WO2024007801A1 - Combination therapy of receptor tyrosine kinase inhibitor and biphenyl cyclooctadiene lignan and use thereof - Google Patents
Combination therapy of receptor tyrosine kinase inhibitor and biphenyl cyclooctadiene lignan and use thereof Download PDFInfo
- Publication number
- WO2024007801A1 WO2024007801A1 PCT/CN2023/098571 CN2023098571W WO2024007801A1 WO 2024007801 A1 WO2024007801 A1 WO 2024007801A1 CN 2023098571 W CN2023098571 W CN 2023098571W WO 2024007801 A1 WO2024007801 A1 WO 2024007801A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- formula
- salt
- compound represented
- biphenylcyclooctadiene
- lignan
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/075—Ethers or acetals
- A61K31/085—Ethers or acetals having an ether linkage to aromatic ring nuclear carbon
- A61K31/09—Ethers or acetals having an ether linkage to aromatic ring nuclear carbon having two or more such linkages
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/335—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
- A61K31/357—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having two or more oxygen atoms in the same ring, e.g. crown ethers, guanadrel
- A61K31/36—Compounds containing methylenedioxyphenyl groups, e.g. sesamin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic 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
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/517—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
Definitions
- the present invention relates to the field of medicine. Specifically, the present invention relates to the combined use of receptor tyrosine kinase inhibitors and biphenylcyclooctadiene lignans and their uses. More specifically, the present invention relates to a pharmaceutical composition, Single dosage form, drug combination, kit, administration method of the compound represented by formula I or its salt, method of extending the half-life of the compound represented by formula I or its salt, reducing the compound represented by formula I or its salt to form the compound represented by formula II Methods.
- (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate and its salts are not An inhibitor (EGFR-TKI) that is limited to oral administration and can inhibit the activity of epidermal growth factor receptor tyrosine kinase with activating mutations, initially developed for the treatment of non-small cell lung cancer (NSCLC) with central nervous system metastasis (CNS) ).
- NSCLC non-small cell lung cancer
- CNS central nervous system metastasis
- CYP450 enzymes metabolized in the human body are CYP3A4 and CYP3A5 subtypes. Their drug efficacy increases with the increase in drug dosage. However, the occurrence of adverse reactions limits its further clinical application. The occurrence of adverse reactions may be related to the regular use. CYP450 enzymes, such as its subtype CYP3A metabolizing enzyme, are related to the generation of metabolites, and the generation of some metabolites increases at a higher rate than the administered dose.
- (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate or its salt is An effective EGFR inhibitor that can penetrate the central nervous system, biphenylcyclooctadienol is a CYP450 enzyme inhibitor and can reduce serum alanine aminotransferase and play a role in protecting the liver.
- the invention provides a pharmaceutical composition.
- the pharmaceutical composition includes: a compound represented by Formula I or a salt thereof and biphenylcyclooctadienolignan.
- the pharmaceutical composition according to the embodiment of the present invention can reduce the content of alanine aminotransferase in serum, play a protective role on the liver, and compared with the single use of the compound represented by formula I or its salt, the pharmaceutical composition can effectively prolong the The duration of drug efficacy of the compound represented by formula I or its salt in the body, and significantly reducing the concentration of the metabolite represented by formula II during the metabolic process, increasing the exposure of the compound represented by formula I or its salt in the body and the dose in brain tissue, Reducing the adverse reactions caused by the metabolites represented by formula II improves the efficacy and safety of the compounds represented by formula I or salts thereof, thereby effectively treating or preventing cancer.
- the above-mentioned pharmaceutical composition may further include at least one of the following additional technical features:
- the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
- the salt of the compound represented by Formula I is a hydrochloride.
- the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
- the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
- the biphenylcyclooctadiene lignan is schisandrin A.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3), the half-life of the compound represented by formula I or its salt is extended.
- the content of the metabolite shown in Formula II during the metabolic process is lower than that of the compound shown in Formula I or its salt alone, and it has strong anti-tumor ability and high safety.
- the dosages for mice and humans can be converted, and adding the above two substances using the converted values can effectively protect humans from or alleviate tumors.
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6)
- the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite shown in formula II during the metabolic process is significantly lower than that of the compound shown in formula I or its salt alone, and it has strong anti-tumor ability and high safety.
- the dosages for mice and humans can be converted, and adding the above two substances using the converted values can effectively protect humans from or alleviate tumors.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5), the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite represented by formula II during the metabolic process is significantly lower than that of the compound represented by formula I or its salt alone, and the pharmaceutical composition has strong anti-tumor ability and high safety.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125.
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125
- the compound represented by formula I or its salt is The half-life is significantly extended, and the content of the metabolite represented by formula II during metabolism is significantly lower than when the compound represented by formula I or its salt is used alone.
- the pharmaceutical composition has strong anti-tumor ability and high safety.
- the pharmaceutical composition is an oral preparation.
- the pharmaceutical composition may be in the form of taking preparations, injection preparations, pills, sustained-release preparations, implants or aerosols, etc., and is not particularly limited.
- the pharmaceutical composition when it is an oral preparation, the half-life of the single dosage form is longer, and the content of the metabolite shown in Formula II during metabolism is significantly lower than when the compound shown in Formula I or its salt is used alone, which is beneficial to improving the penetration of the compound shown in Formula I or its salt.
- the brain dose further improves the drug efficacy. Therefore, the pharmaceutical composition has stronger anti-tumor ability and higher safety.
- the pharmaceutical composition includes the compound represented by Formula I or a salt thereof provided herein, and Schisandrin A, as well as a pharmaceutically acceptable carrier.
- “Pharmaceutically acceptable carrier” may include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Specific examples may be one or more of water, saline, phosphate buffered saline, glucose, glycerin, ethanol, etc., and combinations thereof.
- pharmaceutical compositions include isotonic agents, such as sugars, polyols (such as mannitol, sorbitol) or sodium chloride.
- pharmaceutically acceptable carriers may also include trace amounts of auxiliary substances, such as wetting agents or emulsifiers, preservatives or buffers, to extend the shelf life or effectiveness of the antibody.
- the compound represented by formula I of the present invention or a salt thereof and biphenylcyclooctadienolignan can be incorporated into a pharmaceutical composition suitable for parenteral administration (such as intravenous, subcutaneous, intraperitoneal, intramuscular) .
- parenteral administration such as intravenous, subcutaneous, intraperitoneal, intramuscular
- these pharmaceutical compositions can be prepared in various forms. Examples include liquid, semi-solid, and solid dosage forms, including, but not limited to, liquid solutions (eg, injection solutions and infusion solutions), dispersions or suspensions, tablets, pills, powders, liposomes, and suppositories.
- Typical pharmaceutical compositions are in the form of oral preparations, injection solutions, or infusion solutions.
- the pharmaceutical composition may be administered orally, intravenously, or by injection, or intramuscularly or subcutaneously.
- a single dosage form includes: the compound represented by formula I or its salt and biphenylcyclooctadiene lignan.
- biphenylcyclooctadiene lignans such as Schisandrin A
- the single The dosage form has strong anti-tumor ability and high safety. According to the description of the embodiments of this application, through the exchange of dosage between mice and humans, Calculation, using this ratio for conversion can effectively protect humans from or alleviate tumors.
- the above-mentioned single dosage form may further include at least one of the following additional technical features:
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3).
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6).
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5), the half-life of the single dosage form is significantly extended, and the metabolism process of formula II
- the content of the metabolites shown is significantly lower than that of the compound shown in formula I or its salt alone, thereby increasing the brain-penetrating dose of the compound shown in formula I or its salt and improving the efficacy. Therefore, the single dosage form has strong anti-bacterial activity. tumor capability and higher safety.
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125.
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125.
- the half-life of the single dosage form is significantly extended, and the metabolism In the process, the content of the metabolite represented by formula II is significantly lower than that of the compound represented by formula I or its salt alone, thereby increasing the brain-penetrating dose of the compound represented by formula I or its salt and improving the drug efficacy. Therefore, the single dosage form has very high Strong anti-tumor ability and high safety.
- the single dosage form includes 200 to 300 mg of the compound represented by Formula I or a salt thereof.
- the above-mentioned single dosage form can be used 2 times, 3 times or 4 times a day, preferably 2 times. Through the conversion of dosages for mice and humans, this dosage can effectively protect humans after conversion. Avoid or alleviate tumors.
- the specifications of the single dosage form are not particularly limited.
- the specifications of the single dosage form can be set according to the characteristics of different target groups.
- the specifications of the single dosage form can be set according to the weight of the target group, such as a body weight of 40kg.
- the compound represented by Formula I or its salt described in this application can be used according to the characteristics of the target population and the Set the mass ratio of elements.
- the single dosage form is an oral preparation.
- the single dosage form may be an oral preparation, an injection preparation, a pill, a sustained-release preparation, an implant or an aerosol, and is not particularly limited.
- when the single dosage form is oral The half-life of the single dosage form is longer during preparation, and the content of the metabolite represented by formula II during the metabolism process is significantly lower than that of the compound represented by formula I or its salt alone, thereby increasing the penetration of the compound represented by formula I or its salt. Brain dose improves drug efficacy. Therefore, the single dosage form has stronger anti-tumor ability and higher safety.
- the invention proposes a drug combination.
- the compound of formula I or a salt thereof is included as the first active ingredient and biphenylcyclooctadiene lignan as the second active ingredient.
- Schisandrin A not only has the function of protecting the liver, but also can reduce the metabolism of the compound represented by Formula I or its salt by inhibiting CYP450 enzymes, such as subtype CYP3A, and prolong its retention in the body.
- the drug combination according to the embodiment of the present invention can significantly extend the half-life and brain-penetrating dose of the compound represented by Formula I or its salt, can effectively inhibit tumor growth, and has high safety.
- the above-mentioned drug combination may further include at least one of the following additional technical features:
- the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
- the salt of the compound represented by Formula I is a hydrochloride.
- the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
- the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
- the biphenylcyclooctadiene lignan is schisandrin A.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3), the half-life of the compound represented by formula I or its salt is extended.
- the content of the metabolite shown in Formula II during the metabolic process is lower than that of the compound shown in Formula I or its salt alone, and it has strong anti-tumor ability and high safety.
- the dosages for mice and humans can be converted, and adding the above two substances using the converted values can effectively protect humans from or alleviate tumors.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6)
- the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite shown in formula II during the metabolic process is significantly lower than that of the compound shown in formula I or its salt alone, thereby increasing the brain-penetrating dose of the compound shown in formula I or its salt, and improving the drug efficacy, as described after drug combination.
- the compound represented by formula I or its salt has stronger anti-tumor ability and higher safety. According to the description of the embodiments of the present application, by converting the dosage between mice and humans, using this ratio can effectively protect humans from or alleviate tumors.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5), the half-life of the compound represented by formula I or its salt is significantly improved.
- the content of the metabolite represented by formula II is significantly lower than that of the compound represented by formula I or its salt alone, thereby increasing the brain-penetrating dose of the compound represented by formula I or its salt, and improving the drug efficacy.
- formula I is described The indicated compound or its salt has stronger anti-tumor ability and higher safety.
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125.
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:
- When 125 the half-life of the compound represented by formula I or its salt is significantly extended, and the content of the metabolite represented by formula II is significantly lower than that of the compound represented by formula I or its salt alone, thereby increasing the concentration of the compound represented by formula I or its salt.
- the brain-penetrating dose of the salt can improve the drug efficacy. After drug combination, the compound represented by formula I or its salt has stronger anti-tumor ability and higher safety.
- the compound represented by Formula I or its salt and the biphenylcyclooctadiene lignan are formulated together or separately.
- the compound represented by formula I or its salt and the biphenylcyclooctadiene lignan are used simultaneously or separately.
- the drug combination includes combinations that are separated in time and/or space, as long as the compound represented by formula I or its salt and biphenylcyclooctadienolignan can cooperate to achieve the purpose of the present invention.
- the components contained in the pharmaceutical combination can be administered to the subject or the sample to be tested as a whole, or separately.
- the individual components may be administered to the subject or sample to be tested simultaneously or sequentially.
- the biphenylcyclooctadienolignan is administered prior to the compound represented by Formula I or a salt thereof.
- the order of administration of the biphenylcyclooctadienol and the compound represented by formula I or its salt is not particularly limited, as long as the diphenylcyclooctadienol It is sufficient that the compound can play a role in the metabolic cycle of the compound represented by formula I or its salt.
- the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt
- the biphenylcyclooctadiene lignan is administered after the compound represented by formula I or its salt
- the biphenylcyclooctadienolignan and the compound represented by formula I or its salt are administered Simultaneous administration is within the scope of protection of this application.
- the biphenylcyclooctadienolignan precedes the (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquin when oxazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride is administered, the half-life of the compound represented by formula I or its salt can be significantly extended, and during its metabolism The content of the metabolite shown in the formula II produced is significantly reduced, so that the compound shown in the formula or its salt has a higher brain penetration rate, which increases the brain penetration dose and further improves the drug efficacy.
- the drug combination has a good anti-tumor effect and security.
- the second active ingredient is in an oral dosage form.
- the second active ingredient may be in the form of taking preparations, injection preparations, pills, sustained-release preparations, implants or aerosols, etc., and is not particularly limited.
- the half-life of the single dosage form is longer, and the content of the metabolite shown in Formula II during metabolism is significantly lower than when the compound shown in Formula I or its salt is used alone, and the compound shown in Formula I or its salt has Stronger anti-tumor ability and higher safety.
- the invention provides a medicine box.
- the pharmaceutical kit includes: a compound represented by Formula I or a salt thereof as the first active ingredient and biphenylcyclooctadienolignan as the second active ingredient.
- biphenylcyclooctadienolignan reduces the metabolism of the compound represented by formula I or its salt by inhibiting CYP450 enzymes, such as its subtype CYP3A enzyme, prolongs its residence time in the body and enhances the therapeutic effect.
- the above-mentioned medicine box may further include at least one of the following additional technical features:
- the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
- the salt of the compound represented by Formula I is a hydrochloride.
- the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
- the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
- the biphenylcyclooctadiene lignan is schisandrin A.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3), the half-life of the compound represented by formula I or its salt is extended.
- the content of the metabolite shown in Formula II during the metabolic process is lower than that of the compound shown in Formula I or its salt alone, and it has strong anti-tumor ability and high safety.
- the dosages for mice and humans can be converted, and adding the above two substances using the converted values can effectively protect humans from or alleviate tumors.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6)
- the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite of the metabolite shown in formula II during the metabolic process is significantly lower than that of the compound shown in formula I or its salt alone, and it has strong anti-tumor ability and high safety.
- the dosages for mice and humans can be converted, and adding the above two substances using the converted values can effectively protect humans from or alleviate tumors.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5), the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite of the metabolite shown in Formula II during the metabolic process is significantly lower than that of the compound shown in Formula I or its salt alone, and the kit has strong anti-tumor ability and high safety.
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125.
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125
- the compound represented by formula I or its salt is The half-life is significantly extended, and the content of the metabolite represented by formula II during metabolism is significantly lower than when the compound represented by formula I or its salt is used alone.
- the pharmaceutical composition has strong anti-tumor ability and high safety.
- the compound represented by Formula I or its salt and the biphenylcyclooctadiene lignan are formulated together or separately.
- the preparation method of the compound represented by formula I or its salt and the biphenylcyclooctadiene lignan is not particularly limited, and they can be prepared separately or together.
- the compound represented by formula I or its salt and the biphenylcyclooctadiene lignan are used simultaneously or separately. It should be noted that in the method, the compound represented by formula I or its salt and the biphenylcyclooctadiene lignan are used separately in time and/or space, as long as the compound represented by formula I or its salt is used separately in time and/or space.
- the salt and biphenylcyclooctadiene lignan salt can cooperate to achieve the purpose of the present invention.
- the compound represented by Formula I or a salt thereof and the biphenylcyclooctadienolignan can be administered to the subject or the sample to be tested as a whole, or separately to the subject or the sample to be tested.
- each component may be administered to the subject or sample to be tested simultaneously or sequentially. sample.
- the biphenylcyclooctadienolignan is administered prior to the compound represented by Formula I or a salt thereof.
- the order of administration of the biphenylcyclooctadienol and the compound represented by formula I or its salt is not particularly limited, as long as the diphenylcyclooctadienol It is sufficient that the compound can play a role in the metabolic cycle of the compound represented by formula I or its salt.
- the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt
- the biphenylcyclooctadiene lignan is administered after the compound represented by formula I or its salt
- the biphenylcyclooctadienolignan and the compound represented by formula I or its salt are administered Simultaneous administration is within the scope of protection of this application.
- the half-life of the compound represented by formula I or its salt can be It is significantly prolonged, and the content of the metabolite shown in formula II produced during its metabolism is significantly reduced, and it has good anti-tumor effect and safety.
