WO2023017937A1 - Novel inhibitor for multiple protein kinases - Google Patents

Novel inhibitor for multiple protein kinases Download PDF

Info

Publication number
WO2023017937A1
WO2023017937A1 PCT/KR2022/003249 KR2022003249W WO2023017937A1 WO 2023017937 A1 WO2023017937 A1 WO 2023017937A1 KR 2022003249 W KR2022003249 W KR 2022003249W WO 2023017937 A1 WO2023017937 A1 WO 2023017937A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
kmu
indolin
methylene
pyrazin
Prior art date
Application number
PCT/KR2022/003249
Other languages
French (fr)
Korean (ko)
Inventor
김신
이진호
홍빅터석봉
백혜숙
정승익
추현성
Original Assignee
계명대학교 산학협력단
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 계명대학교 산학협력단 filed Critical 계명대학교 산학협력단
Publication of WO2023017937A1 publication Critical patent/WO2023017937A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A23V2200/306Foods, ingredients or supplements having a functional effect on health having an effect on bone mass, e.g. osteoporosis prevention
    • 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
    • A23V2250/00Food ingredients
    • A23V2250/30Other Organic compounds

Definitions

  • the present invention relates to novel multiple protein kinase inhibitors, and to novel indolin-2-one derivatives exhibiting excellent anti-inflammatory activity.
  • Inflammation is an important and fundamental immune system response of the body that protects the host from harmful stimuli and maintains tissue homeostasis.
  • An effective inflammatory response allows you to survive infection or injury, but excessive or persistent inflammation can lead to various pathological conditions such as asthma, cancer, chronic pain, gout, mental disorders, nervous breakdown, psoriasis, rheumatoid arthritis and vasculitis. .
  • TLRs Toll-like receptors
  • LPS Lipopolysaccharide
  • NF- ⁇ B transcription factor kappa-light-chain-enhancer of activated B cells
  • cytokines include interleukin (IL)-1 ⁇ , IL-6, and tumor necrosis factor- ⁇ (TNF- ⁇ ).
  • IL interleukin
  • TNF- ⁇ tumor necrosis factor- ⁇
  • the inflammasome a complex of inflammatory molecules, is an innate immune response to pathogenic infections and plays an important role in the formation of proinflammatory cytokines.
  • NLRP3 pyrin domain-containing protein 3
  • -like protein containing a CARD (ASC) adapter molecule procaspase-1 and NIMA-related kinase 7.
  • Protein kinase (protein kinase) is responsible for phosphorylation of proteins, which plays an important role in intracellular signal transduction in inflammation. Protein kinases are very important drug targets in drug development for the treatment of inflammatory diseases. For example, tofacitinib, a potent Janus-associated kinase (JAK) 1 and JAK 3 antagonist developed by Pfizer, reduces the signs and symptoms of rheumatoid arthritis (RA) and is improved bodily functions. Moreover, the p38 kinase inhibitor semapimod improved clinical outcomes in patients with Crohn's disease.
  • TOK Janus-associated kinase
  • JAK 3 antagonist developed by Pfizer
  • BAY 61-3606 a spleen tyrosine kinase inhibitor, has been approved by the Food and Drug Administration (FDA) as an indication for allergic asthma patients.
  • FDA Food and Drug Administration
  • various Bruton's tyrosine kinase (BTK) inhibitors are in clinical trials. As such, the need for development of promising protein kinase inhibitors is increasing.
  • An object of the present invention is to provide novel compounds having excellent protein kinase inhibitory activity.
  • Another object of the present invention is to provide a composition for treating inflammatory diseases comprising the compound.
  • the present invention provides a compound represented by Formula 1 below or a pharmaceutically acceptable salt thereof.
  • R 1 is selected from hydrogen, hydroxy, halogen, (C1-C4) alkyl, (C3-C10) cycloalkyl, or adamantanyl
  • R 2 is imidazolyl , pyrazolyl, pyrrolyl or pyrrolidinyl.
  • the present invention provides a pharmaceutical composition for preventing or treating inflammatory diseases containing the compound or a pharmaceutically acceptable salt thereof as an active ingredient.
  • the present invention provides a health functional food composition for preventing or improving inflammatory diseases containing the compound or a pharmaceutically acceptable salt thereof as an active ingredient.
  • novel compound according to the present invention strongly inhibits the activity of various protein kinases related to inflammation, it can be used as a multi-protein kinase inhibitor.
  • the compound since the compound regulates cytokine and NLRP3 inflammasome activity and has a superior anti-inflammatory effect compared to other anti-inflammatory agents, it can be used for the development of therapeutic agents for inflammatory diseases.
  • FIG. 1 is a schematic diagram showing a synthesis process of a novel KMU-series compound synthesized according to an embodiment of the present invention.
  • Figure 2 confirms the inhibitory effect of KMU-series compounds on LPS-induced up-regulated pro-inflammatory cytokines in THP-1 cells according to an experimental example of the present invention (*p ⁇ 0.05, **p ⁇ 0.01, #p ⁇ 0.001 compared to LPS alone)
  • Figure 3 confirms the inhibitory effect of KMU-11342 on RANKL-induced osteoclastogenesis in RAW 264.7 cells (**p ⁇ 0.01, #p ⁇ 0.001 compared to RANKL alone).
  • Figure 4 is a novel compound synthesized according to an embodiment of the present invention, KMU-11170 (hereinafter referred to as "KMU-1170" in the drawings and experimental results) and LPS-induced iNOS and COX-induced iNOS and COX- in the human monocyte cell line THP-1.
  • (A) is a schematic diagram showing the synthesis process of KMU-1170
  • (B) confirms the cytotoxic effect of KMU-1170 in THP-1 cells through XTT analysis
  • (C) to (I) are graphs confirming changes in mRNA or protein expression of iNOS, COX-1, and COX-2 using western blotting and Image-J software ( ⁇ p ⁇ 0.05, *p ⁇ 0.01, and #p ⁇ 0.001 compared to LPS).
  • Figure 5 confirms the inhibitory effect of KMU-1170 on LPS-induced up-regulated pro-inflammatory cytokines in THP-1 cells according to an experimental example of the present invention.
  • the protein expression levels of pro-IL-1 ⁇ , TNF- ⁇ , IL-6, and ⁇ -actin were measured by blotting, (B) using Image-J software, pro-IL-1 ⁇ , TNF- ⁇ , And analyzing the relative optical density of the IL-6 band, (C) extracting total RNA and confirming the mRNA expression levels of IL-1 ⁇ , TNF- ⁇ , and IL-6 by RT-PCR, (D ) is the analysis of the relative optical density of IL-1 ⁇ , TNF- ⁇ , and IL-6 bands using Image-J software, (E) to (G) are total RNA extracted and IL-1 ⁇ by real-time PCR , TNF- ⁇ , and IL-6 mRNA expression levels were confirmed ( ⁇ p ⁇ 0.05, *p ⁇ 0.01, and #p ⁇ 0.001 compared to LPS).
  • Figure 6 confirms the effect of KMU-1170 on LPS-induced TLR4 signaling-related proteins in THP-1 cells according to an experimental example of the present invention.
  • MyD88, and ⁇ -actin protein expression levels were measured
  • (B) analyzed the relative optical density of TLR4 and MyD88 bands using Image-J software
  • (C) was p-TAK1 by western blot
  • TAK1, and ⁇ -actin protein expression levels were measured
  • D analyzed the relative optical density of the p-TAK1 band using Image-J software
  • E was p-ERK by Western blot , ERK, p-JNK, JNK, p-p38, p38, and measuring the protein expression levels of ⁇ -actin
  • the relative optical density of the p38 band was analyzed (#p ⁇ 0.001 compared to LPS).
  • Figure 7 confirms the effect of KMU-1170 on LPS-induced phosphorylation of IKK ⁇ / ⁇ and NF- ⁇ B and nuclear translocation of NF- ⁇ B in THP-1 cells according to an experimental example of the present invention
  • A The protein expression levels of p-IKK ⁇ / ⁇ , IKK ⁇ / ⁇ , p-NF- ⁇ B p65, NF- ⁇ B p65, and ⁇ -actin were measured by Western blotting
  • (B) was obtained using Image-J software.
  • Figure 8 relates to the inhibitory effect of KMU-1170 on LPS-induced NLRP3 inflammasome activation and anti-inflammatory potential of KMU-1170 in THP-1 cells according to an experimental example of the present invention
  • A is for cells Lysate and medium supernatant were separated, and the protein expression levels of pro-IL-1 ⁇ and IL-1 ⁇ in the supernatant and NLRP3, ASC, pro-caspase-1, pro- Measuring the protein expression levels of IL-1 ⁇ and ⁇ -actin
  • (B) is the relative ratio of NLRP3, ASC, pro-caspase-1, and pro-IL-1 ⁇ bands in the lysate using Image-J software The optical density was analyzed,
  • C the protein expression levels of pro-IL-1 ⁇ and ⁇ -actin were measured in the lysate by Western blot,
  • D was pro-IL-1 ⁇ using Image-J software.
  • the relative optical density of the 1 ⁇ band was analyzed ( ⁇ p ⁇ 0.05, *p ⁇ 0.0
  • Figure 9 confirms the inhibitory effect of KMU-1170 on the LPS-induced inflammatory response in primary human osteoarthritis (OA) fibroblast-like synovial cells (FLS) according to an experimental example of the present invention
  • A) to (C) is to extract total RNA and measure IL-1 ⁇ , TNF- ⁇ , and IL-6 mRNA expression levels by real-time PCR
  • D) to (F) isolate whole cell lysates and use Western blot, respectively pro - protein expression levels of IL-1 ⁇ , TNF- ⁇ , IL-6, and ⁇ -actin (D); protein expression levels of iNOS, COX-2, and ⁇ -actin (E);
  • the protein expression levels (F) of p-IKK ⁇ / ⁇ , p-NF- ⁇ B p65, and ⁇ -actin; were measured (*p ⁇ 0.01 and #p ⁇ 0.001 compared to LPS).
  • Figure 10 is a schematic diagram of the anti-inflammatory effect mechanism of KMU-1170 confirmed according to an experimental example of the present invention.
  • Protein kinase is an enzyme that phosphorylates a protein molecule by attaching a phosphoryl group to a serine, threonine or tyrosine residue, and binding to ATP is essential for kinase activity.
  • Recently, reports on the involvement of protein kinases in various human diseases including cancer and inflammatory diseases have led to the development of various protein kinase inhibitors.
  • the present inventors synthesized KMU-series compounds, which are indolin-2-one derivatives, as novel multi-target protein kinase inhibitors, and synthesized human monocyte cell line THP-1 and primary human osteoarthritis fibroblast-like synovial membrane. By using the cell to confirm its anti-inflammatory mechanism, the present invention was completed.
  • the present invention provides a compound represented by Formula 1 below or a pharmaceutically acceptable salt thereof.
  • R 1 may be selected from hydrogen, hydroxy, halogen, (C1-C4) alkyl, (C3-C10) cycloalkyl, or adamantanyl
  • R 2 is imidazolyl ( imidazolyl), pyrazolyl, pyrrolyl, and pyrrolidinyl.
  • R 1 may be selected from (C3-C8) cycloalkyl or adamantanyl
  • R 2 may be selected from imidazolyl or pyrrolyl. , but is not limited thereto.
  • the compound or salt thereof is (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopropylamino)pyrazin-2-yl)indoline- 2-one [(Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopropylamino)pyrazin-2-yl)indolin-2-one] (4a; KMU-11170) , (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopentylamino)pyrazin-2-yl)indolin-2-one [ (Z)-3- ((1H-imidazol-5-yl)methylene)-5-(6-(cyclopentylamino)pyrazin-2-yl)indolin-2-one] ( 4b; KMU-11342), (Z)-3-((1H -imidazol-5-yl)methylene
  • the compound may be used in the form of a pharmaceutically acceptable salt within a range having the same efficacy.
  • pharmaceutically acceptable means a salt that is not toxic to cells or humans exposed to the composition and has a safety and efficacy profile suitable for administration to humans.
  • the salt may be used in the form of either a pharmaceutically acceptable basic salt or acid salt.
  • the basic salt can be used in the form of any one of an organic basic salt and an inorganic basic salt, and includes sodium salt, potassium salt, calcium salt, lithium salt, magnesium salt, cesium salt, aminium salt, ammonium salt, triethylaminium salt, and pyrol. It may be selected from the group consisting of dinium salts.
  • an acid addition salt formed by a free acid is useful.
  • Inorganic acids and organic acids can be used as free acids, and hydrochloric acid, hydrobromic acid, sulfuric acid, sulfurous acid, phosphoric acid, double phosphoric acid, and nitric acid can be used as inorganic acids, and citric acid, acetic acid, maleic acid, malic acid, and fumaric acid can be used as organic acids.
  • glucoic acid methanesulfonic acid, benzenesulfonic acid, camphorsulfonic acid, oxalic acid, malonic acid, glutaric acid, acetic acid, glycolic acid, succinic acid, tartaric acid, 4-toluenesulfonic acid, galacturonic acid, embonic acid, Glutamic acid, citric acid, aspartic acid, stearic acid, etc.
  • salts formed using various inorganic acids and organic acids commonly used in the art may all be included.
  • the compound may include all salts, hydrates, solvates, derivatives, and the like that can be prepared by conventional methods, as well as the salts.
  • the addition salt can be prepared by a conventional method, and is dissolved in a water-miscible organic solvent such as acetone, methanol, ethanol, or acetonitrile, and an excess organic base is added or an aqueous solution of an inorganic base is added, followed by precipitation or crystallization. can be manufactured by Alternatively, the solvent or excess base may be evaporated from the mixture and then dried to obtain an addition salt, or the precipitated salt may be suction filtered.
  • a water-miscible organic solvent such as acetone, methanol, ethanol, or acetonitrile
  • the compound or salt thereof may inhibit various inflammation-related protein kinases.
  • the compound or salt thereof is MAPK (mitogen-activated protein kinase), TAK1 (TGF-beta activated kinase 1), Lck (lymphocyte-specific protein tyrosine kinase), TYK2 (non-receptor tyrosine-protein kinase 2) , JAK3 (janus kinase 3), and Txk (tyrosine-protein kinase) may inhibit one or more protein kinases selected from the group consisting of, but is not limited thereto.
  • MAPK mitogen-activated protein kinase
  • TAK1 TGF-beta activated kinase 1
  • Lck lymphocyte-specific protein tyrosine kinase
  • TYK2 non-receptor tyrosine-protein kinase 2
  • JAK3 janus kinase 3
  • Txk tyrosine-protein kinase
  • the compound is related to various cellular processes including apoptosis, differentiation, immunity, inflammation and survival in THP-1 cells, particularly related to LPS-TLR4-TAK1 signaling of pro-inflammatory cytokines. It was confirmed that TAK1 inhibits LPS-induced phosphorylation of ERK and JNK.
  • the compound or salt thereof may inhibit the activity of NLRP3 inflammasome and inhibit LPS-induced up-regulation of various cytokines, and thus may have excellent anti-inflammatory activity.
  • the compound inhibits iNOS belonging to the family of catalytic enzymes necessary for the production of nitric oxide (NO), an important mediator of the inflammatory response, and various diseases such as inflammatory diseases, neoplastic diseases and Alzheimer's disease.
  • iNOS belonging to the family of catalytic enzymes necessary for the production of nitric oxide (NO)
  • various diseases such as inflammatory diseases, neoplastic diseases and Alzheimer's disease.
  • LPS-induced IKK ⁇ / ⁇ and NF- ⁇ B p65 associated with NF- ⁇ B factor a key mediator of the immune system that selectively inhibits COX-2 associated with human diseases and regulates key factors such as cytokines and inflammasomes in the inflammatory response It was confirmed that phosphorylation of , nuclear translocation of NF- ⁇ B p65, and upregulation of IL-1 ⁇ , TNF- ⁇ , and IL-6 were inhibited.
  • the compound did not inhibit the LPS-mediated upregulation of TLR4 and MyD88 in THP-1 cells, and these results suggest that the compound has an anti-inflammatory effect downstream of TLR4/MyD88 in the LPS-mediated inflammatory response of THP-1 cells. shows that it has
  • COX-2 inhibitors celecoxib
  • BTK inhibitors ibrutinib
  • JAK inhibitors ruxolitinib
  • methotrexate was significantly superior to other drugs. It was confirmed that it exhibited an excellent anti-inflammatory effect.
  • the compound or salt thereof may be used as a composition for preventing or treating inflammatory diseases.
  • the present invention provides a pharmaceutical composition for preventing or treating inflammatory diseases containing the compound or a pharmaceutically acceptable salt thereof as an active ingredient.
  • the inflammatory disease may be any one selected from the group consisting of osteoarthritis, rheumatoid arthritis, osteoporosis, Paget's disease of bone, and ankylosing spondylitis, but is not limited thereto.
  • the compound inhibits the LPS-induced inflammatory response in osteoarthritis FLS, which plays an important role in the progression of osteoarthritis characterized by continuous synovial inflammation and progressive cartilage degradation. Osteoarthritis can be prevented, treated or ameliorated.
  • the pharmaceutical composition according to the present invention can be prepared according to conventional methods in the pharmaceutical field.
  • the pharmaceutical composition may be formulated with an appropriate pharmaceutically acceptable carrier according to the formulation and, if necessary, may further contain excipients, diluents, dispersants, emulsifiers, buffers, stabilizers, binders, disintegrants, solvents, and the like. there is.
  • the appropriate carrier and the like do not inhibit the activity and characteristics of the compound according to the present invention or a pharmaceutically acceptable salt thereof, and may be selected differently depending on the dosage form and dosage form.
  • Examples of carriers, excipients, and diluents that may be included in the pharmaceutical composition include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxy benzoate, propylhydroxy benzoate, talc, magnesium stearate and mineral oil.
  • diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants.
  • the pharmaceutical composition can be applied in any dosage form, and specifically, according to conventional methods, oral dosage forms such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, external preparations, suppositories and sterile injection solutions. It may be formulated into a form and used, preferably formulated into a unit dosage form suitable for oral administration.
  • oral dosage forms such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, external preparations, suppositories and sterile injection solutions. It may be formulated into a form and used, preferably formulated into a unit dosage form suitable for oral administration.
  • the solid dosage form of the oral dosage form is in the form of tablets, pills, powders, granules, capsules, etc., and includes at least one excipient such as starch, calcium carbonate, sucrose, lactose, sorbitol, and mannitol. , cellulose, gelatin, etc. may be mixed, and lubricants such as magnesium stearate and talc may be included in addition to simple excipients.
  • a liquid carrier such as fatty oil may be further included in addition to the above-mentioned materials.
  • liquid formulations include suspensions, solutions for internal use, emulsions, syrups, etc.
  • various excipients such as wetting agents, sweeteners, aromatics, and preservatives may be included. there is.
  • the parenteral formulation may include a sterilized aqueous solution, a non-aqueous solvent, a suspension, an emulsion, an injection, a lyophilized formulation, a suppository, and the like.
  • Propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate may be used as non-aqueous solvents and suspending agents.
  • As a base for the suppository witepsol, macrogol, Tween 61, cacao butter, laurin fat, glycerogeratin, and the like may be used. It is not limited thereto, and all suitable agents well known in the art may be used.
  • an antioxidant may be further added to the pharmaceutical composition to enhance therapeutic efficacy.
  • the antioxidants include thiamin (vitamin B1), riboflavin (vitamin B2), niacin (vitamin B3), pantothenic acid (vitamin B5), pyridoxine (vitamin B6) and cobalamin (cobalamin, Vitamin B group compounds such as vitamin B12), vitamin C, vitamin D, vitamin E, etc. may be used, but are not limited thereto, and suitable preparations widely known in the art may be used.
  • composition according to the present invention can be administered in a pharmaceutically effective amount.
  • pharmaceutically effective amount means an amount that is sufficient to treat a disease with a reasonable benefit/risk ratio applicable to medical treatment and does not cause side effects.
  • the effective dosage level of the pharmaceutical composition depends on the purpose of use, the patient's age, sex, weight and health condition, disease type, severity, drug activity, drug sensitivity, administration method, administration time, administration route and excretion rate, treatment Duration, combination, or factors including drugs used concurrently and other factors well known in the medical arts may be determined differently. For example, although not constant, generally 0.001 to 100 mg/kg, preferably 0.01 to 10 mg/kg, may be administered once or several times a day. The dosage is not intended to limit the scope of the present invention in any way.
