WO2015184661A1 - 化合物及其制备方法和用途 - Google Patents

化合物及其制备方法和用途 Download PDF

Info

Publication number
WO2015184661A1
WO2015184661A1 PCT/CN2014/080887 CN2014080887W WO2015184661A1 WO 2015184661 A1 WO2015184661 A1 WO 2015184661A1 CN 2014080887 W CN2014080887 W CN 2014080887W WO 2015184661 A1 WO2015184661 A1 WO 2015184661A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
disease
substituted
unsubstituted
Prior art date
Application number
PCT/CN2014/080887
Other languages
English (en)
French (fr)
Inventor
许勇
王学海
李莉娥
黄璐
田华
杨仲文
夏庆丰
肖强
郭涤亮
涂荣华
余艳平
于静
黄翔
范昭泽
何震宇
张毅
杨菁
Original Assignee
人福医药集团股份公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 人福医药集团股份公司 filed Critical 人福医药集团股份公司
Publication of WO2015184661A1 publication Critical patent/WO2015184661A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/16Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two nitrogen atoms

Definitions

  • the present invention relates to the field of medicine, and in particular, the present invention relates to a compound, a preparation method thereof and use thereof, and more particularly, to a compound, a preparation method thereof, a pharmaceutical composition, a compound and a pharmaceutical composition for use in the preparation of a medicament.
  • Background technique
  • BTK Bruton's tyrosine kinase
  • BCR B cell receptor
  • the BTK inhibitor ibrutinib was approved by the FDA for the treatment of mantle cell lymphoma in November 2013 and was approved for the treatment of chronic lymphocytic leukemia in February 2014. Ibrutinib is capable of selectively and irreversibly forming a strong covalent bond with the cysteine residue on BTK, inhibiting the transmission of overactive cell survival signals in malignant B cells to achieve anticancer effects. ibrutinib is currently the only one. Listed BTK inhibitors. However, ibrutinib has low solubility, high plasma protein binding rate (PPB), and low oral bioavailability, which directly leads to poor efficacy after oral administration.
  • PPB plasma protein binding rate
  • the present invention is directed to solving at least some of the above technical problems or at least providing a useful commercial option. To this end, it is an object of the present invention to provide a compound for use as a kinase inhibitor.
  • Another object of the present invention is to provide a process for the preparation of a compound of formula I, or a stereoisomer, tautomer, pharmaceutically active metabolite, pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof, and intermediate thereof body.
  • Another object of the present invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention, the pharmaceutical composition further comprising one or more pharmaceutically acceptable excipients and a therapeutically effective amount of at least one of the present invention
  • Another object of the present invention is to provide a compound of the present invention, or a stereoisomer, tautomer, pharmaceutically active metabolite, pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof for use in the preparation of a medicament .
  • the medicament as a BTK inhibitor, can treat a disease mediated by B cells, and is used for preparing a tumor disease, a proliferative disease, an allergic disease, an autoimmune disease, and an inflammatory disease. drug.
  • It is yet another object of the invention to provide a method of treating a disease mediated by a B cell.
  • the compound or pharmaceutical composition of the present invention has high BTK kinase inhibitory activity and is effective for treating a disease mediated by B cells.
  • the invention proposes a compound.
  • the compound is a stereoisomer, tautomer, pharmaceutically active metabolite, pharmaceutically acceptable salt, hydrate, solvate or a compound of formula I or a compound of formula I or
  • X is one selected from the group consisting of substituted or unsubstituted phenyl, substituted or unsubstituted 3 to 7 membered saturated or unsaturated carbocyclic ring, substituted or unsubstituted 8 to 10 membered saturated or unsaturated double ring Or an aryl ring, a substituted or unsubstituted 5- to 6-membered monocyclic heteroaryl ring having 1 to 4 heteroatoms, a substituted or unsubstituted 4 to 7 membered saturated with 1 to 3 heteroatoms or Unsaturated heterocyclic ring, substituted or unsubstituted 7- to 10-membered saturated or unsaturated bicyclic heterocyclic ring having 1 to 5 hetero atoms, substituted or unsubstituted 8- to 10-membered bicyclic ring having 1 to 5 hetero atoms Heteroaryl ring; selected from -R, halogen, -OR, -0(CH 2 )aOR,
  • the chemical bond is a point at which a moiety or substituent is attached to a core structure or a skeletal structure.
  • stereoisomer as used in the present invention may include a compound which is an optical isomer by possessing one or more chiral atoms, and is optically isomerized by limited rotation around one or more bonds. Compound of the body.
  • the definition of a compound of the invention encompasses all possible stereoisomers and mixtures thereof. The racemic form and the isolated optical isomers having specific activities are very specifically contemplated.
  • the racemic form can be resolved by physical methods including, but not limited to, fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography.
  • the individual optical isomers can be obtained from the racemate by conventional methods including, but not limited to, formation of a salt with an optically active acid followed by crystallization.
  • the compounds of the formula I and their salts may exist in their tautomeric form, in which the hydrogen atom is transferred to other parts of the molecule, and the atoms in the molecule The chemical bond between them is thus rearranged. It should be understood that all tautomeric forms (as long as they may be present) are included in the present invention. Further, the compound of the formula I of the present invention may have a trans isomer and a cis isomer.
  • pharmaceutically acceptable salt is a conventional non-toxic salt formed by reacting a compound of the formula I with an inorganic or organic acid.
  • the conventional non-toxic salt can be obtained by reacting a compound of the formula I with an inorganic or organic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, aminosulfonic acid and phosphoric acid.
  • the organic acid includes citric acid, tartaric acid, lactic acid, pyruvic acid, acetic acid, benzenesulfonic acid, p-toluenesulfonic acid, methanesulfonic acid, naphthalenesulfonic acid, ethanesulfonic acid, naphthalene disulfonic acid, maleic acid, malic acid , malonic acid, fumaric acid, succinic acid, propionic acid, oxalic acid, trifluoroacetic acid, stearic acid, pamoic acid, hydroxymaleic acid, phenylacetic acid, benzoic acid, salicylic acid, glutamic acid, ascorbic acid, Aminobenzenesulfonic acid, 2-acetoxybenzoic acid, isethionate, etc.; or a compound of formula I with propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid,
  • prodrug means that once the compound is administered to a subject, the compound is chemically converted by metabolic or chemical processes to provide a compound of formula I and/or Salt and / or solvate. Any compound that can be converted in vivo to provide a biologically active substance (ie, a compound of Formula I) is in the present invention Prodrugs within the scope and subject matter. For example, a compound containing a carboxyl group can form a physiologically hydrolyzable ester which is prepared by hydrolysis in vivo to give the compound of formula I itself.
  • the prodrug is preferably administered orally because hydrolysis occurs in many cases primarily under the influence of digestive enzymes. Parenteral administration can be used when the ester itself is active or hydrolysis occurs in the blood.
  • hydrates, solvates e.g., methanolates, DMSOs
  • Methods of solvation are well known in the art.
  • the invention provides a compound of formula I, or a stereoisomer, tautomer, pharmaceutically active metabolite, pharmaceutically acceptable salt, hydrate, solvate or former thereof.
  • Methods and intermediates for pharmaceuticals are described in detail below.
  • a method of preparing a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically active metabolite, pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof comprises: The compound of the formula 1 is contacted with the compound of the formula 2 to obtain the compound of the formula 3; (2) the compound of the formula 3 is contacted with the compound of the formula 4 (acryloyl chloride), To obtain a compound of the formula 5; (3) contacting the compound of the formula 5 with a compound of the formula 6a to obtain a compound of the formula I.
  • the inventors have found that the compounds of formula I, or their stereoisomers, tautomers, pharmaceutically active metabolites, pharmaceutically acceptable salts, hydrates, solvates or pre-forms, can be prepared rapidly and efficiently using the process of the invention.
  • the medicine has short synthetic route, high environmental friendliness, high yield and purity of target product, easy availability of raw materials, simple operation and post-treatment, and suitable for industrial production.
  • the compound of formula 1 is contacted with a compound of formula 2 to obtain a compound of formula 3; Specifically, a compound of the formula 1, a compound of the formula 2, and tetrahydrofuran (THF) are added to a three-necked flask, and then cesium carbonate is added thereto, and the mixture is refluxed for 12 to 24 hours. After the reaction is completed, the oil is concentrated to dissolve in methanol. It was recrystallized from acetone, and the solid was precipitated, filtered, and dried to give the compound of formula 3 without further purification.
  • THF tetrahydrofuran
  • the compound of the formula 3 is contacted with the compound of the formula 4 to obtain a compound of the formula 5; specifically, the compound of the formula 3 and N-methylpyrrolidone (NMP) are at -5 ° C. mixing at ⁇ 5 ° C, was slowly added the compound of formula 4, and incubated the reaction was stirred at 0 ° C 60 min, then water was added and stirring was continued for half an hour, followed by addition of a saturated NaHC0 3 aqueous solution was basified, then extracted with ethyl acetate Extract 3 times. Washed with water, washed with brine and the combined ethyl acetate extracts were dried, Na 2 S0 4 and concentrated under reduced pressure to obtain the compound of formula 5.
  • NMP N-methylpyrrolidone
  • the compound of the formula 5 is contacted with the compound of the formula 6a to obtain a compound of the formula I; specifically, the compound of the formula 5, the compound of the formula 6a (the compound of the formula 5 and the compound of the formula 6a)
  • the molar ratio of the compound is 1:1.5).
  • Add the solution of N-methylpyrrolidone to microwave irradiation 100 ° C ⁇ 20 (TC, 5-30 min). Cool the reaction mixture, dilute with water, and use acetic acid The ester was extracted three times. The combined ethyl acetate extracts were washed with water and brine, dried over Na 2 SO 4 and concentrated under reduced pressure, then dissolved in isopropyl alcohol and recrystallized at -5 ° C to 5 ° C. Filtration gave a white solid which was dried under reduced pressure to give the compound of formula I.
  • the compound of formula I may be: a compound of formula I or a stereoisomer, tautomer, pharmaceutically active metabolite, pharmaceutically acceptable salt, hydrate, solvent thereof Compound or prodrug.
  • the inventors of the present invention have found that the compounds of the present invention also exhibit improved solubility and properties which are less likely to emanate from cells, thereby improving bioavailability.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I or a stereoisomer, tautomer, pharmaceutically active metabolite, pharmaceutically acceptable salt thereof, At least one of a hydrate, a solvate or a prodrug.
  • the pharmaceutical composition may further comprise a pharmaceutically acceptable excipient.
  • the pharmaceutical composition may further comprise conventional additives such as odorants, flavoring agents and the like.
  • the pharmaceutical composition provided by the present invention preferably contains 0.1% to 99% by weight of the compound of the formula I as an active ingredient, and more preferably, the compound of the formula I as an active ingredient accounts for 10% by weight of the total weight of the pharmaceutical composition. ⁇ 40%, the remainder being a pharmaceutically acceptable carrier and/or conventional additives.
  • the compounds and pharmaceutical compositions provided by the present invention may be in various forms such as tablets, capsules, injections, powders for injection, powders, syrups, solutions, suspensions and aerosols, and the like, and may be present in suitable solids. Or a liquid carrier or diluent is neutralized in a suitable sterilizing device for injection or drip.
  • compositions of the present invention can be prepared according to conventional methods of preparation in the pharmaceutical arts.
  • the compounds and pharmaceutical compositions of the invention may be used clinically in mammals, including humans and animals, through the mouth, nose, skin, lungs or Administration by the gastrointestinal tract or the like.
  • the optimal dosage for the individual should be based on the specific treatment regimen. Usually starting with a small dose, gradually increase the dose until the most suitable dose is found. The most preferred route of administration is oral.
  • the invention provides a compound of formula I, or a stereoisomer, tautomer, pharmaceutically active metabolite, pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof, in the preparation of Use in medicine.
  • the medicament is used as a kinase inhibitor.
  • the compounds of the invention may modulate kinase activity, including modulation of BTK, and may be useful as kinase inhibitors, and other types of kinase activities that may be modulated by the compounds of the invention include, but are not limited to, members of the Tec family, such as BMX, BTK, ITK, Txk and Tec and their mutants. Therefore, the above drugs are effective as BTK inhibitors, and are capable of treating B cell mediated diseases for treating tumor diseases, proliferative diseases, allergic diseases, autoimmune diseases, and inflammatory diseases.
  • the compound of the formula I of the present invention has a higher BTK kinase inhibitory activity than the existing BTK inhibitor, ibrutinib. Further, after the compound of the formula I of the present invention was pretreated with recombinant BTK, it was repeatedly washed with a medium containing no inhibitor, and its activity was not restored, indicating that BTK was irreversibly inhibited by the compound of the present invention.
  • the medicament of the present invention is useful for treating one or more diseases associated with BTK activity, including but not limited to solid tumors, lymphomas, soft tissue sarcomas, lymphocytic lymphomas, mantle cell lymphomas. , melanoma, multiple myeloma, acute lymphocytic leukemia, chronic lymphocytic leukemia, acute myeloid leukemia, chronic myelogenous leukemia, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, systemic lupus erythematosus, Psoriasis, rheumatoid spondylitis, gouty arthritis, etc.
  • diseases associated with BTK activity including but not limited to solid tumors, lymphomas, soft tissue sarcomas, lymphocytic lymphomas, mantle cell lymphomas.
  • melanoma multiple myeloma
  • acute lymphocytic leukemia chronic lymphoc
  • the compounds of the examples of the present invention exhibited better therapeutic effects than ibrutinib.
  • the compound of formula I used as a kinase inhibitor according to the present invention successfully overcomes the defects of low solubility and low oral bioavailability of the existing BTK inhibitor ibrutinib, and has good clinical application. And medical use.
  • the invention provides a method of treating a disease mediated by B cells.
  • the method comprises: administering to a patient a compound or pharmaceutical composition as hereinbefore described.
  • the inventors have found that the compound or pharmaceutical composition described above has a high BTK kinase inhibitory activity and can be effectively used for the treatment of diseases mediated by B cells.
  • the kind of the disease mediated by B cells is not particularly limited.
  • the B cell mediated disease may be at least one selected from the group consisting of a tumor disease, a proliferative disease, an allergic disease, an autoimmune disease, and an inflammatory disease. Thereby, a better therapeutic effect can be obtained.
  • the B cell mediated disease may be selected from the group consisting of solid tumor, lymphoma, soft tissue sarcoma, lymphocytic lymphoma, mantle cell lymphoma, melanoma, multiple myeloma, acute lymphocytic leukemia. , slow Lymphocytic leukemia, acute myeloid leukemia, chronic myelogenous leukemia, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, systemic lupus erythematosus, psoriasis, rheumatoid spondylitis and gouty arthritis At least one. Thereby, the therapeutic effect is better.
  • An embodiment of the invention provides a compound of formula I, or a stereoisomer, tautomer, pharmaceutically active metabolite, pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof, for the preparation of a compound of formula I Or methods, intermediates, pharmaceutical compositions, and compounds of the invention, or stereoisomers, tautomers, pharmaceutically active metabolites, pharmaceutically acceptable salts, hydrates, solvates or prodrugs thereof
  • a compound of the formula 1 (17.0 g, 0.1 mol), a compound of the formula 2 (11.1 g, 0.11 mol), and tetrahydrofuran (800 ml) were added to a three-necked flask, followed by cesium carbonate (39.1 g, 0.12 mol). After refluxing for 15 hours, after completion of the reaction, the oily solution was concentrated, firstly dissolved in methanol (60 liters), and then recrystallized by stirring with acetone (180 liters) to precipitate a solid, which was filtered and dried to give the compound of formula (18.7) g, yield 74.0%), without further purification.
  • a compound of the formula 1 (17.0 g, 0.1 mol), a compound of the formula 2 (10.6 g, 0.105 mol), and tetrahydrofuran (800 ml) were added to a three-necked flask, followed by cesium carbonate (39.1 g, 0.12 mol). After refluxing for 24 hours, after completion of the reaction, the oil was evaporated to dryness eluting with EtOAc (EtOAc) (EtOAc) Rate 72.8%) without further purification.
  • EtOAc EtOAc
  • the compound of formula 3 (25.3 g, 0.1 mol) was mixed with N-methylpyrrolidone (300 ml) at -5 ° C to 5 ° C, and the compound of formula 4 (19.9 g, 0.22 mol) was slowly added.
  • the reaction was stirred at 0 ° C for 60 minutes, then water (50 ml) was added and stirring was continued for half an hour, followed by addition of a saturated NaHCO 3 solution to alkalinize the aqueous solution, followed by extraction with ethyl acetate three times, 300 liters each time. . Washed with water, washed with brine and the combined ethyl acetate extracts were dried, Na 2 S0 4 and concentrated under reduced pressure to give compound (23.9 g, yield 66.3%) of formula 5.
  • Example 5 Compound represented by -1
  • a compound of the formula (36.1 g, 0.1 mol) and a compound of the formula 6a-1 (15.2 g, 0.15 mol) were added to a solution of N-methylpyrrolidone (600 ml) for microwave irradiation (100°). C, 30 minutes).
  • the reaction mixture was cooled, diluted with water (500 mL) and then th
  • the combined ethyl acetate extracts were washed with water and brine, dried over Na 2 SO 4 and concentrated under reduced pressure, then dissolved in 300 ml of isopropyl alcohol, recrystallized at -5 ° C to 5 ° C, and filtered.
  • the white solid was dried under reduced pressure to give a compound of formula 1-1 (yield: 34.5 g, yield: 78.0%).
  • a compound of the formula (36.1 g, 0.1 mol) and a compound of the formula 6a-2 (30.8 g, 0.15 mol) were added to a solution of N-methylpyrrolidone (500 ml), and subjected to microwave irradiation (200 ° C, 5 minutes) .
  • the workup was carried out in the same manner as in Example 5 to give the compound of formula 1-3 (yield: 38.1 g, yield: 72.0%).
  • the preparation method of the compound of the formula 1-4 to the compound of the formula 1-26 is the same as the preparation of the compound of the formula 1-1 to the formula 1-3 in the same manner as in the example 5 to the embodiment.
  • Method of the indicated compound The molar ratio of the compound of the formula 5 to the compound of the formula 6a is 1:1.5.
  • a compound of the formula 6a-15 is used as a starting material, and a compound of the formula 5 is contacted with a compound of the formula 6a-15, and finally a corresponding compound of the formula 1-15 is obtained.
  • Specific compounds and data are shown in Table 1 below. %e'8z,
  • Human B cells were purified from 150 ml of blood. Specifically, the blood was diluted 1/2 with PBS and centrifuged through a Ficoll density gradient. B cells were isolated from monocytes by negative selection using B Cell Isolation Kit II from Milenyi (Auburn, CA). Then, 50000 B cells per well in a 96-well plate were stimulated with 10 g/ml goat F(ab')2 anti-human IgM antibody (Jackson ImmunoResearch Laboratories, West Grove, PA). The compound of the formula 1-1 to the formula 1-26 obtained in the preparation of Example 1-7 was diluted with DMSO (dimethyl sulfoxide) and added to the cells. The final concentration of DMSO was 0.5%. Proliferation was measured 3 days later using PromegaCellTiter-Glo (Madison, WI). As a result, it was found that the compound of the formula I of the present invention was tested to be active.
  • DMSO dimethyl sulfoxide
  • Test compounds human recombinant BTK (InM, Invitrogen Corporation), fluoresceinated peptide (1.5 ⁇ ), ATP (20 ⁇ ), and assay buffer (20 mM HEPES (pH 7.4) were added to a V-bottom 384-well plate. , 10 mM MgCl 2 , 0.015 % Brij 35 and 4 mM DTT in 1.6% DMSO) with a final volume of 30 L. After incubating for 60 minutes at room temperature, by adding 45 ⁇ 135 ⁇ to each sample Stop the reaction. The reaction mixture was analyzed by electrophoretic separation of the fluorogenic substrate and the pity acidified product on a Caliper LabChip 3000 (Caliper, Hopkinton, MA).
  • Inhibition data was calculated by comparison with an enzyme-free control reaction as 100% inhibition and a non-inhibitor control as 0% inhibition.
  • a dose response curve was generated to determine the concentration (IC50) required to inhibit 50% of kinase activity.
  • the compound was dissolved in dimethyl sulfoxide (DMSO) at 10 mM and then evaluated at 11 concentrations. Using this assay, the IC50 values of the compounds of the invention and the existing BTK inhibitor compound, ibrutinib, were determined. The results are shown in Table 2.
  • Table 2 shows the activity of selected compounds of the invention in an in vitro BTK kinase inhibition assay.
  • the compound number corresponds to the compound number in Table 1.
  • Compounds designated as "A” for activity provide IC50 ⁇ 10 nM; compounds designated as “B” for activity provide an IC50 of 10-100 nM; compounds designated as “C” for activity provide IC50 of 100-1000 nM;
  • the compound designated as "D” provides an IC50 of 1000-10000 nM; and the compound designated as ' ⁇ " has an IC50 > 10000 nM.
  • the compound of formula I of the present invention has a higher specificity than ibrutinib.
  • BTK kinase inhibitory activity after pretreatment of recombinant BTK with the compound of the present invention, repeated washing with an inhibitor-free medium did not restore its activity, indicating that BTK was irreversibly inhibited by these compounds.
  • Preparation of the emulsion The bovine collagen was dissolved in acetic acid to prepare a bovine collagen solution with a concentration of 8 mg/ml, and the temperature was 4 ° C overnight. Add an equal volume of complete Freund's adjuvant (CFA, 1 mg/ml) to the bovine collagen solution to prepare a bovine collagen CFA mixture with a final concentration of 4 mg/ml, and then mix it on a wet ice with a high-speed homogenizer. Slurry until a white emulsion is formed.
  • CFA complete Freund's adjuvant
  • mice On day 0, rats were injected with an emulsion of type II bovine collagen in complete Freund's adjuvant (CFA) and injected intradermally (i.d.) 50 microliters at several locations on the back. A booster injection (i.d.) of 50 microliters of collagen emulsion was provided at an alternative site at the base or back of the tail on day 7. Arthritis is usually observed from 12 days to 14 days after the initial collagen injection. From the 14th day, the progress of arthritis in animals can be evaluated as described below (evaluation of arthritis).
  • CFA complete Freund's adjuvant
  • Group 6 (Ibrutinib, 10 mg/kg) and Group 2 of the present invention (Compound of Formula I-2, 5 mg/kg), Group 5 (Compound of Formula 1-18, The inhibition rate was equivalent to 10 mg/kg.
  • the inhibition rate of Group 4 (Compound of Formula 1-11, 10 mg/kg) of the present invention was significantly higher than that of Group 6 (Ibrutinib, 10 mg/kg).
  • the water solubility of the compound 1-2 of the present invention was compared with that of ibrutinib.
  • the measurement method is as follows:
  • the equilibrium solubility was determined in a buffered aqueous solution of pH 7.4.
  • the buffer solution having a pH of 7.4 was adjusted to pH 7.4 by using a sodium hydroxide solution of 0.07 M with 10 mol/liter of sodium hydroxide, and the ionic strength of the solution was 0.15.
  • the sample was then filtered through a 0.45 ⁇ nylon syringe filter saturated with the sample and sampled continuously from the filtrate twice.
  • the filtrate was determined by HPLC using a standard solution prepared in 50% methanol as a reference.
  • the solubility of Compound 1-2 was 44 mg / ml, and the solubility of Ibrutinib was 12 mg /
  • Example 12 Capsules for oral administration
  • the ingredients in the above table were mixed, and the active ingredient (the compound of the present invention) was dispensed in a capsule, and a total of 1000 g of powder was prepared and filled into capsules, and the active ingredient of each capsule was 200 mg.
  • Example 13 Capsules for oral administration Active ingredient 40.0%
  • Magnesium stearate 0.5% The ingredients in the above table were mixed, and the active ingredient (the compound of the present invention) was dispensed in a capsule, and a total of 1000 g of powder was prepared and filled into capsules, and the active ingredient of each capsule was 400 mg.
  • Example 14 Tablets for oral administration
  • the ingredients in the above table are mixed and granulated using a solvent such as ethanol.
  • the preparation was then dried, prepared into a total of 1000 g of a powder, and formed into tablets by a suitable tableting machine, and the active ingredient (the compound of the present invention) per tablet was 200 mg.
  • the active ingredient (the compound of the present invention) is dissolved in a part of water for injection. A sufficient amount of sodium chloride is then added with stirring to make the solution isotonic. The solution was made up to the weight with the remaining water for injection, filtered through a 0.2 micron membrane filter, and packaged under aseptic conditions to give an injection.
  • the description of the terms “one embodiment”, “some embodiments”, “example”, “specific example” or “some examples” and the like means a specific feature described in connection with the embodiment or example, A structure, material or feature is included in at least one embodiment or example of the invention.
  • the schematic representation of the above terms does not necessarily mean the same embodiment or example.
  • the particular features, structures, materials, or characteristics described may be combined in a suitable manner in any one or more embodiments or examples.

