KR101286153B1 - Composition for Anti-cancer Effect Comprising Gallic Acid Derivatives - Google Patents

Composition for Anti-cancer Effect Comprising Gallic Acid Derivatives Download PDF

Info

Publication number
KR101286153B1
KR101286153B1 KR20100114412A KR20100114412A KR101286153B1 KR 101286153 B1 KR101286153 B1 KR 101286153B1 KR 20100114412 A KR20100114412 A KR 20100114412A KR 20100114412 A KR20100114412 A KR 20100114412A KR 101286153 B1 KR101286153 B1 KR 101286153B1
Authority
KR
South Korea
Prior art keywords
cancer
gallic acid
composition
formula
delete delete
Prior art date
Application number
KR20100114412A
Other languages
Korean (ko)
Other versions
KR20120053245A (en
Inventor
윤호근
나영화
김미정
이유현
Original Assignee
연세대학교 산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 연세대학교 산학협력단 filed Critical 연세대학교 산학협력단
Priority to KR20100114412A priority Critical patent/KR101286153B1/en
Publication of KR20120053245A publication Critical patent/KR20120053245A/en
Application granted granted Critical
Publication of KR101286153B1 publication Critical patent/KR101286153B1/en

Links

Images

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Emergency Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Mycology (AREA)
  • Nutrition Science (AREA)
  • Engineering & Computer Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)

Abstract

The present invention relates to a novel gallic acid derivative prepared by using gallic acid as a basic structure, a preparation method thereof, and an anticancer composition using the same.
The gallic acid derivative of the present invention exhibits effective anticancer activity by inhibiting the activity of HAT (histone acetyltransferase), and is safe because it has a basic structure of natural acid gallic acid. The present invention also provides basic data of medicines and foods of gallic acid derivatives having anticancer activity.

Description

Composition for Anti-cancer Effect Comprising Gallic Acid Derivatives

The present invention relates to an anticancer composition containing a gallic acid derivative as an active ingredient.

More specifically, the present invention inhibits histone acetyltransferase, and particularly a pharmaceutical composition for preventing or treating cancer or a cancer composition containing a gallic acid derivative having an excellent anticancer effect in androgen receptor mediated prostate cancer as an active ingredient or It relates to a food composition for improvement.

Prostate cancer, which is rapidly growing in Korea, is the number one cause of death for adult males in the United States, and it is known that diet and lifestyle have a significant effect on cancer. Androgen receptors belonging to the steroid hormone receptor superfamily are regulated by androgen, a male hormone, and these receptors play an essential role in the development and progression of prostate cancer. [Park, SK et al. Prostate . 66 (12): 1285-91 (2006); Taplin ME et al. J Cell Biochem. 15; 91 (3): 483-90 (2004); Chen, L. et al. Mol Cancer Ther . 4 (9): 1311-9 (2005); Yoon, HG et al. Mol Endocrinol . 20 (5): 1048-60 (2006).

Histones are tightly bound to DNA with genetic information to form nucleosomes in the nucleus, and post-translational modifications of these DNA-binding proteins can regulate gene expression and signaling. The acetylation of histones during this transformation is largely regulated by two enzymes, histone acetyltransferase (HAT) and histone deacetylase (HDAC), which are transcription factors and signal transduction parameters. It can also regulate the activity of the same nonhistone protein. HAT proteins increase the acetylation of hormone receptors, and acetylated hormone receptors overexpress each target protein, causing cancer growth and development. As a result, inhibition of hyperacetylation in hormone receptor-related cancers is recognized as a molecular target for the search for new cancer inhibitors. In particular, hyperacetylation of androgen receptors and histones is associated with the development and proliferation of androgen receptor-independent prostate cancer. Related to cancer cell lines in In vitro experiments have reported apoptosis suppression mechanisms [Kang, J. et al. Biochem Pharmacol . 69 (8): 1205-13 (2005); Stimson, L. et al. Mol Cancer Ther . 4 (10): 1521-32 (2005); Balasubramanya, K. et al. J Biol Chem . 279 (49): 51163-71 (2004); Druesne, N. et al. Carcinogenesis . 25 (7): 1227-36 (2004); Fu, M. et al. Biochem Pharmacol . 68 (6): 1199-208 (2004).

In order to control such acetylation, HDAC activity inhibitors have been released so far mainly using HDAC protein as a target protein for cancer therapy. Relatively limited HAT inhibitors compared to these various HDAC inhibitors have been published. Inhibitors studied to date include synthetic peptide-CoA conjugates, isothiazolone, polyprenylated benzophenone, curcumin, and anacardic acid [Kang, J et al. Biochem Pharmacol . 69 (8): 1205-13 (2005); Inche, AG Drug Discov Today . 11 (3-4): 97-109 (2006); Lau, OD et al. Mol Cell . 5 (3): 589-95 (2000); Debes, JD et al. Cancer Res. 63 (22): 7638-40 (2003); Sun, Y. et al, FEBS Lett. 580 (18): 4353-6 (2006). Recently, researches on plant-derived inhibitors that are easier to secure safety than synthetic compounds such as peptide-CoA conjugate or isothiazolone have been actively conducted. Curcuminn in turmeric mentioned above, anacardic acid from cashew nuts, and Garcinia benzophenone in indica ) is an example of a plant-derived HAT inhibitor. Securing a new trace material produced on the basis of such a plant extract containing the HAT inhibitory activity and a single substance derived therefrom or a single substance thereof is continuously required for the development of a new anticancer agent.

Gallic acid (3,4,5-trihydroxybenzoic acid) is evenly distributed in the roots, stems, leaves, and fruits of nuts, teas, and various other plants, and is used in various pharmaceutical industries and natural products It is being used. Gallic acid (gallic acid) is known by various papers and patents to be excellent in inhibiting microbial and viral growth, antioxidant, anticancer and antidiabetic effect.

Numerous papers and patent documents are referenced and cited throughout this specification. The disclosures of the cited papers and patent documents are incorporated herein by reference in their entirety to better understand the state of the art to which the present invention pertains and the content of the present invention.

The present inventors specifically inhibited HAT activity against compounds synthesized based on trace elements obtained from natural products in order to ensure safety and high medicinal activity in the development of functional foods, food drugs and medicines for inhibiting hormone receptor-related cancer. And while searching for the anti-cancer activity, the gallic acid derivatives synthesized with a basic structure of the conventionally known gallic acid as an antioxidant and anti-inflammatory substance has a high HAT inhibitory activity and high anti-cancer activity to confirm the present invention It was completed.

Accordingly, an object of the present invention is to provide a novel gallic acid derivative prepared based on gallic acid.

Another object of the present invention is to provide a method for producing the novel gallic acid derivative based on the gallic acid.

Another object of the present invention to provide a pharmaceutical composition for preventing and treating cancer containing the gallic acid derivative as an active ingredient.

Another object of the present invention to provide a food composition for preventing and improving cancer containing the gallic acid derivative as an active ingredient.

Other objects and advantages of the present invention will become more apparent from the following detailed description of the invention, claims and drawings.

According to one aspect of the present invention, the present invention provides a gallic acid derivative represented by any one of the following Chemical Formulas 1 to 3:

Formula 1

Figure 112010075100198-pat00001

(2)

Figure 112010075100198-pat00002

(3)

Figure 112010075100198-pat00003

In Chemical Formulas 1 to 3, the ROne To R3Are independently of each other hydrogen, straight chain or substituted substituted or unsubstituted COne -5Alkyl, substituted or unsubstituted C6 -10of Aryl, or substituted or unsubstituted benzyl; The R4 To R6Are independently of each other hydrogen, straight or crushed, substituted or unsubstituted C1-10Alkyl, substituted or unsubstituted C6-10of Aryl, or substituted or unsubstituted benzyl.

The present inventors specifically inhibited HAT activity against compounds synthesized based on trace elements obtained from natural products in order to ensure safety and high medicinal activity in the development of functional foods, food drugs and medicines for inhibiting hormone receptor-related cancer. And while searching for the anti-cancer activity, the gallic acid derivatives synthesized with a basic structure of the conventionally known gallic acid as an antioxidant and anti-inflammatory substance has a high HAT inhibitory activity and high anti-cancer activity to confirm the present invention It was completed.

As used herein, the term “alkyl” refers to a straight or tricyclic unsubstituted or substituted saturated hydrocarbon group, and includes, for example, methyl, ethyl, propyl, isobutyl, pentyl and the like. C 1 -C 5 alkyl means an alkyl group having an alkyl unit of 1 to 5 carbon atoms, and when C 1 -C 5 alkyl is substituted, the number of carbon atoms of the substituent is not included. C 1 - C 10 in the alkyl means a alkyl group having an alkylene unit having a carbon number of 1 to 10 carbon atoms in the case where the C 1 -C 5 alkyl substituted with a substituent is not included.

In Formula 1, R 1 , R 2 or R 3 C 1 -C 5 of position Alkyl is preferably an alkyl of C 1 -C 3, R 4 position of the alkyl is preferably a substituted alkyl, more preferably of a substituted C 1 -10 alkyl group, more preferably a dialkyl Amino groups or morpholino groups (

Figure 112010075100198-pat00004
) Is substituted alkyl.

The term “aryl” refers to a substituted or unsubstituted monocyclic or polycyclic carbon ring which is wholly or partially unsaturated. C 6 - C 10 Aryl means an aryl group having a carbon ring atom and having 6 to 10 carbon atoms, C 6 - it does not contain the carbon atoms of the substituent when the aryl is a substituted C 10. Preferably aryl is monoaryl or biaryl. It is preferable that monoaryl has 5-6 carbon atoms, and it is preferable that biaryl has 9-10 carbon atoms. Most preferably said aryl is substituted or unsubstituted phenyl. When monoaryl, such as phenyl, is substituted, substitutions may be made by various substituents at various positions, such as halo, hydroxy, nitro, cyano, C 1 -C 4 Substituted or unsubstituted straight or branched chain alkyl or C 1 -C 4 It may be substituted by straight chain or branched alkoxy.

Preferred embodiments of the present invention clerical script, wherein R 1 to R 3 in the formula (1) independently represent hydrogen, or straight-chain or substituted or unsubstituted C 1 of the crushing each other - an alkyl of C 5, wherein R 4 is an amino group is substituted a C 1 - C 10 in the alkyl, or 4- (alkoxycarbonyl) benzyl, and more preferably a dialkylamino group or a morpholinyl group (

Figure 112010075100198-pat00005
) Is substituted alkyl.

In a preferred another embodiment, the R 1 to R 5 in the formula (2) independently represent hydrogen, or straight-chain or substituted or unsubstituted C 1 of the crushing-is a C 5 alkyl.

In another preferred embodiment, the R 1 to R 4 of the general formula (3) And R 6 is independently hydrogen, or straight or branched chain of substituted or unsubstituted C 1 to each other - and a C 5 alkyl; R 5 is hydrogen, straight-chain or substituted or unsubstituted ring of grinding C 1 - is a C 5 alkyl, or substituted or unsubstituted benzyl.

More preferably, the gallic acid derivative of the present invention is represented by any one of the following formulas (4) to (10):

Formula 4

Figure 112010075100198-pat00006

Formula 5

Figure 112010075100198-pat00007

6

Figure 112010075100198-pat00008

Formula 7

Figure 112010075100198-pat00009

Formula 8

Figure 112010075100198-pat00010

Formula 9

Figure 112010075100198-pat00011

10

Figure 112010075100198-pat00012

The gallic acid derivative compounds of the present invention may have one or more chiral centers and / or geometric isomeric centers, and thus the present invention provides all stereoisomers represented by any one of Chemical Formulas 1 to 10, that is, optical isomers. , Diastereomers and geometric isomers.

According to another aspect of the present invention, the present invention provides a method for synthesizing the gallic acid derivative comprising coupling a compound of Formula 11 with a compound of Formula 12 or Formula 13.

Formula 11

Figure 112010075100198-pat00013

Formula 12

X- R 4

Formula 13

Figure 112010075100198-pat00014

In the formula (11) to (13), wherein R 1 to R 3 are independently hydrogen, substituted or unsubstituted straight or branched chain of one another unsubstituted C 1 - of C 10 - C 5 alkyl, a substituted or unsubstituted C 6 of Aryl, or substituted or unsubstituted benzyl; R 4 above And R 5 is independently hydrogen, substituted or unsubstituted straight or branched chain of one another unsubstituted C 1 - of C 10 - C 10 alkyl, substituted or unsubstituted C 6 of Aryl, or substituted or unsubstituted benzyl; And X is halogen.

The method for preparing a gallic acid derivative of the present invention relates to a method for chemically synthesizing a gallic acid derivative represented by any one of Chemical Formulas 1 to 10, and since the subject is common, common contents in relation to the gallic acid derivative are In order to avoid excessive complexity of the present specification, the description is omitted.

In a preferred embodiment of the invention, the formula (11) wherein R 1 to R 3 are independently H, or straight-chain or substituted or unsubstituted C 1 of the crushing of each other - in and of C 5 alkyl, the formula 12 R 4 C 1 is the amino group is substituted with - of C 10 alkyl, or a 4- (alkoxycarbonyl) benzyl, and more preferably a dialkylamino group or a morpholinyl group (

Figure 112010075100198-pat00015
) Is substituted alkyl.

In a preferred another embodiment, the R 4 in the formula (3) is hydrogen, straight-chain or substituted or unsubstituted in the grinding ring with C 1 - and a C 5 alkyl, or substituted or unsubstituted benzyl, wherein R 5 is hydrogen, or straight-chain or a substituted or unsubstituted ring of grinding C 1 - is a C 5 alkyl.

The gallic acid derivative of the present invention may be prepared by introducing various substituents into a coupling reaction using the gallic acid basic structure represented by Formula 11 as a backbone, wherein Formula 12 or 13 is introduced into the gallic acid basic structure. It is a substituent. The coupling reaction is preferably a nucleophilic substitution 2 (SN2) reaction, but is not necessarily limited thereto.

In addition, the method for preparing a gallic acid derivative of the present invention may further include a hydrogenolysis step after the coupling reaction step. Hydrolysis refers to a reaction in which a carbon-carbon single bond or a carbon-heteroatom single bond is decomposed to hydrogen, and in the present invention, in the basic structure of gallic acid represented by Formula 11, R 1 to R 3 If any one of the hydrogen is not hydrogenated after the coupling reaction (hydrogenolysis) can be additionally converted to the alkyl gallate or benzyl gallate derivatives to gallate derivatives.

According to another aspect of the present invention, the present invention provides a pharmaceutical composition comprising (a) a pharmaceutically effective amount of the gallic acid derivative or a pharmaceutically acceptable salt thereof; And (b) provides a pharmaceutical composition for the prevention or treatment of cancer comprising a pharmaceutically acceptable carrier.

Derivatives of the present invention prepared based on gallic acid exhibit an effect of inhibiting HAT (histone acetyltransferase) activity, and the mechanisms and contents of HAT related to anticancer activity of various cancers including prostate cancer are known in the art. Because it is widely known [Stimson, L. et al. Mol Cancer Ther. 4 (10): 1521-32, 2005; Balasubramanyam, K. et al. J Biol Chem. 279 (49): 51163-71, 2004], the gallic acid derivative of the present invention can be used as an active ingredient of a pharmaceutical composition for preventing or treating cancer.

Examples of the cancers to be prevented or treated include pituitary adenoma, glioma, brain tumors, glandular cancer, laryngeal cancer, thymoma, mesothelioma, breast cancer, lung cancer, gastric cancer, esophageal cancer, colon cancer, liver cancer, pancreatic cancer, pancreatic endocrine tumor, gallbladder cancer, and penile cancer. , Ureter cancer, renal cell carcinoma, bladder cancer, non-Hodgkin's lymphoma, myelodysplastic syndrome, multiple myeloma, plasmacytoma, leukemia, childhood cancer, skin cancer, ovarian cancer, cervical cancer and prostate cancer, but are not necessarily limited thereto. Preferably, it may be prostate cancer.

Gallic acid derivatives of the present invention may be included in the compositions of the present invention in the form of the compounds of formulas (1) to (10) per se, as well as pharmaceutically acceptable salts thereof. The term "pharmaceutically acceptable salts" refers to formulations of formulations of a compound that do not cause serious irritation to the organism to which the compound is administered and do not impair the biological activity and properties of the compound, for example, ammonium salts, alkalis. Metal salts, alkaline earth metal salts, transition metal salts, quaternary amine salts or amino acid salts, but are not necessarily limited thereto. The term “pharmaceutically effective amount” means an amount necessary to properly induce anticancer activity and to exhibit a pharmacological effect as a cancer prevention or treatment agent.

The composition of the present invention may further include a pharmaceutically acceptable carrier in addition to the above-mentioned gallic acid derivatives included as an active ingredient.

Pharmaceutically acceptable carriers included in the pharmaceutical compositions of the present invention are those commonly used in the preparation, such as lactose, dextrose, sucrose, sorbitol, mannitol, starch, acacia rubber, calcium phosphate, alginate, gelatin, Calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil, and the like It doesn't happen. In addition to the above components, the pharmaceutical composition of the present invention may further include a lubricant, a humectant, a sweetener, a flavoring agent, an emulsifier, a suspending agent, a preservative, and the like. Suitable pharmaceutically acceptable carriers and formulations include, but are not limited to, Remington's Pharmaceutical Sciences (19th ed., 1995).

The pharmaceutical composition of the present invention may be administered parenterally, and in the case of parenteral administration, it may be administered by intravenous injection, subcutaneous injection, intramuscular injection, intraperitoneal injection, transdermal administration, or the like.

The appropriate dosage of the pharmaceutical composition of the present invention may vary depending on factors such as the formulation method, administration method, age, body weight, sex, pathological condition, food, administration time, administration route, excretion rate, . On the other hand, the dosage of the pharmaceutical composition of the present invention is preferably 0.001-100 mg / kg (body weight) per day.

The pharmaceutical composition of the present invention may be formulated into a unit dose form by formulating it using a pharmaceutically acceptable carrier and / or excipient according to a method which can be easily carried out by a person having ordinary skill in the art to which the present invention belongs. Or by intrusion into a multi-dose container. The formulations may be in the form of solutions, suspensions or emulsions in oils or aqueous media, or in the form of excipients, powders, granules, tablets or capsules, and may additionally contain dispersing or stabilizing agents.

The present invention is intended to include within its scope all preparations containing gallic acid derivatives as active ingredients.

According to another aspect of the invention, the present invention provides a food composition for preventing or improving cancer containing a gallic acid derivative as an active ingredient.

The cancers to be prevented or ameliorated include pituitary adenoma, glioma, brain tumor, epiglottis cancer, laryngeal cancer, thymoma, mesothelioma, breast cancer, lung cancer, gastric cancer, esophageal cancer, colon cancer, liver cancer, pancreatic cancer, pancreatic endocrine tumor, gallbladder cancer, penile cancer, Ureter cancer, renal cell cancer, bladder cancer, non-Hodgkin's lymphoma, myelodysplastic syndrome, multiple myeloma, plasmacytoma, leukemia, childhood cancer, skin cancer, ovarian cancer, cervical cancer and prostate cancer, but are not necessarily limited thereto, Preferably it may be prostate cancer.

Gallic acid derivatives, which are the raw materials of the present invention, may be formulated in the form of granules, tablets, capsules or drinks by conventional methods known in the art. Moreover, in order to make storage and handling easy, you may add the carrier used for normal formulation, such as dextrin and cyclodextrin, and other arbitrary preparations. In addition, auxiliary raw materials or additives conventionally added to foods are not particularly limited, but for example, glucose, fructose, sucrose, maltose, sorbitol, stevioside, robsoside, corn syrup, lactose, citric acid, tartaric acid, malic acid, succinic acid , Lactic acid, L-ascorbic acid, d1-α-tocopherol, sodium ellisulfate, glycerin, propylene glycol, glycerin fatty acid ester, polyglycerin fatty acid ester, sucrose fatty acid ester, sorbitan fatty acid ester, gum arabic, colorazine, casein , Gelatin, pectin, agar, vitamin B, nicotinic acid amide, calcium panthenate, amino acids, calcium salts, pigments, fragrances, preservatives and the like.

In addition, the present invention includes a health food comprising the gallic acid derivative as an active ingredient. Health foods are foods made by adding gallic acid derivatives to food materials such as beverages, teas, spices, gums and confectionery, or by encapsulating, powdering, and suspensions. Meaning, unlike the general medicine has the advantage that there is no side effect that can occur when taking a long-term use of the drug as a food raw material. Since the health food of this invention obtained in this way can be ingested routinely, high anticancer and cancer prevention effect can be expected, and it is very useful.

Although the addition amount of the gallic acid derivative in such a health food cannot be uniformly prescribed | regulated depending on the kind of the health food which is object, what is necessary is just to add in the range which does not impair the original taste of food, and it is usually 0.01 to 50% by weight, preferably in the range of 0.1 to 20% by weight. In the case of food in the form of granules, tablets or capsules, it is usually added in the range of 0.1 to 100% by weight, preferably 5 to 100% by weight.

In addition, the gallic acid derivative which is an effective ingredient of the HAT activity inhibitor of the present invention is appropriately administered so that the daily intake amount of adult is 1 to 3000 mg. In addition, the dosage can be appropriately increased or decreased depending on age, symptoms, and the like.

The features and advantages of the present invention are summarized as follows:

(Iii) The present invention provides a novel gallic acid derivative prepared by using gallic acid as a basic structure, a preparation method thereof, and an anticancer composition using the same.

(Ii) The gallic acid derivatives of the present invention exhibit effective anticancer activity by inhibiting the activity of HAT (histone acetyltransferase), and are safe because their natural structure is made of gallic acid, which is a natural substance.

(Iii) The present invention provides basic data as medicines and foods of gallic acid derivatives having anticancer activity.

1 is a diagram showing the anti-HAT activity effect of gallic acid derivatives.
Figure 2 is a chart measuring the effect of each concentration of the superior anti-HAT activity of the gallic acid derivatives.
Figure 3 is a chart comparing the prostate cancer cell viability of the gallic acid derivatives in prostate cancer cell line.

Hereinafter, the present invention will be described in more detail with reference to Examples. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not to be construed as limiting the scope of the present invention. It will be self-evident.

Example

Garlic acid  Derivative production

Synthesis of Gallate derivatives was performed using 3,4,5-tribenzyloxybenzoic acid (Compound 1 ') as a starting material. Compound 1 'was synthesized by the method of Dodo, K. et al. Bioorg . Med . Chem . 16: 7975-7982 (2008). Compound 1 'was alkylated with an alkyl or aryl halide in the presence of a K 2 CO 3 base to give compound 2' or compound 3 '. The structure of the compounds was confirmed by spectroscopic method, and the synthesis method of compounds 2 'and 3' is shown in Figure 1 and 2, respectively.

Figure 1. Synthesis of Compound 2 '

Figure 112010075100198-pat00016

Figure 2. Compound 3 ’( Gal  Synthesis method of 2)

Figure 112010075100198-pat00017

Benzylgallate compounds were removed by benzyl group by hydrogenolysis (hydrogenolysis) with 10% -Pd / C. In the 1 H-NMR and 13 C-NMR spectra, it was confirmed that the benzyl groups were the desired compounds. The synthesis method and structure of the final gallate compounds are shown in Figure 3.

Figure 3. Final Galate  Synthesis Method and Structure of Compounds

Figure 112010075100198-pat00018

Experimental conditions

The solvents and reagents used in the experiments were the highest commercially available, and the solvents were used immediately without purification unless otherwise stated. Alkyl or aryl halides reacted with compound 1 'were all purchased from Sigma Chemical Co. (St. Louis, Mo.). TLC plates were Kieselgel 60 F 254 (Art A715, Merck) and Silica Gel 60 for columns (0.040-0.063 mm ASTM, Merck). 1 H and 13 C NMR data were measured using a Varian NMR AS 400 MHz or Bruker AMX 250 machine, and chemical shifts (δ) were measured in parts per million (ppm) using tetramethylsilane as the internal standard. Coupling constants ( J values) are expressed in Hertz. Mass data was measured using a GC: 7890A MS: 5975C MSD (Agilent, USA) machine with an electron ionization (EI) device. Melting points were taken using a Barnstead International MEL-TEMP1202D machine and the values were not corrected separately.

Tribenzyloxygallate  Compound 2 'and compound 3' ( Gal  General synthesis method of 2) (synthesis method 1)

In a round flask, a mixture of 3,4,5-tribenzyloxybenzoic acid, alkyl bromide or aryl bromide and K 2 CO 3 in DMF was reacted at 40-100 ° C., water was added thereto, followed by extraction with CH 2 Cl 2 or EtOAc. , Organic solvents were collected, washed with sat-NaHOC 3 , and water was removed with Na 2 SO 4 . The remaining residue was purified by silica gel chromatography.

4-( Methoxycarbonyl Benzyl 3,4,5- Tris (benzyloxy) benzoate (Compound 2 ′) Synthesis

3,4,5-tribenzyloxybenzoic acid (3.0 g, 6.82 mmol), methyl bromomethylbenzoate (2.51 g, 10.96 mmol) in a round flask using Synthesis 1 ) (Sigma Chemical Co. (St. Louis, MO)) and K 2 CO 3 (1.01 g, 7.31 mmol) in DMF (50 mL) were stirred at 40C (overnight) and extracted with EtOAc. The residue was purified by silica gel chromatography (eluent: EtOAc / n -hexane = 1: 5 EtOAc) to obtain the compound EtOAc / n - by treatment with hexane to obtain a white solid (2.72 g, 62.8%). mp: 120-122 C; R f = 0.44 (EtOAc / n- hexane = 1: 3) 1 H-NMR (CDCl 3 , 400 MHz) 3.95 (s, 3H), 5.15 (s, 6H), 5.38 (s, 2H), 7.28 (d , J = 1.6 Hz, 2H), 7.34-7.47 (m, 17H), 8.07 (d, J = 8.4 Hz, 2H); 13 C-NMR (CDCl 3 , 100 MHz) 52.4, 66.1, 71.4, 75.3, 109.4, 124.9, 127.8, 128.2, 128.3, 128.8, 130.1, 136.8, 137.5, 141.3, 142.8, 152.7, 165.9, 166.9 ppm.

One- Methoxy -One- Oxopropane 2-yl 3,4,5- Tris (benzyloxy) benzoate (Compound 3 ′) Synthesis

3,4,5-tribenzyloxybenzoic acid (3.0 g, 6.82 mmol), methyl 2-bromopropionate, 2.44 in a round flask using Synthesis Method 1. g, 14.62 mmol) (Sigma Chemical Co. (St. Louis, MO)) and K 2 CO 3 (1.01 g, 7.31 mmol) in DMF (50 mL) were stirred at 40 ° C. (overnight), CH Extracted with 2 Cl 2 . The residue was purified by silica gel chromatography (eluent: EtOAc / n- hexane = 1: 5 1: 3) to obtain a white solid (3.35 g, 92.3%). mp: 78-80 C; R f = 0.42 (EtOAc / n- hexane = 1: 3) 1 H-NMR (CDCl 3 , 400 MHz) 3.95 (s, 3H), 5.15 (s, 6H), 5.38 (s, 2H), 7.28 (d , J = 1.6 Hz, 2H), 7.34-7.47 (m, 17H), 8.07 (d, J = 8.4 Hz, 2H); 13 C-NMR (CDCl 3 , 100 MHz) 52.4, 66.1, 71.4, 75.3, 109.4, 124.9, 127.8, 128.2, 128.3, 128.8, 130.1, 136.8, 137.5, 141.3, 142.8, 152.7, 165.9, 166.9 ppm.

Trihydrooxygallate  General Synthesis of Compounds 1,3-12 (Synthesis Method 2)

A benzylgallate compound synthesized from 3,4,5-tribenzyloxy benzoic acid was added to a round flask using Synthesis Method 1, and then dissolved by adding ethyl acetate. 10% -Pd / C was added to the reaction solution, and a hot air balloon filled with H 2 gas was connected and stirred at room temperature for 3-24 hours. After the reaction was completed, the solid was filtered using Celite, the solvent was removed under reduced pressure to give the product.

4-( Methoxycarbonyl Benzyl 3,4,5- Trihydroxybenzoate ( Gal  1) Synthesis

Compound 2 '(0.2 g, 0.34 mmol) was dissolved in EtOAc (10 mL) and 10% -Pd / C (35 mg) was added and stirred at room temperature for 12 hours. The solvent was removed under reduced pressure, and the obtained brown residue was purified by silica gel column chromatography (eluent: EtOAc / n- hexane = 1: 1) to obtain a white solid (32.0 mg, 30.9%). mp: 168-169 C; R f = 0.37 (MeOH / CH 2 Cl 2 = 1: 9) 1 H-NMR (CD 3 OD, 400 MHz) 3.80 (s, 3H), 5.24 (s, 2H), 7.01 (s, 2H), 7.42 (d, J = 7.6 Hz, 2H), 7.91 (d, J = 7.6 Hz, 2H); 13 C-NMR (CD 3 OD, 100 MHz) 51.5, 65.3, 108.9, 119.9, 127.3, 129.6, 129.7, 138.8, 142.3, 145.4, 166.8, 167.0 ppm.

methyl  2- (3,4,5- Trihydroxybenzamido ) Propanoate ( Gal  3) synthetic

Compound 3 '(0.2 g, 0.38 mmol) was dissolved in EtOAc (8 mL) and 10% -Pd / C (50.00 mg) was added and stirred at room temperature for 12 hours. The solvent was removed under reduced pressure and the brown residue obtained was purified by silica gel column chromatography (MeOH / CH 2 Cl 2 = 1: 9) to obtain a white solid (82.9 mg, 85.5%). R f = 0.25 (MeOH / CH 2 Cl 2 = 1: 9, 1% -AcOH) 1 H-NMR (DMSO-d 6 , 250 MHz) 1.33 (d, J = 7.2 Hz, 3H), 3.60 (s, 3H), 4.34-4.36 (m, 1H), 6.84 (s, 2H), 8.40 (d, J = 6.73 Hz, 1H), 8.73 (s, 1H), 9.08 (s, 2H); 13 C-NMR (DMSO-d 6 , 62.5 MHz) 17.0, 48.4, 52.0, 107.2, 124.2, 136.7, 145.6, 166.6, 173.7 ppm.

methyl  4- methyl -2- (3,4,5- Trihydroxybenzamido ) Pentanoate ( Gal  4) synthetic

Benzyl-bonded benzylgallate compound (0.15 g, 0.26 mmol) synthesized from 3,4,5-tribenzyloxy benzoic acid in the same manner as described above (synthesis method 1) was dissolved in EtOAc (5 mL) and 10% -Pd / C (45.0 mg) was added and stirred at room temperature for 15 hours. The solvent was removed under reduced pressure and the brown residue obtained was purified by silica gel column chromatography (eluent: EtOAc / n- hexane = 1: 1 EtOAc) to obtain a white solid (67.0 mg, 86.7%). R f = 0.15 (MeOH / CH 2 Cl 2 = 1: 15); 1 H-NMR (CDCl 3 , 250 MHz) 0.78 (d, J = 3.2 Hz, 6H), 1.64-1.66 (m, 3H), 3.57 (s, 3H), 4.58-4.60 (m, 1H), 6.78 ( s, 2H); 13 C-NMR (CDCl 3 , 62.5 MHz) 21.6, 21.7, 40.5, 51.8, 52.6, 107.7, 124.1, 136.3, 144.3, 168.9, 174.7 ppm.

methyl  3- Phenyl -2- (3,4,5- Trihydroxybenzamido ) Propanoate ( Gal  5) synthetic

Benzyl-bonded benzylgallate compound (0.15 g, 0.25 mmol) synthesized from 3,4,5-tribenzyloxy benzoic acid by the same method as described above (Synthesis Method 1) was dissolved in EtOAc (5 mL) and 10% -Pd / C (48.0 mg) was added and stirred at room temperature for 11 hours. The solvent was removed under reduced pressure to give a white solid (83.0 mg, quantitative). R f = 0.27 (EtOAc / n- hexane = 3: 1) 1 H-NMR (CDCl 3 , 250 MHz) 3.05 (brs, 2H), 3.51 (s, 3H), 4.82-4.84 (m, 1H), 6.75 (s, 2H), 7.01-7.13 (m, 5H); 13 C-NMR (CDCl 3 , 62.5 MHz) 37.2, 52.5, 54.3, 107.5, 124.0, 127.0, 128.5, 129.1, 135.9, 136.3, 144.5, 168.5, 172.9 ppm.

3- Morpholinopropyl  3,4,5- Trihydroxybenzoate ( Gal  6) synthetic

3-morpholinopropyl 3,4,5-tribenzyloxybenzoate benzylgallate compound (0.15 g, 0.26 mmol) synthesized in the same manner as described above (Synthesis Method 1) was dissolved in EtOAc (5 mL) and 10% -Pd / C (40.0 mg) was added followed by stirring at room temperature for 12 hours. After removing the solvent under reduced pressure, the obtained residue was purified by silica gel column chromatography (MeOH / CH 2 Cl 2 = 1: 6) to obtain a white solid (39.5 mg, 51.1%). R f = 0.43 (MeOH / CH 2 Cl 2 = 1: 5) 1 H-NMR (DMSO-d 6 , 250 MHz) 1.81 (quint, J = 6.2 Hz, 2H), 2.34-2.40 (m, 6H), 3.50-3.60 (m, 4H), 4.18 (t, J = 6.2 Hz, 2H), 6.92 (s, 2H), 8.95 (s, 1H), 9.27 (s, 2H); 13 C-NMR (DMSO-d 6 , 62.5 MHz) 25.5, 53.5, 55.1, 62.7, 66.3, 108.7, 119.7, 138.6, 145.8, 166.1 ppm.

3- (dimethylamino) propyl 3,4,5- Trihydroxybenzoate ( Gal  7) synthetic

3- (dimethylamino) propyl 3,4,5-tribenzyloxybenzoate benzylgallate compound (0.16 g, 0.29 mmol) synthesized in the same manner as described above (synthesis method 1) was dissolved in EtOAc (5 mL), and % -Pd / C (45.0 mg) was added followed by stirring at room temperature for 6 hours. The solvent was removed under reduced pressure to give a brown solid (34.6 mg, 46.7%). R f = 0.27 (EtOAc / n- hexane = 3: 1) 1 H-NMR (DMSO-d 6 , 250 MHz) 1.78 (quint, J = 6.0 Hz, 2H), 2.16 (s, 6H), 2.35 9t, J = 6.0 Hz, 2H), 4.16 (t, J = 6.0 Hz, 2H), 6.92 (6.89) (s, 2H); 13 C-NMR (DMSO-d 6 , 62.5 MHz) 26.3, 45.0, 55.7, 62.5, 108.7, 119.7, 138.6, 145.8, 166.1 ppm.

Naming of the Compound

Compound 3 ′ synthesized using Synthesis 1 was named GAL 2 and the derivatives synthesized by Synthesis 2 using the compounds produced by Synthesis 1 were GAL 1, GAL 3, GAL 4, GAL 5, and GAL 6, respectively. And GAL 7, GAL 8, GAL 9, GAL 10, GAL 11 and GAL 12, respectively.

Example  One: Garlic acid  For derivatives HAT  Confirm inhibitory activity

LNCaP cells (ATCC, Rockville, Md, USA), a human prostate cancer cell line, were grown to complete confluence on 150 mm plates, harvested, and centrifuged at 1000 rpm for 3 minutes. Pellets were collected. 20 ml of cold buffer A (10 mM pH 7.9 Hepes, 1.5 mM MgCl 2 , 10 mM KCl, 0.5 mM DTT, and 0.2 mM PMSF) was added to the collected pellets, and the mixture was left on ice for 10 minutes, followed by 5 minutes at 4 ° C and 3000 rpm. Centrifugation was carried out, the pellets were collected again, and homogenized with 6 ml of the buffer A as described above. The prepared cell homogenate was centrifuged at 4 ° C. and 12500 rpm for 30 minutes to collect pellets, and buffer C (20 mM pH 7.9 Hepes, 25% glycerol, 0.42 M NaCl, 1.5 mM MgCl 2 , 0.2 mM EDTA, 0.5 mM DTT, 4 ml of 0.2 mM PMSF) was added, resuspended, and left to stand on ice for 30 minutes, followed by centrifugation at 4 ° C and 12500 rpm for 30 minutes. Finally, the supernatant obtained by centrifuging the supernatant at 4 ° C and 30000 rpm for 1 hour was collected and used for the experiment. In order to measure the HAT inhibitory activity, the absorbance was measured at 440 nm using the HAT Activity Chromatography Kit (Biovision, CA, USA) with 50 μg of the nuclear extract.

As a result of the above method, when the gallic acid derivatives were treated, the anti-HAT activity was shown to be about 2 times as compared with that of gallic acid (FIG. 1). Here, HAT activity inhibition capacity (%) was converted according to the following equation (1).

Equation  One

% Inhibition of HAT activity = [1- (As-Ab / Ac-Ab)] X 100

Ac: absorbance at distilled water treatment

Ab: absorbance of blank

As: absorbance of sample treatment

Example  2: HAT  Restraining effect Galate  By concentration of derivatives HAT  Confirm inhibitory activity

As a result of measuring the HAT activity by the above method, the most effective derivatives (Gal 1 and Gal 2) were selected, and gallate was used as a control to measure the HAT activity by concentration. The same as the above method, the final concentration of the sample was 5, 25, 50 and 100 (μm) and the control gallate was 100 μm (Fig. 2).

Example  3: inhibitory activity of growth and proliferation in prostate cancer cell line

To examine the activity of gallic acid and its derivatives on the growth and proliferation of prostate cancer, the prostate cancer cell lines LNCaP and PC3 (ATCC, Rockville, Md, USA) were inoculated in 96 well-plates at 1.5 x 10 4 cells / well, respectively. After 18 hours incubation at 37 ℃ gallic acid and its derivatives were treated with a final concentration of 100 ㎍ / ㎖ 48 hours after MTT [3- (4, 5-dimethylthiazol-2-yl) -2, 5-diphenyltetrazolium bromide: 1 mg / mL] was added and reacted at 37 ° C. for 2 hours. Formazan (formazane) formed was measured by measuring absorbance at 570 nm and reference 630 nm. As a result of the above method, 1 and 2 of the gallic acid derivatives showed an effect on inhibiting cancer cell growth, and it was confirmed that these two derivatives had an excellent anticancer effect (FIG. 3).

Formulation example  1: preparation of tablets

The following formulation tablets (based on 200 parts by weight of total composition) were prepared by a conventional tablet press.

20 parts by weight of gallic acid derivative powder

Dextrin 72 parts by weight

80 parts by weight per minute

8 parts by weight of glycerin fatty acid ester

Mixing and tableting of raw materials were easy, and refined refinement | purification was obtained.

Formulation example  2: Preparation of capsules

By the usual method, the capsule which has the following compositions was manufactured. In addition, No. 1 hard gelatin capsule was used for the capsule.

Composition in 1 Capsule (200 mg)

Gallic acid derivative powder 5 mg

Cornstarch 60.0 mg

Lactose 100.0 mg

Calcium Lactate 10.0 mg

Hydroxypropylcellulose (HPC-L) 10.0 mg

Formulation example  3: Preparation of Granules

By conventional methods, instant tea granules of the following formulations were prepared by fluid bed granulator (based on a total composition of 970 parts by weight).

20 parts by weight of gallic acid derivative powder

40 parts by weight of oligosaccharide

50 parts by weight of citric acid

50 parts by weight of sugar

Dextrin 810 parts by weight

Mixing of the raw materials and granulation with a fluidized bed granulator were easy, and a fine grain of instant tea was obtained.

Formulation example  5: beverage manufacturing

After dissolving 50 mg of the gallic acid derivative according to the present invention in an appropriate amount of water, vitamin C as an auxiliary component, citric acid, sodium citrate, and oligosaccharides as an auxiliary component are added, and an appropriate amount of sodium benzoate is added as a preservative, followed by the addition of water. To 100 ml to prepare a beverage composition. At this time, taurine, myo-inositol, folic acid, pantothenic acid, etc. may be added alone or together.

While the present invention has been particularly shown and described with reference to exemplary embodiments thereof, it is to be understood that the same is by way of illustration and example only and is not to be construed as limiting the scope of the present invention. Accordingly, the actual scope of the present invention will be defined by the appended claims and their equivalents.

Claims (15)

delete delete delete delete delete delete delete delete (a) a pharmaceutically effective amount of one or more gallic acid derivatives or pharmaceutically acceptable salts thereof selected from the group consisting of: And (b) a pharmaceutically acceptable carrier, wherein the pharmaceutical composition for preventing or treating cancer comprises:
Formula 4
Figure 112013009938827-pat00022

Formula 5
Figure 112013009938827-pat00023

In Formula 5, Bn is a benzyl group.
The composition of claim 9, wherein the composition inhibits proliferation of cancer cells by inhibiting histone acetyltransferase (HAT) activity.
10. The method of claim 9, wherein the cancer is pituitary adenoma, glioma, brain tumor, epiglottis cancer, laryngeal cancer, thymoma, mesothelioma, breast cancer, lung cancer, gastric cancer, esophageal cancer, colon cancer, liver cancer, pancreatic cancer, pancreatic endocrine tumor, gallbladder cancer, penile cancer, Ureter cancer, renal cell cancer, bladder cancer, non-Hodgkin's lymphoma, myelodysplastic syndrome, multiple myeloma, plasmacytoma, leukemia, childhood cancer, skin cancer, ovarian cancer, cervical cancer and prostate cancer Composition.
12. The composition of claim 11, wherein the cancer is prostate cancer.
A food composition for preventing or improving cancer comprising one or more gallic acid derivatives selected from the group consisting of Formulas 4 to 5:
Formula 4
Figure 112013009938827-pat00034

Formula 5
Figure 112013009938827-pat00035

In Formula 5, Bn is a benzyl group.
The method according to claim 13, wherein the cancer is pituitary adenoma, glioma, brain tumor, epiglottis cancer, laryngeal cancer, thymoma, mesothelioma, breast cancer, lung cancer, gastric cancer, esophageal cancer, colon cancer, liver cancer, pancreatic cancer, pancreatic endocrine tumor, gallbladder cancer, penile cancer, Ureter cancer, renal cell cancer, bladder cancer, non-Hodgkin's lymphoma, myelodysplastic syndrome, multiple myeloma, plasmacytoma, leukemia, childhood cancer, skin cancer, ovarian cancer, cervical cancer and prostate cancer Composition.
15. The composition of claim 14, wherein the cancer is prostate cancer.
KR20100114412A 2010-11-17 2010-11-17 Composition for Anti-cancer Effect Comprising Gallic Acid Derivatives KR101286153B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR20100114412A KR101286153B1 (en) 2010-11-17 2010-11-17 Composition for Anti-cancer Effect Comprising Gallic Acid Derivatives

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR20100114412A KR101286153B1 (en) 2010-11-17 2010-11-17 Composition for Anti-cancer Effect Comprising Gallic Acid Derivatives

Publications (2)

Publication Number Publication Date
KR20120053245A KR20120053245A (en) 2012-05-25
KR101286153B1 true KR101286153B1 (en) 2013-07-26

Family

ID=46269503

Family Applications (1)

Application Number Title Priority Date Filing Date
KR20100114412A KR101286153B1 (en) 2010-11-17 2010-11-17 Composition for Anti-cancer Effect Comprising Gallic Acid Derivatives

Country Status (1)

Country Link
KR (1) KR101286153B1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102924318B (en) * 2012-11-26 2014-06-04 武汉大学 L-leucine derivatives blocking entrance of H5N1 avian influenza virus and preparation method for same
WO2017066965A1 (en) * 2015-10-22 2017-04-27 The University Of Hong Kong Gallic acid-l-leucine conjugate for resolution of inflammation and sepsis
KR101651605B1 (en) 2015-12-10 2016-08-26 대봉엘에스 주식회사 Novel phenolic acid derivatives, and uses of the same
CN108017585A (en) * 2017-12-15 2018-05-11 广西壮族自治区中医药研究院 A kind of gallic acid sulphone amide derivative and its application in anti-liver cancer and anti-
KR102100460B1 (en) * 2018-07-16 2020-04-13 대봉엘에스 주식회사 Syringic acid derivatives, and uses of the same

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Chen et al. Gallic acid, a major component of Toona sinensis leaf extracts, contains a ROS-mediated anti-cancer activity in human prostate cancer cells. Cancer Letters, Vol.286, 2009, pp.161-171. *
Chen et al. Gallic acid, a major component of Toona sinensis leaf extracts, contains a ROS-mediated anti-cancer activity in human prostate cancer cells. Cancer Letters, Vol.286, 2009, pp.161-171.*
Fiuza et al. Phenolic acid derivatives with potential anticancer properties - a structure - activity relationship study. Bioorganic & Medicinal Chemistry, Vol.12, 2004, pp.3581-3589. *
Fiuza et al. Phenolic acid derivatives with potential anticancer properties - a structure - activity relationship study. Bioorganic & Medicinal Chemistry, Vol.12, 2004, pp.3581-3589.*
Kubo et al. Molecular design of antibrowning agents : antioxidative tyrosinase inhibitors. Food Chemistry, Vol.81, 2003, pp.241-247. *
Kubo et al. Molecular design of antibrowning agents : antioxidative tyrosinase inhibitors. Food Chemistry, Vol.81, 2003, pp.241-247.*
Rivero-Cruz et al. Effect of natural and synthetic benzyl benzoates on calmodulin. Phytochemistry, Vol.68, 2007, pp.1147-1155. *
Rivero-Cruz et al. Effect of natural and synthetic benzyl benzoates on calmodulin. Phytochemistry, Vol.68, 2007, pp.1147-1155.*

Also Published As

Publication number Publication date
KR20120053245A (en) 2012-05-25

Similar Documents

Publication Publication Date Title
KR101286153B1 (en) Composition for Anti-cancer Effect Comprising Gallic Acid Derivatives
WO2009096655A1 (en) Composition comprising the compound isolated from the flower extract of daphne genkwa for preventing and treating cancer disease and the use thereof
KR101424360B1 (en) Composition comprising an hydroxychalcone compounds as an active ingredient for anti-cancer activity
JP2010070541A (en) Tg synthesis inhibitor
KR101669759B1 (en) Composition for Preventing or Treating inflammatory bowel disease comprising Hydroxybenzilidene Chromanone based Compounds
KR100902094B1 (en) Pharmaceutical composition for the prevention and treatment of cancers containings extracts, fractions and isolated 2?arylbenzofuran compounds of Mori Cortex Radicis as an active ingredient
JP6855263B2 (en) Dipeptidylpeptidase-IV inhibitor
KR101734650B1 (en) Novel Benzylidene Dihydro Indenone based Compounds and Composition for Preventing or Treating Inflammatory Bowel Disease comprising the Same
KR20120036146A (en) Composition for anti-cancer effect comprising pomegranate shell extract
KR101512095B1 (en) Composition for preventing or treating renal disease containing extract of amorpha fruticosa
KR101703827B1 (en) Novel Curcumin Derivatives and Uses Thereof
KR101724425B1 (en) Composition preventing or treating cancer comprising (Z)-2-acetamido-3-(4-hydroxy-3-methoxyphenyl)acrylic acid
KR101697062B1 (en) Novel geranyl flavonoid derivative having improved solubility in water, a method for preparing the same and a pharmaceutical composition comprising the same for prevention and treatment of cancer
KR102565433B1 (en) A composition for treating, preventing and improving bone associated diseases containing an ulmus extracts or an effective single compound isolated therefrom
KR101712708B1 (en) Composition for preventing or treating inflammatory bowel disease comprising Indeno Pyridinium chloride compound as an active ingredient
US11168092B2 (en) Compounds and method of preventing or treating cancer using the same
KR101892659B1 (en) Pharmaceutical composition or functional food containing malaxinic acid for improvement of insulin sensitivity
CN113304129B (en) Application of mono-ketene monocarbonyl curcumin analogue in preparing antioxidant drugs
KR100983318B1 (en) Use in anti-cancer agent of [(meso-Bis(o-hydroxyphenyl)bis(o-Hydroxy)salen, 3,3'-(1E,1'E)-((1R,2S)-1,2-bis(2-hydroxyphenyl)ethane-1,2-diyl)bis(azan-1-yl-1-ylidene)bis(methan-1-yl-1-ylidene)dibenzene-1,2-diol)]
JP2017132762A (en) MRNA maturation inhibitor
KR101112678B1 (en) Composition comprising glycitin as an effective ingredient for treatment and prevention of inflammatory diseases, immune diseases or cancers
KR101426210B1 (en) Composition comprising an hydroxychalcone compounds as an active ingredient for preventing and treating angiogenesis relating diseases
KR101024399B1 (en) Inhibitor of histone acetyltransferase comprising extract of Rubus coreanus fruit as an active ingradient
KR20100138398A (en) Composition comprising 3-o-glucose-isorhamnetin as an effective ingredient for treatment and prevention of inflammatory diseases, immune diseases or cancers
KR100983319B1 (en) Use in anti-cancer agent of [meso-Bis(o-hydroxyphenyl)bis(p-nitro)salen, 2,2'-(1E,1'E)-((1R,2S)-1,2-bis(2-hydroxyphenyl)ethane-1,2-diyl)bis(azan-1-yl-1-ylidene)bis(methan-1-yl-1-ylidene)bis(4-nitrophenol)]

Legal Events

Date Code Title Description
A201 Request for examination
E902 Notification of reason for refusal
AMND Amendment
E601 Decision to refuse application
AMND Amendment
E902 Notification of reason for refusal
AMND Amendment
X701 Decision to grant (after re-examination)
GRNT Written decision to grant
FPAY Annual fee payment

Payment date: 20160629

Year of fee payment: 4

FPAY Annual fee payment

Payment date: 20180702

Year of fee payment: 6

FPAY Annual fee payment

Payment date: 20190708

Year of fee payment: 7