WO2022120512A1 - 一种高生物利用度的索拉非尼药物组合物及应用 - Google Patents

一种高生物利用度的索拉非尼药物组合物及应用 Download PDF

Info

Publication number
WO2022120512A1
WO2022120512A1 PCT/CN2020/134164 CN2020134164W WO2022120512A1 WO 2022120512 A1 WO2022120512 A1 WO 2022120512A1 CN 2020134164 W CN2020134164 W CN 2020134164W WO 2022120512 A1 WO2022120512 A1 WO 2022120512A1
Authority
WO
WIPO (PCT)
Prior art keywords
sorafenib
sodium
oral solid
solid preparation
preparation
Prior art date
Application number
PCT/CN2020/134164
Other languages
English (en)
French (fr)
Inventor
贾慧娟
张加晏
侯鑫
李衍
Original Assignee
天津睿创康泰生物技术有限公司
北京睿创康泰医药研究院有限公司
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 天津睿创康泰生物技术有限公司, 北京睿创康泰医药研究院有限公司 filed Critical 天津睿创康泰生物技术有限公司
Priority to CN202080055010.9A priority Critical patent/CN114916221B/zh
Priority to JP2022516367A priority patent/JP7428356B2/ja
Priority to US17/630,709 priority patent/US20230310393A1/en
Priority to EP20946527.7A priority patent/EP4032529A4/en
Priority to CA3155855A priority patent/CA3155855A1/en
Priority to PCT/CN2020/134164 priority patent/WO2022120512A1/zh
Publication of WO2022120512A1 publication Critical patent/WO2022120512A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • 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
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • 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

Definitions

  • Nexavar Sorafenib is the first multi-targeted oral tumor-targeted drug jointly developed by Bayer and Onyx for the treatment of advanced liver cancer, renal cell carcinoma and thyroid cancer. Sorafenib can act on tumor cells and intratumor blood vessels at the same time, and play a role in inhibiting tumor growth through mechanisms such as anti-cell proliferation, promotion of apoptosis, and inhibition of angiogenesis. On the one hand, sorafenib exerts an anti-cell proliferation effect by inhibiting c-Raf kinase and downstream signaling, hindering the phosphorylation process of MEK and ERK, and reducing the phosphorylation level of ERK.
  • initiation factor-4E initiation factor-4E
  • Mcl-1 anti-apoptotic protein
  • sorafenib The structure of sorafenib is as follows:
  • sorafenib in adults and children, the inter-individual plasma concentration and AUC vary greatly, indicating that after oral administration, various factors in the gastrointestinal tract affect the dissolution rate of the drug, which in turn affects the drug's efficacy. Absorption is the main cause of high drug variability.
  • sorafenib In order to improve the drug dissolution rate of sorafenib, the prior art uses a polymer carrier to prepare a sorafenib solid dispersion pharmaceutical composition, which initially has a higher dissolution rate and dissolution rate, but because the solubility of sorafenib is very high. At low levels (pH 1, 0.0034 mg/ml, pH 4.5, 0.00013 mg/ml), sorafenib crystallized rapidly with increasing dissolution time, resulting in a sharp decrease in dissolution rate.
  • the prior art also has the reports of cosolvents such as tocopherol compounds, polyalcohol fatty acid esters, polyalkoxylated fatty alcohol ethers, hydrogenated castor oil, etc., because these cosolvents have lower melting points. On the one hand, it improves the difficulty of the preparation and production process, and on the other hand, it will lead to a decrease in the physical stability of the sorafenib solid dispersion. During the long-term storage of the pharmaceutical composition, the amorphous sorafenib with poor thermodynamic stability It is easy to be induced by a cosolvent to accelerate the crystallization, so that the purpose of greatly improving the bioavailability of the drug and reducing the dosage cannot be achieved.
  • cosolvents such as tocopherol compounds, polyalcohol fatty acid esters, polyalkoxylated fatty alcohol ethers, hydrogenated castor oil, etc.
  • the technical problem to be solved by the present invention is to provide a sorafenib pharmaceutical composition with high bioavailability and its application, and the prepared sorafenib pharmaceutical composition has high dissolution rate and stability.
  • the present invention provides a high bioavailability Sorafenib pharmaceutical composition, comprising:
  • Vectors include VA64 and HPMCAS.
  • the sorafenib is its tosylate salt, namely sorafenib tosylate.
  • the mass ratio of VA64 and HPMCAS in the component a) and the component b) is 1:(0.1-5):(0.01-5), more preferably 1:(0.5-3) : (0.1 to 1).
  • the mass ratio of VA64 and HPMCAS in the component a) and the component b) is 1:1:0.1 to 1:1:0.5, more preferably 1:1:0.25.
  • the sorafenib pharmaceutical composition is a solid dispersion.
  • the solid dispersion is prepared according to a spray drying method, and the solvent of the spray drying method is a mixed solvent of methanol and dichloromethane.
  • the volume ratio of methanol and dichloromethane is 1:(1-4).
  • the mass-to-volume ratio (w/v) of the component a) and the methanol-dichloromethane mixed solvent is preferably 1:25-300, more preferably 1:50-150, and most preferably 1:100.
  • the inlet air temperature of the spray drying method is preferably 120°C, and the outlet air temperature is preferably 60°C, and nitrogen is preferably used as the carrier gas.
  • the present invention also provides a low-dose oral solid preparation of sorafenib, comprising:
  • the present invention adopts the above-mentioned crystal inhibitor, especially polyvinylpyrrolidone, and the solid dispersion of sorafenib to form a composition to prepare a solid preparation, which ensures that the preparation has a good dissolution rate and avoids the crystallization of sorafenib.
  • the polyvinylpyrrolidone is preferably PVPK30, PVPK25, PVPK60, PVPK90, PVPK12, PVPK15 and PVPK17, more preferably PVPK30 and PVPK25.
  • the mass content of the polyvinylpyrrolidone is 1% to 40%; more preferably 1% to 20%, further preferably 10% to 20%, and most preferably 12.5%.
  • the crystal inhibitor can also be selected from hydroxypropyl methylcellulose acetate succinate (HPMCAS), including but not limited to HPMCAS HG (or the same specification, such as 126G), HPMCAS MG (or the same specification, such as 912G), HPMCAS LG (or the same specification, such as 716G).
  • HPMCAS HG or the same specification, such as 126G
  • HPMCAS MG or the same specification, such as 912G
  • HPMCAS LG or the same specification, such as 716G.
  • the mass content of the hydroxypropyl methylcellulose acetate succinate is 1% to 40%; more preferably 1% to 20%, and most preferably 1 to 10%.
  • the crystal inhibitor can also be selected from bile salt compounds, including but not limited to: sodium glycocholate, sodium taurocholate, sodium deoxyglycocholate, sodium chenodeoxyglycocholate, One or more in sodium ursodeoxyglycylcholate, sodium deoxytaurocholate, sodium ursodeoxytaurocholate and sodium dodecylsulfonate, preferably any one in STC, SDC, SGC or more.
  • bile salt compounds including but not limited to: sodium glycocholate, sodium taurocholate, sodium deoxyglycocholate, sodium chenodeoxyglycocholate, One or more in sodium ursodeoxyglycylcholate, sodium deoxytaurocholate, sodium ursodeoxytaurocholate and sodium dodecylsulfonate, preferably any one in STC, SDC, SGC or more.
  • the mass content of the bile salt compound is 1%-40%; more preferably 1%-20%, most preferably 1-10%.
  • the sorafenib solid dispersion comprises:
  • Sorafenib or a salt, hydrate, solvate, hydrate or solvate of a salt thereof;
  • the vector includes VA64.
  • the mass ratio of the component a) and VA64 is 1:(0.1-5), more preferably 1:(0.5-3), and even more preferably 1:(1-3).
  • the mass ratio of the component a) and VA64 is 1:1.
  • the sorafenib solid dispersion comprises:
  • Sorafenib or a salt, hydrate, solvate, hydrate or solvate of a salt thereof;
  • the present invention adopts VA64 and HPMCAS as the carrier of the sorafenib solid dispersion, and the prepared solid dispersion has high stability and dissolution rate.
  • the mass ratio of the component a) to VA64 and HPMCAS is 1:(0.1 ⁇ 5):(0.01 ⁇ 5), more preferably 1:(0.5 ⁇ 3):(0.1 ⁇ 1) .
  • the mass ratio of the component a) to VA64 and HPMCAS is 1:1:(0.1-0.5), more preferably 1:1:0.25.
  • the solid dispersion is prepared according to a spray drying method, and the solvent of the spray drying method is a mixed solvent of methanol and dichloromethane.
  • the volume ratio of methanol and dichloromethane is 1:(1-4).
  • the present invention adopts the mixed solvent of methanol and dichloromethane as the solvent of the spray drying method to prepare the sorafenib solid dispersion, so that the prepared solid dispersion has better stability.
  • methanol and dichloromethane mixed solvent especially methanol-dichloromethane mixed solvent with a volume ratio of 1:3, sorafenib has high solubility, and it is placed in this organic solvent for 30min and 24h. Substances did not change significantly.
  • the mass-to-volume ratio (w/v) of the component a) and the methanol-dichloromethane mixed solvent is preferably 1:25-300, more preferably 1:50-150, and most preferably 1:100.
  • the inlet air temperature of the spray drying method is preferably 120°C, and the outlet air temperature is preferably 60°C, and nitrogen is preferably used as the carrier gas.
  • the unit dose of the component a) is 70-200 mg; preferably 70-140 mg; more preferably 70-130 mg.
  • the unit dose refers to the amount of the main drug contained in the smallest unit of the drug; for example, the weight of the drug contained in one tablet, one capsule, or one bag of granules.
  • the unit of the unit dose is mg/tablet.
  • the present invention reduces the dosage of sorafenib by improving the stability and dissolution of sorafenib, and patients can achieve the same treatment as Nexavar tablets by taking 35% to 70% of the dosage of oral Nexavar tablets. Effect.
  • the adjuvant is selected from one or more of fillers, disintegrants, binders, glidants, lubricants, and flavoring agents.
  • the mass content of the filler is preferably 20%-50%, more preferably 30%-40%.
  • the disintegrant is selected from corn starch, partially ⁇ -starch, hydroxypropyl starch, carboxymethyl cellulose, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, sodium carboxymethyl starch , one or more of low-substituted hydroxypropyl cellulose, croscarmellose sodium, crospovidone; more preferably crospovidone, sodium carboxymethyl starch or low-substituted hydroxypropyl cellulose white.
  • the mass content of the disintegrant is preferably 3%-20%, more preferably 5%-10%.
  • the binder is selected from one or more of hydroxypropyl cellulose, hypromellose, povidone, starch pulp, and sodium carboxymethyl cellulose.
  • the mass content of the binder is preferably 0-10%, more preferably 0-5%.
  • the glidant is selected from one or more of talc and silicon dioxide; preferably silicon dioxide.
  • the mass content of the silica is preferably 2%-20%, more preferably 5%-15%, and most preferably 10%.
  • the disintegration of the preparation is accelerated, thereby improving the dissolution rate of the preparation.
  • the lubricant is selected from magnesium stearate, stearic acid, calcium stearate, sodium stearyl fumarate, polyethylene glycol, hydrogenated vegetable oil, polyethylene glycol, lauryl sulfate One or more of sodium, talc, and silicon dioxide.
  • the mass content of the lubricant is preferably 0.3%-5%, more preferably 0.5%-2%, and most preferably 0.5%-1%.
  • the dosage form of the oral solid preparation is tablet, granule, dry suspension, capsule or film.
  • Preparation process Weigh the raw and auxiliary materials in the prescribed amount, pass the added raw and auxiliary materials together through a 50-mesh sieve for 6 times and mix, dry granulation, weigh the weight of the granules, convert the amount of the added materials, weigh the added materials, mix for 5min, and press into tablets. Cumulative dissolution was measured.
  • the formulation with VA-64 as the carrier and the addition of bile salts greatly improved the dissolution of sorafenib solid dispersion tablets within 60 minutes, but decreased to the same level as Nexavar after 60 minutes.
  • the crystal inhibition effect does not last as long as the PVP series.
  • Preparation process Weigh the raw and auxiliary materials in the prescribed amount, pass the added raw and auxiliary materials together through a 50-mesh sieve for 6 times and mix, dry granulation, weigh the weight of the granules, convert the amount of the added materials, weigh the added materials, mix for 5min, and press into tablets. Check the dissolution profile.
  • the dissolution method is the same as before.
  • Tablets were prepared according to the formulation shown in Table 21.
  • the preparation method is the same as before.
  • the mass ratio of SLFN and VA64 is 1:1.
  • Tablets were prepared according to the formulation shown in Table 23.
  • the preparation method is the same as before.
  • the mass ratio of SLFN-VA64-HPMCAS is 1:1:0.25.
  • Preparation process Weigh the raw and auxiliary materials in the prescribed amount, pass the added raw and auxiliary materials together through a 50-mesh sieve for 6 times and mix, dry granulation, weigh the weight of the granules, convert the amount of the added materials, weigh the added materials, mix for 5min, and press into tablets.
  • Preparation process Weigh the raw and auxiliary materials in the prescribed amount, pass the added raw and auxiliary materials together through a 50-mesh sieve for 6 times and mix, dry granulation, weigh the weight of the granules, convert the amount of the added materials, weigh the added materials, mix for 5min, and press into tablets.
  • Animal Adult male Beagle (8-11kg), can be Divide into 4 groups with 2 beagle dogs in each group.
  • Administration route and administration frequency 1 tablet orally by oral gavage, fasting for more than 16 hours before administration, and free food after administration for 4 hours; washout period is 4 days. 4 cycles of cross-dosing.
  • LC/MS/MS method was used to detect blood drug concentration, and Phoenix WinNonlin 7.0 was used to calculate pharmacokinetic parameters (AUC 0-t , AUC 0- ⁇ , C max , T 1/2 , T max and the arithmetic mean of these parameters ( ⁇ SD) and geometric mean).
  • 20200509-1 and 20200519-3 are significantly higher than 20200509-2.
  • the 100mg dose of the two is basically the same as the reference preparation 200mg. Comparing the CVs of the reference and the test product, 20200509-1 and 20200509-3 were significantly better than the reference preparation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Nutrition Science (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Physiology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

一种低剂量索拉非尼口服固体制剂,包括:a)索拉非尼固体分散体;b)抑晶剂;c)其他药学上可接受的辅剂。上述低剂量索拉非尼口服固体制剂具有较高的生物利用度,降低了索拉非尼的服用剂量,病人口服多吉美片剂给药剂量的35%~70%即可达到与多吉美片剂相同的治疗效果;同时具有更高的稳定性,更安全、副作用发生率更少;具有更低的C max、AUC 0-t变异,更高的溶出度,随胃肠道pH的升高,析晶速率低;片剂体积小,病人易于服用;崩解速度快,溶出效果好;易于实现产业化。

Description

一种高生物利用度的索拉非尼药物组合物及应用 技术领域
本发明涉及药物制剂技术领域,尤其涉及一种高生物利用度的索拉非尼药物组合物及应用。
背景技术
Nexavar索拉非尼片是由德国拜耳公司与Onyx公司联合开发的用于治疗晚期肝癌、肾细胞癌和甲状腺癌的第一个多靶点口服肿瘤靶向药物。索拉非尼可同时作用于肿瘤细胞和肿瘤内血管,通过抗细胞增殖、促进细胞凋亡、抑制血管生成等机制而发挥抑制肿瘤生长的作用。索拉非尼一方面通过抑制c-Raf激酶及下游信号传导、阻碍MEK和ERK的磷酸化过程、降低ERK磷酸化水平,发挥抗细胞增殖的作用。另一方面索拉非尼通过抑制血管内皮细胞生长因子受体-2(VEGFR-2)、血管内皮细胞生长因子受体-3(VEGFR-3)和血小板衍生生长因子受体-β(PDGFR-β),抑制络氨酸激酶受体的自身磷酸化过程而发挥抗血管生成的作用。它具有双重的抗肿瘤作用:既可通过阻断由RAF/MEK/ERK介导的细胞信号传导通路而直接抑制肿瘤细胞的增殖,还可通过抑制VEGFR和血小板衍生生长因子(PDGF)受体而阻断肿瘤新生血管的形成。同时抑制起始因子-4E(eIF4E)的磷酸化过程、下调体内抗凋亡蛋白Mcl-1的水平,发挥促细胞凋亡的作用。
索拉非尼是一种双芳基脲类口服多激酶抑制剂,其化学名为4-{4-[3-(4-氯-3-三氟甲基-苯基)酰脲]-苯氧基}-吡啶-2-羧酸甲胺,相对分子质量为464.8,结构式如式(1)所示。Nexavar索拉非尼片使用的活性成分为索拉非尼的甲苯磺酸盐,分子式为C 21H 16ClF 3N 4O 3·C 7H 8O 3S见式(2)。
索拉非尼结构如下:
Figure PCTCN2020134164-appb-000001
根据生物药剂学分类系统,索拉非尼为BCS II类药物,为低溶解性高渗透性药物。索拉非尼在pH1.2~pH7.4的水溶液中的溶解度均非常低,这导致索拉非尼片剂在胃肠道中溶出速率非常低,并成为吸收的限速步骤,再加上首过 代谢作用,索拉非尼片剂口服后生物利用度较低,与口服溶液相比,索拉非尼片剂的平均相对生物利用度仅为38%-49%。因此,多吉美索拉非尼片的规格为200mg,每次服用两片,一日两次,日最大服用剂量为800mg。根据Nexavar索拉非尼片说明书,健康志愿者口服100mg 14C放射性标记的索拉非尼,给药剂量的77%经粪便排出,并且排泄物中原型药物占51%以上,这说明Nexavar索拉非尼片口服生物利用度低,有一半以上给药剂量的药物因为溶解度原因而经粪便排出体外。
Oncologist 2007;12:426–37和Ann Oncol 2005;16:1688–94披露的索拉非尼临床实验结果显示,索拉非尼人体药代动力学参数具有高的个体变异性,口服200mg或400mg,一日两次,Cmax和AUC的变异系数分别为33%–88%和5%–83%。Oncotarget,2017,Vol.8,(26),pp:43458-43469披露一项由94个肾细胞癌病人开展的临床试验,其中74人口服400mg索拉非尼片,分两次服用,另外18人口服600mg索拉非尼片,分三次服用;还有2人口服400mg索拉非尼片,一次服用。治疗2周后,索拉非尼稳态血药(血浆)浓度为881 to 12,526ng/mL,主要不良反应的发生率如下:腹泻(76.5%)、手足综合症(68.99%)、疲劳(55.32%),这些严重不良反应均与索拉非尼的血药浓度有关。那些发生严重不良反应的病人的血药浓度均超过10000ng/ml,而且这些严重的不良反应会随着给药剂量降低或停药而减少。这说明,这些严重的副作用均与给药剂量相关。Invest New Drugs.2012 April;30(2):524–528对成年恒河猴静脉注射索拉非尼,评价血浆和脑脊液中索拉非尼的药代动力学行为,恒河猴单次静脉注射索拉非尼,脑脊液中最大血药浓度为0.00045~0.00058μg/mL,AUC 0-24h为0.0048to 0.0016μg·h/mL,个体间变异很小。相比之下,成人和儿童口服索拉非尼后个体间的血药浓度和AUC却变异很大,说明口服给药后,胃肠道中多种因素影响药物的溶出速率,并进而影响药物的吸收是造成药物高变异的主要原因。
Cancer Treatment Reviews,2016指出溶出速率低是口服索拉非尼片后生物利用度低、血药浓度、暴漏量高变异、安全性或无效的主要因素。
现有技术为了提高索拉非尼片剂的口服生物利用度进行了各种方法的尝试和研究,但至今Nexavar索拉非尼片的核心专利基本都已经失效,尚未有一 款新的产品进入商业化。
为提高索拉非尼的药物溶出度,现有技术采用高分子载体制备索拉非尼固体分散体药物组合物,开始有较高的溶出速率和溶出度,但由于索拉非尼的溶解度非常低(pH1,0.0034mg/ml、pH4.5,0.00013mg/ml),随着溶出时间延长,索拉非尼快速结晶析出,从而导致溶出度急剧降低。在含0.1%SDS的pH6.8磷酸盐缓冲液作为溶出介质的溶出实验中,37℃,2h-4h时,累积溶出度下降到与多吉美索拉非尼片基本一致,甚至更低,从而导致固体分散体的药物组合物体内生物利用度提高的并不显著,也无法大幅降低日服药剂量。
现有技术还有固体分散体再加助溶剂,如生育酚类化合物,多元醇脂肪酸酯、聚烷氧基化脂肪醇醚、氢化蓖麻油等助溶剂的报道,由于这些助溶剂熔点较低,一方面提高了制备生产工艺的难度,另一方面会导致索拉非尼固体分散体的物理稳定性降低,药物组合物在长期贮存过程中,热力学稳定性不好的无定形索拉非尼容易受助溶剂诱导而加速结晶析出,从而无法达到大幅度提高药物的生物利用度,降低给药剂量的目的。
索拉非尼本身稳定性较差,容易降解生成诱变杂质CTF-苯胺,制备成固体分散体后更易产生CTF-苯胺。CTF-苯胺的结构见式(3):
Figure PCTCN2020134164-appb-000002
另外,Nexavar索拉非尼片规格较大,制备成固体分散体需加入大量高分子材料,导致制剂成型难度较大,如片剂、胶囊剂崩解时间大大延长,而为了改善崩解,需加入大量制剂辅料,导致片剂或胶囊内容物体积、重量较大,做成的制剂不利于患者服用,导致顺应性差。
因为以上问题的存在,导致针对Nexavar索拉非尼片的改良制剂产品一直无法进入产业化而用于临床。
发明内容
有鉴于此,本发明要解决的技术问题在于提供一种高生物利用度的索拉非尼药物组合物及应用,制备的索拉非尼药物组合物具有较高的溶出度和稳定性。
为达到上述目的,本发明提供了一种高生物利用度的索拉非尼药物组合物,包括:
a)索拉非尼、或其盐、水合物、溶剂化物、盐的水合物或溶剂化物;
b)载体:所述载体包括VA64和HPMCAS。
在本发明的一些具体实施例中,所述索拉非尼为其甲苯磺酸盐,即甲苯磺酸索拉非尼。
本发明优选的,所述组分a),和组分b)中的VA64、HPMCAS的质量比为1:(0.1~5):(0.01~5),更优选为1:(0.5~3):(0.1~1)。
更优选的,所述组分a),和组分b)中的VA64、HPMCAS的质量比为1:1:0.1~1:1:0.5,进一步优选为1:1:0.25。
本发明优选的,所述索拉非尼药物组合物为固体分散体。
本发明优选的,所述固体分散体按照喷雾干燥法制备,所述喷雾干燥法的溶剂为甲醇和二氯甲烷的混合溶剂。
本发明优选的,所述甲醇和二氯甲烷的体积比为1:(1~4)。
更优选为1:3。
本发明采用甲醇和二氯甲烷的混合溶剂作为喷雾干燥法的溶剂,制备索拉非尼固体分散体,使制备的固体分散体具有更好的稳定性。实验表明,采用甲醇和二氯甲烷混合溶剂,尤其是体积比1:3的甲醇-二氯甲烷混合溶剂,索拉非尼具有较高的溶解度,且在此有机溶剂中放置30min和24h,有关物质均无明显变化。
所述组分a),和甲醇-二氯甲烷混合溶剂的质量体积比(w/v)优选为1:25~300,更优选为1:50~150,最优选为1:100。
本发明中,所述喷雾干燥法的进风温度优选为120℃,出风温度优选为60℃,优选采用氮气作为载气。
本发明还提供了一种低剂量索拉非尼口服固体制剂,包括:
a)索拉非尼固体分散体;
b)抑晶剂;
c)其他药学上可接受的辅剂。
本发明优选的,所述抑晶剂,选自聚乙烯吡咯烷酮、醋酸羟丙甲基纤维素 琥珀酸酯(HPMCAS)、甘氨胆酸钠、牛磺胆酸钠、脱氧甘氨胆酸钠、鹅脱氧甘氨胆酸钠、熊脱氧甘氨胆酸钠、脱氧牛磺胆酸钠和熊脱氧牛磺胆酸钠及十二烷基磺酸钠中的一种或多种。
本发明采用上述抑晶剂,尤其是聚乙烯吡咯烷酮,与索拉非尼固体分散体形成组合物,制备固体制剂,保证了制剂具有很好的溶出度,避免了索拉非尼的结晶析出。
本发明中,所述聚乙烯吡咯烷酮优选为PVPK30、PVPK25、PVPK60、PVPK90、PVPK12、PVPK15、PVPK17,更优选为PVPK30、PVPK25。
本发明优选的,所述聚乙烯吡咯烷酮的质量含量为1%~40%;更优选为1%~20%,进一步优选为10%~20%,最优选为12.5%。
本发明中,所述抑晶剂还可以选自醋酸羟丙甲基纤维素琥珀酸酯(HPMCAS),包括但不限于HPMCAS HG(或相同规格,如126G)、HPMCAS MG(或相同规格,如912G)、HPMCAS LG(或相同规格,如716G)中的一种或多种。
本发明优选的,所述醋酸羟丙甲基纤维素琥珀酸酯(HPMCAS)的质量含量为1%~40%;更优选为1%~20%,最优选为1~10%。
本发明中,所述抑晶剂还可以选自胆盐类化合物,包括但不限于:甘氨胆酸钠、牛磺胆酸钠、脱氧甘氨胆酸钠、鹅脱氧甘氨胆酸钠、熊脱氧甘氨胆酸钠、脱氧牛磺胆酸钠和熊脱氧牛磺胆酸钠及十二烷基磺酸钠中的一种或多种,优选为STC、SDC、SGC中的任意一种或多种。
本发明优选的,所述胆盐类化合物的质量含量为1%~40%;更优选为1%~20%,最优选为1~10%。
本发明优选的,所述索拉非尼固体分散体包括:
a)索拉非尼、或其盐、水合物、溶剂化物、盐的水合物或溶剂化物;
b)载体:所述载体包括VA64。
本发明采用VA64作为索拉非尼固体分散体的载体,制备的固体分散体具有较高的稳定性。
本发明优选的,所述组分a),和VA64的质量比为1:(0.1~5),更优选为1:(0.5~3),进一步优选为1:(1-3)。
更优选的,所述组分a),和VA64的质量比为1:1。
本发明优选的,所述索拉非尼固体分散体包括:
a)索拉非尼、或其盐、水合物、溶剂化物、盐的水合物或溶剂化物;
b)载体:所述载体包括VA64和HPMCAS。
本发明采用VA64和HPMCAS作为索拉非尼固体分散体的载体,制备的固体分散体具有较高的稳定性和溶出度。
本发明优选的,所述组分a),和VA64、HPMCAS的质量比为1:(0.1~5):(0.01~5),更优选为1:(0.5~3):(0.1~1)。
更优选的,所述组分a),和VA64、HPMCAS的质量比为1:1:(0.1~0.5),进一步优选为1:1:0.25。
本发明优选的,所述固体分散体按照喷雾干燥法制备,所述喷雾干燥法的溶剂为甲醇和二氯甲烷的混合溶剂。
本发明优选的,所述甲醇和二氯甲烷的体积比为1:(1~4)。
更优选为1:3。
本发明采用甲醇和二氯甲烷的混合溶剂作为喷雾干燥法的溶剂,制备索拉非尼固体分散体,使制备的固体分散体具有更好的稳定性。实验表明,采用甲醇和二氯甲烷混合溶剂,尤其是体积比1:3的甲醇-二氯甲烷混合溶剂,索拉非尼具有较高的溶解度,且在此有机溶剂中放置30min和24h,有关物质均无明显变化。
所述组分a),和甲醇-二氯甲烷混合溶剂的质量体积比(w/v)优选为1:25~300,更优选为1:50~150,最优选为1:100。
本发明中,所述喷雾干燥法的进风温度优选为120℃,出风温度优选为60℃,优选采用氮气作为载气。
本发明优选的,所述索拉非尼口服固体制剂中,所述组分a)的单位剂量为70~200mg;优选70~140mg;更优选为70-130mg。
本发明中,所述单位剂量指药品最小单元所含主药的量;如一片,一颗胶囊,一袋颗粒中所含的药品的重量。例如,当所述复方制剂为片剂时,所述单位剂量的单位是mg/片。
本发明通过提高索拉非尼稳定性和溶出度,降低了索拉非尼的服用剂量, 病人口服多吉美片剂给药剂量的35%~70%即可达到与多吉美片剂相同的治疗效果。
本发明优选的,所述辅剂选自填充剂、崩解剂、粘合剂、助流剂、润滑剂、矫味剂中的一种或多种。
本发明优选的,所述填充剂选自甘露醇、预胶化淀粉、乳糖、磷酸氢钙、淀粉、微晶纤维素、预胶化淀粉、部分预胶化淀粉、硫酸镁、硫酸钙中的一种或多种。
所述填充剂的质量含量优选为20%-50%,更优选为30%-40%。
本发明优选的,所述崩解剂选自玉米淀粉、部分α化淀粉、羟丙基淀粉、羧甲基纤维素、羧甲基纤维素钠、羧甲基纤维素钙、羧甲基淀粉钠、低取代羟丙基纤维素、交联羧甲基纤维素钠、交聚维酮中的一种或多种;更优选为交聚维酮、羧甲基淀粉钠或低取代羟丙基纤维素。
所述崩解剂的质量含量优选为3%-20%,更优选为5%-10%。
本发明优选的,所述粘合剂选自羟丙纤维素、羟丙甲纤维素、聚维酮、淀粉浆、羧甲纤维素钠中的一种或多种。
所述粘合剂的质量含量优选为0-10%,更优选为0-5%。
本发明优选的,所述助流剂选自滑石粉、二氧化硅中的一种或多种;优选为二氧化硅。
所述二氧化硅的质量含量优选为2%-20%,更优选为5%-15%,最优选为10%。
本发明通过增大二氧化硅的用量,使得制剂崩解加快,进而提高了制剂的溶出度。
本发明优选的,所述润滑剂选自硬脂酸镁、硬脂酸、硬脂酸钙、硬脂富马酸钠、聚乙二醇、氢化植物油、聚乙二醇、十二烷基硫酸钠、滑石粉、二氧化硅中的一种或多种。
所述润滑剂的质量含量优选为0.3%-5%,更优选为0.5%-2%,最优选为0.5%-1%。
本发明优选的,所述口服固体制剂的剂型为片剂、颗粒剂、干混悬剂、胶囊剂或膜剂。
本发明优选的,上述口服固体制剂采用干法制粒工艺制备。
本发明提供了上述索拉非尼固体分散体或上述索拉非尼口服固体制剂在制备用于预防、治疗或减轻肝癌、肾细胞癌、甲状腺癌的药物中的应用。
本发明提供了一种预防、治疗或减轻肝癌、肾细胞癌、甲状腺癌的方法,包括将上述索拉非尼固体分散体或上述索拉非尼口服固体制剂,与生物样本接触。
与现有技术相比,本发明提供了一种低剂量索拉非尼口服固体制剂,包括:a)索拉非尼固体分散体;b)抑晶剂;c)其他药学上可接受的辅剂。本发明提供的上述低剂量索拉非尼口服固体制剂获得了以下有益效果:
1、具有较高的生物利用度,降低了索拉非尼的服用剂量,病人口服多吉美片剂给药剂量的35%~70%即可达到与多吉美片剂相同的治疗效果;
2、具有更高的稳定性,更安全、副作用发生率更少;
3、具有更低的C max、AUC 0-t变异;
4、具有更高的溶出度,以含0.1%SDS(十二烷基硫酸钠)的pH6.8磷酸盐缓冲液做溶出介质,比上市产品“多吉美”溶出度高2-7倍(以溶出曲线下面积计算);
5、随胃肠道pH的升高,析晶速率低;
6、片剂体积小,病人易于服用;
7、崩解速度快,溶出效果好;
8、易于实现产业化。
附图说明
图1为不同比例VA64固体分散体制备的片剂pH6.8磷酸盐缓冲液(0.1%SDS)中溶出曲线图;
图2为复合载体固体分散体制备的片剂pH6.8磷酸盐缓冲液(0.1%SDS)中溶出曲线图。
具体实施方式
为了进一步说明本发明,下面结合实施例对本发明提供的高生物利用度的索拉非尼药物组合物及应用进行详细描述。
实施例1索拉非尼在不同有机溶剂中的溶解度
将适量索拉非尼原料加入25ml有机溶剂中,室温下摇床振摇不同时间,取样检测溶解的药物量,结果如下表1所示:
表1索拉非尼在不同有机溶剂中的溶解度
Figure PCTCN2020134164-appb-000003
采用HPLC法检测其有关物质,结果如下表2所示:
表2索拉非尼在不同有机溶剂中的稳定性
Figure PCTCN2020134164-appb-000004
Figure PCTCN2020134164-appb-000005
由以上结果可知索拉非尼在甲醇-二氯甲烷(1:3)中的溶解度较大,且在此有机溶剂中放置30min和24h,有关物质较API相比均无明显变化,故优选甲醇-二氯甲烷为索拉非尼喷雾干燥法制备固体分散体的有机溶剂。
实施例2固体分散体(SD)溶解度和稳定性
1、SD制备
将索拉非尼(SLFN)/甲苯磺酸索拉非尼(TSSLFN)加入甲醇-二氯甲烷(1:3)混合溶剂,30-33℃搅拌溶解,主药:混合溶剂为1:100。再加入载体溶解,载体用量为主药:载体为1:3。
喷雾干燥:进风温度120℃,出风温度设定60℃,氮气为载气。结果如表3所示。
表3不同载体及不同载体与主药比例制备的固体分散体DSC检测结果
Figure PCTCN2020134164-appb-000006
Figure PCTCN2020134164-appb-000007
根据以上结果,不同载体制备的样品均形成固体分散体。
2、溶解度检测
加适量固体分散体(SD)于pH1.0盐酸溶液和pH6.8磷酸盐缓冲液中,保持SD过量状态,于37℃摇床300rpm振摇,不同时间取样检测索拉非尼含量;对VA64和PVPK30为载体的固体分散体同时考察人工肠液和人工胃液中的溶解度,结果如下:
表4索拉非尼固体分散体溶解度测定结果
Figure PCTCN2020134164-appb-000008
Figure PCTCN2020134164-appb-000009
Figure PCTCN2020134164-appb-000010
由以上结果可知,虽然在pH1.0盐酸介质中固体分散体的初始溶解度(0.5h、2h、4h)有所提高,但随时间延长溶解度逐渐降低,试管壁上出现索拉非尼晶型药物,在24h时同甲苯磺酸索拉非尼相差不大。在pH6.8磷酸盐介质中,除以HPMC AS 716G为载体的固体分散体外,其他高分子材料做载体制备的固体分散体的溶解度均低于定量限。
以高分子VA64、PVPK30为载体制备的固体分散体在人工胃液(FaSSGF)中30分钟和2小时,测得的溶解度无明显差异,约为7μg/mL;转至人工肠液(FaSSIF)后1小时测得的VA64载体固体分散体的溶解度为350μg/mL,2小时后快速下降至12.38μg/mL;而以PVP K30做载体的固体分散体,转至人工肠液后1小时测得的溶解度为131μg/mL,2小时后快速下降至3.6μg/mL。
3、稳定性测定
将以上制备的SD铝箔袋密封,放入60℃高温考察10天,30天,HPLC法检测有关物质,结果见下表5所示。
表5不同载体制备的索拉非尼固体分散体高温60℃影响因素检测结果
Figure PCTCN2020134164-appb-000011
Figure PCTCN2020134164-appb-000012
由以上结果可知,以VA64为载体制备的SD化学稳定性最好,以HPMCAS为载体的固体分散体化学稳定性较差;分别以索拉非尼游离碱和甲苯磺酸索拉非尼为主药,用相同比例的高分子载体制备固体分散体,经高温60℃放置30天,甲苯磺酸索拉非尼的固体分散体化学稳定性略差。
实施例3载体比例筛选
1、VA64载体比例筛选
按上述喷雾干燥方法制备不同载体比例(质量比)的索拉非尼固体分散体,并进行DSC检测确认是否完全形成固体分散体。结果如表6所示。
表6不同VA64与主药比例制备的固体分散体DSC检测结果
主药 载体 比例 DSC图中晶型药物吸热峰
SLFN VA64 1:0.5 消失
SLFN VA64 1:1 消失
SLFN VA64 1:1.5 消失
SLFN VA64 1:2 消失
将上述不同载体比例的索拉非尼固体分散体按表7所示处方制备片剂:
表7索拉非尼固体分散片处方
处方组成 用量(mg/片)
SD用量(用量以SLFN计) 100
硅化微晶纤维素(SMCC) 218.50
十二烷基硫酸钠(SDS) 4.50
交联羧甲基纤维素钠(CCNa) 22.50
硬脂酸镁(MS) 4.50
称取处方量的原辅料,除MS外将其他原辅料一起过50目筛6遍混合,干法制粒,称量颗粒重量,折算外加MS用量,称取MS,混合5min,压片。检测溶出曲线。溶出曲线如图1所示。
溶出方法:桨法,pH6.8磷酸盐缓冲液+0.1%SDS,900mL,100rpm,37℃。
取样和样品处理方法:取10mL,用PES滤膜(直径25mm,孔径0.45μm)过滤,弃去滤液7mL,取续滤液2~3mL,并补10mL介质于溶出杯中。过滤后样品用移液管取1mL于10mL容量瓶中,加流动相至刻度线,稀释10倍。
表8索拉非尼固体分散体片的溶出曲线(n=3)
Figure PCTCN2020134164-appb-000013
注:此处崩解时间为片剂在溶出杯中完全崩散/溶蚀时间。下同。
根据以上结果可知,随VA64用量增加,片剂的崩解时间明显延长。因高VA64用量崩解时间延长,导致溶出并未随VA64比例增加而线性增加,而是先升高后降低,最佳用量为1:1。不同固体分散体片剂虽在溶出15min累积溶出度均显著高于原研多吉美,但却在15min~60min迅速下降,也就是说索拉非尼药物溶出后马上又结晶析出,60min~120min时累积溶出度与多吉美片剂无明显差别。
2、复合载体筛选
按表9配比制备固体分散体。
表9不同复合载体与主药制备的固体分散体DSC检测结果
Figure PCTCN2020134164-appb-000014
将上述不同载体比例制备的固体分散体按表10所示处方压片。
表10索拉非尼固体分散片处方
处方组成 用量(mg/片)
SD用量(用量以SLFN计) 100
硅化微晶纤维素(SMCC) 218.50
十二烷基硫酸钠(SDS) 4.50
交联羧甲基纤维素钠(CCNa) 22.50
硬脂酸镁(MS) 4.50
称取处方量的原辅料,除MS外将其他原辅料一起过50目筛6遍混合,干法制粒,称量颗粒重量,折算外加MS用量,称取MS,混合5min,压片。检测溶出曲线,如图2所示。
溶出方法:桨法,pH6.8磷酸盐缓冲液+0.1%SDS,900mL,100rpm,37℃。
取样和样品处理方法:取10mL,用PES滤膜(直径25mm,孔径0.45μm)过滤,弃去滤液7mL,取续滤液2~3mL,并补10mL介质于溶出杯中。过滤后样品用移液管取1mL于10mL容量瓶中,加流动相至刻度线,稀释10倍。
表11复合载体SD制备的片剂溶出曲线(n=3)
Figure PCTCN2020134164-appb-000015
Figure PCTCN2020134164-appb-000016
将以上片剂铝箔袋密封,放入60℃考察10天,HPLC法检测有关物质,结果如下表12所示:
表12不同复合载体的固体分散体制备的片剂高温60℃有关物质检测结果
Figure PCTCN2020134164-appb-000017
Figure PCTCN2020134164-appb-000018
由以上数据可知,载体VA64+HPMCAS,随HPMCAS用量增加,后期溶出降低较慢,但稳定性变差,综合溶出结果与化学稳定性结果,最佳比例为1:1:0.25。其他复合载体效果不明显。
实施例4抑晶剂筛选
称取索拉非尼或甲苯磺酸索拉非尼各适量,分别加二甲基亚砜溶解并稀释制成每1ml约含索拉非尼15mg的高浓度贮备溶液。
以含0.1%十二烷基硫酸钠(SDS)的pH6.8磷酸盐缓冲溶液作为基础介质(pH6.8磷酸盐+0.1%SDS),分别称取高分子载体聚乙烯吡咯烷酮(PVP)PVP K25、PVP K30、PVP K90;乙烯基吡咯烷酮-醋酸乙烯酯的共聚物,如PVP VA64;醋酸羟丙甲基纤维素琥珀酸酯(HPMCAS),如HPMCAS HG(或126G)、HPMCAS MG(912G)、HPMCAS LG(或716G);羟丙基甲基纤维素,如HPMC K100LV;聚乙二醇,如PEG 1000、PEG 3350、PEG4000、PEG 6000及PEG 8000、甘氨胆酸钠(SGC)、牛磺胆酸钠(STC)、脱氧胆酸钠(SDC)、脱氧甘氨胆酸钠(SGDC)、鹅脱氧甘氨胆酸钠(SGCDC)、熊脱氧甘氨胆酸钠(SGUDC)、脱氧牛磺胆酸钠(STDC)和熊脱氧牛磺胆酸钠(STUDC)及十二烷基磺酸钠(SDS)等各适量,分别用pH6.8磷酸盐+0.1%SDS溶解制成每1ml含上述高分子载体约为0.5%的高分子载体介质或含胆盐0.3%的介质,备用。
分别量取50ml上述高分子载体介质,置100ml具塞试管中,于37℃摇床 中振摇1小时后,分别向各具塞试管中加入索拉非尼贮备液0.4ml,37℃超声分散均匀后,转入37℃恒温振荡摇床,并于0.5h、1h、2h、3h、4h和6h取样,HPLC法检测各高分子载体介质中索拉非尼的浓度,结果见下表:
表13高分子载体、胆盐、表面活性剂类抑晶剂的筛选
Figure PCTCN2020134164-appb-000019
根据上述筛选结果,聚乙烯吡咯烷酮系列、醋酸羟丙甲基纤维素琥珀酸酯(HPMCAS)系列高分子载体抑晶效果显著,并且抑晶效果不随时间延长而下降。PEG 1000、PEG8000在1小时内抑晶效果较好,但随时间延长析晶严重。HPMC K100LV、PEG 3350、PEG4000基本没有抑晶效果。
胆盐是人体胆汁分泌入肠道的生理表面活性剂,当含有PVP-VA64的药物组合物制成的制剂口服后,进入肠道,随着胃肠蠕动,与胆盐混合,不会导致药物的快速结晶析出。
实施例5抑晶剂处方筛选
1、SLFN-VA64(1:1)SD片剂处方中抑晶剂筛选
(1)按下表13筛选高分子载体抑晶剂/助溶剂,与抑晶剂PVP系列进行对比。
表14抑晶剂筛选处方
Figure PCTCN2020134164-appb-000020
其中,SLFN、VA64质量比1:1。
抑晶剂/助溶剂:HPMC E5、HPMC K4M、聚氧乙烯氢化蓖麻油RH40、黄原胶、TPGS1000、SOLUPLUS、PVP K25、PVP K30、HPMCAS 126G。
制备工艺:称取处方量的原辅料,将内加原辅料一起过50目筛6遍混合,干法制粒,称量颗粒重量,折算外加物料用量,称取外加物料,混合5min,压片。检测累积溶出度。
溶出方法同前。
表15不同抑晶剂/助溶剂处方累积溶出度(n=3)
Figure PCTCN2020134164-appb-000021
由以上结果可知,抑晶剂PVPK 30、PVP K25明显优于其他助溶剂/抑晶 剂。
将以上片剂铝箔袋密封,进行高温60℃影响因素考察,HPLC检测有关物质,DSC检测物理稳定性,结果如下表15所示:
表16加入不同抑晶剂的固体分散体片剂高温60℃稳定性结果
Figure PCTCN2020134164-appb-000022
Figure PCTCN2020134164-appb-000023
由以上数据可知,低熔点助溶剂在放置过程中主药发生转晶,其物理稳定性较差。
(2)胆盐抑晶剂筛选
表17抑晶剂筛选处方
Figure PCTCN2020134164-appb-000024
其中,SLFN、VA64质量比1:1。
抑晶剂/助溶剂:SGC、SDC。
制备工艺:称取处方量的原辅料,将内加原辅料一起过50目筛6遍混合,干法制粒,称量颗粒重量,折算外加物料用量,称取外加物料,混合5min,压片。检测累积溶出度。
溶出方法同前,同时与PVPK25作为抑晶剂的处方对比。
表18 SLFN-VA64固体分散体-胆盐抑晶剂溶出结果(n=3)
Figure PCTCN2020134164-appb-000025
根据上述结果,以VA-64做载体的处方,外加胆盐,对索拉非尼固体分散体片剂60分钟内的溶出有很大改善,但60分钟后下降到与多吉美相同的水平,抑晶效果保持的时间没有PVP系列长。
2、以SLFN-VA64-HPMCAS(1:1:0.25)为载体的固体分散体制备片剂中抑晶剂筛选
按下表19筛选抑晶剂/助溶剂。
表19抑晶剂筛选-载体SLFN-VA64-HPMCAS(1:1:0.25)
Figure PCTCN2020134164-appb-000026
Figure PCTCN2020134164-appb-000027
制备工艺:称取处方量的原辅料,将内加原辅料一起过50目筛6遍混合,干法制粒,称量颗粒重量,折算外加物料用量,称取外加物料,混合5min,压片。检测溶出曲线。
溶出方法同前。
表20不同抑晶剂处方溶出曲线(n=3)
Figure PCTCN2020134164-appb-000028
Figure PCTCN2020134164-appb-000029
由以上结果可知,PVP K25、PVPK30抑制析晶的效果优于其它三种抑晶剂。
实施例6制剂处方筛选
5.1、二氧化硅对制剂崩解和溶出的影响
按照表21所示处方制备片剂。制备方法同前。
表21二氧化硅用量考察处方
Figure PCTCN2020134164-appb-000030
Figure PCTCN2020134164-appb-000031
其中,所述SLFN、VA64的质量比为1:1。
溶出结果如表22所示。
表22不同SiO 2对溶出的影响(n=3)
Figure PCTCN2020134164-appb-000032
由以上结果可知,随二氧化硅用量增大,崩解变快,溶出提高。
5.2、填充剂考察
按照表23所示处方制备片剂。制备方法同前。
表23填充剂用量考察处方
Figure PCTCN2020134164-appb-000033
Figure PCTCN2020134164-appb-000034
其中,所述SLFN-VA64-HPMCAS质量比为1:1:0.25。
溶出结果如下表24所示:
表24不同填充剂对溶出的影响(n=3)
Figure PCTCN2020134164-appb-000035
Figure PCTCN2020134164-appb-000036
以上结果显示,不同填充剂对溶出结果稍有影响,但同原研制剂相比溶出均较高。
5.3崩解剂考察
按照表25所示处方制备片剂。制备方法同前。
表25崩解剂用量考察处方
Figure PCTCN2020134164-appb-000037
Figure PCTCN2020134164-appb-000038
其中,所述SLFN-VA64-HPMCAS质量比为1:1:0.25。
溶出结果如下表26所示:
表26不同崩解剂对溶出的影响(n=3)
Figure PCTCN2020134164-appb-000039
由以上结果可知,不同崩解剂虽有差异,但溶出均较好。
5.4不同规格样品制备
表27不同规格样品处方(100片/批)
Figure PCTCN2020134164-appb-000040
Figure PCTCN2020134164-appb-000041
制备工艺:称取处方量的原辅料,将内加原辅料一起过50目筛6遍混合,干法制粒,称量颗粒重量,折算外加物料用量,称取外加物料,混合5min,压片。
溶出结果如下表28所示:
表28不同规格样品溶出结果(n=3)
Figure PCTCN2020134164-appb-000042
可以看出,随规格增大,溶出百分比稍有降低,但均远远高于参比制剂多吉美。
实施例7动物实验对比
6.1、动物实验样品制备
表29动物实验样品处方(1000片/批)
Figure PCTCN2020134164-appb-000043
Figure PCTCN2020134164-appb-000044
制备工艺:称取处方量的原辅料,将内加原辅料一起过50目筛6遍混合,干法制粒,称量颗粒重量,折算外加物料用量,称取外加物料,混合5min,压片。
将以上样品(规格100mg)和参比制剂多吉美(规格200mg)进行动物实验。
动物:成年雄性比格犬(8-11kg),可以是
Figure PCTCN2020134164-appb-000045
共分4组,每组2只比格犬。
给药途径及给药频率:单次灌胃口服1片,给药前禁食16小时以上,给药4小时后自由取食;清洗期4天。4周期交叉给药。
取血时间点:0,1.5,2,2.5,3,4,5,6 8,12,24,48h。
采用LC/MS/MS方法检测血药浓度,采用Phoenix WinNonlin 7.0计算药代动力学参数(AUC 0-t、AUC 0-∞、C max、T 1/2、T max及这些参数的算术平均(±SD)和几何平均)。
表30比格犬实验结果
Figure PCTCN2020134164-appb-000046
由以上结果可知,20200509-1、20200519-3明显高于20200509-2。二者100mg剂量即同参比制剂200mg基本一致。对比参比和供试品的CV可知,20200509-1、20200509-3明显好于参比制剂。
以上实施例的说明只是用于帮助理解本发明的方法及其核心思想。应当指 出,对于本技术领域的普通技术人员来说,在不脱离本发明原理的前提下,还可以对本发明进行若干改进和修饰,这些改进和修饰也落入本发明权利要求的保护范围内。

Claims (17)

  1. 一种高生物利用度的索拉非尼药物组合物,其特征在于,包括:
    a)索拉非尼、或其盐、水合物、溶剂化物、盐的水合物或溶剂化物;
    b)载体:所述载体包括VA64和HPMCAS。
  2. 根据权利要求1所述的索拉非尼药物组合物,其特征在于,所述组分a)和组分b)中的VA64、HPMCAS的质量比为1:(0.1~5):(0.01~5),优选1:(0.5~3):(0.1~1)。
  3. 根据权利要求1所述的索拉非尼药物组合物,其特征在于,所述组分a),和组分b)中的VA64、HPMCAS的质量比为1:1:(0.1~0.5),优选1:1:0.25。
  4. 根据权利要求1所述的索拉非尼药物组合物,其特征在于,所述索拉非尼药物组合物为固体分散体,所述固体分散体按照喷雾干燥法制备,所述喷雾干燥法的溶剂为甲醇和二氯甲烷的混合溶剂。
  5. 根据权利要求4所述的索拉非尼药物组合物,其特征在于,所述甲醇和二氯甲烷的体积比为1:(1~4);优选1:3。
  6. 一种低剂量索拉非尼口服固体制剂,包括:
    a)索拉非尼固体分散体;
    b)抑晶剂;
    c)其他药学上可接受的辅剂。
  7. 根据权利要求6所述的低剂量索拉非尼口服固体制剂,其特征在于,所述索拉非尼固体分散体包括:
    a)索拉非尼、或其盐、水合物、溶剂化物、盐的水合物或溶剂化物;
    b)载体:所述载体包括VA64。
  8. 根据权利要求6所述的低剂量索拉非尼口服固体制剂,其特征在于,所述索拉非尼固体分散体包括:
    a)索拉非尼、或其盐、水合物、溶剂化物、盐的水合物或溶剂化物;
    b)载体:所述载体包括VA64和HPMCAS。
  9. 根据权利要求6所述的索拉非尼口服固体制剂,其特征在于,其中, 组分a)的单位剂量为70~200mg;优选70~140mg。
  10. 根据权利要求6所述的索拉非尼口服固体制剂,其特征在于,所述抑晶剂,选自聚乙烯吡咯烷酮、醋酸羟丙甲基纤维素琥珀酸酯(HPMCAS)、甘氨胆酸钠、牛磺胆酸钠、脱氧甘氨胆酸钠、鹅脱氧甘氨胆酸钠、熊脱氧甘氨胆酸钠、脱氧牛磺胆酸钠和熊脱氧牛磺胆酸钠及十二烷基磺酸钠中的一种或多种。
  11. 根据权利要求6所述的索拉非尼口服固体制剂,其特征在于,所述抑晶剂的质量含量为1%~40%;优选1%~20%。
  12. 根据权利要求6所述的索拉非尼口服固体制剂,其特征在于,所述辅剂选自填充剂、崩解剂、粘合剂、助流剂、润滑剂、矫味剂中的一种或多种。
  13. 根据权利要求12所述的索拉非尼口服固体制剂,其特征在于,所述填充剂选自甘露醇、预胶化淀粉、乳糖、磷酸氢钙、淀粉、微晶纤维素、预胶化淀粉、部分预胶化淀粉、硫酸镁、硫酸钙中的一种或多种;
    所述崩解剂选自玉米淀粉、部分α化淀粉、羟丙基淀粉、羧甲基纤维素、羧甲基纤维素钠、羧甲基纤维素钙、羧甲基淀粉钠、低取代羟丙基纤维素、交联羧甲基纤维素钠、交聚维酮中的一种或多种;
    所述粘合剂选自羟丙纤维素、羟丙甲纤维素、聚维酮、淀粉浆、羧甲纤维素钠中的一种或多种;
    所述助流剂选自滑石粉、二氧化硅中的一种或多种;
    所述润滑剂选自硬脂酸镁、硬脂酸、硬脂酸钙、硬脂富马酸钠、聚乙二醇、氢化植物油、聚乙二醇、十二烷基硫酸钠、滑石粉、二氧化硅中的一种或多种。
  14. 根据权利要求12所述的索拉非尼口服固体制剂,其特征在于,所述助流剂为二氧化硅。
  15. 根据权利要求13所述的索拉非尼口服固体制剂,其特征在于,所述二氧化硅的质量含量为2%~20%;优选5%-15%;更优选为10%。
  16. 根据权利要求6所述的索拉非尼口服固体制剂,其特征在于,所述口服固体制剂的剂型为片剂、颗粒剂、干混悬剂、胶囊剂或膜剂。
  17. 权利要求1~5任一项所述的高生物利用度的索拉非尼药物组合物或权利要求6~16任一项所述的索拉非尼口服固体制剂在制备用于预防、治疗或减 轻肝癌、肾细胞癌、甲状腺癌的药物中的应用。
PCT/CN2020/134164 2020-12-07 2020-12-07 一种高生物利用度的索拉非尼药物组合物及应用 WO2022120512A1 (zh)

Priority Applications (6)

Application Number Priority Date Filing Date Title
CN202080055010.9A CN114916221B (zh) 2020-12-07 2020-12-07 一种高生物利用度的索拉非尼药物组合物及应用
JP2022516367A JP7428356B2 (ja) 2020-12-07 2020-12-07 高いバイオアベイラビリティを有するソラフェニブの医薬組成物、ソラフェニブ経口固形製剤、及びその使用
US17/630,709 US20230310393A1 (en) 2020-12-07 2020-12-07 Sorafenib pharmaceutical composition with high bioavailability and use thereof
EP20946527.7A EP4032529A4 (en) 2020-12-07 2020-12-07 SORAFENIB PHARMACEUTICAL COMPOSITION WITH HIGH BIOAVAILABILITY AND USE THEREOF
CA3155855A CA3155855A1 (en) 2020-12-07 2020-12-07 Sorafenib pharmaceutical composition with high bioavailability and use thereof
PCT/CN2020/134164 WO2022120512A1 (zh) 2020-12-07 2020-12-07 一种高生物利用度的索拉非尼药物组合物及应用

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2020/134164 WO2022120512A1 (zh) 2020-12-07 2020-12-07 一种高生物利用度的索拉非尼药物组合物及应用

Publications (1)

Publication Number Publication Date
WO2022120512A1 true WO2022120512A1 (zh) 2022-06-16

Family

ID=81927003

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/134164 WO2022120512A1 (zh) 2020-12-07 2020-12-07 一种高生物利用度的索拉非尼药物组合物及应用

Country Status (6)

Country Link
US (1) US20230310393A1 (zh)
EP (1) EP4032529A4 (zh)
JP (1) JP7428356B2 (zh)
CN (1) CN114916221B (zh)
CA (1) CA3155855A1 (zh)
WO (1) WO2022120512A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115813861A (zh) * 2022-12-28 2023-03-21 药源生物科技(启东)有限公司 一种提高难溶性药物溶解度的固体分散体及制备方法
WO2023155182A1 (zh) * 2022-02-21 2023-08-24 北京睿创康泰医药研究院有限公司 一种低服用剂量高药物暴露量的索拉非尼或多纳非尼口服制剂及其应用

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101048140A (zh) * 2004-08-27 2007-10-03 拜耳制药公司 用于治疗癌症的新药物组合物
WO2008008733A2 (en) * 2006-07-10 2008-01-17 Elan Pharma International Ltd. Nanoparticulate sorafenib formulations
WO2009100176A2 (en) * 2008-02-07 2009-08-13 Abbott Laboratories Pharmaceutical dosage form for oral administration of tyrosine kinase inhibitor
CN101594851A (zh) * 2006-11-09 2009-12-02 阿伯特有限及两合公司 用于口服给药的酪氨酸激酶抑制剂的药物剂型
CN105126111A (zh) * 2015-09-30 2015-12-09 清华大学 提高索拉非尼生物利用度的制剂
CN107115317A (zh) * 2012-01-13 2017-09-01 X喷雾微粒公司 包含稳定的、无定形杂化纳米颗粒的药物组合物
WO2018211336A2 (en) * 2018-09-07 2018-11-22 Alvogen Malta Operations (Row) Ltd Solid dosage form containing sorafenib tosylate

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104971045A (zh) * 2014-04-11 2015-10-14 上海宣泰医药科技有限公司 泊沙康唑药物组合物及其制备方法和药物制剂
US11980610B2 (en) * 2017-10-31 2024-05-14 Samyang Holdings Corporation Oral solid dosage form composition having improved disintegration and preparation method therefor

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101048140A (zh) * 2004-08-27 2007-10-03 拜耳制药公司 用于治疗癌症的新药物组合物
WO2008008733A2 (en) * 2006-07-10 2008-01-17 Elan Pharma International Ltd. Nanoparticulate sorafenib formulations
CN101594851A (zh) * 2006-11-09 2009-12-02 阿伯特有限及两合公司 用于口服给药的酪氨酸激酶抑制剂的药物剂型
WO2009100176A2 (en) * 2008-02-07 2009-08-13 Abbott Laboratories Pharmaceutical dosage form for oral administration of tyrosine kinase inhibitor
CN107115317A (zh) * 2012-01-13 2017-09-01 X喷雾微粒公司 包含稳定的、无定形杂化纳米颗粒的药物组合物
CN105126111A (zh) * 2015-09-30 2015-12-09 清华大学 提高索拉非尼生物利用度的制剂
WO2018211336A2 (en) * 2018-09-07 2018-11-22 Alvogen Malta Operations (Row) Ltd Solid dosage form containing sorafenib tosylate

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
ANN ONCOL, vol. 16, 2005, pages 1688 - 94
INVEST NEW DRUGS., vol. 30, no. 2, April 2012 (2012-04-01), pages 524 - 528
LIU CHENGYU, XU CONG-QIAO, YU JUNGUANG, PUI YIPSHU, CHEN HUIJUN, WANG SHAN, ZHU ALAN DONGHUA, LI JUN, QIAN FENG: "Impact of a Single Hydrogen Substitution by Fluorine on the Molecular Interaction and Miscibility between Sorafenib and Polymers", MOLECULAR PHARMACEUTICS, vol. 16, no. 1, 7 January 2019 (2019-01-07), US , pages 318 - 326, XP055907949, ISSN: 1543-8384, DOI: 10.1021/acs.molpharmaceut.8b00970 *
LIU, CHENGYU ET AL.: "Improving Oral Bioavailability of Sorafenib by Optimizing the ''Spring '' and ''Parachute'' Based on Molecular Interaction Mechanisms", MOLECULAR PHARMACEUTICS, vol. 13, 28 December 2015 (2015-12-28), pages 599 - 608, XP055662728, DOI: 10.1021/acs.molpharmaceut.5b00837 *
ONCOLOGIST, vol. 12, 2007, pages 426 - 37
ONCOTARGET, vol. 8, no. 26, 2017, pages 43458 - 43469

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023155182A1 (zh) * 2022-02-21 2023-08-24 北京睿创康泰医药研究院有限公司 一种低服用剂量高药物暴露量的索拉非尼或多纳非尼口服制剂及其应用
CN115813861A (zh) * 2022-12-28 2023-03-21 药源生物科技(启东)有限公司 一种提高难溶性药物溶解度的固体分散体及制备方法
CN115813861B (zh) * 2022-12-28 2024-06-14 药源生物科技(启东)有限公司 一种提高难溶性药物溶解度的固体分散体及制备方法

Also Published As

Publication number Publication date
JP2023509560A (ja) 2023-03-09
CN114916221A (zh) 2022-08-16
EP4032529A1 (en) 2022-07-27
EP4032529A4 (en) 2022-11-16
JP7428356B2 (ja) 2024-02-06
CA3155855A1 (en) 2022-06-07
CN114916221B (zh) 2024-01-30
US20230310393A1 (en) 2023-10-05

Similar Documents

Publication Publication Date Title
US20190060294A1 (en) Pharmaceutical compositions
US9326945B2 (en) Apixaban formulations
EP2170295B1 (en) Improved pharmaceutical composition containing dihydropyridine calcium channel antagonist and method for the preparation thereof
US20230381194A1 (en) Suspension for oral administration comprising amorphous tolvaptan
JP2011516613A (ja) 好ましくはポサコナゾールおよびhpmcasを含む固体分散物中の経口用薬学的組成物
WO2022120512A1 (zh) 一种高生物利用度的索拉非尼药物组合物及应用
KR102512868B1 (ko) 올라파립의 용해도 및 생체이용율이 개선된 조성물
US10493034B2 (en) Pharmaceutical composition for treatment of HIV infection
US10888519B2 (en) Immediate release pharmaceutical composition of iron chelating agents
CN108096251B (zh) 一种吉非替尼药物组合物及其制备方法
JP2021518422A (ja) レナリドミドを含む医薬組成物
WO2018202224A1 (en) Film-coated tablets of deferasirox
CN115397416B (zh) 包含仑伐替尼的分子水平的药物组合物及其制备方法和应用
WO2023155182A1 (zh) 一种低服用剂量高药物暴露量的索拉非尼或多纳非尼口服制剂及其应用
JP4603803B2 (ja) 放出制御医薬組成物およびこれを用いる製剤
WO2021176083A1 (en) Pharmaceutical compositions comprising dasatinib anhydrous and uses thereof
EA035891B1 (ru) Быстродиспергируемая фармацевтическая композиция, включающая ингибитор тирозинкиназы

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2020946527

Country of ref document: EP

Effective date: 20220129

ENP Entry into the national phase

Ref document number: 2022516367

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE