WO2022042738A1 - Pharmaceutical composition containing cdk4/6 inhibitor - Google Patents

Pharmaceutical composition containing cdk4/6 inhibitor Download PDF

Info

Publication number
WO2022042738A1
WO2022042738A1 PCT/CN2021/115486 CN2021115486W WO2022042738A1 WO 2022042738 A1 WO2022042738 A1 WO 2022042738A1 CN 2021115486 W CN2021115486 W CN 2021115486W WO 2022042738 A1 WO2022042738 A1 WO 2022042738A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical composition
fluoro
microcrystalline cellulose
compound
lactose monohydrate
Prior art date
Application number
PCT/CN2021/115486
Other languages
French (fr)
Chinese (zh)
Inventor
尹磊
姚郑林
Original Assignee
甘李药业股份有限公司
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 甘李药业股份有限公司 filed Critical 甘李药业股份有限公司
Priority to CN202180053613.XA priority Critical patent/CN116113628A/en
Publication of WO2022042738A1 publication Critical patent/WO2022042738A1/en

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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the invention belongs to the field of pharmaceutical preparations, in particular to a kind of 5-fluoro-4-(7'-fluoro-2'-methyl spiro[cyclopentane-1,3'-indol]-5'-yl)-N
  • the cell cycle is an important part of cell life activities. In the normal cell growth process, the realization of the cell cycle process depends on the precise and tight regulation of the cell cycle by various regulatory factors.
  • the core of these regulatory factors is cyclin-dependent protein kinase (Cyclin Dependent Kinase, CDK) and its positive and negative regulators - cyclin (Cyclin) and cyclin-dependent protein kinase inhibitor (CDI).
  • CDK-Cyclin complex formed by cyclin-dependent protein kinases and cyclins is involved in cell growth, proliferation, dormancy or entering apoptosis.
  • cyclins are periodically and continuously expressed and degraded, and respectively bind to their transiently activated CDKs. Through CDK activity, they catalyze the phosphorylation of different substrates to achieve the advancement of different phases of the cell cycle. and transformation.
  • CDK1-CDK13 13 members of the CDK family have been found, namely CDK1-CDK13; CDK1, CDK2, CDK3, CDK4 and CDK6 are involved in regulating cell proliferation, and CDK7, CDK8, CDK9, CDK11, CDK12 and CDK13 are involved in regulating transcription.
  • Cyclin is divided into A-L subtypes, and different CDKs are connected to different subtypes of Cyclin.
  • Cyclin D family (Cyclin D1, D2, D3) begins to express in G1 phase, binds and activates CDK4 and CDK6, and forms CDK4/6-Cyclin D complex, including retinoblastoma protein (Rb) phosphorylation of a series of substrates. After Rb is phosphorylated, it releases the proteins that bind to it and are inhibited by it, mainly transcription factors E2F, etc.
  • Rb retinoblastoma protein
  • E2F activates and transcribes some genes necessary for entering the S phase (Ma Ke, Research Progress in Anti-tumor Effects of CDK4/6 Inhibitors, "Foreign Studies” Medicine and Antibiotics Volume, 2013, 34(5): 197-202). If the balance is disturbed due to various factors, whether the signal that promotes cell proliferation is increased, or the signal that inhibits cell proliferation is weakened to a certain extent, cell proliferation will be out of control, and tumors will appear. Studies have found that about 80% of human tumors have abnormalities in the Cyclin D-CDK4/6-INK4-Rb pathway (1.Malumbres M, Barbacid M., To cycle or not to cycle: a critical decision in cancer[J]. Nature Reviews Cancer, 2001, 1(3): 222; 2.
  • CDK4/6 as an anti-tumor target are: (1) Most proliferating cells depend on CDK2 or CDK4/6 for proliferation, but CDK4/6 inhibitors do not show the cytotoxicity of "pan-CDK inhibitors", such as Myelosuppression and intestinal reaction; (2) Preclinical experiments have shown that if the level of Cyclin D in cells is increased or p16INK4a is inactivated, the sensitivity of cells to drugs can be increased. increased drug targeting.
  • a technical problem to be solved by the present invention is to provide a pharmaceutical composition based on compound 1 as an active ingredient that meets the needs of oral administration.
  • Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient with improved solubility properties and lower risk of food effects.
  • Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient with reduced hygroscopicity.
  • Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient with improved fluidity.
  • Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient with improved dissolution properties.
  • Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient with improved stability.
  • Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient having two or more of the above properties (preferably having all of the above properties).
  • Another technical problem to be solved by the present invention is to provide a solid oral pharmaceutical preparation made from the above-mentioned pharmaceutical composition.
  • the present invention provides a pharmaceutical composition using Compound 1 or a pharmaceutically acceptable salt thereof as an active ingredient, the pharmaceutical composition dissolves rapidly, has improved solubility, stability, fluidity and reduced moisture absorption Sexual and food effect risks, and the quality is stable and controllable.
  • the present invention provides a pharmaceutical composition containing, as an active ingredient, 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3] '-Indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine or a pharmaceutically acceptable salt thereof and dilution It is characterized in that the diluent is selected from one or more of microcrystalline cellulose, lactose or its hydrate, and sugar alcohol.
  • the pharmaceutically acceptable salt is selected from 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5' -yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine fumarate, maleate, adipate and succinic acid one or more of the salts.
  • the pharmaceutically acceptable salt is 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5' -yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine fumarate (hereinafter referred to as "Compound A").
  • the solubility of the pharmaceutical compositions of the present invention will be significantly improved relative to the use of the free base of Compound 1, while allowing the pharmaceutical compositions of the present invention to have a reduced risk of food effects , and compared to the use of other salts of compound 1 such as hydrochloride and mesylate, the obtained pharmaceutical composition can also have improved hygroscopicity.
  • the content of Compound 1 or a pharmaceutically acceptable salt thereof is 3.0% or more, preferably 4.0% or more, preferably 4%- 80%, preferably 5%-62%, preferably 6%-50%, more preferably 6%-25%, most preferably 8%-23%.
  • the pharmaceutical composition of the present invention contains different dosages of active pharmaceutical ingredients, which can meet the needs of different patients, and choose different dosages of medicines according to the severity of different diseases, which is helpful for individualized treatment and avoids adverse effects caused by improper medication.
  • the diluent is selected from one or more of microcrystalline cellulose, lactose monohydrate, mannitol and sorbitol.
  • the diluent is a mixture of microcrystalline cellulose and lactose monohydrate, or a mixture of microcrystalline cellulose and mannitol.
  • the microcrystalline cellulose is microcrystalline cellulose PH101 or microcrystalline cellulose PH102; preferably, the lactose monohydrate is lactose monohydrate FlowLac 100 or lactose monohydrate Tablettose 80; preferably, the mannitol It is mannitol 100SD.
  • the diluent is a mixture of mannitol 100SD and microcrystalline cellulose PH101, a mixture of lactose monohydrate Tablettose 80 and microcrystalline cellulose PH102, or a mixture of microcrystalline cellulose PH102 and lactose monohydrate FlowLac 100 mixture.
  • the weight ratio of lactose monohydrate and microcrystalline cellulose is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5:1-1:1.5, most preferably Preferably it is 1:1; or the weight ratio of the mannitol and microcrystalline cellulose is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5:1-1:1.5, most preferably 1:1.
  • the further selection of the diluent and the dosage ratio of the diluent in the present invention is beneficial to improve the dissolution rate of the pharmaceutical composition and has good dissolution consistency.
  • the content of the diluent is more than 20% of the total weight of the pharmaceutical composition, preferably more than 30%, preferably more than 40%, preferably 20-90%, preferably 50-90% , preferably 60-90%, more preferably 60%-85%, further preferably 65%-80%.
  • the pharmaceutical composition may further include a glidant, the glidant is selected from one or both of talc and colloidal silicon dioxide, more preferably, the glidant For colloidal silica.
  • the content of the glidant is 0.5%-15% of the total weight of the pharmaceutical composition, preferably 1%-10%, more preferably 1%-3%, most preferably 1% , 2% or 3%.
  • the addition of the glidant in the invention is beneficial to improve the fluidity of the pharmaceutical composition, improve the stability of the filling amount of the filling capsule, and the difference in the filling amount is small.
  • the pharmaceutical composition may further comprise a disintegrant, preferably, the disintegrant is selected from the group consisting of croscarmellose sodium, sodium carboxymethyl starch, crospovidone and low Substituting one or more of hydroxypropyl cellulose, more preferably, the disintegrant is croscarmellose sodium.
  • the disintegrant is selected from the group consisting of croscarmellose sodium, sodium carboxymethyl starch, crospovidone and low Substituting one or more of hydroxypropyl cellulose, more preferably, the disintegrant is croscarmellose sodium.
  • the content of the disintegrant is 0.5%-15% of the total weight of the pharmaceutical composition, preferably 1%-10%, more preferably 1%-5%, more preferably 1% -3%, most preferably 1%, 2%, 3%, 4%, or 5%.
  • the disintegrant and the selection of the dosage of the present invention help to improve the dissolution rate of the pharmaceutical composition and have good dissolution consistency.
  • the pharmaceutical composition may further include a lubricant, preferably, the lubricant is selected from one of magnesium stearate, stearic acid, sodium stearoyl fumarate and zinc stearate One or more, more preferably, the lubricant is magnesium stearate.
  • the content of the lubricant is 0.1%-5% of the total weight of the pharmaceutical composition, preferably 0.5%-3%, more preferably 1%-2%, most preferably 1%.
  • the selection of the lubricant and its dosage of the present invention further reduces the friction between the powders and between the powders and the mold wall, and is helpful for filling capsules or tableting.
  • the present invention provides a pharmaceutical composition containing the following components by weight:
  • the diluent is a mixture of microcrystalline cellulose and lactose monohydrate a mixture, or a mixture of microcrystalline cellulose and mannitol;
  • glidant is colloidal silica
  • disintegrant preferably, the disintegrant is selected from one of croscarmellose sodium, sodium carboxymethyl starch, crospovidone and low-substituted hydroxypropyl cellulose or various; and
  • the lubricant is selected from magnesium stearate, stearic acid , one or more of sodium stearoyl fumarate and zinc stearate.
  • the weight ratio of lactose monohydrate and microcrystalline cellulose is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5 : 1-1: 1.5, most preferably 1: 1; or the weight ratio of the mannitol and microcrystalline cellulose is 5: 1-1: 5, preferably 3: 1-1: 3, more preferably 1.5: 1-1:1.5, most preferably 1:1.
  • the pharmaceutical composition in the pharmaceutical composition, 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5'- yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine free base, the pharmaceutical composition comprises 0.1-1000 mg, preferably 1- 100 mg, more preferably 8 to 50 mg of active ingredient.
  • the present invention also provides a solid oral pharmaceutical preparation made from the pharmaceutical composition, preferably a powder, a capsule, a tablet or a granule, more preferably a capsule, wherein the capsule is made of the pharmaceutical composition as filler.
  • the solid oral pharmaceutical preparation comprises 0.1 ⁇ 1000mg, preferably 1 to 100 mg, more preferably 8 to 50 mg of active ingredient.
  • the capsules are prepared using a powder direct filling process.
  • the capsules of the present invention are loaded according to the free base content of compound 1, and a capsule filling machine is used to fill the medicinal powder, that is, the pharmaceutical composition of the present invention, into hard capsule shells of different specifications, and the differences in the loading of the capsules are investigated to avoid Affect product content and uniformity.
  • the present invention also provides the pharmaceutical composition of the present invention or the solid oral pharmaceutical preparation of the present invention, which is used for the treatment of CDK4/6-mediated disorders or diseases.
  • the present invention also provides the use of the pharmaceutical composition of the present invention or the solid oral pharmaceutical preparation of the present invention in preparing a medicament for treating CDK4/6-mediated disorders or diseases.
  • the present invention also provides a method for treating a CDK4/6-mediated disorder or disease, comprising administering the pharmaceutical composition of the present invention or the solid oral pharmaceutical formulation of the present invention to a subject in need thereof.
  • the CDK4/6 mediated disorder or disease is cancer, AIDS, atherosclerosis or restenosis after stent implantation, wherein the cancer is preferably a malignant solid tumor or a malignant non-solid tumor, more Breast cancer, lung cancer, prostate cancer, leukemia, brain cancer or gastric cancer are preferred, and glioma is even more preferred.
  • the present invention can significantly improve the fluidity of the obtained pharmaceutical composition powder, the obtained pharmaceutical composition has a larger bulk density, a smaller Carr index and an angle of repose, and the pharmaceutical composition powder has a uniform color and no color difference. .
  • the solubility of the pharmaceutical composition of the present invention will be further improved relative to the use of the free base of compound 1, and the pharmaceutical composition of the present invention will have a reduced risk of food effects, and is relatively
  • the pharmaceutical composition of the present invention has high stability under the experimental conditions of high temperature (60°C), high humidity (75%RH), strong light (5000lux) and acceleration (40°C/75%RH).
  • the pharmaceutical composition of the present invention can realize the preparation of capsules by directly filling the capsules with powder, the process is simple, the filling volume stability of the capsules prepared by filling is good, and the filling volume difference is small.
  • Figure 1 is a 1 H-NMR chart of the fumarate salt of compound 1 in CD 3 OD.
  • Figure 2 is a 1 H-NMR chart of the maleate salt of compound 1 in CD 3 Cl.
  • Figure 3 is a 1 H-NMR chart of the adipate salt of Compound 1 in CD 3 OD.
  • Figure 4 is a 1 H-NMR chart of the succinate salt of compound 1 in CDCl 3 .
  • Figure 5 is a 1 H-NMR chart of the mesylate salt of Compound 1 in CD 3 OD.
  • Figure 6 is the dissolution curve of the 10 mg specification pilot batch of Test Example 11 in pH 1.2 hydrochloric acid buffer.
  • Figure 7 is the dissolution curve of the 50 mg specification pilot batch of Test Example 11 in pH 1.2 hydrochloric acid buffer.
  • Mannitol 100SD Mannitol 100SD.
  • Microcrystalline Cellulose PH101 Microcrystalline Cellulose
  • Microcrystalline Cellulose PH102 Microcrystalline Cellulose
  • Lactose Monohydrate FlowLac 100 Lactose Monohydrate
  • Lactose Monohydrate Tablettose 80 Lactose Monohydrate
  • Colloidal Silica 200 Colloidal Silica
  • diluent refers to an excipient used to increase the weight and/or volume of a pharmaceutical composition to facilitate molding and sub-dosing.
  • the diluent described in the present invention is selected from one or more of microcrystalline cellulose, lactose or its hydrate, and sugar alcohol, preferably selected from one or more of microcrystalline cellulose, lactose monohydrate, mannitol and sorbitol one or more.
  • the diluent may be a single diluent or a mixture of two diluents.
  • the diluent is a mixture of two diluents, it is preferably microcrystalline cellulose (eg, microcrystalline cellulose PH101 or microcrystalline cellulose PH102) and lactose monohydrate (eg, lactose monohydrate FlowLac 100 or lactose monohydrate).
  • microcrystalline cellulose eg, microcrystalline cellulose PH101 or microcrystalline cellulose PH102
  • lactose monohydrate eg, lactose monohydrate FlowLac 100 or lactose monohydrate
  • Tablettose 80 or a mixture of microcrystalline cellulose (eg, microcrystalline cellulose PH101 or microcrystalline cellulose PH102) and mannitol (eg, mannitol 100SD), and the weight of the lactose monohydrate and microcrystalline cellulose
  • the ratio is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5:1-1:1.5, most preferably 1:1; or the weight ratio of the mannitol and microcrystalline cellulose It is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5:1-1:1.5, most preferably 1:1.
  • the "glidant" in the present invention refers to an excipient used to increase the fluidity of a material.
  • the glidant of the present invention is selected from one or both of talc and colloidal silicon dioxide, more preferably, the glidant is colloidal silicon dioxide.
  • the "disintegrant” in the present invention refers to an excipient used to promote the disintegration of the pharmaceutical composition in the gastrointestinal tract and increase the dissolution of the active ingredient.
  • the disintegrant of the present invention is selected from one or more of croscarmellose sodium, sodium carboxymethyl starch, crospovidone and low-substituted hydroxypropyl cellulose, more preferably, The disintegrating agent is croscarmellose sodium.
  • the "lubricant" used in the present invention refers to an excipient used to reduce the frictional force between particles of a pharmaceutical composition and improve the transmission and distribution of force.
  • the lubricant of the present invention is selected from one or more of magnesium stearate, stearic acid, sodium stearoyl fumarate and zinc stearate, more preferably, the lubricant is stearic acid magnesium.
  • compositions of the present invention may also contain other excipients other than diluents, glidants, disintegrants and lubricants.
  • excipients other than diluents, glidants, disintegrants and lubricants.
  • specific examples thereof include binders, flavoring agents, dispersing agents, coloring agents, fragrances, and the like.
  • solid oral pharmaceutical preparation in the present invention refers to a solid pharmaceutical preparation for oral administration.
  • the present invention relates to a solid oral pharmaceutical preparation made from the above-mentioned pharmaceutical composition, and its specific dosage form is preferably powder, capsule, tablet or granule, more preferably capsule, wherein the capsule is filled with the pharmaceutical composition .
  • the pharmaceutical composition of the present invention or its solid oral pharmaceutical preparation may contain 0.1-1000 mg, preferably 1-100 mg, more preferably 8-50 mg of active ingredients in the form of 5-fluoro-4-(7'-fluoro-2'-methylspiro) [Cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine free base content meter.
  • the content of the active ingredient in the pharmaceutical composition of the present invention or its solid oral pharmaceutical preparation can be 0.1 mg, 0.2 mg, 0.25 mg, 0.5 mg, 1 mg, 2 mg, 2.5 mg, 5 mg, 8 mg, 10 mg, 20 mg, 25 mg, 40mg, 50mg, 100mg, 150mg, 200mg, 250mg, 300mg, 500mg, 1000mg, etc.
  • the administration subject may be a human or a non-human mammal, more preferably a human.
  • the term “comprising” or similar terms “comprising” and “comprising” generally express an open meaning, but also includes a closed meaning defined by “consisting of” within its scope. condition.
  • a pharmaceutical composition comprising also includes the case of "a pharmaceutical composition consisting of”.
  • the above solution was filtered through a microporous filter, and transferred to the reactor, stirred, and dichloromethane and ethanol were distilled off at normal pressure, and then the temperature of the reactor was kept to 80 ⁇ 5 degrees Celsius, and the fumaric acid ( 1.0eq) of ethanol solution (12 volumes) was slowly dropped into the reaction kettle through a microporous filter, and kept stirring overnight. Cool to 20-30 degrees Celsius, continue stirring for at least 1 hour, centrifuge, and collect the filter cake.
  • the purpose of this test example is to compare the solubility of various salts (also called salt forms) and free bases obtained by the present invention.
  • the sample and the solvent were mixed in a centrifuge tube (the initial feeding amount was 10 mg/ml), and then the centrifuge tube was sealed and fixed on a rotating disk with a rotation speed of 25 rpm, and rotated and mixed at 37 °C. Samples were taken after 24 hours. The turbid samples were centrifuged, and the filtered supernatant was used to measure the HPLC concentration. If the sample dissolves, test the concentration of the resulting solution.
  • Test subjects free base, maleate, fumarate, adipate and succinate of compound 1.
  • Test results See Tables 1-3, solubility of different salt forms and free bases in water, simulated fasting intestinal fluid (FaSSIF) and simulated fed intestinal fluid (FeSSIF).
  • the solubility of maleate, fumarate, adipate and succinate of compound 1 in water was 12.9, 6.3, 6.5 and 7.5 mg/ml, respectively, which was significantly higher than that of the free base in water. Solubility in water (0.075 mg/ml). This suggests that when the above-mentioned salt of the present invention is used to formulate the pharmaceutical composition of the present invention, the solubility of the active ingredient Compound 1 can be significantly improved.
  • This test example compares the hygroscopicity of various salt forms and free bases of compound 1 provided by the present invention.
  • the dynamic moisture adsorption (DVS) test was used to test the moisture adsorption capacity of the salt and free base of compound 1 of the present invention when the humidity reached 80% RH, so as to compare the hygroscopicity.
  • Test subjects Compound 1 free base, maleate, fumarate, adipate, succinate, mesylate and hydrochloride.
  • the preparation method of the pharmaceutical composition powder of embodiment 2-3 is as follows:
  • step (2) the pretreatment powder obtained in step (2) is mixed with mannitol 100SD, microcrystalline cellulose PH101, and the winner is mixed powder for subsequent use;
  • step (4) adding magnesium stearate to the main mixed powder prepared in step (4) and mixing to obtain a total mixed powder of pharmaceutical composition powder.
  • Compound A is 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-( The fumarate salt of 1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine.
  • the bulk density of the pharmaceutical composition of Example 2-3 is significantly increased compared to the single active ingredient raw material Compound A, and the angle of repose and Carr's index are significantly reduced, indicating that the pharmaceutical composition of Example 2-3 is combined.
  • the fluidity of the powder is significantly improved, and it can realize the preparation of capsules by a simple powder direct filling process, reduce the risk of large differences in the filling volume of the capsules, and meet the requirements of capsule filling and industrialization. production requirements.
  • the pharmaceutical composition powders of Examples 4-6 were prepared by adopting the steps similar to those in Example 2-3.
  • the bulk density of the pharmaceutical composition powder of Example 4-6 is significantly increased compared to the single active ingredient raw material Compound A, and the angle of repose and Carr's index are significantly reduced, indicating that the medicine of Example 4-6 Compared with the single active ingredient compound A, the fluidity of the composition powder is significantly improved, and when it is subsequently used to prepare a preparation in the form of capsules, it will be able to reduce the risk of large differences in the filling volume of the capsules, and meet the requirements and requirements of capsule filling. requirements for industrial production.
  • the preparation method of the capsule of embodiment 7-9 is as follows:
  • step (2) (4) mixing the pretreated powder obtained in step (2) with microcrystalline cellulose PH102, lactose monohydrate FLOWLAC100 and croscarmellose sodium, and the winner is mixed powder for subsequent use;
  • step (4) adding magnesium stearate to the main mixed powder prepared in step (4) and mixing to obtain a total mixed powder of pharmaceutical composition powder.
  • 50mg capsules are based on 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5'-yl)-N-( The content of the free base of 5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine.
  • Compound A is 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1-methyl)
  • the dissolution rate of the prepared capsules of Examples 7-9 was determined. Using 900 ml of PH1.2 hydrochloric acid buffer as the medium, the water temperature was 37 ⁇ 0.5 °C, and the rotation speed was 50 rpm, and samples were taken at 6 time points such as 5 min, 15 min, 30 min, 45 min, 60 min, and 75 min, respectively. Each sampling volume was 10 ml and was filtered with a PFS filter (0.45 ⁇ m to 10 ⁇ m). For each dissolution test, repeat 3 capsules or 6 capsules. The dissolution results are shown in Table 10 below.
  • the pharmaceutical composition powders of Examples 10-13 were obtained by the steps similar to those of Examples 2-3.
  • the pharmaceutical compositions of Examples 11-13 with colloidal silicon dioxide added to the formula As can be seen from the data in Table 12, compared to the pharmaceutical composition of Example 10 without colloidal silicon dioxide, the pharmaceutical compositions of Examples 11-13 with colloidal silicon dioxide added to the formula, the angle of repose is further reduced. , the fluidity is further improved, and when the amount of colloidal silicon dioxide is in the range of about 1% to 3%, the fluidity of the pharmaceutical composition powder has no significant change.
  • the dissolution rate of the capsules obtained in Examples 14-15 was measured according to the method of Test Example 5, and the test results are shown in Table 14 below.
  • 10mg capsules are based on 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5'-yl)-N-( The content of the free base of 5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine.
  • Compound A is 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1-methyl)
  • the dissolution rate of the capsules obtained in Example 16 was measured according to the method of Test Example 5, and the test results are shown in Table 16.
  • Example 7 50mg specification
  • Example 16 10 capsules were sampled respectively, and the weight of a single capsule was detected (the first, middle and last three stages respectively) Refers to 5 minutes after the start of capsule filling, about half of the filling is completed, and 5 minutes before the end of filling), according to the "Chinese Pharmacopoeia" 2015 edition four 0103 capsules under the difference inspection method detection, the test results are shown in Tables 17 and 18.
  • Example 7 50 mg strength
  • Example 16 10 mg strength
  • the two specifications of the samples were placed under the same conditions for 10 days and 30 days under accelerated conditions of 40°C/75%RH (opening), respectively.
  • the free base content of compound A in the capsules was determined by HPLC, and the method of Test Example 5 was used. Determination of dissolution; respectively at 60°C (open), 25°C/75%RH (open), light (intensity 5000lux) (open), 40°C/75%RH (open), 40°C/75%RH (closed)
  • the changes of related substances were determined by HPLC.
  • the test results of changes in capsule content, dissolution results and related substances are as follows:
  • Capsule content means "free base content of compound A in capsules”.
  • Capsules were prepared using the formulations of Example 7 (50 mg strength) and Example 16 (10 mg strength), and 10,000 capsules were prepared each, and were divided into three batches.
  • the packaging adopts oral solid medicinal high density polyethylene bottle and oral solid medicinal child safety cap.
  • the drug properties were observed by visual observation, the free base content of Compound A and the changes of related substances were determined by HPLC, the dissolution was determined by the first method (basket method) of "Chinese Pharmacopoeia” 2015 Edition Part Four 0931 Dissolution Determination Method, and the "Chinese Pharmacopoeia” 2015 Edition Chinese Pharmacopoeia 2015 edition four 0832 moisture determination method first method (Fischer's method) to measure moisture absorption weight gain, the result is shown in table 23-26.
  • test results show that the capsules of the present invention obtained through enlarged production are stable after long-term storage under the conditions of high temperature (60° C.), strong light (illumination 5000 lux), and high humidity (75% RH), and the results of each test item are not significant. changes to meet quality standard limits. It is further proved that under the conditions of scaled-up production, the obtained pharmaceutical composition of the present invention also has high stability, and the preparation production process adopted in the present invention is simple and stable, and can meet the needs of large-scale industrial production.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

