WO2013135189A1 - Solid dispersion improving absorption performance and preparation of same - Google Patents

Solid dispersion improving absorption performance and preparation of same Download PDF

Info

Publication number
WO2013135189A1
WO2013135189A1 PCT/CN2013/072645 CN2013072645W WO2013135189A1 WO 2013135189 A1 WO2013135189 A1 WO 2013135189A1 CN 2013072645 W CN2013072645 W CN 2013072645W WO 2013135189 A1 WO2013135189 A1 WO 2013135189A1
Authority
WO
WIPO (PCT)
Prior art keywords
solid dispersion
active ingredient
povidone
compound
polymer carrier
Prior art date
Application number
PCT/CN2013/072645
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 WO2013135189A1 publication Critical patent/WO2013135189A1/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, in particular to a solid dispersion for improving absorption properties and its preparation. Background technique
  • the compound is a colored powder which is odorless and tasteless. Very soluble in dimethyl sulfoxide or dimethylformamide, slightly soluble in tetrahydrofuran, methanol, slightly soluble in acetone, absolute ethanol or glacial acetic acid, but almost insoluble in water.
  • the compound of formula I is extremely poor in water solubility, it is a poorly soluble drug, has a low solubility and dissolution rate in physiological body fluids, and has a low dissolution rate, which ultimately leads to low bioavailability and seriously affects mammalian absorption of the compound of formula I. 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
  • An object of the present invention is to provide a solid dispersion containing a hydrazine compound or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof as an active ingredient, which is remarkably improved in bioavailability.
  • 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, including
  • 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 I, or a pharmaceutically acceptable salt, hydrate or solvent thereof
  • 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: 9.
  • 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 has a cumulative dissolution of more than 70%, preferably 75-95%, at 37 ° C in a pH 4.5 aqueous solution of sodium acetate buffer for 10 minutes;
  • the cumulative dissolution of the solid dispersion at 120 ° C in a pH 4.5 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, Where 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, dichloromethane, tetrahydrofuran, or a combination thereof.
  • 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 the dissolution profile of the compound of formula I, the physical mixture of the compound of formula I with PVP K30 and solid dispersion 1.
  • Figure 6 shows the dissolution profile of the compound of formula I, the physical mixture of the compound of formula I with PVP K30 and solid dispersion 2.
  • Figure 7 shows an XRPD pattern of solid dispersion 10.
  • Figure 8 shows the dissolution profile of the solid dispersion 10.
  • Figure 9 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 9. From the top to the bottom, the compound of the formula I, the newly prepared solid dispersion 1, solid XRPD pattern when Dispersion 1 was left for one month, solid dispersion 1 was placed for two months, and solid dispersion 1 was placed for three months.
  • Figure 10 shows the XRPD comparison chart of the solid dispersion 2 after 1, 2, 3 months accelerated stability test, and the results are shown in Figure 10: from top to bottom are the compound of formula I, freshly prepared solid dispersion 2, solid XRPD pattern when Dispersion 2 was placed for one month, Solid Dispersion 2 was placed for two months, and Solid Dispersion 2 was placed for three months.
  • Figure 11 shows the XRPD comparison chart of the solid dispersion 3 after 1, 2, 3 months accelerated stability test, and the results are shown in Figure 11. From the top to the bottom, the compound of the formula I, the newly prepared solid dispersion 3 XRPD pattern when solid dispersion 3 was left for one month, solid dispersion 3 was placed for two months, and solid dispersion 3 was placed for three months.
  • Figure 12 shows a comparison of XRPD of the solid dispersion 4 after 1, 2, 3 months accelerated stability test, and the results are shown in Figure 12: from top to bottom are the compound of formula I, freshly prepared solid dispersion 4, solid The XRPD pattern at which the dispersion 4 was left for one month, the solid dispersion 4 was left for two months, and the solid dispersion 4 was placed 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, hydrate or solvate thereof, wherein R is CD 3 or CH 3 ;
  • the term also encompasses various crystalline forms, hydrates or solvates of the compounds of formula A or derivatives thereof.
  • the compound of formula A refers to a compound of formula I or a compound of formula II, or a pharmaceutically acceptable salt thereof.
  • 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 for the solid dispersion the most commonly used are water-soluble carriers, poorly soluble carriers, and enteric carriers.
  • the “carrier” according to the present invention is a hydrophilic polymer carrier, preferably a povidone or a povidone-containing polymer carrier, or a povidone K30 or a povidone K30-containing polymer carrier, wherein The ketene K30 is contained in an amount of 10 to 100% by weight based on the total weight of the polymer carrier. More preferably, the povidone K30-containing polymer carrier used in the present invention refers to a carrier in which the povidone K30 has a weight content exceeding 20% by weight of the carrier; or the povidone K30 has a weight content exceeding 40% of the total weight of the carrier. The carrier; or the carrier of the povidone K30 in an amount of more than 60% by weight based on the total weight of the carrier.
  • water soluble carriers may also be included, including, for example, but not limited to: polyethylene glycols, povidones, cellulosics, surfactants, mannitol, or combinations thereof.
  • the polyethylene glycols are selected from the group consisting of polyethylene glycol 4000 (PEG 4000), polyethylene glycol 6000 (PEG 6000), polyethylene glycol 8000 (PEG 8000), or combinations thereof;
  • 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
  • HPMC E15 hydroxypropyl cellulose 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.
  • the weight ratio of the (A) and (B) is 1:9-1:2.
