[go: up one dir, main page]

WO2023085645A1 - Novel ramalin derivative and use thereof for prevention or treatment of neurodegenerative diseases - Google Patents

Novel ramalin derivative and use thereof for prevention or treatment of neurodegenerative diseases Download PDF

Info

Publication number
WO2023085645A1
WO2023085645A1 PCT/KR2022/016326 KR2022016326W WO2023085645A1 WO 2023085645 A1 WO2023085645 A1 WO 2023085645A1 KR 2022016326 W KR2022016326 W KR 2022016326W WO 2023085645 A1 WO2023085645 A1 WO 2023085645A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
disease
ramalin
derivative
degenerative brain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/KR2022/016326
Other languages
French (fr)
Korean (ko)
Inventor
임정한
한세종
조동규
김민주
홍주미
김일찬
김태경
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Korea Institute of Ocean Science and Technology KIOST
Original Assignee
Korea Institute of Ocean Science and Technology KIOST
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 Korea Institute of Ocean Science and Technology KIOST filed Critical Korea Institute of Ocean Science and Technology KIOST
Publication of WO2023085645A1 publication Critical patent/WO2023085645A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES, NOT OTHERWISE PROVIDED FOR; PREPARATION OR TREATMENT THEREOF
    • 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
    • A61K31/13Amines
    • A61K31/15Oximes (>C=N—O—); Hydrazines (>N—N<); Hydrazones (>N—N=) ; Imines (C—N=C)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C241/00Preparation of compounds containing chains of nitrogen atoms singly-bound to each other, e.g. hydrazines, triazanes
    • C07C241/02Preparation of hydrazines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C243/00Compounds containing chains of nitrogen atoms singly-bound to each other, e.g. hydrazines, triazanes
    • C07C243/24Hydrazines having nitrogen atoms of hydrazine groups acylated by carboxylic acids
    • C07C243/26Hydrazines having nitrogen atoms of hydrazine groups acylated by carboxylic acids with acylating carboxyl groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C243/34Hydrazines having nitrogen atoms of hydrazine groups acylated by carboxylic acids with acylating carboxyl groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of a carbon skeleton further substituted by nitrogen atoms
    • 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
    • A23V2200/322Foods, ingredients or supplements having a functional effect on health having an effect on the health of the nervous system or on mental function

Definitions

  • the present invention relates to a ramalin derivative and its use for the prevention or treatment of degenerative brain diseases, and more particularly, a derivative obtained by replacing a hydroxyl group of a phenyl ring of ramalin with a methyl group or a fluorine atom is effective against BACE-1. It relates to a novel ramalin derivative used for preventing or treating degenerative brain diseases by exhibiting inhibitory activity, antioxidant and anti-inflammatory activity without toxicity, and a use thereof for preventing or treating degenerative brain diseases.
  • AD Alzheimer's Disease
  • a ⁇ ⁇ -amyloid
  • a ⁇ aggregation and accumulation in the brain could be the result of increased A ⁇ production, decreased A ⁇ protease activity, or altered A ⁇ transport across the BBB (blood-brain barrier) (Mucke, L. Alzheimer's disease. Nature , 2019, 461, 895-897).
  • BACE-1 ⁇ -site APP cleavage enzyme 1 associated with increased production of A ⁇ is considered a target for the prevention and treatment of Alzheimer's disease (Vassar, R. Bace 1. J. Mol. Neurosci., 2004, 23, 105- 113).
  • a ⁇ is also produced through sequential cleavage of amyloid precursor protein (APP) by BACE-1 and BACE-2, and BACE-1 is a key enzyme initiating this process (Roberson, E. D. et al., Science, 2006, 314, 781-784).
  • APP amyloid precursor protein
  • BACE-1 is a key enzyme initiating this process.
  • Experimental evidence based on studies in human APP transgenic mice suggests that memory deficits can be prevented by deletion of the BACE-1 gene (Oakley, H. et al., J. Neurosci., 2006, 26; 10129-10140).
  • a ⁇ aggregates can act as pathogenic mediators and induce oxidative stress, neuroinflammation, and BACE-1 expression. This further aggravates AD (Tamagno, E. et al., Free Radic. Biol.
  • oxidative stress and inflammation can enhance A ⁇ production.
  • plaques were observed in the brains of monkeys with chronic inflammation (Philippens, I. H. et al., J. Alzheimer's Dis., 2017, 55, 101-113). Therefore, given that inflammation increases the production of reactive oxygen species (ROS) that cause oxidative damage to lipids, proteins, and nucleic acids, reducing inflammation based on the use of anti-inflammatory drugs can significantly lower the prevalence of Alzheimer's disease ( McGeer, E. G. et al., Exp. Gerontol., 1998, 33, 371-378).
  • ROS reactive oxygen species
  • NF- ⁇ B associated with inflammation is involved in the expression of BACE-1 regulated by various mechanisms (Roßner, S. et al., Prog. Neurobiol., 2006, 79, 95-111).
  • BACE-1, inflammation and ROS are all associated with A ⁇ formation from the onset of Alzheimer's disease to the post-onset period.
  • oxidative stress by ROS appears like the development of Alzheimer's disease, and it has been reported to be the cause of early onset and exacerbation of Alzheimer's disease (Smith, M. A. et al., Biochim. Biophys. Acta Mol. Basis Dis. , 2000, 1502, 139-144).
  • Mitochondrial ROS produced under normal conditions are balanced by antioxidants and related enzymes.
  • dysfunction due to oxidative damage in this regard results in high levels of ROS production and release of apoptosis-inducing proteins (Dumont, M. et al., Free Radic Biol Med., 2011, 51, 1014-1026; Wang , X. et al., Biochim. Biophys. Acta Mol.
  • Alzheimer's disease exhibits neuropathological features of extracellular beta-amyloid ( ⁇ -amyloid, A ⁇ ) plaques and intracellular nerve fiber tangles.
  • ⁇ -secretase mediates the first step in beta-amyloid production by ⁇ -cleavage of APP, which releases a large soluble extracellular fragment (APP ⁇ ).
  • APP-CTF C-terminal fragment
  • C99 C-terminal fragment of APP
  • beta-amyloids are produced by continuous cleavage of APP mediated by BACE1 (beta-site APP-cleaving enzyme 1, beta-secretase-1) and presenilin/ ⁇ -secretase.
  • BACE1 beta-site APP-cleaving enzyme 1, beta-secretase-1
  • presenilin/ ⁇ -secretase presenilin/ ⁇ -secretase.
  • BACE1 knockout mice show complete absence of beta amyloid production without obvious side effects (Luo Y et al., Neurobiol Dis. Vol. 14(1), pp. 81-88, 2003), and adult Alzheimer's disease. It has been reported that when BACE1 is deleted in a mouse model, amyloid deposition is eliminated and synaptic function is restored (Xiangyou Hu et al., J. Exp. Med. Vol. 215, No. 3, pp. 927-940, 2018), inhibition of BACE1 has become a target for the development of new therapeutic agents for the treatment of Alzheimer's disease.
  • Alzheimer's disease has a complex pathophysiology including pathological protein aggregation, neurotransmission disorders, increased oxidative stress, and microglia-mediated neuroinflammation.
  • Therapeutics targeting only one of the molecular targets associated with AD have been reported.
  • therapeutics with high BACE-1 inhibitory activity can successfully inhibit and eliminate A ⁇ production, but cannot restore cognitive function. Therefore, when developing a treatment for Alzheimer's disease, it is important to focus on multi-target drugs rather than single-target drugs.
  • Ramalin which has antioxidant, BACE-1 inhibitory and anti-inflammatory activities, can serve as a promising therapeutic agent with multiple activities that can address the complex nature of Alzheimer's disease.
  • its application is limited due to low stability in an aqueous environment and cytotoxicity at high concentrations.
  • a substituent such as a methyl group or a fluorine atom
  • Alzheimer's disease can act as a therapeutic agent for Alzheimer's disease because it shows activity against various targets related to Alzheimer's disease without toxicity, such as showing antioxidant activity that plays a role and inhibitory effect of inflammation that exacerbates Alzheimer's disease, and the present invention was completed.
  • An object of the present invention is to provide a novel derivative obtained by replacing a functional group (phenolic hydroxyl group) of a phenyl ring of a ramalin compound isolated from the Antarctic lichen Ramalina terebrata with an alkyl group or a halogen atom and a method for preparing the same. there is.
  • Another object of the present invention is to provide a pharmaceutical composition for preventing or treating degenerative brain diseases containing a ramalin derivative as an active ingredient.
  • Another object of the present invention is to provide a food composition for preventing or improving degenerative brain diseases containing a ramalin derivative as an active ingredient.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano, provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 3 , R 4 and R 5 are hydrogen atoms and R 2 is hydroxy; is excluded.
  • the present invention also provides a pharmaceutical composition for preventing or treating degenerative brain diseases containing the ramalin derivative represented by Chemical Formula 1 as an active ingredient.
  • the present invention also provides a food composition for preventing or alleviating degenerative brain diseases containing the ramalin derivative represented by Chemical Formula 1 as an active ingredient.
  • the present invention also provides a method for treating or preventing degenerative brain disease comprising the step of administering to a subject a pharmaceutical composition for preventing or treating degenerative brain disease comprising the ramalin derivative represented by Chemical Formula 1 as an active ingredient.
  • the present invention also provides a use of a composition for preventing or treating degenerative brain diseases comprising the ramalin derivative represented by Formula 1 as an active ingredient.
  • the present invention also provides a use in the manufacture of a drug for preventing or treating degenerative brain diseases comprising the ramalin derivative represented by Formula 1 as an active ingredient.
  • FIG. 1 is a diagram showing a preferred embodiment of the ramalin derivative of the present invention.
  • FIG. 2 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-2Me, a ramalin derivative of the present invention.
  • FIG. 3 is a diagram showing a 13 C NMR (100 MHz) spectrum of RA-2Me, a ramalin derivative of the present invention.
  • FIG. 4 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-2Me of the present invention.
  • FIG. 5 is a diagram showing the COZY spectrum of the Ramalin derivative RA-2Me of the present invention.
  • FIG. 6 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-3Me, a ramalin derivative of the present invention.
  • FIG. 7 is a diagram showing a 13 C NMR (100 MHz) spectrum of RA-3Me, a ramalin derivative of the present invention.
  • FIG. 8 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-3Me of the present invention.
  • FIG. 9 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-4Me, a ramalin derivative of the present invention.
  • FIG. 10 is a diagram showing a 13 C NMR (100 MHz) spectrum of RA-4Me, a ramalin derivative of the present invention.
  • FIG. 11 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-4Me of the present invention.
  • FIG. 12 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-25Me, a ramalin derivative of the present invention.
  • FIG. 13 is a diagram showing a 13 C NMR (100 MHz) spectrum of RA-25Me, a ramalin derivative of the present invention.
  • FIG. 14 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-25Me of the present invention.
  • 15 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-34Me, a ramalin derivative of the present invention.
  • 16 is a diagram showing a 13 C NMR (100 MHz) spectrum of RA-34Me, a ramalin derivative of the present invention.
  • 17 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-34Me of the present invention.
  • 19 is a diagram showing the HSQC spectrum of the Ramalin derivative RA-34Me of the present invention.
  • FIG. 20 is a diagram showing a 1 H NMR (400 MHz) spectrum of the Ramalin derivative RA-2F of the present invention.
  • 21 is a diagram showing a 13 C NMR (100 MHz) spectrum of the Ramalin derivative RA-2F of the present invention.
  • FIG. 22 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-2F of the present invention.
  • FIG. 23 is a diagram showing the HSQC spectrum of the Ramalin derivative RA-2F of the present invention.
  • FIG. 24 is a diagram showing a 1 H NMR (400 MHz) spectrum of the Ramalin derivative RA-4F of the present invention.
  • 25 is a diagram showing a 13 C NMR (100 MHz) spectrum of the Ramalin derivative RA-4F of the present invention.
  • 26 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-4F of the present invention.
  • FIG. 27 is a diagram showing the 1 H NMR (400 MHz) spectrum of the Ramalin derivative RA-24F of the present invention.
  • 29 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-24F of the present invention.
  • FIG. 30 is a diagram showing the HSQC spectrum of the Ramalin derivative RA-24F of the present invention.
  • FIG. 31 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-PF, a ramalin derivative of the present invention.
  • FIG. 32 is a diagram showing a 13 C NMR (100 MHz) spectrum of the Ramalin derivative RA-PF of the present invention.
  • the phenyl ring of ramalin (ramalin, ⁇ -glutamyl-N'-(2-hydroxyphenyl)hydrazide), a compound of molecular formula C 11 H 16 N 3 O 4 isolated from the Antarctic lichen Ramalina terebrata A derivative obtained by replacing the functional group (phenolic hydroxyl group) with a methyl group or a fluorine atom shows an inhibitory activity against BACE-1, which causes A ⁇ accumulation based on structural transformation, and plays an important role in the prevention and treatment of Alzheimer's disease by removing active oxygen It was found that it can act as a therapeutic agent for Alzheimer's disease because it shows activity against various targets related to Alzheimer's disease without toxicity, such as antioxidant activity that causes Alzheimer's disease and inhibition of inflammation that exacerbates Alzheimer's disease.
  • ramalin ⁇ -glutamyl-N'-(2-hydroxyphenyl)hydrazide
  • the present invention relates to a ramalin derivative represented by Chemical Formula 1.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.
  • each of R 1 , R 2 , R 3 , R 4 and R 5 in Formula 1 is independently a hydrogen atom, C 1 -C 6 alkyl or halogen.
  • Preferred examples of the ramalin derivative according to the present invention may be selected from Chemical Formulas 2 to 10, and are shown in FIG. 1.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano, provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 3 , R 4 and R 5 are hydrogen atoms and R 2 is hydroxy; is excluded.
  • the step (a) is performed by adding triethylamine (TEA) to a solution of 1-benzyl-N-benzyloxycarbonyl-L-glutamic acid dissolved in dichloromethane (DCM) at a temperature of -5 ° C. ) and ethylchloroformate (ECF) were sequentially added, the reaction mixture was stirred for 2 hours, and phenylhydrazine HCl salt was added while maintaining the temperature to perform a coupling reaction.
  • TAA triethylamine
  • DCM dichloromethane
  • ECF ethylchloroformate
  • the deprotection in step (b) may be performed in an atmosphere of Pd/C and H 2 gas.
  • the present invention relates to a pharmaceutical composition for preventing or treating degenerative brain diseases containing a ramalin derivative represented by Formula 1 as an active ingredient.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.
  • each of R 1 , R 2 , R 3 , R 4 and R 5 in Formula 1 is independently a hydrogen atom, C 1 -C 6 alkyl or halogen.
  • Ramalin derivatives according to the present invention may be selected from Formulas 2 to 10, as discussed above.
  • the degenerative brain disease may be selected from the group consisting of Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, Pick's disease, Creutzfeldt-Jakob disease, Huntington's disease and dementia, and particularly preferably Alzheimer's disease.
  • BACE1 expression can be inhibited.
  • the composition containing the ramalin derivative may additionally contain an appropriate pharmaceutically acceptable carrier, excipient or diluent according to a conventional method.
  • Carriers, excipients and diluents that may be included in the composition comprising the compound include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum acacia, alginates, gelatin, calcium phosphate, calcium silicate. , cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.
  • composition containing the ramalin derivative of the present invention is a powder, pill, granule, capsule, suspension, internal solution, emulsion, syrup, sterilized aqueous solution, non-aqueous solution, suspension, freeze-dried agent and suppository according to a conventional method. It may have any one formulation selected from.
  • Solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and these solid preparations contain at least one excipient such as starch, calcium carbonate, sucrose ( It is prepared by mixing sucrose, lactose, or gelatin. In addition to simple excipients, lubricants such as magnesium stearate and talc are also used.
  • Liquid preparations for oral use include suspensions, solutions for oral use, emulsions, syrups, etc.
  • Formulations for parenteral administration include sterilized aqueous solutions, non-aqueous solvents, suspensions, emulsions, freeze-dried formulations, and suppositories.
  • 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 suppositories witepsol, macrogol, tween 60, cacao butter, laurin paper, glycerogelatin, and the like may be used.
  • the pharmaceutical composition of the present invention may be administered orally or parenterally (eg, intravenous, subcutaneous, intraperitoneal or topical application) depending on the desired method, and the dosage is determined by the patient's health condition, weight, age, gender, The range varies depending on diet, excretion rate, severity of disease, drug form, administration time, administration route and administration period, but can be appropriately selected by those skilled in the art.
  • the compound of the present invention is preferably administered at 0.001 to 1000 mg/kg per day, preferably at 0.01 to 100 mg/kg. Administration may be administered once a day, or may be administered in several divided doses.
  • the dosage is not intended to limit the scope of the present invention in any way.
  • the pharmaceutical composition of the present invention can be used alone or in combination with methods using surgery, radiation therapy, hormone therapy, chemotherapy, and biological response modifiers for the prevention or treatment of Alzheimer's.
  • the present invention relates to a food composition for preventing or alleviating degenerative brain diseases containing a ramalin derivative represented by Chemical Formula 1 as an active ingredient.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.
  • each of R 1 , R 2 , R 3 , R 4 and R 5 in Formula 1 is independently a hydrogen atom, C 1 -C 6 alkyl or halogen.
  • Ramalin derivatives according to the present invention may be selected from Formulas 2 to 10, as discussed above.
  • the degenerative brain disease may be selected from the group consisting of Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, Pick's disease, Creutzfeldt-Jakob disease, Huntington's disease and dementia, and particularly preferably Alzheimer's disease.
  • the food composition containing the ramalin derivative may further include additives, excipients, or flavoring agents acceptable in food science.
  • the food of the present invention includes all forms such as functional food, nutritional supplement, health food and food additives.
  • the health functional food of the above type can be prepared in various forms according to conventional methods known in the art.
  • the ramalin derivative of the present invention can be prepared in the form of tea, juice, or drink to be consumed, or granulated, encapsulated, or powdered to be consumed.
  • functional foods include beverages (including alcoholic beverages), fruits and their processed foods (e.g. canned fruits, canned fruits, jams, marmalades, etc.), fish, meat and their processed foods (e.g.
  • ham sausage corned beef, etc.
  • Bread and noodles e.g. udon, buckwheat noodles, ramen, spaghetti, macaroni, etc.
  • fruit juice various drinks, cookies, taffy, dairy products (e.g. butter, cheese, etc.), edible vegetable oil, margarine, vegetable protein, retort food, It can be prepared by adding the ramalin derivative of the present invention to frozen foods, various seasonings (eg, soybean paste, soy sauce, sauce, etc.).
  • the health functional food also includes various forms such as functional foods, nutritional supplements, health foods, food additives, etc. as food compositions, and various forms such as Ramalin mentioned above are prepared according to conventional methods known in the art. Manufactured in the form of tea, juice, or drink, or granulated, encapsulated, or powdered, or added to beverages, fruits and processed foods, fish, meat and its processed foods, breads, noodles, and seasonings with these compounds or extracts It can be provided by manufacturing.
  • the present invention relates to a method for preventing or treating a degenerative brain disease comprising administering a composition containing a ramalin derivative represented by Chemical Formula 1 as an active ingredient.
  • the present invention relates to the use of a composition containing the ramalin derivative represented by Chemical Formula 1 as an active ingredient for the prevention or treatment of degenerative brain diseases.
  • the inhibitory activity of Ramalin derivatives on BACE-1 which causes A ⁇ accumulation
  • the antioxidant effect that plays an important role in the prevention and treatment of Alzheimer's disease by removing active oxygen and the aggravation of Alzheimer's disease
  • the anti-inflammatory effect was investigated, and as a result, the inhibitory activity of the Ramalin derivatives on BACE-1 tended to decrease in a concentration-dependent manner.
  • ramalin derivatives showed antioxidant activity similar to that of ramalin, and showed anti-inflammatory effects without toxicity in RAW 264.7 cells stimulated with LPS. Accordingly, it was confirmed that ramalin derivatives, like ramalin, show activity against various targets related to Alzheimer's disease, and therefore can act as a therapeutic agent for Alzheimer's disease.
  • treatment or treatment of degenerative brain disease comprising the step of administering to a subject a pharmaceutical composition for preventing or treating degenerative brain disease comprising a ramalin derivative represented by Chemical Formula 1 as an active ingredient; It relates to a preventive method, uses of a composition for the prevention or treatment of degenerative brain diseases comprising a ramalin derivative represented by Formula 1 as an active ingredient, and a composition for preventing or treating degenerative brain diseases comprising a ramalin derivative represented by Formula 1 as an active ingredient It relates to use in the manufacture of a prophylactic or therapeutic medicament.
  • Ramalin was successfully synthesized according to a method developed in a previous study (Yim, JH et al., WO201308959A1, March 26, 2013).
  • the synthesis was performed by coupling phenylhydrazine to the starting material, 1-benzyl-N-Cbz-L-glutamic acid. After lowering the temperature to -5°C, triethylamine (TEA) and ethyl chloroformate (ECF) were sequentially added to a solution in which the starting material was dissolved in dichloromethane (DCM). Then, the reaction mixture was stirred for about 2 hours, and the coupling reaction was completed by adding hydrazine HCl salt while maintaining the temperature.
  • TEA triethylamine
  • ECF ethyl chloroformate
  • Ramalin is stable at room temperature (i.e., 25° C.) when in dry solid form. However, it is unstable in aqueous solution. This is considered a disadvantage (Pagire, SH et al., Bioorg. Med. Chem. Lett., 2018, 28, 529-532). Therefore, to improve stability and BBB penetration, the structure was modified by replacing the phenolic hydroxyl group at position 2 with a methyl group or a fluorine atom and changing the position. Thus, several derivatives were obtained.
  • RA-2Me, RA-3Me, and RA-4Me were synthesized by exchanging the position of RA-25Me and RA-34Me, which have two methyl groups in the electron-donating methyl group and the phenyl ring. It was synthesized by introducing an extra methyl group.
  • RA-2F, RA-4F, RA-24F and RA-pentafluoride (PF) were also successfully synthesized to investigate the effect of electron-withdrawing fluorine atoms as opposed to methyl groups.
  • ramalin and its derivatives are low molecular weight antioxidants, they have a high possibility of penetrating the BBB (Gilgun-Sherki, Y.
  • Ramalin and its derivatives are water-soluble low-molecular-weight compounds and have a balance between lipophilicity and hydrophilicity, their effect on AlogP exhibiting lipophilicity was not significant. It is also considered favorable in terms of BBB penetration when considering PSA values (90 ⁇ 2 ⁇ PSA ⁇ 140 ⁇ 2 ) (Rankovic, Z., J. Med. Chem., 2015, 58, 2584-2608; Waring, MJ, Bioorg. Med. Chem. Lett., 2009, 19, 2844-2851).
  • Macrophage-like mouse cell line RAW 264.7 (KCLB No. 40071; Korean Cell Line Bank, Seoul, Korea) was cultured in 10% heat-inactivated fetal bovine serum (FBS, Invitrogen, Burlington, ON, Canada) and 5% CO 2 at 37°C. were cultured in Dulbecco's modified Eagle's medium (DMEM, Sigma-Aldrich, St. Louis, MO, USA) supplemented with 1% (w/v) antibiotic-antifungal solution (Invitrogen, Grand Island, NY, USA).
  • FBS heat-inactivated fetal bovine serum
  • DMEM Dulbecco's modified Eagle's medium
  • Cytotoxicity was determined by MTT (Amresco, Solon, OH, USA) colorimetric assay.
  • RAW 264.7 cells were seeded in a 96-well plate at a density of 2 ⁇ 10 5 cells/mL and incubated for 24 hours in the presence of various concentrations of Ramalin and its derivatives. After incubation with the test substance, MTT solution (5 ⁇ L of 5 mg/mL in PBS) was added to the wells and incubated at 37° C. for 4 hours. Then, 100 ⁇ L of fresh DMSO was treated to dissolve the crystals for 10 minutes, and the cells were detected with a microplate reader (Thermo Scientific Inc., San Diego, CA, USA) and absorbance was measured at 570 nm. Relative cell viability was calculated by comparing the absorbance of the untreated control. All experiments were performed in triplicate.
  • Nitrite accumulation was used as an indicator of NO production in the medium and nitrite levels were measured using Griess reagent (1% sulfanilamide, 0.1% N-(1-napatyl)-ethylenediamine dihydrochloride and 5% phosphoric acid).
  • Griess reagent 1% sulfanilamide, 0.1% N-(1-napatyl)-ethylenediamine dihydrochloride and 5% phosphoric acid.
  • 1 ⁇ 10 6 cells/mL were inoculated in a 96-well plate, and the concentration of Ramalin and its derivatives was displayed at 37° C. for 1 hour, followed by stimulation with 0.5 ⁇ g/mL.
  • LPS 0.5 ⁇ g/mL, Sigma-Aldrich, CA, USA
  • RA-25Me, RA-34Me, RA-2F, RA-4F, RA-24F and RA-PF also showed concentration-dependent NO inhibition, whereas RA-2Me, RA-3Me and RA-4Me showed weak anti-inflammatory show effect. Also, among these derivatives, RA-24F showed a stronger anti-inflammatory effect than Ramalin without cytotoxicity.
  • BACE-1 inhibition assay was performed using the ⁇ -Secretase FRET kit (BACE-1, Thermo Fisher Scientific, San Diego, CA, USA) according to the manufacturer's instructions. The assay was performed according to the manufacturer's protocol as previously described. Stocks of Ramalin and its derivatives were prepared (20 mM) in deionized distilled water (DDW). Samples were further diluted in assay buffer and a black 96-well microplate was mixed with 10 ⁇ L of BACE-1 substrate. The reaction was then started by adding 10 ⁇ L of 3xBACE-1 enzyme to each well. Plates were incubated for 60 minutes at room temperature in the dark.
  • BACE-1 inhibition is a promising target for AD inhibition.
  • the BACE-1 inhibitory activity of Ramalin and its derivatives was confirmed using PanVera ® BACE-1 Fluorescence Resonance Energy Transfer Assay (FRET) kit (P2985, Madison, WI, USA) (Niu, Y. et al., Chem. Biol. Drug Des., 2012, 80, 775-780).
  • FRET Fluorescence Resonance Energy Transfer Assay
  • Ramalin and all other derivatives were observed to exhibit IC50 values at micromolar concentrations. Specifically, Ramalin's BACE-1 inhibitory activity was 17.66 ⁇ 2.74 ⁇ M, and RA-25Me showed the highest activity among the derivatives, 9.81 ⁇ 1.21 ⁇ M.
  • Alzheimer's disease has a complex pathophysiology that includes pathological protein aggregation, impaired neurotransmission, increased oxidative stress, and microglia-mediated neuroinflammation.
  • Therapeutics targeting only one of the molecular targets associated with AD have been reported.
  • therapeutics with high BACE-1 inhibitory activity can successfully inhibit and eliminate A ⁇ production, but cannot restore cognitive function. Therefore, when developing a treatment for Alzheimer's disease, it is important to focus on multi-target drugs rather than single-target drugs.
  • Ramalin which has antioxidant, BACE-1 inhibitory and anti-inflammatory activities, can serve as a promising therapeutic agent with multiple activities that can address the complex nature of Alzheimer's disease.
  • its application is limited due to low stability in an aqueous environment and cytotoxicity at high concentrations.
  • RA-25Me and RA-34Me showed activities similar to Ramalin, highlighting the possibility of improving these disadvantages. Therefore, improvements in physical properties and activity of Ramalin through the synthesis of additional derivatives may be applied to the treatment and prevention of Alzheimer's disease in the future.
  • RA-24F showed a higher anti-inflammatory effect than Ramalin. No cytotoxicity.
  • Ramalin and its derivatives exhibited BACE-1 inhibitory activity in a concentration-dependent manner.
  • RA-25Me showed the highest BACE-1 inhibitory activity at a concentration of 9.81 ⁇ 1.21 ⁇ M.
  • memoquin which exhibits inhibitory activities and antioxidant effects on BACE-1 and AChE, is a suitable example of a multi-target compound (Capurro, V. et al., PLoS One, 2013, 8, e56870). Spatial and episodic memory deficits in mice with scopolamine-induced amnesia resolved after treatment with memoquine. Although further confirmation is needed in this regard, antioxidant effects are required for substantial cognitive improvement as well as elimination and inhibition of A ⁇ production in AD. Prior to Alzheimer's disease onset through A ⁇ accumulation, ROS-induced oxidative damage accelerates Alzheimer's disease induction.
  • ROS causes oxidative damage to nucleic acids, lipids, and proteins related to the induction of Alzheimer's disease, and these damaging substances were initially identified in the brains of Alzheimer's disease patients (Choi, D. Y. et al., Brain Res. Bull., 2012, 87, 144-153).
  • oxidative damage induces neuroinflammation and mediates BACE-1 activity (Zameer, S. et al., Neurotoxicology, 2019, 70, 122-134; Reddy, P. H., J. Neurochem., 2006 , 96, 1-13).
  • a ⁇ generated by BACE-1 can in turn induce ROS production, further exacerbating the symptoms of AD (Tamagno, E.
  • Ramalin derivatives according to the present invention have inhibitory activity against BACE-1, which induces A ⁇ accumulation, and exhibit antioxidant effects that play an important role in preventing and treating Alzheimer's disease by removing active oxygen. In addition, it exhibits an anti-inflammatory effect that exacerbates Alzheimer's disease, and shows an anti-inflammatory effect without toxicity in RAW 264.7 cells stimulated with LPS. As such, ramalin derivatives are active against various targets related to Alzheimer's disease, so they can act as a therapeutic agent for Alzheimer's disease. can
  • Alzheimer's disease is a complex condition that includes pathological protein aggregation, neurotransmission disorders, increased oxidative stress, and microglia-mediated neuroinflammation. have menstruation Therapeutics targeting only one of the molecular targets associated with AD have been reported. For example, therapeutics with high BACE-1 inhibitory activity can successfully inhibit and eliminate A ⁇ production, but cannot restore cognitive function.
  • Ramalin which has antioxidant, BACE-1 inhibitory and anti-inflammatory activities, can serve as a promising therapeutic agent with multiple activities that can address the complex nature of Alzheimer's disease.
  • its application is limited due to low stability in an aqueous environment and cytotoxicity at high concentrations.
  • RA-25Me and RA-34Me showed activities similar to Ramalin, highlighting the possibility of improving these disadvantages. Therefore, improvements in physical properties and activity of Ramalin through the synthesis of additional derivatives may be applied to the treatment and prevention of Alzheimer's disease in the future.
  • RA-24F showed a higher anti-inflammatory effect than Ramalin. No cytotoxicity.
  • Ramalin and its derivatives exhibited BACE-1 inhibitory activity in a concentration-dependent manner.
  • RA-25Me showed the highest BACE-1 inhibitory activity at a concentration of 9.81 ⁇ 1.21 ⁇ M.
  • memoquin which exhibits inhibitory activities and antioxidant effects on BACE-1 and AChE, is a suitable example of a multi-target compound (Capurro, V. et al., PLoS One, 2013, 8, e56870). Spatial and episodic memory deficits in mice with scopolamine-induced amnesia resolved after treatment with memoquine. Although further confirmation is needed in this regard, antioxidant effects are required for substantial cognitive improvement as well as elimination and inhibition of A ⁇ production in AD. Prior to Alzheimer's disease onset through A ⁇ accumulation, ROS-induced oxidative damage accelerates Alzheimer's disease induction.
  • ROS causes oxidative damage to nucleic acids, lipids, and proteins related to the induction of Alzheimer's disease, and these damaging substances were initially identified in the brains of Alzheimer's disease patients (Choi, D. Y. et al., Brain Res. Bull., 2012, 87, 144-153).
  • oxidative damage induces neuroinflammation and mediates BACE-1 activity (Zameer, S. et al., Neurotoxicology, 2019, 70, 122-134; Reddy, P. H., J. Neurochem., 2006 , 96, 1-13).
  • a ⁇ generated by BACE-1 can in turn induce ROS production, further exacerbating the symptoms of AD (Tamagno, E.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Neurosurgery (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Neurology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Nutrition Science (AREA)
  • Polymers & Plastics (AREA)
  • Food Science & Technology (AREA)
  • Hospice & Palliative Care (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Psychiatry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a ramalin derivative and use thereof for the prevention or treatment of neurodegenerative diseases and, more specifically, to use of a ramalin derivative for the prevention or treatment of neurodegenerative diseases, in which a derivative obtained by substituting a hydroxyl group of a phenyl ring of ramalin with a methyl group or a fluorine atom exhibits inhibitory activity against BACE-1 and antioxidant and anti-inflammatory activities without toxicity.

Description

신규한 라말린 유도체 및 이의 퇴행성 뇌질환의 예방 또는 치료 용도Novel Ramalin derivatives and their use for prevention or treatment of degenerative brain diseases

본 발명은 라말린 유도체 및 이의 퇴행성 뇌질환의 예방 또는 치료 용도에 관한 것으로, 더욱 상세하게는 더욱 상세하게는 라말린의 페닐 고리의 수산기를 메틸기 또는 불소 원자로 대체하여 얻은 유도체가 BACE-1에 대한 억제 활성, 항산화 및 항염 활성을 독성 없이 나타냄으로써 퇴행성 뇌질환의 예방 또는 치료 용도에 사용하는 신규한 라말린 유도체 및 이의 퇴행성 뇌질환의 예방 또는 치료 용도에 관한 것이다.The present invention relates to a ramalin derivative and its use for the prevention or treatment of degenerative brain diseases, and more particularly, a derivative obtained by replacing a hydroxyl group of a phenyl ring of ramalin with a methyl group or a fluorine atom is effective against BACE-1. It relates to a novel ramalin derivative used for preventing or treating degenerative brain diseases by exhibiting inhibitory activity, antioxidant and anti-inflammatory activity without toxicity, and a use thereof for preventing or treating degenerative brain diseases.

2016년 전 세계 치매 환자는 2,380만 명으로 인구 고령화와 인구 증가로 매년 증가 추세다. 따라서 치매 환자에 대한 돌봄과 지원은 가족, 의료 시스템 및 사회 전체에 광범위한 영향을 미친다(Nichols, E. et al., Lancet Neurol., 2019, 18, 88-106; Etters, L. et al., J. Am. Assoc. Nurse Parct., 2008, 20, 423-428). 치매는 기대수명 단축과 관련이 있는 것으로 알려져 있으며, 이로 인해 선진국 사망 원인 5위가 된다(Dumurgier, J. et al., Lancet Healthy Longev., 2021, 2, e449-e450). 또한, 이 사망자 수의 절반 이상을 차지하는 알츠하이머병(Alzheimer’s Disease, 이후 AD라 기재함)은 치매의 가장 흔한 원인이다. 알츠하이머병 진단을 받은 환자는 기억 장애, 언어 장애, 시공간 장애 및 망상 및 환각을 포함한 정신 장애를 포함한 증상이 점차 악화된다. 또한, 알츠하이머병의 발병기전은 다양하고 복잡하다. 시냅스 틈의 낮은 아세틸콜린(ACh) 농도, 베타아밀로이드(β-amyloid, 이후 Aβ라 기재함) 펩티드 응집 및 축적, 미세소관 관련 단백질 타우의 얽힘, 산화 스트레스가 알츠하이머병의 주요 원인으로 간주된다. 또한 뇌에서 Aβ 응집 및 축적은 Aβ 생성 증가, Aβ 프로테아제 활성 감소 또는 BBB(blood-brain barrier, 혈액뇌장벽)를 통한 Aβ 수송 변경의 결과일 수 있다는 것이 관찰되었다(Mucke, L. Alzheimer's disease. Nature, 2019, 461, 895-897). 또한, Aβ 생성 증가와 관련된 β-부위 APP 절단 효소 1(BACE-1)은 알츠하이머병 예방 및 치료의 표적으로 여겨진다(Vassar, R. Bace 1. J. Mol. Neurosci., 2004, 23, 105-113). 또한 Aβ는 BACE-1과 BACE-2에 의한 아밀로이드 전구체 단백질(APP)의 순차적 절단을 통해 생성되며, BACE-1은 이 과정을 시작하는 핵심 효소이다(Roberson, E. D. et al., Science, 2006, 314, 781-784). 인간 APP 형질전환 마우스를 대상으로 한 연구에 기반한 실험적 증거는 기억력 결핍이 BACE-1 유전자 결실을 통해 예방될 수 있음을 시사하고(Oakley, H. et al., J. Neurosci., 2006, 26, 10129-10140). 보고된 바에 따르면 Aβ 응집체가 병원성 매개체로 작용하여 산화 스트레스, 신경 염증 및 BACE-1 발현을 유도할 수 있다. 이는 AD를 더욱 악화시킨다(Tamagno, E. et al., Free Radic. Biol. Med., 2006, 41, 202-212). BACE-1을 통한 Aβ 축적 외에도 산화 스트레스와 염증은 Aβ 생산을 향상시킬 수 있다. Aβ 형성과 염증 사이의 연관성이 있다는 연구에서 만성 염증이 있는 원숭이의 뇌에서 플라크가 관찰되었다(Philippens, I. H. et al., J. Alzheimer's Dis., 2017, 55, 101-113). 따라서 염증이 지질, 단백질 및 핵산에 산화적 손상을 일으키는 활성산소종(ROS)의 생성을 증가시킨다는 점을 감안할 때, 항염증제 사용을 기반으로 염증을 감소시키는 것은 알츠하이머병의 유병률을 상당히 낮출 수 있다(McGeer, E. G. et al., Exp. Gerontol., 1998, 33, 371-378). 또한 염증과 관련된 NF-κB는 다양한 기전에 의해 조절되는 BACE-1의 발현에 관여한다(Roßner, S. et al., Prog. Neurobiol., 2006, 79, 95-111). 따라서 BACE-1, 염증 및 ROS는 모두 알츠하이머병 발병부터 발병 후 기간까지 Aβ 형성과 관련이 있다.In 2016, the number of dementia patients worldwide was 23.8 million, which is increasing every year due to population aging and population growth. Thus, care and support for people with dementia have far-reaching implications for families, the health system, and society as a whole (Nichols, E. et al., Lancet Neurol., 2019, 18, 88-106; Etters, L. et al., J. Am. Assoc. Nurse Parct., 2008, 20, 423-428). Dementia is known to be associated with reduced life expectancy, making it the 5th leading cause of death in developed countries (Dumurgier, J. et al., Lancet Healthy Longev., 2021, 2, e449-e450). In addition, Alzheimer's Disease (hereinafter referred to as AD), which accounts for more than half of these deaths, is the most common cause of dementia. Patients diagnosed with Alzheimer's disease experience progressively worse symptoms, including memory impairment, speech impairment, visuospatial disturbances, and mental disorders including delusions and hallucinations. In addition, the pathogenesis of Alzheimer's disease is diverse and complex. Low concentration of acetylcholine (ACh) in the synaptic cleft, aggregation and accumulation of β-amyloid (hereinafter referred to as Aβ) peptide, entanglement of the microtubule-associated protein tau, and oxidative stress are considered to be the main causes of Alzheimer's disease. It was also observed that Aβ aggregation and accumulation in the brain could be the result of increased Aβ production, decreased Aβ protease activity, or altered Aβ transport across the BBB (blood-brain barrier) (Mucke, L. Alzheimer's disease. Nature , 2019, 461, 895-897). In addition, β-site APP cleavage enzyme 1 (BACE-1) associated with increased production of Aβ is considered a target for the prevention and treatment of Alzheimer's disease (Vassar, R. Bace 1. J. Mol. Neurosci., 2004, 23, 105- 113). Aβ is also produced through sequential cleavage of amyloid precursor protein (APP) by BACE-1 and BACE-2, and BACE-1 is a key enzyme initiating this process (Roberson, E. D. et al., Science, 2006, 314, 781-784). Experimental evidence based on studies in human APP transgenic mice suggests that memory deficits can be prevented by deletion of the BACE-1 gene (Oakley, H. et al., J. Neurosci., 2006, 26; 10129-10140). Reportedly, Aβ aggregates can act as pathogenic mediators and induce oxidative stress, neuroinflammation, and BACE-1 expression. This further aggravates AD (Tamagno, E. et al., Free Radic. Biol. Med., 2006, 41, 202-212). In addition to Aβ accumulation through BACE-1, oxidative stress and inflammation can enhance Aβ production. In a study of the association between Aβ formation and inflammation, plaques were observed in the brains of monkeys with chronic inflammation (Philippens, I. H. et al., J. Alzheimer's Dis., 2017, 55, 101-113). Therefore, given that inflammation increases the production of reactive oxygen species (ROS) that cause oxidative damage to lipids, proteins, and nucleic acids, reducing inflammation based on the use of anti-inflammatory drugs can significantly lower the prevalence of Alzheimer's disease ( McGeer, E. G. et al., Exp. Gerontol., 1998, 33, 371-378). In addition, NF-κB associated with inflammation is involved in the expression of BACE-1 regulated by various mechanisms (Roßner, S. et al., Prog. Neurobiol., 2006, 79, 95-111). Thus, BACE-1, inflammation and ROS are all associated with Aβ formation from the onset of Alzheimer's disease to the post-onset period.

다른 관점에서 보면 알츠하이머병의 발달과 같이 ROS에 의한 산화 스트레스가 나타나며 알츠하이머병의 조기 발병과 악화의 원인이 되는 것으로 보고되고 있다(Smith, M. A. et al., Biochim. Biophys. Acta Mol. Basis Dis., 2000, 1502, 139-144). 정상적인 조건에서 생성되는 미토콘드리아 ROS는 항산화제 및 관련 효소에 의해 균형을 이룬다. 그러나 이와 관련하여 산화 손상으로 인한 기능 장애는 높은 수준의 ROS 생성과 세포자살 유도 단백질의 방출을 초래한다(Dumont, M. et al., Free Radic Biol Med., 2011, 51, 1014-1026; Wang, X. et al., Biochim. Biophys. Acta Mol. Basis Dis., 2014, 1842, 1240-1247; Takuma, K. et al., J. Pharmacol. Sci., 2005, 97, 312-316). 또한 산화 스트레스로 인한 미토콘드리아 소포체 기능 장애는 UPR(Unfolded protein response)(Smith, M. A. et al., Biochim. Biophys. Acta Mol. Basis Dis., 2000, 1502, 139-144; Oakley, H. et al., J. Neurosci., 2006, 26, 10129-10140; Konno, T. et al., Cells, 2021, 10, 233)로 알려진 세포 스트레스 반응을 유도하여 독성 단백질의 축적으로부터 세포를 보호할 수 있지만 지속되면, 신경 퇴행으로 이어질 수 있다(Wang, X. et al., Biochim. Biophys. Acta Mol. Basis Dis., 2014, 1842, 1240-1247; Hoozemans, J. J. M. et al., Acta Neuropathol., 2005, 110, 165-172).From another point of view, oxidative stress by ROS appears like the development of Alzheimer's disease, and it has been reported to be the cause of early onset and exacerbation of Alzheimer's disease (Smith, M. A. et al., Biochim. Biophys. Acta Mol. Basis Dis. , 2000, 1502, 139-144). Mitochondrial ROS produced under normal conditions are balanced by antioxidants and related enzymes. However, dysfunction due to oxidative damage in this regard results in high levels of ROS production and release of apoptosis-inducing proteins (Dumont, M. et al., Free Radic Biol Med., 2011, 51, 1014-1026; Wang , X. et al., Biochim. Biophys. Acta Mol. Basis Dis., 2014, 1842, 1240-1247; Takuma, K. et al., J. Pharmacol. Sci., 2005, 97, 312-316). In addition, mitochondrial endoplasmic reticulum dysfunction due to oxidative stress is UPR (Unfolded protein response) (Smith, M. A. et al., Biochim. Biophys. Acta Mol. Basis Dis., 2000, 1502, 139-144; Oakley, H. et al. , J. Neurosci., 2006, 26, 10129-10140; Konno, T. et al., Cells, 2021, 10, 233) can protect cells from accumulation of toxic proteins by inducing a cellular stress response, but , can lead to neurodegeneration (Wang, X. et al., Biochim. Biophys. Acta Mol. Basis Dis., 2014, 1842, 1240-1247; Hoozemans, J. J. M. et al., Acta Neuropathol., 2005, 110 , 165-172).

알츠하이머병은 세포외 베타아밀로이드(β-amyloid, Aβ) 플라크(plaque) 및 세포 내 신경섬유 엉킴의 신경 병리학적 특징을 나타낸다. 베타아밀로이드를 생성하는 아밀로이드 전구체 단백질 (amyloid precursor protein: APP)의 내부 단백질 분해 절단은 베타세크레타제(β-secretase) 및 감마세크레타제(γ-secretase)의 두 가지 프로테아제의 연속적인 작용을 포함하고 있는데, 베타세크레타제는 큰 수용성 세포 외 단편(soluble APPβ)을 유리시키는 APP의 β 절단에 의해 베타아밀로이드 생산의 첫 단계를 중재한다. APP(APP-CTF, C99)의 C 말단 단편(carboxyl terminal fragment: CTF)은 몇 가지 위치에서 감마세크레타제에 의해 절단되어 병원성 종 Aβ42 및 Aβ40이 생성되는 것으로 알려져 있다.Alzheimer's disease exhibits neuropathological features of extracellular beta-amyloid (β-amyloid, Aβ) plaques and intracellular nerve fiber tangles. Internal proteolytic cleavage of amyloid precursor protein (APP), which produces beta-amyloid, involves the sequential action of two proteases, β-secretase and γ-secretase. β-secretase mediates the first step in beta-amyloid production by β-cleavage of APP, which releases a large soluble extracellular fragment (APPβ). It is known that the C-terminal fragment (CTF) of APP (APP-CTF, C99) is cleaved by gamma secretase at several positions to generate the pathogenic species Aβ42 and Aβ40.

