WO2021136089A1 - 一种抗肿瘤药物组合物以及提高化合物的溶解度的方法 - Google Patents

一种抗肿瘤药物组合物以及提高化合物的溶解度的方法 Download PDF

Info

Publication number
WO2021136089A1
WO2021136089A1 PCT/CN2020/139414 CN2020139414W WO2021136089A1 WO 2021136089 A1 WO2021136089 A1 WO 2021136089A1 CN 2020139414 W CN2020139414 W CN 2020139414W WO 2021136089 A1 WO2021136089 A1 WO 2021136089A1
Authority
WO
WIPO (PCT)
Prior art keywords
acid
pharmaceutical composition
pharmaceutically acceptable
formula
compound represented
Prior art date
Application number
PCT/CN2020/139414
Other languages
English (en)
French (fr)
Inventor
鹿宁
程智慧
杨红振
胡怀忠
Original Assignee
北京康辰药业股份有限公司
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 北京康辰药业股份有限公司 filed Critical 北京康辰药业股份有限公司
Priority to CN202080063242.9A priority Critical patent/CN114364386B/zh
Publication of WO2021136089A1 publication Critical patent/WO2021136089A1/zh

Links

Images

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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the invention belongs to the technical field of pharmacy, and specifically relates to a quinoline anti-tumor drug composition containing phosphorus substituents and a method for improving the solubility of bulk drugs in the tumor drug composition.
  • Compound 1 is a quinoline anti-tumor drug containing phosphorus substituents, which mainly acts on hepatocyte growth factor receptor (HGFR or mesenchymal-epithelial transition factor, c-Met) and vascular endothelial growth factor 2.
  • HGFR hepatocyte growth factor receptor
  • c-Met mesenchymal-epithelial transition factor
  • vascular endothelial growth factor 2 VEGFR2 or KDR
  • Compound 1 is a dual inhibitor of c-Met and VEGFR2, which can effectively block both c-Met and VEGFR2 signaling pathways that play different roles in tumors, thus having unique pharmacological effects and clinical application prospects.
  • Compound 1 has a broad-spectrum anti-tumor effect, and shows excellent anti-tumor effect and safety range in animal models of gastric cancer, prostate cancer bone metastasis, medullary thyroid cancer, non-small cell lung cancer, liver cancer, kidney cancer and other tumors.
  • compound 1 is: 1-[[4-[2-fluoro-4-[[1-[(4-fluorophenyl)carbamoyl]cyclopropanecarbonyl]amino]phenoxy]-6-methyl Oxy-7-quinolinyl]oxymethyl]piperidinyl]methyl diethyl phosphate.
  • the present invention provides an anti-tumor pharmaceutical composition, the pharmaceutical composition comprising a compound represented by Formula 1 and a pharmaceutically acceptable acid;
  • the composition further includes a coating layer that coats the surface of the compound represented by Formula 1.
  • the material of the coating layer can be pharmaceutically acceptable pharmaceutical excipients commonly used in the art, such as silicon dioxide, titanium dioxide, hypromellose, hydroxypropyl cellulose, talc, polyethylene glycol, polyethylene Alcohol or a combination of one or more of them; preferably, the material of the coating layer is selected from silica.
  • the main reason for isolating the drug substance from the pharmaceutically acceptable acid is that the drug substance of compound 1 has poor stability under acidic conditions and is easily degraded after prolonged contact with acid.
  • the isolation effect of the coating layer can help Ensure the stability of compound 1 during preparation and storage.
  • silica has a small particle size and a large specific surface area, and is easily adsorbed on the surface of the particles to isolate the acid from the bulk drug. Therefore, silicon dioxide can be used as an ideal coating material.
  • the pharmaceutically acceptable acid in the pharmaceutical composition can enable the compound represented by formula 1 to have a saturated solubility of 0.5 mg/ml or more, preferably 1.0 mg/ml or more, in water with pH ⁇ 3.0 .
  • a saturated solubility of 0.5 mg/ml or more, preferably 1.0 mg/ml or more, in water with pH ⁇ 3.0 .
  • the pharmaceutically acceptable acid in the composition can make the compound represented by formula 1 have a saturated solubility of more than 0.5 mg/ml in water of pH ⁇ 3.0, preferably a saturated solubility of more than 1.0 mg/ml .
  • the pharmaceutically acceptable acid may be an inorganic acid or an organic acid, as long as it is suitable as a pharmaceutical excipient.
  • the pharmaceutically acceptable acid may be an inorganic acid or an organic acid, as long as it is suitable as a pharmaceutical excipient.
  • the present invention there is no restriction on the form of the acid, and it may be a solid acid or a liquid acid.
  • a requirement is that under the same acid concentration, the lower the pH of the acid, the better.
  • all acids with a pH value below 3.0 can be used after being dissolved in water.
  • strong acids for example, hydrochloric acid, acetic acid, etc.
  • they are generally not suitable for use as pharmaceutical excipients.
  • the acids that can be used include, but are not limited to, inorganic acids such as phosphoric acid, and organic acids such as alginic acid, citric acid, fumaric acid, glutamic acid, tartaric acid, malic acid, and the like.
  • citric acid is preferably used.
  • the main reasons include: (1) The dissolution rate of citric acid in water is fast, and only a small amount of citric acid needs to be added to reduce the pH of the water to below 3.0. Considering that the active drug compound 1 enters the stomach, the gastric emptying rate is fast, so a pharmaceutically acceptable acid that can be quickly dissolved and quickly lowers the pH of the gastric juice is required. From this perspective, citric acid meets this requirement.
  • Citric acid is widely used and is a pharmaceutical excipient approved by the FDA with a large allowable dosage and has high safety.
  • the addition ratio of the compound represented by formula 1 of the bulk drug to the pharmaceutically acceptable acid is preferably 1:2 to 1:6 by weight, More preferably, it is 1:4.
  • the specific addition ratio will vary depending on the type of acid.
  • the pharmaceutically acceptable excipients can be various pharmaceutical excipients suitable for the preparation of compound 1 in the art, such as fillers, disintegrants, lubricants, etc., for tablets. .
  • the pharmaceutically acceptable excipient is selected from the group consisting of microcrystalline cellulose, lactose, crospovidone, hydroxypropyl cellulose, magnesium stearate, poly One or more of vitamin ketone, croscarmellose sodium, and sodium carboxymethyl starch.
  • the use of these adjuvants facilitates the preparation of the drug into a suitable dosage form and facilitates the administration of the gastrointestinal tract.
  • composition of the present invention can be made into solids such as tablets, capsules, granules, pills or powders. preparation.
  • another aspect of the present invention provides a method for improving the solubility of the compound represented by Formula 1 in water, the method comprising adding a pharmaceutically acceptable acid to the compound represented by Formula 1.
  • the method includes: before adding the pharmaceutically acceptable acid, after the compound represented by formula 1 is subjected to wet granulation, adding a package
  • the coating material is mixed to coat the compound represented by formula 1; wherein, the coating material is selected from the group consisting of silicon dioxide, titanium dioxide, hypromellose, hydroxypropyl cellulose, talc, polyethylene two At least one of alcohol and polyvinyl alcohol is preferably silica.
  • the added pharmaceutically acceptable acid needs to be able to make the compound represented by formula 1 have a saturated solubility of 0.5 mg/ml or more, preferably 1.0 mg/ml or more, in water with pH ⁇ 3.0 .
  • the addition ratio of the compound represented by formula 1 to the pharmaceutically acceptable acid is 1:2 to 1:6, more preferably 1:4.
  • the inventor found that the solubility of the API of compound 1 is pH-dependent, and the solubility of compound 1 under acidic conditions is much higher than that under neutral and alkaline conditions. .
  • Compound 1 is an anti-tumor drug, which is more commonly used in older middle-aged and elderly patients with tumor patients, and the pH value of the digestive system (stomach) of middle-aged and elderly patients is generally high. According to research literature, the pH value of gastric juice in middle-aged and elderly people can reach pH 4-6, which is not conducive to the dissolution of this product.
  • Tie Zhenjun measured the acidity of the gastric juice of the elderly over 60, which proved that the gastric acid concentration of most elderly people is lower than that of young people, and the pH value of gastric juice can reach pH 4 or more.
  • the active ingredient of the medicine can release acid after entering the gastric juice, thereby reducing the pH value of the gastric juice and increasing the acidity of the gastric juice.
  • the decrease of the pH value of the gastric juice provides a favorable environment for the dissolution of the drug compound 1, thereby promoting the dissolution and absorption of the drug, and improving the bioavailability of the active drug in the body.
  • Figure 1 is a graph showing the relationship between the pH value of a drug solution and the saturated solubility of compound 1 represented by formula 1;
  • Figure 2 is a graph showing changes in mouse blood drug concentration over time.
  • the present invention improves the solubility of the drug in dissolution media with different pH values by adding acid to the bulk drug (compound 1).
  • the effect of several acids on the saturated solubility of compound 1 will be studied.
  • Table 3 lists the amount of acid used when several common acid excipients are dissolved in 100 ml of water and adjusted to a pH value around 3.0.
  • the drug prescription A used in this experiment is shown in Table 4 below.
  • Plasma samples were taken before administration and 15min, 30min, 1h, 2h, 3h, 4h, 6h, 8h, 24h, 48h after administration.
  • the inventors took citric acid as an example to study the effect of adding citric acid to tablets on the bioavailability of compound 1.
  • three tablet prescriptions were designed: prescription A, prescription A 1 and prescription C.
  • Formulation A, Formulation A 1 , and Formulation C have changed the proportion of fillers, binders, and disintegrants in the formulations while keeping the proportion of compound 1 in the formulation of the raw material basically unchanged.
  • the prescription of the tablet is shown in Table 6.
  • the formulation were A, and a tablet formulation for the samples A 1 C prescription grinding, normal saline, vortexed, ultrasound, formulated as a suspension 1mg / ml, the mice by oral gavage, the animals 6 sample. At 5min, 15min, 30min, 1h, 2h, 4h, 8h, 12h, 24h after administration, plasma samples were taken for testing.
  • polysorbate 80 as a surfactant may promote the absorption of compound 1 in the body, but the data of this experiment (Table 6 and Table 7) prove that polysorbate 80 does not affect the absorption of compound 1 in the body.
  • Pretreatment Micronization of the raw material of compound 1; anhydrous citric acid through 40 meshes for use.
  • Pre-mixing the raw material of compound 1, microcrystalline cellulose 101, lactose monohydrate, and croscarmellose sodium are gradually mixed, and mixed through a 40-mesh sieve 3 times to obtain a mixed powder.
  • Granulation Add 10% hydroxypropyl cellulose aqueous solution to the mixed powder, pass through a 20-mesh sieve and granulate to obtain wet granules.
  • Drying Wet particles are placed in an oven at 60°C and dried until the moisture content of the material is less than 3.0% to obtain dry particles.
  • Granulation The dry granules are passed through a 20-mesh sieve and granulated.
  • Total mixing The dry granules are gradually mixed with silicon dioxide, and then sodium starch glycolate, microcrystalline cellulose 102, anhydrous citric acid, and magnesium stearate are added and mixed evenly.
  • Pressing sheet 11mm round dimple punching sheet.
  • Table 11 Stability of prescription D (compound 1 raw material drug after granulation, plus silica and citric acid)

Abstract

一种含磷取代基的喹啉类抗肿瘤药物组合物以及提高该药物组合物中的原料药的溶解度的方法,所述药物组合物包括原料药以及药物学上可接受的酸。通过在原料药中加入酸化合物,可以使药物进入胃液之后,释放出酸,从而提高胃液的酸性,降低胃液pH值。胃液pH值的降低为药物的溶解提供了有利的环境,促进了原料药在胃内的溶解和吸收,提高了药物在体内的生物利用度。还提供了一种提高含磷取代基的喹啉类抗肿瘤药物在水中的溶解度的方法。

Description

一种抗肿瘤药物组合物以及提高化合物的溶解度的方法
本申请要求于2019年12月30日提交中国专利局、申请号为201911399012.5、发明名称为“一种抗肿瘤药物组合物以及提高化合物的溶解度的方法”的中国专利申请的优先权,其全部内容通过引用结合在本申请中。
技术领域
本发明属于制药技术领域,具体地涉及一种含磷取代基的喹啉类抗肿瘤药物组合物以及涉及提高该肿瘤药物组合物中的原料药的溶解度的方法。
背景技术
化合物1是一种含磷取代基的喹啉类抗肿瘤药物,主要作用于肝细胞生长因子受体(hepatocyte growth factor receptor,HGFR或mesenchymal–epithelial transition factor,c-Met)和血管内皮生长因子2(vascular endothelial growth factor 2,VEGFR2或KDR)的新型多靶点受体酪氨酸激酶抑制剂。化合物1是c-Met和VEGFR2的双重抑制剂,能有效地同时阻断c-Met和VEGFR2这两种在肿瘤中发挥不同作用的信号通路,从而具有独特的药理作用和临床应用前景。化合物1具有广谱抗肿瘤效果,对胃癌、前列腺癌骨转移、甲状腺髓样癌、非小细胞肺癌、肝癌、肾癌等肿瘤的动物模型中显示出优良的抗肿瘤效果和安全范围。
化合物1的化学名称为:1-[[4-[2-氟-4-[[1-[(4-氟苯基)氨基甲酰基]环丙烷羰基]氨基]苯氧基]-6-甲氧基-7-喹啉基]氧甲基]哌啶基]甲基磷酸二乙酯。
化合物1的化学结构如下式1所示。
Figure PCTCN2020139414-appb-000001
在临床研究中发现,现有的化合物1的制剂溶解度差,在人体内的生物利用度低且个体差异大,导致临床效果不稳定。为了解决这一问题,发明人对化合物1作了进一步的研究。
发明内容
针对现有技术存在的问题,申请人进行了深入的研究,以提高化合物1的溶解度和生物利用度。
为了实现本发明的目的,本发明提供一种抗肿瘤药物组合物,所述药物组合物包括式1所示的化合物以及药物学上可接受的酸;
Figure PCTCN2020139414-appb-000002
在本发明所述药物组合物的一种实施方式中,所述组合物还包括包覆式1所示的化合物的表面的包覆层。包覆层的材料是可以是本领域常用的药学上可接受的药用辅料,例如二氧化硅、二氧化钛、羟丙甲纤维素、羟丙基纤维素、滑石粉、聚乙二醇、聚乙烯醇或它们中一种或多种的组合;优选地,所述包覆层的材料选自二氧化硅。
将原料药与药学上可接受的酸进行隔离的主要原因是化合物1的原料药在酸性条件下稳定性较差,与酸长时间接触后容易降解,而通过包覆层的隔离的作用,可以保障化合物1在制备和存储过程中的稳定性。在这些包覆层材料中,二氧化硅粒径小、比表面积大,容易吸附在颗粒表面而起到将酸与原料药进行隔离的作用。因此,二氧化硅可作为理想的包覆层的 材料。
在本发明的药物组合物中,药物组合物中的药学上可接受的酸能够使式1所示的化合物在pH≤3.0的水中具有0.5mg/ml以上,优选1.0mg/ml以上的饱和溶解度。根据临床考察,以不超过200mg化合物1的剂量的临床最大用量计,在一般临床服药方式中,会以100ml-200ml的温水送服,在pH=3.0的水中化合物1具有0.5mg/ml的饱和溶解度就能达到提高生物利用度的预期,从而满足临床需求。当pH值增大时,化合物1在水中的饱和溶解度太小,难以满足临床需求。因此,出于临床疗效的考虑,组合物中的药学上可接受的酸能够使式1所示的化合物在pH≤3.0的水中具有0.5mg/ml以上,优选具有1.0mg/ml以上的饱和溶解度。