  • 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, or a surface dispersion method.
  • the preparation method is a melting method, or a solvent method.
  • the melting method is a method in which (A) an active ingredient, and (B) a hydrophilic polymer carrier are uniformly mixed, heated to a melt in a water bath or an oil bath, and then cooled to a solid, and pulverized to obtain a powdery form.
  • 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 pulverized to obtain the active ingredient in a powder form.
  • the solvent method is to dissolve the (A) active ingredient and (B) the hydrophilic polymer carrier in a solvent, and after the mixture is uniform, the solvent in the solution is removed, so that the active ingredient and the hydrophilicity are high.
  • the molecular 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.
  • Some carriers have strong diffraction peaks.
  • PEG4000 and PEG6000 have strong diffraction peaks at 19.20 ⁇ 0.1 ° and 23.30 ⁇ 0.1 °. ? 010 & 11 ⁇ " 407 has strong diffraction peaks at 19.16 ⁇ 0.1 ° and 23.33 ⁇ 0.1 °.
  • Some carriers have no strong diffraction peaks.
  • PVP K30, HPMC E5, and HPC-L have no strong diffraction peaks.
  • 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 solid dispersion of the present invention and a pharmaceutical composition containing the solid dispersion of the present invention as a main component can be used for the treatment, prevention, and alleviation of the phosphoric acid Kinases (Kinase M are listed as raft chymase mediated diseases.
  • the compounds of the invention are useful in the treatment of the following diseases: cancer, cardiovascular disease, obesity, diabetes and the like.
  • 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 it is meant that the amount of the solid dispersion of the present invention 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.), gelatin, talc, solid lubricants (such as hard Fatty 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.
  • gelatin such as talc
  • solid lubricants such as hard Fatty acid, magnesium stearate
  • 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 dispersions of the invention are admixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or mixed with the following ingredients: (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 gum arabic; (c) humectants, for example, glycerin; (d) disintegrants, for example, agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, cross-linked carboxymethyl Cellulose sodium and sodium carbonate; (e) slow solvent, such as stone (f) an absorption accelerator,
  • 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, syrups 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, dimethyl
  • 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 dispersion is 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
  • Example 5-9 Solid Dispersion 5-9
  • the preparation method is the same as that of the example 1, and the specific conditions are shown in Table 1.
  • a physical mixture of a compound of formula I, a compound of formula I and PVP K30 (compound of formula I and PVP) K30 weighs 50mg and 450mg), solid dispersion l (500mg), paddle speed is 100rpm, sample 5mL at 2min, 5min, 10min, 15min, 20min, 30min, 45min, 60min, 120miri, with 0.45 ⁇ microporous The membrane was filtered, and the clarified filtrate was diluted to determine the drug concentration, and the dissolution of the three was calculated.
  • the dissolution test method was the same as in Example 10.
  • the dissolution of the compound of formula I is ⁇ 5%
  • the physical mixture of the compound of formula I and the carrier is slightly better eluted than the compound of formula I, but ⁇ 10%
  • the dissolution of the solid dispersion of the compound of formula I with the carrier is about 80%. .
  • the solid dispersion 2 was selected for in vivo pharmacoassay with the corresponding API (i.e., the compound of formula I).
  • 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 samples were separated and 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. See Table 2 for parameters.
  • the preparation method is the same as that in Example 1, except that the compound of the formula II is substituted for the compound of the formula I, and the compound of the formula II is
  • the PVP K30 has a weight ratio of 1:4.
  • the dissolution test method was the same as in Example 10.
  • sample was configured in 2 parts (sample 1 and sample 2, respectively).
  • the respective dissolution rates were tested as described above and the average value (i.e., average) was calculated.
  • the solid dispersion 10 is selected for in vivo pharmacoassay with the corresponding API (i.e., the compound of formula II).
  • test method is the same as in the embodiment 12.
  • the solid dispersion 1 was selected for accelerated stability test (40 ° C, 75% H), and taken out at 1, 2, and 3 months for X-ray powder diffraction.
  • the solid dispersion 2 was selected for accelerated stability test (40 ° C, 75% H), and taken out at 1, 2, and 3 months for X-ray powder diffraction.
  • the solid dispersion 3 was selected for accelerated stability test (40 ° C, 75% H), and taken out at 1, 2, and 3 months for X-ray powder diffraction.
  • Fig. 11 The XRPD results are shown in Fig. 11. It shows 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 there is no crystal form.
  • the formation i.e., the original amorphous or molecular state is unchanged) remains a solid dispersion that exists in an amorphous or molecular state.
  • the solid dispersion 4 was selected for accelerated stability test (40 ° C, 75% H), and taken out at 1, 2, and 3 months for X-ray powder diffraction.
  • the preparation method was the same as in Example 1, except that the mixed carrier was subjected to the conditions shown in Table 4.
  • the XRPD test results show that in the solid dispersion, the diffraction peaks (characteristic peaks) of the compound of formula I are all in an amorphous form.
  • the solid dispersion comprising the active ingredient and the PVP K30-containing carrier prepared by the present invention is very stable; and the solid dispersion of the present invention has significantly improved dissolution compared to the active ingredient, especially the solid dispersion of the present invention.
  • the area under the curve of the body is 6-20 times, preferably 7-15 times, more preferably 9-14 times the area under the curve of the drug when the active ingredient is administered alone.