또한, 이러한 베타아밀로이드는 BACE1(beta-site APP-cleaving enzyme 1, beta-secretase-1) 및 프리시닐린/감마세크레타제(presenilin/γ-secretase)에 의해 매개되는, APP의 연속 분열에 의해 생성되며, 베타아밀로이드의 생성 및 응집은 신경퇴화, 신경섬유의 엉킴 현상, 염증 및 뉴런의 손실을 일으키는 복잡한 병리학적 유발을 촉발시키는 중요한 역할을 한다.In addition, these beta-amyloids are produced by continuous cleavage of APP mediated by BACE1 (beta-site APP-cleaving enzyme 1, beta-secretase-1) and presenilin/γ-secretase. The production and aggregation of beta-amyloid play an important role in triggering complex pathological events that cause neurodegeneration, entanglement of nerve fibers, inflammation, and loss of neurons.

또한, BACE1 녹아웃 생쥐가 명백한 부작용 없이 베타아밀로이드 생산의 완전한 부재를 나타내고 있다는 내용이 보고되었고(Luo Y et al., Neurobiol Dis. Vol. 14(1), pp. 81-88, 2003), 성체 알츠하이머 마우스 모델에서 BACE1을 결실시킬 경우, 아밀로이드의 침착(deposition) 현상이 제거되고, 시냅스의 기능이 회복된다는 점이 보고되면서(Xiangyou Hu et al., J. Exp. Med. Vol. 215, No. 3, pp. 927-940, 2018), BACE1의 억제는 알츠하이머병의 치료를 위한 새로운 치료제 개발을 위한 타겟이 되고 있다.In addition, it has been reported that BACE1 knockout mice show complete absence of beta amyloid production without obvious side effects (Luo Y et al., Neurobiol Dis. Vol. 14(1), pp. 81-88, 2003), and adult Alzheimer's disease. It has been reported that when BACE1 is deleted in a mouse model, amyloid deposition is eliminated and synaptic function is restored (Xiangyou Hu et al., J. Exp. Med. Vol. 215, No. 3, pp. 927-940, 2018), inhibition of BACE1 has become a target for the development of new therapeutic agents for the treatment of Alzheimer's disease.

한편, 알츠하이머병은 병리학적 단백질 응집, 신경 전달 장애, 산화 스트레스 증가, 미세아교세포 매개 신경 염증을 포함하는 복잡한 병태생리를 가지고 있다. AD와 관련된 분자 표적 중 하나만을 표적으로 하는 치료제가 보고되었다. 예를 들어, BACE-1 억제 활성이 높은 치료제는 Aβ 생성을 성공적으로 억제하고 제거할 수 있지만 인지 기능을 회복할 수는 없다. 따라서 알츠하이머병 치료제 개발 시 단일 표적 약물보다는 다중 표적 약물에 집중하는 것이 중요하다. 항산화, BACE-1 억제 및 항염증 활성을 갖는 라말린은 알츠하이머병의 복잡한 특성을 해결할 수 있는 다중 활성을 가진 유망한 치료제 역할을 할 수 있다. 그러나 수성 환경에서 낮은 안정성과 높은 농도의 세포 독성으로 인해 적용에 제한이 있다.Meanwhile, Alzheimer's disease has a complex pathophysiology including pathological protein aggregation, neurotransmission disorders, increased oxidative stress, and microglia-mediated neuroinflammation. Therapeutics targeting only one of the molecular targets associated with AD have been reported. For example, therapeutics with high BACE-1 inhibitory activity can successfully inhibit and eliminate Aβ production, but cannot restore cognitive function. Therefore, when developing a treatment for Alzheimer's disease, it is important to focus on multi-target drugs rather than single-target drugs. Ramalin, which has antioxidant, BACE-1 inhibitory and anti-inflammatory activities, can serve as a promising therapeutic agent with multiple activities that can address the complex nature of Alzheimer's disease. However, its application is limited due to low stability in an aqueous environment and cytotoxicity at high concentrations.

다양한 장애로 인해 AD에 대한 치료법은 지금까지 확립되지 않았다. 또한, 질병의 발병 기전이 아직 불분명하고 완전히 이해되지 않았으며, 치료에 사용하거나 진행을 멈출 수 있는 승인된 약물이 없다. 따라서 이 문제를 해결하기 위해 특정 작용기전만을 표적으로 하는 것이 아니라 AD 치료제로 사용할 수 있는 항산화, 항염, BACE-1 억제 활성 등의 다양한 생리활성을 나타내는 약물이 필요하다.Because of its various disorders, no cure has been established so far for AD. In addition, the pathogenesis of the disease is still unclear and not fully understood, and there are no approved drugs available to treat or halt its progression. Therefore, to solve this problem, it is necessary to develop a drug that exhibits various physiological activities such as antioxidant, anti-inflammatory, and BACE-1 inhibitory activity that can be used as an AD treatment rather than targeting a specific mechanism of action.

한편, 남극의 지의류인 Ramalina terebrata에서 분리된 라말린은 강력한 항산화 및 항균 효과를 발휘한다(Paudel, B. et al., Z. Naturforsch. C., 2010, 65, 34-38; Paudel, B. et al., Phytomedicine, 2011, 18, 1285-1290). 이전 연구에서 산화성 질환 치료제, 노화방지 기능성 식품, 미백 및 주름개선 기능성 화장품으로 개발하기까지 라말린의 다양한 생리활성을 조사하였다(Yim, J. H. et al., WO2010053327A2, May 14, 2010). 또한 강력한 항산화력으로 인해 항염증(Park, B. et al., Biosci. Biotechnol. Biochem., 2015, 79, 539-552), 지질 축적 감소(Kim, B. et al., Yonsei Med. J., 2018, 59, 85-91) 및 항간 섬유증(Kim, M. K. et al., Bioche.Biophys. Res. Commun., 2018, 504, 25-33) 활성도 나타내는 것으로 나타났다. BACE-1 억제 효과는 물론 항산화 및 항염 효과가 확인되어 알츠하이머병 치료제로서의 가능성을 시사하고 있다.On the other hand, ramalin isolated from Ramalina terebrata, an Antarctic lichen, exerts strong antioxidant and antibacterial effects (Paudel, B. et al., Z. Naturforsch. C., 2010, 65, 34-38; Paudel, B. et al., Phytomedicine, 2011, 18, 1285-1290). In previous studies, various physiological activities of Ramalin were investigated until it was developed as an oxidative disease treatment, anti-aging functional food, and whitening and wrinkle-improving functional cosmetics (Yim, J. H. et al., WO2010053327A2, May 14, 2010). In addition, anti-inflammatory (Park, B. et al., Biosci. Biotechnol. Biochem., 2015, 79, 539-552) and reduction of lipid accumulation (Kim, B. et al., Yonsei Med. J. , 2018, 59, 85-91) and anti-hepatic fibrosis (Kim, M. K. et al., Bioche. Biophys. Res. Commun., 2018, 504, 25-33) activity. Antioxidant and anti-inflammatory effects as well as BACE-1 inhibitory effects were confirmed, suggesting potential as a treatment for Alzheimer's disease.

이에, 본 발명자들은 상기 문제점을 해결하고자 예의 노력한 결과, 남극 지의류 라말리나 테레브라타(Ramalina terebrata)로부터 분리한 분자식 C11H16N3O4의 화합물 라말린(ramalin, γ-glutamyl-N'-(2-hydroxyphenyl)hydrazide)의 페닐 고리에 메틸기 또는 불소 원자와 같은 치환기를 결합시켜 얻은 유도체가 구조 변형을 기반으로 Aβ 축적을 유발하는 BACE-1에 대한 억제 활성을 나타내고, 활성산소를 제거함으로써 알츠하이머병 예방 및 치료에 중요한 역할을 하는 항산화 활성과 알츠하이머병을 악화시키는 염증의 억제 효과를 나타내는 등 독성 없이 알츠하이머병과 관련된 여러 표적에 대해 활성을 나타내므로 알츠하이머병의 치료제로 작용할 수 있다는 것을 확인하고, 본 발명을 완성하게 되었다.Accordingly, the present inventors have made diligent efforts to solve the above problems, and as a result, a compound of molecular formula C11H16N3O4 isolated from the Antarctic lichen Ramalina terebrata (ramalin, γ-glutamyl-N'-(2-hydroxyphenyl)hydrazide) ), a derivative obtained by binding a substituent such as a methyl group or a fluorine atom to the phenyl ring of ) exhibits inhibitory activity against BACE-1, which causes Aβ accumulation based on structural transformation, and removes active oxygen, which is important for the prevention and treatment of Alzheimer's disease. It was confirmed that it can act as a therapeutic agent for Alzheimer's disease because it shows activity against various targets related to Alzheimer's disease without toxicity, such as showing antioxidant activity that plays a role and inhibitory effect of inflammation that exacerbates Alzheimer's disease, and the present invention was completed.

발명의 요약Summary of Invention

본 발명의 목적은 남극 지의류 라말리나 테레브라타(Ramalina terebrata)로부터 분리한 라말린 화합물의 페닐 고리의 작용기(페놀성 수산기)를 알킬기 또는 할로겐 원자로 대체하여 얻은 신규한 유도체 및 그 제조방법을 제공하는데 있다.An object of the present invention is to provide a novel derivative obtained by replacing a functional group (phenolic hydroxyl group) of a phenyl ring of a ramalin compound isolated from the Antarctic lichen Ramalina terebrata with an alkyl group or a halogen atom and a method for preparing the same. there is.

본 발명의 다른 목적은 라말린 유도체를 유효성분으로 함유하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물을 제공하는데 있다.Another object of the present invention is to provide a pharmaceutical composition for preventing or treating degenerative brain diseases containing a ramalin derivative as an active ingredient.

본 발명의 다른 목적은 라말린 유도체를 유효성분으로 함유하는 퇴행성 뇌질환의 예방 또는 개선용 식품 조성물을 제공하는데 있다.Another object of the present invention is to provide a food composition for preventing or improving degenerative brain diseases containing a ramalin derivative as an active ingredient.

상기 목적을 달성하기 위하여, 본 발명은 화학식 1로 표시되는 것을 특징으로 하는 라말린 유도체를 제공한다:In order to achieve the above object, the present invention provides a ramalin derivative represented by Formula 1:

[화학식 1][Formula 1]

Figure PCTKR2022016326-appb-img-000001
Figure PCTKR2022016326-appb-img-000001

화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬, C3-C6 사이클로알킬, 할로겐, 하이드록시, 카복실, 아미노 또는 시아노이고, 단, R1, R2, R3, R4 및 R5는 동시에 수소원자이거나 R1, R2, R3 및 R4는 수소원자이고, R5는 하이드록시인 경우는 제외된다.In Formula 1, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.

본 발명은 또한, (a) 출발 물질인 화학식 11의 1-벤질-N-벤질옥시카보닐-L-글루탐산과 화학식 12의 페닐하이드라진의 커플링 반응을 수행하여 화학식 13의 화합물을 수득하는 단계; 및 (b) 화학식 13의 글루탐산 하이드레이트의 벤질기(Bn) 및 벤질옥시카보닐기(Cbz)의 탈보호화를 수행한 다음 수득한 조생성물을 재결정화하여 화학식 1의 라말린 유도체를 수득하는 단계를 포함하는 상기 라말린 유도체의 제조방법을 제공한다:(a) obtaining a compound of Formula 13 by performing a coupling reaction between 1-benzyl-N-benzyloxycarbonyl-L-glutamic acid of Formula 11 as a starting material and phenylhydrazine of Formula 12; and (b) deprotecting the benzyl group (Bn) and benzyloxycarbonyl group (Cbz) of the glutamic acid hydrate of Formula 13 and then recrystallizing the obtained crude product to obtain a Ramalin derivative of Formula 1. Provided is a method for preparing the ramalin derivative comprising:

[화학식 1][Formula 1]

Figure PCTKR2022016326-appb-img-000002
Figure PCTKR2022016326-appb-img-000002

[화학식 11][Formula 11]

Figure PCTKR2022016326-appb-img-000003
Figure PCTKR2022016326-appb-img-000003

[화학식 12][Formula 12]

Figure PCTKR2022016326-appb-img-000004
Figure PCTKR2022016326-appb-img-000004

[화학식 13][Formula 13]

Figure PCTKR2022016326-appb-img-000005
Figure PCTKR2022016326-appb-img-000005

화학식 1, 12 및 13에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬, C3-C6 사이클로알킬, 할로겐, 하이드록시, 카복실, 아미노 또는 시아노이고, 단, R1, R2, R3, R4 및 R5는 동시에 수소원자이거나 R1, R3, R4 및 R5는 수소원자이고, R2는 하이드록시인 경우는 제외된다.In Formulas 1, 12 and 13, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano, provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 3 , R 4 and R 5 are hydrogen atoms and R 2 is hydroxy; is excluded.

본 발명은 또한, 화학식 1로 표시되는 라말린 유도체를 유효성분으로 함유하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물을 제공한다.The present invention also provides a pharmaceutical composition for preventing or treating degenerative brain diseases containing the ramalin derivative represented by Chemical Formula 1 as an active ingredient.

본 발명은 또한, 화학식 1로 표시되는 라말린 유도체를 유효성분으로 함유하는 퇴행성 뇌질환의 예방 또는 개선용 식품 조성물을 제공한다.The present invention also provides a food composition for preventing or alleviating degenerative brain diseases containing the ramalin derivative represented by Chemical Formula 1 as an active ingredient.

본 발명은 또한, 상기 화학식 1로 표시되는 라말린 유도체를 유효성분으로 포함하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물을 객체(subject)에 투여하는 단계를 포함하는 퇴행성 뇌질환의 치료 또는 예방방법을 제공한다.The present invention also provides a method for treating or preventing degenerative brain disease comprising the step of administering to a subject a pharmaceutical composition for preventing or treating degenerative brain disease comprising the ramalin derivative represented by Chemical Formula 1 as an active ingredient. provides

본 발명은 또한, 상기 화학식 1로 표시되는 화학식 1로 표시되는 라말린 유도체를 유효성분으로 포함하는 퇴행성 뇌질환의 예방 또는 치료용 조성물의 용도를 제공한다.The present invention also provides a use of a composition for preventing or treating degenerative brain diseases comprising the ramalin derivative represented by Formula 1 as an active ingredient.

본 발명은 또한, 상기 화학식 1로 표시되는 라말린 유도체를 유효성분으로 포함하는 퇴행성 뇌질환의 예방 또는 치료용 약제의 제조에 있어서의 용도를 제공한다.The present invention also provides a use in the manufacture of a drug for preventing or treating degenerative brain diseases comprising the ramalin derivative represented by Formula 1 as an active ingredient.

도 1은 본 발명의 라말린 유도체의 바람직한 실시예를 도시한 도면이다.1 is a diagram showing a preferred embodiment of the ramalin derivative of the present invention.

도 2는 본 발명의 라말린 유도체 RA-2Me의 1H NMR (400 MHz) 스펙트럼을 나타낸 도면이다.2 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-2Me, a ramalin derivative of the present invention.

도 3은 본 발명의 라말린 유도체 RA-2Me의 13C NMR (100 MHz) 스펙트럼을 나타낸 도면이다.3 is a diagram showing a 13 C NMR (100 MHz) spectrum of RA-2Me, a ramalin derivative of the present invention.

도 4는 본 발명의 라말린 유도체 RA-2Me의 DEPT 스펙트럼을 나타낸 도면이다.4 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-2Me of the present invention.

도 5는 본 발명의 라말린 유도체 RA-2Me의 COSY 스펙트럼을 나타낸 도면이다.5 is a diagram showing the COZY spectrum of the Ramalin derivative RA-2Me of the present invention.

도 6은 본 발명의 라말린 유도체 RA-3Me의 1H NMR (400 MHz) 스펙트럼을 나타낸 도면이다.6 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-3Me, a ramalin derivative of the present invention.

도 7은 본 발명의 라말린 유도체 RA-3Me의 13C NMR (100 MHz) 스펙트럼을 나타낸 도면이다.7 is a diagram showing a 13 C NMR (100 MHz) spectrum of RA-3Me, a ramalin derivative of the present invention.

도 8은 본 발명의 라말린 유도체 RA-3Me의 DEPT 스펙트럼을 나타낸 도면이다.8 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-3Me of the present invention.

도 9는 본 발명의 라말린 유도체 RA-4Me의 1H NMR (400 MHz) 스펙트럼을 나타낸 도면이다.9 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-4Me, a ramalin derivative of the present invention.

도 10은 본 발명의 라말린 유도체 RA-4Me의 13C NMR (100 MHz) 스펙트럼을 나타낸 도면이다.10 is a diagram showing a 13 C NMR (100 MHz) spectrum of RA-4Me, a ramalin derivative of the present invention.

도 11은 본 발명의 라말린 유도체 RA-4Me의 DEPT 스펙트럼을 나타낸 도면이다.11 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-4Me of the present invention.

도 12는 본 발명의 라말린 유도체 RA-25Me의 1H NMR (400 MHz) 스펙트럼을 나타낸 도면이다.12 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-25Me, a ramalin derivative of the present invention.

도 13은 본 발명의 라말린 유도체 RA-25Me의 13C NMR (100 MHz) 스펙트럼을 나타낸 도면이다.13 is a diagram showing a 13 C NMR (100 MHz) spectrum of RA-25Me, a ramalin derivative of the present invention.

도 14는 본 발명의 라말린 유도체 RA-25Me의 DEPT 스펙트럼을 나타낸 도면이다.14 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-25Me of the present invention.

도 15는 본 발명의 라말린 유도체 RA-34Me의 1H NMR (400 MHz) 스펙트럼을 나타낸 도면이다.15 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-34Me, a ramalin derivative of the present invention.

도 16은 본 발명의 라말린 유도체 RA-34Me의 13C NMR (100 MHz) 스펙트럼을 나타낸 도면이다.16 is a diagram showing a 13 C NMR (100 MHz) spectrum of RA-34Me, a ramalin derivative of the present invention.

도 17은 본 발명의 라말린 유도체 RA-34Me의 DEPT 스펙트럼을 나타낸 도면이다.17 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-34Me of the present invention.

도 18은 본 발명의 라말린 유도체 RA-34Me의 COSY 스펙트럼을 나타낸 도면이다.18 is a diagram showing the COZY spectrum of the Ramalin derivative RA-34Me of the present invention.

도 19는 본 발명의 라말린 유도체 RA-34Me의 HSQC 스펙트럼을 나타낸 도면이다.19 is a diagram showing the HSQC spectrum of the Ramalin derivative RA-34Me of the present invention.

도 20는 본 발명의 라말린 유도체 RA-2F의 1H NMR (400 MHz) 스펙트럼을 나타낸 도면이다.20 is a diagram showing a 1 H NMR (400 MHz) spectrum of the Ramalin derivative RA-2F of the present invention.

도 21은 본 발명의 라말린 유도체 RA-2F의 13C NMR (100 MHz) 스펙트럼을 나타낸 도면이다.21 is a diagram showing a 13 C NMR (100 MHz) spectrum of the Ramalin derivative RA-2F of the present invention.

도 22는 본 발명의 라말린 유도체 RA-2F의 DEPT 스펙트럼을 나타낸 도면이다.22 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-2F of the present invention.

도 23은 본 발명의 라말린 유도체 RA-2F의 HSQC 스펙트럼을 나타낸 도면이다.23 is a diagram showing the HSQC spectrum of the Ramalin derivative RA-2F of the present invention.

도 24는 본 발명의 라말린 유도체 RA-4F의 1H NMR (400 MHz) 스펙트럼을 나타낸 도면이다.24 is a diagram showing a 1 H NMR (400 MHz) spectrum of the Ramalin derivative RA-4F of the present invention.

도 25는 본 발명의 라말린 유도체 RA-4F의 13C NMR (100 MHz) 스펙트럼을 나타낸 도면이다.25 is a diagram showing a 13 C NMR (100 MHz) spectrum of the Ramalin derivative RA-4F of the present invention.

도 26은 본 발명의 라말린 유도체 RA-4F의 DEPT 스펙트럼을 나타낸 도면이다.26 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-4F of the present invention.

도 27은 본 발명의 라말린 유도체 RA-24F의 1H NMR (400 MHz) 스펙트럼을 나타낸 도면이다.27 is a diagram showing the 1 H NMR (400 MHz) spectrum of the Ramalin derivative RA-24F of the present invention.

도 28은 본 발명의 라말린 유도체 RA-24F의 13C NMR (100 MHz) 스펙트럼을 나타낸 도면이다.28 is a diagram showing a 13 C NMR (100 MHz) spectrum of the Ramalin derivative RA-24F of the present invention.

도 29는 본 발명의 라말린 유도체 RA-24F의 DEPT 스펙트럼을 나타낸 도면이다.29 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-24F of the present invention.

도 30은 본 발명의 라말린 유도체 RA-24F의 HSQC 스펙트럼을 나타낸 도면이다.30 is a diagram showing the HSQC spectrum of the Ramalin derivative RA-24F of the present invention.

도 31은 본 발명의 라말린 유도체 RA-PF의 1H NMR (400 MHz) 스펙트럼을 나타낸 도면이다.31 is a diagram showing a 1 H NMR (400 MHz) spectrum of RA-PF, a ramalin derivative of the present invention.

도 32는 본 발명의 라말린 유도체 RA-PF의 13C NMR (100 MHz) 스펙트럼을 나타낸 도면이다.32 is a diagram showing a 13 C NMR (100 MHz) spectrum of the Ramalin derivative RA-PF of the present invention.

도 33은 본 발명의 라말린 유도체 RA-PF의 DEPT 스펙트럼을 나타낸 도면이다.33 is a diagram showing the DEPT spectrum of the Ramalin derivative RA-PF of the present invention.

발명의 상세한 설명 및 구체적인 구현예DETAILED DESCRIPTION OF THE INVENTION AND SPECIFIC EMBODIMENTS

다른 식으로 정의되지 않는 한, 본 명세서에서 사용된 모든 기술적 및 과학적 용어들은 본 발명이 속하는 기술 분야에서 숙련된 전문가에 의해서 통상적으로 이해되는 것과 동일한 의미를 갖는다. 일반적으로, 본 명세서에서 사용된 명명법은 본 기술 분야에서 잘 알려져 있고 통상적으로 사용되는 것이다.Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In general, the nomenclature used herein is one well known and commonly used in the art.

본 발명에서는 남극 지의류 라말리나 테레브라타(Ramalina terebrata)로부터 분리한 분자식 C11H16N3O4의 화합물 라말린(ramalin, γ-glutamyl-N'-(2-hydroxyphenyl)hydrazide)의 페닐 고리의 작용기(페놀성 수산기)를 메틸기 또는 불소 원자로 대체하여 얻은 유도체가 구조 변형을 기반으로 Aβ 축적을 유발하는 BACE-1에 대한 억제 활성을 나타내고, 활성산소를 제거함으로써 알츠하이머병 예방 및 치료에 중요한 역할을 하는 항산화 활성과 알츠하이머병을 악화시키는 염증의 억제 효과를 나타내는 등 독성 없이 알츠하이머병과 관련된 여러 표적에 대해 활성을 나타내므로 알츠하이머병의 치료제로 작용할 수 있다는 것을 규명하였다.In the present invention, the phenyl ring of ramalin (ramalin, γ-glutamyl-N'-(2-hydroxyphenyl)hydrazide), a compound of molecular formula C 11 H 16 N 3 O 4 isolated from the Antarctic lichen Ramalina terebrata A derivative obtained by replacing the functional group (phenolic hydroxyl group) with a methyl group or a fluorine atom shows an inhibitory activity against BACE-1, which causes Aβ accumulation based on structural transformation, and plays an important role in the prevention and treatment of Alzheimer's disease by removing active oxygen It was found that it can act as a therapeutic agent for Alzheimer's disease because it shows activity against various targets related to Alzheimer's disease without toxicity, such as antioxidant activity that causes Alzheimer's disease and inhibition of inflammation that exacerbates Alzheimer's disease.

따라서, 본 발명은 일 관점에서 화학식 1로 표시되는 것을 특징으로 하는 라말린 유도체에 관한 것이다.Accordingly, in one aspect, the present invention relates to a ramalin derivative represented by Chemical Formula 1.

[화학식 1][Formula 1]

Figure PCTKR2022016326-appb-img-000006
Figure PCTKR2022016326-appb-img-000006

화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬, C3-C6 사이클로알킬, 할로겐, 하이드록시, 카복실, 아미노 또는 시아노이고, 단, R1, R2, R3, R4 및 R5는 동시에 수소원자이거나 R1, R2, R3 및 R4는 수소원자이고, R5는 하이드록시인 경우는 제외된다.In Formula 1, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.

본 발명에 있어서, 화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬 또는 할로겐인 것이 바람직하다.In the present invention, it is preferable that each of R 1 , R 2 , R 3 , R 4 and R 5 in Formula 1 is independently a hydrogen atom, C 1 -C 6 alkyl or halogen.

본 발명에 의한 라말린 유도체의 바람직한 실례는 화학식 2 내지 화학식 10에서 선택될 수 있으며, 도 1에 도시하였다.Preferred examples of the ramalin derivative according to the present invention may be selected from Chemical Formulas 2 to 10, and are shown in FIG. 1.

[화학식 2][Formula 2]

Figure PCTKR2022016326-appb-img-000007
Figure PCTKR2022016326-appb-img-000007

[화학식 3][Formula 3]

Figure PCTKR2022016326-appb-img-000008
Figure PCTKR2022016326-appb-img-000008

[화학식 4][Formula 4]

Figure PCTKR2022016326-appb-img-000009
Figure PCTKR2022016326-appb-img-000009

[화학식 5][Formula 5]

Figure PCTKR2022016326-appb-img-000010
Figure PCTKR2022016326-appb-img-000010

[화학식 6][Formula 6]

Figure PCTKR2022016326-appb-img-000011
Figure PCTKR2022016326-appb-img-000011

[화학식 7][Formula 7]

Figure PCTKR2022016326-appb-img-000012
Figure PCTKR2022016326-appb-img-000012

[화학식 8][Formula 8]

Figure PCTKR2022016326-appb-img-000013
Figure PCTKR2022016326-appb-img-000013

[화학식 9][Formula 9]

Figure PCTKR2022016326-appb-img-000014
Figure PCTKR2022016326-appb-img-000014

[화학식 10][Formula 10]

Figure PCTKR2022016326-appb-img-000015
Figure PCTKR2022016326-appb-img-000015

본 발명은 다른 관점에서 (a) 출발 물질인 화학식 11의 1-벤질-N-벤질옥시카보닐-L-글루탐산과 화학식 12의 페닐하이드라진의 커플링 반응을 수행하여 화학식 13의 화합물을 수득하는 단계; 및 (b) 화학식 13의 글루탐산 하이드레이트의 벤질기(Bn) 및 벤질옥시카보닐기(Cbz)의 탈보호화를 수행한 다음 수득한 조생성물을 재결정화하여 화학식 1의 라말린 유도체를 수득하는 단계를 포함하는 상기 라말린 유도체의 제조방법에 관한 것이다.In another aspect of the present invention, (a) performing a coupling reaction between 1-benzyl-N-benzyloxycarbonyl-L-glutamic acid of Formula 11 as a starting material and phenylhydrazine of Formula 12 to obtain a compound of Formula 13 ; and (b) deprotecting the benzyl group (Bn) and benzyloxycarbonyl group (Cbz) of the glutamic acid hydrate of Formula 13 and then recrystallizing the obtained crude product to obtain a Ramalin derivative of Formula 1. It relates to a method for preparing the ramalin derivative.

[화학식 1][Formula 1]

Figure PCTKR2022016326-appb-img-000016
Figure PCTKR2022016326-appb-img-000016

[화학식 11][Formula 11]

Figure PCTKR2022016326-appb-img-000017
Figure PCTKR2022016326-appb-img-000017

[화학식 12][Formula 12]

Figure PCTKR2022016326-appb-img-000018
Figure PCTKR2022016326-appb-img-000018

[화학식 13][Formula 13]

Figure PCTKR2022016326-appb-img-000019
Figure PCTKR2022016326-appb-img-000019

화학식 1, 12 및 13에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬, C3-C6 사이클로알킬, 할로겐, 하이드록시, 카복실, 아미노 또는 시아노이고, 단, R1, R2, R3, R4 및 R5는 동시에 수소원자이거나 R1, R3, R4 및 R5는 수소원자이고, R2는 하이드록시인 경우는 제외된다.In Formulas 1, 12 and 13, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano, provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 3 , R 4 and R 5 are hydrogen atoms and R 2 is hydroxy; is excluded.

본 발명에 있어서, 상기 (a) 단계는 -5℃의 온도에서 1-벤질-N-벤질옥시카보닐-L-글루탐산을 디클로로메탄(dichloromethane, DCM)에 녹인 용액에 트리에틸아민(trimethylamine, TEA)과 에틸클로로포르메이트(ethylchloroformate, ECF)를 차례로 첨가한 다음, 반응 혼합물을 2시간 동안 교반하고, 온도를 유지하면서 페닐하이드라진 HCl 염을 첨가하여 커플링 반응을 수행할 수 있다.In the present invention, the step (a) is performed by adding triethylamine (TEA) to a solution of 1-benzyl-N-benzyloxycarbonyl-L-glutamic acid dissolved in dichloromethane (DCM) at a temperature of -5 ° C. ) and ethylchloroformate (ECF) were sequentially added, the reaction mixture was stirred for 2 hours, and phenylhydrazine HCl salt was added while maintaining the temperature to perform a coupling reaction.

본 발명에 있어서, 상기 (b) 단계의 탈보호화는 Pd/C 및 H2 가스의 분위기에서 수행될 수 있다.In the present invention, the deprotection in step (b) may be performed in an atmosphere of Pd/C and H 2 gas.

본 발명은 또 다른 관점에서 화학식 1로 표시되는 라말린 유도체를 유효성분으로 함유하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물에 관한 것이다.In another aspect, the present invention relates to a pharmaceutical composition for preventing or treating degenerative brain diseases containing a ramalin derivative represented by Formula 1 as an active ingredient.

[화학식 1][Formula 1]

Figure PCTKR2022016326-appb-img-000020
Figure PCTKR2022016326-appb-img-000020

화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬, C3-C6 사이클로알킬, 할로겐, 하이드록시, 카복실, 아미노 또는 시아노이고, 단, R1, R2, R3, R4 및 R5는 동시에 수소원자이거나 R1, R2, R3 및 R4는 수소원자이고, R5는 하이드록시인 경우는 제외된다.In Formula 1, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.

본 발명에 있어서, 화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬 또는 할로겐인 것이 바람직하다.In the present invention, it is preferable that each of R 1 , R 2 , R 3 , R 4 and R 5 in Formula 1 is independently a hydrogen atom, C 1 -C 6 alkyl or halogen.

본 발명에 의한 라말린 유도체의 바람직한 실례는 화학식 2 내지 화학식 10에서 선택될 수 있으며, 상기에 논한 바와 같다.Preferred examples of Ramalin derivatives according to the present invention may be selected from Formulas 2 to 10, as discussed above.

본 발명에 있어서, 상기 퇴행성 뇌질환은 알츠하이머병, 파킨슨병, 루게릭병, 픽크병, 크로이츠펠트-야콥병, 헌팅턴병 및 치매로 구성된 군에서 선택될 수 있으며, 특히 바람직하게는 알츠하이머병일 수 있다.In the present invention, the degenerative brain disease may be selected from the group consisting of Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, Pick's disease, Creutzfeldt-Jakob disease, Huntington's disease and dementia, and particularly preferably Alzheimer's disease.

본 발명에 있어서, BACE1 발현을 억제할 수 있다.In the present invention, BACE1 expression can be inhibited.

본 발명에 있어서, 상기 라말린 유도체를 함유하는 조성물은 통상의 방법에 따른 적절한 약학적으로 허용되는 담체, 부형제 또는 희석제를 추가적으로 포함할 수 있다. 화합물을 포함하는 조성물에 포함될 수 있는 담체, 부형제 및 희석제로는 락토즈, 덱스트로즈, 수크로스, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로즈, 메틸 셀룰로즈, 미정질 셀룰로스, 폴리비닐 피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유를 들 수 있다.In the present invention, the composition containing the ramalin derivative may additionally contain an appropriate pharmaceutically acceptable carrier, excipient or diluent according to a conventional method. Carriers, excipients and diluents that may be included in the composition comprising the compound include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum acacia, alginates, gelatin, calcium phosphate, calcium silicate. , cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.

본 발명의 라말린 유도체를 함유하는 조성물은 통상의 방법에 따라 산제, 환제, 과립제, 캡슐제, 현탁액, 내용액제, 유제, 시럽제, 멸균된 수용액, 비수용액제, 현탁액, 동결 건조제 및 좌제로 이루어진 군으로부터 선택되는 어느 하나의 제형을 가질 수 있다.The composition containing the ramalin derivative of the present invention is a powder, pill, granule, capsule, suspension, internal solution, emulsion, syrup, sterilized aqueous solution, non-aqueous solution, suspension, freeze-dried agent and suppository according to a conventional method. It may have any one formulation selected from.

제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제된다. 경구투여를 위한 고형제제에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 이러한 고형제제는 상기 화합물에 적어도 하나 이상의 부형제, 예를 들면, 전분, 칼슘카보네이트(calcium carbonate), 수크로스(sucrose) 또는 락토오스(lactose), 젤라틴 등을 섞어 조제된다. 또한 단순한 부형제 이외에 마그네슘 스테아레이트, 탈크 같은 윤활제들도 사용된다. 경구를 위한 액상 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데 흔히 사용되는 단순희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성 용제, 현탁제, 유제, 동결건조 제제, 좌제가 포함된다. 비수성용제, 현탁제로는 프로필렌글리콜(propylene glycol), 폴리에틸렌글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔(witepsol), 마크로골, 트윈(tween) 60, 카카오지, 라우린지, 글리세로 젤라틴 등이 사용될 수 있다.When formulated, it is prepared using diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants. Solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and these solid preparations contain at least one excipient such as starch, calcium carbonate, sucrose ( It is prepared by mixing sucrose, lactose, or gelatin. In addition to simple excipients, lubricants such as magnesium stearate and talc are also used. Liquid preparations for oral use include suspensions, solutions for oral 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. . Formulations for parenteral administration include sterilized aqueous solutions, non-aqueous solvents, suspensions, emulsions, freeze-dried formulations, and suppositories. 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 suppositories, witepsol, macrogol, tween 60, cacao butter, laurin paper, glycerogelatin, and the like may be used.

본 발명의 약학 조성물은 목적하는 방법에 따라 경구 투여하거나 비경구 투여(예를 들어 정맥 내, 피하, 복강 내 또는 국소 적용)할 수 있으며, 투여량은 환자의 건강상태, 체중, 연령, 성별, 식이, 배설율, 질병의 정도, 약물형태, 투여시간, 투여경로 및 투여기간에 따라 그 범위가 다양하지만, 당업자에 의해 적절하게 선택될 수 있다. 그러나, 바람직한 효과를 위해서, 본 발명의 화합물은 1일 0.001~1000mg/kg으로, 바람직하게는 0.01~100mg/kg으로 투여하는 것이 좋다. 투여는 하루에 한번 투여할 수도 있고, 수회 나누어 투여할 수도 있다. 상기 투여량은 어떠한 면으로든 본 발명의 범위를 한정하는 것은 아니다.The pharmaceutical composition of the present invention may be administered orally or parenterally (eg, intravenous, subcutaneous, intraperitoneal or topical application) depending on the desired method, and the dosage is determined by the patient's health condition, weight, age, gender, The range varies depending on diet, excretion rate, severity of disease, drug form, administration time, administration route and administration period, but can be appropriately selected by those skilled in the art. However, for desirable effects, the compound of the present invention is preferably administered at 0.001 to 1000 mg/kg per day, preferably at 0.01 to 100 mg/kg. Administration may be administered once a day, or may be administered in several divided doses. The dosage is not intended to limit the scope of the present invention in any way.

본 발명의 약학 조성물은 알츠하이머의 예방 또는 치료를 위하여 단독으로, 또는 수술, 방사선 치료, 호르몬 치료, 화학치료 및 생물학적 반응 조절제를 사용하는 방법들과 병용하여 사용할 수 있다.The pharmaceutical composition of the present invention can be used alone or in combination with methods using surgery, radiation therapy, hormone therapy, chemotherapy, and biological response modifiers for the prevention or treatment of Alzheimer's.

본 발명은 또 다른 관점에서, 화학식 1로 표시되는 라말린 유도체를 유효성분으로 함유하는 퇴행성 뇌질환의 예방 또는 개선용 식품 조성물에 관한 것이다.In another aspect, the present invention relates to a food composition for preventing or alleviating degenerative brain diseases containing a ramalin derivative represented by Chemical Formula 1 as an active ingredient.

[화학식 1][Formula 1]

Figure PCTKR2022016326-appb-img-000021
Figure PCTKR2022016326-appb-img-000021

화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬, C3-C6 사이클로알킬, 할로겐, 하이드록시, 카복실, 아미노 또는 시아노이고, 단, R1, R2, R3, R4 및 R5는 동시에 수소원자이거나 R1, R2, R3 및 R4는 수소원자이고, R5는 하이드록시인 경우는 제외된다.In Formula 1, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.

본 발명에 있어서, 화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬 또는 할로겐인 것이 바람직하다.In the present invention, it is preferable that each of R 1 , R 2 , R 3 , R 4 and R 5 in Formula 1 is independently a hydrogen atom, C 1 -C 6 alkyl or halogen.

본 발명에 의한 라말린 유도체의 바람직한 실례는 화학식 2 내지 화학식 10에서 선택될 수 있으며, 상기에 논한 바와 같다.Preferred examples of Ramalin derivatives according to the present invention may be selected from Formulas 2 to 10, as discussed above.

본 발명에 있어서, 상기 퇴행성 뇌질환은 알츠하이머병, 파킨슨병, 루게릭병, 픽크병, 크로이츠펠트-야콥병, 헌팅턴병 및 치매로 구성된 군에서 선택될 수 있으며, 특히 바람직하게는 알츠하이머병일 수 있다.In the present invention, the degenerative brain disease may be selected from the group consisting of Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, Pick's disease, Creutzfeldt-Jakob disease, Huntington's disease and dementia, and particularly preferably Alzheimer's disease.

본 발명에 있어서, 상기 라말린 유도체를 함유하는 식품 조성물은 식품학적으로 허용되는 첨가제, 부형제 또는 향미제를 추가로 포함할 수 있다. 본 발명의 식품은 기능성 식품(functional food), 영양 보조제(nutritional supplement), 건강식품(health food) 및 식품 첨가제(food additives) 등의 모든 형태를 포함한다. 상기 유형의 건강기능 식품은 당업계에 공지된 통상적인 방법에 따라 다양한 형태로 제조할 수 있다. 예를 들면, 건강식품으로는 본 발명의 라말린 유도체를 차, 쥬스 및 드링크의 형태로 제조하여 음용하도록 하거나, 과립화, 캡슐화 및 분말화하여 섭취할 수 있다. 또한, 기능성 식품으로는 음료(알콜성 음료 포함), 과실 및 그의 가공식품(예: 과일통조림, 병조림, 잼, 마말레이드 등), 어류, 육류 및 그 가공식품(예: 햄, 소시지 콘비프 등), 빵류 및 면류(예: 우동, 메밀국수, 라면, 스파게티, 마카로니 등), 과즙, 각종 드링크, 쿠키, 엿, 유제품(예: 버터, 치즈 등), 식용식물유지, 마가린, 식물성 단백질, 레토르트 식품, 냉동식품, 각종 조미료(예: 된장, 간장, 소스 등) 등에 본 발명의 라말린 유도체를 첨가하여 제조할 수 있다.In the present invention, the food composition containing the ramalin derivative may further include additives, excipients, or flavoring agents acceptable in food science. The food of the present invention includes all forms such as functional food, nutritional supplement, health food and food additives. The health functional food of the above type can be prepared in various forms according to conventional methods known in the art. For example, as a health food, the ramalin derivative of the present invention can be prepared in the form of tea, juice, or drink to be consumed, or granulated, encapsulated, or powdered to be consumed. In addition, functional foods include beverages (including alcoholic beverages), fruits and their processed foods (e.g. canned fruits, canned fruits, jams, marmalades, etc.), fish, meat and their processed foods (e.g. ham, sausage corned beef, etc.), Bread and noodles (e.g. udon, buckwheat noodles, ramen, spaghetti, macaroni, etc.), fruit juice, various drinks, cookies, taffy, dairy products (e.g. butter, cheese, etc.), edible vegetable oil, margarine, vegetable protein, retort food, It can be prepared by adding the ramalin derivative of the present invention to frozen foods, various seasonings (eg, soybean paste, soy sauce, sauce, etc.).

상기 건강 기능식품 또한, 식품조성물로서 기능성 식품, 영양보조제, 건강 식품, 식품 첨가제 등의 다양한 형태를 포함하는 것이며, 당업계에 공지된 통상적인 방법에 따라 다양한 형태, 예컨대, 앞서 언급한 라말린을 차, 쥬스, 드링크의 형태로 제조하거나, 과립화, 캡슐화, 분말화 하거나, 이러한 화합물 또는 추출물을 음료, 과실 및 가공식품, 어류, 육류 및 그 가공식품, 빵류, 면류, 조미료 등 각종 식품에 첨가하여 제조함으로써 제공될 수 있다.The health functional food also includes various forms such as functional foods, nutritional supplements, health foods, food additives, etc. as food compositions, and various forms such as Ramalin mentioned above are prepared according to conventional methods known in the art. Manufactured in the form of tea, juice, or drink, or granulated, encapsulated, or powdered, or added to beverages, fruits and processed foods, fish, meat and its processed foods, breads, noodles, and seasonings with these compounds or extracts It can be provided by manufacturing.

본 발명은 또 다른 관점에서, 화학식 1로 표시되는 라말린 유도체를 유효성분으로 함유하는 조성물을 투여하는 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 치료 방법에 관한 것이다.In another aspect, the present invention relates to a method for preventing or treating a degenerative brain disease comprising administering a composition containing a ramalin derivative represented by Chemical Formula 1 as an active ingredient.

본 발명은 또 다른 관점에서, 화학식 1로 표시되는 라말린 유도체를 유효성분으로 함유하는 조성물을 퇴행성 뇌질환의 예방 또는 치료에 사용하는 용도에 관한 것이다.In another aspect, the present invention relates to the use of a composition containing the ramalin derivative represented by Chemical Formula 1 as an active ingredient for the prevention or treatment of degenerative brain diseases.

본 발명에서, 구체적으로 Aβ 축적을 유발하는 BACE-1에 대한 라말린 유도체의 억제 활성을 평가하고, 또한 활성산소를 제거하여 알츠하이머병 예방 및 치료에 중요한 역할을 하는 항산화 효과와 알츠하이머병을 악화시키는 염증 억제 효과를 조사하였으며, 그 결과 BACE-1에 대한 라말린 유도체의 억제 활성이 농도 의존적으로 감소하는 경향을 보였다. 또한 라말린 유도체는 라말린과 유사한 항산화 활성을 보였으며, LPS로 자극된 RAW 264.7 세포에서도 독성 없이 항염 효과를 보였다. 따라서 라말린 유도체도 라말린과 마찬가지로 알츠하이머병과 관련된 여러 표적에 대해 활성을 나타내므로 알츠하이머병의 치료제로 작용할 수 있다는 것을 확인하였다.In the present invention, specifically, the inhibitory activity of Ramalin derivatives on BACE-1, which causes Aβ accumulation, is evaluated, and the antioxidant effect that plays an important role in the prevention and treatment of Alzheimer's disease by removing active oxygen and the aggravation of Alzheimer's disease The anti-inflammatory effect was investigated, and as a result, the inhibitory activity of the Ramalin derivatives on BACE-1 tended to decrease in a concentration-dependent manner. In addition, ramalin derivatives showed antioxidant activity similar to that of ramalin, and showed anti-inflammatory effects without toxicity in RAW 264.7 cells stimulated with LPS. Accordingly, it was confirmed that ramalin derivatives, like ramalin, show activity against various targets related to Alzheimer's disease, and therefore can act as a therapeutic agent for Alzheimer's disease.

또한, 본 발명의 다른 관점에서 화학식 1로 표시되는 라말린 유도체를 유효성분으로 포함하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물을 객체(subject)에 투여하는 단계를 포함하는 퇴행성 뇌질환의 치료 또는 예방방법에 관한 것이고, 화학식 1로 표시되는 라말린 유도체를 유효성분으로 포함하는 퇴행성 뇌질환의 예방 또는 치료용 조성물의 용도 및 화학식 1로 표시되는 라말린 유도체를 유효성분으로 포함하는 퇴행성 뇌질환의 예방 또는 치료용 약제의 제조에 있어서의 용도에 관한 것이다.In addition, in another aspect of the present invention, treatment or treatment of degenerative brain disease comprising the step of administering to a subject a pharmaceutical composition for preventing or treating degenerative brain disease comprising a ramalin derivative represented by Chemical Formula 1 as an active ingredient; It relates to a preventive method, uses of a composition for the prevention or treatment of degenerative brain diseases comprising a ramalin derivative represented by Formula 1 as an active ingredient, and a composition for preventing or treating degenerative brain diseases comprising a ramalin derivative represented by Formula 1 as an active ingredient It relates to use in the manufacture of a prophylactic or therapeutic medicament.

이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 예시하기 위한 것으로, 본 발명의 범위가 이들 실시예에 의해 제한되지 않는 것은 당업계에서 통상의 지식을 가진 자에게 있어서 자명할 것이다.Hereinafter, the present invention will be described in more detail through examples. These examples are only for exemplifying the present invention, and it will be apparent to those skilled in the art that the scope of the present invention is not limited by these examples.

[실시예][Example]

모든 용매 및 시약은 상용 공급업체인 Merck(Darmstadt, Germany) 또는 TCI(Tokyo, Japan)에서 얻었고 추가 정제 없이 사용하였다. 모든 유리는 건조 오븐(60℃) 또는 화염에서 완전히 건조되었고 사용 직전에 건조 아르곤 증기 하에서 냉각되었다. 필터는 상업용 공급업체인 GE Healthcare(GF/F, 0.7 μm, Whatman, UK)에서 구입하였다. 모든 반응은 아르곤의 불활성 분위기에서 수행되었다. 용매와 액체 시약은 주사기를 사용하여 옮겼다. 유기 추출물을 건조제인 Na2SO4로 건조시키고 회전 증발기(Eyela, Tokyo, Japan)를 사용하여 감압하에 농축시켰다. 잔류 용매를 고진공 하에서 제거하였다(Vacuubrand RZ 2.5, Wertheim, Germany, 1 x 10-2 mbar). 직접 주입 모드의 인터페이스가 있는 AB Sciex Triple TOF 4600(Framingham, MA, USA) 기기를 사용하여 정확한 질량 스펙트럼을 얻었다. NMR 데이터는 D2O(0.01 mg/mL DSS 포함)-아세톤-d6(6:1 v/v) 또는 DMSO-d6의 혼합물을 사용하여 Jeol JNM ECP-400 분광계(Jeol Ltd., Tokyo, Japan)에서 수집되었다. 용매로서 내부 참조 또는 잔류 용매 신호는 [D2O(DSS 포함)-아세톤- d 6: dH 0.00/dC 29.8, DMSO-d6: dH 2.50/dC 39.5]를 참조하는 데 사용되었다. 피크 분할 패턴은 각각 m(다중선), s(단일선), d(이중선) 및 t(삼중선)로 약칭되었다. 마이크로플레이트 리더(Thermo Scientific Inc., San Diego, CA, USA) 및 다중모드 플레이트 리더(MultistkanTM GO, Thermo Scientific, Waltham, MA, USA)를 흡광도 분석에 사용하였다.All solvents and reagents were obtained from commercial suppliers Merck (Darmstadt, Germany) or TCI (Tokyo, Japan) and used without further purification. All glasses were completely dried in a drying oven (60° C.) or flame and cooled under dry argon vapor immediately before use. Filters were purchased from a commercial supplier, GE Healthcare (GF/F, 0.7 μm, Whatman, UK). All reactions were performed in an inert atmosphere of argon. Solvents and liquid reagents were transferred using a syringe. The organic extract was dried with Na 2 SO 4 as a drying agent and concentrated under reduced pressure using a rotary evaporator (Eyela, Tokyo, Japan). Residual solvent was removed under high vacuum (Vacuubrand RZ 2.5, Wertheim, Germany, 1 x 10 -2 mbar). Accurate mass spectra were obtained using an AB Sciex Triple TOF 4600 (Framingham, MA, USA) instrument with interface in direct injection mode. NMR data were obtained using a mixture of D 2 O (with 0.01 mg/mL DSS)-acetone- d6 (6:1 v/v) or DMSO-d6 on a Jeol JNM ECP-400 spectrometer (Jeol Ltd., Tokyo, Japan). ) were collected from An internal reference or residual solvent signal as solvent was used to reference [D 2 O (with DSS) - acetone - d 6 : d H 0.00 / d C 29.8, DMSO - d 6 : d H 2.50 / d C 39.5] . Peak splitting patterns were abbreviated as m (multiplet), s (singlet), d (doublet) and t (triplet), respectively. A microplate reader (Thermo Scientific Inc., San Diego, CA, USA) and a multimode plate reader (Multistkan TM GO, Thermo Scientific, Waltham, MA, USA) were used for absorbance analysis.

실시예 1: 라말린 및 그 유도체의 합성 및 확인Example 1: Synthesis and identification of Ramalin and its derivatives

p-Glu-Hyd 유사체의 합성Synthesis of p-Glu-Hyd analogues

자기 교반 막대가 장착된 250mL 둥근 바닥 플라스크에 DCM(50mL) 용매 중 1-벤질-N-Cbz-L-글루탐산(2.0g, 5.39mmol)을 채웠다. 반응 혼합물을 -5℃로 냉각시키고 TEA(1.2 eq, 6.47 mmol, 902 μL)를 천천히 첨가하였다. 10분 후, ECF(1.2 eq, 6.47 mmol, 615 μL)를 혼합물에 1시간에 걸쳐 적가하였다. 반응 혼합물을 -5℃에서 4시간 동안 교반하였다. 다른 100mL 배 플라스크에 페닐 히드라진 HCl 염(1.2eq, 6.47mmol) 및 TEA(1.2eq, 6.47mmol, 902μL)를 채운 다음 -5℃에서 1시간 동안 주 반응 플라스크에 천천히 첨가하였다. 히드라진 첨가가 완료되면, 반응 혼합물을 실온으로 가온하고 16시간 동안 교반하였다. 반응 종료 후 유기층을 증류수, 1N HCl, 0.5N NaHCO3, 증류수 순으로 세척하여 층을 분리하고 유기층을 회수하였다. 유기상을 Na2SO4로 건조시키고 회전 증발기에서 농축시켰다. 에틸 아세테이트/n-헥산(1:5)으로부터 재결정화를 수행하여 정제하였다.A 250 mL round bottom flask equipped with a magnetic stir bar was charged with 1-benzyl-N-Cbz-L-glutamic acid (2.0 g, 5.39 mmol) in DCM (50 mL) solvent. The reaction mixture was cooled to -5 °C and TEA (1.2 eq, 6.47 mmol, 902 μL) was added slowly. After 10 min, ECF (1.2 eq, 6.47 mmol, 615 μL) was added dropwise to the mixture over 1 h. The reaction mixture was stirred at -5 °C for 4 hours. Another 100mL pear flask was charged with phenyl hydrazine HCl salt (1.2eq, 6.47mmol) and TEA (1.2eq, 6.47mmol, 902μL) and then slowly added to the main reaction flask at -5°C for 1 hour. When the hydrazine addition was complete, the reaction mixture was warmed to room temperature and stirred for 16 hours. After completion of the reaction, the organic layer was washed with distilled water, 1N HCl, 0.5N NaHCO 3 , and distilled water in order to separate the layers and recover the organic layer. The organic phase was dried over Na 2 SO 4 and concentrated on a rotary evaporator. Purification was performed by recrystallization from ethyl acetate/n-hexane (1:5).

라말린 유도체의 합성 및 확인Synthesis and identification of Ramalin derivatives

자기 교반 막대가 장착된 500mL 둥근 바닥 플라스크에 적절한 p-Glu-Hyd 유사체(4.5mmol), MeOH 200mL 중 탄소 상의 팔라듐(10중량%)을 채웠다. 혼합물을 수소 분위기(1 atm, 수소 풍선) 하에 16시간 동안 교반하였다. 완료되면, 반응 혼합물을 유리 극세사 여과지(0.7㎛)를 통해 여과하였다. 여액을 회전 증발기에서 농축시켰다. MeOH/에틸 아세테이트(1:5)로부터 재결정화에 의한 정제를 수행하였다. 화학식 2 내지 화학식 10의 라말린 유도체의 1H NMR 데이터는 다음과 같으며, 각 화합물의 1H NMR (400 NHz), 13C NMR (100 MHz), DEPT, HSQC 스펙트럼은 도 2 내지 도 33에 도시하였다.A 500 mL round bottom flask equipped with a magnetic stir bar was charged with the appropriate p-Glu-Hyd analogue (4.5 mmol), palladium on carbon (10% by weight) in 200 mL of MeOH. The mixture was stirred for 16 hours under a hydrogen atmosphere (1 atm, hydrogen balloon). Upon completion, the reaction mixture was filtered through glass microfiber filter paper (0.7 μm). The filtrate was concentrated on a rotary evaporator. Purification was performed by recrystallization from MeOH/ethyl acetate (1:5). 1 H NMR data of the Ramalin derivatives of Chemical Formulas 2 to 10 are as follows, and 1 H NMR (400 NHz), 13 C NMR (100 MHz), DEPT, and HSQC spectra of each compound are shown in FIGS. 2 to 33 shown

N 5 -(o-tolylamino)-L-glutamine (RA-2Me). From Benzyl-N-Cbz-L-glutamic acid; 0.94 g, 70%, white solid; 1H NMR (400 MHz, D2O/Acetone-d 6 (6/1)): δ 7.17 (m, 2H, PhH), 6.90 (td, J = 7.6, 1.2, 1H, PhH), 6.84 (dd, J = 8.0, 0.8, 1H, PhH), 3.81 (t, J = 6.0, 1H, H-2), 2.56 (m, 2H, H-4), 2.22 (s, 3H, CH3), 2.21 (m, 2H, H-3); 13C NMR (100 MHz D2O/Acetone-d 6 (6/1)): δ 174.8, 174.0, 145.2, 131.0, 127.4, 123.9, 121.3, 112.2, 54.5, 29.9, 26.5, 16.5; HRESIMS m/z 252.1352 [M + H]+ (calcd for C12H18N3O3, 252.1348). N 5 -(o-tolylamino)-L-glutamine (RA-2Me). From Benzyl-N-Cbz-L-glutamic acid; 0.94 g, 70%, white solid; 1 H NMR (400 MHz, D 2 O/Acetone- d 6 (6/1)): δ 7.17 (m, 2H, PhH), 6.90 (td, J = 7.6, 1.2, 1H, PhH), 6.84 (dd , J = 8.0, 0.8, 1H, PhH), 3.81 (t, J = 6.0, 1H, H-2), 2.56 (m, 2H, H-4), 2.22 (s, 3H, CH 3 ), 2.21 ( m, 2H, H-3); 13 C NMR (100 MHz D 2 O/Acetone- d 6 (6/1)): δ 174.8, 174.0, 145.2, 131.0, 127.4, 123.9, 121.3, 112.2, 54.5, 29.9, 26.5, 16.5; HRESIMS m/z 252.1352 [M + H] + (calcd for C 12 H 18 N 3 O 3 , 252.1348).

N 5 -(m-tolylamino)-L-glutamine (RA-3Me). From Benzyl-N-Cbz-L-glutamic acid; 0.97 g, 72%, white solid, 1H NMR (400 MHz, D2O/Acetone-d 6 (6/1)): δ 7.19 (t, J = 7.6, 1H, PhH), 6.79 (d, J = 7.6, 1H, PhH), 6.72 (s, 1H, PhH), 6.69 (d, J = 8.0, 1H, PhH), 3.80 (t, J = 6.4, 1H, H-2), 2.54 (m, 2H, H-4), 2.27 (s, 3H, CH3), 2.20 (m, 2H, H-3); 13C NMR (100 MHz D2O/Acetone-d 6 (6/1)): δ 175.0, 174.1, 147.7, 140.3, 129.8, 122.3, 114.2, 110.9, 54.5, 29.9, 26.5, 20.9; HRESIMS m/z 252.1367 [M + H]+ (calcd for C12H18N3O3, 252.1348). N 5 -(m-tolylamino)-L-glutamine (RA-3Me). From Benzyl-N-Cbz-L-glutamic acid; 0.97 g, 72%, white solid, 1 H NMR (400 MHz, D 2 O/Acetone- d 6 (6/1)): δ 7.19 (t, J = 7.6, 1H, PhH), 6.79 (d, J = 7.6, 1H, PhH), 6.72 (s, 1H, PhH), 6.69 (d, J = 8.0, 1H, PhH), 3.80 (t, J = 6.4, 1H, H-2), 2.54 (m, 2H , H-4), 2.27 (s, 3H, CH 3 ), 2.20 (m, 2H, H-3); 13 C NMR (100 MHz D 2 O/Acetone- d 6 (6/1)): δ 175.0, 174.1, 147.7, 140.3, 129.8, 122.3, 114.2, 110.9, 54.5, 29.9, 26.5, 20.9; HRESIMS m/z 252.1367 [M + H] + (calcd for C 12 H 18 N 3 O 3 , 252.1348).

N 5 -(p-tolylamino)-L-glutamine (RA-4Me). From Benzyl-N-Cbz-L-glutamic acid; 0.90 g, 67%, white solid, 1H NMR (400 MHz, D2O/Acetone-d 6 (6/1)): δ 7.15 (d, J = 8.4, 2H, PhH), 6.82 (d, J = 8.0, 2H, PhH), 3.79 (t, J = 6.4, 1H, H-2), 2.52 (m, 2H, H-4), 2.25 (s, 3H, CH3), 2.18 (m, 2H, H-3); 13C NMR (100 MHz D2O/Acetone-d 6 (6/1)): δ 174.9, 174.1, 145.1, 131.5, 130.2, 114.1, 54.5, 29.9, 26.5, 19.9; HRESIMS m/z 252.1367 [M + H]+ (calcd for C12H18N3O3, 252.1348). N 5 -(p-tolylamino)-L-glutamine (RA-4Me). From Benzyl-N-Cbz-L-glutamic acid; 0.90 g, 67%, white solid, 1 H NMR (400 MHz, D 2 O/Acetone- d 6 (6/1)): δ 7.15 (d, J = 8.4, 2H, PhH), 6.82 (d, J = 8.0, 2H, PhH), 3.79 (t, J = 6.4, 1H, H-2), 2.52 (m, 2H, H-4), 2.25 (s, 3H, CH 3 ), 2.18 (m, 2H, H-3); 13 C NMR (100 MHz D 2 O/Acetone- d 6 (6/1)): δ 174.9, 174.1, 145.1, 131.5, 130.2, 114.1, 54.5, 29.9, 26.5, 19.9; HRESIMS m/z 252.1367 [M + H] + (calcd for C 12 H 18 N 3 O 3 , 252.1348).

N 5 -((2,5-dimethylphenyl)amino)-L-glutamine (RA-25Me). From Benzyl-N-Cbz-L-glutamic acid; 1.12 g, 78%, white solid, 1H NMR (400 MHz, D2O/Acetone-d 6 (6/1)): δ 6.89 (d, J = 7.6, 1H, PhH), 6.57 (d, J = 7.6, 1H, PhH), 6.52 (s, 1H, PhH), 3.67 (t, J = 6.4, 1H, H-2), 2.43 (m, 2H, H-4), 2.10 (s, 3H, CH3), 2.06 (m, 2H, H-3), 2.03 (s, 3H, CH3); 13C NMR (100 MHz D2O/Acetone-d 6 (6/1)): δ 174.4, 173.7, 144.9, 137.1, 130.7, 121.5, 120.4, 112.5, 54.2, 29.6, 26.3, 20.4, 15.8; HRESIMS m/z 266.1504 [M + H]+ (calcd for C13H20N3O3, 266.1505). N 5 -((2,5-dimethylphenyl)amino)-L-glutamine (RA-25Me). From Benzyl-N-Cbz-L-glutamic acid; 1.12 g, 78%, white solid, 1 H NMR (400 MHz, D 2 O/Acetone- d 6 (6/1)): δ 6.89 (d, J = 7.6, 1H, PhH), 6.57 (d, J = 7.6, 1H, PhH), 6.52 (s, 1H, PhH), 3.67 (t, J = 6.4, 1H, H-2), 2.43 (m, 2H, H-4), 2.10 (s, 3H, CH 3 ), 2.06 (m, 2H, H-3), 2.03 (s, 3H, CH 3 ); 13 C NMR (100 MHz D 2 O/Acetone- d 6 (6/1)): δ 174.4, 173.7, 144.9, 137.1, 130.7, 121.5, 120.4, 112.5, 54.2, 29.6, 26.3, 20.4, 15.8; HRESIMS m/z 266.1504 [M + H] + (calcd for C 13 H 20 N 3 O 3 , 266.1505).

N 5 -((3,4-dimethylphenyl)amino)-L-glutamine (RA-34Me). From Benzyl-N-Cbz-L-glutamic acid; 1.01 g, 70%, white solid, 1H NMR (400 MHz, D2O/Acetone-d 6 (6/1)): δ 7.06 (d, J = 8.0, 1H, PhH), 6.71 (d, J = 2.4, 1H, PhH), 6.64 (dd, J = 8.0, 2.4, 1H, PhH), 3.80 (t, J = 6.4, 1H, H-2), 2.53 (m, 2H, H-4), 2.20 (m, 2H, H-3), 2.19 (s, 3H, CH3). 2.15 (s, 3H, CH3); 13C NMR (100 MHz D2O/Acetone-d 6 (6/1)): δ 174.7, 173.9, 145.7, 138.3, 130.6, 129.9, 115.3, 111.4, 54.5, 29.9, 26.6, 19.4, 18.3; HRESIMS m/z 266.1508 [M + H]+ (calcd for C13H20N3O3, 266.1505). N 5 -((3,4-dimethylphenyl)amino)-L-glutamine (RA-34Me). From Benzyl-N-Cbz-L-glutamic acid; 1.01 g, 70%, white solid, 1 H NMR (400 MHz, D 2 O/Acetone- d 6 (6/1)): δ 7.06 (d, J = 8.0, 1H, PhH), 6.71 (d, J = 2.4, 1H, PhH), 6.64 (dd, J = 8.0, 2.4, 1H, PhH), 3.80 (t, J = 6.4, 1H, H-2), 2.53 (m, 2H, H-4), 2.20 (m, 2H, H-3), 2.19 (s, 3H, CH 3 ). 2.15 (s, 3H, CH 3 ); 13 C NMR (100 MHz D 2 O/Acetone- d 6 (6/1)): δ 174.7, 173.9, 145.7, 138.3, 130.6, 129.9, 115.3, 111.4, 54.5, 29.9, 26.6, 19.4, 18.3; HRESIMS m/z 266.1508 [M + H] + (calcd for C 13 H 20 N 3 O 3 , 266.1505).

N 5 -((2-fluorophenyl)amino)-L-glutamine (RA-2F). From Benzyl-N-Cbz-L-glutamic acid; 0.85 g, 62%, white solid, 1H NMR (400 MHz, D2O/Acetone-d 6 (6/1)): δ 7.15-7.08 (m, 2H, PhH), 6.97-6.90 (m, 2H, PhH), 3.80 (t, J = 6.0, 1H, H-2), 2.55 (m, 2H, H-4), 2.20 (m, 2H, H-3); 13C NMR (100 MHz D2O/Acetone-d 6 (6/1)): δ 175.0, 174.0, 151.5, 135.3, 125.2, 121.7, 115.7, 114.9, 54.5, 29.9, 26.5; HRESIMS m/z 256.1059 [M + H]+ (calcd for C11H15FN3O3, 256.1097). N 5 -((2-fluorophenyl)amino)-L-glutamine (RA-2F). From Benzyl-N-Cbz-L-glutamic acid; 0.85 g, 62%, white solid, 1 H NMR (400 MHz, D 2 O/Acetone- d 6 (6/1)): δ 7.15-7.08 (m, 2H, PhH), 6.97-6.90 (m, 2H , PhH), 3.80 (t, J = 6.0, 1H, H-2), 2.55 (m, 2H, H-4), 2.20 (m, 2H, H-3); 13 C NMR (100 MHz D 2 O/Acetone- d 6 (6/1)): δ 175.0, 174.0, 151.5, 135.3, 125.2, 121.7, 115.7, 114.9, 54.5, 29.9, 26.5; HRESIMS m/z 256.1059 [M + H] + (calcd for C 11 H 15 FN 3 O 3 , 256.1097).

N 5 -((4-fluorophenyl)amino)-L-glutamine (RA-4F). From Benzyl-N-Cbz-L-glutamic acid; 0.90 g, 65%, white solid, 1H NMR (400 MHz, D2O/Acetone-d 6 (6/1)): δ 7.09-7.04 (m, 2H, PhH), 6.92-6.88 (m, 2H, PhH), 3.82 (td, J = 6.0, 1.2, 1H, H-2), 2.54 (m, 2H, H-4), 2.21 (m, 2H, H-3); 13C NMR (100 MHz D2O/Acetone-d 6 (6/1)): δ 175.0, 174.1, 158.0 143.7, 116.1, 115.2, 54.4, 29.8, 26.4; HRESIMS m/z 256.1106 [M + H]+ (calcd for C11H15FN3O3, 256.1097). N 5 -((4-fluorophenyl)amino)-L-glutamine (RA-4F). From Benzyl-N-Cbz-L-glutamic acid; 0.90 g, 65%, white solid, 1 H NMR (400 MHz, D 2 O/Acetone- d 6 (6/1)): δ 7.09-7.04 (m, 2H, PhH), 6.92-6.88 (m, 2H , PhH), 3.82 (td, J = 6.0, 1.2, 1H, H-2), 2.54 (m, 2H, H-4), 2.21 (m, 2H, H-3); 13 C NMR (100 MHz D 2 O/Acetone- d 6 (6/1)): δ 175.0, 174.1, 158.0 143.7, 116.1, 115.2, 54.4, 29.8, 26.4; HRESIMS m/z 256.1106 [M + H] + (calcd for C 11 H 15 FN 3 O 3 , 256.1097).

N 5 -((2,4-difluorophenyl)amino)-L-glutamine (RA-24F). From Benzyl-N-Cbz-L-glutamic acid; 1.10 g, 75%, white solid, 1H NMR (400 MHz, D2O/Acetone-d 6 (6/1)): δ 6.99 (m, 1H, PhH), 6.94 (m, 1H, PhH), 6.89 (m, 1H, PhH), 3.79 (t, J = 6.4, 1H, H-2), 2.52 (m, 2H, H-4), 2.18 (m, 2H, H-3); 13C NMR (100 MHz D2O/Acetone-d 6 (6/1)): δ 175.1, 174.1, 157.1, 151.2, 131.8, 115.8, 111.4, 104.3, 54.5, 29.8, 26.4; HRESIMS m/z 274.0963 [M + H]+ (calcd for C11H14F2N3O3, 274.1003). N 5 -((2,4-difluorophenyl)amino)-L-glutamine (RA-24F). From Benzyl-N-Cbz-L-glutamic acid; 1.10 g, 75%, white solid, 1 H NMR (400 MHz, D 2 O/Acetone- d 6 (6/1)): δ 6.99 (m, 1H, PhH), 6.94 (m, 1H, PhH), 6.89 (m, 1H, PhH), 3.79 (t, J = 6.4, 1H, H-2), 2.52 (m, 2H, H-4), 2.18 (m, 2H, H-3); 13 C NMR (100 MHz D 2 O/Acetone- d 6 (6/1)): δ 175.1, 174.1, 157.1, 151.2, 131.8, 115.8, 111.4, 104.3, 54.5, 29.8, 26.4; HRESIMS m/z 274.0963 [M + H] + (calcd for C 11 H 14 F 2 N 3 O 3 , 274.1003).

N 5 -((perfluorophenyl)amino)-L-glutamine (RA-PF). From Benzyl-N-Cbz-L-glutamic acid; 1.39 g, 79%, white solid, 1H NMR (400 MHz, DMSO-d 6): δ 3.20 (t, J = 6.4, 1H, H-2), 2.27 (m, 2H, H-4), 1.85 (m, 2H, H-3); 13C NMR (100 MHz DMSO-d 6): δ 172.0, 169.5, 137.3 (m, 4C), 133.5 (m, 1C), 124.8 (m, 1C), 53.5, 29.6, 26.9; HRESIMS m/z 328.0725 [M + H]+ (calcd for C11H14F2N3O3, 328.0721). N 5 -((perfluorophenyl)amino)-L-glutamine (RA-PF). From Benzyl-N-Cbz-L-glutamic acid; 1.39 g, 79%, white solid, 1 H NMR (400 MHz, DMSO- d 6 ): δ 3.20 (t, J = 6.4, 1H, H-2), 2.27 (m, 2H, H-4), 1.85 (m, 2H, H-3); 13 C NMR (100 MHz DMSO- d 6 ): δ 172.0, 169.5, 137.3 (m, 4C), 133.5 (m, 1C), 124.8 (m, 1C), 53.5, 29.6, 26.9; HRESIMS m/z 328.0725 [M + H] + (calcd for C 11 H 14 F 2 N 3 O 3 , 328.0721).

Blois, M.S. et al. 방법에 따라 라말린 및 그 유도체의 DPPH 라디칼 소거 활성을 평가하였다(Blois, M. S., Nature, 1958, 181, 1199-1200). 간단히 말해서, MeOH에 10, 5, 2.5, 1 μM 농도의 라말린, 유도체 및 BHA 150 μL를 MeOH에 용해된 0.1 mM DPPH 50 μL와 혼합한 다음 실온에서 30분 동안 암실에 두었다. 그 다음, 혼합물을 540 nm에서 측정하였다.Blois, MS et al. The DPPH radical scavenging activity of Ramalin and its derivatives was evaluated according to the method (Blois, MS, Nature , 1958, 181 , 1199-1200). Briefly, 150 μL of Ramalin, derivatives and BHA at concentrations of 10, 5, 2.5, and 1 μM in MeOH were mixed with 50 μL of 0.1 mM DPPH dissolved in MeOH and then left in the dark at room temperature for 30 min. The mixture was then measured at 540 nm.

라말린은 이전 연구에서 개발된 방법에 따라 성공적으로 합성되었다(Yim, J. H. et al., WO201308959A1, March 26, 2013). 합성은 출발 물질인 1-벤질-N-Cbz-L-글루탐산에 페닐히드라진을 커플링하여 수행하였다. -5℃로 온도를 낮춘 후, 출발물질을 디클로로메탄(DCM)에 녹인 용액에 트리에틸아민(TEA)과 에틸클로로포르메이트(ECF)를 차례로 첨가하였다. 그 후, 반응 혼합물을 약 2시간 동안 교반하고, 온도를 유지하면서 히드라진 HCl 염을 첨가하여 커플링 반응을 완료시켰다. 이어서 Pd/C 및 H2 가스를 사용하여 벤질 및 Cbz기의 탈보호를 수행한 후, 조 생성물을 재결정화하여 라말린을 수득하였다. 라말린 유도체를 얻기 위해 유사한 반응 조건을 사용하였다(반응식 1).Ramalin was successfully synthesized according to a method developed in a previous study (Yim, JH et al., WO201308959A1, March 26, 2013). The synthesis was performed by coupling phenylhydrazine to the starting material, 1-benzyl-N-Cbz-L-glutamic acid. After lowering the temperature to -5°C, triethylamine (TEA) and ethyl chloroformate (ECF) were sequentially added to a solution in which the starting material was dissolved in dichloromethane (DCM). Then, the reaction mixture was stirred for about 2 hours, and the coupling reaction was completed by adding hydrazine HCl salt while maintaining the temperature. Then, after deprotection of benzyl and Cbz groups was performed using Pd/C and H 2 gas, the crude product was recrystallized to obtain ramalin. Similar reaction conditions were used to obtain ramalin derivatives (Scheme 1).

[반응식 1][Scheme 1]

Figure PCTKR2022016326-appb-img-000022
Figure PCTKR2022016326-appb-img-000022

보고된 바에 따르면 라말린은 건조한 고체 형태일 때 실온(즉, 25℃)에서 안정적이다. 그러나 수용액에서는 불안정하다. 이것은 단점으로 간주된다(Pagire, S. H. et al., Bioorg. Med. Chem. Lett., 2018, 28, 529-532). 따라서 안정성과 BBB 침투력을 향상시키기 위해 위치 2의 페놀성 수산기를 메틸기 또는 불소 원자로 교체하고 위치를 변경하여 구조를 수정하였다. 따라서 여러 유도체를 얻었다. 구체적으로, RA-2Me, RA-3Me, RA-4Me는 전자공여성기인 메틸기, 페닐고리에 2개의 메틸기가 있는 RA-25Me, RA-34Me의 위치를 바꾸어 합성하였다. 여분의 메틸 그룹을 도입하여 합성되었다. 메틸기와 반대로 전자를 끄는 불소 원자의 효과를 조사하기 위해 RA-2F, RA-4F, RA-24F 및 RA-펜타플루오라이드(PF)도 성공적으로 합성되었다. 또한 라말린과 그 유도체가 저분자량 항산화제라는 점을 감안하면 BBB에 침투할 가능성이 높다(Gilgun-Sherki, Y. et al., Neuropharmacology, 2001, 40, 959-975). 알츠하이머병 관련 치료제는 BBB를 관통하여 목표에 도달해야 한다. 따라서 BBB 투과능을 추정하기 위하여 라말린 및 그 유도체의 친유성, 수소결합수, 극성표면적(PSA), 분자량(MW) 등을 조사하였다(표 1)(Rankovic, Z., J. Med. Chem., 2015, 58, 2584-2608). 1983년부터 2002년 사이에 도입된 중추신경계에 작용하는 약물의 평균 MW는 약 310 Da이며, 이러한 약물의 요구사항으로 MW < 450 Da가 제안되었다(Rankovic, Z., J. Med. Chem., 2015, 58, 2584-2608). 라말린 및 그 유도체는 수용성 저분자 화합물이고 친유성과 친수성이 균형을 이룬다는 점을 감안할 때 친유성을 나타내는 AlogP에 미치는 영향은 크지 않았다. 또한 PSA 값(90 Å2 < PSA < 140 Å2)을 고려할 때 BBB 침투 측면에서 유리한 것으로 간주된다(Rankovic, Z., J. Med. Chem., 2015, 58, 2584-2608; Waring, M. J., Bioorg. Med. Chem. Lett., 2009, 19, 2844-2851).Reportedly, Ramalin is stable at room temperature (i.e., 25° C.) when in dry solid form. However, it is unstable in aqueous solution. This is considered a disadvantage (Pagire, SH et al., Bioorg. Med. Chem. Lett., 2018, 28, 529-532). Therefore, to improve stability and BBB penetration, the structure was modified by replacing the phenolic hydroxyl group at position 2 with a methyl group or a fluorine atom and changing the position. Thus, several derivatives were obtained. Specifically, RA-2Me, RA-3Me, and RA-4Me were synthesized by exchanging the position of RA-25Me and RA-34Me, which have two methyl groups in the electron-donating methyl group and the phenyl ring. It was synthesized by introducing an extra methyl group. RA-2F, RA-4F, RA-24F and RA-pentafluoride (PF) were also successfully synthesized to investigate the effect of electron-withdrawing fluorine atoms as opposed to methyl groups. In addition, given that ramalin and its derivatives are low molecular weight antioxidants, they have a high possibility of penetrating the BBB (Gilgun-Sherki, Y. et al., Neuropharmacology, 2001, 40, 959-975). Alzheimer's-related therapies must penetrate the BBB to reach their targets. Therefore, in order to estimate the BBB permeability, the lipophilicity, number of hydrogen bonds, polar surface area (PSA), and molecular weight (MW) of Ramalin and its derivatives were investigated (Table 1) (Rankovic, Z., J. Med. Chem ., 2015, 58, 2584-2608). The average MW of drugs acting on the central nervous system introduced between 1983 and 2002 is about 310 Da, and a MW < 450 Da has been proposed as a requirement for these drugs (Rankovic, Z., J. Med. Chem., 2015, 58, 2584-2608). Considering that Ramalin and its derivatives are water-soluble low-molecular-weight compounds and have a balance between lipophilicity and hydrophilicity, their effect on AlogP exhibiting lipophilicity was not significant. It is also considered favorable in terms of BBB penetration when considering PSA values (90 Å 2 < PSA < 140 Å 2 ) (Rankovic, Z., J. Med. Chem., 2015, 58, 2584-2608; Waring, MJ, Bioorg. Med. Chem. Lett., 2009, 19, 2844-2851).

Figure PCTKR2022016326-appb-img-000023
Figure PCTKR2022016326-appb-img-000023

실시예 2: 라말린 및 그 유도체의 라말린의 항산화 효과Example 2: Antioxidant effect of ramalin and its derivatives

라말린의 시험관내 항산화 효과(유리기 소거능, Fe3+ 환원력, 과산화물 음이온 소거능, 티로시나제 억제 효과)는 이전 연구에서 확인되었다((Paudel, B. et al., Phytomedicine, 2011, 18, 1285-1290). DPPH 에세이(2,2-diphenyl-1-picryl-hydrazyl-hydrate assay)를 통해 새로 합성된 라말린 유도체의 항산화 효과를 확인하였다(표 1). 얻은 결과에 따르면, 라말린 유도체는 시중에서 판매되는 부틸화 하이드록시아니솔(BHA)보다 항산화 효율이 더 높은 것으로 나타났다. 시험한 유도체 중 RA-4Me의 항산화 효과는 라말린과 가장 유사한 것으로 나타났다. 또한, RA-PF의 항산화 효과는 상대적으로 높은 IC50 값을 나타내었다. 그러나 페닐 양성자가 없을 때 항산화 활성이 감소하는지 여부를 확인하려면 추가 실험이 필요하다.The in vitro antioxidant effects of Ramalin (free radical scavenging ability, Fe 3+ reducing power, superoxide anion scavenging ability, tyrosinase inhibitory effect) were confirmed in previous studies (Paudel, B. et al., Phytomedicine, 2011, 18, 1285-1290). The antioxidant effect of the newly synthesized ramalin derivatives was confirmed through a DPPH assay (2,2-diphenyl-1-picryl-hydrazyl-hydrate assay) (Table 1). Among the tested derivatives, the antioxidant effect of RA-4Me was the most similar to that of Ramalin. In addition, the antioxidant effect of RA-PF was relatively high. IC50 values are shown, but further experiments are needed to determine whether antioxidant activity is reduced in the absence of phenyl protons.

실시예 3: 라말린 및 그 유도체의 항염 활성 분석Example 3: Analysis of anti-inflammatory activity of Ramalin and its derivatives

세포 배양cell culture

대식세포 유사 쥐 세포주 RAW 264.7(KCLB 번호 40071; 한국 세포주 은행, 서울, 한국)을 10% 열-불활성화된 태아 소 혈청(FBS, Invitrogen, Burlington, ON, Canada) 및 5% CO2 및 37℃에서 1%(w/v) 항생제-항진균 용액(Invitrogen, Grand Island, NY, USA)이 보충된 둘베코 변형된 이글 배지(DMEM, Sigma-Aldrich, St. Louis, MO, USA)에서 배양하였다.Macrophage-like mouse cell line RAW 264.7 (KCLB No. 40071; Korean Cell Line Bank, Seoul, Korea) was cultured in 10% heat-inactivated fetal bovine serum (FBS, Invitrogen, Burlington, ON, Canada) and 5% CO 2 at 37°C. were cultured in Dulbecco's modified Eagle's medium (DMEM, Sigma-Aldrich, St. Louis, MO, USA) supplemented with 1% (w/v) antibiotic-antifungal solution (Invitrogen, Grand Island, NY, USA).

세포독성 분석Cytotoxicity assay

세포 독성은 MTT(Amresco, Solon, OH, USA) 비색 분석에 의해 결정되었다. RAW 264.7 세포를 96-웰 플레이트에 2 x 105 cells/mL의 밀도로 시딩하고 다양한 농도의 라말린 및 그 유도체의 존재하에 24시간 동안 인큐베이션하였다. 시험 물질과 함께 인큐베이션한 후, MTT 용액(PBS 중 5 mg/mL의 5 μL)을 웰에 첨가하고 37℃에서 4시간 동안 인큐베이션하였다. 그런 다음 100 μL의 신선한 DMSO를 처리하여 10분 동안 결정을 용해시키고 마이크로플레이트 판독기(Thermo Scientific Inc., San Diego, CA, USA)로 세포를 검출하여 570 nm에서 흡광도를 측정하였다. 처리되지 않은 대조군의 흡광도를 비교하여 상대적 세포 생존율을 계산하였다. 모든 실험은 삼중으로 수행되었다.Cytotoxicity was determined by MTT (Amresco, Solon, OH, USA) colorimetric assay. RAW 264.7 cells were seeded in a 96-well plate at a density of 2×10 5 cells/mL and incubated for 24 hours in the presence of various concentrations of Ramalin and its derivatives. After incubation with the test substance, MTT solution (5 μL of 5 mg/mL in PBS) was added to the wells and incubated at 37° C. for 4 hours. Then, 100 μL of fresh DMSO was treated to dissolve the crystals for 10 minutes, and the cells were detected with a microplate reader (Thermo Scientific Inc., San Diego, CA, USA) and absorbance was measured at 570 nm. Relative cell viability was calculated by comparing the absorbance of the untreated control. All experiments were performed in triplicate.

산화질소 생산의 측정Measurement of Nitric Oxide Production

아질산염(nitrite) 축적은 배지에서 NO 생성의 지표로 사용되었고 아질산염 수준은 Griess 시약(1% 설파닐아미드, 0.1% N-(1-나파틸)-에틸렌디아민 디히드로클로라이드 및 5% 인산 산). 아질산염의 양을 측정하기 위해 96-웰 플레이트에 1×106 cells/mL를 접종한 후 37℃에서 1시간 동안 라말린과 그 유도체의 농도를 표시한 후 0.5 ㎍/mL로 자극하였다. 200 μL의 최종 부피에서 24시간 동안 LPS(0.5 ㎍/mL, Sigma-Aldrich, CA, USA). 그런 다음, 100 μL의 세포 배양 상층액을 96-웰 플레이트에서 100 μL의 Griess 시약과 혼합하였다. 아질산나트륨을 사용하여 표준 곡선을 생성하고 아질산염의 농도를 마이크로플레이트 판독기를 사용하여 540 nm에서의 흡광도로 측정하였다. 모든 결정은 삼중으로 수행되었다.Nitrite accumulation was used as an indicator of NO production in the medium and nitrite levels were measured using Griess reagent (1% sulfanilamide, 0.1% N-(1-napatyl)-ethylenediamine dihydrochloride and 5% phosphoric acid). In order to measure the amount of nitrite, 1×10 6 cells/mL were inoculated in a 96-well plate, and the concentration of Ramalin and its derivatives was displayed at 37° C. for 1 hour, followed by stimulation with 0.5 μg/mL. LPS (0.5 μg/mL, Sigma-Aldrich, CA, USA) for 24 h in a final volume of 200 μL. Then, 100 μL of cell culture supernatant was mixed with 100 μL of Griess reagent in a 96-well plate. A standard curve was generated using sodium nitrite and the concentration of nitrite was determined by absorbance at 540 nm using a microplate reader. All determinations were performed in triplicate.

라말린 및 그 유도체의 항염 활성Anti-inflammatory activity of Ramalin and its derivatives

알츠하이머병의 발병기전에서 염증의 중요성이 인식되어 왔다. 또한 염증이 알츠하이머병 병리에서 역할을 하고 질병을 악화시키는 것으로 확인되었다(Heppner, F. L. et al., Nat. Rev. Neurosci., 2015, 16, 358-372). 이에 라말린과 그 유도체의 세포독성을 조사하고 산화질소 농도를 측정하여 항염증 활성을 확인하였다. 라말린은 최대 농도에서 약간 세포독성이 있는 것으로 밝혀졌다(도 2). 역으로, 그 유도체는 이들 세포에서 세포독성 효과를 나타내지 않았다. 또한, 라말린은 농도 의존적으로 NO 억제 활성을 나타냈다. RA-25Me, RA-34Me, RA-2F, RA-4F, RA-24F 및 RA-PF를 포함한 이의 유도체도 농도 의존적 NO 억제를 보인 반면, RA-2Me, RA-3Me 및 RA-4Me는 약한 항염 효과를 나타낸다. 또한 이들 유도체 중 RA-24F는 세포독성 없이 라말린보다 더 강력한 항염 효과를 보였다.The importance of inflammation in the pathogenesis of Alzheimer's disease has been recognized. Inflammation has also been shown to play a role in Alzheimer's disease pathology and exacerbate the disease (Heppner, F. L. et al., Nat. Rev. Neurosci., 2015, 16, 358-372). Accordingly, the cytotoxicity of Ramalin and its derivatives was investigated, and the anti-inflammatory activity was confirmed by measuring the nitric oxide concentration. Ramalin was found to be slightly cytotoxic at maximum concentration (FIG. 2). Conversely, the derivative showed no cytotoxic effect in these cells. In addition, Ramalin exhibited NO inhibitory activity in a concentration-dependent manner. Its derivatives, including RA-25Me, RA-34Me, RA-2F, RA-4F, RA-24F and RA-PF, also showed concentration-dependent NO inhibition, whereas RA-2Me, RA-3Me and RA-4Me showed weak anti-inflammatory show effect. Also, among these derivatives, RA-24F showed a stronger anti-inflammatory effect than Ramalin without cytotoxicity.

실시예 4: 라말린 및 그 유도체의 β-분비효소(β-Secretase, BACE-1) 억제 활성 분석Example 4: Analysis of β-secretase (BACE-1) inhibitory activity of Ramalin and its derivatives

BACE-1 억제 분석은 제조업체 지침에 따라 β-Secretase FRET 키트(BACE-1, Thermo Fisher Scientific, San Diego, CA, USA)를 사용하여 수행되었다. 분석은 앞서 설명한 대로 제조업체의 프로토콜에 따라 수행되었다. 탈이온 증류수(DDW)에서 라말린 및 그 유도체의 스톡을 준비하였다(20mM). 샘플을 분석 완충액에 추가로 희석하고 검은색 96웰 마이크로플레이트를 10μL의 BACE-1 기질과 혼합하였다. 그런 다음 각 웰에 10 μL의 3xBACE-1 효소를 첨가하여 반응을 시작하였다. 플레이트를 암실에서 실온에서 60분 동안 인큐베이션하였다. 인큐베이션 후, 10 μL의 2.5 mM 아세트산나트륨을 각 웰에 첨가하여 반응을 정지시켰다. 마지막으로, 545 nm의 여기 파장 및 585 nm의 방출 파장에서 다중 모드 플레이트 판독기(MultistkanTM GO, Thermo Scientific, Waltham, MA, USA)에 다중 웰 분광 형광계 기기를 사용하였다. 반값 최대 억제 농도(IC50)는 억제제 농도의 대수에 대해 수득된 시간당 상대 형광 단위(RFU/h)를 플롯팅하여 계산하였다. 모든 결정은 삼중으로 수행되었다.BACE-1 inhibition assay was performed using the β-Secretase FRET kit (BACE-1, Thermo Fisher Scientific, San Diego, CA, USA) according to the manufacturer's instructions. The assay was performed according to the manufacturer's protocol as previously described. Stocks of Ramalin and its derivatives were prepared (20 mM) in deionized distilled water (DDW). Samples were further diluted in assay buffer and a black 96-well microplate was mixed with 10 μL of BACE-1 substrate. The reaction was then started by adding 10 μL of 3xBACE-1 enzyme to each well. Plates were incubated for 60 minutes at room temperature in the dark. After incubation, the reaction was stopped by adding 10 μL of 2.5 mM sodium acetate to each well. Finally, a multi-well spectrofluorometer instrument was used on a multimode plate reader (Multistkan TM GO, Thermo Scientific, Waltham, MA, USA) at an excitation wavelength of 545 nm and an emission wavelength of 585 nm. The half-maximal inhibitory concentration (IC 50 ) was calculated by plotting the obtained relative fluorescence units per hour (RFU/h) against the logarithm of the inhibitor concentration. All determinations were performed in triplicate.

약간의 BACE-1 발현은 알츠하이머병을 유발하는 Aβ의 축적으로 이어질 수 있으며, 이는 알츠하이머병이 만성 질환이라는 점을 감안할 때 치명적일 수 있다. 따라서 BACE-1 억제는 AD 억제를 위한 유망한 표적이다. PanVera®의 BACE-1 형광 공명 에너지 전달 분석(FRET) 키트(P2985, Madison, WI, USA)를 사용하여 라말린과 그 유도체의 BACE-1 억제 활성을 확인하였다(Niu, Y. et al., Chem. Biol. Drug Des., 2012, 80, 775-780). 라말린 및 이의 합성된 유도체의 해당 IC50 값은 표 2에 나와 있다. 실험은 모든 화합물에 대해 각각 3회 수행되었다. 따라서, RA-24F를 제외하고 라말린 및 기타 모든 유도체는 마이크로몰 농도에서 IC50 값을 나타내는 것으로 관찰되었다. 구체적으로 라말린의 BACE-1 저해 활성은 17.66 ± 2.74 μM이었으며, 유도체 중 RA-25Me가 9.81 ± 1.21 μM으로 가장 높은 활성을 보였다.Slight BACE-1 expression can lead to accumulation of Aβ that causes Alzheimer's disease, which can be fatal given that Alzheimer's disease is a chronic disease. Thus, BACE-1 inhibition is a promising target for AD inhibition. The BACE-1 inhibitory activity of Ramalin and its derivatives was confirmed using PanVera ® BACE-1 Fluorescence Resonance Energy Transfer Assay (FRET) kit (P2985, Madison, WI, USA) (Niu, Y. et al., Chem. Biol. Drug Des., 2012, 80, 775-780). The corresponding IC 50 values of Ramalin and its synthesized derivatives are shown in Table 2. Experiments were performed in triplicate each for all compounds. Thus, except for RA-24F, Ramalin and all other derivatives were observed to exhibit IC50 values at micromolar concentrations. Specifically, Ramalin's BACE-1 inhibitory activity was 17.66 ± 2.74 μM, and RA-25Me showed the highest activity among the derivatives, 9.81 ± 1.21 μM.

