CN103301066B - It is a kind of improve absorbent properties solid dispersions and its preparation - Google Patents

It is a kind of improve absorbent properties solid dispersions and its preparation Download PDF

Info

Publication number
CN103301066B
CN103301066B CN201210068420.4A CN201210068420A CN103301066B CN 103301066 B CN103301066 B CN 103301066B CN 201210068420 A CN201210068420 A CN 201210068420A CN 103301066 B CN103301066 B CN 103301066B
Authority
CN
China
Prior art keywords
solid dispersions
compound
formula
active constituent
pharmaceutical composition
Prior art date
Legal status (The legal status 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 status listed.)
Active
Application number
CN201210068420.4A
Other languages
Chinese (zh)
Other versions
CN103301066A (en
Inventor
易必慧
尚晓芳
陆惠萍
盛泽林
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Suzhou Zehuang Biopharmaceutical Co., Ltd.
Original Assignee
Suzhou Zelgen Biopharmaceutical Co Ltd
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 Suzhou Zelgen Biopharmaceutical Co Ltd filed Critical Suzhou Zelgen Biopharmaceutical Co Ltd
Priority to CN201210068420.4A priority Critical patent/CN103301066B/en
Priority to PCT/CN2013/072645 priority patent/WO2013135189A1/en
Publication of CN103301066A publication Critical patent/CN103301066A/en
Application granted granted Critical
Publication of CN103301066B publication Critical patent/CN103301066B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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

Abstract

The present invention relates to a kind of solid dispersions for improving absorbent properties and its preparations.Specifically, the present invention provides a kind of solid dispersions, the solid dispersions include the formula A compound of (A) as active constituent or derivatives thereof or its pharmaceutically acceptable salt, hydrate or solvate, and (B) hydrophilic macromolecular carrier.Wherein the weight ratio of (A) and (B) are 1: 1-1: 40.Solid dispersions provided by the invention can improve the dissolution in vitro of active constituent well, significantly improve absorption of the mammal to the active constituent, to efficiently reduce its dosage.