Abstract

Provided are a solid dispersion improving absorption performance and a method for preparing same. Specifically, a solid dispersion is provided, comprising (A): a compound of Formula A as an active ingredient or a derivant thereof, or a pharmaceutically acceptable salt, a hydrate or a solvate thereof; and (B): a hydrophilic high polymer carrier. A weight ratio of (A) to (B) ranges from 1:1 to 1:40. The solid dispersion according to the present invention can significantly improve a vitro dissolution of the active ingredient and remarkably improve absorption of a mammal for the active ingredient, thereby effectively reducing the dosage thereof in use.

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, in particular to a solid dispersion for improving absorption properties and its preparation. Background technique
4-(4-(3-(4-氯 -3- (三氟甲基)苯基]酰脲) -3-氟-苯氧基) -2-( N-l,,r,r-三氘代甲基) 吡啶酰胺, 即式 I化合物, 所述化合物或其药学上可接受的盐对磷酸激酶 (Kinase) 例如 raf^酶具有优 病。
Figure imgf000002_0001
4-(4-(3-(4-chloro-3-(trifluoromethyl)phenyl) ureide)-3-fluoro-phenoxy)-2-(Nl,,r,r-triindole Methyl)pyridine amide, i.e., a compound of formula I, which is superior to a phosphate kinase (Kinase) such as raf^ enzyme.
Figure imgf000002_0001
该化合物为有色粉末, 无臭无味。 在二甲亚砜或二甲基甲酰胺中极易溶解, 在四氢呋喃、 甲醇中略溶, 在丙酮、 无水乙醇或冰醋酸中微溶, 但是在水中几乎 不溶。  The compound is a colored powder which is odorless and tasteless. Very soluble in dimethyl sulfoxide or dimethylformamide, slightly soluble in tetrahydrofuran, methanol, slightly soluble in acetone, absolute ethanol or glacial acetic acid, but almost insoluble in water.
由于式 I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, 严重影响哺乳动物对式 I化合 物的吸收, 不利于充分发挥该化合物的疾病治疗效果。  Because the compound of formula I is extremely poor in water solubility, it is a poorly soluble drug, has a low solubility and dissolution rate in physiological body fluids, and has a low dissolution rate, which ultimately leads to low bioavailability and seriously affects mammalian absorption of the compound of formula I. 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 K3o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, CN101632630A in terms of more probucol, probucol, and PVP K 3 o of the solid dispersion, the bioavailability was only 5.8 fold increased relative dogs.
因此, 开发一种生物利用度显著提高的式 I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 I with a markedly improved bioavailability is necessary to address 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 Α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 An object of the present invention is to provide a solid dispersion containing a hydrazine compound or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof as an active ingredient, which is remarkably improved in bioavailability.
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, including
(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
Figure imgf000003_0001
(A) a compound of the formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, as an active ingredient, and
Figure imgf000003_0001
式中, R为 CD3或 CH3 ; 以及 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 I所示的化合物, 或其药学上可接受的盐、 水合物或溶剂
Figure imgf000003_0002
In another preferred embodiment, the active ingredient is a compound of formula I, or a pharmaceutically acceptable salt, hydrate or solvent thereof
Figure imgf000003_0002
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: 9。  In another preferred embodiment, the weight ratio of the (A) and (B) is 1: 2-1: 9.
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: 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; In another preferred embodiment, there are one or more of the following features: 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 has a cumulative dissolution of more than 70%, preferably 75-95%, at 37 ° C in a pH 4.5 aqueous solution of sodium acetate buffer 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, 较佳地为 15-30倍。  3 The cumulative dissolution of the solid dispersion at 120 ° C in a pH 4.5 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,
Figure imgf000004_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 imgf000004_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.
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)如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。 图 4显示了固体分散体 4的 XRPD图。 Figure 3 shows an XRPD pattern of Solid Dispersion 3. Figure 4 shows an XRPD pattern of Solid Dispersion 4.
图 5显示了式 I化合物、 式 I化合物与 PVP K30的物理混合物和固体分散体 1 的溶出曲线。  Figure 5 shows the dissolution profile of the compound of formula I, the physical mixture of the compound of formula I with PVP K30 and solid dispersion 1.
图 6显示了式 I化合物、 式 I化合物与 PVP K30的物理混合物和固体分散体 2 的溶出曲线。  Figure 6 shows the dissolution profile of the compound of formula I, the physical mixture of the compound of formula I with PVP K30 and solid dispersion 2.
图 7显示了固体分散体 10的 XRPD图。  Figure 7 shows an XRPD pattern of solid dispersion 10.
图 8显示了固体分散体 10的溶出曲线。  Figure 8 shows the dissolution profile of the solid dispersion 10.
图 9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 果图 9所示: 从上往下依次是式 I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 9 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 9. From the top to the bottom, the compound of the formula I, the newly prepared solid dispersion 1, solid XRPD pattern when Dispersion 1 was left for one month, solid dispersion 1 was placed for two months, and solid dispersion 1 was placed for three months.
图 10显示了固体分散体 2经 1、 2、 3个月加速稳定性试验后 XRPD对比图, 结 果图 10所示: 从上往下依次是式 I化合物、 新制备的固体分散体 2、 固体分散体 2 放置一个月时、固体分散体 2放置两个月时、固体分散体 2放置三个月时的 XRPD 图。  Figure 10 shows the XRPD comparison chart of the solid dispersion 2 after 1, 2, 3 months accelerated stability test, and the results are shown in Figure 10: from top to bottom are the compound of formula I, freshly prepared solid dispersion 2, solid XRPD pattern when Dispersion 2 was placed for one month, Solid Dispersion 2 was placed for two months, and Solid Dispersion 2 was placed for three months.
