WO2021075691A1 - Pyrimidine derivative, method of preparing same, and pharmaceutical composition for preventing or treating cancer, comprising same as effective component - Google Patents

Pyrimidine derivative, method of preparing same, and pharmaceutical composition for preventing or treating cancer, comprising same as effective component Download PDF

Info

Publication number
WO2021075691A1
WO2021075691A1 PCT/KR2020/010657 KR2020010657W WO2021075691A1 WO 2021075691 A1 WO2021075691 A1 WO 2021075691A1 KR 2020010657 W KR2020010657 W KR 2020010657W WO 2021075691 A1 WO2021075691 A1 WO 2021075691A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
amino
egfr
phenyl
formula
Prior art date
Application number
PCT/KR2020/010657
Other languages
French (fr)
Korean (ko)
Inventor
이광호
조병철
두기랄라크리시나바부
최길돈
윤지연
박채원
채종학
정명은
Original Assignee
한국화학연구원
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 한국화학연구원 filed Critical 한국화학연구원
Publication of WO2021075691A1 publication Critical patent/WO2021075691A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/308Foods, ingredients or supplements having a functional effect on health having an effect on cancer prevention

Definitions

  • the present invention relates to a pharmaceutical composition for preventing or treating cancer, and specifically to a pharmaceutical composition for preventing or treating cancer containing a pyrimidine derivative as an active ingredient.
  • Cancer incidence is related to changes in various environmental factors including chemicals, radiation, and viruses, oncogenes, tumor suppressor genes, and genes related to apoptosis and DNA repair.
  • a new treatment, targeted chemotherapy became possible.
  • Targeted therapies are generally made to target molecules characteristic of cancer cells to exert their effects, and molecular targets are cancer cell signal transduction pathways, angiogenesis, and interstitial cells. They are genes related to (matrix), cell cycle regulator, and apoptosis.
  • Currently used as important target therapeutics in treatment include tyrosine kinase inhibitors, signaling pathway inhibitors, and angiogenesis inhibitors.
  • EGFR epidermal growth factor receptor
  • a receptor tyrosine kinase of the erbB family is a non-small cell lung cancer. It is abnormally activated in many epithelial cell tumors, including NSCLC, breast cancer, glioma, squamous cell carcinoma of the head and neck, colon cancer, rectal carcinoma, head and neck cancer, gastric cancer, and prostate cancer. It has been known that activation causes continuous cell proliferation, invasion of surrounding tissues, distant metastasis, and blood vessel formation, and increases cell survival.
  • the EGFR is one of the ErbB tyrosine kinase receptors family (EGFR, HER-2, ErbB-3, ErbB-4), an extracelluar ligand-binding domain and a tyrosine kinase region It is a transmembrane tyrosine kinase that has an intracellular domain including (tyrosine kinase domain).
  • PI3K phosphatidylinositol 3-kinase
  • EGFR is overexpressed in more than half of non-small cell lung cancer (NSCLC), and many studies have been conducted as a target of treatment.
  • EGFR TKI tyrosine kinase inhibitor
  • representative drugs include gefitinib (IRESSATM), erlotinib (TARCEVATM), and lapatinib (TYKERBTM, TYVERBTM).
  • the EGFR mutation is largely classified into a sensitizing mutation and a resistant mutation.
  • the deletion of exon 19 and the L858R point mutation of exon 21 are the most important sensitive mutations. %, and exon 19 del mutations are known to be more sensitive to TKI.
  • the T790M point mutation of exon 20 is the most important resistance mutation and is known to be found in more than 50% of acquired resistance patients (Clin Cancer Res 2006;12:6494-6501.).
  • Somatic mutations identified so far include in-frame deletions within exon 19 or insertions in exon 20, as well as point mutations in which a single nucleic acid residue is modified in the expressed protein (e.g., L858R, G719S, G719C, G719A, L861Q) ( Fukuoka et al. JCO 2003; Kris et al JAMA 2003 and Shepherd et al NEJM 2004).
  • EGFR mutations EGFR_del19 or EGFR_L858R
  • their therapeutic drugs Iressa and Taseba
  • Iressa and Taseba their therapeutic drugs
  • Iressa and Taseba their therapeutic drugs
  • this is the main cause of actual drug resistance.
  • the first generation EGFR inhibitor is used for an average of 10 months, the acquired resistance of the T790M mutation located at the gatekeeper of the EGFR kinase occurs, and the first generation EGFR inhibitors do not take effect.
  • EGFR_del19_T790M or EGFR_L858R_T790M double mutation occurs, preventing the existing treatment from showing efficacy.
  • EGFR Exon20insertion mutations There is a wide variety of EGFR Exon20insertion mutations, including EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV and EGFR D770_N771insSVD.
  • the pyrimidine derivatives according to the present invention exhibit relatively low inhibition against wild-type EGFR, while exhibiting high inhibitory ability against EGFR mutations, thereby preventing cancer.
  • the present invention was completed by finding that it can be usefully used for treatment.
  • Another object of the present invention is to provide a method for preparing the pyrimidine derivative.
  • Another object of the present invention is to provide a pharmaceutical composition for the prevention or treatment of cancer containing the pyrimidine derivative, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
  • Another object of the present invention is to provide a health functional food composition for preventing or improving cancer containing the pyrimidine derivative, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
  • Another object of the present invention is to provide a pharmaceutical composition for preventing or treating diseases related to EGFR mutations containing the pyrimidine derivative, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
  • R is -OR a or -NR b1 R b2 ,
  • R a is -(CH 2 ) m -NR b1 R b2 , where m is 1 to 3,
  • R b1 and R b2 are each independently hydrogen or unsubstituted or straight or branched chain C 1-6 alkyl substituted with NR c1 R c2 , or R b1 and R b2 are N, O together with the nitrogen to which they are attached. And to form an unsubstituted or straight or branched chain C 1-6 alkyl containing one or more heteroatoms selected from the group consisting of S or a 5 to 7 membered heterocycloalkyl substituted with NR c1 R c2,
  • R c1 and R c2 are each independently hydrogen or straight or branched chain C 1-6 alkyl, or, R c1 and R c2 are selected from the group consisting of N, O and S together with the nitrogen to which they are attached. 5 to 7 membered heterocycloalkyl substituted with unsubstituted or straight or branched C 1-6 alkyl containing one or more atoms.
  • R is as defined in Chemical Formula 1.
  • a pharmaceutical composition for preventing or treating cancer containing the compound represented by Chemical Formula 1, an isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.
  • a health functional food for preventing or improving cancer containing the compound represented by Formula 1, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
  • a pharmaceutical composition for preventing or treating diseases related to EGFR mutations containing the compound represented by Formula 1, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
  • cancer or EGFR comprising administering to a subject in need a pharmaceutical composition or a dietary supplement composition containing the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient It provides a method of preventing or treating mutation-related diseases.
  • a pharmaceutical composition or a dietary supplement composition containing a compound represented by Formula 1 or a pharmaceutically acceptable salt thereof in the prevention or treatment of cancer or EGFR mutation-related diseases Provides.
  • the pyrimidine derivative according to the present invention exhibits a relatively weak inhibitory effect on EGFR activity against wild-type EGFR, and exhibits a high inhibitory ability against EGFR mutations, so it can be usefully used in the treatment of cancer in which EGFR mutations have occurred.
  • 1 is a graph showing the results of measuring the cell growth inhibitory ability of the existing EGFR TKI (tyrosine kinase inhibitor) drug osimertinib against the Ba/F3 EGFR exon20 insertion mutation.
  • EGFR TKI tyrosine kinase inhibitor
  • Example 2 is a graph showing the results of measuring the cell growth inhibitory ability of the compound represented by Example 1 against the Ba/F3 EGFR exon20 insertion mutation.
  • the present invention provides a compound represented by the following formula (1), an isomer thereof, or a pharmaceutically acceptable salt thereof.
  • R is -OR a or -NR b1 R b2 ,
  • R a is -(CH 2 ) m -NR b1 R b2 , where m is 1 to 3,
  • R b1 and R b2 are each independently hydrogen or unsubstituted or straight or branched chain C 1-6 alkyl substituted with NR c1 R c2 , or R b1 and R b2 are N, O together with the nitrogen to which they are attached. And to form an unsubstituted or straight or branched chain C 1-6 alkyl containing one or more heteroatoms selected from the group consisting of S or a 5 to 7 membered heterocycloalkyl substituted with NR c1 R c2,
  • R c1 and R c2 are each independently hydrogen or straight or branched chain C 1-6 alkyl, or, R c1 and R c2 are selected from the group consisting of N, O and S together with the nitrogen to which they are attached. 5 to 7 membered heterocycloalkyl substituted with unsubstituted or straight or branched C 1-6 alkyl containing one or more atoms.
  • R is , , , , , , or Can be
  • Examples of the compound represented by Formula 1 according to the present invention include the following compounds:
  • the compound represented by Formula 1 of the present invention can be used in the form of a pharmaceutically acceptable salt, and an acid addition salt formed by a pharmaceutically acceptable free acid is useful.
  • Acid addition salts include inorganic acids such as hydrochloric acid, nitric acid, phosphoric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, nitrous acid, phosphorous acid, etc., aliphatic mono and dicarboxylates, phenyl-substituted alkanoates, hydroxy alkanoates and alkanes.
  • non-toxic organic acids such as dioate, aromatic acids, aliphatic and aromatic sulfonic acids, etc., acetate, benzoic acid, citric acid, lactic acid, maleic acid, gluconic acid, methanesulfonic acid, 4-toluenesulfonic acid, tartaric acid, fumaric acid, and the like.
  • pharmaceutically non-toxic salts include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogen phosphate, dihydrogen phosphate, metaphosphate, pyrophosphate chloride, bromide, i.
  • the acid addition salt according to the present invention can be prepared by a conventional method, for example, a precipitate formed by dissolving the derivative of Formula 1 in an organic solvent such as methanol, ethanol, acetone, methylene chloride, acetonitrile, etc. and adding an organic or inorganic acid It can be prepared by filtration and drying, or it can be prepared by distilling a solvent and an excess of acid under reduced pressure and then drying to crystallize under an organic solvent.
  • an organic solvent such as methanol, ethanol, acetone, methylene chloride, acetonitrile, etc.
  • a pharmaceutically acceptable metal salt can be made using a base.
  • the alkali metal or alkaline earth metal salt is obtained, for example, by dissolving the compound in an excess alkali metal hydroxide or alkaline earth metal hydroxide solution, filtering the undissolved compound salt, and evaporating and drying the filtrate. At this time, it is pharmaceutically suitable to prepare sodium, potassium or calcium salt as the metal salt.
  • the corresponding salt is obtained by reacting an alkali metal or alkaline earth metal salt with a suitable negative salt (eg, silver nitrate).
  • the present invention includes not only the compound represented by Formula 1 and its pharmaceutically acceptable salts, but also solvates, isomers, hydrates, etc. that may be prepared therefrom.
  • solvate refers to a compound of the present invention or a salt thereof containing a stoichiometric or non-stoichiometric amount of a solvent bound by non-covalent intermolecular forces.
  • Preferred solvents therefor include volatile, non-toxic, and/or suitable solvents for administration to humans. In this case, when the solvent is water, it is referred to as "hydrate”.
  • isomers refers to a compound of the present invention or a salt thereof having the same chemical formula or molecular formula, but structurally or sterically different.
  • isomers include structural isomers such as tautomers, R or S isomers having an asymmetric carbon center, stereoisomers such as geometric isomers (trans, cis), and optical isomers. All these isomers and mixtures thereof are also included within the scope of the present invention.
  • It provides a method for preparing a compound represented by Formula 1, comprising the step of preparing a compound represented by Formula 1 by reacting a compound represented by Formula 2 with a compound represented by Formula 3.
  • R is as defined in Chemical Formula 1.
  • step 1 is a step of preparing a compound represented by Formula 1 by reacting a compound represented by Formula 2 with a compound represented by Formula 3. Specifically, this is a step in which the compound represented by Formula 1 is formed by reacting the halogen of the compound represented by Formula 2 with the primary amine of the compound represented by Formula 3.
  • the reaction of Scheme 1 is not particularly limited as long as it is a condition capable of forming an amine bond by combining a halogen and an amine.
  • an acid condition was used, and trifluoroacetic acid (TFA) was used as the acid, but the present invention is not limited thereto.
  • the concentration of the acid may be 1 N, but is not limited thereto.
  • the solvent usable in Reaction Scheme 1 is not particularly limited, but lower alcohols including isopropanol, methanol, ethanol, propanol and butanol; Tetrahydrofuran (THF); Dioxane; Ether solvents including ethyl ether and 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethyl sulfoxide (DMSO), methylene chloride, dichloroethane, water, acetonase sulfonate, toluene sulfonate, chlorobenzene sulfonate, xylene sulfonate, ethyl acetate, phenyl acetate, phenyl propionate , Phenylbutyrate, Cycrate, lactate, hydroxybutyrate, glycolate, malate, tartrate, methanesulfonate, propanesulfonate, naphthalene-1-sulfonate, na
  • reaction temperature in Scheme 1 may be 80° C. to 95° C., but is not limited thereto, and the reaction temperature may be appropriately adjusted according to the degree to which the reaction proceeds.
  • reaction time in Scheme 1 may be 10 minutes to 24 hours, but is not limited thereto, and the reaction time may be appropriately adjusted according to the degree to which the reaction proceeds.
  • the present invention provides a pharmaceutical composition for preventing or treating cancer containing the compound represented by Formula 1, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
  • the cancer is pseudomyxoma, intrahepatic biliary tract cancer, hepatoblastoma, liver cancer, thyroid cancer, colon cancer, testicular cancer, myelodysplastic syndrome, glioblastoma, oral cancer, cleft lip cancer, mycological sarcoma, acute myeloid leukemia, acute lymphocytic leukemia, basal cell Cancer, ovarian epithelial cancer, ovarian germ cell carcinoma, male breast cancer, brain cancer, pituitary adenoma, multiple myeloma, gallbladder cancer, biliary tract cancer, colon cancer, chronic myelogenous leukemia, chronic lymphocytic leukemia, retinoblastoma, choroidal melanoma, barter bulge cancer, bladder cancer, Peritoneal cancer, parathyroid cancer, adrenal cancer, non-sinus cancer, non-small cell lung cancer, tongue cancer, astrocytoma, small cell lung cancer, pediatric brain cancer
  • the EGFR mutations are EGFR del19, EGFR L858R, EGFR del19/T790M, EGFR L858R/T790M, EGFR del19/T790M/C797S, EGFR L858R/T790M/C797S, EGFR D770delinsGY, EGFR H773_V774ins D770ASH, EGFR EGFR and EGFR D770ASH, EGFR and EGFR D770ASH, EGFR, and EGFR del19/T790M_EGFR L858R/T790M It may be one or more selected from the group consisting of.
  • the compound represented by Formula 1 of the present invention exhibits a relatively weak inhibitory effect on EGFR activity against wild-type EGFR, while selectively exhibiting a high inhibitory ability against EGFR mutations, and in particular, exhibits a high inhibitory ability against EGFR del19/T790M, a double mutation. (See Experimental Example 1 and Table 2).
  • the compound represented by Formula 1 of the present invention exhibits a selectively high cell growth inhibitory ability against EGFR mutations in Ba/F3 cell lines.
  • the existing EGFR against triple mutations EGFR del19/T790M/C797S or EGFR L858R/T790M/C797S Since it exhibits a higher cell growth inhibitory ability than Osimertinib, which is a tyrosine kinase inhibitor (TKI) drug, it can be seen that it exhibits superior anticancer effects than osimertinib (see Experimental Example 2, Table 3 and Table 4).
  • TKI tyrosine kinase inhibitor
  • Example 1 compound of the present invention selectively shows high cell growth inhibitory ability against EGFR mutations in Ba/F3 cell lines, specifically EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV or EGFR D770_N771insSVD Since it exhibits a higher cell growth inhibitory ability than mertinib, it can be seen that it exhibits superior anticancer effects than osimertinib (see Experimental Example 3 and Table 5).
  • the compound represented by Formula 1 according to the present invention exhibits high inhibitory ability against EGFR mutations, EGFR del19, EGFR L858R, EGFR del19/T790M, EGFR L858R/T790M, EGFR del19/T790M/C797S, EGFR L858R/ T790M/C797S, EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV, EGFR D770_N771insSVD can be usefully used in the treatment of cancers expressing EGFR mutations.
  • the inhibitory ability against T790M/C797S is remarkably excellent, it can be usefully used in the treatment of cancers expressing EGFR del19/T790M/C797S or EGFR L858R/T790M/C797S.
  • the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof may be administered in various oral and parenteral formulations upon clinical administration.
  • formulation it is prepared using diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrants, and surfactants that are usually used.
  • Solid preparations for oral administration include tablets, pills, powders, granules, capsules, and the like, and these solid preparations include at least one excipient in one or more compounds, such as starch, calcium carbonate, sucrose, or lactose ( lactose), gelatin, etc.
  • lubricants such as magnesium stearate and talc are also used.
  • Liquid preparations for oral administration include suspensions, liquid solutions, emulsions, syrups, etc., but may include various excipients, such as wetting agents, sweeteners, fragrances, and preservatives, in addition to water and liquid paraffin, which are commonly used simple diluents. have.
  • Formulations for parenteral administration include sterilized aqueous solutions, non-aqueous solutions, suspensions, and emulsions.
  • the non-aqueous solvent and the suspension solvent propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate may be used.
  • a pharmaceutical composition containing the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient can be administered parenterally, and parenteral administration is by subcutaneous injection, intravenous injection, intramuscular injection, or intrathoracic injection. It depends on how to do it.
  • the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof is mixed in water together with a stabilizer or buffer to prepare a solution or suspension, and the ampoule or vial unit dosage form It can be manufactured with.
  • the composition may be sterilized and/or contain adjuvants such as preservatives, stabilizers, hydrating agents or emulsification accelerators, salts and/or buffers for controlling osmotic pressure, and other therapeutically useful substances, which are conventional methods of mixing and granulation. It can be formulated according to the method of painting or coating.
  • Formulations for oral administration include, for example, tablets, pills, hard/soft capsules, solutions, suspensions, emulsifiers, syrups, granules, elixirs, and troches. , Dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine), lubricants (such as silica, talc, stearic acid and magnesium or calcium salts thereof and/or polyethylene glycol). Tablets may contain a binder such as magnesium aluminum silicate, starch paste, gelatin, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidine, and in some cases boron such as starch, agar, alginic acid or sodium salt thereof. It may contain release or boiling mixtures and/or absorbents, colorants, flavoring agents, and sweetening agents.
  • a binder such as magnesium aluminum silicate, starch paste, gelatin, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidine,
  • the present invention provides a health functional food for the prevention or improvement of cancer containing the compound represented by Chemical Formula 1, its isomer, or a pharmaceutically acceptable salt thereof as an active ingredient.
  • the cancer is pseudomyxoma, intrahepatic biliary tract cancer, hepatoblastoma, liver cancer, thyroid cancer, colon cancer, testicular cancer, myelodysplastic syndrome, glioblastoma, oral cancer, cleft lip cancer, mycosis fungoides, acute myelogenous leukemia, acute lymphocytic leukemia, basal cell cancer, Ovarian epithelial cancer, ovarian germ cell carcinoma, male breast cancer, brain cancer, pituitary adenoma, multiple myeloma, gallbladder cancer, biliary tract cancer, colon cancer, chronic myelogenous leukemia, chronic lymphocytic leukemia, retinoblastoma, choroidal melanoma, barter bulge cancer, bladder cancer, peritoneal cancer , Parathyroid cancer, adrenal cancer, non-sinus cancer, non-small cell lung cancer, tongue cancer, astrocytoma, small cell lung cancer, pediatric brain cancer,
  • the compound represented by Formula 1 according to the present invention is a health functional food composition for the prevention or improvement of cancer, in particular, cancer in which EGFR mutation is expressed, by showing a high inhibitory ability against EGFR mutation. Can be added to.
  • the compound represented by Formula 1 according to the present invention may be added to food as it is or may be used together with other foods or food ingredients, and may be appropriately used according to a conventional method.
  • the mixing amount of the active ingredient may be appropriately determined depending on the purpose of use (for prevention or improvement).
  • the amount of the compound in the health food may be added in an amount of 0.1 to 90 parts by weight of the total food weight.
  • the amount may be less than the above range, and there is no problem in terms of safety, so the active ingredient may be used in an amount above the above range.
  • the health functional beverage composition of the present invention is not particularly limited to other ingredients other than containing the compound as an essential ingredient in the indicated ratio, and may contain various flavoring agents or natural carbohydrates, etc. as additional ingredients, as in ordinary beverages.
  • natural carbohydrates include monosaccharides such as glucose, fructose, and the like; Disaccharides such as maltose, sucrose, and the like; And polysaccharides, for example, common sugars such as dextrin and cyclodextrin, and sugar alcohols such as xylitol, sorbitol, and erythritol.
  • natural flavoring agents tacmatin, stevia extract (for example, rebaudioside A, glycyrrhizin, etc.)
  • synthetic flavoring agents sacharin, aspartame, etc.
  • the proportion of the natural carbohydrate is generally about 1 to 20 g, preferably about 5 to 12 g per 100 g of the composition of the present invention.
  • the compound represented by Chemical Formula 1 according to the present invention is a variety of nutrients, vitamins, minerals (electrolytes), flavoring agents such as synthetic flavors and natural flavoring agents, coloring agents and heavy weight agents (cheese, chocolate, etc.), pects. Acids and salts thereof, alginic acid and salts thereof, organic acids, protective colloidal thickeners, pH adjusters, stabilizers, preservatives, glycerin, alcohols, carbonates used in carbonated beverages, and the like.
  • the compound represented by Chemical Formula 1 of the present invention may contain flesh for the manufacture of natural fruit juice and fruit juice beverages and vegetable beverages.
  • the present invention provides a pharmaceutical composition for preventing or treating diseases related to EGFR mutations containing the compound represented by Formula 1, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
  • the EGFR mutation-related disease may be at least one selected from the group consisting of cancer, psoriasis, eczema, atherosclerosis, stomatous rash, dry skin, chronic diarrhea, liver cirrhosis, myocardial fibrosis, and chronic renal failure.
  • the present invention is a cancer or EGFR mutation-related disease comprising the step of administering to a subject in need a pharmaceutical composition or a dietary supplement composition containing the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient It provides a method of preventing or treating.
  • the present invention provides a use of a pharmaceutical composition or a health functional food composition containing the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof in the prevention or treatment of cancer or EGFR mutation-related diseases. .
  • N-(2-((2,5-dichloropyrimidin-4-yl)amino)phenyl)methanesulfonamide was prepared through the method represented by Scheme 2 below.
  • the organic layer was washed with water, brine, dried over Na 2 SO 4 and evaporated under reduced pressure.
  • the obtained crude was dissolved in methanol (50 mL), K 2 CO 3 (2.0 g) was added, and stirred for 12 hours. The stirred liquid was evaporated under reduced pressure. Subsequently, the obtained stock solution was diluted with H 2 O (100 mL) and extracted with DCM (100 mL). The aqueous layer was separated, acidified with 1N HCl (pH ⁇ 5-4), and extracted with DCM.
  • the product stock solution was purified by flash silica chromatography, and was eluted with DCM containing 1 to 10% MeOH. The pure fractions were evaporated and dried to obtain 4-fluoro-2-methoxy-5-nitroaniline (4.5 g, 76% yield, yellow solid).
  • Step 2 Preparation of tert-butyl (4-fluoro-2-methoxy-5-nitrophenyl) carbamate
  • the obtained stock solution was purified using column chromatography in which the eluent was hexanes/ethyl acetate (7:3), and tert-butyl (4-fluoro-2-methoxy -5-nitrophenyl)carbamate (4.0 g, 58% yield, yellow solid) was obtained.
  • N-(5-amino-2((2-(dimethylamino)ethyl(methyl)amino)-4-methoxyphenyl)acrylamide was prepared through the method represented by Scheme 4 below.
  • Step 1 Preparation of tert-butyl (4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)carbamate
  • the tert-butyl (4-fluoro-2-methoxy-5-nitrophenyl) carbamate (300 mg, 1.04 mmol) prepared in Preparation Example 1-2 was dissolved in DMF (5 mL) and added to the stirred solution.
  • N 1 ,N 1 ,N 2 -Trimethylethane-1,2-diamine (106 mg, 1.04 mmol) and cesium carbonate (183 mg, 1.56 mmol) were added at room temperature.
  • the reaction mixture was stirred at 70° C. overnight. Thereafter, the reaction mixture was cooled to room temperature and quenched with ice water.
  • Step 2 Preparation of tert-butyl(5-amino-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)carbamate
  • Step 3 Preparation of tert-butyl (5-acrylamido-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)carbamate
  • Step 4 Preparation of N-(5-amino-2((2-(dimethylamino)ethyl(methyl)amino)-4-methoxyphenyl)acrylamide
  • Step 4 of Preparation Example 1-3 except that tert-butyl (5-acrylamido-2-methoxy-4-morpholinophenyl) carbamate prepared in Preparation Example 4-1 was used as a precursor. According to the following, N-(5-amino-4-methoxy-2-morpholinophenyl)acrylamide was prepared.
  • Steps 1 to 3 of Preparation Example 1-3 except that N-methyl-2-morpholinoethan-1-amine was used instead of N 1 ,N 1 ,N 2 -trimethylethane-1,2-diamine According to the following, tert-butyl (5-acrylamido-2-methoxy-4-(methyl(2-morpholinoethyl)amino)phenyl)carbamate (69% yield) was prepared.
  • N-5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino-2-((2- (Dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide was prepared in the same manner as shown in Scheme 5 below.
  • N-(5-amino-2((2-(dimethylamino)ethyl(methyl)amino)-4-methoxyphenyl)acrylic prepared in Preparation Example 1-3 above in a solution dissolved in (30 mL) and stirred Amide (71.5 mg, 0.3 mmol) was added at room temperature The reaction tube was sealed with a Teflon-lined cap, and the reaction mixture was stirred overnight at 90° C.
  • the pyrimidine derivative compound according to the present invention N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-(4 -(Dimethylamino)piperidin-1-yl)-4-methoxyphenyl)acrylamide, prepared in Preparation Example 2-2, N-(5-amino-2-(4-(dimethylamino)pi) It was prepared according to Example 1, except that peridin-1-yl)-4-methoxyphenyl)acrylamide was used (yield 27%).
  • N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy -2-(4-(4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)acrylamide was prepared in Preparation Example 3-2. It was prepared according to Example 1, except that oxy-2-(4-(4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)acrylamide was used (yield 30%).
  • the pyrimidine derivative compound according to the present invention N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy -2-morpholinophenyl)acrylamide, except that the N-(5-amino-4-methoxy-2-morpholinophenyl)acrylamide prepared in Preparation Example 4-2 was used. It was prepared according to Example 1 (yield 31%).
  • the pyrimidine derivative compound according to the present invention N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy -2-(4-methylpiperazin-1-yl)phenyl)acrylamide, prepared in Preparation Example 5-2, N-(5-amino-4-methoxy-2-(4-methylpiperazine-) It was prepared according to Example 1, except that 1-yl)phenyl)acrylamide was used (yield 41%).
  • the pyrimidine derivative compound according to the present invention N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-(2 -(Dimethylamino)ethoxy)-4-methoxyphenyl)acrylamide, prepared in Preparation Example 6-2, N-(5-amino-2-(2-(dimethylamino)ethoxy)-4- It was prepared according to Example 1 except for methoxyphenyl)acrylamide (yield 29%).
  • N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy -2-(methyl(2-morpholinoethyl)amino)phenyl)acrylamide was prepared in Preparation Example 7-2 above. N-(5-amino-4-methoxy-2-(2-morpholino) It was prepared according to Example 1, except that ethoxy)phenyl)acrylamide was used (yield 22%).
  • N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy -2-(Methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)acrylamide was prepared in Preparation Example 8-2 above N-(5-amino-4-methoxy- It was prepared according to Example 1, except that 2-(methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)acrylamide was used (yield 27%).
  • the pyrimidine derivative compound according to the present invention N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amido)pyrimidin-2-yl)amino)-4-me N-(5-amino-4-methoxy-2-(methyl(2-morpholino) prepared in Preparation Example 9-2) of oxy-2-(2-morpholinoethoxy)phenyl)acrylamide It was prepared according to Example 1, except that ethyl) amino) phenyl) acrylamide was used (yield 26%).
  • the activity measurement for wild-type and EGFR mutant enzymes for the compounds of the present invention was tested as follows using the HTRF system sold by Cisbio. EGFR wild-type and EGFR del19/T790M mutant enzymes were used by purchasing a recombinant protein provided by Carna Biosciences.
  • the composition of the assay buffer used for the activity measurement was 50 mM Tris-HCl pH 7.5, 100 mM NaCl, 7.5 mM MgCl 2 , 3 mM KCl, 0.01% Tween 20, 0.1% BSA, and 1 mM DTT.
  • the enzyme reaction was carried out using a peptide substrate labeled with 50 mM ATP and 0.5 mM biotin.
  • the analysis of the EGFR activity inhibitory effect of the compound was performed according to the following analytical reaction recipe.
  • Component 1 4 mL of EGFR wild-type or mutant enzyme
  • Component 3 4 mL ATP and biotin-labeled peptide
  • Enzymatic reaction starts by mixing component 1 and component 2 first, and then adding component 3 to it. After 2 hours reaction at 37°C, 10 mL of a measurement solution consisting of streptavidin-XL665 and europium-labeled anti-phosphotyrosine antibody provided by Cisbio was added to the enzyme reaction solution and reacted at room temperature for 1 hour. Finally, the ratio of fluorescence values at 615 nm and 665 nm is calculated using Perkin-Elmer's Envision equipment, and the enzyme activity is quantitatively measured and the inhibitory ability of the compound is confirmed. The measured values measured at the concentrations of the seven compounds were analyzed using the Prism program (version 5.01, Graphpad Software, Inc.), and the IC 50 value, an index of the inhibitory ability of the compound, was calculated.
  • All the compounds of the present invention exhibit a relatively weak EGFR activity inhibitory effect against wild-type EGFR, and selectively exhibit high inhibitory ability against EGFR mutations, which is similar to the case of osimertinib, a conventional EGFR tyrosine kinase inhibitor (TKI) drug. Similar things were confirmed.
  • TKI EGFR tyrosine kinase inhibitor
  • the pyrimidine derivative compound according to the present invention can effectively inhibit the EGFR mutation, and thus can be usefully used as a pharmaceutical composition for preventing or treating cancer.
  • the /C797S mutant cell line was used by purchasing a cell line provided by Crown Bioscience.
  • the /C797S mutant cell line was cultured in an RPMI containing 10% FBS and 1% penicillin-streptomycin with 1 ug of puromycine in an incubator at 37°C and 5% CO 2.
  • 2500 cells/90 ⁇ L were cultured by passage in a 96 well cell culture plate, and after 24 hours, the compound represented by Formula 1 was treated with 0, 0.01, 0.03, 0.1, 0.3, 1, 3, 10 ( ⁇ M). After the reaction for 72 hours, the plate treated with the compound was allowed to stand at room temperature for 30 minutes, and then 100 ⁇ L of the reagent was further treated and shaking at room temperature for 10 minutes. Finally, the ratio of the fluorescence value at 570 nm is calculated using an equipment, and the quantitative measurement is performed, and the ability of the compound to inhibit cell growth is confirmed. The measured values measured at the concentrations of the 8 compounds were analyzed using the Prism program (version 5.01, Graphpad Software, Inc.), and the IC 50 value, an indicator of the cell growth inhibitory ability of the compound, was calculated.
  • All the compounds of the present invention exhibited relatively weak inhibitory effect against wild-type EGFR in Ba/F3 cell lines, and selectively against EGFR mutations including triple mutations EGFR del19/T790M/C797S, EGFR L858R/T790M/C797S. It showed high cell growth inhibitory ability, and was found to be particularly superior to the case of osimertinib, an existing EGFR TKI drug.
  • the pyrimidine derivative compound according to the present invention can effectively inhibit the EGFR mutation, and thus can be usefully used as a pharmaceutical composition for preventing or treating cancer.
  • Example 1 In order to confirm the ability of the compound represented by Example 1 according to the present invention to inhibit cell growth against the Ba/F3 EGFR exon20 insertion mutation in the Ba/F3 cell line, the following experiment was performed. The results are shown in Table 5 below.
  • the activity measurement of the mutant Ba/F3 EGFR cell line for the compound of the present invention was tested as follows using the CellTiter-Glo system sold by Promega.
  • CellTiter-Glo assay is a method to check cell viability by measuring ATP present in cells in the cell culture state.
  • Ba/F3 EGFR D770delinsGY, H773_V774insH, Y764_V765insHH, V769_D770insASV, D770_N771insSVD mutant cell lines were purchased and used in Pasi A Janne laboratory of Dana-Farber Cancer Institute.
  • Ba/F3 D770delinsGY, H773_V774insH, Y764_V765insHH, V769_D770insASV, D770_N771insSVD mutant cell lines were cultured in an incubator at 37° C., 5% CO 2 by adding 1 ⁇ g/ml of puromycine to RPMI containing 10% FBS and 1% penicillin-streptomycin.
  • Ba/F3 EGFR V769_D770insASV IC 50 ( ⁇ M) Ba/F3 EGFR D770delinsGY IC 50 ( ⁇ M) Ba/F3 EGFR H773_V774insH IC 50 ( ⁇ M) Ba/F3 EGFR Y764_V765insHH IC 50 ( ⁇ M) Ba/F3 EGFR D770_N771insSVD IC 50 ( ⁇ M) Osimertinib 0.124 0.159 0.191 0.136 0.149 Example 1 0.026 0.021 0.06 0.029 0.027
  • the compound represented by Example 1 according to the present invention exhibits high cell growth inhibitory ability against Ba/F3 EGFR exon20 insertion mutations in Ba/F3 cell lines, which is the case of osimertinib, an existing EGFR tyrosine kinase inhibitor (TKI) drug. It was confirmed that it is more excellent.
  • TKI EGFR tyrosine kinase inhibitor
  • the pyrimidine derivative compound according to the present invention can effectively inhibit the EGFR mutation, and thus can be usefully used as a pharmaceutical composition for preventing or treating cancer.
  • the above ingredients were mixed and filled in an airtight cloth to prepare a powder.
  • tablets were prepared by tableting according to a conventional tablet preparation method.
  • a gelatin capsule was filled according to a conventional capsule preparation method to prepare a capsule.
  • injections were prepared by containing the above ingredients in the indicated amount.
  • the ingredients suitable for health food are mixed and formulated as a preferred formulation example, but the mixing ratio may be arbitrarily modified.
  • the resulting solution After mixing the above ingredients according to a conventional health drink manufacturing method, after stirring and heating at 85° C. for about 1 hour, the resulting solution is filtered and obtained in a sterilized container, sealed and sterilized, and then stored in a refrigerator. It was used in preparation.
  • composition ratio is a mixture of ingredients suitable for a relatively preferred beverage in a preferred embodiment, but the mixing ratio may be arbitrarily modified according to regional and ethnic preferences such as the demand class, the country of demand, and the purpose of use.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Mycology (AREA)
  • Nutrition Science (AREA)
  • Engineering & Computer Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a pyrimidine derivative, a method of preparing same, and a pharmaceutical composition for preventing or treating cancer, comprising the pyrimidine derivative as an effective component, wherein the derivative exhibits a relatively weak inhibitory effect on EGFR activity with respect to wild-type EGFR while exhibiting a high inhibitory effect with respect to EGFR mutations, and thus can be effectively used for the treatment of cancer with EGFR mutations.