Description

It is a kind of improve absorbent properties solid dispersions and its preparation
Technical field
The invention belongs to field of medicaments, it is related to a kind of solid dispersions and its preparation, specifically a kind of improvement absorbent properties Solid dispersions and its preparation.
Background technique
4- (4- (3- (4- chloro- 3- (trifluoromethyl) phenyl] uride) the fluoro- phenoxy group of -3-) -2- (N-1 ', 1 ', 1 '-three deuterium For methyl) picolinamide, i.e. compound of formula I, the compound or its pharmaceutically acceptable salt are to phosphokinase (Kinase) Such as raf kinases has excellent inhibition, can be used for treating tumour and its related disease.
The compound is colored powder, odorless, tasteless.It is easily dissolved in dimethyl sulfoxide or dimethylformamide, in tetrahydro It is slightly molten in furans, methanol, the slightly soluble in acetone, dehydrated alcohol or glacial acetic acid, but it is almost insoluble in water.
Insoluble drug since compound of formula I water solubility is very poor, the solubility and solution rate in physiological fluid compared with Low, dissolution rate is relatively low, eventually leads to that bioavilability is lower, seriously affects absorption of the mammal to compound of formula I, unfavorable In the disease therapeuticing effect for giving full play to the compound.
Solid dispersions can be improved the bioavilability of insoluble drug, but existing solid dispersions, and highest is only The area under the drug-time curve (AUC) of drug can be improved 2-6 times.For example, in CN101632630A, it is general for probucol It spreads out and examines and PVP K30Solid dispersions, dog relative bioavailability also only improves only 5.8 times.
Therefore, the solid dispersions for developing a kind of compound of formula I that bioavilability significantly improves, to solution mammal It is necessary to the problem of its active ingredient draws difference.
Summary of the invention
It is an object of the present invention to provide a kind of bioavilability significantly improve with formula A compound or derivatives thereof, Or its pharmaceutically acceptable salt, hydrate or solvate are the solid dispersions of active constituent.
It is a further object of the present invention to provide the preparation methods of solid dispersions of the present invention.
Another object of the present invention is to provide the pharmaceutical composition of the solid dispersions.
First aspect present invention provides a kind of solid dispersions, including
(A) as formula A compound of active constituent or derivatives thereof or its pharmaceutically acceptable salt, hydrate or molten Object is closed in agent, and
In formula, R CD3Or CH3;And
(B) hydrophilic macromolecular carrier;
Wherein, the weight ratio of (A) and (B) are 1: 1-1: 40.
In another preferred example, the hydrophilic macromolecular carrier is povidone or the macromolecule carrier containing povidone.
In another preferred example, the hydrophilic macromolecular carrier is PVP K30 or the macromolecule containing PVP K30 Carrier, wherein the content of PVP K30 is 10-100wt%, by the total weight of macromolecule carrier.
In another preferred example, the active constituent is Formulas I compound represented or its pharmaceutically acceptable salt, water Close object or solvate:
In another preferred example, the macromolecule carrier containing PVP K30 also includes other water solubilitys selected from the group below Carrier: polyethylene glycols, povidone class, cellulose family, it is surfactant-based, or combinations thereof;
Wherein, the polyethylene glycols are selected from the group: Macrogol 4000, Macrogol 6000, PEG 8000 or A combination thereof;
The povidone class is selected from the group: 30 POVIDONE K 30 BP/USP 25,30 POVIDONE K 30 BP/USP 90, povidone S630, or combinations thereof;
The cellulose family is selected from the group: hydroxypropyl methylcellulose E5, hydroxypropyl methylcellulose E15, hydroxypropylcellulose L or its Combination;
It is described surfactant-based to be selected from the group: poloxamer188, PLURONICS F87, or combinations thereof.
In another preferred example, the macromolecule carrier containing PVP K30 includes to be selected from 30 POVIDONE K 30 BP/USP 25, povidone K90, povidone S630, or combinations thereof povidone class carrier.
In another preferred example, the weight ratio of the macromolecule carrier containing PVP K30 and other water-solubility carriers It is 1: 4-4: 1;Preferably 1: 2-4: 1;It is more preferably 1: 1-4: 1.
In another preferred example, the weight ratio of described (A) and (B) are 1: 1-1: 20.
In another preferred example, the weight ratio of described (A) and (B) are 1: 2-1: 9.
In another preferred example, in the X-ray powder diffraction figure of the solid dispersions, do not have the solid and disperse The X-ray diffraction characteristic peak of the active constituent of body.
In another preferred example, there are following one or more features:
1. the area under the drug-time curve of the solid dispersions is that the active constituent of the solid dispersions is administered alone 6-20 times, preferably 7-15 times of area under the drug-time curve;
2. accumulation dissolution rate is big when the solid dispersions are 10 minutes in 37 DEG C, the sodium acetate buffer aqueous solution of pH4.5 In 70%, preferably 75-95%;
3. accumulation dissolution rate is when the solid dispersions are 120 minutes in 37 DEG C, the sodium acetate buffer aqueous solution of pH4.5 10-40 times, preferably 15-30 times of the active constituent accumulation dissolution rate of the solid dispersions.
In another preferred example, the area under the drug-time curve is measured in following animal: rat, mouse or Dog.
Second aspect of the present invention provides a kind of preparation method of solid dispersions described in first aspect present invention, including side Method:
(a) fusion method 1, comprising steps of
(a1) mixed active ingredient and hydrophilic macromolecular carrier, to obtain a mixture containing active constituent;
(a2) mixture containing active constituent that heating melting steps (a1) obtain, thus the mixture after being melted; With
(a3) mixture after the melting that cooling step (a2) obtains, to obtain solid described in first aspect present invention Dispersion;
Or
(b) fusion method 2, comprising steps of
(b1) heating melting hydrophilic macromolecular carrier, thus the carrier after being melted;
(b2) by after melting that step (b1) obtains carrier and active constituent mix after melt, thus after being melted Mixture;With
(b3) mixture after the melting that cooling step (b2) obtains, to obtain solid described in first aspect present invention Dispersion;
Or
(c) solvent method, comprising steps of
(c1) by active constituent and hydrophilic macromolecular carrier co-dissolve in solvent, to obtain a mixture;With
(c2) solvent in the mixture that step (c1) is obtained is removed, to obtain solid described in first aspect present invention Dispersion;
Wherein, the active constituent be formula A compound or derivatives thereof or its pharmaceutically acceptable salt, hydrate or Solvate,
In formula, R CD3Or CH3,
The hydrophilic macromolecular carrier is the hydrophilic macromolecular carrier containing PVP K30,
The solvent is selected from the group: methanol, ethyl alcohol, isopropanol, acetone, methylene chloride, tetrahydrofuran, or combinations thereof.
In another preferred example, the step (c2) comprising steps of
The solvent in the mixture that step (c1) is obtained is removed with Rotary Evaporators;Or
The solvent in the mixture that step (c1) obtains is dried and removed with spray dryer.
In another preferred example, the technological parameter of the drying are as follows: 110 DEG C~120 DEG C of inlet air temperature, power of fan 55% ~65%, wriggling pump power 50%~55%.
In another preferred example, the solid dispersions are further handled at least one other procedure of processing, described Procedure of processing is to grind, sieve, roll, mill, screen, mix or combinations thereof.
Third aspect present invention provides a kind of pharmaceutical composition, and the composition includes:
(1) solid dispersions described in first aspect present invention;With
(2) pharmaceutically acceptable excipient.
In another preferred example, the excipient include: filler, diluent, sweetener, corrigent, antioxidant, Toner, preservative, lubricant, adhesive, disintegrating agent or its mixing.
In another preferred example, the composition is capsule, tablet, pill, powder and granule.
In another preferred example, described pharmaceutical composition, for inhibiting the pharmaceutical composition of phosphokinase (such as raf kinases) Object.