图 1 1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 果图 1 1所示: 从上往下依次是式 I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。  Figure 11 shows the XRPD comparison chart of the solid dispersion 3 after 1, 2, 3 months accelerated stability test, and the results are shown in Figure 11. From the top to the bottom, the compound of the formula I, the newly prepared solid dispersion 3 XRPD pattern when solid dispersion 3 was left for one month, solid dispersion 3 was placed for two months, and solid dispersion 3 was placed for three months.
图 12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 果图 12所示: 从上往下依次是式 I化合物、 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。 具体实施方式  Figure 12 shows a comparison of XRPD of the solid dispersion 4 after 1, 2, 3 months accelerated stability test, and the results are shown in Figure 12: from top to bottom are the compound of formula I, freshly prepared solid dispersion 4, solid The XRPD pattern at which the dispersion 4 was left for one month, the solid dispersion 4 was left for two months, and the solid dispersion 4 was placed 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, 极其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 extremely 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"是指式 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, 式中, R为 CD3或 CH3 ; The term "active ingredient" as used herein refers to a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, wherein R is CD 3 or CH 3 ;
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 物。  The term also encompasses various crystalline forms, hydrates or solvates of the compounds of formula A or derivatives thereof.
优选地, 所述式 A化合物指式 I化合物或式 II化合物、 或其药学上可接受的 盐。  Preferably, the compound of formula A refers to a compound of formula I or a compound of formula II, or a pharmaceutically acceptable salt thereof.
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 载体。  As the carrier for the solid dispersion, the most commonly used are water-soluble carriers, poorly soluble carriers, and enteric carriers.
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。 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。 The "carrier" according to the present invention is a hydrophilic polymer carrier, preferably a povidone or a povidone-containing polymer carrier, or a povidone K30 or a povidone K30-containing polymer carrier, wherein The ketene K30 is contained in an amount of 10 to 100% by weight based on the total weight of the polymer carrier. More preferably, the povidone K30-containing polymer carrier used in the present invention refers to a carrier in which the povidone K30 has a weight content exceeding 20% by weight of the carrier; or the povidone K30 has a weight content exceeding 40% of the total weight of the carrier. The carrier; or the carrier of the povidone K30 in an amount of more than 60% by weight based on the total weight of the carrier.
当然, 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、 甘露醇、 或其组合。  Of course, other water soluble carriers may also be included, including, for example, but not limited to: polyethylene glycols, povidones, cellulosics, surfactants, mannitol, 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 6000)、 聚乙二醇 8000(PEG 8000)或其组合;  The polyethylene glycols are selected from the group consisting of polyethylene glycol 4000 (PEG 4000), polyethylene glycol 6000 (PEG 6000), polyethylene glycol 8000 (PEG 8000), or combinations 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 The cellulose is selected from the group consisting of hypromellose E5 (HPMC E5), hypromellose
E15(HPMC E15)、 羟丙纤维素 L(HPC-L)、 或其组合; E15 (HPMC E15), hydroxypropyl cellulose 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 : 9-1: 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 1:9-1:2.
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、 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, or a surface dispersion method. Preferably, the preparation method is a melting 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。 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 Usually, the melting method is a method in which (A) an active ingredient, and (B) a hydrophilic polymer carrier are uniformly mixed, heated to a melt in a water bath or an oil bath, and then cooled to a solid, and pulverized to obtain a powdery form. A solid dispersion of the active ingredient. 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 pulverized to obtain the active ingredient in a powder form. Solid dispersion. 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 to dissolve the (A) active ingredient and (B) the hydrophilic polymer carrier in a solvent, and after the mixture is uniform, the solvent in the solution is removed, so that the active ingredient and the hydrophilicity are high. The molecular 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.
式 I化合物, 其在 7·24±0· 、 13·38±0· 、 14·53±0· 、 14·74±0· 、 15·61±0· 、 17·23±0·1ο、 18·57±0·1ο、 18·84±0·1ο、 19·81±0·1ο、 20·01±0·1ο、 23·42±0·1ο、 24·24±0·1ο、 25·85±0·1。、 26·39±0·1。、 27.43±0. 有强衍射峰。 a compound of formula I, which is at 7.24±0·, 13·38±0·, 14·53±0·, 14·74±0·, 15.61±0·, 17·23±0·1 ο , 18·57±0·1 ο , 18·84±0·1 ο , 19·81±0·1 ο , 20·01±0·1 ο , 23·42±0·1 ο , 24·24±0 ·1 ο , 25·85±0·1. , 26·39±0·1. , 27.43 ± 0. There are strong diffraction peaks.
式 II化合物, 其在 7·20±0·1ο、 13·34±0·1ο、 14·49±0·1ο、 14·72±0·1ο、 15·55±0·1。、 16·44±0·1ο、 17·17±0·1ο、 18.53±0.1。、 18·81±0·1ο、 19·76±0·1。、 19·97±0·1。、 23.41±0.1°、 23·75±0·1。、 24·22±0·1。、 24·75±0·1。、 25·90±0·1。、 26·35±0·1。、 27·42±0·1。、 31·54±0·1。 有强衍射峰。 载体的特征峰 a compound of formula II, which is at 7.20 ±0·1 ο , 13·34±0·1 ο , 14·49±0·1 ο , 14·72±0·1 ο , 15·55±0·1. , 16·44±0·1 ο , 17·17±0·1 ο , 18.53±0.1. , 18·81±0·1 ο , 19·76±0·1. , 19.97±0·1. , 23.41 ± 0.1 °, 23 · 75 ± 0 · 1. , 24·22±0·1. , 24·75±0·1. , 25·90±0·1. , 26·35±0·1. , 27.42±0·1. , 31·54±0·1. There are strong diffraction peaks. Characteristic peak of the carrier
有些载体有强衍射峰, 例如, PEG4000和 PEG6000在 19.20±0.1°、 23.30±0.1°有 强衍射峰。?010 &11^" 407在19.16±0.1°、 23.33±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 19.20 ± 0.1 ° and 23.30 ± 0.1 °. ? 010 & 11^" 407 has strong diffraction peaks at 19.16 ± 0.1 ° and 23.33 ± 0.1 °. Some carriers have no strong diffraction peaks. For example, PVP K30, HPMC E5, and HPC-L have no strong diffraction peaks. Solid dispersions Characteristic peaks
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.
由于本发明化合物具有优异的对磷酸激酶 (Kinase)例如 raf激酶的抑制活 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 present invention has excellent inhibitory activity against a kinase such as ratase, the solid dispersion of the present invention and a pharmaceutical composition containing the solid dispersion of the present invention as a main component can be used for the treatment, prevention, and alleviation of the phosphoric acid Kinases (Kinase M are listed as raft chymase mediated diseases. According to the prior art, the compounds of the invention are useful in the treatment of the following diseases: cancer, cardiovascular disease, obesity, diabetes and the like.
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: 本发 明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" it is meant that the amount of the solid dispersion of the present invention is sufficient to significantly improve the condition without causing serious side effects. Generally, 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)、 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.), gelatin, talc, solid lubricants (such as hard Fatty 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, 本发明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 硫酸钠和单硬脂酸甘油酯; (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 dispersions of the invention are admixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or mixed with the following ingredients: (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 gum arabic; (c) humectants, for example, glycerin; (d) disintegrants, for example, agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, cross-linked carboxymethyl Cellulose sodium and sodium carbonate; (e) slow solvent, such as stone (f) an absorption accelerator, for example, a quaternary amine compound; (g) a wetting agent such as cetyl alcohol, sodium dodecyl sulfate and glyceryl monostearate; (h) an adsorbent, for example, kaolin And (i) a lubricant, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium dodecyl sulfate, or a mixture 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、 糖浆 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, syrups 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。 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 dispersion is 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的固体分散体,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 I化合物与 PVP K30, 重量比例为 1 : 9, 溶剂 (无水乙醇)法  The compound of formula I and PVP K30, the weight ratio is 1: 9, solvent (anhydrous ethanol) method
称取 1重量份的式 I化合物和 9重量份的 PVP K30置同一烧瓶中, 烧瓶置 80°C油 浴中, 加入无水乙醇至烧瓶中, 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, 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 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 2 固体分散体 2 Weigh 1 part by weight of the compound of formula I and 9 parts by weight of PVP K30 in the same flask, place the flask in an oil bath at 80 ° C, add absolute ethanol to the flask, and stir the plug under reflux of the condensed water until the compound Both were dissolved in PVP K30 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, and finely 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 I disappeared completely, and existed in an amorphous or molecular form. Example 2 Solid Dispersion 2
式 I化合物与 PVP K30, 重量比例为 1 : 4, 溶剂 (无水乙醇)法  Formula I compound and PVP K30, weight ratio of 1: 4, solvent (anhydrous ethanol) method
制备方法同实施例 1, 不同点在于式 I化合物与 PVP K30的重量比例为 1 : 4。 其 XRPD测试结果如图 2所示, 固体分散体 2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 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 I to the PVP K30 was 1:4. 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 I disappeared completely, and existed in an amorphous or molecular form. Example 3 Solid dispersion 3
式 I化合物与 PVP K30, 重量比例为 1 : 3, 溶剂 (无水乙醇)法  Formula I compound and PVP K30, weight ratio of 1: 3, solvent (anhydrous ethanol) method
制备方法同实施例 1, 不同点在于式 I化合物与 PVP K30的重量比例为 1 : 3。 其 XRPD测试结果如图 3所示, 固体分散体 3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 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 I to the PVP K30 was 1:3. 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 I disappeared completely, and existed in an amorphous or molecular form. Example 4 Solid dispersion 4
式 I化合物与 PVP K30, 重量比例为 1 : 2, 溶剂 (无水乙醇)法  Formula I compound and PVP K30, weight ratio of 1: 2, solvent (anhydrous ethanol) method
制备方法同实施例 1, 不同点在于式 I化合物与 PVP K30的重量比例为 1 : 2。 其 XRPD测试结果如图 4所示, 固体分散体 4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 5-9 固体分散体 5-9  The preparation method was the same as in Example 1, except that the weight ratio of the compound of the formula I to the PVP K30 was 1:2. The XRPD test results are shown in Fig. 4. In the solid dispersion 4, the diffraction peaks (characteristic peaks) of the compound of the formula I disappeared completely, and existed in an amorphous or molecular form. Example 5-9 Solid Dispersion 5-9
制备方法同实施例 1, 具体条件见表 1。  The preparation method is the same as that of the example 1, and the specific conditions are shown in Table 1.
表 1  Table 1
Figure imgf000013_0001
Figure imgf000013_0001
实施例 10 固体分散体 1的溶出度测试  Example 10 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 I化合物、 式 I化合物和 PVP K30的物理混合物 (式 I化合物和 PVP K30的重量分别为 50mg和 450mg)、 固体分散体 l(500mg), 桨速为 100rpm, 分别 于 2min、 5min、 10min、 15min、 20min、 30min、 45min、 60min、 120miri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。 A physical mixture of a compound of formula I, a compound of formula I and PVP K30 (compound of formula I and PVP) K30 weighs 50mg and 450mg), solid dispersion l (500mg), paddle speed is 100rpm, sample 5mL at 2min, 5min, 10min, 15min, 20min, 30min, 45min, 60min, 120miri, with 0.45μιη microporous The membrane was filtered, and the clarified filtrate was diluted to determine the drug concentration, and the dissolution of the three was calculated.