在本发明的药物组合物中,所述药学上可接受的酸可以是无机酸,也可以是有机酸,只要其适用作药用辅料即可。同时,在本发明中,对酸的形态也没有限制,可以是固态酸,也可以是液态酸。为了实现本发明的目的,对于酸的选择,一个要求是在相同的酸浓度下,酸的pH值越低越好。理论上,在水中溶解后pH值为3.0以下的酸都可用。虽然强酸(例如,盐酸、醋酸等)的效果更好,但强酸一般不适合用作药物辅料。在本发明中,可以使用的酸包括但不限于无机酸例如磷酸等,有机酸例如海藻酸、枸橼酸、富马酸、谷氨酸、酒石酸、苹果酸等。在这些酸中,优选使用枸椽酸,主要原因包括:(1)枸橼酸在水中的溶解速率快,且只需要添加少量枸橼酸就可以使水的pH值降到3.0以下。考虑到活性药物化合物1进入胃后,胃排空速度快,因此一种需要能够快速溶解,迅速降低胃液pH的药学上可接受的酸,从这个角度而言,枸橼酸符合这一要求。(2)枸橼酸使用广泛,是经FDA认证的允许用量较大的药用辅料,具有很高的安全性。
在本发明的药物组合物中,从理论上来说,只要在处方中添加酸,即可对式1所示的化合物在体内的溶解和吸收产生促进作用。为了更好地适应胃的环境以及达到理想的生物利用度,原料药式1所示的化合物与所述药学上可接受的酸的添加比例优选为以重量计的1∶2~1∶6,更优选为1∶4。具体的添加比例会因酸的种类而有所区别。
在本发明的药物组合物中,所述药学上可接受的辅料可以是本领域适用于化合物1的制剂的各种药用辅料,例如用作片剂的填充剂、崩解剂、润滑剂等。在本发明所述药物组合物一种优选实施方案中,所述药学上可 接受的辅料选自微晶纤维素、乳糖、交联聚维酮、羟丙基纤维素、硬脂酸镁、聚维酮、交联羧甲基纤维素钠、羧甲基淀粉钠中的一种或多种,采用这些辅料有利于药物的做成合适的剂型且有利于经胃肠道给药。
鉴于式1所示的化合物的特性以及使本发明所述的药物组合物更适于胃肠道给药,本发明的组合物可以制成片剂、胶囊剂、颗粒剂、丸剂或散剂等固体制剂。
因此,本发明的另一方面提供了一种提高式1所示的化合物在水中的溶解度的方法,所述方法包括在式1所示的化合物中添加药学上可接受的酸。
在本发明制备所述的药物组合物方法的一种实施例中,所述方法包括:在加入所述药学上可接受的酸之前,在将式1所示的化合物进行湿法制粒后添加包覆材料,混合,以将式1所示的化合物进行包覆;其中,所述包覆材料选自二氧化硅、二氧化钛、羟丙甲纤维素、羟丙基纤维素、滑石粉、聚乙二醇、聚乙烯醇中的至少一种,优选二氧化硅。通过包覆层的隔离作用,可以将原料药化合物1与药学上可接受的酸进行隔离。
为了更好地实现本发明的目的,所添加的药学上可接受的酸需要能够使式1所示的化合物在pH≤3.0的水中具有0.5mg/ml以上,优选1.0mg/ml以上的饱和溶解度。优选地,式1所示的化合物与所述药学上可接受的酸的添加比例为1∶2~1∶6,更优选1∶4。
发明人在对抗肿瘤药物化合物1的研究过程中发现,化合物1的原料药(API)的溶解度呈pH依赖性,化合物1在酸性条件下溶解度远远高于在中性及碱性条件下的溶解度。化合物1为抗肿瘤药物,其更多地应用于年纪较大的中老年患者肿瘤患者,而中老年人的消化系统(胃)的pH值普遍偏高。根据研究文献记载,中老年人胃液的pH值可达pH4-6,更不利于本品的溶解。在《老年人常见消化疾病的诊治进展》中,铁振军对60岁以上老年人胃液酸度测定,证明大多数老年人胃酸浓度低于青年人,胃液的pH值可达pH4以上。
本发明通过在药物组合物中加入适用作辅料的酸,可以使药物有效成分进入胃液之后,释放出酸,从而降低胃液的pH值,提高胃液的酸性。胃液pH值的降低为药物化合物1的溶解提供了有利的环境,从而促进了药物的溶解和吸收,提高了活性药物在体内的生物利用度。
附图说明
图1是表示药物溶液的pH值与式1所示化合物1的饱和溶解度关系的曲线;
图2是示出小鼠血药浓度随时间变化的曲线图。
具体实施方式
下面通过实施例和对比例进一步说明本发明,这些实施例只是用于说明本发明,本发明不限于以下实施例。凡是对本发明技术方案进行修改或者等同替换,而不脱离本发明技术方案的范围,均应涵盖在本发明的保护范围中。
实施例1
药物组合物的体外溶解度测定实验
本发明通过在原料药(化合物1)中加入酸,提高药物在具有不同pH值的溶解介质中的溶解度。下面就几种酸对化合物1的饱和溶解度的影响进行研究。
表1化合物1在具有不同pH值的介质中的饱和溶解度
介质 饱和溶解度(mg/ml)
pH1.0盐酸溶液 106.2
pH2.0盐酸溶液 64.1
pH3.0磷酸氢二钠-枸橼酸缓冲液 1.0
pH4.5醋酸盐缓冲液 3.1×10 -3
0.6×10 -3
pH6.8磷酸盐缓冲液 0.7×10 -3
将溶解介质的pH与药物的饱和溶解度的关系制作成曲线,如图1所示。
从图1可以看出,在pH值为3处,原料药化合物1在溶解介质中的溶解度已经变得很小。根据临床考察,以不超过200mg化合物1的剂量的临床最大用量计,一般在临床服药方式会以100ml-200ml温水送服,故在pH3.0的介质中化合物1具有1mg/ml的饱和溶解度即可满足临床需求。pH值增大时,化合物1的饱和溶解度太小,难以满足临床需求。
另外,发明人进一步测试了在pH值为3.1至pH值为4.0的数值范围内溶解介质的pH与药物的饱和溶解度的关系,结果如下表2所示。
表2
介质 饱和溶解度(mg/ml)
pH3.1磷酸氢二钠-枸橼酸缓冲液 0.450
pH3.3磷酸氢二钠-枸橼酸缓冲液 0.212
pH3.5磷酸氢二钠-枸橼酸缓冲液 0.093
pH3.8磷酸氢二钠-枸橼酸缓冲液 0.034
pH3.8醋酸盐缓冲液 0.024
pH4.0醋酸盐缓冲液 9×10 -3
由表1、表2和图1可以看出,化合物1在酸性条件下溶解度较高,且随着pH值的降低,溶解度呈现增大的趋势,由此可知化合物1的溶解度具有明显的pH依赖性。
下面的表3列出了几种常见的酸辅料在100ml的水中溶解并调节至3.0附近的pH值时所用的酸的量。在选取测试所用的pH值过程中,发明人发现日常饮料可口可乐的pH为约2.4,因此在测量所用酸量是基于pH=2.40时的测量结果。
表3
Figure PCTCN2020139414-appb-000003
由表1、表2和表3可知,所用的多种酸溶液以及图1的曲线可以推测,在处方中,本发明列出的其他药学上可接受的酸也能实现提高抗肿瘤药物化合物1的体内生物利用度的目的。从表3可以看到,为了使水的pH值降低至2.40,需要添加的酒石酸和富马酸的用量均超过口服最大用量,需要考虑其安全性。另外,增加谷氨酸的量也能使pH值降至3.0以下,但是由 于谷氨酸溶解速度慢,不适合实际的临床需求。
药物在不同pH的药代动力学研究实验
本实验使用的药物处方A如下表4所示。
表4处方A的组成
Figure PCTCN2020139414-appb-000004
实验1
将化合物1的样品加入0.9%NaCl注射液中,涡旋5min,搅拌5min后,用1N HCl调pH到2.0,超声30min后,再用1N NaOH调pH到3.0,超声10min,得浓度为5mg/ml的白色混悬液。后经口灌胃至犬,动物样本数6只。分别于给药前及给药后15min、30min、1h、1.5h、2h、3h、4h、6h、8h、24h、48h取血浆样品检测。
实验2
将化合物1的样品研磨后,加入0.9%NaCl注射液中,搅拌5min,超声20min,得浓度为5mg/ml的白色混悬液。后经口灌胃至犬,动物样本数3只。分别于给药前及给药后15min、30min、1h、2h、3h、4h、6h、8h、24h、48h取血浆样品检测。
表5犬药代结果对比
Figure PCTCN2020139414-appb-000005
Figure PCTCN2020139414-appb-000006
由表5可知,处方A在溶液中加入一定量的盐酸后(调节pH值不大于3.0),化合物1的样品在犬体内的生物利用度有较明显的提升,且个体差异更小。
实施例2
发明人以枸橼酸为例,研究了在片剂中添加枸橼酸后对化合物1的生物利用度的影响。在该实验中,设计了三种片剂处方:处方A、处方A 1和处方C。处方A与处方A 1、处方C在保持化合物1的原料药处方比例基本不变的情况下,对处方中填充剂的比例和粘合剂、崩解剂的种类进行了变更。片剂的处方如表6所示。
表6
Figure PCTCN2020139414-appb-000007
Figure PCTCN2020139414-appb-000008
采用由处方A、处方A 1和处方C制成的片剂,测量化合物1在小鼠体内的生物利用度。
分别取处方A、处方A 1和处方C的片剂样品进行研磨,加入生理盐水,涡旋,超声,配制成1mg/ml的混悬液,经口灌胃小鼠,动物样本6只。在给药后的5min、15min、30min、1h、2h、4h、8h、12h、24h的时刻,取血浆进行样品检测。
采用处方A与处方A 1进行测量时的血药浓度-时间的数据(10mg/kg小鼠)见下面的表7。
表7血药浓度-时间数据(10mg/kg小鼠)
Figure PCTCN2020139414-appb-000009
根据表7绘制药物化合物1的平均血药浓度与时间关系的曲线,具体的曲线见图2。
由表7和图2可知,处方A与处方A 1在小鼠体内24h血药浓度变化,两个处方的片剂样品在相同时间段的血药浓度值在同一水平线上,未有特别明显的差别,故可认为处方中的这几个辅料(包括聚山梨酯80)的变化不会影响化合物1在动物体内的药代动力学。即,在辅料中去除聚山梨酯80 也不影响化合物1本身的体内吸收。
一般认为,聚山梨酯80作为表面活性剂可能会促进化合物1在体内的吸收,但本实验的数据(表6和表7)证明了聚山梨酯80并不会影响化合物1的体内吸收,为从处方C中去除聚山梨酯80,避免聚山梨酯80的使用提供了依据。
在确定处方中的几种辅料对化合物1在动物体内的药代动力学未有显著影响的前提下,对根据处方A和处方C制备的片剂在犬体内的药代动力学进行对比研究,结果见表8。
表8处方A与处方C的犬药代动力学实验结果对比
Figure PCTCN2020139414-appb-000010
由表8可知,在相同剂量下,处方C(添加了枸橼酸)较处方A的C max、AUC均有较大程度的提升,此结果可说明在化合物1的制剂中添加枸橼酸,化合物1的原料药的体内生物利用度更高。
另外,综合表7和表8的数据,可以证明片剂中增加的辅料枸橼酸是影响化合物1在动物体内的生物利用度的关键参数。因此,本发明通过化合物1与酸的结合,可以有效增加化合物1在体内的溶解度和生物利用度,有利于提高药物的临床应用效果。基于上面公开的内容,可以推断,当药学上可接受的酸用在胶囊剂、颗粒剂、丸剂或散剂等固体制剂中时,也能够产生提高化合物1在体内的溶解度和生物利用度。
实施例3
表9处方C和处方D的组成
原辅料名称 处方C 处方D
  (g/1000片)  
化合物1的原料药 25 25
一水乳糖 / 42.5
无水枸橼酸 100 100
微晶纤维素101 100 25
羟丙基纤维素 4 2
交联羧甲基纤维素钠 25 2.5
微晶纤维素102 100 150
羧甲基淀粉钠 25 25
二氧化硅 / 3
硬脂酸镁 3 3
处方D的制备过程如下:
预处理:化合物1的原料药微粉化;无水枸橼酸过40目,备用。
预混:化合物1的原料药、微晶纤维素101、一水乳糖、交联羧甲基纤维素钠递加混合,过40目筛3次混合,得混合粉末。
制粒:向混合粉末中加入10%的羟丙基纤维素水溶液,过20目筛制粒,得湿颗粒。
干燥:湿颗粒放入60℃烘箱中,烘干至物料水分小于3.0%,得干颗粒。
整粒:干颗粒过20目筛,整粒。
总混:干颗粒与二氧化硅递加混合,后加入羧甲淀粉钠、微晶纤维素102、无水枸橼酸、硬脂酸镁,混合均匀。
压片:11mm圆形浅凹冲压片。
对由处方C和处方D得到的片剂的稳定性进行测试,测试结果分别如表10和表11所示。
表10处方C(化合物1的原料药与柠檬酸直接混合制粒)的稳定性
Figure PCTCN2020139414-appb-000011
Figure PCTCN2020139414-appb-000012
表11处方D(化合物1的原料药制粒后外加二氧化硅、枸橼酸)稳定性
Figure PCTCN2020139414-appb-000013
Figure PCTCN2020139414-appb-000014
从表10和表11可以看出,在化合物1的原料药制粒后加入二氧化硅对原料药进行包覆以将原料药与酸隔离的片剂中,其杂质状况明显优于未添加二氧化硅的处方C。其主要原因在于,二氧化硅吸附在制料后的原料药颗粒表面,形成包覆层,起到将原料药与酸进行隔离的作用。从实验数据可以判断,化合物1的原料药与酸直接接触时,稳定性较差,而通过对原料药颗粒的外表面进行包覆,将原料药与酸隔离开,不仅可以提高化合物1的溶解度,还可以提高药物在存储和运输过程中的稳定性。

Claims (10)

  1. 一种抗肿瘤药物组合物,其特征在于,所述药物组合物包括式1所示的化合物以及药物学上可接受的酸;
    Figure PCTCN2020139414-appb-100001
  2. 根据权利要求1所述的药物组合物,其特征在于,所述药物组合物还包括包覆式1所示的化合物的表面的包覆层;
    优选地,所述包覆层的材料选自二氧化硅、二氧化钛、羟丙甲纤维素、羟丙基纤维素、滑石粉、聚乙二醇、聚乙烯醇的一种或多种。
  3. 根据权利要求1所述的药物组合物,其特征在于,所述药学上可接受的酸能够使式1所示的化合物在pH≤3.0的水中具有0.5mg/ml以上,优选具有1.0mg/ml以上的饱和溶解度。
  4. 根据权利要求1所述的药物组合物,其特征在于,所述药学上可接受的酸选自磷酸、海藻酸、枸椽酸、富马酸、谷氨酸、酒石酸、苹果酸中的一种或多种。
  5. 根据权利要求1所述的药物组合物,其特征在于,以重量计,式1所示的化合物与所述药学上可接受的酸的比例为1∶2~1∶6,优选1∶4。
  6. 根据权利要求1至5任一项所述的药物组合物,其特征在于,所述药物组合物还包括药学上可接受的辅料;
    优选地,所述辅料选自微晶纤维素、乳糖、交联聚维酮、羟丙基纤维素、硬脂酸镁、聚维酮、交联羧甲基纤维素钠、羧甲基淀粉钠中的一种或多种。
  7. 根据权利要求1至5任一项所述的药物组合物,其特征在于,所述 药物组合物为片剂、胶囊剂、颗粒剂、丸剂或散剂。
  8. 一种提高式1所示化合物在水中的溶解度的方法,其特征在于,所述方法包括在式1所示的化合物中加入药学上可接受的酸;
    Figure PCTCN2020139414-appb-100002
    优选地,所述方法还包括:在加入所述药学上可接受的酸之前,将式1所示的化合物进行湿法制粒后添加包覆材料,混合,以将式1所示的化合物进行包覆;其中,所述包覆材料选自二氧化硅、二氧化钛、羟丙甲纤维素、羟丙基纤维素、滑石粉、聚乙二醇、聚乙烯醇中的至少一种。
  9. 根据权利要求8所述的方法,其特征在于,所述药学上可接受的酸选自磷酸、海藻酸、枸椽酸、富马酸、谷氨酸、酒石酸、苹果酸中一种或多种;优选地,所述药学上可接受的酸使式1所示的化合物在pH≤3.0的水中具有1.0mg/ml以上的饱和溶解度。
  10. 根据权利要求8所述的方法,其特征在于,以重量计,式1所示的化合物与所述药学上可接受的酸的添加比例为1∶2~1∶6,优选1∶4。
PCT/CN2020/139414 2019-12-30 2020-12-25 一种抗肿瘤药物组合物以及提高化合物的溶解度的方法 WO2021136089A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080063242.9A CN114364386B (zh) 2019-12-30 2020-12-25 一种抗肿瘤药物组合物以及提高化合物的溶解度的方法

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201911399012.5 2019-12-30
CN201911399012.5A CN113116894A (zh) 2019-12-30 2019-12-30 一种抗肿瘤药物组合物以及提高化合物的溶解度的方法

Publications (1)

Publication Number Publication Date
WO2021136089A1 true WO2021136089A1 (zh) 2021-07-08

Family

ID=76686464

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/139414 WO2021136089A1 (zh) 2019-12-30 2020-12-25 一种抗肿瘤药物组合物以及提高化合物的溶解度的方法

Country Status (2)

Country Link
CN (2) CN113116894A (zh)
WO (1) WO2021136089A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1197456A (zh) * 1995-09-21 1998-10-28 帕斯医药股份有限公司 新的含有对酸不稳定的奥美拉唑的组合物及其制备方法
US20100093727A1 (en) * 2008-10-14 2010-04-15 Ning Xi Compounds and methods of use
CN102093421A (zh) * 2011-01-28 2011-06-15 广州盈升生物科技有限公司 一种含磷取代基的喹啉类化合物及其制备方法、以及含有该化合物的药物组合物及其应用
CN102408411A (zh) * 2011-09-19 2012-04-11 广州盈升生物科技有限公司 一种含喹啉基的羟肟酸类化合物及其制备方法、以及含有该化合物的药物组合物及其应用
CN106290600A (zh) * 2015-06-12 2017-01-04 北京康辰药业股份有限公司 一种用液相色谱法分离康尼替尼及有关物质的方法

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20120116422A (ko) * 2009-12-29 2012-10-22 코와 가부시키가이샤 경구 투여용 의약 조성물

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1197456A (zh) * 1995-09-21 1998-10-28 帕斯医药股份有限公司 新的含有对酸不稳定的奥美拉唑的组合物及其制备方法
US20100093727A1 (en) * 2008-10-14 2010-04-15 Ning Xi Compounds and methods of use
CN102093421A (zh) * 2011-01-28 2011-06-15 广州盈升生物科技有限公司 一种含磷取代基的喹啉类化合物及其制备方法、以及含有该化合物的药物组合物及其应用
CN102408411A (zh) * 2011-09-19 2012-04-11 广州盈升生物科技有限公司 一种含喹啉基的羟肟酸类化合物及其制备方法、以及含有该化合物的药物组合物及其应用
CN106290600A (zh) * 2015-06-12 2017-01-04 北京康辰药业股份有限公司 一种用液相色谱法分离康尼替尼及有关物质的方法

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHEN YOU-SHENG,ZHANG JIAN-HONG: "Research on How to Improve Solubility of Water-Insoluable Drugs", STRAIT PHARMACEUTICAL JOURNAL, vol. 20, no. 7, 15 July 2008 (2008-07-15), pages 25 - 27, XP055825978, ISSN: 1006-3765 *
YU LI, QIAN ZHAO, PEI HU , JI JIANG: "CX1003 Quantification by Ultra-Performance LC–MS/MS in Human Plasma and its Application to a Pharmacokinetic Study in Solid Tumor Patients", BIOANALYSIS, vol. 11, no. 16, 3 September 2019 (2019-09-03), pages 1483 - 1493, XP009528826, ISSN: 1757-6180, DOI: 10.4155/bio-2019-0096 *

Also Published As

Publication number Publication date
CN113116894A (zh) 2021-07-16
CN114364386A (zh) 2022-04-15
CN114364386B (zh) 2023-06-16

Similar Documents

Publication Publication Date Title
EP2580206B1 (en) Pharmaceutical composition comprising amide derivative or pharmaceutically acceptable salt thereof
TWI744224B (zh) 固形製劑
TW201418244A (zh) 醫藥組合物
EP3862318A1 (en) Porous silica particle composition
WO2021238978A1 (zh) 含硝羟喹啉前药的药物组合物及其制备方法和应用
EP2540318B1 (en) Sustained-release solid preparation for oral use
US20110091535A1 (en) Solid pharmaceutical formulation
EP2590652A1 (en) Pharmaceutical compositions containing vanoxerine
EP4032529A1 (en) Sorafenib pharmaceutical composition having high bioavailability and application
WO2018095403A1 (zh) 一种吡啶酮类衍生物药物组合物及其制备方法
TW201821063A (zh) 包含兩種不同活性成分的醫藥組成物
WO2021136089A1 (zh) 一种抗肿瘤药物组合物以及提高化合物的溶解度的方法
CN110404079A (zh) 一种不含碳酸盐、低基因毒性杂质含量的喹啉衍生物或其盐的药物组合物
JP2020518611A (ja) 水溶解度及びバイオアベイラビリティが改善された組成物
JP7370126B2 (ja) エルロチニブを有効成分とする医薬錠剤
TWI356711B (en) Saquinavir mesylate oral dosage form
WO2011079764A1 (zh) 一种右旋佐匹克隆固体制剂及其制备方法
WO2019230937A1 (ja) 溶出性に優れた経口固形製剤
WO2018049989A1 (zh) 一种瑞舒伐他汀钙药物组合物及其制备方法
US20130035344A1 (en) Pharmaceutical composition for oral administration
EP2694039B1 (en) Solid preparation
TWI278316B (en) Highly absorptive solid preparation
CN114727965B (zh) 一种jak激酶抑制剂药物组合物
WO2022042646A1 (zh) 盐酸鲁拉西酮组合物及其制备方法
KR20180060705A (ko) 에제티미브 및 로수바스타틴을 포함하는 경구용 복합정제

Legal Events

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

Ref document number: 20909713

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20909713

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 20909713

Country of ref document: EP

Kind code of ref document: A1