AU2021257523A1 - Ginsenoside composition having alcoholic fatty liver-preventing and -treating function - Google Patents

Ginsenoside composition having alcoholic fatty liver-preventing and -treating function Download PDF

Info

Publication number
AU2021257523A1
AU2021257523A1 AU2021257523A AU2021257523A AU2021257523A1 AU 2021257523 A1 AU2021257523 A1 AU 2021257523A1 AU 2021257523 A AU2021257523 A AU 2021257523A AU 2021257523 A AU2021257523 A AU 2021257523A AU 2021257523 A1 AU2021257523 A1 AU 2021257523A1
Authority
AU
Australia
Prior art keywords
ginsenoside
pharmaceutical composition
composition
weight
alcoholic fatty
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.)
Granted
Application number
AU2021257523A
Other versions
AU2021257523B2 (en
Inventor
Zhiguang DUAN
Daidi Fan
Jing He
Jianya YAN
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.)
Shaanxi Giant Biotechnology Co Ltd
Original Assignee
Shaanxi Giant Biotechnology Co Ltd
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 Shaanxi Giant Biotechnology Co Ltd filed Critical Shaanxi Giant Biotechnology Co Ltd
Publication of AU2021257523A1 publication Critical patent/AU2021257523A1/en
Application granted granted Critical
Publication of AU2021257523B2 publication Critical patent/AU2021257523B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

Disclosed in the present invention is a ginsenoside composition having an alcoholic fatty liver-preventing and -treating function. The pharmaceutical composition able to be used to prevent and/or treat alcoholic fatty liver of the present invention contains a prophylactically and/or therapeutically effective dose of ginsenoside Rk3 and ginsenoside Rh4 as active ingredients, and a pharmaceutically acceptable carrier. In the pharmaceutical composition, the mass ratio between the ginsenoside Rk3 and the ginsenoside Rh4 is 1:0.5-2, and is preferably 1:0.9-1.1. The ginsenoside composition of the present invention is able to markedly improve liver function index abnormalities caused by a large amount of alcohol consumption, reducing the generation of inflammatory infiltrates and fat vacuoles in liver tissue. The present invention provides a synergistically interacting pharmaceutical composition containing ginsenosides Rk3 and Rh4 and having alcoholic fatty liver-preventing and/or -treating effects.

Description

W O202202 1209070 A 1 ||1||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| (8p AT-j 4)) M): ARIPO (BW, GH, GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, TZ, UG, ZM, ZW), Skl (AM, AZ, BY, KG, KZ, RU, TJ, TM), [III (AL, AT, BE, BG, CH, CY, CZ, DE, DK, EE, ES, Fl, FR, GB, GR, HR, HU, IE, IS, IT, LT, LU, LV, MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, SM, TR), OAPI (BF, BJ, CF, CG, CI, CM, GA, GN, GQ, GW, KM, ML, MR, NE, SN, TD, TG).
- # K[I Tf*R(* M21*(3))
GINSENOSIDE COMPOSITION HAVING ALCOHOLIC FATTY LIVER-PREVENTING AND -TREATING FUNCTION FIELD OF THE INVENTION
The application relates to the field of biomedicine, in particular to a ginsenoside Rk3/Rh4
composition for preventing and treating alcoholic fatty liver.
BACKGROUND OF THE INVENTION
Fatty liver disease is a common disease. According to the history of heavy drinking, it can be divided
into alcoholic fatty liver (AFL) disease and non-alcoholic fatty liver (NAFLD) disease. AFL is
alcoholic liver damage caused by long-term alcohol consumption. The main reason for the alcoholic
liver damage is the toxic metabolites, oxidative stress and metabolic disorders caused by alcohol
metabolism in liver cells. The pathogenesis of NAFLD is mainly related to IR, abnormal liver fat
metabolism, mitochondrial dysfunction and oxidative stress, genetic variation and metabolic changes,
and susceptibility to cell damage. Although the major cause of the liver diseases is viral hepatitis,
with the rising status of alcohol in the human diet, the incidence of alcoholic fatty liver disease is
getting higher and higher, and the chances of developing liver cirrhosis and liver cancer are much
higher than NAFLD. It has become the second leading cause of liver damage after viral hepatitis.
Polyene phosphatidylcholine is a commonly used drug for adjuvant treatment of alcoholic fatty liver
disease. It can effectively reduce oxygen free radicals, protect liver cell membrane, delay liver
fibrosis, and thus promote the recovery of patients. However, it is clinically believed that single-drug
therapy will cause slower recovery of patients, which is likely to cause intestinal disorders and lead
to diarrhea.
Ginsenoside, the active ingredient in ginseng, is a steroid compound that can affect multiple
metabolic pathways in the body. Studies have reported that ginsenosides Rk3 and Rh4 have obvious
anti-tumor, anti-oxidant, anti-inflammatory, and anti-apoptotic effects. There are also reports in the
literature that ginsenosides CK, Rhl and compositions thereof can be used to improve non-alcoholic
fatty liver fibrosis and insulin resistance drugs, but no research has been conducted on alcoholic fatty
liver; ginsenoside Rgl has a certain effect in the treatment of metabolic disorders caused by drinking,
but whether it has a protective effect on the liver has not been studied, while the combination of ginsenoside Rk3/Rh4 has never been used to improve alcohol-induced fatty liver, liver disease, and hepatic antioxidant reduction, etc.
SUMMARY OF THE INVENTION
The purpose of the present application is to provide a ginsenoside Rk3/Rh4 composition with the
function of preventing and treating alcoholic fatty liver disease.
The inventors found that the combination of ginsenoside Rk3/Rh4 can effectively improve the degree
of liver damage and steatosis caused by drinking, and thus the ginsenoside Rk3/Rh4 composition was
further applied to the treatment and remission of alcoholic fatty liver disease, and it is the first time
that the ginsenoside Rk3/Rh4 composition is used as the main component in the prevention and
treatment of alcoholic fatty liver disease, and the composition has good treatment and improvement
effects on alcoholic fatty liver disease. In a mouse model of alcoholic fatty liver disease, the
Rk3/Rh4 composition can significantly improve the survival rate of drinking mice, reduce the
content of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST), and reduce
the content of cholesterol (TC), triglyceride (TG), low density lipoprotein (LDL) in serum and liver,
reduce lipid accumulation (oil droplets), inflammatory infiltration of liver tissue and fat vacuoles in
liver caused by alcohol consumption.
That is, the present application comprises:
1. A pharmaceutical composition for the preventing and/or treating alcoholic fatty liver disease,
comprising prophylactically and/or therapeutically effective amount of ginsenoside Rk3 and
ginsenoside Rh4 as active ingredients, and a pharmaceutically acceptable carrier,
wherein in the pharmaceutical composition, the weight ratio of the ginsenoside Rk3 to the
ginsenoside Rh4 is 1:0.5-2, preferably 1:0.9~1.1.
2. The pharmaceutical composition according to item 1, wherein in the pharmaceutical composition,
the weight ratio of the ginsenoside Rk3 to the ginsenoside Rh4 is 1:1.
3. The pharmaceutical composition according to item 1, wherein, based on 100 parts by weight of the
total ginsenosides comprised in the pharmaceutical composition, the total amount of the ginsenoside
Rk3 and the ginsenoside Rh4 comprised in the pharmaceutical composition is 10 parts by weight or
more.
4. The pharmaceutical composition according to item 1, wherein, based on 100 parts by weight of
total ginsenosides comprised in the pharmaceutical composition, the total amount of the ginsenoside
Rk3 and the ginsenoside Rh4 comprised in the pharmaceutical composition is 100 parts by weight.
5. The pharmaceutical composition according to item 1, wherein the pharmaceutical composition
does not comprise other active ingredients other than the ginsenoside Rk3 and the ginsenoside Rh4
for preventing and/or treating alcoholic fatty liver disease.
6. Use of ginsenoside Rk3 and ginsenoside Rh4 in the preparation of a pharmaceutical composition
for preventing and/or treating alcoholic fatty liver disease, wherein, in the pharmaceutical
composition, the weight ratio of the ginsenoside Rk3 to the ginsenoside Rh4 is 1:0.5-2, preferably
1:0.9~1.1.
7. The use according to item 6, wherein, in the pharmaceutical composition, the weight ratio of the
ginsenoside Rk3 to the ginsenoside Rh4 is 1:1.
8. The use according to item 6, wherein, based on 100 parts by weight of total ginsenosides
comprised in the pharmaceutical composition, the total amount of the ginsenoside Rk3 and the
ginsenoside Rh4 comprised in the pharmaceutical composition is 10 parts by weight or more.
9. The use according to item 6, wherein, based on 100 parts by weight of total ginsenosides
comprised in the pharmaceutical composition, the total amount of the ginsenoside Rk3 and the
ginsenoside Rh4 comprised in the pharmaceutical composition is 100 parts by weight.
10. The use according to item 6, wherein the pharmaceutical composition does not comprise other
active ingredients other than the ginsenoside Rk3 and the ginsenoside Rh4 for preventing and/or
treating alcoholic fatty liver disease.
The ginsenoside Rk3/Rh4 composition for preventing and treating alcoholic fatty liver disease
according to the present application can be either a medicine or any other preparation form; it can be
used for the purpose of treating related diseases, or it can be used to improve or prevent fatty liver
caused by alcohol consumption. The preparation can be ginsenoside Rk3/Rh4, or a compound
preparation composed of ginsenoside Rk3/Rh4 and other components, wherein ginsenoside Rk3/Rh4
is used as the main component.
The ginsenoside Rk3/Rh4 of the present application can be obtained by extraction, transformation
and separation of ginseng or American ginseng, or by chemical synthesis. Specifically, the ginsenoside Rk3/Rh4 can be obtained by extracting, transforming and separating from the roots, stems and/or leaves of ginseng or American ginseng by utilizing the process methods of the prior art.
In addition, the inventors also found that in the mouse model of alcoholic fatty liver disease, Rk3 or
Rh4 can improve the survival rate of drinking mice, reduce the content of alanine aminotransferase
(ALT) and aspartate aminotransferase (AST) in serum, and reduce the content of cholesterol (TC),
triglyceride (TG), and low-density lipoprotein (LDL) in serum and liver, reduce alcohol-induced
lipid accumulation (oil droplets), inflammatory infiltration of liver tissue, and fat vacuoles in the
liver.
Therefore, the present application also includes:
11.Use of ginsenoside Rk3 in the preparation of a pharmaceutical composition for preventing and/or
treating alcoholic fatty liver disease.
12.The use according to item 11, wherein based on 100 parts by weight of total ginsenosides
comprised in the pharmaceutical composition, the amount of the ginsenoside Rk3 comprised in the
pharmaceutical composition is 5 parts by weight or more, preferably 10 parts by weight or more
, more preferably 20 parts by weight or more, more preferably 50 parts by weight or more, more
preferably 70 parts by weight or more, more preferably 80 parts by weight or more, more preferably
parts by weight or more, more preferably 95 parts by weight or more, more preferably 99 parts by
weight or more, more preferably, 100 parts by weight.
13.The use according to item 11, wherein the pharmaceutical composition does not comprise other
active ingredients other than the ginsenoside Rk3 for preventing and/or treating alcoholic fatty liver
disease.
14.Use of ginsenoside Rh4 in the preparation of a pharmaceutical composition for preventing and/or
treating alcoholic fatty liver disease.
15.The use according to item 14, wherein, based on 100 parts by weight of total ginsenosides
comprised in the pharmaceutical composition, the amount of the ginsenoside Rh4 comprised in the
pharmaceutical composition is 5 parts by weight or more, preferably 10 parts by weight or more,
more preferably 20 parts by weight or more, more preferably 50 parts by weight or more, more
preferably 70 parts by weight or more, more preferably 80 parts by weight or more, more preferably
parts by weight or more, more preferably 95 parts by weight or more, more preferably 99 parts by
weight or more, more preferably, 100 parts by weight.
16.The use according to item 14, wherein the pharmaceutical composition does not comprise other
active ingredients of preventing and/or treating alcoholic fatty liver disease other than the
ginsenoside Rh4.
The above-mentioned pharmaceutical composition may be, for example, an oral preparation or an
injection. The oral preparation can be, for example, hard capsules, soft capsules, slow-release
capsules, sugar-coated tablets, powders, granules, tablets, medicinal granules, dropping pills,
honeydew pills, syrups or oral liquids; the injections are solution type, mixed suspension type,
emulsion type or lyophilized powder. The above-mentioned pharmaceutical compositions may also
be in external dosage forms, such as ointments, gels, sprays or patches. The above-mentioned
pharmaceutical compositions may comprise adjuvants or other pharmaceutically acceptable carriers.
The ratio of the adjuvant to the active ingredient of the drug can be, for example, the active
ingredient accounts for 30% to 50% by weight, and the adjuvant accounts for 50% to 70% by weight.
The adjuvant can be, for example, one or more of sodium hyaluronate, magnesium stearate, sodium
alginate, starch, microcrystalline cellulose, chitosan, stachyose, adhesive or collagen.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the chemical structural formula of the ginsenoside Rk3
Figure 2 is the chemical structural formula of the ginsenoside Rh4
Figure 3 is a graph showing the results of HE staining of mouse liver tissue in Example 4. Wherein,
1: a graph showing HE staining of liver tissue of mice in control group;
2: a graph showing HE staining of liver tissue of mice in model group;
3: a graph showing HE staining of liver tissue of mice in positive drug control group;
4: a graph showing HE staining of liver tissue of mice in ginsenoside Rk3 group;
: a graph showing HE staining of liver tissue of mice in ginsenoside Rh4 group;
6: a graph showing HE staining of liver tissue of mice in ginsenoside Rk3/Rh4 composition group 1;
7: a graph showing HE staining of liver tissue of mice in ginsenoside Rk3/Rh4 composition group 2;
8: a graph showing HE staining of liver tissue of mice in ginsenoside Rk3/Rh4 composition 3
groups;
9: a graph showing HE staining of liver tissue of mice in ginsenoside Rgl group;
: a graph showing HE staining of liver tissue of mice in ginsenoside CK/Rhl group.
DETAIL DESCRIPTION OF THE INVENTION
First, in one aspect, the present application provides a pharmaceutical composition that can be used
for the prevention and/or treatment of alcoholic fatty liver disease, comprising prophylactically
and/or therapeutically effective amount of ginsenoside Rk3 and ginsenoside Rh4 as active
ingredients, and a pharmaceutically acceptable carrier,
In the pharmaceutical composition, the weight ratio of the ginsenoside Rk3 to the ginsenoside Rh4 is
1:0.5-2.
In this specification, the ginsenoside Rk3 refers to the compound shown in Figure 1, and its
molecular formula is C 3 6H 6 0 0 8 .
The above-mentioned ginsenoside Rk3 is a known compound, and can be prepared by methods
known in the art, for example, by adding an organic acid solution to to triol group ginsenosides in a
directional conversion reaction under high temperature and high pressure conditions to obtain a
higher-purity ginsenoside Rk3 crude product, and then purified by high-performance liquid phase
separation and other methods to obtain pure ginsenoside Rk3 with a purity of >98%.
In this specification, the ginsenoside Rh4 refers to the compound shown in Figure 2, and its
molecular formula is C 3 6H 6 0 0 8 .
The above-mentioned ginsenoside Rh4 is a known compound and can be prepared by methods
known in the art. For example, triol group ginsenosides are added with organic acid solutions, and
after a directional conversion reaction under high temperature and high pressure conditions, the pure
ginsenoside Rh4 with a purity of >98% is obtained by separation and purification.
The inventor found that the combination of the ginsenoside Rk3 and the ginsenoside Rh4 according
to the ratio range defined in the present application showed synergistic effect in the prevention and/or
treatment of alcoholic fatty liver disease (CIvalue is less than 1, preferably CI value is less than 0.8,
more preferably less than 0.7). The inventors also found that within the above ratio range, the
ginsenoside Rk3 and the ginsenoside Rh4 are also low in toxicity. Moreover, the inventors found that
if the contents of the ginsenoside Rk3 and the ginsenoside Rh4 in the pharmaceutical composition
are not within the ratio range defined in the present application, there is no synergistic effect.
Considering the significant synergistic effect, in the composition, the weight ratio of the ginsenoside
Rk3 and the ginsenoside Rh4 is preferably 1:0.9~1.1, more preferably 1:1.
In the pharmaceutical composition of the present application, other ginsenosides may or may not be
comprised. When the pharmaceutical composition of the present application comprises other
ginsenosides, from the perspective of better playing the role of synergy, the content of the
ginsenoside Rk3 and the ginsenoside Rh4 is preferably 50 parts by weight or more, more preferably
parts by weight or more, more preferably 70 parts by weight or more, more preferably 80 parts by
weight or more, more preferably 90 parts by weight or more, more preferably 95 parts by weight or
more, more preferably 99 parts by weight or more, more preferably 100 parts by weight (that is, the
pharmaceutical composition comprises only the above two ginsenosides), based on 100 parts by
weight of total ginsenosides comprised in the pharmaceutical composition. The purity of ginsenoside
Rk3 used in the pharmaceutical composition of the present application can be more than 98%, and
the purity of ginsenoside Rh4 used in the pharmaceutical composition of the present application can
be more than 98%.
The content of the total ginsenosides can be determined by the vanillin method, and the contents of
the ginsenosides Rk3 and Rh4 can be determined by the HPLC method.
The pharmaceutical composition of the present application may comprise other active ingredients for
preventing and/or treating alcoholic fatty liver disease, or may not comprise active ingredients for
preventing and/or treating alcoholic fatty liver disease (that is, using the ginsenoside Rk3 and the
ginsenoside Rh4 as the only active ingredient for preventing and/or treating alcoholic fatty liver
disease).
On the other hand, the inventors found that the pharmaceutical composition of the present application
has a significant preventive and/or therapeutic effect on alcoholic fatty liver disease. Therefore, the
present application also provides the use of the pharmaceutical composition of the present
application in the preparation of a medicament for preventing and/or treating alcoholic fatty liver
disease.
Pharmaceutically acceptable carriers, such as adjuvants, may be comprised in the pharmaceutical
compositions of the present application. There are no special restrictions on the adjuvants in the
pharmaceutical composition of the present application, for example, the adjuvants commonly used in
medicines or health products in the technical field can be used. Specifically, the adjuvants are starch,
dextrin, lactose, mannitol, sodium hypromellose, xanthan gum, protein sugar, etc.
The dosage form of the pharmaceutical composition of the present application is not particularly
limited, for example, it may be an oral dosage form or an injection dosage form. The oral dosage
form can be a liquid dosage form or a solid dosage form. The oral dosage forms can be, for example,
hard capsules, soft capsules, slow-release capsules, compressed tablets, sugar-coated tablets, powders,
granules, dropping pills, honeydew pills, syrups or oral liquids; the injection dosage forms can be, for
example, solution forms, suspension type, emulsion type or lyophilized powder. The administration
mode of the pharmaceutical composition for preventing and/or treating alcoholic fatty liver disease
can be, for example, oral administration, drip or injection.
When preparing a solid preparation for oral use, after adding an excipient and optionally a binder, a
disintegrating agent, a lubricant, a coloring agent, a flavoring agent, etc. to the main drug, tablets,
coated tablets, granules, fine granules, powders, capsules, etc can be prepared according to a
conventional method.
As an excipient, for example, lactose, corn starch, white sugar, glucose, sorbitol, crystalline cellulose,
silicon dioxide, etc. can be used; as a binder, for example, polyvinyl alcohol, ethyl cellulose, methyl
cellulose, Arabic gum, hydroxypropyl cellulose, hydroxypropyl methylcellulose, etc. can be used; as
a lubricant, for example, magnesium stearate, talc, silicon dioxide, etc. can be used; as a coloring
agent, coloring agents allowed to be added in drugs can be used; as a flavoring agent, cocoa powder,
menthol, aromatic acid, peppermint oil, borneol, cinnamon powder can be used. Of course, the
above-mentioned tablets and granules can also be coated with sugar coating, gelatin coating, and
other necessary outer coatings.
When preparing injections, pH adjusters, buffers, suspending aids, solubilizers, stabilizers, isotonic
agents, preservatives, etc. can be added to the main drug as required, and then intravenous,
subcutaneous, and intramuscular injections can be prepared according to conventional methods. At
this time, if necessary, a freeze-dried product can be prepared by a conventional method.
Examples of the suspending aid include methyl cellulose, Tween 80, hydroxyethyl cellulose, Arabic
gum, tragacanth gum powder, sodium carboxymethyl cellulose, polyoxyethylene sorbitan
monolaurate, and the like.
Examples of the solubilizer include polyoxyethylene hydrogenated castor oil, Tween 80, niacinamide,
polyoxyethylene sorbitan monolaurate, polyethylene glycol, castor oil fatty acid ethyl ester, and the
like.
Moreover, examples of the stabilizer include sodium sulfite, sodium metasulfite, etc., examples of the preservative include methylparaben, ethylparaben, sorbic acid, phenol, cresol, chlorocresol, etc.
Tumors can be treated by administering the pharmaceutical composition of the present application to
a subject. The subject may be a mammal, such as a human, a rat, a rabbit, a sheep, a pig, a cow, a cat,
a dog, a monkey, etc., preferably a human.
The pharmaceutical composition of the present application can be administered orally or
parenterally.
The dosage varies depending on the degree of symptoms, patient age, sex, body weight, difference in
sensitivity, administration method, administration period, administration interval, the nature of the
pharmaceutical preparation, the type of active ingredient, etc. There is no particular limitation, but
usually the dosage for an adult (with body weight 60Kg) is g-30000mg daily, preferably 10tg~
3000mg daily, more preferably 100g~2000mg daily, more preferably 1mg~1000mg daily, more
preferably 5~900mg daily, more preferably 10mg~500mg daily, more preferably 100mg~300mg
daily (based on the total amount of the ginsenoside Rk3 and the ginsenoside Rh4), and the
above-mentioned dosage can usually be administered 1 to 3 times a day.
Examples The present application will be further described below through specific examples. The following embodiments describe the present application by way of example only, but the protection scope of the present application is not limited thereto.
Example 1 Preparation of the ginsenoside Rk3/Rh4 composition tablets for preventing and treating alcoholic fatty liver disease 100 g of ginsenoside Rk3 with a purity of > 98%, 200 g of ginsenoside Rh4 with a purity of > 98%, and 500 g of medicinal adjuvants commonly used in tablet preparation were taken, mixed evenly, pressed into tablets, dried and packed to obtain 2000 tablets of the present application, each tablet comprising 150mg of active ingredients.
Example 2 Preparation of the ginsenoside Rk3/Rh4 composition capsules for preventing and treating alcoholic fatty liver disease
100 g of ginsenoside Rk3 with a purity of >98%, 100 g of ginsenoside Rh4 with a purity of >98%,
and 600 g of commonly used medicinal adjuvants were taken and mixed evenly, and then put into
capsules currently on the market to obtain 2000 capsules of the present application, each capsule
comprising 100mg of active ingredients.
Example 3 Preparation of the ginsenoside Rk3/Rh4 composition oral liquid for preventing and
treating alcoholic fatty liver disease
g of ginsenoside Rk3 with a purity of > 98% and 80 g of ginsenoside Rh4 with a purity of > 98%
were taken, mixed evenly, and then an appropriate amount of flavorings, such as crystalline fructose,
stachyose, citric acid, and pectin, and diluent water were added by conventional methods. The
resulting mixture was heated to dissolve, homogenized, filled, and sterilized to prepare 1,000 bottles
of the oral liquid of the present application, each bottle of the oral liquid comprising 120 mg of active
ingredients.
Example 4 Efficacy verification test of ginsenoside Rk3/Rh4 composition in preventing and
treating alcoholic fatty liver disease
100 adult male mice (18-22 grams) were randomly divided into normal control group; model group;
drug administration group: the composition group 1 of the present application in Example 2 (Rk3
mg/kg, Rh4 15mg); the composition group 2 of the present application (Rk3 7.5mg/kg, Rh4
22.5mg), the composition group 3 of the present application (Rk3 22.5mg/kg, Rh4 7.5mg),
ginsenoside Rk3 (30mg/kg) group, ginsenoside Rh4 (30mg/kg) kg) group, polyene
phosphatidylcholine (30mg/kg) positive drug control group, ginsenoside Rgl (30mg/kg) group,
ginsenoside CK/Rhl (CK 15mg/kg, Rhl 15mg/kg) group, a total of 10 groups, except for the normal
control group, the mice in the other groups were given 50% ethanol by gavage once a day, and the
ethanol dose was 12 mL/kg BW for 4 consecutive weeks. Mice in the normal control group were
given an equal volume of purified water by gavage. The test drug was prepared into a suspension
with purified water, and it was administered by gavage once a day. The normal control group and the
model control group were given the same amount of purified water by gavage, and the gavage
volume was 10ml/kg/d. During the experiment, the mice had free access to food and water, and were
weighed twice a week, and the tested sample amount was adjusted according to body weight. The model control group and each sample group were given 50% ethanol by gavage at the end of the experiment, and the mice were given 12ml/kg BW by gavage. The negative control group was given purified water and fasted for 16 hours. After the animals were anesthetized by intraperitoneal injection of 60 mg/kg BW pentobarbital sodium solution, blood was collected from the abdominal aorta, and liver tissue was collected to detect various indicators: serum ALT, AST and LDL, serum
TC, serum TG and LDL content, liver tissue TG content and histopathological examination. The test
results were expressed as mean plus or minus standard deviation( xs), and the statistical processing
method adopts one-way analysis of variance; the two-drug interaction index CI=AB/(AxB) was
calculated, wherein T was the measured index value of the drug administration group, and C was the
measured index value of the model group, AB was the T/C value of the two-drug combination group,
and A and B were the T/C values of the single-drug action group. When CI < 1, the two drugs had
synergistic effect, and when CI < 0.7, the synergistic effect was very significant (refer to David
H.Kem, Carol R.Morgan, and Susanne U.Hildebrand-Zanki. In vitro and in vivo interaction between
cisplatin and topotecan in ovarian carcinoma systems[J]. Cancer Research, 1988, 48.; and Li Xingqi.
High potency of lidamycin on glioma suppression and its synergism with temozolomide:
involvement of enhanced antiangiogenesis and apoptosis induction[D]. China Union Medical
College, 2009.).
The experimental results were as follows:
Table 1. The influence of the composition of the present application on mouse body weight, liver
weight, liver coefficient (xs)
group dose (mg/kg) weight (g) liver weight (g) liver coefficient(%)
blank control group 36.35±1.20 1.42±0.21AA 3.91±0.20AA
model group 35.62±2.18 1.81±0.37** 5.08±0.33**
polyene 30 36.02±2.13 1.67±0.36A 4.63±0.36*A phosphatidylcholine
Ginsenoside Rk3 30 36.21±1.43 1.75±0.31*A 4.82±0.55*A
Ginsenoside Rh4 30 35.83±1.35 1.74±0.28*A 4.85±0.44*A
Composition 1 30 36.12±1.09 1.46±0.23AA 4.04±0.36AA
Composition 2 30 36.08±1.23 1.71±0.21*A 4.76±0.52*A
Composition 3 30 35.96±1.31 1.77±0.26*A 4.71±0.34*A
Ginsenoside Rgl 30 36.18±1.25 1.72±0.19*A 4.75±0.36A
Ginsenoside CK/Rh1 30 35.92±1.92 1.75±0.23A 4.88±0.29A
CI index of Composition - - - - 0.88 0.88 1
CI index of Composition - - - - 1.02 1.03 2
CI index of Composition - - - - 1.05 1.02 3
Note: **P<0.01, *P<0.05 vs. blank control; AAP<0.01,AP<0.05 vs. model group
There was no significant difference in body weight among the groups (P>0.05); compared with the
blank control group, the liver weight and liver coefficient of the model group were significantly
increased (P<0.01). Compared with the model group, the liver coefficient of each administration
group decreased significantly (P < 0.05), among which the liver coefficient of the Rk3/Rh4 group
decreased extremely significantly (P<0.01). The results of this experiment showed that each drug
administration group had obvious inhibitory effect on the hepatomegaly of mice caused by alcohol,
and under the same dose, the composition 1 of the present application was more effective than each
single component, and had a synergistic effect (CI value < 1); compared with polyene
phosphatidylcholine, ginsenoside Rgl and ginsenoside CK/Rhl, the effect of reducing the liver
coefficient of the composition 1 of the present application was more significant.
Table 2. The influence of the composition of the present application on serum ALT, and AST
activity of mice with alcoholic fatty liver disease( xs)
dose ALT activity AST activity group (mg/kg) (U/L) (U/L)
blank control group 32.43±3.07AA 52.17±8.29AA model group 61.82±8.62** 126.93±19.45** polyene 30 43.93±4.16AA 82.12±11.33AA phosphatidylcholine
Ginsenoside Rk3 30 53.29±7.3 1A 91.23±11.85A*
Ginsenoside Rh4 30 51.87±6.92A 93.58±19.96A*
Composition 1 30 40.28±3.59AA 62.98±9.73AA
Composition 2 30 49.87±4.96A 83.59±19.01A*
Composition 3 30 50.06±6.38A 77.42±11.98A*
Ginsenoside Rg1 30 54.84±6.11A 94.73±9.87A
Ginsenoside CK/Rhl 30 49.72±4.93AA 93.09±11.36A
CI index of Composition 1 0.89 0.94
CI index of Composition 2 1.24
CI index of Composition 3 1.15
Note: **P<0.01, *P<0.05 vs. blank control; AAP<0.01,AP<0.05 vs. model group
ALT and AST are the landmark indicators of liver damage. The content of ALT and AST in normal
serum is very low. Only after liver damage, ALT and AST in hepatocytes are released into the blood
by cell rupture, and the reactive oxygen species generated during the metabolism of ethanol by the
liver can directly cause oxidative damage to liver cells. As could be seen from Table 2, the animal
serum ALT and AST activities of each administration group and the model group had significant
differences (P<0.05), and the animal serum ALT and AST activities of the composition Rk3/Rh4
group of the present application were extremely significant different from those of the model group
(P<0.01), significant lower than those of the model group, but not significantly different from those
of the blank control group (P>0.05). The test results showed that the composition of the present
application could significantly reduce the activities of abnormally elevated ALT and AST in the
serum of mice with alcoholic fatty liver disease, under the same dose, the composition 1 of the
present application had a stronger effect on reducing the activity of ALT and AST than each single
component, as well as polyene phosphatidylcholine, ginsenoside Rgl, and ginsenoside CK/Rhl.
After calculation, the combination index CI of composition 1 of the ginsenoside Rk3 and the ginsenoside Rh4 was less than 1, while the combination index CI of composition 2 and composition
3 was greater than 1, indicating that the composition 1 of the present application had a good
synergistic effect and had a good protective effect on liver cell damage caused by drinking.
Table 3. The influence of composition of the present application on serum TC, TG, LDL content and
liver tissue TG content of mice with alcoholic fatty liver disease (xs) Serum TC Serum TG Serum LDL liver tissue TG group (mmol/L) (mmol/L) (mmol/L) (mmol/L)
blank control group 8.84±0.28AA 1.97±0.12AA 7.23±0.56AA 1.82±0.12AA
model group 12.59±0.27** 3.02±0.17** 13.28±0.67** 2.98±0.21**
polyene 10.29±0.21A 2.47±0.20AA 9.72±0.35A 2.61±0.18A phosphatidylcholine
Ginsenoside Rk3 11.39±0.25 2.83±0.18A 10.98±0.62*A 2.72±0.17*
Ginsenoside Rh4 11.25±0.17 2.76±0.21A 10.82±0.59*A 2.65±0.16*A
Composition 1 9.08±0.15AA 2.09±0.15AA 8.12±0.41AA 1.97±0.12AA
Composition 2 10.95±0.19 2.66±0.24A 9.87±0.53*A 2.61±0.13*A
Composition 3 11.03±0.18 2.59±0.19A 9.38±0.51*A 2.47±0.12*A
Ginsenoside Rgl 10.21±0.23A 2.44±0.19AA 9.62±0.16A 2.69±0.21A
Ginsenoside CK/Rh1 9.91±0.31A 2.69±0.21A 9.81±0.29A 2.33±0.18AA
CI index of Composition 1 0.89 0.81 0.91 0.81
CI index of Composition 2 1.08 1.03 1.10 1.08
CI index of Composition 3 1.08 1.00 1.05 1.02
Note: **P<0.01, *P<0.05 vs. blank control; AAPS0.01,AP<0.05 vs. model group
Long-term ingestion of alcohol will abnormally increase the serum TC and TG content of mice, and
the liver accumulates cholesterol and triglycerides. As shown in Table 3, the serum TC, TG, LDL
contents and liver tissue TG content of animals in each drug administration group were significantly
different from those in the model group (P<0.05), wherein the serum TC, TG, LDL contents and
liver tissue TG content of animals in the composition group of the present application were extremely significantly different from those of the model group (P<0.01). After calculation, the combination index (CI) of composition 1 was less than 1, while the combination index (CI) of composition 2 and composition 3 was greater than 1, indicating that the composition 1 of the present application had a good synergistic effect, while outside the ratio range of the composition of the present application, the combination of the ginsenoside Rk3 and the ginsenoside Rh4 had no synergistic effect, and the damage of alcohol to liver cells was achieved through lipid peroxidation mediated by free radicals. Composition 1 of the present application could effectively reduce the contents of TC and TG in the serum of mice, improved LDL levels, inhibited the synthesis of endogenous TC and TG, and improved lipid distribution in the body and deposition in internal organs.
From the pathological sections of the mouse livers, it could be clearly seen that the livers of the mice
in the control group were normal in size, red, soft and sharp; the livers of the mice in the model
group were significantly enlarged, gray-white, grainy, and greasy on the cut surface; the liver
morphology of each treatment group was improved compared with that of the model group, but the
liver morphology of the Rk3/Rh4 composition group was closer to that of the control group
compared to the other treatment groups. The results of HE staining of the liver (Figures 3, 1-10)
showed that the hepatocytes in the blank control group were normal in size and shape, neatly
arranged, with large and round nuclei in the center, and the cytoplasm was uniformly colored. There
was no lipid droplet deposition in the hepatocytes, the morphology and structure of the hepatocytes
were normal, and there was no steatosis and inflammatory changes; the hepatocytes in the model
group were swollen and the boundaries were unclear. A large number of vacuolar-like lipid droplets
could be seen in the cells with different sizes. Steatotic hepatocytes were distributed in foci. Liver
tissue was almost replaced by fat vacuoles. Ballooning change of hepatocyte took place, and there
was inflammatory cell infiltration in the portal area; in ginsenoside Rk3 group, ginsenoside Rh4
group, composition 2 group, composition 3 group, positive drug blank control group, ginsenoside
Rgl group and ginsenoside CK/Rhl group, the fat vacuoles in the liver tissue showed scattered
distribution, lipid accumulation (oil droplets) became smaller, and no obvious inflammatory
inflammatory cells were found. The mice fed with the Rk3/Rh4 composition 1 of the present
application had almost no lipid droplet deposition in the liver tissue, the morphological structure of
the hepatocytes were close to normal, andthere was no steatosis. The above effects were better at medium doses. It showed that the disclosed Rk3/Rh4 composition of the present application could better improve the degree of alcohol-induced liver steatosis in mice.
Example 5 Exploration on the mechanism of ginsenoside Rk3/Rh4 composition in preventing
and treating alcoholic fatty liver disease
In Example 4, the normal control group; model group; Rk3 group, Rh4 group, and Rk3/Rh4 group
were measured for liver superoxide dismutase (SOD), liver reduced glutathione (GSH), and liver
propylene glycol aldehyde (MDA) levels; and the expression levels of related genes in hepatocytes
were determination by molecular chip screening. The test results were expressed as mean plus or
minus standard deviation( xs), and the statistical processing method adopted one-way analysis of
variance;
The experimental results were as follows:
Table 4. The influence of the composition of the present application on liver SOD, GSH and MDA of
mice with alcoholic fatty liver disease( xs) SOD GSH MDA group (U/mg) (mmol/mg) (jtmol/mg)
blank control group 322.4±22.01AA 62.11±0.62AA 1.82±0.12AA
model group 154.34±17.3** 27.73±0.55** 2.98±0.21**
Ginsenoside Rk3 234.13±25.97AA 47.83±0.18AA 2.12±0.17AA
Ginsenoside Rh4 208.23±23.22AA 42.76±0.21*AA 2.25±0.16*AA
Ginsenoside Rk3/Rh4 189.02±20.25AA 52.02±0.72AA 2.03±0.18AA
Note: **P<0.01, *P<0.05vs blank control; AAP<0.01, AP<0.05vs model group
The liver cells of alcoholic fatty liver were very susceptible to oxidative stress, and the antioxidant
capacity of the mice in the model group was weakened, which was manifested as increased levels of
MDA, a harmful product of alcohol metabolism, decreased SOD activity in liver tissue, and
decreased GSH content of reduced glutathione. Compared with the model group, the activities of
SOD and GSH content in liver tissues of the Rk3 group, Rh4 group and Rk3/Rh4 group were significantly increased (P<0.01), and the MDA content was extremely significantly decreased
(P<0.01), indicating that Rk3 and Rh4 had antioxidant activities, which could effectively improve
liver oxidative damage caused by drinking.
Table 5. Statistical table of the effect of the composition of the present application on the relative
expression levels of genes related to important metabolic pathways in mice with alcoholic fatty liver
disease
group Fasn LpL Scdl PPARu
model group vs. blank t4.17 13.18 t6.43 12.96 control group
Rk3 group vs. model 12.92 t 1.13 15.92 t1.84 group
Rh4 group vs. model 12.18 t2.68 13.57 t 1.97 group
Rk3/Rh4 group vs. 13.38 t2.97 17.18 t2.63 model group
Note: In the table, I indicates the down-regulated fold of the corresponding gene in hepatocyte mRNA expression,
and t indicates the up-regulated fold of the corresponding gene in the hepatocyte mRNA expression.
Fatty acid synthase (Fasn), which plays a crucial role in fatty acid synthesis and is responsible for all
catalytic steps in the de novo synthesis of a long-chain palmitate (palmitate) from acetyl-CoA and
malonyl-CoA. The gene expression directly affects the amount of fatty acid synthase and plays an
important role in controlling animal fat deposition. Lipoprotein lipase (LpL) is one of the key
enzymes in fat metabolism. It acts on lipoproteins and mainly decomposes triacylglycerols in
lipoproteins. It is the rate-limiting enzyme for removing triacylglycerols in plasma and promotes the
transfer of cholesterol from lipoproteins, phospholipids and apolipoproteins; LpL also has the ability
to increase the binding of chylomicrons to LP receptors, promoting chylomicron uptake. Sterol
coenzyme A desaturase 1 (Scdl) is the rate-limiting enzyme in the biosynthesis of monounsaturated
fatty acids, plays a central regulatory role in fatty acid metabolism, and is also one of the target genes for leptin; leptin is closely related to diabetes, obesity, and fatty liver, and is one of the hot areas of metabolic diseases in recent years.
Under the experimental conditions, the model group can increase the synthesis of Fasn and decrease
the synthesis of LpL, which is the main culprit leading to excessive deposition of fat in liver cells and
the primary factor causing fatty liver. Rh4 and the Rk3/Rh4 composition can significantly increase
the synthesis of LpL, reduce the synthesis of fatty acid in the liver tissue, strengthen the
decomposition of fat, and have positive significance for relieving liver steatosis. In addition, the
up-regulation of Scdl gene expression in the model group may be one of the most important
molecular mechanisms of alcoholic steatosis, but each drug administration group can significantly
down-regulate the expression of Scdl gene.
It can be known from the above pharmacodynamic experiments that the composition of the present
application has the effect of preventing and treating alcoholic fatty liver disease, and long-term
administration has no side effects.
Finally, it should be noted that the above description is only a preferred embodiment of the present
application, and under the enlightenment of the application, those ordinary skilled in the art can make
a variety of similar expressions without departing from the purpose and claims of the application,
and such transformations fall within the scope of protection of the application..
Industrial Applicability
The present application provides a pharmaceutical composition comprising ginsenosides Rk3 and
Rh4, which is synergistic and has the effect of preventing and/or treating alcoholic fatty liver disease.

Claims (10)

1. A pharmaceutical composition for the preventing and/or treating alcoholic fatty liver disease,
comprising prophylactically and/or therapeutically effective amount of ginsenoside Rk3 and
ginsenoside Rh4 as active ingredients, and a pharmaceutically acceptable carrier,
wherein in the pharmaceutical composition, the weight ratio of the ginsenoside Rk3 to the
ginsenoside Rh4 is 1:0.5-2, preferably 1:0.9~1.1.
2. The pharmaceutical composition according to claim 1, wherein in the pharmaceutical composition,
the weight ratio of the ginsenoside Rk3 to the ginsenoside Rh4 is 1:1.
3. The pharmaceutical composition according to claim 1, wherein based on 100 parts by weight of
the total ginsenosides comprised in the pharmaceutical composition, the total amount of the
ginsenoside Rk3 and the ginsenoside Rh4 comprised in the pharmaceutical composition is 10 parts
by weight or more.
4. The pharmaceutical composition according to claim 1, wherein based on 100 parts by weight of
total ginsenosides comprised in the pharmaceutical composition, the total amount of the ginsenoside
Rk3 and the ginsenoside Rh4 comprised in the pharmaceutical composition is 100 parts by weight.
5. The pharmaceutical composition according to claim 1, wherein the pharmaceutical composition
does not comprise other active ingredients other than the ginsenoside Rk3 and the ginsenoside Rh4
for preventing and/or treating alcoholic fatty liver disease.
6. Use of ginsenoside Rk3 and ginsenoside Rh4 in the preparation of a pharmaceutical composition
for preventing and/or treating alcoholic fatty liver disease, wherein in the pharmaceutical
composition, the weight ratio of the ginsenoside Rk3 to the ginsenoside Rh4 is 1:0.5-2, preferably
1:0.9~1.1.
7. The use according to claim 6, wherein in the pharmaceutical composition, the weight ratio of the
ginsenoside Rk3 to the ginsenoside Rh4 is 1:1.
8. The use according to claim 6, wherein based on 100 parts by weight of total ginsenosides
comprised in the pharmaceutical composition, the total amount of the ginsenoside Rk3 and the
ginsenoside Rh4 comprised in the pharmaceutical composition is 10 parts by weight or more.
9. The use according to claim 6, wherein based on 100 parts by weight of total ginsenosides
comprised in the pharmaceutical composition, the total amount of the ginsenoside Rk3 and the
ginsenoside Rh4 comprised in the pharmaceutical composition is 100 parts by weight.
10. The use according to claim 6, wherein the pharmaceutical composition does not comprise other
active ingredients other than the ginsenoside Rk3 and the ginsenoside Rh4 for preventing and/or
treating alcoholic fatty liver disease.
TFE00387AU 1/2
OH H
H
H
OH H O O OH
OH O OH HO O
OH
OH
Figure 1 5
OH H
H
H
OH H O O OH
OH O OH HO O
OH
OH Figure 2 10
15
1005 96.7
AU2021257523A 2020-04-17 2021-04-19 Ginsenoside composition having alcoholic fatty liver-preventing and -treating function Active AU2021257523B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202010305247.X 2020-04-17
CN202010305247.XA CN111388490B (en) 2020-04-17 2020-04-17 Ginsenoside composition with function of preventing and treating alcoholic fatty liver
PCT/CN2021/088142 WO2021209070A1 (en) 2020-04-17 2021-04-19 Ginsenoside composition having alcoholic fatty liver-preventing and -treating function

Publications (2)

Publication Number Publication Date
AU2021257523A1 true AU2021257523A1 (en) 2022-12-22
AU2021257523B2 AU2021257523B2 (en) 2024-03-21

Family

ID=71417622

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2021257523A Active AU2021257523B2 (en) 2020-04-17 2021-04-19 Ginsenoside composition having alcoholic fatty liver-preventing and -treating function

Country Status (3)

Country Link
CN (1) CN111388490B (en)
AU (1) AU2021257523B2 (en)
WO (1) WO2021209070A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111388490B (en) * 2020-04-17 2021-07-02 陕西巨子生物技术有限公司 Ginsenoside composition with function of preventing and treating alcoholic fatty liver
CN116023423B (en) * 2023-03-29 2023-06-27 中国农业科学院特产研究所 Ginsenoside Rk3 and preparation and application thereof in preparation of folliculitis medicines

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103054906B (en) * 2012-11-30 2014-09-10 周斌 Propolis ethanol extract for alleviating hangover and preparation method thereof, and application of propolis ethanol extract in producing buccal tablets
KR101577111B1 (en) * 2013-08-13 2015-12-11 주식회사 한국인삼공사 Pharmaceutical compositions for anti-inflammation or anti-oxidation containing ginsenoside rh4-enriched extraction
CN106109483B (en) * 2016-07-29 2020-02-07 陕西巨子生物技术有限公司 Diol/triol rare ginsenoside composition with anti-tumor activity
CN108685931A (en) * 2018-07-02 2018-10-23 南京类人生物材料有限公司 Panaxsaponin composition with hypoglycemic activity and its application
CN109394703A (en) * 2018-07-05 2019-03-01 西安巨子生物基因技术股份有限公司 3beta,6alpha,12beta-Trihydroxydammar-20's intravenous formulations and preparation method
CN111388490B (en) * 2020-04-17 2021-07-02 陕西巨子生物技术有限公司 Ginsenoside composition with function of preventing and treating alcoholic fatty liver

Also Published As

Publication number Publication date
CN111388490B (en) 2021-07-02
AU2021257523B2 (en) 2024-03-21
WO2021209070A1 (en) 2021-10-21
CN111388490A (en) 2020-07-10

Similar Documents

Publication Publication Date Title
AU2021257523B2 (en) Ginsenoside composition having alcoholic fatty liver-preventing and -treating function
US10702519B2 (en) Use of methoxatin, derivative and/or salt thereof in Sjogren&#39;s syndrome and pharmaceutical composition
CN107847513B (en) Composition for preventing and treating obesity or impaired glucose tolerance comprising nicotinamide adenine dinucleotide
AU2021246790B2 (en) Anti-tumor composition containing rare ginsenosides Rk2, CK, and PPT
Salama et al. Experimental and therapeutic trials of amygdalin
US20110039796A1 (en) Natural Composition for Anti-Angiogenesis and Anti-Obesity
WO2007065314A1 (en) Compound preparation for treating virus hepatopathy
CN108815218B (en) Pharmaceutical composition and use thereof
WO2014171583A1 (en) Composition for preventing or treating stroke or degenerative brain disease
US10039724B2 (en) 7-hydroxy cannabidiol (7-OH-CBD) for use in the treatment of non-alcoholic fatty liver disease (NAFLD)
US9072762B2 (en) Natural composition to decrease effects of a high fat diet
AU2020358184B2 (en) Preparation of drug for treating Alzheimer&#39;s disease
WO2019083264A1 (en) Composition for preventing, alleviating or treating liver diseases, containing fermented product of aged sprout ginseng extract as active ingredient
EP2286819A1 (en) Pharmceuttical composition for treating cardio-cerebro vascular diseases and preparative method and kit thereof
CN111265514B (en) Use of ketones for the treatment of fatty liver
US20150328271A1 (en) Composition for Preventing or Treating Oxidative Brain Damage and Brain Dysfunction, and Production Method for Same
CN111467385B (en) Use of composition in preventing or treating neurodegenerative disease
US11083767B2 (en) Pharmaceutical composition comprising an extract of platycodon grandiflorum and method for preventing or treating of obesity using the same
CN106822152B (en) Pharmaceutical composition and application thereof
WO2017113775A1 (en) Application of triglyceride compound for use in preparing medicine for treating neurodegenerative diseases
TWI621440B (en) Use of Ceylon Seven-fingered Fern, Diosgenin and Flavonoids for the Treatment or Prevention of Metabolic Diseases
CN112691102A (en) Application of baicalein in preventing and treating Parkinson&#39;s disease/Parkinson&#39;s syndrome depression symptoms
KR101572311B1 (en) A composition for preventing or treating obesity comprising 2-amino-2-norbornanecarboxylic acid
CA2973962A1 (en) Pharmaceutical composition comprising platycodon grandiflorum extract for preventing or treating obesity
CN112656794B (en) Application of pyrroloquinoline quinone or salt thereof in preparation of medicines for preventing and treating prostatic hyperplasia and pharmaceutical composition