结果如图 5所示,式 I化合物在 lOmin时的溶出度 <2%,式 I化合物和 PVP K30 的物理混合物的溶出度 < 5%, 固体分散体 1的溶出度为 78%, 以 PVP K30为基质 的固体分散体极大地改善了式 I化合物的溶出度。 实施例 11 固体分散体 2的溶出度测试  The results are shown in Figure 5. The dissolution of the compound of formula I at 10 min is < 2%, the dissolution of the physical mixture of the compound of formula I and PVP K30 is < 5%, and the dissolution of solid dispersion 1 is 78%, with PVP K30. The solid dispersion of the matrix greatly improves the dissolution of the compound of formula I. Example 11 Dissolution test of solid dispersion 2
溶出度测试方法同实施例 10。  The dissolution test method was the same as in Example 10.
测试结果如图 6所示, 30min时,式 I化合物的溶出度 <2%,式 I化合物和 PVP K30的物理混合物的溶出度 < 5%, 固体分散体 2(载体为 PVP K30)的溶出度为 78%。  The test results are shown in Figure 6. At 30 min, the dissolution of the compound of formula I is <2%, the dissolution of the physical mixture of the compound of formula I and PVP K30 is < 5%, and the dissolution of solid dispersion 2 (carrier is PVP K30). It is 78%.
120min时, 式 I化合物的溶出≤5%, 式 I化合物与载体的物理混合物的溶出 略好于式 I化合物的, 但≤10%, 式 I化合物与载体形成的固体分散体的溶出约 80%。  At 120 min, the dissolution of the compound of formula I is ≤ 5%, the physical mixture of the compound of formula I and the carrier is slightly better eluted than the compound of formula I, but ≤ 10%, and the dissolution of the solid dispersion of the compound of formula I with the carrier is about 80%. .
结果表明: 固体分散体的溶出度要明显好于式 I化合物及式 I化合物与载体 的物理混合物。 此外, 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 (30分钟时溶出度 65-70%)。 实施例 12 固体分散体 2的体内药代研究  The results show that the dissolution of the solid dispersion is significantly better than the physical mixture of the compound of formula I and the compound of formula I with the carrier. In addition, the solid dispersion of PVP K30 as a carrier greatly improved the dissolution of the compound of formula I, which was significantly better than other commonly used carriers (such as PEG 6000, hypromellose E5) and PVP K90 and K25 (dissolution 65-70% at 30 minutes). Example 12 In vivo pharmacokinetic study of solid dispersion 2
选取固体分散体 2与对应的 API (即式 I化合物)进行体内药代试验。  The solid dispersion 2 was selected for in vivo pharmacoassay with the corresponding API (i.e., the compound of formula I).
8只雄性 6周龄 Wistar大鼠, 体重 170 g左右, 分为 2组, 每组 4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。  Eight male 6-week-old Wistar rats weighing approximately 170 g were divided into 2 groups of 4 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 samples were separated and 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。 参数参见表 2。  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. See Table 2 for parameters.
结果如表 2所示: 式 I化合物与 PVP K30按 1 : 4重量比制得的固体分散体 2体 内药代的 AUCQ_约为式 I化合物的 9.03倍。 表 2 The results are shown in Table 2: Compounds of formula I and PVP K30 1: 4 ratio of weight of the dispersion produced was 9.03 times the solid compound 2 Pharmacokinetics AUC Q _ of formula I is about. Table 2
Figure imgf000015_0001
实施例 13 固体分散体 10
Figure imgf000015_0001
Example 13 Solid Dispersion 10
式 II化合物与 PVP K30 , 重量比例为 1 : 4, 溶剂 (无水乙醇)法  Compound of formula II and PVP K30 in a weight ratio of 1:4, solvent (anhydrous ethanol)
制备方法同实施例 1, 不同点在于式 II化合物代替了式 I化合物, 式 II化合物与 The preparation method is the same as that in Example 1, except that the compound of the formula II is substituted for the compound of the formula I, and the compound of the formula II is
PVP K30的重量比例为 1 : 4。 The PVP K30 has a weight ratio of 1:4.
其 XRPD测试结果如图 7所示, 固体分散体 10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 14 固体分散体 10的溶出度测试  The XRPD test results are shown in Fig. 7. In the solid dispersion 10, the diffraction peaks (characteristic peaks) of the compound of the formula II disappeared completely, and existed in an amorphous or molecular form. Example 14 Dissolution Test of Solid Dispersion 10
溶出度测试方法同实施例 10。  The dissolution test method was the same as in Example 10.
具体地, 将样品配置 2份 (分别为 sample 1和 sample 2)。按照上述方法测试各 自的溶出度, 并计算平均值 (即为 average)。  Specifically, the sample was configured in 2 parts (sample 1 and sample 2, respectively). The respective dissolution rates were tested as described above and the average value (i.e., average) was calculated.
测试结果如图 8所示, 30min时, 固体分散体 10的溶出度约为 72.5%, 120min 时, 固体分散体 10的溶出约为 94%。 可见, 式 II化合物与载体 PVP K30形成的固 体分散体 10的溶出度很好。 实施例 15 固体分散体 10的体内药代研究 The test results are shown in Fig. 8. At 30 minutes, the dissolution rate of the solid dispersion 10 was about 72.5%, and at 120 minutes, the dissolution of the solid dispersion 10 was about 94%. It can be seen that the compound of formula II forms a solid with the carrier PVP K30. The dissolution of the bulk dispersion 10 is good. Example 15 In vivo pharmacokinetic study of solid dispersion 10
选取固体分散体 10与对应的 API (即式 II化合物)进行体内药代试验。  The solid dispersion 10 is selected for in vivo pharmacoassay with the corresponding API (i.e., the compound of formula II).
测试方法同实施例 12。  The test method is the same as in the embodiment 12.
结果如表 3所示: 式 II化合物与 PVP K30按 1 : 4重量比例制备的固体分散体 10体内药代的 AUClast约为式 II化合物的 13.5倍。 The results are shown in Table 3: The solid dispersion 10 of the compound of the formula II and the PVP K30 in a weight ratio of 1:4 was subjected to an AUC last of about 13.5 times that of the compound of the formula II.
表 3  table 3
Figure imgf000016_0001
实施例 16 固体分散体 1加速稳定性试验
Figure imgf000016_0001
Example 16 Solid dispersion 1 accelerated 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), and taken out at 1, 2, and 3 months for X-ray powder diffraction.
XRPD结果如图 9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 Figure 9, which indicates that the solid dispersion 1 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 is unchanged), still in the form of a solid dispersion in an amorphous or molecular state.
结果图 9所示: 从上往下依次是式 I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 17 固体分散体 2加速稳定性试验 The results are shown in Fig. 9. From the top to the bottom, the compound of the formula I, the freshly prepared solid dispersion 1, the solid dispersion 1 was left for one month, the solid dispersion 1 was placed for two months, and the solid dispersion 1 was placed three times. XRPD chart at the time of month. Example 17 Solid dispersion 2 accelerated stability test