Figure PCTKR2022016326-appb-img-000024
Figure PCTKR2022016326-appb-img-000024

알츠하이머병은 병리학적 단백질 응집, 신경 전달 장애, 산화 스트레스 증가, 미세아교세포 매개 신경 염증을 포함하는 복잡한 병태생리를 가지고 있다. AD와 관련된 분자 표적 중 하나만을 표적으로 하는 치료제가 보고되었다. 예를 들어, BACE-1 억제 활성이 높은 치료제는 Aβ 생성을 성공적으로 억제하고 제거할 수 있지만 인지 기능을 회복할 수는 없다. 따라서 알츠하이머병 치료제 개발 시 단일 표적 약물보다는 다중 표적 약물에 집중하는 것이 중요하다. 항산화, BACE-1 억제 및 항염증 활성을 갖는 라말린은 알츠하이머병의 복잡한 특성을 해결할 수 있는 다중 활성을 가진 유망한 치료제 역할을 할 수 있다. 그러나 수성 환경에서 낮은 안정성과 높은 농도의 세포 독성으로 인해 적용에 제한이 있다. RA-25Me와 RA-34Me는 라말린과 유사한 활성을 보이며 이러한 단점을 개선할 수 있는 가능성을 강조하였다. 따라서 추가적인 유도체 합성을 통한 라말린의 물성 및 활성 개선은 향후 알츠하이머병 치료 및 예방에 응용될 수 있을 것이다.Alzheimer's disease has a complex pathophysiology that includes pathological protein aggregation, impaired neurotransmission, increased oxidative stress, and microglia-mediated neuroinflammation. Therapeutics targeting only one of the molecular targets associated with AD have been reported. For example, therapeutics with high BACE-1 inhibitory activity can successfully inhibit and eliminate Aβ production, but cannot restore cognitive function. Therefore, when developing a treatment for Alzheimer's disease, it is important to focus on multi-target drugs rather than single-target drugs. Ramalin, which has antioxidant, BACE-1 inhibitory and anti-inflammatory activities, can serve as a promising therapeutic agent with multiple activities that can address the complex nature of Alzheimer's disease. However, its application is limited due to low stability in an aqueous environment and cytotoxicity at high concentrations. RA-25Me and RA-34Me showed activities similar to Ramalin, highlighting the possibility of improving these disadvantages. Therefore, improvements in physical properties and activity of Ramalin through the synthesis of additional derivatives may be applied to the treatment and prevention of Alzheimer's disease in the future.

라말린 및 그 유도체의 항산화, BACE-1 억제 및 항염 활성을 평가하였다. 9개의 라말린 유도체는 라말린 합성에 사용된 것과 유사한 조건에서 비교적 높은 수율(62-79%)로 합성되었다. 새로 합성된 메틸기나 플루오로 원자를 가진 유도체는 라말린과 비슷하거나 더 나은 항산화 효과를 나타냈다. 또한, 유도체의 항산화 효과는 메틸기의 위치에 따라 다르며, F 원자를 갖는 유도체는 상대적으로 낮은 항산화 효과를 보였다. 특징적으로 RA-2Me는 라말린보다 높은 항산화 효과를 보였다. 또한, RA-2Me, RA-3Me, RA-4Me를 제외한 합성된 유도체의 대부분은 세포독성 없이 농도 의존적으로 NO를 억제하였으며, 구체적으로 RA-24F가 더 높은 항염증 효과를 나타내나, 라말린보다 세포 독성이 없다. 또한, RA-24F를 제외하고 라말린 및 그 유도체는 농도 의존적으로 BACE-1 억제 활성을 나타냈다. 이 중 RA-25Me는 9.81 ± 1.21 μM 농도에서 가장 높은 BACE-1 저해 활성을 보였다.Antioxidant, BACE-1 inhibitory and anti-inflammatory activities of Ramalin and its derivatives were evaluated. Nine derivatives of ramalin were synthesized in relatively high yields (62-79%) under conditions similar to those used for the synthesis of ramalin. The newly synthesized derivatives with methyl or fluoro atoms showed similar or better antioxidant effects than Ramalin. In addition, the antioxidant effect of the derivative differs depending on the location of the methyl group, and the derivative having an F atom showed a relatively low antioxidant effect. Characteristically, RA-2Me showed a higher antioxidant effect than Ramalin. In addition, most of the synthesized derivatives, except for RA-2Me, RA-3Me, and RA-4Me, inhibited NO in a concentration-dependent manner without cytotoxicity, and specifically, RA-24F showed a higher anti-inflammatory effect than Ramalin. No cytotoxicity. In addition, except for RA-24F, Ramalin and its derivatives exhibited BACE-1 inhibitory activity in a concentration-dependent manner. Among them, RA-25Me showed the highest BACE-1 inhibitory activity at a concentration of 9.81 ± 1.21 μM.

전자-트론 인출 또는 공여 그룹으로 구성된 방향족 시스템에서는 명확한 전자 효과가 관찰되지 않는다. 그러나 RA-2Me와 RA-2F를 비교했을 때 RA-2Me는 우수한 항산화 효과를 보였고 RA-2F는 우수한 항염 효과를 보였다. 또한, BACE-1 저해 활성도 두 물질 모두 유사한 결과를 보였다. 합성된 유도체 중 RA-25Me와 RA-34Me는 항산화, 항염, BACE-1 억제 활성을 나타내어 라말린과 유사하나 세포독성 및 안정성 측면에서 라말린보다 우수하였다. 반대로, RA-24F는 높은 항염 활성을 나타냈으나 BACE-1 억제 활성은 확인할 수 없었다.No clear electron effect is observed in aromatic systems composed of electron-tron withdrawing or donating groups. However, when RA-2Me and RA-2F were compared, RA-2Me showed excellent antioxidant effect and RA-2F showed excellent anti-inflammatory effect. Also, the BACE-1 inhibitory activity of both substances showed similar results. Among the synthesized derivatives, RA-25Me and RA-34Me showed antioxidant, anti-inflammatory, and BACE-1 inhibitory activities similar to Ramalin, but were superior to Ramalin in terms of cytotoxicity and stability. Conversely, RA-24F showed high anti-inflammatory activity, but no BACE-1 inhibitory activity could be confirmed.

BBB를 관통하여 뇌에 도달할 수 있는 충분한 억제 활성을 갖는 소분자 BACE-1 억제제를 개발하기 위해 집중적인 노력이 이루어져 왔다(Coimbra, J. R. et al., Front. Chem., 2018, 6, 178). 큰 제약사들이 선호하는 BACE-1 억제를 표적으로 하는 치료제가 Aβ 수치를 감소시킬 수 있지만 알츠하이머병 환자에서 인지 기능의 개선이 관찰되지 않았다(Voytyuk, I. et al., Biol. Psychiatry, 2018, 83, 320-327). 이는 알츠하이머병의 조기 진단과 치료가 중요함을 시사한다. 최근 연구에서는 알츠하이머병의 복잡성을 고려한 BACE-1을 중심으로 한 다중 표적 접근법을 가진 저분자의 개발이 보고되고 있다(Prati, F. et al., J. Med. Chem., 2018, 61, 619-637). 이러한 분자의 예로는 BACE-1 및 AChE의 억제제인 쿠마린 유도체(Piazzi, L. et al., Bioorg. Med. Chem. Lett., 2008, 18, 423-426)와 BACE-1 및 GSK-3β의 억제제인 커큐민 유도체(Di Martino, R. M. C. et al., J. Med. Chem., 2016, 59, 531-544)가 있다. 따라서 BACE-1 억제 활성만을 나타내는 약물에는 한계가 있다는 점에서 다중 표적 활성을 갖는 화합물이 각광받고 있다. 또한, BACE-1 및 AChE에 대한 억제 활성과 항산화 효과를 나타내는 memoquin은 다중 표적 화합물의 적합한 예이다(Capurro, V. et al., PLoS One, 2013, 8, e56870). 스코폴라민에 의해 유발된 기억상실증이 있는 생쥐의 공간 및 일화적 기억력 결핍은 메모퀸 치료 후 해결되었다. 이와 관련하여 추가 확인이 필요하지만 AD에서 Aβ 생성의 제거 및 억제뿐만 아니라 실질적인 인지 개선을 위해서는 항산화 효과가 필요하다. Aβ 축적을 통한 알츠하이머병 발병 이전에, ROS 유도 산화 손상은 알츠하이머병 유도를 가속화한다. 또한, ROS는 알츠하이머병 유도와 관련된 핵산, 지질, 단백질에 산화적 손상을 일으키며 이러한 손상 물질은 알츠하이머병 환자의 뇌에서 초기에 확인되었다(Choi, D. Y. et al., Brain Res. Bull., 2012, 87, 144-153). 보고된 바에 따르면, 산화적 손상은 신경 염증을 유발하고 BACE-1 활성을 매개하며(Zameer, S. et al., Neurotoxicology, 2019, 70, 122-134; Reddy, P. H., J. Neurochem., 2006, 96, 1-13). BACE-1에 의해 생성된 Aβ는 차례로 ROS 생성을 유도하여 AD의 증상을 더욱 악화시킬 수 있다(Tamagno, E. et al., Free Radic. Biol. Med., 2006, 41, 202-212; Reddy, P. H. et al., Trends Mol. Med., 2008, 14, 45-53; Lin, M. T. et al., Nature, 2006, 443, 787-795). 따라서 강력한 항산화제인 라말린은 다양한 생리활성을 바탕으로 알츠하이머병의 다중표적 치료제로서 상당한 잠재력을 갖고 있다. 그러나 추가적인 AD 유도제 관련 활성 및 유도체에 대한 추가 연구가 필요하다. 따라서 여기에 제시된 결과를 바탕으로 활성이 향상된 유도체의 합성을 계속해야 한다.Intensive efforts have been made to develop small molecule BACE-1 inhibitors with sufficient inhibitory activity to penetrate the BBB and reach the brain (Coimbra, J. R. et al., Front. Chem., 2018, 6, 178). Therapies targeting BACE-1 inhibition, favored by big pharma, can reduce Aβ levels, but no improvement in cognitive function has been observed in Alzheimer's disease patients (Voytyuk, I. et al., Biol. Psychiatry, 2018, 83 , 320-327). This suggests that early diagnosis and treatment of Alzheimer's disease is important. Recent studies have reported the development of small molecules with a multi-target approach centered on BACE-1 considering the complexity of Alzheimer's disease (Prati, F. et al., J. Med. Chem., 2018, 61, 619- 637). Examples of such molecules are coumarin derivatives, inhibitors of BACE-1 and AChE (Piazzi, L. et al., Bioorg. Med. Chem. Lett., 2008, 18, 423-426) and inhibitors of BACE-1 and GSK-3β. Curcumin derivatives (Di Martino, R. M. C. et al., J. Med. Chem., 2016, 59, 531-544) are inhibitors. Therefore, compounds with multi-target activity are in the spotlight in that drugs exhibiting only BACE-1 inhibitory activity have limitations. In addition, memoquin, which exhibits inhibitory activities and antioxidant effects on BACE-1 and AChE, is a suitable example of a multi-target compound (Capurro, V. et al., PLoS One, 2013, 8, e56870). Spatial and episodic memory deficits in mice with scopolamine-induced amnesia resolved after treatment with memoquine. Although further confirmation is needed in this regard, antioxidant effects are required for substantial cognitive improvement as well as elimination and inhibition of Aβ production in AD. Prior to Alzheimer's disease onset through Aβ accumulation, ROS-induced oxidative damage accelerates Alzheimer's disease induction. In addition, ROS causes oxidative damage to nucleic acids, lipids, and proteins related to the induction of Alzheimer's disease, and these damaging substances were initially identified in the brains of Alzheimer's disease patients (Choi, D. Y. et al., Brain Res. Bull., 2012, 87, 144-153). Reportedly, oxidative damage induces neuroinflammation and mediates BACE-1 activity (Zameer, S. et al., Neurotoxicology, 2019, 70, 122-134; Reddy, P. H., J. Neurochem., 2006 , 96, 1-13). Aβ generated by BACE-1 can in turn induce ROS production, further exacerbating the symptoms of AD (Tamagno, E. et al., Free Radic. Biol. Med., 2006, 41, 202-212; Reddy , P. H. et al., Trends Mol. Med., 2008, 14, 45-53; Lin, M. T. et al., Nature, 2006, 443, 787-795). Therefore, Ramalin, a powerful antioxidant, has considerable potential as a multi-target treatment for Alzheimer's disease based on its various physiological activities. However, further studies on additional AD inducer-related activities and derivatives are needed. Therefore, based on the results presented here, the synthesis of derivatives with improved activity should continue.

본 발명에 따른 라말린 유도체는 Aβ 축적을 유발하는 BACE-1에 대하여 억제 활성을 가지며, 활성산소를 제거하여 알츠하이머병 예방 및 치료에 중요한 역할을 하는 항산화 효과를 나타낸다. 또한, 알츠하이머병을 악화시키는 염증 억제 효과를 나타내며, LPS로 자극된 RAW 264.7 세포에서도 독성 없이 항염 효과를 보임에 따라 라말린 유도체는 알츠하이머병과 관련된 여러 표적에 대해 활성을 나타내므로 알츠하이머병의 치료제로 작용할 수 있다.Ramalin derivatives according to the present invention have inhibitory activity against BACE-1, which induces Aβ accumulation, and exhibit antioxidant effects that play an important role in preventing and treating Alzheimer's disease by removing active oxygen. In addition, it exhibits an anti-inflammatory effect that exacerbates Alzheimer's disease, and shows an anti-inflammatory effect without toxicity in RAW 264.7 cells stimulated with LPS. As such, ramalin derivatives are active against various targets related to Alzheimer's disease, so they can act as a therapeutic agent for Alzheimer's disease. can

이상으로 본 발명 내용의 특정한 부분을 상세히 기술하였는 바, 당업계의 통상의 지식을 가진 자에게 있어서 이러한 구체적 기술은 단지 바람직한 실시 양태일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백할 것이다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다고 할 것이다.알츠하이머병은 병리학적 단백질 응집, 신경 전달 장애, 산화 스트레스 증가, 미세아교세포 매개 신경 염증을 포함하는 복잡한 병태생리를 가지고 있다. AD와 관련된 분자 표적 중 하나만을 표적으로 하는 치료제가 보고되었다. 예를 들어, BACE-1 억제 활성이 높은 치료제는 Aβ 생성을 성공적으로 억제하고 제거할 수 있지만 인지 기능을 회복할 수는 없다. 따라서 알츠하이머병 치료제 개발 시 단일 표적 약물보다는 다중 표적 약물에 집중하는 것이 중요하다. 항산화, BACE-1 억제 및 항염증 활성을 갖는 라말린은 알츠하이머병의 복잡한 특성을 해결할 수 있는 다중 활성을 가진 유망한 치료제 역할을 할 수 있다. 그러나 수성 환경에서 낮은 안정성과 높은 농도의 세포 독성으로 인해 적용에 제한이 있다. RA-25Me와 RA-34Me는 라말린과 유사한 활성을 보이며 이러한 단점을 개선할 수 있는 가능성을 강조하였다. 따라서 추가적인 유도체 합성을 통한 라말린의 물성 및 활성 개선은 향후 알츠하이머병 치료 및 예방에 응용될 수 있을 것이다.Having described specific parts of the present invention in detail above, it will be 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. will be. Accordingly, the substantial scope of the present invention will be defined by the appended claims and their equivalents. Alzheimer's disease is a complex condition that includes pathological protein aggregation, neurotransmission disorders, increased oxidative stress, and microglia-mediated neuroinflammation. have menstruation Therapeutics targeting only one of the molecular targets associated with AD have been reported. For example, therapeutics with high BACE-1 inhibitory activity can successfully inhibit and eliminate Aβ production, but cannot restore cognitive function. Therefore, when developing a treatment for Alzheimer's disease, it is important to focus on multi-target drugs rather than single-target drugs. Ramalin, which has antioxidant, BACE-1 inhibitory and anti-inflammatory activities, can serve as a promising therapeutic agent with multiple activities that can address the complex nature of Alzheimer's disease. However, its application is limited due to low stability in an aqueous environment and cytotoxicity at high concentrations. RA-25Me and RA-34Me showed activities similar to Ramalin, highlighting the possibility of improving these disadvantages. Therefore, improvements in physical properties and activity of Ramalin through the synthesis of additional derivatives may be applied to the treatment and prevention of Alzheimer's disease in the future.

라말린 및 그 유도체의 항산화, BACE-1 억제 및 항염 활성을 평가하였다. 9개의 라말린 유도체는 라말린 합성에 사용된 것과 유사한 조건에서 비교적 높은 수율(62-79%)로 합성되었다. 새로 합성된 메틸기나 플루오로 원자를 가진 유도체는 라말린과 비슷하거나 더 나은 항산화 효과를 나타냈다. 또한, 유도체의 항산화 효과는 메틸기의 위치에 따라 다르며, F 원자를 갖는 유도체는 상대적으로 낮은 항산화 효과를 보였다. 특징적으로 RA-2Me는 라말린보다 높은 항산화 효과를 보였다. 또한, RA-2Me, RA-3Me, RA-4Me를 제외한 합성된 유도체의 대부분은 세포독성 없이 농도 의존적으로 NO를 억제하였으며, 구체적으로 RA-24F가 더 높은 항염증 효과를 나타내나, 라말린보다 세포 독성이 없다. 또한, RA-24F를 제외하고 라말린 및 그 유도체는 농도 의존적으로 BACE-1 억제 활성을 나타냈다. 이 중 RA-25Me는 9.81 ± 1.21 μM 농도에서 가장 높은 BACE-1 저해 활성을 보였다.Antioxidant, BACE-1 inhibitory and anti-inflammatory activities of Ramalin and its derivatives were evaluated. Nine derivatives of ramalin were synthesized in relatively high yields (62-79%) under conditions similar to those used for the synthesis of ramalin. The newly synthesized derivatives with methyl or fluoro atoms showed similar or better antioxidant effects than Ramalin. In addition, the antioxidant effect of the derivative differs depending on the location of the methyl group, and the derivative having an F atom showed a relatively low antioxidant effect. Characteristically, RA-2Me showed a higher antioxidant effect than Ramalin. In addition, most of the synthesized derivatives, except for RA-2Me, RA-3Me, and RA-4Me, inhibited NO in a concentration-dependent manner without cytotoxicity, and specifically, RA-24F showed a higher anti-inflammatory effect than Ramalin. No cytotoxicity. In addition, except for RA-24F, Ramalin and its derivatives exhibited BACE-1 inhibitory activity in a concentration-dependent manner. Among them, RA-25Me showed the highest BACE-1 inhibitory activity at a concentration of 9.81 ± 1.21 μM.

전자-트론 인출 또는 공여 그룹으로 구성된 방향족 시스템에서는 명확한 전자 효과가 관찰되지 않는다. 그러나 RA-2Me와 RA-2F를 비교했을 때 RA-2Me는 우수한 항산화 효과를 보였고 RA-2F는 우수한 항염 효과를 보였다. 또한, BACE-1 저해 활성도 두 물질 모두 유사한 결과를 보였다. 합성된 유도체 중 RA-25Me와 RA-34Me는 항산화, 항염, BACE-1 억제 활성을 나타내어 라말린과 유사하나 세포독성 및 안정성 측면에서 라말린보다 우수하였다. 반대로, RA-24F는 높은 항염 활성을 나타냈으나 BACE-1 억제 활성은 확인할 수 없었다.No clear electron effect is observed in aromatic systems composed of electron-tron withdrawing or donating groups. However, when RA-2Me and RA-2F were compared, RA-2Me showed excellent antioxidant effect and RA-2F showed excellent anti-inflammatory effect. Also, the BACE-1 inhibitory activity of both substances showed similar results. Among the synthesized derivatives, RA-25Me and RA-34Me showed antioxidant, anti-inflammatory, and BACE-1 inhibitory activities similar to Ramalin, but were superior to Ramalin in terms of cytotoxicity and stability. Conversely, RA-24F showed high anti-inflammatory activity, but no BACE-1 inhibitory activity could be confirmed.

BBB를 관통하여 뇌에 도달할 수 있는 충분한 억제 활성을 갖는 소분자 BACE-1 억제제를 개발하기 위해 집중적인 노력이 이루어져 왔다(Coimbra, J. R. et al., Front. Chem., 2018, 6, 178). 큰 제약사들이 선호하는 BACE-1 억제를 표적으로 하는 치료제가 Aβ 수치를 감소시킬 수 있지만 알츠하이머병 환자에서 인지 기능의 개선이 관찰되지 않았다(Voytyuk, I. et al., Biol. Psychiatry, 2018, 83, 320-327). 이는 알츠하이머병의 조기 진단과 치료가 중요함을 시사한다. 최근 연구에서는 알츠하이머병의 복잡성을 고려한 BACE-1을 중심으로 한 다중 표적 접근법을 가진 저분자의 개발이 보고되고 있다(Prati, F. et al., J. Med. Chem., 2018, 61, 619-637). 이러한 분자의 예로는 BACE-1 및 AChE의 억제제인 쿠마린 유도체(Piazzi, L. et al., Bioorg. Med. Chem. Lett., 2008, 18, 423-426)와 BACE-1 및 GSK-3β의 억제제인 커큐민 유도체(Di Martino, R. M. C. et al., J. Med. Chem., 2016, 59, 531-544)가 있다. 따라서 BACE-1 억제 활성만을 나타내는 약물에는 한계가 있다는 점에서 다중 표적 활성을 갖는 화합물이 각광받고 있다. 또한, BACE-1 및 AChE에 대한 억제 활성과 항산화 효과를 나타내는 memoquin은 다중 표적 화합물의 적합한 예이다(Capurro, V. et al., PLoS One, 2013, 8, e56870). 스코폴라민에 의해 유발된 기억상실증이 있는 생쥐의 공간 및 일화적 기억력 결핍은 메모퀸 치료 후 해결되었다. 이와 관련하여 추가 확인이 필요하지만 AD에서 Aβ 생성의 제거 및 억제뿐만 아니라 실질적인 인지 개선을 위해서는 항산화 효과가 필요하다. Aβ 축적을 통한 알츠하이머병 발병 이전에, ROS 유도 산화 손상은 알츠하이머병 유도를 가속화한다. 또한, ROS는 알츠하이머병 유도와 관련된 핵산, 지질, 단백질에 산화적 손상을 일으키며 이러한 손상 물질은 알츠하이머병 환자의 뇌에서 초기에 확인되었다(Choi, D. Y. et al., Brain Res. Bull., 2012, 87, 144-153). 보고된 바에 따르면, 산화적 손상은 신경 염증을 유발하고 BACE-1 활성을 매개하며(Zameer, S. et al., Neurotoxicology, 2019, 70, 122-134; Reddy, P. H., J. Neurochem., 2006, 96, 1-13). BACE-1에 의해 생성된 Aβ는 차례로 ROS 생성을 유도하여 AD의 증상을 더욱 악화시킬 수 있다(Tamagno, E. et al., Free Radic. Biol. Med., 2006, 41, 202-212; Reddy, P. H. et al., Trends Mol. Med., 2008, 14, 45-53; Lin, M. T. et al., Nature, 2006, 443, 787-795). 따라서 강력한 항산화제인 라말린은 다양한 생리활성을 바탕으로 알츠하이머병의 다중표적 치료제로서 상당한 잠재력을 갖고 있다. 그러나 추가적인 AD 유도제 관련 활성 및 유도체에 대한 추가 연구가 필요하다. 따라서 여기에 제시된 결과를 바탕으로 활성이 향상된 유도체의 합성을 계속해야 한다.Intensive efforts have been made to develop small molecule BACE-1 inhibitors with sufficient inhibitory activity to penetrate the BBB and reach the brain (Coimbra, J. R. et al., Front. Chem., 2018, 6, 178). Therapies targeting BACE-1 inhibition, favored by big pharma, can reduce Aβ levels, but no improvement in cognitive function has been observed in Alzheimer's disease patients (Voytyuk, I. et al., Biol. Psychiatry, 2018, 83 , 320-327). This suggests that early diagnosis and treatment of Alzheimer's disease is important. Recent studies have reported the development of small molecules with a multi-target approach centered on BACE-1 considering the complexity of Alzheimer's disease (Prati, F. et al., J. Med. Chem., 2018, 61, 619- 637). Examples of such molecules are coumarin derivatives, inhibitors of BACE-1 and AChE (Piazzi, L. et al., Bioorg. Med. Chem. Lett., 2008, 18, 423-426) and inhibitors of BACE-1 and GSK-3β. Curcumin derivatives (Di Martino, R. M. C. et al., J. Med. Chem., 2016, 59, 531-544) are inhibitors. Therefore, compounds with multi-target activity are in the spotlight in that drugs exhibiting only BACE-1 inhibitory activity have limitations. In addition, memoquin, which exhibits inhibitory activities and antioxidant effects on BACE-1 and AChE, is a suitable example of a multi-target compound (Capurro, V. et al., PLoS One, 2013, 8, e56870). Spatial and episodic memory deficits in mice with scopolamine-induced amnesia resolved after treatment with memoquine. Although further confirmation is needed in this regard, antioxidant effects are required for substantial cognitive improvement as well as elimination and inhibition of Aβ production in AD. Prior to Alzheimer's disease onset through Aβ accumulation, ROS-induced oxidative damage accelerates Alzheimer's disease induction. In addition, ROS causes oxidative damage to nucleic acids, lipids, and proteins related to the induction of Alzheimer's disease, and these damaging substances were initially identified in the brains of Alzheimer's disease patients (Choi, D. Y. et al., Brain Res. Bull., 2012, 87, 144-153). Reportedly, oxidative damage induces neuroinflammation and mediates BACE-1 activity (Zameer, S. et al., Neurotoxicology, 2019, 70, 122-134; Reddy, P. H., J. Neurochem., 2006 , 96, 1-13). Aβ generated by BACE-1 can in turn induce ROS production, further exacerbating the symptoms of AD (Tamagno, E. et al., Free Radic. Biol. Med., 2006, 41, 202-212; Reddy , P. H. et al., Trends Mol. Med., 2008, 14, 45-53; Lin, M. T. et al., Nature, 2006, 443, 787-795). Therefore, Ramalin, a powerful antioxidant, has considerable potential as a multi-target treatment for Alzheimer's disease based on its various physiological activities. However, further studies on additional AD inducer-related activities and derivatives are needed. Therefore, based on the results presented here, the synthesis of derivatives with improved activity should continue.

Claims (18)

화학식 1로 표시되는 것을 특징으로 하는 라말린 유도체:Ramalin derivatives characterized by being represented by Formula 1: [화학식 1][Formula 1]
Figure PCTKR2022016326-appb-img-000025
Figure PCTKR2022016326-appb-img-000025
화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬, C3-C6 사이클로알킬, 할로겐, 하이드록시, 카복실, 아미노 또는 시아노이고, 단, R1, R2, R3, R4 및 R5는 동시에 수소원자이거나 R1, R2, R3 및 R4는 수소원자이고, R5는 하이드록시인 경우는 제외된다.In Formula 1, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.
제1항에 있어서, 화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬 또는 할로겐인 것을 특징으로 하는 라말린 유도체.The Ramalin derivative according to claim 1, wherein each of R 1 , R 2 , R 3 , R 4 and R 5 in Formula 1 is independently a hydrogen atom, C 1 -C 6 alkyl or halogen. 제1항에 있어서, 화학식 2 내지 화학식 10에서 선택되는 것을 특징으로 하는 라말린 유도체:The Ramalin derivative according to claim 1, characterized in that it is selected from Formulas 2 to 10: [화학식 2][Formula 2]
Figure PCTKR2022016326-appb-img-000026
Figure PCTKR2022016326-appb-img-000026
[화학식 3][Formula 3]
Figure PCTKR2022016326-appb-img-000027
Figure PCTKR2022016326-appb-img-000027
[화학식 4][Formula 4]
Figure PCTKR2022016326-appb-img-000028
Figure PCTKR2022016326-appb-img-000028
[화학식 5][Formula 5]
Figure PCTKR2022016326-appb-img-000029
Figure PCTKR2022016326-appb-img-000029
[화학식 6][Formula 6]
Figure PCTKR2022016326-appb-img-000030
Figure PCTKR2022016326-appb-img-000030
[화학식 7][Formula 7]
Figure PCTKR2022016326-appb-img-000031
Figure PCTKR2022016326-appb-img-000031
[화학식 8][Formula 8]
Figure PCTKR2022016326-appb-img-000032
Figure PCTKR2022016326-appb-img-000032
[화학식 9][Formula 9]
Figure PCTKR2022016326-appb-img-000033
Figure PCTKR2022016326-appb-img-000033
[화학식 10][Formula 10]
Figure PCTKR2022016326-appb-img-000034
Figure PCTKR2022016326-appb-img-000034
다음 단계를 포함하는 제1항의 라말린 유도체의 제조방법:A method for preparing the ramalin derivative of claim 1 comprising the following steps: (a) 출발 물질인 화학식 11의 1-벤질-N-벤질옥시카보닐-L-글루탐산과 화학식 12의 페닐하이드라진의 커플링 반응을 수행하여 화학식 13의 화합물을 수득하는 단계; 및(a) obtaining a compound of Formula 13 by performing a coupling reaction between 1-benzyl-N-benzyloxycarbonyl-L-glutamic acid of Formula 11 as a starting material and phenylhydrazine of Formula 12; and (b) 화학식 13의 글루탐산 하이드레이트의 벤질기(Bn) 및 벤질옥시카보닐기(Cbz)의 탈보호화를 수행한 다음 수득한 조생성물을 재결정화하여 화학식 1의 라말린 유도체를 수득하는 단계.(b) deprotecting the benzyl group (Bn) and benzyloxycarbonyl group (Cbz) of the glutamic acid hydrate of Formula 13 and then recrystallizing the obtained crude product to obtain the Ramalin derivative of Formula 1. [화학식 1][Formula 1]
Figure PCTKR2022016326-appb-img-000035
Figure PCTKR2022016326-appb-img-000035
[화학식 11][Formula 11]
Figure PCTKR2022016326-appb-img-000036
Figure PCTKR2022016326-appb-img-000036
[화학식 12][Formula 12]
Figure PCTKR2022016326-appb-img-000037
Figure PCTKR2022016326-appb-img-000037
[화학식 13][Formula 13]
Figure PCTKR2022016326-appb-img-000038
Figure PCTKR2022016326-appb-img-000038
화학식 1, 12 및 13에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬, C3-C6 사이클로알킬, 할로겐, 하이드록시, 카복실, 아미노 또는 시아노이고, 단, R1, R2, R3, R4 및 R5는 동시에 수소원자이거나 R1, R3, R4 및 R5는 수소원자이고, R2는 하이드록시인 경우는 제외된다.In Formulas 1, 12 and 13, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano, provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 3 , R 4 and R 5 are hydrogen atoms and R 2 is hydroxy; is excluded.
제4항에 있어서, 상기 (a) 단계는 -5℃의 온도에서 1-벤질-N-벤질옥시카보닐-L-글루탐산을 디클로로메탄(dichloromethane, DCM)에 녹인 용액에 트리에틸아민(trimethylamine, TEA)과 에틸클로로포르메이트(ethylchloroformate, ECF)를 차례로 첨가한 다음, 반응 혼합물을 2시간 동안 교반하고, 온도를 유지하면서 페닐하이드라진 HCl 염을 첨가하여 커플링 반응을 수행하는 것을 특징으로 하는 라말린 유도체의 제조방법.The method of claim 4, wherein step (a) is performed in a solution of 1-benzyl-N-benzyloxycarbonyl-L-glutamic acid dissolved in dichloromethane (DCM) at -5 ° C. TEA) and ethylchloroformate (ECF) were sequentially added, the reaction mixture was stirred for 2 hours, and phenylhydrazine HCl salt was added while maintaining the temperature to perform a coupling reaction. Method for preparing derivatives. 제4항에 있어서, 상기 (b) 단계의 탈보호화는 Pd/C 및 H2 가스의 분위기에서 수행되는 것을 특징으로 하는 라말린 유도체의 제조방법.5. The method of claim 4, wherein the deprotection in step (b) is performed in an atmosphere of Pd/C and H 2 gas. 화학식 1로 표시되는 라말린 유도체를 유효성분으로 함유하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물:A pharmaceutical composition for preventing or treating degenerative brain diseases containing a ramalin derivative represented by Formula 1 as an active ingredient: [화학식 1][Formula 1]
Figure PCTKR2022016326-appb-img-000039
Figure PCTKR2022016326-appb-img-000039
화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬, C3-C6 사이클로알킬, 할로겐, 하이드록시, 카복실, 아미노 또는 시아노이고, 단, R1, R2, R3, R4 및 R5는 동시에 수소원자이거나 R1, R2, R3 및 R4는 수소원자이고, R5는 하이드록시인 경우는 제외된다.In Formula 1, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.
제7항에 있어서, 화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬 또는 할로겐인 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물.The method of claim 7, wherein in Formula 1, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 Alkyl or halogen, characterized in that prevention or treatment of degenerative brain disease pharmaceutical composition for use. 제7항에 있어서, 화학식 2 내지 화학식 10에서 선택되는 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물:The pharmaceutical composition for preventing or treating degenerative brain diseases according to claim 7, wherein the composition is selected from Formulas 2 to 10: [화학식 2][Formula 2]
Figure PCTKR2022016326-appb-img-000040
Figure PCTKR2022016326-appb-img-000040
[화학식 3][Formula 3]
Figure PCTKR2022016326-appb-img-000041
Figure PCTKR2022016326-appb-img-000041
[화학식 4][Formula 4]
Figure PCTKR2022016326-appb-img-000042
Figure PCTKR2022016326-appb-img-000042
[화학식 5][Formula 5]
Figure PCTKR2022016326-appb-img-000043
Figure PCTKR2022016326-appb-img-000043
[화학식 6][Formula 6]
Figure PCTKR2022016326-appb-img-000044
Figure PCTKR2022016326-appb-img-000044
[화학식 7][Formula 7]
Figure PCTKR2022016326-appb-img-000045
Figure PCTKR2022016326-appb-img-000045
[화학식 8][Formula 8]
Figure PCTKR2022016326-appb-img-000046
Figure PCTKR2022016326-appb-img-000046
[화학식 9][Formula 9]
Figure PCTKR2022016326-appb-img-000047
Figure PCTKR2022016326-appb-img-000047
[화학식 10][Formula 10]
Figure PCTKR2022016326-appb-img-000048
Figure PCTKR2022016326-appb-img-000048
제7항에 있어서, 상기 퇴행성 뇌질환은 알츠하이머병, 파킨슨병, 루게릭병, 픽크병, 크로이츠펠트-야콥병, 헌팅턴병 및 치매로 구성된 군에서 선택되는 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물.The method of claim 7, wherein the degenerative brain disease is selected from the group consisting of Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, Pick's disease, Creutzfeldt-Jakob disease, Huntington's disease, and dementia. composition. 제7항에 있어서, 상기 퇴행성 뇌질환은 알츠하이머병인 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물.The pharmaceutical composition for preventing or treating degenerative brain disease according to claim 7, wherein the degenerative brain disease is Alzheimer's disease. 제7항에 있어서, BACE1 발현을 억제하는 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물.The pharmaceutical composition for the prevention or treatment of degenerative brain diseases according to claim 7, wherein the BACE1 expression is inhibited. 제7항에 있어서, 약학적으로 허용되는 담체, 부형제 또는 희석제를 추가로 포함하는 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 치료용 약학 조성물.The pharmaceutical composition for preventing or treating degenerative brain disease according to claim 7, further comprising a pharmaceutically acceptable carrier, excipient or diluent. 화학식 1로 표시되는 라말린 유도체를 유효성분으로 함유하는 퇴행성 뇌질환의 예방 또는 개선용 식품 조성물:A food composition for preventing or alleviating degenerative brain diseases containing a ramalin derivative represented by Formula 1 as an active ingredient: [화학식 1][Formula 1]
Figure PCTKR2022016326-appb-img-000049
Figure PCTKR2022016326-appb-img-000049
화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬, C3-C6 사이클로알킬, 할로겐, 하이드록시, 카복실, 아미노 또는 시아노이고, 단, R1, R2, R3, R4 및 R5는 동시에 수소원자이거나 R1, R2, R3 및 R4는 수소원자이고, R5는 하이드록시인 경우는 제외된다.In Formula 1, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 alkyl, C 3- C 6 cycloalkyl, halogen, hydroxy, carboxyl, amino or cyano , provided that R 1 , R 2 , R 3 , R 4 and R 5 are both hydrogen atoms or R 1 , R 2 , R 3 and R 4 are hydrogen atoms and R 5 is hydroxy.
제14항에 있어서, 화학식 1에서 R1, R2, R3, R4 및 R5는 각각 독립적으로 수소원자, C1-C6 알킬 또는 할로겐인 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 개선용 식품 조성물.The method of claim 14, wherein in Formula 1, R 1 , R 2 , R 3 , R 4 and R 5 are each independently a hydrogen atom, C 1- C 6 Alkyl or halogen to prevent or improve degenerative brain diseases, characterized in that food composition for use. 제14항에 있어서, 화학식 2 내지 화학식 10에서 선택되는 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 개선용 식품 조성물:[Claim 15] The food composition for preventing or improving degenerative brain diseases according to claim 14, which is selected from Formulas 2 to 10: [화학식 2][Formula 2]
Figure PCTKR2022016326-appb-img-000050
Figure PCTKR2022016326-appb-img-000050
[화학식 3][Formula 3]
Figure PCTKR2022016326-appb-img-000051
Figure PCTKR2022016326-appb-img-000051
[화학식 4][Formula 4]
Figure PCTKR2022016326-appb-img-000052
Figure PCTKR2022016326-appb-img-000052
[화학식 5][Formula 5]
Figure PCTKR2022016326-appb-img-000053
Figure PCTKR2022016326-appb-img-000053
[화학식 6][Formula 6]
Figure PCTKR2022016326-appb-img-000054
Figure PCTKR2022016326-appb-img-000054
[화학식 7][Formula 7]
Figure PCTKR2022016326-appb-img-000055
Figure PCTKR2022016326-appb-img-000055
[화학식 8][Formula 8]
Figure PCTKR2022016326-appb-img-000056
Figure PCTKR2022016326-appb-img-000056
[화학식 9][Formula 9]
Figure PCTKR2022016326-appb-img-000057
Figure PCTKR2022016326-appb-img-000057
[화학식 10][Formula 10]
Figure PCTKR2022016326-appb-img-000058
Figure PCTKR2022016326-appb-img-000058
제14항에 있어서, 상기 퇴행성 뇌질환은 알츠하이머병, 파킨슨병, 루게릭병, 픽크병, 크로이츠펠트-야콥병, 헌팅턴병 및 치매로 구성된 군에서 선택되는 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 개선용 식품 조성물.The food for preventing or improving degenerative brain disease according to claim 14, wherein the degenerative brain disease is selected from the group consisting of Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, Pick's disease, Creutzfeldt-Jakob disease, Huntington's disease and dementia. composition. 제14항에 있어서, 식품학적으로 허용되는 첨가제, 부형제 또는 향미제를 추가로 포함하는 것을 특징으로 하는 퇴행성 뇌질환의 예방 또는 개선용 식품 조성물.[Claim 15] The food composition for preventing or improving degenerative brain disease according to claim 14, further comprising a food-acceptable additive, excipient or flavoring agent.
PCT/KR2022/016326 2021-11-15 2022-10-25 Novel ramalin derivative and use thereof for prevention or treatment of neurodegenerative diseases Ceased WO2023085645A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020210156546A KR20230070702A (en) 2021-11-15 2021-11-15 Novel Ramalin Derivatives and Their Uses for Prevention or Treatment of Degenerative Brain Diseases
KR10-2021-0156546 2021-11-15

Publications (1)

Publication Number Publication Date
WO2023085645A1 true WO2023085645A1 (en) 2023-05-19

Family

ID=86336375

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2022/016326 Ceased WO2023085645A1 (en) 2021-11-15 2022-10-25 Novel ramalin derivative and use thereof for prevention or treatment of neurodegenerative diseases

Country Status (2)

Country Link
KR (1) KR20230070702A (en)
WO (1) WO2023085645A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3274232A (en) * 1961-08-04 1966-09-20 Upjohn Co Arylhydrazides
WO1992007562A1 (en) * 1990-10-29 1992-05-14 The Children's Medical Center Corporation Controlling glutamine/glutamate related neuronal injury
KR20120007130A (en) * 2010-07-14 2012-01-20 한국해양연구원 Synthesis of Ramalin
KR20130085306A (en) * 2012-01-19 2013-07-29 한국해양과학기술원 Synthesis method of ramalin and ramalin analogues using glutamic acid derivative and hydroxy aniline or hydroxy aniline with protected hydroxy group
JP2017533261A (en) * 2014-10-24 2017-11-09 コリア インスティテュート オブ オーシャン サイエンス アンド テクノロジーKorea Institute Of Ocean Science And Technology Composition for preventing or treating degenerative brain disease containing lamarine
KR20190048846A (en) * 2017-10-31 2019-05-09 재단법인 유타 인하 디디에스 및 신의료기술개발 공동연구소 Glutamic acid derivatives and a composition comprising the same

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3274232A (en) * 1961-08-04 1966-09-20 Upjohn Co Arylhydrazides
WO1992007562A1 (en) * 1990-10-29 1992-05-14 The Children's Medical Center Corporation Controlling glutamine/glutamate related neuronal injury
KR20120007130A (en) * 2010-07-14 2012-01-20 한국해양연구원 Synthesis of Ramalin
KR20130085306A (en) * 2012-01-19 2013-07-29 한국해양과학기술원 Synthesis method of ramalin and ramalin analogues using glutamic acid derivative and hydroxy aniline or hydroxy aniline with protected hydroxy group
JP2017533261A (en) * 2014-10-24 2017-11-09 コリア インスティテュート オブ オーシャン サイエンス アンド テクノロジーKorea Institute Of Ocean Science And Technology Composition for preventing or treating degenerative brain disease containing lamarine
KR20190048846A (en) * 2017-10-31 2019-05-09 재단법인 유타 인하 디디에스 및 신의료기술개발 공동연구소 Glutamic acid derivatives and a composition comprising the same

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
KIM TAI KYOUNG, HONG JU-MI, KIM KYUNG HEE, HAN SE JONG, KIM IL-CHAN, OH HYUNCHEOL, YIM JOUNG HAN: "Potential of Ramalin and Its Derivatives for the Treatment of Alzheimer’s Disease", MOLECULES, vol. 26, no. 21, pages 6445, XP093066133, DOI: 10.3390/molecules26216445 *
KINOSHITA, T. ET AL.: "The Sythesis of Anthglutin and Its Analogues", BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN, CHEMICAL SOCIETY OF JAPAN,NIPPON KAGAKUKAI, JP, vol. 54, 1 January 1981 (1981-01-01), JP , pages 2219 - 2220, XP008147669, ISSN: 0009-2673, DOI: 10.1246/bcsj.54.2219 *
SUVARNA H. PAGIRE, EUNHYE LEE, HAUSHABHAU S. PAGIRE, EUN JUNG BAE, SOO JUNG RYU, DAHYE LEE, MIN HEE KIM, GEUM RAN KIM, KYU-SEOK HW: "Design, synthesis and biological evaluation of glutamic acid derivatives as anti-oxidant and anti-inflammatory agents", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, ELSEVIER, AMSTERDAM NL, vol. 28, no. 3, 1 February 2018 (2018-02-01), Amsterdam NL , pages 529 - 532, XP055613860, ISSN: 0960-894X, DOI: 10.1016/j.bmcl.2017.11.012 *

Also Published As

Publication number Publication date
KR20230070702A (en) 2023-05-23

Similar Documents

Publication Publication Date Title
WO2011155803A2 (en) Novel compound having hearing protection effects
WO2020122391A1 (en) Composition for preventing or treating cellular senescence-associated diseases, containing homoharringtonine as active ingredient
WO2018106002A1 (en) Composition for preventing and treating male infertility, containing compound combination comprising flavonoid derivative and iridoid derivative as active ingredient, and use thereof
WO2020218720A1 (en) Composition for preventing or treating muscular disorders or improving muscular functions, containing leonurus japonicus extract or leonurine
WO2023214836A1 (en) Novel berberine analogue and use thereof
WO2023085645A1 (en) Novel ramalin derivative and use thereof for prevention or treatment of neurodegenerative diseases
WO2021225363A1 (en) Anti-inflammatory or antidiabetic composition comprising metabolite of marine-drived fungus penicillium glabrum sf-7123
WO2022005201A1 (en) Composition for preventing, ameliorating, or treating diseases caused by nitration of tyrosine in protein, containing tyrosine as active ingredient
WO2013122344A1 (en) Pharmaceutical composition containing as active ingredient extract from bark of liriodendron tulipifera
WO2021034111A1 (en) Novel sesquiterpene derivative and use thereof
WO2013025004A2 (en) Pharmaceutical composition for preventing or treating cancer comprising radix lithospermi seu arnebiae extract as an active ingredient
WO2022182141A1 (en) Composition, for preventing, relieving, or treating disease caused by nitration of protein, containing peptide having tyrosine located at terminal as active ingredient
WO2018105998A1 (en) Composition for preventing and treating male infertility, containing flavonoid derivative compound as active ingredient, and use thereof
WO2013100270A1 (en) Pharmaceutical composition for the prevention or treatment cancer, apoptosis inducing agent, and health functional food including aruncus dioicus var. kamtschaticus extract or aruncin b as an active ingredient
WO2022235057A1 (en) Pharmaceutical composition for prevention, alleviation, or treatment of cancer
WO2023090479A1 (en) Novel transglutaminase2 inhibitor and use thereof
WO2024242530A1 (en) Compound for neuroprotection, or treating diseases related to alpha-synuclein and/or neuro-inflammation
WO2022270760A1 (en) Method for treating non-alcoholic steatohepatitis through co-administration of curcumin derivative and tgf-β receptor inhibitor
WO2022220512A1 (en) Selective mtorc2 inhibitor and uses thereof
WO2020101302A1 (en) Anticancer composition
WO2015072667A1 (en) Pharmaceutical composition for preventing or treating degenerative cranial nerve diseases
WO2021086101A1 (en) Use of novel compound, for preventing, improving or treating amyotrophic lateral sclerosis
WO2020122392A1 (en) Composition for preventing or treating cellular senescence-related diseases comprising zotarolimus as active ingredient
WO2025075465A1 (en) Novel compound derived from myristica fragrans, and anticancer composition comprising same
WO2024162814A1 (en) Novel bicyclic compound 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: 22893068

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 22893068

Country of ref document: EP

Kind code of ref document: A1