  • the pharmaceutical composition may be administered to any animal that may develop an inflammatory disease, and the animal may include, for example, humans and primates as well as livestock such as cattle, pigs, horses, and dogs.
  • the pharmaceutical composition may be administered by an appropriate administration route according to the formulation form, and may be administered through various oral or parenteral routes as long as it can reach the target tissue.
  • the method of administration is not particularly limited, and may be administered by conventional methods such as oral, rectal or intravenous, intramuscular, skin application, intraventricular inhalation, intrauterine dural or intracerebroventricular injection. there is.
  • the pharmaceutical composition may be used alone for the prevention or treatment of inflammatory diseases, or may be used in combination with surgery or other drug treatment.
  • the present invention provides a health functional food composition for preventing or improving inflammatory diseases containing a compound or a pharmaceutically acceptable salt thereof as an active ingredient.
  • the inflammatory disease may be any one selected from the group consisting of osteoarthritis, rheumatoid arthritis, osteoporosis, Paget's disease of bone, and ankylosing spondylitis, but is not limited thereto.
  • the health functional food may be prepared in powder, granule, tablet, capsule, syrup or beverage for the purpose of preventing or improving inflammatory diseases.
  • the health functional food can take, and it can be formulated in the same way as the pharmaceutical composition and used as a functional food or added to various foods.
  • the health functional food may include all foods in a conventional sense.
  • beverages and various drinks, fruits and their processed foods such as canned fruit, jam, etc.
  • fish, meat and their processed foods such as ham, bacon, etc.
  • breads and noodles such as cookies and snacks
  • dairy products such as dairy products (butter, cheese, etc.) ), etc.
  • food used as feed for animals may also be included.
  • the health functional food composition may be prepared by further including food additives (food additives) commonly used in the art and appropriate other auxiliary components.
  • food additives food additives
  • the suitability as a food additive can be determined according to the standards and standards for the item in accordance with the general rules of the Food Additive Code and general test methods approved by the Ministry of Food and Drug Safety, unless otherwise specified.
  • Examples of the items listed in the 'Food Additive Code' include, for example, chemical compounds such as ketones, glycine, calcium citrate, nicotinic acid, and cinnamic acid; natural additives such as persimmon pigment, licorice extract, crystalline cellulose, kaoliang pigment, and guar gum; mixed preparations such as sodium L-glutamate preparations, noodle-added alkali preparations, preservative preparations, and tar color preparations; and the like.
  • chemical compounds such as ketones, glycine, calcium citrate, nicotinic acid, and cinnamic acid
  • natural additives such as persimmon pigment, licorice extract, crystalline cellulose, kaoliang pigment, and guar gum
  • mixed preparations such as sodium L-glutamate preparations, noodle-added alkali preparations, preservative preparations, and tar color preparations; and the like.
  • the other auxiliary ingredients include, for example, flavoring agents, natural carbohydrates, sweeteners, vitamins, electrolytes, colorants, pectic acid, alginic acid, organic acids, protective colloidal thickeners, pH adjusting agents, stabilizers, preservatives, glycerin, alcohols, carbonating agents, etc. may additionally contain.
  • natural carbohydrate monosaccharides such as glucose and fructose, disaccharides such as maltose and sucrose, polysaccharides such as dextrin and cyclodextrin, and sugar alcohols such as xylitol, sorbitol, and erythritol may be used.
  • natural sweeteners such as thaumatin and stevia extract or synthetic sweeteners such as saccharin and aspartame may be used.
  • An effective dose of the compound or salt thereof contained in the health functional food according to the present invention may be appropriately adjusted according to the purpose of use, such as preventing or improving inflammatory diseases.
  • the health functional food composition uses food as a raw material and has the advantage of not having side effects that can occur when taking general medicines for a long time, and has excellent portability, so it can be taken as an adjuvant for preventing or improving inflammatory diseases.
  • the KMU-series compounds were synthesized from 5-bromoindoline-2,3-dione under the following conditions:
  • Compound 1 was obtained by reduction and borylation [(a) i) NH 2 NH 2 , NaOH, EtOH; ii) bis(pincholato)diboron, PdCl 2 (dppf), KOAc, 1,4-dioxane: EtOH (1: 1.5), microwave], compound 1 and pyrazine derivative compound 2 [(b) K 2 CO 3 , amines or alcohols, DMF] to obtain compound 3 [(c) PdCl 2 (PPh 3 ) 2 , 2M K 2 CO 3 , 1,4-dioxane : EtOH (1: 1.5), microwave].
  • KMU-series compounds were synthesized through a Knoevenagel condensation reaction with 1H-imidazole-4-carbaldehyde [(d) piperidine, 1H-imidazole- 4-carbaldehyde, EtOH, microwave].
  • a microwave vessel was prepared with 5-bromoindoline-2,3-dione (0.50 g, 2.2 mmol), hydrazine hydrate (0.14 g, 4.4 mmol), and ethanol (EtOH, 2 mL) was filled. 100 W microwave was irradiated to the mixture at 100° C. for 10 minutes. After adding sodium hydroxide (0.18 g, 4.4 mmol), the mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The mixture was acidified with 2 M hydrochloric acid (HCl) and cold water was added to form a precipitate, which was collected by filtration, washed with 2 M HCl and washed again with water. Drying under vacuum gave 0.42 g of 5-bromoindolin-2-one (89% yield), which was used in the next step without further purification.
  • HCl hydrochloric acid
  • 1-adamantanamine (1-adamantanamine; 0.99 mmol, 0.15 g) and potassium tert-butyrate (potassium tert-butoxide, KOtbu; 4.0 mmol, 0.45 g) was added at room temperature and refluxed for 10 minutes under argon (Ar) gas.
  • a microwave vessel was charged with compound 1 (0.45 g, 1.8 mmol), compound 2a (0.20 g, 1.2 mmol), 1,4-dioxane (2.0 mL), ethanol (0.40 mL), and 2 M aqueous potassium carbonate (1.8 mL, 2.54 mL). mmol) was filled. bis(triphenylphosphine) palladium(II) dichloride, PdCl 2 (PPh 3 ) 2 in N 2 atmosphere; 0.041 g, 0.059 mmol] was added, and the mixture was irradiated with 100 W microwave at 110° C. for 10 minutes.
  • a microwave vessel was charged with compound 1 (0.23 g, 0.91 mmol), compound 2b (0.15 g, 0.76 mmol), 1,4-dioxane (2.0 mL), ethanol (0.40 mL), and 2 M aqueous potassium carbonate (0.91 mL, 1.5 mL). mmol) was filled.
  • Bis(triphenylphosphine)palladium( II ) dichloride [PdCl 2 (PPh 3 ) 2 ; 0.027 g, 0.038 mmol] was added, and the mixture was irradiated with 100 W microwave at 110° C. for 10 minutes.
  • a microwave vessel was charged with compound 1 (0.12 g, 0.47 mmol), compound 2c (0.12 g, 0.56 mmol), 1,4-dioxane (2.0 mL), ethanol (0.40 mL), and 2 M aqueous sodium carbonate (0.70 mL). , 1.4 mmol).
  • Bis(triphenylphosphine)palladium( II ) dichloride [PdCl 2 (PPh 3 ) 2 ; 0.017 g, 0.024 mmol] was added, and the mixture was irradiated with 100 W microwave at 110° C. for 10 minutes.
  • a microwave vessel was charged with compound 1 (0.062 g, 0.24 mmol), compound 2d (0.052 g, 0.20 mmol), 1,4-dioxane (2.0 mL), ethanol (0.40 mL), and 2 M aqueous sodium carbonate (0.30 mL, 0.6 mmol).
  • compound 1 0.062 g, 0.24 mmol
  • compound 2d 0.052 g, 0.20 mmol
  • 1,4-dioxane 2.0 mL
  • ethanol 0.40 mL
  • 2 M aqueous sodium carbonate 0.30 mL, 0.6 mmol
  • a microwave vessel was charged with compound 1 (0.12 g, 0.46 mmol), compound 2e (0.13 g, 0.56 mmol), 1,4-dioxane (2.0 mL), ethanol (0.40 mL), and 2 M aqueous sodium carbonate (0.69 mL, 1.4 mmol).
  • compound 1 (0.12 g, 0.46 mmol
  • compound 2e (0.13 g, 0.56 mmol
  • 1,4-dioxane 2.0 mL
  • ethanol 0.40 mL
  • 2 M aqueous sodium carbonate 0.69 mL, 1.4 mmol
  • a microwave vessel was charged with compound 3b (0.080 g, 0.27 mmol), piperidine (6.0 ⁇ L, 0.05 mmol), 1H-imidazole-4-carboaldehyde (0.031 g, 0.33 mmol), and ethanol (2.0 mL); The mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The solvent was removed in vacuo and the residue was purified by silica gel chromatography using 80:10 DCM/MeOH to give 0.042 g (22%) of the title compound (4b).
  • a microwave vessel was charged with compound 3c (0.056 g, 0.18 mmol), piperidine (1.8 ⁇ L, 0.018 mmol), 1H-imidazole-4-carboaldehyde (0.021 g, 0.22 mmol), and ethanol (2.0 mL); The mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The solvent was removed under reduced pressure, and the resulting solid was filtered by adding distilled water. The residue was purified by silica gel chromatography using 100:5:0.5 chloroform (CHCl 3 )/MeOH/ammonia water (NH 4 OH) to give 0.053 g (75%) of the title compound (4c).
  • a microwave vessel was charged with compound 3c (0.079 g, 0.26 mmol), piperidine (2.5 ⁇ L, 0.025 mmol), 1H-imidazole-4-carboaldehyde (0.029 g, 0.31 mmol), and ethanol (2.0 mL); The mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The solvent was removed under reduced pressure, and the resulting solid was filtered by adding distilled water. The residue was purified by silica gel chromatography using 100:3:0.3 CHCl 3 /MeOH/NH 4 OH to give 0.068 g (68%) of the title compound (4c′).
  • a microwave vessel was charged with compound 3d (0.034 g, 0.094 mmol), piperidine (0.9 ⁇ L, 0.0091 mmol), 1H-imidazole-4-carboaldehyde (0.011 g, 0.11 mmol), and ethanol (1.0 mL), The mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The solvent was removed under reduced pressure, and the resulting solid was filtered by adding distilled water. The residue was purified by silica gel chromatography using 90:10 DCM/MeOH to give 0.030 g (72%) of the title compound (4d).
  • a microwave vessel was charged with compound 3e (0.12 g, 0.7 mmol), piperidine (4.1 ⁇ L, 0.037 mmol), 1H-imidazole-4-carboaldehyde (0.042 g, 0.44 mmol), and ethanol (2.0 mL); The mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The solvent was removed under reduced pressure, and the resulting solid was filtered by adding distilled water. The residue was purified by silica gel chromatography using 100:3:0.3 CHCl 3 /MeOH/NH 4 OH to give 0.062 g (42%) of the title compound (4e).
  • LPS Escherichia coli serotype
  • Celecoxib, ibrutinib, methotrexate, and ruxolitinib were purchased from Selleck Chemicals (Houston, TX, USA).
  • Receptor activator of NF- ⁇ B ligands (RANKL) was purchased from PeproTech (Rocky Hill, NH, USA).
  • Anti-IL-1 ⁇ antibody was purchased from Novus Biologicals (Centennial, CO, USA).
  • Antibodies against IL-6, COX-1, COX-2, TAK1, p-p38, and p38 were respectively Santa Cruz Bio-technology (Santa Cruz, CA, USA) and Sigma-Aldrich (St. Louis, MO, USA).
  • Anti-TNF- ⁇ antibody was purchased from Abcam (Cambridge, MA, USA).
  • Anti- ⁇ -actin antibody was purchased from Sigma-Aldrich (St. Louis, MO, USA).
  • Anti-mouse IgG-horseradish peroxidase (HRP) and anti-mouse IgG-HRP antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA).
  • Human monocytic cell line THP-1 cells (Korean Cell Line Bank, Seoul, Korea) were cultured in RPMI1640 medium (Welgene Inc., Gyeongsan, Korea), 10% fetal bovine serum (FBS; Welgene Inc.). ., Gyeongsan, Korea), and 1% antibiotic-antimycotic solution (Welgene Inc., Gyeongsan, Korea). Cells were cultured at 37.5° C., 5% CO 2 in completely humid air.
  • alpha-MEM was developed by Welgene Inc. (Gyeongsan, Korea).
  • RAW 264.7 cells were grown in osteoclast differentiation medium (alpha-MEM+50 ng/ml RANKL) to generate osteoclast-like cells. After differentiation, multinucleated cells (MNCs) were visualized using a TRAP staining kit (TaKaRa, Japan) according to the manufacturer's protocol.
  • MNCs multinucleated cells
  • OA human osteoarthritis
  • FLS fibroblast-like synoviocytes
  • Osteoarthritis tissue was pulverized into 2-3 mm pieces, and then the minced tissue was treated with 0.5 mg/mL type II collagenase in Dulbecco's modified Eagle's medium (DMEM; Welgene Inc., Gyeongsan, Korea) at 37.5°C under 5% CO 2 . (collagenases; Thermo Fisher Scientific, Waltham, MA, USA).
  • DMEM Dulbecco's modified Eagle's medium
  • FBS FBS
  • antibiotic-antimycotic solution 1% antibiotic-antimycotic solution
  • adherent cells were cultured in DMEM, 10% FBS, and 1% antibiotic-antimycotic solution at 37.5° C. under 5% CO 2 , and the medium was changed every 3 days.
  • the medium is diluted 1:3 with fresh culture.
  • kinase-profiling service was performed by Eurofins Cerep S.A.
  • Kinase-profiling assays were performed with ATP concentrations of 1 ⁇ M of compound and K values for each individual kinase and kinase substrate according to Eurofin's protocol.
  • XTT assay was performed using XTT assay (Welgene Inc., Gyeongsan, Korea). Briefly, 2 ⁇ 10 5 THP-1 cells were plated in 96-well plates for 24 hours, and then cells were treated with various concentrations of KMU-1170 and cultured for 24 hours at 37° C. under 5% CO 2 . . Thereafter, a 0.5 mg/mL XTT solution was added to the culture medium, and cultured at 37° C. for 3 hours under 5% CO 2 . Optical density (OD) was measured with a microplate reader (BMG Labtech, Ortenberg, Germany) at a wavelength of 450 nM.
  • THP-1 cells (2 ⁇ 10 6 cells/well in 60-mm dishes) and FLS (2 ⁇ 10 6 cells/well in 100-mm dishes) were plated. After culturing for 24 hours, the cells were dissolved in RIPA buffer (20 mM HEPES and 0.5% Triton X-100, pH 7.6), and the supernatant fraction was collected. Protein concentration was measured using the BCA assay kit (Thermo Fisher Scientific, Waltham, MA, USA). Equal amounts of protein were electrophoresed on SDS-PAGE and transferred to a nitrocellulose membrane (GE Healthcare Life Science, Pittsburgh, PA, USA).
  • the membrane was incubated for 1 hour with blocking buffer (0.05% Tween 20 with 5% non-fat dry milk in Tris-buffered saline (TBS)) and then incubated for 24 hours with an appropriately diluted primary antibody against a specific target protein. cultured for a while. Next, the membrane was washed in TBS with Tween 20 and incubated with the appropriate secondary antibody for 1 hour at room temperature.
  • blocking buffer 0.05% Tween 20 with 5% non-fat dry milk in Tris-buffered saline (TBS)
  • TBS Tris-buffered saline
  • RNA isolation and reverse transcription Reverse Transcription-Polymerase Chain Reaction RT- PCR
  • qPCR real-time quantification PCR
  • Table 1 below shows the primer sequences used in this experimental example.
  • THP-1 cells (1 ⁇ 10 3 ) were cultured on 8-chamber glass slides for 24 hours, treated with 1 ⁇ mol/L of KMU-1170 for 1 hour, and then stimulated with LPS for 6 hours. Cells were washed with phosphate-buffered saline (PBS), fixed with 4% formaldehyde, permeabilized with PBS containing 0.2% Triton X-100, and treated with bovine serum albumin for 1 hour. Incubation was performed to block non-specific binding.
  • PBS phosphate-buffered saline
  • the cells were incubated with the primary antibody for 24 hours at 4°C and then FITC-conjugated goat anti-mouse IgG (Thermo Fisher Scientific, Waltham, MA, USA) and 4',6-diamidino-2 After treatment with -phenylindole (4',6-diamidino-2-phenylindole; Thermo Fisher Scientific, Wal-tham, MA, USA; for nuclear staining), the stained cells were examined under a fluorescence microscope.
  • KMU-series compounds as anti-inflammatory agents, the anti-inflammatory effects of KMU-series compounds on LPS-mediated inflammation in THP-1 cells were analyzed.
  • THP-1 cells were differentiated into macrophages using 100 nM of phorbol-12-myristate-13-acetate (PMA) for 24 hours.
  • the cells were treated with KMU-11421, 11426, 11427, 11361, 11342, 11170 (1 ⁇ M), and Ruxolitinib (50 ⁇ M) for 1 hour, then LPS (1 ⁇ g/mL) was added for 3 hours .
  • Total RNA was extracted and mRNA expression levels of IL-1 ⁇ , TNF- ⁇ , and IL-6 were determined using real-time PCR.
  • KMU-11342 On RANKL-induced osteoclast differentiation, after treatment with KMU-11342 (0.01, 0.25, 0.5 ⁇ M) for 1 hour, RAW 264.7 cells were treated with RANKL (50 ng/mL) for 5 days. The treated cells were differentiated into osteoclasts, and the cells were stained using a TRAP staining kit. TRAP-positive multinucleated cells (MNC) were stained purple red, and the number of TRAP-stained cells indicating the degree of osteoclast differentiation increased in a time-dependent manner for 5 days.
  • MNC multinucleated cells
  • KMU -11170 (Hereinafter, in the experimental results and drawings, " KMU -1170") Analysis of various activities of the compound
  • Table 2 shows the kinase activities of 15 protein kinases according to 1 ⁇ M KMU-1170 treatment.
  • KMU-1170 inhibits various kinases related to the inflammatory response, in particular, mitogen-activated protein kinase 1 (MAPK1), Lck, TYK2 , JAK3, and Txk.
  • MAPK1 mitogen-activated protein kinase 1
  • Lck Lck
  • TYK2 TYK2
  • JAK3 Txk
  • THP-1 cells were differentiated into macrophages using 100 nM of PMA for 24 hours, and then the cells were treated with various concentrations of KMU-1170 (0.01, 0.1, 0.5, 1, 5, and 10 ⁇ M), Fig. 4B As shown, KMU-1170 showed no toxicity up to 1 ⁇ M concentration.
  • KMU-1170 (0.1, 0.5, and 1 ⁇ M) was pretreated for 1 hour, followed by LPS (1 ⁇ g/mL) for 6 hours to confirm the upregulated changes in iNOS and COX-2, 1 ⁇ M of KMU-1170 inhibited LPS-induced up-regulation of inducible nitric oxide synthase (iNOS) mRNA and protein (Fig. 4C, D, E), and as a pretreatment, LPS-induced cyclook Upregulation of cyclooxygenase-2 (COX-2) mRNA and protein was suppressed, but no significant changes were observed in COX-1 mRNA and protein (FIG. 4F, G, H, I).
  • iNOS inducible nitric oxide synthase
  • THP-1 cells were differentiated into macrophages using PMA (100 nM) for 24 hours, followed by pretreatment with KMU-1170 (0.1, 0.5, and 1 ⁇ M) for 1 hour followed by LPS (1 ⁇ g/mL) for 6 hours.
  • KMU-1170 0.1, 0.5, and 1 ⁇ M
  • LPS 1 ⁇ g/mL
  • THP-1 cells were differentiated into macrophages using PMA (100 nM) for 24 hours, followed by pretreatment with KMU-1170 (0.1, 0.5, and 1 ⁇ M) for 1 hour followed by LPS (1 ⁇ g/mL) for 6 hours.
  • PMA 100 nM
  • KMU-1170 0.1, 0.5, and 1 ⁇ M
  • LPS 1 ⁇ g/mL
  • TAK1 transforming growth factor- ⁇ -activated kinase 1
  • THP-1 cells were differentiated into macrophages using PMA (100 nM) for 24 hours, treated with or without KMU-1170 for 24 hours, and induced with LPS (1 ⁇ g/mL).
  • PMA 100 nM
  • KMU-1170 a monopeptide kinase
  • LPS 1 ⁇ g/mL
  • 6E and 6F among MAPKs, only ERK (extracellular signal-regulated kinases) and JNK (c-Jun N-terminal kinases) were phosphorylated in response to LPS, and pretreatment with 1 ⁇ M KMU-1170 It was shown to inhibit LPS-induced phosphorylation of ERK and JNK in 1 cells.
  • THP-1 cells were differentiated into macrophages using PMA (100 nM) for 24 hours, and then pretreated with 1 ⁇ M KMU-1170 for 1 hour. After that, LPS (1 ⁇ g/mL) and/or ATP (1 mM) were treated for 6 hours.
  • pretreatment of THP-1 cells with 1 ⁇ M KMU-1170 inhibited LPS-induced pro-IL-1 ⁇ and IL-1 ⁇ cytoplasmic release. Furthermore, pretreatment with 1 ⁇ M KMU-1170 upregulated NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), pro-caspase-1, and pro-IL-1 ⁇ induced by LPS and co-treatment with LPS and ATP. control was weakened.
  • ASC apoptosis-associated speck-like protein containing a CARD
  • THP-1 cells were differentiated into macrophages using PMA (100 nM) for 24 hours, followed by KMU-1170 (1 ⁇ M), celecoxib (25 ⁇ M), ibrutinib (5 ⁇ M), Luxority After pre-treatment with nip (luxolitinib, 50 ⁇ M) and methotrexate (1 ⁇ M) for 1 hour, LPS (1 ⁇ g/mL) was treated for 6 hours.
  • OA human osteoarthritis
  • FLS fibroblast-like synovial cells
  • KMU-1170 pretreated FLS showed LPS-induced up-regulated IL-1 ⁇ , TNF- ⁇ , and IL-6 mRNA and protein levels (FIG. 9A to 9D), iNOS and COX -2 protein levels (FIG. 9E).
  • KMU-1170 pretreatment inhibited LPS-induced phosphorylation of IKK ⁇ / ⁇ and NF- ⁇ B p65 in FLS (FIG. 9F).
  • the present invention is a novel multi-protein kinase inhibitor KMU-1170 is anti-inflammatory effect by regulating the p-TAK1 / p-IKK ⁇ / ⁇ / p-NF- ⁇ B / pro-inflammatory cytokine signaling pathway and NLRP3 inflammasome It was confirmed that it had (FIG. 10). These results suggest that the KMU-series compounds are very advantageous compounds for the development of potent anti-inflammatory drugs.

Landscapes

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

Abstract

The present invention relates to a novel inhibitor for multiple protein kinases, and provides: a novel indolin-2-one compound or a pharmaceutically acceptable salt thereof that exhibits excellent anti-inflammatory activity; and a composition for treating inflammatory diseases, containing same as an active ingredient. The novel compound strongly inhibits the activity of various protein kinases related to inflammation, and thus can be used as an inhibitor for multiple protein kinases, and has an excellent anti-inflammatory effect as compared to other anti-inflammatory drugs, and thus can be used in the development of therapeutic agents for inflammatory diseases.

Description

신규한 다중 단백질 키나아제 억제제Novel multiple protein kinase inhibitors
본 발명은 신규한 다중 단백질 인산화효소 억제제에 관한 것으로, 우수한 항염증 활성을 나타내는 인돌린-2-온 신규 유도체에 관한 것이다. The present invention relates to novel multiple protein kinase inhibitors, and to novel indolin-2-one derivatives exhibiting excellent anti-inflammatory activity.
염증은 유해한 자극으로부터 숙주를 보호하고 조직 항상성을 유지하는 신체의 중요하고 기본적인 면역 체계 반응이다. 효과적인 염증 반응은 감염 또는 손상으로부터 살아남을 수 있도록 하지만, 과도하거나 지속적인 염증은 천식, 암, 만성 통증, 통풍, 정신 장애, 신경 쇠약, 건선, 류마티스 관절염 및 혈관염과 같은 다양한 병리학적 상태로 이어질 수 있다. 더욱이, 유해 자극에 대한 염증 반응은 자극이 제거될 때 종료되는 것도 똑같이 중요하다. 그러므로, 효과적인 염증 반응 및 항염증 반응 사이의 균형은 건강한 상태를 유지하는 가장 중요한 조건 중 하나이다.Inflammation is an important and fundamental immune system response of the body that protects the host from harmful stimuli and maintains tissue homeostasis. An effective inflammatory response allows you to survive infection or injury, but excessive or persistent inflammation can lead to various pathological conditions such as asthma, cancer, chronic pain, gout, mental disorders, nervous breakdown, psoriasis, rheumatoid arthritis and vasculitis. . Moreover, it is equally important that the inflammatory response to a noxious stimulus ends when the stimulus is removed. Therefore, a balance between an effective inflammatory response and an anti-inflammatory response is one of the most important conditions for maintaining a healthy state.
톨-유사 수용체(Toll-like receptors, TLRs)는 다양한 병원균 및 손상된 조직을 인식하는 선척적 면역 시스템에서 중요한 수용체 군을 구성한다. TLRs는 구조, 세포 내 위치 및 인식 분자 특성과 같은 특이성에 따라 여러 유형으로 나누어진다. 그람-음성 박테리아의 외벽을 구성하는 박테리아의 내독소(endotoxin)인 지질다당류(lipopolysaccharide, LPS)는 TLR4에 의해 인식된다. LPS가 TLR4와 결합할 때, 염증 관련 유전자 발현이 전사 인자 NF-κB(nuclear factor kappa-light-chain-enhancer of activated B cells)를 통해 조절된다. 염증은 사이토카인(cytokines)이라고 불리는 수용성 면역 신호 분자에 의해 조절되고, 인터류킨(interleukin (IL)-1β), IL-6, 및 종양 괴사 인자-α(tumor necrosis factor-α, TNF-α)와 같은 염증 사이토카인은 LPS에 의해 유도된다. 이러한 사이토카인은 다양한 염증 질환에 관여하며, 이들의 억제는 염증 질환 치료에 상당한 도움이 된다.Toll-like receptors (TLRs) constitute an important receptor family in the innate immune system that recognizes various pathogens and damaged tissues. TLRs are divided into several types according to specificities such as structure, subcellular localization and recognition molecular properties. Lipopolysaccharide (LPS), an endotoxin of bacteria constituting the outer wall of Gram-negative bacteria, is recognized by TLR4. When LPS binds to TLR4, inflammation-related gene expression is regulated through the transcription factor NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells). Inflammation is regulated by soluble immune signaling molecules called cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α). The same inflammatory cytokines are induced by LPS. These cytokines are involved in various inflammatory diseases, and their inhibition is of great help in the treatment of inflammatory diseases.
염증 분자의 복합체인 인플라마좀(inflammasome)은 병원성 감염에 대한 선천 면역 반응으로, 전염증성 사이토카인의 형성에 중요한 역할을 한다. 이들 중에서, NOD-, LRR- 및 pyrin 도메인-포함 단백질 3 (NLRP3) 인플라마좀의 기능은 많이 연구되어 왔으며, 필수적으로 조절 단백질 NLRP3, CARD를 포함하는 아폽토시스 관련 speck-유사 단백질(apoptosis-associated speck-like protein containing a CARD, ASC) 연결 분자(adaptor molecule), 프로카스파제(procaspase)-1 및 NIMA-관련 키나아제 7로 구성된다. NF-κB에 의한 NLRP3 및 IL-1β의 전사 및 번역 후, 활성 caspase-1는 pro-IL-1β를 성숙한 분비 형태로 처리할 수 있다. 그러므로, 염증성 질환에서 NLRP3 인플라마좀-관련 신호를 효과적으로 조절하는 것이 매우 중요하다.The inflammasome, a complex of inflammatory molecules, is an innate immune response to pathogenic infections and plays an important role in the formation of proinflammatory cytokines. Among them, the functions of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome have been extensively studied, and the apoptosis-associated speck-like proteins (including the regulatory protein NLRP3, CARD) have been indispensable. -like protein containing a CARD (ASC) adapter molecule, procaspase-1 and NIMA-related kinase 7. After transcription and translation of NLRP3 and IL-1β by NF-κB, active caspase-1 can process pro-IL-1β into its mature secreted form. Therefore, it is very important to effectively modulate NLRP3 inflammasome-related signaling in inflammatory diseases.
단백질 인산화효소(단백질 키나아제, protein kinase)는 단백질의 인산화(phosphorylation)를 담당하며, 이는 염증에서 세포 내 신호 전달에 중요한 역할을 한다. 단백질 키나아제는 염증성 질환의 치료를 위한 약물 개발에서 매우 중요한 약물 표적이다. 예를 들어, 화이자(Pfizer)가 개발한 강력한 JAK(Janus-associated kinase) 1 및 JAK 3 길항체인 토파시티닙(tofacitinib)은 류마티스 관절염(rheumatoid arthritis, RA)의 징후와 증상을 줄이고, RA 환자의 신체 기능을 개선시켰다. 게다가, p38 키나아제 억제제인 semapimod는 크론병(Crohn’s disease) 환자에서 임상 결과를 개선시켰다. 비장 티로신 키나아제 억제제인 BAY 61-3606는 알레르기성 천식 환자에 대한 적응증(indication)으로 미국 식품의약국(Food and Drug Administration, FDA)에 승인되었다. 또한, 다양한 브루톤 티로신 카나아제(Bruton’s tyrosine kinase, BTK) 억제제는 임상 시험 중에 있다. 이렇듯 유망한 단백질 키나아제 억제제에 대한 개발 필요성이 높아지고 있다.Protein kinase (protein kinase) is responsible for phosphorylation of proteins, which plays an important role in intracellular signal transduction in inflammation. Protein kinases are very important drug targets in drug development for the treatment of inflammatory diseases. For example, tofacitinib, a potent Janus-associated kinase (JAK) 1 and JAK 3 antagonist developed by Pfizer, reduces the signs and symptoms of rheumatoid arthritis (RA) and is improved bodily functions. Moreover, the p38 kinase inhibitor semapimod improved clinical outcomes in patients with Crohn's disease. BAY 61-3606, a spleen tyrosine kinase inhibitor, has been approved by the Food and Drug Administration (FDA) as an indication for allergic asthma patients. In addition, various Bruton's tyrosine kinase (BTK) inhibitors are in clinical trials. As such, the need for development of promising protein kinase inhibitors is increasing.
본 발명의 목적은 우수한 단백질 키나아제 억제 활성을 가지는 신규 화합물을 제공하는 데에 있다.An object of the present invention is to provide novel compounds having excellent protein kinase inhibitory activity.
본 발명의 다른 목적은 상기 화합물을 포함하는 염증성 질환 치료 조성물을 제공하는 데에 있다.Another object of the present invention is to provide a composition for treating inflammatory diseases comprising the compound.
상기의 목적을 달성하기 위하여, 본 발명은 하기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 제공한다.In order to achieve the above object, the present invention provides a compound represented by Formula 1 below or a pharmaceutically acceptable salt thereof.
<화학식 1><Formula 1>
Figure PCTKR2022003249-appb-img-000001
Figure PCTKR2022003249-appb-img-000001
상기 화학식 1에서, R1은 수소, 하이드록시, 할로겐, (C1-C4) 알킬, (C3-C10) 사이클로알킬, 또는 아다만타닐(adamantanyl)에서 선택되고, R2는 이미다졸일(imidazolyl), 피라졸일(pyrazolyl), 피롤일(pyrrolyl) 또는 피롤리딘일(pyrrolidinyl)에서 선택된다.In Formula 1, R 1 is selected from hydrogen, hydroxy, halogen, (C1-C4) alkyl, (C3-C10) cycloalkyl, or adamantanyl, and R 2 is imidazolyl , pyrazolyl, pyrrolyl or pyrrolidinyl.
본 발명은 상기 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 염증성 질환 예방 또는 치료용 약학 조성물을 제공한다.The present invention provides a pharmaceutical composition for preventing or treating inflammatory diseases containing the compound or a pharmaceutically acceptable salt thereof as an active ingredient.
또한, 본 발명은 상기 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 염증성 질환 예방 또는 개선용 건강기능식품 조성물을 제공한다.In addition, the present invention provides a health functional food composition for preventing or improving inflammatory diseases containing the compound or a pharmaceutically acceptable salt thereof as an active ingredient.
본 발명에 따른 신규 화합물은 염증과 관련된 다양한 단백질 키나아제의 활성을 강력하게 억제하는 바, 다중 단백질 키나아제 억제제로 활용할 수 있다.Since the novel compound according to the present invention strongly inhibits the activity of various protein kinases related to inflammation, it can be used as a multi-protein kinase inhibitor.
또한, 상기 화합물은 사이토카인 및 NLRP3 인플라마좀 활성을 조절하고, 다른 항염증제들과 비교하여 보다 우수한 항염증 효과를 가지는 바, 이를 염증성 질환의 치료제 개발에 활용할 수 있다.In addition, since the compound regulates cytokine and NLRP3 inflammasome activity and has a superior anti-inflammatory effect compared to other anti-inflammatory agents, it can be used for the development of therapeutic agents for inflammatory diseases.
도 1은 본 발명의 일 실시예에 따라 합성된 신규의 KMU-시리즈 화합물의 합성 과정을 나타낸 모식도이다.1 is a schematic diagram showing a synthesis process of a novel KMU-series compound synthesized according to an embodiment of the present invention.
도 2는 본 발명의 일 실험예에 따라 THP-1 세포에서 LPS-유도 상향 조절된 전염증성 사이토카인에 대한 KMU-시리즈 화합물의 억제 효과를 확인한 것이다 (*p < 0.05, **p < 0.01 , #p < 0.001 compared to LPS alone)Figure 2 confirms the inhibitory effect of KMU-series compounds on LPS-induced up-regulated pro-inflammatory cytokines in THP-1 cells according to an experimental example of the present invention (*p < 0.05, **p < 0.01, #p < 0.001 compared to LPS alone)
도 3은 RAW 264.7 세포의 RANKL 유도 파골세포 형성(osteoclastogenesis)에 대한 KMU-11342의 억제 효과를 확인한 것이다 (**p < 0.01, #p < 0.001 compared to RANKL alone).Figure 3 confirms the inhibitory effect of KMU-11342 on RANKL-induced osteoclastogenesis in RAW 264.7 cells (**p < 0.01, #p < 0.001 compared to RANKL alone).
도 4는 본 발명의 일 실시예에 따라 합성된 신규 화합물인 KMU-11170 (이하, 도면 및 실험결과에서는 "KMU-1170"으로 표기) 및 인간 단핵구 세포주 THP-1에서 LPS-유도 iNOS 및 COX-2에 대한 KMU-1170의 영향에 관한 것으로, (A)는 KMU-1170의 합성 과정을 나타낸 모식도, (B)는 XTT 분석을 통해 THP-1 세포에서 KMU-1170의 세포 독성 효과를 확인한 것, (C) 내지 (I)는 웨스턴 블롯 및 Image-J software를 사용하여 iNOS, COX-1, 및 COX-2의 mRNA 또는 단백질의 발현 변화를 확인한 그래프이다 (†p < 0.05, *p < 0.01, 및 #p < 0.001 compared to LPS).Figure 4 is a novel compound synthesized according to an embodiment of the present invention, KMU-11170 (hereinafter referred to as "KMU-1170" in the drawings and experimental results) and LPS-induced iNOS and COX-induced iNOS and COX- in the human monocyte cell line THP-1. 2, (A) is a schematic diagram showing the synthesis process of KMU-1170, (B) confirms the cytotoxic effect of KMU-1170 in THP-1 cells through XTT analysis, (C) to (I) are graphs confirming changes in mRNA or protein expression of iNOS, COX-1, and COX-2 using western blotting and Image-J software (†p < 0.05, *p < 0.01, and #p < 0.001 compared to LPS).
도 5는 본 발명의 일 실험예에 따라 THP-1 세포에서 LPS-유도 상향 조절된 전염증성 사이토카인에 대한 KMU-1170의 억제 효과를 확인한 것으로, (A)는 전체 세포 용해물을 분리하여 웨스턴 블롯으로 pro-IL-1β, TNF-α, IL-6, 및 β-actin의 단백질 발현 수준을 측정한 것, (B)는 Image-J software를 사용하여 pro-IL-1β, TNF-α, 및 IL-6 밴드의 상대적 광학 밀도를 분석한 것, (C)는 총 RNA를 추출하여, RT-PCR로 IL-1β, TNF-α, 및 IL-6의 mRNA 발현 수준을 확인한 것, (D)는 Image-J software를 사용하여 IL-1β, TNF-α, 및 IL-6 밴드의 상대적 광학 밀도를 분석한 것, (E) 내지 (G)는 총 RNA를 추출하여 실시간 PCR로 IL-1β, TNF-α, 및 IL-6의 mRNA 발현 수준을 확인한 것이다 (†p < 0.05, *p < 0.01, 및 #p < 0.001 compared to LPS).Figure 5 confirms the inhibitory effect of KMU-1170 on LPS-induced up-regulated pro-inflammatory cytokines in THP-1 cells according to an experimental example of the present invention. The protein expression levels of pro-IL-1β, TNF-α, IL-6, and β-actin were measured by blotting, (B) using Image-J software, pro-IL-1β, TNF-α, And analyzing the relative optical density of the IL-6 band, (C) extracting total RNA and confirming the mRNA expression levels of IL-1β, TNF-α, and IL-6 by RT-PCR, (D ) is the analysis of the relative optical density of IL-1β, TNF-α, and IL-6 bands using Image-J software, (E) to (G) are total RNA extracted and IL-1β by real-time PCR , TNF-α, and IL-6 mRNA expression levels were confirmed (†p < 0.05, *p < 0.01, and #p < 0.001 compared to LPS).
도 6은 본 발명의 일 실험예에 따라 THP-1 세포에서 LPS-유도 TLR4 신호전달관련 단백질에 대한 KMU-1170의 효과를 확인한 것으로, (A)는 전체 세포 용해물을 분리하여 웨스턴 블롯으로 TLR4, MyD88, 및 β-actin의 단백질 발현 수준을 측정한 것, (B)는 Image-J software를 사용하여 TLR4 및 MyD88 밴드의 상대적 광학 밀도를 분석한 것, (C)는 웨스턴 블롯으로 p-TAK1, TAK1, 및 β-actin의 단백질 발현 수준을 측정한 것, (D)는 Image-J software를 사용하여 p-TAK1 밴드의 상대적 광학 밀도를 분석한 것, (E)는 웨스턴 블롯으로 p-ERK, ERK, p-JNK, JNK, p-p38, p38, 및 β-actin의 단백질 발현 수준을 측정한 것, (F)는 Image-J software를 사용하여 p-ERK, p-JNK, 및 p-p38 밴드의 상대적 광학 밀도를 분석한 것이다 (#p < 0.001 compared to LPS).Figure 6 confirms the effect of KMU-1170 on LPS-induced TLR4 signaling-related proteins in THP-1 cells according to an experimental example of the present invention. , MyD88, and β-actin protein expression levels were measured, (B) analyzed the relative optical density of TLR4 and MyD88 bands using Image-J software, (C) was p-TAK1 by western blot , TAK1, and β-actin protein expression levels were measured, (D) analyzed the relative optical density of the p-TAK1 band using Image-J software, (E) was p-ERK by Western blot , ERK, p-JNK, JNK, p-p38, p38, and measuring the protein expression levels of β-actin, (F) using Image-J software to measure p-ERK, p-JNK, and p- The relative optical density of the p38 band was analyzed (#p < 0.001 compared to LPS).
도 7은 본 발명의 일 실험예에 따라 THP-1 세포에서 LPS-유도 IKKα/β 및 NF-κB의 인산화 및 NF-κB의 핵 전좌에 대한 KMU-1170의 효과를 확인한 것으로, (A)는 웨스턴 블롯으로 p-IKKα/β, IKKα/β, p-NF-κB p65, NF-κB p65, 및 β-actin의 단백질 발현 수준을 측정한 것, (B)는 Image-J software를 사용하여 p-IKKα/β 및 p-NF-κB p65 밴드의 상대적인 광학 밀도를 분석한 것, (C)는 세포를 NF-κB p65 (녹색) 및 4’,6-디아미디노-2-페닐인돌(4’,6-diamidino-2-phenylindole) (청색)의 항체로 염색하여 형광 현미경으로 확인한 것으로, 화살표는 400배의 확대를 나타낸다 (*p < 0.01 및 #p < 0.001 compared to LPS).Figure 7 confirms the effect of KMU-1170 on LPS-induced phosphorylation of IKKα / β and NF-κB and nuclear translocation of NF-κB in THP-1 cells according to an experimental example of the present invention, (A) The protein expression levels of p-IKKα/β, IKKα/β, p-NF-κB p65, NF-κB p65, and β-actin were measured by Western blotting, (B) was obtained using Image-J software. Analysis of relative optical densities of -IKKα/β and p-NF-κB p65 bands, (C) shows cells were treated with NF-κB p65 (green) and 4',6-diamidino-2-phenylindole (4 ',6-diamidino-2-phenylindole) (blue) and confirmed by fluorescence microscopy, arrows indicate 400-fold magnification (*p < 0.01 and #p < 0.001 compared to LPS).
도 8은 본 발명의 일 실험예에 따라 THP-1 세포에서 LPS-유도 NLRP3 인플라마좀 활성화에 대한 KMU-1170의 억제 효과 및 KMU-1170의 항염증 잠재력에 관한 것으로, (A)는 세포 용해물(Lysate) 및 배지 상등액(Sup)을 분리하여 웨스턴 블롯으로 상등액에 대한 pro-IL-1β 및 IL-1β의 단백질 발현 수준과 세포 용해물에 대한 NLRP3, ASC, pro-caspase-1, pro-IL-1β, 및 β-actin의 단백질 발현 수준을 측정한 것, (B)는 Image-J software를 사용하여 용해물에서 NLRP3, ASC, pro-caspase-1, 및 pro-IL-1β 밴드의 상대적 광학 밀도를 분석한 것, (C)는 웨스턴 블롯으로 용해물에서 pro-IL-1β 및 β-actin의 단백질 발현 수준을 측정한 것, (D)는 Image-J software를 사용하여 pro-IL-1β 밴드의 상대적 광학 밀도를 분석한 것이다 (†p < 0.05, *p < 0.01, 및 #p < 0.001 compared to LPS).Figure 8 relates to the inhibitory effect of KMU-1170 on LPS-induced NLRP3 inflammasome activation and anti-inflammatory potential of KMU-1170 in THP-1 cells according to an experimental example of the present invention, (A) is for cells Lysate and medium supernatant were separated, and the protein expression levels of pro-IL-1β and IL-1β in the supernatant and NLRP3, ASC, pro-caspase-1, pro- Measuring the protein expression levels of IL-1β and β-actin, (B) is the relative ratio of NLRP3, ASC, pro-caspase-1, and pro-IL-1β bands in the lysate using Image-J software The optical density was analyzed, (C) the protein expression levels of pro-IL-1β and β-actin were measured in the lysate by Western blot, (D) was pro-IL-1β using Image-J software. The relative optical density of the 1β band was analyzed (†p < 0.05, *p < 0.01, and #p < 0.001 compared to LPS).
도 9는 본 발명의 일 실험예에 따라 원발성 인간 골관절염(OA) 섬유아세포 유사 활막세포(FLS)에서 LPS-유도 염증 반응에 대한 KMU-1170의 억제 효과를 확인한 것으로, (A) 내지 (C)는 총 RNA를 추출하여 실시간 PCR로 IL-1 β, TNF-α, 및 IL-6 mRNA 발현 수준을 측정한 것, (D) 내지 (F)는 전체 세포 용해물을 분리하여 웨스턴 블롯으로 각각 pro-IL-1β, TNF-α, IL-6, 및 β-actin의 단백질 발현 수준 (D); iNOS, COX-2, 및 β-actin의 단백질 발현 수준 (E); p-IKKα/β, p-NF-κB p65, 및 β-actin의 단백질 발현 수준 (F);을 측정한 것이다 (*p < 0.01 및 #p < 0.001 compared to LPS).Figure 9 confirms the inhibitory effect of KMU-1170 on the LPS-induced inflammatory response in primary human osteoarthritis (OA) fibroblast-like synovial cells (FLS) according to an experimental example of the present invention, (A) to (C) is to extract total RNA and measure IL-1 β, TNF-α, and IL-6 mRNA expression levels by real-time PCR, (D) to (F) isolate whole cell lysates and use Western blot, respectively pro - protein expression levels of IL-1β, TNF-α, IL-6, and β-actin (D); protein expression levels of iNOS, COX-2, and β-actin (E); The protein expression levels (F) of p-IKKα/β, p-NF-κB p65, and β-actin; were measured (*p < 0.01 and #p < 0.001 compared to LPS).
도 10은 본 발명의 일 실험예에 따라 확인한 KMU-1170의 항염증 효과 매커니즘을 도식화한 것이다.Figure 10 is a schematic diagram of the anti-inflammatory effect mechanism of KMU-1170 confirmed according to an experimental example of the present invention.
이하, 본 발명을 상세하게 설명하기로 한다.Hereinafter, the present invention will be described in detail.
단백질 키나아제는 포스포릴기(phosphoryl group)를 세린(serine), 트레오닌(threonine) 또는 티로신(tyrosine) 잔기에 부착하여 단백질 분자를 인산화하는 효소로, ATP와의 결합은 키나아제 활성에 필수적이다. 최근, 단백질 키나아제가 암 및 염증성 질환을 포함하는 다양한 인간 질병에 관련있음에 대한 보고들로 다양한 단백질 키나아제 억제제의 개발로 이어지고 있다.Protein kinase is an enzyme that phosphorylates a protein molecule by attaching a phosphoryl group to a serine, threonine or tyrosine residue, and binding to ATP is essential for kinase activity. Recently, reports on the involvement of protein kinases in various human diseases including cancer and inflammatory diseases have led to the development of various protein kinase inhibitors.
이에, 본 발명자는 신규의 다중 표적 단백질 키나아제 억제제로 인돌린-2-온(indolin-2-one) 유도체인 KMU-시리즈 화합물을 합성하였고, 인간 단핵구 세포주 THP-1 및 원발성 인간 골관절염 섬유아세포 유사 활막세포를 이용하여 이의 항염증 메커니즘을 확인함으로써, 본 발명을 완성하였다.Accordingly, the present inventors synthesized KMU-series compounds, which are indolin-2-one derivatives, as novel multi-target protein kinase inhibitors, and synthesized human monocyte cell line THP-1 and primary human osteoarthritis fibroblast-like synovial membrane. By using the cell to confirm its anti-inflammatory mechanism, the present invention was completed.
본 발명은 하기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 제공한다.The present invention provides a compound represented by Formula 1 below or a pharmaceutically acceptable salt thereof.
<화학식 1><Formula 1>
Figure PCTKR2022003249-appb-img-000002
Figure PCTKR2022003249-appb-img-000002
상기 화학식 1에서, R1은 수소, 하이드록시, 할로겐, (C1-C4) 알킬, (C3-C10) 사이클로알킬, 또는 아다만타닐(adamantanyl)에서 선택될 수 있고, R2는 이미다졸일(imidazolyl), 피라졸일(pyrazolyl), 피롤일(pyrrolyl) 및 피롤리딘일(pyrrolidinyl)로 이루어진 헤테로 고리화합물에서 선택될 수 있다.In Formula 1, R 1 may be selected from hydrogen, hydroxy, halogen, (C1-C4) alkyl, (C3-C10) cycloalkyl, or adamantanyl, and R 2 is imidazolyl ( imidazolyl), pyrazolyl, pyrrolyl, and pyrrolidinyl.
바람직하게는, 상기 R1은 (C3-C8) 사이클로알킬, 또는 아다만타닐(adamantanyl)에서 선택될 수 있고, 상기 R2는 이미다졸일(imidazolyl) 또는 피롤일(pyrrolyl)에서 선택될 수 있으나, 이에 제한되는 것은 아니다.Preferably, R 1 may be selected from (C3-C8) cycloalkyl or adamantanyl, and R 2 may be selected from imidazolyl or pyrrolyl. , but is not limited thereto.
보다 바람직하게는, 상기 화합물 또는 이의 염은 (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(사이클로프로필아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopropylamino)pyrazin-2-yl)indolin-2-one] (4a; KMU-11170), (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(사이클로펜틸아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopentylamino)pyrazin-2-yl)indolin-2-one] (4b; KMU-11342), (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(사이클로헥실아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H-Imidazol-5-yl)methylene)-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one] (4c; KMU-11361), (Z)-3-((1H-피롤-2-일)메틸렌)-5-(6-(사이클로헥실아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H- pyrrol -2- yl ) methylene )-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one] (4c'; KMU-11426), (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(아다만탄-1-일아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H-Imidazol-5-yl)methylene)-5-(6-(adamantan-1-ylamino)pyrazin-2-yl)indolin-2-one] (4d; KMU-11421), 및 (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(사이클로헵틸아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H- Imidazol -5- yl ) methylene )-5-(6-(cycloheptylamino)pyrazin-2-yl)indolin-2-one] (4e; KMU-11427)로 이루어진 군에서 선택될 수 있으나, 이에 제한되는 것은 아니다.More preferably, the compound or salt thereof is (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopropylamino)pyrazin-2-yl)indoline- 2-one [(Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopropylamino)pyrazin-2-yl)indolin-2-one] (4a; KMU-11170) , (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopentylamino)pyrazin-2-yl)indolin-2-one [ (Z)-3- ((1H-imidazol-5-yl)methylene)-5-(6-(cyclopentylamino)pyrazin-2-yl)indolin-2-one] ( 4b; KMU-11342), (Z)-3-((1H -imidazol-5-yl)methylene)-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one [ (Z)-3-((1H-Imidazol-5-yl) methylene)-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one ] (4c; KMU-11361), (Z)-3-((1H-pyrrol-2-yl)methylene)- 5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one [ (Z)-3-((1H- pyrrol -2- yl ) methylene )-5-(6-(cyclohexylamino) pyrazin-2-yl) indolin-2-one ] (4c'; KMU-11426), (Z) -3-((1H-imidazol-5-yl) methylene) -5-(6-(adamantane) -1-ylamino) pyrazin-2-yl) indolin-2-one [ (Z)-3-((1H-Imidazol-5-yl)methylene)-5-(6-(adamantan-1-ylamino) pyrazin-2-yl)indolin-2-one ] (4d; KMU-11421), and (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cycloheptylamino) )pyrazin-2-yl)indolin-2-one [ (Z)-3-((1H- Imidazol -5- yl ) methylene )-5-(6-(cycloheptylamino)pyrazin-2-yl)indolin -2-one ] (4e; KMU-11427), but is not limited thereto.
본 발명에 있어서, 상기 화합물은 이와 동일한 효능을 갖는 범위 내에서 약학적으로 허용가능한 염의 형태로 사용할 수 있다.In the present invention, the compound may be used in the form of a pharmaceutically acceptable salt within a range having the same efficacy.
본 명세서에서, "약학적으로 허용가능한"이란, 상기 조성물에 노출되는 세포나 인간에게 독성이 없어, 인간에게 투여하기에 적합한 안전성 및 효능 프로파일을 갖는 염을 의미한다. As used herein, "pharmaceutically acceptable" means a salt that is not toxic to cells or humans exposed to the composition and has a safety and efficacy profile suitable for administration to humans.
상기 염은 약학적으로 허용가능한 염기성 염 또는 산성염 중 어느 하나의 형태로 사용할 수 있다. 염기성염은 유기 염기염, 무기 염기염 중 어느 하나의 형태로 사용할 수 있으며, 나트륨염, 칼륨염, 칼슘염, 리튬염, 마그네슘염, 세슘염, 아미늄염, 암모늄염, 트리에칠아미늄염 및 피리디늄염으로 이루어진 군에서 선택될 수 있다.The salt may be used in the form of either a pharmaceutically acceptable basic salt or acid salt. The basic salt can be used in the form of any one of an organic basic salt and an inorganic basic salt, and includes sodium salt, potassium salt, calcium salt, lithium salt, magnesium salt, cesium salt, aminium salt, ammonium salt, triethylaminium salt, and pyrol. It may be selected from the group consisting of dinium salts.
산성염은 유리산(free acid)에 의해 형성된 산부가염이 유용하다. 유리산으로는 무기산과 유기산을 사용할 수 있으며, 무기산으로는 염산, 브롬산, 황산, 아황산, 인산, 이중 인산, 질산 등을 사용할 수 있고, 유기산으로는 구연산, 초산, 말레산, 말산, 퓨마르산, 글루코산, 메탄설폰산, 벤젠설폰산, 캠퍼설폰산, 옥살산, 말론산, 글루타릭산, 아세트산, 글리콘산, 석신산, 타타르산, 4-톨루엔설폰산, 갈락투론산, 엠본산, 글루탐산, 시트르산, 아스파르탄산, 스테아르산 등을 사용할 수 있으나, 이에 제한되지 않고 당업계에서 통상적으로 사용되는 다양한 무기산 및 유기산을 이용하여 형성되는 염이 모두 포함될 수 있다.As the acid salt, an acid addition salt formed by a free acid is useful. Inorganic acids and organic acids can be used as free acids, and hydrochloric acid, hydrobromic acid, sulfuric acid, sulfurous acid, phosphoric acid, double phosphoric acid, and nitric acid can be used as inorganic acids, and citric acid, acetic acid, maleic acid, malic acid, and fumaric acid can be used as organic acids. , glucoic acid, methanesulfonic acid, benzenesulfonic acid, camphorsulfonic acid, oxalic acid, malonic acid, glutaric acid, acetic acid, glycolic acid, succinic acid, tartaric acid, 4-toluenesulfonic acid, galacturonic acid, embonic acid, Glutamic acid, citric acid, aspartic acid, stearic acid, etc. may be used, but are not limited thereto, and salts formed using various inorganic acids and organic acids commonly used in the art may all be included.
또한, 상기 화합물은 상기 염뿐만 아니라, 통상의 방법에 의해 제조될 수 있는 모든 염, 수화물, 용매화물, 유도체 등을 모두 포함할 수 있다. 부가염은 통상의 방법으로 제조할 수 있고, 수혼화성 유기용매, 예를 들면 아세톤, 메탄올, 에탄올, 또는 아세토니트릴 등에 녹여 과량의 유기염기를 가하거나 무기염기의 염기 수용액을 가한 후 침전시키거나 결정화시켜서 제조할 수 있다. 또는 이 혼합물에서 용매나 과량의 염기를 증발시킨 후 건조시켜서 부가염을 얻거나 또는 석출된 염을 흡인 여과시켜 제조할 수 있다.In addition, the compound may include all salts, hydrates, solvates, derivatives, and the like that can be prepared by conventional methods, as well as the salts. The addition salt can be prepared by a conventional method, and is dissolved in a water-miscible organic solvent such as acetone, methanol, ethanol, or acetonitrile, and an excess organic base is added or an aqueous solution of an inorganic base is added, followed by precipitation or crystallization. can be manufactured by Alternatively, the solvent or excess base may be evaporated from the mixture and then dried to obtain an addition salt, or the precipitated salt may be suction filtered.
본 발명에 있어서, 상기 화합물 또는 이의 염은 다양한 염증 관련 단백질 키나아제를 억제할 수 있다.In the present invention, the compound or salt thereof may inhibit various inflammation-related protein kinases.
바람직하게는, 상기 화합물 또는 이의 염은 MAPK (mitogen-activated protein kinase), TAK1 (TGF-beta activated kinase 1), Lck(lymphocyte-specific protein tyrosine kinase), TYK2 (non-receptor tyrosine-protein kinase 2), JAK3 (janus kinase 3), 및 Txk (tyrosine-protein kinase)로 이루어진 군에서 선택되는 하나 이상의 단백질 키나아제를 억제할 수 있으나, 이에 제한되는 것은 아니다.Preferably, the compound or salt thereof is MAPK (mitogen-activated protein kinase), TAK1 (TGF-beta activated kinase 1), Lck (lymphocyte-specific protein tyrosine kinase), TYK2 (non-receptor tyrosine-protein kinase 2) , JAK3 (janus kinase 3), and Txk (tyrosine-protein kinase) may inhibit one or more protein kinases selected from the group consisting of, but is not limited thereto.
본 발명의 일 실험예에 따르면, 상기 화합물은 THP-1 세포에서 세포 사멸, 분화, 면역, 염증 및 생존을 포함한 다양한 세포 과정과 관련된, 특히 전염증성 사이토카인의 LPS-TLR4-TAK1 신호전달과 관련된 TAK1 ERK, 및 JNK의 LPS-유도 인산화를 억제함을 확인할 수 있었다.According to one experimental example of the present invention, the compound is related to various cellular processes including apoptosis, differentiation, immunity, inflammation and survival in THP-1 cells, particularly related to LPS-TLR4-TAK1 signaling of pro-inflammatory cytokines. It was confirmed that TAK1 inhibits LPS-induced phosphorylation of ERK and JNK.
상기 화합물 또는 이의 염은 NLRP3 인플라마좀의 활성을 억제할 수 있으며, 다양한 사이토카인의 LPS-유도 상향 조절을 억제하는 바, 우수한 항염증 활성을 가질 수 있다. The compound or salt thereof may inhibit the activity of NLRP3 inflammasome and inhibit LPS-induced up-regulation of various cytokines, and thus may have excellent anti-inflammatory activity.
본 발명의 일 실험예에 따르면, 상기 화합물은 염증 반응의 중요한 매개체인, 산화 질소(NO) 생성에 필요한 촉매적 효소 계열에 속하는 iNOS를 억제하고, 염증성 질환, 종양성 질환 및 알츠하이머 질환과 같은 다양한 인간 질환과 관련된 COX-2를 선택적으로 억제하며, 염증 반응에서 사이토카인 및 인플라마좀과 같은 주요 인자를 조절하는 면역계의 주요 매개체인 NF-κB 인자와 관련된 LPS 유도 IKKα/β 및 NF-κB p65의 인산화, NF-κB p65의 핵 전좌(nuclear translocation), 및 IL-1β, TNF-α, IL-6의 상향조절을 억제함을 확인할 수 있었다. 반면, 상기 화합물은 THP-1 세포에서 LPS-매개 TLR4 및 MyD88의 상향 조절을 억제하지 않은 바, 이러한 결과는 상기 화합물이 THP-1 세포의 LPS 매개 염증 반응에서 TLR4/MyD88의 downstream에 항염증 효과를 가짐을 보여준다.According to one experimental example of the present invention, the compound inhibits iNOS belonging to the family of catalytic enzymes necessary for the production of nitric oxide (NO), an important mediator of the inflammatory response, and various diseases such as inflammatory diseases, neoplastic diseases and Alzheimer's disease. LPS-induced IKKα/β and NF-κB p65 associated with NF-κB factor, a key mediator of the immune system that selectively inhibits COX-2 associated with human diseases and regulates key factors such as cytokines and inflammasomes in the inflammatory response It was confirmed that phosphorylation of , nuclear translocation of NF-κB p65, and upregulation of IL-1β, TNF-α, and IL-6 were inhibited. On the other hand, the compound did not inhibit the LPS-mediated upregulation of TLR4 and MyD88 in THP-1 cells, and these results suggest that the compound has an anti-inflammatory effect downstream of TLR4/MyD88 in the LPS-mediated inflammatory response of THP-1 cells. shows that it has
또한, 상기 화합물의 항염증 효과를 다른 표적 항염증제와 비교한 결과, COX-2 억제제 (celecoxib), BTK 억제제 (ibrutinib), JAK 억제제 (ruxolitinib), 및 메토트레세이트를 포함하는 다른 약물과 비교하여 현저히 우수한 항염증 효과를 나타냄을 확인할 수 있었다.In addition, as a result of comparing the anti-inflammatory effect of the compound with other targeted anti-inflammatory drugs, COX-2 inhibitors (celecoxib), BTK inhibitors (ibrutinib), JAK inhibitors (ruxolitinib), and methotrexate were significantly superior to other drugs. It was confirmed that it exhibited an excellent anti-inflammatory effect.
이에, 상기 화합물 또는 이의 염은 염증성 질환 예방 또는 치료를 위한 조성물로 활용될 수 있다.Accordingly, the compound or salt thereof may be used as a composition for preventing or treating inflammatory diseases.
본 발명은 상기 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 염증성 질환 예방 또는 치료용 약학 조성물을 제공한다.The present invention provides a pharmaceutical composition for preventing or treating inflammatory diseases containing the compound or a pharmaceutically acceptable salt thereof as an active ingredient.
상기 염증성 질환은 골관절염, 류마티스관절염, 골다공증, 골 파제트병, 및 강직성척추염으로 이루어진 군에서 선택되는 어느 하나일 수 있으나, 이에 제한되는 것은 아니다.The inflammatory disease may be any one selected from the group consisting of osteoarthritis, rheumatoid arthritis, osteoporosis, Paget's disease of bone, and ankylosing spondylitis, but is not limited thereto.
본 발명의 일 실험예에 따르면, 지속적인 활막 염증 및 진행성 연골 분해를 특징으로 하는 골관절염 진행에서 중요한 역할을 하는 골관절염 FLS에서 상기 화합물이 LPS-유도 염증 반응을 억제함을 확인할 수 있는 바, 이를 활용하여 골관절염을 예방, 치료 또는 개선시킬 수 있다.According to an experimental example of the present invention, it can be confirmed that the compound inhibits the LPS-induced inflammatory response in osteoarthritis FLS, which plays an important role in the progression of osteoarthritis characterized by continuous synovial inflammation and progressive cartilage degradation. Osteoarthritis can be prevented, treated or ameliorated.
본 발명에 따른 약학 조성물은 약학적 분야의 통상적인 방법에 따라 제조될 수 있다. 상기 약학 조성물은 제형에 따라 약학적으로 허용가능한 적절한 담체와 배합될 수 있고, 필요에 따라, 부형제, 희석제, 분산제, 유화제, 완충제, 안정제, 결합제, 붕해제, 용제 등을 더 포함하여 제조될 수 있다. 상기 적절한 담체 등은 본 발명에 따른 화합물, 또는 이의 약학적으로 허용가능한 염의 활성 및 특성을 저해하지 않는 것으로, 투여 형태 및 제형에 따라 달리 선택될 수 있다.The pharmaceutical composition according to the present invention can be prepared according to conventional methods in the pharmaceutical field. The pharmaceutical composition may be formulated with an appropriate pharmaceutically acceptable carrier according to the formulation and, if necessary, may further contain excipients, diluents, dispersants, emulsifiers, buffers, stabilizers, binders, disintegrants, solvents, and the like. there is. The appropriate carrier and the like do not inhibit the activity and characteristics of the compound according to the present invention or a pharmaceutically acceptable salt thereof, and may be selected differently depending on the dosage form and dosage form.
상기 약학 조성물에 포함될 수 있는 담체, 부형제, 희석제 등으로는 락토오스, 덱스트로오스, 수크로오스, 소르비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로오스, 메틸 셀룰로오스, 미정질 셀룰로오스, 폴리비닐 피롤리돈, 물, 메틸히드록시 벤조에이트, 프로필히드록시 벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유 등이 있다. 상기 조성물을 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제될 수 있다.Examples of carriers, excipients, and diluents that may be included in the pharmaceutical composition include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxy benzoate, propylhydroxy benzoate, talc, magnesium stearate and mineral oil. When formulating the composition, it may be prepared using diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants.
상기 약학 조성물은 어떠한 제형으로도 적용될 수 있고, 상세하게는 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 현탁액, 에멀젼, 시럽, 에어로졸 등의 경구형 제형, 외용제, 좌제 및 멸균 주사용액의 형태로 제형화하여 사용될 수 있으며, 바람직하게는 경구 투여에 적합한 단위투여형의 제제로 제형화시켜 사용될 수 있다.The pharmaceutical composition can be applied in any dosage form, and specifically, according to conventional methods, oral dosage forms such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, external preparations, suppositories and sterile injection solutions. It may be formulated into a form and used, preferably formulated into a unit dosage form suitable for oral administration.
보다 상세하게는, 상기 경구형 제형 중 고형 제형은 정제, 환제, 산제, 과립제, 캡슐제 등의 형태로, 적어도 하나 이상의 부형제, 예를 들면, 전분, 칼슘카보네이트, 수크로스, 락토오스, 솔비톨, 만니톨, 셀룰로오스, 젤라틴 등을 섞어 조제할 수 있고, 단순한 부형제 이외에 마그네슘 스테아레이트, 탈크 같은 윤활제들도 포함될 수 있다. 또한, 캡술제형의 경우 상기 언급한 물질 외에도 지방유와 같은 액체 담체를 더 포함할 수 있다.More specifically, the solid dosage form of the oral dosage form is in the form of tablets, pills, powders, granules, capsules, etc., and includes at least one excipient such as starch, calcium carbonate, sucrose, lactose, sorbitol, and mannitol. , cellulose, gelatin, etc. may be mixed, and lubricants such as magnesium stearate and talc may be included in addition to simple excipients. In addition, in the case of a capsule formulation, a liquid carrier such as fatty oil may be further included in addition to the above-mentioned materials.
상기 경구형 제형 중 액상 제형은 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.Among the oral formulations, liquid formulations include suspensions, solutions for internal use, emulsions, syrups, etc. In addition to water and liquid paraffin, which are commonly used simple diluents, various excipients such as wetting agents, sweeteners, aromatics, and preservatives may be included. there is.
상기 비경구 제형은 멸균된 수용액, 비수성용제, 현탁제, 유제, 주사제, 동결건조 제제, 좌제 등이 포함될 수 있다. 비수성용제, 현탁제로는 프로필렌글리콜, 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔(witepsol), 마크로골, 트윈 61, 카카오지, 라우린지, 글리세로제라틴 등이 사용될 수 있다. 이에 제한되지 않고, 당업계에 널리 공지된 적합한 제제를 모두 사용 가능하다.The parenteral formulation may include a sterilized aqueous solution, a non-aqueous solvent, a suspension, an emulsion, an injection, a lyophilized formulation, a suppository, and the like. Propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate may be used as non-aqueous solvents and suspending agents. As a base for the suppository, witepsol, macrogol, Tween 61, cacao butter, laurin fat, glycerogeratin, and the like may be used. It is not limited thereto, and all suitable agents well known in the art may be used.
또한, 상기 약학 조성물은 치료 효능의 증진을 위해 항산화제를 더 첨가할 수 있다. 상기 항산화제로는 티아민(thiamin, 비타민 B1), 리보플라빈(riboflavin, 비타민 B2), 나이아신(niacin, 비타민 B3), 판토펜산(pantothenic acid, 비타민 B5), 피리독신(pyridoxine, 비타민 B6) 및 코발라민(cobalamin, 비타민 B12) 등의 비타민 B군의 화합물과 비타민 C, 비타민 D, 비타민 E 등이 사용될 수 있으나, 이에 제한되지 않고, 당업계에 널리 공지된 적합한 제제를 모두 사용 가능하다.In addition, an antioxidant may be further added to the pharmaceutical composition to enhance therapeutic efficacy. The antioxidants include thiamin (vitamin B1), riboflavin (vitamin B2), niacin (vitamin B3), pantothenic acid (vitamin B5), pyridoxine (vitamin B6) and cobalamin (cobalamin, Vitamin B group compounds such as vitamin B12), vitamin C, vitamin D, vitamin E, etc. may be used, but are not limited thereto, and suitable preparations widely known in the art may be used.
본 발명에 따른 약학 조성물은 약학적으로 유효한 양으로 투여될 수 있다. The pharmaceutical composition according to the present invention can be administered in a pharmaceutically effective amount.
본 명세서에서, "약학적으로 유효한 양"이란, 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분하며 부작용을 일으키지 않을 정도의 양을 의미한다.In the present specification, "pharmaceutically effective amount" means an amount that is sufficient to treat a disease with a reasonable benefit/risk ratio applicable to medical treatment and does not cause side effects.
상기 약학 조성물의 유효 용량 수준은 사용 목적, 환자의 연령, 성별, 체중 및 건강 상태, 질환의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 방법, 투여 시간, 투여 경로 및 배출 비율, 치료기간, 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 달리 결정될 수 있다. 예를 들어, 일정하지는 않지만 일반적으로 0.001 내지 100mg/kg으로, 바람직하게는 0.01 내지 10mg/kg을 일일 1회 내지 수회 투여될 수 있다. 상기 투여량은 어떠한 면으로든 본 발명의 범위를 한정하는 것은 아니다.The effective dosage level of the pharmaceutical composition depends on the purpose of use, the patient's age, sex, weight and health condition, disease type, severity, drug activity, drug sensitivity, administration method, administration time, administration route and excretion rate, treatment Duration, combination, or factors including drugs used concurrently and other factors well known in the medical arts may be determined differently. For example, although not constant, generally 0.001 to 100 mg/kg, preferably 0.01 to 10 mg/kg, may be administered once or several times a day. The dosage is not intended to limit the scope of the present invention in any way.
상기 약학 조성물은 염증성 질환이 발생할 수 있는 임의의 동물에 투여될 수 있고, 상기 동물은 예를 들어, 인간 및 영장류뿐만 아니라 소, 돼지, 말, 개 등의 가축 등을 포함할 수 있다.The pharmaceutical composition may be administered to any animal that may develop an inflammatory disease, and the animal may include, for example, humans and primates as well as livestock such as cattle, pigs, horses, and dogs.
상기 약학 조성물은 제제 형태에 따른 적당한 투여 경로로 투여될 수 있고, 목적 조직에 도달할 수 있는 한 경구 또는 비경구의 다양한 경로를 통하여 투여될 수 있다. 투여 방법은 특히 한정할 필요 없이, 예를 들면, 경구, 직장 또는 정맥, 근육, 피부 도포, 호흡기내 흡입, 자궁내 경막 또는 뇌혈관내(intracere-broventricular) 주사 등의 통상적인 방법으로 투여될 수 있다.The pharmaceutical composition may be administered by an appropriate administration route according to the formulation form, and may be administered through various oral or parenteral routes as long as it can reach the target tissue. The method of administration is not particularly limited, and may be administered by conventional methods such as oral, rectal or intravenous, intramuscular, skin application, intraventricular inhalation, intrauterine dural or intracerebroventricular injection. there is.
상기 약학 조성물은 염증성 질환의 예방 또는 치료를 위하여 단독으로 사용될 수 있고, 수술 또는 다른 약물치료 등과 병용하여 사용될 수 있다.The pharmaceutical composition may be used alone for the prevention or treatment of inflammatory diseases, or may be used in combination with surgery or other drug treatment.
또한, 본 발명은 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 염증성 질환 예방 또는 개선용 건강기능식품 조성물을 제공한다.In addition, the present invention provides a health functional food composition for preventing or improving inflammatory diseases containing a compound or a pharmaceutically acceptable salt thereof as an active ingredient.
상기 염증성 질환은 골관절염, 류마티스관절염, 골다공증, 골 파제트병, 및 강직성척추염으로 이루어진 군에서 선택되는 어느 하나일 수 있으나, 이에 제한되는 것은 아니다.The inflammatory disease may be any one selected from the group consisting of osteoarthritis, rheumatoid arthritis, osteoporosis, Paget's disease of bone, and ankylosing spondylitis, but is not limited thereto.
이에 상응하는 특징들은 상술된 부분에서 대신할 수 있다.Corresponding features may be substituted in the foregoing.
본 발명에 따른 건강기능식품 조성물에 있어서, 상기 건강기능식품은 염증성 질환의 예방 또는 개선 목적으로, 분말, 과립, 정제, 캡슐, 시럽 또는 음료 등으로 제조될 수 있다. 상기 건강기능식품이 취할 수 있는 형태에는 제한이 없으며, 상기 약학 조성물과 동일한 방식으로 제제화되어 기능성 식품으로 이용하거나, 각종 식품에 첨가될 수 있다. In the health functional food composition according to the present invention, the health functional food may be prepared in powder, granule, tablet, capsule, syrup or beverage for the purpose of preventing or improving inflammatory diseases. There is no limit to the form that the health functional food can take, and it can be formulated in the same way as the pharmaceutical composition and used as a functional food or added to various foods.
상기 건강기능식품 조성물에 있어서, 상기 건강기능식품은 통상적인 의미의 식품을 모두 포함할 수 있다. 예를 들어, 음료 및 각종 드링크, 과실 및 그의 가공식품(과일통조림, 잼 등), 어류, 육류 및 그 가공식품(햄, 베이컨 등), 빵류 및 면류, 쿠키 및 스낵류, 유제품(버터, 치즈 등) 등이 가능하며, 통상적인 의미에서의 기능성 식품을 모두 포함할 수 있다. 또한, 동물을 위한 사료로 이용되는 식품도 포함할 수 있다.In the health functional food composition, the health functional food may include all foods in a conventional sense. For example, beverages and various drinks, fruits and their processed foods (canned fruit, jam, etc.), fish, meat and their processed foods (ham, bacon, etc.), breads and noodles, cookies and snacks, dairy products (butter, cheese, etc.) ), etc., and may include all functional foods in a conventional sense. In addition, food used as feed for animals may also be included.
상기 건강기능식품 조성물은 당업계에서 통상적으로 사용되는 식품학적으로 허용가능한 식품 첨가제(식품 첨가물) 및 적절한 기타 보조 성분을 더 포함하여 제조될 수 있다. 식품 첨가물로서의 적합 여부는 다른 규정이 없는 한, 식품의약품안전처에 승인된 식품첨가물공전의 총칙 및 일반시험법 등에 따라 해당 품목에 관한 규격 및 기준에 의하여 판정할 수 있다. 상기 '식품첨가물공전'에 수재된 품목으로는 예를 들어, 케톤류, 글리신, 구연산칼슘, 니코틴산, 계피산 등의 화학적 합성물; 감색소, 감초추출물, 결정셀룰로오스, 고량색소, 구아검 등의 천연첨가물; L-글루타민산나트륨 제제, 면류첨가알칼리제, 보존료 제제, 타르색소제제 등의 혼합 제제류 등을 들 수 있다. The health functional food composition may be prepared by further including food additives (food additives) commonly used in the art and appropriate other auxiliary components. The suitability as a food additive can be determined according to the standards and standards for the item in accordance with the general rules of the Food Additive Code and general test methods approved by the Ministry of Food and Drug Safety, unless otherwise specified. Examples of the items listed in the 'Food Additive Code' include, for example, chemical compounds such as ketones, glycine, calcium citrate, nicotinic acid, and cinnamic acid; natural additives such as persimmon pigment, licorice extract, crystalline cellulose, kaoliang pigment, and guar gum; mixed preparations such as sodium L-glutamate preparations, noodle-added alkali preparations, preservative preparations, and tar color preparations; and the like.
상기 기타 보조 성분은 예를 들어, 향미제, 천연 탄수화물, 감미제, 비타민, 전해질, 착색제, 펙트산, 알긴산, 유기산, 보호성 콜로이드 증점제, pH 조절제, 안정화제, 방부제, 글리세린, 알콜, 탄산화제 등을 추가로 함유할 수 있다. 특히, 상기 천연 탄수화물로는 포도당, 과당과 같은 모노사카라이드, 말토스, 수크로오스와 같은 디사카라이드, 및 덱스트린, 사이클로덱스트린과 같은 폴리사카라이드, 자일리톨, 소르비톨, 에리트리톨 등의 당알콜을 사용할 수 있으며, 감미제로서는 타우마틴, 스테비아 추출물과 같은 천연 감미제나 사카린, 아스파르탐과 같은 합성 감미제 등을 사용할 수 있다.The other auxiliary ingredients include, for example, flavoring agents, natural carbohydrates, sweeteners, vitamins, electrolytes, colorants, pectic acid, alginic acid, organic acids, protective colloidal thickeners, pH adjusting agents, stabilizers, preservatives, glycerin, alcohols, carbonating agents, etc. may additionally contain. In particular, as the natural carbohydrate, monosaccharides such as glucose and fructose, disaccharides such as maltose and sucrose, polysaccharides such as dextrin and cyclodextrin, and sugar alcohols such as xylitol, sorbitol, and erythritol may be used. As the sweetener, natural sweeteners such as thaumatin and stevia extract or synthetic sweeteners such as saccharin and aspartame may be used.
본 발명에 따른 건강기능식품에 함유된 상기 화합물 또는 이의 염의 유효용량은 염증성 질환 예방 또는 개선 등 그 사용 목적에 따라 적절하게 조절될 수 있다. An effective dose of the compound or salt thereof contained in the health functional food according to the present invention may be appropriately adjusted according to the purpose of use, such as preventing or improving inflammatory diseases.