选取固体分散体 2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 2 was selected for accelerated stability test (40 ° C, 75% H), and taken out at 1, 2, and 3 months for X-ray powder diffraction.
XRPD结果如图 10所示, 表明固体分散体 2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 10. It shows that the solid dispersion 2 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.
结果图 10所示: 从上往下依次是式 I化合物、 新制备的固体分散体 2、 固体 分散体 2放置一个月时、 固体分散体 2放置两个月时、 固体分散体 2放置三个月 时的 XRPD图。 实施例 18 固体分散体 3加速稳定性试验  The results are shown in Fig. 10: From the top to the bottom, the compound of the formula I, the freshly prepared solid dispersion 2, the solid dispersion 2 are left for one month, the solid dispersion 2 is placed for two months, and the solid dispersion 2 is placed three times. XRPD chart at the time of month. Example 18 Solid dispersion 3 accelerated 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), and taken out at 1, 2, and 3 months for X-ray powder diffraction.
XRPD结果如图 1 1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 Fig. 11. It shows 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 there is no crystal form. The formation (i.e., the original amorphous or molecular state is unchanged) remains a solid dispersion that exists in an amorphous or molecular state.
结果图 1 1所示: 从上往下依次是式 I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 19 固体分散体 4加速稳定性试验  The results are shown in Fig. 11. From the top to the bottom, the compound of the formula I, the freshly prepared solid dispersion 3, the solid dispersion 3 are left for one month, the solid dispersion 3 is placed for two months, and the solid dispersion 3 is placed three. XRPD chart at the time of month. Example 19 Solid Dispersion 4 Accelerated 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), and taken out at 1, 2, and 3 months for X-ray powder diffraction.
XRPD结果如图 12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 12, which indicates that the solid dispersion 4 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.
结果图 12所示: 从上往下依次是式 I化合物、 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 20-24 固体分散体 11-15 活性成分: 式 I化合物 The results are shown in Fig. 12: from the top to the bottom, the compound of the formula I, the freshly prepared solid dispersion 4, the solid dispersion 4 was left for one month, the solid dispersion 4 was left for two months, and the solid dispersion 4 was placed three times. XRPD chart at the time of month. Example 20-24 Solid Dispersion 11-15 Active Ingredient: Compound of Formula I
载体: PVP K30和其他载体的混合载体  Vector: Mixed carrier of PVP K30 and other carriers
制备方法同实施例 1, 不同点在于混合载体采用如表 4所示的条件。  The preparation method was the same as in Example 1, except that the mixed carrier was subjected to the conditions shown in Table 4.
表 4  Table 4
Figure imgf000018_0001
Figure imgf000018_0001
其 XRPD测试结果表明, 固体分散体中, 式 I化合物的衍射峰 (特征峰)全部消 以无定型形态存在。  The XRPD test results show that in the solid dispersion, the diffraction peaks (characteristic peaks) of the compound of formula I are all in an amorphous form.
固体分散体 11-15的体内药代试验 (操作同前)表明: 固体分散体的体内药代 的 AUClast约为 API的 7- 15倍。 (Before the operation is the same) of a solid dispersion 11-15 Pharmacokinetics test sample showed: solid dispersion of Pharmacokinetics AUC last about 7-15 times the API.
可见, 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; 且较活性成分而言, 本发明的固体分散体具有显著提高的溶出度, 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, 更佳地为 9-14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。  It can be seen that the solid dispersion comprising the active ingredient and the PVP K30-containing carrier prepared by the present invention is very stable; and the solid dispersion of the present invention has significantly improved dissolution compared to the active ingredient, especially the solid dispersion of the present invention. The area under the curve of the body is 6-20 times, preferably 7-15 times, more preferably 9-14 times the area under the curve of the drug when the active ingredient is administered alone. 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, 和  (A) a compound of the formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, as an active ingredient, and
Figure imgf000019_0001
Figure imgf000019_0001
式中, R为 CD3或 CH: 以及 Where R is CD 3 or CH : and
(B) 亲水性高分子载体;  (B) a hydrophilic polymer carrier;
其中, (A)和 (B)的重量比 1-1: 40。  Among them, the weight ratio of (A) and (B) is 1-1:40.
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 I所示的 化合物, 或其药学上  The solid dispersion according to claim 1, wherein the active ingredient is a compound represented by Formula I, or a pharmaceutical thereof
Figure imgf000019_0002
Figure imgf000019_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 :The solid dispersion according to claim 1, wherein the weight ratio of the (A) and (B) is 1:
1-1: 20。 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 in a pH 4.5 aqueous solution of sodium acetate buffer 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 120 ° 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,  Wherein the active ingredient is a compound of formula A or a derivative thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof,
Figure imgf000020_0001
Figure imgf000020_0001
式中, R为 CD3或 CH: 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, 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。 Where R is CD 3 or CH : The hydrophilic polymer carrier is a hydrophilic polymer carrier containing povidone K30, and the solvent is selected from the group consisting of methanol, ethanol, isopropanol, acetone, chloroform, 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.
1 1.一种如权利要求 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. The pharmaceutical composition is described.
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^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., raf^ enzyme); 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/CN2013/072645 2012-03-15 2013-03-14 Solid dispersion improving absorption performance and preparation of same WO2013135189A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201210068420.4 2012-03-15
CN201210068420.4A CN103301066B (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
WO2013135189A1 true WO2013135189A1 (en) 2013-09-19