Description

피리미딘 유도체, 이의 제조방법 및 이를 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물Pyrimidine derivative, preparation method thereof, and pharmaceutical composition for preventing or treating cancer containing the same as an active ingredient
본 발명은 암의 예방 또는 치료용 약학적 조성물에 관한 것으로, 구체적으로 피리미딘 유도체를 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물에 관한 것이다.The present invention relates to a pharmaceutical composition for preventing or treating cancer, and specifically to a pharmaceutical composition for preventing or treating cancer containing a pyrimidine derivative as an active ingredient.
암의 발생은 화학물질, 방사선, 바이러스를 포함하는 여러 가지 환경적인 요인과 종양 유전자, 종양 억제 유전자, 세포사멸(apoptosis)과 DNA 복구에 관련된 유전자의 변화 등에 관련되어 있는데, 최근 이러한 암의 분자적 메커니즘을 이해함에 따라 새로운 치료법인 표적 항암치료가 가능하게 되었다.Cancer incidence is related to changes in various environmental factors including chemicals, radiation, and viruses, oncogenes, tumor suppressor genes, and genes related to apoptosis and DNA repair. By understanding the mechanism, a new treatment, targeted chemotherapy, became possible.
표적 치료제들은 일반적으로 암세포가 특징적으로 가지고 있는 분자를 표적으로 하여 그 효과를 나타낼 수 있도록 만들어지며, 분자적 표적이 되는 것은 암세포의 신호전달경로(signal transduction pathway), 혈관신생(angiogenesis), 세포간질(matrix), 세포주기조절인자(cell cycle regulator), 세포사멸(apoptosis) 등에 관련된 유전자들이다. 현재 치료에서 중요한 표적 치료제로 사용되고 있는 것으로는 티로신 키나아제(tyrosine kinase) 억제제를 비롯한 '신호전달경로 억제제'와 '신생혈관생성 억제제'들이 있다.Targeted therapies are generally made to target molecules characteristic of cancer cells to exert their effects, and molecular targets are cancer cell signal transduction pathways, angiogenesis, and interstitial cells. They are genes related to (matrix), cell cycle regulator, and apoptosis. Currently used as important target therapeutics in treatment include tyrosine kinase inhibitors, signaling pathway inhibitors, and angiogenesis inhibitors.
단백질 티로신 키나아제(protein tyrosine kinase)는 많은 악성 종양에서 중요한 역할을 하는 것으로 밝혀졌으며, 특히 erbB 패밀리의 수용체 티로신 키나아제(receptor tyrosine kinase)인 상피 성장 인자 수용체(epidermal growth factor receptor, EGFR)는 비소세포폐암종(NSCLC), 유방암, 신경교종, 두경부의 편평 세포 암종, 대장암, 곧창자 생암종, 두경부암, 위암 및 전립선암을 포함한 많은 상피세포 종양에서 비정상적으로 활성화되어 있고, 상기 EGFR-티로신 키나아제의 활성화가 지속적인 세포 증식, 주변 조직에 대한 침범, 원격 전이, 혈관 형성을 일으키며 세포 생존을 증가시킴이 알려진 바 있다.Protein tyrosine kinase has been shown to play an important role in many malignancies. In particular, epidermal growth factor receptor (EGFR), a receptor tyrosine kinase of the erbB family, is a non-small cell lung cancer. It is abnormally activated in many epithelial cell tumors, including NSCLC, breast cancer, glioma, squamous cell carcinoma of the head and neck, colon cancer, rectal carcinoma, head and neck cancer, gastric cancer, and prostate cancer. It has been known that activation causes continuous cell proliferation, invasion of surrounding tissues, distant metastasis, and blood vessel formation, and increases cell survival.
구체적으로, 상기 EGFR은 ErbB 티로신키나아제 수용체군(tyrosine kinase receptors family; EGFR, HER-2, ErbB-3, ErbB-4) 중의 하나로, 세포외 리간드결합영역(extracelluar ligand-binding domain)과 티로신 키나아제영역(tyrosine kinase domain)을 포함한 세포내 영역(intracellular domain)을 가지고 있는 막경유 티로신키나아제(transmembrane tyrosine kinase)이다. 호모다이머(homodimer) 또는 헤테로다이머(heterodimer)를 이룬 수용체에 리간드가 결합하면 세포내의 티로신키나아제가 활성화되고 이렇게 EGFR에 의해 자극된 신호는 포스파티딜이노지톨 3-키나아제(phosphatidylinositol 3-kinase(PI3K)/AKT/mTOR, RAS/RAF/MAPK, JAK/STAT) 신호전달 경로를 활성화한다(Nat Rev Cancer 2007;7:169-81).Specifically, the EGFR is one of the ErbB tyrosine kinase receptors family (EGFR, HER-2, ErbB-3, ErbB-4), an extracelluar ligand-binding domain and a tyrosine kinase region It is a transmembrane tyrosine kinase that has an intracellular domain including (tyrosine kinase domain). When a ligand is bound to a receptor that forms a homodimer or heterodimer, the intracellular tyrosine kinase is activated, and the signal stimulated by EGFR is phosphatidylinositol 3-kinase (PI3K)/ AKT/mTOR, RAS/RAF/MAPK, JAK/STAT) signaling pathways are activated (Nat Rev Cancer 2007; 7:169-81).
EGFR은 특히, 비소세포폐암(non-small cell lung cancer, NSCLC)의 절반이상에서 과발현되어 치료의 표적으로 많은 연구들이 시행되었다. EGFR 티로신키나아제 활성을 억제하는 EGFR TKI(tyrosine kinase inhibitor)가 개발되었으며, 대표적인 약제로는 제피티닙(IRESSA™), 에를로티닙(TARCEVA™), 라파티닙(TYKERB™, TYVERB™)이 있다.In particular, EGFR is overexpressed in more than half of non-small cell lung cancer (NSCLC), and many studies have been conducted as a target of treatment. EGFR TKI (tyrosine kinase inhibitor), which inhibits EGFR tyrosine kinase activity, has been developed, and representative drugs include gefitinib (IRESSA™), erlotinib (TARCEVA™), and lapatinib (TYKERB™, TYVERB™).
한편, 2004년 EGFR의 활성화 돌연변이가 비소세포 폐암(NSCLC:non-small-cell lung cancer)에서 제피티닙 요법에 대한 반응과 상관관계가 있다는 것이 보고되었다(Science [2004] Vol.304, 1497-500 및 New England Journal of Medicine [2004] Vol. 350, 2129-39). Meanwhile, in 2004, it was reported that the activating mutation of EGFR correlates with the response to gefitinib therapy in non-small-cell lung cancer (NSCLC) (Science [2004] Vol.304, 1497- 500 and New England Journal of Medicine [2004] Vol. 350, 2129-39).
구체적으로, 상기 EGFR 돌연변이는 크게 민감성(sensitizing) 돌연변이와 내성(resistant) 돌연변이로 구분되는데 엑손 19의 결손(deletion)과 엑손 21의 L858R 점 돌연변이(point mutation)가 가장 중요한 민감성 돌연변이로서 약 85∼90%를 차지하고 있고 엑손 19 del 돌연변이가 TKI에 대한 민감성이 더 좋은 것으로 알려져 있다. 반면 엑손 20의 T790M 점 돌연변이는 가장 중요한 내성 돌연변이로서 획득 내성 환자의 50% 이상에서 발견되는 것으로 알려져 있다(Clin Cancer Res 2006;12:6494-6501.).Specifically, the EGFR mutation is largely classified into a sensitizing mutation and a resistant mutation. The deletion of exon 19 and the L858R point mutation of exon 21 are the most important sensitive mutations. %, and exon 19 del mutations are known to be more sensitive to TKI. On the other hand, the T790M point mutation of exon 20 is the most important resistance mutation and is known to be found in more than 50% of acquired resistance patients (Clin Cancer Res 2006;12:6494-6501.).
지금까지 동정된 체세포 돌연변이에는 엑손 19 내 틀내 결손 또는 엑손 20 내 삽입뿐만 아니라, 발현된 단백질 내에서 단일 핵산 잔기가 변형된 점 돌연변이(예컨대, L858R, G719S, G719C, G719A, L861Q)가 포함된다(Fukuoka et al. JCO 2003; Kris et al JAMA 2003 and Shepherd et al NEJM 2004).Somatic mutations identified so far include in-frame deletions within exon 19 or insertions in exon 20, as well as point mutations in which a single nucleic acid residue is modified in the expressed protein (e.g., L858R, G719S, G719C, G719A, L861Q) ( Fukuoka et al. JCO 2003; Kris et al JAMA 2003 and Shepherd et al NEJM 2004).
EGFR 돌연변이를 갖는 NSCLC 환자에게서 제피티닙/에를로티닙의 초기 임상 효과에도 불구하고, 대부분의 환자에게서 결국에는 이들 제제에 대한 요법을 받는 동안 진행성 암이 발병한다. 재발된 표본의 초기연구에서 제피티닙 및 에를로티닙을 EGFR 키나아제 활성의 비효과적인 억제제가 되게 하는 이차 EGFR 돌연변이, T790M가 동정되었다(Kobayashi et al NEJM 2005 and Pao et al PLOS Medicine 2005). EGFR T790M 돌연변이가 제피티닙 또는 에를로티닙에 대해 내성을 획득한 환자 유래 종양의 대략 50%(24/48)에서 발견됨이 후속 연구에서 입증되었다(Kosaka et al CCR 2006; Balak et al CCR 2006 and Engelman et al Science 2007). 이러한 이차 유전적 변형은 키나아제 억제제로 치료된 환자에게서 '게이트키퍼(gatekeeper)' 잔기 및 이것과 연관된 이차 내성 대립 유전자와 유사한 위치에서 야기된다(예를 들어, 이마티닙 내성 CML에서 ABL 내 T315I).Despite the initial clinical effects of gefitinib/erlotinib in NSCLC patients with EGFR mutations, most patients eventually develop advanced cancer during therapy with these agents. In an initial study of relapsed specimens, a secondary EGFR mutation, T790M, was identified that renders gefitinib and erlotinib ineffective inhibitors of EGFR kinase activity (Kobayashi et al NEJM 2005 and Pao et al PLOS Medicine 2005). It was demonstrated in subsequent studies that the EGFR T790M mutation was found in approximately 50% (24/48) of tumors from patients who acquired resistance to gefitinib or erlotinib (Kosaka et al CCR 2006; Balak et al CCR 2006 and Engelman et al Science 2007). This secondary genetic modification results from a position similar to the'gatekeeper' residue and the secondary resistance allele associated with it in patients treated with kinase inhibitors (e.g., T315I in ABL in imatinib resistant CML).
EGFR 돌연변이인 EGFR_del19 또는 EGFR_L858R이 비소세포폐암과 두경부암의 주요한 원인이라는 것은 오래 전부터 알려져 왔고, 이들의 치료약물인 이레사, 타세바가 개발되어 현재 임상에서 사용되고 있다. 하지만, 이러한 약물을 환자에 사용하였을 때 약물의 구조에 기반을 두는 EGFR 2차 돌연변이가 생기는 획득내성(acquired resistance)이 관찰되었고, 이것이 실제 약제내성의 주요원인이라는 것도 밝혀졌다. EGFR 1세대 저해제를 평균 10개월 정도 사용하게 되면 EGFR 키나아제의 게이트키퍼(gatekeeper)에 위치한 T790M 돌연변이라는 획득내성이 발생하여 EGFR 1세대 저해제들이 약효를 내지 못하는 것이다. 즉, EGFR_del19_T790M 또는 EGFR_L858R_T790M 이중돌연변이가 발생하여 기존 치료제가 약효를 나타내지 못하게 된다.It has long been known that EGFR mutations, EGFR_del19 or EGFR_L858R, are the main causes of non-small cell lung cancer and head and neck cancer, and their therapeutic drugs, Iressa and Taseba, have been developed and are currently used in clinical practice. However, when these drugs were used in patients, acquired resistance, which caused the EGFR secondary mutation based on the structure of the drug, was observed, and it was also found that this is the main cause of actual drug resistance. When the first generation EGFR inhibitor is used for an average of 10 months, the acquired resistance of the T790M mutation located at the gatekeeper of the EGFR kinase occurs, and the first generation EGFR inhibitors do not take effect. In other words, EGFR_del19_T790M or EGFR_L858R_T790M double mutation occurs, preventing the existing treatment from showing efficacy.
이러한 사실을 기반으로 약효가 우수하면서 새로운 구조를 가지고 있는 2세대, 3세대 약물 개발에 대한 필요성이 대두되고 있다.Based on these facts, the need for the development of 2nd and 3rd generation drugs with excellent drug efficacy and new structures is emerging.
또한 EGFR_del19 또는 EGFR_L858R 돌연변이가 아닌 희귀 EGFR 돌연변이에 대한 적절한 치료약제도 없는 상황인데, 특히 EGFR Exon20insertion 돌연변이에 대한 적절한 치료약제가 없다. EGFR Exon20insertion 돌연변이는 매우 다양한데 EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV 및 EGFR D770_N771insSVD 등이 포함된다.In addition, there is no suitable therapeutic agent for rare EGFR mutations other than EGFR_del19 or EGFR_L858R mutations. In particular, there is no suitable therapeutic agent for EGFR Exon20insertion mutation. There is a wide variety of EGFR Exon20insertion mutations, including EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV and EGFR D770_N771insSVD.
이에, 야생형(WT) EGFR에 대해 상대적으로 낮은 억제를 보임과 동시에, 특정 활성화 또는 내성 돌연변이체 형태의 EGFR에 대해 더 높은 억제를 나타내는 억제제의 개발이 요구되고 있다.Accordingly, there is a need for the development of inhibitors that exhibit relatively low inhibition against wild-type (WT) EGFR and higher inhibition against specific activated or resistant mutant forms of EGFR.
이에, EGFR 다중돌연변이를 억제하는 암 치료제를 개발하기 위하여 노력하던 중, 본 발명에 따른 피리미딘 유도체가 야생형 EGFR에 대하여 상대적으로 낮은 억제를 나타내면서, EGFR 돌연변이에 대하여 높은 억제능을 나타냄으로써, 암의 예방 또는 치료에 유용하게 사용될 수 있음을 알아내어 본 발명을 완성하였다.Accordingly, while trying to develop a cancer therapeutic agent that inhibits EGFR multiple mutations, the pyrimidine derivatives according to the present invention exhibit relatively low inhibition against wild-type EGFR, while exhibiting high inhibitory ability against EGFR mutations, thereby preventing cancer. Alternatively, the present invention was completed by finding that it can be usefully used for treatment.
본 발명의 목적은 피리미딘 유도체, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 제공하는 것이다.It is an object of the present invention to provide a pyrimidine derivative, an isomer thereof, or a pharmaceutically acceptable salt thereof.
본 발명의 다른 목적은 상기 피리미딘 유도체의 제조방법을 제공하는 것이다.Another object of the present invention is to provide a method for preparing the pyrimidine derivative.
본 발명의 다른 목적은 상기 피리미딘 유도체, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition for the prevention or treatment of cancer containing the pyrimidine derivative, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
본 발명의 다른 목적은 상기 피리미딘 유도체, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 암의 예방 또는 개선용 건강기능식품 조성물을 제공하는 것이다.Another object of the present invention is to provide a health functional food composition for preventing or improving cancer containing the pyrimidine derivative, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
본 발명의 다른 목적은 상기 피리미딘 유도체, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 EGFR 돌연변이 관련 질환의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition for preventing or treating diseases related to EGFR mutations containing the pyrimidine derivative, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
상기 목적을 달성하기 위하여,To achieve the above object,
본 발명의 일 측면에 따라, 하기 화학식 1로 표시되는 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 제공한다:According to an aspect of the present invention, there is provided a compound represented by the following formula (1), an isomer thereof, or a pharmaceutically acceptable salt thereof:
[화학식 1][Formula 1]
Figure PCTKR2020010657-appb-img-000001
Figure PCTKR2020010657-appb-img-000001
상기 화학식 1에서,In Formula 1,
R은 -OR a 또는 -NR b1R b2이고,R is -OR a or -NR b1 R b2 ,
R a는 -(CH 2) m-NR b1R b2이고, 여기서 m은 1 내지 3이고,R a is -(CH 2 ) m -NR b1 R b2 , where m is 1 to 3,
R b1 및 R b2는 각각 독립적으로 수소 또는 비치환된 또는 NR c1R c2로 치환된 직쇄 또는 분지쇄 C 1-6알킬이고, 또는, R b1 및 R b2는 이들이 결합된 질소와 함께 N, O 및 S로 이루어진 군에서 선택되는 하나 이상의 헤테로 원자를 하나 이상 포함하는 비치환된 또는 직쇄 또는 분지쇄 C 1-6알킬 또는 NR c1R c2로 치환된 5 내지 7 원자의 헤테로시클로알킬을 형성하고,R b1 and R b2 are each independently hydrogen or unsubstituted or straight or branched chain C 1-6 alkyl substituted with NR c1 R c2 , or R b1 and R b2 are N, O together with the nitrogen to which they are attached. And to form an unsubstituted or straight or branched chain C 1-6 alkyl containing one or more heteroatoms selected from the group consisting of S or a 5 to 7 membered heterocycloalkyl substituted with NR c1 R c2,
R c1 및 R c2는 각각 독립적으로 수소 또는 직쇄 또는 분지쇄 C 1-6알킬이고, 또는, R c1 및 R c2는 이들이 결합된 질소와 함께 N, O 및 S로 이루어진 군에서 선택되는 하나 이상의 헤테로 원자를 하나 이상 포함하는 비치환된 또는 직쇄 또는 분지쇄 C 1-6알킬로 치환된 5 내지 7 원자의 헤테로시클로알킬을 형성한다.R c1 and R c2 are each independently hydrogen or straight or branched chain C 1-6 alkyl, or, R c1 and R c2 are selected from the group consisting of N, O and S together with the nitrogen to which they are attached. 5 to 7 membered heterocycloalkyl substituted with unsubstituted or straight or branched C 1-6 alkyl containing one or more atoms.
본 발명의 다른 측면에 따라, 하기 반응식 1에 나타낸 바와 같이,According to another aspect of the present invention, as shown in Scheme 1 below,
화학식 2로 표시되는 화합물과 화학식 3으로 표시되는 화합물을 반응시켜 화학식 1로 표시되는 화합물을 제조하는 단계를 포함하는 상기 화학식 1로 표시되는 화합물의 제조방법을 제공한다:It provides a method for preparing a compound represented by Formula 1, comprising the step of preparing a compound represented by Formula 1 by reacting a compound represented by Formula 2 with a compound represented by Formula 3:
[반응식 1][Scheme 1]
Figure PCTKR2020010657-appb-img-000002
Figure PCTKR2020010657-appb-img-000002
상기 반응식 1에서 R은 상기 화학식 1에서 정의한 바와 같다.In Reaction Scheme 1, R is as defined in Chemical Formula 1.
본 발명의 다른 측면에 따라, 상기 화학식 1로 표시되는 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물을 제공한다.According to another aspect of the present invention, there is provided a pharmaceutical composition for preventing or treating cancer containing the compound represented by Chemical Formula 1, an isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.
본 발명의 다른 측면에 따라, 상기 화학식 1로 표시되는 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 암의 예방 또는 개선용 건강기능식품을 제공한다.According to another aspect of the present invention, there is provided a health functional food for preventing or improving cancer containing the compound represented by Formula 1, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
본 발명의 다른 측면에 따라, 상기 화학식 1로 표시되는 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 EGFR 돌연변이 관련 질환의 예방 또는 치료용 약학적 조성물을 제공한다.According to another aspect of the present invention, there is provided a pharmaceutical composition for preventing or treating diseases related to EGFR mutations containing the compound represented by Formula 1, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
본 발명의 다른 측면에 따라, 상기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 약학적 조성물 또는 건강기능식품 조성물을 필요한 대상에게 투여하는 단계를 포함하는 암 또는 EGFR 돌연변이 관련 질환의 예방 또는 치료 방법을 제공한다.According to another aspect of the present invention, cancer or EGFR comprising administering to a subject in need a pharmaceutical composition or a dietary supplement composition containing the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient It provides a method of preventing or treating mutation-related diseases.
본 발명의 다른 측면에 따라, 암 또는 EGFR 돌연변이 관련 질환의 예방 또는 치료에 있어서의, 상기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 함유하는 약학적 조성물 또는 건강기능식품 조성물의 용도를 제공한다.According to another aspect of the present invention, use of a pharmaceutical composition or a dietary supplement composition containing a compound represented by Formula 1 or a pharmaceutically acceptable salt thereof in the prevention or treatment of cancer or EGFR mutation-related diseases Provides.
본 발명에 따른 피리미딘 유도체는 야생형 EGFR에 대하여 상대적으로 약한 EGFR 활성억제효과를 나타내면서, EGFR 돌연변이에 대하여 높은 억제능을 나타내므로, EGFR 돌연변이가 발생된 암의 치료에 유용하게 사용될 수 있다.The pyrimidine derivative according to the present invention exhibits a relatively weak inhibitory effect on EGFR activity against wild-type EGFR, and exhibits a high inhibitory ability against EGFR mutations, so it can be usefully used in the treatment of cancer in which EGFR mutations have occurred.
도 1은 기존 EGFR TKI(tyrosine kinase inhibitor) 약물인 오시머티닙(osimertinib)의 Ba/F3 EGFR exon20 insertion 돌연변이에 대한 세포성장 억제능을 측정한 결과를 나타내는 그래프이다.1 is a graph showing the results of measuring the cell growth inhibitory ability of the existing EGFR TKI (tyrosine kinase inhibitor) drug osimertinib against the Ba/F3 EGFR exon20 insertion mutation.
도 2는 실시예 1로 표시되는 화합물의 Ba/F3 EGFR exon20 insertion 돌연변이에 대한 세포성장 억제능을 측정한 결과를 나타내는 그래프이다.2 is a graph showing the results of measuring the cell growth inhibitory ability of the compound represented by Example 1 against the Ba/F3 EGFR exon20 insertion mutation.
이하, 본 발명을 상세히 설명한다. Hereinafter, the present invention will be described in detail.
본 발명은 하기 화학식 1로 표시되는 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 제공한다.The present invention provides a compound represented by the following formula (1), an isomer thereof, or a pharmaceutically acceptable salt thereof.
[화학식 1][Formula 1]
Figure PCTKR2020010657-appb-img-000003
Figure PCTKR2020010657-appb-img-000003
상기 화학식 1에서,In Formula 1,
R은 -OR a 또는 -NR b1R b2이고,R is -OR a or -NR b1 R b2 ,
R a는 -(CH 2) m-NR b1R b2이고, 여기서 m은 1 내지 3이고,R a is -(CH 2 ) m -NR b1 R b2 , where m is 1 to 3,
R b1 및 R b2는 각각 독립적으로 수소 또는 비치환된 또는 NR c1R c2로 치환된 직쇄 또는 분지쇄 C 1-6알킬이고, 또는, R b1 및 R b2는 이들이 결합된 질소와 함께 N, O 및 S로 이루어진 군에서 선택되는 하나 이상의 헤테로 원자를 하나 이상 포함하는 비치환된 또는 직쇄 또는 분지쇄 C 1-6알킬 또는 NR c1R c2로 치환된 5 내지 7 원자의 헤테로시클로알킬을 형성하고,R b1 and R b2 are each independently hydrogen or unsubstituted or straight or branched chain C 1-6 alkyl substituted with NR c1 R c2 , or R b1 and R b2 are N, O together with the nitrogen to which they are attached. And to form an unsubstituted or straight or branched chain C 1-6 alkyl containing one or more heteroatoms selected from the group consisting of S or a 5 to 7 membered heterocycloalkyl substituted with NR c1 R c2,
R c1 및 R c2는 각각 독립적으로 수소 또는 직쇄 또는 분지쇄 C 1-6알킬이고, 또는, R c1 및 R c2는 이들이 결합된 질소와 함께 N, O 및 S로 이루어진 군에서 선택되는 하나 이상의 헤테로 원자를 하나 이상 포함하는 비치환된 또는 직쇄 또는 분지쇄 C 1-6알킬로 치환된 5 내지 7 원자의 헤테로시클로알킬을 형성한다.R c1 and R c2 are each independently hydrogen or straight or branched chain C 1-6 alkyl, or, R c1 and R c2 are selected from the group consisting of N, O and S together with the nitrogen to which they are attached. 5 to 7 membered heterocycloalkyl substituted with unsubstituted or straight or branched C 1-6 alkyl containing one or more atoms.
상기 화학식 1에 있어서,In Formula 1,
상기 R은
Figure PCTKR2020010657-appb-img-000004
,
Figure PCTKR2020010657-appb-img-000005
,
Figure PCTKR2020010657-appb-img-000006
,
Figure PCTKR2020010657-appb-img-000007
,
Figure PCTKR2020010657-appb-img-000008
,
Figure PCTKR2020010657-appb-img-000009
,
Figure PCTKR2020010657-appb-img-000010
,
Figure PCTKR2020010657-appb-img-000011
또는
Figure PCTKR2020010657-appb-img-000012
일 수 있다.
R is
Figure PCTKR2020010657-appb-img-000004
,
Figure PCTKR2020010657-appb-img-000005
,
Figure PCTKR2020010657-appb-img-000006
,
Figure PCTKR2020010657-appb-img-000007
,
Figure PCTKR2020010657-appb-img-000008
,
Figure PCTKR2020010657-appb-img-000009
,
Figure PCTKR2020010657-appb-img-000010
,
Figure PCTKR2020010657-appb-img-000011
or
Figure PCTKR2020010657-appb-img-000012
Can be
본 발명에 따른 상기 화학식 1로 표시되는 화합물의 예로는 하기의 화합물들을 들 수 있다:Examples of the compound represented by Formula 1 according to the present invention include the following compounds:
<1> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-2-((2-(디메틸아미노)에틸)(메틸)아미노)-4-메톡시페닐)아크릴아미드;<1> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-((2-(dimethylamino) Ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide;
<2> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-2-(4-(디메틸아미노)피페리딘-1-일)-4-메톡시페닐)아크릴아미드;<2> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-(4-(dimethylamino)pi Peridin-1-yl)-4-methoxyphenyl)acrylamide;
<3> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)아크릴아미드;<3> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-(4- (4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)acrylamide;
<4> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-모폴리노페닐)아크릴아미드;<4> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-morpholino Phenyl)acrylamide;
<5> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(4-메틸피페라진-1-일)페닐)아크릴아미드;<5> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-(4- Methylpiperazin-1-yl)phenyl)acrylamide;
<6> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-2-(2-(디메틸아미노)에톡시)-4-메톡시페닐)아크릴아미드;<6> to N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-(2-(dimethylamino) Oxy)-4-methoxyphenyl)acrylamide;
<7> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(메틸(2-모폴리노에틸)아미노)페닐)아크릴아미드;<7> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-(methyl( 2-morpholinoethyl)amino)phenyl)acrylamide;
<8> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)아크릴아미드; 및<8> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-(methyl( 2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)acrylamide; And
<9> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미도)피리미딘-2-일)아미노)-4-메톡시-2-(2-모폴리노에톡시)페닐)아크릴아미드.<9> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amido)pyrimidin-2-yl)amino)-4-methoxy-2-(2 -Morpholinoethoxy)phenyl)acrylamide.
본 발명의 상기 화학식 1로 표시되는 화합물은 약학적으로 허용가능한 염의 형태로 사용할 수 있으며, 염으로는 약학적으로 허용가능한 유리산(free acid)에 의해 형성된 산 부가염이 유용하다. 산 부가염은 염산, 질산, 인산, 황산, 브롬화수소산, 요드화수소산, 아질산, 아인산 등과 같은 무기산류, 지방족 모노 및 디카르복실레이트, 페닐-치환된 알카노에이트, 하이드록시 알카노에이트 및 알칸디오에이트, 방향족 산류, 지방족 및 방향족 설폰산류 등과 같은 무독성 유기산, 아세테이트, 안식향산, 구연산, 젖산, 말레인산, 글루콘산, 메탄설폰산, 4-톨루엔설폰산, 주석산, 푸마르산 등과 같은 유기산으로부터 얻는다. 이러한 약학적으로 무독한 염의 종류로는 설페이트, 피로설페이트, 바이설페이트, 설파이트, 바이설파이트, 니트레이트, 포스페이트, 모노하이드로겐 포스페이트, 디하이드로겐 포스페이트, 메타포스페이트, 피로포스페이트 클로라이드, 브로마이드, 아이오다이드, 플루오라이드, 아세테이트, 프로피오네이트, 데카노에이트, 카프릴레이트, 아크릴레이트, 포메이트, 이소부티레이트, 카프레이트, 헵타노에이트, 프로피올레이트, 옥살레이트, 말로네이트, 석시네이트, 수베레이트, 세바케이트, 푸마레이트, 말리에이트, 부틴-1,4-디오에이트, 헥산-1,6-디오에이트, 벤조에이트, 클로로벤조에이트, 메틸벤조에이트, 디니트로 벤조에이트, 하이드록시벤조에이트, 메톡시벤조에이트, 프탈레이트, 테레프탈레이트, 벤젠설포네이트, 톨루엔설포네이트, 클로로벤젠설포네이트, 크실렌설포네이트, 페닐아세테이트, 페닐프로피오네이트, 페닐부티레이트, 시트레이트, 락테이트, β-하이드록시부티레이트, 글리콜레이트, 말레이트, 타트레이트, 메탄설포네이트, 프로판설포네이트, 나프탈렌-1-설포네이트, 나프탈렌-2-설포네이트, 만델레이트 등을 포함한다.The compound represented by Formula 1 of the present invention can be used in the form of a pharmaceutically acceptable salt, and an acid addition salt formed by a pharmaceutically acceptable free acid is useful. Acid addition salts include inorganic acids such as hydrochloric acid, nitric acid, phosphoric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, nitrous acid, phosphorous acid, etc., aliphatic mono and dicarboxylates, phenyl-substituted alkanoates, hydroxy alkanoates and alkanes. It is obtained from non-toxic organic acids such as dioate, aromatic acids, aliphatic and aromatic sulfonic acids, etc., acetate, benzoic acid, citric acid, lactic acid, maleic acid, gluconic acid, methanesulfonic acid, 4-toluenesulfonic acid, tartaric acid, fumaric acid, and the like. Examples of such pharmaceutically non-toxic salts include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogen phosphate, dihydrogen phosphate, metaphosphate, pyrophosphate chloride, bromide, i. Odide, fluoride, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, sube Rate, sebacate, fumarate, maleate, butine-1,4-dioate, hexane-1,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitro benzoate, hydroxybenzoate, Methoxybenzoate, phthalate, terephthalate, benzenesulfonate, toluenesulfonate, chlorobenzenesulfonate, xylenesulfonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, β-hydroxybutyrate, Glycolate, malate, tartrate, methanesulfonate, propanesulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, mandelate, and the like.