Abstract

一种化合物及其制备方法和用途被公开,该化合物为式I所示化合物或式I所示化合物的立体异构体、互变异构体、药物活性代谢物、可药用盐、水合物、溶剂化物或前药,其中X、R1,R2如说明书所定义。

Description

化合物及其制备方法和用途 技术领域
本发明涉及医药领域, 具体的, 本发明涉及化合物及其制备方法和用途, 更具体的, 本发明涉及化合物及其制备方法、药物组合物、化合物及药物组合物在制备药物中的用途。 背景技术
Bruton酪氨酸激酶 (Bruton's tyrosine kinase, BTK)是 Tec激酶家族的一种非受体酪氨酸 激酶, 是 B细胞发育、 激活、 信号传导和存活的关键调节物, 在 B细胞受体(BCR)信号 转导中发挥重要作用。 在激活 BCR时, BTK首先被其他酪氨酸激酶 (如 Lyn与 SYK)活化, 导致 B细胞增殖与分化的必需转录因子获得活化。除此之外, BTK也参与了与 B细胞迁移 及黏附相关的受体信号转导, 其中包括趋化因子受体 CXCR4、 CXCR5与黏附分子 (整合 素) 。 BTK激酶活跃失控则导致细胞无规则繁殖而引发或增进癌变。
BTK抑制剂 ibrutinib于 2013年 11月获 FDA批准用于治疗套细胞淋巴瘤, 2014年 2 月获批治疗慢性淋巴细胞白血病。 Ibrutinib能够与 BTK上的半胱氨酸残基选择性、 不可逆 性的形成强有力共价键, 抑制恶性 B细胞中过度活跃的细胞生存信号的传输从而达到抗癌 的功效, ibrutinib也是目前唯一一个上市的 BTK抑制剂。 但是 ibrutinib溶解度低、 与血浆 蛋白的结合率 (PPB) 高、 口服生物利用度低, 直接导致了患者口服后疗效不高。
因此, 目前的 BTK抑制剂仍有待改进。 发明内容
本发明旨在至少在一定程度上解决上述技术问题之一或至少提供一种有用的商业选 择。 为此, 本发明的一个目的在于提出一种用作激酶抑制剂的化合物。
本发明的另一个目的是提供制备式 I所示化合物、 或其立体异构体、 互变异构体、 药 物活性代谢物、 可药用盐、 水合物、 溶剂化物或前药的方法及中间体。
本发明的另一个目的是提供一种包含本发明的化合物的药物组合物, 所述药物组合物 进一歩包含一种或多种药学上可接受的赋形剂和治疗有效量的至少一种本发明的化合物、 或其立体异构体、 互变异构体、 药物活性代谢物、 可药用盐、 水合物、 溶剂化物或前药。
本发明的另一个目的是提供本发明的化合物、 或其立体异构体、 互变异构体、 药物活 性代谢物、 可药用盐、 水合物、 溶剂化物或前药在制备药物中的用途。 根据本发明的实施 例, 所述药物作为 BTK抑制剂, 可治疗由 B细胞介导的疾病, 用于制备***疾病、增 殖性疾病、 ***反应性疾病、 自身免疫性疾病和炎症性疾病的药物。 本发明的再一个目的是提供治疗由 B细胞介导的疾病的方法。 根据本发明的实施例, 本发明的化合物或药物组合物具有较高的 BTK激酶抑制活性,能够有效用于治疗由 B细胞 介导的疾病。
本发明所示化合物的这些和其它目的、 特点和优点在随后本专利公开的详细说明中披 下面详细描述本发明:
在本发明的第一方面, 本发明提出了一种化合物。 根据本发明的实施例, 所述化合物 为式 I所示化合物或式 I所示化合物的立体异构体、互变异构体、药物活性代谢物、可药用 盐、 水合物、 溶剂化物或
Figure imgf000004_0001
其中, X为选自下列之一: 取代或未取代的苯基、 取代或未取代的 3元到 7元饱和或 不饱和碳环、取代或未取代的 8元到 10元饱和或不饱和双环或芳基环、取代或未取代的具 有 1到 4个杂原子的 5元到 6元单环杂芳基环、 取代或未取代的具有 1到 3个杂原子的 4 元到 7元饱和或不饱和杂环、取代或未取代的具有 1到 5个杂原子的 7元到 10元饱和或不 饱和双环杂环、取代或未取代的具有 1到 5个杂原子的 8元到 10元双环杂芳基环; 为选 自 -R、 卤素、 -OR、 -0(CH2)aOR、 -CN、 -N02、 -S02R、 -S02N(R)2、 -SOR、 -C(0)R、 -C02R、 -C(0)N(R)2、 -NRC(0)R、 - RC(0) R2> -NRS02R和 -N(R)2中的任意一种; R2为选自 -R、 卤素、 -OR、 -0(CH2)aOR、 -CN、 -N02、 -S02R、 -S02N(R)2、 -SOR、 -C(0)R、 -C02R、 -C(0)N(R)2、 - RC(0)R -NRC(0)NR2 -NRS02R和 -N(R)2中的任意一种; 其中, a为 1至 5的整数, R 基团各自独立地为选自下列之一: 氢、 取代或未取代的 脂肪族基、 取代或未取代的苯 基、 取代或未取代的具有 1到 2个杂原子的 4元到 7元杂环、 取代或未取代的具有 1到 4 个杂原子的 5元到 6元单环杂芳基环, 所述杂原子各自独立地选自氮、氧和硫的任意一种。
进一步的, 根据本发明的实施例, ' 为选自下列之 
Figure imgf000005_0001
于描绘化学键, 所述化学键为部分或取代基与核心结构或骨架结构相连的点。
由此, 在本说明书通篇中, 本领域技术人员可对 基团及其取代基进行选择, 以提供本发明的实施例中所述的、 稳定的式 I所示化合物或其立体异构体、 互变异构体、 药物活性代谢物、 可药用盐、 水合物、 溶剂化物或前药。
在本发明中所使用的术语, 将本发明化合物的所有立体异构体 (无论是混合物形式还 是纯的形式或基本纯的形式) 都考虑在内。 在本发明中所使用的术语"立体异构体"可包括 通过拥有一个或多个手性原子而为光学异构体的化合物, 以及通过围绕一个或多个键受限 旋转而为光学异构体的化合物。 本发明化合物的定义涵盖所有可能的立体异构体和它们的 混合物。 非常具体地涵盖外消旋形式和具有特定活性的经分离的光学异构体。 可通过物理 方法拆分外消旋形式, 所述物理方法包括但不限于分级结晶、 非对映异构衍生物的分离或 结晶或通过手性柱色谱法分离。 可通过常规方法由外消旋体获得单独的光学异构体, 所述 常规方法包括但不限于与光学活性酸形成盐, 然后结晶。
在本发明中所使用的术语, 式 I所示化合物及其盐可按它们的互变异构形式存在, 在 所述互变异构形式中氢原子转移到分子的其它部分, 并且分子中原子之间的化学键因此发 生重排。 应该理解的是, 所有互变异构形式 (只要它们可以存在) , 就包括在本发明中。 此外, 本发明式 I所示化合物可具有反式异构体和顺式异构体。
在本发明中所使用的术语, "可药用盐"为通式 I所示化合物与无机酸或有机酸反应形成 的常规的无毒盐。 例如, 所述常规的无毒盐可通过通式 I所示化合物与无机酸或有机酸反 应制得, 所述无机酸包括盐酸、 氢溴酸、 硫酸、 硝酸、 胺基磺酸和磷酸等, 以及所述有机 酸包括柠檬酸、 酒石酸、 乳酸、 丙酮酸、 乙酸、 苯磺酸、 对甲苯磺酸、 甲磺酸、 萘磺酸、 乙磺酸、 萘二磺酸、 马来酸、 苹果酸、 丙二酸、 富马酸、 琥珀酸、 丙酸、 草酸、 三氟乙酸、 硬酯酸、 扑酸、 羟基马来酸、 苯乙酸、 苯甲酸、 水杨酸、 谷氨酸、 抗坏血酸、 对胺基苯磺 酸、 2-乙酰氧基苯甲酸和羟乙磺酸等; 或者通式 I所示化合物与丙酸、 草酸、丙二酸、琥珀 酸、 富马酸、 马来酸、 乳酸、 苹果酸、 酒石酸、 柠檬酸、 天冬氨酸或谷氨酸形成酯后再与 无机碱形成的钠盐、 钾盐、 钙盐、 铝盐或铵盐; 或者通式 I所示化合物与有机碱形成的甲 胺盐、 乙胺盐或乙醇胺盐; 或者通式 I所示化合物与赖氨酸、 精氨酸、 鸟氨酸形成酯后再 与盐酸、 氢溴酸、 氢氟酸、 硫酸、 硝酸、 磷酸形成的对应的无机酸盐或与甲酸、 乙酸、 苦 味酸、 甲磺酸和乙磺酸形成的对应的有机酸盐。
在本发明中所使用的术语, "前药 "表示一旦将所述化合物给予受试者, 所述化合物就 通过代谢过程或化学过程来进行化学转化, 从而得到式 I所示化合物和 /或其盐和 /或溶剂化 物。 可在体内转化以提供生物活性物质 (即式 I所示化合物) 的任何化合物是在本发明的 范围和主旨内的前药。 例如, 含有羧基的化合物可形成生理上可水解的酯, 其通过在体内 水解以得到式 I所示化合物本身而充当前药。 所述前药优选口服给药, 这是因为水解在许 多情况下主要在消化酶的影响下发生。 当酯本身具有活性或水解发生在血液中时, 可使用 肠胃外给药。
还应该理解的是, 本发明式 I所示化合物的水合物、溶剂化物(例如甲醇化物、 DMSO 化物) 也在本发明的范围内。 溶剂化的方法是本领域公知的。
在本发明的第二方面, 本发明提出了一种制备式 I所示化合物或其立体异构体、 互变 异构体、 药物活性代谢物、 可药用盐、 水合物、 溶剂化物或前药的方法和中间体。
根据本发明的实施例, 制备式 I所示化合物或其立体异构体、 互变异构体、 药物活性 代谢物、 可药用盐、 水合物、 溶剂化物或前药的方法包括: (1 )使所述式 1所示化合物与 式 2所示化合物进行接触, 以便获得式 3所示化合物; (2) 使所述式 3所示化合物与式 4 所示化合物 (丙烯酰氯)进行接触, 以便获得式 5所示化合物; (3 ) 使所述式 5所示化合 物与式 6a所示化合物进行接触, 以便获得式 I所示化合物。
Figure imgf000007_0001
其中, X、 以及 R2为如前面所定义的。
发明人发现, 利用本发明的该方法能够快速有效地制备式 I所示化合物或其立体异构 体、 互变异构体、 药物活性代谢物、 可药用盐、 水合物、 溶剂化物或前药, 且合成路线短、 环境友好、 目标产物的收率和纯度较高, 原料易得、 操作及后处理简单、 适合工业化生产。
下面对本发明的实施例中所采用的制备式 I所示化合物的一般方法进行描述:
1 ) 式 3所示化合物 (中间体) 的制备
使式 1所示化合物与式 2所示化合物进行接触, 以便获得式 3所示化合物; 具体地, 在三口瓶中加入式 1所示化合物、 式 2所示化合物、 以及四氢呋喃 (THF), 再加入碳酸铯, 回流 12 24小时, 反应结束后, 浓縮得油状液, 先用甲醇溶解, 再用丙酮 重结晶, 析出固体, 过滤, 干燥得式 3所示化合物, 无需进一步纯化。
2) 式 5所示化合物 (中间体 ) 的制备
使所述式 3所示化合物与式 4所示化合物进行接触, 以便获得式 5所示化合物; 具体地, 将式 3所示化合物与 N-甲基吡咯垸酮 (NMP)在 -5 °C~5 °C下混合, 缓慢加入 式 4所示化合物, 并在 0°C下保温搅拌反应 60分钟, 然后加入水继续搅拌半小时, 接着加 入饱和 NaHC03溶液碱化水溶液, 随后用乙酸乙酯萃取 3次。 先后用水、 盐水洗涤合并的 乙酸乙酯萃取物, Na2S04干燥并在减压下浓缩, 得到式 5所示化合物。
3 ) 式 I所示化合物的制备
使所述式 5所示化合物与式 6a所示化合物进行接触, 以便获得式 I所示化合物; 具体地, 将式 5所示化合物、 式 6a所示化合物 (式 5所示化合物与式 6a所示化合物 的摩尔比为 1 : 1.5 ) 加入 N-甲基吡咯垸酮的溶液中, 进行微波照射 (100°C~20(TC, 5-30 分钟) 。 冷却反应混合物, 用水稀释, 并用乙酸乙酯萃取 3次。 先后用水、 盐水洗涤合并 的乙酸乙酯萃取物, Na2S04干燥并在减压下浓缩, 再用异丙醇溶解, 在 -5°C~5°C下重结晶, 过滤, 得白色固体, 减压干燥, 即得式 I所示化合物。
在本发明的实施例中, 所述式 I所示化合物可为: 式 I所示化合物或其立体异构体、互 变异构体、 药物活性代谢物、 可药用盐、 水合物、 溶剂化物或前药。
本发明的发明人发现, 本发明的化合物还显示出提高的溶解性以及更不易从细胞中流 出的性质, 从而提高了生物利用度。
在本发明的第三方面, 本发明提供一种药物组合物, 所述组合物包含式 I所示化合物 或其立体异构体、 互变异构体、 药物活性代谢物、 可药用盐、 水合物、 溶剂化物或前药中 的至少一种。 根据本发明的实施例, 所述药物组合物可以进一步包括药学上可接受的赋形 剂。 该药用组合物还可以进一步包含气味剂、 香味剂等常规添加剂。
本发明所提供的药物组合物优选含有重量比为 0.1%~99%的式 I所示化合物作为活性成 份, 更优选的是, 式 I所示化合物作为活性成分占药物组合物总重量的 10%〜40%, 其余 部分为药学上可接受的载体和 /或常规添加剂。
本发明所提供的化合物和药物组合物可以是多种形式, 如片剂、 胶囊、 注射剂、 注射 用粉针、 粉剂、 糖浆、 溶液状、 悬浮液和气雾剂等, 并可以存在于适宜的固体或液体的载 体或稀释液中和适宜的用于注射或滴注的消毒器具中。
本发明的药物组合物的各种剂型可按照药学领域的常规制备方法制备。 本发明的化合 物和药物组合物可对哺乳动物临床使用, 包括人和动物, 可以通过口、 鼻、 皮肤、 肺或者 胃肠道等的途径给药。 不管采用何种服用方法, 个人的最佳剂量应依据具体的治疗方案而 定。 通常情况下是从小剂量开始, 逐渐增加剂量一直到找到最适合的剂量。 最优选的给药 途径为口服。
在本发明的第四方面, 本发明提出了式 I所示化合物或其立体异构体、 互变异构体、 药物活性代谢物、 可药用盐、 水合物、 溶剂化物或前药在制备药物中的用途。 根据本发明 的实施例, 所述药物用作激酶抑制剂。 根据本发明的实施例, 本发明的化合物可调节激酶 活性, 包括对 BTK进行调节, 可用作激酶抑制剂, 可由本发明化合物调节的其它类型的激 酶活性包括但不限于 Tec家族的成员, 如 BMX、 BTK、 ITK、 Txk和 Tec及它们的突变体。 因此, 上述药物能够有效作为 BTK抑制剂, 进而可治疗由 B细胞介导的疾病, 用于治疗肿 瘤疾病、 增殖性疾病、 ***反应性疾病、 自身免疫性疾病和炎症性疾病。
进一步的, 本发明所述式 I所示化合物比现有 BTK抑制剂依鲁替尼具有更高的 BTK 激酶抑制活性。 而且, 本发明的式 I所示化合物预处理重组 BTK后, 用不含抑制剂的培养 基重复洗涤, 其活性不会恢复, 表明 BTK被本发明的化合物不可逆抑制。
具体的, 本发明所述药物适用于治疗一种或一种以上与 BTK活性有关的疾病, 所述疾 病包括但不限于实体瘤、 淋巴瘤、 软组织肉瘤、 淋巴细胞性淋巴瘤、 套细胞淋巴瘤、 黑色 素瘤、 多发性骨髓瘤、 急性淋巴细胞白血病、 慢性淋巴细胞白血病、 急性骨髓性白血病、 慢性骨髓性白血病、 类风湿性关节炎、 银屑病关节炎、 骨关节炎、 ***性红斑狼疮、 牛皮 癣、 类风湿性脊椎炎、 痛风性关节炎等。
进一步的, 在牛胶原蛋白诱导的鼠关节炎模型(CIA)实验中, 本发明实施例的化合物 表现出了比依鲁替尼更好的疗效。
本发明所述的用作激酶抑制剂的式 I所示化合物, 其作为 BTK抑制剂, 成功克服了现 有 BTK抑制剂 ibrutinib药物本身溶解度低、 口服生物利用度低的缺陷, 具有良好的临床应 用和医药用途。