Family

ID=49127077

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2013/072645 WO2013135189A1 (en) 2012-03-15 2013-03-14 Solid dispersion improving absorption performance and preparation of same

Country Status (2)

Country Link
CN (1) CN103301066B (en)
WO (1) WO2013135189A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114014804A (en) * 2013-10-25 2022-02-08 苏州泽璟生物制药股份有限公司 Fluorine-containing deuterated omega-diphenylurea hydrate and crystal form thereof
WO2015085888A1 (en) * 2013-12-09 2015-06-18 Jiangsu Medolution Limited 4-(4-(3-(4-chloro-3-(trifluoromethyl)phenyl)ureido)-3-fluorophenoxy)-n-d3-methylpicolinamide monohydrate
CN104434809B (en) * 2014-12-10 2018-10-16 北京科莱博医药开发有限责任公司 A kind of olaparib solid dispersion preparation and preparation method thereof
CN107661296A (en) * 2016-07-27 2018-02-06 江苏先声药业有限公司 A kind of Rui Gefeini solid dispersions and preparation method thereof
CN109276543B (en) * 2017-07-19 2021-01-08 深圳微芯生物科技股份有限公司 Sitaglipta solid dispersion and preparation method and application thereof
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 (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008089388A2 (en) * 2007-01-19 2008-07-24 Bayer Healthcare Llc Treatment of cancers having resistance to chemotherapeutic agents
WO2009156070A1 (en) * 2008-06-25 2009-12-30 Bayer Schering Pharma Aktiengesellschaft Diaryl urea for treating heart failure
WO2010144499A2 (en) * 2009-06-09 2010-12-16 Medolution Limited Urea derivatives as kinase inhibitors
CN102266290A (en) * 2010-05-27 2011-12-07 四川大学 4-(4-benzamido-phenoxyl)-2-(methyl-carbamyl) pyridine derivative or solid dispersion of salt derivative, as well as preparation method of derivative or solid dispersion

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
MY191349A (en) * 2004-08-27 2022-06-17 Bayer Pharmaceuticals Corp New pharmaceutical compositions for the treatment of hyper-proliferative disorders
CN101676266B (en) * 2008-09-19 2011-10-26 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

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008089388A2 (en) * 2007-01-19 2008-07-24 Bayer Healthcare Llc Treatment of cancers having resistance to chemotherapeutic agents
WO2009156070A1 (en) * 2008-06-25 2009-12-30 Bayer Schering Pharma Aktiengesellschaft Diaryl urea for treating heart failure
WO2010144499A2 (en) * 2009-06-09 2010-12-16 Medolution Limited Urea derivatives as kinase inhibitors
CN102266290A (en) * 2010-05-27 2011-12-07 四川大学 4-(4-benzamido-phenoxyl)-2-(methyl-carbamyl) pyridine derivative or solid dispersion of salt derivative, as well as preparation method of derivative or solid dispersion

Also Published As

Publication number Publication date
CN103301066B (en) 2018-12-07
CN103301066A (en) 2013-09-18

Similar Documents

Publication Publication Date Title
JP6612200B2 (en) Choline salts of anti-inflammatory substituted cyclobutenedione compounds
EP2068839B2 (en) Pharmaceutical compositions comprising nilotinib or its salt
WO2013135189A1 (en) Solid dispersion improving absorption performance and preparation of same
US10406137B2 (en) Polymorphic forms of ST-246 and methods of preparation
SG190210A1 (en) Pharmaceutical compositions
US20140341993A1 (en) Solid pharmaceutical composition comprising an antibiotic from the quinolone family and method of production thereof
JP2003534296A (en) Pranlukast solid dispersion with improved dissolution and method for producing the same
WO2011079766A1 (en) Aripiprazole solid preparation and the production method thereof
WO2013135187A1 (en) Solid dispersion of improved adsorption performance and preparation thereof
US20220323441A1 (en) Therapeutic for gout or hyperuricemia
US20110262540A1 (en) Solid Pharmaceutical Composition Comprising Exemestane
US20210205281A1 (en) Therapy for treating malignancies
US20220241208A1 (en) Pharmaceutical compositions for the treatment of hbv
US20110160213A1 (en) Pharmaceutical compositions for the treatment of inflammatory and allergic disorders
AU2017251803A1 (en) Choline salt of an anti-inflammatory substituted cyclobutenedione compound
AU2017202359A1 (en) Polymorphic forms st-246 and methods of preparation
AU2012303675A1 (en) Choline salt of an anti - inflammatory substituted cyclobutenedione compound

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: 13761661

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: 13761661

Country of ref document: EP

Kind code of ref document: A1