본 발명에 따른 산 부가염은 통상의 방법으로 제조할 수 있으며, 예를 들면 화학식 1의 유도체를 메탄올, 에탄올, 아세톤, 메틸렌클로라이드, 아세토니트릴 등과 같은 유기용매에 녹이고 유기산 또는 무기산을 가하여 생성된 침전물을 여과, 건조시켜 제조하거나, 용매와 과량의 산을 감압 증류한 후 건조시켜 유기용매 하에서 결정화시켜셔 제조할 수 있다. The acid addition salt according to the present invention can be prepared by a conventional method, for example, a precipitate formed by dissolving the derivative of Formula 1 in an organic solvent such as methanol, ethanol, acetone, methylene chloride, acetonitrile, etc. and adding an organic or inorganic acid It can be prepared by filtration and drying, or it can be prepared by distilling a solvent and an excess of acid under reduced pressure and then drying to crystallize under an organic solvent.
또한, 염기를 사용하여 약학적으로 허용가능한 금속염을 만들 수 있다. 알칼리 금속 또는 알칼리 토금속 염은 예를 들면 화합물을 과량의 알칼리 금속 수산화물 또는 알칼리 토금속 수산화물 용액 중에 용해하고, 비용해 화합물 염을 여과하고, 여액을 증발, 건조시켜 얻는다. 이때, 금속염으로는 나트륨, 칼륨 또는 칼슘염을 제조하는 것이 제약상 적합하다. 또한, 이에 대응하는 염은 알칼리 금속 또는 알칼리 토금속 염을 적당한 음염(예, 질산은)과 반응시켜 얻는다.In addition, a pharmaceutically acceptable metal salt can be made using a base. The alkali metal or alkaline earth metal salt is obtained, for example, by dissolving the compound in an excess alkali metal hydroxide or alkaline earth metal hydroxide solution, filtering the undissolved compound salt, and evaporating and drying the filtrate. At this time, it is pharmaceutically suitable to prepare sodium, potassium or calcium salt as the metal salt. In addition, the corresponding salt is obtained by reacting an alkali metal or alkaline earth metal salt with a suitable negative salt (eg, silver nitrate).
나아가, 본 발명은 상기 화학식 1로 표시되는 화합물 및 이의 약학적으로 허용가능한 염뿐만 아니라, 이로부터 제조될 수 있는 용매화물, 이성질체, 수화물 등을 모두 포함한다.Furthermore, the present invention includes not only the compound represented by Formula 1 and its pharmaceutically acceptable salts, but also solvates, isomers, hydrates, etc. that may be prepared therefrom.
용어 "용매화물(solvate)"은 비공유적 분자간력에 의해 결합된 화학양론적 또는 비화학양론적 양의 용매를 포함하고 있는 본 발명의 화합물 또는 그것의 염을 의미한다. 그에 관한 바람직한 용매들로는 휘발성, 비독성, 및/또는 인간에게 투여되기에 적합한 용매들이 있다. 이때, 상기 용매가 물인 경우는 "수화물"이라 지칭한다.The term "solvate" refers to a compound of the present invention or a salt thereof containing a stoichiometric or non-stoichiometric amount of a solvent bound by non-covalent intermolecular forces. Preferred solvents therefor include volatile, non-toxic, and/or suitable solvents for administration to humans. In this case, when the solvent is water, it is referred to as "hydrate".
용어 "이성질체(isomer)"는 동일한 화학식 또는 분자식을 가지지만 구조적 또는 입체적으로 다른 본 발명의 화합물 또는 그것의 염을 의미한다. 이러한 이성질체에는 호변이성질체(tautomer) 등의 구조 이성질체와, 비대칭 탄소 중심을 가지는 R 또는 S 이성체, 기하이성질체(트랜스, 시스) 등의 입체 이성질체, 광학 이성질체(enantiomer)가 모두 포함된다. 이들 모든 이성체 및 그것의 혼합물들 역시 본 발명의 범위에 포함된다.The term "isomer" refers to a compound of the present invention or a salt thereof having the same chemical formula or molecular formula, but structurally or sterically different. Such isomers include structural isomers such as tautomers, R or S isomers having an asymmetric carbon center, stereoisomers such as geometric isomers (trans, cis), and optical isomers. All these isomers and mixtures thereof are also included within the scope of the present invention.
또한, 본 발명은 하기 반응식 1에 나타난 바와 같이,In addition, the present invention, as shown in the following Scheme 1,
화학식 2로 표시되는 화합물과 화학식 3으로 표시되는 화합물을 반응시켜 화학식 1로 표시되는 화합물을 제조하는 단계를 포함하는 상기 화학식 1로 표시되는 화합물의 제조방법을 제공한다.It provides a method for preparing a compound represented by Formula 1, comprising the step of preparing a compound represented by Formula 1 by reacting a compound represented by Formula 2 with a compound represented by Formula 3.
[반응식 1][Scheme 1]
Figure PCTKR2020010657-appb-img-000013
Figure PCTKR2020010657-appb-img-000013
상기 반응식 1에서 R은 상기 화학식 1에서 정의한 바와 같다.In Reaction Scheme 1, R is as defined in Chemical Formula 1.
이하, 상기 반응식 1로 표시되는 제조방법에 대하여 상세히 설명한다.Hereinafter, the manufacturing method represented by Scheme 1 will be described in detail.
본 발명에 따른 상기 반응식 1로 표시되는 제조방법에 있어서, 상기 단계 1은 화학식 2로 표시되는 화합물과 화학식 3으로 표시되는 화합물을 반응시켜, 화학식 1로 표시되는 화합물을 제조하는 단계이다. 구체적으로, 화학식 2로 표시되는 화합물의 할로겐과 화학식 3으로 표시되는 화합물의 1차 아민이 반응하여 화학식 1로 표시되는 화합물이 형성되는 단계이다.In the manufacturing method represented by Scheme 1 according to the present invention, step 1 is a step of preparing a compound represented by Formula 1 by reacting a compound represented by Formula 2 with a compound represented by Formula 3. Specifically, this is a step in which the compound represented by Formula 1 is formed by reacting the halogen of the compound represented by Formula 2 with the primary amine of the compound represented by Formula 3.
상기 반응식 1의 반응은 할로겐과 아민을 결합시켜 아민본드를 형성할 수 있는 조건이라면 특히 한정되지 않는다.The reaction of Scheme 1 is not particularly limited as long as it is a condition capable of forming an amine bond by combining a halogen and an amine.
본 발명에서는 산 조건을 사용하였으며, 상기 산으로는 TFA(Trifluoroacetic acid)을 사용하였으나, 이에 한정되는 것은 아니다. 또한 상기 산의 농도는 1 N일 수 있으나, 이에 한정되는 것은 아니다.In the present invention, an acid condition was used, and trifluoroacetic acid (TFA) was used as the acid, but the present invention is not limited thereto. In addition, the concentration of the acid may be 1 N, but is not limited thereto.
상기 반응식 1에서 사용가능한 용매는 특히 한정되는 것은 아니나, 아이소프로판올, 메탄올, 에탄올, 프로판올 및 부탄올을 포함하는 저급 알코올; 테트라하이드로퓨란(THF); 디옥산; 에틸에테르, 1,2-다이메톡시에탄 등을 포함하는 에테르용매; 디메틸포름아미드(DMF), 디메틸설폭사이드(DMSO), 메틸렌클로라이드, 디클로로에탄, 물, 아세토나젠설포네이트, 톨루엔설포네이트, 클로로벤젠설포네이트, 크실렌설포네이트, 에틸아세테이트, 페닐아세테이트, 페닐프로피오네이트, 페닐부티레이트, 시크레이트, 락테이트, 하이드록시부티레이트, 글리콜레이트, 말레이트, 타트레이트, 메탄설포네이트, 프로판설포네이트, 나프탈렌-1-설포네이트, 나프탈렌-2-설포네이트, 만델레이트, 아세토나이트릴 등이 있으며, 이를 단독 또는 혼합하여 사용할 수 있다. 본 발명에서는 저급 알코올인 n-부탄올를 사용하였으나, 이에 한정되는 것은 아니다.The solvent usable in Reaction Scheme 1 is not particularly limited, but lower alcohols including isopropanol, methanol, ethanol, propanol and butanol; Tetrahydrofuran (THF); Dioxane; Ether solvents including ethyl ether and 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethyl sulfoxide (DMSO), methylene chloride, dichloroethane, water, acetonase sulfonate, toluene sulfonate, chlorobenzene sulfonate, xylene sulfonate, ethyl acetate, phenyl acetate, phenyl propionate , Phenylbutyrate, Cycrate, lactate, hydroxybutyrate, glycolate, malate, tartrate, methanesulfonate, propanesulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, mandelate, acetonite There are reels and the like, and these may be used alone or in combination. In the present invention, n-butanol, which is a lower alcohol, was used, but the present invention is not limited thereto.
또한, 상기 반응식 1의 반응온도는 80℃ 내지 95℃일 수 있으나, 이에 한정되는 것은 아니며, 반응이 진행되는 정도에 따라 적절히 반응온도를 조절할 수 있다.In addition, the reaction temperature in Scheme 1 may be 80° C. to 95° C., but is not limited thereto, and the reaction temperature may be appropriately adjusted according to the degree to which the reaction proceeds.
그리고, 상기 반응식 1의 반응시간은 10분 내지 24시간일 수 있으나, 이에 한정되는 것은 아니며, 반응이 진행되는 정도에 따라 적절히 반응시간을 조절할 수 있다.In addition, the reaction time in Scheme 1 may be 10 minutes to 24 hours, but is not limited thereto, and the reaction time may be appropriately adjusted according to the degree to which the reaction proceeds.
나아가, 본 발명은 상기 화학식 1로 표시되는 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물을 제공한다.Furthermore, the present invention provides a pharmaceutical composition for preventing or treating cancer containing the compound represented by Formula 1, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
이때, 상기 암은 가성점액종, 간내 담도암, 간모세포종, 간암, 갑상선암, 결장암, 고환암, 골수이형성증후군, 교모세포종, 구강암, 구순암, 균상식육종, 급성골수성백혈병, 급성림프구성백혈병, 기저세포암, 난소상피암, 난소생식세포암, 남성유방암, 뇌암, 뇌하수체선종, 다발성골수종, 담낭암, 담도암, 대장암, 만성골수성백혈병, 만성림프구백혈병, 망막모세포종, 맥락막흑색종, 바터팽대부암, 방광암, 복막암, 부갑상선암, 부신암, 비부비동암, 비소세포폐암, 설암, 성상세포종, 소세포폐암, 소아뇌암, 소아림프종, 소아백혈병, 소장암, 수막종, 식도암, 신경교종, 신우암, 신장암, 심장암, 십이지장암, 악성 연부조직 암, 악성골암, 악성림프종, 악성중피종, 악성흑색종, 안암, 외음부암, 요관암, 요도암, 원발부위불명암, 위림프종, 위암, 위유암종, 위장관간질암, 윌름스암, 유방암, 육종, 음경암, 인두암, 임신융모질환, 자궁경부암, 자궁내막암, 자궁육종, 전립선암, 전이성 골암, 전이성뇌암, 종격동암, 직장암, 직장유암종, 질암, 척수암, 청신경초종, 췌장암, 침샘암, 카포시 육종, 파제트병, 편도암, 편평상피세포암, 폐선암, 폐암, 폐편평상피세포암, 피부암, 항문암, 횡문근육종, 후두암, 흉막암, 혈액암 및 흉선암으로 이루어진 군으로부터 선택되는 1종 이상이며, 상기 암은 EGFR에 대하여 돌연변이가 발현된 암일 수 있다.At this time, the cancer is pseudomyxoma, intrahepatic biliary tract cancer, hepatoblastoma, liver cancer, thyroid cancer, colon cancer, testicular cancer, myelodysplastic syndrome, glioblastoma, oral cancer, cleft lip cancer, mycological sarcoma, acute myeloid leukemia, acute lymphocytic leukemia, basal cell Cancer, ovarian epithelial cancer, ovarian germ cell carcinoma, male breast cancer, brain cancer, pituitary adenoma, multiple myeloma, gallbladder cancer, biliary tract cancer, colon cancer, chronic myelogenous leukemia, chronic lymphocytic leukemia, retinoblastoma, choroidal melanoma, barter bulge cancer, bladder cancer, Peritoneal cancer, parathyroid cancer, adrenal cancer, non-sinus cancer, non-small cell lung cancer, tongue cancer, astrocytoma, small cell lung cancer, pediatric brain cancer, pediatric lymphoma, pediatric leukemia, small intestine cancer, meningioma, esophageal cancer, glioma, kidney cancer, kidney cancer, heart Cancer, duodenal cancer, malignant soft tissue cancer, malignant bone cancer, malignant lymphoma, malignant mesothelioma, malignant melanoma, eye cancer, vulvar cancer, ureteral cancer, urethral cancer, primary site unknown cancer, gastric lymphoma, gastric cancer, gastric carcinoma, gastrointestinal interstitial cancer , Wilms cancer, breast cancer, sarcoma, penile cancer, pharyngeal cancer, pregnancy chorionic disease, cervical cancer, endometrial cancer, uterine sarcoma, prostate cancer, metastatic bone cancer, metastatic brain cancer, mediastinal cancer, rectal cancer, rectal carcinoma, vaginal cancer, spinal cord cancer, Auditory schwannoma, pancreatic cancer, salivary gland cancer, Kaposi's sarcoma, Paget's disease, tonsil cancer, squamous cell carcinoma, lung adenocarcinoma, lung cancer, lung squamous cell carcinoma, skin cancer, anal cancer, rhabdomyosarcoma, laryngeal cancer, pleural cancer, blood cancer, and It is one or more selected from the group consisting of thymic cancer, and the cancer may be a cancer in which a mutation is expressed for EGFR.
이때, 상기 EGFR 돌연변이는 EGFR del19, EGFR L858R, EGFR del19/T790M, EGFR L858R/T790M, EGFR del19/T790M/C797S, EGFR L858R/T790M/C797S, EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV 및 EGFR D770_N771insSVD로 이루어진 군으로부터 선택되는 하나 이상일 수 있다.In this case, the EGFR mutations are EGFR del19, EGFR L858R, EGFR del19/T790M, EGFR L858R/T790M, EGFR del19/T790M/C797S, EGFR L858R/T790M/C797S, EGFR D770delinsGY, EGFR H773_V774ins D770ASH, EGFR EGFR and EGFR D770ASH, EGFR and EGFR D770ASH, EGFR, and EGFR del19/T790M_EGFR L858R/T790M It may be one or more selected from the group consisting of.
본 발명의 화학식 1로 표시되는 화합물은 야생형 EGFR에 대하여 상대적으로 약한 EGFR 활성억제효과를 나타내면서, EGFR 돌연변이에 대하여 선택적으로 높은 억제능을 나타내고, 특히, 이중 돌연변이인 EGFR del19/T790M에 대하여 높은 억제능을 나타낸다(실험예 1 및 표 2 참조).The compound represented by Formula 1 of the present invention exhibits a relatively weak inhibitory effect on EGFR activity against wild-type EGFR, while selectively exhibiting a high inhibitory ability against EGFR mutations, and in particular, exhibits a high inhibitory ability against EGFR del19/T790M, a double mutation. (See Experimental Example 1 and Table 2).
본 발명의 화학식 1로 표시되는 화합물은 Ba/F3세포주에서 EGFR 돌연변이에 대하여 선택적으로 높은 세포성장 억제능을 나타내는데, 특히, 삼중 돌연변이인 EGFR del19/T790M/C797S 또는 EGFR L858R/T790M/C797S에 대하여 기존 EGFR TKI(tyrosine kinase inhibitor) 약물인 오시머티닙(Osimertinib)보다 높은 세포성장 억제능을 나타내므로, 오시머티닙보다 우수한 항암효과를 나타냄을 알 수 있다(실험예 2, 표 3 및 표 4 참조).The compound represented by Formula 1 of the present invention exhibits a selectively high cell growth inhibitory ability against EGFR mutations in Ba/F3 cell lines.In particular, the existing EGFR against triple mutations EGFR del19/T790M/C797S or EGFR L858R/T790M/C797S Since it exhibits a higher cell growth inhibitory ability than Osimertinib, which is a tyrosine kinase inhibitor (TKI) drug, it can be seen that it exhibits superior anticancer effects than osimertinib (see Experimental Example 2, Table 3 and Table 4).
본 발명의 실시예 1 화합물은 Ba/F3세포주에서 EGFR 돌연변이에 대하여 선택적으로 높은 세포성장 억제능을 나타내는데, 구체적으로 EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV 또는 EGFR D770_N771insSVD에 대하여 기존 EGFR TKI 약물인 오시머티닙보다 높은 세포성장 억제능을 나타내므로, 오시머티닙보다 우수한 항암효과를 나타냄을 알 수 있다(실험예 3 및 표 5 참조).Example 1 compound of the present invention selectively shows high cell growth inhibitory ability against EGFR mutations in Ba/F3 cell lines, specifically EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV or EGFR D770_N771insSVD Since it exhibits a higher cell growth inhibitory ability than mertinib, it can be seen that it exhibits superior anticancer effects than osimertinib (see Experimental Example 3 and Table 5).
따라서, 본 발명에 따른 화학식 1로 표시되는 화합물은, EGFR 돌연변이에 대하여 높은 억제능을 나타내므로, EGFR del19, EGFR L858R, EGFR del19/T790M, EGFR L858R/T790M, EGFR del19/T790M/C797S, EGFR L858R/T790M/C797S, EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV, EGFR D770_N771insSVD 등의 EGFR 돌연변이가 발현된 암의 치료에 유용하게 사용될 수 있고, 특히, 삼중 돌연변이인 EGFR del19/T790M/C797S 또는 EGFR L858R/T790M/C797S에 대한 억제능이 현저히 우수한바, EGFR del19/T790M/C797S 또는 EGFR L858R/T790M/C797S가 발현된 암의 치료에도 유용하게 사용될 수 있다.Therefore, since the compound represented by Formula 1 according to the present invention exhibits high inhibitory ability against EGFR mutations, EGFR del19, EGFR L858R, EGFR del19/T790M, EGFR L858R/T790M, EGFR del19/T790M/C797S, EGFR L858R/ T790M/C797S, EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV, EGFR D770_N771insSVD can be usefully used in the treatment of cancers expressing EGFR mutations. Since the inhibitory ability against T790M/C797S is remarkably excellent, it can be usefully used in the treatment of cancers expressing EGFR del19/T790M/C797S or EGFR L858R/T790M/C797S.
상기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염은 임상 투여시에 경구 및 비경구의 여러 가지 제형으로 투여될 수 있다. 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제된다. 경구투여를 위한 고형제제에는 정제, 환제, 산제, 과립제, 캡슐제 등 이 포함되며, 이러한 고형제제는 하나 이상의 화합물에 적어도 하나 이상의 부형제 예를 들면, 전분, 탄산칼슘, 수크로오스(sucrose) 또는 락토오스(lactose), 젤라틴 등을 섞어 조제된다. 또한 단순한 부형제 이외에 스테아린산 마그네슘, 탈크 등과 같은 윤활제들도 사용된다. 경구투여를 위한 액상제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데 흔히 사용되는 단순 희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. 비경구투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제가 포함된다. 비수성용제, 현탁용제로는 프로필렌글리콜(propylene glycol), 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테로 등이 사용될 수 있다.The compound represented by Formula 1 or a pharmaceutically acceptable salt thereof may be administered in various oral and parenteral formulations upon clinical administration. In the case of formulation, it is prepared using diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrants, and surfactants that are usually used. Solid preparations for oral administration include tablets, pills, powders, granules, capsules, and the like, and these solid preparations include at least one excipient in one or more compounds, such as starch, calcium carbonate, sucrose, or lactose ( lactose), gelatin, etc. In addition, in addition to simple excipients, lubricants such as magnesium stearate and talc are also used. Liquid preparations for oral administration include suspensions, liquid solutions, emulsions, syrups, etc., but may include various excipients, such as wetting agents, sweeteners, fragrances, and preservatives, in addition to water and liquid paraffin, which are commonly used simple diluents. have. Formulations for parenteral administration include sterilized aqueous solutions, non-aqueous solutions, suspensions, and emulsions. As the non-aqueous solvent and the suspension solvent, propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate may be used.
상기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 유효 성분으로 하는 약학적 조성물은 비경구 투여할 수 있으며, 비경구 투여는 피하주사, 정맥주사, 근육 내 주사 또는 흉부 내 주사를 주입하는 방법에 의한다. A pharmaceutical composition containing the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient can be administered parenterally, and parenteral administration is by subcutaneous injection, intravenous injection, intramuscular injection, or intrathoracic injection. It depends on how to do it.
이때, 비경구 투여용 제형으로 제제화하기 위하여 상기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 안정제 또는 완충제와 함께 물에 혼합하여 용액 또는 현탁액으로 제조하고, 이를 앰플 또는 바이알 단위 투여형으로 제조할 수 있다. 상기 조성물은 멸균되고/되거나 방부제, 안정화제, 수화제 또는 유화 촉진제, 삼투압 조절을 위한 염 및/또는 완충제 등의 보조제, 및 기타 치료적으로 유용한 물질을 함유할 수 있으며, 통상적인 방법인 혼합, 과립화 또는 코팅 방법에 따라 제제화할 수 있다.At this time, in order to formulate a formulation for parenteral administration, the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof is mixed in water together with a stabilizer or buffer to prepare a solution or suspension, and the ampoule or vial unit dosage form It can be manufactured with. The composition may be sterilized and/or contain adjuvants such as preservatives, stabilizers, hydrating agents or emulsification accelerators, salts and/or buffers for controlling osmotic pressure, and other therapeutically useful substances, which are conventional methods of mixing and granulation. It can be formulated according to the method of painting or coating.
경구 투여용 제형으로는 예를 들면 정제, 환제, 경/연질 캅셀제, 액제, 현탁제, 유화제, 시럽제, 과립제, 엘릭시르제, 트로키제 등이 있는데, 이들 제형은 유효성분 이외에 희석제(예: 락토즈, 덱스트로즈, 수크로즈, 만니톨, 솔비톨, 셀룰로즈 및/또는 글리신), 활택제(예: 실리카, 탈크, 스테아르산 및 그의 마그네슘 또는 칼슘염 및/또는 폴리에틸렌 글리콜)를 함유하고 있다. 정제는 마그네슘 알루미늄 실리케이트, 전분 페이스트, 젤라틴, 메틸셀룰로즈, 나트륨 카복시메틸셀룰로즈 및/또는 폴리비닐피롤리딘 등과 같은 결합제를 함유할 수 있으며, 경우에 따라 전분, 한천, 알긴산 또는 그의 나트륨 염 등과 같은 붕해제 또는 비등 혼합물 및/또는 흡수제, 착색제, 향미제, 및 감미제를 함유할 수 있다.Formulations for oral administration include, for example, tablets, pills, hard/soft capsules, solutions, suspensions, emulsifiers, syrups, granules, elixirs, and troches. , Dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine), lubricants (such as silica, talc, stearic acid and magnesium or calcium salts thereof and/or polyethylene glycol). Tablets may contain a binder such as magnesium aluminum silicate, starch paste, gelatin, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidine, and in some cases boron such as starch, agar, alginic acid or sodium salt thereof. It may contain release or boiling mixtures and/or absorbents, colorants, flavoring agents, and sweetening agents.
또한, 본 발명은 상기 화학식 1로 표시되는 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 암의 예방 또는 개선용 건강기능식품을 제공한다.In addition, the present invention provides a health functional food for the prevention or improvement of cancer containing the compound represented by Chemical Formula 1, its isomer, or a pharmaceutically acceptable salt thereof as an active ingredient.
상기 암은 가성점액종, 간내 담도암, 간모세포종, 간암, 갑상선암, 결장암, 고환암, 골수이형성증후군, 교모세포종, 구강암, 구순암, 균상식육종, 급성골수성백혈병, 급성림프구성백혈병, 기저세포암, 난소상피암, 난소생식세포암, 남성유방암, 뇌암, 뇌하수체선종, 다발성골수종, 담낭암, 담도암, 대장암, 만성골수성백혈병, 만성림프구백혈병, 망막모세포종, 맥락막흑색종, 바터팽대부암, 방광암, 복막암, 부갑상선암, 부신암, 비부비동암, 비소세포폐암, 설암, 성상세포종, 소세포폐암, 소아뇌암, 소아림프종, 소아백혈병, 소장암, 수막종, 식도암, 신경교종, 신우암, 신장암, 심장암, 십이지장암, 악성 연부조직 암, 악성골암, 악성림프종, 악성중피종, 악성흑색종, 안암, 외음부암, 요관암, 요도암, 원발부위불명암, 위림프종, 위암, 위유암종, 위장관간질암, 윌름스암, 유방암, 육종, 음경암, 인두암, 임신융모질환, 자궁경부암, 자궁내막암, 자궁육종, 전립선암, 전이성 골암, 전이성뇌암, 종격동암, 직장암, 직장유암종, 질암, 척수암, 청신경초종, 췌장암, 침샘암, 카포시 육종, 파제트병, 편도암, 편평상피세포암, 폐선암, 폐암, 폐편평상피세포암, 피부암, 항문암, 횡문근육종, 후두암, 흉막암, 혈액암 및 흉선암으로 이루어진 군으로부터 선택되는 1종 이상이며, 상기 암은 EGFR에 대하여 돌연변이가 발현된 암일 수 있다.The cancer is pseudomyxoma, intrahepatic biliary tract cancer, hepatoblastoma, liver cancer, thyroid cancer, colon cancer, testicular cancer, myelodysplastic syndrome, glioblastoma, oral cancer, cleft lip cancer, mycosis fungoides, acute myelogenous leukemia, acute lymphocytic leukemia, basal cell cancer, Ovarian epithelial cancer, ovarian germ cell carcinoma, male breast cancer, brain cancer, pituitary adenoma, multiple myeloma, gallbladder cancer, biliary tract cancer, colon cancer, chronic myelogenous leukemia, chronic lymphocytic leukemia, retinoblastoma, choroidal melanoma, barter bulge cancer, bladder cancer, peritoneal cancer , Parathyroid cancer, adrenal cancer, non-sinus cancer, non-small cell lung cancer, tongue cancer, astrocytoma, small cell lung cancer, pediatric brain cancer, pediatric lymphoma, pediatric leukemia, small intestine cancer, meningioma, esophageal cancer, glioma, renal cancer, kidney cancer, heart cancer, Duodenal cancer, malignant soft tissue cancer, malignant bone cancer, malignant lymphoma, malignant mesothelioma, malignant melanoma, eye cancer, vulvar cancer, ureteral cancer, urethral cancer, primary site unknown cancer, gastric lymphoma, gastric cancer, gastric carcinoma, gastrointestinal interstitial cancer, Wilm Cancer, breast cancer, sarcoma, penile cancer, pharyngeal cancer, gestational chorionic disease, cervical cancer, endometrial cancer, uterine sarcoma, prostate cancer, metastatic bone cancer, metastatic brain cancer, mediastinal cancer, rectal cancer, rectal carcinoma, vaginal cancer, spinal cord cancer, auditory nerve sheath , Pancreatic cancer, salivary gland cancer, Kaposi's sarcoma, Paget's disease, tonsil cancer, squamous cell carcinoma, lung adenocarcinoma, lung cancer, lung squamous cell carcinoma, skin cancer, anal cancer, rhabdomyosarcoma, laryngeal cancer, pleural cancer, hematologic cancer and thymic cancer It is one or more selected from the group consisting of, and the cancer may be a cancer in which a mutation is expressed for EGFR.
본 발명에 따른 화학식 1로 표시되는 화합물은, EGFR 돌연변이에 대하여 높은 억제능을 나타냄으로써, 암 특히, EGFR 돌연변이가 발현된 암의 예방 또는 개선용 건강기능식품 조성물로 식품, 음료 등의 건강기능보조 식품에 첨가할 수 있다.The compound represented by Formula 1 according to the present invention is a health functional food composition for the prevention or improvement of cancer, in particular, cancer in which EGFR mutation is expressed, by showing a high inhibitory ability against EGFR mutation. Can be added to.
본 발명에 따른 상기 화학식 1로 표시되는 화합물은 식품에 그대로 첨가하거나 다른 식품 또는 식품 성분과 함께 사용될 수 있고, 통상적인 방법에 따라 적절하게 사용될 수 있다. 유효성분의 혼합량은 그의 사용 목적(예방 또는 개선용)에 따라 적합하게 결정될 수 있다. 일반적으로, 건강식품 중의 상기 화합물의 양은 전체 식품 중량의 0.1 내지 90 중량부로 가할 수 있다. 그러나 건강 및 위생을 목적으로 하거나 또는 건강 조절을 목적으로 하는 장기간 섭취의 경우에는 상기 양은 상기 범위 이하일 수 있으며, 안전성 면에서 아무런 문제가 없기 때문에 유효성분은 상기 범위 이상의 양으로도 사용될 수 있다.The compound represented by Formula 1 according to the present invention may be added to food as it is or may be used together with other foods or food ingredients, and may be appropriately used according to a conventional method. The mixing amount of the active ingredient may be appropriately determined depending on the purpose of use (for prevention or improvement). In general, the amount of the compound in the health food may be added in an amount of 0.1 to 90 parts by weight of the total food weight. However, in the case of long-term intake for the purpose of health and hygiene or for the purpose of health control, the amount may be less than the above range, and there is no problem in terms of safety, so the active ingredient may be used in an amount above the above range.
또한, 본 발명의 건강 기능성 음료 조성물은 지시된 비율로 필수 성분으로서 상기 화합물을 함유하는 외에는 다른 성분에는 특별한 제한이 없으며 통상의 음료와 같이 여러 가지 향미제 또는 천연 탄수화물 등을 추가 성분으로서 함유할 수 있다. 상술한 천연 탄수화물의 예는 모노사카라이드, 예를 들어, 포도당, 과당 등; 디사카라이드, 예를 들어 말토스, 슈크로스 등; 및 폴리사카라이드, 예를 들어 덱스트린, 시클로덱스트린 등과 같은 통상적인 당, 및 자일리톨, 소르비톨, 에리트리톨 등의 당알콜이다. 상술한 것 이외의 향미제로서 천연 향미제(타우마틴, 스테비아 추출물(예를 들어 레바우디오시드 A, 글리시르히진등) 및 합성 향미제(사카린, 아스파르탐 등)를 유리하게 사용할 수 있다. 상기 천연 탄수화물의 비율은 본 발명의 조성물 100 g당 일반적으로 약 1 내지 20 g, 바람직하게는 약 5 내지 12 g이다.In addition, the health functional beverage composition of the present invention is not particularly limited to other ingredients other than containing the compound as an essential ingredient in the indicated ratio, and may contain various flavoring agents or natural carbohydrates, etc. as additional ingredients, as in ordinary beverages. have. Examples of the above-described natural carbohydrates include monosaccharides such as glucose, fructose, and the like; Disaccharides such as maltose, sucrose, and the like; And polysaccharides, for example, common sugars such as dextrin and cyclodextrin, and sugar alcohols such as xylitol, sorbitol, and erythritol. As flavoring agents other than those described above, natural flavoring agents (taumatin, stevia extract (for example, rebaudioside A, glycyrrhizin, etc.)) and synthetic flavoring agents (saccharin, aspartame, etc.) can be advantageously used. The proportion of the natural carbohydrate is generally about 1 to 20 g, preferably about 5 to 12 g per 100 g of the composition of the present invention.