在本发明的第五方面, 本发明提供了一种治疗由 B细胞介导的疾病的方法。 根据本发 明的实施例, 该方法包括: 对患者给药前面所述的化合物或药物组合物。 发明人发现, 前 面所述的化合物或药物组合物具有较高的 BTK激酶抑制活性,能够有效用于治疗由 B细胞 介导的疾病。
根据本发明的实施例, 由 B细胞介导的疾病的种类不受特别限制。 根据本发明的实施 例, 由 B细胞介导的疾病可以为选自肿瘤疾病、 增殖性疾病、 ***反应性疾病、 自身免疫 性疾病和炎症性疾病的至少一种。 由此, 能够获得较好的治疗效果。
根据本发明的实施例, 由 B细胞介导的疾病可以为选自实体瘤、淋巴瘤、软组织肉瘤、 淋巴细胞性淋巴瘤、 套细胞淋巴瘤、 黑色素瘤、 多发性骨髓瘤、 急性淋巴细胞白血病、 慢 性淋巴细胞白血病、 急性骨髓性白血病、 慢性骨髓性白血病、 类风湿性关节炎、 银屑病关 节炎、 骨关节炎、 ***性红斑狼疮、 牛皮癣、 类风湿性脊椎炎和痛风性关节炎中的至少一 种。 由此, 治疗效果较佳。
本发明的附加方面和优点将在下面的描述中部分给出, 部分将从下面的描述中变得明 显, 或通过本发明的实践了解到。 具体实施方式
下面详细描述本发明的实施例。 下面描述的实施例是示例性的, 仅用于解释本发明, 而不能理解为对本发明的限制。 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。
本发明的实施例提供了式 I所示化合物或其立体异构体、 互变异构体、 药物活性代谢 物、 可药用盐、 水合物、 溶剂化物或前药, 制备式 I所示化合物或其立体异构体、 互变异 构体、 药物活性代谢物、 可药用盐、 水合物、 溶剂化物或前药的方法和中间体、 药物组合 物、 以及本发明的化合物
Figure imgf000010_0001
实施例 1 : 制备式 3所示化合物 (中间体)
在三口瓶中加入式 1所示化合物(17.0克, 0.1摩尔)、 式 2所示化合物(11.1克, 0.11 摩尔)、 以及四氢呋喃 (800毫升), 再加入碳酸铯 (39.1克, 0.12摩尔), 回流 15小时, 反应结束后, 浓縮得油状液, 先用甲醇 (60亳升) 溶解, 再用丙酮 (180亳升) 搅拌重结 晶, 析出固体, 过滤, 干燥得式 3所示化合物 (18.7克, 收率 74.0%), 无需进一步纯化。
实施例 2: 制备式 3所示化合物 (中间体)
在三口瓶中加入式 1所示化合物(17.0克, 0.1摩尔)、式 2所示化合物(15.2克, 0.15 摩尔)、 以及四氢呋喃 (800毫升), 再加入碳酸铯 (48.9克, 0.15摩尔), 回流 12小时, 反应结束后, 浓縮得油状液, 先用甲醇 (60亳升) 溶解, 再用丙酮 (180亳升) 搅拌重结 晶, 析出固体, 过滤, 干燥得式 3所示化合物 (18.2克, 收率 72.1%), 无需进一步纯化。 实施例 3 : 制备式 3所示化合物 (中间体)
在三口瓶中加入式 1所示化合物(17.0克, 0.1摩尔)、式 2所示化合物(10.6克, 0.105 摩尔)、 以及四氢呋喃 (800毫升), 再加入碳酸铯 (39.1克, 0.12摩尔), 回流 24小时, 反应结束后, 浓缩得油状液, 先用甲醇 (60毫升) 溶解, 再用丙酮 (180毫升) 搅拌重结 晶, 析出固体, 过滤, 干燥得式 3所示化合物 (18.4克, 收率 72.8%), 无需进一步纯化。
实施例 4: 制备式 5所示化合物 (中间体)
将式 3所示化合物(25.3克, 0.1摩尔)与 N-甲基吡咯垸酮(300毫升)在 -5°C〜5°C下 混合, 缓慢加入式 4所示化合物 ( 19.9克, 0.22摩尔), 并在 0°C下保温搅拌反应 60分钟, 然后加入水 (50毫升)继续搅拌半小时, 接着加入饱和 NaHC03溶液碱化水溶液, 随后用 乙酸乙酯萃取 3次, 每次 300亳升。 先后用水、 盐水洗涤合并的乙酸乙酯萃取物, Na2S04 干燥并在减压下浓缩, 得到式 5所示化合物 (23.9克, 收率 66.3%)。
实施例 5: -1所示化合物
Figure imgf000011_0001
根据下面所述的方案、 步骤制备式 1-1所示化合物。 具体如下:
将式 5所示化合物 (36.1克, 0.1摩尔)、 式 6a-l所示化合物 (15.2克, 0.15摩尔)加 入 N-甲基吡咯垸酮 (600毫升) 的溶液中, 进行微波照射 (100°C, 30分钟) 。 冷却反应 混合物, 用水 500毫升稀释, 并用乙酸乙酯萃取 3次, 每次 400亳升。 先后用水、 盐水洗 涤合并的乙酸乙酯萃取物, Na2S04干燥并在减压下浓缩, 再用异丙醇 300 毫升溶解, 在 -5°C~5°C下重结晶, 过滤, 得白色固体, 减压干燥, 即得式 1-1所示化合物 (得量 34.5克, 收率 78.0%)。
实施例 6: 制备式 1-2所示化合物
Figure imgf000012_0001
将式 5所示化合物 (36.1克, 0.1摩尔)、 式 6a-2所示化合物 (25.1克, 0.15摩尔)加 入 N-甲基吡咯烷酮 (600亳升) 的溶液中, 进行微波照射 (160°C, 10分钟)。 后处理同实 施例 5, 得式 1-2所示化合物 (得量 40.2克, 收率 81.6%)。
Figure imgf000012_0002
将式 5所示化合物 (36.1克, 0.1摩尔)、 式 6a-2所示化合物 (30.8克, 0.15摩尔)加 入 N-甲基吡咯烷酮 (500毫升) 的溶液中, 进行微波照射 (200°C, 5分钟) 。 后处理同实 施例 5, 得式 1-3所示化合物 (得量 38.1克, 收率 72.0%)。
根据本发明的实施例, 同理, 式 1-4所示化合物至式 1-26所示化合物的制备方法同实 施例 5至实施例 Ί中制备式 1-1所示化合物至式 1-3所示化合物的方法。其中式 5所示化合 物与式 6a所示化合物的投料摩尔比为 1 : 1.5。 例如, 以式 6a-15所示化合物为起始原料, 用式 5所示化合物与式 6a-15所示化合物进行接触, 最后得到的是对应的式 1-15所示化合 物。 具体化合物及数据见下表 1。
Figure imgf000012_0003
Figure imgf000013_0001
%e'8z,
%5Ό8
%8'
%69L
%e 9
%Z 18
¾V£8
Zl
.88080/M0ZN3/X3d I99 /SI0Z OAV
Figure imgf000015_0001
.88080/M0ZN3/X3d I99 /SI0Z OAV
Figure imgf000016_0001
实施例 8: 人类 B细胞刺激的方案
人类 B细胞从 150 ml血液纯化。 具体地, 血液用 PBS稀释 1/2, 经 Ficoll密度梯度离 心。 使用来自 Milenyi (Auburn, CA) 的 B细胞分离试剂盒 II通过阴性选择而从单核细胞 分离 B细胞。 然后, 在 96孔板中每孔 50000B细胞用 10 g/ml山羊 F(ab')2抗人 IgM抗体 (Jackson ImmunoResearch Laboratories, West Grove, PA)刺激。 实施例 1-7中制备获得的 式 1-1〜式 1-26所示化合物用 DMSO (二甲基亚砜)稀释并添加至细胞。 DMSO的终浓度为 0.5 %。 3天后使用 PromegaCellTiter-Glo (Madison, WI) 测量增殖。 结果发现本发明所述 式 I所示化合物被测试均是有活性的。