상기 건강기능식품 조성물은 식품을 원료로 하여 일반 약품의 장기 복용 시 발생할 수 있는 부작용 등이 없는 장점이 있고, 휴대성이 뛰어나, 염증성 질환 예방 또는 개선을 위한 보조제로 섭취될 수 있다.The health functional food composition uses food as a raw material and has the advantage of not having side effects that can occur when taking general medicines for a long time, and has excellent portability, so it can be taken as an adjuvant for preventing or improving inflammatory diseases.
이하, 본 발명의 이해를 돕기 위하여 실시예를 들어 상세하게 설명하기로 한다. 다만 하기의 실시예는 본 발명의 내용을 예시하는 것일 뿐 본 발명의 범위가 하기 실시예에 한정되는 것은 아니다. 본 발명의 실시예는 당업계에서 평균적인 지식을 가진 자에게 본 발명을 보다 완전하게 설명하기 위해 제공되는 것이다.Hereinafter, examples will be described in detail to aid understanding of the present invention. However, the following examples are merely illustrative of the contents of the present invention, but the scope of the present invention is not limited to the following examples. The embodiments of the present invention are provided to more completely explain the present invention to those skilled in the art.
<< 실시예Example 1> 1> KMUKMU -series (-series ( KMUKMU -11421, 11426, 11427, 11361, 11342, 11170) 화합물의 합성-11421, 11426, 11427, 11361, 11342, 11170) Synthesis of compounds
1. 합성 시약 1. Synthesis Reagents 및 기기and device
모든 반응은 제조업체가 명시한 적절한 조건에서 추가 정제없이 시판되는 시약 및 용매를 사용하여 수행되었다. CEM Discover BenchMate는 마이크로파-지원 반응에 사용되었다. 반응의 완료는 E. Merck 실리카 겔 F254 TLC 플레이트에서 모니터링되었다. 합성된 화합물의 정제는 Merck Silica Gel 60 (230-400 mesh)을 사용한 플래쉬 컬럼 크로마토그래프로 수행되었다. 합성된 화합물은 1H on Bruker AVANCE 400 (1H: 400 MHz) 및 JEOL ECA 500 (1H: 500 MHz) spectrometer로 특정되었고, 화학적 이동(chemical shift) δ 값은 TMS를 참조 표준으로 하여 ppm 단위로 측정되었다. 질량 스펙트럼은 Waters ACQUI-TY UPLC, Micromass Quattro microTM API를 사용하여 획득하였다.All reactions were performed using commercially available reagents and solvents without further purification under appropriate conditions specified by the manufacturer. A CEM Discover BenchMate was used for microwave-assisted reactions. Completion of the reaction was monitored on E. Merck silica gel F254 TLC plates. Purification of the synthesized compound was performed by flash column chromatography using Merck Silica Gel 60 (230-400 mesh). The synthesized compound was characterized by 1 H on Bruker AVANCE 400 ( 1 H: 400 MHz) and JEOL ECA 500 ( 1 H: 500 MHz) spectrometer, and the chemical shift δ value was in ppm units with TMS as the reference standard. was measured as Mass spectra were acquired using Waters ACQUI-TY UPLC, Micromass Quattro microTM API.
2. 합성 방법2. Synthesis method
KMU-시리즈 화합물은 도 1에 나타난 바와 같이 5-브로모인돌린-2,3-디온(5-bromoindoline-2,3-dione)에서 하기와 같은 조건에서 합성되었다: As shown in Figure 1, the KMU-series compounds were synthesized from 5-bromoindoline-2,3-dione under the following conditions:
화합물 1은 환원 및 보릴화(borylation)로 얻어졌고 [(a) i) NH2NH2, NaOH, EtOH; ii) bis(pincholato)diboron, PdCl2(dppf), KOAc, 1,4-dioxane: EtOH(1: 1.5), microwave], 화합물 1과 피라진 유도체인 화합물 2[(b) K2CO3, amines or alcohols, DMF]의 Suzuki 커플링 반응으로 화합물 3이 수득되었다[(c) PdCl2(PPh3)2, 2M K2CO3, 1,4-dioxane : EtOH (1: 1.5), microwave]. 마지막으로, 1H-이미다졸-4-카보알데하이드(1H-imidazole-4-carbaldehyde)와의 크네페나겔 축합(Knoevenagel condensation) 반응을 통해 KMU-시리즈 화합물을 합성하였다 [(d) piperidine, 1H-imidazole-4-carbaldehyde, EtOH, microwave]. Compound 1 was obtained by reduction and borylation [(a) i) NH 2 NH 2 , NaOH, EtOH; ii) bis(pincholato)diboron, PdCl 2 (dppf), KOAc, 1,4-dioxane: EtOH (1: 1.5), microwave], compound 1 and pyrazine derivative compound 2 [(b) K 2 CO 3 , amines or alcohols, DMF] to obtain compound 3 [(c) PdCl 2 (PPh 3 ) 2 , 2M K 2 CO 3 , 1,4-dioxane : EtOH (1: 1.5), microwave]. Finally, KMU-series compounds were synthesized through a Knoevenagel condensation reaction with 1H-imidazole-4-carbaldehyde [(d) piperidine, 1H-imidazole- 4-carbaldehyde, EtOH, microwave].
2-1. 5-(4,4,5,5-2-1. 5-(4,4,5,5- 테트라메틸tetramethyl -1,3,2--1,3,2- 디옥사보롤란Dioxaborolane -2-일)-2 days) 인돌린indoline -2-온 [-2-on [ 5-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)indolin-2-one5-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)indolin-2-one ] 합성 : 화합물 (1)] Synthesis: Compound (1)
<화합물 1><Compound 1>
Figure PCTKR2022003249-appb-img-000003
Figure PCTKR2022003249-appb-img-000003
마이크로파 용기를 5-브로모인돌린-2,3-디온(5-bromoindoline-2,3-dione; 0.50 g, 2.2 mmol), 하이드라진 수화물(hydrazine hydrate; 0.14 g, 4.4 mmol), 및 에탄올(EtOH, 2 mL)로 채웠다. 상기 혼합물에 100℃에서 10분간 100 W 마이크로파를 조사하였다. 수산화나트륨(sodium hydroxide; 0.18 g, 4.4 mmol) 첨가 후, 혼합물에 80℃에서 10분간 100 W 마이크로파를 조사하였다. 2 M 염산(HCl)을 사용하여 상기 혼합물을 산성화하고 냉수를 첨가하여 형성된 침전물을 여과를 통해 수집한 다음, 2 M HCl로 세척 후 물로 다시 세척하였다. 진공 하에서 건조하여 0.42 g의 5-브로모인돌린-2-온(5-bromoindolin-2-one, 89% 수율)을 얻었고, 이는 추가 정제없이 다음 단계에 사용되었다. A microwave vessel was prepared with 5-bromoindoline-2,3-dione (0.50 g, 2.2 mmol), hydrazine hydrate (0.14 g, 4.4 mmol), and ethanol (EtOH, 2 mL) was filled. 100 W microwave was irradiated to the mixture at 100° C. for 10 minutes. After adding sodium hydroxide (0.18 g, 4.4 mmol), the mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The mixture was acidified with 2 M hydrochloric acid (HCl) and cold water was added to form a precipitate, which was collected by filtration, washed with 2 M HCl and washed again with water. Drying under vacuum gave 0.42 g of 5-bromoindolin-2-one (89% yield), which was used in the next step without further purification.
마이크로파 용기에 5-브로모인돌린-2-온(5-bromoindoline-2-one; 0.40 g, 1.9 mmol) 및 1,4-다이옥산(1,4-dioxane; 2.0 mL)을 채운 다음, 상기 혼합물에 비스(피나콜라토)디보론[bis(pinacolato)diboron; 0.72 g, 3.8 mmol] 및 아세트산 칼륨(potassium acetate, KOAc; 0.56 g, 5.7 mmol)을 추가하여 5분 동안 N2 가스로 제거(purge)하였다. 반응 혼합물에 N2 가스 내 [1,1′-비스(디페닐포스피노)페로센]디클로로팔라듐(II)([1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium (II), PdCl2(dppf); 0.069 g, 0.094 mmol)를 첨가하고, 혼합물에 110℃에서 10분간 100 W 마이크로파를 조사하였다. 진공에서 용매를 제거한 후, 잔여물은 1:1 헥산(HEX)/에틸아세테이트(EA)를 사용한 실리카 겔 크로마토그래피로 정제하여, 0.410 g (82 %)의 표제 화합물 (1)을 수득하였다.After filling 5-bromoindoline-2-one (0.40 g, 1.9 mmol) and 1,4-dioxane (2.0 mL) in a microwave container, add the mixture to the mixture. bis (pinacolato) diboron [bis (pinacolato) diboron; 0.72 g, 3.8 mmol] and potassium acetate (KOAc; 0.56 g, 5.7 mmol) were added and purged with N 2 gas for 5 minutes. [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), PdCl 2 (dppf) in N 2 gas in the reaction mixture ; After removal of the solvent in vacuo, the residue was purified by silica gel chromatography using 1:1 hexane (HEX)/ethyl acetate (EA) to give 0.410 g (82%) of the title compound (1).
2-2-1. 6-2-2-1. 6- 클로로Chloro -N--N- 사이클로프로필피라진Cyclopropylpyrazine -2--2- 아민amine [ [ 6-6- ChloroChloro -N-cyclopropylpyrazin-2-amine-N-cyclopropylpyrazin-2-amine ] 합성 : 화합물 (2a)] Synthesis: compound (2a)
<화합물 2a><Compound 2a>
Figure PCTKR2022003249-appb-img-000004
Figure PCTKR2022003249-appb-img-000004
사이클로프로필아민(cyclopropylamine; 5.1 mmol, 0.35 mL) 및 탄산칼륨(potassium carbonate, K2CO3; 6.7 mmol, 0.93 g)을 5.0 mL의 N,N-디메틸포름아미드(N,N-dimethylformamide, DMF)에 첨가하고 이 혼합물을 30분 동안 실온에서 교반하였다. 2,6-디클로로피라진(2,6-dichloropyrazine; 3.4 mmol, 0.50 g) 첨가 후, 반응 혼합물을 실온에서 밤새 교반한 다음, 진공에서 용매를 제거하였다. 잔여물을 디클로로메탄(dichloromethane, DCM)으로 처리하고 셀라이트(celite)를 이용하여 여과한 다음, 여액을 수집하고 진공에서 용매를 제거하였다. 최종 잔여물은 5:1 DCM/EA를 사용한 실리카 겔 크로마토그래피로 정제하여, 0.21 g (36 %)의 표제 화합물 (2a)를 수득하였다. Cyclopropylamine (5.1 mmol, 0.35 mL) and potassium carbonate (K 2 CO 3 ; 6.7 mmol, 0.93 g) were mixed with 5.0 mL of N,N-dimethylformamide (DMF). was added and the mixture was stirred at room temperature for 30 minutes. After addition of 2,6-dichloropyrazine (3.4 mmol, 0.50 g), the reaction mixture was stirred at room temperature overnight, then the solvent was removed in vacuo. The residue was treated with dichloromethane (DCM) and filtered using celite, then the filtrate was collected and the solvent removed in vacuo. The final residue was purified by silica gel chromatography using 5:1 DCM/EA to give 0.21 g (36%) of the title compound (2a).
2-2-2. 6-2-2-2. 6- 클로로Chloro -N--N- 사이클로펜틸피라진Cyclopentylpyrazine -2--2- 아민amine [ [ 6-6- ChloroChloro -N-cyclopentylpyrazin-2-amine-N-cyclopentylpyrazin-2-amine ] 합성 : 화합물 (2b)] Synthesis: compound (2b)
<화합물 2b><Compound 2b>
Figure PCTKR2022003249-appb-img-000005
Figure PCTKR2022003249-appb-img-000005
사이클로펜틸아민(cyclopentylamine; 5.0 mmol, 0.50 mL) 및 K2CO3 (6.7 mmol, 0.93 g)를 5.0 mL의 DMF에 첨가하고, 이 혼합물을 실온에서 30분 동안 교반하였다. 2,6-디클로로피라진 (3.4 mmol, 0.50 g)을 첨가 후, 반응 혼합물을 실온에서 밤새 교반한 다음, 진공에서 용매를 제거하였다. 잔여물을 DCM으로 처리하고 셀라이트를 이용하여 여과한 다음, 여액을 수집하고 진공에서 용매를 제거하였다. 최종 잔여물은 5:1 DCM/EA를 사용한 실리카 겔 크로마토그래피로 정제하여, 0.43 g (65 %)의 표제 화합물 (2b)를 수득하였다.Cyclopentylamine (5.0 mmol, 0.50 mL) and K 2 CO 3 (6.7 mmol, 0.93 g) were added to 5.0 mL of DMF and the mixture was stirred at room temperature for 30 minutes. After adding 2,6-dichloropyrazine (3.4 mmol, 0.50 g), the reaction mixture was stirred at room temperature overnight, then the solvent was removed in vacuo. The residue was treated with DCM and filtered using celite, then the filtrate was collected and the solvent removed in vacuo. The final residue was purified by silica gel chromatography using 5:1 DCM/EA to give 0.43 g (65%) of the title compound (2b).
2-2-3. 6-2-2-3. 6- 클로로Chloro -N--N- 사이클로헥실피라진Cyclohexylpyrazine -2--2- 아민amine [ [ 6-6- ChloroChloro -N-cyclohexylpyrazin-2-amine-N-cyclohexylpyrazin-2-amine ] 합성 : 화합물 (2c)] Synthesis: compound (2c)
<화합물 2c><Compound 2c>
Figure PCTKR2022003249-appb-img-000006
Figure PCTKR2022003249-appb-img-000006
사이클로헥실아민(cyclohexylamine; 5.0 mmol, 0.58 mL) 및 K2CO3 (6.7 mmol, 0.93 g)를 5.0 mL의 DMF에 첨가하고, 이 혼합물을 실온에서 30분 동안 교반하였다. 2,6-디클로로피라진 (3.4 mmol, 0.50 g)을 첨가 후, 반응 혼합물을 실온에서 밤새 교반한 다음, 진공에서 용매를 제거하였다. 잔여물을 DCM으로 처리하고 셀라이트를 이용하여 여과한 다음, 여액을 수집하고 진공에서 용매를 제거하였다. 최종 잔여물은 40:1 DCM/EA를 사용한 실리카 겔 크로마토그래피로 정제하여, 0.25 g (35 %)의 표제 화합물 (2c)를 수득하였다.Cyclohexylamine (5.0 mmol, 0.58 mL) and K 2 CO 3 (6.7 mmol, 0.93 g) were added to 5.0 mL of DMF, and the mixture was stirred at room temperature for 30 minutes. After adding 2,6-dichloropyrazine (3.4 mmol, 0.50 g), the reaction mixture was stirred at room temperature overnight, then the solvent was removed in vacuo. The residue was treated with DCM and filtered using celite, then the filtrate was collected and the solvent removed in vacuo. The final residue was purified by silica gel chromatography using 40:1 DCM/EA to give 0.25 g (35%) of the title compound (2c).
2-2-4. N-(2-2-4. N-( 아다만탄Adamantane -1-일)-6--1-day)-6- 클로로피라진Chloropyrazine -2--2- 아민amine [ [ N-(N-( AdamantanAdamantan -1--One- ylyl )-6-chloropyrazin-2-amine)-6-chloropyrazin-2-amine ] 합성 : 화합물 (2d)] synthesis: compound (2d)
<화합물 2d><Compound 2d>
Figure PCTKR2022003249-appb-img-000007
Figure PCTKR2022003249-appb-img-000007
2,6-디클로로피라진 (1.5 mmol, 0.22 g)에 용해된 톨루엔(toluene; 50 mL) 용액에 1-아다만탄아민(1-adamantanamine; 0.99 mmol, 0.15 g) 및 칼륨터트-부티르산염(potassium tert-butoxide, KOtbu; 4.0 mmol, 0.45 g)을 실온에서 첨가하여 아르곤(Ar) 가스 하에서 10분 동안 환류시켰다. (2-바이페닐)터트-부틸포스핀[(2-biphenyl)di-tert-butylphosphine (JohnPhos); 0.027 mmol, 0.0079 g] 및 트리스(디벤질리덴아세톤)디팔라듐(0)[tris(dibenzylideneacetone)dipalladium(0) (Pd2(dba)3); 0.026 mmol, 0.024 g]을 첨가한 후, 반응 혼합물을 아르곤 가스에서 4시간 동안 환류시킨 다음, 진공에서 용매를 제거하였다. 잔여물을 EA로 처리하고 셀라이트를 이용하여 여과한 다음, 여액을 수집하고 감압 하에서 용매를 제거하였다. 최종 잔여물은 실리카 겔 크로마토그래피 DCM으로 정제하여, 0.033 g (13 %)의 표제 화합물 (2d)를 수득하였다.1-adamantanamine (1-adamantanamine; 0.99 mmol, 0.15 g) and potassium tert-butyrate (potassium tert-butoxide, KOtbu; 4.0 mmol, 0.45 g) was added at room temperature and refluxed for 10 minutes under argon (Ar) gas. (2-biphenyl)tert-butylphosphine [(2-biphenyl)di-tert-butylphosphine (JohnPhos); 0.027 mmol, 0.0079 g] and tris(dibenzylideneacetone)dipalladium(0) (Pd 2 (dba) 3 ); 0.026 mmol, 0.024 g], the reaction mixture was refluxed in argon gas for 4 hours and then the solvent was removed in vacuo. The residue was treated with EA and filtered using celite, then the filtrate was collected and the solvent was removed under reduced pressure. The final residue was purified by silica gel chromatography DCM to give 0.033 g (13%) of the title compound (2d).
2-2-5. 6-2-2-5. 6- 클로로Chloro -N--N- 사이클로헵틸피라진Cycloheptylpyrazine -2--2- 아민amine [ [ 6-6- ChloroChloro -N-cycloheptylpyrazin-2-amine-N-cycloheptylpyrazin-2-amine ] 합성 : 화합물 (2e)] synthesis: compound (2e)
<화합물 2e><Compound 2e>
Figure PCTKR2022003249-appb-img-000008
Figure PCTKR2022003249-appb-img-000008
사이클로헵틸아민(cycloheptylamine; 4.0 mmol, 0.31 mL) 및 K2CO3 (6.7 mmol, 0.93 g)를 5.0 mL의 DMF에 첨가하고, 이 혼합물을 실온에서 30분 동안 교반하였다. 2,6-디클로로피라진 (3.4 mmol, 0.50 g)을 첨가 후, 반응 혼합물을 실온에서 밤새 교반한 다음, 진공에서 용매를 제거하였다. 잔여물을 DCM으로 처리하고 셀라이트를 이용하여 여과한 다음, 여액을 수집하고 진공에서 용매를 제거하였다. 최종 잔여물은 실리카 겔 크로마토그래피 DCM으로 정제하여, 0.33 g (44 %)의 표제 화합물 (2e)를 수득하였다.Cycloheptylamine (4.0 mmol, 0.31 mL) and K 2 CO 3 (6.7 mmol, 0.93 g) were added to 5.0 mL of DMF and the mixture was stirred at room temperature for 30 minutes. After adding 2,6-dichloropyrazine (3.4 mmol, 0.50 g), the reaction mixture was stirred at room temperature overnight, then the solvent was removed in vacuo. The residue was treated with DCM and filtered using celite, then the filtrate was collected and the solvent removed in vacuo. The final residue was purified by silica gel chromatography DCM to give 0.33 g (44%) of the title compound (2e).
2-3-1. 5-(6-(2-3-1. 5-(6-( 사이클로프로필아미노cyclopropylamino )) 피라진pyrazine -2-일)-2 days) 인돌린indoline -2-온 [-2-on [ 5-(6-(Cyclopropylamino)pyrazin-2-yl)indolin-2-one5-(6-(Cyclopropylamino)pyrazin-2-yl)indolin-2-one ] 합성 : 화합물 (3a)] Synthesis: compound (3a)
<화합물 3a><Compound 3a>
Figure PCTKR2022003249-appb-img-000009
Figure PCTKR2022003249-appb-img-000009
마이크로파 용기를 화합물 1 (0.45 g, 1.8 mmol), 화합물 2a (0.20 g, 1.2 mmol), 1,4-다이옥산 (2.0 mL), 에탄올 (0.40 mL), 및 2 M 수성 탄산칼륨 (1.8 mL, 2.54 mmol)으로 채웠다. N2 분위기에서 비스(트리페닐포스핀)팔라듐(Ⅱ) 디클로라이드[bis(triphenylphosphine) palladium(Ⅱ) dichloride, PdCl2(PPh3)2; 0.041 g, 0.059 mmol]을 첨가한 후, 혼합물에 110℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 진공에서 제거하고 잔여물은 DCM으로 처리한 다음, 셀라이트를 이용하여 여과하고 여액을 수집하였으며, 용매를 진공에서 제거하였다. 잔여물은 5:1 DCM/EA를 사용한 실리카 겔 크로마토그래피로 정제하여, 0.19 g (41 %)의 표제 화합물 (3a)를 수득하였다. A microwave vessel was charged with compound 1 (0.45 g, 1.8 mmol), compound 2a (0.20 g, 1.2 mmol), 1,4-dioxane (2.0 mL), ethanol (0.40 mL), and 2 M aqueous potassium carbonate (1.8 mL, 2.54 mL). mmol) was filled. bis(triphenylphosphine) palladium(II) dichloride, PdCl 2 (PPh 3 ) 2 in N 2 atmosphere; 0.041 g, 0.059 mmol] was added, and the mixture was irradiated with 100 W microwave at 110° C. for 10 minutes. The solvent was removed in vacuo and the residue was treated with DCM, filtered using celite and the filtrate was collected, and the solvent was removed in vacuo. The residue was purified by silica gel chromatography using 5:1 DCM/EA to give 0.19 g (41%) of the title compound (3a).
2-3-2. 5-(6-(2-3-2. 5-(6-( 사이클로펜틸아미노Cyclopentylamino )) 피라진pyrazine -2-일)-2 days) 인돌린indoline -2-온 [-2-on [ 5-(6-(Cyclopentylamino)pyrazin-2-yl)indolin-2-one5-(6-(Cyclopentylamino)pyrazin-2-yl)indolin-2-one ] 합성 : 화합물 (3b)] Synthesis: compound (3b)
<화합물 3b><Compound 3b>
Figure PCTKR2022003249-appb-img-000010
Figure PCTKR2022003249-appb-img-000010
마이크로파 용기를 화합물 1 (0.23 g, 0.91 mmol), 화합물 2b (0.15 g, 0.76 mmol), 1,4-다이옥산 (2.0 mL), 에탄올 (0.40 mL), 및 2 M 수성 탄산칼륨 (0.91 mL, 1.5 mmol)으로 채웠다. N2 분위기에서 비스(트리페닐포스핀)팔라듐(Ⅱ) 디클로라이드[PdCl2(PPh3)2; 0.027 g, 0.038 mmol]을 첨가한 후, 혼합물에 110℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 진공에서 제거하고 잔여물은 DCM으로 처리한 다음, 셀라이트를 이용하여 여과하고 여액을 수집하였으며, 용매를 진공에서 제거하였다. 잔여물은 95:5 DCM/MeOH을 사용한 실리카 겔 크로마토그래피로 정제하여, 0.11 g (49 %)의 표제 화합물 (3b)를 수득하였다. A microwave vessel was charged with compound 1 (0.23 g, 0.91 mmol), compound 2b (0.15 g, 0.76 mmol), 1,4-dioxane (2.0 mL), ethanol (0.40 mL), and 2 M aqueous potassium carbonate (0.91 mL, 1.5 mL). mmol) was filled. Bis(triphenylphosphine)palladium( II ) dichloride [PdCl 2 (PPh 3 ) 2 ; 0.027 g, 0.038 mmol] was added, and the mixture was irradiated with 100 W microwave at 110° C. for 10 minutes. The solvent was removed in vacuo and the residue was treated with DCM, filtered using celite and the filtrate was collected, and the solvent was removed in vacuo. The residue was purified by silica gel chromatography using 95:5 DCM/MeOH to give 0.11 g (49%) of the title compound (3b).
2-3-3. 5-(6-(2-3-3. 5-(6-( 사이클로헥실아미노cyclohexylamino )) 피라진pyrazine -2-일)-2 days) 인돌린indoline -2-온 [-2-on [ 5-(6-(Cyclohexylamino)pyrazin-2-yl)indolin-2-one5-(6-(Cyclohexylamino)pyrazin-2-yl)indolin-2-one ] 합성 : 화합물 (3c)] synthesis: compound (3c)
<화합물 3c><Compound 3c>
Figure PCTKR2022003249-appb-img-000011
Figure PCTKR2022003249-appb-img-000011
마이크로파 용기를 화합물 1 (0.12 g, 0.47 mmol), 화합물 2c (0.12 g, 0.56 mmol), 1,4-다이옥산 (2.0 mL), 에탄올 (0.40 mL), 및 2 M 수성 탄산나트륨(sodium carbonate; 0.70 mL, 1.4 mmol)으로 채웠다. N2 분위기에서 비스(트리페닐포스핀)팔라듐(Ⅱ) 디클로라이드[PdCl2(PPh3)2; 0.017 g, 0.024 mmol]을 첨가한 후, 혼합물에 110℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 진공에서 제거하고 잔여물은 DCM으로 처리한 다음, 셀라이트를 이용하여 여과하고 여액을 수집하였으며, 용매를 진공에서 제거하였다. 잔여물은 95:5 DCM/MeOH을 사용한 실리카 겔 크로마토그래피로 정제하여, 0.070 g (48 %)의 표제 화합물 (3c)를 수득하였다. A microwave vessel was charged with compound 1 (0.12 g, 0.47 mmol), compound 2c (0.12 g, 0.56 mmol), 1,4-dioxane (2.0 mL), ethanol (0.40 mL), and 2 M aqueous sodium carbonate (0.70 mL). , 1.4 mmol). Bis(triphenylphosphine)palladium( II ) dichloride [PdCl 2 (PPh 3 ) 2 ; 0.017 g, 0.024 mmol] was added, and the mixture was irradiated with 100 W microwave at 110° C. for 10 minutes. The solvent was removed in vacuo and the residue was treated with DCM, filtered using celite and the filtrate was collected, and the solvent was removed in vacuo. The residue was purified by silica gel chromatography using 95:5 DCM/MeOH to give 0.070 g (48%) of the title compound (3c).
2-3-4. 5-(6-(2-3-4. 5-(6-( 아다만탄Adamantane -1--One- 일아미노ylamino )) 피라진pyrazine -2-일)-2 days) 인돌린indoline -2-온 [-2-on [ 5-(6-(Adamantan-1-ylamino)pyrazin-2-yl)indolin-2-one5-(6-(Adamantan-1-ylamino)pyrazin-2-yl)indolin-2-one ] 합성 : 화합물 (3d)] synthesis: compound (3d)
<화합물 3d><Compound 3d>
Figure PCTKR2022003249-appb-img-000012
Figure PCTKR2022003249-appb-img-000012
마이크로파 용기를 화합물 1 (0.062 g, 0.24 mmol), 화합물 2d (0.052 g, 0.20 mmol), 1,4-다이옥산 (2.0 mL), 에탄올 (0.40 mL), 및 2 M 수성 탄산나트륨 (0.30 mL, 0.6 mmol)으로 채웠다. N2 분위기에서 비스(트리페닐포스핀)팔라듐(Ⅱ) 디클로라이드[PdCl2(PPh3)2; 0.0070 g, 0.010 mmol]을 첨가한 후, 혼합물에 110℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 진공에서 제거하고 잔여물은 실리카 겔 상에서 90:10 DCM/MeOH을 사용한 건식 컬럼 진공 크로마토그래피(dry column vacuum chromatography, DCVC)로 정제하였다. 진공에서 용매를 제거한 후, 잔여물은 95:5 DCM/MeOH을 사용한 실리카 겔 크로마토그래피로 정제하여, 0.034 g (47 %)의 표제 화합물 (3d)를 수득하였다. A microwave vessel was charged with compound 1 (0.062 g, 0.24 mmol), compound 2d (0.052 g, 0.20 mmol), 1,4-dioxane (2.0 mL), ethanol (0.40 mL), and 2 M aqueous sodium carbonate (0.30 mL, 0.6 mmol). ) was filled with Bis(triphenylphosphine)palladium( II ) dichloride [PdCl 2 (PPh 3 ) 2 ; 0.0070 g, 0.010 mmol] was added, and the mixture was irradiated with 100 W microwave at 110° C. for 10 minutes. The solvent was removed in vacuo and the residue was purified by dry column vacuum chromatography (DCVC) on silica gel using 90:10 DCM/MeOH. After removal of the solvent in vacuo, the residue was purified by silica gel chromatography using 95:5 DCM/MeOH to give 0.034 g (47%) of the title compound (3d).
2-3-5. 5-(6-(2-3-5. 5-(6-( 사이클로헵틸아미노cycloheptylamino )) 피라진pyrazine -2-일)-2 days) 인돌린indoline -2-온 [-2-on [ 5-(6-(Cycloheptylamino)pyrazin-2-yl)indolin-2-one5-(6-(Cycloheptylamino)pyrazin-2-yl)indolin-2-one ] 합성 : 화합물 (3e)] synthesis: compound (3e)
<화합물 3e><Compound 3e>
Figure PCTKR2022003249-appb-img-000013
Figure PCTKR2022003249-appb-img-000013
마이크로파 용기를 화합물 1 (0.12 g, 0.46 mmol), 화합물 2e (0.13 g, 0.56 mmol), 1,4-다이옥산 (2.0 mL), 에탄올 (0.40 mL), 및 2 M 수성 탄산나트륨 (0.69 mL, 1.4 mmol)으로 채웠다. N2 분위기에서 비스(트리페닐포스핀)팔라듐(Ⅱ) 디클로라이드[PdCl2(PPh3)2; 0.016 g, 0.023 mmol]을 첨가한 후, 혼합물에 110℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 진공에서 제거하고 잔여물은 97:3 DCM/MeOH로 처리한 다음, 셀라이트를 이용하여 여과하고 여액을 수집하였으며, 용매를 진공에서 제거하였다. 잔여물은 97:3 DCM/MeOH 및 95:5 DCM/MeOH을 사용한 실리카 겔 크로마토그래피로 정제하여, 0.12 g (79 %)의 표제 화합물 (3e)를 수득하였다. A microwave vessel was charged with compound 1 (0.12 g, 0.46 mmol), compound 2e (0.13 g, 0.56 mmol), 1,4-dioxane (2.0 mL), ethanol (0.40 mL), and 2 M aqueous sodium carbonate (0.69 mL, 1.4 mmol). ) was filled with Bis(triphenylphosphine)palladium( II ) dichloride [PdCl 2 (PPh 3 ) 2 ; 0.016 g, 0.023 mmol] was added, and the mixture was irradiated with 100 W microwave at 110° C. for 10 minutes. The solvent was removed in vacuo and the residue was treated with 97:3 DCM/MeOH, filtered using celite and the filtrate was collected, and the solvent was removed in vacuo. The residue was purified by silica gel chromatography using 97:3 DCM/MeOH and 95:5 DCM/MeOH to give 0.12 g (79%) of the title compound (3e).
2-4-1. (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(2-4-1. (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-( 사이클로프로필아미노cyclopropylamino )) 피라진pyrazine -2-일)인돌린-2-온 [-2-day) indoline-2-one [ (Z)-3-((1H-(Z)-3-((1H- imidazolimidazol -5--5- ylyl )) methylenemethylene )-5-(6-(cyclopropylamino)pyrazin-2-yl)indolin-2-one)-5-(6-(cyclopropylamino)pyrazin-2-yl)indolin-2-one ] 합성 : 화합물 (4a; ] Synthesis: compound (4a; KMUKMU -11170)-11170)
<화합물 4a><Compound 4a>
Figure PCTKR2022003249-appb-img-000014
Figure PCTKR2022003249-appb-img-000014
마이크로파 용기를 화합물 3a (0.15 g, 0.056 mmol), 피페리딘(piperidine; 6.0 μL, 0.06 mmol), 1H-이미다졸-4-카보알데하이드(1H-imidazole-4-carbaldehyde; 0.081 g, 0.85 mmol), 및 에탄올 (2.0 mL)로 채우고, 혼합물에 80℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 진공에서 제거하고 잔여물은 95:5 DCM/MeOH을 사용한 실리카 겔 크로마토그래피로 정제하여, 0.042 g (22 %)의 표제 화합물 (4a)를 수득하였다.In a microwave container, compound 3a (0.15 g, 0.056 mmol), piperidine (6.0 μL, 0.06 mmol), 1H-imidazole-4-carbaldehyde (1H-imidazole-4-carbaldehyde; 0.081 g, 0.85 mmol) , and ethanol (2.0 mL), and the mixture was irradiated with 100 W microwave at 80° C. for 10 minutes. The solvent was removed in vacuo and the residue was purified by silica gel chromatography using 95:5 DCM/MeOH to give 0.042 g (22%) of the title compound (4a).
1H-NMR (500 MHz, DMSO-d6) δ 11.27-11.14 (1H), 8.41 (s, 1H), 8.38 (s, 1H), 8.06 (s, 1H), 7.99 (s, 1H), 7.96 (d, J = 8.0 Hz, 1H), 7.95 (s, 1H), 7.69 (s, 1H), 7.29 (s, 1H), 7.01 (d, J = 8.0 Hz, 1H), 2.71 (s, 1H), 0.79 (m, 2H), 0.57-0.45 (m, 2H); ESI MS: m/z = 345[M+H]+, 173[(M+H)/2]+. 1 H-NMR (500 MHz, DMSO-d 6 ) δ 11.27-11.14 (1H), 8.41 (s, 1H), 8.38 (s, 1H), 8.06 (s, 1H), 7.99 (s, 1H), 7.96 (d, J = 8.0 Hz, 1H), 7.95 (s, 1H), 7.69 (s, 1H), 7.29 (s, 1H), 7.01 (d, J = 8.0 Hz, 1H), 2.71 (s, 1H) , 0.79 (m, 2H), 0.57–0.45 (m, 2H); ESI MS: m/z = 345[M+H] + , 173[(M+H)/2] + .
2-4-2. (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(2-4-2. (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-( 사이클로펜틸아미노Cyclopentylamino )) 피라진pyrazine -2-일)인돌린-2-온 [-2-day) indoline-2-one [ (Z)-3-((1H-(Z)-3-((1H- imidazolimidazol -5--5- ylyl )) methylenemethylene )-5-(6-(cyclopentylamino)pyrazin-2-yl)indolin-2-one)-5-(6-(cyclopentylamino)pyrazin-2-yl)indolin-2-one ] 합성 : 화합물 (4b; ] Synthesis: compound (4b; KMUKMU -11342)-11342)
<화합물 4b><Compound 4b>
Figure PCTKR2022003249-appb-img-000015
Figure PCTKR2022003249-appb-img-000015
마이크로파 용기를 화합물 3b (0.080 g, 0.27 mmol), 피페리딘 (6.0 μL, 0.05 mmol), 1H-이미다졸-4-카보알데하이드 (0.031 g, 0.33 mmol), 및 에탄올 (2.0 mL)로 채우고, 혼합물에 80℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 진공에서 제거하고 잔여물은 80:10 DCM/MeOH을 사용한 실리카 겔 크로마토그래피로 정제하여, 0.042 g (22 %)의 표제 화합물 (4b)를 수득하였다.A microwave vessel was charged with compound 3b (0.080 g, 0.27 mmol), piperidine (6.0 μL, 0.05 mmol), 1H-imidazole-4-carboaldehyde (0.031 g, 0.33 mmol), and ethanol (2.0 mL); The mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The solvent was removed in vacuo and the residue was purified by silica gel chromatography using 80:10 DCM/MeOH to give 0.042 g (22%) of the title compound (4b).
1H-NMR (500 MHz, DMSO-d6) δ 11.19 (s, 1H), 8.37 (s, 1H), 8.30 (s, 1H), 8.03 (s, 1H), 8.00 (s, 1H), 7.97 (d, J = 8.0 Hz, 1H), 7.83 (s, 1H), 7.09 (d, J = 6.3 Hz, 1H), 7.01 (d, J = 8.0 Hz, 1H), 4.26 (q, J = 6.1 Hz, 1H), 2.18-1.91 (m, 2H), 1.82-1.67 (m, 2H), 1.67-1.57 (m, 2H), 1.57-1.32 (m, 2H); ESI MS: m/z = 373[M+H]+, 187[(M+H)/2]+. 1 H-NMR (500 MHz, DMSO-d6) δ 11.19 (s, 1H), 8.37 (s, 1H), 8.30 (s, 1H), 8.03 (s, 1H), 8.00 (s, 1H), 7.97 ( d, J = 8.0 Hz, 1H), 7.83 (s, 1H), 7.09 (d, J = 6.3 Hz, 1H), 7.01 (d, J = 8.0 Hz, 1H), 4.26 (q, J = 6.1 Hz, 1H), 2.18-1.91 (m, 2H), 1.82-1.67 (m, 2H), 1.67-1.57 (m, 2H), 1.57-1.32 (m, 2H); ESI MS: m/z = 373[M+H] + , 187[(M+H)/2] + .
2-4-3-1. (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(2-4-3-1. (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-( 사이클로헥실아미노cyclohexylamino )) 피라진pyrazine -2-일)인돌린-2-온 [-2-day) indoline-2-one [ (Z)-3-((1H-(Z)-3-((1H- ImidazolImidazol -5--5- ylyl )) methylenemethylene )-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one)-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one ] 합성 : 화합물 (4c; ] Synthesis: compound (4c; KMUKMU -11361)-11361)
<화합물 4c><Compound 4c>
Figure PCTKR2022003249-appb-img-000016
Figure PCTKR2022003249-appb-img-000016
마이크로파 용기를 화합물 3c (0.056 g, 0.18 mmol), 피페리딘 (1.8 μL, 0.018 mmol), 1H-이미다졸-4-카보알데하이드 (0.021 g, 0.22 mmol), 및 에탄올 (2.0 mL)로 채우고, 혼합물에 80℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 감압 하에서 제거하고 생성된 고체는 증류수를 첨가하여 여과하였다. 잔여물은 100:5:0.5 클로로포름(CHCl3)/MeOH/암모니아수(NH4OH)를 사용한 실리카 겔 크로마토그래피로 정제하여, 0.053 g (75 %)의 표제 화합물 (4c)를 수득하였다.A microwave vessel was charged with compound 3c (0.056 g, 0.18 mmol), piperidine (1.8 μL, 0.018 mmol), 1H-imidazole-4-carboaldehyde (0.021 g, 0.22 mmol), and ethanol (2.0 mL); The mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The solvent was removed under reduced pressure, and the resulting solid was filtered by adding distilled water. The residue was purified by silica gel chromatography using 100:5:0.5 chloroform (CHCl 3 )/MeOH/ammonia water (NH 4 OH) to give 0.053 g (75%) of the title compound (4c).
1H-NMR (500 MHz, DMSO-d6): δ 8.31 (s, 1H), 8.24 (s, 1H), 7.98 (s, 1H), 7.93 (s, 1H), 7.89 (d, J = 8.6 Hz, 1H), 7.78 (s, 1H), 6.96 (d, J = 8.6 Hz, 1H), 6.91 (d, J = 7.4 Hz, 1H), 3.88-3.74 (m, 1H), 2.00-1.89 (m, 2H), 1.76-1.65 (m, 2H), 1.61-1.52 (m, 1H), 1.41-1.30 (m, 2H), 1.28-1.13 (m, 4H); ESI MS: m/z = 387[M+H]+, 194[(M+H)/2]+. 1 H-NMR (500 MHz, DMSO-d6): δ 8.31 (s, 1H), 8.24 (s, 1H), 7.98 (s, 1H), 7.93 (s, 1H), 7.89 (d, J = 8.6 Hz , 1H), 7.78 (s, 1H), 6.96 (d, J = 8.6 Hz, 1H), 6.91 (d, J = 7.4 Hz, 1H), 3.88–3.74 (m, 1H), 2.00–1.89 (m, 2H), 1.76-1.65 (m, 2H), 1.61-1.52 (m, 1H), 1.41-1.30 (m, 2H), 1.28-1.13 (m, 4H); ESI MS: m/z = 387[M+H] + , 194[(M+H)/2] + .
2-4-3-2-4-3- 2.(2.( Z)-3-((1H-피롤-2-일)메틸렌)-5-(6-(Z)-3-((1H-pyrrol-2-yl)methylene)-5-(6-( 사이클로헥실아미노cyclohexylamino )) 피라진pyrazine -2-일)인돌린-2-온 [-2-day) indoline-2-one [ (Z)-3-((1H-(Z)-3-((1H- pyrrolpyrrol -2--2- ylyl )) methylenemethylene )-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one)-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one ] 합성 : 화합물 (4c'; ] Synthesis: compound (4c'; KMUKMU -11426) -11426)
<화합물 4c'><Compound 4c'>
Figure PCTKR2022003249-appb-img-000017
Figure PCTKR2022003249-appb-img-000017
마이크로파 용기를 화합물 3c (0.079 g, 0.26 mmol), 피페리딘 (2.5 μL, 0.025 mmol), 1H-이미다졸-4-카보알데하이드 (0.029 g, 0.31 mmol), 및 에탄올 (2.0 mL)로 채우고, 혼합물에 80℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 감압 하에서 제거하고 생성된 고체는 증류수를 첨가하여 여과하였다. 잔여물은 100:3:0.3 CHCl3/MeOH/NH4OH를 사용한 실리카 겔 크로마토그래피로 정제하여, 0.068 g (68 %)의 표제 화합물 (4c')을 수득하였다.A microwave vessel was charged with compound 3c (0.079 g, 0.26 mmol), piperidine (2.5 μL, 0.025 mmol), 1H-imidazole-4-carboaldehyde (0.029 g, 0.31 mmol), and ethanol (2.0 mL); The mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The solvent was removed under reduced pressure, and the resulting solid was filtered by adding distilled water. The residue was purified by silica gel chromatography using 100:3:0.3 CHCl 3 /MeOH/NH 4 OH to give 0.068 g (68%) of the title compound (4c′).
1H-NMR (500 MHz, DMSO-d6): δ 11.03 (s, 1H), 8.28 (s, 1H), 8.24 (s, 1H), 7.85 (m, 2H), 7.77 (s, 1H), 7.35 (br, 1H), 6.95 (d, J = 8.6 Hz, 1H), 6.91 (d, J = 7.4 Hz, 1H), 6.82 (t, J = 1.7 Hz, 1H), 6.39-6.32 (m, 1H), 3.85-3.75 (m, 1H), 1.99-1.90 (m, 2H), 1.71 (dt, J = 13.5, 3.5 Hz, 2H), 1.58 (dt, J = 12.5, 4.0 Hz, 1H), 1.36 (q, J = 11.1 Hz, 2H), 1.28-1.14 (m, 3H); ESI MS: m/z = 386[M+H]+. 1 H-NMR (500 MHz, DMSO-d6): δ 11.03 (s, 1H), 8.28 (s, 1H), 8.24 (s, 1H), 7.85 (m, 2H), 7.77 (s, 1H), 7.35 (br, 1H), 6.95 (d, J = 8.6 Hz, 1H), 6.91 (d, J = 7.4 Hz, 1H), 6.82 (t, J = 1.7 Hz, 1H), 6.39–6.32 (m, 1H) , 3.85–3.75 (m, 1H), 1.99–1.90 (m, 2H), 1.71 (dt, J = 13.5, 3.5 Hz, 2H), 1.58 (dt, J = 12.5, 4.0 Hz, 1H), 1.36 (q , J = 11.1 Hz, 2H), 1.28–1.14 (m, 3H); ESI MS: m/z = 386[M+H] + .
2-4-4. (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(2-4-4. (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-( 아다만탄Adamantane -1--One- 일아미노ylamino )) 피라진pyrazine -2-일)인돌린-2-온 [-2-day) indoline-2-one [ (Z)-3-((1H-(Z)-3-((1H- ImidazolImidazol -5--5- ylyl )) methylenemethylene )-5-(6-()-5-(6-( adamantanadamantan -1-ylamino)pyrazin-2-yl)indolin-2-one-1-ylamino) pyrazin-2-yl) indolin-2-one ] 합성 : 화합물 (4d; ] synthesis: compound (4d; KMUKMU -11421)-11421)
<화합물 4d><Compound 4d>
Figure PCTKR2022003249-appb-img-000018
Figure PCTKR2022003249-appb-img-000018
마이크로파 용기를 화합물 3d (0.034 g, 0.094 mmol), 피페리딘 (0.9 μL, 0.0091 mmol), 1H-이미다졸-4-카보알데하이드 (0.011 g, 0.11 mmol), 및 에탄올 (1.0 mL)로 채우고, 혼합물에 80℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 감압 하에서 제거하고 생성된 고체는 증류수를 첨가하여 여과하였다. 잔여물은 90:10 DCM/MeOH를 사용한 실리카 겔 크로마토그래피로 정제하여, 0.030 g (72 %)의 표제 화합물 (4d)를 수득하였다.A microwave vessel was charged with compound 3d (0.034 g, 0.094 mmol), piperidine (0.9 μL, 0.0091 mmol), 1H-imidazole-4-carboaldehyde (0.011 g, 0.11 mmol), and ethanol (1.0 mL), The mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The solvent was removed under reduced pressure, and the resulting solid was filtered by adding distilled water. The residue was purified by silica gel chromatography using 90:10 DCM/MeOH to give 0.030 g (72%) of the title compound (4d).
1H-NMR (500 MHz, DMSO-d6): δ 11.20 (br, 1H), 8.38 (s, 1H), 8.27 (s, 1H), 8.05 (s, 1H), 7.97 (d, J = 8.0 Hz, 1H), 7.91 (s, 1H), 7.80 (s, 1H), 7.61 (s, 1H), 7.03 (d, J = 8.0 Hz, 1H), 6.75 (s, 1H), 2.27-2.14 (m, 6H), 2.14-2.04 (m, 3H), 1.83-1.64 (m, 6H); ESI MS: m/z = 439[M+H]+, 220[(M+H)/2]+. 1 H-NMR (500 MHz, DMSO-d6): δ 11.20 (br, 1H), 8.38 (s, 1H), 8.27 (s, 1H), 8.05 (s, 1H), 7.97 (d, J = 8.0 Hz , 1H), 7.91 (s, 1H), 7.80 (s, 1H), 7.61 (s, 1H), 7.03 (d, J = 8.0 Hz, 1H), 6.75 (s, 1H), 2.27–2.14 (m, 6H), 2.14-2.04 (m, 3H), 1.83-1.64 (m, 6H); ESI MS: m/z = 439[M+H] + , 220[(M+H)/2] + .
2-4-5. (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(2-4-5. (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-( 사이클로헵틸아미노cycloheptylamino )) 피라진pyrazine -2-일)인돌린-2-온 [-2-day) indoline-2-one [ (Z)-3-((1H-(Z)-3-((1H- ImidazolImidazol -5--5- ylyl )) methylenemethylene )-5-(6-(cycloheptylamino)pyrazin-2-yl)indolin-2-one)-5-(6-(cycloheptylamino)pyrazin-2-yl)indolin-2-one ] 합성 : 화합물 (4e; ] Synthesis: compound (4e; KMUKMU -11427)-11427)
<화합물 4e><Compound 4e>
Figure PCTKR2022003249-appb-img-000019
Figure PCTKR2022003249-appb-img-000019
마이크로파 용기를 화합물 3e (0.12 g, 0.7 mmol), 피페리딘 (4.1 μL, 0.037 mmol), 1H-이미다졸-4-카보알데하이드 (0.042 g, 0.44 mmol), 및 에탄올 (2.0 mL)로 채우고, 혼합물에 80℃에서 10분간 100 W 마이크로파를 조사하였다. 용매는 감압 하에서 제거하고 생성된 고체는 증류수를 첨가하여 여과하였다. 잔여물은 100:3:0.3 CHCl3/MeOH/NH4OH를 사용한 실리카 겔 크로마토그래피로 정제하여, 0.062 g (42 %)의 표제 화합물 (4e)을 수득하였다.A microwave vessel was charged with compound 3e (0.12 g, 0.7 mmol), piperidine (4.1 μL, 0.037 mmol), 1H-imidazole-4-carboaldehyde (0.042 g, 0.44 mmol), and ethanol (2.0 mL); The mixture was irradiated with 100 W microwave at 80°C for 10 minutes. The solvent was removed under reduced pressure, and the resulting solid was filtered by adding distilled water. The residue was purified by silica gel chromatography using 100:3:0.3 CHCl 3 /MeOH/NH 4 OH to give 0.062 g (42%) of the title compound (4e).
1H-NMR (500 MHz, DMSO-d6): δ 11.20 (s, 1H), 8.38 (s, 1H), 8.28 (s, 1H), 8.06 (s, 1H), 7.99 (s, 1H), 7.96 (d, J = 8.0 Hz, 1H), 7.83 (s, 1H), 7.66 (s, 1H), 7.09-6.98 (m, 2H), 4.14-3.94 (m, 1H), 2.06-1.89 (m, 2H), 1.76-1.65 (m, 2H), 1.65-1.47 (m, 8H); ESI MS: m/z = 401[M+H]+, 201[(M+H)/2]+. 1 H-NMR (500 MHz, DMSO-d6): δ 11.20 (s, 1H), 8.38 (s, 1H), 8.28 (s, 1H), 8.06 (s, 1H), 7.99 (s, 1H), 7.96 (d, J = 8.0 Hz, 1H), 7.83 (s, 1H), 7.66 (s, 1H), 7.09–6.98 (m, 2H), 4.14–3.94 (m, 1H), 2.06–1.89 (m, 2H) ), 1.76-1.65 (m, 2H), 1.65-1.47 (m, 8H); ESI MS: m/z = 401[M+H] + , 201[(M+H)/2] + .
<실험방법><Experiment method>
1. 실험 및 분석방법1. Experiment and analysis method
1-1. 시약1-1. reagent
LPS (Escherichia coli serotype)는 Sigma-Aldrich (St. Louis, MO, USA)에서 구입하였다. 셀레콕시브(celecoxib), 이브루티닙(ibrutinib), 메토트렉세이트(methotrexate), 및 룩소리티닙(ruxolitinib)은 Selleck Chemicals (Houston, TX, USA)에서 구입하였다. NF-κB 리간드의 수용체 활성제 (RANKL)는 PeproTech (Rocky Hill, NH, USA)에서 구입하였다.LPS ( Escherichia coli serotype) was purchased from Sigma-Aldrich (St. Louis, MO, USA). Celecoxib, ibrutinib, methotrexate, and ruxolitinib were purchased from Selleck Chemicals (Houston, TX, USA). Receptor activator of NF-κB ligands (RANKL) was purchased from PeproTech (Rocky Hill, NH, USA).
iNOS, TLR4, MyD88, p-TAK1 (Ser412), p-ERK, ERK, p-JNK, p-IKKα/β, IKKα/β, p-NF-κB p65, NF-κB, ASC, caspase-1, 및 NLRP3에 대한 1차 항체는 Cell Signaling Technology (Beverly, MA, USA)에서 구입하였다. Anti-IL-1β 항체는 Novus Biologicals (Centennial, CO, USA)에서 구입하였다. IL-6, COX-1, COX-2, TAK1, p-p38, 및 p38에 대한 항체는 각각 Santa Cruz Bio-technology (Santa Cruz, CA, USA) 및 Sigma-Aldrich (St. Louis, MO, USA)에서 구입하였다. Anti-TNF-α 항체는 Abcam (Cambridge, MA, USA)에서 구입하였다. Anti-β-actin 항체는 Sigma-Aldrich (St. Louis, MO, USA)에서 구입하였다. Anti-mouse IgG-horseradish peroxidase (HRP) 및 anti-mouse IgG-HRP 항체는 Santa Cruz Biotechnology (Santa Cruz, CA, USA)에서 구입하였다.iNOS, TLR4, MyD88, p-TAK1 (Ser412), p-ERK, ERK, p-JNK, p-IKKα/β, IKKα/β, p-NF-κB p65, NF-κB, ASC, caspase-1, and primary antibodies against NLRP3 were purchased from Cell Signaling Technology (Beverly, MA, USA). Anti-IL-1β antibody was purchased from Novus Biologicals (Centennial, CO, USA). Antibodies against IL-6, COX-1, COX-2, TAK1, p-p38, and p38 were respectively Santa Cruz Bio-technology (Santa Cruz, CA, USA) and Sigma-Aldrich (St. Louis, MO, USA). ) was purchased. Anti-TNF-α antibody was purchased from Abcam (Cambridge, MA, USA). Anti-β-actin antibody was purchased from Sigma-Aldrich (St. Louis, MO, USA). Anti-mouse IgG-horseradish peroxidase (HRP) and anti-mouse IgG-HRP antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA).
1-2. 세포주 및 세포 배양1-2. Cell lines and cell culture
인간 단핵구 세포주(human monocytic cell line) THP-1 세포 (Korean Cell Line Bank, Seoul, Korea)는 RPMI1640 배지 (Welgene Inc., Gyeongsan, Korea), 10% 소태아혈청 (fetal bovine serum, FBS; Welgene Inc., Gyeongsan, Korea), 및 1% 항생제(antibiotic)-항진균제(antimycotic) 용액 (Welgene Inc., Gyeongsan, Korea)에서 배양되었다. 세포는 완전 습한 공기 내 37.5℃, 5% CO2에서 배양되었다.Human monocytic cell line THP-1 cells (Korean Cell Line Bank, Seoul, Korea) were cultured in RPMI1640 medium (Welgene Inc., Gyeongsan, Korea), 10% fetal bovine serum (FBS; Welgene Inc.). ., Gyeongsan, Korea), and 1% antibiotic-antimycotic solution (Welgene Inc., Gyeongsan, Korea). Cells were cultured at 37.5° C., 5% CO 2 in completely humid air.
alpha-MEM은 Welgene Inc. (Gyeongsan, Korea)에서 구입하였다.alpha-MEM was developed by Welgene Inc. (Gyeongsan, Korea).
1-3. 파골세포 분화(1-3. Osteoclast differentiation ( osteoclastosteoclast differentiation) differentiation
RAW 264.7 세포를 파골세포 분화 배지 (alpha-MEM+50 ng/ml RANKL)에서 성장시켜 파골세포 유사 세포를 생성하였다. 분화 후, 제조업체의 프로토콜에 따라 TRAP 염색 키트 (TaKaRa, Japan)를 이용하여 다핵세포(multinucleated cells, MNCs)를 시각화하였다.RAW 264.7 cells were grown in osteoclast differentiation medium (alpha-MEM+50 ng/ml RANKL) to generate osteoclast-like cells. After differentiation, multinucleated cells (MNCs) were visualized using a TRAP staining kit (TaKaRa, Japan) according to the manufacturer's protocol.
1-4. 원발성 인간 골관절염 섬유아세포 유사 1-4. Primary human osteoarthritis fibroblast-like 활막세포의of synovial cells 분리 및 배양 Isolation and culture
인간 골관절염(osteoarthritis, OA) 섬유아세포 유사 활막세포(fibroblast-like synoviocytes, FLS)의 분리는 계명대학교 동산의료원에서 활막절제술(synovectomy)을 받은 골관절염 환자의 활막 조직을 효소 소화하여 수행하였다.Isolation of human osteoarthritis (OA) fibroblast-like synoviocytes (FLS) was performed by enzymatic digestion of synovial tissues of osteoarthritis patients who underwent synovectomy at Keimyung University Dongsan Medical Center.
등록된 골관절염 환자는 미국 류마티스학회(American College of Rheumatology, ACR) 기준을 충족하고 사전 동의(informed consent)를 제공하는 데 동의하였다. 실험 연구는 2020년 11월 20일 계명대학교 동산의료원 기관 심의 위원회(IRB)의 승인을 받았다 [IRB No. 2020-11-031].Enrolled osteoarthritis patients met American College of Rheumatology (ACR) criteria and agreed to provide informed consent. The experimental study was approved by the Institutional Review Board (IRB) of Keimyung University Dongsan Medical Center on November 20, 2020 [IRB No. 2020-11-031].
골관절염 조직을 2-3 mm 조각으로 분쇄한 다음, 다져진 조직을 37.5℃, 5% CO2 하, Dulbecco’s modified Eagle 배지 (DMEM; Welgene Inc., Gyeongsan, Korea)에서 0.5 mg/mL II 타입 콜라게나제(collagenases; Thermo Fisher Scientific, Waltham, MA, USA)로 처리하였다. 분리된 세포는 원심분리 (3,000 rpm, 5 min) 한 후, DMEM, 10% FBS, 및 1% 항생제-항진균제 용액에 현탁시켜, 75 cm2 플라스크에서 교반하였다. 1일 후, 부착 세포는 5% CO2 하 37.5℃, DMEM, 10% FBS 및 1% 항생제-항진균제 용액에서 배양되었고, 배양액은 3일마다 교체하였다. 플라스크 바닥의 90-95%가 섬유아세포로 채워졌을 때, 배지를 새로운 배양물을 사용하여 1:3의 비율로 희석하였다.Osteoarthritis tissue was pulverized into 2-3 mm pieces, and then the minced tissue was treated with 0.5 mg/mL type II collagenase in Dulbecco's modified Eagle's medium (DMEM; Welgene Inc., Gyeongsan, Korea) at 37.5°C under 5% CO 2 . (collagenases; Thermo Fisher Scientific, Waltham, MA, USA). The isolated cells were centrifuged (3,000 rpm, 5 min), suspended in DMEM, 10% FBS, and 1% antibiotic-antimycotic solution, and stirred in a 75 cm 2 flask. After 1 day, adherent cells were cultured in DMEM, 10% FBS, and 1% antibiotic-antimycotic solution at 37.5° C. under 5% CO 2 , and the medium was changed every 3 days. When 90-95% of the bottom of the flask is filled with fibroblasts, the medium is diluted 1:3 with fresh culture.
1-5. 키나아제-프로파일링 분석1-5. Kinase-profiling assay
*신규한 다중-단백질 키나아제 억제제 KMU-1170의 특이성을 평가하기 위하여, Eurofins Cerep S.A.에 의해 키나아제-프로파일링 서비스(kinase-profiling service)를 수행하였다. 키나아제-프로파일링 분석은 1μM의 화합물 및 Eurofin의 프로토콜에 따른 각 개별 키나아제 및 키나아제 기질에 대한 Km 값의 ATP 농도로 수행되었다.*To evaluate the specificity of the novel multi-protein kinase inhibitor KMU-1170, a kinase-profiling service was performed by Eurofins Cerep S.A. Kinase-profiling assays were performed with ATP concentrations of 1 μM of compound and K values for each individual kinase and kinase substrate according to Eurofin's protocol.
1-6. 세포 생존율 분석1-6. Cell viability assay
세포 생존율 분석은 XTT assay (Welgene Inc., Gyeongsan, Korea)을 사용하여 수행되었다. 간략하게, 2×105 THP-1 세포를 96-well plates에 24시간 동안 플레이팅한 다음, 다양한 농도의 KMU-1170을 세포에 처리하여 5% CO2 하, 37℃에서 24시간 동안 배양하였다. 이후, 0.5 mg/mL XTT 용액을 배양액에 첨가하고, 5% CO2 하, 37℃에서 3시간 동안 배양하였다. 450 nM 파장에서 microplate reader (BMG Labtech, Ortenberg, Germany)로 광학 밀도(optical density, OD)를 측정하였다.Cell viability assay was performed using XTT assay (Welgene Inc., Gyeongsan, Korea). Briefly, 2×10 5 THP-1 cells were plated in 96-well plates for 24 hours, and then cells were treated with various concentrations of KMU-1170 and cultured for 24 hours at 37° C. under 5% CO 2 . . Thereafter, a 0.5 mg/mL XTT solution was added to the culture medium, and cultured at 37° C. for 3 hours under 5% CO 2 . Optical density (OD) was measured with a microplate reader (BMG Labtech, Ortenberg, Germany) at a wavelength of 450 nM.
1-7. 1-7. 웨스턴Western 블로팅blotting 분석 analyze
웨스턴 블로팅 분석을 위해 THP-1 세포 (2×106 cells/well in 60-mm dishes) 및 FLS (2×106 cells/well in 100-mm dishes)를 플레이팅하였다. 24시간 동안 배양한 후, 세포를 RIPA buffer (20 mM HEPES and 0.5% Triton X-100, pH 7.6)에 용해시키고, 상등액 분획을 수집하였다. 단백질 농도는 BCA assay kit (Thermo Fisher Scientific, Waltham, MA, USA)를 사용하여 측정되었다. 동량의 단백질을 SDS-PAGE에서 전기영동하고 니트로셀룰로오스 막 (GE Healthcare Life Science, Pittsburgh, PA, USA)으로 옮겼다. 그 후, 막을 blocking buffer (0.05% Tween 20 with 5% non-fat dry milk in Tris-buffered saline (TBS))로 1시간 동안 배양한 다음, 특정 표적 단백질에 대해 적절히 희석된 1차 항체로 24시간 동안 배양하였다. 다음, 막을 Tween 20과 함께 TBS로 세척하고, 적절한 2차 항체와 함께 실온에서 1시간 동안 배양하였다. For Western blotting analysis, THP-1 cells (2×10 6 cells/well in 60-mm dishes) and FLS (2×10 6 cells/well in 100-mm dishes) were plated. After culturing for 24 hours, the cells were dissolved in RIPA buffer (20 mM HEPES and 0.5% Triton X-100, pH 7.6), and the supernatant fraction was collected. Protein concentration was measured using the BCA assay kit (Thermo Fisher Scientific, Waltham, MA, USA). Equal amounts of protein were electrophoresed on SDS-PAGE and transferred to a nitrocellulose membrane (GE Healthcare Life Science, Pittsburgh, PA, USA). Then, the membrane was incubated for 1 hour with blocking buffer (0.05% Tween 20 with 5% non-fat dry milk in Tris-buffered saline (TBS)) and then incubated for 24 hours with an appropriately diluted primary antibody against a specific target protein. cultured for a while. Next, the membrane was washed in TBS with Tween 20 and incubated with the appropriate secondary antibody for 1 hour at room temperature.
특정 단백질은 제조사의 프로토콜에 따라 ECL western blotting kit (EMD Millipore, Darmstadt, Germany)로 검출되었고, 신호 강도는 Chemi Image System Fusion FX (Vilber Lourmat, France)를 사용하여 측정되었다. 단백질 밴드는 Image-J software를 사용하여 정량화되었다. Image-J를 사용한 밴드 밀도의 정량 분석은 세 번의 독립적인 실험의 평균±표준편차(standard deviation, SD)로 표시되었다. Specific proteins were detected with the ECL western blotting kit (EMD Millipore, Darmstadt, Germany) according to the manufacturer's protocol, and signal intensities were measured using the Chemi Image System Fusion FX (Vilber Lourmat, France). Protein bands were quantified using Image-J software. Quantitative analysis of band density using Image-J was expressed as the mean±standard deviation (SD) of three independent experiments.
1-8. RNA 분리 및 1-8. RNA isolation and 역전사reverse transcription 중합효소 연쇄 반응(Reverse Transcription-Polymerase Chain Reaction, RT- Reverse Transcription-Polymerase Chain Reaction (RT- PCRPCR ) 및 실시간 정량 ) and real-time quantification PCRPCR ( ( qPCRqPCR ))
총 세포 RNA는 TRIzol Reagent (Thermo Fisher Scientific, Waltham, MA, USA)을 사용하여 분리되고, 각 RNA는 NanoDrop 1000 (Thermo Fisher Scientific, Waltham, MA, USA)을 사용하여 정량화되었다. 각 cDNA는 제조사의 지침에 따라 Moloney murine leukemia virus reverse transcriptase (Gibco-BRL, Gaithersburg, MD, USA)를 사용하여 1μg의 분리된 RNA로부터 획득하였다.Total cellular RNA was isolated using TRIzol Reagent (Thermo Fisher Scientific, Waltham, MA, USA) and each RNA was quantified using a NanoDrop 1000 (Thermo Fisher Scientific, Waltham, MA, USA). Each cDNA was obtained from 1 μg of isolated RNA using Moloney murine leukemia virus reverse transcriptase (Gibco-BRL, Gaithersburg, MD, USA) according to the manufacturer's instructions.
하기 표 1은 본 실험예에 사용된 프라이머 서열을 나타낸 것이다. Table 1 below shows the primer sequences used in this experimental example.
PrimersPrimers 방향direction Sequences ( 5’→ 3’)Sequences ( 5’→ 3’) 서열번호sequence number

Cathepsin K

Cathepsin K
Forward Forward CAG CAG AAC GGA GGC ATT GACAG CAG AAC GGA GGC ATT GA 1One
ReverseReverse CCT TTG CCG TGG CGT TAT ACCCT TTG CCG TGG CGT TAT AC 22

iNOS

iNOS
ForwardForward CTG TCT GGT TCC TAC GTC ACCCTG TCT GGT TCC TAC GTC ACC 33
ReverseReverse CCC ACG TTA CAT GGG AGG ATACCC ACG TTA CAT GGG AGG ATA 44

COX-1

COX-1
ForwardForward ACC TTG AAG GAG TCA GGC ATG AGACC TTG AAG GAG TCA GGC ATG AG 55
ReverseReverse TGT TCG GTG TCC AGT TCC AAT ATGT TCG GTG TCC AGT TCC AAT A 66

COX-2

COX-2
ForwardForward ATC ACA GGC TTC CAT TGA CCATC ACA GGC TTC CAT TGA CC 77
ReverseReverse TAT CAT CTA GTC CGG AGG GGTAT CAT CTA GTC CGG AGG GG 88

IL-1β

IL-1β
ForwardForward CCT TGG GCC TCA AGG AAA ACCT TGG GCC TCA AGG AAA A 99
ReverseReverse CTC CAG CTG TAG AGT GGG CTT ACTC CAG CTG TAG AGT GGG CTT A 1010

TNF-α

TNF-α
ForwardForward GGA GAA GGG TGA CCG ACT CAGGA GAA GGG TGA CCG ACT CA 1111
ReverseReverse CTG CCC AGA CTC GGC AACTG CCC AGA CTC GGC AA 1212

IL-6

IL-6
ForwardForward ATG GCA CAG TAT CTG GAG GAGATG GCA CAG TAT CTG GAG GAG 1313
ReverseReverse TAA GCT GGA CTC ACT CTC GGATAA GCT GGA CTC ACT CTC GGA 1414

β-actin

β-actin
ForwardForward AAT CTG GCA CCA CAC CTT CTAAAT CTG GCA CCA CAC CTT CTA 1515
ReverseReverse ATA GCA CAG CCT GGA TAG CAAATA GCA CAG CCT GGA TAG CAA 1616
PCR의 경우, DNA 중합효소는 IL-1β, TNF-α, 및 IL-6를 표적으로 하는 프라이머와 함께 사용되었다. 증폭된 PCR 산물은 2% 아가로스 겔에서 전기영동으로 분리되고 자외선 하에서 검출되었다. 특이적 프라이머 및 SYBR GREEN Premix (Toyobo, Osaka, Japan) 사용에 의한 qPCR의 경우, LightCycler® 480 real-time PCR system (Roche Diagnostics, Mannheim, Germany)로 수행되었다. 실험은 3회 수행되었고, 각 표적 mRNA의 threshold cycle number (Ct)는 β-actin으로 정규화하였다. 각 유전자의 delta-delta Ct(ΔΔ Ct) 값은 세 번의 독립적인 실험의 평균±표준편차로 표시되었다.For PCR, DNA polymerase was used with primers targeting IL-1β, TNF-α, and IL-6. Amplified PCR products were electrophoretically separated on a 2% agarose gel and detected under ultraviolet light. For qPCR using specific primers and SYBR GREEN Premix (Toyobo, Osaka, Japan), it was performed with a LightCycler® 480 real-time PCR system (Roche Diagnostics, Mannheim, Germany). The experiment was performed three times, and the threshold cycle number (Ct) of each target mRNA was normalized to β-actin. The delta-delta Ct (ΔΔ Ct) values of each gene were expressed as the mean±standard deviation of three independent experiments.
1-9. 면역 형광 염색1-9. immunofluorescence staining
THP-1 세포 (1×103)를 8-챔버 유리 슬라이드에서 24시간 동안 배양하고, KMU-1170 1μmol/L로 1시간 동안 처리한 다음, 6시간 동안 LPS로 자극하였다. 세포를 phosphate-buffered saline (PBS)으로 세척하고 4% 포름알데히드(formaldehyde)로 고정한 다음, 0.2% Triton X-100이 용해된 PBS를 사용하여 투과시키고 소 혈청 알부민(bovine serum albumin)으로 1시간 동안 배양하여 비특이적 결합을 차단하였다. 그 후, 세포는 4℃에서 24시간 동안 1차 항체와 함께 배양한 다음, FITC-conjugated goat anti-mouse IgG (Thermo Fisher Scientific, Waltham, MA, USA) 및 4’,6-디아미디노-2-페닐인돌(4’,6-diamidino-2-phenylindole; Thermo Fisher Scientific, Wal-tham, MA, USA; for nuclear staining) 처리한 다음, 염색된 세포를 현광 현미경으로 확인하였다.THP-1 cells (1×10 3 ) were cultured on 8-chamber glass slides for 24 hours, treated with 1 μmol/L of KMU-1170 for 1 hour, and then stimulated with LPS for 6 hours. Cells were washed with phosphate-buffered saline (PBS), fixed with 4% formaldehyde, permeabilized with PBS containing 0.2% Triton X-100, and treated with bovine serum albumin for 1 hour. Incubation was performed to block non-specific binding. Then, the cells were incubated with the primary antibody for 24 hours at 4°C and then FITC-conjugated goat anti-mouse IgG (Thermo Fisher Scientific, Waltham, MA, USA) and 4',6-diamidino-2 After treatment with -phenylindole (4',6-diamidino-2-phenylindole; Thermo Fisher Scientific, Wal-tham, MA, USA; for nuclear staining), the stained cells were examined under a fluorescence microscope.
1-10. 통계적 분석1-10. statistical analysis
정량적 결과는 평균 및 표준편차로 표시되었다. 데이터는 Statisti-cal Package for Social Science 26.0 software (SPSS Inc.; Chicago, IL, USA)를 사용하여 one-was ANOVA 및 post-hoc comparisons (Student-Newman-Keuls)에 의해 분석되었다. 0.05 이하의 p 값은 중요한 것으로 간주되었다.Quantitative results were expressed as mean and standard deviation. Data were analyzed by one-was ANOVA and post-hoc comparisons (Student-Newman-Keuls) using the Statisti-cal Package for Social Science 26.0 software (SPSS Inc.; Chicago, IL, USA). A p value of 0.05 or less was considered significant.
<< 실험예Experimental example 1> 1> 실시예Example 1에 따른 according to 1 KMUKMU -시리즈 화합물의 항염 활성 분석-Analysis of anti-inflammatory activity of series compounds
KMU-시리즈 화합물의 항염증제로서의 가능성을 조사하기 위하여, THP-1 세포에서 LPS-매개 염증에 대한 KMU-시리즈의 항염증 효과를 분석하였다. To investigate the potential of KMU-series compounds as anti-inflammatory agents, the anti-inflammatory effects of KMU-series compounds on LPS-mediated inflammation in THP-1 cells were analyzed.
THP-1 세포를 PMA(phorbol-12-myristate-13-acetate) 100 nM을 사용하여 24시간 동안 대식세포로 분화시켰다. 상기 세포에 KMU-11421, 11426, 11427, 11361, 11342, 11170 (1μM), 및 룩소리티닙(Ruxolitinib) (50μM)을 1시간 동안 처리한 다음, LPS (1μg/mL)를 3시간 동안 첨가하였다. 총 RNA를 추출하여 실시간 PCR을 이용해 IL-1β, TNF-α, 및 IL-6의 mRNA 발현 수준을 결정하였다.THP-1 cells were differentiated into macrophages using 100 nM of phorbol-12-myristate-13-acetate (PMA) for 24 hours. The cells were treated with KMU-11421, 11426, 11427, 11361, 11342, 11170 (1 μM), and Ruxolitinib (50 μM) for 1 hour, then LPS (1 μg/mL) was added for 3 hours . Total RNA was extracted and mRNA expression levels of IL-1β, TNF-α, and IL-6 were determined using real-time PCR.
그 결과, 도 2A 및 2B에 나타난 바와 같이, KMU-11426을 제외한 KMU-시리즈 화합물 및 룩소리티닙(JAK 억제제)로 세포를 전처리하면, IL-1β 및 TNF-α의 LPS-유도 상향 조절이 약화되었다. 반면, 도 2C에 나타난 바와 같이, 모든 KMU-시리즈 화합물 및 룩소리티닙은 IL-6의 LPS-유도 상향 조절을 억제하였다.As a result, as shown in Figures 2A and 2B, when cells were pretreated with KMU-series compounds other than KMU-11426 and ruxolitinib (JAK inhibitor), LPS-induced upregulation of IL-1β and TNF-α was attenuated. It became. On the other hand, as shown in Figure 2C, all KMU-series compounds and ruxolitinib inhibited the LPS-induced upregulation of IL-6.
<< 실험예Experimental example 2> 2> KMUKMU -11342의 of -11342 RANKLRANKL -유도 파골세포 분화 억제 확인-Confirmation of inhibition of induced osteoclast differentiation
RANKL-유도 파골세포 분화에서 KMU-11342의 효과를 평가하기 위하여, KMU-11342 (0.01, 0.25, 0.5μM)을 1시간 동안 처리한 다음, RAW 264.7 세포를 RANKL (50ng/mL)로 5일 동안 처리하여 파골세포로 분화시켜 TRAP 염색 키트를 사용하여 세포를 염색하였다. TRAP-양성 다핵세포(MNC)는 자줏빛 붉은색으로 염색되었고, 파골세포 분화 정도를 나타내는 TRAP 염색된 세포의 수가 5일 동안 시간 의존적으로 증가하였다.To evaluate the effect of KMU-11342 on RANKL-induced osteoclast differentiation, after treatment with KMU-11342 (0.01, 0.25, 0.5 μM) for 1 hour, RAW 264.7 cells were treated with RANKL (50 ng/mL) for 5 days. The treated cells were differentiated into osteoclasts, and the cells were stained using a TRAP staining kit. TRAP-positive multinucleated cells (MNC) were stained purple red, and the number of TRAP-stained cells indicating the degree of osteoclast differentiation increased in a time-dependent manner for 5 days.
그 결과, 도 3A에 나타난 바와 같이, KMU-11342의 농도가 높을수록, TRAP-양성 MNC가 더 적게 관찰되었으며, 이는 KMU-11342가 RAW 264.7 세포의 RANKL-유도 파골세포 분화를 용량 의존적으로 억제함을 나타낸다.As a result, as shown in Figure 3A, the higher the concentration of KMU-11342, the fewer TRAP-positive MNCs were observed, indicating that KMU-11342 inhibited RANKL-induced osteoclast differentiation of RAW 264.7 cells in a dose-dependent manner. indicates
그 다음, 총 RNA를 추출하여 실시간 PCR을 이용해 파골세포 분화의 최종 조절 단계에 관여하는 파골세포 특이적 바이오마커 카뎁신 K (cathepsin K)의 발현에 대한 KMU-11342의 효과를 분석한 결과, 도 3B에 나타난 바와 같이, KMU-11342는 카뎁신 K mRNA 발현 수준의 RANKL-유도 상향 조절을 억제하였다.Then, total RNA was extracted and the effect of KMU-11342 on the expression of cathepsin K, a biomarker specific to osteoclasts involved in the final regulatory step of osteoclast differentiation, was analyzed using real-time PCR. As shown in 3B, KMU-11342 inhibited RANKL-induced upregulation of cathepsin K mRNA expression levels.
<< 실험예Experimental example 3> 3> KMUKMU -11170 (이하, 실험결과 및 도면에서는 "-11170 (Hereinafter, in the experimental results and drawings, " KMUKMU -1170"으로 표기) 화합물의 여러 가지 활성 분석-1170") Analysis of various activities of the compound
3-1. 다양한 단백질 키나아제의 활성에 대한 3-1. activity of various protein kinases. KMUKMU -1170의 영향 확인Check the impact of -1170
하기 표 2는 1μM KMU-1170 처리에 따른 15개 단백질 키나아제의 키나아제 활성을 나타낸 것이다.Table 2 below shows the kinase activities of 15 protein kinases according to 1 μM KMU-1170 treatment.
하기 표 2를 참조하면, 단백질 키나아제 분석은 KMU-1170이 염증 반응과 관련된 다양한 키나아제를 억제함을 보여주며, 특히 미토겐-활성 단백질 키나아제 1(mitogen-activated protein kinase 1, MAPK1), Lck, TYK2, JAK3, 및 Txk에 대해 강한 억제를 나타내었다.Referring to Table 2 below, protein kinase analysis shows that KMU-1170 inhibits various kinases related to the inflammatory response, in particular, mitogen-activated protein kinase 1 (MAPK1), Lck, TYK2 , JAK3, and Txk.
Figure PCTKR2022003249-appb-img-000020
Figure PCTKR2022003249-appb-img-000020
3-2. 3-2. THPTHP -1 세포에서 in -1 cell KMUKMU -1170의 -1170's LPSLPS -유도 상향 조절된 -induced upregulated iNOSiNOS 및 COX-2의 억제 효과 확인 and confirmation of the inhibitory effect of COX-2
THP-1 세포에서 KMU-1170의 독성을 조사하기 위하여, 2,3-비스-(2-메톡시-4-니트로-5-설포페닐)-2H-테트라졸리움-5-카르복스아닐리드[2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide, XTT] 분석을 수행하였다.To investigate the toxicity of KMU-1170 in THP-1 cells, 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide [2, 3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide, XTT] analysis was performed.
THP-1 세포를 PMA 100 nM을 사용하여 24시간 동안 대식세포로 분화시킨 다음, 세포에 다양한 농도의 KMU-1170 (0.01, 0.1, 0.5, 1, 5, 및 10μM)을 처리한 결과, 도 4B에 나타난 바와 같이, KMU-1170은 1μM 농도까지 독성을 나타내지 않았다.THP-1 cells were differentiated into macrophages using 100 nM of PMA for 24 hours, and then the cells were treated with various concentrations of KMU-1170 (0.01, 0.1, 0.5, 1, 5, and 10 μM), Fig. 4B As shown, KMU-1170 showed no toxicity up to 1 μM concentration.
이 후, KMU-1170 (0.1, 0.5, 및 1μM)을 1시간 동안 전처리한 후, 6시간 동안 LPS (1μg/mL)를 처리하여 이에 따라 상향 조절된 iNOS 및 COX-2의 변화를 확인한 결과, 1μM의 KMU-1170은 LPS-유도된 유도성 산화질소 합성효소(inducible nitric oxide synthase, iNOS) mRNA 및 단백질의 상향 조절을 억제하였고 (도 4C, D, E), 전처리로 LPS-유도된 사이클로옥시게나제-2(cyclooxygenase-2, COX-2) mRNA 및 단백질의 상향 조절을 억제하였으나, COX-1의 mRNA 및 단백질은 유의미한 변화를 확인하지 못하였다 (도 4F, G, H, I).After that, KMU-1170 (0.1, 0.5, and 1μM) was pretreated for 1 hour, followed by LPS (1μg/mL) for 6 hours to confirm the upregulated changes in iNOS and COX-2, 1 μM of KMU-1170 inhibited LPS-induced up-regulation of inducible nitric oxide synthase (iNOS) mRNA and protein (Fig. 4C, D, E), and as a pretreatment, LPS-induced cyclook Upregulation of cyclooxygenase-2 (COX-2) mRNA and protein was suppressed, but no significant changes were observed in COX-1 mRNA and protein (FIG. 4F, G, H, I).
3-3. 3-3. THPTHP -1 세포에서 in -1 cell KMUKMU -1170의 -1170's LPSLPS -유도 -Judo 전염증성pro-inflammatory 사이토카인 생성 억제 효과 확인 Confirmation of cytokine production inhibitory effect
KMU-1170의 항염증 작용 기전을 평가하기 위해, IL-1β, TNF-α, 및 IL-6과 같은 전염증성 사이토카인(proinflammatory cytokines)의 수준 조절에 대해 조사하였다. THP-1 세포를 PMA (100 nM)을 사용하여 24시간 동안 대식세포로 분화시킨 다음, KMU-1170 (0.1, 0.5, 및 1μM)를 1시간 동안 전처리한 후 6시간 동안 LPS (1μg/mL)를 처리하여 확인한 결과, 도 5에 나타난 바와 같이, 1μM KMU-1170으로 전처리된 세포는 LPS-유도로 상향 조절된 IL-1β(interleukin-1β), TNF-α(tumor necrosis factor-α), 및 IL-6를 억제하는 것으로 나타났다.To evaluate the anti-inflammatory mechanism of action of KMU-1170, the regulation of the levels of proinflammatory cytokines such as IL-1β, TNF-α, and IL-6 was investigated. THP-1 cells were differentiated into macrophages using PMA (100 nM) for 24 hours, followed by pretreatment with KMU-1170 (0.1, 0.5, and 1 μM) for 1 hour followed by LPS (1 μg/mL) for 6 hours. As a result of confirming by processing, as shown in FIG. 5, cells pretreated with 1 μM KMU-1170 showed LPS-induced up-regulated IL-1β (interleukin-1β), TNF-α (tumor necrosis factor-α), and It has been shown to inhibit IL-6.
3-4. 3-4. THPTHP -1 세포에서 in -1 cell KMUKMU -1170의 -1170's LPSLPS -유도 -Judo TAK1TAK1 and MAPKs의MAPKs 인산화 억제 효과 확인 Confirmation of phosphorylation inhibitory effect
KMU-1170이 TLR4 및 MyD88 발현에 영향을 미치는지 조사하였다. THP-1 세포를 PMA (100 nM)을 사용하여 24시간 동안 대식세포로 분화시킨 다음, KMU-1170 (0.1, 0.5, 및 1μM)를 1시간 동안 전처리한 후 6시간 동안 LPS (1μg/mL)를 처리하여 확인한 결과, 도 6A 및 6B에 나타난 바와 같이, 1μM KMU-1170로 전처리된 THP-1 세포는 TLR4(toll-like receptor 4) 및 MyD88(myeloid differentiation factor 88)의 LPS-유도 상향 조절을 약화시키지 않는 것으로 나타났다. The effects of KMU-1170 on TLR4 and MyD88 expression were investigated. THP-1 cells were differentiated into macrophages using PMA (100 nM) for 24 hours, followed by pretreatment with KMU-1170 (0.1, 0.5, and 1 μM) for 1 hour followed by LPS (1 μg/mL) for 6 hours. 6A and 6B, as shown in FIGS. 6A and 6B, THP-1 cells pretreated with 1 μM KMU-1170 exhibited LPS-induced upregulation of toll-like receptor 4 (TLR4) and myeloid differentiation factor 88 (MyD88). did not appear to weaken.
염증에서 KMU-1170의 잠재적인 분자 표적을 확인하기 위하여, MAPKs 및 NF-κB의 기능을 조절하는 전환 성장 인자-베타-활성화 키나아제 1(transforming growth factor-β-activated kinase 1, TAK1)의 인산화를 조사한 결과, 도 6C 및 도 6D에 나타난 바와 같이, THP-1 세포에서 KMU-1170의 전처리는 LPS-유도 TAK1의 인산화를 억제하는 것으로 나타났다. To identify potential molecular targets of KMU-1170 in inflammation, phosphorylation of transforming growth factor-β-activated kinase 1 (TAK1), which regulates the function of MAPKs and NF-κB, was investigated. As a result of the investigation, as shown in FIGS. 6C and 6D , pretreatment with KMU-1170 in THP-1 cells inhibited LPS-induced phosphorylation of TAK1.
또한, THP-1 세포를 PMA (100 nM)을 사용하여 24 시간 동안 대식세포로 분화시킨 다음, 24시간 동안 KMU-1170의 유무에 관계없이 처리하고 LPS (1μg/mL)로 유도한 결과, 도 6E 및 도 6F에 나타난 바와 같이, MAPKs 중에서는 오직 ERK(extracellular signal-regulated kinases) 및 JNK(c-Jun N-terminal kinases)만이 LPS에 대한 반응으로 인산화되었고, 1μM KMU-1170의 전처리는 THP-1 세포에서 LPS 유도 ERK 및 JNK의 인산화를 억제하는 것으로 나타났다.In addition, THP-1 cells were differentiated into macrophages using PMA (100 nM) for 24 hours, treated with or without KMU-1170 for 24 hours, and induced with LPS (1 μg/mL). 6E and 6F, among MAPKs, only ERK (extracellular signal-regulated kinases) and JNK (c-Jun N-terminal kinases) were phosphorylated in response to LPS, and pretreatment with 1 μM KMU-1170 It was shown to inhibit LPS-induced phosphorylation of ERK and JNK in 1 cells.
3-5. THP-1 세포에서 KMU-1170의 LPS-유도 NF-κB의 활성 억제능 확인3-5. Confirmation of KMU-1170's ability to inhibit LPS-induced NF-κB activity in THP-1 cells
THP-1 세포에서 LPS는 NF-κB kinase α/β 억제제 (inhibitor NF-κB kinase α/β, IKKα/β) 및 NF-κB p65의 인산화를 유도했지만, 도 7A 및 도 7B를 참조하면, KMU-1170로 전처리하면 LPS-유도 인산화가 억제되는 것으로 나타났다. In THP-1 cells, LPS induced phosphorylation of NF-κB kinase α/β inhibitors (inhibitor NF-κB kinase α/β, IKKα/β) and NF-κB p65, but referring to FIGS. 7A and 7B, KMU It was shown that pretreatment with -1170 inhibits LPS-induced phosphorylation.
형광 현미경을 사용하여 KMU-1170이 NF-κB p65의 핵 전좌(nuclear translocation)에 영향을 미치는지 여부를 조사한 결과, 도 7C에 나타난 바와 같이, KMU-1170의 전처리가 THP-1 세포에서 NF-κB p65의 LPS-유도 핵 전좌를 억제하는 것으로 나타났다.As a result of examining whether KMU-1170 affects nuclear translocation of NF-κB p65 using a fluorescence microscope, as shown in FIG. 7C , pretreatment with KMU-1170 inhibited NF-κB It has been shown to inhibit LPS-induced nuclear translocation of p65.
3-6. THP-1 세포에서 KMU-1170의 LPS-유도 NLRP3의 활성 약화 확인3-6. Confirmation of attenuation of LPS-induced NLRP3 activity of KMU-1170 in THP-1 cells
KMU-1170이 NLRP3 인플라마좀 신호에 영향을 미치는지 확인하기 위해, THP-1 세포를 PMA (100 nM)을 사용하여 24시간 동안 대식세포로 분화시킨 다음, KMU-1170 1μM를 1시간 동안 전처리한 후 6시간 동안 LPS (1μg/mL) 및/또는 ATP (1 mM)를 처리하였다.To confirm whether KMU-1170 affects NLRP3 inflammasome signaling, THP-1 cells were differentiated into macrophages using PMA (100 nM) for 24 hours, and then pretreated with 1 μM KMU-1170 for 1 hour. After that, LPS (1 μg/mL) and/or ATP (1 mM) were treated for 6 hours.
그 결과, 도 8A와 같이, THP-1 세포에 1μM KMU-1170의 전처리는 LPS-유도 pro-IL-1β 및 IL-1β의 세포질 방출을 억제하는 것으로 나타났다. 게다가, 1μM KMU-1170의 전처리는 LPS 및 LPS와 ATP 공동 처리에 의해 유도된 NLRP3, ASC(apoptosis-associated speck-like protein containing a CARD), pro-caspase-1, 및 pro-IL-1β의 상향 조절을 약화시켰다. As a result, as shown in FIG. 8A , pretreatment of THP-1 cells with 1 μM KMU-1170 inhibited LPS-induced pro-IL-1β and IL-1β cytoplasmic release. Furthermore, pretreatment with 1 μM KMU-1170 upregulated NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), pro-caspase-1, and pro-IL-1β induced by LPS and co-treatment with LPS and ATP. control was weakened.
이러한 정량적 결과는 KMU-1170이 NLRP3 인플라마좀 활성을 유의하게 억제할 수 있음을 보여준다 (도 8B).These quantitative results show that KMU-1170 can significantly inhibit NLRP3 inflammasome activity (FIG. 8B).
3-7. THP-1 세포에서 KMU-1170의 강력한 항염증 활성 확인3-7. Confirmation of strong anti-inflammatory activity of KMU-1170 in THP-1 cells
항염증제로서 KMU-1170의 작용을 고려하여, KMU-1170의 항염증 효능의 잠재적 우수성을 다른 다양한 항염증제와 비교하여 분석하였다. THP-1 세포를 PMA (100 nM)을 사용하여 24시간 동안 대식세포로 분화시킨 다음, KMU-1170 (1μM), 셀레콕시브(celecoxib, 25μM), 이브루티닙(ibrutinib, 5μM), 룩소리티닙(luxolitinib, 50μM), 및 메토트렉세이트(metotrexate, 1μM)로 1시간 동안 전처리한 후, 6시간 동안 LPS (1μg/mL)를 처리하였다.Considering the action of KMU-1170 as an anti-inflammatory agent, the potential superiority of anti-inflammatory efficacy of KMU-1170 compared to various other anti-inflammatory agents was analyzed. THP-1 cells were differentiated into macrophages using PMA (100 nM) for 24 hours, followed by KMU-1170 (1 μM), celecoxib (25 μM), ibrutinib (5 μM), Luxority After pre-treatment with nip (luxolitinib, 50 μM) and methotrexate (1 μM) for 1 hour, LPS (1 μg/mL) was treated for 6 hours.
그 결과, 도 8C 및 8D에 나타난 바와 같이, 1μM KMU-1170는 THP-1 세포에서 LPS-유도로 증가된 IL-1β 단백질을 유의적으로 억제한 반면, 셀레콕시브 (COX-2 억제제), 이브루티닙 (BTK 억제제), 룩소리티닙 (JAK 억제제) 및 메토트렉세이트 (RA의 항염증제)와 같은 다른 시험된 약물은 고 농도에서도 이러한 효과가 확인되지 않았다. 1μM 메토트렉세이트 만이 KMU-1170을 처리한 경우와 유사하게 LPS-유도로 상향 조절된 IL-1β를 억제하는 것으로 나타났다.As a result, as shown in FIGS. 8C and 8D, 1 μM KMU-1170 significantly suppressed the LPS-induced increased IL-1β protein in THP-1 cells, whereas celecoxib (COX-2 inhibitor), Other tested drugs such as ibrutinib (a BTK inhibitor), ruxolitinib (a JAK inhibitor) and methotrexate (an anti-inflammatory in RA) did not show this effect even at high concentrations. Only 1 μM methotrexate was shown to inhibit LPS-induced up-regulation of IL-1β, similar to treatment with KMU-1170.
3-8. 골관절염 섬유아세포 유사 3-8. Osteoarthritis fibroblast-like 활막세포에서in synovial cells KMUKMU -1170의 -1170's LPSLPS -유도 염증 억제 활성 확인-Confirmation of induced inflammation suppression activity
마지막으로, 인간 골관절염(OA) 섬유아세포 유사 활막세포(FLS)를 사용하여 골관절염에 대한 KMU-1170의 항염증 활성을 확인하였다. 무릎 골관절염 환자의 활액 조직에서 인간 골관절염 섬유아세포 유사 활막세포를 획득하였고, 이 세포에 KMU-1170 (0.1, 0.5, 및 1μM)를 1시간 동안 전처리한 후 6시간 동안 LPS (1μg/mL)를 처리하였다.Finally, the anti-inflammatory activity of KMU-1170 against osteoarthritis was confirmed using human osteoarthritis (OA) fibroblast-like synovial cells (FLS). Human osteoarthritis fibroblast-like synovial cells were obtained from the synovial tissues of patients with knee osteoarthritis, pretreated with KMU-1170 (0.1, 0.5, and 1 μM) for 1 hour and then treated with LPS (1 μg/mL) for 6 hours. did
그 결과, 도 9에 나타난 바와 같이, KMU-1170 전처리된 FLS는 LPS-유도 상향 조절된 IL-1β, TNF-α, 및 IL-6의 mRNA 및 단백질 수준 (도 9A 내지 9D)과 iNOS 및 COX-2 단백질 수준 (도 9E)을 억제하였다. 또한, KMU-1170 전처리는 FLS에서 IKKα/β 및 NF-κB p65의 LPS-유도 인산화를 억제하였다 (도 9F).As a result, as shown in FIG. 9, KMU-1170 pretreated FLS showed LPS-induced up-regulated IL-1β, TNF-α, and IL-6 mRNA and protein levels (FIG. 9A to 9D), iNOS and COX -2 protein levels (FIG. 9E). In addition, KMU-1170 pretreatment inhibited LPS-induced phosphorylation of IKKα/β and NF-κB p65 in FLS (FIG. 9F).
즉, 본 발명은 새로운 다중-단백질 키나아제 억제제 KMU-1170이 p-TAK1/ p-IKKα/β/p-NF-κB/전염증성 사이토카인 신호 조절 경로 및 NLRP3 인플라마좀을 조절하여 항염증 효과를 가짐을 확인하였다 (도 10). 이러한 결과는 KMU-시리즈 화합물이 강력한 항염증제 개발에 대해 매우 유리한 화합물임을 시사한다.That is, the present invention is a novel multi-protein kinase inhibitor KMU-1170 is anti-inflammatory effect by regulating the p-TAK1 / p-IKKα / β / p-NF-κB / pro-inflammatory cytokine signaling pathway and NLRP3 inflammasome It was confirmed that it had (FIG. 10). These results suggest that the KMU-series compounds are very advantageous compounds for the development of potent anti-inflammatory drugs.
이상으로 본 발명 내용의 특정한 부분을 상세히 기술하였는 바, 당업계의 통상의 지식을 가진 자에게 있어서, 이러한 구체적 기술은 단지 바람직한 실시양태일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백하다. 즉, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다.Having described specific parts of the present invention in detail above, it is clear to those skilled in the art that these specific descriptions are only preferred embodiments, and the scope of the present invention is not limited thereby. do. That is, the substantial scope of the present invention is defined by the appended claims and their equivalents.

Claims (10)

  1. 하기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염:A compound represented by Formula 1 or a pharmaceutically acceptable salt thereof:
    <화학식 1><Formula 1>
    Figure PCTKR2022003249-appb-img-000021
    Figure PCTKR2022003249-appb-img-000021
    상기 화학식 1에서,In Formula 1,
    R1은 수소, 하이드록시, 할로겐, (C1-C4) 알킬, (C3-C10) 사이클로알킬, 또는 아다만타닐(adamantanyl)에서 선택되고, R 1 is selected from hydrogen, hydroxy, halogen, (C1-C4) alkyl, (C3-C10) cycloalkyl, or adamantanyl;
    R2는 이미다졸일(imidazolyl), 피라졸일(pyrazolyl), 피롤일(pyrrolyl) 또는 피롤리딘일(pyrrolidinyl)에서 선택됨. R 2 is selected from imidazolyl, pyrazolyl, pyrrolyl or pyrrolidinyl.
  2. 제 1 항에 있어서,According to claim 1,
    상기 화합물 또는 이의 염은,The compound or salt thereof,
    상기 R1이 (C3-C8) 사이클로알킬, 또는 아다만타닐(adamantanyl)에서 선택되고, wherein R 1 is selected from (C3-C8) cycloalkyl or adamantanyl;
    상기 R2는 이미다졸일(imidazolyl) 또는 피롤일(pyrrolyl)에서 선택되는 것을 특징으로 하는, 화합물 또는 이의 약학적으로 허용가능한 염.The R 2 is characterized in that selected from imidazolyl (imidazolyl) or pyrrolyl (pyrrolyl), a compound or a pharmaceutically acceptable salt thereof.
  3. 제 1 항에 있어서, According to claim 1,
    상기 화합물 또는 이의 염은,The compound or salt thereof,
    (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(사이클로프로필아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopropylamino)pyrazin-2-yl)indolin-2-one] (4a; KMU-11170), (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(사이클로펜틸아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopentylamino)pyrazin-2-yl)indolin-2-one] (4b; KMU-11342), (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(사이클로헥실아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H-Imidazol-5-yl)methylene)-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one] (4c; KMU-11361), (Z)-3-((1H-피롤-2-일)메틸렌)-5-(6-(사이클로헥실아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H-pyrrol-2-yl)methylene)-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one] (4c'; KMU-11426), (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(아다만탄-1-일아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H-Imidazol-5-yl)methylene)-5-(6-(adamantan-1-ylamino)pyrazin-2-yl)indolin-2-one] (4d; KMU-11421), 및 (Z)-3-((1H-이미다졸-5-일)메틸렌)-5-(6-(사이클로헵틸아미노)피라진-2-일)인돌린-2-온 [(Z)-3-((1H-Imidazol-5-yl)methylene)-5-(6-(cycloheptylamino)pyrazin-2-yl)indolin-2-one] (4e; KMU-11427)로 이루어진 군에서 선택되는 것을 특징으로 하는, 화합물 또는 이의 약학적으로 허용가능한 염.(Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cyclopropylamino)pyrazin-2-yl)indolin-2-one [(Z)-3-( (1H-imidazol-5-yl)methylene)-5-(6-(cyclopropylamino)pyrazin-2-yl)indolin-2-one] (4a; KMU-11170), (Z)-3-((1H- imidazol-5-yl)methylene)-5-(6-(cyclopentylamino)pyrazin-2-yl)indolin-2-one [(Z)-3-((1H-imidazol-5-yl)methylene )-5-(6-(cyclopentylamino)pyrazin-2-yl)indolin-2-one] (4b; KMU-11342), (Z)-3-((1H-imidazol-5-yl)methylene)- 5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one [(Z)-3-((1H-Imidazol-5-yl)methylene)-5-(6-(cyclohexylamino) pyrazin-2-yl)indolin-2-one] (4c; KMU-11361), (Z)-3-((1H-pyrrol-2-yl)methylene)-5-(6-(cyclohexylamino)pyrazine -2-yl)indolin-2-one [(Z)-3-((1H-pyrrol-2-yl)methylene)-5-(6-(cyclohexylamino)pyrazin-2-yl)indolin-2-one ] (4c'; KMU-11426), (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(adamantan-1-ylamino)pyrazin-2-yl )indolin-2-one [(Z)-3-((1H-Imidazol-5-yl)methylene)-5-(6-(adamantan-1-ylamino)pyrazin-2-yl)indolin-2-one ] (4d; KMU-11421), and (Z)-3-((1H-imidazol-5-yl)methylene)-5-(6-(cycloheptylamino)pyrazin-2-yl)indolin-2 -one [(Z)-3-((1H-Imidazol-5-yl)methylene)-5-(6-(cycloheptylamino)pyrazin-2-yl)indolin-2-one] (4e; KMU-11427) characterized in that selected from the group consisting of , A compound or a pharmaceutically acceptable salt thereof.
  4. 제 1 항에 있어서,According to claim 1,
    상기 화합물 또는 염은,The compound or salt,
    MAPK, TAK1, Lck, TYK2, JAK3, 및 Txk로 이루어진 군에서 선택되는 하나 이상의 단백질 키나아제를 억제하는 것을 특징으로 하는, 화합물 또는 이의 약학적으로 허용가능한 염.A compound or pharmaceutically acceptable salt thereof, characterized in that it inhibits at least one protein kinase selected from the group consisting of MAPK, TAK1, Lck, TYK2, JAK3, and Txk.
  5. 제 1 항에 있어서,According to claim 1,
    상기 화합물 또는 염은,The compound or salt,
    NLRP3 인플라마좀의 활성을 억제하는 것을 특징으로 하는 화합물 또는 이의 약학적으로 허용가능한 염.A compound or a pharmaceutically acceptable salt thereof characterized in that it inhibits the activity of NLRP3 inflammasome.
  6. 제 1 항에 있어서,According to claim 1,
    상기 화합물 또는 염은,The compound or salt,
    항염증 활성을 가지는 것을 특징으로 하는 화합물 또는 이의 약학적으로 허용가능한 염.A compound characterized by having anti-inflammatory activity or a pharmaceutically acceptable salt thereof.
  7. 제 1 항에 따른 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 염증성 질환 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating inflammatory diseases containing the compound according to claim 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  8. 제 7 항에 있어서,According to claim 7,
    상기 염증성 질환은,The inflammatory disease,
    골관절염, 류마티스관절염, 골다공증, 골 파제트병, 및 강직성척추염으로 이루어진 군에서 선택되는 어느 하나인 것을 특징으로 하는 염증성 질환 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating inflammatory diseases, characterized in that it is any one selected from the group consisting of osteoarthritis, rheumatoid arthritis, osteoporosis, Paget's disease of bone, and ankylosing spondylitis.
  9. 제 1 항에 따른 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 염증성 질환 예방 또는 개선용 건강기능식품 조성물.A health functional food composition for preventing or improving inflammatory diseases containing the compound according to claim 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  10. 제 9 항에 있어서,According to claim 9,
    상기 염증성 질환은,The inflammatory disease,
    골관절염, 류마티스관절염, 골다공증, 골 파제트병, 및 강직성척추염으로 이루어진 군에서 선택되는 어느 하나인 것을 특징으로 하는 염증성 질환 예방 또는 개선용 건강기능식품 조성물.Osteoarthritis, rheumatoid arthritis, osteoporosis, bone Paget's disease, and ankylosing spondylitis, characterized in that any one selected from the group consisting of inflammatory disease prevention or improvement functional health food composition.
PCT/KR2022/003249 2021-08-10 2022-03-08 Novel inhibitor for multiple protein kinases WO2023017937A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2021-0105096 2021-08-10
KR1020210105096A KR102566028B1 (en) 2021-08-10 2021-08-10 Novel multi-protein kinase inhibitor

Publications (1)

Publication Number Publication Date
WO2023017937A1 true WO2023017937A1 (en) 2023-02-16

Family

ID=85200781

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2022/003249 WO2023017937A1 (en) 2021-08-10 2022-03-08 Novel inhibitor for multiple protein kinases

Country Status (2)

Country Link
KR (1) KR102566028B1 (en)
WO (1) WO2023017937A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6268391B1 (en) * 1997-08-06 2001-07-31 Glaxo Wellcome Inc. Benzylidene-1,3-dihydro-indol-2-one derivatives a receptor tyrosine kinase inhibitors, particularly of Raf kinases
US20130310340A1 (en) * 2012-05-16 2013-11-21 Rigel Pharmaceuticals, Inc. Method of treating muscular degradation
KR20160093073A (en) * 2013-12-19 2016-08-05 사노피 Oxindole derivatives, preparation thereof and therapeutic use in the treatment of ampk-related diseases
WO2017007658A1 (en) * 2015-07-07 2017-01-12 Rigel Pharmaceuticals, Inc. A combination for immune mediated cancer treatment
WO2020076723A1 (en) * 2018-10-08 2020-04-16 Revolution Medicines, Inc. Shp2 inhibitor compositions for use in treating cancer

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20040026657A (en) 2001-05-30 2004-03-31 주식회사 엘지생명과학 Inhibitors Of Protein Kinase For The Treatment Of Disease

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6268391B1 (en) * 1997-08-06 2001-07-31 Glaxo Wellcome Inc. Benzylidene-1,3-dihydro-indol-2-one derivatives a receptor tyrosine kinase inhibitors, particularly of Raf kinases
US20130310340A1 (en) * 2012-05-16 2013-11-21 Rigel Pharmaceuticals, Inc. Method of treating muscular degradation
KR20160093073A (en) * 2013-12-19 2016-08-05 사노피 Oxindole derivatives, preparation thereof and therapeutic use in the treatment of ampk-related diseases
WO2017007658A1 (en) * 2015-07-07 2017-01-12 Rigel Pharmaceuticals, Inc. A combination for immune mediated cancer treatment
WO2020076723A1 (en) * 2018-10-08 2020-04-16 Revolution Medicines, Inc. Shp2 inhibitor compositions for use in treating cancer

Also Published As

Publication number Publication date
KR102566028B1 (en) 2023-08-10
KR20230023206A (en) 2023-02-17

Similar Documents

Publication Publication Date Title
WO2019112344A1 (en) Novel pyrimidine derivative having effect of inhibiting cancer cell growth and pharmaceutical composition containing same
WO2011111880A1 (en) Pharmaceutical composition for treating or preventing diseases caused by the nuclear export of gsk3, including a compound for inhibiting the nuclear export of gsk3
WO2021230600A1 (en) Anticancer composition comprising metabolic anticancer drug
WO2020149715A1 (en) Pyrrolopyridine derivative and use thereof in prevention and treatment of protein kinase-related disease
WO2020149723A1 (en) Pyrrolopyrimidine derivative, and pharmaceutical composition for preventing or treating protein kinase-related disease comprising same as active ingredient
WO2021010715A1 (en) Pharmaceutical composition for preventing or treating bone diseases
WO2011159137A2 (en) Novel thiourea or urea derivative, preparation method thereof, and pharmaceutical composition for preventing or treating aids, containing same as active ingredient
WO2015160192A1 (en) Pharmaceutical composition for treating and preventing leukemia, containing thienopyrimidine derivative or pharmaceutically acceptable salt thereof
WO2015037778A1 (en) Composition containing lignan compound as active ingredient for preventing or treating cancer
WO2023017937A1 (en) Novel inhibitor for multiple protein kinases
WO2019231261A1 (en) Novel biphenyl derivative compound and use thereof
WO2020096416A1 (en) Compound inhibiting yap-tead binding, and pharmaceutical composition for preventing or treating cancer, comprising compound as active ingredient
WO2019147089A1 (en) Pharmaceutical composition for preventing or treating cancer comprising, as active ingredient, calcium channel inhibitor or pharmaceutically acceptable salt thereof
WO2018101762A1 (en) Pharmaceutical composition for preventing or treating ischemic acute kidney injury, containing tricyclic derivative or pharmaceutically acceptable salt thereof
WO2019035522A1 (en) Composition for preventing or treating cancer containing triazolopyridine-based derivative as active ingredient
WO2021230710A1 (en) Novel ido/tdo inhibitor, anticancer use thereof, and anticancer combination therapy thereof
WO2021182903A1 (en) Composition comprising pi3 kinase inhibitor and btk inhibitor
WO2020040326A1 (en) Phenylacetic acid derivative, and composition for preventing or treating autoimmune diseases, containing same as active ingredient
KR20240013180A (en) Arginine methyltransferase inhibitors and uses thereof
WO2021040502A1 (en) Imidazopyridine derivative and pharmaceutical composition comprising same as active ingredient
WO2021085888A1 (en) Novel heterocyclic-substituted pyrimidine derivative exhibiting cancer cell growth inhibitory effect, and pharmaceutical composition containing same
EP3386988A1 (en) Novel dihydropyranopyrimidinone derivatives, and use thereof
WO2020080641A1 (en) Composition for preventing or treating ampk-related diseases, comprising gynostemma longipes vk1 extract or compound isolated therefrom as active ingredient
EP3867249A1 (en) Novel (isopropyl-triazolyl)pyridinyl-substituted benzooxazinone or benzothiazinone derivatives and use thereof
WO2023277331A1 (en) Dyrk1 inhibitor pyrrolo[3,2-c]pyridine derivative and use thereof

Legal Events

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

Ref document number: 22855939

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE