US20190290617A1 - Novel use of sesquiterpene derivative - Google Patents

Novel use of sesquiterpene derivative Download PDF

Info

Publication number
US20190290617A1
US20190290617A1 US16/301,583 US201616301583A US2019290617A1 US 20190290617 A1 US20190290617 A1 US 20190290617A1 US 201616301583 A US201616301583 A US 201616301583A US 2019290617 A1 US2019290617 A1 US 2019290617A1
Authority
US
United States
Prior art keywords
bond
methyl
compound
preparation example
trimethyl
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.)
Abandoned
Application number
US16/301,583
Inventor
Sangtaek OH
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.)
Industry Academic Cooperation Foundation of Kookmin University
Original Assignee
Industry Academic Cooperation Foundation of Kookmin University
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 Industry Academic Cooperation Foundation of Kookmin University filed Critical Industry Academic Cooperation Foundation of Kookmin University
Assigned to KOOKMIN UNIVERSITY INDUSTRY ACADEMY COOPERATION FOUNDATION reassignment KOOKMIN UNIVERSITY INDUSTRY ACADEMY COOPERATION FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: OH, Sangtaek
Publication of US20190290617A1 publication Critical patent/US20190290617A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/423Oxazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/01Hydrocarbons
    • A61K31/015Hydrocarbons carbocyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/136Amines having aromatic rings, e.g. ketamine, nortriptyline having the amino group directly attached to the aromatic ring, e.g. benzeneamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/145Amines having sulfur, e.g. thiurams (>N—C(S)—S—C(S)—N< and >N—C(S)—S—S—C(S)—N<), Sulfinylamines (—N=SO), Sulfonylamines (—N=SO2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/235Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids having an aromatic ring attached to a carboxyl group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health

Definitions

  • the present disclosure relates to a novel use of a sesquiterpene derivative, more particularly to a composition for preventing, improving or treating macular degeneration or macular edema caused by vascular leakage in the eye, the composition containing a sesquiterpene derivative compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • Macular edema refers to the swelling of the macula lutea.
  • the edema is caused by fluid leaking from the retinal blood vessel. Blood is leaked from weak blood vessel walls and flown into the macula lutea packed with retinal cones which sense color and are responsible for vision during the day. Then, images are blurred at the center or right side of the central region. The vision worsens gradually over several months. All age-related macular degeneration (AMD) is associated with macular edema.
  • AMD age-related macular degeneration
  • Vascular leakage in the eye occurs due to various causes. For example, continued increase in blood pressure in in hypertensive patients causes breakdown of the blood-retinal barrier and the damage to the blood-retinal barrier causes retinal edema due to vascular leakage.
  • the macula lutea is often damaged by macula tumentia following the removal of the eye lens for treatment of cataract.
  • Laser photocoagulation alleviates macula tumentia by blocking the blood vessels where fluid leakage occurs.
  • care should be taken to avoid the fovea when irradiating laser because it is extremely vulnerable. If the fovea is damaged during the operation, the central vision may be impaired.
  • more than one laser treatments are often necessary to remove the swelling.
  • Vitrectomy is employed when the laser treatment is ineffective, but this method is often associated with the high risk of tissue invasion which causes postoperative complications.
  • the intravitreal administration of steroids may cause ocular hypertension, steroid-induced glaucoma and posterior subcapsular cataract. In addition, the intravitreal administration of steroids often cause postoperative complications.
  • the inventors of the present disclosure have researched to develop a medication exhibiting an excellent therapeutic effect for macular edema or macular degeneration related thereto, which reduces the inconvenience of administration and can be administered for a long time. In doing so, they have identified that a compound represented by Chemical Formula 1 exhibits therapeutic effect in a macular edema or macular degeneration animal model by effectively preventing vascular leakage in the eye and that the compound is targeted to the eye even when it is administered orally.
  • the present disclosure is directed to providing a pharmaceutical composition for preventing or treating macular degeneration or macular edema caused by vascular leakage in the eye.
  • the present disclosure is also directed to providing a food composition for preventing or improving macular degeneration or macular edema caused by vascular leakage in the eye.
  • the present disclosure provides a pharmaceutical composition for preventing or treating macular degeneration or macular edema, which contains a compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient:
  • R 1 is H or CH 3 ,
  • R 3 is a functional group selected from a group consisting of R 3a through R 3d ,
  • each of R 4 and R 7 is OH or OCH 3 and R 5 , R 6 and R 8 are H; or
  • R 5 is COOCH 3 , R 7 is H or OH, R 8 is OH and R 4 and R are H,
  • R 9 is a functional group selected from a group consisting of H, NH 2 , C 1 -C 8 alkoxy and R 9a through R 9j and R 10 is H or OH,
  • each of R 11 and R 12 is OH or OAc and R 13 is H; or
  • each of R 11 and R 12 is OH or OCH 3 and R 13 is CH 3 and
  • R 14 is OCH 3 and R 15 and R 16 are CH 3 .
  • the present disclosure also provides a pharmaceutical composition for inhibiting vascular leakage in the eye, which contains the compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient
  • the present disclosure also provides a food composition for preventing or improving macular degeneration or macular edema, which contains the compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • FIG. 1 shows a result of investigating the inhibitory effect of a compound of the present disclosure (Preparation Example 30 or Preparation Example 31) on 3-catenin expression in HEK293 cells, in which the Wnt/ ⁇ -catenin pathway is activated by treating with Wnt-3a CM, by western blot.
  • FIG. 2 shows a result of investigating the inhibitory effect of a compound of the present disclosure (Preparation Example 32) on 3-catenin expression in HEK293 cells, in which the Wnt/ ⁇ -catenin pathway is activated by treating with Wnt-3a CM, by western blot.
  • FIG. 3 shows a result of investigating the inhibitory effect of a compound of the present disclosure (Preparation Example 33 or Preparation Example 34) on 3-catenin expression in human retinal epithelial cells, in which the Wnt/3-catenin pathway is activated by treating with Wnt-3a CM, by western blot.
  • FIG. 4 shows a result of investigating the inhibitory effect of intravitreal administration of a compound of the present disclosure (Preparation Example 33) on vascular leakage in a macular edema mouse model by fluorescein angiography and optical coherence tomography (A and B are images for a control group (compound-untreated) obtained after DMSO injection and C and D are images for a test group (Preparation Example 33) obtained after injection.
  • the arrows indicate blood vessels).
  • FIG. 5 shows a result of investigating the inhibitory effect of intraperitoneal injection of a compound of the present disclosure (Preparation Example 33) on vascular leakage in a macular edema mouse model by optical coherence tomography (A: vehicle-administered group, B: Preparation Example 33 compound 1 mg/kg administered group).
  • FIG. 6 shows a result of orally administering a compound of the present disclosure (Preparation Example 33) to an ICR mouse and measuring distribution of the compound in target tissues (particularly, eye).
  • the inventors of the present disclosure have researched to develop a medication exhibiting an excellent therapeutic effect for macular edema or macular degeneration related thereto, which reduces the inconvenience of administration and can be administered for a long time. In doing so, they have identified that a compound represented by Chemical Formula 1 exhibits therapeutic effect for macular edema or macular degeneration diseases by effectively preventing vascular leakage in the eye, particularly in the retina.
  • the present disclosure relates to a pharmaceutical composition for preventing or treating macular degeneration or macular edema, which contains a compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • R 1 is H or CH 3 ,
  • R 3 is a functional group selected from a group consisting of R 3a through R 3d ,
  • each of R 4 and R 7 is OH or OCH 3 and R 5 , R 6 and R 8 are H; or
  • R 5 is COOCH 3 , R 7 is H or OH, R 8 is OH and R 4 and R 6 are H,
  • R 9 is a functional group selected from a group consisting of H, NH 2 , C 1 -C 8 alkoxy and R 9a through R 9j and R 10 is H or OH,
  • each of R 11 and R 12 is OH or OAc and R 13 is H; or
  • each of R 11 and R 12 is OH or OCH 3 and R 13 is CH 3 and
  • R 14 is OCH 3 and R 15 and R 16 are CH 3 .
  • the term alkoxy group refers to an alkyl group bonded to oxygen (O-alkyl group).
  • the alkoxy group may be a C 1 -C 8 alkoxy group selected from a group consisting of a methoxy group (C 1 ), an ethoxy group (C 2 ), a propoxy group (C 3 ), a butoxy group (C 4 ), a pentyloxy group (C 5 ), a hexyloxy group (C 6 ), a heptyloxy group (C 7 ) and an octyloxy group (C 8 ), although not being limited thereto.
  • the alkoxy group of the present disclosure may be a methoxy group or an ethoxy group.
  • the compound of the present disclosure of Chemical Formula 1 may be the compounds described in [Table 1], although not being limited thereto.
  • the sesquiterpene derivative compound of the present disclosure may be one wherein, in Chemical Formula 1, if the bond between C-3 and C-4 and the bond between C-5 and C-6 are single bonds, R 2b is nonexistent, R 2a is CH 2 and R 3 is a functional group selected from a group consisting of R 3b through R 3d .
  • R 9 may be selected from a group consisting of ethoxy, methoxy and R 9a .
  • R 11 may be OH
  • R 12 may be OCH 3
  • R 13 may be CH 3 .
  • R 14 may be OCH 3 and R 15 and R 16 may be CH 3 .
  • the compound of Chemical Formula 1 of the present disclosure may be a compound selected from a group consisting of:
  • the compound of Chemical Formula 1 of the present disclosure may be extracted from sponge.
  • the compound of Chemical Formula 1 for preventing or treating macular degeneration or macular edema caused by vascular leakage in the eye may be obtained by a method including a step of extracting one or more sponge selected from a group consisting of Rhopaloeides sp., Spongia sp., Smenospongia sp., Hippospongia sp., Dactylospongia sp., Verongula sp., Dysidea sp., sponge SS-1047, sponge SS-265 and sponge SS-1208 by adding a C 1 -C 6 organic solvent.
  • the C 1 -C 6 organic solvent may be selected from a group consisting of a C 1 -C 6 alcohol (methanol, ethanol, propanol, butanol, pentanol, hexanol), acetone, an ether, benzene, chloroform, ethyl acetate, methylene chloride, hexane, cyclohexane, acetonitrile, dichloromethane and petroleum ether.
  • a C 1 -C 6 alcohol methanol, ethanol, propanol, butanol, pentanol, hexanol
  • acetone an ether
  • benzene chloroform
  • ethyl acetate methylene chloride
  • hexane hexane
  • cyclohexane acetonitrile
  • dichloromethane and petroleum ether.
  • the compound of the present disclosure of Chemical Formula 1 may be obtained by a method including: a step of extracting sponge by adding water, a C 1 -C 4 alcohol or a mixture thereof as a solvent, thereby preparing a sponge extract; and a step of fractionating the extract by adding a second solvent and separating the same through chromatography.
  • any one known to those skilled in the art can be used without limitation, including silica gel column chromatography, LH-20 column chromatography, ion-exchange chromatography, medium pressure liquid chromatography, thin-layer chromatography (TLC), silica gel vacuum liquid chromatography, high-performance liquid chromatography, etc.
  • the C 1 -C 4 alcohol used to prepare the sponge extract may be selected from a group consisting of methanol, ethanol, propanol, isopropanol, butanol and isobutanol.
  • a C 1 -C 4 alcohol n-hexane, methylene chloride, acetone, chloroform, dichloromethane, ethyl acetate, acetonitrile or a mixture thereof may be used.
  • the compound of the present disclosure of Chemical Formula 1 includes a pharmaceutically acceptable salt thereof.
  • pharmaceutically acceptable means being physiologically acceptable and not causing allergic reactions such as gastroenteric trouble, dizziness, etc. or similar reactions when administered to human.
  • the pharmaceutically acceptable salt includes an acid addition salt with an inorganic acid or an organic acid.
  • an acid addition salt formed by a pharmaceutically acceptable free acid is useful.
  • an inorganic acid or an organic acid may be used.
  • the inorganic acid hydrochloric acid, bromic acid, sulfuric acid, phosphoric acid, etc. may be used.
  • organic acid citric acid, acetic acid, lactic acid, tartaric acid, fumaric acid, formic acid, propionic acid, oxalic acid, trifluoroacetic acid, methanesulfonic acid, benzenesulfonic acid, maleic acid, benzoic acid, gluconic acid, glycolic acid, succinic acid, 4-morpholinoethanesulfonic acid, camphorsulfonic acid, 4-nitrobenzenesulfonic acid, hydroxy-O-sulfonic acid, 4-toluenesulfonic acid, galacturonic acid, embonic acid, glutamic acid, aspartic acid, etc.
  • organic acid citric acid, acetic acid, lactic acid, tartaric acid, fumaric acid, formic acid, propionic acid, oxalic acid, trifluoroacetic acid, methanesulfonic acid, benzenesulfonic acid, maleic acid, benzoic acid, gluconic acid,
  • the compound of the present disclosure of Chemical Formula 1 inhibits ⁇ -catenin in vitro, suggesting that it can inhibit vascular leakage by inhibiting the Wnt/0-catenin mechanism.
  • the present disclosure compound exhibits therapeutic effect by inhibiting vascular leakage in a macular edema animal model in vivo.
  • the present disclosure relates to a pharmaceutical composition for inhibiting vascular leakage in the eye, which contains the compound of Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • vascular leakage refers to the leakage of body fluid or blood plasma due to damage to the integrity of blood vessels.
  • the vascular leakage in the eye constitutes the major pathological conditions of various eye diseases.
  • vascular leakage in the eye refers to vascular leakage in various tissues (choroid, retina, etc.) constituting the eye. Specifically, it may refer to vascular leakage in the retina, although not being limited thereto.
  • the pharmaceutical composition of the present disclosure has preventive or therapeutic effect for a disease caused by vascular leakage in the eye.
  • the disease caused by vascular leakage in the eye may be any one known in the art. For example, it include retinal degeneration, macular degeneration, retinal edema and macular edema.
  • the disease caused by vascular leakage in the eye may be macular degeneration or macular edema.
  • the pharmaceutical composition according to the present disclosure may contain only the sesquiterpene derivative compound of Chemical Formula 1 or a pharmaceutically acceptable salt thereof or may further contain one or more pharmaceutically acceptable carrier, excipient or diluent.
  • the pharmaceutically acceptable carrier may further contain, for example, a carrier for oral administration or a carrier for parenteral administration.
  • the carrier for oral administration may include lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, etc.
  • the carrier for parenteral administration may include water, suitable oils, physiological saline, water-soluble glucose, glycol, etc.
  • the pharmaceutical composition of the present disclosure may further contain a stabilizer and a preservative.
  • a suitable stabilizer includes sodium bisulfite, sodium sulfite or an antioxidant such as ascorbic acid.
  • a suitable preservative includes benzalkonium chloride, methyl- or propylparaben and chlorobutanol.
  • the pharmaceutical composition of the present disclosure may further contain a lubricant, a humectant, a sweetener, a flavorant, an emulsifier, a suspending agent, etc.
  • a lubricant for other pharmaceutically acceptable carriers, reference can be made to the literature (Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Company, Easton, Pa., 1995).
  • composition of the present disclosure may be administered to mammals including human by any means.
  • it may be administered orally or parenterally.
  • the parenteral administration method may include intravenous, intramuscular, intraarterial, intramedullary, intradural, intracardiac, intraocular, intravitreal, transdermal, subcutaneous, intraabdominal, intranasal, intraintestinal, topical, sublingual or intrarectal administration, although not being limited thereto.
  • the pharmaceutical composition of the present disclosure may be prepared into a formulation for oral administration or parenteral administration depending on the administration routes.
  • the composition of the present disclosure may be formulated into a powder, a granule, a tablet, a pill, a sugar-coated tablet, a capsule, a solution, a gel, a syrup, a slurry, a suspension, etc. using the method known in the art.
  • a tablet or a sugar-coated tablet may be prepared by mixing the active ingredient with a solid excipient, pulverizing the mixture, adding a suitable adjuvant and then processing into a granule mixture.
  • the suitable excipient may include sugars including lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, etc., a starches including corn starch, wheat starch, rice starch, potato starch, etc., celluloses including cellulose, methyl cellulose, sodium carboxymethyl cellulose, hydroxypropylmethyl cellulose, etc. and fillers such as gelatin, polyvinylpyrrolidone, etc. If necessary, crosslinked polyvinylpyrrolidone, agar, alginic acid, sodium alginate, etc. may be added as a disintegrant.
  • the pharmaceutical composition of the present disclosure may further contain an antiagglomerant, a lubricant, a humectant, a flavor, an emulsifier, an antiseptic, etc.
  • the composition may be formulated into an injection, an eye drop, an ointment, a cream, a lotion, an oil, a gel, an aerosol or a nasal inhaler using the method known in the art.
  • formulations are described in the literature generally known in the field of pharmaceutical chemistry (Remington's Pharmaceutical Science, 15th Edition, 1975. Mack Publishing Company, Easton, Pa. 18042, Chapter 87: Blaug, Seymour).
  • the pharmaceutical composition of the present disclosure may be prepared into a formulation selected from a group consisting of an oral medication, an injection, an eye drop and an ointment.
  • the total effective amount of the sesquiterpene derivative compound of the present disclosure or a pharmaceutically acceptable salt thereof may be administered to a patient with a single dose or a multiple dose according to the fractionated treatment protocol for long-term administration.
  • the content of the active ingredient of the pharmaceutical composition of the present disclosure may vary depending on the severity of a disease.
  • the effective administration dosage of the compound or a pharmaceutically acceptable salt thereof is determined in consideration of various factors including the route and number of administration of the pharmaceutical composition, the age, body weight, health condition and sex of a patient, the severity of a disease, diet, excretion rate, etc.
  • the pharmaceutical composition according to the present disclosure is not specially limited in formulation, administration route and administration method as long as the effect of the present disclosure can be achieved.
  • the present disclosure relates to a food composition for preventing or improving macular degeneration or macular edema, which contains the compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • the food composition of the present disclosure includes all forms such as a functional food, a nutritional supplement, a health food, a food additive, a feed, etc. and is provided for animals including human or livestock.
  • the food composition may be prepared into various forms according to the method known in the art.
  • the health food may be prepared by preparing the sesquiterpene derivative of the present disclosure or a sponge extract containing the sesquiterpene into tea, juice or a drink for drinking or into a granule, a capsule or a powder.
  • a composition may be prepared by mixing the sesquiterpene derivative of the present disclosure or a sponge extract containing the sesquiterpene with an active ingredient known to be effective in improving and preventing macular edema or macular degeneration.
  • the functional food be prepared by adding the sesquiterpene derivative of the present disclosure or a sponge extract containing the sesquiterpene to beverages (including alcoholic beverage, fruit or processed fruit (e.g., canned fruit, bottling, jam, marmalade, etc.), fish, meat or processed foodstuffs thereof (e.g., ham, sausage corn beef, etc.), bread or noodles (e.g., udon, buckwheat noodle, instant noodle, spaghetti, macaroni, etc.), fruit juice, drinks, cookies, taffy, dairy products (e.g., butter, cheese, etc.), vegetable fats and oils, margarine, vegetable proteins, retort foods, frozen foods, condiments (e.g., soybean paste, soy sauce, etc.), etc.
  • beverages including alcoholic beverage, fruit or processed fruit (e.g., canned fruit, bottling, jam, marmalade, etc.), fish, meat or processed foodstuffs thereof (e.g., ham, sausage corn
  • sesquiterpene derivative of the present disclosure or a sponge extract containing the sesquiterpene may be prepared into a powder or a concentrate for use as a food additive.
  • the obtained compound of Preparation Example 1 had the following physicochemical properties and was identified as ‘methyl 3-[[(1R,2S,4aR,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4,5-dihydroxybenzoate’.
  • the obtained compound of Preparation Example 2 had the following physicochemical properties and was identified as ‘3-[[(1S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-2,5-dihydroxycyclohexa-2,5-diene-1,4-dione’.
  • the obtained compound of Preparation Example 3 had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(3-methylbutylamino)cyclohexa-3,5-diene-1,2-dione’.
  • a compound of Preparation Example 4 was prepared in the same manner as in Preparation Example 3.
  • the obtained compound (Preparation Example 4) had the following physicochemical properties and was identified as ‘3-[[(1S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(2-methylpropylamino)cyclohexa-3,5-diene-1,2-dione’.
  • MeOH and DCM crude extracts of Hippospongia sp. were combined and fractionated with MeOH, DCM, hexane and BuOH. Among them, the hexane, DCM and MeOH fractions were subjected to flash column chromatography and semi-preparative RP-HPLC to obtain a compound of Preparation Example 5.
  • the obtained compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-5-amino-4-hydroxycyclohexa-3,5-diene-1,2-dione’.
  • the prepared Preparation Example 6 compound had the following physicochemical properties and was identified as ‘2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]acetic acid’.
  • a compound of Preparation Example 7 was prepared in the same manner as in Preparation Example 6.
  • the prepared compound (Preparation Example 7) had the following physicochemical properties and was identified as ‘3-[[5-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]propanoic acid’.
  • a compound of Preparation Example 8 was prepared in the same manner as in Preparation Example 6.
  • the prepared compound (Preparation Example 8) had the following physicochemical properties and was identified as ‘7-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-1,3-benzoxazole-5,6-diol’.
  • the obtained compound of Preparation Example 9 (2.3 mg, 0.007%) had the following physicochemical properties and was identified as ‘[7-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-6-acetyloxy-1,3-benzoxazol-5-yl] acetate’.
  • a sponge (Spongiidae SS-1047, 0.30 kg, wet weight) was obtained as described in the literature ‘Yohei Takahashi et al., 2010’ and extracted. Briefly, EtOAc-soluble substances (1.2 g) were fractionated using a silica gel column (n-hexane/EtOAc) and a fraction 1, a fraction 2 and a fraction 3 of low polarity and a polar fraction 4 were prepared.
  • the fraction 3 was fractionated and purified by C 18 column (MeOH/H 2 O) and C 18 HPLC (Luna 5u Phenyl-Hexyl, 250 ⁇ 10 mm; eluent, MeOH/H 2 O/CF 3 CO 2 H, 85:15:0.05; flow rate, 2.5 mL/min; UV detection at 320 nm) to obtain compounds of Preparation Example 10 and Preparation Example 11.
  • the obtained Preparation Example 10 compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-2-hydroxy-5-methoxycyclohexa-2,5-diene-1,4-dione’.
  • a compound of Preparation Example 11 was prepared in the same manner as in Preparation Example 10.
  • the obtained compound (Preparation Example 11) had the following physicochemical properties and was identified as ‘2-hydroxy-5-methoxy-3-[[(1R,2S)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]cyclohexa-2,5-diene-1,4-dione’.
  • the fraction 1 obtained in Preparation Example 10 was refractionated by Cis column (MeOH/H 2 O) and C 18 HPLC (Wakosil-II 5C18AR, Wako Pure Chemical Industries, Ltd., 250 ⁇ 10 mm; eluent, MeCN/H 2 O/CF 3 CO 2 H, 90:10:0.05; flow rate, 2.0 mL/min; UV detection at 300 nm) to obtain a compound of Preparation Example 11 (2.8 mg, 0.00093% wet weight) and a compound of Preparation Example 12 (24.7 mg, 0.0082%).
  • the obtained Preparation Example 12 compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,8aS)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione’.
  • a compound of Preparation Example 13 was prepared in the same manner as in Preparation Example 12.
  • the obtained compound (Preparation Example 13) had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione’.
  • the fraction 2 obtained in Preparation Example 10 was subjected to C 18 column (MeOH/H 2 O/CF 3 CO 2 H) and C 18 HPLC (Luna 5u Phenyl-Hexyl, Phenomenex, 250 ⁇ 10 mm; eluent, MeCN/H 2 O/CF 3 CO 2 H, 80:20:0.05; flow rate, 2.0 mL/min; UV detection at 300 nm) to obtain a compound of Preparation Example 14 (0.9 mg, 0.00030%), a fraction y and a fraction 6.
  • the obtained compound of Preparation Example 14 had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(2-methylpropylamino)cyclohexa-3,5-diene-1,2-dione’.
  • the fraction ⁇ obtained in Preparation Example 14 was purified by C 18 HPLC (Luna 5u C18(2), Phenomenex, 250 ⁇ 10 mm; MeOH/H 2 O/Et 2 NH, 70:30:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm) to obtain a compound of Preparation Example 15 (1.4 mg, 0.00047%) and a compound of Preparation Example 16 (4.0 mg, 0.0013%).
  • the compound of Preparation Example 15 had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-[[(2S)-2-methylbutyl]amino]cyclohexa-3,5-diene-1,2-dione’.
  • a compound of Preparation Example 16 was prepared in the same manner as in Preparation Example 15. It had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(3-methylbutylamino)cyclohexa-3,5-diene-1,2-dione’.
  • the fraction 6 obtained in Preparation Example 14 was refractionated by Cis HPLC (Luna 5u Phenyl-Hexyl, 250 ⁇ 10 mm; MeOH/H 2 O/Et 2 NH, 65:35:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm) to obtain a compound of Preparation Example 17 (0.7 mg, 0.00023%) and a compound of Preparation Example 18 (1.6 mg, 0.00053%).
  • the obtained Preparation Example 17 compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,8aS)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-[[(2S)-2-methylbutyl]amino]cyclohexa-3,5-diene-1,2-dione’.
  • a compound of Preparation Example 18 was prepared in the same manner as in Preparation Example 17. It had the following physicochemical properties and was identified as ‘3-[[(1R,2S,8aS)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(3-methylbutylamino)cyclohexa-3,5-diene-1,2-dione’ Compound
  • a sponge (Spongiidae SS-265) was obtained as described in the literature ‘Yohei Takahashi et al., 2010’ and extracted. Briefly, an extract was prepared by adding MeOH (4.3 and 3.2 L) to sponge SS-265 (1.4 kg, wet weight). The MeOH extract (68.4 g) was fractionated with CHCl 3 and H 2 O.
  • the CHCl 3 -soluble substances (2.3 g) were subjected to silica gel column (n-hexane/EtOAc), C 18 column (MeOH/H 2 O), silica gel column (n-hexane/acetone) and C 18 HPLC (Wakosil-II 5C18AR, 250 ⁇ 10 mm; eluent, MeCN/H 2 O/CF 3 CO 2 H, 90:10:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm and Luna 5u C18(2), 250 ⁇ 10 mm; MeOH/H 2 O/Et 2 NH, 70:30:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm) repeatedly to obtain a compound of Preparation Example 19 (1.8 mg, 0.00013%).
  • the obtained Preparation Example 19 compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-[[(2S)-2-methylbutyl]amino]cyclohexa-3,5-diene-1,2-dione’.
  • a sponge (Spongiidae SS-1208) was obtained as described in the literature ‘Yohei Takahashi et al., 2010’ and extracted. Briefly, an extract was prepared by adding MeOH (3 ⁇ 0.8 L) and MeOH/toluene (3:1, 1 ⁇ 0.8 L) to sponge SS-1208 (0.4 kg, wet weight). The extract mixture (15.9 g) was fractionated with CHCl 3 and H 2 O (3 ⁇ 500 mL).
  • the CHCl 3 -soluble fraction (2.7 g) was subjected to silica gel column (n-hexane/EtOAc and CHCl 3 /MeOH), C 18 column (MeOH/H 2 O/CF 3 CO 2 H) and C 18 HPLC (Luna 5u Phenyl-Hexyl, 250 ⁇ 10 mm; eluent, MeCN/H 2 O/CF 3 CO 2 H, 70:30:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm and Wakosil-II 5C18AR, 250 ⁇ 10 mm; eluent, MeCN/H 2 O/CF 3 CO 2 H, 75:25:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm) repeatedly to obtain a compound of Preparation Example 20 (0.8 mg, 0.00020%).
  • the obtained Preparation Example 20 compound had the following physicochemical properties and was identified as ‘2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]ethanesulfonic acid’.
  • the obtained Preparation Example 21 compound had the following physicochemical properties and was identified as ‘methyl 3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxybenzoate’.
  • a compound of Preparation Example 22 was prepared in the same manner as in Preparation Example 21. It had the following physicochemical properties and was identified as ‘methyl 3-[[(1 S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4,5-dihydroxybenzoate’.
  • a heated ylide solution was added dropwise to a solution of 212 mg (0.62 mmol) of the ketone ( ⁇ )-(1R,4aS,8aS)-1 ⁇ ,2 ⁇ , 4a ⁇ -trimethyl-1 ⁇ [(2′,5′-dimethoyphenyl)methyl]-1,2,3,4,4a,5,6,7,8,8a ⁇ -decahydronaphthalen-5-one dissolved in 3 mL of benzene. After heat-treating further for 22 hours, the reaction mixture was cooled and diluted by stirring fast while sequentially adding 10 mL of ether and 3 mL of H 2 O.
  • the obtained Preparation Example 23 compound had the following physicochemical properties and was identified as ‘( ⁇ )-(1R,4aS,8aS)-1 ⁇ ,2 ⁇ ,4 ⁇ ,-trimethyl-1 ⁇ [(2′,5′-dimethoxyphenyl)methyl]-5-exo-methylene-(3H)-1, 4,4a,5,6,7,8,8a ⁇ -octahydronaphthalene’.
  • the obtained Preparation Example 28 compound had the following physicochemical properties and was identified as ‘(2S)-2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]-3-hydroxypropanoic acid’.
  • the obtained Preparation Example 29 compound had the following physicochemical properties and was identified as ‘(2S)-2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]-3-hydroxybutanoic acid’.
  • the fraction 10 (39.3 g) was fractionated further with hexane-acetone mixtures (95:5, 90:10, 85:15, 80:20), MeOH (100%) and MeOH—H 2 O (50:50) using a silica gel VLC (12 (H) ⁇ 17.5 (D) cm) into 9 fractions (Fr. 10-1 to 10-9).
  • the fraction 10-7 (3.7 g) was subjected to C 18 MPLC (15.5 ⁇ 4 cm) under an isocratic condition of MeOH—H 2 O (85:15) to prepare 6 subfractions (Fr. 10-7-1 to 10-7-6).
  • the fraction 10-7-3 (115.8 mg) was subjected to C 18 HPLC (250 ⁇ 21.20 mm, 10 ⁇ m) chromatography using MeOH—H 2 O (83:17) to prepare 3 fractions (Fr. 10-7-3-1 to 10-7-3-3).
  • the fraction 10-7-3-2 (12.4 mg) was subjected to C 18 HPLC (250 ⁇ 4.60 mm and 150 ⁇ 4.60 mm, 5 ⁇ m, connected in line) using MeOH—H 2 O (75:25) to obtain a compound of Preparation Example 30 and an epimer mixture thereof.
  • the obtained substance had the following physicochemical properties and was identified as ‘18-methoxy-22,22-dimethyl-16-[ ⁇ (5R,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl ⁇ methyl]benzo[d]-oxazol-17(2H)-one’.
  • the obtained compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione’.
  • the compound was identified by HPLC (Agilent Technologies 1260 Infinity) using a UV spectrophotometer (203 nm) and a Bluespher AB2 (150 ⁇ 2 mm) column.
  • HPLC was conducted at a flow rate of 1 mL/min and 40° C. using water-methanol (78:22) as a mobile phase and the peaks of the compound were detected at 114 minutes.
  • the obtained compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-2-hydroxy-5-methoxycyclohexa-2,5-diene-1,4-dione’.
  • the obtained final compound had the following physicochemical properties and was identified as ‘3-[[(1S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-5-ethoxy-2-hydroxycyclohexa-2,5-diene-1,4-dione’.
  • HEK293 cells human embryonic kidney cells
  • Wnt3a-secreting L cells were obtained from the ATCC (American Type Culture Collection, USA) and were cultured in DMEM (Dulbecco's modified Eagle's medium) supplemented with 10% FBS (fetal bovine serum), 120 ⁇ g/mL penicillin and 200 ⁇ g/mL streptomycin.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • streptomycin fetal bovine serum
  • Wnt3a-CM Wnt3a-conditioned medium
  • DMEM fetal bovine serum
  • FBS fetal bovine serum
  • the compound of Preparation Example 30 or the compound of Preparation Example 31 (10, 20 or 40 ⁇ M) for 15 hours and extracting cytoplasmic proteins from the cells
  • the amount of ⁇ -catenin regulating the CRT ( ⁇ -catenin response transcription) of the Wnt/ ⁇ -catenin pathway in the cells was investigated by western blot using a ⁇ -catenin antibody (BD Transduction Laboratories, USA) and the ECL system (Santa Cruz Biotechnology). The result is shown in FIG. 1 .
  • the cells treated with the Wnt3a-CM showed increased 3-catenin expression in the cytoplasm but the cells treated with the Preparation Example 30 compound or the Preparation Example 31 compound of the present disclosure showed decreased level of ⁇ -catenin.
  • the Wnt/1-Catenin Pathway Inhibitory Activity of the Preparation Example 32 Compound was evaluated in the same manner as in Example ⁇ 1-1>. Briefly, after treating HEK293 cells with the Wnt3a-CM or the compound of Preparation Example 32 (10 or 20 ⁇ M) for 15 hours and extracting cytoplasmic proteins from the cells, the amount of ⁇ -catenin regulating the CRT ( ⁇ -catenin response transcription) of the Wnt/ ⁇ -catenin pathway in the cells was investigated by western blot using a ⁇ -catenin antibody (BD Transduction Laboratories, USA) and the ECL system (Santa Cruz Biotechnology). The result is shown in FIG. 2 .
  • a ⁇ -catenin antibody BD Transduction Laboratories, USA
  • ECL system Santa Cruz Biotechnology
  • the cells treated with the Wnt3a-CM showed increased 3-catenin expression in the cytoplasm but the cells treated with the Preparation Example 32 compound of the present disclosure showed decreased level of ⁇ -catenin.
  • ARPE-19 cells human retinal epithelial cells
  • Wnt3a-secreting L cells were obtained from the ATCC (American Type Culture Collection, USA) and were cultured in DMEM (Dulbecco's modified Eagle's medium) supplemented with 10% FBS, 120 ⁇ g/mL penicillin and 200 ⁇ g/mL streptomycin.
  • Wnt3a-CM Wnt3a-conditioned medium
  • DMEM fetal bovine serum
  • FBS fetal bovine serum
  • the compound of Preparation Example 33 or the compound of Preparation Example 34 (3 or 6 ⁇ M) for 24 hours and extracting cytoplasmic proteins from the cells
  • the amount of ⁇ -catenin regulating the CRT ( ⁇ -catenin response transcription) of the Wnt/3-catenin pathway in the cells was investigated by western blot using a ⁇ -catenin antibody (BD Transduction Laboratories, USA) and the ECL system (Santa Cruz Biotechnology). The result is shown in FIG. 3 .
  • the cells treated with the Wnt3a-CM showed increased 3-catenin expression in the cytoplasm but the cells treated with the Preparation Example 33 compound or Preparation Example 34 compound of the present disclosure showed decreased level of ⁇ -catenin, suggesting that the compounds inhibit the Wnt/0-catenin pathway in the human retinal epithelial cells. Meanwhile, this activity was not identified in the sponge ethanol extract used to isolate the compound of Preparation Example 33 (data not shown).
  • Example 33 For the compound of Preparation Example 33 which showed good activity in Example 1, the inhibitory activity on vascular leakage, which is a cause of macular degeneration or macular edema, was investigated in a macular edema-induced mouse model.
  • Macular edema was induced in a 10-week-old C57BL/6 mouse by irradiating a laser. After anesthetizing the mouse with ketamine (70 mg/kg) and xylazine (30 mg/kg), the pupil was dilated with 1% tropicamide. Hydroxypropylmethyl cellulose was dropped onto the eye and a microscope cover glass was used as a contact lens.
  • FIG. 4 shows the images of the control group and the compound-treated group.
  • test group ( FIG. 4 , C, D) showed distinctly decreased vascular leakage, which is a cause of macular degeneration or macular edema, as compared to the control group ( FIG. 4 , A, B).
  • the compound of the present disclosure inhibited vascular leakage, which is a cause of macular degeneration or macular edema, even when it was injected intraperitoneally.
  • mice Male ICR mice (8 weeks, 30-35 g) were purchased from Samtako Co. (Osan, Korea). The test animals were acclimatized for a week under the following conditions: temperature 23 ⁇ 2° C., relative humidity 55 ⁇ 10%, illumination intensity 150-300 lux, ventilation frequency 15-20 times/h, illumination cycle 12 h (07:00-19:00). All the animal experiments were approved by the Animal Care and Use Committee of Kyungpook National University (Study No. 2016-0043).
  • mice were fasted for 12 hours before drug administration. Feed and water were supplied ad libitum.
  • blood samples were taken from the abdominal artery. 50 mL of a plasma sample obtained after centrifuging the blood sample at 13,000 rpm for 5 minutes was stored at ⁇ 80° C. until use for analysis. An eye sample taken from the mouse was homogenized with 9-fold saline to obtain a 10% cell homogenate. The obtained 50-mL aliquots were stored at ⁇ 80° C. until use for analysis.
  • the compound of the present disclosure In order to investigate the distribution of the compound of the present disclosure in the target tissue, the compound was orally administered at a dose of 10 mg/kg and the concentration of the compound in the blood plasma and eye was measured. The sampling times were determined as 0.5 hour and 2 hours based on the peak plasma concentration and the distribution phase. As seen from FIG. 6 , the compound of the present disclosure (particularly, the compound of Preparation Example 33) showed high permeability for the target tissue and was found to be highly targeted in the eye even when it was administered orally. Through this result, it was confirmed that the compound of Preparation Example 33 of the present disclosure can exhibit therapeutic effect even when it is administered via different administration routes (oral administration, intraperitoneal injection, intravenous injection, etc.) other than being administered directly into the vitreous cavity. That is to say, whereas the currently available eye disease-related therapeutic agents cause inconvenience, pain and side effects because they have to be administered directly into the vitreous cavity, it was confirmed that the compound of the present disclosure can be administered orally.
  • This experiment was conducted to determine acute (within 24 hours) toxicity and lethality when the compound of Preparation Example 33 was administered in excessive amounts in a short period of time.
  • 20 normal ICR mice were divided into a control group and a test group, with 10 mice per each.
  • the control group was administered with PEG 400:Tween 80:ethanol (8:1:1, v:v:v) only and the test group was orally administered with the compound of Preparation Example 33 dissolved in PEG 400:Tween 80:ethanol (8:1:1, v:v:v).
  • lethality was investigated 24 hours after the administration, all the mice in the control group and the mice in the test group administered with the Preparation Example 33 compound at a dose of 2 g/kg/day survived.
  • a long-term toxicity test was conducted by administering the compound of Preparation Example 33 at different doses to C57BL/6J mice (10 mice per group) for 8 weeks.
  • blood was taken from the animals of the test group to which the compound of Preparation Example 33 was administered and the control group to which only PEG 400:Tween 80:ethanol (8:1:1, v:v:v) was administered 8 weeks later and the level of GPT (glutamate-pyruvate transferase) and BUN (blood urea nitrogen) in the blood was measured using Select E (Vital Scientific NV, Netherlands).
  • 200 g of the compound of the present disclosure was mixed with 175.9 g of lactose, 180 g of potato starch and 32 g of colloidal silicate. After adding a 10% gelatin solution, the mixture was pulverized and passed through a 14-mesh sieve. A mixture obtained by drying the same and adding 160 g of potato starch, 50 g of talc and 5 g of magnesium stearate thereto was prepared into a tablet.
  • the present disclosure relates to a novel use of a sesquiterpene derivative, more particularly to a composition for preventing, improving or treating macular degeneration or macular edema caused by vascular leakage in the eye, which contains the sesquiterpene derivative compound represented by Chemical Formula 1 of the present disclosure or a pharmaceutically acceptable salt thereof as an active ingredient.
  • the compound of the present disclosure of Chemical Formula 1 has therapeutic effect for a disease caused by vascular leakage in the eye, such as macular edema, macular degeneration, etc., by inhibiting the vascular leakage in the eye, particularly in the retina.
  • vascular leakage in the eye such as macular edema, macular degeneration, etc.
  • the intraocular disease-related treating agents available in the market should be injected directly into the vitreous cavity, thus causing pain and side effects
  • the sesquiterpene derivative compound of the present disclosure is delivered to the target tissue (eye) via different administration routes (oral, intraperitoneal, etc.) other than the intravitreal route. Accordingly, the sesquiterpene derivative compound provides excellent therapeutic effect without being restricted by the administration routes. Accordingly, it is highly industrially applicable.
  • the compound of the present disclosure of Chemical Formula 1 has therapeutic effect for a disease caused by vascular leakage in the eye, such as macular edema, macular degeneration, etc., by inhibiting the vascular leakage in the eye, particularly in the retina.
  • vascular leakage in the eye such as macular edema, macular degeneration, etc.
  • the intraocular disease-related treating agents available in the market should be injected directly into the vitreous cavity, thus causing pain and side effects
  • the sesquiterpene derivative compound of the present disclosure is delivered to the target tissue (eye) via different administration routes (oral, intraperitoneal, etc.) other than the intravitreal route. Accordingly, the sesquiterpene derivative compound provides excellent therapeutic effect without being restricted by the administration routes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Nutrition Science (AREA)
  • Ophthalmology & Optometry (AREA)
  • Mycology (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Emergency Medicine (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Coloring Foods And Improving Nutritive Qualities (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present disclosure relates to a novel use of a sesquiterpene derivative, more particularly to a composition for preventing, improving or treating macular degeneration or macular edema caused by vascular leakage in the eye, the composition containing a sesquiterpene derivative compound represented by Chemical Formula 1 of the present disclosure or a pharmaceutically acceptable salt thereof as an active ingredient. Whereas the intraocular disease-related treating agents available in the market should be injected directly into the vitreous cavity, thus causing pain and side effects, the sesquiterpene derivative compound of the present disclosure is delivered to the target tissue (eye) via different administration routes (oral, intraperitoneal, etc.) other than the intravitreal route. Accordingly, the sesquiterpene derivative compound provides excellent therapeutic effect without being restricted by the administration routes.

Description

    TECHNICAL FIELD
  • The present disclosure relates to a novel use of a sesquiterpene derivative, more particularly to a composition for preventing, improving or treating macular degeneration or macular edema caused by vascular leakage in the eye, the composition containing a sesquiterpene derivative compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • BACKGROUND ART
  • Macular edema refers to the swelling of the macula lutea. The edema is caused by fluid leaking from the retinal blood vessel. Blood is leaked from weak blood vessel walls and flown into the macula lutea packed with retinal cones which sense color and are responsible for vision during the day. Then, images are blurred at the center or right side of the central region. The vision worsens gradually over several months. All age-related macular degeneration (AMD) is associated with macular edema. Vascular leakage in the eye occurs due to various causes. For example, continued increase in blood pressure in in hypertensive patients causes breakdown of the blood-retinal barrier and the damage to the blood-retinal barrier causes retinal edema due to vascular leakage. The macula lutea is often damaged by macula tumentia following the removal of the eye lens for treatment of cataract.
  • For treatment of macular edema, laser photocoagulation, vitrectomy or systemic, intravitreal or sub-Tenon of steroids, etc. have been employed. Laser photocoagulation alleviates macula tumentia by blocking the blood vessels where fluid leakage occurs. However, care should be taken to avoid the fovea when irradiating laser because it is extremely vulnerable. If the fovea is damaged during the operation, the central vision may be impaired. Also, more than one laser treatments are often necessary to remove the swelling. Vitrectomy is employed when the laser treatment is ineffective, but this method is often associated with the high risk of tissue invasion which causes postoperative complications. The intravitreal administration of steroids may cause ocular hypertension, steroid-induced glaucoma and posterior subcapsular cataract. In addition, the intravitreal administration of steroids often cause postoperative complications.
  • In addition, other drugs administered directly into the vitreous cavity are known to require repeated administrations with 4-6 week intervals. The administration directly into the vitreous cavity also causes inconvenience of administration, pain and side effects.
  • The inventors of the present disclosure have researched to develop a medication exhibiting an excellent therapeutic effect for macular edema or macular degeneration related thereto, which reduces the inconvenience of administration and can be administered for a long time. In doing so, they have identified that a compound represented by Chemical Formula 1 exhibits therapeutic effect in a macular edema or macular degeneration animal model by effectively preventing vascular leakage in the eye and that the compound is targeted to the eye even when it is administered orally.
  • REFERENCES OF RELATED ART Non-Patent Documents
    • (Non-patent document 1) Joo-Hyun Kim et al., “Wnt5a attenuates the pathogenic effects of the Wnt/β-catenin pathway in human retinal pigment epithelial cells via down-regulating β-catenin and Snail”, BMB Rep. 2015; 48(9): 525-530.
    • (Non-patent document 2) Bokjun Ji et al., “Increased Levels of Dickkopf 3 in the Aqueous Humor of Patients With Diabetic Macular Edema”, Invest Ophthalmol Vis Sci. April 2016; 57; 2296-2304.
    SUMMARY
  • The present disclosure is directed to providing a pharmaceutical composition for preventing or treating macular degeneration or macular edema caused by vascular leakage in the eye.
  • The present disclosure is also directed to providing a food composition for preventing or improving macular degeneration or macular edema caused by vascular leakage in the eye.
  • The above objects of the present disclosure can be achieved by the present disclosure as described below.
  • The present disclosure provides a pharmaceutical composition for preventing or treating macular degeneration or macular edema, which contains a compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient:
  • Figure US20190290617A1-20190926-C00001
  • wherein
  • the broken line denotes a single bond or a double bond, wherein
  • i) if the bond between C-3 and C-4 and the bond between C-5 and C-6 are single bonds, R2b is nonexistent and R2a is CH2;
  • ii) if the bond between C-3 and C-4 is a double bond, the bond between C-5 and C-6 is a single bond, R2b is nonexistent and R2a is CH3; or
  • iii) if the bond between C-5 and C-6 is a double bond, the bond between C-3 and C-4 is a single bond and R2a and R2b are CH3,
  • R1 is H or CH3,
  • R3 is a functional group selected from a group consisting of R3a through R3d,
  • Figure US20190290617A1-20190926-C00002
  • in R3a,
  • i) each of R4 and R7 is OH or OCH3 and R5, R6 and R8 are H; or
  • ii) R5 is COOCH3, R7 is H or OH, R8 is OH and R4 and R are H,
  • in R3b,
  • R9 is a functional group selected from a group consisting of H, NH2, C1-C8 alkoxy and R9a through R9j and R10 is H or OH,
  • Figure US20190290617A1-20190926-C00003
  • in R3c,
  • each of R11 and R12 is OH or OAc and R13 is H; or
  • each of R11 and R12 is OH or OCH3 and R13 is CH3 and
  • in R3d,
  • R14 is OCH3 and R15 and R16 are CH3.
  • The present disclosure also provides a pharmaceutical composition for inhibiting vascular leakage in the eye, which contains the compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient
  • The present disclosure also provides a food composition for preventing or improving macular degeneration or macular edema, which contains the compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1 shows a result of investigating the inhibitory effect of a compound of the present disclosure (Preparation Example 30 or Preparation Example 31) on 3-catenin expression in HEK293 cells, in which the Wnt/β-catenin pathway is activated by treating with Wnt-3a CM, by western blot.
  • FIG. 2 shows a result of investigating the inhibitory effect of a compound of the present disclosure (Preparation Example 32) on 3-catenin expression in HEK293 cells, in which the Wnt/β-catenin pathway is activated by treating with Wnt-3a CM, by western blot.
  • FIG. 3 shows a result of investigating the inhibitory effect of a compound of the present disclosure (Preparation Example 33 or Preparation Example 34) on 3-catenin expression in human retinal epithelial cells, in which the Wnt/3-catenin pathway is activated by treating with Wnt-3a CM, by western blot.
  • FIG. 4 shows a result of investigating the inhibitory effect of intravitreal administration of a compound of the present disclosure (Preparation Example 33) on vascular leakage in a macular edema mouse model by fluorescein angiography and optical coherence tomography (A and B are images for a control group (compound-untreated) obtained after DMSO injection and C and D are images for a test group (Preparation Example 33) obtained after injection. The arrows indicate blood vessels).
  • FIG. 5 shows a result of investigating the inhibitory effect of intraperitoneal injection of a compound of the present disclosure (Preparation Example 33) on vascular leakage in a macular edema mouse model by optical coherence tomography (A: vehicle-administered group, B: Preparation Example 33 compound 1 mg/kg administered group).
  • FIG. 6 shows a result of orally administering a compound of the present disclosure (Preparation Example 33) to an ICR mouse and measuring distribution of the compound in target tissues (particularly, eye).
  • BEST MODE
  • The inventors of the present disclosure have researched to develop a medication exhibiting an excellent therapeutic effect for macular edema or macular degeneration related thereto, which reduces the inconvenience of administration and can be administered for a long time. In doing so, they have identified that a compound represented by Chemical Formula 1 exhibits therapeutic effect for macular edema or macular degeneration diseases by effectively preventing vascular leakage in the eye, particularly in the retina.
  • Accordingly, in an aspect, the present disclosure relates to a pharmaceutical composition for preventing or treating macular degeneration or macular edema, which contains a compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • Figure US20190290617A1-20190926-C00004
  • the broken line denotes a single bond or a double bond, wherein
  • i) if the bond between C-3 and C-4 and the bond between C-5 and C-6 are single bonds, R2b is nonexistent and R2a is CH2;
  • ii) if the bond between C-3 and C-4 is a double bond, the bond between C-5 and C-6 is a single bond, R2b is nonexistent and R2a is CH3; or
  • iii) if the bond between C-5 and C-6 is a double bond, the bond between C-3 and C-4 is a single bond and R2a and R2b are CH3,
  • R1 is H or CH3,
  • R3 is a functional group selected from a group consisting of R3a through R3d,
  • Figure US20190290617A1-20190926-C00005
  • in R3a,
  • i) each of R4 and R7 is OH or OCH3 and R5, R6 and R8 are H; or
  • ii) R5 is COOCH3, R7 is H or OH, R8 is OH and R4 and R6 are H,
  • in R3b,
  • R9 is a functional group selected from a group consisting of H, NH2, C1-C8 alkoxy and R9a through R9j and R10 is H or OH,
  • Figure US20190290617A1-20190926-C00006
  • in R3c,
  • each of R11 and R12 is OH or OAc and R13 is H; or
  • each of R11 and R12 is OH or OCH3 and R13 is CH3 and
  • in R3d,
  • R14 is OCH3 and R15 and R16 are CH3.
  • In the present disclosure, the term alkoxy group refers to an alkyl group bonded to oxygen (O-alkyl group). In the present disclosure, the alkoxy group may be a C1-C8 alkoxy group selected from a group consisting of a methoxy group (C1), an ethoxy group (C2), a propoxy group (C3), a butoxy group (C4), a pentyloxy group (C5), a hexyloxy group (C6), a heptyloxy group (C7) and an octyloxy group (C8), although not being limited thereto. Specifically, the alkoxy group of the present disclosure may be a methoxy group or an ethoxy group.
  • In Chemical Formula 1,
  • i) the structure wherein, if the bond between C-3 and C-4 and the the bond between C-5 and C-6 are single bonds, R2b is nonexistent and R2a is CH2 may be represented by <Chemical Formula 1-1>;
  • ii) the structure wherein, if the bond between C-3 and C-4 is a double bond, the bond between C-5 and C-6 is a single bond, R2b is nonexistent and R2a is CH3 may be represented by <Chemical Formula 1-2>; and
  • iii) the structure wherein, if the bond between C-5 and C-6 is a double bond, the bond between C-3 and C-4 is a single bond and R2a and R2b are CH3 may be represented by <Chemical Formula 1-3>.
  • Figure US20190290617A1-20190926-C00007
  • Specifically, the compound of the present disclosure of Chemical Formula 1 may be the compounds described in [Table 1], although not being limited thereto.
  • TABLE 1
    Preparation
    Example Structure
     1
    Figure US20190290617A1-20190926-C00008
     2
    Figure US20190290617A1-20190926-C00009
     3
    Figure US20190290617A1-20190926-C00010
     4
    Figure US20190290617A1-20190926-C00011
     5
    Figure US20190290617A1-20190926-C00012
     6
    Figure US20190290617A1-20190926-C00013
     7
    Figure US20190290617A1-20190926-C00014
     8
    Figure US20190290617A1-20190926-C00015
     9
    Figure US20190290617A1-20190926-C00016
    10
    Figure US20190290617A1-20190926-C00017
    11
    Figure US20190290617A1-20190926-C00018
    12
    Figure US20190290617A1-20190926-C00019
    13
    Figure US20190290617A1-20190926-C00020
    14
    Figure US20190290617A1-20190926-C00021
    15
    Figure US20190290617A1-20190926-C00022
    16
    Figure US20190290617A1-20190926-C00023
    17
    Figure US20190290617A1-20190926-C00024
    18
    Figure US20190290617A1-20190926-C00025
    19
    Figure US20190290617A1-20190926-C00026
    20
    Figure US20190290617A1-20190926-C00027
    21
    Figure US20190290617A1-20190926-C00028
    22
    Figure US20190290617A1-20190926-C00029
    23
    Figure US20190290617A1-20190926-C00030
    24
    Figure US20190290617A1-20190926-C00031
    25
    Figure US20190290617A1-20190926-C00032
    26
    Figure US20190290617A1-20190926-C00033
    27
    Figure US20190290617A1-20190926-C00034
    28
    Figure US20190290617A1-20190926-C00035
    29
    Figure US20190290617A1-20190926-C00036
    30
    Figure US20190290617A1-20190926-C00037
    31
    Figure US20190290617A1-20190926-C00038
    32
    Figure US20190290617A1-20190926-C00039
    33
    Figure US20190290617A1-20190926-C00040
    34
    Figure US20190290617A1-20190926-C00041
    Figure US20190290617A1-20190926-C00042
  • Specifically, the sesquiterpene derivative compound of the present disclosure may be one wherein, in Chemical Formula 1, if the bond between C-3 and C-4 and the bond between C-5 and C-6 are single bonds, R2b is nonexistent, R2a is CH2 and R3 is a functional group selected from a group consisting of R3b through R3d.
  • Specifically, in R3b, R9 may be selected from a group consisting of ethoxy, methoxy and R9a.
  • Specifically, in R3c, R11 may be OH, R12 may be OCH3 and R13 may be CH3.
  • Specifically, in R3d, R14 may be OCH3 and R15 and R16 may be CH3.
  • Most specifically, the compound of Chemical Formula 1 of the present disclosure may be a compound selected from a group consisting of:
    • 3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione;
    • 3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-2-hydroxy-5-methoxycyclohexa-2,5-diene-1,4-dione;
    • 3-[[(1S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-5-ethoxy-2-hydroxycyclohexa-2,5-diene-1,4-dione;
    • 18-methoxy-22-methyl-16-[{(5 S,8 S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17-ol; and
    • 18-methoxy-22,22-dimethyl-16-[{(5R,8 S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17(2H)-one.
  • The compound of Chemical Formula 1 of the present disclosure may be extracted from sponge. Specifically, the compound of Chemical Formula 1 for preventing or treating macular degeneration or macular edema caused by vascular leakage in the eye may be obtained by a method including a step of extracting one or more sponge selected from a group consisting of Rhopaloeides sp., Spongia sp., Smenospongia sp., Hippospongia sp., Dactylospongia sp., Verongula sp., Dysidea sp., sponge SS-1047, sponge SS-265 and sponge SS-1208 by adding a C1-C6 organic solvent.
  • The C1-C6 organic solvent may be selected from a group consisting of a C1-C6 alcohol (methanol, ethanol, propanol, butanol, pentanol, hexanol), acetone, an ether, benzene, chloroform, ethyl acetate, methylene chloride, hexane, cyclohexane, acetonitrile, dichloromethane and petroleum ether.
  • Specifically, the compound of the present disclosure of Chemical Formula 1 may be obtained by a method including: a step of extracting sponge by adding water, a C1-C4 alcohol or a mixture thereof as a solvent, thereby preparing a sponge extract; and a step of fractionating the extract by adding a second solvent and separating the same through chromatography.
  • As the chromatography, any one known to those skilled in the art can be used without limitation, including silica gel column chromatography, LH-20 column chromatography, ion-exchange chromatography, medium pressure liquid chromatography, thin-layer chromatography (TLC), silica gel vacuum liquid chromatography, high-performance liquid chromatography, etc.
  • The C1-C4 alcohol used to prepare the sponge extract may be selected from a group consisting of methanol, ethanol, propanol, isopropanol, butanol and isobutanol.
  • As the second solvent for fractionating the sponge extract, a C1-C4 alcohol, n-hexane, methylene chloride, acetone, chloroform, dichloromethane, ethyl acetate, acetonitrile or a mixture thereof may be used.
  • The compound of the present disclosure of Chemical Formula 1 includes a pharmaceutically acceptable salt thereof. In the present disclosure, the term ‘pharmaceutically acceptable’ means being physiologically acceptable and not causing allergic reactions such as gastroenteric trouble, dizziness, etc. or similar reactions when administered to human.
  • The pharmaceutically acceptable salt includes an acid addition salt with an inorganic acid or an organic acid. As the acid addition salt, an acid addition salt formed by a pharmaceutically acceptable free acid is useful. As the free acid, an inorganic acid or an organic acid may be used. As the inorganic acid, hydrochloric acid, bromic acid, sulfuric acid, phosphoric acid, etc. may be used. And, as the organic acid, citric acid, acetic acid, lactic acid, tartaric acid, fumaric acid, formic acid, propionic acid, oxalic acid, trifluoroacetic acid, methanesulfonic acid, benzenesulfonic acid, maleic acid, benzoic acid, gluconic acid, glycolic acid, succinic acid, 4-morpholinoethanesulfonic acid, camphorsulfonic acid, 4-nitrobenzenesulfonic acid, hydroxy-O-sulfonic acid, 4-toluenesulfonic acid, galacturonic acid, embonic acid, glutamic acid, aspartic acid, etc. may be used.
  • In an example of the present disclosure, it was found out that the compound of the present disclosure of Chemical Formula 1 inhibits β-catenin in vitro, suggesting that it can inhibit vascular leakage by inhibiting the Wnt/0-catenin mechanism. In another example of the present disclosure, it was confirmed that the present disclosure compound exhibits therapeutic effect by inhibiting vascular leakage in a macular edema animal model in vivo. In addition, it was confirmed that it exhibits therapeutic effect regardless of administration routes because the compound was distributed in the target tissue (eye) even when it was administered through other administration routes (oral adminstration, intraperitoneal injection, etc.) rather than directly into the vitreous cavity.
  • Accordingly, in another aspect, the present disclosure relates to a pharmaceutical composition for inhibiting vascular leakage in the eye, which contains the compound of Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • In the present disclosure, the term ‘vascular leakage’ refers to the leakage of body fluid or blood plasma due to damage to the integrity of blood vessels. The vascular leakage in the eye constitutes the major pathological conditions of various eye diseases. In the present disclosure, the term ‘vascular leakage in the eye’ refers to vascular leakage in various tissues (choroid, retina, etc.) constituting the eye. Specifically, it may refer to vascular leakage in the retina, although not being limited thereto.
  • The pharmaceutical composition of the present disclosure has preventive or therapeutic effect for a disease caused by vascular leakage in the eye. The disease caused by vascular leakage in the eye may be any one known in the art. For example, it include retinal degeneration, macular degeneration, retinal edema and macular edema. Specifically, in the present disclosure, the disease caused by vascular leakage in the eye may be macular degeneration or macular edema.
  • The pharmaceutical composition according to the present disclosure may contain only the sesquiterpene derivative compound of Chemical Formula 1 or a pharmaceutically acceptable salt thereof or may further contain one or more pharmaceutically acceptable carrier, excipient or diluent.
  • As the pharmaceutically acceptable carrier, it may further contain, for example, a carrier for oral administration or a carrier for parenteral administration. The carrier for oral administration may include lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, etc. And, the carrier for parenteral administration may include water, suitable oils, physiological saline, water-soluble glucose, glycol, etc. The pharmaceutical composition of the present disclosure may further contain a stabilizer and a preservative. A suitable stabilizer includes sodium bisulfite, sodium sulfite or an antioxidant such as ascorbic acid. A suitable preservative includes benzalkonium chloride, methyl- or propylparaben and chlorobutanol. In addition to these ingredients, the pharmaceutical composition of the present disclosure may further contain a lubricant, a humectant, a sweetener, a flavorant, an emulsifier, a suspending agent, etc. For other pharmaceutically acceptable carriers, reference can be made to the literature (Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Company, Easton, Pa., 1995).
  • The composition of the present disclosure may be administered to mammals including human by any means. For example, it may be administered orally or parenterally. The parenteral administration method may include intravenous, intramuscular, intraarterial, intramedullary, intradural, intracardiac, intraocular, intravitreal, transdermal, subcutaneous, intraabdominal, intranasal, intraintestinal, topical, sublingual or intrarectal administration, although not being limited thereto.
  • The pharmaceutical composition of the present disclosure may be prepared into a formulation for oral administration or parenteral administration depending on the administration routes.
  • For oral administration, the composition of the present disclosure may be formulated into a powder, a granule, a tablet, a pill, a sugar-coated tablet, a capsule, a solution, a gel, a syrup, a slurry, a suspension, etc. using the method known in the art. For example, as the formulation for oral administration, a tablet or a sugar-coated tablet may be prepared by mixing the active ingredient with a solid excipient, pulverizing the mixture, adding a suitable adjuvant and then processing into a granule mixture. Examples of the suitable excipient may include sugars including lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, etc., a starches including corn starch, wheat starch, rice starch, potato starch, etc., celluloses including cellulose, methyl cellulose, sodium carboxymethyl cellulose, hydroxypropylmethyl cellulose, etc. and fillers such as gelatin, polyvinylpyrrolidone, etc. If necessary, crosslinked polyvinylpyrrolidone, agar, alginic acid, sodium alginate, etc. may be added as a disintegrant. In addition, the pharmaceutical composition of the present disclosure may further contain an antiagglomerant, a lubricant, a humectant, a flavor, an emulsifier, an antiseptic, etc.
  • For parenteral administration, the composition may be formulated into an injection, an eye drop, an ointment, a cream, a lotion, an oil, a gel, an aerosol or a nasal inhaler using the method known in the art. These formulations are described in the literature generally known in the field of pharmaceutical chemistry (Remington's Pharmaceutical Science, 15th Edition, 1975. Mack Publishing Company, Easton, Pa. 18042, Chapter 87: Blaug, Seymour).
  • Specifically, the pharmaceutical composition of the present disclosure may be prepared into a formulation selected from a group consisting of an oral medication, an injection, an eye drop and an ointment.
  • The total effective amount of the sesquiterpene derivative compound of the present disclosure or a pharmaceutically acceptable salt thereof may be administered to a patient with a single dose or a multiple dose according to the fractionated treatment protocol for long-term administration. The content of the active ingredient of the pharmaceutical composition of the present disclosure may vary depending on the severity of a disease. The effective administration dosage of the compound or a pharmaceutically acceptable salt thereof is determined in consideration of various factors including the route and number of administration of the pharmaceutical composition, the age, body weight, health condition and sex of a patient, the severity of a disease, diet, excretion rate, etc. Those of ordinary skill in the art will be able to determine the adequate effective administration dosage of the sesquiterpene derivative or a pharmaceutically acceptable salt thereof for prevention or treatment of a disease caused by vascular leakage in the eye in consideration of these factors. The pharmaceutical composition according to the present disclosure is not specially limited in formulation, administration route and administration method as long as the effect of the present disclosure can be achieved.
  • In another aspect, the present disclosure relates to a food composition for preventing or improving macular degeneration or macular edema, which contains the compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient.
  • The food composition of the present disclosure includes all forms such as a functional food, a nutritional supplement, a health food, a food additive, a feed, etc. and is provided for animals including human or livestock. The food composition may be prepared into various forms according to the method known in the art.
  • For example, the health food may be prepared by preparing the sesquiterpene derivative of the present disclosure or a sponge extract containing the sesquiterpene into tea, juice or a drink for drinking or into a granule, a capsule or a powder. In addition, a composition may be prepared by mixing the sesquiterpene derivative of the present disclosure or a sponge extract containing the sesquiterpene with an active ingredient known to be effective in improving and preventing macular edema or macular degeneration.
  • Also, the functional food be prepared by adding the sesquiterpene derivative of the present disclosure or a sponge extract containing the sesquiterpene to beverages (including alcoholic beverage, fruit or processed fruit (e.g., canned fruit, bottling, jam, marmalade, etc.), fish, meat or processed foodstuffs thereof (e.g., ham, sausage corn beef, etc.), bread or noodles (e.g., udon, buckwheat noodle, instant noodle, spaghetti, macaroni, etc.), fruit juice, drinks, cookies, taffy, dairy products (e.g., butter, cheese, etc.), vegetable fats and oils, margarine, vegetable proteins, retort foods, frozen foods, condiments (e.g., soybean paste, soy sauce, etc.), etc.
  • In addition, the sesquiterpene derivative of the present disclosure or a sponge extract containing the sesquiterpene may be prepared into a powder or a concentrate for use as a food additive.
  • MODE FOR INVENTION
  • Hereinafter, the present disclosure will be described in detail through examples. However, the following examples are for illustrative purposes only and it will be apparent to those of ordinary skill in the art that the scope of the present disclosure is not limited by the examples.
  • EXAMPLES <Preparation Example 1> Preparation of methyl 3-[[(1R,2S,4aR,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4,5-dihydroxybenzoate
  • A Hyrtios sp. (38 g dry weight) sample preserved in EtOH was extracted completely using MeOH. After evaporating the MeOH extract in vacuo, the remaining residue (15.6 g) was fractionated using water and a CH2Cl2 solvent. The organic phase was evaporated in vacuo and a gum (5.32 g) was obtained. 2.37 g of the gum was subjected to flash chromatography using a Si gel column and using hexane and EtOAc of increasing concentrations as eluents. Some of the resulting fractions were subjected to flash chromatography using a Si gel column and using hexane/EtOAc (100:0 to 50:50). Two UV-positive fractions were obtained and further purified by HPLC (UV detection at 210 nm, eluent 90:10 MeOH/H2O) to obtain a compound of Preparation Example 1 (3 mg).
  • The obtained compound of Preparation Example 1 had the following physicochemical properties and was identified as ‘methyl 3-[[(1R,2S,4aR,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4,5-dihydroxybenzoate’.
  • Amorphous Solid.
  • IR (film) 3339, 1680, 1303 cm1.
  • UV (CH3OH) λmax 221 (17440), 269 (7460), 305 nm (3341, sh).
  • UV (CH3OH/NaOH) λmax 210 (18520), 241 (13176), 284 (4310), 322 nm (6950).
  • 1H NMR (600 MHz) δ 7.49 (1H, d, 1.5), 7.45 (1H, d, 1.5), 5.32 (1H, bs), 3.87 (3H, s), 2.84 (1H, d, 14) and 2.60 (1H, d, 14) AB system, 1.64 (3H, bs), 0.98 (3H, d, 6), 0.95 (3H, s), 0.90 (3H, s).
  • 13C NMR (CDCl3, 150.87 MHz): see [Table 2].
  • TABLE 2
    Carbon Preparation Example 1 (δ ppm)
    C-1 19.7
    C-2 25
    C-3 124.3
    C-4 140
    C-5 37.7
    C-6 37.8
    C-7 29.5
    C-8 37.6
    C-9 43.9
    C-10 44.9
    C-11 38.1
    C-12 17.3
    C-13 18.4
    C-14 33
    C-15 20.5
    C-16 126.2
    C-17 149.2
    C-18 143.1
    C-19 114.6
    C-20 121.3
    C-21 128.1
    C-22 168.2
    C-23 52.8
  • <Preparation Example 2> Preparation of 3-[[(1S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-2,5-dihydroxycyclohexa-2,5-diene-1,4-dione
  • A sponge (Smenospongia sp.) sample (2 kg) was immersed in MeOH and extracted with CHCl3/MeOH (1/1 mixture). The extract was evaporated under reduced pressure and an aqueous suspension thereof was extracted with CH2Cl2 (extract A). The extract A (8 g) was subjected to silica gel (CHCl3/MeOH of increasing concentrations) chromatography. A fraction 1 eluted with 2% MeOH (in CHCl3) and a fraction 2 eluted with 5% MeOH (in CHCl3) were prepared therefrom. The fraction 1 was eluted with 30% AcOEt (in hexane) to obtain a compound of Preparation Example 2 (20 mg).
  • The obtained compound of Preparation Example 2 had the following physicochemical properties and was identified as ‘3-[[(1S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-2,5-dihydroxycyclohexa-2,5-diene-1,4-dione’.
  • C21H28O4.
  • m.p.>350° C.
  • SM m/e (%): 191 (40), 154 (12), 135 (44), 121 (65), 109 (56), 107 (87), 95 (100).
  • UV (EtOH) λmax nm (c): 214, 286.
  • IR (KBr) ν cm−1: 3324, 2940, 1645, 1535.
  • 1H NMR (MeOD, 80 MHz) δ ppm: 5.71 (1H, s), 4.76 (2H, br s), 2.40 (2H, br s), 1.01 (3H, s), 0.92 (3H, d, J=7 HZ), 0.78 (3H, s).
  • 13C NMR (δ ppm, CD3OD, 20.115 MHz): see [Table 3].
  • <Preparation Example 3> Preparation of 3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(3-methylbutylamino)cyclohexa-3,5-diene-1,2-dione
  • 44 g of dried sponge (Smenospongia sp.) was extracted with CH2Cl2 and then with MeOH (extract B). The extract B (4 g) was subjected to chromatography using a silica gel column (CHCl3/increasing amounts MeOH) to obtain a fraction A eluted with 2% MeOH (in CHCl3) and a fraction B eluted with 5% MeOH (in HCl3). The faction A was purified with a Sephadex LH 20 (MeOH/CHCl3: 60/40) column to obtain a compound of Preparation Example 3 (20 mg) and a compound of Preparation Example 4 (5 mg).
  • The obtained compound of Preparation Example 3 had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(3-methylbutylamino)cyclohexa-3,5-diene-1,2-dione’.
  • C26H39NO3.
  • m.p.: 170-172° C.
  • SM m/e (%): 413 (4), 311 (8), 283 (12), 223 (100), 191 (11), 167 (22), 153 (27), 149 (15), 135 (14), 121 (16), 109 (18), 107 (12), 95 (79).
  • m/e 191.179, calc. 191.179 for C14H23; m/e 223.119, calc. 223.120 for C12H17NO3.
  • UV (EtOH) λmax nm (c): 204 (27230), 324 (14070).
  • IR (KBr) ν cm−1: 3417, 3275, 1640, 1592.
  • 1H NMR (CDCl3 0.200 MHz) δ ppm: 8.41 (1H exch., s), 6.41 (1H exch., t). 5.36 (1H, s), 4.43 (br s), 3.20 (2H, dt), 2.48 (d), 2.37 (d) (AB syst.), 2.31 (dt), 2.07 (2H, m), 1.85 (1H, m), 1.80-1.05 (11H, m), 1.04 (3H, s), 0.95 (9H, 3d overlapped), 0.83 (3H, s), 0.78 (1H, dd).
  • 13C NMR (δ ppm, CDCl3, 20.115 MHz): see [Table 3].
  • TABLE 3
    Preparation
    Carbon Example 2 (δ ppm) Preparation Example 3 (δ ppm)
    1  24.49 t  23.20 t
    2  29.48 t  27.98 t
    3  38.30 t  36.85 t
    4 162.35 s 160.33 s
    5  41.61 s  40.41 s
    6  33.70 t  32.97 t
    7  30.05 t  28.63 t
    8  39.20 d   37.% d
    9  43.65 s  42.89 s
    10  51.46 d  50.01 d
    11 102.58 t 102.39 t
    12  21.06 q  20.48 q
    13  18.88 q  17.80 q
    14  17.85 q  17.16 q
    15  33.94 t  32.57 t
    16 114.45 s 113.53 s
    17 188.82 s* 157.06 s
    18 179.65 s* 182.77 s
    19 101.79 d 91.484 d
    20 174.72 s* 150.21 s
    21 166.80 s* 177.97 s
    *may be reversed
  • <Preparation Example 4> Preparation of 3-[[(1S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(2-methylpropylamino)cyclohexa-3,5-diene-1,2-dione
  • A compound of Preparation Example 4 was prepared in the same manner as in Preparation Example 3. The obtained compound (Preparation Example 4) had the following physicochemical properties and was identified as ‘3-[[(1S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(2-methylpropylamino)cyclohexa-3,5-diene-1,2-dione’.
  • C25H37NO3.
  • SM m/e (%): 399 (5). 209 (100). 191(17), 166 (36). 152 (18), 135 (11), 121 (15), 109 (15), 107 (12), 95 (66).
  • UV (EtOH) λmax nm (c): 210 (14000), 329 (20150).
  • IR (KBr) ν cm−1: 3417, 3275, 1640, 1592.
  • 1H NMR (CDCl3, 200 MHz) δ ppm: 6.53 (1H, s). 5.41 (1H, s), 4.45 (2H, br s), 2.95 (2H, dt), 2.48 (1H, d), 2.45 (1H, d, J=13 Hz), 1.03 (3H, s), 0.97 (9H, 3d overlapped), 0.82 (3H, s). 0.76 (1H, dd).
  • <Preparation Example 5> Preparation of 3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-5-amino-4-hydroxycyclohexa-3,5-diene-1,2-dione
  • MeOH and DCM crude extracts of Hippospongia sp. were combined and fractionated with MeOH, DCM, hexane and BuOH. Among them, the hexane, DCM and MeOH fractions were subjected to flash column chromatography and semi-preparative RP-HPLC to obtain a compound of Preparation Example 5.
  • The obtained compound (Preparation Example 5) had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-5-amino-4-hydroxycyclohexa-3,5-diene-1,2-dione’.
  • Purple Solid.
  • C21H30O3N (HRESIMS m/z 344.2295, [M+H]+).
  • UV (MeOH) λmax (log ε) 315 (3.58) nm.
  • IR (KBr) 3835, 3566, 1624, 1536 cm−1.
  • 1H NMR (400 MHz, CD3OD) δ: 2.17/1.43 (2H, m H-1), 1.39 (2H, m, H-2), 1.50/1.38 (2H, m, H-3), 2.34/2.05 (2H, m, H-6), 1.23/1.82 (2H, m, H-7), 1.23 (1H, m, H-8), 0.82 (1H, m, H-10), 4.44 (2H, s, H-11), 1.05 (3H, s, H-12), 0.98 (3H, d, J=6.4 Hz H-13), 0.84 (3H, s, H-14), 2.47/2.40 (2H, dd, J=13.7 Hz, H-15), 5.51 (1H, s, H-19).
  • 13C NMR (100 MHz, CD3OD) δ: 22.8 (t, C-1), 27.5 (t, C-2), 36.4 (t, C-3), 160.1 (s, C-4), 39.9 (s, C-5), 32.5 (t, C-6), 28.3 (t, C-7), 37.6 (d, C-8), 42.1 (s, C-9), 49.7 (d, C-10), 101.1 (t, C-11), 19.4 (q, C-12), 16.9 (q, C-13), 16.2 (q, C-14), 31.6 (t, C-15), 113.7 (s, C-16), 159.4 (s, C-17), 183.2 (s, C-18), 93.6 (d, C-19), 183.2 (s, C-21).
  • <Preparation Example 6> Preparation of 2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien 1-yl]amino]acetic acid
  • After freeze-drying sponge (Dactylospongia elegans), an extract obtained by adding MeOH (3×1 L) to the freeze-dried sponge (33 g dry weight) was concentrated in vacuo and subjected to reversed-phase C18 vacuum liquid chromatography (0%, 20%, 50%, 70%, 90%, 100% MeOH in H2O and 1:1 CH2Cl2/MeOH). 20%, 50% and 70% MeOH fractions were obtained and subjected to C18 preparative HPLC (4 mL/min, gradient elution from 3:7 H2O/MeCN/0.1% formic acid to 100% MeCN/0.1% formic acid over 10 min, through a 150×10 mm, 5 m Phenomenex phenyl hexyl column). A Preparation Example 6 compound (11.7 mg, 0.035%), a Preparation Example 7 compound (1.4 mg, 0.004%) and a Preparation Example 8 compound (0.8 mg, 0.002%) were obtained therefrom.
  • The prepared Preparation Example 6 compound had the following physicochemical properties and was identified as ‘2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]acetic acid’.
  • Amorphous, red solid;
  • [α]D+94.4 (c 0.018, MeOH).
  • UV (PDA, CH3CN/H2O) λmax 218, 311, 494 nm.
  • IR (neat) νmax 3598, 2936, 2064, 1657 cm1′.
  • 1H (300 MHz) and 13C (75 MHz) NMR (CD3OD): see [Table 4].
  • HRESIMS m/z 424.2104 [M+Na]+ (calcd for C23H31NO5Na, 424.2094, Δ 1.0 mmu).
  • TABLE 4
    no. δC
    Figure US20190290617A1-20190926-P00899
    mult
    Figure US20190290617A1-20190926-P00899
    Figure US20190290617A1-20190926-P00899
    δH (J in Hz)
    Figure US20190290617A1-20190926-P00899
    COSY
    Figure US20190290617A1-20190926-P00899
    gHMBC
    Figure US20190290617A1-20190926-P00899
    δC, mult
    Figure US20190290617A1-20190926-P00899
    Figure US20190290617A1-20190926-P00899
    δH (J in Hz)
    Figure US20190290617A1-20190926-P00899
    1 24.2 CH2 2.19 br d(12.7) H
    Figure US20190290617A1-20190926-P00899
    -1, H
    Figure US20190290617A1-20190926-P00899
    -2, H-10
    2.15 22.7, CH2 2.09, m
    1.48, m H
    Figure US20190290617A1-20190926-P00899
    -1, H
    Figure US20190290617A1-20190926-P00899
    -2, H-10
    1.35, m
    2 29.7, CH2 1.84 dd (12.7, 3.0) H
    Figure US20190290617A1-20190926-P00899
    -1, H
    Figure US20190290617A1-20190926-P00899
    -2, H
    Figure US20190290617A1-20190926-P00899
    -3
    28.0, CH2 1.74, m
    1.23, m H
    Figure US20190290617A1-20190926-P00899
    -1, H
    Figure US20190290617A1-20190926-P00899
    -2, H
    Figure US20190290617A1-20190926-P00899
    -3
    1.14, m
    3 34.0, CH2 2.35, ddd (13.8, 5.2, 3.0) H
    Figure US20190290617A1-20190926-P00899
    -2, H
    Figure US20190290617A1-20190926-P00899
    -3, H
    Figure US20190290617A1-20190926-P00899
    -11
    4 32.1, CH2 2.23, m
    2.04, dd (13.8, 5.2) H
    Figure US20190290617A1-20190926-P00899
    -2, H
    Figure US20190290617A1-20190926-P00899
    -3
    1, 2, 4, 11 1.99, m
    4 161.4, C 159.3, C
    5 41.3, C 39.8, C
    6 37.8, CH2 1.51, m H
    Figure US20190290617A1-20190926-P00899
    -6, H
    Figure US20190290617A1-20190926-P00899
    -7
    8, 10 36.4, CH2 1.43, m
    1.41, m H
    Figure US20190290617A1-20190926-P00899
    -6
    5, 7, 8, 12 1.27, m
    7 28.8, CH2 1.40, m H2-6 6, 8, 9 27.6, CH2 1.32, m
    1.11, m
    8 38.9, CH 1.25, m H
    Figure US20190290617A1-20190926-P00899
    -7, H
    Figure US20190290617A1-20190926-P00899
    13
    7, 13 37.3, CH 1.17, m
    9 43.7, C 42.2, C
    10 50.9, CH 0.85, m H2-1 1, 8, 9, 12, 15 49.3, CH 0.74, m
    11 103.1, CH2 4.40 br s H
    Figure US20190290617A1-20190926-P00899
    -3, H2-11
    3, 4, 5 102.9, CH2 4.40, s
    4.37, s
    12 20.9, CH
    Figure US20190290617A1-20190926-P00899
    1.05, s 4, 5, 6, 10 20.2, CH3 0.97, s
    13 18.5, CH
    Figure US20190290617A1-20190926-P00899
    0.98, d (6.3) H-8 7, 8, 9 18.1, CH3 0.92, d (6.2)
    14 17.4, CH
    Figure US20190290617A1-20190926-P00899
    0.84, s 8, 9, 10, 15 17.3, CH3 0.76, s
    15 33.1, CH2 2.50, d (13.8) H
    Figure US20190290617A1-20190926-P00899
    -15
    8, 9, 10, 14, 16, 17, 21 32.0, CH3 2.36, d (13.6)
    2.40, d (13.8) H
    Figure US20190290617A1-20190926-P00899
    -15
    8, 9, 10, 14, 16, 17, 21 2.27, d (13.6)
    16 115.4, C 113.2, C
    17 159.4, C 159.7, C
    18 182.2, C 180.4, C
    19 93.6, CH 5.27, s 17, 18, 21 92.9, CH 5.19, s
    20 150.9, C 149.6, C
    21 183.8, C 181.8, C
    22 7.56, s (5.8)
    23 44.7, CH2 3.94, br s 20, 24 42.1, CH3 3.88, d (5.8)
    24 171.7, C 169.8, C
    *CD, OD,
    Figure US20190290617A1-20190926-P00899
    75 MHz,
    Figure US20190290617A1-20190926-P00899
    300 MHz,
    dHMBC correlations are from proton(s) stated to the indicated carbons,
    Figure US20190290617A1-20190926-P00899
    DMSO-d
    Figure US20190290617A1-20190926-P00899
    .
    Figure US20190290617A1-20190926-P00899
    indicates data missing or illegible when filed
  • <Preparation Example 7> Preparation of 3-[[5-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dienl-yl]amino]propanoic acid
  • A compound of Preparation Example 7 was prepared in the same manner as in Preparation Example 6. The prepared compound (Preparation Example 7) had the following physicochemical properties and was identified as ‘3-[[5-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]propanoic acid’.
  • Amorphous, Red Solid.
  • [α]D+13 (c 0.06, MeOH).
  • UV (PDA, CH3CN/H2O) λmax 233, 314, 494 nm.
  • IR (neat) λmax 3410, 2930, 1686, 1632, 1567 cm−1.
  • HRESIMS m/z 438.2264 [M+Na]+ (calcd for C24H33NO5Na, 438.2251, Δ 1.3 mmu).
  • 1H NMR (CD3OD, 300 MHz) δ 5.38 (1H, s, H-19), 4.40 (2H, s, H2-11), 3.45 (2H, t, J=6.8 Hz, H2-23), 2.59 (2H, t, J=6.8 Hz, H2-24), 2.47 (1H, d, J=13.6 Hz, Ha-15), 2.38 (1H, d, J=13.6 Hz, Hb-15), 2.32 (1H, m, Ha-3), 2.16 (1H, m, Ha-1), 2.04 (1H, m, Hb-3), 1.80 (1H, m, Ha-2), 1.48 (1H, m, Ha-6), 1.43 (1H, m, Hb-1), 1.41 (1H, m, Ha-7), 1.36 (1H, m, Hb-6), 1.35 (1H, m, Hb-7), 1.29 (1H, m, Hb-2), 1.21 (1H, m, H-8), 1.04 (3H, s, H3-12), 0.97 (3H, d, J=6.4 Hz, H3-13), 0.83 (3H, s, H3-14), 0.81 (1H, m, H-10).
  • 13C NMR (CD3OD, 75 MHz) δ 183.4 (C, C-21), 179.6 (C, C-18), 176.0 (C, C-25), 162.9 (C, C-4), 161.1 (C, C-17), 152.0 (C, C-20), 115.5 (C, C-16), 103.3 (CH2, C-11), 92.6 (CH, C-19), 51.1 (CH, C-10), 44.0 (C, C-9), 41.8 (C, C-5), 39.9 (CH2, C-23), 39.0 (CH, C-8), 38.2 (CH2, C-6), 34.5 (CH2, C-24), 34.2 (CH2, C-3), 33.4 (CH2, C-15), 30.0 (CH2, C-2), 29.4 (CH2, C-7), 24.3 (CH2, C-1), 21.4 (CH3, C-12), 18.7 (CH3, C-13), 18.1 (CH3, C-14).
  • <Preparation Example 8> Preparation of 7-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-1,3-benzoxazole-5,6-diol
  • A compound of Preparation Example 8 was prepared in the same manner as in Preparation Example 6. The prepared compound (Preparation Example 8) had the following physicochemical properties and was identified as ‘7-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-1,3-benzoxazole-5,6-diol’.
  • Colorless Solid.
  • [α]D−6.7 (c 0.075, MeOH).
  • UV (PDA, CH3CN/H2O) λmax 236, 297, 323 (sh) nm.
  • IR (neat) νmax 3408, 2927, 1541 cm−1.
  • 1H (300 MHz) and 13C (75 MHz) NMR (CD3OD): see [Table 5].
  • HRESIMS m/z 378.2050 [M+Na]+ (calcd for C22H29NO3Na, 378.2040, Δ 1.0 mmu).
  • TABLE 5
    no. δC, mult.a δN (J in Hz)b COSY gHMBCc
    1  24.8, CH2 2.37, m Hb-1, Ha-2, H-10 2
     1.55, m Ha-1, H-10
    2  30.2, CH2 1.87, m Hb-2, Ha-3
     1.28, m Ha-1, Ha-2, H2-3
    3  34.4, CH2 2.32, m Hb-2, Hb-3 2, 4, 11
     2.03, m Ha-2, Ha-3 5
    4 161.8, C
    5  41.8, C
    6  38.3, CH2 1.45, m 8
    7  29.5, CH2 1.41, m
    8  38.7, CH 1.41, m H3-13
    9  44.2, C
    10  51.1, CH 0.94, m H2-1 5, 9, 12, 14
    11 103.4, CH2 4.35, s 3, 4, 5
    4.32, s 3, 4, 5
    12  20.7, CH3 1.07, s 4, 5, 6, 10
    13  19.2, CH3 1.08, d (6.8) H-8 7, 8, 9
    14  18.3, CH3 0.95, s 8, 9, 10, 15
    15  35.8, CH2 2.94, d (13.9) Hb-15 8, 9, 10, 14,
    16, 17, 21
    2.86, d (13.9) Ha-15 8, 9, 10, 14,
    16, 17, 21
    16 111.0, C
    17 146.7, C
    18 145.2, C
    19 102.6, CH 6.96, s 17, 18, 20, 21
    20 131.7, C
    21 146.7, C
    22 153.2, CH 8.20, s 20, 21
    a75 MHz,
    b300 MHz,
    cHMBC correlations are from proton(s) stated to the indicated carbons.
  • <Preparation Example 9> Preparation of [7-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-6-acetyloxy-1,3-benzoxazol-5-yl] acetate
  • As in Preparation Example 5, after freeze-drying sponge (Dactylospongia elegans), an extract was prepared by adding MeOH (3×1 L) to the freeze-dried sponge (33 g dry weight).
  • The substance insoluble in MeOH (120 mg) was added to pyridine (0.5 mL) and stirred at room temperature for 12 hours after treating with (CH3CO)2O (0.5 mL). The obtained substance was concentrated in vacuo and subjected to sequential reversed-phase HPLC separation (A: H2O/MeCN+0.1% formic acid (3:7) to 100% MeCN+0.1% formic acid over 10 min at 4 mL/min and held for an additional 10 min on a 150×10 mm, 5 μm Phenomenex Luna C18 column; B: H2O/MeOH with 0.1% formic acid (3:7) to 100% MeOH with 0.1% formic acid for 10 min at 4 mL/min and held for an additional 5 min on a 150×10 mm, 5 μm Phenomenex Luna phenyl hexyl column). The obtained compound of Preparation Example 9 (2.3 mg, 0.007%) had the following physicochemical properties and was identified as ‘[7-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-6-acetyloxy-1,3-benzoxazol-5-yl] acetate’.
  • Colorless Oil.
  • [α]D−170 (c 0.003, CHCl3).
  • UV (PDA, CH3CN/H2O) λmax 233, 278, 284, 300 (sh) nm.
  • IR (neat) νmax 3488, 2927, 1775, 1630, 1458 cm−1.
  • HRESIMS m/z 462.2250 [M+Na]+ (calcd for C26H33NO5Na, 462.2251, Δ 0.1 mmu).
  • 1H NMR (CDCl3, 300 MHz) δ 8.07 (1H, s, H-22), 7.54 (1H, s, H-19), 4.42 (1H, d, J=1.6 Hz, Ha-11), 4.38 (1H, d, J=1.6 Hz, Hb-11), 2.83 (1H, d, J=14.2 Hz, Ha-15), 2.76 (1H, d, J=14.2 Hz, Hb-15), 2.35 (1H, m, Ha-3), 2.34 (s, a-OCOCH3), 2.30 (s, b-OCOCH3), 2.08 (1H, m, Hb-3), 1.92 (1H, m, Ha-2), 1.58 (1H, m, Ha-1), 1.49 (1H, m, Ha-6), 1.45 (1H, m, Hb-1), 1.43 (1H, m, H-8), 1.42 (2H, m, H2-7), 1.28 (1H, m, Hb-6), 1.26 (1H, m, Hb-2), 1.07 (3H, s, H3-12), 0.97 (3H, d, J=5.6 Hz, H3-13), 0.94 (3H, s, H3-14), 0.92 (1H, m, H-10).
  • 13C NMR (CDCl3, 150 MHz) δ168.1 (C, a-OCOCH3), 167.9 (C, b-OCOCH3), 159.1 (C, C-4), 152.7 (CH, C-22), 147.9 (C, C-21), 140.4 (C, C-17), 137.4 (C, C-18), 136.6 (C, C-20), 118.3 (C, C-16), 111.9 (CH, C-19), 102.2 (CH2, C-11), 50.1 (CH, C-10), 43.0 (C, C-9), 40.3 (C, C-5), 37.6 (CH, C-8), 36.2 (CH2, C-6), 35.6 (CH2, C-15), 32.6 (CH2, C-3), 28.3 (CH2, C-2), 27.7 (CH2, C-7), 23.1 (CH2, C-1), 20.1 (2×CH3, —OCOCH3), 19.9 (CH3, C-12), 18.1 (CH3, C-13), 16.8 (CH3, C-14).
  • <Preparation Example 10> Preparation of 3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-2-hydroxy-5-methoxycyclohexa-2,5-diene-1,4-dione
  • A sponge (Spongiidae SS-1047, 0.30 kg, wet weight) was obtained as described in the literature ‘Yohei Takahashi et al., 2010’ and extracted. Briefly, EtOAc-soluble substances (1.2 g) were fractionated using a silica gel column (n-hexane/EtOAc) and a fraction 1, a fraction 2 and a fraction 3 of low polarity and a polar fraction 4 were prepared. The fraction 3 was fractionated and purified by C18 column (MeOH/H2O) and C18 HPLC (Luna 5u Phenyl-Hexyl, 250×10 mm; eluent, MeOH/H2O/CF3CO2H, 85:15:0.05; flow rate, 2.5 mL/min; UV detection at 320 nm) to obtain compounds of Preparation Example 10 and Preparation Example 11.
  • The obtained Preparation Example 10 compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-2-hydroxy-5-methoxycyclohexa-2,5-diene-1,4-dione’.
  • m.p.: 95-98° C.
  • [α]20 579+64.4° (c 0.27 CHCl3).
  • IR (film) 3341, 1652, 1645, 1609, 1243 cm1′.
  • UV(CH3OH) λmax 213 (9600), 288 nm (13485).
  • UV (CH3OH/NaOH) λmax 210 (12850), 290 (8930), 526 nm (1650).
  • 1H NMR (600 MHz, CDCl3, 6, J in Hz): see [Table 6].
  • 13C NMR (150.87 MHz, CDCl3): see [Table 6].
  • HREIMS m/z 358.2151 [M+](12, calcd for C22H30O4, 358.2144), 191.1803 (15, calcd for C14H23, 191.1800), 168.0423 (41, calcd for C8H8O4, 168.0422), 121.1013 (12, calcd for C9H13, 121.1017), 107.0859 (30, calcd for C8H11, 107.0861), 95.0861 (100, calcd for C7H11, 95.0861).
  • TABLE 6
    13C NMR
    position 1 H NMR carbon 1a 1
    Figure US20190290617A1-20190926-P00899
    1c
    H2C-1 1.40, m C-1 17.7 19.9 20.6
    1.97, m
    H2C-2 1.82, m C-2 27.1 27.1 27.8
    1.93, m
    HC-3 5.06, bs C-3 121 121 121.6
    H2C-6 0.96 (ax), m C-4 143.9 143.9 144.7
    1.57 (eq), ddd,
    13.0, 3.0, 3.0
    H2C-7 1.25, m C-5 43.1 38.6 39.2
    1.28, m
    HC-8 1.17, m C-6 36.1 36.1 36.7
    HC-10 0.95, bd, 12.0 C-7 28.1 28.1 28.6
    H2C-11 2.42, d, 14.0 C-8 38.1 38.1 38.6
    2.55, d, 14.0
    ABsystem
    H3C-12 0.77, s C-9 38.6 43.1 43.8
    H3C-13 0.90, d, 6.0 C-10 48.2 48.2 48.6
    H3C-14 0.94, s C-11 32.5 32.5 33
    H3C-15 1.47, bs C-12 17.3 17.3 18
    H3C-0 3.79, s C-13 18.1 17.7 18.4
    HC-18 C-14 19.9 20.2 20.9
    HC-19 5.77, s C-15 20.2 18.1 18.9
    HC-21 C-16 117.8 117.8 118.3
    C-17 182.4 182.4 183
    C-18 161.8 161.8 162.5
    C-19 102 102 102.7
    C-20 182 182 182.8
    C-21 153.4 153.4 154
    C-22 56.8 56.8 57.5
    Figure US20190290617A1-20190926-P00899
    indicates data missing or illegible when filed
  • <Preparation Example 11> Preparation of 2-hydroxy-5-methoxy-3-[[(1R,2S)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]cyclohexa-2,5-diene-1,4-dione
  • A compound of Preparation Example 11 was prepared in the same manner as in Preparation Example 10. The obtained compound (Preparation Example 11) had the following physicochemical properties and was identified as ‘2-hydroxy-5-methoxy-3-[[(1R,2S)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]cyclohexa-2,5-diene-1,4-dione’.
  • Pale Yellow Feathery Solid.
  • m.p. 108.5-109.5° C.
  • C22H30O4 (high resolution FABMS (M+358.2146, Δ 0.2 mmu, C22H30O4; MH+ 359.2223, Δ 0.1 mmu, C22H31O4)).
  • 1H NMR (500 MHz, CDCl3) δ: 0.73 (s, 3H), 0.90 (sh, 1H), 0.92 (s, 3H), 0.96 (d, J=7 Hz, 3H), 0.99 (s, 3H), 1.12 (ddd, J=13.5, 13.5, 4.3 Hz, 1H), 1.33-1.45 (complex mult., 4H), 1.73 (mult, 1H), 1.79 (br d, 1H), 1.95 (ddd, J=18, 17.5, 4.5 Hz, 1H), 2.08 (br d, J=13 Hz, 1H), 2.45 (d, cJ=13.0 HZ, 1H), 2.58 (d, J=13.0 Hz, 1H), 3.84 (s, 3H), 5.35 (br s, 1H), 5.84 (s, 1H), 7.45 (s, 1H); 13C NMR (500 MHz, CDCl3), δ (mult., proton assignments): 16.0 (q, 0.73, C-14), 16.5 (q, 0.92, C-11), 22.7 (t, 1.40, 1.46, C-2), 27.9 (q, 0.96, C-13), 29.7 (q, 0.99, C-12). 30.6 (t, 0.90, 1.79, C-l), 31.5 (t, 1.73, 1.95, C-7), 32.7 (t, 2.45, 2.58, C-15), 36.3 (s, C-4), 36.4 (d, 1.36, C-8), 40.9 (s, C-g), 41.2 (t, 1.13, 1.32, C-3), 41.7 (s, 2.08, C-10), 56.8 (q, 3.86, C-22), 102.0 (d, 5.85, C-19), 114.8 (d, 5.35, C-6), 118.3 (s, C-16), 146.3 (s, C-5), 152.8 (s, —OH, C-17), 161.5 (s, C-20), 182.0 (s, C-21), 182.4 (s, C-18).
  • <Preparation Example 12> Preparation of 3-[[(1R,2S,8aS)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione
  • The fraction 1 obtained in Preparation Example 10 was refractionated by Cis column (MeOH/H2O) and C18 HPLC (Wakosil-II 5C18AR, Wako Pure Chemical Industries, Ltd., 250×10 mm; eluent, MeCN/H2O/CF3CO2H, 90:10:0.05; flow rate, 2.0 mL/min; UV detection at 300 nm) to obtain a compound of Preparation Example 11 (2.8 mg, 0.00093% wet weight) and a compound of Preparation Example 12 (24.7 mg, 0.0082%).
  • The obtained Preparation Example 12 compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,8aS)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione’.
  • Purple-Red, Amorphous Solid.
  • [α]23 D−14 (c 0.2, CHCl3).
  • IR (film) νmax 3290, 1730, 1650, 1590, 1510, 1460, 1380, 1360, 1220 cm−1.
  • UV (MeOH) λmax 336 (log 4.28), 507 nm (2.84).
  • 1H NMR (CDCl3): see [Table 7].
  • 13C NMR (CDCl3): see [Table 7].
  • EIMS m/z (%) 447 (M+, 9), 257 (100), 191 (2), 166 (20), 152 (5), 105 (10), 95 (15); HREIMS m/z 447.2790 [M]+ (calcd for C29H37NO3, 447.2773).
  • TABLE 7
    position δC δH (m, J in Hz) HMBC NOESY
    1 30.5 CH2 1.85 (m) 1b, 2a, 10, 14, 15a
    0.95 (m) 1a, 3b
    2 22.8 CH2 1.49 (m) 1a, 1b, 2a, 3b
    1.32 (m) 2a, 3a, 12
    3 41.3 CH2 1.37 (m) 2, 11, 12 3b
    1.16 (ddd, 13.5, 13.5, 4.1) 2, 4, 11, 12 2a, 3a, 11
    4 36.4 C
    5 146.5 C
    6 114.8 CH 5.38 brs 4, 8, 7, 10 7a, 7b, 11
    7 31.6 CH2 1.96 (ddd, 17.3, 5.2, 5.2) 5, 6, 8, 9, 13 7b, 8, 13
    1.77 (dddd, 17.3, 9.5, 2.6, 2.6) 5, 6, 8, 13 7a, 8, 13, 14
    8 36.3 CH 1.39 (m) 7, 9, 13 7a, 10, 13, 15b
    9 40.6 C
    10 41.6 CH 2.10 (m) 5 1a, 2b, 8, 12, 15b
    11 29.7 CH3 1.03b (brs) 3, 4, 5, 12 3a, 3b
    12 28.0 CH3 0.95b (s) 3, 4, 5, 11 2b, 10
    13 16.5 CH3 0.99b (d, 6.7) 7, 8, 9 7a, 7b, 8, 14, 5a, 15b
    14 15.9 CH3 0.74b (s) 8, 9, 10, 15 1a, 1b, 7b, 13, 15a, 15b
    15 32.7 CH3 2.54 (d, 13.4) 8, 9, 10, 14, 16, 17, 21 1a, 13, 14, 15b
    2.41 (d, 13.4) 8, 9, 10, 14, 16, 17, 21 8, 10, 13, 15a
    16 114.7 C
    17 156.5 C
    18 178.5 C
    19 91.8 CH 5.42 (s) 17, 21 22, 23
    20 149.9 C
    21 183.0 C
    22 44.0 CH3 3.44a (td, 6.9, 6.1) 20, 23, 24 19, 23, 25
    23 34.2 CH2 2.95a (t, 6.9) 22, 24, 25 19, 22, 25
    24 137.4 C
    25 128.5
    Figure US20190290617A1-20190926-P00899
    CH
    7.19
    Figure US20190290617A1-20190926-P00899
    (d, 73)
    23, 27 22, 23
    26 128.9
    Figure US20190290617A1-20190926-P00899
    CH
    7.33
    Figure US20190290617A1-20190926-P00899
    (dd, 7.3, 7.3)
    24, 27
    27 127.0 CH 7.26 (t, 7.3) 25
    20-NH 6.54 (brs) 19, 21
    Figure US20190290617A1-20190926-P00899
    Figure US20190290617A1-20190926-P00899
    H
    Figure US20190290617A1-20190926-P00899
    Figure US20190290617A1-20190926-P00899
    H
    Figure US20190290617A1-20190926-P00899
    Figure US20190290617A1-20190926-P00899
    C
    Figure US20190290617A1-20190926-P00899
    indicates data missing or illegible when filed
  • <Preparation Example 13> Preparation of 3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione
  • A compound of Preparation Example 13 was prepared in the same manner as in Preparation Example 12. The obtained compound (Preparation Example 13) had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione’.
  • Purple-Red. Amorphous Solid.
  • [α]25 D+180 (c 0.1, CHCl3).
  • IR (film) λmax 3270, 1730, 1640, 1590, 1510, 1460, 1380, 1210 cm−1.
  • UV (MeOH) λmax 335 (log 4.20), 502 nm (2.74).
  • 1H NMR (CDCl3): see [Table 8].
  • 13C NMR (CDCl3): see [Table 8].
  • EIMS m/z (%) 447 (M+, 25), 257 (100), 209 (17), 191 (18), 168 (45), 166 (48), 152 (17), 119 (42), 105 (40);
  • HREIMS m/z 447.2783 [M]+ (calcd for C29H37NO3, 447.2773).
  • TABLE 8
    Pos. δC δH (m, J in Hz) HMBC NOESY
     1 19.9 CH2 2.03 (m)  5 1b, 2b, 10, 14, 15a
    1.45 (dddd, 12.0, 12.0, 12.0, 6.1) 1a, 2a, 12, 14
     2 27.0 CH2 1.98 (m) 1b, 2a, 2b, 3, 10
    1.81 (m) 1a, 2a, 3
     3 120.8 CH 5.12 (brs) 2a, 2b, 11
     4 144.1 C
     5 38.5 C
     6 36.0 C 1.62 (ddd, 12.2, 3.1, 3.1) 6b, 7
    1.05 (ddd, 12.2, 12.2, 4.8) 6a, 7
     7 28.0 CH2 1.32
    Figure US20190290617A1-20190926-P00899
    (m)
    6a, 6b, 8, 13, 14
     8 37.7 CH 1.26 (m) 7, 10
     9 42.7 C
    10 47.6 CH 1.03 (overlapped) 1a, 2b, 8, 15a
    11 18.2 CH3 1.54b (brs) 3, 4, 5 3, 12
    12 20.1 CH3 1.00b (s) 4, 5, 6, 10 6a, 6b, 14
    13 17.7 CH3 0.95b (d, 6.3) 7, 8, 9 7, 14
    14 17.3 CH3 0.82b (s) 8, 9, 10, 15 1a, 1b, 8, 12, 13, 15a, 15b
    15 32.4 CH2 2.56 (d, 13.9) 8, 9, 14, 16, 17, 21 1a, 10, 14, 15b
    2.41 (d, 13.9) 9, 10, 14, 16, 17, 21 13, 14, 15a
    16 113.9 C
    17 156.9 C
    18 178.3 C
    19 91.8 CH 5.40 (s) 17, 21 22
    20 150.9 C
    21 182.8 C
    22 44.0 CH3 3.42
    Figure US20190290617A1-20190926-P00899
    (1d, 7.1, 6.7)
    20, 23, 24 19, 23, 25, 20-NH
    23 34.3 CH3 2.95
    Figure US20190290617A1-20190926-P00899
    (t, 7.1)
    22, 24, 25 19, 22, 25, 20-NH
    24 137.4 C
    25 128.6b CH 7.18
    Figure US20190290617A1-20190926-P00899
    (d, 7.2)
    23, 27 22, 23, 26
    26 128.9c CH 7.33
    Figure US20190290617A1-20190926-P00899
    (dd, 7.4, 7.2)
    24, 25 25, 27
    27 127.1 CH 7.26 (t, 7.4) 25 26
    17-OH 8.36 (brs) 22, 23
    20-NH 6.46 (brt, 6.7) 22, 23
    a2H,
    b3H,
    c2C.
    Figure US20190290617A1-20190926-P00899
    indicates data missing or illegible when filed
  • <Preparation Example 14> Preparation of 3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(2-methylpropylamino)cyclohexa-3,5-diene-1,2-dione
  • The fraction 2 obtained in Preparation Example 10 was subjected to C18 column (MeOH/H2O/CF3CO2H) and C18 HPLC (Luna 5u Phenyl-Hexyl, Phenomenex, 250×10 mm; eluent, MeCN/H2O/CF3CO2H, 80:20:0.05; flow rate, 2.0 mL/min; UV detection at 300 nm) to obtain a compound of Preparation Example 14 (0.9 mg, 0.00030%), a fraction y and a fraction 6.
  • The obtained compound of Preparation Example 14 had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(2-methylpropylamino)cyclohexa-3,5-diene-1,2-dione’.
  • Purple-Red, Amorphous Solid.
  • [α]23 D+160 (c 0.1, CHCl3).
  • IR (film) λmax 3270, 1730, 1640, 1590, 1510, 1380, 1210 cm1.
  • UV (MeOH) λmax 334 (log 4.29), 509 nm (2.86).
  • 1H NMR (CDCl3): see [Table 9].
  • 13C NMR (CDCl3): see [Table 9].
  • EIMS m/z (%) 399 (M+, 8), 209 (100), 191 (3), 166 (11), 152 (9), 107 (9), 95 (22).
  • HREIMS m/z 399.2790 [M]+ (calcd for C25H37NO3, 399.2773).
  • TABLE 9
    Pos. δC δH (m, J in Hz) HMBC NOESY
     1 19.9 CH2 2.04 (m) 2, 3, 5, 9, 10 1b, 3, 10, 15a
    1.45 (dddd, 12.0, 12.0, 12.0, 6.1) 2, 5, 10 1a, 14
     2 27.1 CH2 1.99 (m) 1b, 2b
    1.86 (m) 2a, 3, 10
     3 120.8 CH 5.12 (brs) 12 1a, 2b, 11
     4 144.1 C
     5 38.5 C
     6 36.0 CH2 1.61 (ddd, 12.7, 3.2, 3.2)  7 6b, 7
    1.04 (m) 6a, 7
     7 27.9 CH2 1.34
    Figure US20190290617A1-20190926-P00899
    (m)
     8 6a, 6b, 13, 14
     8 37.7 CH 1.27 (m) 10, 13
     9 42.6 C
    10 47.6 CH 1.04 (m) 1, 2, 5, 9 1a, 2b, 8
    11 18.1 CH3 1.53b (brs) 3, 4, 5, 6 3, 12
    12 20.1 CH3 1.00b (s) 4, 5, 6, 10 11, 14
    13 17.7 CH3 0.96b (d, 6.1) 7, 8, 9 7, 8, 15b
    1b, 7, 12, 13, 15a
    14 17.3 CH3 0.82b (s) 8, 9 10, 15 15b
    15 32.4 CH2 2.56 (d, 13.9) 8, 9, 14, 16, 17, 21 1a, 14, 15b
    2.42 (d, 13.9) 8, 9, 10, 14, 16, 17, 21 13, 14, 15a
    16 113.9 C
    17 157.1 C
    18 178.1 C
    19 91.6 CH 5.37 (s) 17, 21 22
    20 150.5 C
    21 182.9 C
    22 50.3 CH2 2.97
    Figure US20190290617A1-20190926-P00899
    (dd, 6.4, 6.4)
    20, 23, 24 19, 23, 24, 20-NH
    23 27.6 CH 1.96
    Figure US20190290617A1-20190926-P00899
    (m)
    22, 24
    24 20.2d CH3 098c (d, 6.7) 22, 23 22, 23
    20-NH 6.53 (brs) 22
    a2H,
    b3H,
    c6H, d 2C.
    Figure US20190290617A1-20190926-P00899
    indicates data missing or illegible when filed
  • <Preparation Example 15> Preparation of 3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-[[(2S)-2-methylbutyl]amino]cyclohexa-3,5-diene-1,2-dione
  • The fraction γ obtained in Preparation Example 14 was purified by C18 HPLC (Luna 5u C18(2), Phenomenex, 250×10 mm; MeOH/H2O/Et2NH, 70:30:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm) to obtain a compound of Preparation Example 15 (1.4 mg, 0.00047%) and a compound of Preparation Example 16 (4.0 mg, 0.0013%).
  • The compound of Preparation Example 15 had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-[[(2S)-2-methylbutyl]amino]cyclohexa-3,5-diene-1,2-dione’.
  • Purple-Red, Amorphous Solid.
  • [α]21 D+136 (c 0.25, CHCl3).
  • IR (film) νmax 3270, 1680, 1650, 1590, 1510, 1450, 1380, 1210 cm−1.
  • UV (MeOH) λmax 336 (log 4.09), 505 nm (2.67).
  • 1H NMR (CDCl3): see [Table 10].
  • 13C NMR (CDCl3): see [Table 10].
  • HREIMS m/z 413.2940 [M]+ (calcd for C26H39NO3, 413.2930).
  • TABLE 10
    Pos. δC δH (m, J in Hz) HMBC NOESY
     1 19.9 CH3 2.04 (m) 2, 3, 5, 10 1b, 10, 15a
    1.47 (m) 12, 14
     2 27.1 CH2 1.99 (m) 1b, 2b
    1.85 (m) 2a, 10
     3 120.8 CH 5.12 (brs) 2a, 2b, 11
     4 144.1 C
     5 38.5 C
     6 36.0 CH2 1.62 (ddd, 12.7, 3.0, 3.0) 6b, 7, 11, 12
    1.08 (m) 6a
     7 27.9 CH2 1.35
    Figure US20190290617A1-20190926-P00899
    (m)
    6a, 6b
     8 37.7 CH 1.27 (m) 10, 13
     9 42.7 C
    10 47.6 CH 1.04 (m) 2, 5, 9 1a, 2b, 8
    11 18.1 CH3 1.53b (brs) 3, 4, 5 3, 6a
    12 19.9 CH3 0.99b (s) 4, 5, 6 6a, 14
    13 17.7 CH3 0.96b (d, 6, 8) 7, 8 8, 14, 15b
    14 17.3 CH3 0.82b (s) 8, 9, 10, 15 1b, 7, 12, 13 15a, 15b
    15 32.4 CH2 2.55 (d, 13.9) 8, 9, 14, 16, 17, 21 1a, 14, 15b
    2.42 (d, 13.9) 8, 9, 10, 14, 16, 17, 21 13, 14, 15a
    16 113.8 C
    17 157.2 C
    18 178.1 C
    19 91.5 CH 5.37 (s) 17, 21 22a, 22b, 23
    20 150.6 C
    21 182.9 C
    22 48.7 CH3 3.08 (ddd, 13.2, 6.5, 6.5) 20, 23, 24, 26 19, 26, 20-NH
    2.95 (ddd, 13.2, 6.7, 6.7) 20, 23, 24, 26 19, 26, 20-NH
    23 34.0 CH 1.74 (m) 22, 24 22a, 22b, 24a, 26
    24 27.2 CH2 1.44 (m) 23, 26 23, 24b, 25
    1.24 (m) 26 24a, 25
    25 11.1 CH3 0.93
    Figure US20190290617A1-20190926-P00899
    (t, 7.4)
    23, 24 24a, 24b
    26 17.4 CH3 0.96
    Figure US20190290617A1-20190926-P00899
    (d, 6.8)
    22, 23, 24 22a, 22b, 23
    20-NH 6.53 (brs) 22a, 22b
    Figure US20190290617A1-20190926-P00899
    2H,
    Figure US20190290617A1-20190926-P00899
    3H.
    Figure US20190290617A1-20190926-P00899
    indicates data missing or illegible when filed
  • <Preparation Example 16> Preparation of 3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(3-methylbutylamino)cyclohexa-3,5-diene-1,2-dione
  • A compound of Preparation Example 16 was prepared in the same manner as in Preparation Example 15. It had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(3-methylbutylamino)cyclohexa-3,5-diene-1,2-dione’.
  • Purple-Red, Amorphous Solid.
  • [α]21 D+124 (c 0.25, CHCl3).
  • IR (film) νmax 3270, 1680, 1640, 1590, 1510, 1380, 1210 cm−1.
  • UV (MeOH) λmax 336 (log 4.17), 515 nm (2.55).
  • 1H NMR (CDCl3): see [Table 11].
  • 13C NMR (CDCl3): see [Table 11].
  • EIMS m/z (%) 413 (M+, 7), 223 (100), 191 (3), 166 (8), 152 (9), 107 (8), 95 (18);
  • HREIMS m/z 413.2947 [M]+ (calcd for C26H39NO3, 413.2930).
  • TABLE 11
    Pos. δC δH (m, J in Hz) HMBC NOESY
     1 20.1 CH2 2.03 (m) 2, 3, 5, 9, 10 1b, 15a
    1.44 (dddd, 12.1, 12.1, 12.1, 6.2) 2, 5, 10 1a, 8
     2 27.0 CH2 1.98 (ddd, 17.5, 5.3, 5.3) 2b, 3
    1.85 (m) 2a, 3
     3 120.7 CH 5.10 (brs) 11 2a, 2b, 11
     4 144.0 C
     5 38.4 C
     6 35.9 CH2 1.60 (ddd, 12.8, 3.3, 3.3) 6b, 7
    1.03 (m) 6a, 8
     7 27.9 CH2 1.33
    Figure US20190290617A1-20190926-P00899
    (m)
    5, 6, 8, 9
     8 37.6 CH 1.25 (m) 14 1b
     9 42.6 C
    10 47.5 CH 1.03 (m)  8
    11 18.1 CH3 1.52b (brs) 3, 4, 5 3, 12
    12 19.8 CH3 0.98b (s) 4, 5, 6, 10 1b, 6a, 14
    13 17.7 CH3 0.94b (d, 6.2) 7, 9 7, 15b
    14 17.2 CH3 0.81b (s) 8, 9, 10, 15 1b, 8, 12, 15a, 15b
    15 32.4 CH2 2.54 (d, 14.0) 8, 9, 14, 16, 17, 21 1a, 14, 15a
    2.41 (d, 14.0) 8, 9, 10, 14, 16, 17, 21 13, 14, 15b
    16 113.8 C
    17 157.2 C
    18 178.0 C
    19 91.5 CH 5.37 (s) 17, 21 22, 23, 25
    20 150.3 C
    21 182.8 C
    22 41.1 CH2 3.15
    Figure US20190290617A1-20190926-P00899
    (dt, 7.3, 6.6)
    20, 23, 24 19, 23, 25
    23 36.8 CH2 1.55b (td, 7.3, 6.7) 22, 24, 25 19, 22, 25
    24 25.9 CH 1.66 (d sept, 6.7, 6.7) 23, 25 25
    25 22.3d CH3 0.93c (d, 6.7) 23, 24 19, 23, 24
    17-OH 7.92 (brs)
    20-NH 6.46 (brs) 19, 21 22
    a2H,
    b3H,
    c6H,
    d2C.
    Figure US20190290617A1-20190926-P00899
    indicates data missing or illegible when filed
  • <Preparation Example 17> Preparation of 3-[[(1R,2S,8aS)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-[[(2S)-2-methylbutyl]amino]cyclohexa-3,5-diene-1,2-dione
  • The fraction 6 obtained in Preparation Example 14 was refractionated by Cis HPLC (Luna 5u Phenyl-Hexyl, 250×10 mm; MeOH/H2O/Et2NH, 65:35:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm) to obtain a compound of Preparation Example 17 (0.7 mg, 0.00023%) and a compound of Preparation Example 18 (1.6 mg, 0.00053%).
  • The obtained Preparation Example 17 compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,8aS)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-[[(2S)-2-methylbutyl]amino]cyclohexa-3,5-diene-1,2-dione’.
  • Purple-Red, Amorphous Solid.
  • [α]23 D−42 (c 0.25, CHCl3).
  • IR (film) λmax 3290, 1680, 1650, 1590, 1520, 1460, 1390, 1200 cm−1.
  • UV (MeOH) λmax 338 (log 4.06), 511 nm (2.63).
  • 1H NMR (CDCl3): see [Table 12].
  • 13C NMR (CDCl3): see [Table 12].
  • EIMS m/z (%) 413 (M+, 15), 223 (100), 191 (10), 168 (15), 166 (14), 152 (16), 119 (18).
  • HREIMS m/z 413.2916 [M]+ (calcd for C26H39NO3, 413.2930).
  • TABLE 12
    Pos. δC δH (m, J in Hz) HMBC NOESY
     1 30.6 CH2 1.87 (m) 1b, 2a, 2b, 10, 14, 15a
    0.96 (m) 1a
     2 22.8 CH2 1.50 (m) 1a, 1b, 2b
    1.40 (m) 2a, 3a, 3b
     3 41.4 CH2 1.38 (m) 2a, 2b, 3b
    1.16 (ddd, 13.1, 13.1, 4.4) 2a, 2b, 3a
     4 36.3 C
     5 146.5 C
     6 114.9 CH 5.40 (brs) 4, 7, 8, 10 7a, 7b, 11
     7 31.6 CH2 1.97 (ddd, 17.6, 4.3, 4.3) 5 7b, 8, 13
    1.78 (m) 5, 6 6, 7a
     8 36.4 CH 1.40 (m) 7a, 10, 12
     9 40.6 C
    10 41.6 CH 2.11 (m) 1a, 8, 12, 15b
    11 29.7 CH3 1.03b (brs) 3, 4, 5, 12 3a, 3b
    12 28.0 CH3 0.96b (a) 3, 4, 5, 11 8, 10
    13 16.6 CH3 1.00b (d, 6.7) 7, 8, 9 7a, 8, 14, 15a, 15b
    14 15.9 CH3 0.74b (s) 8, 9, 10, 15 1a, 7b, 13, 15a, 15b
    15 32.8 CH2 2.56 (d, 13.5) 8, 9, 10, 14, 16, 17, 21 1a, 13, 14, 15b
    2.42 (d, 13.5) 8, 9, 10, 14, 16, 17, 21 8, 10, 13, 14, 15a
    16 114.5 C
    17 156.7 C
    18 178.3 C
    19 91.5 CH 5.40 (s) 17, 21 22a, 22b
    20 150.5 C
    21 183.1 C
    22 48.7 CH2 3.11 (ddd, 13.1, 6.4, 6.4) 20, 23, 24, 26 19, 22b, 23, 26
    3.00 (ddd, 13.1, 6.7, 6.7) 20, 23, 24, 26 19, 22a, 23, 26
    23 34.0 CH2 1.76 (m) 22, 24, 25, 26 22a, 22b, 24a, 26
    24 27.2 CH 1.44 (m) 22, 23, 25, 26 23, 24b, 25
    1.23 (m) 22, 23, 25, 26 24a
    25 11.1 CH3 0.93b (t, 7.5) 23, 24 24a
    26 17.3 CH3 0.97b (d, 6.7) 22, 23, 24 22a, 22b, 23
    20-NH 6.58 (brs)
    a2H.
    b3H.
  • <Preparation Example 18> Preparation of 3-[[(1R,2S,8aS)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(3-methylbutylamino)cyclohexa-3,5-diene-1,2-dione
  • A compound of Preparation Example 18 was prepared in the same manner as in Preparation Example 17. It had the following physicochemical properties and was identified as ‘3-[[(1R,2S,8aS)-1,2,5,5-tetramethyl-2,3,6,7,8,8a-hexahydronaphthalen-1-yl]methyl]-4-hydroxy-5-(3-methylbutylamino)cyclohexa-3,5-diene-1,2-dione’ Compound
  • Purple-Red, Amorphous Solid.
  • [α]21D-38 (c 0.2, CHCl3).
  • IR (film) νmax 3270, 1680, 1650, 1590, 1510, 1460, 1380, 1200 cm−1.
  • UV (MeOH) λmax 338 (log 4.21), 515 nm (2.63).
  • 1H NMR (CDCl3): see [Table 13].
  • 13C NMR (CDCl3): see [Table 13].
  • EIMS m/z (%) 413 (M+, 24), 223 (100), 191 (13), 166 (20), 152 (17), 119 (20).
  • HREIMS m/z 413.2947 [M]+ (calcd for C29H37NO3, 413.2930).
  • TABLE 13
    Pos. δC δH (m, J in Hz) HMBC NOESY
     1 30.6 CH2 1.87 (m) 1b, 2a, 2b, 10, 14, 15a
    0.93 (m) 1a, 2a, 2b, 14
     2 22.8 CH2 1.50 (m) 1a, 1b, 2b, 3a, 3b
    1.38 (m) 3a, 3b, 12
     3 41.4 CH2 1.38 (m) 2a, 2b
    1.16 (ddd, 13.0, 13.0, 4.2) 2a, 2b
     4 36.4 C
     5 146.5 C
     6 114.9 CH 5.39 (brs) 4, 7, 8, 10 7a, 7b, 11
     7 31.6 CH2 1.98 (m) 6, 7b, 8, 13
    1.77 (m)  8 6, 7a, 13, 14
     8 36.3 CH 1.38 (m) 7a, 7b, 10, 15b
     9 40.6 C
    10 41.6 CH 2.11 (m) 1a, 8, 12, 15a
    11 29.7 CH3 1.03b (brs) 3, 4, 5, 12  6
    12 28.0 CH3 0.95b (s) 3, 4, 5, 11 2b, 10
    13 16.6 CH3 0.99b (d, 6.7) 7, 8, 9 7a, 7b, 8, 14, 15a, 15b
    14 16.0 CH3 0.74b (s) 8, 9, 10, 15 1a, 1b, 7b, 13, 15a, 15b
    15 32.7 CH2 2.55 (d, 13.4) 8, 9, 10, 14, 16, 17, 21 1a, 13, 14, 15b
    2.42 (d, 13.4) 8, 9, 10, 14, 16, 17, 21 8, 10, 13, 14, 15a
    16 114.5 C
    17 156.7 C
    18 178.3 C
    19 91.5 CH 5.39 (s) 17, 21
    20 150.1 C
    21 183.1 C
    22 41.1 CH2 3.18a (td, 6.7, 6.3) 20, 23, 24 23, 25
    23 36.9 CH2 1.57a (dt, 6.7, 6.7) 22, 24, 25 22, 25
    24 25.9 CH 1.68 (d sept, 6.7, 6.7) 25 25
    25 22.3d CH3 0.95c (d, overlapped) 23, 24 22, 23, 24
    20-NH 6.43 (brs)
    a2H.
    b3H.
    c6H.
    d2C.
  • <Preparation Example 19> Preparation of 3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-[[(2S)-2-methylbutyl]amino]cyclohexa-3,5-diene-1,2-dione
  • A sponge (Spongiidae SS-265) was obtained as described in the literature ‘Yohei Takahashi et al., 2010’ and extracted. Briefly, an extract was prepared by adding MeOH (4.3 and 3.2 L) to sponge SS-265 (1.4 kg, wet weight). The MeOH extract (68.4 g) was fractionated with CHCl3 and H2O. The CHCl3-soluble substances (2.3 g) were subjected to silica gel column (n-hexane/EtOAc), C18 column (MeOH/H2O), silica gel column (n-hexane/acetone) and C18 HPLC (Wakosil-II 5C18AR, 250×10 mm; eluent, MeCN/H2O/CF3CO2H, 90:10:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm and Luna 5u C18(2), 250×10 mm; MeOH/H2O/Et2NH, 70:30:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm) repeatedly to obtain a compound of Preparation Example 19 (1.8 mg, 0.00013%).
  • The obtained Preparation Example 19 compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-[[(2S)-2-methylbutyl]amino]cyclohexa-3,5-diene-1,2-dione’.
  • Purple-Red, Amorphous Solid.
  • [α]22 D+33 (c 0.2, CHCl3).
  • IR (film) λmax 3280, 1640, 1590, 1510, 1380, 1200 cm1.
  • UV (MeOH) λmax 501 (log 2.88), 327 (4.17), 243 (3.86), 208 nm (4.25).
  • 1H NMR (CDCl3): see [Table 14].
  • 13C NMR (CDCl3): see [Table 14].
  • EIMS m/z (%) 413 (M+, 15), 223 (100), 191 (3), 166 (10), 152 (10), 95 (10).
  • HREIMS m/z 413.2934 [M]+ (calcd for C26H39NO3, 413.2930).
  • TABLE 14
    Pos. δC δH (m, J in Hz) HMBC NOESY
     1 23.2 CH2 2.09 (m) 3, 5 1b, 8, 10, 15a
    1.43 (m) 1a, 2a, 3a
     2 28.7 CH2 1.84 (m) 1b, 2b, 3a, 3b
    1.14 (m) 2a, 3b
     3 33.0 CH2 2.32 (ddd, 13.7, 13.7, 5.4) 2, 4, 11 1b, 2a, 3b, 12
    2.05 (m) 1.5 2a, 2b, 3a, 11a
     4 160.5 C
     5 40.4 C
     6 36.7 CH2 1.51 (m) 5, 7, 8, 10 6b, 11b
    1.36 (m) 6a, 11b
     7 28.0 CH2 1.39a (m) 12, 14
     8 37.9 CH 1.18 (m) 10
     9 42.9 C
    10 50.0 CH 0.78 (dd, 11.6, 1.8) 1, 5, 8, 9, 12, 14, 15 1a, 2b, 6b, 8, 15a
    11 102.5 CH2 4.43 (s) 3, 4, 5 3b
    4.42 (s) 3, 4, 5 6a, 6b
    12 20.5 CH3 1.04b (s) 4, 5, 6, 10 3a, 7, 14
    13 17.9c CH3 0.96b,c (d, 6.4) 7, 8, 9 7, 15b
    14 17.2 CH3 0.82b (s) 8, 9, 10, 15 7, 12, 15a, 15b
    15 32.5 CH2 2.48 (d, 14.0) 8, 9, 10, 14, 16, 17, 21 1a, 10, 14, 15b
    2.39 (d, 14.0) 8, 9, 10, 14, 16, 17, 21 8, 13, 14, 15a
    16 113.5 C
    17 157.3 C
    18 178.1 C
    19 91.6 CH 5.36 (s) 17, 21 22a, 22b, 23
    20 150.5 C
    21 182.9 C
    22 48.7 CH2 3.08 (ddd, 13.2, 6.6, 6.6) 20, 23, 24, 26 19, 22b, 23, 24a, 20-NH
    2.95 (ddd, 13.2, 6.7, 6.7) 20, 23, 24, 26 19, 22a, 23, 20-NH
    23 34.0 CH2 1.75 (m) 22, 24, 25, 26 22a, 22b, 24a, 24b, 25, 26
    24 27.2 CH 1.45 (m) 22, 23, 25, 26 22a, 23, 24b, 25
    1.23 (m) 22, 23, 25, 26 23, 24a
    25 11.1 CH3 0.93b (t, 7.4) 23, 24 23, 24a
    26 17.4c CH3 0.96b,c (d, 6.8) 22, 23, 24 23
    20-NH 6.51 (brs) 22a, 22b, 23, 26
    a2H.
    b3H.
    cinterchangeable.
  • <Preparation Example 20> Preparation of 2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dienl-yl]amino]ethanesulfonic acid
  • A sponge (Spongiidae SS-1208) was obtained as described in the literature ‘Yohei Takahashi et al., 2010’ and extracted. Briefly, an extract was prepared by adding MeOH (3×0.8 L) and MeOH/toluene (3:1, 1×0.8 L) to sponge SS-1208 (0.4 kg, wet weight). The extract mixture (15.9 g) was fractionated with CHCl3 and H2O (3×500 mL). The CHCl3-soluble fraction (2.7 g) was subjected to silica gel column (n-hexane/EtOAc and CHCl3/MeOH), C18 column (MeOH/H2O/CF3CO2H) and C18 HPLC (Luna 5u Phenyl-Hexyl, 250×10 mm; eluent, MeCN/H2O/CF3CO2H, 70:30:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm and Wakosil-II 5C18AR, 250×10 mm; eluent, MeCN/H2O/CF3CO2H, 75:25:0.1; flow rate, 2.0 mL/min; UV detection at 300 nm) repeatedly to obtain a compound of Preparation Example 20 (0.8 mg, 0.00020%).
  • The obtained Preparation Example 20 compound had the following physicochemical properties and was identified as ‘2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]ethanesulfonic acid’.
  • Purple-Red, Amorphous Solid.
  • [α]22 D+38 (c 0.2, MeOH).
  • IR (KBr) νmax 3450, 1640, 1600, 1530, 1380, 1210 cm1.
  • UV (MeOH) λmax 237 (log 2.8), 345 (4.00), 513 nm (2.47).
  • 1H NMR (DMSO-d6): see [Table 15].
  • 13C NMR (DMSO-d6): see [Table 15].
  • ESIMS (neg) m/z 450 [M−H].
  • HRESIMS (neg) m/z 450.1955 [M−H] (calcd for C23H32NO6S, 450.1950).
  • TABLE 15
    Pos. δC δH (m, J in Hz) HMBC NOESY
     1 19.4 CH2 1.99 (m) 1b, 15a
    1.33 (m) 1a, 2, 12, 14
     2 26.3 CH2 1.88a (m) 1b, 10
     3 120.8 CH 5.05 (brs) 12  2
     4 143.1 C
     5 37.8 C
     6 35.4 C 1.53 (m) 6b, 7, 12
    0.96 (m) 6a
     7 27.5 CH2 1.27a (m) 6a, 13
     8 37.1 CH 1.22 (m) 13, 14
     9 41.8 C
    10 47.0 CH 0.96 (m) 14 2, 15b
    11 17.9 CH3 1.47b (brs) 3, 4, 5
    12 19.9 CH3 0.93b (s) 4, 5, 6, 10 1b, 6a, 14
    13 17.8 CH3 0.90b (d, 6.0) 7, 8, 9 7, 8
    14 17.2 CH3 0.74b (s) 8, 9, 10, 15 1b, 8, 12, 15a
    15 32.0 CH3 2.41 (d, 13.6) 16, 17, 21 1a, 15b
    2.30 (d, 13.6) 9, 10, 16, 10, 15a
    17, 21
    16 113.6 C
    17 158.8c C
    18 178.0 C
    19 91.6 CH 5.26 (s) 17, 21 22, 23
    20 dC
    21 182.7c C
    22 39.2c CH2 3.33a (overlapped) 23
    23 48.0 CH2 2.69a (brt, 6.4) 22 19, 20-NH
    20-NH 7.96 (brs) 23
    a2H,
    b3H.
    cassigned from HMBC spectrum.
    dnot observed.
  • <Preparation Example 21> Preparation of methyl 3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxybenzoate
  • After freeze-drying a sponge (Dactylospongia elegans), the freeze-dried sponge (2.6 kg, wet weight) was chopped and immersed in MeOH overnight to prepare an extract. The MeOH extract was fractionated with hexane, 90% methanol, n-BuOH and H2O. The 90% methanol fraction was evaporated under reduced pressure to obtain 12 g of a 90% MeOH extract. 2 g of the 90% MeOH extract was separated by SiO2 column (hexane-AcOEt-acetone-MeOH) to obtain three fractions, Fr. A (0.42 g), Fr. B (0.73 g) and Fr. C (0.83 g). Among the fractions, Fr. A and Fr. B were separated by ODS column (MeOH—H2O) or HPLC (Cosmosil 5SL, hexane-AcOEt=7:1) to obtain a compound of Preparation Example 21 (20 mg, 1%) and a compound of Preparation Example 22 (17 mg, 0.85%).
  • The obtained Preparation Example 21 compound had the following physicochemical properties and was identified as ‘methyl 3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxybenzoate’.
  • White Solid.
  • [α]27D+17.3 (c 0.12, CHCl3).
  • 1H NMR (500 MHz, CDCl3) δ: 7.77 (1H, s), 7.77-7.74 (1H, m), 6.77 (1H, d, J=8.0 Hz), 6.01 (1H, s), 4.41 (1H, s), 4.36 (1H, s), 3.87 (3H, s), 2.68 (1H, d, J=14.3 Hz), 2.64 (1H, d, J=14.3 Hz), 2.33 (1H, td, J=13.7, 5.2 Hz), 2.08 (2H, d, J=13.7 Hz), 1.93-1.89 (1H, m), 1.61-1.56 (1H, m), 1.47 (1H, dt, J=12.2, 3.2 Hz), 1.41-1.38 (3H, m), 1.31-1.27 (1H, m), 1.22-1.19 (1H, m), 1.06 (3H, s), 1.02 (3H, d, J=6.9 Hz), 0.96 (1H, dd, J=12.0, 1.7 Hz), 0.88 (3H, s).
  • 13C NMR (125 MHz, CDCl3) δ: 167.6, 160.0, 159.2, 135.0, 129.3, 125.2, 121.6, 115.3, 102.8, 52.0, 48.0, 42.0, 40.2, 37.0, 36.5, 36.3, 33.0, 27.8, 27.7, 23.2, 20.5, 17.62, 17.59.
  • IR (KBr): 3341, 1686, 1601, 1426, 1287 cm1′.
  • MS (ESI-TOF) m/z: 379 [M+Na]+.
  • HRMS (ESI-TOF) m/z: 379.2249 (calcd for C23H32O3Na; found: 379.2266).
  • <Preparation Example 22> Preparation of methyl 3-[[(1 S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4,5-dihydroxybenzoate
  • A compound of Preparation Example 22 was prepared in the same manner as in Preparation Example 21. It had the following physicochemical properties and was identified as ‘methyl 3-[[(1 S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4,5-dihydroxybenzoate’.
  • White Solid.
  • [α]26: D+10.4 (c 0.19, CHCl3).
  • 1H NMR (500 MHz, CDCl3) δ: 7.49 (1H, d, J=2.0 Hz), 7.40 (1H, d, J=2.0 Hz), 5.90 (2H, S), 4.41 (1H, s), 4.37 (1H, s), 3.87 (3H, s), 2.68 (1H, d, J=14.3 Hz), 2.65 (1H, d, J=14.3 Hz), 2.34 (1H, td, J=13.7, 5.0 Hz), 2.09 (2H, d, J=14.3 Hz), 1.93-1.91 (1H, m), 1.60-1.55 (1H, m), 1.47 (1H, dt, J=11.6, 2.7 Hz), 1.43-1.35 (3H, m), 1.33-1.19 (2H, m), 1.06 (3H, s), 1.03 (3H, d, J=6.3 Hz), 0.96 (1H, d, J=11.5 Hz), 0.88 (3H, s).
  • 13C NMR (125 MHz, CDCl3) δ: 167.7, 160.1, 148.7, 142.3, 127.4, 125.2, 120.3, 114.0, 102.8, 52.1, 48.0, 42.1, 40.2, 37.0, 36.5, 36.3, 33.0, 27.9, 27.7, 23.2, 20.6, 17.64, 17.59.
  • IR (KBr): 3341, 1686, 1601, 1426, 1287 cm1.
  • MS (ESI-TOF) m/z: 395 [M+Na]+.
  • HRMS (ESI-TOF) m/z: 395.2198 (calcd for C23H32O4Na; found: 395.2214).
  • <Preparation Example 23> Preparation of (−)-(1R,4aS,8aS)-1β,2β,4β,-trimethyl-1α[(2′,5′-dimethoxyphenyl)methyl]-5-exo-methylene-(3H)-1,4,4a,5,6,7,8,8aα-octahydronaphthalene
  • After adding anhydrous 95% potassium tert-butoxide (217 mg, 1.93 mmol) to 7.5 mL of benzene, a suspension was prepared by stirring the same. Then, 657 mg (0.62 mmol) of methyltriphenylphosphonium bromide was added. The prepared light yellow solution was heated for 30 minutes under reflux. A heated ylide solution was added dropwise to a solution of 212 mg (0.62 mmol) of the ketone (−)-(1R,4aS,8aS)-1β,2β, 4aβ-trimethyl-1α[(2′,5′-dimethoyphenyl)methyl]-1,2,3,4,4a,5,6,7,8,8aα-decahydronaphthalen-5-one dissolved in 3 mL of benzene. After heat-treating further for 22 hours, the reaction mixture was cooled and diluted by stirring fast while sequentially adding 10 mL of ether and 3 mL of H2O. After phase separation was completed, the organic phase was washed with 2 mL of H2O and 3 mL of saturated brine and then dried (MgSO4). An almost colorless oil obtained by concentrating under reduced pressure was separated by silica gel column (10×2.5 cm) chromatography using 5% EtOAc (in hexane) as an eluent to obtain a compound of Preparation Example 23 (180 mg, 85%).
  • The obtained Preparation Example 23 compound had the following physicochemical properties and was identified as ‘(−)-(1R,4aS,8aS)-1β,2β,4β,-trimethyl-1α[(2′,5′-dimethoxyphenyl)methyl]-5-exo-methylene-(3H)-1, 4,4a,5,6,7,8,8aα-octahydronaphthalene’.
  • [α]25 D−40.4° (c 0.5, CH2Cl2).
  • m.p.: 77-78° C.
  • Silica gel TLC Rf 0.70 (15% EtOAc in hexane).
  • 1H NMR (CDCl3) δ 0.86 (s, 3H), 1.01 (d, 3H, J=5.5 Hz), 1.07 (s, 3H), 1.15-1.65 (m, 7H), 1.70-1.95 (m, 2H), 2.05-2.15 (m, 2H), 2.20-2.45 (m, 1H), 2.64 (AB q, 2H, J=14 Hz), 3.72 (s, 3H), 3.75 (s, 3H), 4.33-4.47 (m, 2H), 6.65-6.77 (m, 3H).
  • Anal calcd for C23H34O2: C, 80.65; H, 10.00. found: C, 80.82; H, 10.04.
  • <Preparation Example 24> Preparation of dimethyl ether of 2-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]benzene-1,4-diol
  • The compound of Preparation Example 23 (108.5 mg, 0.317 mmol) and rhodium trichloride hydrate (16.7 mg, 0.06 mmol, 20 mol %) were added to 11 mL of EtOH solution and the prepared mixture was heated under reflux. After heat-treating for 20 hours, the reaction mixture was cooled and quenched by adding 5 mL of H2O. The aqueous phase was extracted three times with 10 mL of CH2Cl2 and the extract was combined, dried (MgSO4) and concentrated to obtain a faintly colored oil. The residue was purified with a silica gel (10% EtOAc in hexane) plug and concentrated to obtain a clear colorless oil. Then, a compound of Preparation Example 24 was obtained by slowly solidifying the same under a high-pressure condition. The obtained Preparation Example 24 compound had the following physicochemical properties and was identified as ‘dimethyl ether of 2-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]benzene-1,4-diol’.
  • [α]25 D+8.88° (c 0.18, CH2Cl2).
  • m.p.: 63-68° C.
  • Silica gel TLC Rf 0.71 (15% EtOAc in hexane), 0.37 (5% EtOAc in hexane).
  • 1H NMR (CDCl3) δ 0.75-1.15 (m, 4H), 0.87 (s, 3H), 1.01 (s, 3H), 1.24-1.65 (m, 9H), 2.0-2.15 (br m, 3H), 2.70 (br s, 2H), 3.72 (s, 3H), 3.75 (s, 3H), 5.15 (br s, 1H), 6.65-6.85 (m, 3H).
  • Mass spectrum (chemical ionization, negative ion), m/z 341 (M−1).
  • <Preparation Example 25> Preparation of 2-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]cyclohexa-2,5-diene-1,4-dione
  • A solution obtained by adding the compound of Preparation Example 24 (70.0 mg, 0.204 mmol) to 3.5 mL of THF was added dropwise to 448 mg (0.82 mmol) of a ceric ammonium nitrate solution (in 3.5 mL of H2O) under stirring. 15 minutes later, the reaction mixture was diluted sequentially with 3 mL of saturated brine and 10 mL of ethyl ether. After phase separation was completed, the aqueous phase fraction was extracted three times with 10 mL of CH2Cl2. The extracted solution was combined, dried (MgSO4), concentrated and purified by silica gel column (15×2 cm) chromatography to obtain an orange oil. It was eluted with 5% EtOAc (in hexane) to obtain a compound of Preparation Example 25 (25 mg, 40%). The obtained compound had the following physicochemical properties and was identified as ‘2-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]cyclohexa-2,5-diene-1,4-dione’.
  • [α]25 D+21° (c 0.02, CH2Cl2).
  • Silica gel TLC Rf 0.55 (15% EtOAc in hexane); λmax (CH3OH) 292 nm.
  • 1H NMR (CDCl3) δ 0.80-2.15 (m, 5H), 0.85 (s, 3H), 0.93 (d, 3H, J=6.5 Hz), 1.00 (s, 3H), 1.53 (br s, 1H), 2.45-2.67 (AB q, 2H, J=13.5 Hz), 5.14 (br s, 1H), 6.51 (br s, 1H), 6.71 (m, 2H).
  • Mass spectrum (chemical ionization) m/z 312 [M+1]+; mass spectrum (electron impact), m/z 311.199 (C21H27O2 requires 311.201).
  • <Preparation Example 26> Preparation of 2-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]benzene-1,4-diol
  • 25 mg (0.08 mmol) of the compound of Preparation Example 25 was dissolved in 2 mL of ethyl ether and a Na2S2O4 solution (56 mg Na2S2O4 in 2 mL of H2O, 0.32 mol) was added dropwise to the resulting solution under vigorous stirring. 45 minutes later, the reaction mixture was diluted with 2 mL of saturated brine and 10 mL of ethyl ether. After phase separation was completed, the extracted aqueous phase fraction was further extracted three times with 10 mL of ethyl ether. The extracted ether solution was combined, dried (Na2SO4), concentrated and purified by silica gel column (18×1 cm) chromatography to obtain an oily residue. It was eluted with a 15% EtOAc (in hexane) solution to obtain a clear colorless oil. The oil was solidified in vacuo to obtain a compound of Preparation Example 26 (23 mg, 92%). The obtained compound had the following physicochemical properties and was identified as ‘2-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]benzene-1,4-diol’.
  • (+)-1: [α]25 D+22.0° (c 1.35, CDCl3).
  • (−)-1: [α]25 D−19.5° (c 1.0, CKCl3).
  • m.p.: 125-127° C.
  • Silica gel TLC Rf 0.10 (15% EtOAc in hexane); λmax (DMSO) 305 nm.
  • 1H NMR (CDCl3) δ 0.86 (s, 3H), 0.99 (d, 3H, J=8 Hz), 1.02 (s, 3H), 1.51 (br s, 3H), 1.2-1.65 (m, 7H), 1.9-2.15 (m, 3H), 2.54-2.70 (AB q, 2H, J=14 HZ), 4.38 (br s, 1H), 4.41 (br s, 1H), 5.14 (br s, 1H) and 6.59 (m, 3H); mass spectrum (chemical ionization), m/z 315 [M+1]+.
  • Mass spectrum (electron impact), m/z 314.225 [M]+ (C21H30O2 requires 314.225).
  • <Preparation Example 27> Preparation of 2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dienl-yl]amino]acetic acid
  • The compound of Preparation Example 10 (3.0 mg, 8.4 μmol) and glycine (0.8 mg, 10 mol) were added to EtOH (1 mL) and stirred for 24 hours at room temperature in the presence of NaHCO3 (11 mg, 130 μmol). A residue prepared through filtration and evaporation was subjected to C18 reversed-phase HPLC (YMC-Pack AM-323, 1.0×25 cm; flow rate 2.5 mL/min; UV detection at 300 nm; eluent CH3CN/H2O/CF3CO2H, 85:15:0.1) to obtain a compound of Preparation Example 27 (1.6 mg, 47%).
  • The obtained Preparation Example 27 compound had the following physicochemical properties and was identified as ‘2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]acetic acid’.
  • m.p.: 156-158° C.
  • [α]20 D−71.7θ (C 1.0, MeOH).
  • IR (KBr) νmax 3300, 1720, 1640, 1580, 1370, 1200 cm1.
  • UV(MeOH) λmax 317 (c 11800) and 488 nm (860).
  • ELMS m/z (%) 401 (M+, 1). 385(1), 357(4), 343(3), 211(20), 191(25) and 95(100).
  • FABMS (positive) m/z 404 [M+2H+H]+; HRFABMS m/z 404.2461 [M+2H+H]+, calcd for C23H34NO5, 404.2437.
  • 1H NMR (CD3OD): see [Table 16].
  • 13C NMR (CD3OD): see [Table 16].
  • TABLE 16
    position 1H a J(Hz) 13Ca H coupled with C b
    1 2.10 m 21.1 t
    1.44 m
    2 1.93c m 28.0 t H-10
    3 5.08 brs 121.9 d H-11
    4 144.9 s H-11, H-12
    5 39.6 s H-10, H-11, H-12
    6 1.63 m 37.4 t H-12
    1.03 m
    7 1.36c m 29.2 t H-13
    8 1.32 m 39.0 d H-10, H-14, H-15
    9 43.6 s H-10, H-13, H-14, H-15
    10 1.10 m 49.9 d H-12, H-14, H-15
    11 1.50 s 18.4 q
    12 1.00 s 20.7 q H-10
    13 0.97 d 7.0 18.4 q
    14 0.82 s 17.8 q H-10, H-15
    15 2.57 d 13.6 33.3 t H-14
    2.42 d 13.6
    16 115.9 s H-15
    17 159.6 s H-15, H-19
    18 180.8 s
    19 5.28 s 93.8 d
    20 151.5 s H-22
    21 184.0 s H-15, H-19
    22 3.96 s 44.9 t
    23 171.9 s H-22
    aδ in ppm
    bHMBC correlations
    c2H
  • <Preparation Example 28> Preparation of (2S)-2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dienl-yl]amino]-3-hydroxypropanoic acid
  • The compound of Preparation Example 10 (3.0 mg, 8.4 μmol) and L-serine (1.3 mg, 10 μmol) were added to EtOH (1 mL) and stirred for 24 hours at 40° C. in the presence of NaHCO3 (27 mg, 34 μmol). A residue prepared through filtration and evaporation was subjected to C18 reversed-phase HPLC (YMC-Pack AM-323, 1.0×25 cm; flow rate 2.5 mL/min; UV detection at 300 nm; eluent CH3CN/H2O/CF3CO2H, 85:15:0.1) to obtain a compound of Preparation Example 28 (1.7 mg, 46%).
  • The obtained Preparation Example 28 compound had the following physicochemical properties and was identified as ‘(2S)-2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]-3-hydroxypropanoic acid’.
  • m.p.: 198-200° C.
  • [α]17 D−71° (c 0.73, EtOH).
  • IR (K Br) νmax 3400, 1670, 1630, 1590, 1540, 1380, 1200 cm1.
  • UV (MeOH) λmax 321 (c 12100) and 498 nm (920).
  • FABMS (negative, diethanolamine matrix) m/z 432 [M+2H−H].
  • HRFABMS m/z 432.2381 [M+2H−H], calcd for C24H34NO6 432.2386.
  • 1H NMR (DMSO-d6): see [Table 17].
  • 13C NMR (DMSO-d6): see [Table 17].
  • TABLE 17
    position 1H a J(Hz) 13Ca H coupled with Cb
     1 2.01 m 18.9 t H-10
    1.39 m H-10
     2 1.92c m 25.8 t
     3 5.05 brs 121.0 d H3-11
     4 142.8 s H3-11, H3-12
     5 37.2 s H-3, H3-11, H3-12
     6 1.53 m 34.9 t H3-12
    0.98 m
     7 1.33c m 26.9 t H3-13
     8 1.30 m 36.8 d H-6, H-10, H3-14, H2-15
     9 41.2 s H-10, H3-13, H3-14, H2-15
    10 1.02 m 46.5 d H3-12, H3-14, H2-15
    11 1.48 s 17.5 q H-3
    12 0.93 s 19.3 q
    13 0.90 d  7.0 17.2 q
    14 0.78 s 16.5 q H-10, H2-15
    15 2.43 d 13.6 31.4 t H3-14
    2.32 d 13.6
    16 113.5 s H2-15
    17 158.5 s H2-15, H-19
    18 178.8 s H-19, NH-20
    19 5.35 s 93.1 d NH-20
    20 147.1 s
    20-NH 7.15 d  8.0
    21 182.0 s H2-15, H-19, NH-20
    22 4.20 m 56.5 d
    23 171.9 s H2-24
    24 3.78 dd 11.4, 2.9 59.8 t
    3.82 dd 11.4, 2.9
    aδ in ppm
    bHMBC correlations
    c2H
  • <Preparation Example 29> Preparation of (2S)-2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien 1-yl]amino]-3-hydroxybutanoic acid
  • The compound of Preparation Example 10 (3.0 mg, 8.4 μmol) and L-threonine (1.3 mg, 13 μmol) were added to EtOH (1 mL) and stirred for 24 hours at 40° C. in the presence of NaHCO3 (11 mg, 130 μmol). A residue prepared through filtration and evaporation was subjected to C18 reversed-phase HPLC (YMC-Pack AM-323, 1.0×25 cm; flow rate 2.5 mL/min; UV detection at 300 nm; eluent CH3CN/H2O/CF3CO2H, 85:15:0.1) to obtain a compound of Preparation Example 29 (1.3 mg, 35%).
  • The obtained Preparation Example 29 compound had the following physicochemical properties and was identified as ‘(2S)-2-[[5-[[(1R,2S,4aS,8aS)-1,2,4a,5-tetramethyl-2,3,4,7,8,8a-hexahydronaphthalen-1-yl]methyl]-6-hydroxy-3,4-dioxocyclohexa-1,5-dien-1-yl]amino]-3-hydroxybutanoic acid’.
  • m.p.: 188-191° C.
  • [α]17 D−183° (c 1.0, EtOH).
  • IR (K Br) νmax 3400, 1670, 1630, 1590, 1540, 1380, 1200 cm−1.
  • UV (MeOH) λmax 317 (c 12600) and 490 nm (1000).
  • FABMS (negative, diethanolamine matrix) m/z 446 [M+2H+H].
  • HRFABMS m/z 446.2524 [M+2H−H], calcd for C25H36NO6, 446.2906.
  • 1H NMR (DMSO-d6): see [Table 18].
  • 13C NMR (DMSO-d6): see [Table 18].
  • TABLE 18
    position 1H a J(Hz) 13Ca H coupled with C b
     1 1.99 m 19.9 t H-10
    1.35 m H-10
     2 1.88c m 26.2 t
     3 5.05 brs 120.5 d H3-11
     4 143.5 s H3-11, H3-12
     5 37.5 s H-3, H3-11, H3-12
     6 1.56 m 35.7 t H3-12
    0.95 m
     7 1.28c m 28.5 t H3-13
     8 1.25 m 37.2 d H-10, H3-13, H3-14, H2-15
     9 41.9 s H 10, H3 13, H3 14, H2 15
    10 0.98 m 46.9 d H3-12, H3-14, H2-15
    11 1.48 s 17.9 q H-3
    12 0.94 s 20.0 q
    13 0.92 d 7.0 18.0 q
    14 0.78 s 17.1 q H-10
    15 2.47 d 13.7 31.7 t H3-14
    2.32 d 13.7
    16 114.1 s H2-15
    17 158.1 s H2-15
    18 179.1 s H-19
    19 5.33 s 93.0 d
    20 149.5 s
    20-NH 6.95 brd 7.0
    21 183.1 s H2-15, H-19
    22 4.07 m 60.2 d NH-20, H3-25
    23 171.0 s
    24 4.27 m 66.5 d H-22
    25 1.09 d 7.0 20.8 q
    aδ in ppm
    bHMBC correlations
    c2H
  • <Preparation Example 30> Preparation of 18-methoxy-22,22-dimethyl-16-[({(5R,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17(2H)-one
  • Smenospongia aurea and Smenospongia cerebriformis were homogenized and incubated with Verongula rigida in ethanol for a week. A dried ethanol extract (3.6 kg) of the mixture of the three sponge species was subjected to silica gel VLC (36 kg, 14 (H)×17.5 (D) cm) and eluted sequentially with hexane (100%), hexane-acetone (80:20, 60:40, 50:50, 40:60, 20:80), acetone (100%), acetone-MeOH (80:20, 60:40, 50:50), MeOH (100%), MeOH—H2O (50:50) and H2O (100%) to obtain 13 fractions (Fr. 1-13). The fraction 10 (39.3 g) was fractionated further with hexane-acetone mixtures (95:5, 90:10, 85:15, 80:20), MeOH (100%) and MeOH—H2O (50:50) using a silica gel VLC (12 (H)×17.5 (D) cm) into 9 fractions (Fr. 10-1 to 10-9). The fraction 10-7 (3.7 g) was subjected to C18 MPLC (15.5×4 cm) under an isocratic condition of MeOH—H2O (85:15) to prepare 6 subfractions (Fr. 10-7-1 to 10-7-6). The fraction 10-7-3 (115.8 mg) was subjected to C18 HPLC (250×21.20 mm, 10 μm) chromatography using MeOH—H2O (83:17) to prepare 3 fractions (Fr. 10-7-3-1 to 10-7-3-3). The fraction 10-7-3-2 (12.4 mg) was subjected to C18 HPLC (250×4.60 mm and 150×4.60 mm, 5 μm, connected in line) using MeOH—H2O (75:25) to obtain a compound of Preparation Example 30 and an epimer mixture thereof.
  • The obtained substance had the following physicochemical properties and was identified as ‘18-methoxy-22,22-dimethyl-16-[{(5R,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17(2H)-one’.
  • Yellow, amorphous solid.
  • [α]25 D+21 (c 0.1, MeOH).
  • UV (MeOH) λmax 297 nm.
  • 1H NMR (150 MHz, CDCl3): see [Table 19];
  • 13C NMR (150 MHz, CDCl3): see [Table 19].
  • HRFABMS m/z 398.2696 [M+H]+ (calcd for C25H36NO3, 398.2695), 420.2509 [M+Na]+ (calcd for C25H35NO3Na, 420.2515).
  • TABLE 19
    position δH, mult. (J in Hz) δ C
     1 ax 1.42, m 23
    eq 2.13, m
     2 ax 1.20, m 28.8
    eq 1.84, m
     3 ax 2.29, dt (14, 5.2) 33.2
    eq 2.04, m
     4 161
     5 40.5
     6 ax 1.30, m 36.9
    eq 1.47, m
     7 ax 1.36, m 28.1
    eq
     8 1.22, m 38.1
     9 42.9
    10 0.80, m 50
    11 a 4.40, br s 102.4
    b 4.38, br s
    12 1.01, s 20.7
    13 0.97, d (6.6) 18
    14 0.81, s 17.5
    15 2.37, d (14) 32.7
    2.53, d (14)
    16 110.3
    17 181.6
    18 158.9
    19 6.14, s 96.9
    20 155.6
    21 161.4
    22 116.2
    23 1.58, s 25.9
    24 1.60, s 26
    OH
    OCH3 3.83, s 56.4
  • <Preparation Example 31> Preparation of 18-methoxy-22-methyl-16-[{(5S,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17-ol
  • During the procedure of Preparation Example 30, the fraction 10-7-3-3 (9.7 mg) was subjected to C18 HPLC (250×4.60 mm and 150×4.60 mm, 5 μm, connected in line) using MeOH—H2O (78:22) and then to C18 HPLC (250×4.60 mm, 5 μm) for 120 minutes with varying concentrations of MeOH—H2O (80:20-100:0) to obtain a compound of Preparation Example 31 (1.8 mg).
  • The obtained compound (Preparation Example 31) had the following physicochemical properties and was identified as ‘18-methoxy-22-methyl-16-[{(5S,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17-ol’.
  • White, Amorphous Solid.
  • [α]25 D−29 (c 0.1, MeOH).
  • UV (MeOH) λmax 295 nm.
  • 1H NMR (150 MHz, CDCl3): see [Table 20].
  • 13C NMR (150 MHz, CDCl3): see [Table 20].
  • HRFABMS m/z 384.2540 [M+H]+ (calcd for C24H34NO3, 384.2539), 406.2357 [M+Na]+ (calcd for C24H33NO3Na, 406.2358).
  • TABLE 20
    δH, mult. (J in
    position Hz) δ C
     1 ax 1.51, m 23.4
    eq 2.27, m
     2 ax 1.22, m 28.9
    eq 1.87, m
     3 ax 2.33, t (7.5) 33.2
    eq 2.02, m
     4 160.5
     5 40.6
     6 ax 1.19, m 36.5
    eq 1.43, m
     7 ax 1.39, m 28.2
    eq
     8 1.40, m 37.4
     9 43.1
    10 0.92, m 49.4
    11 a 4.36, br s 102.7
    b 4.32, br s
    12 1.04, s 20.7
    13 1.04, d 18.5
    14 0.91, s 17.7
    15 2.80, d (14) 34.6
    2.89, d (14)
    16 109.2
    17 143.7
    18 144.6
    19 6.98, s 98.8
    20 132.3
    21 146.6
    22 162
    23 2.54, s 14.6
    24
    OH 5.87, s
    OCH3 3.90, s 56.6
  • <Preparation Example 32> Preparation of 3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione
  • After preparing subfractions Fr. 10-7-1 to 10-7-6 according to the same method as in Preparation Example 30, Fr. 10-7-4 (53 mg) was subjected to C18 HPLC (25×2.1 cm, 10 m) repeatedly under an isocratic condition of MeOH—H2O (87:13) to obtain a compound of Preparation Example 32 (tR=113 min).
  • The obtained compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione’.
  • C29H37NO3.
  • m.p.: 168-170° C.
  • SM m/e (%): 447 (7), 257 (64), 191 (11), 166 (59), 152 (25), 135 (16), 121 (23), 109 (23), 107 (20), 95 (100).
  • m/e 166.0495. calc. 166.0504 for C8H8NO3; m/e 191.1795. calc. 191.1799 for C14H23; m/e 257.104, calc. 257.105 for C15H15NO3.
  • IR (KBr) ν cm−1: 3265, 1600, 1395.
  • 1H NMR (CDCl3, 250 MHz) δ ppm: 6.47 (1H exch., s), 5.41 (1H, s), 4.45 (2H, br s). 3.43 (2H, q), 2.87 (2H, t), 2.52-2.51 (dd, AB syst., J=14 and 2 Hz), 1.05 (3H, s), 0.98 (3H, d, J=7.5 Hz), 0.84 (3H, s), 0.79 (1H, dd, J=11.2 and 2 Hz).
  • 13C NMR (ppm; CDCl3): see [Table 21].
  • TABLE 21
    Preparation Example 32
    Carbon (δ ppm)
    C-1 22.5
    C-2 25.0
    C-3 32.8
    C-4 153.6
    C-5 39.5
    C-6 37.9
    C-7 27.9
    C-8 39.3
    C-9 44.5
    C-10 48.3
    C-11 105.7
    C-12 33.2
    C-13 18.7
    C-14 18.4
    C-15 32.0
    C-16 114.0
    C-17 157.0
    C-18 178.4
    C-19 91.9
    C-20 150.1
    C-21 182.8
  • <Preparation Example 33> Preparation of 3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-2-hydroxy-5-methoxycyclohexa-2,5-diene-1,4-dione
  • Three sponge species Smenospongia aurea, Smenospongia cerebriformis and Verongula rigida were mixed and an ethanol extract was prepared by adding ethanol (98%). The ethanol extract was separated with a silica gel column using hexane, acetone, methanol, water, etc. to obtain a total of 13 fractions. Among them, fractions 4, 5, 6 and 10 were concentrated and separated by C18 RP column chromatography using a mobile phase (methanol:water=1:1 to 3:1) to obtain a pure compound (Preparation Example 33 compound). The compound was identified by HPLC (Agilent Technologies 1260 Infinity) using a UV spectrophotometer (203 nm) and a Bluespher AB2 (150×2 mm) column. The HPLC was conducted at a flow rate of 1 mL/min and 40° C. using water-methanol (78:22) as a mobile phase and the peaks of the compound were detected at 114 minutes.
  • The obtained compound had the following physicochemical properties and was identified as ‘3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-2-hydroxy-5-methoxycyclohexa-2,5-diene-1,4-dione’.
  • Yellow Solid.
  • C22H30O4.
  • Molecular weight: 358.47.
  • m.p.: 72.5° C.
  • IT-TOF/MS: m/z 381.1972 [M+Na]+.
  • 1H-NMR (CDCl3, 600 MHz): 2.08, 1.42 (each 1H, m, H2-1), 1.84, 1.16 (each 1H, m, H2-2), 2.29, 2.05 (each 1H, ddd, J=13.7, 8.6, 5.4, H2-3), 1.49, 1.32 (each 1H, m, H2-6), 1.37 (2H, m, H2-7), 1.14 (1H, m, H-8), 0.74 (1H, d, J=12.0, H-10), 4.43, 4.41 (each 1H, s, H2-11), 1.02 (3H, s, H3-12), 0.96 (3H, d, J=6.4, H3-13), 0.82 (3H, s, H3-14), 2.51, 2.45 (each 1H, d, J=13.7, H2-15), 5.83 (1H, s, H-19), 3.84 (3H, s, H3-22).
  • 13C NMR (CDCl3, 150 MHz): 23.34 (C-1), 28.11 (C-2), 33.13 (C-3), 160.69 (C-4), 40.63 (C-5), 36.82 (C-6), 28.80 (C-7), 38.25 (C-8), 43.50 (C-9), 50.30 (C-10), 102.66 (C-11), 20.73 (C-12), 18.01 (C-13), 17.52 (C-14), 32.52 (C-15), 117.49 (C-16), 153.49 (C-17), 182.51 (C-18), 102.17 (C-19), 161.90 (C-20), 182.20 (C-21), 57.01 (C-22).
  • <Preparation Example 34> Preparation of 3-[[(1S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-5-ethoxy-2-hydroxycyclohexa-2,5-diene-1,4-dione
  • 400 mg of the compound of Preparation Example 33 was dissolved in 20 mL of ethanol in a round flask. After adding 10.5 mL of a 1 M potassium hydroxide (KOH) solution, the reaction mixture was stirred at 70° C. for an hour. After adding a 1 M hydrochloric acid solution to the stirred reaction mixture and concentrating under reduced pressure, the mixture was transferred to a separation funnel and fractionated by dissolving in ethyl acetate and distilled water. The ethyl acetate layer was combined, dehydrated with magnesium sulfate, filtered and then concentrated under reduced pressure. In order to obtain a pure reaction product, the resulting concentrate was separated by silica gel column chromatography using a mobile phase (n-hexane:ethyl acetate=10:1) to obtain the final compound (Preparation Example 34).
  • The obtained final compound (Preparation Example 34) had the following physicochemical properties and was identified as ‘3-[[(1S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-5-ethoxy-2-hydroxycyclohexa-2,5-diene-1,4-dione’.
  • Yellow Semi-Solid.
  • C23H32O4.
  • Molecular weight: 372.5.
  • IT-TOF/MS: m/z 395.2146 [M+Na]+.
  • 1H NMR (CDCl3, 600 MHz): 7.47 (1H, s), 5.83 (1H, s), 4.46, 4.44 (each 1H, s), 4.06 (2H, q, J=7.2 Hz), 2.50 (2H, dd, J=12, 6.0 Hz), 2.33 (1H, dt, J=12, 6.0 Hz), 2.17-1.66 (4H, m), 1.49 (3H, t, J=7.2 Hz), 1.46-1.09 (7H, m), 1.04 (3H, m), 0.98 (3H, d, J=6 Hz), 0.84 (3H, s).
  • 13C NMR (CDCl3, 150 MHz): 182.68, 182.26, 161.19, 160.75, 153.35, 117.45, 102.63, 102.40, 66.10, 50.35, 43.45, 40.63, 38.29, 36.82, 33.14, 32.63, 28.79, 28.12, 23.32, 20.73, 18.46, 18.05, 13.97.
  • <Example 1> Evaluation of Wnt/3-Catenin Pathway Inhibitory Activity
  • It was investigated whether the prepared compounds inhibit Wnt/3-catenin for some structurally representative compounds.
  • <1-1> Wnt/β-catenin pathway inhibitory activity of compounds of Preparation Examples 30 and 31
  • HEK293 cells (human embryonic kidney cells) and Wnt3a-secreting L cells were obtained from the ATCC (American Type Culture Collection, USA) and were cultured in DMEM (Dulbecco's modified Eagle's medium) supplemented with 10% FBS (fetal bovine serum), 120 λg/mL penicillin and 200 λg/mL streptomycin.
  • Wnt3a-CM (Wnt3a-conditioned medium) was prepared by culturing Wnt3a-secreting L cells in DMEM supplemented with 10% [v/v] FBS (fetal bovine serum) for 4 days and recovering the DMEM medium and sterilizing by filtering through a 0.22-μm filter. Then, after adding a fresh DMEM (supplemented with 10% [v/v] FBS) medium to the cells and culturing for 3 days, the medium was recovered by the same method and then combined with the previously prepared Wnt3a-CM.
  • After treating HEK293 cells with the Wnt3a-CM, the compound of Preparation Example 30 or the compound of Preparation Example 31 (10, 20 or 40 μM) for 15 hours and extracting cytoplasmic proteins from the cells, the amount of β-catenin regulating the CRT (β-catenin response transcription) of the Wnt/β-catenin pathway in the cells was investigated by western blot using a β-catenin antibody (BD Transduction Laboratories, USA) and the ECL system (Santa Cruz Biotechnology). The result is shown in FIG. 1.
  • As seen from the western blot result of FIG. 1, the cells treated with the Wnt3a-CM showed increased 3-catenin expression in the cytoplasm but the cells treated with the Preparation Example 30 compound or the Preparation Example 31 compound of the present disclosure showed decreased level of β-catenin.
  • <1-2> Wnt/3-Catenin Pathway Inhibitory Activity of Preparation Example 32 Compound
  • The Wnt/1-Catenin Pathway Inhibitory Activity of the Preparation Example 32 Compound was evaluated in the same manner as in Example <1-1>. Briefly, after treating HEK293 cells with the Wnt3a-CM or the compound of Preparation Example 32 (10 or 20 μM) for 15 hours and extracting cytoplasmic proteins from the cells, the amount of β-catenin regulating the CRT (β-catenin response transcription) of the Wnt/β-catenin pathway in the cells was investigated by western blot using a β-catenin antibody (BD Transduction Laboratories, USA) and the ECL system (Santa Cruz Biotechnology). The result is shown in FIG. 2.
  • As seen from the western blot result of FIG. 2, the cells treated with the Wnt3a-CM showed increased 3-catenin expression in the cytoplasm but the cells treated with the Preparation Example 32 compound of the present disclosure showed decreased level of β-catenin.
  • <1-3> Wnt/3-catenin pathway inhibitory activity of Preparation Example 33 and 34 compounds
  • ARPE-19 cells (human retinal epithelial cells) and Wnt3a-secreting L cells were obtained from the ATCC (American Type Culture Collection, USA) and were cultured in DMEM (Dulbecco's modified Eagle's medium) supplemented with 10% FBS, 120 λg/mL penicillin and 200 λg/mL streptomycin. Wnt3a-CM (Wnt3a-conditioned medium) was prepared by culturing Wnt3a-secreting L cells in DMEM supplemented with 10% [v/v] FBS (fetal bovine serum) for 4 days and recovering the DMEM medium and sterilizing by filtering through a 0.22-μm filter. Then, after adding fresh DMEM (supplemented with 10% [v/v] FBS) to the cells and culturing for 3 days, Wnt3a-CM was recovered.
  • After treating ARPE-19 cells with the Wnt3a-CM, the compound of Preparation Example 33 or the compound of Preparation Example 34 (3 or 6 μM) for 24 hours and extracting cytoplasmic proteins from the cells, the amount of β-catenin regulating the CRT (β-catenin response transcription) of the Wnt/3-catenin pathway in the cells was investigated by western blot using a β-catenin antibody (BD Transduction Laboratories, USA) and the ECL system (Santa Cruz Biotechnology). The result is shown in FIG. 3.
  • As seen from FIG. 3, the cells treated with the Wnt3a-CM showed increased 3-catenin expression in the cytoplasm but the cells treated with the Preparation Example 33 compound or Preparation Example 34 compound of the present disclosure showed decreased level of β-catenin, suggesting that the compounds inhibit the Wnt/0-catenin pathway in the human retinal epithelial cells. Meanwhile, this activity was not identified in the sponge ethanol extract used to isolate the compound of Preparation Example 33 (data not shown).
  • <Example 2> Evaluation of Inhibitory Activity of Vascular Leakage in the Eye
  • <2-1> Intravitreal Administration
  • For the compound of Preparation Example 33 which showed good activity in Example 1, the inhibitory activity on vascular leakage, which is a cause of macular degeneration or macular edema, was investigated in a macular edema-induced mouse model. Macular edema was induced in a 10-week-old C57BL/6 mouse by irradiating a laser. After anesthetizing the mouse with ketamine (70 mg/kg) and xylazine (30 mg/kg), the pupil was dilated with 1% tropicamide. Hydroxypropylmethyl cellulose was dropped onto the eye and a microscope cover glass was used as a contact lens. Then, five laser burns were made in the space between the blood vessels around the optic disc (Zeiss 1149-630, laser power 180 mW, duration 0.1 s, spot size 50 μm). In this state, the blood vessel size and permeability were investigated by fluorescein angiography (FA) and optical coherence tomography (OCT). 24 hours after the laser irradiation, 0.5 μL of DMSO (dimethyl sulfoxide) was injected to a control group and 100 ng/0.5 μL of the Preparation Example 33 compound dissolved in DMSO was injected to a test group, into the vitreous cavity of the mouse. One week later, fluorescein angiography and optical coherence tomography were conducted again. FIG. 4 shows the images of the control group and the compound-treated group.
  • As seen from the fluorescein angiography and optical coherence tomography results of FIG. 4, the test group (FIG. 4, C, D) showed distinctly decreased vascular leakage, which is a cause of macular degeneration or macular edema, as compared to the control group (FIG. 4, A, B).
  • <2-2> Intraperitoneal Administration After inducing macular edema in a C57BL/6 mouse and intraperitoneally administering the compound of Preparation Example 33, vascular leakage in the retina was investigated.
  • Briefly, after anesthetizing an 8-to-12-week-old C57BL/6 mouse with Zoletil (40 mg/kg) and xylazine (5 mg/kg), the pupil was dilated with 1% tropicamide. Hydroxypropylmethyl cellulose was dropped onto the eye and a microscope cover glass was used as a contact lens. Then, 3-5 laser burns were made in the space between the retinal blood vessels around the optic disc (Zeiss 1149-630, laser power 200 mW, duration 0.05 s, spot size 50 m).
  • After the laser irradiation, 10 mL/kg of distilled water as a vehicle was intraperitoneally administered to a control group (FIG. 5, A) and 1 mg/kg of the compound of Preparation Example 33 (dissolved distilled water) was intraperitoneally administered to a test group (FIG. 5, B) every day for 7 days. On day 6, 10% sodium fluorescein was intraperitoneally administered and optical coherence tomography was conducted.
  • As seen from FIG. 5, it was confirmed that the compound of the present disclosure inhibited vascular leakage, which is a cause of macular degeneration or macular edema, even when it was injected intraperitoneally.
  • <Example 3> In Vivo PK (Pharmacokinetics) after Oral Administration
  • Male ICR mice (8 weeks, 30-35 g) were purchased from Samtako Co. (Osan, Korea). The test animals were acclimatized for a week under the following conditions: temperature 23±2° C., relative humidity 55±10%, illumination intensity 150-300 lux, ventilation frequency 15-20 times/h, illumination cycle 12 h (07:00-19:00). All the animal experiments were approved by the Animal Care and Use Committee of Kyungpook National University (Study No. 2016-0043).
  • The mice were fasted for 12 hours before drug administration. Feed and water were supplied ad libitum. The compound of Preparation Example 33 was dissolved in DW:PEG 400 (=60:40 (v/v)) and administered by oral gavage at a dose of 10 mg/kg.
  • 0.5 hour and 2 hours after the oral administration, blood samples were taken from the abdominal artery. 50 mL of a plasma sample obtained after centrifuging the blood sample at 13,000 rpm for 5 minutes was stored at −80° C. until use for analysis. An eye sample taken from the mouse was homogenized with 9-fold saline to obtain a 10% cell homogenate. The obtained 50-mL aliquots were stored at −80° C. until use for analysis.
  • 50 μL of the aliquot was added to 200 μL of an acetonitrile solution containing 0.5 ng/mL propranolol. After vortex mixing for 10 minutes, followed by centrifugation at 13,000 rpm for 10 minutes, the supernatant was transferred to a fresh tube and evaporated under nitrogen gas flow. The residue was added to 150 μL of a mobile phase and a 5-μL aliquot was injected directly into the LC-MS/MS system for analysis.
  • In order to investigate the distribution of the compound of the present disclosure in the target tissue, the compound was orally administered at a dose of 10 mg/kg and the concentration of the compound in the blood plasma and eye was measured. The sampling times were determined as 0.5 hour and 2 hours based on the peak plasma concentration and the distribution phase. As seen from FIG. 6, the compound of the present disclosure (particularly, the compound of Preparation Example 33) showed high permeability for the target tissue and was found to be highly targeted in the eye even when it was administered orally. Through this result, it was confirmed that the compound of Preparation Example 33 of the present disclosure can exhibit therapeutic effect even when it is administered via different administration routes (oral administration, intraperitoneal injection, intravenous injection, etc.) other than being administered directly into the vitreous cavity. That is to say, whereas the currently available eye disease-related therapeutic agents cause inconvenience, pain and side effects because they have to be administered directly into the vitreous cavity, it was confirmed that the compound of the present disclosure can be administered orally.
  • <Example 4> Evaluation of Safety of Compound
  • <4-1> Evaluation of Acute Toxicity
  • This experiment was conducted to determine acute (within 24 hours) toxicity and lethality when the compound of Preparation Example 33 was administered in excessive amounts in a short period of time. 20 normal ICR mice were divided into a control group and a test group, with 10 mice per each. The control group was administered with PEG 400:Tween 80:ethanol (8:1:1, v:v:v) only and the test group was orally administered with the compound of Preparation Example 33 dissolved in PEG 400:Tween 80:ethanol (8:1:1, v:v:v). When lethality was investigated 24 hours after the administration, all the mice in the control group and the mice in the test group administered with the Preparation Example 33 compound at a dose of 2 g/kg/day survived.
  • <4-2> Evaluation of Tissue Toxicity
  • A long-term toxicity test was conducted by administering the compound of Preparation Example 33 at different doses to C57BL/6J mice (10 mice per group) for 8 weeks. In order to investigate the effect on different organs (tissues) of the animals, blood was taken from the animals of the test group to which the compound of Preparation Example 33 was administered and the control group to which only PEG 400:Tween 80:ethanol (8:1:1, v:v:v) was administered 8 weeks later and the level of GPT (glutamate-pyruvate transferase) and BUN (blood urea nitrogen) in the blood was measured using Select E (Vital Scientific NV, Netherlands). As a result, there was no significant difference between the control group and the test group in GPT, which is known to be related with liver toxicity, and BUN, which is known to be related with kidney toxicity. In addition, no special abnormality was observed in the liver and kidney tissues taken from the animals when they were prepared into tissue sections and observed under an optical microscope according to the common method.
  • <Formulation Examples> Preparation of Pharmaceutical Formulations
  • <Formulation Example 1> Preparation of Tablet
  • 200 g of the compound of the present disclosure was mixed with 175.9 g of lactose, 180 g of potato starch and 32 g of colloidal silicate. After adding a 10% gelatin solution, the mixture was pulverized and passed through a 14-mesh sieve. A mixture obtained by drying the same and adding 160 g of potato starch, 50 g of talc and 5 g of magnesium stearate thereto was prepared into a tablet.
  • <Formulation Example 2> Preparation of Injection 1 g of the compound of the present disclosure, 0.6 g of sodium chloride and 0.1 g of ascorbic acid were dissolved in distilled water to make 100 mL. The resulting solution was put in a bottle and sterilized by heating at 20° C. for 30 minutes.
  • As described above, the present disclosure relates to a novel use of a sesquiterpene derivative, more particularly to a composition for preventing, improving or treating macular degeneration or macular edema caused by vascular leakage in the eye, which contains the sesquiterpene derivative compound represented by Chemical Formula 1 of the present disclosure or a pharmaceutically acceptable salt thereof as an active ingredient.
  • The compound of the present disclosure of Chemical Formula 1 has therapeutic effect for a disease caused by vascular leakage in the eye, such as macular edema, macular degeneration, etc., by inhibiting the vascular leakage in the eye, particularly in the retina. In addition, whereas the intraocular disease-related treating agents available in the market should be injected directly into the vitreous cavity, thus causing pain and side effects, the sesquiterpene derivative compound of the present disclosure is delivered to the target tissue (eye) via different administration routes (oral, intraperitoneal, etc.) other than the intravitreal route. Accordingly, the sesquiterpene derivative compound provides excellent therapeutic effect without being restricted by the administration routes. Accordingly, it is highly industrially applicable.
  • INDUSTRIAL APPLICABILITY
  • The compound of the present disclosure of Chemical Formula 1 has therapeutic effect for a disease caused by vascular leakage in the eye, such as macular edema, macular degeneration, etc., by inhibiting the vascular leakage in the eye, particularly in the retina. Whereas the intraocular disease-related treating agents available in the market should be injected directly into the vitreous cavity, thus causing pain and side effects, the sesquiterpene derivative compound of the present disclosure is delivered to the target tissue (eye) via different administration routes (oral, intraperitoneal, etc.) other than the intravitreal route. Accordingly, the sesquiterpene derivative compound provides excellent therapeutic effect without being restricted by the administration routes.
  • Those skilled in the art will appreciate that the conceptions and specific embodiments disclosed in the foregoing description may be readily utilized as a basis for modifying or designing other embodiments for carrying out the same purposes of the present disclosure. Those skilled in the art will also appreciate that such equivalent embodiments do not depart from the spirit and scope of the disclosure as set forth in the appended claims.

Claims (13)

1. A pharmaceutical composition for preventing or treating macular degeneration or macular edema, comprising a compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient:
Figure US20190290617A1-20190926-C00043
wherein
the broken line denotes a single bond or a double bond, wherein
i) if the bond between C-3 and C-4 and the bond between C-5 and C-6 are single bonds, R2b is nonexistent and R2a is CH2;
ii) if the bond between C-3 and C-4 is a double bond, the bond between C-5 and C-6 is a single bond, R2b is nonexistent and R2a is CH3; or
iii) if the bond between C-5 and C-6 is a double bond, the bond between C-3 and C-4 is a single bond and R2a and R2b are CH3,
R1 is H or CH3,
R3 is a functional group selected from a group consisting of R3a through R3d,
Figure US20190290617A1-20190926-C00044
in R3a,
i) each of R4 and R7 is OH or OCH3 and R5, R6 and R8 are H; or
ii) R5 is COOCH3, R is H or OH, R8 is OH and R4 and R are H,
in R3b,
R9 is a functional group selected from a group consisting of H, NH2, C1-C8 alkoxy and R9a through R9j and R10 is H or OH,
Figure US20190290617A1-20190926-C00045
in R3c,
each of R11 and R12 is OH or OAc and R3 is H; or
each of R11 and R12 is OH or OCH3 and R13 is CH3 and
in R3d,
R14 is OCH3 and R15 and R16 are CH3.
2. The pharmaceutical composition according to claim 1, wherein, if the bond between C-3 and C-4 and the bond between C-5 and C-6 are single bonds, R2b is nonexistent, R2a is CH2 and R3 is a functional group selected from a group consisting of R3b through R3d.
3. The pharmaceutical composition according to claim 2, wherein, in the functional group selected from a group consisting of R3b through R3d,
in R3b, R9 is selected from a group consisting of ethoxy, methoxy and R9a;
in R3c, R11 is OH, R12 is OCH3 and R13 is CH3; or
in R3d, R14 is OCH3 and R15 and R16 are CH3.
4. The pharmaceutical composition according to claim 3, wherein the compound of Chemical Formula 1 is a compound selected from a group consisting of:
3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione;
3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-2-hydroxy-5-methoxycyclohexa-2,5-diene-1,4-dione;
3-[[(1 S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-5-ethoxy-2-hydroxycyclohexa-2,5-diene-1,4-dione;
18-methoxy-22-methyl-16-[{(5 S,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17-ol; and
18-methoxy-22,22-dimethyl-16-[{(5R,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17(2H)-one.
5. The pharmaceutical composition according to claim 1, wherein the composition is prepared into a formulation selected from a group consisting of an oral medication, an injection, an eye drop and an ointment.
6. A pharmaceutical composition for inhibiting vascular leakage in the eye, comprising a compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient:
Figure US20190290617A1-20190926-C00046
wherein
the broken line denotes a single bond or a double bond,
wherein
i) if the bond between C-3 and C-4 and the bond between C-5 and C-6 are single bonds, R2b is nonexistent and R2a is CH2;
ii) if the bond between C-3 and C-4 is a double bond, the bond between C-5 and C-6 is a single bond, R2b is nonexistent and R2a is CH3; or
iii) if the bond between C-5 and C-6 is a double bond, the bond between C-3 and C-4 is a single bond and R2a and R2b are CH3,
R1 is H or CH3,
R3 is a functional group selected from a group consisting of R3a through R3d,
Figure US20190290617A1-20190926-C00047
in R3a,
i) each of R4 and R7 is OH or OCH3 and R5, R6 and R8 are H; or
ii) R5 is COOCH3, R7 is H or OH, R8 is OH and R4 and R6 are H,
in R3b,
R9 is a functional group selected from a group consisting of H, NH2, C1-C8 alkoxy and R9a through R9j and R10 is H or OH,
Figure US20190290617A1-20190926-C00048
in R3c,
each of R11 and R12 is OH or OAc and R13 is H; or
each of R11 and R12 is OH or OCH3 and R13 is CH3 and
in R3d,
R14 is OCH3 and R15 and R16 are CH3.
7. The pharmaceutical composition according to claim 6, wherein, if the bond between C-3 and C-4 and the bond between C-5 and C-6 are single bonds, R2b is nonexistent, R2a is CH2 and R3 is a functional group selected from a group consisting of R3b through R3d.
8. The pharmaceutical composition according to claim 7, wherein, in the functional group selected from a group consisting of R3b through R3d,
in R3b, R9 is selected from a group consisting of ethoxy, methoxy and R9a;
in R3c, R11 is OH, R12 is OCH3 and R13 is CH3; or
in R3d, R14 is OCH3 and R15 and R16 are CH3.
9. The pharmaceutical composition according to claim 8, wherein the compound of Chemical Formula 1 is a compound selected from a group consisting of:
3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione;
3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-2-hydroxy-5-methoxycyclohexa-2,5-diene-1,4-dione;
3-[[(1 S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-5-ethoxy-2-hydroxycyclohexa-2,5-diene-1,4-dione;
18-methoxy-22-methyl-16-[{(5 S,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17-ol; and
18-methoxy-22,22-dimethyl-16-[{(5R,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17(2H)-one.
10. A food composition for preventing or improving macular degeneration or macular edema, comprising a compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient:
Figure US20190290617A1-20190926-C00049
wherein
the broken line denotes a single bond or a double bond, wherein
i) if the bond between C-3 and C-4 and the bond between C-5 and C-6 are single bonds, R2b is nonexistent and R2a is CH2;
ii) if the bond between C-3 and C-4 is a double bond, the bond between C-5 and C-6 is a single bond, R2b is nonexistent and R2a is CH3; and
iii) if the bond between C-5 and C-6 is a double bond, the bond between C-3 and C-4 is a single bond and R2a and R2b are CH3,
R1 is H or CH3,
R3 is a functional group selected from a group consisting of R3a through R3d,
Figure US20190290617A1-20190926-C00050
in R3a,
i) each of R4 and R7 is OH or OCH3 and R5, R6 and R8 are H; or
ii) R5 is COOCH3, R7 is H or OH, R8 is OH and R4 and R6 are H,
in R3b,
R9 is a functional group selected from a group consisting of H, NH2, C1-C8 alkoxy and R9a through R9j and R10 is H or OH,
Figure US20190290617A1-20190926-C00051
in R3c,
each of R11 and R12 is OH or OAc and R13 is H; or
each of R11 and R12 is OH or OCH3 and R13 is CH3 and
in R3d,
R14 is OCH3 and R15 and R16 are CH3.
11. The food composition according to claim 10, wherein, if the bond between C-3 and C-4 and the bond between C-5 and C-6 are single bonds, R2b is nonexistent, R2a is CH2 and R3 is a functional group selected from a group consisting of R3b through R3d.
12. The food composition according to claim 11, wherein, in the functional group selected from a group consisting of R3b through R3d,
in R3b, R9 is selected from a group consisting of ethoxy, methoxy and R9a;
in R3c, R11 is OH, R12 is OCH3 and R13 is CH3; or
in R3d, R14 is OCH3 and R15 and R16 are CH3.
13. The food composition according to claim 12, wherein the compound of Chemical Formula 1 is a compound selected from a group consisting of:
3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-4-hydroxy-5-(2-phenylethylamino)cyclohexa-3,5-diene-1,2-dione;
3-[[(1R,2S,4aS,8aS)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-2-hydroxy-5-methoxycyclohexa-2,5-diene-1,4-dione;
3-[[(1 S,2R,4aR,8aR)-1,2,4a-trimethyl-5-methylidene-3,4,6,7,8,8a-hexahydro-2H-naphthalen-1-yl]methyl]-5-ethoxy-2-hydroxycyclohexa-2,5-diene-1,4-dione;
18-methoxy-22-methyl-16-[{(5 S,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17-ol; and
18-methoxy-22,22-dimethyl-16-[{(5R,8S,9R,10S)-5,8,9-trimethyl-4-methylenedecahydronaphthalen-9-yl}methyl]benzo[d]-oxazol-17(2H)-one.
US16/301,583 2016-07-07 2016-10-14 Novel use of sesquiterpene derivative Abandoned US20190290617A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
KR10-2016-0086331 2016-07-07
KR1020160086331A KR101674622B1 (en) 2016-07-07 2016-07-07 Novel Use of Sesquiterpene Derivatives
PCT/KR2016/011558 WO2018008803A1 (en) 2016-07-07 2016-10-14 Novel use of sesquiterpene derivative