实施例 9: 人重组体 BTK酶测定
向 V形底 384孔板中加入测试化合物、人重组体 BTK ( InM, Invitrogen Corporation) 、 荧光素化肽 (fluoresceinated peptide) ( 1.5μΜ)、 ATP (20μΜ)和测定缓冲液(20mM HEPES (pH 7.4) 、 10mM MgCl2、 0.015 % Brij35和 4mM DTT于 1.6%DMSO中的溶液) , 其中 最终体积为 30 L。在室温孵育 60分钟后, 通过向每个样品中加入 45μ 135ηιΜ ΕϋΤΑ来终 止反应。 在 Caliper LabChip3000 (Caliper, Hopkinton, MA) 上通过对荧光底物和憐酸化 产物进行电泳分离来对反应混合物进行分析。 通过与作为 100 %抑制的无酶对照反应和作 为 0 %抑制的无抑制剂对照进行比较来计算抑制数据。 生成剂量响应曲线以确定抑制 50 % 激酶活性所需的浓度 (IC50)。 将化合物以 10 mM溶于二甲基亚砜 (DMSO)中, 然后以 11 个浓度进行评价。使用该测定, 确定本发明所述化合物以及现有 BTK抑制剂化合物依鲁替 尼的 IC50值。 结果见表 2。
表 2显示了选定的本发明化合物在体外 BTK激酶抑制测定中的活性。化合物编号对应 于表 1中的化合物编号。 活性指定为" A"的化合物提供的 IC50≤10 nM; 活性指定为 "B "的 化合物提供的 IC50为 10-100 nM; 活性指定为" C"的化合物提供的 IC50为 100-1000 nM; 活性指定为" D"的化合物提供的 IC50为 1000-10000 nM; 而活性指定为 'Έ"的化合物提供的 IC50> 10000 nM。 本发明所述式 I所示化合物比依鲁替尼具有更高的 BTK激酶抑制活性。 而且, 本发明的化合物预处理重组 BTK后, 用不含抑制剂的培养基重复洗涤, 其活性不会 恢复, 表明 BTK被这些化合物不可逆抑制。
表 2: BTK抑制数据
化合物编号 抑制 BTK的 50(μΜ;> 活性指定
1-1 0.018 A
1-2 0.025 A
1-3 0.016 A
1-4 0.54 A
1-5 1.2 A
1-6 0.41 A
1-7 0.94 A
1-8 1.2 A
1-9 1.7 A
1-10 0.45 A
1-11 0.58 A
1-12 0.028 A
1-13 0.049 A
1-14 0.80 A
1-15 1.7 A
1-16 2.3 A
1-17 0+44 A 1-18 0.050 A
1-19 0.045 A
1-20 0.39 A
1-21 0.038 A
1-22 1.3 A
1-23 0.50 A
1-24 0.80 A
1-25 0.016 A
1-26 0.48 A
依鲁替尼 2.5 A
实施例 10: 牛胶原蛋白诱导的鼠关节炎模型 (CIA)
1、 乳剂配制: 称取牛胶原溶于乙酸中, 配成浓度为 8 mg/ml的牛胶原溶液, 4摄氏度过 夜。 向牛胶原溶液中添加等体积的完全弗氏佐剂(CFA, 1 mg/ml ), 配成终浓度为 4 mg/ml 的牛胶原 CFA混合液, 再在湿冰上用高速匀浆器匀浆, 直到形成白色乳剂。
在第 0天, 将大鼠用 II型牛胶原在完全弗氏佐剂 (CFA) 中的乳剂注射, 在背部上的几 个位置皮内注射 (i.d. ) 50微升。 在约第 7天在尾巴基部或背部的备选位点提供胶原乳剂的 加强注射 (i.d. ) 50微升。 在初始胶原注射之后的 12天至 14天通常观察到关节炎。 从第 14天 起, 如下面所述 (关节炎的评价)可以评价动物的关节炎的进展。
关节炎的评价:
在两种模型中, 使用评分***对爪和肢关节的炎症发展进行定量, 所述的评分***包 括按照下面所述的标准对 4个爪的评估, 评分标准见表 3。
表 3 : 评分标准
Figure imgf000018_0001
第 21天, 对所有动物进行分组, 使每组动物体重、 发病率 (即动物发病个数占总数的 百分比)基本一致。 在第 21天开始口服灌胃给药, 每天一次(QD ) , 每组动物给药剂量见 表 4。 给药 14天后, 计算各组药物对关节炎大鼠的抑制率。 表 4:
Figure imgf000019_0002
注: * p<0.01
从表 4可看出,第 6组(依鲁替尼, 10mg/kg)与本发明的第 2组(式 I -2化合物, 5 mg/kg), 第 5组(式 1 -18化合物, 10 mg/kg)抑制率相当。 但是本发明的第 4组(式 1 -11化合物, 10 mg/kg) 的抑制率明显高于第 6组 (依鲁替尼, 10mg/kg) 。
实施例 11: 水溶解性测定
以化合物 1-2为例, 将本发明所述化合物 1-2的水溶解度与依鲁替尼作比较。 测定方法 如下:
在 pH为 7.4的缓冲水溶液中测定平衡溶解度。 pH为 7.4的缓冲溶液通过用 10摩尔 / 升的氢氧化钠将 0.07M的磷酸二氢钠溶液的 pH调节至 7.4, 该溶液的离子强度为 0.15。将 至少 1毫克的粉末与 1毫升的缓冲液混合, 制备浓度大于 1毫克 /毫升的混合物。 将这些样 品振荡超过 2小时, 并且在室温下放置过夜。 然后通过先用样品饱和的 0.45 μηι的尼龙注 射器过滤器过滤样品, 从滤液中连续取样 2次。 以在 50%甲醇中制备的标准溶液作参比, 通过 HPLC测定滤液。 化合物 1-2的溶解度为 44毫克 /毫升, 依鲁替尼的溶解度为 12毫克 / 实施例 12: 用于口服给药的胶囊
Figure imgf000019_0003
将上表中的成分混合,并且活性成分(本发明的化合物)分配在胶囊中,共制备成 1000 克粉末, 灌装成胶囊, 每粒胶囊的活性成分为 200mg。
实施例 13: 用于口服给药的胶囊
Figure imgf000019_0001
活性成分 40.0%
预胶化玉米淀粉 59.5%
硬脂酸镁 0.5% 将上表中的成分混合,并且活性成分(本发明的化合物)分配在胶囊中,共制备成 1000 克粉末, 灌装成胶囊, 每粒胶囊的活性成分为 400mg。
实施例 14: 用于口服给药的片剂
Figure imgf000020_0001
将上表中的成分混合, 并且使用溶剂如乙醇制粒。然后将制剂干燥, 共制备成 1000克 粉末, 并且用合适的压片机形成为片剂, 每片的活性成分 (本发明的化合物) 为 200 mg。
实施例 15 : 用于注射给药的注射液
Figure imgf000020_0002
将活性成分 (本发明的化合物) 溶解在部分注射用水中。 然后在搅拌下加入足够量的 氯化钠, 以使溶液等渗。 用剩余的注射用水将该溶液补足重量, 用 0.2微米膜过滤器过滤, 并且在无菌条件下包装, 得注射液。 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一定 指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何的 一个或多个实施例或示例中以合适的方式结合。
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不脱离本发明的原理和宗旨的情况 下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。