Disclosed is a pharmaceutical composition containing 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5'-yl)-N-(5-(1-methylpiperidine-4-yl)pyridine-2-yl)pyrimidine-2-amine as an active ingredient or a pharmaceutically acceptable salt thereof and a diluent. The pharmaceutical composition is quick to dissolve, has improved solubility, stability and fluidity, has reduced moisture absorption and food effect risks, and has stable and controllable quality. In addition, the pharmaceutical composition has a simple preparation formula and process and is more suitable for industrial mass production.

Description

一种含CDK4/6抑制剂的药物组合物A kind of pharmaceutical composition containing CDK4/6 inhibitor 技术领域technical field
本发明属于药物制剂领域,具体涉及一种5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺或其药学上可接受盐的药物组合物。The invention belongs to the field of pharmaceutical preparations, in particular to a kind of 5-fluoro-4-(7'-fluoro-2'-methyl spiro[cyclopentane-1,3'-indol]-5'-yl)-N A pharmaceutical composition of -(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine or a pharmaceutically acceptable salt thereof.
背景技术Background technique
细胞周期是细胞生命活动的重要部分,在正常的细胞生长过程中,细胞周期进程的实现有赖于各级调控因子对细胞周期精确而严密的调控。这些调控因子的核心是细胞周期蛋白依赖性蛋白激酶(Cyclin Dependent Kinase,CDK)及其正、负性调控因子——细胞周期蛋白(Cyclin)和细胞周期蛋白依赖性蛋白激酶抑制剂(CDI)。细胞周期蛋白依赖性蛋白激酶和细胞周期蛋白形成的CDK-Cyclin复合物,参与细胞的生长、增殖、休眠或者进入凋亡。在细胞周期的过程中,细胞周期蛋白周期性连续的表达和降解,并分别结合到由它们瞬时活化的CDK上,通过CDK活性,催化不同底物磷酸化,实现对细胞周期不同时相的推进和转化作用。The cell cycle is an important part of cell life activities. In the normal cell growth process, the realization of the cell cycle process depends on the precise and tight regulation of the cell cycle by various regulatory factors. The core of these regulatory factors is cyclin-dependent protein kinase (Cyclin Dependent Kinase, CDK) and its positive and negative regulators - cyclin (Cyclin) and cyclin-dependent protein kinase inhibitor (CDI). The CDK-Cyclin complex formed by cyclin-dependent protein kinases and cyclins is involved in cell growth, proliferation, dormancy or entering apoptosis. During the cell cycle, cyclins are periodically and continuously expressed and degraded, and respectively bind to their transiently activated CDKs. Through CDK activity, they catalyze the phosphorylation of different substrates to achieve the advancement of different phases of the cell cycle. and transformation.
目前CDK家族已经发现了13个成员,分别是CDK1-CDK13;其中CDK1、CDK2、CDK3、CDK4和CDK6与调节细胞增殖有关,CDK7、CDK8、CDK9、CDK11、CDK12和CDK13参与调控转录。At present, 13 members of the CDK family have been found, namely CDK1-CDK13; CDK1, CDK2, CDK3, CDK4 and CDK6 are involved in regulating cell proliferation, and CDK7, CDK8, CDK9, CDK11, CDK12 and CDK13 are involved in regulating transcription.
Cyclin分为A-L亚型,不同的CDK分别连接不同亚型的Cyclin。其中,Cyclin D家族(Cyclin D1、D2、D3)在G1期开始表达,结合并激活CDK4和CDK6,形成CDK4/6-Cyclin D复合物,使包括视网膜母细胞瘤蛋白(Retinoblastomaprotein,Rb)在内的一系列底物磷酸化。Rb磷酸化后释放与其结合并被其抑制的蛋白,主要是转录因子E2F等,E2F激活并转录进入S期所必须的一些基因(马珂,CDK4/6抑制剂抗肿瘤作用研究进展,《国外医药·抗生素分册》,2013,34(5):197-202)。如果由于各种因素使平衡被打破,无论是促进细胞增殖的信号增强,或者是抑制细胞增殖的信号减弱到某种程度,细胞增殖都会失控,进而出现肿瘤。研究发现,大约80%的人类肿瘤中存在Cyclin D-CDK4/6-INK4-Rb通路的异常(1.Malumbres M,Barbacid M.,To  cycle or not to cycle:a critical decision in cancer[J].Nature Reviews Cancer,2001,1(3):222;2.Shapiro GI.,Cyclin-dependent kinase pathways as targets for cancer treatment[J].J Clinical Oncology,2006,24(11):1770)。这条通路的改变,加速了G1期进程,使得肿瘤细胞增殖加快而获得生存优势。因此,对其的干预成为一种治疗策略,CDK4/6因此成为潜在的抗肿瘤的靶点之一。Cyclin is divided into A-L subtypes, and different CDKs are connected to different subtypes of Cyclin. Among them, Cyclin D family (Cyclin D1, D2, D3) begins to express in G1 phase, binds and activates CDK4 and CDK6, and forms CDK4/6-Cyclin D complex, including retinoblastoma protein (Rb) phosphorylation of a series of substrates. After Rb is phosphorylated, it releases the proteins that bind to it and are inhibited by it, mainly transcription factors E2F, etc. E2F activates and transcribes some genes necessary for entering the S phase (Ma Ke, Research Progress in Anti-tumor Effects of CDK4/6 Inhibitors, "Foreign Studies" Medicine and Antibiotics Volume, 2013, 34(5): 197-202). If the balance is disturbed due to various factors, whether the signal that promotes cell proliferation is increased, or the signal that inhibits cell proliferation is weakened to a certain extent, cell proliferation will be out of control, and tumors will appear. Studies have found that about 80% of human tumors have abnormalities in the Cyclin D-CDK4/6-INK4-Rb pathway (1.Malumbres M, Barbacid M., To cycle or not to cycle: a critical decision in cancer[J]. Nature Reviews Cancer, 2001, 1(3): 222; 2. Shapiro GI., Cyclin-dependent kinase pathways as targets for cancer treatment [J]. J Clinical Oncology, 2006, 24(11): 1770). Alteration of this pathway accelerates the G1 phase process, allowing tumor cells to proliferate faster and gain a survival advantage. Therefore, its intervention has become a therapeutic strategy, and CDK4/6 has thus become one of the potential anti-tumor targets.
CDK4/6作为抗肿瘤靶点的优势在于:(1)大多数增殖的细胞依赖CDK2或者CDK4/6增殖,但CDK4/6的抑制剂不表现出“泛-CDK抑制剂”的细胞毒性,如骨髓抑制和肠道反应;(2)临床前实验表明,如果细胞Cyclin D水平升高或者p16INK4a失活,能够增加细胞对药物的敏感性,由于肿瘤细胞相对于正常细胞存在上述现象,所以一定程度上增加了药物的靶向。The advantages of CDK4/6 as an anti-tumor target are: (1) Most proliferating cells depend on CDK2 or CDK4/6 for proliferation, but CDK4/6 inhibitors do not show the cytotoxicity of "pan-CDK inhibitors", such as Myelosuppression and intestinal reaction; (2) Preclinical experiments have shown that if the level of Cyclin D in cells is increased or p16INK4a is inactivated, the sensitivity of cells to drugs can be increased. increased drug targeting.
申请人申请了一系列新的取代的2-(吡啶-2-基)氨基嘧啶类化合物(WO2017/092635A1),它们显示出很好的针对CDK4/6的高选择性的活性。其中,专利文献WO2017/092635A1公开了式(I)的化合物The applicant applied for a series of new substituted 2-(pyridin-2-yl)aminopyrimidine compounds (WO2017/092635A1), which showed good and highly selective activities against CDK4/6. Among them, the patent document WO2017/092635A1 discloses the compound of formula (I)
Figure PCTCN2021115486-appb-000001
Figure PCTCN2021115486-appb-000001
其化学名称为:5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺(以下简称“化合物1”),合成方法具体描述于实施例17中。Its chemical name is: 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1 -Methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine (hereinafter referred to as "Compound 1"), the synthesis method is specifically described in Example 17.
然而,上述专利文献中没有公开如何获得溶出迅速且稳定的药物组合物。化合物1的溶解性较差。同时,化合物1的某些盐具有吸湿性的特征,且有流动性差、堆密度小等缺陷,因此需要提供一种溶出迅速且具有提高的溶解性、稳定性、流动性以及降低的吸湿性的药物组合物,从而解决其应用、存储及放大生产的工艺问题。However, the above-mentioned patent documents do not disclose how to obtain a pharmaceutical composition with rapid dissolution and stability. Compound 1 has poor solubility. At the same time, some salts of compound 1 have the characteristics of hygroscopicity, and have the defects of poor fluidity and low bulk density, so it is necessary to provide a compound that dissolves rapidly and has improved solubility, stability, fluidity and reduced hygroscopicity. Pharmaceutical compositions, thereby solving the process problems of their application, storage and scale-up production.
发明内容SUMMARY OF THE INVENTION
本发明所要解决的一个技术问题是提供一种满足口服给药需要的基于作为活性成分的化合物1的药物组合物。A technical problem to be solved by the present invention is to provide a pharmaceutical composition based on compound 1 as an active ingredient that meets the needs of oral administration.
本发明所要解决的另一个技术问题是提供一种具有改善的溶解性能和较低的食物效应风险的基于作为活性成分的化合物1的药物组合物。Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient with improved solubility properties and lower risk of food effects.
本发明所要解决的另一个技术问题是提供一种具有降低的吸湿性的基于作为活性成分的化合物1的药物组合物。Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient with reduced hygroscopicity.
本发明所要解决的另一个技术问题是提供一种具有改善的流动性的基于作为活性成分的化合物1的药物组合物。Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient with improved fluidity.
本发明所要解决的另一个技术问题是提供一种具有改善的溶出性能的基于作为活性成分的化合物1的药物组合物。Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient with improved dissolution properties.
本发明所要解决的另一个技术问题是提供一种具有改善的稳定性的基于作为活性成分的化合物1的药物组合物。Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient with improved stability.
本发明所要解决的另一个技术问题是提供一种具有以上特性中的两种或更多种(优选具有全部以上特性)的基于作为活性成分的化合物1的药物组合物。Another technical problem to be solved by the present invention is to provide a pharmaceutical composition based on Compound 1 as an active ingredient having two or more of the above properties (preferably having all of the above properties).
本发明所要解决的另一个技术问题是提供一种由上述药物组合物制成的固体口服药物制剂。Another technical problem to be solved by the present invention is to provide a solid oral pharmaceutical preparation made from the above-mentioned pharmaceutical composition.
在致力于获得期望的基于作为活性成分的化合物1的药物组合物及其固体口服药物制剂的研究过程中,发明人对药物组合物的配方进行了缜密的筛选实验,并且已经发现本发明所述的特定的药物组合物的配方组成能够解决以上技术问题,从而完成了本发明。据此,本发明提供了一种以化合物1或其药学上可接受的盐作为活性成分的药物组合物,该药物组合物溶出迅速,具有提高的溶解性、稳定性、流动性以及降低的吸湿性和食物效应风险,且质量稳定可控。In the course of research to obtain the desired pharmaceutical composition based on Compound 1 as an active ingredient and solid oral pharmaceutical preparations thereof, the inventors have carried out careful screening experiments on the formulation of the pharmaceutical composition, and have found that the The formula composition of the specific pharmaceutical composition can solve the above technical problems, thus completing the present invention. Accordingly, the present invention provides a pharmaceutical composition using Compound 1 or a pharmaceutically acceptable salt thereof as an active ingredient, the pharmaceutical composition dissolves rapidly, has improved solubility, stability, fluidity and reduced moisture absorption Sexual and food effect risks, and the quality is stable and controllable.
具体而言,本发明提供了一种药物组合物,所述药物组合物含有作为活性成分的5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺或其药学上可接受的盐以及稀释剂,其特征在于,所述稀释剂选自微晶纤维素、乳糖或其水合物、和糖醇中的一种或多种。Specifically, the present invention provides a pharmaceutical composition containing, as an active ingredient, 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3] '-Indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine or a pharmaceutically acceptable salt thereof and dilution It is characterized in that the diluent is selected from one or more of microcrystalline cellulose, lactose or its hydrate, and sugar alcohol.
发明人发现,当使用本发明的所述稀释剂时,能够显著提高所获得的药物组合物粉末的流动性,所获得的药物组合物具有较大的堆密度及较小的卡尔指数和休止角,且药物组合物粉末颜色均一、无色差。The inventors found that when the diluent of the present invention is used, the fluidity of the obtained pharmaceutical composition powder can be significantly improved, and the obtained pharmaceutical composition has a larger bulk density and smaller Carr's index and angle of repose , and the powder color of the pharmaceutical composition is uniform and no color difference.
在一个实施方案中,所述药学上可接受的盐选自5-氟-4-(7’-氟-2’- 甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺的富马酸盐、马来酸盐、己二酸盐和琥珀酸盐中的一种或多种。在另一个实施方案中,所述药学上可接受的盐为5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺富马酸盐(以下简称“化合物A”)。In one embodiment, the pharmaceutically acceptable salt is selected from 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5' -yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine fumarate, maleate, adipate and succinic acid one or more of the salts. In another embodiment, the pharmaceutically acceptable salt is 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5' -yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine fumarate (hereinafter referred to as "Compound A").
发明人发现,当采用本发明的所述盐时,相对于使用化合物1的游离碱,将显著提高本发明的药物组合物的溶解性,同时使得本发明的药物组合物具有降低的食物效应风险,且相对于使用化合物1的其它盐如盐酸盐和甲磺酸盐,还能使所获得的药物组合物具有改善的吸湿性。The inventors have found that when the salts of the present invention are used, the solubility of the pharmaceutical compositions of the present invention will be significantly improved relative to the use of the free base of Compound 1, while allowing the pharmaceutical compositions of the present invention to have a reduced risk of food effects , and compared to the use of other salts of compound 1 such as hydrochloride and mesylate, the obtained pharmaceutical composition can also have improved hygroscopicity.
在一个实施方案中,以化合物1的游离碱的含量计,化合物1或其药学上可接受的盐的含量为所述药物组合物总重的3.0%以上、优选4.0%以上、优选4%-80%、优选5%-62%、优选6%-50%、更优选为6%-25%、最优选为8%-23%。本发明的药物组合物中含有不同用量的药物活性成分,可以满足不同患者使用,根据不同疾病严重程度选择不同用量的药物,有助于进行个性化治疗,以免产生用药不当引起的不良作用。In one embodiment, based on the content of the free base of Compound 1, the content of Compound 1 or a pharmaceutically acceptable salt thereof is 3.0% or more, preferably 4.0% or more, preferably 4%- 80%, preferably 5%-62%, preferably 6%-50%, more preferably 6%-25%, most preferably 8%-23%. The pharmaceutical composition of the present invention contains different dosages of active pharmaceutical ingredients, which can meet the needs of different patients, and choose different dosages of medicines according to the severity of different diseases, which is helpful for individualized treatment and avoids adverse effects caused by improper medication.
在一个实施方案中,所述稀释剂选自微晶纤维素、一水乳糖、甘露醇和山梨醇中的一种或多种。在更优选的实施方案中,所述稀释剂为微晶纤维素和一水乳糖的混合物、或微晶纤维素和甘露醇的混合物。优选地,所述微晶纤维素为微晶纤维素PH101或微晶纤维素PH102;优选地,所述一水乳糖为一水乳糖FlowLac 100或一水乳糖Tablettose 80;优选地,所述甘露醇为甘露醇100SD。在进一步优选的实施方案中,所述稀释剂为甘露醇100SD和微晶纤维素PH101的混合物、一水乳糖Tablettose 80和微晶纤维素PH102的混合物、或微晶纤维素PH102和一水乳糖FlowLac 100的混合物。In one embodiment, the diluent is selected from one or more of microcrystalline cellulose, lactose monohydrate, mannitol and sorbitol. In a more preferred embodiment, the diluent is a mixture of microcrystalline cellulose and lactose monohydrate, or a mixture of microcrystalline cellulose and mannitol. Preferably, the microcrystalline cellulose is microcrystalline cellulose PH101 or microcrystalline cellulose PH102; preferably, the lactose monohydrate is lactose monohydrate FlowLac 100 or lactose monohydrate Tablettose 80; preferably, the mannitol It is mannitol 100SD. In a further preferred embodiment, the diluent is a mixture of mannitol 100SD and microcrystalline cellulose PH101, a mixture of lactose monohydrate Tablettose 80 and microcrystalline cellulose PH102, or a mixture of microcrystalline cellulose PH102 and lactose monohydrate FlowLac 100 mixture.
在一个实施方案中,所述一水乳糖和微晶纤维素的重量比为5∶1-1∶5,优选为3∶1-1∶3,更优选为1.5∶1-1∶1.5,最优选为1∶1;或所述甘露醇和微晶纤维素的重量比为5∶1-1∶5,优选为3∶1-1∶3,更优选为1.5∶1-1∶1.5,最优选为1∶1。本发明对稀释剂及其用量配比的进一步选择,有利于提高药物组合物的溶出度,具有良好的溶出一致性。In one embodiment, the weight ratio of lactose monohydrate and microcrystalline cellulose is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5:1-1:1.5, most preferably Preferably it is 1:1; or the weight ratio of the mannitol and microcrystalline cellulose is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5:1-1:1.5, most preferably 1:1. The further selection of the diluent and the dosage ratio of the diluent in the present invention is beneficial to improve the dissolution rate of the pharmaceutical composition and has good dissolution consistency.
在一个实施方案中,所述稀释剂的含量为所述药物组合物总重的20%以上,优选为30%以上,优选为40%以上,优选为20-90%,优选为 50-90%,优选为60-90%,更优选为60%-85%,进一步优选为65%-80%。In one embodiment, the content of the diluent is more than 20% of the total weight of the pharmaceutical composition, preferably more than 30%, preferably more than 40%, preferably 20-90%, preferably 50-90% , preferably 60-90%, more preferably 60%-85%, further preferably 65%-80%.
在一个实施方案中,所述药物组合物还可以包括助流剂,所述助流剂选自滑石粉和胶态二氧化硅中的一种或两种,更优选地,所述助流剂为胶态二氧化硅。In one embodiment, the pharmaceutical composition may further include a glidant, the glidant is selected from one or both of talc and colloidal silicon dioxide, more preferably, the glidant For colloidal silica.
在一个实施方案中,所述助流剂的含量为所述药物组合物总重的0.5%-15%,优选为1%-10%,更优选为1%-3%,最优选为1%、2%或3%。本发明加入助流剂,有利于改善药物组合物的流动性,提高了灌装胶囊的装量稳定性,装量差异小。In one embodiment, the content of the glidant is 0.5%-15% of the total weight of the pharmaceutical composition, preferably 1%-10%, more preferably 1%-3%, most preferably 1% , 2% or 3%. The addition of the glidant in the invention is beneficial to improve the fluidity of the pharmaceutical composition, improve the stability of the filling amount of the filling capsule, and the difference in the filling amount is small.
在一个实施方案中,所述药物组合物还可以包括崩解剂,优选地,所述崩解剂选自交联羧甲基纤维素钠、羧甲基淀粉钠、交联聚乙烯吡咯烷酮和低取代羟丙基纤维素中的一种或多种,更优选地,所述崩解剂为交联羧甲基纤维素钠。In one embodiment, the pharmaceutical composition may further comprise a disintegrant, preferably, the disintegrant is selected from the group consisting of croscarmellose sodium, sodium carboxymethyl starch, crospovidone and low Substituting one or more of hydroxypropyl cellulose, more preferably, the disintegrant is croscarmellose sodium.
在一个实施方案中,所述崩解剂的含量为所述药物组合物总重的0.5%-15%,优选为1%-10%,更优选为1%-5%,更优选为1%-3%,最优选为1%、2%、3%、4%、或5%。本发明的崩解剂及其用量的选择,有助于提高药物组合物的溶出度,并且具有良好的溶出一致性。In one embodiment, the content of the disintegrant is 0.5%-15% of the total weight of the pharmaceutical composition, preferably 1%-10%, more preferably 1%-5%, more preferably 1% -3%, most preferably 1%, 2%, 3%, 4%, or 5%. The disintegrant and the selection of the dosage of the present invention help to improve the dissolution rate of the pharmaceutical composition and have good dissolution consistency.
在一个实施方案中,所述药物组合物还可以包括润滑剂,优选地,所述润滑剂选自硬脂酸镁、硬脂酸、硬脂酰富马酸钠和硬脂酸锌中的一种或多种,更优选地,所述润滑剂为硬脂酸镁。In one embodiment, the pharmaceutical composition may further include a lubricant, preferably, the lubricant is selected from one of magnesium stearate, stearic acid, sodium stearoyl fumarate and zinc stearate One or more, more preferably, the lubricant is magnesium stearate.
在一个实施方案中,所述润滑剂的含量为所述药物组合物总重的0.1%-5%,优选为0.5%-3%,更优选为1%-2%,最优选为1%。本发明的润滑剂及其用量的选择,进一步减低粉末之间及粉末与模壁之间的摩擦力,有助于灌装胶囊或压片。In one embodiment, the content of the lubricant is 0.1%-5% of the total weight of the pharmaceutical composition, preferably 0.5%-3%, more preferably 1%-2%, most preferably 1%. The selection of the lubricant and its dosage of the present invention further reduces the friction between the powders and between the powders and the mold wall, and is helpful for filling capsules or tableting.
作为优选的实施方案,本发明提供了一种药物组合物,所述药物组合物含有以重量计的如下成分:As a preferred embodiment, the present invention provides a pharmaceutical composition containing the following components by weight:
(1)5%-40%、优选8%-35%,更优选10%-30%的作为活性成分的5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺富马酸盐;(1) 5%-40%, preferably 8%-35%, more preferably 10%-30% as active ingredient 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane] -1,3'-Indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine fumarate;
(2)50%-90%、优选60%-90%,优选60%-85%,更优选65%-80%的稀释剂,优选,所述稀释剂为微晶纤维素和一水乳糖的混合物、或微晶纤维素和甘露醇的混合物;(2) 50%-90%, preferably 60%-90%, preferably 60%-85%, more preferably 65%-80% diluent, preferably, the diluent is a mixture of microcrystalline cellulose and lactose monohydrate a mixture, or a mixture of microcrystalline cellulose and mannitol;
(3)0.5%-15%,优选为1%-10%,更优选为1%-3%,更优选为1%、 2%、或3%的助流剂,优选,所述助流剂为胶态二氧化硅;(3) 0.5%-15%, preferably 1%-10%, more preferably 1%-3%, more preferably 1%, 2%, or 3% of a glidant, preferably, the glidant is colloidal silica;
(4)0.5%-15%,优选为1%-10%,更优选为1%-5%,更优选为1%-3%,更优选为1%、2%、3%、4%、或5%的崩解剂,优选,所述崩解剂选自交联羧甲基纤维素钠、羧甲基淀粉钠、交联聚乙烯吡咯烷酮和低取代羟丙基纤维素中的一种或多种;和(4) 0.5%-15%, preferably 1%-10%, more preferably 1%-5%, more preferably 1%-3%, more preferably 1%, 2%, 3%, 4%, Or 5% disintegrant, preferably, the disintegrant is selected from one of croscarmellose sodium, sodium carboxymethyl starch, crospovidone and low-substituted hydroxypropyl cellulose or various; and
(5)0.1%-5%,优选为0.5%-3%,更优选为1%-2%,最优选为1%的润滑剂,优选所述润滑剂选自硬脂酸镁、硬脂酸、硬脂酰富马酸钠和硬脂酸锌中的一种或多种。(5) 0.1%-5%, preferably 0.5%-3%, more preferably 1%-2%, most preferably 1% lubricant, preferably the lubricant is selected from magnesium stearate, stearic acid , one or more of sodium stearoyl fumarate and zinc stearate.
在一个实施方案中,在所述药物组合物中,所述一水乳糖和微晶纤维素的重量比为5∶1-1∶5,优选为3∶1-1∶3,更优选为1.5∶1-1∶1.5,最优选为1∶1;或所述甘露醇和微晶纤维素的重量比为5∶1-1∶5,优选为3∶1-1∶3,更优选为1.5∶1-1∶1.5,最优选为1∶1。In one embodiment, in the pharmaceutical composition, the weight ratio of lactose monohydrate and microcrystalline cellulose is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5 : 1-1: 1.5, most preferably 1: 1; or the weight ratio of the mannitol and microcrystalline cellulose is 5: 1-1: 5, preferably 3: 1-1: 3, more preferably 1.5: 1-1:1.5, most preferably 1:1.
在一个实施方案中,在所述药物组合物中,以5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺游离碱的含量计,所述药物组合物包含0.1~1000mg,优选1~100mg,更优选8~50mg活性成分。In one embodiment, in the pharmaceutical composition, 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5'- yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine free base, the pharmaceutical composition comprises 0.1-1000 mg, preferably 1- 100 mg, more preferably 8 to 50 mg of active ingredient.
本发明还提供了一种由所述的药物组合物制成的固体口服药物制剂,优选散剂、胶囊剂、片剂或颗粒剂,更优选胶囊剂,其中所述胶囊剂以所述药物组合物作为填充物。The present invention also provides a solid oral pharmaceutical preparation made from the pharmaceutical composition, preferably a powder, a capsule, a tablet or a granule, more preferably a capsule, wherein the capsule is made of the pharmaceutical composition as filler.
在一个实施方案中,在所述固体口服药物制剂中,以5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺游离碱的含量计,所述固体口服药物制剂包含0.1~1000mg,优选1~100mg,更优选8~50mg活性成分。In one embodiment, in the solid oral pharmaceutical formulation, 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5' -yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine free base content, the solid oral pharmaceutical preparation comprises 0.1~1000mg, preferably 1 to 100 mg, more preferably 8 to 50 mg of active ingredient.
在一个实施方案中,所述胶囊剂采用粉末直接灌装工艺制备。In one embodiment, the capsules are prepared using a powder direct filling process.
本发明的胶囊剂是按化合物1的游离碱含量折算装量,使用胶囊填充机将药粉即本发明的药物组合物装入不同规格的硬胶囊壳中,并考察了胶囊的装量差异,避免影响产品含量及均匀度。The capsules of the present invention are loaded according to the free base content of compound 1, and a capsule filling machine is used to fill the medicinal powder, that is, the pharmaceutical composition of the present invention, into hard capsule shells of different specifications, and the differences in the loading of the capsules are investigated to avoid Affect product content and uniformity.
本发明还提供了本发明所述的药物组合物或本发明所述的固体口服药物制剂,其用于治疗CDK4/6介导的障碍或疾病。The present invention also provides the pharmaceutical composition of the present invention or the solid oral pharmaceutical preparation of the present invention, which is used for the treatment of CDK4/6-mediated disorders or diseases.
本发明还提供了本发明所述的药物组合物或本发明所述的固体口服药物制剂在制备用于治疗CDK4/6介导的障碍或疾病的药物中的用途。The present invention also provides the use of the pharmaceutical composition of the present invention or the solid oral pharmaceutical preparation of the present invention in preparing a medicament for treating CDK4/6-mediated disorders or diseases.
本发明还提供了一种用于治疗CDK4/6介导的障碍或疾病的方法, 其包括向需要其的对象施用本发明所述的药物组合物或本发明所述的固体口服药物制剂。The present invention also provides a method for treating a CDK4/6-mediated disorder or disease, comprising administering the pharmaceutical composition of the present invention or the solid oral pharmaceutical formulation of the present invention to a subject in need thereof.
在一个实施方案中,所述CDK4/6介导的障碍或疾病是癌症、艾滋病、动脉粥样硬化或血管支架植入后再狭窄,其中所述癌症优选恶性实体瘤或恶性非实体瘤,更优选乳腺癌、肺癌、前列腺癌、白血病、脑癌或胃癌,再更优选胶质瘤。In one embodiment, the CDK4/6 mediated disorder or disease is cancer, AIDS, atherosclerosis or restenosis after stent implantation, wherein the cancer is preferably a malignant solid tumor or a malignant non-solid tumor, more Breast cancer, lung cancer, prostate cancer, leukemia, brain cancer or gastric cancer are preferred, and glioma is even more preferred.
本发明的有益效果:Beneficial effects of the present invention:
1)本发明能够显著提高所获得的药物组合物粉末的流动性,所获得的药物组合物具有较大的堆密度及较小的卡尔指数和休止角,且药物组合物粉末颜色均一、无色差。1) The present invention can significantly improve the fluidity of the obtained pharmaceutical composition powder, the obtained pharmaceutical composition has a larger bulk density, a smaller Carr index and an angle of repose, and the pharmaceutical composition powder has a uniform color and no color difference. .
2)本发明的药物组合物溶出迅速,并且具有良好的溶出一致性。2) The pharmaceutical composition of the present invention dissolves rapidly and has good dissolution consistency.
3)采用本发明的所述盐时,相对于使用化合物1的游离碱,将进一步提高本发明的药物组合物的溶解性,同时使得本发明的药物组合物具有降低的食物效应风险,且相对于使用化合物1的其它盐如盐酸盐和甲磺酸盐,还能使所获得的药物组合物具有改善的吸湿性。3) When the salt of the present invention is used, the solubility of the pharmaceutical composition of the present invention will be further improved relative to the use of the free base of compound 1, and the pharmaceutical composition of the present invention will have a reduced risk of food effects, and is relatively The use of other salts of Compound 1, such as hydrochloride and mesylate, also results in improved hygroscopicity of the obtained pharmaceutical composition.
4)本发明的药物组合物在高温(60℃)、高湿(75%RH)、强光(5000lux)、加速(40℃/75%RH)的实验条件下均具有较高的稳定性。4) The pharmaceutical composition of the present invention has high stability under the experimental conditions of high temperature (60°C), high humidity (75%RH), strong light (5000lux) and acceleration (40°C/75%RH).
5)本发明的药物组合物能够实现以粉末直接灌装胶囊工艺制备胶囊,工艺简单,灌装制备的胶囊的装量稳定性好,装量差异小。5) The pharmaceutical composition of the present invention can realize the preparation of capsules by directly filling the capsules with powder, the process is simple, the filling volume stability of the capsules prepared by filling is good, and the filling volume difference is small.
附图说明Description of drawings
图1为化合物1的富马酸盐在CD 3OD中的 1H-NMR图。 Figure 1 is a 1 H-NMR chart of the fumarate salt of compound 1 in CD 3 OD.
图2为化合物1的马来酸盐在CD 3Cl中的 1H-NMR图。 Figure 2 is a 1 H-NMR chart of the maleate salt of compound 1 in CD 3 Cl.
图3为化合物1的己二酸盐在CD 3OD的 1H-NMR图。 Figure 3 is a 1 H-NMR chart of the adipate salt of Compound 1 in CD 3 OD.
图4为化合物1的琥珀酸盐在CDCl 3中的 1H-NMR图。 Figure 4 is a 1 H-NMR chart of the succinate salt of compound 1 in CDCl 3 .
图5为化合物1的甲磺酸盐在CD 3OD中的 1H-NMR图。 Figure 5 is a 1 H-NMR chart of the mesylate salt of Compound 1 in CD 3 OD.
图6为试验例11的10mg规格中试批次在pH 1.2盐酸缓冲液中的溶出曲线。Figure 6 is the dissolution curve of the 10 mg specification pilot batch of Test Example 11 in pH 1.2 hydrochloric acid buffer.
图7为试验例11的50mg规格中试批次在pH 1.2盐酸缓冲液中的溶出曲线。Figure 7 is the dissolution curve of the 50 mg specification pilot batch of Test Example 11 in pH 1.2 hydrochloric acid buffer.
具体实施方式detailed description
以下通过具体实施方式对本发明作进一步的详细说明,但不应将此理解为本发明的范围仅限于以下的实施例。在不脱离本发明上述方案的思想的情况下,根据本领域普通技术知识和惯用手段做出的各种替换或变更,均应包含在本发明的范围内。The present invention will be further described in detail below through specific embodiments, but it should not be understood that the scope of the present invention is limited to the following examples. Without departing from the idea of the above solutions of the present invention, various substitutions or changes made according to common technical knowledge in the art and conventional means should all be included within the scope of the present invention.
缩略语或定义abbreviation or definition
甘露醇100SD:甘露醇
Figure PCTCN2021115486-appb-000002
100SD。
Mannitol 100SD: Mannitol
Figure PCTCN2021115486-appb-000002
100SD.
微晶纤维素PH101:微晶纤维素
Figure PCTCN2021115486-appb-000003
Microcrystalline Cellulose PH101: Microcrystalline Cellulose
Figure PCTCN2021115486-appb-000003
微晶纤维素PH102:微晶纤维素
Figure PCTCN2021115486-appb-000004
Microcrystalline Cellulose PH102: Microcrystalline Cellulose
Figure PCTCN2021115486-appb-000004
一水乳糖FlowLac 100:一水乳糖
Figure PCTCN2021115486-appb-000005
Lactose Monohydrate FlowLac 100: Lactose Monohydrate
Figure PCTCN2021115486-appb-000005
一水乳糖Tablettose 80:一水乳糖
Figure PCTCN2021115486-appb-000006
Lactose Monohydrate Tablettose 80: Lactose Monohydrate
Figure PCTCN2021115486-appb-000006
胶态二氧化硅200:胶态二氧化硅
Figure PCTCN2021115486-appb-000007
Colloidal Silica 200: Colloidal Silica
Figure PCTCN2021115486-appb-000007
本发明所述的“稀释剂”是指用于增加药物组合物的重量和/或体积,以便于成型和分剂量的赋形剂。本发明所述的稀释剂选自微晶纤维素、乳糖或其水合物、和糖醇中的一种或多种,优选选自微晶纤维素、一水乳糖、甘露醇和山梨醇中的一种或多种。所述稀释剂可以为单一稀释剂,也可为两种稀释剂的混合物。当所述稀释剂为两种稀释剂的混合物时,其优选为微晶纤维素(例如微晶纤维素PH101或微晶纤维素PH102)和一水乳糖(例如一水乳糖FlowLac 100或一水乳糖Tablettose 80)的混合物、或微晶纤维素(例如微晶纤维素PH101或微晶纤维素PH102)和甘露醇(例如甘露醇100SD)的混合物,且所述一水乳糖和微晶纤维素的重量比为5∶1-1∶5,优选为3∶1-1∶3,更优选为1.5∶1-1∶1.5,最优选为1∶1;或所述甘露醇和微晶纤维素的重量比为5∶1-1∶5,优选为3∶1-1∶3,更优选为1.5∶1-1∶1.5,最优选为1∶1。The term "diluent" as used herein refers to an excipient used to increase the weight and/or volume of a pharmaceutical composition to facilitate molding and sub-dosing. The diluent described in the present invention is selected from one or more of microcrystalline cellulose, lactose or its hydrate, and sugar alcohol, preferably selected from one or more of microcrystalline cellulose, lactose monohydrate, mannitol and sorbitol one or more. The diluent may be a single diluent or a mixture of two diluents. When the diluent is a mixture of two diluents, it is preferably microcrystalline cellulose (eg, microcrystalline cellulose PH101 or microcrystalline cellulose PH102) and lactose monohydrate (eg, lactose monohydrate FlowLac 100 or lactose monohydrate). Tablettose 80), or a mixture of microcrystalline cellulose (eg, microcrystalline cellulose PH101 or microcrystalline cellulose PH102) and mannitol (eg, mannitol 100SD), and the weight of the lactose monohydrate and microcrystalline cellulose The ratio is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5:1-1:1.5, most preferably 1:1; or the weight ratio of the mannitol and microcrystalline cellulose It is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5:1-1:1.5, most preferably 1:1.
本发明所述的“助流剂”是指用于增加物料的流动性的赋形剂。本发明所述的助流剂选自滑石粉和胶态二氧化硅中的一种或两种,更优选地,所述助流剂为胶态二氧化硅。The "glidant" in the present invention refers to an excipient used to increase the fluidity of a material. The glidant of the present invention is selected from one or both of talc and colloidal silicon dioxide, more preferably, the glidant is colloidal silicon dioxide.
本发明所述的“崩解剂”是指用于促进药物组合物在胃肠道中的崩解和增加活性成分的溶出的赋形剂。本发明所述的崩解剂选自交联羧甲基纤维素钠、羧甲基淀粉钠、交联聚乙烯吡咯烷酮和低取代羟丙基纤维素中的一种或多种,更优选地,所述崩解剂为交联羧甲基纤维素钠。The "disintegrant" in the present invention refers to an excipient used to promote the disintegration of the pharmaceutical composition in the gastrointestinal tract and increase the dissolution of the active ingredient. The disintegrant of the present invention is selected from one or more of croscarmellose sodium, sodium carboxymethyl starch, crospovidone and low-substituted hydroxypropyl cellulose, more preferably, The disintegrating agent is croscarmellose sodium.
本发明所述的“润滑剂”是指用于减少药物组合物的颗粒间的摩擦 力,改善力的传递和分布的赋形剂。本发明所述的润滑剂选自硬脂酸镁、硬脂酸、硬脂酰富马酸钠和硬脂酸锌中的一种或多种,更优选地,所述润滑剂为硬脂酸镁。The "lubricant" used in the present invention refers to an excipient used to reduce the frictional force between particles of a pharmaceutical composition and improve the transmission and distribution of force. The lubricant of the present invention is selected from one or more of magnesium stearate, stearic acid, sodium stearoyl fumarate and zinc stearate, more preferably, the lubricant is stearic acid magnesium.
本发明的药物组合物还可含有除稀释剂、助流剂、崩解剂和润滑剂以外的其它赋形剂。作为其具体实例,可以举出:粘合剂、矫味剂、分散剂、着色剂及香料等。The pharmaceutical compositions of the present invention may also contain other excipients other than diluents, glidants, disintegrants and lubricants. Specific examples thereof include binders, flavoring agents, dispersing agents, coloring agents, fragrances, and the like.
本发明所述的“固体口服药物制剂”是指用于口服施用的呈固态的药物制剂。本发明涉及由上述药物组合物制成的固体口服药物制剂,其具体剂型优选散剂、胶囊剂、片剂或颗粒剂,更优选胶囊剂,其中所述胶囊剂以所述药物组合物作为填充物。The "solid oral pharmaceutical preparation" in the present invention refers to a solid pharmaceutical preparation for oral administration. The present invention relates to a solid oral pharmaceutical preparation made from the above-mentioned pharmaceutical composition, and its specific dosage form is preferably powder, capsule, tablet or granule, more preferably capsule, wherein the capsule is filled with the pharmaceutical composition .
本发明的药物组合物或其固体口服药物制剂可含有0.1~1000mg,优选1~100mg,更优选8~50mg活性成分,以5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺游离碱的含量计。例如,本发明的药物组合物或其固体口服药物制剂中活性成分的含量可以为0.1mg、0.2mg、0.25mg、0.5mg、1mg、2mg、2.5mg、5mg、8mg、10mg、20mg、25mg、40mg、50mg、100mg、150mg、200mg、250mg、300mg、500mg、1000mg等。The pharmaceutical composition of the present invention or its solid oral pharmaceutical preparation may contain 0.1-1000 mg, preferably 1-100 mg, more preferably 8-50 mg of active ingredients in the form of 5-fluoro-4-(7'-fluoro-2'-methylspiro) [Cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine free base content meter. For example, the content of the active ingredient in the pharmaceutical composition of the present invention or its solid oral pharmaceutical preparation can be 0.1 mg, 0.2 mg, 0.25 mg, 0.5 mg, 1 mg, 2 mg, 2.5 mg, 5 mg, 8 mg, 10 mg, 20 mg, 25 mg, 40mg, 50mg, 100mg, 150mg, 200mg, 250mg, 300mg, 500mg, 1000mg, etc.
除非另外指出,否则所有的百分数都以药物组合物及其固体口服药物制剂的总重量的重量%给出。All percentages are given in weight % of the total weight of the pharmaceutical composition and its solid oral pharmaceutical formulation unless otherwise indicated.
在本发明中,给药对象可以是人或非人哺乳动物,更优选人。In the present invention, the administration subject may be a human or a non-human mammal, more preferably a human.
在本发明中,术语“含有”或类似术语“包含”和“包括”等一般表示开放性的含义,但是在其范围内也包括“由......组成”所限定的封闭性的情况。例如,“一种药物组合物,所述药物组合物含有......”也包括“一种药物组合物,其由......组成”的情况。In the present invention, the term "comprising" or similar terms "comprising" and "comprising" generally express an open meaning, but also includes a closed meaning defined by "consisting of" within its scope. condition. For example, "a pharmaceutical composition comprising..." also includes the case of "a pharmaceutical composition consisting of".
在本发明的范围中,任一特征的各种选项可以与其它特征的各种选项相互组合,从而构成许多不同的实施方案。本发明意在包括由所有技术特征的各种选项所组成的所有可能的实施方案。Within the scope of the present invention, various options of any one feature can be combined with each other with various options of other features to form many different embodiments. The present invention is intended to include all possible embodiments consisting of various options of all technical features.
实施例1Example 1
化合物1的不同盐型的制备方法如下:The preparation methods of different salt forms of compound 1 are as follows:
(1)化合物1富马酸盐的制备(1) Preparation of Compound 1 Fumarate
氮气保护下,室温,向反应釜中加入二氯甲烷(22.0体积)和乙醇 (22.0体积),边搅拌边加入5-氟-4-(7′-氟-2′-甲基螺[环戊烷-1,3′-吲哚]-5′-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺(2.070kg),升温至30-40摄氏度,搅拌直至全部溶解,降温至室温,将溶液转移至溶剂桶中备用。在氮气保护下,将上述溶液通过微孔过滤器过滤,并转移至反应釜中,搅拌,常压蒸出二氯甲烷和乙醇,然后保持反应釜温度至80±5摄氏度,将富马酸(1.0eq)的乙醇溶液(12体积)通过微孔过滤器缓慢滴入反应釜中,保温搅拌过夜。降温至20-30摄氏度,继续搅拌至少1个小时,离心,收集滤饼。将滤饼置于真空干燥箱中,干燥过夜,得到1.605kg 5-氟-4-(7′-氟-2′-甲基螺[环戊烷-1,3′-吲哚]-5′-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺富马酸盐(化合物A),收率63.6%。其 1H-NMR数据如图1所示。 Under nitrogen protection, at room temperature, dichloromethane (22.0 vol) and ethanol (22.0 vol) were added to the reaction kettle, and 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane] was added while stirring. Alkyl-1,3'-indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine (2.070kg), Heat up to 30-40 degrees Celsius, stir until all dissolved, cool to room temperature, and transfer the solution to a solvent bucket for later use. Under nitrogen protection, the above solution was filtered through a microporous filter, and transferred to the reactor, stirred, and dichloromethane and ethanol were distilled off at normal pressure, and then the temperature of the reactor was kept to 80 ± 5 degrees Celsius, and the fumaric acid ( 1.0eq) of ethanol solution (12 volumes) was slowly dropped into the reaction kettle through a microporous filter, and kept stirring overnight. Cool to 20-30 degrees Celsius, continue stirring for at least 1 hour, centrifuge, and collect the filter cake. The filter cake was placed in a vacuum oven and dried overnight to obtain 1.605 kg of 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5' -yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine fumarate (compound A), yield 63.6%. Its 1 H-NMR data are shown in FIG. 1 .
(2)化合物1马来酸盐的制备(2) Preparation of compound 1 maleate
称取353.5毫克5-氟-4-(7′-氟-2′-甲基螺[环戊烷-1,3′-吲哚]-5′-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺至20毫升玻璃瓶中,加入5毫升异丙醇(IPA)/H 2O(19∶1,v/v),将含有83.7毫克马来酸的IPA/H 2O(19∶1,v/v;3.75毫升)溶液滴入玻璃瓶中,室温搅拌4天,离心分离固体,50摄氏度干燥5小时,得到405.1毫克马来酸盐,收率92.7%。其 1H-NMR数据如图2所示。 Weigh out 353.5 mg of 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1- Methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine to a 20 mL glass bottle, add 5 mL isopropanol (IPA)/ H2O (19:1, v/v), A solution of IPA/H 2 O (19:1, v/v; 3.75 ml) containing 83.7 mg of maleic acid was dropped into a glass bottle, stirred at room temperature for 4 days, centrifuged to separate the solid, and dried at 50 degrees Celsius for 5 hours to obtain 405.1 mg Maleate, yield 92.7%. Its 1 H-NMR data are shown in FIG. 2 .
(3)化合物1己二酸盐的制备(3) Preparation of Compound 1 Adipate
称取350.6毫克5-氟-4-(7′-氟-2′-甲基螺[环戊烷-1,3′-吲哚]-5′-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺至20毫升玻璃瓶中,加入5毫升四氢呋喃(THF),将含有105.4毫克己二酸的THF溶液(3.75毫升)滴入玻璃瓶中,室温搅拌4天,离心分离固体,50摄氏度干燥5小时,得到362.4毫克己二酸盐,收率79.5%。其 1H-NMR数据如图3所示。 Weigh out 350.6 mg of 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1- Methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine to a 20 mL glass bottle, add 5 mL of tetrahydrofuran (THF), drop a solution of 105.4 mg of adipic acid in THF (3.75 mL) put into a glass bottle, stirred at room temperature for 4 days, centrifuged to separate the solid, and dried at 50 degrees Celsius for 5 hours to obtain 362.4 mg of adipate with a yield of 79.5%. Its 1 H-NMR data are shown in FIG. 3 .
(4)化合物1琥珀酸盐的制备(4) Preparation of Compound 1 Succinate
将351.6毫克5-氟-4-(7′-氟-2′-甲基螺[环戊烷-1,3′-吲哚]-5′-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺和5毫升丙酮加入到反应瓶中,磁力搅拌,然后将含有85.1毫克琥珀酸的3.75毫升丙酮溶液滴加入反应瓶中,室温搅拌4天,离心分离固体,50摄氏度干燥5小时,得到377.7毫克固体,即琥珀酸盐,收率86.5%。其 1H-NMR数据如图4所示。 351.6 mg of 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1-methyl) Piperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine and 5 ml of acetone were added to the reaction flask, stirred magnetically, and then 3.75 ml of acetone solution containing 85.1 mg of succinic acid was added dropwise to the reaction flask, It was stirred at room temperature for 4 days, the solid was separated by centrifugation, and dried at 50 degrees Celsius for 5 hours to obtain 377.7 mg of solid, namely succinate, with a yield of 86.5%. Its 1 H-NMR data are shown in FIG. 4 .
(5)化合物1甲磺酸盐的制备(5) Preparation of compound 1 mesylate
将61.1毫克甲磺酸和305.3毫克5-氟-4-(7′-氟-2′-甲基螺[环戊烷-1,3′-吲哚]-5′-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺加入到反应瓶中,加入10毫升乙酸乙酯,室温搅拌3天,离心分离固体,50摄氏度干燥3小时,得到370.2毫克固体,即甲磺酸盐,产率101.0%。其 1H-NMR数据如图5所示。 61.1 mg of methanesulfonic acid and 305.3 mg of 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-( 5-(1-Methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine was added to the reaction flask, 10 mL of ethyl acetate was added, stirred at room temperature for 3 days, centrifuged to separate the solid, and dried at 50 degrees Celsius After 3 hours, 370.2 mg of solid, the mesylate salt, was obtained in 101.0% yield. Its 1 H-NMR data are shown in FIG. 5 .
(6)化合物1盐酸盐的制备(6) Preparation of Compound 1 Hydrochloride
称取352.2毫克5-氟-4-(7′-氟-2′-甲基螺[环戊烷-1,3′-吲哚]-5′-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺至20毫升玻璃瓶中,并加入5毫升THF,得到化合物1游离碱的悬浊液。称取60微升37%盐酸并用3.75毫升THF稀释,然后将得到的酸溶液逐步加入游离碱悬浊液中,于室温搅拌4天。离心分离固体,50℃干燥5小时,收集361.4毫克固体,即得到盐酸盐,产率83.3%。Weigh out 352.2 mg of 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1- Methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine was added to a 20 mL glass vial and 5 mL of THF was added to give a suspension of compound 1 free base. 60 μl of 37% hydrochloric acid was weighed and diluted with 3.75 ml of THF, and the resulting acid solution was gradually added to the free base suspension and stirred at room temperature for 4 days. The solid was separated by centrifugation, dried at 50° C. for 5 hours, and 361.4 mg of the solid was collected to obtain the hydrochloride salt with a yield of 83.3%.
试验例1 溶解度试验Test Example 1 Solubility Test
本试验例的目的在于对比本发明得到的各类盐(也可以称为盐型)以及游离碱的溶解度。The purpose of this test example is to compare the solubility of various salts (also called salt forms) and free bases obtained by the present invention.
试验方法:溶解度测试在水、模拟禁食肠液(FaSSIF)和模拟喂食肠液(FeSSIF)三种溶媒中进行,用以评估本发明的盐的溶解度及食物效应风险。Test Methods: Solubility tests were performed in three vehicles, water, simulated fasting intestinal fluid (FaSSIF) and simulated fed intestinal fluid (FeSSIF), to assess the solubility and food effect risk of the salts of the present invention.
试验中,分别将样品与溶媒在离心管中混合(起始投料量为10毫克/毫升),再将离心管密封固定在转速为25转/分钟的旋转盘上,在37℃条件下旋转混合24小时后取样。对浑浊样品离心分离,取过滤后的上清液测定HPLC浓度。若样品溶清,则测试所得溶液浓度。In the test, the sample and the solvent were mixed in a centrifuge tube (the initial feeding amount was 10 mg/ml), and then the centrifuge tube was sealed and fixed on a rotating disk with a rotation speed of 25 rpm, and rotated and mixed at 37 °C. Samples were taken after 24 hours. The turbid samples were centrifuged, and the filtered supernatant was used to measure the HPLC concentration. If the sample dissolves, test the concentration of the resulting solution.
试验对象:化合物1的游离碱、马来酸盐、富马酸盐、己二酸盐和琥珀酸盐。Test subjects: free base, maleate, fumarate, adipate and succinate of compound 1.
试验结果:见表1-3,不同盐型和游离碱在水中、模拟禁食肠液(FaSSIF)和模拟喂食肠液(FeSSIF)中的溶解度。Test results: See Tables 1-3, solubility of different salt forms and free bases in water, simulated fasting intestinal fluid (FaSSIF) and simulated fed intestinal fluid (FeSSIF).
表1 化合物1的不同盐型和游离碱在水中的溶解度Table 1 Solubility of different salt forms and free bases of compound 1 in water
样品名称sample name 溶解度(mg/ml)Solubility (mg/ml)
化合物1的游离碱Free base of compound 1 0.0750.075
化合物1的马来酸盐Maleate of Compound 1 12.912.9
化合物1的富马酸盐Fumarate of compound 1 6.36.3
化合物1的己二酸盐Adipate salt of compound 1 6.56.5
化合物1的琥珀酸盐Succinate of Compound 1 7.57.5
如表1所示,化合物1的马来酸盐、富马酸盐、己二酸盐和琥珀酸盐在水中的溶解度分别为12.9、6.3、6.5和7.5毫克/毫升,显著高于游离碱在水中的溶解度(0.075毫克/毫升)。这提示在采用本发明的上述盐配制本发明的药物组合物时,会显著改善活性成分化合物1的溶解性能。As shown in Table 1, the solubility of maleate, fumarate, adipate and succinate of compound 1 in water was 12.9, 6.3, 6.5 and 7.5 mg/ml, respectively, which was significantly higher than that of the free base in water. Solubility in water (0.075 mg/ml). This suggests that when the above-mentioned salt of the present invention is used to formulate the pharmaceutical composition of the present invention, the solubility of the active ingredient Compound 1 can be significantly improved.
表2 化合物1的不同盐型和游离碱在模拟禁食肠液(FaSSIF)中的溶解度Table 2 Solubility of different salt forms and free bases of compound 1 in simulated fasting intestinal fluid (FaSSIF)
样品名称sample name 溶解度(mg/ml)Solubility (mg/ml)
化合物1的游离碱Free base of compound 1 0.190.19
化合物1的马来酸盐Maleate of Compound 1 7.17.1
化合物1的富马酸盐Fumarate of compound 1 7.67.6
化合物1的己二酸盐Adipate salt of compound 1 6.86.8
化合物1的琥珀酸盐Succinate of Compound 1 6.46.4
如表2所示,化合物1的马来酸盐、富马酸盐、己二酸盐和琥珀酸盐在模拟禁食肠液(FaSSIF)中的溶解度分别显著高于游离碱在模拟禁食肠液(FaSSIF)中的溶解度(0.19毫克/毫升)。As shown in Table 2, the solubility of maleate, fumarate, adipate and succinate of compound 1 in simulated fasting intestinal fluid (FaSSIF) was significantly higher than that of free base in simulated fasting intestinal fluid (FaSSIF), respectively ( FaSSIF) (0.19 mg/ml).
表3 化合物1的不同盐型和游离碱在模拟喂食肠液(FeSSIF)中的溶解度Table 3 Solubility of different salt forms and free bases of compound 1 in simulated fed intestinal fluid (FeSSIF)
样品名称sample name 溶解度(mg/ml)Solubility (mg/ml)
化合物1的游离碱Free base of compound 1 10.210.2
化合物1的马来酸盐Maleate of Compound 1 7.87.8
化合物1的富马酸盐Fumarate of compound 1 8.08.0
化合物1的己二酸盐Adipate salt of compound 1 11.611.6
化合物1的琥珀酸盐Succinate of Compound 1 10.710.7
如表3所示,化合物1的马来酸盐、富马酸盐、己二酸盐和琥珀酸盐在模拟喂食肠液(FeSSIF)中的溶解度与化合物1的游离碱在模拟喂食 肠液(FeSSIF)中的溶解度(10.2毫克/毫升)相当,但相对于游离碱,这些盐在模拟禁食肠液(FaSSIF)和模拟喂食肠液(FeSSIF)中的溶解度接近,这提示本发明的上述盐具有较低的食物效应风险。As shown in Table 3, the solubility of maleate, fumarate, adipate and succinate of Compound 1 in simulated fed intestinal fluid (FeSSIF) was comparable to the free base of Compound 1 in simulated fed intestinal fluid (FeSSIF) The solubility of these salts in simulated fasting intestinal fluid (FaSSIF) and simulated fed intestinal fluid (FeSSIF) is similar relative to the free base, suggesting that the above salts of the present invention have lower food effect risk.
试验例2 吸湿性试验Test Example 2 Hygroscopicity Test
本试验例对比了本发明所提供的化合物1的各种盐型与游离碱的吸湿性。This test example compares the hygroscopicity of various salt forms and free bases of compound 1 provided by the present invention.
25℃条件下,通过动态水分吸附(DVS)试验,测试本发明的化合物1的盐及游离碱在湿度达到80%RH时的水分吸附量,以进行吸湿性比对。Under the condition of 25°C, the dynamic moisture adsorption (DVS) test was used to test the moisture adsorption capacity of the salt and free base of compound 1 of the present invention when the humidity reached 80% RH, so as to compare the hygroscopicity.
试验对象:化合物1的游离碱、马来酸盐、富马酸盐、己二酸盐、琥珀酸盐、甲磺酸盐和盐酸盐。Test subjects: Compound 1 free base, maleate, fumarate, adipate, succinate, mesylate and hydrochloride.
实验结果:测试结果如表4所示。其中,吸湿性(以试验对象在湿度达到80%RH时的水分吸附量计)>15%为非常吸湿,2~15%为吸湿,0.2~2%为略有吸湿性,<0.2%为几乎无吸湿性。Experimental results: The test results are shown in Table 4. Among them, hygroscopicity (measured by the amount of moisture absorbed by the test object when the humidity reaches 80%RH)>15% is very hygroscopic, 2-15% is hygroscopic, 0.2-2% is slightly hygroscopic, <0.2% is almost hygroscopic Not hygroscopic.
表4 化合物1的不同盐型和游离碱的吸湿性Table 4 Hygroscopicity of different salt forms and free bases of compound 1
样品名称sample name 吸湿性(%)Hygroscopicity (%)
化合物1的游离碱Free base of compound 1 0.100.10
化合物1的马来酸盐Maleate of Compound 1 2.002.00
化合物1的富马酸盐Fumarate of compound 1 0.350.35
化合物1的己二酸盐Adipate salt of compound 1 0.220.22
化合物1的琥珀酸盐Succinate of Compound 1 0.330.33
化合物1的甲磺酸盐Mesylate of Compound 1 12.0212.02
化合物1的盐酸盐 Compound 1 hydrochloride 9.129.12
结果显示:化合物1的马来酸盐、富马酸盐、己二酸盐和琥珀酸盐在25℃/80%RH条件下水分吸附量为0.10~2.00%,仅具有轻微吸湿性,几乎与游离碱的吸湿性相当;而盐酸盐和甲磺酸盐在25℃/80%RH条件下水分吸附量分别为9.12%和12.02%,具有较高的吸湿性。这提示,采用化合物1的马来酸盐、富马酸盐、己二酸盐和琥珀酸盐作为药物组合物的活性成分时,相对于化合物1的甲磺酸盐和盐酸盐会显著改善药物组合物的吸湿性。The results showed that the water adsorption capacity of maleate, fumarate, adipate and succinate of compound 1 was 0.10-2.00% under the condition of 25℃/80%RH. The hygroscopicity of the free base is comparable; while the hydrochloric acid salt and the mesylate salt have higher hygroscopicity at 25℃/80%RH, the water adsorption capacity is 9.12% and 12.02%, respectively. This suggests that when the maleate, fumarate, adipate and succinate of Compound 1 are used as active ingredients in the pharmaceutical composition, significant improvements will be seen relative to the methanesulfonate and hydrochloride of Compound 1 Hygroscopicity of pharmaceutical compositions.
实施例2-3Example 2-3
实施例2-3的药物组合物粉末的制备方法如下:The preparation method of the pharmaceutical composition powder of embodiment 2-3 is as follows:
(1)按照表5中的配方用量,分别称取化合物A、甘露醇100SD、微晶纤维素PH101、胶态二氧化硅200和硬脂酸镁;(1) according to the formula consumption in table 5, take by weighing compound A, mannitol 100SD, microcrystalline cellulose PH101, colloidal silicon dioxide 200 and magnesium stearate respectively;
(2)将化合物A与胶态二氧化硅200混合均匀过筛,得预处理粉;(2) compound A and colloidal silica 200 are mixed and sieved to obtain pretreated powder;
(3)将甘露醇100SD、微晶纤维素PH101、硬脂酸镁分别过筛,备用;(3) sieve mannitol 100SD, microcrystalline cellulose PH101, magnesium stearate respectively, for subsequent use;
(4)将步骤(2)所得预处理粉与甘露醇100SD、微晶纤维素PH101混合均匀,得主混粉备用;(4) the pretreatment powder obtained in step (2) is mixed with mannitol 100SD, microcrystalline cellulose PH101, and the winner is mixed powder for subsequent use;
(5)将硬脂酸镁加入到步骤(4)制得的主混粉中混合,得到药物组合物粉末总混粉。(5) adding magnesium stearate to the main mixed powder prepared in step (4) and mixing to obtain a total mixed powder of pharmaceutical composition powder.
表5table 5
Figure PCTCN2021115486-appb-000008
Figure PCTCN2021115486-appb-000008
*注:化合物A为5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺的富马酸盐。*Note: Compound A is 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-( The fumarate salt of 1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine.
试验例3 流动性试验Test Example 3 Fluidity Test
采用智能粉体特性测试仪测定活性成分原料化合物A及实施例2-3的药物组合物的休止角(符合国家标准GB11986-89关于《表面活性剂粉体和颗粒休止角的测量》的规定)、堆密度、振实密度和卡尔指数。其中,卡尔指数=(振实密度-堆密度)/振实密度*100%,结果如表6所示。The angle of repose of the active ingredient raw material Compound A and the pharmaceutical compositions of Examples 2-3 was measured by an intelligent powder characteristic tester (in compliance with the national standard GB11986-89 on "Measurement of the angle of repose of surfactant powders and particles") , bulk density, tap density, and Carr's index. Wherein, Carr's index=(tap density-bulk density)/tap density*100%, and the results are shown in Table 6.
表6Table 6
Figure PCTCN2021115486-appb-000009
Figure PCTCN2021115486-appb-000009
由表6的数据可知,实施例2-3的药物组合物相对于单一的活性成分原料化合物A,堆密度有显著增加,休止角和卡尔指数明显减小,表明实施例2-3的药物组合物粉末相对于单一的活性成分原料化合物A,流动性显著提高,能够实现以简单的粉末直接灌装工艺制备胶囊,降低灌装胶囊时装量差异较大的风险,满足胶囊灌装的要求及工业化生产的要求。As can be seen from the data in Table 6, the bulk density of the pharmaceutical composition of Example 2-3 is significantly increased compared to the single active ingredient raw material Compound A, and the angle of repose and Carr's index are significantly reduced, indicating that the pharmaceutical composition of Example 2-3 is combined. Compared with the single active ingredient raw material Compound A, the fluidity of the powder is significantly improved, and it can realize the preparation of capsules by a simple powder direct filling process, reduce the risk of large differences in the filling volume of the capsules, and meet the requirements of capsule filling and industrialization. production requirements.
实施例4-6Examples 4-6
按照表7中的配方用量,采用与实施例2-3类似的步骤,制备得到实施例4-6的药物组合物粉末。According to the formulation dosage in Table 7, the pharmaceutical composition powders of Examples 4-6 were prepared by adopting the steps similar to those in Example 2-3.
表7Table 7
Figure PCTCN2021115486-appb-000010
Figure PCTCN2021115486-appb-000010
试验例4 流动性试验Test Example 4 Fluidity Test
按照试验例3的方法测定实施例4-6所获得的药物组合物粉末的休止角、堆密度、振实密度和卡尔指数,结果如下表8所示。According to the method of Test Example 3, the angle of repose, bulk density, tap density and Carr's index of the pharmaceutical composition powder obtained in Examples 4-6 were measured, and the results are shown in Table 8 below.
表8Table 8
Figure PCTCN2021115486-appb-000011
Figure PCTCN2021115486-appb-000011
由表8的数据可知,实施例4-6的药物组合物粉末相对于单一的活性成分原料化合物A,堆密度有显著增加,休止角和卡尔指数明显减小,表明实施例4-6的药物组合物粉末相对于单一的活性成分原料化合物A,流动性显著提高,在后续用于制备胶囊形式的制剂时,将能够降低灌装胶囊时装量差异较大的风险,满足胶囊灌装的要求及工业化生产的要求。As can be seen from the data in Table 8, the bulk density of the pharmaceutical composition powder of Example 4-6 is significantly increased compared to the single active ingredient raw material Compound A, and the angle of repose and Carr's index are significantly reduced, indicating that the medicine of Example 4-6 Compared with the single active ingredient compound A, the fluidity of the composition powder is significantly improved, and when it is subsequently used to prepare a preparation in the form of capsules, it will be able to reduce the risk of large differences in the filling volume of the capsules, and meet the requirements and requirements of capsule filling. requirements for industrial production.
实施例7-9Examples 7-9
实施例7-9的胶囊的制备方法如下:The preparation method of the capsule of embodiment 7-9 is as follows:
(1)按照表9中的用量,分别称取化合物A、微晶纤维素PH102、一水乳糖FLOWLAC100、交联羧甲基纤维素钠、胶态二氧化硅200和硬脂酸镁;(1) according to the consumption in table 9, take by weighing compound A, microcrystalline cellulose PH102, lactose monohydrate FLOWLAC100, croscarmellose sodium, colloidal silicon dioxide 200 and magnesium stearate respectively;
(2)将化合物A与胶态二氧化硅200混合均匀过筛,得预处理粉;(2) compound A and colloidal silica 200 are mixed and sieved to obtain pretreated powder;
(3)将微晶纤维素PH102、一水乳糖FLOWLAC100、交联羧甲基纤维素钠、硬脂酸镁分别过筛,备用;(3) sieving microcrystalline cellulose PH102, lactose monohydrate FLOWLAC100, croscarmellose sodium, magnesium stearate, respectively, for subsequent use;
(4)将步骤(2)所得预处理粉与微晶纤维素PH102、一水乳糖FLOWLAC100、交联羧甲基纤维素钠混合均匀,得主混粉备用;(4) mixing the pretreated powder obtained in step (2) with microcrystalline cellulose PH102, lactose monohydrate FLOWLAC100 and croscarmellose sodium, and the winner is mixed powder for subsequent use;
(5)将硬脂酸镁加入到步骤(4)制得的主混粉中混合,得到药物组合物粉末总混粉。(5) adding magnesium stearate to the main mixed powder prepared in step (4) and mixing to obtain a total mixed powder of pharmaceutical composition powder.
(6)使用胶囊填充机将药物组合物粉末总混粉填充到空胶囊中。(6) Filling the pharmaceutical composition powder blend powder into empty capsules using a capsule filling machine.
表9Table 9
Figure PCTCN2021115486-appb-000012
Figure PCTCN2021115486-appb-000012
*注:50mg规格胶囊,是以5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺的游离碱的含量计。化合物A为5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺的富马酸盐,分子量为604.66,其游离碱分子量为488.59,换算系数为1.24。*Note: 50mg capsules are based on 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5'-yl)-N-( The content of the free base of 5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine. Compound A is 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1-methyl) The fumarate salt of ylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine, the molecular weight is 604.66, the molecular weight of its free base is 488.59, and the conversion factor is 1.24.
试验例5 溶出试验Test Example 5 Dissolution Test
根据《中国药典》2015年版四部0931溶出度测定法第一法(篮法),对制备得到的实施例7-9的胶囊的溶出度进行测定。采用900毫升的PH1.2盐酸缓冲液为介质,在水温为37±0.5℃,以50rpm的转速,分别在5min、15min、30min、45min、60min、75min等6个时间点取样。每个取样体积为10毫升,并用PFS过滤器(0.45μm~10μm)进行过滤处理。每次溶出测试,重复3粒胶囊或6粒胶囊。溶出结果如下表10所示。According to the first method (basket method) of "Chinese Pharmacopoeia" 2015 Edition Part Four 0931 Dissolution Determination Method, the dissolution rate of the prepared capsules of Examples 7-9 was determined. Using 900 ml of PH1.2 hydrochloric acid buffer as the medium, the water temperature was 37±0.5 °C, and the rotation speed was 50 rpm, and samples were taken at 6 time points such as 5 min, 15 min, 30 min, 45 min, 60 min, and 75 min, respectively. Each sampling volume was 10 ml and was filtered with a PFS filter (0.45 μm to 10 μm). For each dissolution test, repeat 3 capsules or 6 capsules. The dissolution results are shown in Table 10 below.
表10Table 10
Figure PCTCN2021115486-appb-000013
Figure PCTCN2021115486-appb-000013
*注:RSD表示“相对标准偏差”。*Note: RSD stands for "relative standard deviation".
由表10的数据可知,本发明实施例7-9得到的胶囊,在pH1.2的溶出介质中,化合物A均具有较快的溶出速度及良好的溶出一致性,溶出行为没有显著的差异。From the data in Table 10, it can be seen that the capsules obtained in Examples 7-9 of the present invention, in the dissolution medium of pH 1.2, Compound A has a faster dissolution rate and good dissolution consistency, and there is no significant difference in dissolution behavior.
实施例10-13Examples 10-13
按照表11中的配方,采用与实施例2-3类似的步骤,得到实施例10-13的药物组合物粉末。According to the formulations in Table 11, the pharmaceutical composition powders of Examples 10-13 were obtained by the steps similar to those of Examples 2-3.
表11Table 11
Figure PCTCN2021115486-appb-000014
Figure PCTCN2021115486-appb-000014
试验例6 流动性试验Test Example 6 Fluidity Test
按照试验例3的方法测定实施例10-13所获得的药物组合物粉末的休止角,试验结果如表12所示。According to the method of Test Example 3, the angles of repose of the pharmaceutical composition powders obtained in Examples 10-13 were measured, and the test results are shown in Table 12.
表12Table 12
配方formula 休止角(°)Angle of repose (°)
实施例10Example 10 41.9741.97
实施例11Example 11 40.4140.41
实施例12Example 12 40.0540.05
实施例13Example 13 40.8640.86
由表12的数据可知,相比于不加入胶态二氧化硅的实施例10的药物组合物,配方中加入胶态二氧化硅的实施例11-13的药物组合物,休止角进一步减小,流动性有进一步的改善,且胶态二氧化硅的用量在约1%~3%的范围内时,所述药物组合物粉末的流动性无显著变化。As can be seen from the data in Table 12, compared to the pharmaceutical composition of Example 10 without colloidal silicon dioxide, the pharmaceutical compositions of Examples 11-13 with colloidal silicon dioxide added to the formula, the angle of repose is further reduced. , the fluidity is further improved, and when the amount of colloidal silicon dioxide is in the range of about 1% to 3%, the fluidity of the pharmaceutical composition powder has no significant change.
实施例14-15Examples 14-15
按照与实施例7-9类似的步骤,得到配方如表13所示的实施例14-15的胶囊。Following the steps similar to those of Examples 7-9, capsules of Examples 14-15 whose formulations are shown in Table 13 were obtained.
表13Table 13
Figure PCTCN2021115486-appb-000015
Figure PCTCN2021115486-appb-000015
试验例7 溶出试验Test Example 7 Dissolution Test
根据试验例5的方法测定实施例14-15所获得的胶囊的溶出度,试验结果如下表14所示。The dissolution rate of the capsules obtained in Examples 14-15 was measured according to the method of Test Example 5, and the test results are shown in Table 14 below.
表14Table 14
Figure PCTCN2021115486-appb-000016
Figure PCTCN2021115486-appb-000016
由表14的数据可知,本发明实施例14-15得到的胶囊,在pH1.2的溶出介质中,化合物A均具有较快的溶出速度及良好的溶出一致性。It can be seen from the data in Table 14 that the capsules obtained in Examples 14-15 of the present invention, in the dissolution medium of pH 1.2, Compound A has a relatively fast dissolution rate and good dissolution consistency.
实施例1610mg规格胶囊Example 1610mg specification capsule
按照与实施例7-9类似的步骤,得到配方如表15所示的10mg规格胶囊。Following the steps similar to those in Examples 7-9, 10 mg capsules with the formula shown in Table 15 were obtained.
表15Table 15
Figure PCTCN2021115486-appb-000017
Figure PCTCN2021115486-appb-000017
*注:10mg规格胶囊,是以5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺的游离碱的含量计。化合物A为5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺的富马酸盐,分子量为604.66,其游离碱分子量为488.59,换算系数为1.24。*Note: 10mg capsules are based on 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5'-yl)-N-( The content of the free base of 5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine. Compound A is 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1-methyl) The fumarate salt of ylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine, the molecular weight is 604.66, the molecular weight of its free base is 488.59, and the conversion factor is 1.24.
试验例8 溶出试验Test Example 8 Dissolution Test
根据试验例5的方法测定实施例16所获得胶囊的溶出度,试验结果如表16所示。The dissolution rate of the capsules obtained in Example 16 was measured according to the method of Test Example 5, and the test results are shown in Table 16.
表16 10mg规格溶出结果Table 16 Dissolution results of 10mg specification
Figure PCTCN2021115486-appb-000018
Figure PCTCN2021115486-appb-000018
由以上结果可知,本发明10mg规格胶囊具有很好的溶出行为。It can be seen from the above results that the 10 mg capsules of the present invention have good dissolution behavior.
试验例9 胶囊重量检测结果Test Example 9 Capsule weight test results
在实施例7(50mg规格)和实施例16(10mg规格)的胶囊填充的前、中、后三个阶段,分别取样10粒胶囊,并检测单个胶囊重量(前、中、后三个阶段分别指胶囊填充开始后5分钟、填充完成一半左右、填充结束前5分钟),依据《中国药典》2015版四部0103胶囊剂项下装量差异检查法检测,试验结果如表17、18所示。In the three stages before, during and after the capsule filling of Example 7 (50mg specification) and Example 16 (10mg specification), 10 capsules were sampled respectively, and the weight of a single capsule was detected (the first, middle and last three stages respectively) Refers to 5 minutes after the start of capsule filling, about half of the filling is completed, and 5 minutes before the end of filling), according to the "Chinese Pharmacopoeia" 2015 edition four 0103 capsules under the difference inspection method detection, the test results are shown in Tables 17 and 18.
表17 50mg规格胶囊重量检测结果
Figure PCTCN2021115486-appb-000019
Table 17 Weight test results of 50mg capsules
Figure PCTCN2021115486-appb-000019
表18 10mg规格胶囊重量检测结果Table 18 10mg specification capsule weight test results
Figure PCTCN2021115486-appb-000020
Figure PCTCN2021115486-appb-000020
由表17和18的实验结果可知,利用本发明的药物组合物配方使得胶囊填充工艺稳定,得到的胶囊装量差异较小,装量差异均显著小于装量差异限度(±7.5%),在胶囊填充过程中不会对产品含量及含量均匀度产生显著影响。From the experimental results of Tables 17 and 18, it can be known that the capsule filling process is stabilized by using the pharmaceutical composition formula of the present invention, and the difference of the obtained capsule filling is less, and the difference in the filling is all significantly less than the limit of the difference in the filling (±7.5%), and in Product content and content uniformity are not significantly affected during capsule filling.
试验例10 本发明的药物组合物的稳定性Test Example 10 Stability of the pharmaceutical composition of the present invention
为考察本发明得到的药物组合物的稳定性,使用实施例7(50mg规格)和实施例16(10mg规格)的胶囊进行稳定性实验。两个规格样品放置条件相同,分别在40℃/75%RH(开口)的加速条件下放置10天、30天,采用HPLC法测定胶囊中化合物A的游离碱含量变化,采用试验例5的方法测定溶出度;分别在60℃(开口)、25℃/75%RH(开口)、光照(强度5000lux)(开口)、40℃/75%RH(开口)、40℃/75%RH(闭口)条件下放置10天、30天,然后采用HPLC法测定有关物质变化。胶囊含量变化、溶出结果和有关物质变化的检测结果如下:In order to investigate the stability of the pharmaceutical composition obtained by the present invention, the capsules of Example 7 (50 mg strength) and Example 16 (10 mg strength) were used for stability experiments. The two specifications of the samples were placed under the same conditions for 10 days and 30 days under accelerated conditions of 40°C/75%RH (opening), respectively. The free base content of compound A in the capsules was determined by HPLC, and the method of Test Example 5 was used. Determination of dissolution; respectively at 60°C (open), 25°C/75%RH (open), light (intensity 5000lux) (open), 40°C/75%RH (open), 40°C/75%RH (closed) After 10 days and 30 days under the conditions, the changes of related substances were determined by HPLC. The test results of changes in capsule content, dissolution results and related substances are as follows:
表19 胶囊含量变化结果Table 19 Results of changes in capsule content
Figure PCTCN2021115486-appb-000021
Figure PCTCN2021115486-appb-000021
*注:胶囊含量表示“胶囊中化合物A的游离碱含量”。*Note: Capsule content means "free base content of compound A in capsules".
表20 胶囊溶出度结果Table 20 Capsule dissolution results
Figure PCTCN2021115486-appb-000022
Figure PCTCN2021115486-appb-000022
由以上数据可知,2个规格的胶囊在40℃/75%RH(开口)的加速条件下放置30天后活性物质含量稳定,且加速条件(40℃/75%RH)对2个规格胶囊的溶出均无影响。It can be seen from the above data that the active substance content of the capsules of 2 specifications is stable after being placed under the accelerated conditions of 40°C/75%RH (opening) for 30 days, and the accelerated conditions (40°C/75%RH) are effective for the dissolution of the capsules of 2 specifications. No effect.
Figure PCTCN2021115486-appb-000023
Figure PCTCN2021115486-appb-000023
Figure PCTCN2021115486-appb-000024
Figure PCTCN2021115486-appb-000024
试验例11 本发明的中试放大样品的溶出试验和稳定性试验研究Test Example 11 Research on the dissolution test and stability test of the pilot scale-up sample of the present invention
采用实施例7(50mg规格)和实施例16(10mg规格)的配方制备胶囊,各制备10000粒,并分别分三个批次进行。包装采用口服固体药用高密度聚乙烯瓶和口服固体药用儿童安全盖。Capsules were prepared using the formulations of Example 7 (50 mg strength) and Example 16 (10 mg strength), and 10,000 capsules were prepared each, and were divided into three batches. The packaging adopts oral solid medicinal high density polyethylene bottle and oral solid medicinal child safety cap.
1、溶出试验1. Dissolution test
对每个批次随机选取6粒胶囊,采用试验例5的方法测定溶出度,溶出结果曲线如图6-7所示,结果表明:两种规格的药物组合物在pH1.2盐酸缓冲液中,15分钟溶出大于95%,30分钟内均能达到溶出完全。6 capsules were randomly selected from each batch, and the dissolution rate was measured by the method of Test Example 5. The dissolution result curve is shown in Figure 6-7. , the dissolution is greater than 95% in 15 minutes, and the complete dissolution can be achieved within 30 minutes.
2、稳定性试验2. Stability test
针对中试放大得到的样品,在高温(60℃)、强光(光照5000lux)、高湿(75%RH)条件下,测定药物性状、含量和有关物质变化、溶出度、吸湿增重,以进一步确认本发明的药物组合物在放大生产条件下的稳定性。其中,采用目测法观察药物性状,采用HPLC法测定化合物A的游离碱含量和有关物质变化,采用《中国药典》2015年版四部0931溶出度测定法第一法(篮法)测定溶出度,采用《中国药典》2015年版四部0832水分测定法第一法(费休氏法)测定吸湿增重,结果如表23-26所示。For the samples amplified in the pilot scale, under the conditions of high temperature (60°C), strong light (illumination 5000 lux), and high humidity (75% RH), the drug properties, content and changes in related substances, dissolution rate, moisture absorption and weight gain were determined. The stability of the pharmaceutical composition of the present invention under scaled-up production conditions was further confirmed. Among them, the drug properties were observed by visual observation, the free base content of Compound A and the changes of related substances were determined by HPLC, the dissolution was determined by the first method (basket method) of "Chinese Pharmacopoeia" 2015 Edition Part Four 0931 Dissolution Determination Method, and the "Chinese Pharmacopoeia" 2015 Edition Chinese Pharmacopoeia 2015 edition four 0832 moisture determination method first method (Fischer's method) to measure moisture absorption weight gain, the result is shown in table 23-26.
试验结果表明,经由放大生产得到的本发明的胶囊在高温(60℃)、强光(光照5000lux)、高湿(75%RH)条件下,长期放置后均较稳定,各检测项结果没有显著变化,符合质量标准限度要求。进一步证明,在放大生产条件下,得到的本发明的药物组合物同样具有较高的稳定性,且本发明采用的制剂生产工艺简单、稳定,能够满足工业化大生产的需求。The test results show that the capsules of the present invention obtained through enlarged production are stable after long-term storage under the conditions of high temperature (60° C.), strong light (illumination 5000 lux), and high humidity (75% RH), and the results of each test item are not significant. changes to meet quality standard limits. It is further proved that under the conditions of scaled-up production, the obtained pharmaceutical composition of the present invention also has high stability, and the preparation production process adopted in the present invention is simple and stable, and can meet the needs of large-scale industrial production.
Figure PCTCN2021115486-appb-000025
Figure PCTCN2021115486-appb-000025
表24 10mg规格胶囊稳定性试验(高湿75%RH)Table 24 10mg specification capsule stability test (high humidity 75%RH)
Figure PCTCN2021115486-appb-000026
Figure PCTCN2021115486-appb-000026
Figure PCTCN2021115486-appb-000027
Figure PCTCN2021115486-appb-000027
表26 50mg规格胶囊稳定性试验(高湿75%RH)Table 26 Stability test of 50mg capsules (high humidity 75%RH)
Figure PCTCN2021115486-appb-000028
Figure PCTCN2021115486-appb-000028

Claims (21)

  1. 一种药物组合物,所述药物组合物含有作为活性成分的5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺或其药学上可接受的盐以及稀释剂,其特征在于,所述稀释剂选自微晶纤维素、乳糖或其水合物、和糖醇中的一种或多种。A pharmaceutical composition containing 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]-5' as an active ingredient -yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine or a pharmaceutically acceptable salt thereof and a diluent, wherein the The diluent is selected from one or more of microcrystalline cellulose, lactose or its hydrate, and sugar alcohol.
  2. 如权利要求1所述的药物组合物,其特征在于,所述药学上可接受的盐选自5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺的富马酸盐、马来酸盐、己二酸盐和琥珀酸盐中的一种或多种;优选,所述药学上可接受的盐为5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺富马酸盐。The pharmaceutical composition of claim 1, wherein the pharmaceutically acceptable salt is selected from the group consisting of 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1, 3'-Indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine fumarate, maleic acid one or more of salts, adipates and succinates; preferably, the pharmaceutically acceptable salt is 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopenta] Alk-1,3'-indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine fumarate.
  3. 如权利要求1-2任一项所述的药物组合物,其特征在于,以5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺游离碱的含量计,5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺或其药学上可接受的盐的含量为所述药物组合物总重的3.0%以上、优选4.0%以上、优选4%-80%、优选5%-62%、优选6%-50%、更优选为6%-25%、最优选为8%-23%。The pharmaceutical composition according to any one of claims 1-2, characterized in that, with 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indone] Indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine free base content, 5-fluoro-4-( 7'-Fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridine- The content of 2-yl)pyrimidin-2-amine or its pharmaceutically acceptable salt is 3.0% or more, preferably 4.0% or more, preferably 4%-80%, preferably 5%-62% of the total weight of the pharmaceutical composition , preferably 6%-50%, more preferably 6%-25%, most preferably 8%-23%.
  4. 如权利要求1-3任一项所述的药物组合物,其特征在于,所述稀释剂选自微晶纤维素、一水乳糖、甘露醇和山梨醇中的一种或多种;The pharmaceutical composition of any one of claims 1-3, wherein the diluent is selected from one or more of microcrystalline cellulose, lactose monohydrate, mannitol and sorbitol;
    优选地,所述稀释剂为微晶纤维素和一水乳糖的混合物、或微晶纤维素和甘露醇的混合物;Preferably, the diluent is a mixture of microcrystalline cellulose and lactose monohydrate, or a mixture of microcrystalline cellulose and mannitol;
    优选地,所述微晶纤维素为微晶纤维素PH101或微晶纤维素PH102;Preferably, the microcrystalline cellulose is microcrystalline cellulose PH101 or microcrystalline cellulose PH102;
    优选地,所述一水乳糖为一水乳糖FlowLac 100或一水乳糖Tablettose 80;Preferably, described lactose monohydrate is lactose monohydrate FlowLac 100 or lactose monohydrate Tablettose 80;
    优选地,所述甘露醇为甘露醇100SD;Preferably, the mannitol is mannitol 100SD;
    进一步优选地,所述稀释剂为甘露醇100SD和微晶纤维素PH101的混合物、一水乳糖Tablettose 80和微晶纤维素PH102的混合物、或微晶纤维素PH102和一水乳糖FlowLac 100的混合物。Further preferably, the diluent is a mixture of mannitol 100SD and microcrystalline cellulose PH101, a mixture of lactose monohydrate Tablettose 80 and microcrystalline cellulose PH102, or a mixture of microcrystalline cellulose PH102 and lactose monohydrate FlowLac 100.
  5. 如权利要求1-4任一项所述的药物组合物,其特征在于,所述一水乳糖和微晶纤维素的重量比为5∶1-1∶5,优选为3∶1-1∶3,更优选为 1.5∶1-1∶1.5,最优选为1∶1;或The pharmaceutical composition according to any one of claims 1-4, wherein the weight ratio of lactose monohydrate and microcrystalline cellulose is 5:1-1:5, preferably 3:1-1: 3, more preferably 1.5:1 to 1:1.5, most preferably 1:1; or
    所述甘露醇和微晶纤维素的重量比为5∶1-1∶5,优选为3∶1-1∶3,更优选为1.5∶1-1∶1.5,最优选为1∶1。The weight ratio of mannitol and microcrystalline cellulose is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5:1-1:1.5, most preferably 1:1.
  6. 如权利要求1-5任一项所述的药物组合物,其特征在于,所述稀释剂的含量为所述药物组合物总重的20%以上,优选为30%以上,优选为40%以上,优选为20-90%,优选为50-90%,优选为60-90%,更优选为60%-85%,进一步优选为65%-80%。The pharmaceutical composition according to any one of claims 1-5, wherein the content of the diluent is 20% or more, preferably 30% or more, preferably 40% or more of the total weight of the pharmaceutical composition , preferably 20-90%, preferably 50-90%, preferably 60-90%, more preferably 60-85%, further preferably 65-80%.
  7. 如权利要求1-6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含助流剂,优选地,所述助流剂选自滑石粉和胶态二氧化硅中的一种或两种,更优选地,所述助流剂为胶态二氧化硅。The pharmaceutical composition according to any one of claims 1-6, wherein the pharmaceutical composition further comprises a glidant, preferably, the glidant is selected from talc and colloidal silicon dioxide One or both of the above, more preferably, the glidant is colloidal silica.
  8. 如权利要求7所述的药物组合物,其特征在于,所述助流剂的含量为所述药物组合物总重的0.5%-15%,优选为1%-10%,更优选为1%-3%,最优选为1%、2%、或3%。The pharmaceutical composition according to claim 7, wherein the content of the glidant is 0.5%-15% of the total weight of the pharmaceutical composition, preferably 1%-10%, more preferably 1% -3%, most preferably 1%, 2%, or 3%.
  9. 如权利要求1-8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含崩解剂,优选地,所述崩解剂选自交联羧甲基纤维素钠、羧甲基淀粉钠、交联聚乙烯吡咯烷酮和低取代羟丙基纤维素中的一种或多种,更优选地,所述崩解剂为交联羧甲基纤维素钠。The pharmaceutical composition according to any one of claims 1-8, wherein the pharmaceutical composition further comprises a disintegrating agent, preferably, the disintegrating agent is selected from croscarmellose sodium, One or more of sodium carboxymethyl starch, cross-linked polyvinyl pyrrolidone and low-substituted hydroxypropyl cellulose, more preferably, the disintegrant is cross-linked sodium carboxymethyl cellulose.
  10. 如权利要求9所述的药物组合物,其特征在于,所述崩解剂的含量为所述药物组合物总重的0.5%-15%,优选为1%-10%,优选为1%-5%,更优选为1%-3%,更优选为1%、2%、3%、4%、或5%。The pharmaceutical composition of claim 9, wherein the content of the disintegrant is 0.5%-15%, preferably 1%-10%, preferably 1%-15% of the total weight of the pharmaceutical composition 5%, more preferably 1%-3%, more preferably 1%, 2%, 3%, 4%, or 5%.
  11. 如权利要求1-10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含润滑剂,优选地,所述润滑剂选自硬脂酸镁、硬脂酸、硬脂酰富马酸钠和硬脂酸锌中的一种或多种,更优选地,所述润滑剂为硬脂酸镁。The pharmaceutical composition according to any one of claims 1-10, wherein the pharmaceutical composition further comprises a lubricant, preferably, the lubricant is selected from magnesium stearate, stearic acid, stearic acid One or more of sodium acyl fumarate and zinc stearate, more preferably, the lubricant is magnesium stearate.
  12. 如权利要求11所述的药物组合物,其特征在于,所述润滑剂的含量为所述药物组合物总重的0.1%-5%,优选为0.5%-3%,更优选为1%-2%,最优选为1%。The pharmaceutical composition according to claim 11, wherein the content of the lubricant is 0.1%-5% of the total weight of the pharmaceutical composition, preferably 0.5%-3%, more preferably 1%- 2%, most preferably 1%.
  13. 一种药物组合物,其特征在于,所述药物组合物含有以重量计的如下成分:A pharmaceutical composition, characterized in that the pharmaceutical composition contains the following components by weight:
    (1)5%-40%,优选8%-35%,更优选10%-30%的作为活性成分的5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺富马酸盐;(1) 5%-40%, preferably 8%-35%, more preferably 10%-30% as active ingredient 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane] -1,3'-Indol]-5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine fumarate;
    (2)50%-90%,优选60%-90%,优选60%-85%,更优选65%-80%的稀释剂,优选,所述稀释剂为微晶纤维素和一水乳糖的混合物、或微晶纤维素和甘露醇的混合物;(2) 50%-90%, preferably 60%-90%, preferably 60%-85%, more preferably 65%-80% diluent, preferably, the diluent is microcrystalline cellulose and lactose monohydrate a mixture, or a mixture of microcrystalline cellulose and mannitol;
    (3)0.5%-15%,优选为1%-10%,更优选为1%-3%,更优选为1%、2%、或3%的助流剂,优选,所述助流剂为胶态二氧化硅;(3) 0.5%-15%, preferably 1%-10%, more preferably 1%-3%, more preferably 1%, 2%, or 3% of a glidant, preferably, the glidant is colloidal silica;
    (4)0.5%-15%,优选为1%-10%,更优选为1%-5%,更优选为1%-3%,更优选为1%、2%、3%、4%、或5%的崩解剂,优选,所述崩解剂选自交联羧甲基纤维素钠、羧甲基淀粉钠、交联聚乙烯吡咯烷酮和低取代羟丙基纤维素中的一种或多种;和(4) 0.5%-15%, preferably 1%-10%, more preferably 1%-5%, more preferably 1%-3%, more preferably 1%, 2%, 3%, 4%, Or 5% disintegrant, preferably, the disintegrant is selected from one of croscarmellose sodium, sodium carboxymethyl starch, crospovidone and low-substituted hydroxypropyl cellulose or various; and
    (5)0.1%-5%,优选为0.5%-3%,更优选为1%-2%,最优选为1%的润滑剂,优选所述润滑剂选自硬脂酸镁、硬脂酸、硬脂酰富马酸钠和硬脂酸锌中的一种或多种。(5) 0.1%-5%, preferably 0.5%-3%, more preferably 1%-2%, most preferably 1% lubricant, preferably the lubricant is selected from magnesium stearate, stearic acid , one or more of sodium stearoyl fumarate and zinc stearate.
  14. 如权利要求13所述的药物组合物,其特征在于,所述一水乳糖和微晶纤维素的重量比为5∶1-1∶5,优选为3∶1-1∶3,更优选为1.5∶1-1∶1.5,最优选为1∶1;或所述甘露醇和微晶纤维素的重量比为5∶1-1∶5,优选为3∶1-1∶3,更优选为1.5∶1-1∶1.5,最优选为1∶1。The pharmaceutical composition of claim 13, wherein the weight ratio of lactose monohydrate to microcrystalline cellulose is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5:1-1:1.5, most preferably 1:1; or the weight ratio of the mannitol and microcrystalline cellulose is 5:1-1:5, preferably 3:1-1:3, more preferably 1.5 :1-1:1.5, most preferably 1:1.
  15. 如权利要求1-14任一项所述的药物组合物,其中以5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺游离碱的含量计,所述药物组合物包含0.1~1000mg,优选1~100mg,更优选8~50mg活性成分。The pharmaceutical composition of any one of claims 1-14, wherein 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indole]- 5'-yl)-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine free base content, the pharmaceutical composition comprises 0.1-1000mg, Preferably 1 to 100 mg, more preferably 8 to 50 mg of active ingredient.
  16. 一种由权利要求1-15任一项所述的药物组合物制成的固体口服药物制剂,优选散剂、胶囊剂、片剂或颗粒剂,更优选胶囊剂,其中所述胶囊剂以所述药物组合物作为填充物。A solid oral pharmaceutical preparation made from the pharmaceutical composition of any one of claims 1-15, preferably powder, capsule, tablet or granule, more preferably capsule, wherein the capsule is in the form of the Pharmaceutical compositions as fillers.
  17. 如权利要求16所述的固体口服药物制剂,其中以5-氟-4-(7’-氟-2’-甲基螺[环戊烷-1,3’-吲哚]-5’-基)-N-(5-(1-甲基哌啶-4-基)吡啶-2-基)嘧啶-2-胺游离碱的含量计,所述固体口服药物制剂包含0.1~1000mg,优选1~100mg,更优选8~50mg活性成分。The solid oral pharmaceutical formulation of claim 16, wherein 5-fluoro-4-(7'-fluoro-2'-methylspiro[cyclopentane-1,3'-indol]-5'-yl )-N-(5-(1-methylpiperidin-4-yl)pyridin-2-yl)pyrimidin-2-amine free base content, the solid oral pharmaceutical preparation comprises 0.1~1000mg, preferably 1~ 100 mg, more preferably 8 to 50 mg of active ingredient.
  18. 如权利要求16或17所述的固体口服药物制剂,其特征在于,所述胶囊剂采用粉末直接灌装工艺制备。The solid oral pharmaceutical preparation according to claim 16 or 17, wherein the capsule is prepared by a powder direct filling process.
  19. 如权利要求1-15任一项所述的药物组合物或权利要求16-18任一项所述的固体口服药物制剂,其用于治疗CDK4/6介导的障碍或疾病。The pharmaceutical composition of any one of claims 1-15 or the solid oral pharmaceutical formulation of any one of claims 16-18, for use in the treatment of a CDK4/6 mediated disorder or disease.
  20. 如权利要求1-15任一项所述的药物组合物或权利要求16-18任一项所述的固体口服药物制剂在制备用于治疗CDK4/6介导的障碍或疾病的药物中的用途。Use of the pharmaceutical composition according to any one of claims 1-15 or the solid oral pharmaceutical preparation according to any one of claims 16-18 in the manufacture of a medicament for the treatment of CDK4/6 mediated disorders or diseases .
  21. 一种用于治疗CDK4/6介导的障碍或疾病的方法,其包括向需要其的对象施用如权利要求1-15任一项所述的药物组合物或权利要求16-18任一项所述的固体口服药物制剂。A method for treating a CDK4/6-mediated disorder or disease, comprising administering the pharmaceutical composition according to any one of claims 1-15 or any one of claims 16-18 to a subject in need thereof solid oral pharmaceutical preparations.
PCT/CN2021/115486 2020-08-31 2021-08-31 Pharmaceutical composition containing cdk4/6 inhibitor WO2022042738A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180053613.XA CN116113628A (en) 2020-08-31 2021-08-31 Pharmaceutical composition containing CDK4/6 inhibitor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010893836.4 2020-08-31
CN202010893836 2020-08-31

Publications (1)

Publication Number Publication Date
WO2022042738A1 true WO2022042738A1 (en) 2022-03-03

Family

ID=80354702

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/115486 WO2022042738A1 (en) 2020-08-31 2021-08-31 Pharmaceutical composition containing cdk4/6 inhibitor

Country Status (2)

Country Link
CN (1) CN116113628A (en)
WO (1) WO2022042738A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102007124A (en) * 2008-02-15 2011-04-06 里格尔制药公司 Pyrimidine-2-amine compounds and their use as inhibitors of jak kinases
CN102264725A (en) * 2008-12-22 2011-11-30 伊莱利利公司 Protein kinase inhibitors
CN106810536A (en) * 2015-11-30 2017-06-09 甘李药业股份有限公司 A kind of kinases inhibitor and preparation method thereof and medical usage
WO2020253875A1 (en) * 2019-06-21 2020-12-24 甘李药业股份有限公司 Salts of compound, crystalline forms thereof, preparation method therefor and application thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102007124A (en) * 2008-02-15 2011-04-06 里格尔制药公司 Pyrimidine-2-amine compounds and their use as inhibitors of jak kinases
CN102264725A (en) * 2008-12-22 2011-11-30 伊莱利利公司 Protein kinase inhibitors
CN106810536A (en) * 2015-11-30 2017-06-09 甘李药业股份有限公司 A kind of kinases inhibitor and preparation method thereof and medical usage
WO2020253875A1 (en) * 2019-06-21 2020-12-24 甘李药业股份有限公司 Salts of compound, crystalline forms thereof, preparation method therefor and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BRONNER, S. M. ET AL.: "Design of a brain-penetrant CDK4/6 inhibitor for glioblastoma", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 29, 26 June 2019 (2019-06-26), XP085759050, DOI: 10.1016/j.bmcl.2019.06.021 *
LEI YIN, HENG LI, WENJIAN LIU, ZHENGLIN YAO, ZHENZHEN CHENG, HUABEI ZHANG, HUI ZOU: "A highly potent CDK4/6 inhibitor was rationally designed to overcome blood brain barrier in gliobastoma therapy", EUROEPAN JOURNAL OF MEDICINAL CHEMISTRY, ELSEVIER, pages 1 - 28, XP055766971, [retrieved on 20210120] *

Also Published As

Publication number Publication date
CN116113628A (en) 2023-05-12

Similar Documents

Publication Publication Date Title
TW201328725A (en) Pharmaceutical compositions of N-methyl-2-3-((E)-2-pyridin-2-yl-vinyl)-1H-indazol-6-ylsulfanyl-benzamide
CZ297954B6 (en) Ziprasidone-based pharmaceutical composition and process for preparing large crystals of ziprasidone hydrochloride monohydrate
CN102988296A (en) Celecoxib solid dispersion and preparation method thereof
JP2022548688A (en) Sustained release dosage form of TYK2 inhibitor
CN104887641A (en) Palbociclib gastric-floating tablet and preparation method thereof
CN113508114B (en) Oral pharmaceutical composition with azetidine derivative as active ingredient, preparation method and application thereof
CN110446705A (en) (S)-[the fluoro- 5- of the chloro- 4- of 2- (7- morpholine -4- base quinazoline -4- base) phenyl] solid form of-(6- methoxy-pyridazine -3- base) methanol
WO2022042738A1 (en) Pharmaceutical composition containing cdk4/6 inhibitor
WO2023213878A1 (en) Solid tablet dosage form of ridinilazole
JP2022524424A (en) Compound forms and formulations thereof with improved bioavailability
CN109963565B (en) Pharmaceutical composition and preparation method thereof
TWI738729B (en) A preparation method of pharmaceutical composition comprising pyrrolo six-member heterocyclic compound or its medicinal salt thereof
CN114028400B (en) Pharmaceutical composition containing cyclin kinase inhibitor and preparation method thereof
US20220226249A1 (en) Solid tablet dosage form of ridinilazole
CN106176752A (en) ceritinib pharmaceutical composition
KR102486126B1 (en) Pharmaceutical compositions comprising alpelisib
US10973807B2 (en) Pharmaceutical composition comprising pyrrolo-fused six-membered heterocyclic compound
TW202038918A (en) Pharmaceutical composition
JPWO2019230937A1 (en) Oral solid preparation with excellent dissolution
CN110664812B (en) Pharmaceutical composition containing imidazo isoindole derivative
WO2023186031A1 (en) Pharmaceutical composition of multi-target protein kinase inhibitor, use thereof, and method for preparing same
WO2023085300A1 (en) Pharmaceutical composition having excellent elution property
CN107648237B (en) Pharmaceutical composition of aminopyrimidine compounds and preparation method thereof
CN114727965A (en) JAK kinase inhibitor pharmaceutical composition
CN117357531A (en) Pharmaceutical composition, preparation method and application thereof

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: 21860570

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: 21860570

Country of ref document: EP

Kind code of ref document: A1