AU2021107213A4 - Use of sphondin as an effective component in preparing medicine for treating hepatitis b - Google Patents

Use of sphondin as an effective component in preparing medicine for treating hepatitis b Download PDF

Info

Publication number
AU2021107213A4
AU2021107213A4 AU2021107213A AU2021107213A AU2021107213A4 AU 2021107213 A4 AU2021107213 A4 AU 2021107213A4 AU 2021107213 A AU2021107213 A AU 2021107213A AU 2021107213 A AU2021107213 A AU 2021107213A AU 2021107213 A4 AU2021107213 A4 AU 2021107213A4
Authority
AU
Australia
Prior art keywords
hbv
treatment
hepatitis
sphondin
drugs
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.)
Active
Application number
AU2021107213A
Inventor
Juan Chen
Shengtao Cheng
Ailong HUANG
Fang REN
Jihua Ren
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.)
Chongqing Medical University
Original Assignee
Chongqing Medical University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chongqing Medical University filed Critical Chongqing Medical University
Application granted granted Critical
Publication of AU2021107213A4 publication Critical patent/AU2021107213A4/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
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • A61K31/37Coumarins, e.g. psoralen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Communicable Diseases (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention belongs to the technical field of biomedicine, and specifically discloses a use of sphondin as an effective component in preparation of drugs for treatment of hepatitis B. It is found for the first time in the present invention that sphondin (Sph for short) can inhibit secretion of HBsAg from HBV, the transcription level of HBV RNAs, the replication level of HBV core DNA, the transcription activity of cccDNA and levels of HBV RNAs, HBV DNA, an HBV S protein and an HBV X protein in liver tissues, has an obvious function in treatment of hepatitis B, can be used to prepare new anti-HBV drugs which can reduce the HBsAg level and inhibit transcription of cccDNA, and has a broad application prospect in treatment of hepatitis B. SEQUENCE LISTING <110> Chongqing Medical University <120> Use of sphondin as an effective component in preparation of drugs for treatment of hepatitis B <130> 210429 <160>6 <170> PatentIn version 3.5 <210> 1 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> total HBV RNAs forward <400> 1 accgaccttg aggcatactt 20 1

Description

SEQUENCE LISTING
<110> Chongqing Medical University
<120> Use of sphondin as an effective component in preparation of drugs for treatment of hepatitis B
<130> 210429
<160>6
<170> PatentIn version 3.5
<210> 1
<211> 20
<212> DNA
<213> Artificial Sequence
<220>
<223> total HBV RNAs forward
<400> 1
accgaccttg aggcatactt 20
USE OF SPHONDIN AS AN EFFECTIVE COMPONENT IN PREPARING MEDICINE FOR TREATING HEPATITIS B
Background of the Present Invention
Field of Invention
The present invention relates to the technical field of biomedicine, and in particular, to a use
of sphondin as an effective component in preparation of drugs for treatment of hepatitis B.
Description of Related Arts
Hepatitis B virus (HBV), a prototype member of the hepadnaviridae family, has chronically
infected more than 250 million people worldwide. HBV infection is closely related to the occurrence
and development of acute and chronic hepatitis, cirrhosis and liver cancer.
According to statistics, nearly two billion people worldwide have been infected with HBV,
of which about 350 million people are chronically infected. Chronic HBV infection is a global public
health burden. Annually, about 1 million deaths from severe disease are caused by HBV infection,
such as hepatic failure, liver cirrhosis and primary hepatocellular carcinoma.
Mother-to-child transmission can be difficult to prevent, and 10% of adults and 5% of minors
cannot get vaccine immunity and thus become the population with the highest potential infection risk.
Hepatitis B virus infection and resulting diseases not only seriously endanger people's health, but also
become a serious social public health problem. Therefore, treatment of chronic HBV infection and
prevention of development of hepatitis B-related diseases are important tasks related to global public
health.
At present, there are only two types of drugs approved by US Food and Drug Administration
(FDA) for treatment of chronic hepatitis B (CHB): nucleoside analogs (NAs) targeting a pol protein
and interferons (IFNs) targeting viral transcription, which exert an antiviral effect through immune
regulation and interference with HBV replication respectively. However, IFN-a has many application
restrictions due to its low response rate, many adverse reactions, low tolerance, high price, limited treatment targets and the like. In recent years, NAs drugs have been developed rapidly and become the first choice of antiviral therapeutic drugs and have been applied widely in the clinic practice.
Although NAs drugs have a strong ability to inhibit viral DNA, HBsAg and an HBV transcription
template cccDNA (covalently closed circular DNA) in patients cannot be completely removed by NAs.
Futhermore, NAs drugs need to be taken for a long time, drug resistance may be caused due to
mutation of an HBV polymerase, the viral load rebounds after discontinuance of most NAs drugs, and
applications of such drugs are greatly limited. Therefore, it is necessary to develop anti-HBV drugs
with new action mechanisms and action targets.
HBV has a complicated life cycle. When being in contact with liver cell membranes, virus
particles enter cells under mediation of a receptor (NTCP), and relaxed circular DNA (rc DNA)
wrapped by a viral core protein is released after internalization. After rcDNA undergoes a series of
excision repair reactions under the action of a host DNA polymerase and a topoisomerase, cccDNA
with a superhelical structure is finally formed. As a key molecule to establish persistent infection,
cccDNA is subjected to transcription under the action of a host RNA polymerase II to produce 4 kinds
of RNA transcripts. Precore RNA is used to encode an HBV core protein and form HBeAg; 2.4/2.1
kb RNA is used to form an envelope protein (L, M, S); 0.7-kb RNA is used to finally form HBx. A
largest pregenomic RNA (pgRNA) forms rcDNA under the action of an HBV polymerase and
continues to replenish a cccDNA pool. HBV is difficult to cure and is closely related to long-term
stable existence of cccDNA. In addition, the envelope protein can be integrated into host genomic
DNA. Therefore, a large amount of HBsAg in the serum of a patient is difficult to turn negative. Based
on this, the present invention intends to find new anti-HBV drugs which can effectively reduce HBsAg
and stop transcription of cccDNA.
Summary of the Present Invention
In view of the shortcomings of the prior art, the present invention provideS a use of sphondin
as an effective component in preparation of drugs for treatment of hepatitis B, so as to provide a new
anti-HBV drug and a treatment method for effectively reducing HBsAg and stopping transcription of
cccDNA.
Accordingly, in a first aspect, the present invention provides a use of sphondin as an effective component in preparation of drugs for treatment of hepatitis B.
Further, the drugs for treatment of hepatitis B have at least one of the following functions:
inhibiting secretion of HBsAg from HBV, inhibiting the transcription level of HBV RNAs,
inhibiting the replication level of HBV core DNA, inhibiting the transcription activity of cccDNA,
and inhibiting levels of HBV RNAs, HBV DNA, an HBV S protein and an HBV X protein in liver
tissues.
Further, the drugs for treatment of hepatitis B necessarily include sphondin, and sphondin is
used as an effective component to achieve the functions above.
Further, in the drugs for treatment of hepatitis B, the effective component to achieve the
functions above may only be sphondin and may also contain other molecules which may achieve
similar functions.
Further, sphondin is used as one of the effective components or the only effective component
of the drugs for treatment of hepatitis B.
Further, the drugs for treatment of hepatitis B may be single-component substances or multi
component substances.
Further, the drugs for treatment of hepatitis B are not particularly limited in form and may be
in the form of solid, liquid, gel, semi-liquid, aerosol and other substance forms.
Further, the drugs for treatment of hepatitis B are mainly used to treat mammals, such as
rodents and primates.
In a second aspect, the present invention provides a medicinal preparation for treatment of
hepatitis B, and the medicinal preparation includes a safe and effective dose of sphondin.
Further, the medicinal preparation for treatment of hepatitis B also includes a
pharmaceutically acceptable carrier and/or an adjuvant.
Further, the medicinal preparation for treatment of hepatitis B necessarily includes sphondin,
and sphondin is used as an effective component to achieve the functions above.
Further, in the medicinal preparation for treatment of hepatitis B, the effective component to
achieve the functions above may only be sphondin and may also contain other molecules which may achieve similar functions.
Further, sphondin is used as one of the effective components or the only effective component
of the medicinal preparation for treatment of hepatitis B.
Further, the medicinal preparation for treatment of hepatitis B may be a single-component
substance or a multi-component substance.
Further, the medicinal preparation for treatment of hepatitis B is not particularly limited in
form and may be in the form of solid, liquid, gel, semi-liquid, aerosol and other substance forms.
Further, the medicinal preparation for treatment of hepatitis B is mainly used to treat
mammals, such as rodents and primates.
In a third aspect, the present invention provides a method for treatment of hepatitis B, and
the method includes administering sphondin to a subject.
Further, the subject may be mammals or hepatitis B cells of mammals. The mammals are
preferably rodents, artiodactyls, perissodactyls, lagomorphs, primates and the like. The primates are
preferably monkeys, apes or humans. The hepatitis B cells may be in-vitro hepatitis B cells.
Further, the subject may be a patient suffering from hepatitis B or a hepatitis B individual
expecting treatment. Or, the subject is hepatitis B cells of a patient suffering from hepatitis B or a
hepatitis B individual expecting treatment.
Further, sphondin may be administered to the subject before, during and after treatment of
hepatitis B.
In a fourth aspect, the present invention provides a drug combination for combined treatment
of hepatitis B, and the drug combination includes a safe and effective dose of sphondin, at least one
other drug for treatment of hepatitis B and the balance of a pharmaceutically acceptable carrier and/or
an adjuvant.
Further, the drug combination for combined treatment of hepatitis B may be any one of the
following forms:
(1) Sphondin and other drugs for treatment of hepatitis B are separately prepared into
independent preparations, where dosage forms of the preparations may be the same or different, and administration routes may also be the same or different.
When other drugs for treatment of hepatitis B are antibodies, a parenteral administration route is generally used. When other drugs for treatment of hepatitis B are chemical drugs, there are many administration forms including gastrointestinal administration or non-gastrointestinal administration. It is generally recommended to administer the chemical drugs through already known administration routes.
(2) Sphondin and other drugs for treatment of hepatitis B are prepared into a compound preparation, where sphondin and other drugs for treatment of hepatitis B are administered at the same time in the same administration route, and the combination of both may be prepared into a compound preparation.
In a fifth aspect, the present invention provides a method for treatment of hepatitis B, and the method includes administering an effective dose of sphondin and an effective dose of other drugs for treatment of hepatitis B to a subject, and/or implementing other means for treatment of hepatitis B to the subject.
Further, an effective dose of sphondin and an effective dose of at least one of other drugs for treatment of hepatitis B may be administered simultaneously or sequentially.
Since sphondin is a drug for treatment of hepatitis B discovered for the first time by the present invention, at least an improved treatment effect can be achieved when sphondin is combined with other drugs for treatment of hepatitis B, and a treatment effect on hepatitis B is further improved.
Further, other drugs for treatment of hepatitis B include but are not limited to: antibody drugs, chemical drugs or targeted drugs and the like.
Further, sphondin may be administered in a gastrointestinal administration route or a parenteral administration route, and other drugs for treatment of hepatitis B may be administered in a gastrointestinal administration route or a parenteral administration route. The antibody drugs are generally administered in a parenteral administration route.
In a sixth aspect, the present invention provides uses of sphondin in preparation of substances having any one or more of the following effects: a use for preparing a substance for inhibiting secretion of HBsAg from HBV, a use for preparing a substance for inhibiting the transcription level of HBV RNAs, a use for preparing a substance for inhibiting the replication level of HBV core DNA, a use for preparing a substance for inhibiting the transcription activity of cccDNA, and a use for preparing a substance for inhibiting levels of HBV RNAs, HBV DNA, an HBV S protein and an HBV X protein in liver tissues.
As described above, in the present invention, the use of sphondin as an effective component
in preparation of drugs for treatment of hepatitis B has the following beneficial effects:
it is found for the first time by the present invention that sphondin (Sph for short) can inhibit
secretion of HBsAg from HBV, the transcription level of HBV RNAs, the replication level of HBV
core DNA, the transcription activity of cccDNA and levels of HBV RNAs, HBV DNA, an HBV S
protein and an HBV X protein in liver tissues, has an obvious function in treatment of hepatitis B, can
be used to prepare new anti-HBV drugs which can reduce the HBsAg level and inhibit transcription
of cccDNA, and has a broad application prospect in treatment of hepatitis B.
Brief Description of the Drawings
FIGs. 1A-IF show MTT assay showing the cell viability of different cell lines after a series
of Sph concentration treatment.
FIG. IG is a RNA-seq analysis of the expressions of host factors after 90 M Sph treatment
compared to DMSO treatment.
FIGs. 2A-2B show ELISA assay showing HBsAg and HBeAg secretion levels in cell
supernatant in HBV-infected PHHs (A) and HepG2-NTCP cells (B) after Sph treatment.
FIG. 2C is dot blot assay to detect HBsAg levels in cell supernatant of HBV-infected PHHs
and HepG2-NTCP cells after Sph treatment.
FIGs. 2D-2E show western blotting assay to detect the expression of intracellular HBsAg in
HBV-infected PHHs (D) and HepG2-NTCP cells (E) after Sph treatment.
FIGs. 3A-3D show detection of HBV RNAs species by RT-PCR and northern blotting in
HBV-infected PHHs (A and C) and HepG2-NTCP cells (B and D).
FIGs. 4A-4D show detection of HBV DNA by quantitative PCR and southern blotting assay
in HBV-infected PHHs (A and C) and HepG2-NTCP cells (B and D). HBV DNA was significantly
inhibited both in HBV-infected PHHs and HepG2-NTCP cells.
FIGs. 5A-5D show detection of the inhibitory effects of Sph in different treating period.
Supernatant of HBV-infected PHHs (A) and HepG2-NTCP cells (C) were collected at different points
of time to measure HBsAg levels. Western blotting detected intracellular HBsAg in HBV-infected
PHHs (B) and HepG2-NTCP cells (D) 3, 6, 9 and 12 days after Sph treatment.
FIGs. 6A-6D show detection of the expressions of intracellular HBV RNAs in HBV-infected
PHHs (A and B) and HepG2-NTCP cells (C and D) by RT-PCR and northern blotting 3, 6, 9 and 12
days after Sph treatment.
FIGs. 7A-7C show Actinomycin D assay to detect the effects of Sph on HBV RNAs
degradation in HBV-infected HepG2-NTCP cells. Results showed there was no effect on HBV RNA
stability after Sph treatment.
FIGs. 8A-8C show the nascent RNA synthesis assay to detect the effect of Sph on the
synthesis of HBV RNAs in HBV-infected HepG2-NTCP cells. Results showed Sph significantly
reduced the level of nascent HBV RNAs.
FIGs. 9A-9C show detection of HBV cccDNA in HepAD38 cells and HBV-infected HepG2
NTCP cells by quantitative PCR (A) and southern blotting assay (B-C), and results showed no
significant difference of the HBV cccDNA levels after Sph treatment compared to DMSO in both cell
lines.
FIG. 9D shows the ratio of total RNAs/cccDNA and 3.5 kb RNA/cccDNA, and results
suggested the ratios were reduced in a dose-dependent manner in HBV-infected HepG2-NTCP cells
after Sph treatment.
FIG. 9E is RNA-Seq to detect the effect of Sph on HBV transcripts, and results suggested
that Sph treatment significantly decreased the HBV transcription level.
FIGs. 10A-OB show detection of the combined effect of Sph and ETV on HBV DNA in
HBV-infected HepG2-NTCP cells. (A) RT-PCR showed the inhibitory effect of Sph combined ETV
on HBV DNA. (B) Dose response matrix showed strong inhibitory effect of Sph combined ETV on
HBV DNA.
FIGs. 11A-IIF show detection of the effect of Sph on exogenous and endogenous HBV
proteins. (A-E) Huh-7 cells were transfected with plasmids expressing core (Flag-HBc), small surface
(Flag-HBs), polymerase (Flag-Pol) or HBx (Flag-HBx and HA-HBx), respectively. Cells were treated
with Sph for 72 hours, western blotting analysis was performed using anti-Flag antibody. (F)
Detection of expression of endogenous HBV X protein in HBV-infected HepG2-NTCP cells after Sph
treatment.
FIGs. 12A-12E show detection of antiviral activity of Sph in a humanised liver mouse model
of HBV infection. HBV infected Human liver-chimeric uPA/SCID mice were randomly divided into
three groups, vehicle group, Sph monotherapy group and Sph combined ETV group. (A) Serum
albumin levels of the three groups. (B) Body weight of three groups of mice. (C) Serum HBsAg levels
of the three groups. (D) Serum HBV DNA levels of the three groups.
FIGs. 13A-13D show detection of total HBV RNAs (A), 3.5 kb HBV RNA (B), HBV DNA
(C) and HBV cccDNA (D) levels in liver tissue of the three groups.
FIG.14 shows analysis of the ratio of total RNAs/cccDNA and 3.5 kb RNA/cccDNA in liver
tissues, and results suggested the ratios were significantly reduced after Sph treatment which
compared to vehicles.
FIG.15 is an immunohistochemistry staining to detect the expressions of intrahepatic HBsAg
and HBx levels. Results suggested that Sph could significantly inhibit the expressions of intrahepatic
HBsAg and HBx levels compared to vehicle group while had no influence on albumin levels.
Detailed Description of the Preferred Embodiments
Sphondin, Sph for short, is also named as 6-methoxyangelicin. In the present invention, it is
found through a high-throughput ELISA experiment that Sph can effectively inhibit secretion of
HBsAg from HBV and has no obvious toxicity to multiple liver cancer cell lines and primary human
hepatocytes (PHH). It is confirmed again through Western blot and Dot blot that Sph can effectively
reduce intracellular and extracellular HBsAg. It is proved through an RT-PCR experiment and a
Northern blot experiment that Sph can inhibit the HBV RNA level. At the same time, it is also found
in the present invention that Sph also has a concentration-dependent inhibiting effect on HBV core
DNA. However, it is not found in the present invention that Sph has an obvious inhibiting effect on a
transcription template cccDNA of HBV, but it is interesting that it is found in the present invention
that Sph can effectively reduce the transcription activity of cccDNA. In addition, a human liver
chimeric mouse model is used to perform an in-vivo experiment in the present invention. After mice
are infected with HBV and continuously cultured for 8 weeks, HBV is stably replicated and then
treated with Sph. It is found by testing virus indexes in serum and liver tissues that Sph can
significantly reduce levels of HBsAg and HBV DNA in the serum of the mice and levels of HBV
RNAs, HBV DNA, an HBV S protein and an HBV X protein in the liver tissues.
It is shown through the results above that Sph affects generation of HBV RNAs by inhibiting
the transcription activity of cccDNA, virology indexes such as a viral protein and DNA are further
reduced, and Sph is a potential and new anti-HBV drug which can reduce the HBsAg level and inhibit
transcription of cccDNA at the same time.
Based on this, the present invention provides sphondin as an effective component in
preparation of drugs for treatment of hepatitis B. Generally, the drugs for treatment of hepatitis B
include not only a safe and effective dose of sphondin, but also one or more of pharmaceutically
acceptable carriers or adjuvants according to needs of different dosage forms.
"Pharmaceutically acceptable" indicates that when a molecular ontology and a composition
are properly administered to animals or humans, adverse, allergic or other adverse reactions are not
caused.
"Pharmaceutically acceptable carriers or adjuvants" should be compatible with sphondin, and
that is to say, these substances can be mixed with sphondin without greatly reducing the effect of a
pharmaceutical composition under normal circumstances. Specific examples of some substances
which can be used as pharmaceutically acceptable carriers or adjuvants include sugars, such as lactose,
glucose and sucrose; starches, such as corn starch and potato starch; cellulose and derivatives thereof,
such as sodium methylcellulose, ethyl cellulose and methyl cellulose; tragacanth powder; malt; gelatin;
talc; solid lubricants, such as stearic acid and magnesium stearate; calcium sulfate; vegetable oils,
such as peanut oil, cottonseed oil, sesame oil, olive oil, corn oil and cocoa butter; polyols, such as propylene glycol, glycerin, sorbitol, mannitol and polyethylene glycol; alginic acid; emulsifiers, such as Tween; wetting agents, such as sodium lauryl sulfate; coloring agents; flavoring agents; tableting agents, stabilizers; antioxidants; preservatives; pyrogen-free water; isotonic salt solutions and phosphate buffers. These substances are used as needed to improve the stability, activity or biological effectiveness of a formula or to produce an acceptable mouthfeel or a smell under oral administration.
In the present invention, unless otherwise specified, drugs are not particularly limited in
dosage forms and can be prepared in the dosage forms such as injection, oral liquid, tablet, capsule,
dripping pill and spray through conventional methods. The dosage forms of the drugs need to be
matched with the administration routes.
In addition, the present invention also provides a drug combination for combined treatment
of hepatitis B and an administration method. The drug combination for combined treatment of
hepatitis B may be any one of the following forms:
(1) Sphondin and other drugs for treatment of hepatitis B are separately prepared into
independent preparations, where dosage forms of the preparations may be the same or different, and
administration routes may also be the same or different. During use, several drugs may be used
simultaneously or sequentially. When several drugs are used sequentially, other drugs are
administered to the body during the period when the former drug is still effective on the body.
When other drugs for treatment of hepatitis B are antibodies, a parenteral administration route
is generally used, such as intravenous injection, intravenous drip or arterial infusion. The prior art may
be taken as a reference for usage and dosage.
When other drugs for treatment of hepatitis B are chemical drugs, there are many
administration forms including gastrointestinal administration or non-gastrointestinal administration.
It is generally recommended to administer the chemical drugs through already known administration
routes.
(2) Sphondin and other drugs for treatment of hepatitis B are prepared into a compound
preparation, where sphondin and other drugs for treatment of hepatitis B are administered at the same
time in the same administration route, and the combination of both may be prepared into a compound
preparation.
It should be noted that combined administration in the present invention refers to reasonable combined administration based on a basic principle of improving the treatment effect and/or reducing adverse reactions. During combined administration, interactions of drugs should include an interaction affecting pharmacokinetics and an interaction affecting pharmacodynamics. During combined administration, drug types should be minimized, so as to reduce adverse drug reactions caused by the drug interactions and avoid reduction of the treatment effect of the drugs, increase of toxicity and opposite results.
Before further describing the specific embodiments of the present disclosure, it is understood that the scope of the present disclosure is not limited to the specific embodiments described below. It is also to be understood that the terminology of the disclosure is used to describe the specific embodiments, and not to limit the scope of the disclosure. The test methods without specific conditions noted in the following embodiments are generally based on conventional conditions or the conditions recommended by the manufacturers.
When the numerical values are given by the embodiments, it is to be understood that the two endpoints of each numerical range and any one between the two may be selected unless otherwise stated. Unless otherwise defined, all technical and scientific terms used in the present disclosure have the same meaning as commonly understood by one skill in the art. In addition to the specific method, equipment and material used in the embodiments, any method, equipment and material in the existing technology similar or equivalent to the method, equipment and material mentioned in the embodiments of the present disclosure may be used to realize the invention according to the grasp of the existing technology and the record of the invention by those skilled in the art.
Unless otherwise stated, the experimental methods, detection methods, and preparation methods disclosed in the present disclosure all employ conventional techniques of molecular biology, biochemistry, chromatin structure and analysis, analytical chemistry, cell culture, recombinant DNA technology in the technical field and related fields.
Embodiment 1
One. Experimental methods
1. Cell culture
HepG2 cells were purchased from American Type Culture Collection (ATCC) and Huh-7
cells were purchased from Health Science Research Resource Bank. HepG2-NTCP cells were kindly
provided by Prof. Ningshao Xia (The Xiamen University, Fujian, China). HepAD38 cells were
purchased from Shanghai Second Military University. Primary human hepatocytes (PHHs) were
purchased from ScienCell (San Diego, USA) and maintained in hepatocyte medium (catalogue no.
5201; Sciencell). Huh-7 and HepG2-NTCP cells were maintained in DMEM supplemented with 10%
fetal bovine serum and 100U/ml penicillin and 100ug/ml streptomycin (Thermo, USA). HepAD38
cells were maintained in DMEM supplemented with 10% fetal bovine serum and1OOU/ml penicillin
and 100ug/ml streptomycin in the presence of 400ug/ml G418 (Merck, German). HepG2-NTCP cells
were maintained in DMEM supplemented with 10% fetal bovine serum and 100U/ml penicillin and
100ug/ml streptomycin in the presence of 2ug/ml Doxycycline (Solarbio, China).All cells were
cultured in a humidified incubator at 37°C with 5% C02.
2. Drugs and plasmids
The natural compounds library including 1124 Chinese medicine monomeric compounds
constructed by ourselves. Sphondin (CAS Number: 483-66-9) was dissolved into 50mM for storage,
and diluted into 1 M to 90 M in cell culture. Entecavir (selleck, CAS Number: 142217-69-4) was
diluted to 25nM. pCH9/3091 was obtained from Prof. Lin Lan (The Army Medical University,
Chongqing, China). HBx-deleted HBV plasmid (HBV-AHBx) was constructed by introducing a stop
codon at the beginning of the HBx gene based on pCH9/3091 (wild-type HBV, HBV WT). The
expression plasmid for 3 X Flag-HBx was amplified by PCR using pCH9/3091 plasmid as template
and subcloned into the p3 X Flag-CMV-7.1 vector using Hind III and XbaI. Transfection of plasmids
was carried out using Lipofectamine 3000TM (Invitrogen, UAS) according to the manufacturer's
instructions.
3. Virus preparation and cell infection
HBV particles were collected from the supernatant of HepAD38 cells. HBV wild type virus
and HBV HBx-deficient virus was collected from the supernatant of Huh-7 cells transfected with HBV
wild type plasmids (HBV WT) or cotransfected with HBV-AHBx and Flag-HBx plasmids.
Concentration of the virus particles was carried out as previously described. Cells were induced 24
hours with PMM before infection, then 1000 MOI of HBV particles or 500 MOI of HBV particles were added to HepG2-NTCP cells or PHHs, respectively in the presence of 5% (v/v) PEG 8000.
4. MTT experiment
Sph was dissolved in DMSO and prepared into a 50 mM stock solution. 1.5*104 Huh-7,
PLC/PRF/5, PHH cells or 2*104 HepG2-NTCP cells and the like were inoculated into a 96-well plate,
and after 24 hours, the Sph stock solution was subjected to doubling dilution with a growth culture
medium to sequentially obtain 9 concentrations including 500 [M, 250 [M, 125 [M, 62.5 [M, 31.25
ptM, 15.6 [M, 7.8 [M, 3.9 M and 1.95 [M; a drug-free treatment group was used as a control, a pure
culture medium was used as a blank, 3 replicate wells were set for each concentration, and 100 [ of
a drug solution was added into each well. After 72 hours, 10 [ of an MTT reagent was added for
incubation at 37C for 4 hours, a supernatant was completely absorbed, 100 [ of a DMSO solution
was added for shaking oscillation for 10 minutes, the OD value was tested at 490 nm, and the CC50
value was calculated.
5. Detection of HBsAg and HBeAg
The levels of HBsAg and HBeAg from cell supernatant were detected using commercial
enzyme-linked immunosorbent assay kits (KHB) according to the manufacturer's instructions. The
serum HBsAg level was quantified by using the Abbott Architect System with HBsAg Reagent Kit
(08P0877, Abbott, Chicago, USA) according to the manufacturer's instructions.
6. Western blotting and Dot blot assay
Cells were lysed by radio immunoprecipitation assay (RIPA) lysis buffer at 4 °C for 20 min.
Then, the cell lysates were subjected to 15% SDS-PAGE and transferred to poly-vinylidene difluoride
(PVDF) membranes. After blocked in 5% nonfat milk, the membrane was incubated with primary
antibody at 4 °C overnight. Next day the membrane was incubated with secondary antibody and the
Immunoreactive signals were detected by ECL western blot reagents (WBKLS0500, Millipore,
Massachusetts, USA). GAPDH was used as a loading control.
For dot blot assay, 4 microliters of the supernatant were spotted onto a nitrocellulose
membrane (GE Healthcare, Buckinghamshire, UK), air dried, soaked in 2.5% formaldehyde-PBS for
min, rinsed with water at room temperature for 5 min, and then soaked in 50% methanol for 30
min. The following procedures were the same as western blotting.
7. Extraction and detection of RNA
After cells were washed with PBS, total RNAs of the cells were extracted by using a TRizol
method, 1 g of RNA was used for reverse transcription by using a Fast KingRT Kit of TIANGEN
company to synthesize cDNA, and then qRT-PCR was performed to detect a target gene.
Total HBV RNAs forward: 5'- ACCGACCTTGAGGCATACTT-3' (SEQ ID NO. 1), and
total HBV RNAs reverse: 5'- GCCTACAGCCTCCTAGTACA-3' (SEQ ID NO. 2); HBV 3.5-kb
mRNA forward: 5'- GCCTTAGAGTCTCCTGAGCA-3'(SEQ ID NO.3), and HBV 3.5-kb mRNA
reverse: 5'-GAGGGAGTTCTTCTTCTAGG-3'(SEQ ID NO.4).
3 replicate wells were set for each sample, and each experiment was repeated 3 times. P- actin
served as an internal reference.
8. Extraction and detection of HBV core DNA
After cells were washed with PBS, 0.5 ml of a cell lysis buffer (containing 10 mM Tris-HCl
with a pH of 8.0, 1 mM EDTA, 1% of NP-40 and 2% of sucrose) was added and uniformly mixed,
the mixture was incubated at 37°C for 15 minutes, and 15000 g of a cell lysis buffer was collected and
centrifuged for 5 minutes. A supernatant was transferred, 40 U/ml of DNaseI and 10 mM MgCl2 were
added, the mixture was incubated at 37°C for 4 hours, 200 1 of 35% PEG8000 (containing 1.5 mol/L
of NaCl) was added, treatment with an ice bath was performed for 1 hour, 11,000 g of the mixture
was centrifuged at 4°C for 10 minutes, a supernatant was removed, and 0.5 ml of a proteinase K
digestion solution (containing 0.5% of SDS, 150 mM NaCl, 25 mM Tris-HCl with a pH of 8.0 and 10
mM EDTA) and 0.5 mg/ml of a proteinase K (promega) were added for a water bath at 45°C overnight.
On the next day, phenol and chloroform equal in volume were used for extraction, 70% ethanol was
used for precipitation, and sterilized ddH20 was used to dissolve HBV DNA. A product was
quantified by using q-PCR. An HBV DNA primer, F: CCTAGTAGTCAGTTATGTCAAC (SEQ ID
NO. 5), R: TCTATAAGCTGGAGGAGTGCGA (SEQ ID NO. 6). 3 replicate wells were set for each
sample, and each experiment was repeated 3 times.
Southern blotting was performed following the DIG-High Prime DNA Labeling and
Detection Starter Kit (11585614910, Roche, Germany). Briefly, DNA samples were separated on a
0.9% agarose gel and then transferred onto a nylon membrane overnight (Roche, Germany). After UV crosslinking and pre-hybridization, the membrane was hybridized with DIG-labeled full-length HBV genome probe at 42 °C overnight. Next day, the membrane was washed in different concentration of
SSC/SDS, washing buffer, detection buffer. After blocking and incubating with anti-DIG antibody,
then placed membrane with DNA side facing up on a development folder and applied 1 ml CSPD
ready-to-use. After incubating for 5 min at +15 to +25°C, the signal was detected using X-ray film.
9. Hirt cccDNA extraction and detection
Cells were lysed in 500 L cell lysis buffer (50 mM Tris-HCl, pH 8.0, 10 mM EDTA, 150
mM NaCl, 1% SDS) at 37C for 20 min. Then, 125 1 of 2.5 M KCl was added to the cell lysate, and
the solution was stationary overnight at 4°C. The next day, after centrifugation at 12,000 g for 20 min,
twice, the supernatant was collected and extracted by using phenol/chloroform/isopropanol reagent
for twice and DNA was precipitated by adding the same volume of isopropanol. Then, the DNA
precipitation was washed with 70% ethanol and dissolved in double distilled water.
HBV cccDNA levels were detected by Taq-man probe qRT-PCR. Before used as the
templates for Taq-man probe qRT-PCR, the sample was treated as follows: 1) Denaturate rcDNA:
heat at 800 C for 5 minutes, immediately place on ice to cool; 2) Digest rcDNA: incubating with
exonuclease V (M0345S, New England Biolabs, MA, USA) at 37 C for 30 min; 3) Denature
cccDNA: Heating at 1000 C for 20 min to denature cccDNA. After the above processing, samples
were subjected to Taq-man probe qRT-PCR for cccDNA quantification. The PCR reaction contains
0.8 M of primers and 0.2 M of probe and the thermal cycling conditions are as follow: 10 min at
°C, 45 cycles of 15 s at 95 °C and 30 s at 64 °C.
10. Northern blotting
Total RNA was detected by DIG Northern Starter Kit (12039672910, Roche, Mannheim,
Germany) following manufacturer's instructions. The extracted RNA was separated by 1.4%
formaldehyde-agarose gel. The RNA was transferred onto nylon membrane by capillary siphon
method overnight. After UV-crosslinking, the membrane was hybridized with DIG-labeled HBV
RNA probe, and washed in different concentration of SSC/SDS, washing buffer and detection buffer.
Then blocked and incubated with secondary antibody, next placed membrane with RNA side facing
up on a development folder and applied 1 ml CDP ready-to-use. After incubating for 5 min at +15 to
+25°C, the signal was detected using X-ray film. The 28S/18S RNA was set as internal control.
11. Nascent RNA synthesis assay and Actinomycin D assay
HepG2-NTCP cells in a 12-well plate were labelled by addition of 5-ethynyl uridine (EU, an
alkyne-modified nucleoside) at a final concentration of 0.5 mM to the cell culture medium. Cells were
incubated with -/+ EU at 37C for 1 h and washed once with 1x PBS. Then, the cells were lysed in
TRNzol Reagent (TIANGEN, China), and total RNA was extracted according to the manufacturer's
instructions. To enrich, EU-labelled nascent RNA was biotinylated through a click reaction. Then, the
biotinylated EU-labelled nascent RNA was precipitated by incubation overnight at -70°C. Streptavidin
TI was bound to magnetic beads, and 50 L of Dynabeads@ MyOneTM Streptavidin TI magnetic
beads was mixed with 500 ng of biotinylated total RNA. The beads were washed 3 times with 500 p
L of Click-iT@ reaction wash buffer 2 (Thermo). After the last wash, RNA was eluted into 40 pl
DEPC-treated water by heating at 95°C for 10 minutes. First-strand cDNA was synthesized from 1 g
of RNA using an iScriptTM cDNA Synthesis kit (Bio-Rad). Relative RNA expression levels were
quantified with FastStart Universal SYBR Green Master Mix (Roche), and p-actin mRNA was used
as an endogenous control. Values were analysed using the 2^-AACt method. (QuantStudio 6 Flex,
Applied Biosystems).
12. RNA-seq
After the RNA concentration and quality were determined with a [email protected] Fluorometer
(Life Technologies, U.S.A.) and a Nanodrop One spectrophotometer (Thermo Fisher Scientific Inc.,
U.S.A.), total RNA was isolated using the RNeasy mini kit (Qiagen, Germany). The integrity of total
RNA was assessed using an Agilent 2100 Bioanalyzer (Agilent Technologies Inc., U.S.A.), and
samples with RNA integrity number (RIN) values above 7.0 were used for sequencing. One
microgram of RNA was used as input material for RNA sample preparations. RNA-seq strand-specific
libraries were constructed using a VAHTS Total RNA-seq(H/M/R) Library Prep Kit (Vazyme, China)
according to the manufacturer's instructions. Briefly, RNA was purified with magnetic beads after
removal of rRNA. Then, the RNA was fragmented into small pieces using divalent cations for 8 min
at 94oC. The cleaved RNA fragments were copied into first strand cDNA using reverse transcriptase
and random primers. Second strand cDNA synthesis was subsequently performed using DNA
Polymerase I and RNase H. These cDNA fragments were then subjected to the end repair process, addition of a single 'A' base, and ligation of the adapters. The products were purified and enriched via PCR to create the final cDNA library. Purified libraries were quantified and validated using the [email protected] Fluorometer and Agilent 2100 Bioanalyzer to confirm the insert size and calculate the molar concentration. Clusters were generated with cBot after the library was diluted to 10 pM, and then were sequenced on the Illumina NovaSeq 6000platform (Illumina, U.S.A.). The library construction and sequencing were performed by Sinotech Genomics Co., Ltd (Shanghai, China).
13. Mouse model construction and drug treatment
15 human liver-chimeric Alb-uPA/SCID mice were purchased from Beijing Vitalstar Biotechnology Co., Ltd. (Beijing, China). Fresh serum of a patient was injected into the mice through a caudal vein, the replication level of HBV in the mice was tested after the mice were subjected to normal feeding for 8 weeks, and the mice were randomly divided into 3 groups: a control group (injected with an equal volume of a solute without drugs), an Sph group (2.5 mg/kg/2 days, abdominal administration) and an ETV+Sph group (ETV: 0.02 mg/kg/2 days, intragastric administration). Blood collection was performed from the eye orbits every 4 days, the serum was separated and cryopreserved at -80°C, the mice were killed by cervical dislocation at the 7th week, blunt dissection was performed on livers of the mice, a large and complete tissue piece was cut out for paraffin-embedded sectioning, the remaining tissues were to be ground, and DNA and RNA were extracted.
14. Extraction of mouse serum virus DNA
A Biospin virus DNA extraction kit (BioFlux) was used, 10 pl of serum was added into 190 ul of normal saline, 10 1 of a proteinase K was added into a new 1.5 ml centrifuge tube, and then 200 1 of a lysis buffer was added for uniform mixing and shaking for 5-10 seconds. The mixture was incubated at 56°C for 15 minutes. 200 1 of absolute ethanol was added into the centrifuge tube above for thorough and uniform mixing and shaking for 5-10 seconds. Viral DNA was extracted with a purification column in the kit according to manufacturer's instructions of the kit, and a DNA sample was preserved at -20°C.
15. Extraction of tissue DNA and RNA