Claims

权利要求书
1、 一种化合物, 其特征在于, 所述化合物为式 I所示化合物或式 I所示化合物的立体 异构体、 互变异构体、 药 物或前药,
Figure imgf000021_0001
其中,
X为选自下列之一: 取代或未取代的苯基、 取代或未取代的 3元到 7元饱和或不饱和 碳环、取代或未取代的 8元到 10元饱和或不饱和双环或芳基环、取代或未取代的具有 1到 4个杂原子的 5元到 6元单环杂芳基环、 取代或未取代的具有 1到 3个杂原子的 4元到 Ί 元饱和或不饱和杂环、取代或未取代的具有 1到 5个杂原子的 Ί元到 10元饱和或不饱和双 环杂环、 取代或未取代的具有 1到 5个杂原子的 8元到 10元双环杂芳基环;
Ri为选自 -R、卤素、 -OR、 -0(CH2)aOR、 -CN、 -N02、 -S02R、 -S02N(R)2、 -SOR、 -C(0)R、 -C02R、 -C(0)N(R)2、 -NRC(0)R、 -NRC(0) R2 -NRS02R和 -N(R)2中的任意一种;
R2为选自 -R、卤素、 -OR、 -0(CH2)aOR、 -CN、 -N02、 -S02R、 -S02N(R)2、 -SOR、 -C(0)R、 -C02R、 -C(0)N(R)2、 - RC(0)R、 -NRC(0) R2、 -NRS02R和 -N(R)2中的任意一种;
其中, a为 1至 5的整数,
R基团各自独立地为选自下列之一: 氢、 取代或未取代的 脂肪族基、 取代或未取 代的苯基、 取代或未取代的具有 1到 2个杂原子的 4元到 7元杂环、 取代或未取代的具有 1到 4个杂原子的 5元到 6元单环杂芳基环,
所述杂原子各自独立地为选自氮、 氧和硫的任意
2、 根据权利要求 1所述的化合物, 其特征在于,
Figure imgf000021_0002
自下列之
Figure imgf000022_0001
Figure imgf000022_0002
、 一种制备权利要求 1或 2所述的化合物的方法, 其特征在于, 包括:
( 1 ) 使式 1所示化合物与式 2所示化合物进行接触, 以便获得式 3所示化合物; (2) 使所述式 3所示化合物与式 4所示化合物进行接触, 以便获得式 5所示化合物;
( 化合物,
Figure imgf000023_0001
4、 一种药物组合物, 其特征在于, 包括:
权利要求 1或 2所述的化合物。
5、 根据权利要求 4所述的药物组合物, 其特征在于, 进一歩包括:
药学上可以接受的赋形剂。
6、权利要求 1或 2所述的化合物或权利要求 4或 5所述的药物组合物在制备药物中的 用途, 所述药物用作激酶抑制剂。
7、 根据权利要求 6所述的用途, 其特征在于, 所述药物用作 BTK抑制剂。
8、根据权利要求 6所述的用途,其特征在于,所述药物用于治疗由 B细胞介导的疾病。
9、 根据权利要求 6所述的用途, 其特征在于, 所述药物用于***疾病、 增殖性疾 病、 ***反应性疾病、 自身免疫性疾病和炎症性疾病的至少一种。
10、 根据权利要求 6所述的用途, 其特征在于, 所述药物用于治疗实体瘤、 淋巴瘤、 软组织肉瘤、 淋巴细胞性淋巴瘤、 套细胞淋巴瘤、 黑色素瘤、 多发性骨髓瘤、 急性淋巴细 胞白血病、 慢性淋巴细胞白血病、 急性骨髓性白血病、 慢性骨髓性白血病、 类风湿性关节 炎、 银屑病关节炎、 骨关节炎、 ***性红斑狼疮、 牛皮癣、 类风湿性脊椎炎和痛风性关节 炎中的至少一种。
11、 一种治疗由 B细胞介导的疾病的方法, 其特征在于, 对患者给药权利要求 1或 2 所述的化合物或者权利要求 4或 5所述的药物组合物。
12、根据权利要求 11所述的方法, 其特征在于, 所述由 B细胞介导的疾病为选自肿瘤 疾病、 增殖性疾病、 ***反应性疾病、 自身免疫性疾病和炎症性疾病的至少一种。
13、根据权利要求 11所述的方法, 其特征在于, 所述由 B细胞介导的疾病为选自实体 瘤、 淋巴瘤、 软组织肉瘤、 淋巴细胞性淋巴瘤、 套细胞淋巴瘤、 黑色素瘤、 多发性骨髓瘤、 急性淋巴细胞白血病、 慢性淋巴细胞白血病、 急性骨髓性白血病、 慢性骨髓性白血病、 类 风湿性关节炎、 银屑病关节炎、 骨关节炎、 ***性红斑狼疮、 牛皮癣、 类风湿性脊椎炎和 痛风性关节炎中的至少一种。
PCT/CN2014/080887 2014-06-06 2014-06-26 化合物及其制备方法和用途 WO2015184661A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201410251204.2 2014-06-06
CN201410251204.2A CN104086551B (zh) 2014-06-06 2014-06-06 化合物及其制备方法和用途

Publications (1)

Publication Number Publication Date
WO2015184661A1 true WO2015184661A1 (zh) 2015-12-10

Family

ID=51634339

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2014/080887 WO2015184661A1 (zh) 2014-06-06 2014-06-26 化合物及其制备方法和用途

Country Status (2)

Country Link
CN (1) CN104086551B (zh)
WO (1) WO2015184661A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11865120B2 (en) 2013-08-23 2024-01-09 Neupharma, Inc. Substituted quinazolines for inhibiting kinase activity

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113582995B (zh) * 2021-08-17 2022-08-16 西安交通大学 9-位含有丙烯酰氨基片段的-9h-嘌呤类化合物及其盐和应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1231611A (zh) * 1996-08-02 1999-10-13 Cv治疗公司 依赖细胞周期蛋白的激酶2和IKB-α的嘌呤抑制剂
WO2001049688A1 (en) * 2000-01-07 2001-07-12 Universitaire Instelling Antwerpen Purine derivatives, process for their preparation and use thereof
CN101715453A (zh) * 2007-03-23 2010-05-26 安姆根有限公司 杂环化合物及其应用

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101315610B1 (ko) * 2006-09-22 2013-10-10 파마시클릭스, 인코포레이티드 브루톤 티로신 키나제 억제제
JP2010522241A (ja) * 2007-03-21 2010-07-01 ブリストル−マイヤーズ スクイブ カンパニー 増殖性疾患、アレルギー性疾患、自己免疫疾患または炎症性疾患として有用な縮合ヘテロ環化合物
CA2874756C (en) * 2007-03-28 2018-05-29 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
CA2723185A1 (en) * 2008-04-22 2009-10-29 Portola Pharmaceuticals, Inc. Inhibitors of protein kinases
KR20110025224A (ko) * 2008-06-27 2011-03-09 아빌라 테라퓨틱스, 인크. 헤테로아릴 화합물 및 이의 용도
JP2011529073A (ja) * 2008-07-24 2011-12-01 ブリストル−マイヤーズ スクイブ カンパニー キナーゼ調節因子として有用な縮合ヘテロ環化合物
US7718662B1 (en) * 2009-10-12 2010-05-18 Pharmacyclics, Inc. Pyrazolo-pyrimidine inhibitors of bruton's tyrosine kinase
EP2694486B1 (en) * 2011-04-01 2018-01-10 University of Utah Research Foundation Substituted n-(3-(pyrimidin-4-yl)phenyl)acrylamide analogs as tyrosine receptor kinase btk inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1231611A (zh) * 1996-08-02 1999-10-13 Cv治疗公司 依赖细胞周期蛋白的激酶2和IKB-α的嘌呤抑制剂
WO2001049688A1 (en) * 2000-01-07 2001-07-12 Universitaire Instelling Antwerpen Purine derivatives, process for their preparation and use thereof
CN101715453A (zh) * 2007-03-23 2010-05-26 安姆根有限公司 杂环化合物及其应用

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11865120B2 (en) 2013-08-23 2024-01-09 Neupharma, Inc. Substituted quinazolines for inhibiting kinase activity

Also Published As

Publication number Publication date
CN104086551A (zh) 2014-10-08
CN104086551B (zh) 2016-09-21

Similar Documents

Publication Publication Date Title
JP7383652B2 (ja) B-rafキナーゼのマレイン酸塩、結晶形、調整方法、及びその使用
CA3004372C (en) Pyrimidine derivative and use thereof
JP2023071709A (ja) Jakファミリーのキナーゼの低分子阻害物質
JP2019108346A (ja) リジン特異的なデメチラーゼ−1の阻害剤
CN113490495A (zh) Helios的小分子降解剂及其使用方法
JP2020519596A (ja) Irak−4阻害剤として有用なチエノピリジンおよびベンゾチオフェン
KR102575246B1 (ko) 페닐-2-히드록시-아세틸아미노-2-메틸-페닐 화합물
TW201716406A (zh) 丙烯酸類衍生物、其製備方法及其在醫藥上的用途
CN109890388B (zh) 毒蕈碱型乙酰胆碱受体m4的正向别构调节剂
KR20230049584A (ko) 아미노산 화합물을 사용한 호흡기 질환의 치료
CN109862893B (zh) 毒蕈碱型乙酰胆碱受体m4的正向别构调节剂
JP2018514524A (ja) 5−芳香族アルキニル基置換ベンズアミド系化合物並びにその製造方法、薬物組成物及び使用
JP7481435B2 (ja) Crac阻害剤としての2h-ベンゾピラン誘導体
JP2021176819A (ja) キナゾリン化合物を有効成分とする医薬組成物
JP6522665B2 (ja) 新規化合物
EP4073065A1 (en) New methylquinazolinone derivatives
JP2014533262A (ja) 慢性骨髄性白血病を治療するためのn−メチル−2−[3−((e)−2−ピリジン−2−イル−ビニル)−1h−インダゾール−6−イルスルファニル]−ベンズアミド
CN112292374A (zh) 一种新型磷酸肌醇3-激酶抑制剂及其制备方法和用途
WO2020156479A1 (zh) 环丙烯并苯并呋喃取代的氮杂芳基化合物、其中间体、制备方法及应用
JP6847851B2 (ja) Nmda受容体のモジュレーターとしてのピリドピリミジノン及びその使用
CN108676009B (zh) 作为her2酪氨酸激酶抑制剂的嘧啶衍生物及其应用
KR20230035311A (ko) Nek7 키나제의 억제제
EA027882B1 (ru) Этинильные производные в качестве антагонистов метаботропного глутаматного рецептора
WO2015184661A1 (zh) 化合物及其制备方法和用途
WO2014086102A1 (zh) 作为酪氨酸激酶抑制剂的吲哚满酮衍生物

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: 14894128

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: 14894128

Country of ref document: EP

Kind code of ref document: A1