- the second active ingredient is in an oral dosage form.
- the second active ingredient may be in the form of taking preparations, injection preparations, pills, sustained-release preparations, implants or aerosols, etc., and is not particularly limited.
- the half-life of the kit is longer, and the content of the metabolite shown in Formula II during metabolism is significantly lower than when the compound shown in Formula I or its salt is used alone, and the kit has strong anti-tumor ability. and higher security.
- the present invention provides a method of administering the compound represented by Formula I or a salt thereof.
- the method includes combining the compound represented by Formula I or a salt thereof with biphenylcyclooctadienolignan.
- the above method further includes at least one of the following additional technical features:
- the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
- the salt of the compound represented by Formula I is a hydrochloride.
- the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
- the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
- the biphenylcyclooctadiene lignan is schisandrin A.
- the combined medication includes administering the compound represented by formula I or its salt and biphenylcyclooctadienolignan simultaneously or separately.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3). According to the description of the embodiments of the present application, by converting the dosage between mice and humans, this ratio conversion can effectively protect humans from or alleviate tumors.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (1:10) to (5:6).
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (7:50) to (4:5).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan during the medication process is 3:20, 1:5, 81:250 or 81:125.
- the biphenylcyclooctadienolignan is administered prior to the compound represented by Formula I or a salt thereof.
- the order of administration of the biphenylcyclooctadienol and the compound represented by formula I or its salt is not particularly limited, as long as the diphenylcyclooctadienol It is sufficient that the compound can play a role in the metabolic cycle of the compound represented by formula I or its salt.
- the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt
- the biphenylcyclooctadiene lignan is administered after the compound represented by formula I or its salt
- the biphenylcyclooctadienolignan and the compound represented by formula I or its salt are administered Simultaneous administration is within the scope of protection of this application.
- the biphenylcyclooctadiene lignan is administered orally.
- the biphenylcyclooctadiene lignans can be in the form of oral preparations, injection preparations, pills, sustained-release preparations, implants or aerosols, etc., and are not particularly limited.
- the biphenylcyclooctadiene lignan is an oral preparation, the half-life of the compound represented by formula I or its salt is longer, and the content of metabolite 1 during metabolism is significantly lower than that of the compound alone.
- the administration method has stronger anti-tumor ability and higher safety.
- the dosage of the biphenylcyclooctadiene lignan is 0 to 25 mg/kg.
- the dosage of the biphenylcyclooctadiene lignan is 5 to 25 mg/kg.
- the half-life of the compound represented by formula I or its salt can be significantly extended, and the The content of the metabolites shown in formula II produced during the metabolism process is significantly reduced, which increases the exposure of the compound shown in formula I or its salt in the body and the dose in brain tissue, and reduces the adverse reactions caused by the metabolites shown in formula II, so that The efficacy and safety of the compound represented by formula I or its salt are improved, and the administration method can more effectively inhibit tumor growth and has higher safety.
- the present invention proposes a method for extending the half-life of the compound represented by formula I or its salt.
- the method includes combining the compound represented by Formula I or a salt thereof with biphenylcyclooctadiene lignan.
- biphenylcyclooctadienolignans reduce the metabolism of the compound represented by formula I or its salt by inhibiting CYP450 enzymes, such as its subtype CYP3A enzyme, prolonging its residence time in the body and enhancing the therapeutic effect. , Therefore, the method according to the embodiment of the present invention can effectively extend the half-life of the compound represented by formula I or its salt.
- the above method further includes at least one of the following additional technical features:
- the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
- the salt of the compound represented by Formula I is a hydrochloride.
- the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
- the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
- the biphenylcyclooctadiene lignan is schisandrin A.
- the combined medication includes administering the compound represented by formula I or its salt and biphenylcyclooctadienolignan simultaneously or separately.
- the method of combined administration will not be described in detail, and reference may be made to the method of combined administration in the administration method of the compound represented by formula I or its salt described in the fifth aspect.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3). According to the description of the embodiments of the present application, by converting the dosage between mice and humans, using this ratio conversion can effectively extend the efficacy time of protecting humans from or alleviating tumors.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (1:10) to (5:6).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (7:10) to (4:5).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan during the medication process is 3:20, 1:5, 81:250 or 81:125.
- the biphenylcyclooctadienolignan is administered prior to the compound represented by Formula I or a salt thereof.
- the order of administration of the biphenylcyclooctadienolignan and the compound represented by Formula I or its salt is not particularly limited, as long as the biphenylcyclooctadienolignan can It only needs to be effective within the metabolic cycle of the compound represented by formula I or its salt.
- the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt, or the The biphenylcyclooctadiene lignan is administered after the compound represented by the formula I or its salt, or the biphenylcyclooctadiene lignan and the compound represented by the formula I or its salt are administered simultaneously. All drugs are within the protection scope of this application.
- the biphenylcyclooctadiene lignan is administered orally.
- the dosage of the biphenylcyclooctadiene lignan is 0 to 25 mg/kg.
- the dosage of the biphenylcyclooctadiene lignan is 5 to 25 mg/kg.
- the half-life of the compound represented by formula I or its salt can be significantly extended, and the The content of the metabolites shown in formula II produced during the metabolic process is significantly reduced, and the method can more effectively extend the The half-life of the compound represented by formula I or its salt is increased, and its brain penetration rate is increased to improve the drug efficacy.
- the present invention proposes a method for reducing the compound represented by formula I or its salt to produce the compound represented by formula II.
- the method includes combining the compound represented by Formula I or a salt thereof with biphenylcyclooctadienolignan.
- biphenylcyclooctadienolignan inhibits CYP450 enzymes, such as its subtype CYP3A enzyme, thereby reducing the metabolism of the compound represented by formula I or its salt, and reducing the concentration of the metabolite represented by formula II in the serum.
- the above method further includes at least one of the following additional technical features:
- the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
- the salt of the compound represented by Formula I is a hydrochloride.
- the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
- the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
- the biphenylcyclooctadiene lignan is schisandrin A.
- the combined medication includes administering the compound represented by formula I or its salt and biphenylcyclooctadienolignan simultaneously or separately.
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3).
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (1:10) to (5:6).
- the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (7:50) to (4:5).
- the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan during the medication process is 3:20, 1:5, 81:250 or 81:125.
- the biphenylcyclooctadienolignan is administered prior to the compound represented by Formula I or a salt thereof.
- the order of administration of the biphenylcyclooctadienolignan and the compound represented by Formula I or its salt is not particularly limited, as long as the biphenylcyclooctadienolignan can It only needs to be effective within the metabolic cycle of the compound represented by formula I or its salt.
- the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt, or the The biphenylcyclooctadiene lignan is administered after the compound represented by the formula I or its salt, or the biphenylcyclooctadiene lignan and the compound represented by the formula I or its salt are administered simultaneously. All drugs are within the protection scope of this application.
- the biphenylcyclooctadiene lignan is administered orally.
- the dosage of the biphenylcyclooctadiene lignan is 0 to 25 mg/kg.
- the dosage of the biphenylcyclooctadiene lignan is 5 to 25 mg/kg.
- the dosage of Schisandrin A is 5 to 25 mg/kg
- the compound represented by Formula I or its salt can more effectively reduce the amount of Formula II produced in the serum during the metabolism process. Therefore, the method according to the embodiment of the present invention can more significantly reduce the content of the metabolite shown in Formula II during the metabolism of the compound shown in Formula I or its salt.
- single dosage form refers to a dosage form that is used up after a single administration of a preparation.
- the drug combination refers to the simultaneous or sequential application of two or more drugs to achieve therapeutic purposes. The result is mainly to increase the efficacy of the drug or to reduce the toxic and side effects of the drug.
- the experimental system uses human liver microsomes.
- liver microsomes There are 4 groups of liver microsomes each, namely 1) (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl. 2,4-Dimethylpiperazine-1-carboxylate hydrochloride group (test drug alone group); 2) (R)-4-(3-chloro-2-fluorophenylamino)-7 -Methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride combined with Schizandrin A (WWZ) group (test drug + WWZ group); 3) ( R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride with water Silybin group (test drug + silibinin group), 4) (R)-4-(3-ch
- liver microsomes (HLM) was 20 mg/mL, and it was diluted with PBS buffer solution to a liver microsome working solution with a molar concentration of 2 mg/mL. Take each of the above working solutions and prepare according to Table 1.
- this experiment mainly investigated the original drug (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-di Differences in metabolic stability of methylpiperazine-1-carboxylate hydrochloride in liver microsome incubation system, and changes in metabolic stability after combined use with the inhibitor WWZ.
- the experimental system uses human liver microsomes. There are three groups of liver microsomes, each of which is 1) (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl.
- Verapamil was used as a positive control (sampling time points were 30 min and 2 h after incubation), and verapamil without NADPH (replaced by PBS) was used as a negative control.
- the drug-free (replaced by PBS) group served as a blank control.
- This example is carried out on the basis of Examples 1 and 2, mainly investigating the test drug ((R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazoline- Changes in metabolic kinetics in SD rats after combined administration of 6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride and schizandrin.
- test drug ((R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazoline- Changes in metabolic kinetics in SD rats after combined administration of 6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride and schizandrin.
- 9 healthy growing male rats aged 6-8 weeks and weighing 220-300g were divided into 3 groups: the test drug (5mg/kg) group was administered by gavage; the group was administered 25 mg of Schisandrin A by gavage.
- the whole blood of the rats in the above three groups was collected at the following time points: before administration and 5min, 15min, 30min, 1h, 2h, 4h, 8h, 12h, 24h, and 36h after administration.
- Blood collection site and volume About 0.3 mL of blood is collected from the retroocular venous plexus.
- the obtained whole blood sample was centrifuged (centrifugal force 2000g, 4°C) for 10 min, and the upper plasma was taken. Plasma samples were processed using organic solvent protein precipitation.
- the blood drug concentration-time change data of the test drugs in each group of animals are shown in Table 5, and the main pharmacokinetic parameters are shown in Tables 6 and 7.
- Tables 6 and 7 The blood drug concentration-time change data of the test drugs in each group of animals are shown in Table 5, and the main pharmacokinetic parameters are shown in Tables 6 and 7.
- Tables 6 and 7 Based on the in vivo metabolism results, it can be seen that the in vivo PK behavior of the test drug compounds is affected after taking WWZ. This effect is related to the administration method of co-administration of WWZ. Intravenous co-administration did not change the clearance rate of the test drug, while oral co-administration improved the bioavailability of the test drug. From this, the inventor speculates that drug interactions with WWZ cannot simply be explained by the inhibitory effect of WWZ on liver metabolic enzymes, but are mainly manifested in the absorption process.
- WWZ mainly acts on the intestines and liver in the absorption phase, improving bioavailability by increasing the absorption of the test drug or reducing the first pass of the liver and intestines.
- (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylic The average exposure AUCinf and half-life t 1/2 h of acid ester hydrochloride were significantly higher than those of (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazoline alone -6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride group.
- the concentration-time change results of metabolite 1 in the plasma of SD rats in each group are shown in Table 7.
- Table 7 and Table 8 show that the metabolic production pattern of metabolite 1 has changed after taking WWZ; whether it is taken orally or When WWZ was administered intravenously together, within 8 hours after its administration, the concentration of metabolite 1 was lower than that of the test drug alone group, indicating that the combined administration of WWZ can effectively inhibit the production of metabolite 1.
- the blood exposure Cmax of metabolite 1 in the drug alone group was as high as 261ng/mL 4 hours after administration.
- this example continues to study the test drugs and WWZ.
- the PC9 tumor tissue is taken out under sterile conditions and cut into tumor pieces of 2.0mm ⁇ 2.0mm ⁇ 2.0mm, uniformly Inoculate into children weighing 16.0-18.0g BALB/cAnSlacNifdc-nu male nude mice were injected subcutaneously on the left axillary back, and the day of inoculation was recorded as D0.
- the tumors are randomly divided into groups according to the tumor volume: solvent control group 1 (25% PEG400 + pH 5.5 water), solvent control group 2 (pH 5.5 water), WWZ ( 25.0 mg/kg) group, test drug [(R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiper Azine-1-carboxylate hydrochloride] group (8.1 mg/kg) group, test drug [(R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazole Phenolin-6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride] group (16.2 mg/kg), WWZ (25.0 mg/kg) + test drug group (8.1 mg/kg) WWZ (25.0mg/kg) + test drug group (16.2mg/kg), a total of 7 groups, 10 animals in each group;
- the effective doses were converted into human doses of 0.7 mg/kg and 1.4 mg/kg based on the body surface area dose. .
- Instructions on the dosage of WWZ The current drug name of WWZ in human clinical use is Wuester Capsule (TCM), and its preparation is a capsule, each capsule containing WWZ 11.25mg.
- the clinical usage and dosage is two capsules each time, three times a day, and the daily dosage is 67.5mg.
- This dose is the highest clinically used daily dose for adults (60kg).
- This highest dose is converted into a dose for mice in this study by body surface area dose. In this study, mice were given a dose group of WWZ. , that is, the highest effective dose is 25.0mg/kg.
- Tumor volume 0.5 ⁇ long diameter ⁇ short diameter 2 .
- Tumor growth inhibition rate (TGI, %) (1-T (tumor volume of treatment group)/C (tumor volume of solvent control group)) ⁇ 100.
- Relative tumor proliferation rate (T/C, %) RTV of treatment group/RTV of solvent control group.
- the data in Table 9 shows that the groups of WWZ (25.0mg/kg), test drug (8.1mg/kg), test drug (16.2mg/kg), WWZ (25.0mg/kg) + test drug were administered intragastrically. (8.1mg/kg), WWZ (25.0mg/kg) + test drug (16.2mg/kg) group, on D34, except for the WWZ (25.0mg/kg) group, the tumor growth inhibition rates of the other treatment groups (according to tumor Volume and tumor weight) were both greater than 60%, and T/C were both less than 40%.
- test drug 8 mg/kg
- test drug 16.2mg/kg
- WWZ 25.0mg/kg
- test drug 8.1mg/kg
- WWZ 25.0mg/kg
- references to the terms “one embodiment,” “some embodiments,” “an example,” “specific examples,” or “some examples” or the like means that specific features are described in connection with the embodiment or example. , structures, materials or features are included in at least one embodiment or example of the invention. In this specification, the schematic expressions of the above terms are not necessarily directed to the same embodiment or example. Furthermore, the specific features, structures, materials or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, those skilled in the art may combine and combine different embodiments or examples and features of different embodiments or examples described in this specification unless they are inconsistent with each other.
Landscapes
- Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
Abstract
Description
本发明涉及医药领域,具体地,本发明涉及受体酪氨酸激酶抑制剂与联苯环辛二稀木脂素的联合用药及其用途,更具体地,本发明涉及一种药物组合物,单剂型,药物联合,药盒,式Ⅰ所示化合物或其盐的给药方法,延长式Ⅰ所示化合物或其盐半衰期的方法,降低式Ⅰ所示化合物或其盐生成式Ⅱ所示化合物的方法。The present invention relates to the field of medicine. Specifically, the present invention relates to the combined use of receptor tyrosine kinase inhibitors and biphenylcyclooctadiene lignans and their uses. More specifically, the present invention relates to a pharmaceutical composition, Single dosage form, drug combination, kit, administration method of the compound represented by formula I or its salt, method of extending the half-life of the compound represented by formula I or its salt, reducing the compound represented by formula I or its salt to form the compound represented by formula II Methods.
(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯及其盐不局限于口服,可以抑制带有活化突变的表皮生长因子受体酪氨酸激酶活性的抑制剂(EGFR-TKI),最初开发用于治疗有中枢神经系统转移(CNS)的非小细胞肺癌(NSCLC)。(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯及其盐在人体内代谢的主要CYP450酶是CYP3A4和CYP3A5亚型,其药效随着用药剂量的升高而升高,但不良反应的出现限制了其在临床上的进一步应用,不良反应的出现可能与经CYP450酶,如其亚型CYP3A代谢酶作用后的代谢产物的生成有关,部分代谢产物的生成量以高于给药剂量的比例增加。现有研究结果显示,(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐可以100%浓度的形式透过血脑屏障,在代谢过程中生成的同样具有活性的代谢产物穿透血脑屏障的能力显著低于该药物,使得颅内药效降低。此外,不同的CYP450酶抑制剂与(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯或其盐联合时可能产生药物间相互作用,因此,仍需研究探索一种能够提高(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯及其盐的药效,且安全可靠的用药方法。(R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate and its salts are not An inhibitor (EGFR-TKI) that is limited to oral administration and can inhibit the activity of epidermal growth factor receptor tyrosine kinase with activating mutations, initially developed for the treatment of non-small cell lung cancer (NSCLC) with central nervous system metastasis (CNS) ). (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate and its salts in The main CYP450 enzymes metabolized in the human body are CYP3A4 and CYP3A5 subtypes. Their drug efficacy increases with the increase in drug dosage. However, the occurrence of adverse reactions limits its further clinical application. The occurrence of adverse reactions may be related to the regular use. CYP450 enzymes, such as its subtype CYP3A metabolizing enzyme, are related to the generation of metabolites, and the generation of some metabolites increases at a higher rate than the administered dose. Existing research results show that (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylic The acid ester hydrochloride can penetrate the blood-brain barrier in the form of 100% concentration. The ability of the equally active metabolites generated during metabolism to penetrate the blood-brain barrier is significantly lower than that of the drug, resulting in reduced intracranial efficacy. Furthermore, different CYP450 enzyme inhibitors have been compared with (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine- Drug-drug interactions may occur when 1-carboxylic acid esters or their salts are combined. Therefore, research is still needed to explore a method that can improve (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxy. It is a safe and reliable method of medication.