In another preferred example, the pharmaceutical composition is for treating and preventing cancer.
Fourth aspect present invention provides a kind of preparation method of third aspect present invention described pharmaceutical composition, this is sent out Solid dispersions described in bright first aspect and the mixing of pharmaceutically acceptable excipient, so that the pharmaceutical composition be made Object.
Fifth aspect present invention provides a kind of purposes of solid dispersions described in first aspect present invention, is used to prepare Inhibit the drug of phosphokinase (such as raf kinases);Or it is used to prepare the drug for the treatment of tumour.
Sixth aspect present invention provides a kind for the treatment of method, by solid dispersions described in (i) first aspect present invention Or (ii) pharmaceutical composition as described in the third aspect of the present invention, it is applied to patient in need for the treatment of.
It should be understood that above-mentioned each technical characteristic of the invention and having in below (eg embodiment) within the scope of the present invention It can be combined with each other between each technical characteristic of body description, to form a new or preferred technical solution.As space is limited, exist This no longer tires out one by one states.
Detailed description of the invention
Fig. 1 shows the XRPD figure of solid dispersions 1.
Fig. 2 shows the XRPD figure of solid dispersions 2.
Fig. 3 shows the XRPD figure of solid dispersions 3.
Fig. 4 shows the XRPD figure of solid dispersions 4.
Fig. 5 shows the dissolution of compound of formula I, the physical mixture of compound of formula I and PVP K30 and solid dispersions 1 Curve.
Fig. 6 shows the dissolution of compound of formula I, the physical mixture of compound of formula I and PVP K30 and solid dispersions 2 Curve.
Fig. 7 shows the XRPD figure of solid dispersions 10.
Fig. 8 shows the dissolution curve of solid dispersions 10.
Fig. 9 shows the XRPD comparison diagram after 1,2,3 month accelerated stability test of solid dispersions 1,9 institute of result figure Show: when being successively from top to bottom that compound of formula I, freshly prepd solid dispersions 1, solid dispersions 1 are placed one month, solid point XRPD figure when granular media 1 is placed two months, when solid dispersions 1 are placed three months.
Figure 10 shows the XRPD comparison diagram after 1,2,3 month accelerated stability test of solid dispersions 2,10 institute of result figure Show: when being successively from top to bottom that compound of formula I, freshly prepd solid dispersions 2, solid dispersions 2 are placed one month, solid point XRPD figure when granular media 2 is placed two months, when solid dispersions 2 are placed three months.
Figure 11 shows the XRPD comparison diagram after 1,2,3 month accelerated stability test of solid dispersions 3,11 institute of result figure Show: when being successively from top to bottom that compound of formula I, freshly prepd solid dispersions 3, solid dispersions 3 are placed one month, solid point XRPD figure when granular media 3 is placed two months, when solid dispersions 3 are placed three months.
Figure 12 shows the XRPD comparison diagram after 1,2,3 month accelerated stability test of solid dispersions 4,12 institute of result figure Show: when being successively from top to bottom that compound of formula I, freshly prepd solid dispersions 4, solid dispersions 4 are placed one month, solid point XRPD figure when granular media 4 is placed two months, when solid dispersions 4 are placed three months.
Specific embodiment
The present inventor passes through long-term in-depth study, it has unexpectedly been found that, one kind includes the formula A of (A) as active constituent Compound or derivatives thereof or its pharmaceutically acceptable salt, hydrate or solvate and (B) hydrophilic macromolecular carrier The solid dispersions of (especially containing the macromolecule carrier of PVP K30), for active constituent, the solid dispersions Area under the drug-time curve be about 6-20 times of active constituent;The solid dispersions significantly improve the external molten of its active constituent Out-degree significantly improves absorption of the mammal to active constituent, to effectively reduce its dosage.In addition, the solid Dispersion also has extraordinary stability, is extremely applicable to the pharmaceutical composition of preparation related disease.On this basis, it invents People completes the present invention.
Solid dispersions
Solid dispersions (solid dispersion), which refer to for active constituent to be highly dispersed in (solid) carrier, to be formed A kind of decentralized system existing in solid form.
Active constituent
As used herein, term " active constituent " refers to formula A compound or derivatives thereof or its is pharmaceutically acceptable Salt, hydrate or solvate, in formula, R CD3Or CH3
The term further includes and various crystalline forms, hydrate or the solvate of formula A compound or derivatives thereof.
Preferably, the formula A compound refers to compound of formula I or Formula II compound or its pharmaceutically acceptable salt.
As used herein, term " pharmaceutically acceptable salt " refers to that the compounds of this invention and acid or alkali are formed by suitable use Make the salt of drug.Pharmaceutically acceptable salt includes inorganic salts and organic salt.A kind of preferred salt is the compounds of this invention and acid The salt of formation.The acid for suitably forming salt includes but is not limited to: hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid etc. are inorganic Acid, formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, Picric acid, methanesulfonic acid, benzene methanesulfonic acid, the organic acids such as benzene sulfonic acid;And the acidic amino acids such as aspartic acid, glutamic acid.
Preferably, the pharmaceutically acceptable salt is toluenesulfonate.
Carrier
Carrier used in solid dispersions, it is most common to have water-solubility carrier, slightly solubility carrier and enteric solubility carrier.
" carrier " of the present invention is hydrophilic macromolecular carrier, and preferably povidone or the macromolecule containing povidone carries Body or PVP K30 or macromolecule carrier containing PVP K30, wherein the content of PVP K30 is 10-100wt%, is pressed The total weight of macromolecule carrier.
It is highly preferred that the macromolecule carrier used in the present invention containing PVP K30 refers to that the weight content of PVP K30 is super Cross the carrier of total weight of carrier 20%;Or the weight content of PVP K30 is more than the carrier of total weight of carrier 40%;Or povidone The weight content of K30 is more than the carrier of total weight of carrier 60%.
It is, of course, also possible to contain other water-solubility carriers, for example including (but being not limited to): polyethylene glycols, povidone class, Cellulose family, surfactant-based, mannitol, or combinations thereof.
The polyethylene glycols are selected from the group: Macrogol 4000 (PEG 4000), Macrogol 6000 (PEG6000), PEG 8000 (PEG 8000) or combinations thereof;
The povidone class is selected from the group: 30 POVIDONE K 30 BP/USP 90 (PVP K90), 30 POVIDONE K 30 BP/USP 25 (PVP K25), povidone S630, or combinations thereof;
The cellulose family is selected from the group: hydroxypropyl methylcellulose E5 (HPMC E5), hydroxypropyl methylcellulose E15 (HPMC E15), hydroxypropylcellulose L (HPC-L), or combinations thereof;
It is described surfactant-based to be selected from the group: poloxamer188 (Poloxamer 407), PLURONICS F87 (Poloxamer 188), or combinations thereof.
Composition
Solid dispersions provided by the invention include
(A) as formula A compound of active constituent or derivatives thereof or its pharmaceutically acceptable salt, hydrate or molten Object is closed in agent, wherein formula A compound is the same as the above.With
(B) hydrophilic macromolecular carrier;
Wherein, the weight ratio of (A) and (B) are 1: 1-1: 40.
Preferably, the weight ratio of described (A) and (B) are 1: 20-1: 1, preferably, the weight ratio of (A) and (B) are 1: 9-1∶2。
Preparation method
Solid dispersions of the present invention can be made by following method, however the condition of this method, such as carrier, molten Following explanation is not limited to the time required to agent, the amount of each ingredient, preparation temperature, preparation etc..Solid dispersions of the present invention are also Optionally various synthetic methods describing in the present specification or known in the art can be combined and be easily made, this The combination of sample can readily be carried out by those skilled in the art in the invention.