나아가, 상기 외에 본 발명에 따른 화학식 1로 표시되는 화합은 여러 가지 영양제, 비타민, 광물(전해질), 합성 풍미제 및 천연 풍미제 등의 풍미제, 착색제 및 중진제(치즈, 초콜릿 등), 펙트산 및 그의 염, 알긴산 및 그의 염, 유기산, 보호성 콜로이드 증점제, pH 조절제, 안정화제, 방부제, 글리세린, 알코올, 탄산음료에 사용되는 탄산화제 등을 함유할 수 있다. 그 밖에 본 발명의 화학식 1로 표시되는 화합물은 천연 과일 쥬스 및 과일 쥬스 음료 및 야채 음료의 제조를 위한 과육을 함유할 수 있다.Further, in addition to the above, the compound represented by Chemical Formula 1 according to the present invention is a variety of nutrients, vitamins, minerals (electrolytes), flavoring agents such as synthetic flavors and natural flavoring agents, coloring agents and heavy weight agents (cheese, chocolate, etc.), pects. Acids and salts thereof, alginic acid and salts thereof, organic acids, protective colloidal thickeners, pH adjusters, stabilizers, preservatives, glycerin, alcohols, carbonates used in carbonated beverages, and the like. In addition, the compound represented by Chemical Formula 1 of the present invention may contain flesh for the manufacture of natural fruit juice and fruit juice beverages and vegetable beverages.
그리고, 본 발명은 상기 화학식 1로 표시되는 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 EGFR 돌연변이 관련 질환의 예방 또는 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for preventing or treating diseases related to EGFR mutations containing the compound represented by Formula 1, isomers thereof, or pharmaceutically acceptable salts thereof as an active ingredient.
상기 EGFR 돌연변이 관련 질환은 암, 건선, 습진, 아테롬성 동맥 경화증, 구농포성 발진, 피부건조증, 만성 설사, 간 경화증, 심근섬유증 및 만성 신부전으로 이루어진 군으로부터 선택되는 1종 이상일 수 있다.The EGFR mutation-related disease may be at least one selected from the group consisting of cancer, psoriasis, eczema, atherosclerosis, stomatous rash, dry skin, chronic diarrhea, liver cirrhosis, myocardial fibrosis, and chronic renal failure.
또한, 본 발명은 상기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 약학적 조성물 또는 건강기능식품 조성물을 필요한 대상에게 투여하는 단계를 포함하는 암 또는 EGFR 돌연변이 관련 질환의 예방 또는 치료 방법을 제공한다.In addition, the present invention is a cancer or EGFR mutation-related disease comprising the step of administering to a subject in need a pharmaceutical composition or a dietary supplement composition containing the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient It provides a method of preventing or treating.
또한, 본 발명은 암 또는 EGFR 돌연변이 관련 질환의 예방 또는 치료에 있어서의, 상기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 함유하는 약학적 조성물 또는 건강기능식품 조성물의 용도를 제공한다.In addition, the present invention provides a use of a pharmaceutical composition or a health functional food composition containing the compound represented by Formula 1 or a pharmaceutically acceptable salt thereof in the prevention or treatment of cancer or EGFR mutation-related diseases. .
이하, 본 발명을 실시예 및 실험예에 의해 상세히 설명한다.Hereinafter, the present invention will be described in detail by examples and experimental examples.
단, 하기 실시예 및 실험예는 본 발명을 예시하는 것일 뿐, 본 발명의 내용이 하기 실시예 및 실험예에 의해 한정되는 것은 아니다.However, the following Examples and Experimental Examples are merely illustrative of the present invention, and the contents of the present invention are not limited by the following Examples and Experimental Examples.
<제조예 1-1> N-(2-((2,5-디클로로피리미딘-4-일)아미노)페닐)메탄설폰아미드의 제조<Preparation Example 1-1> Preparation of N-(2-((2,5-dichloropyrimidin-4-yl)amino)phenyl)methanesulfonamide
하기 반응식 2로 표시되는 방법을 통해 N-(2-((2,5-디클로로피리미딘-4-일)아미노)페닐)메탄설폰아미드을 제조하였다.N-(2-((2,5-dichloropyrimidin-4-yl)amino)phenyl)methanesulfonamide was prepared through the method represented by Scheme 2 below.
[반응식 2][Scheme 2]
Figure PCTKR2020010657-appb-img-000014
Figure PCTKR2020010657-appb-img-000014
단계 1: 2,5-디클로로-N-(2-니트로페닐)피리미딘-4-아민의 제조Step 1: Preparation of 2,5-dichloro-N-(2-nitrophenyl)pyrimidin-4-amine
2-니트로아닐린(10 g, 72.4 mmol)을 DMF(100 mL)에 용해하여 교반한 용액에 NaH(3.47 g, 144.8 mmol)를 첨가하고, 0℃에서 30분 동안 교반하였다. 이후 2,4,5-트리클로로피리미딘(16.0 g, 86.8 mmol)을 0℃에서 첨가하였다. 상기 반응혼합물을 얼음물(300 mL)로 급냉(quench)시키고, 15분 동안 교반하였다. 침전된 고체를 여과하고, 진공 하 건조하여, 2,5-디클로로-N-(2-니트로페닐)피리미딘-4-아민(17 g, 85% 수율, 황색 고체)을 얻었다.2-nitroaniline (10 g, 72.4 mmol) was dissolved in DMF (100 mL) and NaH (3.47 g, 144.8 mmol) was added to the stirred solution, followed by stirring at 0° C. for 30 minutes. Then 2,4,5-trichloropyrimidine (16.0 g, 86.8 mmol) was added at 0°C. The reaction mixture was quenched with ice water (300 mL) and stirred for 15 minutes. The precipitated solid was filtered and dried under vacuum to give 2,5-dichloro-N-(2-nitrophenyl)pyrimidin-4-amine (17 g, 85% yield, yellow solid).
1H NMR (300 MHz, DMSO- d 6) δ 10.23 (s, 1 H), 8.49 (s, 1 H), 8.10 (dd, J = 8.2, 1.4 Hz, 1 H), 7.91 - 7.71 (m, 2 H), 7.53 - 7.43 (m, 1 H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 10.23 (s, 1 H), 8.49 (s, 1 H), 8.10 (dd, J = 8.2, 1.4 Hz, 1 H), 7.91-7.71 (m, 2H), 7.53-7.43 (m, 1H).
단계 2 : N-(2,5-디클로로피리미딘-4-일)벤젠-1,2-디아민의 제조Step 2: Preparation of N-(2,5-dichloropyrimidin-4-yl)benzene-1,2-diamine
상기 단계 1에서 제조한 2,5-디클로로-N-(2-니트로페닐)피리미딘-4-아민(17 g, 59.6 mmol)을 THF/H 2O(1:1, 300 mL)에 용해하여 교반한 용액에 iron powder(16.6 g, 298 mmol) 및 NH 4Cl(15.9 g, 298.1 mmol)을 상온에서 첨가하였다. 상기 반응혼합물을 5시간 동안 65℃까지 가열하였다. 본 반응은 TLC로 관찰하였다. 가열 후의 반응혼합물을 실온까지 식히고, 셀라이트(Celite)로 여과하고, 에틸 아세테이트(ethyl acetate)로 세척하였다. 유기층은 분리되고, brine으로 세척되었으며, Na 2SO 4로 건조되고, 감압 하에 증발되어 N-(2,5-디클로로피리미딘-4-일)벤젠-1,2-디아민(10 g, 65% 수율, 황색 고체)를 얻었다.Dissolving 2,5-dichloro-N-(2-nitrophenyl)pyrimidin-4-amine (17 g, 59.6 mmol) prepared in step 1 in THF/H 2 O (1:1, 300 mL) Iron powder (16.6 g, 298 mmol) and NH 4 Cl (15.9 g, 298.1 mmol) were added to the stirred solution at room temperature. The reaction mixture was heated to 65° C. for 5 hours. This reaction was observed by TLC. The reaction mixture after heating was cooled to room temperature, filtered through Celite, and washed with ethyl acetate. The organic layer was separated, washed with brine, dried over Na 2 SO 4 , evaporated under reduced pressure and N-(2,5-dichloropyrimidin-4-yl)benzene-1,2-diamine (10 g, 65% Yield, yellow solid).
1H NMR (500 MHz, DMSO- d 6) δ 9.02 (s, 1 H), 8.26 (s, 1 H), 7.06 - 6.95 (m, 2 H), 6.77 (dd, J = 8.1, 1.4 Hz, 1 H), 6.58 (td, J = 7.5, 1.4 Hz, 1 H), 4.98 (s, 2 H); LCMS: 256.8 [M+H +]. 1 H NMR (500 MHz, DMSO- d 6 ) δ 9.02 (s, 1 H), 8.26 (s, 1 H), 7.06-6.95 (m, 2 H), 6.77 (dd, J = 8.1, 1.4 Hz, 1H), 6.58 (td, J = 7.5, 1.4 Hz, 1H), 4.98 (s, 2H); LCMS: 256.8 [M+H + ].
단계 3 : N-(2-((2,5-디클로로피리미딘-4-일)아미노)페닐)메탄설폰아미드의 제조Step 3: Preparation of N-(2-((2,5-dichloropyrimidin-4-yl)amino)phenyl)methanesulfonamide
상기 단계 2에서 제조한 N-(2,5-디클로로피리미딘-4-일)벤젠-1,2-디아민(3 g, 11.5 mmol)을 DCM(50 mL)에 용해하여 교반한 용액에 Et 3N(5 mL, 35.0 mmol)을 상온에서 첨가하였다. 상기 반응혼합물을 0℃까지 식히고, 메실 클로라이드(mesyl chloride)(2.3 mL, 23.0 mmol) 적가방식(dropwise)으로 첨가하였다. 이후 반응혼합물을 상온까지 따뜻하게 하고, 14시간 동안 두었다. 본 반응은 TLC로 관찰하였다. 상기 반응혼합물을 포화된 NaHCO 3 수용액(30 mL)으로 급냉(quench)시키고, DCM(30 mL)로 추출하였다. 유기층은 물, brine으로 세척되고, Na 2SO 4로 건조되고, 감압 하에 증발되었다. 수득된 원액(crude)는 메탄올(50 mL)에 용해되고, K 2CO 3(2.0 g)이 첨가되고, 12시간 동안 교반되었다. 상기 교반액은 감압 하에 증발되었다. 이후 수득된 원액은 H 2O(100 mL)로 희석되고, DCM(100 mL)로 추출되었다. 수용액층은 분리되고, 1N HCl(pH ~ 5 내지 4)로 산성화되고, DCM으로 추출되었다. 유기층이 분리되고, Na 2SO 4로 건조되고, 감압농축되어, N-(2-((2,5-디클로로피리미딘-4-일)아미노)페닐)메탄설폰아미드(2.5 g, 64% 수율, 엷은 적색 고체(pale red solid))를 얻었다.Dissolve N-(2,5-dichloropyrimidin-4-yl)benzene-1,2-diamine (3 g, 11.5 mmol) prepared in step 2 in DCM (50 mL) and add Et 3 to the stirred solution. N (5 mL, 35.0 mmol) was added at room temperature. The reaction mixture was cooled to 0°C, and mesyl chloride (2.3 mL, 23.0 mmol) was added dropwise. After that, the reaction mixture was warmed to room temperature and left for 14 hours. This reaction was observed by TLC. The reaction mixture was quenched with saturated NaHCO 3 aqueous solution (30 mL), and extracted with DCM (30 mL). The organic layer was washed with water, brine, dried over Na 2 SO 4 and evaporated under reduced pressure. The obtained crude was dissolved in methanol (50 mL), K 2 CO 3 (2.0 g) was added, and stirred for 12 hours. The stirred liquid was evaporated under reduced pressure. Subsequently, the obtained stock solution was diluted with H 2 O (100 mL) and extracted with DCM (100 mL). The aqueous layer was separated, acidified with 1N HCl (pH ~ 5-4), and extracted with DCM. The organic layer was separated , dried over Na 2 SO 4 , concentrated under reduced pressure, and N-(2-((2,5-dichloropyrimidin-4-yl)amino)phenyl)methanesulfonamide (2.5 g, 64% yield) , A pale red solid) was obtained.
1H NMR (500 MHz, DMSO- d 6) δ 10.03 (s, 1 H), 8.33 (s, 1 H), 8.24 (dd, J = 8.0, 1.5 Hz, 1 H), 7.19 (dd, J = 8.0, 1.3 Hz, 1 H), 6.83 (td, J = 7.7, 1.6 Hz, 1 H), 6.58 (td, J = 7.6, 1.4 Hz, 1 H), 2.64 (s, 3 H); LCMS: 334.8 [M+H +]. 1 H NMR (500 MHz, DMSO- d 6 ) δ 10.03 (s, 1 H), 8.33 (s, 1 H), 8.24 (dd, J = 8.0, 1.5 Hz, 1 H), 7.19 (dd, J = 8.0, 1.3 Hz, 1H), 6.83 (td, J = 7.7, 1.6 Hz, 1H), 6.58 (td, J = 7.6, 1.4 Hz, 1H), 2.64 (s, 3H); LCMS: 334.8 [M+H + ].
<제조예 1-2> tert-부틸(4-플루오로-2-메톡시-5-니트로페닐)카바메이트의 제조<Production Example 1-2> Preparation of tert-butyl (4-fluoro-2-methoxy-5-nitrophenyl) carbamate
하기 반응식 3로 표시되는 방법을 통해 tert-부틸(4-플루오로-2-메톡시-5-니트로페닐)카바메이트를 제조하였다.Tert-butyl (4-fluoro-2-methoxy-5-nitrophenyl) carbamate was prepared through the method represented by Scheme 3 below.
[반응식 3][Scheme 3]
Figure PCTKR2020010657-appb-img-000015
Figure PCTKR2020010657-appb-img-000015
단계 1 : 4-플루오로-2-메톡시-5-니트로아닐린의 제조Step 1: Preparation of 4-fluoro-2-methoxy-5-nitroaniline
농축 H 2SO 4(50 mL)에 4-플루오로-2-메톡시아닐린(4.7 g, 33.0 mmol)을 portionwise로 첨가하였고, 얼음물 수조 내에서 식혔고, 온도가 15℃ 미만을 유지하도록 했다. 상기 혼합물을 고체가 용해될 때까지 교반하였다. 포타슘 니트레이트(potassium nitrate)(3.36 g, 33.0 mmol)을 portion wise로 첨가하고, 온도가 10℃ 미만을 유지하도록 했다. 이후 혼합물을 밤새 교반하고, 얼음/물에 부었다. 이후 혼합물은 농축 NH 4OH로 염기성화되었다. 고체가 여과되었고, 이를 DCM에 용해하였으며, 물로 세척하고, Na 2SO 4로 건조되며, 감압농축되었다. 생성물인 원액은 플래시 실리카 크로마토그래피(flash silica chromatography)로 정제되고, MeOH 1 내지 10%를 성분으로 하는 DCM으로 용출되었다. 순수 분획물을 증발하여 건조함으로써, 4-플루오로-2-메톡시-5-니트로아닐린(4.5 g, 76% 수율, 황색 고체)을 얻었다.To concentrated H 2 SO 4 (50 mL) 4-fluoro-2-methoxyaniline (4.7 g, 33.0 mmol) was added portionwise, cooled in an ice water bath, and the temperature was kept below 15°C. The mixture was stirred until the solid dissolved. Potassium nitrate (3.36 g, 33.0 mmol) was added portionwise, and the temperature was kept below 10°C. The mixture was then stirred overnight and poured onto ice/water. The mixture was then basified with concentrated NH 4 OH. The solid was filtered, dissolved in DCM, washed with water, dried over Na 2 SO 4 and concentrated under reduced pressure. The product stock solution was purified by flash silica chromatography, and was eluted with DCM containing 1 to 10% MeOH. The pure fractions were evaporated and dried to obtain 4-fluoro-2-methoxy-5-nitroaniline (4.5 g, 76% yield, yellow solid).
1H NMR (400 MHz, DMSO- d 6) δ 7.38 - 7.29 (d, J = 7.8 Hz, 1 H), 7.11 - 6.97 (d, J = 13.4 Hz, 1 H), 5.24 (s, 2 H), 3.91 (s, 3 H); LCMS: 187.2 [M+H +]. 1 H NMR (400 MHz, DMSO- d 6 ) δ 7.38-7.29 (d, J = 7.8 Hz, 1 H), 7.11-6.97 (d, J = 13.4 Hz, 1 H), 5.24 (s, 2 H) , 3.91 (s, 3H); LCMS: 187.2 [M+H + ].
단계 2 : tert-부틸(4-플루오로-2-메톡시-5-니트로페닐)카바메이트의 제조Step 2: Preparation of tert-butyl (4-fluoro-2-methoxy-5-nitrophenyl) carbamate
상기 단계 1에서 제조한 4-플루오로-2-메톡시-5-니트로아닐린(4.5 g, 24.1 mmol) 및 DMAP(300 mg, 2.41 mmol)을 DCM에 용해시켜 교반한 용액에 (Boc) 2O(5.8 g, 26.2 mmol)을 0℃에서 첨가하였다. 상기 반응혼합물은 상온으로 천천히 따뜻하게 하고, 12시간 동안 두었다. 이후 반응혼합물을 DCM으로 희석하고, 포화 NaHCO 3 수용액으로 급냉하였다. 분리된 유기층은 물 및 brine으로 세척되고, Na 2SO 4로 건조되고, 감압농축되었다. 수득된 원액은 용리제(eluent)가 헥세인(hexanes)/에틸 아세테이트(ethyl acetate)(7:3)인 컬럼 크로마토그래피를 사용하여 정제되었고, tert-부틸(4-플루오로-2-메톡시-5-니트로페닐)카바메이트(4.0 g, 58% 수율, 황색 고체)을 얻었다.4-fluoro-2-methoxy-5-nitroaniline (4.5 g, 24.1 mmol) and DMAP (300 mg, 2.41 mmol) prepared in step 1 were dissolved in DCM and added to the stirred solution (Boc) 2 O (5.8 g, 26.2 mmol) was added at 0°C. The reaction mixture was slowly warmed to room temperature and left for 12 hours. Thereafter, the reaction mixture was diluted with DCM and quenched with saturated NaHCO 3 aqueous solution. The separated organic layer was washed with water and brine, dried over Na 2 SO 4 , and concentrated under reduced pressure. The obtained stock solution was purified using column chromatography in which the eluent was hexanes/ethyl acetate (7:3), and tert-butyl (4-fluoro-2-methoxy -5-nitrophenyl)carbamate (4.0 g, 58% yield, yellow solid) was obtained.
1H NMR (400 MHz, Chloroform- d) δ 8.91 (d, J = 8.0 Hz, 1 H), 6.99 (s, 1 H), 6.78 - 6.68 (d, J = 12.1 Hz, 1 H), 4.00 (s, 3 H), 1.56 (s, 9 H); LCMS: 287.2 [M+H +]. 1 H NMR (400 MHz, Chloroform- d ) δ 8.91 (d, J = 8.0 Hz, 1 H), 6.99 (s, 1 H), 6.78-6.68 (d, J = 12.1 Hz, 1 H), 4.00 ( s, 3H), 1.56 (s, 9H); LCMS: 287.2 [M+H + ].
<제조예 1-3> N-(5-아미노-2((2-(디메틸아미노)에틸(메틸)아미노)-4-메톡시페닐)아크릴아미드의 제조<Production Example 1-3> Preparation of N-(5-amino-2((2-(dimethylamino)ethyl(methyl)amino)-4-methoxyphenyl)acrylamide
하기 반응식 4로 표시되는 방법을 통해 N-(5-아미노-2((2-(디메틸아미노)에틸(메틸)아미노)-4-메톡시페닐)아크릴아미드를 제조하였다.N-(5-amino-2((2-(dimethylamino)ethyl(methyl)amino)-4-methoxyphenyl)acrylamide was prepared through the method represented by Scheme 4 below.
[반응식 4][Scheme 4]
Figure PCTKR2020010657-appb-img-000016
Figure PCTKR2020010657-appb-img-000016
단계 1 : tert-부틸 (4-((2-(디메틸아미노)에틸)(메틸)아미노)-2-메톡시-5-니트로페닐)카바메이트의 제조Step 1: Preparation of tert-butyl (4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)carbamate
상기 제조예 1-2에서 제조한 tert-부틸(4-플루오로-2-메톡시-5-니트로페닐)카바메이트(300 mg, 1.04 mmol)를 DMF(5 mL)에 용해시키고 교반한 용액에 N 1,N 1,N 2-트리메틸에테인-1,2-디아민(106 mg, 1.04 mmol) 및 세슘 카보네이트(cesium carbonate)(183 mg, 1.56 mmol)을 상온에서 첨가하였다. 상기 반응혼합물을 70℃에서 밤새 교반하였다. 이후 반응혼합물을 상온으로 식히고, 얼음물로 급냉하였다. 침전된 고체를 여과하고, 진공 하 건조하여, tert-부틸 (4-((2-(디메틸아미노)에틸)(메틸)아미노)-2-메톡시-5-니트로페닐)카바메이트(250 mg, 65% 수율, 황색 고체)를 얻었다.The tert-butyl (4-fluoro-2-methoxy-5-nitrophenyl) carbamate (300 mg, 1.04 mmol) prepared in Preparation Example 1-2 was dissolved in DMF (5 mL) and added to the stirred solution. N 1 ,N 1 ,N 2 -Trimethylethane-1,2-diamine (106 mg, 1.04 mmol) and cesium carbonate (183 mg, 1.56 mmol) were added at room temperature. The reaction mixture was stirred at 70° C. overnight. Thereafter, the reaction mixture was cooled to room temperature and quenched with ice water. The precipitated solid was filtered and dried under vacuum, tert-butyl (4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)carbamate (250 mg, 65% yield, yellow solid).
1H NMR (400 MHz, DMSO- d 6) δ 8.16 (s, 1 H), 8.11 (s, 1 H), 6.74 (s, 1 H), 3.90 (s, 3 H), 3.24 - 3.19 (t, J = 6.8 Hz, 2 H), 2.80 (s, 3 H), 2.46 - 2.40 (t, J = 6.8 Hz, 2 H), 2.13 (s, 6 H), 1.45 (s, 9 H); LCMS: 369.0 [M+H +]. 1 H NMR (400 MHz, DMSO- d 6 ) δ 8.16 (s, 1 H), 8.11 (s, 1 H), 6.74 (s, 1 H), 3.90 (s, 3 H), 3.24-3.19 (t , J = 6.8 Hz, 2H), 2.80 (s, 3H), 2.46-2.40 (t, J = 6.8 Hz, 2H), 2.13 (s, 6H), 1.45 (s, 9H); LCMS: 369.0 [M+H + ].
단계 2 : tert-부틸(5-아미노-4-((2-(디메틸아미노)에틸)(메틸)아미노)-2-메톡시페닐)카바메이트의 제조Step 2: Preparation of tert-butyl(5-amino-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)carbamate
상기 단계 1에서 제조한 tert-부틸 (4-((2-(디메틸아미노)에틸)(메틸)아미노)-2-메톡시-5-니트로페닐)카바메이트(250 mg, 0.64 mmol)를 메탄올(15 mL)에 용해하여 교반한 용액에 아르곤(Ar, Argon) 대기 하에서 10% Pd/C(6.8 mg, 0.06 mmol)을 첨가했다. 상기 반응혼합물을 상온에서 4시간 동안 1 atm 수소 가스 압력 하에서 교반했고, 감압농축하여, tert-부틸(5-아미노-4-((2-(디메틸아미노)에틸)(메틸)아미노)-2-메톡시페닐)카바메이트(210 mg, 91% 수율, 갈색 고체)를 얻었다.Tert-butyl (4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)carbamate (250 mg, 0.64 mmol) prepared in step 1 was added to methanol ( 15 mL), 10% Pd/C (6.8 mg, 0.06 mmol) was added to the stirred solution under an argon (Ar, Argon) atmosphere. The reaction mixture was stirred at room temperature for 4 hours under 1 atm hydrogen gas pressure, and concentrated under reduced pressure, tert-butyl(5-amino-4-((2-(dimethylamino)ethyl)(methyl)amino)-2- Methoxyphenyl) carbamate (210 mg, 91% yield, brown solid) was obtained.
LCMS: 339.2 [M+H +]LCMS: 339.2 [M+H + ]
단계 3 : tert-부틸 (5-아크릴아미도-4-((2-(디메틸아미노)에틸)(메틸)아미노)-2-메톡시페닐)카바메이트의 제조Step 3: Preparation of tert-butyl (5-acrylamido-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)carbamate
상기 단계 2에서 제조한 tert-부틸(5-아미노-4-((2-(디메틸아미노)에틸)(메틸)아미노)-2-메톡시페닐)카바메이트(200 mg, 0.59 mmol) 및 트리에틸아민(0.16 mL, 1.18 mmol)을 DCM에 용해하여 교반한 용액에 아크릴로일 클로라이드(acryloyl chloride)(0.047 mL, 0.59 mmol)을 -30℃에서 첨가하였다. 상기 반응혼합물은 상온으로 천천히 따뜻하게 하였고, 3시간 동안 두었다. 이후 반응혼합물은 DCM으로 희석되고, 포화 NaHCO 3 수용액으로 급냉되었다. 유기층이 분리되고, 물 및 brine으로 세척되고, Na 2SO 4로 건조되며, 감압농축되었다. 수득된 원액은 용리제가 DCM/MeOH(9.5:0.5)인 컬럼 크로마토그래피를 사용하여 정제되었고, tert-부틸 (5-아크릴아미도-4-((2-(디메틸아미노)에틸)(메틸)아미노)-2-메톡시페닐)카바메이트(150 mg, 65% 수율, 황백색 고체(off white solid))를 얻었다. Tert-butyl (5-amino-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)carbamate (200 mg, 0.59 mmol) and triethyl prepared in step 2 Amine (0.16 mL, 1.18 mmol) was dissolved in DCM, and acryloyl chloride (0.047 mL, 0.59 mmol) was added to the stirred solution at -30°C. The reaction mixture was slowly warmed to room temperature and left for 3 hours. Thereafter, the reaction mixture was diluted with DCM and quenched with saturated NaHCO 3 aqueous solution. The organic layer was separated, washed with water and brine, dried over Na 2 SO 4 , and concentrated under reduced pressure. The obtained stock solution was purified using column chromatography in which the eluent was DCM/MeOH (9.5:0.5), and tert-butyl (5-acrylamido-4-((2-(dimethylamino)ethyl)(methyl)amino )-2-methoxyphenyl)carbamate (150 mg, 65% yield, off white solid) was obtained.
1H NMR (400 MHz, DMSO- d 6) δ 10.03 (s, 1 H), 8.47 (s, 1 H), 7.83 (s, 1 H), 6.93 (s, 1 H), 6.49 - 6.34 (m, 1 H), 6.27 - 6.18 (dd, J = 17.0, 2.2 Hz, 1 H), 5.78 - 5.69 (dd, J = 10.0, 2.2 Hz, 1 H), 3.79 (s, 3 H), 2.91 - 2.82 (t, J = 5.9 Hz, 2 H), 2.66 (s, 3 H), 2.33 (bs, 2 H), 2.22 (s, 6 H), 1.45 (s, 9 H); LCMS: 393.0 [M+H +]. 1 H NMR (400 MHz, DMSO- d 6 ) δ 10.03 (s, 1 H), 8.47 (s, 1 H), 7.83 (s, 1 H), 6.93 (s, 1 H), 6.49-6.34 (m , 1 H), 6.27-6.18 (dd, J = 17.0, 2.2 Hz, 1 H), 5.78-5.69 (dd, J = 10.0, 2.2 Hz, 1 H), 3.79 (s, 3 H), 2.91-2.82 (t, J = 5.9 Hz, 2H), 2.66 (s, 3H), 2.33 (bs, 2H), 2.22 (s, 6H), 1.45 (s, 9H); LCMS: 393.0 [M+H + ].
단계 4 : N-(5-아미노-2((2-(디메틸아미노)에틸(메틸)아미노)-4-메톡시페닐)아크릴아미드의 제조Step 4: Preparation of N-(5-amino-2((2-(dimethylamino)ethyl(methyl)amino)-4-methoxyphenyl)acrylamide
상기 단계 3에서 제조한 tert-부틸 (5-아크릴아미도-4-((2-(디메틸아미노)에틸)(메틸)아미노)-2-메톡시페닐)카바메이트(150.0 mg, 0.38 mmol)을 DCM에 용해시켜 교반한 용액에 DCM(1 mL)에 용해된 25% TFA를 상온에서 첨가하였다. 상기 혼합물은 12시간 동안 상온에서 교반하였다. 반응혼합물은 TLC로 관찰되었다. 이후, 원액상태인 N-(5-아미노-2((2-(디메틸아미노)에틸(메틸)아미노)-4-메톡시페닐)아크릴아미드는 감압농축되었고, 다른 정제는 없었다.Tert-butyl (5-acrylamido-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)carbamate (150.0 mg, 0.38 mmol) prepared in step 3 25% TFA dissolved in DCM (1 mL) was added to the stirred solution in DCM at room temperature. The mixture was stirred at room temperature for 12 hours. The reaction mixture was observed by TLC. Thereafter, N-(5-amino-2((2-(dimethylamino)ethyl(methyl)amino)-4-methoxyphenyl)acrylamide as a stock solution was concentrated under reduced pressure, and there was no other purification.
LCMS: 293.2 [M+H +].LCMS: 293.2 [M+H + ].
<제조예 2-1> tert-부틸 (5-아크릴아미도-4-(4-(디메틸아미노)피페리딘-1-일)-2-메톡시페닐)카바메이트의 제조<Preparation Example 2-1> Preparation of tert-butyl (5-acrylamido-4-(4-(dimethylamino) piperidin-1-yl)-2-methoxyphenyl) carbamate
[tert-부틸 (5-아크릴아미도-4-(4-(디메틸아미노)피페리딘-1-일)-2-메톡시페닐)카바메이트][tert-butyl (5-acrylamido-4-(4-(dimethylamino) piperidin-1-yl)-2-methoxyphenyl) carbamate]
Figure PCTKR2020010657-appb-img-000017
Figure PCTKR2020010657-appb-img-000017
N 1,N 1,N 2-트리메틸에테인-1,2-디아민 대신 N,N-디메틸피페리딘-4-아민을 사용한 것을 제외하고는, 상기 제조예 1-3의 단계 1 내지 3에 따라 tert-부틸 (5-아크릴아미도-4-(4-(디메틸아미노)피페리딘-1-일)-2-메톡시페닐)카바메이트(78% 수율)를 제조하였다.According to steps 1 to 3 of Preparation Example 1-3, except that N,N-dimethylpiperidin-4-amine was used instead of N 1 ,N 1 ,N 2 -trimethylethane-1,2-diamine tert-butyl (5-acrylamido-4-(4-(dimethylamino)piperidin-1-yl)-2-methoxyphenyl)carbamate (78% yield) was prepared.
* 1H NMR (400 MHz, DMSO- d 6) δ 8.95 (s, 1 H), 8.16 (s, 1 H), 7.83 (s, 1 H), 6.76 (s, 1 H), 6.69 - 6.57 (m, 1 H), 6.26 - 6.17 (dd, J = 16.9, 2.0 Hz, 1 H), 5.78 - 5.67 (m, 1 H), 3.79 (s, 3 H), 3.05 - 2.96 (m, 2 H), 2.69 - 2.57 (m, 2 H), 2.28 (s, 7 H), 1.88 - 1.79 (m, 2 H), 1.76 - 1.61 (m, 2 H), 1.45 (s, 9 H); LCMS: 419.0 [M+H +].* 1 H NMR (400 MHz, DMSO- d 6 ) δ 8.95 (s, 1 H), 8.16 (s, 1 H), 7.83 (s, 1 H), 6.76 (s, 1 H), 6.69-6.57 ( m, 1 H), 6.26-6.17 (dd, J = 16.9, 2.0 Hz, 1 H), 5.78-5.67 (m, 1 H), 3.79 (s, 3 H), 3.05-2.96 (m, 2 H) , 2.69-2.57 (m, 2H), 2.28 (s, 7H), 1.88-1.79 (m, 2H), 1.76-1.61 (m, 2H), 1.45 (s, 9H); LCMS: 419.0 [M+H + ].
<제조예 2-2> N-(5-아미노-2-(4-(디메틸아미노)피페리딘-1-일)-4-메톡시페닐)아크릴아미드의 제조<Production Example 2-2> Preparation of N-(5-amino-2-(4-(dimethylamino)piperidin-1-yl)-4-methoxyphenyl)acrylamide
[N-(5-아미노-2-(4-(디메틸아미노)피페리딘-1-일)-4-메톡시페닐)아크릴아미드][N-(5-amino-2-(4-(dimethylamino)piperidin-1-yl)-4-methoxyphenyl)acrylamide]
Figure PCTKR2020010657-appb-img-000018
Figure PCTKR2020010657-appb-img-000018
전구체로 상기 제조예 2-1에서 제조한 tert-부틸 (5-아크릴아미도-4-(4-(디메틸아미노)피페리딘-1-일)-2-메톡시페닐)카바메이트를 사용한 것을 제외하고, 상기 제조예 1-3의 단계 4에 따라 N-(5-아미노-2-(4-(디메틸아미노)피페리딘-1-일)-4-메톡시페닐)아크릴아미드를 제조하였다.Using tert-butyl (5-acrylamido-4-(4-(dimethylamino) piperidin-1-yl)-2-methoxyphenyl) carbamate prepared in Preparation Example 2-1 as a precursor Except, N-(5-amino-2-(4-(dimethylamino)piperidin-1-yl)-4-methoxyphenyl)acrylamide was prepared according to Step 4 of Preparation Example 1-3. .
LCMS: 319.0 [M+H +].LCMS: 319.0 [M+H + ].
<제조예 3-1> tert-부틸 (5-아크릴아미도-2-메톡시-4-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)카바메이트의 제조<Production Example 3-1> tert-butyl (5-acrylamido-2-methoxy-4-(4-(4-methylpiperazin-1-yl) piperidin-1-yl) phenyl) carbamate Manufacture of
[tert-부틸 (5-아크릴아미도-2-메톡시-4-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)카바메이트][tert-butyl (5-acrylamido-2-methoxy-4-(4-(4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)carbamate]
Figure PCTKR2020010657-appb-img-000019
Figure PCTKR2020010657-appb-img-000019
N 1,N 1,N 2-트리메틸에테인-1,2-디아민 대신 1-메틸-4-(피페리딘-4-일)피페라진을 사용한 것을 제외하고는, 상기 제조예 1-3의 단계 1 내지 3에 따라 tert-부틸 (5-아크릴아미도-2-메톡시-4-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)카바메이트(65% 수율)를 제조하였다.The steps of Preparation Example 1-3, except that 1-methyl-4-(piperidin-4-yl)piperazine was used instead of N 1 ,N 1 ,N 2 -trimethylethane-1,2-diamine Tert-butyl (5-acrylamido-2-methoxy-4-(4-(4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)carbamate (65 % Yield).
1H NMR (400 MHz, DMSO- d 6) δ 8.94 (s, 1H), 8.16 (s, 1 H), 7.85 (s, 1 H), 6.75 (s, 1 H), 6.71 - 6.57 (m, 1 H), 6.27 - 6.16 (dd, J = 16.9, 2.1 Hz, 1 H), 5.75 - 5.67 (dd, J = 10.3, 2.0 Hz, 1 H), 3.79 (s, 3 H), 3.06 - 2.95 (d, J = 11.4 Hz, 2 H), 2.91 - 2.58 (m, 10 H), 1.93 - 1.78 (bs, 2 H), 1.77 - 1.62 (bs, 2 H), 1.45 (s, 9 H); LCMS: 474.2 [M+H +]. 1 H NMR (400 MHz, DMSO- d 6 ) δ 8.94 (s, 1H), 8.16 (s, 1 H), 7.85 (s, 1 H), 6.75 (s, 1 H), 6.71-6.57 (m, 1 H), 6.27-6.16 (dd, J = 16.9, 2.1 Hz, 1 H), 5.75-5.67 (dd, J = 10.3, 2.0 Hz, 1 H), 3.79 (s, 3 H), 3.06-2.95 ( d, J = 11.4 Hz, 2H), 2.91-2.58 (m, 10H), 1.93-1.78 (bs, 2H), 1.77-1.62 (bs, 2H), 1.45 (s, 9H); LCMS: 474.2 [M+H + ].
<제조예 3-2> N-(5-아미노-4-메톡시-2-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)아크릴아미드의 제조<Production Example 3-2> Preparation of N-(5-amino-4-methoxy-2-(4-(4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)acrylamide
[N-(5-아미노-4-메톡시-2-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)아크릴아미드][N-(5-amino-4-methoxy-2-(4-(4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)acrylamide]
Figure PCTKR2020010657-appb-img-000020
Figure PCTKR2020010657-appb-img-000020
전구체로 상기 제조예 3-1에서 제조한 tert-부틸 (5-아크릴아미도-2-메톡시-4-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)카바메이트를 사용한 것을 제외하고, 상기 제조예 1-3의 단계 4에 따라 N-(5-아미노-4-메톡시-2-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)아크릴아미드를 제조하여, 정제 없이 이후 단계에 사용하였다.Tert-butyl prepared in Preparation Example 3-1 as a precursor (5-acrylamido-2-methoxy-4-(4-(4-methylpiperazin-1-yl)piperidin-1-yl) N-(5-amino-4-methoxy-2-(4-(4-methylpiperazin-1-yl)pi according to Step 4 of Preparation Example 1-3 above, except that phenyl) carbamate was used. Peridine-1-yl)phenyl)acrylamide was prepared and used in the later step without purification.
<제조예 4-1> tert-부틸 (5-아크릴아미도-2-메톡시-4-모폴리노페닐)카바메이트의 제조<Production Example 4-1> Preparation of tert-butyl (5-acrylamido-2-methoxy-4-morpholinophenyl) carbamate
[tert-부틸 (5-아크릴아미도-2-메톡시-4-모폴리노페닐)카바메이트][tert-butyl (5-acrylamido-2-methoxy-4-morpholinophenyl) carbamate]
Figure PCTKR2020010657-appb-img-000021
Figure PCTKR2020010657-appb-img-000021
N 1,N 1,N 2-트리메틸에테인-1,2-디아민 대신 모폴린을 사용한 것을 제외하고는, 상기 제조예 1-3의 단계 1 내지 3에 따라 tert-부틸 (5-아크릴아미도-2-메톡시-4-모폴리노페닐)카바메이트(77% 수율)를 제조하였다.Tert-butyl (5-acrylamido-) according to steps 1 to 3 of Preparation Example 1-3, except that morpholine was used instead of N 1 ,N 1 ,N 2 -trimethylethane-1,2-diamine 2-methoxy-4-morpholinophenyl)carbamate (77% yield) was prepared.
1H NMR (400 MHz, Chloroform- d) δ 6.99 (s, 1 H), 6.73 (s, 1 H), 6.51 - 6.42 (d, J = 16.5 Hz, 1 H), 6.27 - 6.17 (m, 1 H), 5.82 - 5.74 (m, 1 H), 3.99 (bs, 4 H), 3.89 (s, 3 H), 2.12 - 2.93(bs, 4 H), 1.54 (s, 9 H); LCMS: 378.0 [M+H +]. 1 H NMR (400 MHz, Chloroform- d ) δ 6.99 (s, 1 H), 6.73 (s, 1 H), 6.51-6.42 (d, J = 16.5 Hz, 1 H), 6.27-6.17 (m, 1 H), 5.82-5.74 (m, 1H), 3.99 (bs, 4H), 3.89 (s, 3H), 2.12-2.93 (bs, 4H), 1.54 (s, 9H); LCMS: 378.0 [M+H + ].
<제조예 4-2> N-(5-아미노-4-메톡시-2-모폴리노페닐)아크릴아미드의 제조<Production Example 4-2> Preparation of N-(5-amino-4-methoxy-2-morpholinophenyl)acrylamide
[N-(5-아미노-4-메톡시-2-모폴리노페닐)아크릴아미드][N-(5-amino-4-methoxy-2-morpholinophenyl)acrylamide]
Figure PCTKR2020010657-appb-img-000022
Figure PCTKR2020010657-appb-img-000022
전구체로 상기 제조예 4-1에서 제조한 tert-부틸 (5-아크릴아미도-2-메톡시-4-모폴리노페닐)카바메이트를 사용한 것을 제외하고, 상기 제조예 1-3의 단계 4에 따라 N-(5-아미노-4-메톡시-2-모폴리노페닐)아크릴아미드를 제조하였다.Step 4 of Preparation Example 1-3, except that tert-butyl (5-acrylamido-2-methoxy-4-morpholinophenyl) carbamate prepared in Preparation Example 4-1 was used as a precursor. According to the following, N-(5-amino-4-methoxy-2-morpholinophenyl)acrylamide was prepared.
LCMS: 278.0.2 [M+H +].LCMS: 278.0.2 [M+H + ].
<제조예 5-1> tert-부틸 (5-아크릴아미도-2-메톡시-4-(4-메틸피페라진-1-일)페닐)카바메이트의 제조<Preparation Example 5-1> Preparation of tert-butyl (5-acrylamido-2-methoxy-4-(4-methylpiperazin-1-yl)phenyl)carbamate
[tert-부틸 (5-아크릴아미도-2-메톡시-4-(4-메틸피페라진-1-일)페닐)카바메이트][tert-butyl (5-acrylamido-2-methoxy-4-(4-methylpiperazin-1-yl)phenyl)carbamate]
Figure PCTKR2020010657-appb-img-000023
Figure PCTKR2020010657-appb-img-000023
N 1,N 1,N 2-트리메틸에테인-1,2-디아민 대신 1-메틸피페라진을 사용한 것을 제외하고는, 상기 제조예 1-3의 단계 1 내지 3에 따라 tert-부틸 (5-아크릴아미도-2-메톡시-4-(4-메틸피페라진-1-일)페닐)카바메이트(77% 수율)를 제조하였다.Tert-butyl (5-acrylic) according to steps 1 to 3 of Preparation Example 1-3, except that 1-methylpiperazine was used instead of N 1 ,N 1 ,N 2 -trimethylethane-1,2-diamine. Amido-2-methoxy-4-(4-methylpiperazin-1-yl)phenyl)carbamate (77% yield) was prepared.
1H NMR (400 MHz, Chloroform- d) δ 9.09 (s, 1 H), 8.31 (s, 1 H), 6.94 (s, 1 H), 6.74 (s, 1 H), 6.49 - 6.35 (d, J = 16.8 Hz, 1 H), 6.33 - 6.22 (dd, J = 17.4, 10.0 Hz, 1 H), 5.80 - 5.68 (d, J = 10.2 Hz, 1 H), 3.85 (s, 3 H), 3.03 (bs, 4 H), 2.96 - 2.77 (m, 4 H), 2.55 (s, 3 H), 1.54 (s, 9 H); LCMS: 391.0 [M+H +]. 1 H NMR (400 MHz, Chloroform- d ) δ 9.09 (s, 1 H), 8.31 (s, 1 H), 6.94 (s, 1 H), 6.74 (s, 1 H), 6.49-6.35 (d, J = 16.8 Hz, 1 H), 6.33-6.22 (dd, J = 17.4, 10.0 Hz, 1 H), 5.80-5.68 (d, J = 10.2 Hz, 1 H), 3.85 (s, 3 H), 3.03 (bs, 4H), 2.96-2.77 (m, 4H), 2.55 (s, 3H), 1.54 (s, 9H); LCMS: 391.0 [M+H + ].
<제조예 5-2> N-(5-아미노-4-메톡시-2-(4-메틸피페라진-1-일)페닐)아크릴아미드의 제조<Production Example 5-2> Preparation of N-(5-amino-4-methoxy-2-(4-methylpiperazin-1-yl)phenyl)acrylamide
[N-(5-아미노-4-메톡시-2-(4-메틸피페라진-1-일)페닐)아크릴아미드][N-(5-amino-4-methoxy-2-(4-methylpiperazin-1-yl)phenyl)acrylamide]
Figure PCTKR2020010657-appb-img-000024
Figure PCTKR2020010657-appb-img-000024
전구체로 상기 제조예 5-1에서 제조한 tert-부틸 (5-아크릴아미도-2-메톡시-4-(4-메틸피페라진-1-일)페닐)카바메이트를 사용한 것을 제외하고, 상기 제조예 1-3의 단계 4에 따라 N-(5-아미노-4-메톡시-2-(4-메틸피페라진-1-일)페닐)아크릴아미드를 제조하였다.Except for using tert-butyl (5-acrylamido-2-methoxy-4- (4-methylpiperazin-1-yl) phenyl) carbamate prepared in Preparation Example 5-1 as a precursor, the above According to Step 4 of Preparation Example 1-3, N-(5-amino-4-methoxy-2-(4-methylpiperazin-1-yl)phenyl)acrylamide was prepared.
LCMS: 291.2 [M+H +].LCMS: 291.2 [M+H + ].
<제조예 6-1> tert-부틸 (5-아크릴아미도-4-(2-(디메틸아미노)에톡시)-2-메톡시페닐)카바메이트의 제조<Preparation Example 6-1> Preparation of tert-butyl (5-acrylamido-4-(2-(dimethylamino)ethoxy)-2-methoxyphenyl)carbamate
[tert-부틸 (5-아크릴아미도-4-(2-(디메틸아미노)에톡시)-2-메톡시페닐)카바메이트][tert-butyl (5-acrylamido-4-(2-(dimethylamino)ethoxy)-2-methoxyphenyl)carbamate]
Figure PCTKR2020010657-appb-img-000025
Figure PCTKR2020010657-appb-img-000025
N 1,N 1,N 2-트리메틸에테인-1,2-디아민 대신 2-(디메틸아미노)에탄-1-올을 사용한 것을 제외하고는, 상기 제조예 1-3의 단계 1 내지 3에 따라 tert-부틸 (5-아크릴아미도-4-(2-(디메틸아미노)에톡시)-2-메톡시페닐)카바메이트(71% 수율)를 제조하였다.Tert according to steps 1 to 3 of Preparation Example 1-3, except that 2-(dimethylamino)ethan-1-ol was used instead of N 1 ,N 1 ,N 2 -trimethylethane-1,2-diamine -Butyl (5-acrylamido-4-(2-(dimethylamino)ethoxy)-2-methoxyphenyl)carbamate (71% yield) was prepared.
1H NMR (300 MHz, Chloroform- d) δ 9.75 (s, 1 H), 9.00 (s, 1 H), 6.83 (s, 1 H), 6.57 (s, 1 H), 6.48 - 6.39 (dd, J = 16.9, 1.9 Hz, 1 H), 6.34 - 6.21 (dd, J = 16.9, 10.0 Hz, 1 H), 5.71 - 5.65 (dd, J = 10.0, 1.9 Hz, 1 H), 4.14 - 4.07 (m, 2 H), 3.83 (s, 3 H), 2.65 - 2.55 (m, 3 H), 2.37 (s, 6 H), 1.53 (s, 12 H); LCMS: 380.2 [M+H +]. 1 H NMR (300 MHz, Chloroform- d ) δ 9.75 (s, 1 H), 9.00 (s, 1 H), 6.83 (s, 1 H), 6.57 (s, 1 H), 6.48-6.39 (dd, J = 16.9, 1.9 Hz, 1 H), 6.34-6.21 (dd, J = 16.9, 10.0 Hz, 1 H), 5.71-5.65 (dd, J = 10.0, 1.9 Hz, 1 H), 4.14-4.07 (m , 2H), 3.83 (s, 3H), 2.65-2.55 (m, 3H), 2.37 (s, 6H), 1.53 (s, 12H); LCMS: 380.2 [M+H + ].
<제조예 6-2> N-(5-아미노-2-(2-(디메틸아미노)에톡시)-4-메톡시페닐)아크릴아미드의 제조<Production Example 6-2> Preparation of N-(5-amino-2-(2-(dimethylamino)ethoxy)-4-methoxyphenyl)acrylamide
[N-(5-아미노-2-(2-(디메틸아미노)에톡시)-4-메톡시페닐)아크릴아미드][N-(5-amino-2-(2-(dimethylamino)ethoxy)-4-methoxyphenyl)acrylamide]
Figure PCTKR2020010657-appb-img-000026
Figure PCTKR2020010657-appb-img-000026
전구체로 상기 제조예 6-1에서 제조한 tert-부틸 (5-아크릴아미도-4-(2-(디메틸아미노)에톡시)-2-메톡시페닐)카바메이트를 사용한 것을 제외하고, 상기 제조예 1-3의 단계 4에 따라 N-(5-아미노-2-(2-(디메틸아미노)에톡시)-4-메톡시페닐)아크릴아미드를 제조하여, 정제 없이 이후 단계에 사용하였다.Preparation above except that tert-butyl (5-acrylamido-4-(2-(dimethylamino)ethoxy)-2-methoxyphenyl)carbamate prepared in Preparation Example 6-1 was used as a precursor. N-(5-amino-2-(2-(dimethylamino)ethoxy)-4-methoxyphenyl)acrylamide was prepared according to step 4 of Example 1-3, and used in the subsequent step without purification.
<제조예 7-1> tert-부틸 (5-아크릴아미도-2-메톡시-4-(2-모폴리노에톡시)페닐)카바메이트의 제조<Production Example 7-1> Preparation of tert-butyl (5-acrylamido-2-methoxy-4- (2-morpholinoethoxy) phenyl) carbamate
[tert-부틸 (5-아크릴아미도-2-메톡시-4-(2-모폴리노에톡시)페닐)카바메이트][tert-butyl (5-acrylamido-2-methoxy-4-(2-morpholinoethoxy)phenyl) carbamate]
Figure PCTKR2020010657-appb-img-000027
Figure PCTKR2020010657-appb-img-000027
N 1,N 1,N 2-트리메틸에테인-1,2-디아민 대신 2-모폴리노에탄-1-올을 사용한 것을 제외하고는, 상기 제조예 1-3의 단계 1 내지 3에 따라 tert-부틸 (5-아크릴아미도-2-메톡시-4-(2-모폴리노에톡시)페닐)카바메이트(79% 수율)를 제조하였다.Tert- according to steps 1 to 3 of Preparation Example 1-3, except that 2-morpholinoethan-1-ol was used instead of N 1 ,N 1 ,N 2 -trimethylethane-1,2-diamine Butyl (5-acrylamido-2-methoxy-4- (2-morpholinoethoxy) phenyl) carbamate (79% yield) was prepared.
1H NMR (300 MHz, Chloroform- d) δ 8.88 (s, 1 H), 6.80 (s, 1 H), 6.51 (s, 1 H), 6.49 - 6.39 (d, J = 15.9 Hz, 1 H), 6.27 - 6.17 (m, 1 H), 5.81 - 5.67 (d, J = 11.8 Hz, 1H), 4.24 (s, 2 H), 3.90 (bs, 4 H), 3.84 (s, 3 H), 3.09 - 2.55 (bs, 6 H), 1.53 (s, 9 H); LCMS: 422.0 [M+H +]. 1 H NMR (300 MHz, Chloroform- d ) δ 8.88 (s, 1 H), 6.80 (s, 1 H), 6.51 (s, 1 H), 6.49-6.39 (d, J = 15.9 Hz, 1 H) , 6.27-6.17 (m, 1H), 5.81-5.67 (d, J = 11.8 Hz, 1H), 4.24 (s, 2H), 3.90 (bs, 4H), 3.84 (s, 3H), 3.09 -2.55 (bs, 6H), 1.53 (s, 9H); LCMS: 422.0 [M+H + ].
<제조예 7-2> N-(5-아미노-4-메톡시-2-(2-모폴리노에톡시)페닐)아크릴아미드의 제조<Production Example 7-2> Preparation of N-(5-amino-4-methoxy-2-(2-morpholinoethoxy)phenyl)acrylamide
[N-(5-아미노-4-메톡시-2-(2-모폴리노에톡시)페닐)아크릴아미드][N-(5-amino-4-methoxy-2-(2-morpholinoethoxy)phenyl)acrylamide]
*
Figure PCTKR2020010657-appb-img-000028
*
Figure PCTKR2020010657-appb-img-000028
전구체로 상기 제조예 7-1에서 제조한 tert-부틸 (5-아크릴아미도-2-메톡시-4-(2-모폴리노에톡시)페닐)카바메이트를 사용한 것을 제외하고, 상기 제조예 1-3의 단계 4에 따라 N-(5-아미노-4-메톡시-2-(2-모폴리노에톡시)페닐)아크릴아미드를 제조하였다.Preparation Example, except that tert-butyl (5-acrylamido-2-methoxy-4- (2-morpholinoethoxy) phenyl) carbamate prepared in Preparation Example 7-1 was used as a precursor. N-(5-amino-4-methoxy-2-(2-morpholinoethoxy)phenyl)acrylamide was prepared according to Step 4 of 1-3.
LCMS: 322.2 [M+H +].LCMS: 322.2 [M+H + ].
<제조예 8-1> tert-부틸 (5-아크릴아미도-2-메톡시-4-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)카바메이트의 제조<Preparation Example 8-1> Preparation of tert-butyl (5-acrylamido-2-methoxy-4-(methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)carbamate
[tert-부틸 (5-아크릴아미도-2-메톡시-4-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)카바메이트][tert-butyl (5-acrylamido-2-methoxy-4-(methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)carbamate]
Figure PCTKR2020010657-appb-img-000029
Figure PCTKR2020010657-appb-img-000029
N 1,N 1,N 2-트리메틸에테인-1,2-디아민 대신 N-메틸-2-(4-메틸피페라진-1-일)에탄-1-아민을 사용한 것을 제외하고는, 상기 제조예 1-3의 단계 1 내지 3에 따라 tert-부틸 (5-아크릴아미도-2-메톡시-4-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)카바메이트(70% 수율)를 제조하였다.The above Preparation Example, except that N-methyl-2-(4-methylpiperazin-1-yl)ethan-1-amine was used instead of N 1 ,N 1 ,N 2 -trimethylethane-1,2-diamine Tert-butyl (5-acrylamido-2-methoxy-4-(methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)carba according to steps 1 to 3 of 1-3 Mate (70% yield) was prepared.
1H NMR (300 MHz, Chloroform- d) δ 9.15 (s, 1 H), 9.02 (s, 1 H), 6.94 (s, 1 H), 6.70 (s, 1 H), 6.52 - 6.45 (d, J = 3.4 Hz, 1 H), 6.24 - 6.08 (m, 1 H), 5.81 - 5.70 (m, 1 H), 3.85 (s, 3 H), 3.06 - 2.98 (m, 2 H), 2.89 - 2.69 (m, 8 H), 2.65 (s, 3 H), 2.53 (s, 2 H), 2.50 (s, 3 H), 1.54 (s, 9 H); LCMS: 447.0 [M+H +]. 1 H NMR (300 MHz, Chloroform- d ) δ 9.15 (s, 1 H), 9.02 (s, 1 H), 6.94 (s, 1 H), 6.70 (s, 1 H), 6.52-6.45 (d, J = 3.4 Hz, 1 H), 6.24-6.08 (m, 1 H), 5.81-5.70 (m, 1 H), 3.85 (s, 3 H), 3.06-2.98 (m, 2 H), 2.89-2.69 (m, 8H), 2.65 (s, 3H), 2.53 (s, 2H), 2.50 (s, 3H), 1.54 (s, 9H); LCMS: 447.0 [M+H + ].
<제조예 8-2> N-(5-아미노-4-메톡시-2-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)아크릴아미드의 제조<Preparation Example 8-2> Preparation of N-(5-amino-4-methoxy-2-(methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)acrylamide
[N-(5-아미노-4-메톡시-2-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)아크릴아미드][N-(5-amino-4-methoxy-2-(methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)acrylamide]
Figure PCTKR2020010657-appb-img-000030
Figure PCTKR2020010657-appb-img-000030
전구체로 상기 제조예 8-1에서 제조한 tert-부틸 (5-아크릴아미도-2-메톡시-4-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)카바메이트를 사용한 것을 제외하고, 상기 제조예 1-3의 단계 4에 따라 N-(5-아미노-4-메톡시-2-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)아크릴아미드를 제조하였다.Tert-butyl (5-acrylamido-2-methoxy-4-(methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl) prepared in Preparation Example 8-1 as a precursor N-(5-amino-4-methoxy-2-(methyl(2-(4-methylpiperazin-1-yl)ethyl) according to Step 4 of Preparation Example 1-3 above, except that carbamate was used. )Amino)phenyl)acrylamide was prepared.
LCMS: 348.2 [M+H +].LCMS: 348.2 [M+H + ].
<제조예 9-1> tert-부틸 (5-아크릴아미도-2-메톡시-4-(메틸(2-모폴리노에틸)아미노)페닐)카바메이트의 제조<Preparation Example 9-1> Preparation of tert-butyl (5-acrylamido-2-methoxy-4- (methyl (2-morpholinoethyl) amino) phenyl) carbamate
[tert-부틸 (5-아크릴아미도-2-메톡시-4-(메틸(2-모폴리노에틸)아미노)페닐)카바메이트][tert-butyl (5-acrylamido-2-methoxy-4-(methyl(2-morpholinoethyl)amino)phenyl)carbamate]
Figure PCTKR2020010657-appb-img-000031
Figure PCTKR2020010657-appb-img-000031
N 1,N 1,N 2-트리메틸에테인-1,2-디아민 대신 N-메틸-2-모폴리노에탄-1-아민을 사용한 것을 제외하고는, 상기 제조예 1-3의 단계 1 내지 3에 따라 tert-부틸 (5-아크릴아미도-2-메톡시-4-(메틸(2-모폴리노에틸)아미노)페닐)카바메이트(69% 수율)를 제조하였다.Steps 1 to 3 of Preparation Example 1-3, except that N-methyl-2-morpholinoethan-1-amine was used instead of N 1 ,N 1 ,N 2 -trimethylethane-1,2-diamine According to the following, tert-butyl (5-acrylamido-2-methoxy-4-(methyl(2-morpholinoethyl)amino)phenyl)carbamate (69% yield) was prepared.
1H NMR (300 MHz, Chloroform- d) δ 9.15 (s, 1 H), 9.12 (s, 1 H), 6.94 (s, 1 H), 6.52 - 6.42 (m, 1 H), 6.25 - 6.07 (m, 1 H), 5.74 (d, J = 11.8 Hz, 1 H), 3.84 (s, 3 H), 3.81 - 3.71 (t, J = 5.9 Hz, 4 H), 3.41 (s, 3 H), 3.04 (s, 2 H), 2.74 - 2.65 (m, 2 H), 2.64 (s, 3 H), 2.45 (bs, 4 H), 1.54 (s, 9 H); LCMS: 435.2 [M+H +]. 1 H NMR (300 MHz, Chloroform- d ) δ 9.15 (s, 1 H), 9.12 (s, 1 H), 6.94 (s, 1 H), 6.52-6.42 (m, 1 H), 6.25-6.07 ( m, 1 H), 5.74 (d, J = 11.8 Hz, 1 H), 3.84 (s, 3 H), 3.81-3.71 (t, J = 5.9 Hz, 4 H), 3.41 (s, 3 H), 3.04 (s, 2H), 2.74-2.65 (m, 2H), 2.64 (s, 3H), 2.45 (bs, 4H), 1.54 (s, 9H); LCMS: 435.2 [M+H + ].
<제조예 9-2> N-(5-아미노-4-메톡시-2-(메틸(2-모폴리노에틸)아미노)페닐)아크릴아미드의 제조<Preparation Example 9-2> Preparation of N-(5-amino-4-methoxy-2-(methyl(2-morpholinoethyl)amino)phenyl)acrylamide
[N-(5-아미노-4-메톡시-2-(메틸(2-모폴리노에틸)아미노)페닐)아크릴아미드][N-(5-amino-4-methoxy-2-(methyl(2-morpholinoethyl)amino)phenyl)acrylamide]
Figure PCTKR2020010657-appb-img-000032
Figure PCTKR2020010657-appb-img-000032
전구체로 상기 제조예 9-1에서 제조한 tert-부틸 (5-아크릴아미도-2-메톡시-4-(메틸(2-모폴리노에틸)아미노)페닐)카바메이트를 사용한 것을 제외하고, 상기 제조예 1-3의 단계 4에 따라 N-(5-아미노-4-메톡시-2-(메틸(2-모폴리노에틸)아미노)페닐)아크릴아미드를 제조하였다.Except for using tert-butyl (5-acrylamido-2-methoxy-4- (methyl (2-morpholinoethyl) amino) phenyl) carbamate prepared in Preparation Example 9-1 as a precursor, According to Step 4 of Preparation Example 1-3, N-(5-amino-4-methoxy-2-(methyl(2-morpholinoethyl)amino)phenyl)acrylamide was prepared.
LCMS: 335.2 [M+H +].LCMS: 335.2 [M+H + ].
<실시예 1> N-5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노-2-((2-(디메틸아미노)에틸)(메틸)아미노)-4-메톡시페닐)아크릴아미드의 제조<Example 1> N-5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino-2-((2-(dimethylamino) Preparation of ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide
본 발명에 따른 피리미딘 유도체 화합물인 N-5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노-2-((2-(디메틸아미노)에틸)(메틸)아미노)-4-메톡시페닐)아크릴아미드를 하기 반응식 5에 나타난 바와 같은 방법으로 제조하였다.The pyrimidine derivative compound according to the present invention, N-5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino-2-((2- (Dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide was prepared in the same manner as shown in Scheme 5 below.
[반응식 5][Scheme 5]
Figure PCTKR2020010657-appb-img-000033
Figure PCTKR2020010657-appb-img-000033
상기 제조예 1-1에서 제조한 N-(2-((2,5-디클로로피리미딘-4-일)아미노)페닐)메탄설폰아미드(100 mg, 0.3 mmol)를 1N TFA를 포함한 n-부탄올(30 mL)에 용해시켜 교반한 용액에 상기 제조예 1-3에서 제조한 N-(5-아미노-2((2-(디메틸아미노)에틸(메틸)아미노)-4-메톡시페닐)아크릴아미드(71.5 mg, 0.3 mmol)을 상온에서 첨가하였다. 반응 튜브를 테플론-라인드 캡(Teflon-lined cap)으로 봉하고, 반응혼합물을 90℃에서 밤새 교반하였다. 교반액을 감압농축하였다. 잔여물을 DCM에 용해하고, 포화 NaHCO 3 수용액, 물 및 brine으로 세척하고, Na 2SO 4로 건조하고, 감압 하에 증발하였다. 수득된 원액은 컬럼 크로마토그래피(DCM:MeOH/9:1 내지 8.5:1.5)로 정제하여, N-5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노-2-((2-(디메틸아미노)에틸)(메틸)아미노)-4-메톡시페닐)아크릴아미드(40 mg, 23% 수율, 황백색 고체)를 얻었다.N-(2-((2,5-dichloropyrimidin-4-yl)amino)phenyl)methanesulfonamide (100 mg, 0.3 mmol) prepared in Preparation Example 1-1 was added to n-butanol containing 1N TFA. N-(5-amino-2((2-(dimethylamino)ethyl(methyl)amino)-4-methoxyphenyl)acrylic prepared in Preparation Example 1-3 above in a solution dissolved in (30 mL) and stirred Amide (71.5 mg, 0.3 mmol) was added at room temperature The reaction tube was sealed with a Teflon-lined cap, and the reaction mixture was stirred overnight at 90° C. The stirring solution was concentrated under reduced pressure. Was dissolved in DCM, washed with saturated NaHCO 3 aqueous solution, water and brine, dried over Na 2 SO 4 and evaporated under reduced pressure The obtained stock solution was subjected to column chromatography (DCM:MeOH/9:1 to 8.5:1.5 ) Purified with N-5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino-2-((2-(dimethylamino) Ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide (40 mg, 23% yield, off-white solid) was obtained.
1H NMR (400 MHz, DMSO- d 6) δ 10.03 (s, 1H), 9.29 (s, 1 H), 8.53 (s, 1 H), 8.39 (s, 1 H), 8.16 (s, 1 H), 8.12 (s, 1 H), 8.07 - 8.01 (m, 1 H), 7.431 - 7.22 (m, 1 H), 7.06 - 7.04 (dd, J = 6.6, 3.1 Hz, 2 H), 6.96 (s, 1 H), 6.42 - 6.32 (m, 1 H), 6.23 - 6.19 (dd, J = 16.9, 2.1 Hz, 1 H), 5.76 - 5.73 (dd, J = 10.1, 2.1 Hz, 1 H), 3.76 (s, 3H), 2.94 (s, 3 H), 2.91 - 2.88 (t, J = 5.8 Hz, 2 H), 2.70 (s, 3 H), 2.38 (s, 3 H), 2.26 (s, 6 H); LCMS: 590.0 [M+H +]. 1 H NMR (400 MHz, DMSO- d 6 ) δ 10.03 (s, 1H), 9.29 (s, 1 H), 8.53 (s, 1 H), 8.39 (s, 1 H), 8.16 (s, 1 H ), 8.12 (s, 1 H), 8.07-8.01 (m, 1 H), 7.431-7.22 (m, 1 H), 7.06-7.04 (dd, J = 6.6, 3.1 Hz, 2 H), 6.96 (s , 1 H), 6.42-6.32 (m, 1 H), 6.23-6.19 (dd, J = 16.9, 2.1 Hz, 1 H), 5.76-5.73 (dd, J = 10.1, 2.1 Hz, 1 H), 3.76 (s, 3H), 2.94 (s, 3H), 2.91-2.88 (t, J = 5.8 Hz, 2H), 2.70 (s, 3H), 2.38 (s, 3H), 2.26 (s, 6 H); LCMS: 590.0 [M+H + ].
<실시예 2> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-2-(4-(디메틸아미노)피페리딘-1-일)-4-메톡시페닐)아크릴아미드의 제조<Example 2> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-(4-(dimethylamino )Piperidin-1-yl)-4-methoxyphenyl)acrylamide
본 발명에 따른 피리미딘 유도체 화합물인 N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-2-(4-(디메틸아미노)피페리딘-1-일)-4-메톡시페닐)아크릴아미드를, 상기 제조예 2-2에서 제조한 N-(5-아미노-2-(4-(디메틸아미노)피페리딘-1-일)-4-메톡시페닐)아크릴아미드를 사용한 것을 제외하고는 상기 실시예 1에 따라 제조하였다(수율 27%).The pyrimidine derivative compound according to the present invention, N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-(4 -(Dimethylamino)piperidin-1-yl)-4-methoxyphenyl)acrylamide, prepared in Preparation Example 2-2, N-(5-amino-2-(4-(dimethylamino)pi) It was prepared according to Example 1, except that peridin-1-yl)-4-methoxyphenyl)acrylamide was used (yield 27%).
1H NMR (400 MHz, DMSO- d 6) δ 8.97 (s, 1 H), 8.58 (s, 1 H), 8.17 - 8.08 (m, 3 H), 8.08 - 7.99 (m, 1 H), 7.33-7.26 (dd, J = 7.3, 2.2 Hz, 1 H), 7.13 - 7.01 (m, 2 H), 6.79 (s, 1 H), 6.72 - 6.61 (dd, J = 16.9, 10.2 Hz, 1 H), 6.25 - 6.14 (dd, J = 17.0, 2.1 Hz, 1 H), 5.77 - 5.67 (dd, J = 10.0, 2.0 Hz, 1 H), 3.76 (s, 3 H), 3.10 - 3.00 (m, 2 H), 2.74 - 2.59 (m, 2 H), 2.31 (s, 7 H), 1.91 - 1.80 (d, J = 11.9 Hz, 2 H), 1.78 - 1.63 (m, 2 H); LCMS: 615.2 [M+H +]. 1 H NMR (400 MHz, DMSO- d 6 ) δ 8.97 (s, 1 H), 8.58 (s, 1 H), 8.17-8.08 (m, 3 H), 8.08-7.99 (m, 1 H), 7.33 -7.26 (dd, J = 7.3, 2.2 Hz, 1 H), 7.13-7.01 (m, 2 H), 6.79 (s, 1 H), 6.72-6.61 (dd, J = 16.9, 10.2 Hz, 1 H) , 6.25-6.14 (dd, J = 17.0, 2.1 Hz, 1 H), 5.77-5.67 (dd, J = 10.0, 2.0 Hz, 1 H), 3.76 (s, 3 H), 3.10-3.00 (m, 2 H), 2.74-2.59 (m, 2H), 2.31 (s, 7H), 1.91-1.80 (d, J = 11.9 Hz, 2H), 1.78-1.63 (m, 2H); LCMS: 615.2 [M+H + ].
<실시예 3> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)아크릴아미드의 제조<Example 3> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-( Preparation of 4-(4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)acrylamide
본 발명에 따른 피리미딘 유도체 화합물인 N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)아크릴아미드를, 상기 제조예 3-2에서 제조한 N-(5-아미노-4-메톡시-2-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)아크릴아미드를 사용한 것을 제외하고는 상기 실시예 1에 따라 제조하였다(수율 30%).The pyrimidine derivative compound according to the present invention, N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy -2-(4-(4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)acrylamide was prepared in Preparation Example 3-2. It was prepared according to Example 1, except that oxy-2-(4-(4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)acrylamide was used (yield 30%).
1H NMR (400 MHz, Methanol- d 4) δ 8.11 (s, 1 H), 7.95 (s, 1 H), 7.83 - 7.77 (d, J = 7.4 Hz, 1 H), 7.49 - 7.43 (d, J = 7.5 Hz, 1 H), 7.35 - 7.26 (m, 2 H), 6.93 (s, 1 H), 6.61 - 6.51 (dd, J = 17.0, 10.2 Hz, 1 H), 6.43 - 6.36 (d, J = 16.9 Hz, 1 H), 5.89 - 5.83 (d, J = 10.3 Hz, 1 H), 3.89 (s, 3 H), 3.30 - 3.10 (m, 5 H), 2.99 (s, 3 H), 2.89 (s, 6 H), 2.15 - 2.07 (d, J = 12.2 Hz, 2 H), 1.97 - 1.82 (m, 2 H); LCMS: 670.2 [M+H +]. 1 H NMR (400 MHz, Methanol- d 4 ) δ 8.11 (s, 1 H), 7.95 (s, 1 H), 7.83-7.77 (d, J = 7.4 Hz, 1 H), 7.49-7.43 (d, J = 7.5 Hz, 1 H), 7.35-7.26 (m, 2 H), 6.93 (s, 1 H), 6.61-6.51 (dd, J = 17.0, 10.2 Hz, 1 H), 6.43-6.36 (d, J = 16.9 Hz, 1 H), 5.89-5.83 (d, J = 10.3 Hz, 1 H), 3.89 (s, 3 H), 3.30-3.10 (m, 5 H), 2.99 (s, 3 H), 2.89 (s, 6H), 2.15-2.07 (d, J = 12.2 Hz, 2H), 1.97-1.82 (m, 2H); LCMS: 670.2 [M+H + ].
<실시예 4> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-모폴리노페닐)아크릴아미드의 제조<Example 4> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-mo Preparation of polynophenyl)acrylamide
본 발명에 따른 피리미딘 유도체 화합물인 N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-모폴리노페닐)아크릴아미드를, 상기 제조예 4-2에서 제조한 N-(5-아미노-4-메톡시-2-모폴리노페닐)아크릴아미드를 사용한 것을 제외하고는 상기 실시예 1에 따라 제조하였다(수율 31%).The pyrimidine derivative compound according to the present invention, N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy -2-morpholinophenyl)acrylamide, except that the N-(5-amino-4-methoxy-2-morpholinophenyl)acrylamide prepared in Preparation Example 4-2 was used. It was prepared according to Example 1 (yield 31%).
1H NMR (400 MHz, Methanol- d 4) δ 8.10 (s, 1H), 7.93 (s, 1 H), 7.82 - 7.74 (d, J = 7.8 Hz, 1 H), 7.49 - 7.43 (d, J = 7.6 Hz, 1 H), 7.39 - 7.26 (m, 2 H), 6.92 (s, 1 H), 6.61 - 6.51 (dd, J = 17.0, 10.1 Hz, 1 H), 6.42 - 6.32 (d, J = 17.0 Hz, 1 H), 5.89 - 5.81 (d, J = 10.4 Hz, 1 H), 3.89 (s, 7 H), 3.00 (s, 3 H), 2.98 - 2.91 (t, J = 4.6 Hz, 4 H); LCMS: 575.2 [M+H +]. 1 H NMR (400 MHz, Methanol- d 4 ) δ 8.10 (s, 1H), 7.93 (s, 1 H), 7.82-7.74 (d, J = 7.8 Hz, 1 H), 7.49-7.43 (d, J = 7.6 Hz, 1 H), 7.39-7.26 (m, 2 H), 6.92 (s, 1 H), 6.61-6.51 (dd, J = 17.0, 10.1 Hz, 1 H), 6.42-6.32 (d, J = 17.0 Hz, 1 H), 5.89-5.81 (d, J = 10.4 Hz, 1 H), 3.89 (s, 7 H), 3.00 (s, 3 H), 2.98-2.91 (t, J = 4.6 Hz, 4H); LCMS: 575.2 [M+H + ].
<실시예 5> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(4-메틸피페라진-1-일)페닐)아크릴아미드의 제조<Example 5> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-( Preparation of 4-methylpiperazin-1-yl)phenyl)acrylamide
본 발명에 따른 피리미딘 유도체 화합물인 N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(4-메틸피페라진-1-일)페닐)아크릴아미드를, 상기 제조예 5-2에서 제조한 N-(5-아미노-4-메톡시-2-(4-메틸피페라진-1-일)페닐)아크릴아미드를 사용한 것을 제외하고는 상기 실시예 1에 따라 제조하였다(수율 41%).The pyrimidine derivative compound according to the present invention, N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy -2-(4-methylpiperazin-1-yl)phenyl)acrylamide, prepared in Preparation Example 5-2, N-(5-amino-4-methoxy-2-(4-methylpiperazine-) It was prepared according to Example 1, except that 1-yl)phenyl)acrylamide was used (yield 41%).
1H NMR (400 MHz, Methanol- d 4) δ 8.13 (s, 1 H), 7.85 - 7.78 (m, 1 H), 7.48 - 7.40 (m, 1 H), 7.33 - 7.24 m, 1 H), 6.89 (s, 1 H), 6.63 - 6.52 (dd, J = 17.0, 10.3 Hz, 1 H), 6.43 - 6.33 (d, J = 16.9 Hz, 1 H), 5.90 - 5.84 (d, J = 10.0 Hz, 1 H), 3.91 (s, 3 H), 3.67 - 3.58 (d, J = 11.9 Hz, 2 H), 3.30 - 3.21 (m, 4 H), 3.18 - 3.10 (d, J = 12.0 Hz, 2 H), 3.01 (s, 3 H), 2.98 (s, 3 H); LCMS: 587.2 [M+H +]. 1 H NMR (400 MHz, Methanol- d 4 ) δ 8.13 (s, 1 H), 7.85-7.78 (m, 1 H), 7.48-7.40 (m, 1 H), 7.33-7.24 m, 1 H), 6.89 (s, 1 H), 6.63-6.52 (dd, J = 17.0, 10.3 Hz, 1 H), 6.43-6.33 (d, J = 16.9 Hz, 1 H), 5.90-5.84 (d, J = 10.0 Hz , 1 H), 3.91 (s, 3 H), 3.67-3.58 (d, J = 11.9 Hz, 2 H), 3.30-3.21 (m, 4 H), 3.18-3.10 (d, J = 12.0 Hz, 2 H), 3.01 (s, 3H), 2.98 (s, 3H); LCMS: 587.2 [M+H + ].
<실시예 6> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-2-(2-(디메틸아미노)에톡시)-4-메톡시페닐)아크릴아미드의 제조<Example 6> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-(2-(dimethylamino )Ethoxy)-4-methoxyphenyl)acrylamide preparation
본 발명에 따른 피리미딘 유도체 화합물인 N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-2-(2-(디메틸아미노)에톡시)-4-메톡시페닐)아크릴아미드를, 상기 제조예 6-2에서 제조한 N-(5-아미노-2-(2-(디메틸아미노)에톡시)-4-메톡시페닐)아크릴아미드를 제외하고는 상기 실시예 1에 따라 제조하였다(수율 29%).The pyrimidine derivative compound according to the present invention, N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-(2 -(Dimethylamino)ethoxy)-4-methoxyphenyl)acrylamide, prepared in Preparation Example 6-2, N-(5-amino-2-(2-(dimethylamino)ethoxy)-4- It was prepared according to Example 1 except for methoxyphenyl)acrylamide (yield 29%).
1H NMR (400 MHz, Methanol- d 4) δ 8.11 (s, 1 H), 7.90 (s, 1 H), 7.87 - 7.79 (m, 1 H), 7.47 - 7.40 (d, J = 7.5 Hz, 1 H), 7.31 - 7.21 (m, 2 H), 6.87 (s, 1 H), 6.58 - 6.39 (m, 2 H), 5.93 - 5.85 (d, J = 9.8 Hz, 1 H), 4.56 - 4.58 (t, J = 5.0 Hz, 2 H), 3.95 (s, 3 H), 3.65 - 3.58 (t, J = 4.8 Hz, 2 H), 3.02 (s, 6 H), 2.98 (s, 3 H); LCMS: 576.8 [M+H +]. 1 H NMR (400 MHz, Methanol- d 4 ) δ 8.11 (s, 1 H), 7.90 (s, 1 H), 7.87-7.79 (m, 1 H), 7.47-7.40 (d, J = 7.5 Hz, 1 H), 7.31-7.21 (m, 2 H), 6.87 (s, 1 H), 6.58-6.39 (m, 2 H), 5.93-5.85 (d, J = 9.8 Hz, 1 H), 4.56-4.58 (t, J = 5.0 Hz, 2 H), 3.95 (s, 3 H), 3.65-3.58 (t, J = 4.8 Hz, 2 H), 3.02 (s, 6 H), 2.98 (s, 3 H) ; LCMS: 576.8 [M+H + ].
<실시예 7> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(메틸(2-모폴리노에틸)아미노)페닐)아크릴아미드의 제조<Example 7> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-( Preparation of methyl (2-morpholinoethyl) amino) phenyl) acrylamide
본 발명에 따른 피리미딘 유도체 화합물인 N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(메틸(2-모폴리노에틸)아미노)페닐)아크릴아미드를, 상기 제조예 7-2에서 제조한 N-(5-아미노-4-메톡시-2-(2-모폴리노에톡시)페닐)아크릴아미드를 사용한 것을 제외하고는 상기 실시예 1에 따라 제조하였다(수율 22%).The pyrimidine derivative compound according to the present invention, N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy -2-(methyl(2-morpholinoethyl)amino)phenyl)acrylamide was prepared in Preparation Example 7-2 above. N-(5-amino-4-methoxy-2-(2-morpholino) It was prepared according to Example 1, except that ethoxy)phenyl)acrylamide was used (yield 22%).
1H NMR (400 MHz, Methanol- d 4) δ 8.13 (s, 1 H), 7.83 - 7.75 (m, 2 H), 7.49 - 7.41 (m, 1 H), 7.33 - 7.24 (m, 2 H), 6.88 (s, 1 H), 6.60 - 6.49 (dd, J = 17.0, 9.9 Hz, 1 H), 6.48 - 6.40 (d, J = 16.8 Hz, 1 H), 5.95 - 5.88(d, J = 9.9 Hz, 1 H), 4.61 - 4.54 (t, J = 5.1 Hz, 2 H), 4.03 (bs, 4 H), 3.95 (s, 3 H), 3.67 - 3.60 (t, J = 4.9 Hz, 2 H), 3.33 (bs, 4 H), 2.97 (s, 3 H); LCMS: 619.2 [M+2H +]. 1 H NMR (400 MHz, Methanol- d 4 ) δ 8.13 (s, 1 H), 7.83-7.75 (m, 2 H), 7.49-7.41 (m, 1 H), 7.33-7.24 (m, 2 H) , 6.88 (s, 1 H), 6.60-6.49 (dd, J = 17.0, 9.9 Hz, 1 H), 6.48-6.40 (d, J = 16.8 Hz, 1 H), 5.95-5.88(d, J = 9.9 Hz, 1 H), 4.61-4.54 (t, J = 5.1 Hz, 2 H), 4.03 (bs, 4 H), 3.95 (s, 3 H), 3.67-3.60 (t, J = 4.9 Hz, 2 H ), 3.33 (bs, 4H), 2.97 (s, 3H); LCMS: 619.2 [M+2H + ].
<실시예 8> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)아크릴아미드의 제조<Example 8> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-( Preparation of methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)acrylamide
본 발명에 따른 피리미딘 유도체 화합물인 N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)아크릴아미드를, 상기 제조예 8-2에서 제조한 N-(5-아미노-4-메톡시-2-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)아크릴아미드를 사용한 것을 제외하고는 상기 실시예 1에 따라 제조하였다(수율 27%).The pyrimidine derivative compound according to the present invention, N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy -2-(Methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)acrylamide was prepared in Preparation Example 8-2 above N-(5-amino-4-methoxy- It was prepared according to Example 1, except that 2-(methyl(2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)acrylamide was used (yield 27%).
1H NMR (500 MHz, Methanol- d 4) δ 8.16 (s, 1 H), 7.88 (s, 1 H), 7.82 - 7.76 (d, J = 7.0 Hz, 1 H), 7.50 - 7.44 (m, 1 H), 7.34 - 7.27 (m, 2 H), 7.00 (s, 1 H), 6.64 - 6.55 (dd, J = 16.9, 10.0 Hz, 1 H), 6.52 - 6.46 (d, J = 17.0 Hz, 1 H), 5.99 (d, J = 10.1 Hz), 3.96 (s, 3 H), 3.51 - 3.45 (bs, 2 H), 3.41 - 3.35 (bs, 4 H), 3.23 - 3.14 (bs, 4 H), 3.08 (s, 2 H), 2.97 (s, 3 H), 2.87 (s, 3 H), 2.82 (s, 3 H); LCMS: 645.0 [M+2H +]. 1 H NMR (500 MHz, Methanol- d 4 ) δ 8.16 (s, 1 H), 7.88 (s, 1 H), 7.82-7.76 (d, J = 7.0 Hz, 1 H), 7.50-7.44 (m, 1 H), 7.34-7.27 (m, 2 H), 7.00 (s, 1 H), 6.64-6.55 (dd, J = 16.9, 10.0 Hz, 1 H), 6.52-6.46 (d, J = 17.0 Hz, 1 H), 5.99 (d, J = 10.1 Hz), 3.96 (s, 3 H), 3.51-3.45 (bs, 2 H), 3.41-3.35 (bs, 4 H), 3.23-3.14 (bs, 4 H) ), 3.08 (s, 2H), 2.97 (s, 3H), 2.87 (s, 3H), 2.82 (s, 3H); LCMS: 645.0 [M+2H + ].
<실시예 9> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미도)피리미딘-2-일)아미노)-4-메톡시-2-(2-모폴리노에톡시)페닐)아크릴아미드의 제조<Example 9> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amido)pyrimidin-2-yl)amino)-4-methoxy-2- Preparation of (2-morpholinoethoxy)phenyl)acrylamide
본 발명에 따른 피리미딘 유도체 화합물인 N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미도)피리미딘-2-일)아미노)-4-메톡시-2-(2-모폴리노에톡시)페닐)아크릴아미드를, 상기 제조예 9-2에서 제조한 N-(5-아미노-4-메톡시-2-(메틸(2-모폴리노에틸)아미노)페닐)아크릴아미드를 사용한 것을 제외하고는 상기 실시예 1에 따라 제조하였다(수율 26%).The pyrimidine derivative compound according to the present invention, N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amido)pyrimidin-2-yl)amino)-4-me N-(5-amino-4-methoxy-2-(methyl(2-morpholino) prepared in Preparation Example 9-2) of oxy-2-(2-morpholinoethoxy)phenyl)acrylamide It was prepared according to Example 1, except that ethyl) amino) phenyl) acrylamide was used (yield 26%).
1H NMR (400 MHz, DMSO- d 6) δ 9.34 - 9.21 (m, 2 H), 8.48 (s, 1 H), 8.22 (s, 1 H), 8.17 - 8.09 (m, 2 H), 8.06 - 7.98 (d, J = 7.9 Hz, 1 H), 7.36 - 7.27 (dd, J = 7.7, 1.8 Hz, 1 H), 7.16 - 7.02 (m, 2 H), 6.92 (s, 1 H), 6.67 - 6.54 (m, 1 H), 6.26 - 6.15 (dd, J = 17.0, 2.0 Hz, 1 H), 5.80 - 5.69 (d, J = 12.2 Hz, 1 H), 3.77 (s, 3 H), 3.56 (t, J = 4.7 Hz, 4 H), 2.02 - 2.90 (m, 5 H), 2.68 (s, 3 H), 2.43 - 2.28 (m, 6 H); LCMS: 632.2 [M+H +]. 1 H NMR (400 MHz, DMSO- d 6 ) δ 9.34-9.21 (m, 2 H), 8.48 (s, 1 H), 8.22 (s, 1 H), 8.17-8.09 (m, 2 H), 8.06 -7.98 (d, J = 7.9 Hz, 1 H), 7.36-7.27 (dd, J = 7.7, 1.8 Hz, 1 H), 7.16-7.02 (m, 2 H), 6.92 (s, 1 H), 6.67 -6.54 (m, 1 H), 6.26-6.15 (dd, J = 17.0, 2.0 Hz, 1 H), 5.80-5.69 (d, J = 12.2 Hz, 1 H), 3.77 (s, 3 H), 3.56 (t, J = 4.7 Hz, 4H), 2.02-2.90 (m, 5H), 2.68 (s, 3H), 2.43-2.28 (m, 6H); LCMS: 632.2 [M+H + ].
하기 표 1에 실시예 1 내지 9에서 제조한 화합물의 화학구조식을 정리하여 나타내었다.The chemical structural formulas of the compounds prepared in Examples 1 to 9 are summarized and shown in Table 1 below.
실시예Example 화학구조Chemical structure 실시예Example 화학구조Chemical structure
1One
Figure PCTKR2020010657-appb-img-000034
Figure PCTKR2020010657-appb-img-000034
22
Figure PCTKR2020010657-appb-img-000035
Figure PCTKR2020010657-appb-img-000035
33
Figure PCTKR2020010657-appb-img-000036
Figure PCTKR2020010657-appb-img-000036
44
Figure PCTKR2020010657-appb-img-000037
Figure PCTKR2020010657-appb-img-000037
55
Figure PCTKR2020010657-appb-img-000038
Figure PCTKR2020010657-appb-img-000038
66
Figure PCTKR2020010657-appb-img-000039
Figure PCTKR2020010657-appb-img-000039
77
Figure PCTKR2020010657-appb-img-000040
Figure PCTKR2020010657-appb-img-000040
88
Figure PCTKR2020010657-appb-img-000041
Figure PCTKR2020010657-appb-img-000041
99
Figure PCTKR2020010657-appb-img-000042
Figure PCTKR2020010657-appb-img-000042
<실험예 1> 야생형 EGFR 및 EGFR 돌연변이 효소 억제능<Experimental Example 1> Wild-type EGFR and EGFR mutant enzyme inhibitory ability
본 발명에 따른 화학식 1로 표시되는 화합물의 야생형 EGFR 및 EGFR 돌연변이 효소에 대한 억제능을 확인하기 위하여, 하기와 같은 실험을 수행하였다. 그 결과를 하기 표 2에 나타내었다.In order to confirm the inhibitory ability of the compound represented by Formula 1 according to the present invention against wild-type EGFR and EGFR mutant enzymes, the following experiment was performed. The results are shown in Table 2 below.
본 발명의 화합물에 대한 야생형 및 EGFR 돌연변이 효소에 대한 활성측정은 Cisbio 사에서 판매하는 HTRF 시스템을 활용하여 다음과 같이 실험하였다. EGFR 야생형 및 EGFR del19/T790M 돌연변이 효소는 Carna Biosciences 에서 제공하는 재조합 단백질을 구매하여 사용하였다.The activity measurement for wild-type and EGFR mutant enzymes for the compounds of the present invention was tested as follows using the HTRF system sold by Cisbio. EGFR wild-type and EGFR del19/T790M mutant enzymes were used by purchasing a recombinant protein provided by Carna Biosciences.
활성측정에 사용된 assay buffer의 조성은 50 mM Tris-HCl pH 7.5, 100 mM NaCl, 7.5 mM MgCl 2, 3 mM KCl, 0.01% Tween 20, 0.1% BSA, 1 mM DTT 이었다. 여기에 50 mM 농도의 ATP 와 0.5 mM 농도의 biotin으로 표지된 펩타이드 기질을 사용하여 효소반응을 진행하였다. 화합물의 EGFR 활성억제효과 분석은 하기의 분석 반응 레시피에 따라 진행되었다. The composition of the assay buffer used for the activity measurement was 50 mM Tris-HCl pH 7.5, 100 mM NaCl, 7.5 mM MgCl 2 , 3 mM KCl, 0.01% Tween 20, 0.1% BSA, and 1 mM DTT. Here, the enzyme reaction was carried out using a peptide substrate labeled with 50 mM ATP and 0.5 mM biotin. The analysis of the EGFR activity inhibitory effect of the compound was performed according to the following analytical reaction recipe.
Component 1: 4 mL 의 EGFR 야생형 또는 돌연변이 효소Component 1: 4 mL of EGFR wild-type or mutant enzyme
Component 2: 2 mL 의 화합물 용액Component 2: 2 mL of compound solution
Component 3: 4 mL ATP 와 biotin 표지 펩타이드 Component 3: 4 mL ATP and biotin-labeled peptide
효소반응은 component 1 과 component 2 를 먼저 섞어준 후에 component 3을 첨가하여 시작한다. 37℃에서 2시간 반응 후에 Cisbio 사에서 제공되는 streptavidin-XL665 와 europium 표지된 anti-phosphotyrosine antibody로 이루어진 측정용액 10 mL를 효소반응 용액에 추가하고 1시간 동안 상온에서 반응한다. 최종적으로 Perkin-Elmer 사의 Envision 장비를 이용하여 615 nm 와 665 nm 에서의 형광 값의 비율을 구하여 효소활성을 정량적으로 측정하고 화합물의 억제능을 확인한다. 7가지 화합물 농도에서 측정된 측정값을 Prism 프로그램(버전 5.01, Graphpad Software, Inc.)을 사용하여 분석하고 화합물의 억제능 지표인 IC 50값을 산출하였다. Enzymatic reaction starts by mixing component 1 and component 2 first, and then adding component 3 to it. After 2 hours reaction at 37℃, 10 mL of a measurement solution consisting of streptavidin-XL665 and europium-labeled anti-phosphotyrosine antibody provided by Cisbio was added to the enzyme reaction solution and reacted at room temperature for 1 hour. Finally, the ratio of fluorescence values at 615 nm and 665 nm is calculated using Perkin-Elmer's Envision equipment, and the enzyme activity is quantitatively measured and the inhibitory ability of the compound is confirmed. The measured values measured at the concentrations of the seven compounds were analyzed using the Prism program (version 5.01, Graphpad Software, Inc.), and the IC 50 value, an index of the inhibitory ability of the compound, was calculated.
EGFR wt. IC 50(μM)EGFR wt. IC 50 (μM) EGFR del19/T790M IC 50(μM)EGFR del19/T790M IC 50 (μM)
오시머티닙Osimertinib 0.00310.0031 <0.001<0.001
실시예 1Example 1 <0.001<0.001 <0.001<0.001
실시예 2Example 2 0.0640.064 <0.001<0.001
실시예 3Example 3 0.010.01 <0.001<0.001
실시예 4Example 4 0.370.37 <0.001<0.001
실시예 5Example 5 0.0180.018 <0.001<0.001
실시예 6Example 6 0.00160.0016 <0.001<0.001
실시예 7Example 7 0.00940.0094 <0.001<0.001
실시예 8Example 8 0.0290.029 <0.001<0.001
실시예 9Example 9 0.000650.00065 <0.001<0.001
상기 표 4에 나타낸 바와 같이, As shown in Table 4 above,
본 발명의 모든 실시예 화합물이 야생형 EGFR에 대하여 상대적으로 약한 EGFR 활성억제효과를 나타내면서, EGFR 돌연변이에 대하여 선택적으로 높은 억제능을 나타내고, 이는 기존 EGFR TKI(tyrosine kinase inhibitor) 약물인 오시머티닙의 경우와 유사한 것을 확인하였다.All the compounds of the present invention exhibit a relatively weak EGFR activity inhibitory effect against wild-type EGFR, and selectively exhibit high inhibitory ability against EGFR mutations, which is similar to the case of osimertinib, a conventional EGFR tyrosine kinase inhibitor (TKI) drug. Similar things were confirmed.
상기 결과를 통해, 본 발명에 따른 피리미딘 유도체 화합물은 EGFR 돌연변이를 효과적으로 억제할 수 있으므로, 암의 예방 또는 치료용 약학적 조성물로 유용하게 사용할 수 있음을 알 수 있다.Through the above results, it can be seen that the pyrimidine derivative compound according to the present invention can effectively inhibit the EGFR mutation, and thus can be usefully used as a pharmaceutical composition for preventing or treating cancer.
<실험예 2> Ba/F3 세포주에서의 야생형 Ba/F3 EGFR 및 Ba/F3 EGFR 돌연변이 세포성장 억제능 측정<Experimental Example 2> Measurement of wild-type Ba/F3 EGFR and Ba/F3 EGFR mutant cell growth inhibitory ability in Ba/F3 cell lines
본 발명에 따른 화학식 1로 표시되는 화합물의 Ba/F3 세포주에서의 야생형 Ba/F3 EGFR 및 Ba/F3 EGFR 돌연변이에 대한 세포성장 억제능을 확인하기 위하여, 하기와 같은 실험을 수행하였다. 그 결과를 하기 표 3 및 표 4에 나타내었다.In order to confirm the ability of the compound represented by Formula 1 according to the present invention to inhibit cell growth against wild-type Ba/F3 EGFR and Ba/F3 EGFR mutations in Ba/F3 cell lines, the following experiment was performed. The results are shown in Tables 3 and 4 below.
*본 발명의 화합물의 야생형 및 돌연변이 Ba/F3 EGFR 세포주에 대한 활성측정은 Promega 사에서 판매하는 CellTiter-Glo 시스템을 활용하여 다음과 같이 실험하였다. CellTiter-Glo assay은 세포 배양 상태에서 세포에 존재하는 ATP를 측정하여 세포 viability를 확인하는 방법이다. Ba/F3 EGFR 야생형 및 Ba/F3 EGFR del19, Ba/F3 EGFR L858R, Ba/F3 EGFR del19/T790M, Ba/F3 EGFR L858R/T790M, Ba/F3 del19/T790M/C797S, Ba/F3 EGFR L858R/T790M/C797S 돌연변이 세포주는 크라운 바이오사이언스사에서 제공하는 세포주를 구매하여 사용하였다. Ba/F3 EGFR 야생형 및 Ba/F3 EGFR del19, Ba/F3 EGFR L858R, Ba/F3 EGFR del19/T790M, Ba/F3 EGFR L858R/T790M, Ba/F3 del19/T790M/C797S, Ba/F3 EGFR L858R/T790M/C797S 돌연변이 세포주는 10% FBS, 1% penicillin-streptomycin이 들어있는 RPMI 에 puromycine 1ug 을 넣어 37℃, 5% CO 2 인큐베이터에 배양하였다. * The activity of the compounds of the present invention against wild-type and mutant Ba/F3 EGFR cell lines was tested as follows using the CellTiter-Glo system sold by Promega. CellTiter-Glo assay is a method to check cell viability by measuring ATP present in cells in the cell culture state. Ba/F3 EGFR wild type and Ba/F3 EGFR del19, Ba/F3 EGFR L858R, Ba/F3 EGFR del19/T790M, Ba/F3 EGFR L858R/T790M, Ba/F3 del19/T790M/C797S, Ba/F3 EGFR L858R/T790M The /C797S mutant cell line was used by purchasing a cell line provided by Crown Bioscience. Ba/F3 EGFR wild type and Ba/F3 EGFR del19, Ba/F3 EGFR L858R, Ba/F3 EGFR del19/T790M, Ba/F3 EGFR L858R/T790M, Ba/F3 del19/T790M/C797S, Ba/F3 EGFR L858R/T790M The /C797S mutant cell line was cultured in an RPMI containing 10% FBS and 1% penicillin-streptomycin with 1 ug of puromycine in an incubator at 37°C and 5% CO 2.
화합물의 EGFR 세포성장 억제능 효과 분석은 하기의 분석 반응 레시피에 따라 진행되었다. Analysis of the EGFR cell growth inhibitory effect of the compound was carried out according to the following analytical reaction recipe.
2500 cells/ 90 μL 을 96 well 세포배양 plate 에 계대하여 배양하고 24시간 후에 화학식 1로 표시되는 화합물을 0, 0.01, 0.03, 0.1, 0.3, 1, 3, 10 (μM) 로 처리하였다. 72시간 동안 반응 후 화합물을 처리한 plate를 30분 동안 상온에 방치 한 후 reagent를 100 μL 더 처리 하고 10분 동안 상온에서 shaking하였다. 최종적으로 장비를 이용하여 570 nm 에서의 형광 값의 비율을 구하여 정량적으로 측정하고 화합물의 세포성장 억제능을 확인한다. 8가지 화합물 농도에서 측정된 측정값을 Prism 프로그램(버전 5.01, Graphpad Software, Inc.)을 사용하여 분석하고 화합물의 세포성장 억제능 지표인 IC 50값을 산출하였다. 2500 cells/90 μL were cultured by passage in a 96 well cell culture plate, and after 24 hours, the compound represented by Formula 1 was treated with 0, 0.01, 0.03, 0.1, 0.3, 1, 3, 10 (μM). After the reaction for 72 hours, the plate treated with the compound was allowed to stand at room temperature for 30 minutes, and then 100 μL of the reagent was further treated and shaking at room temperature for 10 minutes. Finally, the ratio of the fluorescence value at 570 nm is calculated using an equipment, and the quantitative measurement is performed, and the ability of the compound to inhibit cell growth is confirmed. The measured values measured at the concentrations of the 8 compounds were analyzed using the Prism program (version 5.01, Graphpad Software, Inc.), and the IC 50 value, an indicator of the cell growth inhibitory ability of the compound, was calculated.
Ba/F3 EGFR wt. IC 50(μM)Ba/F3 EGFR wt. IC 50 (μM) Ba/F3 EGFR del19 IC 50(μM)Ba/F3 EGFR del19 IC 50 (μM) Ba/F3 EGFR L858R IC 50(μM)Ba/F3 EGFR L858R IC 50 (μM)
오시머티닙Osimertinib 0.920.92 0.0020.002 0.0070.007
실시예 1Example 1 1.81.8 0.0010.001 0.0020.002
실시예 2Example 2 0.50.5 0.0090.009 0.0530.053
실시예 3Example 3 0.490.49 0.0040.004 0.0280.028
실시예 4Example 4 0.410.41 0.0050.005 0.0020.002
실시예 5Example 5 0.230.23 0.0080.008 0.0170.017
실시예 6Example 6 0.0240.024 0.0010.001 0.0030.003
실시예 7Example 7 0.40.4 0.0090.009 0.0340.034
실시예 8Example 8 0.270.27 0.0090.009 0.0280.028
실시예 9Example 9 0.0120.012 0.0010.001 0.0010.001
Ba/F3 EGFR del19/T790M IC 50(μM)Ba/F3 EGFR del19/T790M IC 50 (μM) Ba/F3 EGFR L858R/T790M IC 50(μM)Ba/F3 EGFR L858R/T790M IC 50 (μM) Ba/F3 EGFR del19/T790M/C797S IC 50(μM)Ba/F3 EGFR del19/T790M/C797S IC 50 (μM) Ba/F3 EGFR L858R/T790M/C797S IC 50(μM)Ba/F3 EGFR L858R/T790M/C797S IC 50 (μM)
오시머티닙Osimertinib 0.0030.003 0.0020.002 0.330.33 0.810.81
실시예 1Example 1 0.0030.003 0.0080.008 0.120.12 0.120.12
실시예 2Example 2 0.0150.015 0.0350.035 0.240.24 0.390.39
실시예 3Example 3 0.0050.005 0.0120.012 0.160.16 0.220.22
실시예 4Example 4 0.0250.025 0.0170.017 0.320.32 0.470.47
실시예 5Example 5 0.0010.001 0.0210.021 0.0920.092 0.720.72
실시예 6Example 6 0.0020.002 0.0080.008 1One 0.990.99
실시예 7Example 7 0.0060.006 0.010.01 0.990.99 0.10.1
실시예 8Example 8 0.0280.028 0.0130.013 0.010.01 0.840.84
실시예 9Example 9 0.0010.001 0.0010.001 0.960.96 0.90.9
상기 표 3 및 표 4에 나타낸 바와 같이, As shown in Table 3 and Table 4 above,
본 발명의 모든 실시예 화합물이 Ba/F3 세포주에서 야생형 EGFR에 대하여 상대적으로 약한 활성억제효과를 나타내면서, 삼중 돌연변이인 EGFR del19/T790M/C797S, EGFR L858R/T790M/C797S를 포함한 EGFR 돌연변이에 대하여 선택적으로 높은 세포성장 억제능을 나타내고, 특히 기존 EGFR TKI 약물인 오시머티닙의 경우보다 우수한 것을 확인하였다.All the compounds of the present invention exhibited relatively weak inhibitory effect against wild-type EGFR in Ba/F3 cell lines, and selectively against EGFR mutations including triple mutations EGFR del19/T790M/C797S, EGFR L858R/T790M/C797S. It showed high cell growth inhibitory ability, and was found to be particularly superior to the case of osimertinib, an existing EGFR TKI drug.
상기 결과를 통해, 본 발명에 따른 피리미딘 유도체 화합물은 EGFR 돌연변이를 효과적으로 억제할 수 있으므로, 암의 예방 또는 치료용 약학적 조성물로 유용하게 사용할 수 있음을 알 수 있다.Through the above results, it can be seen that the pyrimidine derivative compound according to the present invention can effectively inhibit the EGFR mutation, and thus can be usefully used as a pharmaceutical composition for preventing or treating cancer.
<실험예 3> Ba/F3 세포주에서의 Ba/F3 EGFR exon20 insertion 돌연변이 세포성장 억제능<Experimental Example 3> Ba/F3 EGFR exon20 insertion mutant cell growth inhibitory ability in Ba/F3 cell lines
본 발명에 따른 실시예 1로 표시되는 화합물의 Ba/F3 세포주에서의 Ba/F3 EGFR exon20 insertion 돌연변이에 대한 세포성장 억제능을 확인하기 위하여, 하기와 같은 실험을 수행하였다. 그 결과를 하기 표 5에 나타내었다.In order to confirm the ability of the compound represented by Example 1 according to the present invention to inhibit cell growth against the Ba/F3 EGFR exon20 insertion mutation in the Ba/F3 cell line, the following experiment was performed. The results are shown in Table 5 below.
본 발명의 화합물에 대한 돌연변이 Ba/F3 EGFR 세포주에 대한 활성측정은 Promega 사에서 판매하는 CellTiter-Glo 시스템을 활용하여 다음과 같이 실험하였다. CellTiter-Glo assay은 세포 배양 상태에서 세포에 존재하는 ATP를 측정하여 세포 viability를 확인하는 방법이다. Ba/F3 EGFR D770delinsGY, H773_V774insH, Y764_V765insHH, V769_D770insASV, D770_N771insSVD 돌연변이 세포주는 Dana-Farber Cancer Institute의 Pasi A Janne 실험실에서 구매하여 사용하였다. Ba/F3 D770delinsGY, H773_V774insH, Y764_V765insHH, V769_D770insASV, D770_N771insSVD 돌연변이 세포주는 10% FBS, 1% penicillin-streptomycin이 들어있는 RPMI에 puromycine 1 μg/ml 을 넣어 37℃, 5% CO 2 인큐베이터에 배양하였다.The activity measurement of the mutant Ba/F3 EGFR cell line for the compound of the present invention was tested as follows using the CellTiter-Glo system sold by Promega. CellTiter-Glo assay is a method to check cell viability by measuring ATP present in cells in the cell culture state. Ba/F3 EGFR D770delinsGY, H773_V774insH, Y764_V765insHH, V769_D770insASV, D770_N771insSVD mutant cell lines were purchased and used in Pasi A Janne laboratory of Dana-Farber Cancer Institute. Ba/F3 D770delinsGY, H773_V774insH, Y764_V765insHH, V769_D770insASV, D770_N771insSVD mutant cell lines were cultured in an incubator at 37° C., 5% CO 2 by adding 1 μg/ml of puromycine to RPMI containing 10% FBS and 1% penicillin-streptomycin.
화합물의 BaF3 cells overexpressing exon20 insertion mutations의 세포성장 억제능 효과 분석은 하기의 분석 반응 레시피에 따라 진행되었다.The analysis of the cell growth inhibitory effect of the compound's BaF3 cells overexpressing exon20 insertion mutations was performed according to the following analytical reaction recipe.
1500 cells/ 100 μL을 96 well 세포배양 plate에 계대하여 배양하고 24시간 후에 실시예 1로 표시되는 화합물을 0, 30, 100, 300, 1000 (nM)로 처리하였다. 72시간 동안 반응 후 화합물을 처리한 plate를 30분 동안 상온에 방치한 후 CellTiter Glo 2.0 reagent를 100 μL 더 처리하고 10분 동안 상온에서 shaking 하였다. 최종적으로 Luminometer 장비를 이용하여 luminescence 값을 측정하여 vehicle (control) 대비 화합물에서의 세포성장 억제능을 확인하였다. 5가지 화합물 농도에서 측정된 측정값을 Prism 프로그램(버전 5.01, Graphpad Software, Inc.)을 사용하여 분석하고 화합물의 세포성장 억제능 지표인 IC 50값을 산출하였다. 1500 cells/100 μL were cultured by passage to a 96 well cell culture plate, and after 24 hours, the compound represented by Example 1 was treated with 0, 30, 100, 300, 1000 (nM). After the reaction for 72 hours, the plate treated with the compound was left at room temperature for 30 minutes, and then 100 μL of CellTiter Glo 2.0 reagent was further treated and shaking at room temperature for 10 minutes. Finally, the luminescence value was measured using a luminometer to confirm the ability of the compound to inhibit cell growth compared to the vehicle (control). The measured values measured at the concentrations of the five compounds were analyzed using the Prism program (version 5.01, Graphpad Software, Inc.), and the IC 50 value, an indicator of the cell growth inhibitory ability of the compound, was calculated.
Ba/F3 EGFR V769_D770insASV IC 50(μM)Ba/F3 EGFR V769_D770insASV IC 50 (μM) Ba/F3 EGFR D770delinsGY IC 50(μM)Ba/F3 EGFR D770delinsGY IC 50 (μM) Ba/F3 EGFR H773_V774insH IC 50(μM)Ba/F3 EGFR H773_V774insH IC 50 (μM) Ba/F3 EGFR Y764_V765insHH IC 50(μM)Ba/F3 EGFR Y764_V765insHH IC 50 (μM) Ba/F3 EGFR D770_N771insSVD IC 50(μM)Ba/F3 EGFR D770_N771insSVD IC 50 (μM)
오시머티닙Osimertinib 0.1240.124 0.1590.159 0.1910.191 0.1360.136 0.1490.149
실시예 1Example 1 0.0260.026 0.0210.021 0.060.06 0.0290.029 0.0270.027
상기 표 5에 나타난 바와 같이,As shown in Table 5 above,
본 발명에 따른 실시예 1로 표시되는 화합물은 Ba/F3 세포주에서의 Ba/F3 EGFR exon20 insertion 돌연변이에 대하여 높은 세포성장 억제능을 나타내고, 이는 기존 EGFR TKI(tyrosine kinase inhibitor) 약물인 오시머티닙의 경우보다 우수한 것을 확인하였다.The compound represented by Example 1 according to the present invention exhibits high cell growth inhibitory ability against Ba/F3 EGFR exon20 insertion mutations in Ba/F3 cell lines, which is the case of osimertinib, an existing EGFR tyrosine kinase inhibitor (TKI) drug. It was confirmed that it is more excellent.
상기 결과를 통해, 본 발명에 따른 피리미딘 유도체 화합물은 EGFR 돌연변이를 효과적으로 억제할 수 있으므로, 암의 예방 또는 치료용 약학적 조성물로 유용하게 사용할 수 있음을 알 수 있다.Through the above results, it can be seen that the pyrimidine derivative compound according to the present invention can effectively inhibit the EGFR mutation, and thus can be usefully used as a pharmaceutical composition for preventing or treating cancer.
<제제예 1> 산제의 제조<Formulation Example 1> Preparation of powder
화학식 1로 표시되는 유도체 2g2g of derivatives represented by Formula 1
유당 1g1g lactose
상기의 성분을 혼합하고 기밀포에 충진하여 산제를 제조하였다.The above ingredients were mixed and filled in an airtight cloth to prepare a powder.
<제제예 2> 정제의 제조<Formulation Example 2> Preparation of tablets
화학식 1로 표시되는 유도체 100 ㎎100 mg of derivatives represented by Formula 1
옥수수전분 100 ㎎ Corn starch 100 mg
유당 100 ㎎100 mg lactose
스테아린산 마그네슘 2 ㎎2 mg of magnesium stearate
상기의 성분을 혼합한 후, 통상의 정제의 제조방법에 따라서 타정하여 정제를 제조하였다.After mixing the above ingredients, tablets were prepared by tableting according to a conventional tablet preparation method.
<제제예 3> 캡슐제의 제조<Formulation Example 3> Preparation of capsules
화학식 1로 표시되는 유도체 100 ㎎100 mg of derivatives represented by Formula 1
옥수수전분 100 ㎎ Corn starch 100 mg
유당 100 ㎎100 mg lactose
스테아린산 마그네슘 2 ㎎2 mg of magnesium stearate
상기의 성분을 혼합한 후, 통상의 캡슐제의 제조방법에 따라서 젤라틴 캡슐에 충전하여 캡슐제를 제조하였다.After mixing the above ingredients, a gelatin capsule was filled according to a conventional capsule preparation method to prepare a capsule.
<제제예 4> 주사제의 제조<Formulation Example 4> Preparation of injection
화학식 1로 표시되는 유도체 100 ㎎100 mg of derivatives represented by Formula 1
만니톨 180 ㎎Mannitol 180 mg
Na 2HPO 4ㆍ2H 2O 26 ㎎Na 2 HPO 4 ㆍ2H 2 O 26 mg
증류수 2974 ㎎Distilled water 2974 mg
통상적인 주사제의 제조방법에 따라, 상기 성분들을 제시된 함량으로 함유시켜 주사제를 제조하였다.According to a conventional method for preparing injections, injections were prepared by containing the above ingredients in the indicated amount.
<제제예 5> 건강식품의 제조<Formulation Example 5> Preparation of health food
화학식 1로 표시되는 유도체 500ng500 ng of derivatives represented by Formula 1
비타민 혼합물 적량The right amount of vitamin mixture
비타민 A 아세테이트 70mg 70mg vitamin A acetate
비타민 E 1.0mg1.0mg vitamin E
비타민 0.13mg0.13mg vitamin
비타민 B2 0.15mg0.15mg vitamin B2
비타민 B6 0.5mg0.5mg vitamin B6
비타민 B12 0.2mg0.2mg vitamin B12
비타민 C 10mg10mg vitamin C
비오틴 10mg10mg biotin
니코틴산아미드 1.7mg1.7 mg of nicotinic acid amide
엽산 50mg50mg folic acid
판토텐산 칼슘 0.5mg0.5mg calcium pantothenate
무기질 혼합물 적량Suitable amount of inorganic mixture
황산제1철 1.75mg1.75 mg ferrous sulfate
산화아연 0.82mgZinc oxide 0.82mg
탄산마그네슘 25.3mg25.3mg magnesium carbonate
제1인산칼륨 15mg15 mg of potassium monophosphate
제2인산칼슘 55mgDicalcium Phosphate 55mg
구연산칼륨 90mg90mg potassium citrate
탄산칼슘 100mg100mg calcium carbonate
염화마그네슘 24.8mgMagnesium chloride 24.8mg
상기의 비타민 및 미네랄 혼합물의 조성비는 비교적 건강식품에 적합한 성분을 바람직한 제제예로 혼합 조성하였지만, 그 배합비를 임의로 변형 실시하여도 무방하며, 통상의 건강식품 제조방법에 따라 상기의 성분을 혼합한 다음, 과립을 제조하고, 통상의 방법에 따라 건강식품 조성물 제조에 사용할 수 있다.As for the composition ratio of the vitamin and mineral mixture, the ingredients suitable for health food are mixed and formulated as a preferred formulation example, but the mixing ratio may be arbitrarily modified. , To prepare granules, and can be used for preparing a health food composition according to a conventional method.
<제제예 6> 건강음료의 제조<Formulation Example 6> Preparation of health drink
화학식 1로 표시되는 유도체 500ng500 ng of derivatives represented by Formula 1
구연산 1000mg1000mg citric acid
올리고당 100g100g oligosaccharide
매실농축액 2g2g plum concentrate
타우린 1g1 g taurine
정제수를 가하여 전체 900mlTotal 900ml with purified water
통상의 건강 음료 제조방법에 따라 상기의 성분을 혼합한 다음, 약 1시간 동안 85℃에서 교반 가열한 후, 만들어진 용액을 여과하여 멸균된 용기에 취득하여 밀봉 멸균한 뒤 냉장 보관한 다음 건강 음료 조성물 제조에 사용하였다.After mixing the above ingredients according to a conventional health drink manufacturing method, after stirring and heating at 85° C. for about 1 hour, the resulting solution is filtered and obtained in a sterilized container, sealed and sterilized, and then stored in a refrigerator. It was used in preparation.
상기 조성비는 비교적 기호 음료에 적합한 성분을 바람직한 실시예로 혼합 조성하였지만 수요계층이나, 수요국가, 사용용도 등 지역적, 민족적 기호 도에 따라서 그 배합비를 임의로 변형 실시하여도 무방하다.The composition ratio is a mixture of ingredients suitable for a relatively preferred beverage in a preferred embodiment, but the mixing ratio may be arbitrarily modified according to regional and ethnic preferences such as the demand class, the country of demand, and the purpose of use.

Claims (10)

  1. 하기 화학식 1로 표시되는 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염:A compound represented by the following Formula 1, an isomer thereof, or a pharmaceutically acceptable salt thereof:
    [화학식 1][Formula 1]
    Figure PCTKR2020010657-appb-img-000043
    Figure PCTKR2020010657-appb-img-000043
    상기 화학식 1에서,In Formula 1,
    R은 -OR a 또는 -NR b1R b2이고,R is -OR a or -NR b1 R b2 ,
    R a는 -(CH 2) m-NR b1R b2이고, 여기서 m은 1 내지 3이고,R a is -(CH 2 ) m -NR b1 R b2 , where m is 1 to 3,
    R b1 및 R b2는 각각 독립적으로 수소 또는 비치환된 또는 NR c1R c2로 치환된 직쇄 또는 분지쇄 C 1-6알킬이고, 또는, R b1 및 R b2는 이들이 결합된 질소와 함께 N, O 및 S로 이루어진 군에서 선택되는 하나 이상의 헤테로 원자를 하나 이상 포함하는 비치환된 또는 직쇄 또는 분지쇄 C 1-6알킬 또는 NR c1R c2로 치환된 5 내지 7 원자의 헤테로시클로알킬을 형성하고,R b1 and R b2 are each independently hydrogen or unsubstituted or straight or branched chain C 1-6 alkyl substituted with NR c1 R c2 , or R b1 and R b2 are N, O together with the nitrogen to which they are attached. And to form an unsubstituted or straight or branched chain C 1-6 alkyl containing one or more heteroatoms selected from the group consisting of S or a 5 to 7 membered heterocycloalkyl substituted with NR c1 R c2,
    R c1 및 R c2는 각각 독립적으로 수소 또는 직쇄 또는 분지쇄 C 1-6알킬이고, 또는, R c1 및 R c2는 이들이 결합된 질소와 함께 N, O 및 S로 이루어진 군에서 선택되는 하나 이상의 헤테로 원자를 하나 이상 포함하는 비치환된 또는 직쇄 또는 분지쇄 C 1-6알킬로 치환된 5 내지 7 원자의 헤테로시클로알킬을 형성한다.R c1 and R c2 are each independently hydrogen or straight or branched chain C 1-6 alkyl, or, R c1 and R c2 are selected from the group consisting of N, O and S together with the nitrogen to which they are attached. 5 to 7 membered heterocycloalkyl substituted with unsubstituted or straight or branched C 1-6 alkyl containing one or more atoms.
  2. 제1항에 있어서,The method of claim 1,
    R은
    Figure PCTKR2020010657-appb-img-000044
    ,
    Figure PCTKR2020010657-appb-img-000045
    ,
    Figure PCTKR2020010657-appb-img-000046
    ,
    Figure PCTKR2020010657-appb-img-000047
    ,
    Figure PCTKR2020010657-appb-img-000048
    ,
    Figure PCTKR2020010657-appb-img-000049
    ,
    Figure PCTKR2020010657-appb-img-000050
    ,
    Figure PCTKR2020010657-appb-img-000051
    또는
    Figure PCTKR2020010657-appb-img-000052
    인 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염.
    R is
    Figure PCTKR2020010657-appb-img-000044
    ,
    Figure PCTKR2020010657-appb-img-000045
    ,
    Figure PCTKR2020010657-appb-img-000046
    ,
    Figure PCTKR2020010657-appb-img-000047
    ,
    Figure PCTKR2020010657-appb-img-000048
    ,
    Figure PCTKR2020010657-appb-img-000049
    ,
    Figure PCTKR2020010657-appb-img-000050
    ,
    Figure PCTKR2020010657-appb-img-000051
    or
    Figure PCTKR2020010657-appb-img-000052
    Phosphorus compounds, isomers thereof, or pharmaceutically acceptable salts thereof.
  3. 제1항에 있어서,The method of claim 1,
    상기 화학식 1로 표시되는 화합물은 하기 화합물 군으로부터 선택되는 어느 하나인 것인 화합물, 이의 이성질체 또는 이의 약학적으로 허용가능한 염:The compound represented by Formula 1 is any one selected from the following compound group, an isomer thereof, or a pharmaceutically acceptable salt thereof:
    <1> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-2-((2-(디메틸아미노)에틸)(메틸)아미노)-4-메톡시페닐)아크릴아미드;<1> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-((2-(dimethylamino) Ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide;
    <2> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-2-(4-(디메틸아미노)피페리딘-1-일)-4-메톡시페닐)아크릴아미드;<2> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-(4-(dimethylamino)pi Peridin-1-yl)-4-methoxyphenyl)acrylamide;
    <3> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(4-(4-메틸피페라진-1-일)피페리딘-1-일)페닐)아크릴아미드;<3> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-(4- (4-methylpiperazin-1-yl)piperidin-1-yl)phenyl)acrylamide;
    <4> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-모폴리노페닐)아크릴아미드;<4> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-morpholino Phenyl)acrylamide;
    <5> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(4-메틸피페라진-1-일)페닐)아크릴아미드;<5> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-(4- Methylpiperazin-1-yl)phenyl)acrylamide;
    <6> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-2-(2-(디메틸아미노)에톡시)-4-메톡시페닐)아크릴아미드;<6> to N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-2-(2-(dimethylamino) Oxy)-4-methoxyphenyl)acrylamide;
    <7> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(메틸(2-모폴리노에틸)아미노)페닐)아크릴아미드;<7> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-(methyl( 2-morpholinoethyl)amino)phenyl)acrylamide;
    <8> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미노)피리미딘-2-일)아미노)-4-메톡시-2-(메틸(2-(4-메틸피페라진-1-일)에틸)아미노)페닐)아크릴아미드; 및<8> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxy-2-(methyl( 2-(4-methylpiperazin-1-yl)ethyl)amino)phenyl)acrylamide; And
    <9> N-(5-((5-클로로-4-((2-(메틸설폰아미도)페닐)아미도)피리미딘-2-일)아미노)-4-메톡시-2-(2-모폴리노에톡시)페닐)아크릴아미드.<9> N-(5-((5-chloro-4-((2-(methylsulfonamido)phenyl)amido)pyrimidin-2-yl)amino)-4-methoxy-2-(2 -Morpholinoethoxy)phenyl)acrylamide.
  4. 하기 반응식 1에 나타낸 바와 같이,As shown in Scheme 1 below,
    화학식 2로 표시되는 화합물과 화학식 3으로 표시되는 화합물을 반응시켜 화학식 1로 표시되는 화합물을 제조하는 단계를 포함하는 제1항의 화학식 1로 표시되는 화합물의 제조방법:A method for preparing a compound represented by Formula 1 of claim 1 comprising the step of preparing a compound represented by Formula 1 by reacting a compound represented by Formula 2 with a compound represented by Formula 3:
    [반응식 1][Scheme 1]
    Figure PCTKR2020010657-appb-img-000053
    Figure PCTKR2020010657-appb-img-000053
    상기 반응식 1에서 R은 제1항의 화학식 1에서 정의한 바와 같다.In Reaction Scheme 1, R is as defined in Formula 1 of claim 1.
  5. 제1항의 화학식 1로 표시되는 화합물을 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물.A pharmaceutical composition for preventing or treating cancer containing the compound represented by Formula 1 of claim 1 as an active ingredient.
  6. 제5항에 있어서,The method of claim 5,
    상기 화합물은 EGFR(epidermal growth factor receptor) 돌연변이를 억제하는 것인 약학적 조성물.The pharmaceutical composition that the compound inhibits EGFR (epidermal growth factor receptor) mutation.
  7. 제6항에 있어서,The method of claim 6,
    상기 EGFR 돌연변이는 EGFR del19, EGFR L858R, EGFR del19/T790M, EGFR L858R/T790M, EGFR del19/T790M/C797S, EGFR L858R/T790M/C797S, EGFR D770delinsGY, EGFR H773_V774insH, EGFR Y764_V765insHH, EGFR D770insASV 및 EGFR D770_N771insSVD로 이루어진 군으로부터 선택되는 하나 이상인 것을 특징으로 하는 약학적 조성물.The EGFR mutations are EGFR del19, EGFR L858R, EGFR del19/T790M, EGFR L858R/T790M, EGFR del19/T790M/C797S, EGFR L858R/T790M/C797S, EGFR D770delinsGY, EGFR D770AS H773_V774insVinsH, EGFR del19/T790M, EGFR D770ASH773_V774insH, EGFR D770ASH773, and EGFRVinsD770AS_N, EGFRVinsD770AS Pharmaceutical composition, characterized in that at least one selected from the group.
  8. 제5항에 있어서,The method of claim 5,
    상기 암은 가성점액종, 간내 담도암, 간모세포종, 간암, 갑상선암, 결장암, 고환암, 골수이형성증후군, 교모세포종, 구강암, 구순암, 균상식육종, 급성골수성백혈병, 급성림프구성백혈병, 기저세포암, 난소상피암, 난소생식세포암, 남성유방암, 뇌암, 뇌하수체선종, 다발성골수종, 담낭암, 담도암, 대장암, 만성골수성백혈병, 만성림프구백혈병, 망막모세포종, 맥락막흑색종, 바터팽대부암, 방광암, 복막암, 부갑상선암, 부신암, 비부비동암, 비소세포폐암, 설암, 성상세포종, 소세포폐암, 소아뇌암, 소아림프종, 소아백혈병, 소장암, 수막종, 식도암, 신경교종, 신우암, 신장암, 심장암, 십이지장암, 악성 연부조직 암, 악성골암, 악성림프종, 악성중피종, 악성흑색종, 안암, 외음부암, 요관암, 요도암, 원발부위불명암, 위림프종, 위암, 위유암종, 위장관간질암, 윌름스암, 유방암, 육종, 음경암, 인두암, 임신융모질환, 자궁경부암, 자궁내막암, 자궁육종, 전립선암, 전이성 골암, 전이성뇌암, 종격동암, 직장암, 직장유암종, 질암, 척수암, 청신경초종, 췌장암, 침샘암, 카포시 육종, 파제트병, 편도암, 편평상피세포암, 폐선암, 폐암, 폐편평상피세포암, 피부암, 항문암, 횡문근육종, 후두암, 흉막암, 혈액암 및 흉선암으로 이루어진 군으로부터 선택되는 1종 이상인 것을 특징으로 하는 약학적 조성물.The cancer is pseudomyxoma, intrahepatic biliary tract cancer, hepatoblastoma, liver cancer, thyroid cancer, colon cancer, testicular cancer, myelodysplastic syndrome, glioblastoma, oral cancer, cleft lip cancer, mycosis fungoides, acute myelogenous leukemia, acute lymphocytic leukemia, basal cell cancer, Ovarian epithelial cancer, ovarian germ cell carcinoma, male breast cancer, brain cancer, pituitary adenoma, multiple myeloma, gallbladder cancer, biliary tract cancer, colon cancer, chronic myelogenous leukemia, chronic lymphocytic leukemia, retinoblastoma, choroidal melanoma, barter bulge cancer, bladder cancer, peritoneal cancer , Parathyroid cancer, adrenal cancer, non-sinus cancer, non-small cell lung cancer, tongue cancer, astrocytoma, small cell lung cancer, pediatric brain cancer, pediatric lymphoma, pediatric leukemia, small intestine cancer, meningioma, esophageal cancer, glioma, renal cancer, kidney cancer, heart cancer, Duodenal cancer, malignant soft tissue cancer, malignant bone cancer, malignant lymphoma, malignant mesothelioma, malignant melanoma, eye cancer, vulvar cancer, ureteral cancer, urethral cancer, primary site unknown cancer, gastric lymphoma, gastric cancer, gastric carcinoma, gastrointestinal interstitial cancer, Wilm Cancer, breast cancer, sarcoma, penile cancer, pharyngeal cancer, gestational chorionic disease, cervical cancer, endometrial cancer, uterine sarcoma, prostate cancer, metastatic bone cancer, metastatic brain cancer, mediastinal cancer, rectal cancer, rectal carcinoma, vaginal cancer, spinal cord cancer, auditory schwannoma , Pancreatic cancer, salivary gland cancer, Kaposi's sarcoma, Paget's disease, tonsil cancer, squamous cell carcinoma, lung adenocarcinoma, lung cancer, lung squamous cell carcinoma, skin cancer, anal cancer, rhabdomyosarcoma, laryngeal cancer, pleural cancer, hematologic cancer and thymic cancer Pharmaceutical composition, characterized in that at least one selected from the group consisting of.
  9. 제1항의 화학식 1로 표시되는 화합물을 유효성분으로 함유하는 암의 예방 또는 개선용 건강기능식품.Health functional food for preventing or improving cancer containing the compound represented by the formula 1 of claim 1 as an active ingredient.
  10. 제1항의 화학식 1로 표시되는 화합물을 유효성분으로 함유하는 EGFR 돌연변이 관련 질환의 예방 또는 치료용 약학적 조성물.A pharmaceutical composition for preventing or treating diseases related to EGFR mutations containing the compound represented by Formula 1 of claim 1 as an active ingredient.
PCT/KR2020/010657 2019-10-18 2020-08-12 Pyrimidine derivative, method of preparing same, and pharmaceutical composition for preventing or treating cancer, comprising same as effective component WO2021075691A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2019-0130157 2019-10-18
KR1020190130157A KR102383200B1 (en) 2019-10-18 2019-10-18 pyrimidine derivatives, preparation method thereof, and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient

Publications (1)

Publication Number Publication Date
WO2021075691A1 true WO2021075691A1 (en) 2021-04-22

Family

ID=75537846

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2020/010657 WO2021075691A1 (en) 2019-10-18 2020-08-12 Pyrimidine derivative, method of preparing same, and pharmaceutical composition for preventing or treating cancer, comprising same as effective component

Country Status (2)

Country Link
KR (1) KR102383200B1 (en)
WO (1) WO2021075691A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3226006A1 (en) * 2021-07-13 2023-01-19 Korea Research Institute Of Chemical Technology Novel pyrimidine-2,4-diamine derivatives, preparation method therefor, and pharmaceutical composition containing same as active ingredient for prevention or treatment of cancer
KR102611488B1 (en) * 2021-08-05 2023-12-11 한국화학연구원 Pyrimidine derivatives, preparation method thereof, and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient
CA3228333A1 (en) * 2021-08-05 2023-02-09 Korea Research Institute Of Chemical Technology Pyrimidine derivative, method for preparing same, and pharmaceutical composition for preventing or treating cancer comprising same as active ingredient
KR20230054567A (en) * 2021-10-15 2023-04-25 제이투에이치바이오텍 (주) Compounds inhibiting ALK and/or EGFR mutation kinases and medical use thereof
KR102524856B1 (en) * 2022-06-23 2023-04-24 주식회사 카나프테라퓨틱스 Novel compound for inhibiting epidermal growth factor receptor tyrosine kinase and uses thereof
WO2023163527A1 (en) * 2022-02-23 2023-08-31 주식회사 카나프테라퓨틱스 Novel compound having inhibitory activity against epidermal growth factor receptor tyrosine kinase, and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105503827A (en) * 2014-10-11 2016-04-20 上海翰森生物医药科技有限公司 EGFR (Epidermal growth factor receptor) inhibitor and preparation method and use thereof
CN106187915A (en) * 2015-05-27 2016-12-07 上海翰森生物医药科技有限公司 There is inhibitor of ALK Yu EGFR double activity and its preparation method and application
WO2017101803A1 (en) * 2015-12-15 2017-06-22 合肥中科普瑞昇生物医药科技有限公司 Novel egfr and alk dual inhibitor
KR20170118681A (en) * 2014-10-11 2017-10-25 상하이 한서 바이오메디컬 컴퍼니 리미티드 Egfr inhibitor, and preparation and application thereof
KR20190108079A (en) * 2018-03-13 2019-09-23 보로노이바이오 주식회사 2,4,5-substituted pyrimidine derivatives, preparation method thereof, and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105503827A (en) * 2014-10-11 2016-04-20 上海翰森生物医药科技有限公司 EGFR (Epidermal growth factor receptor) inhibitor and preparation method and use thereof
KR20170118681A (en) * 2014-10-11 2017-10-25 상하이 한서 바이오메디컬 컴퍼니 리미티드 Egfr inhibitor, and preparation and application thereof
CN106187915A (en) * 2015-05-27 2016-12-07 上海翰森生物医药科技有限公司 There is inhibitor of ALK Yu EGFR double activity and its preparation method and application
WO2017101803A1 (en) * 2015-12-15 2017-06-22 合肥中科普瑞昇生物医药科技有限公司 Novel egfr and alk dual inhibitor
KR20190108079A (en) * 2018-03-13 2019-09-23 보로노이바이오 주식회사 2,4,5-substituted pyrimidine derivatives, preparation method thereof, and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient

Also Published As

Publication number Publication date
KR102383200B1 (en) 2022-04-06
KR20210046914A (en) 2021-04-29

Similar Documents

Publication Publication Date Title
WO2021075691A1 (en) Pyrimidine derivative, method of preparing same, and pharmaceutical composition for preventing or treating cancer, comprising same as effective component
WO2018230934A1 (en) N2,n4-diphenylpyrimidine-2,4-diamine derivative, method for preparing same, and pharmaceutical composition containing same as active ingredient for prevention or treatment of cancer
WO2019112344A1 (en) Novel pyrimidine derivative having effect of inhibiting cancer cell growth and pharmaceutical composition containing same
WO2018139903A1 (en) Pyrimidine compound and pharmaceutical use thereof
WO2020171499A1 (en) Novel pyrido[3,4-d]pyrimidin-8-one derivative having protein kinase inhibitory activity, and pharmaceutical composition for preventing, alleviating, or treating cancer, comprising same
WO2018012907A1 (en) Novel quinazolinone derivatives inhibiting pi3k and pharmaceutical composition containing same
WO2009125923A2 (en) Novel indol carboxylic acid bispyridyl carboxamide derivatives, pharmaceutically acceptable salt thereof, preparation method and composition containing the same as an active ingredient
WO2018088749A1 (en) Novel pyrimidine compound, method for preparing same, and pharmaceutical composition containing same as active ingredient for preventing or treating cancer and inflammatory diseases
WO2017142325A1 (en) Novel 2,3,5-substituted thiophene compound as protein kinase inhibitor
WO2018155947A1 (en) Pharmaceutical composition comprising compound capable of penetrating blood-brain barrier as effective ingredient for preventing or treating brain cancer
WO2018004258A1 (en) Novel heterocyclic derivative compound and use thereof
WO2020149715A1 (en) Pyrrolopyridine derivative and use thereof in prevention and treatment of protein kinase-related disease
WO2021145521A1 (en) Pyrido[3,4-d]pyrimidine derivative and therapeutic pharmaceutic composition comprising same
WO2021125803A1 (en) Novel pyrimidin derivative and use thereof
WO2018004213A1 (en) Novel compound having smo inhibitory activity and composition comprising same as active ingredient for preventing or treating cancer
WO2014109530A1 (en) 2-(phenylethynyl)thieno[3,4-b]pyrazine derivative and pharmaceutical composition comprising same for preventing or treating cancer
WO2020036386A1 (en) Isoindolin-1-on derivative, method for preparing same, and pharmaceutical composition comprising same as effective component for preventing or treating cancer
AU2017256488B2 (en) Quinazoline derivative or its salt and pharmaceutical composition comprising the same
WO2020040582A1 (en) Novel benzopyran derivative or pharmaceutically acceptable salt thereof and pharmaceutical composition comprising same as effective ingredient
WO2018056621A1 (en) Novel imidazolyl pyrimidine derivative, method for preparing same, and pharmaceutical composition comprising same as active ingredient for prevention or treatment of cancer
WO2018021826A1 (en) Novel pyrimidine-2,4-diamine derivative and pharmaceutical composition for prevention or treatment of cancer containing same as active ingredient
WO2021096112A1 (en) Pyrrolopyrimidine, pyrrolopyridine and indazole compound derivative, and therapeutic pharmaceutical composition containing same
WO2016093554A2 (en) Novel 4-(aryl)-n-(2-alkoxythieno[3,2-b]pyrazin-3-yl)-piperazine-1-carboxamide derivative, and antiproliferative effect thereof
WO2021101268A1 (en) Benzamide derivative, preparation method therefor, and pharmaceutical composition comprising same as active ingredient for prevention or treatment of cancer
WO2022103149A1 (en) Novel carbazole derivative and pharmaceutical composition for prevention or treatment of cancer comprising same as active ingredient

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: 20876928

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: 20876928

Country of ref document: EP

Kind code of ref document: A1