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2016/011558 A-371-Of-International WO2018008803A1 (en) 2016-07-07 2016-10-14 Novel use of sesquiterpene derivative

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/686,940 Continuation US10918625B2 (en) 2016-07-07 2019-11-18 Use of sesquiterpene derivative

Publications (1)

Publication Number Publication Date
US20190290617A1 true US20190290617A1 (en) 2019-09-26

Family

ID=57528861

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/301,583 Abandoned US20190290617A1 (en) 2016-07-07 2016-10-14 Novel use of sesquiterpene derivative
US16/686,940 Active US10918625B2 (en) 2016-07-07 2019-11-18 Use of sesquiterpene derivative

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/686,940 Active US10918625B2 (en) 2016-07-07 2019-11-18 Use of sesquiterpene derivative

Country Status (8)

Country Link
US (2) US20190290617A1 (en)
EP (1) EP3482750B1 (en)
JP (1) JP6667681B2 (en)
KR (1) KR101674622B1 (en)
CN (1) CN109414416B (en)
AU (1) AU2016413709B2 (en)
ES (1) ES2929708T3 (en)
WO (1) WO2018008803A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101674622B1 (en) * 2016-07-07 2016-11-09 국민대학교산학협력단 Novel Use of Sesquiterpene Derivatives
KR101800346B1 (en) 2016-08-30 2017-11-22 국민대학교산학협력단 Composition for Inhibiting Vascular Leakage in Retina
TWI723344B (en) * 2019-02-20 2021-04-01 安益藥業股份有限公司 Uses of picrotoxane-type sesquiterpenes for preparing pharmaceutical composition with eye care effect

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2100124B1 (en) 1994-08-01 1998-04-01 Pharma Mar Sa IMPROVEMENTS INTRODUCED IN IP 9401697 FOR TERPENO-QUINONAS WITH ANTITUMORAL ACTIVITY.
WO2000015766A1 (en) * 1998-09-16 2000-03-23 Oncopharmaceutical, Inc. Treatment of oncologic tumors with an injectable formulation of a golgi apparatus disturbing agent
US20040072831A1 (en) * 2001-10-12 2004-04-15 Choongwae Pharma Corporation Reverse-turn mimetics and method relating thereto
US20090074668A1 (en) * 2007-09-14 2009-03-19 Farjo Rafal A Vldlr-/- mouse models and related methods
KR100905419B1 (en) * 2008-09-11 2009-07-02 연세대학교 산학협력단 Uses of sesquiterpene derivatives
KR101059321B1 (en) * 2008-11-07 2011-08-24 영남대학교 산학협력단 Composition for the prevention and treatment of diseases caused by overexpression of MPM-X or MMP-2 containing sponge extract fraction as an active ingredient
WO2012033353A2 (en) * 2010-09-07 2012-03-15 서울대학교 산학협력단 Sesterterpene compounds and use thereof
EP2554662A1 (en) * 2011-08-05 2013-02-06 M Maria Pia Cosma Methods of treatment of retinal degeneration diseases
US10010610B2 (en) 2014-01-16 2018-07-03 Retinal Therapies, LLC Compositions and methods for the treatment of intraocular neovascularization and/or leakage
KR101674622B1 (en) * 2016-07-07 2016-11-09 국민대학교산학협력단 Novel Use of Sesquiterpene Derivatives
KR101800346B1 (en) * 2016-08-30 2017-11-22 국민대학교산학협력단 Composition for Inhibiting Vascular Leakage in Retina
KR101692378B1 (en) * 2016-09-20 2017-01-03 국민대학교산학협력단 Novel Use of Sesquiterpene Derivatives

Also Published As

Publication number Publication date
EP3482750A4 (en) 2020-04-15
EP3482750B1 (en) 2022-09-28
KR101674622B1 (en) 2016-11-09
JP6667681B2 (en) 2020-03-18
CN109414416B (en) 2021-03-26
US10918625B2 (en) 2021-02-16
CN109414416A (en) 2019-03-01
JP2019524686A (en) 2019-09-05
US20200078338A1 (en) 2020-03-12
WO2018008803A1 (en) 2018-01-11
ES2929708T3 (en) 2022-12-01
AU2016413709A1 (en) 2019-01-17
AU2016413709B2 (en) 2019-07-11
EP3482750A1 (en) 2019-05-15

Similar Documents

Publication Publication Date Title
US10918625B2 (en) Use of sesquiterpene derivative
KR102338093B1 (en) Composition for preventing, improving or treating eye diseases comprising (7S)-(+)-cyclopentyl carbamic acid, 8,8-dimethyl-2-oxo-6,7-dihydro-2H,8H-pyrano[3,2-g]chromen-7-yl-ester as an active ingredient
US20160067209A1 (en) Pterocarpan compound or pharmaceutically acceptable salt thereof and pharmaceutical composition for prevention or treatment of metabolic disease or complication thereof, or for antioxidant containing the same as an active ingredient
WO1999064424A1 (en) Therapeutic agents
KR101800346B1 (en) Composition for Inhibiting Vascular Leakage in Retina
KR100704299B1 (en) Novel quinoline compound, and composition containing centipede extract or compounds isolated therefrom for prevention and treatment of cardiovascular disease
KR101692378B1 (en) Novel Use of Sesquiterpene Derivatives
WO2021025103A1 (en) Composition for inhibiting reduction of testosterone and/or dihydrotestosterone
US20130041023A1 (en) Composition for Preventing or Treating Rotavirus Infection Comprising Licorice Extract
TW518221B (en) Antiallergic agent and antiallergic foods or drinks comprising 4,5-dihydroxy-2-cyclopenten-1-one or its optical activators or their salts
KR20160137808A (en) Anti-cancer adjuvant comprising panaxadiols compound
JPWO2006030907A1 (en) Retinal protective agent
KR101051085B1 (en) Parkinson&#39;s disease prevention and treatment composition containing cinnamon extract, fractions thereof or trans-cinnaaldehyde isolated from cinnamon as an active ingredient
US8785503B2 (en) Biphenyl compound or pharmaceutically acceptable salt thereof, method for preparing novel biphenyl compound or pharmaceutically acceptable salt thereof, and pharmaceutical composition containing same as active ingredient for preventing or treating diabetes complications
KR101642291B1 (en) Composition for Preventing or Treating Breast Cancer Diseases Comprising Ramalin
US8686031B2 (en) Flavonoids and uses thereof
US11168092B2 (en) Compounds and method of preventing or treating cancer using the same
CA2862807A1 (en) Opsin-binding ligands, compositions and methods of use
US20240216462A1 (en) Pharmaceutical composition for enhancing immunity or anti-cancer activity including extract of pseudolysimachion nakaianum (ohwi) t.yamaz as active ingredient
KR101448116B1 (en) Composition comprising DDMP group soyasaponin for preventing or treating obesity or lipid related metabolic disease
KR20140018706A (en) Composition comprising extract of fermented curcuma longa by natural fermented soybean and curcuminoid derivatives isolated therefrom for treating or preventing liver disease
KR20230073533A (en) Composition containing red ginseng saponin extract for preventing or treating age-related macular degeneration
KR100806226B1 (en) A composition for the prevention and treatment of cardiovascular disease containing extract of Glycine max Roots or polypenol compounds isolated thereof
US20210299103A1 (en) Composition for preventing or treating cancer, comprising sulfonamide derivative as active ingredient
KR101677083B1 (en) Anti-inflammatory active composition and pharmaceutical composition comprising the same

Legal Events

Date Code Title Description
AS Assignment

Owner name: KOOKMIN UNIVERSITY INDUSTRY ACADEMY COOPERATION FO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:OH, SANGTAEK;REEL/FRAME:047499/0927

Effective date: 20181114

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION