或分子形态存在。 实施例 8 固体分散体 8  The XRPD test results are shown in Fig. 7. In the solid dispersion 7, the diffraction peaks (characteristic peaks) of the p-toluenesulfonate of the compound of the formula II disappeared, and existed in an amorphous or molecular form. Example 8 Solid dispersion 8
式 II化合物的对甲基苯磺酸盐与 PVP K30, 重量比例为 1 : 4, 溶剂 (异丙醇) 法  The compound of formula II has p-toluenesulfonate and PVP K30 in a weight ratio of 1:4, solvent (isopropyl alcohol) method
制备方法同实施例 1, 不同点在于采用式 II化合物的对甲基苯磺酸盐代替式 II 化合物, 溶剂为异丙醇。  The preparation method is the same as in Example 1, except that the p-toluenesulfonate of the compound of the formula II is used instead of the compound of the formula II, and the solvent is isopropanol.
其 XRPD测试结果如图 8所示, 固体分散体 8中, 式 II化合物的对甲基苯磺酸 盐的衍射峰 (特征峰)全部消失, 以无定型或分子形态存在。 实施例 9 固体分散体 9(喷雾干燥)  The XRPD test results are shown in Fig. 8. In the solid dispersion 8, the diffraction peaks (characteristic peaks) of the p-toluenesulfonate of the compound of the formula II disappeared, and existed in an amorphous or molecular form. Example 9 Solid Dispersion 9 (spray drying)
式 II化合物的对甲基苯磺酸盐与 PVP K30, 重量比例为 1 : 4, 溶剂 (无水乙醇) 法  The compound of formula II has p-toluenesulfonate and PVP K30 in a weight ratio of 1:4, solvent (anhydrous ethanol) method
将 1重量份式 II化合物的对甲基苯磺酸盐和 4重量份载体 PVP K30混合装入 三口烧瓶中, 烧瓶中加入定量 (式 II化合物的对甲基苯磺酸盐: 溶剂 =lg: 400ml) 的溶剂 (无水乙醇), 烧瓶密塞置 80°C油浴锅中在冷凝水回流条件下搅拌子搅拌 至式 II化合物的对甲基苯磺酸盐和载体全部溶清, 再中速搅拌 30分钟。  1 part by weight of p-toluenesulfonate of the compound of the formula II and 4 parts by weight of the carrier PVP K30 were mixed into a three-necked flask, and the flask was added with a quantitative amount (p-toluenesulfonate of the compound of the formula II: solvent = lg: 400ml) of solvent (anhydrous ethanol), the flask is placed in a 80 ° C oil bath, stirred under condensed water reflux conditions, and the mixture is stirred until the p-toluenesulfonate and the carrier of the compound of formula II are completely dissolved. Stir for 30 minutes.
将溶清的溶液用瑞士产 Buchi 209型喷雾干燥器喷雾干燥。 工艺参数如下: 进风温度 1 10°C〜120°C, 风机功率 55%〜65%, 蠕动泵功率 50%〜55%。 得到白 色均匀细小粉末。 将得到的粉末在 50°C真空干燥箱内真空干燥 24小时, 即得到 固体分散体 9粉末。  The lysed solution was spray dried with a Swiss-made Buchi Model 209 spray dryer. The process parameters are as follows: inlet air temperature 1 10 ° C ~ 120 ° C, fan power 55% ~ 65%, peristaltic pump power 50% ~ 55%. A white fine powder was obtained. The obtained powder was vacuum dried in a vacuum oven at 50 ° C for 24 hours to obtain a solid dispersion 9 powder.
其 XRPD测试结果如图 9所示, 固体分散体 9中, 式 II化合物的对甲基苯磺酸盐 的衍射峰 (特征峰)全部消失, 以无定型或分子形态存在。 实施例 10 固体分散体 10(喷雾干燥)  The XRPD test results are shown in Fig. 9. In the solid dispersion 9, the diffraction peaks (characteristic peaks) of the p-toluenesulfonate of the compound of the formula II disappeared and existed in an amorphous or molecular form. Example 10 Solid Dispersion 10 (spray drying)
式 II化合物的对甲基苯磺酸盐与 PVP K30, 重量比例为 1 : 4, 溶剂 (异丙醇) 法  The compound of formula II has p-toluenesulfonate and PVP K30 in a weight ratio of 1:4, solvent (isopropyl alcohol) method
制备方法同实施例 9, 不同点在于溶剂为异丙醇。  The preparation method was the same as in Example 9, except that the solvent was isopropyl alcohol.
其 XRPD测试结果如图 10所示, 固体分散体 10中, 式 II化合物的对甲基苯磺 酸盐的衍射峰 (特征峰)全部消失, 以无定型或分子形态存在。 实施例 11 固体分散体 11(喷雾干燥) The XRPD test results are shown in Fig. 10. In the solid dispersion 10, the diffraction peaks (characteristic peaks) of the p-toluenesulfonate of the compound of the formula II disappeared and existed in an amorphous or molecular form. Example 11 Solid Dispersion 11 (spray drying)
式 II化合物的对甲基苯磺酸盐与 PVP K30, 重量比例为 1 : 2, 溶剂 (异丙醇) 法  The compound of formula II has p-toluenesulfonate and PVP K30 in a weight ratio of 1:2, solvent (isopropyl alcohol) method
制备方法同实施例 9, 不同点在于采用式 II化合物的对甲基苯磺酸盐与 PVP The preparation method is the same as that of Example 9, except that p-toluenesulfonate and PVP of the compound of formula II are used.
K30的重量比例为 1 : 2, 溶剂为异丙醇。 The weight ratio of K30 is 1:2, and the solvent is isopropyl alcohol.
其 XRPD测试结果如图 11所示, 固体分散体 1 1中, 式 II化合物的对甲基苯磺 酸盐的衍射峰 (特征峰)全部消失, 以无定型或分子形态存在。 对实施例 9-11制备的固体分散体, 经检验可知:  The XRPD test results are shown in Fig. 11. In the solid dispersion 1, the diffraction peaks (characteristic peaks) of the p-toluenesulfonate of the compound of the formula II disappeared and existed in an amorphous or molecular form. The solid dispersion prepared in Examples 9-11 was tested and found to:
在活性成分与载体重量比例为 1 : 2的固体分散体中, API的百分含量是 33.97%;  In the solid dispersion of the active ingredient to the carrier in a weight ratio of 1: 2, the percentage of API is 33.97%;
在活性成分与载体重量比例为 1 : 4的固体分散体中 API的百分含量是 20.61-21.50%,显示,在用喷雾干燥法制备固体分散体的过程中, API没有损失。 实施例 12 固体分散体 1的溶出度测试  The percentage of API in the solid dispersion in which the active ingredient to carrier weight ratio was 1:4 was 20.61-21.50%, indicating that there was no loss in API during the preparation of the solid dispersion by spray drying. Example 12 Dissolution Test of Solid Dispersion 1
参考中国药典 2010版二部附录 XC溶出度测试法,量取 0.1%十二垸基硫酸钠 pH 4.5醋酸-醋酸钠缓冲液 900mL, 置于 lOOOmL溶出杯中, 温度保持 37±0.5°C。  Refer to the Chinese Pharmacopoeia 2010 edition two appendix XC dissolution test method, measure 0.1% sodium decyl sulfate pH 4.5 acetic acid-sodium acetate buffer 900mL, placed in lOOOOmL dissolution cup, the temperature is maintained at 37 ± 0.5 °C.
分别加入式 II化合物、 式 II化合物与 PVP K30的物理混合物 (式 II化合物和 PVP K30的重量分别为 50mg和 200mg)、 固体分散体 l(250mg)各 3份, 桨速为 100rpm, 分另 ll于 2min、 5min、 10min、 15min、 20min、 30min、 45min、 60min、 120min 取样 5mL, 用 0.45μιη 微孔滤膜过滤, 取澄清滤液稀释后测定药物浓度, 计算三 者的溶出度。  The physical mixture of the compound of the formula II, the compound of the formula II and the PVP K30 (the weight of the compound of the formula II and the PVP K30 are 50 mg and 200 mg, respectively) and the solid dispersion 1 (250 mg) are respectively added, and the paddle speed is 100 rpm, respectively. 5 mL was sampled at 2 min, 5 min, 10 min, 15 min, 20 min, 30 min, 45 min, 60 min, 120 min, filtered through a 0.45 μιη microporous membrane filter, and the clarified filtrate was diluted to determine the drug concentration, and the dissolution of the three was calculated.
结果如图 12所示, 式 II化合物在 lOmin时的溶出度 < 1%, 式 II化合物和 PVP K30的物理混合物的溶出度 < 3%, 固体分散体 1的溶出度为 76%, 以 PVP K30为 载体的固体分散体极大的改善了式 II化合物的溶出度。 实施例 13 固体分散体 1的体内药代试验  The results are shown in Figure 12. The dissolution of the compound of formula II at 10 min was < 1%, the dissolution of the physical mixture of the compound of formula II and PVP K30 was < 3%, and the dissolution of solid dispersion 1 was 76%, with PVP K30. The solid dispersion of the carrier greatly improves the dissolution of the compound of formula II. Example 13 In vivo pharmacokinetic test of solid dispersion 1
选取固体分散体 1与对应的 API (即式 II化合物)进行体内药代试验。  The solid dispersion 1 was selected for in vivo pharmacoassay with the corresponding API (i.e., the compound of formula II).
6只雄性 6周龄 Wistar大鼠, 体重 170 g左右, 分为 2组, 每组 3只。 动物饲养 于 IVC动物房内, 12小时明暗交替。 以硬木刨花作为垫料。 动物给予饲料和经 灭菌的自来水, 自由饮食。 在给药前禁食 16 h, 给药后 2 h恢复给食。 禁食过夜 后, 灌胃给予 4 mg/kg固体分散体 1或者对应的 API的 1%CMC悬液, 给药容量为 10 ml/kg o 于给药后 0.5、 1.0、 2.0、 4.0、 6.0、 8.0、 24、 48和 72 h经大鼠眼球后 静脉丛取静脉血 0.2 ml, 置肝素化试管中, 3500 rpm离心 lO min, 分离得到血浆 样品, -20°C保存。 Six male 6-week-old Wistar rats weighing approximately 170 g were divided into 2 groups of 3 animals each. Animals were housed in IVC animal rooms and alternated for 12 hours. Hardwood shavings are used as litter. Animals are given feed and sterilized tap water for free diet. Fasting for 16 h before administration and returning to food 2 h after administration. After fasting overnight, 4 mg/kg solid dispersion 1 or 1% CMC suspension of the corresponding API was administered orally, with a drug volume of 10 ml/kg o 0.5, 1.0, 2.0, 4.0, 6.0 after administration. At 8.0, 24, 48 and 72 h, 0.2 ml of venous blood was taken from the posterior venous plexus of rats, placed in heparinized tubes, centrifuged at 3500 rpm for 10 min, and plasma was separated. Sample, stored at -20 °C.
血浆样品用经过验证的 LC-MS/MS分析方法定量检测 API的含量。药代动力 学参数将基于每只大鼠在不同时间点的血药浓度进行计算。 参数见表 1。  Plasma samples were quantified for API content using a validated LC-MS/MS analytical method. The pharmacokinetic parameters will be calculated based on the plasma concentration of each rat at different time points. The parameters are shown in Table 1.
结果如表 1所示: 固体分散体 1的体内药代的 AUCINF―。 bs约为 API的 13.9倍。 The results are shown in Table 1: AUC INF of the in vivo pharmacogen of solid dispersion 1. Bs is about 13.9 times that of the API.
表 1 Table 1
Figure imgf000016_0001
实施例 14 固体分散体 12
Figure imgf000016_0001
Example 14 Solid Dispersion 12
式 I化合物与 PVPK30, 重量比例为 1: 4, 溶剂 (无水乙醇)法  Formula I compound and PVPK30, weight ratio 1: 4, solvent (anhydrous ethanol) method
制备方法同实施例 1, 不同点在于采用式 I化合物代替式 II化合物, 溶剂为无 水乙醇。  The preparation method was the same as in Example 1, except that the compound of the formula I was used instead of the compound of the formula II, and the solvent was anhydrous ethanol.
其 XRPD测试结果如图 13所示, 固体分散体 12中, 式 I化合物的衍射峰 (特征 峰)全部消失, 以无定型或分子形态存在。 实施例 15 固体分散体 13  The XRPD test results are shown in Fig. 13. In the solid dispersion 12, the diffraction peaks (characteristic peaks) of the compound of the formula I disappeared and existed in an amorphous or molecular form. Example 15 Solid dispersion 13
式 I化合物与 PVPK25, 重量比例为 1: 4, 溶剂 (无水乙醇)法  Formula I compound and PVPK25, weight ratio 1: 4, solvent (anhydrous ethanol) method
制备方法同实施例 14, 不同点在于采用载体 PVPK25代替载体 PVPK30。 实施例 16 固体分散体 14  The preparation method was the same as that of Example 14, except that the carrier PVPK25 was used instead of the carrier PVPK30. Example 16 Solid Dispersion 14
式 I化合物与 PVPK90, 重量比例为 1: 4, 溶剂 (无水乙醇)法  Formula I compound and PVPK90, weight ratio 1: 4, solvent (anhydrous ethanol) method
制备方法同实施例 14, 不同点在于采用载体 PVPK90代替载体 PVPK30。 实施例 17 固体分散体 12的溶出度比较测试  The preparation method was the same as that of Example 14, except that the carrier PVPK90 was used instead of the carrier PVPK30. Example 17 Comparative Test of Dissolution of Solid Dispersion 12
测试方法同实施例 12, 测试固体分散体 12(250mg)的溶出度。 结果表明: 固体分散体 12的在 30分钟时溶出度 80%, PVP K30为载体的 固体分散体均极大的改善了式 I化合物的溶出度,显著优于采用其他常用的载体 (如 PEG 6000、 羟丙甲纤维素 E5), 也优于 PVP K90和 K25。 实施例 18 固体分散体 12的体内药代试验 The test method was the same as in Example 12, and the dissolution rate of the solid dispersion 12 (250 mg) was tested. The results show that the solid dispersion 12 has a dissolution rate of 80% at 30 minutes, and the solid dispersion of PVP K30 as a carrier greatly improves the dissolution of the compound of formula I, which is significantly better than other commonly used carriers (such as PEG 6000). Hypromellose E5) is also superior to PVP K90 and K25. Example 18 In vivo pharmacokinetic test of solid dispersion 12
选取固体分散体 12与对应的 API (即式 I化合物)进行体内药代试验, 测试方法同实施例 13。  The solid dispersion 12 was selected for in vivo pharmacoassay with the corresponding API (i.e., the compound of formula I) in the same manner as in Example 13.
结果如表 2所示: 固体分散体 12的体内药代的 AUCINFbs约为 API的 7.3倍。 The results are shown in Table 2: AUC INF of the in vivo pharmacogen of solid dispersion 12. Bs is about 7.3 times that of the API.
表 2  Table 2
Figure imgf000017_0001
实施例 19 固体分散体 4无定形稳定性试验
Figure imgf000017_0001
Example 19 Solid dispersion 4 amorphous stability test
选取固体分散体 4进行加速稳定性试验 (40°C, 75% H): 1、 2、 3个月时均 取出进行 X-射线粉末衍射。  The solid dispersion 4 was selected for accelerated stability test (40 ° C, 75% H): taken at 1, 2, and 3 months for X-ray powder diffraction.
XRPD结果如图 14所示, 表明固体分散体 4经 1、 2、 3个月加速稳定性试验 后, 非常稳定 (无 API的 X-衍射特征峰, 且曲线基本相同), 均没有晶型生成 (即 原来的无定型或分子状态没有改变), 仍然是以无定形或分子状态存在的固体分 散体。  The XRPD results are shown in Figure 14, which indicates that the solid dispersion 4 is very stable after 1, 2, and 3 months accelerated stability test (no X-ray diffraction characteristic peak of API, and the curves are basically the same), and no crystal form is formed. (ie, the original amorphous or molecular state does not change), still a solid dispersion that exists in an amorphous or molecular state.
结果如图 14所示: 从上往下依次是式 II化合物对甲基苯磺酸盐、 新制备的 固体分散体 4、 固体分散体 4放置一个月时、 固体分散体 4放置两个月时、 固体 分散体 4放置三个月时的 XRPD图。 实施例 20 固体分散体 5无定形稳定性试验  The results are shown in Fig. 14. From the top to the bottom, the compound of the formula II is p-toluenesulfonate, the freshly prepared solid dispersion 4, the solid dispersion 4 is left for one month, and the solid dispersion 4 is placed for two months. The solid dispersion 4 was placed in an XRPD pattern at three months. Example 20 Solid dispersion 5 amorphous stability test
选取固体分散体 5进行加速稳定性试验 (40°C, 75% H): 1、 2、 3个月时均 取出进行 X-射线粉末衍射。 Solid dispersion 5 was selected for accelerated stability test (40 ° C, 75% H): 1, 2, 3 months The X-ray powder diffraction was taken out.
XRPD结果如图 15所示, 表明固体分散体 5经 1、 2、 3个月加速稳定性试验 后, 非常稳定 (无 API的 X-衍射特征峰, 且曲线基本相同), 均没有晶型生成 (即 原来的无定型或分子状态没有改变), 仍然是以无定形或分子状态存在的固体分 散体。  The XRPD results are shown in Figure 15, which indicates that the solid dispersion 5 is very stable after 1, 2, and 3 months accelerated stability test (no X-ray diffraction characteristic peak of API, and the curves are basically the same), and no crystal form is formed. (ie, the original amorphous or molecular state does not change), still a solid dispersion that exists in an amorphous or molecular state.
结果如图 15所示: 从上往下依次是式 II化合物对甲基苯磺酸盐、 新制备的 固体分散体 5、 固体分散体 5放置一个月时、 固体分散体 5放置两个月时、 固体 分散体 5放置三个月时的 XRPD图。 实施例 21 固体分散体 1无定形稳定性试验  The results are shown in Fig. 15 : from top to bottom, the compound of formula II is p-toluenesulfonate, freshly prepared solid dispersion 5, solid dispersion 5 is left for one month, and solid dispersion 5 is placed for two months. , XRPD pattern of solid dispersion 5 placed for three months. Example 21 Solid Dispersion 1 Amorphous Stability Test
选取固体分散体 1进行加速稳定性试验 (40°C, 75% H): 1、 2、 3个月时均 取出进行 X-射线粉末衍射。  The solid dispersion 1 was selected for accelerated stability test (40 ° C, 75% H): 1 , 2, 3 months were taken out for X-ray powder diffraction.
XRPD结果如图 16所示, 表明固体分散体 1经 1、 2、 3个月加速稳定性试验 后, 非常稳定 (无 API的 X-衍射特征峰, 且曲线基本相同), 均没有晶型生成 (即 原来的无定型或分子状态没有改变), 仍然是以无定形或分子状态存在的固体分 散体。  The XRPD results are shown in Fig. 16, which indicates that the solid dispersion 1 is very stable after the 1, 2, and 3 months accelerated stability test (no X-ray diffraction characteristic peak of the API, and the curves are basically the same), and no crystal form is formed. (ie, the original amorphous or molecular state does not change), still a solid dispersion that exists in an amorphous or molecular state.
结果如图 16所示: 从上往下依次是式 II化合物、 新制备的固体分散体 1、 固 体分散体 1放置一个月时、 固体分散体 1放置两个月时、 固体分散体 1放置三个 月时的 XRPD图。 实施例 22 固体分散体 3无定形稳定性试验  The results are shown in Fig. 16: From the top to the bottom, the compound of the formula II, the freshly prepared solid dispersion 1, the solid dispersion 1 was left for one month, the solid dispersion 1 was left for two months, and the solid dispersion 1 was placed three. XRPD chart at the time of month. Example 22 Solid dispersion 3 amorphous stability test
选取固体分散体 3进行加速稳定性试验 (40°C, 75% H): 1、 2、 3个月时均 取出进行 X-射线粉末衍射。  The solid dispersion 3 was selected for accelerated stability test (40 ° C, 75% H): taken at 1, 2, and 3 months for X-ray powder diffraction.
XRPD结果如图 17所示, 表明固体分散体 3经 1、 2、 3个月加速稳定性试验 后, 非常稳定 (无 API的 X-衍射特征峰, 且曲线基本相同), 均没有晶型生成 (即 原来的无定型或分子状态没有改变), 仍然是以无定形或分子状态存在的固体分 散体。  The XRPD results are shown in Figure 17, which indicates that the solid dispersion 3 is very stable after 1, 2, 3 months accelerated stability test (no X-ray diffraction characteristic peak of API, and the curves are basically the same), and no crystal form is formed. (ie, the original amorphous or molecular state does not change), still a solid dispersion that exists in an amorphous or molecular state.
结果如图 17所示: 从上往下依次是式 II化合物、 新制备的固体分散体 3、 固 体分散体 3放置一个月时、 固体分散体 3放置两个月时、 固体分散体 3放置三个 月时的 XRPD图。 实施例 23-27 固体分散体 15-19  The results are shown in Fig. 17. From the top to the bottom, the compound of the formula II, the freshly prepared solid dispersion 3, the solid dispersion 3 were left for one month, the solid dispersion 3 was left for two months, and the solid dispersion 3 was placed three. XRPD chart at the time of month. Example 23-27 Solid Dispersion 15-19
API为式 II化合物;  API is a compound of formula II;
载体为 PVP K30和其它水溶性载体的混合载体 (混合载体见表 3)。  The carrier is a mixed carrier of PVP K30 and other water-soluble carriers (see Table 3 for mixed carriers).
制备方法同实施例 1, 不同点在于混合载体采用如表 3所示的条件, 溶剂为 无水乙醇。 The preparation method was the same as that in Example 1, except that the mixed carrier was subjected to the conditions shown in Table 3, and the solvent was Anhydrous ethanol.
表 3  table 3
Figure imgf000019_0001
Figure imgf000019_0001
其 XRPD测试结果表明, 式 II化合物的衍射峰 (特征峰)全部消失, 以无定型或 分子形态存在。  The XRPD test results show that the diffraction peaks (characteristic peaks) of the compound of formula II disappear completely, and exist in an amorphous or molecular form.
固体分散体 15-19(依次对应实施例 23-27)的体内药代试验 (操作同前)表明: 固体分散体的体内药代的 AUCINF―。 bs约为 API的 7- 15倍。 结论: The in vivo pharmacokinetic test of solid dispersions 15-19 (in turn, corresponding to Examples 23-27) (operation as before) indicates: AUC INF of the in vivo pharmacophore of the solid dispersion. Bs is about 7-15 times the API. in conclusion:
1. 本发明制备的包含活性成分和含 PVP K30的载体的固体分散体非常稳 定。  1. The solid dispersion comprising the active ingredient and the PVP K30-containing carrier prepared according to the present invention is very stable.
2. 较活性成分而言, 本发明制备的固体分散体具有显著提高的溶出度, 体 内药代动力学参数好, 尤其是本发明所述固体分散体的药时曲线下面积是其活性 成分单独给药时的药时曲线下面积的 6-20倍, 较佳地 7-15倍。  2. Compared with the active ingredient, the solid dispersion prepared by the invention has significantly improved dissolution and good in vivo pharmacokinetic parameters, especially the area under the drug-time curve of the solid dispersion of the invention is its active ingredient alone. The area under the curve of the drug at the time of administration is 6-20 times, preferably 7-15 times.
3. 较式 I化合物的固体分散体而言,式 II化合物的固体分散体的药时曲线 下面积的改善程度更明显。式 II化合物的固体分散体的药时曲线下面积是活性 成分式 II化合物的 13.9倍,式 I化合物的固体分散体的药时曲线下面积是活性 成分式 I化合物的 7.3倍。 在本发明提及的所有文献都在本申请中引用作为参考, 就如同每一篇文献 被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后, 本领域技术人员可以对本发明作各种改动或修改, 这些等价形式同样落于本申 请所附权利要求书所限定的范围。  3. The solids dispersion of the compound of formula II has a more pronounced improvement in area under the drug time curve than the solid dispersion of the compound of formula I. The solids dispersion of the solid dispersion of the compound of formula II is 13.9 times the area of the active ingredient of the compound of formula II, and the solid dispersion of the solid dispersion of the compound of formula I is 7.3 times the area of the active ingredient of the compound of formula I. All documents mentioned in the present application are hereby incorporated by reference in their entirety in their entireties in the the the the the the the the the In addition, it should be understood that various modifications and changes may be made to the present invention, and the scope of the invention is defined by the scope of the appended claims.

Claims

权 利 要 求 Rights request
1. 一种固体分散体, 其特征在于, 包括: A solid dispersion characterized by comprising:
(A) 作为活性成分的式 A化合物或其衍生物、 或其药学上可接受的盐、 水合物 或溶剂合物,
Figure imgf000020_0001
式中, R为 CD3或 CH3 ; 以及
(A) a compound of the formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, as an active ingredient,
Figure imgf000020_0001
Wherein R is CD 3 or CH 3 ;
(B) 亲水性高分子载体;  (B) a hydrophilic polymer carrier;
其中, (A)和 (B)的重量比为 1 : 1-1:  Wherein the weight ratio of (A) and (B) is 1: 1-1:
2. 如权利要求 1所述的固体分散体, 其特征在于, 所述的亲水性高分子载体是 聚维酮或含聚维酮的高分子载体。  The solid dispersion according to claim 1, wherein the hydrophilic polymer carrier is povidone or a povidone-containing polymer carrier.
3. 如权利要求 1所述的固体分散体, 其特征在于, 所述的亲水性高分子载体是 聚维酮 K30或含聚维酮 K30的高分子载体, 其中, 聚维酮 K30的含量为 10-100wt%, 按高分子载体的总重量计。  The solid dispersion according to claim 1, wherein the hydrophilic polymer carrier is povidone K30 or a povidone K30-containing polymer carrier, wherein the povidone K30 content It is 10-100% by weight based on the total weight of the polymer carrier.
4. 如权利要求 1所述的固体分散体, 其特征在于, 所述活性成分为式 II所示的 化合物, 或其药
Figure imgf000020_0002
The solid dispersion according to claim 1, wherein the active ingredient is a compound represented by Formula II, or a drug thereof
Figure imgf000020_0002
5. 如权利要求 3所述的固体分散体, 其特征在于, 所述含聚维酮 K30的高分子 载体还包含选自下组的其他水溶性载体: 聚乙二醇类、 聚维酮类、 纤维素类、 表面 活性剂类、 或其组合;  The solid dispersion according to claim 3, wherein the povidone-containing K30-containing polymer carrier further comprises other water-soluble carriers selected from the group consisting of polyethylene glycols and povidones. , celluloses, surfactants, or combinations thereof;
其中,所述聚乙二醇类选自下组:聚乙二醇 4000、聚乙二醇 6000、聚乙二醇 8000 或其组合;  Wherein the polyethylene glycol is selected from the group consisting of polyethylene glycol 4000, polyethylene glycol 6000, polyethylene glycol 8000 or a combination thereof;
所述聚维酮类选自下组:聚维酮 K25、聚维酮 Κ90、聚维酮 S630、或其组合; 所述纤维素类选自下组: 羟丙甲纤维素 E5、 羟丙甲纤维素 E15、 羟丙纤维素 L、 或其组合;  The povidone is selected from the group consisting of povidone K25, povidone oxime 90, povidone S630, or a combination thereof; the cellulose is selected from the group consisting of hypromellose E5, hyprothenol Cellulose E15, hydroxypropyl cellulose L, or a combination thereof;
所述表面活性剂类选自下组: 泊洛沙姆 407、 泊洛沙姆 188、 或其组合。 The surfactant class is selected from the group consisting of poloxamer 407, poloxamer 188, or a combination thereof.
6. 如权利要求 1所述的固体分散体, 其特征在于, 所述 (A)和 (B)的重量比为 1 : 1-1: 20。 The solid dispersion according to claim 1, wherein the weight ratio of the (A) and (B) is 1:1-1:20.
7. 如权利要求 1所述的固体分散体, 其特征在于, 所述固体分散体的 X-射线粉 末衍射图中, 不具有所述固体分散体的活性成分的 X-射线衍射特征峰。 The solid dispersion according to claim 1, wherein the solid dispersion has X-ray powder In the final diffraction pattern, there is no X-ray diffraction characteristic peak of the active component of the solid dispersion.
8. 如权利要求 1所述的固体分散体, 其特征在于, 具有以下一个或多个特征: ① 所述固体分散体的药时曲线下面积是所述固体分散体的活性成分单独给药 时的药时曲线下面积的 6-20倍;  The solid dispersion according to claim 1, which has one or more of the following features: 1 The area under the drug-time curve of the solid dispersion is when the active ingredient of the solid dispersion is administered alone 6-20 times the area under the drug time curve;
② 所述固体分散体在 37°C, pH4.5的醋酸钠缓冲水溶液中 10分钟时累积溶出 度大于 70%;  2 The solid dispersion has a cumulative dissolution of more than 70% at 37 ° C, pH 4.5 in a sodium acetate buffered aqueous solution for 10 minutes;
③ 所述固体分散体在 37°C, pH4.5的醋酸钠缓冲水溶液中 120分钟时累积溶 出度是所述固体分散体的活性成分累积溶出度的 10-40倍。  3 The cumulative dispersion of the solid dispersion at 37 ° C in a pH 4.5 aqueous solution of sodium acetate buffer for 120 minutes is 10-40 times the cumulative dissolution of the active ingredient of the solid dispersion.
9. 一种权利要求 1所述固体分散体的制备方法, 其特征在于, 包括方法: (a)熔融法 1, 包括步骤:  9. A method of preparing a solid dispersion according to claim 1, comprising the method of: (a) melting method 1, comprising the steps of:
(al) 混合活性成分与亲水性高分子载体, 从而得到一含活性成分的混合物; (a2) 加热熔融步骤 (al)得到的含活性成分的混合物, 从而得到熔融后的混合 物; 和  (al) mixing the active ingredient with a hydrophilic polymer carrier to obtain a mixture containing the active ingredient; (a2) heating the molten component (al) to obtain the mixture containing the active ingredient, thereby obtaining a molten mixture;
(a3) 冷却步骤 (a2)得到的熔融后的混合物, 从而得到权利要求 1所述的固体分 散体;  (a3) cooling the molten mixture obtained in the step (a2) to obtain the solid dispersion according to claim 1;
 Or
(b)熔融法 2, 包括步骤:  (b) Melting method 2, including steps:
(bl) 加热熔融亲水性高分子载体, 从而得到熔融后的载体;  (bl) heating and melting the hydrophilic polymer carrier to obtain a molten carrier;
(b2) 将步骤 (bl)得到的熔融后的载体和活性成分混合后熔融,从而得到熔融后 的混合物; 和  (b2) mixing the molten carrier obtained in the step (bl) with the active ingredient and melting the mixture to obtain a molten mixture;
(b3) 冷却步骤 (b2)得到的熔融后的混合物, 从而得到权利要求 1所述的固体分 散体;  (b3) cooling the molten mixture obtained in the step (b2) to obtain the solid dispersion according to claim 1;
 Or
(c) 溶剂法, 包括步骤:  (c) Solvent method, including steps:
(cl) 将活性成分与亲水性高分子载体共同溶解于溶剂中, 从而得到一混合物; 禾口  (cl) co-dissolving the active ingredient and the hydrophilic polymer carrier in a solvent to obtain a mixture;
(c2) 除去步骤 (cl)得到的混合物中的溶剂, 从而得到权利要求 1所述的固体分 散体;  (c2) removing the solvent in the mixture obtained in the step (cl), thereby obtaining the solid dispersion according to claim 1;
其中, 所述活性成分为式 A化合物或其衍生物、 或其药学上可接受的盐、 水合 物或溶剂合物,
Figure imgf000021_0001
式中, R为 CD3或 CH3
Wherein the active ingredient is a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof,
Figure imgf000021_0001
Where R is CD 3 or CH 3 ,
所述亲水性高分子载体为含聚维酮 K30的亲水性高分子载体,  The hydrophilic polymer carrier is a hydrophilic polymer carrier containing povidone K30.
所述溶剂选自下组: 甲醇、 乙醇、 异丙醇、 丙酮、 二氯甲垸、 四氢呋喃, 或其 组合。  The solvent is selected from the group consisting of methanol, ethanol, isopropanol, acetone, dichloromethane, tetrahydrofuran, or a combination thereof.
10. 一种药物组合物, 其特征在于, 所述组合物包括:  10. A pharmaceutical composition, the composition comprising:
(1)权利要求 1-8任一项所述的固体分散体; 和  (1) The solid dispersion of any one of claims 1-8;
(2)药学上可接受的赋形剂。  (2) A pharmaceutically acceptable excipient.
11.一种如权利要求 10所述药物组合物的制备方法, 其特征在于, 将权利要求 1-8任一项所述的固体分散体和药学上可接受的赋形剂混合, 从而制得所述的药物 组合物。  A method for producing a pharmaceutical composition according to claim 10, which comprises mixing the solid dispersion according to any one of claims 1 to 8 and a pharmaceutically acceptable excipient. Said pharmaceutical composition.
12.一种如权利要求 1所述的固体分散体的用途, 其特征在于, 用于制备抑制 磷酸激酶 (如 raf敫酶)的药物; 或用于制备治疗肿瘤的药物。  12. Use of a solid dispersion according to claim 1 for the preparation of a medicament for inhibiting a phosphokinase (e.g., rafase); or for the preparation of a medicament for treating a tumor.
13.一种治疗方法, 其特征在于, 将 (i)权利要求 1-8任一项所述的固体分散体 或 (ii)权利要求 10所述的药物组合物, 施用于需要治疗的患者。  A method of treatment, which comprises applying (i) the solid dispersion according to any one of claims 1 to 8 or (ii) the pharmaceutical composition according to claim 10 to a patient in need of treatment.
PCT/CN2013/072643 2012-03-15 2013-03-14 Solid dispersion of improved adsorption performance and preparation thereof WO2013135187A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201210068698.1 2012-03-15
CN201210068698.1A CN103301067B (en) 2012-03-15 2012-03-15 It is a kind of improve absorbent properties solid dispersions and its preparation