Mouse livers were separated, tissue pieces were ground into dry powder in liquid nitrogen, about 20 mg of the dry powder was weighed and added into a 1.5 ml centrifuge tube, a DNA lysis buffer and Trizol were separately added, and polishing was performed. After polishing and homogenizing were performed, DNA was extracted with a Biospin tissue genomic DNA extraction kit (BioFlux), 600 1 of an FL buffer and 10 1 of a PK solution were added and mixed thoroughly and uniformly, a small disperser (T10 basic ULTRA-TURRAX@) was used for auxiliary lysis, the mixture was incubated at 56°C for 15 minutes to fully lyse tissues, viral DNA was extracted with a purification column in the kit according to manufacturer's instructions of the kit, and a DNA sample was preserved at -20°C. RNA was extracted by using a cell RNA extraction method.
16. Immunohistochemistry
Paraffin-embedded sections were put into an oven for standing overnight at 55°C; on the next
day, the paraffin-embedded sections were continuously baked at 95°C for 10 minutes; then the
paraffin-embedded sections were sequentially put into xylene and concentration gradient of ethanol
for dewaxing treatment. Antigen retrieval was performed twice in a microwave, and then 0.5% Triton
100/PBS was added to permeate a membrane for 20 minutes; an endogenous peroxidase blocker was
added to remove an endogenous peroxidase; goat working serum was added dropwise to block non
specific sites; an HBs antibody was added for incubation overnight; on the next day, after the sections
were washed, a biotin-labeled secondary antibody and a streptavidin-peroxidase reagent were added
dropwise in sequence; finally, a newly prepared DAB color development reagent was added dropwise
for color development for 3-5 minutes; hematoxylin was added for redyeing at room temperature for
1 minute, and the sections were turned blue when rinsed with tap water; the sections were dehydrated,
sealed and observed under a microscope.
17. Statistical processing
The results are expressed as the meanSD. Statistical analyses were performed using either
Student's t-test or the Mann-Whitney U test and by one-way ANOVA. Differences were considered
significant when P<0.05. All statistical analysis was performed by using SPSS 19.0 software.
Two. Experimental results
1. The cytotoxicity of sphondin detected through an MTT experiment
As shown in FIG. 1, it is shown through results of MTT that CC50 of sphondin (Sph) on
multiple liver cancer cell lines and PHH cells is higher than 500 [M, and it is shown through RNA- seq that Sph has no significant effect on human liver specific factors after action, indicating that Sph has little toxicity to cells.
2. Inhibition of the expression of HBsAg and HBeAg by sphondin
HBV-infected PHH and HepG2-NTCP cells are treated with Sph to detect the secretion level
and intracellular expression level of HBsAg and HBeAg after drug treatment. Results in FIGs. 2A-2B
indicated that sphondin could inhibit the expression of HBsAg and HBeAg. Results in FIGs. 2C-2E
suggested sphondin could significantly inhibit the expression of HBsAg both in supernatant and
intracellular in a dose-dependent manner compared to DMSO. Results in FIG. 5 suggested sphondin
decreased the HBsAg both in supernatant and intracellular in a time-dependent manner.
3. Inhibition of the transcription level of HBV RNAs by sphondin
HBsAg is translated from HBV 2.4/2.1-kb RNA and used to detect whether HBV RNAs are
inhibited by Sph, HBV-infected PHH and HepG2-NTCP cells are treated with different concentrations
of Sph, and the cells at different time points are collected to extract total RNAs. It is shown through
results of qRT-PCR and Northern blot that HBV RNAs (including 3.5-kb RNA and 2.4/2.1-kb RNA)
are reduced by Sph in a dose-dependent manner (FIG. 3) and time-dependent manner (FIG. 6), and
EC50 is 23.48 M and 19.24 M (PHHs) and 9.26 M and 6.14 M (HepG2-NTCP cells) respectively
(FIG. 3).
4. Inhibition of the replication level of HBV core DNA by sphondin
HBV 3.5-kb RNA can be further subjected to reverse transcription to generate HBV core
DNA, so as to detect whether Sph can also inhibit the HBV core DNA level; similarly, HBV-infected
PHH and HepG2-NTCP cells are treated with concentration gradient of Sph, and the HBV core DNA
level in the cells is detected by using q-PCR and Southern blot. It is shown through results that the
HBV core DNA level is reduced by Sph in a dose-dependent manner (FIG. 4).
5. Inhibition of the transcription activity of cccDNA by sphondin
After the inhibiting effect of Sph on various virology indexes of HBV is clear, the effect of
Sph on a transcription template cccDNA of HBV is further tested. It is shown through Taq-Man probe
PCR that Sph has no obvious inhibiting effect on the replication level of cccDNA, but it is interesting
that Sph can effectively inhibit the transcription activity of cccDNA; at the same time, it is also found through RNA-Seq that Sph can effectively reduce levels of HBV transcripts (FIG. 9).
6. Inhibition of the synthesis ofnascent HBV RNA by sphondin
Results of nascent RNA synthesis assay indicated that sphondin significantly reduced the
level of nascent HBV RNAs (FIG. 8).
7. Test on in-vivo toxicity of sphondin
In order to explore an in-vivo antiviral effect of Sph, a toxic effect of Sph in mice is tested
first. 15 BALB/c mice at the age of 6-8 weeks are randomly divided into 3 groups, treated with
different doses of Sph (including a 0 mg/kg group, a 2.5 mg/kg group and a 5 mg/kg group) and
administered once every two days through abdominal administration, and the body weight of the mice
is monitored at the same time. After the mice are continuously treated for 28 days, blood collection is
performed from eyeballs, and blood routine indexes and blood biochemical indexes are tested. As
shown in Table 1, treatment with Sph has no significant effect on the body weight, white blood cells,
red blood cells, hemoglobin, platelets, total proteins, albumin, ALT, AST, GGT, total bilirubin, serum
creatinine and blood urea of the mice. It is shown that sph has little toxicity in the body and has no
obvious toxicity to liver and kidney. Therefore, sph with a concentration of 2.5 mg/kg is used in
following experiments (Table 1).
Table 1 Effects of sphondin on the body weight, blood routine indexes and blood biochemical
indexes of mice
Sph Project/Unit 0 mg/kg/2 day 2.5 mg/kg/2 day 5 mg/kg/2 day
NC group Low concentration High concentration
WBC *10 3/ L 7.08+0.17 5.95±1.91ns 6.8±2.1s
RBC *106/ L 11.72+0.26 11.56±0.33"n 11.68±0.38"n
HGB g/L 165.25+1.92 162±4.06"n 162.75±6.3"n
HCT % 52.7±0.48 51.93±1.28"n 51.71±1.39"n
PLT *10 3 4/L 874.6+347.48 584.2±107.06"n 753.55+239.41"s
MCV fL 45±1.01 44.95+0.69"s 44.29±1.44"n
MCH pg 14.1+0.14 14.03±0.18"n 13.96±0.21s
MCHC g/L 313.75±4.6 312±2.24"n 314.85±9.82"s
LYMPH *10 9/FL 4.99+0.19 4.07±1.4Os 4.685±1.53"n
BUN mg/dL 13+2.92 14.75±1.3"n 14±1.87"n
TP g/dL 5.75+0.36 5.45±0.23"n 5.5±0.47"n
ALB g/dL <0.1I <0.1"ns <0.1"ns
ALT U/L 67.75+14.08 69.25±3.77"n 67.25+7.29"s
AST U/L 118.25+5.67 116.75±10.11s 124.25±19.14"n
ALKP U/L 134.75+14.18 115.5+15.79"s 139.75±21s
GGT U/L 0.75+1.3 0.25±0.43"n 1.25±0.43"n
TBIL mg/dL 0.475+0.33 0.2250.11"s 0.23±0.04"n
All values in Table 1 are expressed as mean standard deviation.
Abbreviations in Table 1: WBC, white blood cell; RBC, red blood cell; HGB, hemoglobin;
HCT, hematocrit; PLT, platelets; MCV, mean corpusular volume; MCH, mean corpusular hemoglobin;
BUN, blood urea nitrogen; TP, total protein; ALB, albumin; ALT, alanine transaminase; AST,
aspartate transaminase; ALKP, alkaline phosphatase; GGT, y-glutamyl transpeptidase; TBIL, total
bilirubin.
Compared with the sphondin group with a concentration of 0 mg/kg/2 days, the sphondin
group with a concentration of 2.5 mg/kg/2 days has no significant effect (p>0.05).
8. Test of an in-vivo antiviral effect of sphondin by using a human liver chimeric mouse
model
Human liver chimeric mice infected with HBV for 8 weeks are randomly divided into 3
groups (including a control group, a 2.5 mg/kg group and an ETV+sph group) with 4 mice in each
group. The mice are administered once every 2 days, and blood collection is performed from the eye
orbits every 4 days. The mice are killed by cervical dislocation after treatment for 7 weeks, and then
livers are taken. Levels of HBsAg and HBV DNA in serum of the mice and levels of HBV RNAs,
HBV DNA, HBs and an HBx protein in liver tissues are tested. It is shown through results that Sph
can effectively reduce the levels of HBsAg and HBV DNA in the serum and the levels of HBV RNAs,
HBV DNA and viral proteins in the liver tissues, and especially has an obvious inhibiting effect on
HBV RNAs, HBsAg and HBx; in addition, an antiviral effect can be improved after Sph is combined with ETV (entecavir) (FIGs. 12-15).
The above-mentioned embodiments are merely illustrative of the principle and effects of the
present disclosure instead of limiting the present disclosure.
Modifications or variations of the above-described embodiments may be made by those
skilled in the art without departing from the spirit and scope of the disclosure.
Therefore, all equivalent modifications or changes made by those who have common
knowledge in the art without departing from the spirit and technical concept disclosed by the present
disclosure shall be still covered by the claims of the present disclosure.
Where any or all of the terms "comprise", "comprises", "comprised" or "comprising" are used
in this specification (including the claims) they are to be interpreted as specifying the presence of the
stated features, integers, steps or components, but not precluding the presence of one or more other
features, integers, steps or components.
A reference herein to a patent document or any other matter identified as prior art, is not to
be taken as an admission that the document or other matter was known or that the information it
contains was part of the common general knowledge as at the priority date of any of the claims.

Claims (5)

What is claimed is:
1. The use of sphondin as an effective component in preparation of drugs for treatment of hepatitis
B.
2. The use according to claim 1, wherein the drugs for treatment of hepatitis B have at least one of
the following functions:
inhibiting secretion of HBsAg from HBV;
inhibiting a transcription level of HBV RNAs;
inhibiting a replication level of HBV core DNA in a concentration-dependent manner;
inhibiting a transcription activity of cccDNA;
inhibiting levels of HBV RNAs, HBV DNA, an HBV S protein and an HBV X protein in liver
tissues.
3. A medicinal preparation for treatment of hepatitis B, comprising a safe and effective dose of
sphondin.
4. A drug combination for combined treatment of hepatitis B, comprising a safe and effective dose
of sphondin and at least one other drug for treatment of hepatitis B.
5. Uses of sphondin in preparation of substances having any one or more of the following effects: a
use for preparing a substance for inhibiting secretion of HBsAg from HBV, a use for preparing a
substance for inhibiting a transcription level of HBV RNAs, a use for preparing a substance for
inhibiting a replication level of HBV core DNA, a use for preparing a substance for inhibiting a
transcription activity of cccDNA, and a use for preparing a substance for inhibiting levels of HBV
RNAs, HBV DNA, an HBV S protein and an HBV X protein in liver tissues.
AU2021107213A 2021-05-11 2021-08-25 Use of sphondin as an effective component in preparing medicine for treating hepatitis b Active AU2021107213A4 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021105149667 2021-05-11
CN202110514966.7A CN113069445A (en) 2021-05-11 2021-05-11 Application of bovine ledebouriella seseloides as effective component in preparing medicine for treating hepatitis B

Publications (1)

Publication Number Publication Date
AU2021107213A4 true AU2021107213A4 (en) 2021-12-09

Family

ID=76616475

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2021107213A Active AU2021107213A4 (en) 2021-05-11 2021-08-25 Use of sphondin as an effective component in preparing medicine for treating hepatitis b

Country Status (3)

Country Link
CN (1) CN113069445A (en)
AU (1) AU2021107213A4 (en)
WO (1) WO2022237007A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113069445A (en) * 2021-05-11 2021-07-06 重庆医科大学 Application of bovine ledebouriella seseloides as effective component in preparing medicine for treating hepatitis B

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113304165B (en) * 2021-07-15 2022-07-22 重庆医科大学 Application of monomeric compound Ciliaoside A in preparation of hepatitis B treatment drug

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4939167A (en) * 1988-07-22 1990-07-03 Jean Jacques Goupil Use of 5-methoxypsoralen and other furocoumarins as jet lag suppressants
US6103706A (en) * 1997-04-29 2000-08-15 New York Blood Center, Inc. Methods for treating viral infections
US6242188B1 (en) * 1999-07-30 2001-06-05 Applied Gene Technologies, Inc. Sample processing to release nucleic acids for direct detection
US20060024691A1 (en) * 2004-03-25 2006-02-02 Buck Institute For Age Research Novel pathways in the etiology of cancer
US20090312406A1 (en) * 2008-06-12 2009-12-17 Hsing-Pang Hsieh Coumarin compounds and their use for treating viral infection
US20110268722A1 (en) * 2010-04-22 2011-11-03 Siegelin Markus D Combination therapies with mitochondrial-targeted anti-tumor agents
CN101904839B (en) * 2010-08-06 2012-06-20 中国人民解放军第二军医大学 Application of imperatorin in preparing medicament for preventing and treating hepatitis or liver injury
CN103191105B (en) * 2013-03-18 2015-08-19 中山大学 One class furocoumarin compound is preparing the application in anti-hepatitis B virus (HBV) medicine
CN105616463A (en) * 2014-11-10 2016-06-01 南京浩鼎生物科技有限公司 Side-effect-free conditioning agent for inhibiting hepatitis B virus
CN105456517A (en) * 2015-12-15 2016-04-06 彭国能 Traditional Chinese medicinal composition and preparation for treating chronic hepatitis B and preparation method of preparation
CN107496476A (en) * 2017-09-29 2017-12-22 北京健旭康技术有限公司 The beneficial liver effect of yunna cowparsnip and application technology
CN109364074B (en) * 2018-11-01 2021-05-07 重庆医科大学 Application of 6-aminonicotinamide as effective component in preparing medicament for treating hepatitis B
WO2020118159A1 (en) * 2018-12-07 2020-06-11 The University Of Chicago Methods and compositions comprising an nfkb inhibitor and an adjuvant
CN113069445A (en) * 2021-05-11 2021-07-06 重庆医科大学 Application of bovine ledebouriella seseloides as effective component in preparing medicine for treating hepatitis B
CN113304165B (en) * 2021-07-15 2022-07-22 重庆医科大学 Application of monomeric compound Ciliaoside A in preparation of hepatitis B treatment drug

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113069445A (en) * 2021-05-11 2021-07-06 重庆医科大学 Application of bovine ledebouriella seseloides as effective component in preparing medicine for treating hepatitis B

Also Published As

Publication number Publication date
WO2022237007A1 (en) 2022-11-17
CN113069445A (en) 2021-07-06

Similar Documents

Publication Publication Date Title
Liu et al. Identification of hydrolyzable tannins (punicalagin, punicalin and geraniin) as novel inhibitors of hepatitis B virus covalently closed circular DNA
AU2021107213A4 (en) Use of sphondin as an effective component in preparing medicine for treating hepatitis b
Tatsukawa et al. Role of transglutaminase 2 in liver injury via cross-linking and silencing of transcription factor Sp1
Sekiba et al. Pevonedistat, a neuronal precursor cell‐expressed developmentally down‐regulated protein 8–activating enzyme inhibitor, is a potent inhibitor of hepatitis B virus
JP2012522013A (en) Regulated IRES-mediated translation
Huang et al. Antiviral activity of chemical compound isolated from Artemisia morrisonensis against hepatitis B virus in vitro
CN107635566A (en) Treat the composition and method of HBV infection
CN112675174B (en) New use of poly ADP ribose polymerase inhibitor for treating hepatitis B virus related diseases
US8580759B2 (en) Anti-hepatitis C virus composition
WO2018193902A1 (en) Antiviral effect of microrna against hepatitis b virus
Nio et al. Bardoxolone methyl as a novel potent antiviral agent against hepatitis B and C viruses in human hepatocyte cell culture systems.
Wen et al. Flavonoids derived from licorice suppress LPS-induced acute lung injury in mice by inhibiting the cGAS-STING signaling pathway
US10123997B2 (en) Target for treating hepatitis B virus
EP3395363B1 (en) Compounds for use in treating hbv-and hcv-related conditions
Ren et al. Sphondin efficiently blocks HBsAg production and cccDNA transcription through promoting HBx degradation
CN116327943A (en) Application of KLF15 in alcoholic liver disease
CN114796233A (en) Application of asiatic acid in preparing medicine for treating hepatitis B
JP2023510274A (en) Use of FXR agonists to treat infection by hepatitis D virus
CN109771410B (en) Application of alkannic acid in preparing anti-hepatitis B virus medicine
Yang et al. Fibronectin is essential for hepatitis B virus propagation in vitro: may be a potential cellular target?
Zhang et al. Rapamycin inhibits hepatitis B virus covalently closed circular DNA transcription by enhancing the ubiquitination of HBx
US10378015B2 (en) Targeting hepatitis B virus (HBV) host factors
EP3773575A1 (en) Methods for treating hepatitis b virus (hbv) infection
CN111172165B (en) Application of combination of siRNA and membrane-penetrating peptide in treatment of liver cancer
TWI620933B (en) Method of detecting carbohydrate metabolism disorder and the prevention and treatment

Legal Events

Date Code Title Description
FGI Letters patent sealed or granted (innovation patent)