发明内容Contents of the invention
本申请是基于发明人对下列问题和事实的发现而提出的:This application is filed based on the inventor's discovery of the following issues and facts:
(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯或其盐是一种有效的可渗透中枢神经系统的EGFR抑制剂,联苯环辛二稀木脂素是CYP450酶抑制剂,且具有降低血清谷丙转氨酶,起到保肝护肝的作用。发明人研究(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯或其盐单用或与多种CYP450酶抑制剂联用,如五味子甲素、水飞蓟宾、酮康唑等,发现五味 子甲素相较于其它种类的抑制剂对人肝微粒体细胞色素CYP450酶有明显的抑制作用,且抑制程度适中,相较于(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯或其盐单用,联用后联苯环辛二稀木脂素通过抑制CYP450酶而降低(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯或其盐的代谢,延长其在体内的滞留时间而增强疗效,同时通过抑制CYP450细胞色素代谢酶进而减少代谢产物的浓度或含量,药效和安全性均得到提高。(R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate or its salt is An effective EGFR inhibitor that can penetrate the central nervous system, biphenylcyclooctadienol is a CYP450 enzyme inhibitor and can reduce serum alanine aminotransferase and play a role in protecting the liver. The inventor studied (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate or Its salt is used alone or in combination with various CYP450 enzyme inhibitors, such as schisandrin A, silibinin, ketoconazole, etc. It is found that schisandra Compared with other types of inhibitors, promethin has a significant inhibitory effect on human liver microsomal cytochrome CYP450 enzymes, and the degree of inhibition is moderate. Compared with (R)-4-(3-chloro-2-fluorophenyl) Amino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate or its salt is used alone. After combined use, the passage of biphenylcyclooctadiene lignans is inhibited. (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate by reducing CYP450 enzymes Or the metabolism of its salts, prolonging its residence time in the body to enhance the efficacy, and at the same time reducing the concentration or content of metabolites by inhibiting CYP450 cytochrome metabolic enzymes, thus improving both efficacy and safety.
在本发明的的第一方面,本发明提出了一种药物组合物。根据本发明的实施例,所述药物组合物包括:式Ⅰ所示化合物或其盐和联苯环辛二稀木脂素。根据本发明实施例的所述药物组合物能够降低血清中谷丙转氨酶的含量,对肝脏起到保护作用,且相较于单独使用式Ⅰ所示化合物或其盐,所述药物组合物可以有效延长式Ⅰ所示化合物或其盐在体内的药效时间,并显著降低代谢过程中式Ⅱ所示代谢物的浓度,提高式Ⅰ所示化合物或其盐在体内的暴露量和脑组织中的剂量,降低式Ⅱ所示代谢产物引起的不良反应,使得式Ⅰ所示化合物或其盐的药效和安全性均得到提高,从而有效治疗或预防癌症。In a first aspect of the invention, the invention provides a pharmaceutical composition. According to an embodiment of the present invention, the pharmaceutical composition includes: a compound represented by Formula I or a salt thereof and biphenylcyclooctadienolignan. The pharmaceutical composition according to the embodiment of the present invention can reduce the content of alanine aminotransferase in serum, play a protective role on the liver, and compared with the single use of the compound represented by formula I or its salt, the pharmaceutical composition can effectively prolong the The duration of drug efficacy of the compound represented by formula I or its salt in the body, and significantly reducing the concentration of the metabolite represented by formula II during the metabolic process, increasing the exposure of the compound represented by formula I or its salt in the body and the dose in brain tissue, Reducing the adverse reactions caused by the metabolites represented by formula II improves the efficacy and safety of the compounds represented by formula I or salts thereof, thereby effectively treating or preventing cancer.
根据本发明的实施例,上述药物组合物还可以进一步包括如下附加技术特征中的至少之一:According to embodiments of the present invention, the above-mentioned pharmaceutical composition may further include at least one of the following additional technical features:
根据本发明的实施例,所述式Ⅰ所示化合物的盐包括下列中的至少之一:硫酸盐、磷酸盐、柠檬酸盐、酒石酸盐、富马酸盐、苯甲酸盐、己二酸盐、琥珀酸盐、甲磺酸盐和马来酸盐。According to an embodiment of the present invention, the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为盐酸盐。According to an embodiment of the present invention, the salt of the compound represented by Formula I is a hydrochloride.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐。According to embodiments of the present invention, the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
根据本发明的实施例,所述联苯环辛二稀木脂素包括下列中的至少之一:五味子甲素、五味子酯甲、五味子醇已、五味子醇甲、五味子乙素和五味子酚。According to an embodiment of the present invention, the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
根据本发明的实施例,所述联苯环辛二稀木脂素为五味子甲素。According to an embodiment of the present invention, the biphenylcyclooctadiene lignan is schisandrin A.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:31)~(4:3)。当式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:31)~(4:3)时,所述式Ⅰ所示化合物或其盐的半衰期得到延长,代谢过程中式Ⅱ所示代谢物的含量低于单独使用式Ⅰ所示化合物或其盐,具备较强的抗肿瘤能力和较高的安全性。根据本申请实施例的描述,小鼠与人类给药剂量之间可以进行换算,采用该比例换算后的数值添加上述两种物质能够有效地保护人类免于或者缓解肿瘤。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3). When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3), the half-life of the compound represented by formula I or its salt is extended. , the content of the metabolite shown in Formula II during the metabolic process is lower than that of the compound shown in Formula I or its salt alone, and it has strong anti-tumor ability and high safety. According to the description of the embodiments of this application, the dosages for mice and humans can be converted, and adding the above two substances using the converted values can effectively protect humans from or alleviate tumors.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:10)~(5:6)。当式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:10)~(5:6)时,所述式Ⅰ所示化合物或其盐的半衰期得到显著延长,代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,具备较强的抗肿瘤能力和较高的安全性。根据本申请实施例的描述,小鼠与人类给药剂量之间可以进行换算,采用该比例换算后的数值添加上述两种物质能够有效地保护人类免于或者缓解肿瘤。 According to an embodiment of the present invention, the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6). When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6), the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite shown in formula II during the metabolic process is significantly lower than that of the compound shown in formula I or its salt alone, and it has strong anti-tumor ability and high safety. According to the description of the embodiments of this application, the dosages for mice and humans can be converted, and adding the above two substances using the converted values can effectively protect humans from or alleviate tumors.
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:50)~(4:5)。当式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:50)~(4:5)时,所述式Ⅰ所示化合物或其盐的半衰期得到显著延长,代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,所述药物组合物具备很强的抗肿瘤能力和较高的安全性。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5). When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5), the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite represented by formula II during the metabolic process is significantly lower than that of the compound represented by formula I or its salt alone, and the pharmaceutical composition has strong anti-tumor ability and high safety.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81:125。式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81:125时,所述式Ⅰ所示化合物或其盐的半衰期得到显著延长,代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,所述药物组合物具备很强的抗肿瘤能力和较高的安全性。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125. When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125, the compound represented by formula I or its salt is The half-life is significantly extended, and the content of the metabolite represented by formula II during metabolism is significantly lower than when the compound represented by formula I or its salt is used alone. The pharmaceutical composition has strong anti-tumor ability and high safety.
根据本发明的实施例,所述药物组合物为口服制剂。所述药物组合物可以是服用制剂、注射制剂、丸剂、缓控释制剂、植入剂或气雾剂等类型,并不受特别限制,根据本发明的具体实施例,当所述药物组合物为口服制剂时所述单剂型的半衰期更长,代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,有利于提高式Ⅰ所示化合物或其盐的透脑剂量,进一步提高药效,因此,所述药物组合物具备更强的抗肿瘤能力和较高的安全性。According to an embodiment of the present invention, the pharmaceutical composition is an oral preparation. The pharmaceutical composition may be in the form of taking preparations, injection preparations, pills, sustained-release preparations, implants or aerosols, etc., and is not particularly limited. According to specific embodiments of the present invention, when the pharmaceutical composition When it is an oral preparation, the half-life of the single dosage form is longer, and the content of the metabolite shown in Formula II during metabolism is significantly lower than when the compound shown in Formula I or its salt is used alone, which is beneficial to improving the penetration of the compound shown in Formula I or its salt. The brain dose further improves the drug efficacy. Therefore, the pharmaceutical composition has stronger anti-tumor ability and higher safety.
根据本发明的一些具体实施例,所述药物组合物包括本文提供的式Ⅰ所示化合物或其盐和五味子甲素以及药学上可接受的载体。“药学上可接受的载体”可以包括生理学上相容的任何和所有溶剂、分散介质、包衣、抗细菌剂和抗真菌剂、等渗剂和延迟吸收剂等等。具体实例可以是水、盐水、磷酸盐缓冲盐水、葡萄糖、甘油、乙醇等以及它们的组合物中的一种或多种。有许多情况下,药物组合物中包括等渗剂,例如糖类、多元醇(如甘露醇、山梨醇)或氯化钠等。当然药学上可接受的载体还可包括微量的辅助物质,例如润湿剂或乳化剂、防腐剂或缓冲剂,用来延长抗体的保存限期或效力。According to some specific embodiments of the present invention, the pharmaceutical composition includes the compound represented by Formula I or a salt thereof provided herein, and Schisandrin A, as well as a pharmaceutically acceptable carrier. "Pharmaceutically acceptable carrier" may include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Specific examples may be one or more of water, saline, phosphate buffered saline, glucose, glycerin, ethanol, etc., and combinations thereof. In many cases, pharmaceutical compositions include isotonic agents, such as sugars, polyols (such as mannitol, sorbitol) or sodium chloride. Of course, pharmaceutically acceptable carriers may also include trace amounts of auxiliary substances, such as wetting agents or emulsifiers, preservatives or buffers, to extend the shelf life or effectiveness of the antibody.
例如,本发明的式Ⅰ所示化合物或其盐和联苯环辛二稀木脂素可掺入适用于胃肠外施用(例如静脉内、皮下、腹膜内、肌肉内)的药物组合物中。这些药物组合物可以被制备成各种形式。例如液体、半固体和固体剂型等,包括但不限于液体溶液(例如,注射溶液和输注溶液)、分散剂或悬浮剂、片剂、丸剂、粉末、脂质体和栓剂。典型的药物组合物为口服制剂、注射溶液或输注溶液形式。所述药物组合物可通过口服、静脉输注或注射或肌肉内或皮下注射来施用。For example, the compound represented by formula I of the present invention or a salt thereof and biphenylcyclooctadienolignan can be incorporated into a pharmaceutical composition suitable for parenteral administration (such as intravenous, subcutaneous, intraperitoneal, intramuscular) . These pharmaceutical compositions can be prepared in various forms. Examples include liquid, semi-solid, and solid dosage forms, including, but not limited to, liquid solutions (eg, injection solutions and infusion solutions), dispersions or suspensions, tablets, pills, powders, liposomes, and suppositories. Typical pharmaceutical compositions are in the form of oral preparations, injection solutions, or infusion solutions. The pharmaceutical composition may be administered orally, intravenously, or by injection, or intramuscularly or subcutaneously.
在本发明的第二方面,本发明提出了一种单剂型。根据本发明的实施例,包括:式Ⅰ所示化合物或其盐和联苯环辛二稀木脂素。发明人发现联苯环辛二稀木脂素,如五味子甲素不仅具有保肝护肝的功能,且对肝微粒体细胞色素CYP450酶有明显的抑制作用,进一步地,通过抑制CYP450酶而降低式Ⅰ所示化合物或其盐的代谢,延长其在体内的滞留时间而增强疗效,同时减少同时刻血清中代谢反应产生的式Ⅱ所示代谢物的浓度或含量,提高式Ⅰ所示化合物或其盐在体内的暴露量和脑组织中的剂量,降低式Ⅱ所示代谢产物引起的不良反应,使得式Ⅰ所示化合物或其盐的药效和安全性均得到提高,因此,所述单剂型具备较强的抗肿瘤能力和较高的安全性。根据本申请实施例的描述,通过小鼠与人类给药剂量的换 算,采用该比例进行换算能够有效地保护人类免于或者缓解肿瘤。In a second aspect of the invention, a single dosage form is provided. According to the embodiment of the present invention, it includes: the compound represented by formula I or its salt and biphenylcyclooctadiene lignan. The inventor found that biphenylcyclooctadiene lignans, such as Schisandrin A, not only have the function of protecting the liver, but also have a significant inhibitory effect on liver microsomal cytochrome CYP450 enzymes. Furthermore, by inhibiting CYP450 enzymes, they reduce The metabolism of the compound represented by formula I or its salt prolongs its residence time in the body and enhances the therapeutic effect. At the same time, it reduces the concentration or content of the metabolite represented by formula II produced by the metabolic reaction in the serum at the same time, and increases the concentration or content of the compound represented by formula I or its salt. The exposure of its salt in the body and the dose in brain tissue reduce the adverse reactions caused by the metabolites shown in Formula II, so that the efficacy and safety of the compound shown in Formula I or its salt are improved. Therefore, the single The dosage form has strong anti-tumor ability and high safety. According to the description of the embodiments of this application, through the exchange of dosage between mice and humans, Calculation, using this ratio for conversion can effectively protect humans from or alleviate tumors.
根据本发明的实施例,上述单剂型还可以进一步包括如下附加技术特征中的至少之一:According to embodiments of the present invention, the above-mentioned single dosage form may further include at least one of the following additional technical features:
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:31)~(4:3)。According to embodiments of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3).
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:10)~(5:6)。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6).
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:50)~(4:5)。当式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:50)~(4:5)时,所述单剂型的半衰期得到显著延长,代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,进而提高式Ⅰ所示化合物或其盐的透脑剂量,提高药效,因此,所述单剂型具备很强的抗肿瘤能力和较高的安全性。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5). When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5), the half-life of the single dosage form is significantly extended, and the metabolism process of formula II The content of the metabolites shown is significantly lower than that of the compound shown in formula I or its salt alone, thereby increasing the brain-penetrating dose of the compound shown in formula I or its salt and improving the efficacy. Therefore, the single dosage form has strong anti-bacterial activity. tumor capability and higher safety.
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81:125。式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81:125时,所述单剂型的半衰期得到显著延长,代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,进而提高式Ⅰ所示化合物或其盐的透脑剂量,提高药效,因此,所述单剂型具备很强的抗肿瘤能力和较高的安全性。According to an embodiment of the present invention, the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125. When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125, the half-life of the single dosage form is significantly extended, and the metabolism In the process, the content of the metabolite represented by formula II is significantly lower than that of the compound represented by formula I or its salt alone, thereby increasing the brain-penetrating dose of the compound represented by formula I or its salt and improving the drug efficacy. Therefore, the single dosage form has very high Strong anti-tumor ability and high safety.
根据本发明的实施例,所述单剂型包括200~300mg所述式Ⅰ所示化合物或其盐。根据本申请实施例的描述,上述单剂型每天可以使用2次、3次或4次,优选为2次,通过小鼠与人类给药剂量的换算,采用该剂量进行换算后能够有效地保护人类免于或者缓解肿瘤。本领域技术人员可以理解,所述单剂型的规格不受特别限制,例如,可以根据不同目标人群特点设定所述单剂型的规格,例如,按照目标人群的体重进行设定,如体重为40kg、60kg、70kg、80kg、90kg、100kg等,或,根据本申请所述的式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比进行设定,如3:20,1:6,5:6,4:5,81:250或81:125等,再或者,可以按照目标人群的特点和所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比进行设定。According to an embodiment of the present invention, the single dosage form includes 200 to 300 mg of the compound represented by Formula I or a salt thereof. According to the description of the embodiments of this application, the above-mentioned single dosage form can be used 2 times, 3 times or 4 times a day, preferably 2 times. Through the conversion of dosages for mice and humans, this dosage can effectively protect humans after conversion. Avoid or alleviate tumors. Those skilled in the art can understand that the specifications of the single dosage form are not particularly limited. For example, the specifications of the single dosage form can be set according to the characteristics of different target groups. For example, the specifications of the single dosage form can be set according to the weight of the target group, such as a body weight of 40kg. , 60kg, 70kg, 80kg, 90kg, 100kg, etc., or set according to the mass ratio of the compound represented by Formula I or its salt described in this application to biphenylcyclooctadiene lignan, such as 3:20, 1:6, 5:6, 4:5, 81:250 or 81:125, etc. Or, the compound of formula I or its salt and biphenylcyclooctadienyl lignin can be used according to the characteristics of the target population and the Set the mass ratio of elements.