Publications (1)

Publication Number Publication Date
WO2013135187A1 true WO2013135187A1 (en) 2013-09-19

Family

ID=49127078

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2013/072643 WO2013135187A1 (en) 2012-03-15 2013-03-14 Solid dispersion of improved adsorption performance and preparation thereof

Country Status (2)

Country Link
CN (1) CN103301067B (en)
WO (1) WO2013135187A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105348186B (en) * 2015-10-15 2018-05-22 青岛海洋生物医药研究院股份有限公司 Deuterated substituted bisarylurea compound and preparation method thereof and the application in anti-tumor drug is prepared
WO2023155182A1 (en) 2022-02-21 2023-08-24 北京睿创康泰医药研究院有限公司 Low-dose, high-exposure sorafenib or donafenib oral formulation and use thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101516335A (en) * 2006-07-21 2009-08-26 诺瓦提斯公司 Formulations for benzimidazolyl pyridyl ethers
WO2009106825A1 (en) * 2008-02-27 2009-09-03 Cipla Limited Polymorphs of sorafenib and salts thereof
CN101676266A (en) * 2008-09-19 2010-03-24 苏州泽璟生物制药有限公司 Deuterium-substituted omega-diphenylurea and its derivatives, as well as pharmaceutical composition containing the same

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000042012A1 (en) * 1999-01-13 2000-07-20 Bayer Corporation φ-CARBOXYARYL SUBSTITUTED DIPHENYL UREAS AS RAF KINASE INHIBITORS
CA2578442A1 (en) * 2004-08-27 2006-03-09 Bayer Pharmaceuticals Corporation Pharmaceutical compositions comprising 4{4-[3-(4-chloro-3-trifluoromethylphenyl)-ureido]-phenoxy}-pyridine-2-carboxylic acid methyl amide for the treatment of cancer
EP1920767A1 (en) * 2006-11-09 2008-05-14 Abbott GmbH & Co. KG Melt-processed imatinib dosage form

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101516335A (en) * 2006-07-21 2009-08-26 诺瓦提斯公司 Formulations for benzimidazolyl pyridyl ethers
WO2009106825A1 (en) * 2008-02-27 2009-09-03 Cipla Limited Polymorphs of sorafenib and salts thereof
CN101676266A (en) * 2008-09-19 2010-03-24 苏州泽璟生物制药有限公司 Deuterium-substituted omega-diphenylurea and its derivatives, as well as pharmaceutical composition containing the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ZOU, YANG ET AL.: "Progress of Preparation of Solid Dispersion", CHINESE JOURNAL OF PHARMACEUTICALS, 31 October 2005 (2005-10-31), pages 648 - 651 *

Also Published As

Publication number Publication date
CN103301067B (en) 2018-09-11
CN103301067A (en) 2013-09-18

Similar Documents

Publication Publication Date Title
US11890270B2 (en) Polymorphic forms of ST-246 and methods of preparation
WO2013135189A1 (en) Solid dispersion improving absorption performance and preparation of same
EA006777B1 (en) Pharmaceutical compositions of adsorbates of amorphous drug
KR20160124907A (en) Pharmaceutical compositions of therapeutically active compounds
CN108472291A (en) Combination treatment for treating malignant tumour
WO2015145145A1 (en) Pharmaceutical composition comprising lapatinib
WO2017054741A1 (en) Preparation for improving bioavailability of sorafenib
WO2013097003A1 (en) Solid pharmaceutical composition comprising an antibiotic from the quinolone family and method for the production thereof
WO2011079766A1 (en) Aripiprazole solid preparation and the production method thereof
WO2013135187A1 (en) Solid dispersion of improved adsorption performance and preparation thereof
WO2015145157A1 (en) Pharmaceutical composition comprising pazopanib
JP7058104B2 (en) Pharmaceutical tablets containing aprepitant as an active ingredient
US20210205281A1 (en) Therapy for treating malignancies
US20220241208A1 (en) Pharmaceutical compositions for the treatment of hbv
KR101136075B1 (en) Pharmaceutical composition for treating hypertension
CN116407554A (en) Medicine for reducing intestinal injury caused by targeted antitumor medicine and application thereof
CN115554292A (en) Pharmaceutical compositions and methods for reducing blood glucose and treating conditions associated with glucagon
CN116407549A (en) Cardiovascular and cerebrovascular medicine and its application
AU2017202359A1 (en) Polymorphic forms st-246 and methods of preparation

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13760597

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13760597

Country of ref document: EP

Kind code of ref document: A1