Specifically, the preparation method can be fusion method, solvent method, solvent-fusion method or Dispersion on surface method.It is preferred that Ground, the preparation method are fusion method or solvent method.
Fusion method
Common, the fusion method is to be uniformly mixed (A) active constituent, and (B) hydrophilic macromolecular carrier, uses water-bath Or then oil bath heating is cooled to solid, the solid dispersions of the powdered active constituent is obtained after crushing to melting.
Alternatively, (A) active constituent after (B) hydrophilic macromolecular carrier heating melting, can will be added, then by melting Mixture with vigorous stirring, is quickly cooled to solid, and the solid dispersions of the powdered active constituent are obtained after crushing.
Solvent method
Common, the solvent method is by (A) active constituent, and (B) hydrophilic macromolecular carrier co-dissolve is in solvent In, after mixture is uniform, the solvent in solution is removed, active constituent and hydrophilic macromolecular carrier can be made while being precipitated, crushed The solid dispersions of the powdered active constituent can be obtained afterwards.
Solvent used in the solvent method include: methanol, ethyl alcohol, isopropanol, acetone, tetrahydrofuran, or combinations thereof.
Structural Identification
The Structural Identification method of solid dispersions includes differential scanning calorimetric analysis, X-ray powder diffraction etc..It is right respectively The solid dispersions of active constituent, carrier and the method for the invention preparation have carried out X-ray powder diffraction (XRPD) detection, Compare the XRPD spectrogram of three.
The characteristic peak of active constituent
After solid dispersions of the invention are made, the X-ray diffraction characteristic peak of active constituent substantially or entirely disappears It loses.In other words, in the X-ray powder diffraction figure of solid dispersions of the present invention, the active constituent without the solid dispersions X-ray diffraction characteristic peak.
Compound of formula I, 7.24 ± 0.1 °, 13.38 ± 0.1 °, 14.53 ± 0.1 °, 14.74 ± 0.1 °, 15.61 ± 0.1°、17.23±0.1°、18.57±0.1°、18.84±0.1°、19.81±0.1°、20.01±0.1°、23.42±0.1°、 24.24 ± 0.1 °, 25.85 ± 0.1 °, 26.39 ± 0.1 °, 27.43 ± 0.1 ° have strong diffraction maximum.
Formula II compound, 7.20 ± 0.1 °, 13.34 ± 0.1 °, 14.49 ± 0.1 °, 14.72 ± 0.1 °, 15.55 ± 0.1°、16.44±0.1°、17.17±0.1°、18.53±0.1°、18.81±0.1°、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 ° has strong diffraction maximum.
The characteristic peak of carrier
Some carriers have strong diffraction maximum, for example, PEG4000 and PEG6000 has by force in 19.20 ± 0.1 °, 23.30 ± 0.1 ° Diffraction maximum.Poloxamer 407 has strong diffraction maximum in 19.16 ± 0.1 °, 23.33 ± 0.1 °.Some carriers do not have strong diffraction maximum, For example, PVP K30, HPMC E5 and HPC-L do not detect strong diffraction maximum.
The characteristic peak of solid dispersions
The strong diffraction maximum of active constituent disappears in the XRPD spectrogram of solid dispersions prepared by the present invention, so that it is determined that this hair The bright solid dispersions are successfully prepared, wherein the simple physical mixture of not instead of active constituent and carrier, with nothing Sizing or molecular conformation exist.
Pharmaceutical composition
Term " the compounds of this invention " refers to formula A compound or derivatives thereof or its pharmaceutically acceptable salt.
Term " solid dispersions of the present invention " refers to that active constituent is formula A compound or derivatives thereof or it can pharmaceutically connect The solid dispersion for the salt received.
Since the compounds of this invention has the excellent inhibitory activity to phosphokinase (Kinase) such as raf kinases, because This solid dispersions of the present invention and it can be used for treating, in advance containing solid dispersions of the present invention pharmaceutical composition as main component Anti- and alleviation is by the disease kinase mediated to phosphokinase (Kinase) such as raf.According to the prior art, the compounds of this invention It can be used for treating following disease: cancer, cardiovascular disease, obesity, diabetes etc..
Pharmaceutical composition of the invention include safe and effective amount within the scope of solid dispersions of the present invention or its pharmacologically may be used The salt of receiving and pharmacologically acceptable excipient.Wherein " safe and effective amount " refers to: the amount of solid dispersions of the present invention It is enough to be obviously improved the state of an illness, and is unlikely to generate serious side effect.Consolidate in general, pharmaceutical composition contains the 1-2000mg present invention Body dispersion/agent more preferably contains 10-200mg solid dispersions/agent of the present invention.Preferably, it is described it is " one " be one Capsule or tablet.
" pharmaceutically acceptable excipient " refers to: one or more biocompatible solids or liquid filler or jello Matter, they are suitable for people's use and it is necessary to have enough purity and sufficiently low toxicity." compatibility " referred to herein as combines In object each component energy and solid dispersions of the invention and they between mutually admix, and significantly reduce solid dispersions Drug effect.Pharmaceutically acceptable excipient partial example has cellulose and its derivates (such as sodium carboxymethylcellulose, ethyl fibre Tie up plain sodium, cellulose ethanoate etc.), gelatin, talcum, solid lubricant (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oil (such as soya-bean oil, sesame oil, peanut oil, olive oil), polyalcohol (such as propylene glycol, glycerol, mannitol, sorbierite), emulsifier (such as tween), wetting agent (such as lauryl sodium sulfate), colorant, flavoring agent, stabilizer, antioxidant, preservative, without heat Raw water etc..
The method of application of solid dispersions of the present invention or pharmaceutical composition is not particularly limited, representative method of application packet Include (but being not limited to): in oral, tumor, rectum, parenteral (intravenous, intramuscular or subcutaneous) and local administration.
Solid dosage forms for oral administration includes capsule, tablet, pill, powder and granule.In these solid formulations In type, solid dispersions of the present invention are mixed at least one conventional inert excipients (or carrier), such as sodium citrate or di(2-ethylhexyl)phosphate Calcium, or mixed with following compositions: (a) filler or expanding material, for example, starch, lactose, sucrose, glucose, microcrystalline cellulose, sweet Reveal pure and mild silicic acid;(b) adhesive, for example, hypromellose, alginates, gelatin, polyvinylpyrrolidone, sucrose and Arabic gum;(c) moisturizer, for example, glycerol;(d) disintegrating agent, for example, agar, calcium carbonate, potato starch or tapioca, Alginic acid, certain composition silicates, croscarmellose sodium and sodium carbonate;(e) retarding solvent, such as paraffin;(f) it absorbs and accelerates Agent, for example, quaternary ammonium compound;(g) wetting agent, such as cetanol, lauryl sodium sulfate and glycerin monostearate;(h) it inhales Attached dose, for example, kaolin;(i) lubricant, for example, talcum, calcium stearate, magnesium stearate, solid polyethylene glycol, dodecane Or mixtures thereof base sodium sulphate,.In capsule, tablet and pill, dosage form also may include buffer.
Coating and shell material preparation can be used in solid dosage forms such as tablet, sugar-pill, capsule, pill and granule, such as casing and Other materials well known in the art.They may include opacifying agent, also, the release of solid dispersions can be in this composition The mode of delay discharges in certain a part in the digestive tract.The example of adoptable embedding component is polymeric material and wax class object Matter.When necessary, solid dispersions can also be with one of above-mentioned excipient or a variety of formation microencapsulation forms.
Liquid formulation for oral administration includes pharmaceutically acceptable lotion, solution, suspension, syrup or tincture. Other than solid dispersions, liquid dosage form may include the inert diluent routinely used in this field, such as water or other solvents, increase Solvent and emulsifier, example know, ethyl alcohol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-BDO, dimethyl formyl The mixture of amine and oil, especially cottonseed oil, peanut oil, maize germ, olive oil, castor oil and sesame oil or these substances Deng.
Other than these inert diluents, composition also may include auxiliary agent, such as wetting agent, emulsifier and suspending agent, sweet taste Agent, tender taste agent and fragrance.
Other than solid dispersions, suspension may include suspending agent, for example, ethoxylation isooctadecane alcohol, polyoxyethylene Sorbierite and Isosorbide Dinitrate, microcrystalline cellulose, aluminium methoxide and agar or the mixture of these substances etc..
Composition for parenteral injection may include physiologically acceptable sterile, aqueous or anhydrous solution, dispersion liquid, Suspension or lotion, and the aseptic powdery for re-dissolving into sterile Injectable solution or dispersion liquid.It is suitable aqueous and Nonaqueous carrier, diluent, solvent or excipient include water, ethyl alcohol, polyalcohol and its suitable mixture.
The dosage form of solid dispersions of the present invention for local administration includes ointment, powder, patch, stock solution and sucking Agent.Solid dispersions aseptically with physiologically acceptable carrier and any preservative, buffer, or when necessary may The propellant needed is mixed together.
Solid dispersions of the present invention can be administered alone, or be administered in combination with other pharmaceutically acceptable compounds. When using pharmaceutical composition, be the solid dispersions of the present invention of safe and effective amount are applicable to treatment mammal (such as People), wherein application when dosage be the effective dosage pharmaceutically thought, for the people of 60kg weight, day dosage Usually 1~2000mg, preferably 20~500mg.Certainly, specific dosage be also contemplated that administration route, patient health situation etc. because Element, within the scope of these are all skilled practitioners technical ability.
Present invention has the main advantage that
(1) one kind is provided with formula A compound or derivatives thereof or its pharmaceutically acceptable salt, hydrate or solvent The solid dispersions that object is active constituent are closed, the solid dispersions significantly improve the dissolution in vitro of active constituent, very Be conducive to absorption of the mammal to the active constituent.And itself relative to its active compound component, the solid Dispersion has significantly excellent internal pharmacokinetic parameter (especially area under the drug-time curve), and highly stable.
(2) a kind of preparation method of solid dispersions is provided.
Below with reference to specific implementation, the present invention is further explained.It should be understood that these embodiments be merely to illustrate the present invention and It is not used in and limits the scope of the invention.In the following examples, the experimental methods for specific conditions are not specified, usually according to normal condition, Or according to the normal condition proposed by manufacturer.Unless otherwise stated, otherwise percentage and number are calculated by weight.
" API " refers to active constituent as used herein.
Embodiment
The dissolution test method of solid dispersions
According to the Pharmacopoeia of the People's Republic of China two annex XC the second methods of dissolution method (paddle method) of version in 2010 into Row detection.Condition is as follows: dissolution medium: 0.1% lauryl sodium sulfate pH, 4.5 Acetic acid-sodium acetate buffer 900mL/ dissolution Cup;Temperature: 37 ± 0.5 DEG C;Revolving speed: 100 revs/min.
Specifically, sample is divided into three groups, every group of sample is respectively configured 3 parts.Wherein, the 1st group of sample is active constituent;The 2 groups of samples are that active constituent and carrier are only passed through simple physical method (as stirred) mixed sample.Third group sample Product are the solid dispersions that middle preparation method obtains according to various embodiments.Table specific as follows.
Using the dissolution medium, the dissolution rate of above 3 groups of samples is conventionally detected respectively, every group 3 parts of detection Sample takes its average value.Compare 3 groups of test result.
1 solid dispersions 1 of embodiment
Compound of formula I and PVP K30, weight ratio 1: 9, solvent (dehydrated alcohol) method
The PVP K30 of the compound of formula I and 9 parts by weight that weigh 1 parts by weight is set in same flask, and flask sets 80 DEG C of oil baths In, dehydrated alcohol is added into flask, close plug stirs under the conditions of condensate return, until compound and the equal dissolved clarification of PVPK30, It is stirred for 30 minutes.The liquid of dissolved clarification is quickly evaporated with rotary evaporator at 80 DEG C, solid is obtained.Solid is true at 50 DEG C It is 24 hours dry in empty drying box, it is finely ground to get solid dispersions 1.
Test results are shown in figure 1 by its XRPD, and in solid dispersions 1, the diffraction maximum (characteristic peak) of compound of formula I all disappears It loses, exists with unformed or molecular conformation.
2 solid dispersions 2 of embodiment
Compound of formula I and PVP K30, weight ratio 1: 4, solvent (dehydrated alcohol) method
The preparation method is the same as that of Example 1, and difference is that the weight ratio of compound of formula I and PVP K30 are 1: 4.
Test results are shown in figure 2 by its XRPD, and in solid dispersions 2, the diffraction maximum (characteristic peak) of compound of formula I all disappears It loses, exists with unformed or molecular conformation.
3 solid dispersions 3 of embodiment
Compound of formula I and PVP K30, weight ratio 1: 3, solvent (dehydrated alcohol) method
The preparation method is the same as that of Example 1, and difference is that the weight ratio of compound of formula I and PVP K30 are 1: 3.
Test results are shown in figure 3 by its XRPD, and in solid dispersions 3, the diffraction maximum (characteristic peak) of compound of formula I all disappears It loses, exists with unformed or molecular conformation.
4 solid dispersions 4 of embodiment
Compound of formula I and PVP K30, weight ratio 1: 2, solvent (dehydrated alcohol) method
The preparation method is the same as that of Example 1, and difference is that the weight ratio of compound of formula I and PVP K30 are 1: 2.
Test results are shown in figure 4 by its XRPD, and in solid dispersions 4, the diffraction maximum (characteristic peak) of compound of formula I all disappears It loses, exists with unformed or molecular conformation.
Embodiment 5-9 solid dispersions 5-9
The preparation method is the same as that of Example 1, and actual conditions are shown in Table 1.
Table 1
Embodiment Active constituent/carrier (weight ratio) Solvent
5 Compound of formula I/PVP K90 (1: 4) Dehydrated alcohol
6 Compound of formula I/PVP K25 (1: 4) Dehydrated alcohol
7 Compound of formula I/PVP S630 (1: 4) Dehydrated alcohol
8 Compound of formula I/HPMC E5 (1: 5) Dehydrated alcohol
9 Compound of formula I/HPC-L (1: 5) Acetone
The dissolution rate test of 10 solid dispersions 1 of embodiment
With reference to 2010 editions two annex XC dissolution rate test methods of Chinese Pharmacopoeia, 0.1% lauryl sodium sulfate pH is measured 4.5 Acetic acid-sodium acetate buffer 900mL, are placed in 1000mL stripping rotor, and temperature is kept for 37 ± 0.5 DEG C.
Be separately added into compound of formula I, compound of formula I and PVP K30 physical mixture (compound of formula I and PVPK30's Weight is respectively 50mg and 450mg), solid dispersions 1 (500mg), paddle speed be 100rpm, respectively at 2min, 5min, 10min, 15min, 20min, 30min, 45min, 60min, 120min sample 5mL and take clear filtrate dilute with 0.45 μm of filtering with microporous membrane Drug concentration is measured after releasing, and calculates the dissolution rate of three.
As a result as shown in figure 5, dissolution rate < 2% of the compound of formula I in 10min, the object of compound of formula I and PVP K30 The dissolution rate < 5% of mixture is managed, the dissolution rate of solid dispersions 1 is 78%, using PVP K30 as the solid dispersions pole of matrix The earth improves the dissolution rate of compound of formula I.
The dissolution rate test of 11 solid dispersions 2 of embodiment
Dissolution test method is the same as embodiment 10.
Test results are shown in figure 6, when 30min, the dissolution rate < 2% of compound of formula I, compound of formula I and PVPK30's The dissolution rate < 5% of physical mixture, the dissolution rate of solid dispersions 2 (carrier is PVP K30) are 78%.
When 120min, the dissolution of dissolution≤5% of compound of formula I, the physical mixture of compound of formula I and carrier is slightly better than Compound of formula I, but≤10%, the dissolution about 80% for the solid dispersions that compound of formula I is formed with carrier.
The result shows that: the dissolution rate of solid dispersions will be significantly better than the physics of compound of formula I and compound of formula I and carrier Mixture.In addition, PVP K30 is that the solid dispersions of carrier greatly improve the dissolution rate of compound of formula I, it is significantly better than Using other common carriers (such as PEG 6000, hydroxypropyl methylcellulose E5), (dissolved out at 30 minutes also superior to PVP K90 and K25 Spend 65-70%).
The internal medicine generation research of 12 solid dispersions 2 of embodiment
It chooses solid dispersions 2 and carries out internal medicine generation test with corresponding API (i.e. compound of formula I).
86 week old Wistar rats of male, weight 170g or so are divided into 2 groups, every group 4.Animal feeding is in IVC animal In room, 12 hours light and shade alternatings.Using hardwood wood shavings as padding.Animal gives feed and sterilized tap water, free diet. Fasting 16h before administration, 2h restores to food after administration.After overnight fasting, 4mg/kg solid dispersions 1 or corresponding are given in stomach-filling API 1%CMC suspension, administration capacity be 10ml/kg.0.5,1.0,2.0,4.0,6.0,8.0,24,48 and after administration 72h is set in heparinised tubes through rat eye rear vein beard extracting vein blood 0.2ml, and 3500rpm is centrifuged 10min, isolated blood Slurry samples, -20 DEG C of preservations.
The plasma sample content of the LC-MS/MS analysis method quantitative detection API by verifying.Pharmacokinetic parameter It will be calculated based on the blood concentration of every rat in different time points.Parameter is referring to table 2.
The results are shown in Table 2: compound of formula I and PVP K30 are by medicine generation in 2 body of solid dispersions made from 1: 4 weight ratio AUC0-∞About 9.03 times of compound of formula I.
Table 2
13 solid dispersions 10 of embodiment
Formula II compound and PVP K30, weight ratio 1: 4, solvent (dehydrated alcohol) method
The preparation method is the same as that of Example 1, and difference is Formula II compound instead of compound of formula I, Formula II compound and PVP The weight ratio of K30 is 1: 4.
Test results are shown in figure 7 by its XRPD, and in solid dispersions 10, the diffraction maximum (characteristic peak) of Formula II compound is all It disappears, exists with unformed or molecular conformation.
The dissolution rate test of 14 solid dispersions 10 of embodiment
Dissolution test method is the same as embodiment 10.
Specifically, sample is configured into 2 parts (respectively sample 1 and sample 2).It tests according to the method described above respective Dissolution rate, and calculate average value (as average).
Test results are shown in figure 8, when 30min, when the dissolution rate of solid dispersions 10 is about 72.5%, 120min, Gu The dissolution of body dispersion 10 is about 94%.As it can be seen that the dissolution for the solid dispersions 10 that Formula II compound and carrier PVP K30 are formed Degree is fine.
The internal medicine generation research of 15 solid dispersions 10 of embodiment
It chooses solid dispersions 10 and carries out internal medicine generation test with corresponding API (i.e. Formula II compound).
Test method is the same as embodiment 12.
The results are shown in Table 3: in 10 body of solid dispersions of Formula II compound and PVP K30 by the preparation of 1: 4 weight ratio The AUC in medicine generationlastAbout 13.5 times of Formula II compound.
Table 3
16 solid dispersions of embodiment, 1 accelerated stability test
It chooses solid dispersions 1 and carries out accelerated stability test (40 DEG C, 75%RH), 1,2,3 month when takes out progress X-ray powder diffraction.
XRPD result as shown in figure 9, show solid dispersions 1 after 1,2,3 month accelerated stability test, it is highly stable (the X- diffractive features peak of no API, and curve is essentially identical) generates (i.e. original unformed or molecular state without crystal form Do not change), it is still with solid dispersions existing for amorphous or molecular state.
Shown in result figure 9: being successively that compound of formula I, freshly prepd solid dispersions 1, solid dispersions 1 are put from top to bottom XRPD figure when setting one month, when solid dispersions 1 are placed two months, when solid dispersions 1 are placed three months.
17 solid dispersions of embodiment, 2 accelerated stability test
It chooses solid dispersions 2 and carries out accelerated stability test (40 DEG C, 75%RH), 1,2,3 month when takes out progress X-ray powder diffraction.
The results are shown in Figure 10 by XRPD, show solid dispersions 2 after 1,2,3 month accelerated stability test, very surely Fixed (the X- diffractive features peak of no API, and curve is essentially identical) generates (i.e. original unformed or molecularity without crystal form State does not change), it is still with solid dispersions existing for amorphous or molecular state.
Shown in result figure 10: being successively compound of formula I, freshly prepd solid dispersions 2, solid dispersions 2 from top to bottom XRPD figure when placing one month, when solid dispersions 2 are placed two months, when solid dispersions 2 are placed three months.
18 solid dispersions of embodiment, 3 accelerated stability test
It chooses solid dispersions 3 and carries out accelerated stability test (40 DEG C, 75%RH), 1,2,3 month when takes out progress X-ray powder diffraction.
XRPD result is as shown in figure 11, show solid dispersions 3 after 1,2,3 month accelerated stability test, very surely Fixed (the X- diffractive features peak of no API, and curve is essentially identical) generates (i.e. original unformed or molecularity without crystal form State does not change), it is still with solid dispersions existing for amorphous or molecular state.
Shown in result figure 11: being successively compound of formula I, freshly prepd solid dispersions 3, solid dispersions 3 from top to bottom XRPD figure when placing one month, when solid dispersions 3 are placed two months, when solid dispersions 3 are placed three months.
19 solid dispersions of embodiment, 4 accelerated stability test
It chooses solid dispersions 4 and carries out accelerated stability test (40 DEG C, 75%RH), 1,2,3 month when takes out progress X-ray powder diffraction.
XRPD result is as shown in figure 12, show solid dispersions 4 after 1,2,3 month accelerated stability test, very surely Fixed (the X- diffractive features peak of no API, and curve is essentially identical) generates (i.e. original unformed or molecularity without crystal form State does not change), it is still with solid dispersions existing for amorphous or molecular state.
Shown in result figure 12: being successively compound of formula I, freshly prepd solid dispersions 4, solid dispersions 4 from top to bottom XRPD figure when placing one month, when solid dispersions 4 are placed two months, when solid dispersions 4 are placed three months.
Embodiment 20-24 solid dispersions 11-15
Active constituent: compound of formula I
Carrier: the mixed carrier of PVP K30 and other carriers
The preparation method is the same as that of Example 1, and difference is mixed carrier using condition as shown in table 4.
Table 4
Its XRPD test result shows in solid dispersions that the diffraction maximum (characteristic peak) of compound of formula I all disappears, with Unformed form exists.
The internal medicine generation test (operation is the same) of solid dispersions 11-15 shows: the internal medicine generation of solid dispersions AUClastAbout 7-15 times of API.
As it can be seen that the solid dispersions of the carrier prepared by the present invention comprising active constituent and the K30 containing PVP are highly stable;And For active constituent, solid dispersions of the invention have the dissolution rate significantly improved, solid especially of the present invention point The area under the drug-time curve of granular media is 6-20 times of area under the drug-time curve when its active constituent is administered alone, preferably 7-15 Times, it is more preferably 9-14 times.
All references mentioned in the present invention is incorporated herein by reference, independent just as each document It is incorporated as with reference to such.In addition, it should also be understood that, after reading the above teachings of the present invention, those skilled in the art can To make various changes or modifications to the present invention, such equivalent forms equally fall within model defined by the application the appended claims It encloses.

Claims (15)

1. a kind of solid dispersions, which is characterized in that be made of following components (A) and component (B):
(A) as the Formulas I of active constituent or Formula II compound or its pharmaceutically acceptable salt;
And
(B) hydrophilic macromolecular carrier;The hydrophilic macromolecular carrier is PVP K30;
Wherein, the weight ratio of (A) and (B) are 1:3-1:4.
2. solid dispersions as described in claim 1, which is characterized in that the weight ratio of (A) and (B) are 1:4.
3. solid dispersions as described in claim 1, which is characterized in that the compound is Formula II compound, and (A) and (B) weight ratio is 1:4.
4. solid dispersions as described in claim 1, which is characterized in that the compound is compound of formula I, and (A) and (B) Weight ratio be 1:4.
5. solid dispersions as described in claim 1, which is characterized in that the X-ray powder diffraction figure of the solid dispersions In, the X-ray diffraction characteristic peak of the active constituent without the solid dispersions.
6. a kind of preparation method of solid dispersions described in claim 1, which is characterized in that including method:
(a) fusion method 1, comprising steps of
(a1) mixed active ingredient and hydrophilic macromolecular carrier, to obtain a mixture containing active constituent;
(a2) mixture containing active constituent that heating melting steps (a1) obtain, thus the mixture after being melted;With
(a3) mixture after the melting that cooling step (a2) obtains, to obtain solid dispersions described in claim 1;
Or
(b) fusion method 2, comprising steps of
(b1) heating melting hydrophilic macromolecular carrier, thus the carrier after being melted;
(b2) by after melting that step (b1) obtains carrier and active constituent mix after melt, thus the mixing after being melted Object;With
(b3) mixture after the melting that cooling step (b2) obtains, to obtain solid dispersions described in claim 1;
Or
(c) solvent method, comprising steps of
(c1) by active constituent and hydrophilic macromolecular carrier co-dissolve in solvent, to obtain a mixture;With
(c2) solvent in the mixture that step (c1) is obtained is removed, to obtain solid dispersions described in claim 1;
Wherein, the active constituent is Formulas I or Formula II compound or its pharmaceutically acceptable salt,
The hydrophilic macromolecular carrier is PVP K30,
The solvent is selected from the group: methanol, ethyl alcohol, isopropanol, acetone, methylene chloride, tetrahydrofuran, or combinations thereof.
7. preparation method as claimed in claim 6, which is characterized in that the step (c2) is comprising steps of use Rotary Evaporators Remove the solvent in the mixture that step (c1) is obtained;Or the mixture that step (c1) obtains is dried and removed with spray dryer In solvent;Wherein, the technological parameter of the drying are as follows: 110 DEG C~120 DEG C of inlet air temperature, power of fan 55%~65% is compacted Dynamic pump power 50%~55%.
8. preparation method as claimed in claim 6, which is characterized in that in addition the solid dispersions further use at least one Procedure of processing processing, the procedure of processing is to grind, sieve, roll, mill, screen, mix or combinations thereof.
9. a kind of pharmaceutical composition, which is characterized in that the composition includes:
(1) the described in any item solid dispersions of claim 1-5;With
(2) pharmaceutically acceptable excipient.
10. pharmaceutical composition as claimed in claim 9, which is characterized in that the excipient includes: filler, diluent, sweet tea Taste agent, corrigent, antioxidant, colorant, preservative, lubricant, adhesive, disintegrating agent or its mixing.
11. pharmaceutical composition as claimed in claim 9, which is characterized in that the composition is capsule, tablet, pill, dissipates Agent and granule.
12. pharmaceutical composition as claimed in claim 9, which is characterized in that described pharmaceutical composition, for inhibiting phosphokinase Pharmaceutical composition.
13. pharmaceutical composition as claimed in claim 9, which is characterized in that the pharmaceutical composition is for treating and preventing Cancer.
14. a kind of preparation method of pharmaceutical composition as claimed in claim 9, which is characterized in that by any one of claim 1-5 The solid dispersions and the mixing of pharmaceutically acceptable excipient, so that the pharmaceutical composition be made.
15. a kind of purposes of solid dispersions as described in claim 1, which is characterized in that be used to prepare inhibition phosphokinase Drug;Or it is used to prepare the drug for the treatment of tumour.
CN201210068420.4A 2012-03-15 2012-03-15 It is a kind of improve absorbent properties solid dispersions and its preparation Active CN103301066B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201210068420.4A CN103301066B (en) 2012-03-15 2012-03-15 It is a kind of improve absorbent properties solid dispersions and its preparation
PCT/CN2013/072645 WO2013135189A1 (en) 2012-03-15 2013-03-14 Solid dispersion improving absorption performance and preparation of same

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201210068420.4A CN103301066B (en) 2012-03-15 2012-03-15 It is a kind of improve absorbent properties solid dispersions and its preparation

Publications (2)

Publication Number Publication Date
CN103301066A CN103301066A (en) 2013-09-18
CN103301066B true CN103301066B (en) 2018-12-07

Family

ID=49127077

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201210068420.4A Active CN103301066B (en) 2012-03-15 2012-03-15 It is a kind of improve absorbent properties solid dispersions and its preparation

Country Status (2)

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

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023155307A1 (en) 2022-02-21 2023-08-24 北京睿创康泰医药研究院有限公司 Sorafenib or donafenib oral preparation with low dose and high drug exposure, and application thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104557687A (en) * 2013-10-25 2015-04-29 苏州泽璟生物制药有限公司 Fluorine-containing deuterated omega-diphenylurea hydrate and crystal form substance 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

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101048140A (en) * 2004-08-27 2007-10-03 拜耳制药公司 Novel pharmaceutical compositions for the treatment of cancer
CN101287463A (en) * 2004-08-27 2008-10-15 拜耳制药公司 New pharmaceutical compositions comprising 4-(4-(3-(4-chloro-3-trifluoromethyl-phenyl)-ureido)-3-fluoro-phenoxy)-pyridine-2-carboxylic acid for the treatment of hyper-proliferative disorders
CN101676266A (en) * 2008-09-19 2010-03-24 苏州泽璟生物制药有限公司 Deuterium-substituted omega-diphenylurea and its derivatives, as well as pharmaceutical composition containing the same

Family Cites Families (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
CN102459180B (en) * 2009-06-09 2014-05-14 江苏迈度药物研发有限公司 Urea derivatives as kinase inhibitors
CN102199121B (en) * 2010-05-27 2013-10-30 四川大学 4-(4-(3-trifluoro methyl) benzamido phenoxyl)-2-(methyl carbamyl) pyridine salt, its preparation method and application

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101048140A (en) * 2004-08-27 2007-10-03 拜耳制药公司 Novel pharmaceutical compositions for the treatment of cancer
CN101287463A (en) * 2004-08-27 2008-10-15 拜耳制药公司 New pharmaceutical compositions comprising 4-(4-(3-(4-chloro-3-trifluoromethyl-phenyl)-ureido)-3-fluoro-phenoxy)-pyridine-2-carboxylic acid for the treatment of hyper-proliferative disorders
CN101676266A (en) * 2008-09-19 2010-03-24 苏州泽璟生物制药有限公司 Deuterium-substituted omega-diphenylurea and its derivatives, as well as pharmaceutical composition containing the same

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023155307A1 (en) 2022-02-21 2023-08-24 北京睿创康泰医药研究院有限公司 Sorafenib or donafenib oral preparation with low dose and high drug exposure, and application thereof

Also Published As

Publication number Publication date
WO2013135189A1 (en) 2013-09-19
CN103301066A (en) 2013-09-18

Similar Documents

Publication Publication Date Title
CN103301066B (en) It is a kind of improve absorbent properties solid dispersions and its preparation
US11890270B2 (en) Polymorphic forms of ST-246 and methods of preparation
JP5288791B2 (en) Miniaturized composition containing a hardly water-soluble substance
EP2599486B1 (en) Dronedarone solid dispersion and preparation method thereof
EP3299014B1 (en) Somcl-9112 solid dispersion and preparation method thereof and somcl-9112 solid preparation containing somcl-9112 solid dispersion
US20140039031A1 (en) Pharmaceutical formulations of acetyl-11-keto-b-boswellic acid, diindolylmethane, and curcumin for pharmaceutical applications
NZ505245A (en) Pharmaceutical compositions of itraconazole and a water-soluble polymer to treat fungal infection
CN108472291A (en) Combination treatment for treating malignant tumour
KR100381834B1 (en) Solid dispersion system of pranlukast with improved dissolution, and the method thereof
WO2015145145A1 (en) Pharmaceutical composition comprising lapatinib
CN103301067B (en) It is a kind of improve absorbent properties solid dispersions and its preparation
JP2020518611A (en) Compositions with improved water solubility and bioavailability
EP3981469A1 (en) Therapeutic for gout or hyperuricemia
CA2253769C (en) Pharmaceutical compositions comprising fenofibrate
CN105582546A (en) Compound enteric-coated tablets of entecavir phospholipid complex and diammonium glycyrrhizinate
KR100981750B1 (en) Spray-dried granules and processes for the preparation thereof
ES2231043A1 (en) Instant release pharmaceutical consists of a cellulose disaggregating excipient coated with non micronized solution

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
C10 Entry into substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CP03 Change of name, title or address
CP03 Change of name, title or address

Address after: 215300 Chenfeng Road 209, Yushan Town, Kunshan City, Suzhou City, Jiangsu Province

Patentee after: Suzhou Zehuang Biopharmaceutical Co., Ltd.

Address before: 215300 Chenfeng Road 209, Kunshan City, Suzhou City, Jiangsu Province

Patentee before: Suzhou Zelgen Biopharmaceuticals Co., Ltd.

PP01 Preservation of patent right
PP01 Preservation of patent right

Effective date of registration: 20190613

Granted publication date: 20181207

PD01 Discharge of preservation of patent
PD01 Discharge of preservation of patent

Date of cancellation: 20190816

Granted publication date: 20181207