根据本发明的实施例,所述单剂型为口服制剂。所述单剂型可以是服用制剂、注射制剂、丸剂、缓控释制剂、植入剂或气雾剂等类型,并不受特别限制,根据本发明的具体实施例,当所述单剂型为口服制剂时所述单剂型的半衰期更长,代谢过程中代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,进而提高式Ⅰ所示化合物或其盐的透脑剂量,提高药效,因此,所述单剂型具备更强的抗肿瘤能力和较高的安全性。According to an embodiment of the present invention, the single dosage form is an oral preparation. The single dosage form may be an oral preparation, an injection preparation, a pill, a sustained-release preparation, an implant or an aerosol, and is not particularly limited. According to specific embodiments of the present invention, when the single dosage form is oral The half-life of the single dosage form is longer during preparation, and the content of the metabolite represented by formula II during the metabolism process is significantly lower than that of the compound represented by formula I or its salt alone, thereby increasing the penetration of the compound represented by formula I or its salt. Brain dose improves drug efficacy. Therefore, the single dosage form has stronger anti-tumor ability and higher safety.
在本发明的第三方面,本发明提出了一种药物联合。根据本发明的实施例,包括:式Ⅰ所示化合物或其盐作为第一活性成分和联苯环辛二稀木脂素作为第二活性成分。如前所述,发明人发现五味子甲素不仅具有保肝护肝的功能,且可以通过抑制CYP450酶,如亚型CYP3A而降低式Ⅰ所示化合物或其盐的代谢,延长其在体内的滞留时间而增强疗效,同时减少同时刻血清中脱甲基反应产生的式Ⅱ所示代谢 物,提高式Ⅰ所示化合物或其盐在体内的暴露量和脑组织中的剂量,降低式Ⅱ所示代谢产物引起的不良反应,使得式Ⅰ所示化合物或其盐的药效和安全性均得到提高,因此,根据本发明实施例的药物联合可以显著延长所述式Ⅰ所示化合物或其盐的半衰期及透脑剂量,可以有效的抑制肿瘤生长,并具有较高的安全性。In a third aspect of the invention, the invention proposes a drug combination. According to an embodiment of the present invention, the compound of formula I or a salt thereof is included as the first active ingredient and biphenylcyclooctadiene lignan as the second active ingredient. As mentioned above, the inventor found that Schisandrin A not only has the function of protecting the liver, but also can reduce the metabolism of the compound represented by Formula I or its salt by inhibiting CYP450 enzymes, such as subtype CYP3A, and prolong its retention in the body. time to enhance the therapeutic effect, and at the same time reduce the metabolism of formula II produced by the demethylation reaction in the serum at the same time It can increase the exposure of the compound represented by formula I or its salt in the body and the dose in brain tissue, reduce the adverse reactions caused by the metabolite represented by formula II, and improve the efficacy and safety of the compound represented by formula I or its salt. Therefore, the drug combination according to the embodiment of the present invention can significantly extend the half-life and brain-penetrating dose of the compound represented by Formula I or its salt, can effectively inhibit tumor growth, and has high safety.
根据本发明的实施例,上述药物联合还可以进一步包括如下附加技术特征中的至少之一:According to embodiments of the present invention, the above-mentioned drug combination may further include at least one of the following additional technical features:
根据本发明的实施例,所述式Ⅰ所示化合物的盐包括下列中的至少之一:硫酸盐、磷酸盐、柠檬酸盐、酒石酸盐、富马酸盐、苯甲酸盐、己二酸盐、琥珀酸盐、甲磺酸盐和马来酸盐。According to an embodiment of the present invention, the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为盐酸盐。According to an embodiment of the present invention, the salt of the compound represented by Formula I is a hydrochloride.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐。According to embodiments of the present invention, the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
根据本发明的实施例,所述联苯环辛二稀木脂素包括下列中的至少之一:五味子甲素、五味子酯甲、五味子醇已、五味子醇甲、五味子乙素和五味子酚。According to an embodiment of the present invention, the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
根据本发明的实施例,所述联苯环辛二稀木脂素为五味子甲素。According to an embodiment of the present invention, the biphenylcyclooctadiene lignan is schisandrin A.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:31)~(4:3)。当式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:31)~(4:3)时,所述式Ⅰ所示化合物或其盐的半衰期得到延长,代谢过程中式Ⅱ所示代谢物的含量低于单独使用式Ⅰ所示化合物或其盐,具备较强的抗肿瘤能力和较高的安全性。根据本申请实施例的描述,小鼠与人类给药剂量之间可以进行换算,采用该比例换算后的数值添加上述两种物质能够有效地保护人类免于或者缓解肿瘤。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3). When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3), the half-life of the compound represented by formula I or its salt is extended. , the content of the metabolite shown in Formula II during the metabolic process is lower than that of the compound shown in Formula I or its salt alone, and it has strong anti-tumor ability and high safety. According to the description of the embodiments of this application, the dosages for mice and humans can be converted, and adding the above two substances using the converted values can effectively protect humans from or alleviate tumors.
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:10)~(5:6)。当式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:10)~(5:6)时,所述式Ⅰ所示化合物或其盐的半衰期得到显著延长,代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,进而提高式Ⅰ所示化合物或其盐的透脑剂量,提高药效,药物联合后所述式Ⅰ所示化合物或其盐具备更强的抗肿瘤能力和更高的安全性。根据本申请实施例的描述,通过小鼠与人类给药剂量的换算,采用该比例进行换算能够有效地保护人类免于或者缓解肿瘤。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6). When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6), the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite shown in formula II during the metabolic process is significantly lower than that of the compound shown in formula I or its salt alone, thereby increasing the brain-penetrating dose of the compound shown in formula I or its salt, and improving the drug efficacy, as described after drug combination. The compound represented by formula I or its salt has stronger anti-tumor ability and higher safety. According to the description of the embodiments of the present application, by converting the dosage between mice and humans, using this ratio can effectively protect humans from or alleviate tumors.
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:50)~(4:5)。当式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:50)~(4:5)时,所述式Ⅰ所示化合物或其盐的半衰期得到显著延长,式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,进而提高式Ⅰ所示化合物或其盐的透脑剂量,提高药效,药物联合后所述式Ⅰ所示化合物或其盐具备更强的抗肿瘤能力和更高的安全性。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5). When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5), the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite represented by formula II is significantly lower than that of the compound represented by formula I or its salt alone, thereby increasing the brain-penetrating dose of the compound represented by formula I or its salt, and improving the drug efficacy. After drug combination, formula I is described The indicated compound or its salt has stronger anti-tumor ability and higher safety.
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81:125。式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81: 125时,所述式Ⅰ所示化合物或其盐的半衰期得到显著延长,式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,进而提高式Ⅰ所示化合物或其盐的透脑剂量,提高药效,药物联合后所述式Ⅰ所示化合物或其盐具备更强的抗肿瘤能力和更高的安全性。According to an embodiment of the present invention, the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125. The mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81: When 125, the half-life of the compound represented by formula I or its salt is significantly extended, and the content of the metabolite represented by formula II is significantly lower than that of the compound represented by formula I or its salt alone, thereby increasing the concentration of the compound represented by formula I or its salt. The brain-penetrating dose of the salt can improve the drug efficacy. After drug combination, the compound represented by formula I or its salt has stronger anti-tumor ability and higher safety.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐和所述联苯环辛二稀木脂素共同配制或分开配制。According to an embodiment of the present invention, the compound represented by Formula I or its salt and the biphenylcyclooctadiene lignan are formulated together or separately.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐和所述联苯环辛二稀木脂素同时使用或分开使用。According to an embodiment of the present invention, the compound represented by formula I or its salt and the biphenylcyclooctadiene lignan are used simultaneously or separately.
需要注意的是,所述药物联合包括在时间和/或空间上分开的联合,只要式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素能够共同作用以实现本发明的目的。例如,所述药物联合中所含的成分可以以整体施用于受试者或待测样本,或者分开施用于受试者或待测样本。当所述药物联合中所含的成分分开地施用于受试者或待测样本时,各个成分可以同时或依次施用于受试者或待测样本。It should be noted that the drug combination includes combinations that are separated in time and/or space, as long as the compound represented by formula I or its salt and biphenylcyclooctadienolignan can cooperate to achieve the purpose of the present invention. For example, the components contained in the pharmaceutical combination can be administered to the subject or the sample to be tested as a whole, or separately. When the components contained in the pharmaceutical combination are administered to the subject or sample to be tested separately, the individual components may be administered to the subject or sample to be tested simultaneously or sequentially.
根据本发明的实施例,所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药。本领域技术人员可以理解,所述联苯环辛二稀木脂素,和所述式Ⅰ所示化合物或其盐的给药顺序不受特别限制,只要所述联苯环辛二稀木脂素能够在所述式Ⅰ所示化合物或其盐的代谢周期内发挥作用即可,例如,所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药,或所述联苯环辛二稀木脂素后于所述式Ⅰ所示化合物或其盐给药,或所述联苯环辛二稀木脂素与所述式Ⅰ所示化合物或其盐同时给药均在本申请的保护范围内。根据本发明的一些具体实施例,当所述联苯环辛二稀木脂素先于所述(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐给药时,所述式Ⅰ所示化合物或其盐的半衰期可以得到显著延长,且其代谢过程中产生的式Ⅱ所示代谢物的含量显著降低,使得式所示化合物或其盐具有更高的透脑率,使得透脑剂量增多,进一步提高药效,所述药物联合具有良好的抗肿瘤效果和安全性。According to an embodiment of the present invention, the biphenylcyclooctadienolignan is administered prior to the compound represented by Formula I or a salt thereof. Those skilled in the art can understand that the order of administration of the biphenylcyclooctadienol and the compound represented by formula I or its salt is not particularly limited, as long as the diphenylcyclooctadienol It is sufficient that the compound can play a role in the metabolic cycle of the compound represented by formula I or its salt. For example, the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt, Or the biphenylcyclooctadiene lignan is administered after the compound represented by formula I or its salt, or the biphenylcyclooctadienolignan and the compound represented by formula I or its salt are administered Simultaneous administration is within the scope of protection of this application. According to some specific embodiments of the present invention, when the biphenylcyclooctadienolignan precedes the (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquin When oxazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride is administered, the half-life of the compound represented by formula I or its salt can be significantly extended, and during its metabolism The content of the metabolite shown in the formula II produced is significantly reduced, so that the compound shown in the formula or its salt has a higher brain penetration rate, which increases the brain penetration dose and further improves the drug efficacy. The drug combination has a good anti-tumor effect and security.
根据本发明的实施例,所述第二活性成分为口服剂型。所述第二活性成分可以是服用制剂、注射制剂、丸剂、缓控释制剂、植入剂或气雾剂等类型,并不受特别限制,根据本发明的具体实施例,当所述第二活性成分为口服制剂时所述单剂型的半衰期更长,代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,所述式Ⅰ所示化合物或其盐具备更强的抗肿瘤能力和较高的安全性。According to an embodiment of the invention, the second active ingredient is in an oral dosage form. The second active ingredient may be in the form of taking preparations, injection preparations, pills, sustained-release preparations, implants or aerosols, etc., and is not particularly limited. According to specific embodiments of the present invention, when the second active ingredient When the active ingredient is an oral preparation, the half-life of the single dosage form is longer, and the content of the metabolite shown in Formula II during metabolism is significantly lower than when the compound shown in Formula I or its salt is used alone, and the compound shown in Formula I or its salt has Stronger anti-tumor ability and higher safety.
在本发明的第四方面,本发明提出了一种药盒。根据本发明的实施例,所述药盒包括:式Ⅰ所示化合物或其盐作为第一活性成分和联苯环辛二稀木脂素作为第二活性成分。如前所述,发明人发现联苯环辛二稀木脂素通过抑制CYP450酶,如其亚型CYP3A酶而降低式Ⅰ所示化合物或其盐的代谢,延长其在体内的滞留时间而增强疗效,同时减少同时刻血清中式Ⅱ所示代谢物的浓度或含量,提高式Ⅰ所示化合物或其盐在体内的暴露量和脑组织中的剂量,降低式Ⅱ所示代谢产物引起的不良反应,使得式Ⅰ所示化合物或其盐的药效和安全性均得到提高,可以有效的抑制肿瘤生长,并具有较高的安全性。 In a fourth aspect of the invention, the invention provides a medicine box. According to an embodiment of the present invention, the pharmaceutical kit includes: a compound represented by Formula I or a salt thereof as the first active ingredient and biphenylcyclooctadienolignan as the second active ingredient. As mentioned above, the inventor found that biphenylcyclooctadienolignan reduces the metabolism of the compound represented by formula I or its salt by inhibiting CYP450 enzymes, such as its subtype CYP3A enzyme, prolongs its residence time in the body and enhances the therapeutic effect. , while reducing the concentration or content of the metabolite represented by formula II in the serum at the same time, increasing the exposure of the compound represented by formula I or its salt in the body and the dose in brain tissue, and reducing the adverse reactions caused by the metabolite represented by formula II, The efficacy and safety of the compound represented by formula I or its salt are improved, and the compound can effectively inhibit tumor growth and has high safety.
根据本发明的实施例,上述药盒还可以进一步包括如下附加技术特征中的至少之一:According to an embodiment of the present invention, the above-mentioned medicine box may further include at least one of the following additional technical features:
根据本发明的实施例,所述式Ⅰ所示化合物的盐包括下列中的至少之一:硫酸盐、磷酸盐、柠檬酸盐、酒石酸盐、富马酸盐、苯甲酸盐、己二酸盐、琥珀酸盐、甲磺酸盐和马来酸盐。According to an embodiment of the present invention, the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为盐酸盐。According to an embodiment of the present invention, the salt of the compound represented by Formula I is a hydrochloride.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐。According to embodiments of the present invention, the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
根据本发明的实施例,所述联苯环辛二稀木脂素包括下列中的至少之一:五味子甲素、五味子酯甲、五味子醇已、五味子醇甲、五味子乙素和五味子酚。According to an embodiment of the present invention, the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
根据本发明的实施例,所述联苯环辛二稀木脂素为五味子甲素。According to an embodiment of the present invention, the biphenylcyclooctadiene lignan is schisandrin A.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:31)~(4:3)。当式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:31)~(4:3)时,所述式Ⅰ所示化合物或其盐的半衰期得到延长,代谢过程中式Ⅱ所示代谢物的含量低于单独使用式Ⅰ所示化合物或其盐,具备较强的抗肿瘤能力和较高的安全性。根据本申请实施例的描述,小鼠与人类给药剂量之间可以进行换算,采用该比例换算后的数值添加上述两种物质能够有效地保护人类免于或者缓解肿瘤。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3). When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3), the half-life of the compound represented by formula I or its salt is extended. , the content of the metabolite shown in Formula II during the metabolic process is lower than that of the compound shown in Formula I or its salt alone, and it has strong anti-tumor ability and high safety. According to the description of the embodiments of this application, the dosages for mice and humans can be converted, and adding the above two substances using the converted values can effectively protect humans from or alleviate tumors.
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:10)~(5:6)。当式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:10)~(5:6)时,所述式Ⅰ所示化合物或其盐的半衰期得到显著延长,代谢过程中式Ⅱ所示代谢物代谢产物的含量显著低于单独使用式Ⅰ所示化合物或其盐,具备较强的抗肿瘤能力和较高的安全性。根据本申请实施例的描述,小鼠与人类给药剂量之间可以进行换算,采用该比例换算后的数值添加上述两种物质能够有效地保护人类免于或者缓解肿瘤。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6). When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (1:10) to (5:6), the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite of the metabolite shown in formula II during the metabolic process is significantly lower than that of the compound shown in formula I or its salt alone, and it has strong anti-tumor ability and high safety. According to the description of the embodiments of this application, the dosages for mice and humans can be converted, and adding the above two substances using the converted values can effectively protect humans from or alleviate tumors.
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:50)~(4:5)。当式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:50)~(4:5)时,所述式Ⅰ所示化合物或其盐的半衰期得到显著延长,代谢过程中式Ⅱ所示代谢物代谢产物的含量显著低于单独使用式Ⅰ所示化合物或其盐,所述药盒具备很强的抗肿瘤能力和较高的安全性。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5). When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is (7:50) to (4:5), the half-life of the compound represented by formula I or its salt is significantly improved. Prolonged, the content of the metabolite of the metabolite shown in Formula II during the metabolic process is significantly lower than that of the compound shown in Formula I or its salt alone, and the kit has strong anti-tumor ability and high safety.
根据本发明的实施例,式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81:125。式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81:125时,所述式Ⅰ所示化合物或其盐的半衰期得到显著延长,代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,所述药物组合物具备很强的抗肿瘤能力和较高的安全性。According to an embodiment of the present invention, the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125. When the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan is 3:20, 1:5, 81:250 or 81:125, the compound represented by formula I or its salt is The half-life is significantly extended, and the content of the metabolite represented by formula II during metabolism is significantly lower than when the compound represented by formula I or its salt is used alone. The pharmaceutical composition has strong anti-tumor ability and high safety.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐和所述联苯环辛二稀木脂素共同配制或分开配制。本领域技术人员可以理解,所述式Ⅰ所示化合物或其盐和所述联苯环辛二稀木脂素的配制方法不受特别限制,分开配制或共同配制均可。 According to an embodiment of the present invention, the compound represented by Formula I or its salt and the biphenylcyclooctadiene lignan are formulated together or separately. Those skilled in the art can understand that the preparation method of the compound represented by formula I or its salt and the biphenylcyclooctadiene lignan is not particularly limited, and they can be prepared separately or together.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐和所述联苯环辛二稀木脂素同时使用或分开使用。需要注意的是,所述方法中所述式Ⅰ所示化合物或其盐和所述联苯环辛二稀木脂素包括在时间和/或空间上分开使用,只要式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素盐能够共同作用以实现本发明的目的。例如,式Ⅰ所示化合物或其盐和所述联苯环辛二稀木脂素可以以整体施用于受试者或待测样本,或者分开施用于受试者或待测样本。当所述式Ⅰ所示化合物或其盐和所述联苯环辛二稀木脂素分开地施用于受试者或待测样本时,各个成分可以同时或依次施用于受试者或待测样本。According to an embodiment of the present invention, the compound represented by formula I or its salt and the biphenylcyclooctadiene lignan are used simultaneously or separately. It should be noted that in the method, the compound represented by formula I or its salt and the biphenylcyclooctadiene lignan are used separately in time and/or space, as long as the compound represented by formula I or its salt is used separately in time and/or space. The salt and biphenylcyclooctadiene lignan salt can cooperate to achieve the purpose of the present invention. For example, the compound represented by Formula I or a salt thereof and the biphenylcyclooctadienolignan can be administered to the subject or the sample to be tested as a whole, or separately to the subject or the sample to be tested. When the compound represented by Formula I or a salt thereof and the biphenylcyclooctadienolignan are administered to a subject or a sample to be tested separately, each component may be administered to the subject or sample to be tested simultaneously or sequentially. sample.
根据本发明的实施例,所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药。本领域技术人员可以理解,所述联苯环辛二稀木脂素,和所述式Ⅰ所示化合物或其盐的给药顺序不受特别限制,只要所述联苯环辛二稀木脂素能够在所述式Ⅰ所示化合物或其盐的代谢周期内发挥作用即可,例如,所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药,或所述联苯环辛二稀木脂素后于所述式Ⅰ所示化合物或其盐给药,或所述联苯环辛二稀木脂素与所述式Ⅰ所示化合物或其盐同时给药均在本申请的保护范围内。根据本发明的一些具体实施例,当所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药时,所述式Ⅰ所示化合物或其盐的半衰期可以得到显著延长,且其代谢过程中产生的式Ⅱ所示代谢物的含量显著降低,具有良好的抗肿瘤效果和安全性。According to an embodiment of the present invention, the biphenylcyclooctadienolignan is administered prior to the compound represented by Formula I or a salt thereof. Those skilled in the art can understand that the order of administration of the biphenylcyclooctadienol and the compound represented by formula I or its salt is not particularly limited, as long as the diphenylcyclooctadienol It is sufficient that the compound can play a role in the metabolic cycle of the compound represented by formula I or its salt. For example, the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt, Or the biphenylcyclooctadiene lignan is administered after the compound represented by formula I or its salt, or the biphenylcyclooctadienolignan and the compound represented by formula I or its salt are administered Simultaneous administration is within the scope of protection of this application. According to some specific embodiments of the present invention, when the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt, the half-life of the compound represented by formula I or its salt can be It is significantly prolonged, and the content of the metabolite shown in formula II produced during its metabolism is significantly reduced, and it has good anti-tumor effect and safety.
根据本发明的实施例,所述第二活性成分为口服剂型。所述第二活性成分可以是服用制剂、注射制剂、丸剂、缓控释制剂、植入剂或气雾剂等类型,并不受特别限制,根据本发明的具体实施例,当所述第二活性成分为口服制剂时所述药盒的半衰期更长,代谢过程中式Ⅱ所示代谢物的含量显著低于单独使用式Ⅰ所示化合物或其盐,所述药盒具备较强的抗肿瘤能力和较高的安全性。According to an embodiment of the invention, the second active ingredient is in an oral dosage form. The second active ingredient may be in the form of taking preparations, injection preparations, pills, sustained-release preparations, implants or aerosols, etc., and is not particularly limited. According to specific embodiments of the present invention, when the second active ingredient When the active ingredient is an oral preparation, the half-life of the kit is longer, and the content of the metabolite shown in Formula II during metabolism is significantly lower than when the compound shown in Formula I or its salt is used alone, and the kit has strong anti-tumor ability. and higher security.
在本发明的第五方面,本发明提出了一种式Ⅰ所示化合物或其盐的给药方法。根据本发明的实施例,所述方法包括使所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素联合用药。In the fifth aspect of the present invention, the present invention provides a method of administering the compound represented by Formula I or a salt thereof. According to an embodiment of the present invention, the method includes combining the compound represented by Formula I or a salt thereof with biphenylcyclooctadienolignan.
根据本发明的实施例,上述方法进一步包括如下附加技术特征中的至少之一:According to an embodiment of the present invention, the above method further includes at least one of the following additional technical features:
根据本发明的实施例,所述式Ⅰ所示化合物的盐包括下列中的至少之一:硫酸盐、磷酸盐、柠檬酸盐、酒石酸盐、富马酸盐、苯甲酸盐、己二酸盐、琥珀酸盐、甲磺酸盐和马来酸盐。According to an embodiment of the present invention, the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为盐酸盐。According to an embodiment of the present invention, the salt of the compound represented by Formula I is a hydrochloride.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐。According to embodiments of the present invention, the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
根据本发明的实施例,所述联苯环辛二稀木脂素包括下列中的至少之一:五味子甲素、五味子酯甲、五味子醇已、五味子醇甲、五味子乙素和五味子酚。According to an embodiment of the present invention, the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
根据本发明的实施例,所述联苯环辛二稀木脂素为五味子甲素。According to an embodiment of the present invention, the biphenylcyclooctadiene lignan is schisandrin A.
根据本发明的实施例,所述联合用药包括将式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素同时用药或分开用药。 According to an embodiment of the present invention, the combined medication includes administering the compound represented by formula I or its salt and biphenylcyclooctadienolignan simultaneously or separately.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:31)~(4:3)。根据本申请实施例的描述,通过小鼠与人类给药剂量的换算,采用该比例换算能够有效地保护人类免于或者缓解肿瘤。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3). According to the description of the embodiments of the present application, by converting the dosage between mice and humans, this ratio conversion can effectively protect humans from or alleviate tumors.
根据本发明的实施例,所述用药过程中式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:10)~(5:6)。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (1:10) to (5:6).
根据本发明的实施例,所述用药过程中式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:50)~(4:5)。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (7:50) to (4:5).
根据本发明的实施例,所述用药过程中式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81:125。According to an embodiment of the present invention, the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan during the medication process is 3:20, 1:5, 81:250 or 81:125.
根据本发明的实施例,所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药。本领域技术人员可以理解,所述联苯环辛二稀木脂素,和所述式Ⅰ所示化合物或其盐的给药顺序不受特别限制,只要所述联苯环辛二稀木脂素能够在所述式Ⅰ所示化合物或其盐的代谢周期内发挥作用即可,例如,所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药,或所述联苯环辛二稀木脂素后于所述式Ⅰ所示化合物或其盐给药,或所述联苯环辛二稀木脂素与所述式Ⅰ所示化合物或其盐同时给药均在本申请的保护范围内。According to an embodiment of the present invention, the biphenylcyclooctadienolignan is administered prior to the compound represented by Formula I or a salt thereof. Those skilled in the art can understand that the order of administration of the biphenylcyclooctadienol and the compound represented by formula I or its salt is not particularly limited, as long as the diphenylcyclooctadienol It is sufficient that the compound can play a role in the metabolic cycle of the compound represented by formula I or its salt. For example, the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt, Or the biphenylcyclooctadiene lignan is administered after the compound represented by formula I or its salt, or the biphenylcyclooctadienolignan and the compound represented by formula I or its salt are administered Simultaneous administration is within the scope of protection of this application.
根据本发明的实施例,所述联苯环辛二稀木脂素为口服给药。本领域技术人员可以理解,所述联苯环辛二稀木脂素可以是服用制剂、注射制剂、丸剂、缓控释制剂、植入剂或气雾剂等类型,并不受特别限制,根据本发明的具体实施例,当所述联苯环辛二稀木脂素为口服制剂时所述式Ⅰ所示化合物或其盐的半衰期更长,代谢过程中代谢产物1的含量显著低于单独使用式Ⅰ所示化合物或其盐,所述给药方法具有更强的抗肿瘤能力和较高的安全性。According to an embodiment of the present invention, the biphenylcyclooctadiene lignan is administered orally. Those skilled in the art can understand that the biphenylcyclooctadiene lignans can be in the form of oral preparations, injection preparations, pills, sustained-release preparations, implants or aerosols, etc., and are not particularly limited. According to In specific embodiments of the present invention, when the biphenylcyclooctadiene lignan is an oral preparation, the half-life of the compound represented by formula I or its salt is longer, and the content of metabolite 1 during metabolism is significantly lower than that of the compound alone. Using the compound represented by formula I or its salt, the administration method has stronger anti-tumor ability and higher safety.
根据本发明的实施例,所述联苯环辛二稀木脂素的给药剂量为0~25mg/kg。According to an embodiment of the present invention, the dosage of the biphenylcyclooctadiene lignan is 0 to 25 mg/kg.
根据本发明的实施例,所述联苯环辛二稀木脂素的给药剂量为5~25mg/kg。根据本发明的具体实施例,当所述联苯环辛二稀木脂素的给药剂量为5~25mg/kg时,所述式Ⅰ所示化合物或其盐的半衰期可以得到显著延长,其代谢过程中产生的式Ⅱ所示代谢物的含量显著降低,提高式Ⅰ所示化合物或其盐在体内的暴露量和脑组织中的剂量,降低式Ⅱ所示代谢产物引起的不良反应,使得式Ⅰ所示化合物或其盐的药效和安全性均得到提高,所述给药方法可以更加有效的抑制肿瘤生长,并具有较高的安全性。According to an embodiment of the present invention, the dosage of the biphenylcyclooctadiene lignan is 5 to 25 mg/kg. According to specific embodiments of the present invention, when the dosage of the biphenylcyclooctadiene lignan is 5 to 25 mg/kg, the half-life of the compound represented by formula I or its salt can be significantly extended, and the The content of the metabolites shown in formula II produced during the metabolism process is significantly reduced, which increases the exposure of the compound shown in formula I or its salt in the body and the dose in brain tissue, and reduces the adverse reactions caused by the metabolites shown in formula II, so that The efficacy and safety of the compound represented by formula I or its salt are improved, and the administration method can more effectively inhibit tumor growth and has higher safety.
在本发明的第六方面,本发明提出了一种延长式Ⅰ所示化合物或其盐半衰期的方法。根据本发明的实施例,所述方法包括使所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素联合用药。如前所述,发明人发现联苯环辛二稀木脂素通过抑制CYP450酶,如其亚型CYP3A酶而降低式Ⅰ所示化合物或其盐的代谢,延长其在体内的滞留时间而增强疗效,因此,根据本发明实施例的方法可以有效延长式Ⅰ所示化合物或其盐的半衰期。 In the sixth aspect of the present invention, the present invention proposes a method for extending the half-life of the compound represented by formula I or its salt. According to an embodiment of the present invention, the method includes combining the compound represented by Formula I or a salt thereof with biphenylcyclooctadiene lignan. As mentioned above, the inventor found that biphenylcyclooctadienolignans reduce the metabolism of the compound represented by formula I or its salt by inhibiting CYP450 enzymes, such as its subtype CYP3A enzyme, prolonging its residence time in the body and enhancing the therapeutic effect. , Therefore, the method according to the embodiment of the present invention can effectively extend the half-life of the compound represented by formula I or its salt.
根据本发明的实施例,上述方法进一步包括如下附加技术特征中的至少之一:According to an embodiment of the present invention, the above method further includes at least one of the following additional technical features:
根据本发明的实施例,所述式Ⅰ所示化合物的盐包括下列中的至少之一:硫酸盐、磷酸盐、柠檬酸盐、酒石酸盐、富马酸盐、苯甲酸盐、己二酸盐、琥珀酸盐、甲磺酸盐和马来酸盐。According to an embodiment of the present invention, the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为盐酸盐。According to an embodiment of the present invention, the salt of the compound represented by Formula I is a hydrochloride.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐。According to embodiments of the present invention, the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
根据本发明的实施例,所述联苯环辛二稀木脂素包括下列中的至少之一:五味子甲素、五味子酯甲、五味子醇已、五味子醇甲、五味子乙素和五味子酚。According to an embodiment of the present invention, the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
根据本发明的实施例,所述联苯环辛二稀木脂素为五味子甲素。According to an embodiment of the present invention, the biphenylcyclooctadiene lignan is schisandrin A.
根据本发明的实施例,所述联合用药包括将式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素同时用药或分开用药。所述联合用药的方式不再赘述,可参考第五方面所述式Ⅰ所示化合物或其盐的给药方法中的联合用药方式。According to an embodiment of the present invention, the combined medication includes administering the compound represented by formula I or its salt and biphenylcyclooctadienolignan simultaneously or separately. The method of combined administration will not be described in detail, and reference may be made to the method of combined administration in the administration method of the compound represented by formula I or its salt described in the fifth aspect.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:31)~(4:3)。根据本申请实施例的描述,通过小鼠与人类给药剂量的换算,采用该比例换算能够有效地延长保护人类免于或者缓解肿瘤的药效时间。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3). According to the description of the embodiments of the present application, by converting the dosage between mice and humans, using this ratio conversion can effectively extend the efficacy time of protecting humans from or alleviating tumors.
根据本发明的实施例,所述用药过程中式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:10)~(5:6)。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (1:10) to (5:6).
根据本发明的实施例,所述用药过程中式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:10)~(4:5)。According to an embodiment of the present invention, the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (7:10) to (4:5).
根据本发明的实施例,所述用药过程中式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81:125。According to an embodiment of the present invention, the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan during the medication process is 3:20, 1:5, 81:250 or 81:125.
根据本发明的实施例,所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药。如前所述,所述联苯环辛二稀木脂素,和所述式Ⅰ所示化合物或其盐的给药顺序不受特别限制,只要所述联苯环辛二稀木脂素能够在所述式Ⅰ所示化合物或其盐的代谢周期内发挥作用即可,例如,所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药,或所述联苯环辛二稀木脂素后于所述式Ⅰ所示化合物或其盐给药,或所述联苯环辛二稀木脂素与所述式Ⅰ所示化合物或其盐同时给药均在本申请的保护范围内。According to an embodiment of the present invention, the biphenylcyclooctadienolignan is administered prior to the compound represented by Formula I or a salt thereof. As mentioned above, the order of administration of the biphenylcyclooctadienolignan and the compound represented by Formula I or its salt is not particularly limited, as long as the biphenylcyclooctadienolignan can It only needs to be effective within the metabolic cycle of the compound represented by formula I or its salt. For example, the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt, or the The biphenylcyclooctadiene lignan is administered after the compound represented by the formula I or its salt, or the biphenylcyclooctadiene lignan and the compound represented by the formula I or its salt are administered simultaneously. All drugs are within the protection scope of this application.
根据本发明的实施例,所述联苯环辛二稀木脂素为口服给药。According to an embodiment of the present invention, the biphenylcyclooctadiene lignan is administered orally.
根据本发明的实施例,所述联苯环辛二稀木脂素的给药剂量为0~25mg/kg。According to an embodiment of the present invention, the dosage of the biphenylcyclooctadiene lignan is 0 to 25 mg/kg.
根据本发明的实施例,所述联苯环辛二稀木脂素的给药剂量为5~25mg/kg。根据本发明的具体实施例,当所述联苯环辛二稀木脂素的给药剂量为5~25mg/kg时,所述式Ⅰ所示化合物或其盐的半衰期可以得到显著延长,其代谢过程中产生的式Ⅱ所示代谢物的含量显著降低,所述方法可以更加有效的延长 式Ⅰ所示化合物或其盐的半衰期,并提高其透脑率,以提高药效。According to an embodiment of the present invention, the dosage of the biphenylcyclooctadiene lignan is 5 to 25 mg/kg. According to specific embodiments of the present invention, when the dosage of the biphenylcyclooctadiene lignan is 5 to 25 mg/kg, the half-life of the compound represented by formula I or its salt can be significantly extended, and the The content of the metabolites shown in formula II produced during the metabolic process is significantly reduced, and the method can more effectively extend the The half-life of the compound represented by formula I or its salt is increased, and its brain penetration rate is increased to improve the drug efficacy.
在本发明的第七方面,本发明提出了一种降低式Ⅰ所示化合物或其盐生成式Ⅱ所示化合物的方法。根据本发明的实施例,所述方法包括,使所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素联合用药。如前所述,发明人发现联苯环辛二稀木脂素抑制CYP450酶,如其亚型CYP3A酶而降低式Ⅰ所示化合物或其盐的代谢,并减少血清中式Ⅱ所示代谢物的浓度或含量,提高式Ⅰ所示化合物或其盐在体内的暴露量和脑组织中的剂量,降低式Ⅱ所示代谢产物引起的不良反应,使得式Ⅰ所示化合物或其盐的药效和安全性均得到提高。In the seventh aspect of the present invention, the present invention proposes a method for reducing the compound represented by formula I or its salt to produce the compound represented by formula II. According to an embodiment of the present invention, the method includes combining the compound represented by Formula I or a salt thereof with biphenylcyclooctadienolignan. As mentioned above, the inventor found that biphenylcyclooctadienolignan inhibits CYP450 enzymes, such as its subtype CYP3A enzyme, thereby reducing the metabolism of the compound represented by formula I or its salt, and reducing the concentration of the metabolite represented by formula II in the serum. or content, increase the exposure of the compound represented by formula I or its salt in the body and the dose in brain tissue, reduce the adverse reactions caused by the metabolite represented by formula II, and make the compound represented by formula I or its salt more effective and safe sex has been improved.
根据本发明的实施例,上述方法进一步包括如下附加技术特征中的至少之一:According to an embodiment of the present invention, the above method further includes at least one of the following additional technical features:
根据本发明的实施例,所述式Ⅰ所示化合物的盐包括下列中的至少之一:硫酸盐、磷酸盐、柠檬酸盐、酒石酸盐、富马酸盐、苯甲酸盐、己二酸盐、琥珀酸盐、甲磺酸盐和马来酸盐。According to an embodiment of the present invention, the salt of the compound represented by formula I includes at least one of the following: sulfate, phosphate, citrate, tartrate, fumarate, benzoate, adipic acid Salts, succinates, methanesulfonates and maleates.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为盐酸盐。According to an embodiment of the present invention, the salt of the compound represented by Formula I is a hydrochloride.
根据本发明的实施例,所述式Ⅰ所示化合物的盐为(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐。According to embodiments of the present invention, the salt of the compound represented by Formula I is (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2, 4-Dimethylpiperazine-1-carboxylate hydrochloride.
根据本发明的实施例,所述联苯环辛二稀木脂素包括下列中的至少之一:五味子甲素、五味子酯甲、五味子醇已、五味子醇甲、五味子乙素和五味子酚。According to an embodiment of the present invention, the biphenylcyclooctadiene lignans include at least one of the following: schisandrin A, schisandrin ester A, schisandrol hae, schisandrin alcohol A, schisandrin B, and schisandrol.
根据本发明的实施例,所述联苯环辛二稀木脂素为五味子甲素。According to an embodiment of the present invention, the biphenylcyclooctadiene lignan is schisandrin A.
根据本发明的实施例,所述联合用药包括将式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素同时用药或分开用药。According to an embodiment of the present invention, the combined medication includes administering the compound represented by formula I or its salt and biphenylcyclooctadienolignan simultaneously or separately.
根据本发明的实施例,所述式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:31)~(4:3)。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan is (1:31) to (4:3).
根据本发明的实施例,所述用药过程中式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(1:10)~(5:6)。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (1:10) to (5:6).
根据本发明的实施例,所述用药过程中式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为(7:50)~(4:5)。According to an embodiment of the present invention, the mass ratio of the compound represented by Formula I or its salt to biphenylcyclooctadiene lignan during the medication process is (7:50) to (4:5).
根据本发明的实施例,所述用药过程中式Ⅰ所示化合物或其盐与联苯环辛二稀木脂素的质量比为3:20,1:5,81:250或81:125。According to an embodiment of the present invention, the mass ratio of the compound represented by formula I or its salt to biphenylcyclooctadiene lignan during the medication process is 3:20, 1:5, 81:250 or 81:125.
根据本发明的实施例,所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药。如前所述,所述联苯环辛二稀木脂素,和所述式Ⅰ所示化合物或其盐的给药顺序不受特别限制,只要所述联苯环辛二稀木脂素能够在所述式Ⅰ所示化合物或其盐的代谢周期内发挥作用即可,例如,所述联苯环辛二稀木脂素先于所述式Ⅰ所示化合物或其盐给药,或所述联苯环辛二稀木脂素后于所述式Ⅰ所示化合物或其盐给药,或所述联苯环辛二稀木脂素与所述式Ⅰ所示化合物或其盐同时给药均在本申请的保护范围内。According to an embodiment of the present invention, the biphenylcyclooctadienolignan is administered prior to the compound represented by Formula I or a salt thereof. As mentioned above, the order of administration of the biphenylcyclooctadienolignan and the compound represented by Formula I or its salt is not particularly limited, as long as the biphenylcyclooctadienolignan can It only needs to be effective within the metabolic cycle of the compound represented by formula I or its salt. For example, the biphenylcyclooctadienolignan is administered before the compound represented by formula I or its salt, or the The biphenylcyclooctadiene lignan is administered after the compound represented by the formula I or its salt, or the biphenylcyclooctadiene lignan and the compound represented by the formula I or its salt are administered simultaneously. All drugs are within the protection scope of this application.
根据本发明的实施例,所述联苯环辛二稀木脂素为口服给药。 According to an embodiment of the present invention, the biphenylcyclooctadiene lignan is administered orally.
根据本发明的实施例,所述联苯环辛二稀木脂素的给药剂量为0~25mg/kg。According to an embodiment of the present invention, the dosage of the biphenylcyclooctadiene lignan is 0 to 25 mg/kg.
根据本发明的实施例,所述联苯环辛二稀木脂素的给药剂量为5~25mg/kg。根据本发明的具体实施例,当所述五味子甲素的给药剂量为5~25mg/kg时,所述式Ⅰ所示化合物或其盐代谢过程中可以更加有效的减少血清中产生的式Ⅱ所示代谢物的浓度或含量,因此,根据本发明实施例的方法可以更加显著的降低式Ⅰ所示化合物或其盐代谢过程中式Ⅱ所示代谢物的含量。According to an embodiment of the present invention, the dosage of the biphenylcyclooctadiene lignan is 5 to 25 mg/kg. According to specific embodiments of the present invention, when the dosage of Schisandrin A is 5 to 25 mg/kg, the compound represented by Formula I or its salt can more effectively reduce the amount of Formula II produced in the serum during the metabolism process. Therefore, the method according to the embodiment of the present invention can more significantly reduce the content of the metabolite shown in Formula II during the metabolism of the compound shown in Formula I or its salt.
下面详细描述本发明的实施例,下述实施例是示例性的,旨在用于解释本发明,而不能理解为对本发明的限制。The embodiments of the present invention are described in detail below. The following embodiments are illustrative and are intended to explain the present invention, but should not be understood as limiting the present invention.
在对本发明描述的过程中,对于本文中有关的术语进行了解释和说明,这些解释和说明仅仅是为了方便对于方案的理解,并不能看做是对本发明保护方案的限制。In the process of describing the present invention, the relevant terms in this article have been explained and explained. These explanations and explanations are only for the convenience of understanding of the scheme and cannot be regarded as limiting the protection scheme of the present invention.
本申请中,“式Ⅰ所示化合物”均指以下结构的化合物:
In this application, "compounds represented by formula I" all refer to compounds with the following structures:
本申请中,所述“式Ⅰ所示化合物的盐酸盐”、“(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐”、”供试药物”、“原研药物”的分子式为C22H23ClFN5O3化学结构为:
In this application, the "hydrochloride of the compound represented by formula I", "(R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl The molecular formula of "2,4-dimethylpiperazine-1-carboxylate hydrochloride", "test drug" and "original drug" is C 22 H 23 ClFN 5 O 3. The chemical structure is:
本申请中“式Ⅱ所示代谢物”、“代谢产物1”的分子式为C21H21ClFN5O3,其化学结构为:
In this application, the molecular formula of "metabolite represented by formula II" and "metabolite 1" is C 21 H 21 ClFN 5 O 3 , and its chemical structure is:
本申请中,“五味子甲素”是指分子式为C24H32O6,分子量为416.507的物质,化学结构为:
In this application, "Schisandrin A" refers to a substance with a molecular formula of C 24 H 32 O 6 and a molecular weight of 416.507. Its chemical structure is:
“五味子酯甲”的分子式为C30H32O9,分子量为536.577,化学结构为:
The molecular formula of "Schisandrin A" is C 30 H 32 O 9 , the molecular weight is 536.577, and the chemical structure is:
“五味子醇已”的分子式为C23H28O7,分子量为416.47,化学结构为:
The molecular formula of "Schizandrol" is C 23 H 28 O 7 , the molecular weight is 416.47, and the chemical structure is:
“五味子醇甲”的分子式为C24H32O7,分子量为432.51,化学结构为:
The molecular formula of "Schizandrol Methyl" is C 24 H 32 O 7 , the molecular weight is 432.51, and the chemical structure is:
“五味子乙素”的分子式为C23H28O6,分子量为400.46,化学结构为:
The molecular formula of "Schisandrin B" is C 23 H 28 O 6 , the molecular weight is 400.46, and the chemical structure is:
“五味子酚”的分子式为C23H30O6,分子量为402.48,化学结构为:
The molecular formula of "Schizandrol" is C 23 H 30 O 6 , the molecular weight is 402.48, and the chemical structure is:
本申请中,所述“单剂型”是指一个制剂单次给药使用完的剂型。In this application, the "single dosage form" refers to a dosage form that is used up after a single administration of a preparation.
本申请中,所述“药物联合”是指为了达到治疗目的而采用的两种或两种以上药物同时或先后应用,其结果主要是为了增加药物的疗效或为了减轻药物的毒副作用。In this application, the "drug combination" refers to the simultaneous or sequential application of two or more drugs to achieve therapeutic purposes. The result is mainly to increase the efficacy of the drug or to reduce the toxic and side effects of the drug.
下面将结合实施例对本发明的方案进行解释。本领域技术人员将会理解,下面的实施例仅用于说明本发明,而不应视为限定本发明的范围。实施例中未注明具体技术或条件的,按照本领域内的文献所描述的技术或条件或者按照产品说明书进行。所用试剂或仪器未注明生产厂商者,均为可以通过市购获得的常规产品。The solutions of the present invention will be explained below with reference to examples. Those skilled in the art will understand that the following examples are only used to illustrate the present invention and should not be regarded as limiting the scope of the present invention. If specific techniques or conditions are not specified in the examples, the techniques or conditions described in literature in the field or product instructions will be followed. If the manufacturer of the reagents or instruments used is not indicated, they are all conventional products that can be purchased commercially.
实施例1 CYP450酶抑制剂对CYP450酶抑制能力检测Example 1 Detection of CYP450 Enzyme Inhibitory Ability of CYP450 Enzyme Inhibitors
本申请中发明人对药物(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯 盐酸盐单用及与CYP450酶抑制剂联用进行研究,其中,所述(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐的化学结构为:In this application, the inventor is interested in the drug (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1- Carboxylate Hydrochloride was studied alone and in combination with CYP450 enzyme inhibitors, wherein the (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazoline-6- The chemical structure of 2,4-dimethylpiperazine-1-carboxylate hydrochloride is:
具体的实验操作如下: The specific experimental operations are as follows:
实验体系使用人肝微粒体,肝微粒体各4组,分别为1)(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐组(供试药物单用组);2)(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐伍用五味子甲素(WWZ)组(供试药物+WWZ组);3)(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐伍用水飞蓟宾组(供试药物+水飞蓟宾组),4)(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐伍用酮康唑组(供试药物+酮康唑组)。具体的实验操作步骤如下:The experimental system uses human liver microsomes. There are 4 groups of liver microsomes each, namely 1) (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl. 2,4-Dimethylpiperazine-1-carboxylate hydrochloride group (test drug alone group); 2) (R)-4-(3-chloro-2-fluorophenylamino)-7 -Methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride combined with Schizandrin A (WWZ) group (test drug + WWZ group); 3) ( R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride with water Silybin group (test drug + silibinin group), 4) (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2 , 4-dimethylpiperazine-1-carboxylate hydrochloride combined with ketoconazole group (test drug + ketoconazole group). The specific experimental steps are as follows:
精密量取(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐的储备液(Stock0009)1mg/mL,用PBS缓冲溶液稀释为摩尔浓度为10μM的标准工作溶液。精密量取(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐1mg/mL的储备液(Stock0009),用溶解有五味子甲素、水飞蓟宾或酮康唑,各5μM的PBS缓冲溶液稀释为摩尔浓度为20μM的标准工作溶液,然后得到为10μM的含有5μM的多种CYP450酶抑制剂混合工作液2。称取NADPH(还原型)约7mg,用PBS缓冲溶液稀释为摩尔浓度为10mM的工作液。肝微粒体(HLM)初始浓度为20mg/mL,用PBS缓冲溶液稀释为摩尔浓度为2mg/mL的肝微粒体工作液。取上述各工作液,按表1量取配制。Precisely measure (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate salt The stock solution of acid salt (Stock0009) is 1 mg/mL, and diluted with PBS buffer solution to a standard working solution with a molar concentration of 10 μM. Precisely measure (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate salt The stock solution of acid salt (Stock0009) of 1 mg/mL was diluted into a standard working solution with a molar concentration of 20 μM with a PBS buffer solution containing 5 μM of Schisandrin A, silibinin, or ketoconazole, and then 10 μM was obtained. Mixed working solution 2 containing 5 μM of various CYP450 enzyme inhibitors. Weigh about 7mg of NADPH (reduced form) and dilute it with PBS buffer solution to a working solution with a molar concentration of 10mM. The initial concentration of liver microsomes (HLM) was 20 mg/mL, and it was diluted with PBS buffer solution to a liver microsome working solution with a molar concentration of 2 mg/mL. Take each of the above working solutions and prepare according to Table 1.
表1:肝微粒体孵育体系样品配制方法
Table 1: Preparation method of liver microsome incubation system sample
将4种受试工作液,HLM,PBS缓冲溶液按表中体积在孵育管中混合(平行三份),37℃水浴预热5分钟。同时,10mM的NADPH也在37℃水浴预热。充分预热后,取30μL NADPH加入肝微粒体孵育管,轻轻震荡孵育管,开始孵育。分别于启动孵育2min、15min、1h、2h、4h取40μL孵育管中 加入160μL含有内标乙腈溶液(普萘洛尔20ng/mL)。各样品经振荡,离心10min(离心力20200g),取上清LC/MS/MS进样测定。Mix the four test working solutions, HLM, and PBS buffer solutions in incubation tubes according to the volumes in the table (in triplicate), and preheat in a 37°C water bath for 5 minutes. At the same time, 10mM NADPH was also preheated in a 37°C water bath. After sufficient preheating, add 30 μL NADPH to the liver microsome incubation tube, shake the incubation tube gently, and start incubation. Take 40 μL of incubation tubes at 2 minutes, 15 minutes, 1 hour, 2 hours, and 4 hours after starting the incubation. Add 160 μL of acetonitrile solution containing internal standard (propranolol 20 ng/mL). Each sample was shaken and centrifuged for 10 min (centrifugal force 20200g), and the supernatant was taken and injected for LC/MS/MS analysis.
实验结果如表2所示,通过对药物(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐单用及与多种CYP450酶抑制剂联用进行清除率效果进行对比,发明人发现,五味子甲素相较于其它高、低强度CYP450酶抑制剂,其对CYP450酶抑制程度适中,本研究中选择中等程度的抑制剂WWZ)进行后续研究。The experimental results are shown in Table 2. By treating the drug (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperidine Comparing the clearance effect of oxazine-1-carboxylate hydrochloride alone and in combination with various CYP450 enzyme inhibitors, the inventor found that compared with other high and low intensity CYP450 enzyme inhibitors, Schisandrin A has a higher The degree of inhibition of CYP450 enzymes is moderate. In this study, the moderate inhibitor WWZ) was selected for follow-up research.
表2:
Table 2:
实施例2 (R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐在肝微粒中体外代谢稳定性研究Example 2 (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate salt Study on the metabolic stability of acid salt in liver microparticles in vitro
在实施例1的基础上,本试验主要考察原研药物(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐在肝微粒体孵育体系中代谢稳定性差异,及与抑制剂WWZ伍用后代谢稳定性的变化。实验体系使用人肝微粒体,肝微粒体各三组,分别为1)(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐组(供试药物单用组);2)(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐伍用WWZ组(供试药物+WWZ组);3)阳性对照药物(维拉帕米,VPL)组。具体的实验操作步骤如下:On the basis of Example 1, this experiment mainly investigated the original drug (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-di Differences in metabolic stability of methylpiperazine-1-carboxylate hydrochloride in liver microsome incubation system, and changes in metabolic stability after combined use with the inhibitor WWZ. The experimental system uses human liver microsomes. There are three groups of liver microsomes, each of which is 1) (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl. 2,4-Dimethylpiperazine-1-carboxylate hydrochloride group (test drug alone group); 2) (R)-4-(3-chloro-2-fluorophenylamino)-7 -Methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride combined with WWZ group (test drug + WWZ group); 3) Positive control drug (Vera Pami, VPL) group. The specific experimental steps are as follows:
精密量取(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐的储备液(Stock0009)1mg/mL,用PBS缓冲溶液稀释为摩尔浓度为10μM的标准工作溶液。精密量取(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐1mg/mL的储备液(Stock0009),用溶解有五味子甲素(5μM)的PBS缓冲溶液稀释为摩尔浓度为20μM的标准工作溶液,然后得到为10μM的含有5μM五味子甲素的混合工作液2。量取维拉帕米1mg/mL的储备液(StockVPL)100μL,用PBS缓冲溶液稀释为摩尔浓度为10μM的工作液,作为阳性对照。称取NADPH(还原型)约7mg,用PBS缓冲溶液稀释为摩尔浓度为10mM的工作液。肝微粒体(HLM)初始浓度为20mg/mL,用PBS 缓冲溶液稀释为摩尔浓度为2mg/mL的肝微粒体工作液。取上述各工作液,按表3量取配制。Precisely measure (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate salt The stock solution of acid salt (Stock0009) is 1 mg/mL, and diluted with PBS buffer solution to a standard working solution with a molar concentration of 10 μM. Precisely measure (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate salt The stock solution (Stock0009) of acid salt 1 mg/mL was diluted with PBS buffer solution dissolved with Schisandrin A (5 μM) to a standard working solution with a molar concentration of 20 μM, and then a mixed working solution containing 5 μM Schisandrin A was obtained as 10 μM. 2. Measure 100 μL of verapamil 1 mg/mL stock solution (StockVPL) and dilute it with PBS buffer solution to a working solution with a molar concentration of 10 μM as a positive control. Weigh about 7mg of NADPH (reduced form) and dilute it with PBS buffer solution to a working solution with a molar concentration of 10mM. The initial concentration of liver microsomes (HLM) was 20 mg/mL in PBS. The buffer solution was diluted to a liver microsomal working solution with a molar concentration of 2 mg/mL. Take each of the above working solutions and prepare according to Table 3.
表3:肝微粒体孵育体系样品配制方法
Table 3: Preparation method of liver microsome incubation system sample
将三种受试工作液,HLM,PBS缓冲溶液按表中体积在孵育管中混合(平行三份),37℃水浴预热5分钟。同时,10mM的NADPH也在37℃水浴预热。充分预热后,取30μL NADPH加入肝微粒体孵育管,轻轻震荡孵育管,开始孵育。分别于启动孵育2min、15min、1h、2h、4h取40μL孵育管中加入160μL含有内标乙腈溶液(普萘洛尔20ng/mL)。各样品经振荡,离心10min(离心力20200g),取上清LC/MS/MS进样测定。Mix the three test working solutions, HLM, and PBS buffer solutions in incubation tubes according to the volumes in the table (in triplicate), and preheat in a 37°C water bath for 5 minutes. At the same time, 10mM NADPH was also preheated in a 37°C water bath. After sufficient preheating, add 30 μL NADPH to the liver microsome incubation tube, shake the incubation tube gently, and start incubation. At 2 min, 15 min, 1 h, 2 h, and 4 h after starting the incubation, add 160 μL of acetonitrile solution containing the internal standard (propranolol 20 ng/mL) into a 40 μL incubation tube. Each sample was shaken and centrifuged for 10 min (centrifugal force 20200g), and the supernatant was taken and injected for LC/MS/MS analysis.
维拉帕米作为阳性对照(取样时间点为孵育起始30min和2h),维拉帕米不加NADPH(以PBS替代)作为阴性对照。无药物(以PBS替代)组作为空白对照。Verapamil was used as a positive control (sampling time points were 30 min and 2 h after incubation), and verapamil without NADPH (replaced by PBS) was used as a negative control. The drug-free (replaced by PBS) group served as a blank control.
人肝微粒体(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐体外代谢稳定性检测结果如表4所示,其中,作为阳性对照的维拉帕米,经过2h孵育后,原型药物剩余10%,提示本批次孵育体系酶代谢活性正常。人肝微粒体中孵育供试药物后发生了代谢,原型浓度随着孵育时间增加而减少。同时,(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐伍用WWZ后代谢速率减慢。Human liver microsomal (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate The results of the in vitro metabolic stability test of hydrochloride are shown in Table 4. Among them, for verapamil, which was used as a positive control, 10% of the prototype drug remained after 2 hours of incubation, indicating that the enzyme metabolic activity of this batch of incubation system was normal. The test drug was metabolized after incubation in human liver microsomes, and the prototype concentration decreased with increasing incubation time. At the same time, (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride Metabolic rate slows down after taking WWZ.
表4:
Table 4:
实施例3 (R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐与五味子甲素伍用后SD大鼠体内代谢动力学研究 Example 3 (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate salt Study on the metabolic kinetics in SD rats after combined administration of acid salt and Schisandrin A
本实施例在实施例1和2的基础上进行,主要考察所述供试药物((R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐)与五味子甲素伍用后SD大鼠体内代谢动力学变化。该研究将9只健康生长的6-8周龄体重为220~300g雄性大鼠分为3组,分别为灌胃给药供试药物(5mg/kg)组;灌胃给药五味子甲素25mg/kg后灌胃给药供试药物(5mg/kg)组(A0009+WWZ(PO));静脉注射五味子甲素(10mg/kg)后灌胃给药供试药物(5mg/kg)组(A0009+WWZ(IV))。This example is carried out on the basis of Examples 1 and 2, mainly investigating the test drug ((R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazoline- Changes in metabolic kinetics in SD rats after combined administration of 6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride and schizandrin. In this study, 9 healthy growing male rats aged 6-8 weeks and weighing 220-300g were divided into 3 groups: the test drug (5mg/kg) group was administered by gavage; the group was administered 25 mg of Schisandrin A by gavage. /kg and then intragastric administration of the test drug (5mg/kg) group (A0009+WWZ(PO)); intravenous injection of Schisandrin A (10mg/kg) followed by intragastric administration of the test drug (5mg/kg) group ( A0009+WWZ(IV)).
采集上述3组大鼠全血,采血时间点为:给药前、给药后5min、15min、30min、1h、2h、4h、8h、12h、24h、36h。采血部位及采血量:由眼后静脉丛采血约0.3mL。将获得的全血样品进行离心(离心力2000g,4℃)10min,取上层血浆。使用有机溶剂蛋白沉淀法处理血浆样品。取血浆100μL,加入400μL内标乙腈溶液(普萘洛尔,20ng/mL)涡旋混匀,离心10min(离心力20200g,4℃),取上清LC/MS/MS进样测定。采用Winnonlin药代动力学程序对所测数据进行分析,计算主要药代动力学参数。The whole blood of the rats in the above three groups was collected at the following time points: before administration and 5min, 15min, 30min, 1h, 2h, 4h, 8h, 12h, 24h, and 36h after administration. Blood collection site and volume: About 0.3 mL of blood is collected from the retroocular venous plexus. The obtained whole blood sample was centrifuged (centrifugal force 2000g, 4°C) for 10 min, and the upper plasma was taken. Plasma samples were processed using organic solvent protein precipitation. Take 100 μL of plasma, add 400 μL of internal standard acetonitrile solution (propranolol, 20 ng/mL), vortex and mix evenly, centrifuge for 10 min (centrifugal force 20200g, 4°C), and take the supernatant and inject it for LC/MS/MS analysis. The Winnonlin pharmacokinetic program was used to analyze the measured data and calculate the main pharmacokinetic parameters.
各组动物供试药物的血药浓度-时间的变化数据如表5所示,主要的药代动力学参数如表6和表7所示。综合体内代谢结果可以看出,伍用了WWZ后供试药物化合物的体内PK行为受到影响。这种影响与伍用WWZ的给药方式有关,静脉伍用没有改变供试药物的清除率,而口服伍用提高了供试药物的生物利用度。由此,发明人推测,伍用WWZ的药物相互作用不能简单的以WWZ对肝代谢酶的抑制作用来解释,而主要表现在吸收环节。WWZ主要作用于吸收相的肠道和肝脏,通过增加供试药物的吸收或降低肝肠首过而提高生物利用度。口服伍用WWZ后,(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐的暴露量AUCinf和半衰期t1/2h平均值明显高于单用(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐组。The blood drug concentration-time change data of the test drugs in each group of animals are shown in Table 5, and the main pharmacokinetic parameters are shown in Tables 6 and 7. Based on the in vivo metabolism results, it can be seen that the in vivo PK behavior of the test drug compounds is affected after taking WWZ. This effect is related to the administration method of co-administration of WWZ. Intravenous co-administration did not change the clearance rate of the test drug, while oral co-administration improved the bioavailability of the test drug. From this, the inventor speculates that drug interactions with WWZ cannot simply be explained by the inhibitory effect of WWZ on liver metabolic enzymes, but are mainly manifested in the absorption process. WWZ mainly acts on the intestines and liver in the absorption phase, improving bioavailability by increasing the absorption of the test drug or reducing the first pass of the liver and intestines. After oral administration of WWZ, (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylic The average exposure AUCinf and half-life t 1/2 h of acid ester hydrochloride were significantly higher than those of (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazoline alone -6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride group.
表5:
table 5:
表6:单独给药、伍用WWZ(灌胃、静注)试验中供试药物的药代动力学参数
Table 6: Pharmacokinetic parameters of the test drugs in single administration and combined WWZ (gastric administration, intravenous injection) trials
实施例4 (R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐与WWZ伍用后SD大鼠体内代谢转化研究Example 4 (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate salt Study on metabolic transformation in SD rats after combined use of acid salt and WWZ
为进一步观察伍用WWZ后供试药物代谢过程中的代谢产物1(式Ⅱ所示化合物)在体内给药后的动力学特征,该部分研究利用实施例3获得的SD大鼠血浆样品,检测代谢产物1的浓度-时间变化,并计算药动学参数,具体的实验操作参考实施例2。
In order to further observe the kinetic characteristics of metabolite 1 (compound of formula II) in the metabolism process of the test drug after taking WWZ in vivo, this part of the study used the SD rat plasma samples obtained in Example 3 to detect Concentration-time changes of metabolite 1, and pharmacokinetic parameters were calculated. For specific experimental operations, refer to Example 2.
各组SD大鼠血浆中代谢产物1浓度-时间变化结果如表7所示所示,其中,表7及表8数据表明伍用WWZ后代谢产物1的代谢生成规律发生了改变;无论口服还是静脉伍用WWZ,在其用药后的8h内,代谢产物1的浓度均低于供试药物单用组,表明伍用WWZ可以有效抑制代谢产物1的生成。供试 药物单用组的代谢产物1的血液中的暴露量Cmax于给药后4h高达261ng/mL。口服或是静脉伍用WWZ后,首先降低了血液中代谢产物1暴露量,Cmax分别为190ng/mL和118ng/mL;其次延长了代谢产物1的达峰时间,分别与给药后8h或12h达到Cmax。从代谢产物1的暴露量和达峰时间数据表明,伍用WWZ后具有降低代谢产物1的表征。The concentration-time change results of metabolite 1 in the plasma of SD rats in each group are shown in Table 7. Among them, the data in Table 7 and Table 8 show that the metabolic production pattern of metabolite 1 has changed after taking WWZ; whether it is taken orally or When WWZ was administered intravenously together, within 8 hours after its administration, the concentration of metabolite 1 was lower than that of the test drug alone group, indicating that the combined administration of WWZ can effectively inhibit the production of metabolite 1. for testing The blood exposure Cmax of metabolite 1 in the drug alone group was as high as 261ng/mL 4 hours after administration. After oral or intravenous administration of WWZ, it first reduced the exposure of metabolite 1 in the blood, with Cmax of 190ng/mL and 118ng/mL respectively; secondly, it prolonged the peak time of metabolite 1, which was 8h or 12h after administration. Reach Cmax. Data from the exposure amount and peak time of metabolite 1 show that the use of WWZ has the characteristics of reducing metabolite 1.
表7:SD大鼠血浆样品中代谢产物1的血药浓度
Table 7: Plasma concentration of metabolite 1 in SD rat plasma samples
表8:
Table 8:
实施例5 (R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐与WWZ联合使用对人肺癌PC9裸鼠移植瘤的生长抑制作用Example 5 (R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiperazine-1-carboxylate salt The growth inhibitory effect of combined use of acid salt and WWZ on the growth of human lung cancer PC9 transplanted tumors in nude mice
本实施例在实施例1-4的基础上,继续对所述供试药物以及WWZ进行研究,无菌条件下取出PC9肿瘤组织,切成2.0mm×2.0mm×2.0mm大小的瘤块,均匀接种于体重为16.0-18.0g的 BALB/cAnSlacNifdc-nu雄性裸鼠左侧腋背部皮下,接种当天记为D0。当肿瘤体积达到200.0mm3左右时(D6),按肿瘤体积随机分组,分为:溶剂对照1组(25%PEG400+pH5.5水)、溶剂对照2组(pH5.5水)、WWZ(25.0mg/kg)组、供试药物[(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐]组(8.1mg/kg)组、供试药物[(R)-4-(3-氯-2-氟苯基氨基)-7-甲氧基喹唑啉-6-基2,4-二甲基哌嗪-1-羧酸酯盐酸盐]组(16.2mg/kg)、WWZ(25.0mg/kg)+供试药物组(8.1mg/kg)WWZ(25.0mg/kg)+供试药物组(16.2mg/kg),共7组,每组10只动物;分组当天,所有组别动物开始灌胃给药,每天一次,共饲养28天,给药28次。Based on Examples 1-4, this example continues to study the test drugs and WWZ. The PC9 tumor tissue is taken out under sterile conditions and cut into tumor pieces of 2.0mm×2.0mm×2.0mm, uniformly Inoculate into children weighing 16.0-18.0g BALB/cAnSlacNifdc-nu male nude mice were injected subcutaneously on the left axillary back, and the day of inoculation was recorded as D0. When the tumor volume reaches about 200.0 mm 3 (D6), the tumors are randomly divided into groups according to the tumor volume: solvent control group 1 (25% PEG400 + pH 5.5 water), solvent control group 2 (pH 5.5 water), WWZ ( 25.0 mg/kg) group, test drug [(R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yl 2,4-dimethylpiper Azine-1-carboxylate hydrochloride] group (8.1 mg/kg) group, test drug [(R)-4-(3-chloro-2-fluorophenylamino)-7-methoxyquinazole Phenolin-6-yl 2,4-dimethylpiperazine-1-carboxylate hydrochloride] group (16.2 mg/kg), WWZ (25.0 mg/kg) + test drug group (8.1 mg/kg) WWZ (25.0mg/kg) + test drug group (16.2mg/kg), a total of 7 groups, 10 animals in each group; on the day of grouping, animals in all groups began to be given intragastric administration, once a day, for a total of 28 days. Dosed 28 times.
给药剂量说明:研究表明,所述供试药物在小鼠异种移植肿瘤模型中的体内抗肿瘤研究中,按照游离碱来计算使用剂量为7.5mg/kg和15mg/kg,并证明具有较好的抗肿瘤效果,本研究中继续使用该剂量。根据CoA该批次化合物(批号:CPo121798-04-05-02-34-01-RS)游离碱含量为91.85%,游离碱分子量为459.9,盐酸盐分子量为496.4。按照CoA中盐酸盐含量计算,该研究中小鼠使用剂量分别为8.1mg/kg和16.2mg/kg,该药效剂量经体表面积剂量换算为人的使用剂量为0.7mg/kg和1.4mg/kg。WWZ给药剂量说明:目前WWZ在人用临床上的药品名称为五酯胶囊(中药),其制剂为胶囊剂,每粒含WWZ 11.25mg。临床上用法用量为每次两粒,一日三次,日使用剂量为67.5mg。该剂量为成年人(60kg)每日临床上使用的最高药效剂量,该最高药效剂量经体表面积剂量换算为该研究中小鼠的使用剂量,本研究中选择给与小鼠WWZ一个剂量组,即最高药效剂量为25.0mg/kg。Instructions for dosage: Studies have shown that in the in vivo anti-tumor study of the test drug in mouse xenograft tumor model, the dosage calculated based on the free base is 7.5 mg/kg and 15 mg/kg, and has been proven to have better anti-tumor effect, this dose was continued to be used in this study. According to CoA, the free base content of this batch of compounds (batch number: CPo121798-04-05-02-34-01-RS) is 91.85%, the molecular weight of the free base is 459.9, and the molecular weight of the hydrochloride is 496.4. Calculated based on the hydrochloride content in CoA, the doses used in mice in this study were 8.1 mg/kg and 16.2 mg/kg respectively. The effective doses were converted into human doses of 0.7 mg/kg and 1.4 mg/kg based on the body surface area dose. . Instructions on the dosage of WWZ: The current drug name of WWZ in human clinical use is Wuester Capsule (TCM), and its preparation is a capsule, each capsule containing WWZ 11.25mg. The clinical usage and dosage is two capsules each time, three times a day, and the daily dosage is 67.5mg. This dose is the highest clinically used daily dose for adults (60kg). This highest dose is converted into a dose for mice in this study by body surface area dose. In this study, mice were given a dose group of WWZ. , that is, the highest effective dose is 25.0mg/kg.
实验期间每周2次测量并记录肿瘤体积及体重。实验结束时,统计肿瘤体积计算肿瘤生长抑制率TGI和相对肿瘤增殖率T/C%,每周使用游标卡尺对肿瘤体积进行两次测量,测量肿瘤的长径和短径,其体积计算公式为:肿瘤体积=0.5×长径×短径2。
肿瘤生长抑制率(TGI,%)=(1-T(治疗组肿瘤体积)/C(溶剂对照组肿瘤体积))×100。
相对肿瘤增殖率(T/C,%)=治疗组RTV/溶剂对照组RTV。During the experiment, tumor volume and body weight were measured and recorded twice a week. At the end of the experiment, the tumor volume was counted to calculate the tumor growth inhibition rate TGI and relative tumor proliferation rate T/C%. The tumor volume was measured twice a week using a vernier caliper to measure the long and short diameters of the tumor. The volume calculation formula is: Tumor volume = 0.5 × long diameter × short diameter 2 .
Tumor growth inhibition rate (TGI, %) = (1-T (tumor volume of treatment group)/C (tumor volume of solvent control group)) × 100.
Relative tumor proliferation rate (T/C, %) = RTV of treatment group/RTV of solvent control group.
试验结果如表9所示。The test results are shown in Table 9.
表9:
注:评价标准:T/C(%)>40%为无效;T/C(%)≤40%;TGI:肿瘤抑制率;T/C%:相对肿瘤增值
率。Table 9:
Note: Evaluation criteria: T/C (%) > 40% is invalid; T/C (%) ≤ 40%; TGI: tumor inhibition rate; T/C%: relative tumor growth rate.
表9的数据显示,灌胃给予WWZ(25.0mg/kg)、供试药物(8.1mg/kg)组、供试药物(16.2mg/kg)组、WWZ(25.0mg/kg)+供试药物(8.1mg/kg)、WWZ(25.0mg/kg)+供试药物(16.2mg/kg)组,在D34,除WWZ(25.0mg/kg)组外,其余处理组肿瘤生长抑制率(按肿瘤体积及肿瘤重量计算)均大于60%,T/C均小于40%。提示灌胃给与供试药物(8.1mg/kg)、供试药物(16.2mg/kg)、WWZ(25.0mg/kg)+供试药物(8.1mg/kg)、WWZ(25.0mg/kg)+供试药物(16.2mg/kg)对人肺癌PC9裸鼠移植瘤的肿瘤生长具有显著的抑制作用。The data in Table 9 shows that the groups of WWZ (25.0mg/kg), test drug (8.1mg/kg), test drug (16.2mg/kg), WWZ (25.0mg/kg) + test drug were administered intragastrically. (8.1mg/kg), WWZ (25.0mg/kg) + test drug (16.2mg/kg) group, on D34, except for the WWZ (25.0mg/kg) group, the tumor growth inhibition rates of the other treatment groups (according to tumor Volume and tumor weight) were both greater than 60%, and T/C were both less than 40%. Prompt to administer test drug (8.1mg/kg), test drug (16.2mg/kg), WWZ (25.0mg/kg) + test drug (8.1mg/kg), WWZ (25.0mg/kg) by gavage +The test drug (16.2 mg/kg) has a significant inhibitory effect on the tumor growth of human lung cancer PC9 transplanted tumors in nude mice.
在本说明书的描述中,参考术语“一个实施例”、“一些实施例”、“示例”、“具体示例”、或“一些示例”等的描述意指结合该实施例或示例描述的具体特征、结构、材料或者特点包含于本发明的至少一个实施例或示例中。在本说明书中,对上述术语的示意性表述不必须针对的是相同的实施例或示例。而且,描述的具体特征、结构、材料或者特点可以在任一个或多个实施例或示例中以合适的方式结合。此外,在不相互矛盾的情况下,本领域的技术人员可以将本说明书中描述的不同实施例或示例以及不同实施例或示例的特征进行结合和组合。In the description of this specification, reference to the terms "one embodiment," "some embodiments," "an example," "specific examples," or "some examples" or the like means that specific features are described in connection with the embodiment or example. , structures, materials or features are included in at least one embodiment or example of the invention. In this specification, the schematic expressions of the above terms are not necessarily directed to the same embodiment or example. Furthermore, the specific features, structures, materials or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, those skilled in the art may combine and combine different embodiments or examples and features of different embodiments or examples described in this specification unless they are inconsistent with each other.
尽管上面已经示出和描述了本发明的实施例,可以理解的是,上述实施例是示例性的,不能理解为对本发明的限制,本领域的普通技术人员在本发明的范围内可以对上述实施例进行变化、修改、替换和变型。 Although the embodiments of the present invention have been shown and described above, it can be understood that the above-mentioned embodiments are illustrative and should not be construed as limitations of the present invention. Those of ordinary skill in the art can make modifications to the above-mentioned embodiments within the scope of the present invention. The embodiments are subject to changes, modifications, substitutions and variations.
Claims (54)
A pharmaceutical composition, characterized by comprising: a compound represented by formula I or a salt thereof and biphenylcyclooctadienolignan
A single dosage form, characterized by comprising: a compound represented by formula I or a salt thereof and biphenylcyclooctadienolignan
A drug combination, characterized by comprising: a compound represented by formula I or a salt thereof as the first active ingredient and biphenylcyclooctadienolignan as the second active ingredient
A pharmaceutical kit, characterized by comprising: a compound represented by Formula I or a salt thereof as the first active ingredient and biphenylcyclooctadienolignan as the second active ingredient
A method of administering a compound represented by formula I or a salt thereof, which is characterized in that the compound represented by formula I or a salt thereof is combined with biphenylcyclooctadienolignans
A method for extending the half-life of a compound represented by Formula I or a salt thereof, which is characterized in that the compound represented by Formula I or a salt thereof is combined with biphenylcyclooctadiene lignan
A method for reducing the compound represented by formula I or a salt thereof to produce a compound represented by formula II, characterized in that the compound represented by formula I or a salt thereof is combined with biphenylcyclooctadienolignans
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN202210799853.0 | 2022-07-06 | ||
| CN202210799853.0A CN115813927A (en) | 2022-07-06 | 2022-07-06 | Combined medicine of receptor tyrosine kinase inhibitor and biphenyl cyclooctadiene diluted lignan and application thereof |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024007801A1 true WO2024007801A1 (en) | 2024-01-11 |
Family
ID=85522740
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2023/098571 Ceased WO2024007801A1 (en) | 2022-07-06 | 2023-06-06 | Combination therapy of receptor tyrosine kinase inhibitor and biphenyl cyclooctadiene lignan and use thereof |
Country Status (2)
| Country | Link |
|---|---|
| CN (1) | CN115813927A (en) |
| WO (1) | WO2024007801A1 (en) |
Families Citing this family (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN115813927A (en) * | 2022-07-06 | 2023-03-21 | 江苏晨泰医药科技有限公司 | Combined medicine of receptor tyrosine kinase inhibitor and biphenyl cyclooctadiene diluted lignan and application thereof |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1994289A (en) * | 2006-01-05 | 2007-07-11 | 胡汛 | Application of dibenzocyclootadiene lignans in preparation of tyrosine-inhibiting medicine |
| CN105209456A (en) * | 2013-03-06 | 2015-12-30 | 阿斯利康(瑞典)有限公司 | Quinazoline inhibitors of activating mutant forms of epidermal growth factor receptor |
| CN115813927A (en) * | 2022-07-06 | 2023-03-21 | 江苏晨泰医药科技有限公司 | Combined medicine of receptor tyrosine kinase inhibitor and biphenyl cyclooctadiene diluted lignan and application thereof |
-
2022
- 2022-07-06 CN CN202210799853.0A patent/CN115813927A/en active Pending
-
2023
- 2023-06-06 WO PCT/CN2023/098571 patent/WO2024007801A1/en not_active Ceased
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1994289A (en) * | 2006-01-05 | 2007-07-11 | 胡汛 | Application of dibenzocyclootadiene lignans in preparation of tyrosine-inhibiting medicine |
| CN105209456A (en) * | 2013-03-06 | 2015-12-30 | 阿斯利康(瑞典)有限公司 | Quinazoline inhibitors of activating mutant forms of epidermal growth factor receptor |
| CN115813927A (en) * | 2022-07-06 | 2023-03-21 | 江苏晨泰医药科技有限公司 | Combined medicine of receptor tyrosine kinase inhibitor and biphenyl cyclooctadiene diluted lignan and application thereof |
Non-Patent Citations (7)
| Title |
|---|
| AL-SHAKLIAH NASSER S., KADI ADNAN A., ALJOHAR HAYA I., ALRABIAH HAITHAM, ATTWA MOHAMED W.: "Profiling of in vivo , in vitro and reactive zorifertinib metabolites using liquid chromatography ion trap mass spectrometry", RSC ADVANCES, ROYAL SOCIETY OF CHEMISTRY, GB, vol. 12, no. 32, 14 July 2022 (2022-07-14), GB , pages 20991 - 21003, XP093126388, ISSN: 2046-2069, DOI: 10.1039/D2RA02848D * |
| ATTWA MOHAMED W., AL-SHAKLIAH NASSER S., ALRABIAH HAITHAM, KADI ADNAN A., ABDELHAMEED ALI S.: "Estimation of zorifertinib metabolic stability in human liver microsomes using LC–MS/MS", JOURNAL OF PHARMACEUTICAL AND BIOMEDICAL ANALYSIS, ELSEVIER B.V., AMSTERDAM, NL, vol. 211, 1 March 2022 (2022-03-01), AMSTERDAM, NL , pages 114626, XP093126398, ISSN: 0731-7085, DOI: 10.1016/j.jpba.2022.114626 * |
| GE FENG, ZHAI JIAN-XIU; CHEN WAN-SHENG; XIONG XIAO-JUAN; GAO SHOU-HONG , YANG YANG, XIA TIAN-YI, ZHANG FENG: "Study progress of the interactions between the Schisandra chinensis and the Schisandra sphenanthera as well as their preparations with other drugs", CHINESE JOURNAL OF HOSPITAL PHARMACY, vol. 37, no. 21, 12 October 2017 (2017-10-12), pages 2206 - 2209, 2215, XP093126672 * |
| GUANGJIAN ZHANG, WANG DECAI: "Research progress on cytochrome P4503A4 and drug metabolism", JOURNAL OF TAISHAN MEDICAL COLLEGE, vol. 33, no. 10, 25 October 2012 (2012-10-25), pages 757 - 760, XP093126668 * |
| TING-TING GUO, ZHU SHUANG-YUAN; LI MING; TAN SHAO-YING: "An Effective New Drug in Treatment of Non-small Cell Lung Cancer with Brain Metastases(BM) and Leptomeningeal Metastases(LM): Zorifertinib", GUANGZHOU CHEMISTRY, vol. 46, no. 2, 28 April 2021 (2021-04-28), pages 1 - 5, 59, XP093126675, DOI: 10.16560/j.cnki.gzhx.20210212 * |
| WEN XU, LIU TAO; ZHAO ZHENHUAN; CAO ZHIHONG; LYU ZHIQIANG; MA MIN; MA LIN: "Effects of Wuzhi Soft Capsule Combined with Imatinib Mesylate Tablet on Pharmacokinetics of Imatinib in Rats after Administration", CHINA PHARMACY, ZHONGHUA YIYUAN GUANLI XUEHUI, CN, vol. 30, no. 9, 15 May 2019 (2019-05-15), CN , pages 1198 - 1202, XP093126663, ISSN: 1001-0408, DOI: 10.6039/j.issn.1001-0408.2019.09.10 * |
| YADONG FU, CHEN JIAMEI; LIU WEI; LIU PING: "Research Progress of Schisantherin A in Regulating Hepatic Drug-metabolizing Enzymes and Its Pharmacological Effects", TRADITIONAL CHINESE DRUG RESEARCH AND CLINICAL PHARMACOLOGY, vol. 31, no. 6, 28 May 2020 (2020-05-28), pages 741 - 748, XP093126660, DOI: 10.19378/j.issn.1003-9783.2020.06.019 * |
Also Published As
| Publication number | Publication date |
|---|---|
| CN115813927A (en) | 2023-03-21 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP2013529654A (en) | Pharmaceutical composition comprising levocarnitine and dobesylate | |
| EP1358177B1 (en) | Treatment of affective disorders by the combined action of a nicotinic receptor agonist and a monoaminergic substance | |
| TW202023563A (en) | Novel quinazoline egfr inhibitors | |
| JP5514123B2 (en) | Combination drug containing paclitaxel for the treatment of ovarian cancer | |
| CA2664935A1 (en) | Compositions for the treatment of hepatitis c and methods for using compositions for the treatment of hepatitis c | |
| US11084790B2 (en) | Insoluble complex or solvate thereof, pharmaceutical composition and use thereof | |
| WO2024007801A1 (en) | Combination therapy of receptor tyrosine kinase inhibitor and biphenyl cyclooctadiene lignan and use thereof | |
| US8748488B2 (en) | Methods and compositions for administration of oxybutynin | |
| WO2009015561A1 (en) | The use of leonurine and compositions thereof | |
| EP4541353A1 (en) | Pharmaceutical composition for treating and resisting blood coagulation and use thereof | |
| CN111349111A (en) | Pentazocine prodrug, preparation method and application thereof | |
| EP1655029B1 (en) | Medicinal compositions | |
| JP2025011312A (en) | Method for preventing precipitation of injection solutions containing p-boronophenylalanine | |
| US20230321085A1 (en) | Compound for use in the treatment of dry mouth | |
| KR102905321B1 (en) | Compounds for use in the treatment of dry mouth | |
| US20250387405A1 (en) | Pharmaceutical composition containing platinum drugs or platinum drug cocrystals, and use thereof | |
| WO2013090319A2 (en) | Treatment of type i and type ii diabetes | |
| JPH09500375A (en) | Use of efaroxan and its derivatives for the manufacture of a medicament for the treatment of Parkinson's disease | |
| KR20210046115A (en) | Niacin derivative and pharmaceutical composition containing the same | |
| US20200061053A1 (en) | Pharmaceutical composition and method for acute on chronic liver failure and related liver diseases | |
| WO2009104152A1 (en) | Combination treatment for ovarian cancer | |
| CN119326894A (en) | Use of GSDME inhibitors for manufacturing drugs for preventing and treating peripheral nerve damage during chemotherapy | |
| HK40085950A (en) | Compound for use in the treatment of dry mouth | |
| WO2023093837A1 (en) | Pharmaceutical composition containing platinum drugs or platinum drug co-crystals, and use thereof | |
| CN113599387A (en) | Compound preparation and application thereof in preparing medicament for treating angina |
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: 23834575 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: 23834575 Country of ref document: EP Kind code of ref document: A1 |