WO2016167369A1 - Inhibiteur de sécrétion de virus d'hépatite b - Google Patents

Inhibiteur de sécrétion de virus d'hépatite b Download PDF

Info

Publication number
WO2016167369A1
WO2016167369A1 PCT/JP2016/062225 JP2016062225W WO2016167369A1 WO 2016167369 A1 WO2016167369 A1 WO 2016167369A1 JP 2016062225 W JP2016062225 W JP 2016062225W WO 2016167369 A1 WO2016167369 A1 WO 2016167369A1
Authority
WO
WIPO (PCT)
Prior art keywords
hbv
gene
sugar chain
slc35b1
inhibitor
Prior art date
Application number
PCT/JP2016/062225
Other languages
English (en)
Japanese (ja)
Inventor
清彦 安形
成松 久
晶 栂谷内
佐藤 隆
清顕 伊藤
政志 米田
Original Assignee
国立研究開発法人産業技術総合研究所
学校法人愛知医科大学
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 国立研究開発法人産業技術総合研究所, 学校法人愛知医科大学 filed Critical 国立研究開発法人産業技術総合研究所
Priority to JP2017512609A priority Critical patent/JP6566368B2/ja
Publication of WO2016167369A1 publication Critical patent/WO2016167369A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • 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
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • 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
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/711Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
    • 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
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5067Liver cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/01DNA viruses
    • G01N2333/02Hepadnaviridae, e.g. hepatitis B virus
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2400/00Assays, e.g. immunoassays or enzyme assays, involving carbohydrates
    • G01N2400/10Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters

Definitions

  • the present invention relates to identification of a glycan gene involved in HBV secretion in hepatitis B virus (HBV) -infected hepatocytes, a method for inhibiting HBV secretion targeting the glycan synthesis-related protein or its gene, and the glycan
  • HBV hepatitis B virus
  • the present invention relates to an HBV therapeutic agent and an HBV therapeutic method comprising an activity inhibitor of a synthesis-related protein or a gene expression inhibitor thereof as an active ingredient.
  • Hepatitis B virus belongs to hepadnavirus and has incomplete double-stranded DNA that specifically infects human liver. The infection is caused by acute hepatitis B, chronic hepatitis, cirrhosis, hepatocytes This is one of the major health problems. Currently there are approximately 1.1 to 1.4 million hepatitis B virus (HBV) carriers in Japan, but there are 350 million infected people worldwide (WHO report) ), HBV therapeutics are expected to be developed worldwide. Despite universal vaccination for HBV in most countries and regions, it is considered difficult to prevent the onset of newly infected patients. In addition to conventional maternal-infant infection, horizontal infection is also spreading.
  • HBV hepatitis B virus
  • hepatitis B is mainly treated with interferon (IFN) (injection) and nucleic acid analog preparations (lamivudine [LMV], adefovir [ADV], entecavir [ETV], etc.) (internal use).
  • IFN interferon
  • nucleic acid analog preparations lamivudine [LMV], adefovir [ADV], entecavir [ETV], etc.
  • HBV genomic DNA is easily mutated, and over 1000 types of HBV DNA sequences have been reported so far, and over 8 types of HBV genotypes have been reported.
  • the fact that most of these mutation sites are regions encoding reverse transcriptase is also the main cause of the emergence of resistant strains of nucleic acid analog preparations that inhibit reverse transcription reaction. Therefore, it is essential to search for drug discovery targets that can replace reverse transcriptase.
  • HBV infection inhibitory activity can be prevented by inhibiting capsid formation of HBV pregenomic RNA.
  • Low molecular weight compounds Patent Document 1
  • peptides that inhibit binding of HBsAg surface antigen and HBcAg core antigen by peptides that bind to HBV core antigen, etc. Patent Documents 2 and 3 have been proposed.
  • HBV-inhibiting active substance derived from an extract from a natural product
  • a polysaccharide component of a medicinal fungus Patent Document 4
  • a cyclohexane ketone compound derived from an extract of Benix nokitake Patent Document 5
  • a pro extracted from a plant Anthocyanidin oligomers Patent Document 6
  • sphingolipid conventionally used as a therapeutic agent for antifungal infection and immune disease is useful as a therapeutic agent for HBV infection (Patent Document 7)
  • HBsAg surface antigen and HBcAg core antigen are found.
  • Patent Document 8 A non-natural sialic acid-containing sugar chain compound (Patent Document 8) having an activity of binding to HBsAg and an anti-HBsAg antibody (Patent Document 9) that binds to an HBsAg surface antigen have also been proposed.
  • Non-Patent Document 1 Non-Patent Document 1
  • Non-Patent Documents 3 and 4 RNA interference introduces a complementary sequence of mRNA after transcription into the cell, forms a complex with mRNA, and is destroyed by the activity of ribonucleases such as Dicer. As a result, the mRNA expression level and protein level decrease. cause.
  • dsRNA double-stranded ribonucleic acid
  • Etc. Etc.
  • the present invention provides a method for suppressing the formation and / or secretion of HBV in infected hepatocytes, a method for preventing and treating HBV infection caused thereby, and a pharmaceutical composition therefor.
  • HBV particle formation and / or HBV particle secretion in HBV-infected hepatocytes by inhibiting sugar chain modification activity or inhibiting the expression of sugar chain synthesis-related protein targeting such a sugar chain synthesis-related protein or gene thereof He thought that he could prevent or treat hepatitis B by preventing infection of surrounding hepatocytes as a result.
  • glycoprotein synthesis-related protein genes (hereinafter also simply referred to as “glycan genes”) are localized in the endoplasmic reticulum or Golgi apparatus in hepatocytes, they are efficient when used as target molecules for inhibiting HBV infection. It is considered advantageous for effective inhibition.
  • Non-Patent Document 2 the other to 2014.4.19-20 2 nd TASL-Japan HBV in Taipei, 2014.5.29-30 50th liver General meeting of the Society workshop 3,2014.9.3-6 Molecular Biology of HBV announced at Los Angeles. Based on these results, first, total RNA was prepared from each of normal hepatocytes and two types of liver cancer-derived cell lines, a cDNA library was prepared, and comprehensive sequencing was performed.
  • glycan genes in the glycan-related gene database (GGDB, Japan) are extracted, and approximately 185 types of glycan genes expressed in hepatocytes are identified. did. Expression profiles were obtained by comparative analysis of the expression levels of these 185 sugar chain genes. Based on the expression profiles of the 185 types of sugar chain genes, 86 types that are highly expressed or moderately expressed in hepatocytes are extracted, and consist of 3 types of siRNAs corresponding to each gene 3 A seed mixed siRNA was prepared.
  • HBV S-HBs antigen cDNA was transfected into each cell, and S-HBs secreted into the culture supernatant The expression level of the antigen and the addition of sugar chains were confirmed.
  • the expression of S-HBs antigen was reduced or the S-HBs antigen sugar was reduced when expression was suppressed by siRNA introduction compared to control siRNA introduction. A decrease in chain addition was shown.
  • siRNAs used for SLC35B1 gene knockdown were confirmed to have the ability to reduce both the amount of HBV DNA and the amount of HBs antigen as well as the ability to inhibit SLC35B1 gene expression.
  • the position on the base sequence was identified.
  • siRNA (siSLC35B1 # 1) which has the most ability to inhibit SLC35B1 gene expression among the three species, reduces the amount of HBV DNA and HBs antigen, that is, HBV particle formation and HBV secretion in HBV-infected cells. It was also confirmed that the function was inhibited.
  • siRNA dsRNA
  • the base sequence of the highly effective target region in the SLC35B1 gene sequence is determined by comparing each SLC35B1 gene expression inhibition ability I was able to.
  • RNAi reagents such as shRNA that generate RNAi in the same target region, antisense, and ribozyme can be used as inhibitors of HBV infection.
  • RNAi reagents such as siRNA and shRNA corresponding to the target region can suppress the growth and secretion of HBV in the liver by using a system that transports siRNA specifically in the liver (Patent Document 16, etc.) It can be used as a therapeutic agent for HBV infection.
  • Patent Document 16 Patent Document 16
  • SLC35B1, CHST9 and ST8SIA3 binding peptides are also HBs surface antigens in HBV-infected hepatocytes. Since it acts as an inhibitor of HBV proliferation and secretion by inhibiting the sugar chain transfer activity, it becomes an active ingredient of a therapeutic agent for HBV infection.
  • hepatocyte glycosylation-related proteins such as SLC35B1 that act on the above-mentioned siRNA are necessary for HBV particle formation and secretion, and inhibition of siRNA expression inhibits HBV particle formation and HBV antigen expression. It was also confirmed that it has little effect on the original proliferation of hepatocytes. Obtaining these findings completed the present invention.
  • the present invention includes the following inventions.
  • An inhibitor of HBV particle formation or HBV secretion in HBV-infected hepatocytes comprising as an active ingredient an inhibitor of at least one sugar chain gene expression selected from the SLC35B1 gene, CHST9 gene, and ST8SIA3 gene.
  • the sugar chain gene expression inhibitor is an oligonucleic acid preparation selected from RNAi preparations, antisenses, nucleic acid aptamers, and ribozymes.
  • a nucleic acid comprising the oligonucleic acid preparation comprising a base sequence containing at least 15 consecutive bases in a base sequence of at least one sugar chain gene selected from SLC35B1 gene, CHST9 gene and ST8SIA3 gene
  • the inhibitor according to [2] above which is contained as an active ingredient.
  • the oligonucleic acid preparation is an RNAi preparation, and a base sequence in which at least 15 consecutive bases coincide with the complementary strand of mRNA of at least one sugar chain gene selected from the SLC35B1 gene, CHST9 gene and ST8SIA3 gene
  • Regions and sequences of the oligonucleic acid agent at positions 102 to 121, 161 to 179, 311 to 330, 477 to 495, 576 to 594, and 627 to 644 of the base sequence shown in SEQ ID NO: 3 A nucleotide sequence comprising at least 15 consecutive bases of any one or more of regions selected from the region of positions 806 to 824 of SEQ ID NO: 10 and the region of positions 178 to 196 of SEQ ID NO: 10 or a complementary sequence thereof.
  • the sugar chain gene expression inhibitor specifically binds to a region consisting of a base sequence containing at least 15 consecutive bases of SEQ ID NO: 12 in the base sequence of SLC35B1 mRNA.
  • An inhibitor of HBV particle formation or HBV secretion in HBV-infected hepatocytes comprising an inhibitor of sugar chain modification activity of at least one sugar chain synthesis-related protein selected from SLC35B1, CHST9, and ST8SIA3.
  • an inhibitor of HBV particle formation or HBV secretion in HBV-infected hepatocytes comprising an inhibitor of sugar chain modification activity of at least one sugar chain synthesis-related protein selected from SLC35B1, CHST9, and ST8SIA3.
  • For prevention, suppression or treatment of HBV infection comprising as an active ingredient at least one inhibitor of HBV particle formation or HBV secretion according to any one of [1] to [7] Pharmaceutical composition.
  • a method for screening an inhibitor of HBV particle formation or HBV secretion comprising the following steps (1) to (4); (1) A step of measuring the expression level of at least one sugar chain gene selected from SLC35B1 gene, CHST9 gene and ST8SIA3 gene in cultured hepatocytes, (2) introducing a test compound into cultured hepatocytes or administering the test compound into a medium; (3) measuring the expression level of the sugar chain gene selected in step (1), (4) When the expression level measured in step (3) decreases compared to the expression level measured in step (1), the test compound is used as a candidate for an inhibitor of HBV particle formation or HBV secretion. Process to select.
  • the test compound includes a test oligonucleic acid, and in the case of a test oligonucleic acid, it is preferably introduced into cultured hepatocytes using a vector or a liposome.
  • a method for screening an inhibitor of HBV particle formation or HBV secretion comprising the following steps (1) to (4); (1) a step of measuring the amount or activity of at least one sugar chain synthesis-related protein selected from SLC35B1, CHST9 and ST8SIA3 in cultured hepatocytes, (2) introducing a test compound into cultured hepatocytes or administering the test compound into a medium; (3) a step of measuring the amount or activity of the sugar chain synthesis-related protein selected in step (1), (4) Inhibitor of HBV particle formation or HBV secretion when the amount or activity value measured in step (3) decreases compared to the amount or activity value measured in step (1) Selecting as a candidate.
  • the test compound includes a test oligonucleic acid, and in the case of a test oligonucleic acid, it is preferably introduced into cultured hepatocytes using a vector or a liposome.
  • a test oligonucleic acid in order to measure the activity of a sugar chain synthesis-related protein, in the case of SLC35B1, for example, the concentration of a sugar chain such as UDP-Gal or the concentration of UDP released from UDP-Gal may be measured.
  • the concentration of inorganic phosphate based on CMP released from CMP-NeuAc containing sialic acid (For example, manufactured by R & D Systems).
  • a method for screening an inhibitor of HBV particle formation or HBV secretion comprising the following steps (1) to (4); (1) A step of preparing a transformed cell transformed with at least one sugar chain gene selected from the SLC35B1 gene, CHST9 gene and ST8SIA3 gene, and measuring the expression level of the selected sugar chain gene in the cell , (2) introducing a test compound into the transformed cell or administering the test compound in a medium; (3) a step of measuring the expression level of the selected sugar chain gene in the transformed cell of step (2), (4) When the expression level of the selected glycan gene measured in step (3) decreases compared to the expression level measured in step (1), the test compound is converted into HBV particles or Selecting as a candidate for an inhibitor of HBV secretion.
  • the test compound includes a test oligonucleic acid, and in the case of a test oligonucleic acid, it is preferably introduced into a transformed cell using a vector or a liposome. Further, instead of the step of measuring the expression level of the sugar chain gene in steps (1) and (3), the activity of the sugar chain synthesis-related protein expressed in the transformed cell may be measured and compared.
  • a method for screening an inhibitor of HBV particle formation or HBV secretion comprising the following steps (1) to (4); (1) Prepare transformed cells that express at least one labeled sugar chain synthesis-related protein selected from labeled SLC35B1, CHST9, and ST8SIA3 on the inner membrane surface, and measure the amount of labeling on the inner membrane surface The process of (2) introducing the test compound into the transformed cell or administering the test compound into the medium; (3) a step of measuring the labeling amount on the surface of the intracellular membrane, (4) When the amount of label measured in step (3) decreases compared to the amount of label measured in step (1), the test compound is selected as a candidate for an inhibitor of HBV particle formation or HBV secretion Process.
  • the test compound includes a test oligonucleic acid, and in the case of a test oligonucleic acid, it is preferably introduced into a transformed cell using a vector or a liposome.
  • the transformed cell is a cell that expresses a tagged sugar chain synthesis-related protein, and is a transformed cell in which the inner membrane surface is fluorescently labeled with a fluorescently labeled anti-tag antibody, and the cell to be measured
  • a kit for screening an inhibitor of HBV particle formation or HBV secretion A kit comprising a transformed cell transformed with at least one sugar chain gene selected from SLC35B1 gene, CHST9 gene and ST8SIA3 gene. Furthermore, a kit combining a sugar chain-nucleotide such as UDP-Gal, PAPS, or CMP-NeuAc for measuring the activity of a sugar chain synthesis-related protein may be used.
  • the sugar chain gene is fused with a gene encoding a marker protein, and the transformed cell has at least one sugar chain synthesis-related protein selected from SLC35B1, CHST9, and ST8SIA3 on the inner membrane surface thereof.
  • kits according to [15] above which is a transformed cell expressed in a directly or indirectly labeled state.
  • a gene encoding the labeled protein for example, a fluorescent protein gene such as a GFP gene is preferably used.
  • a kit for screening an inhibitor of HBV particle formation or HBV secretion comprising the following (1) and (2); (1) A transformed cell that expresses at least one sugar chain synthesis-related protein selected from tagged SLC35B1, CHST9, and ST8SIA3, (2) A labeled anti-tag antibody.
  • the tagged sugar chain synthesis-related protein is a Flag-tagged sugar chain synthesis-related protein
  • the anti-tag antibody is an anti-Flag antibody.
  • a kit for screening for inhibitors of HBV particle formation or HBV secretion comprising the following (1) and (2); (1) a substrate on which at least one sugar chain synthesis-related protein selected from SLC35B1, CHST9 and ST8SIA3 is immobilized; (2) A library comprising nucleic acids containing random base sequences and corresponding peptides.
  • the length of the random base sequence is usually 21 to 36 bases (7 to 12 amino acids).
  • it is a peptide library by phage display, and may be a peptide library obtained by translating a nucleic acid library with a cell-free translation system.
  • the method comprises the step of administering an expression inhibitor of at least one sugar chain gene selected from SLC35B1 gene, CHST9 gene and ST8SIA3 gene to HBV-infected hepatocytes.
  • an expression inhibitor of at least one sugar chain gene selected from SLC35B1 gene, CHST9 gene and ST8SIA3 gene to HBV-infected hepatocytes.
  • the step of administering a sugar chain gene expression inhibitor is a step of introducing an oligonucleic acid preparation selected from RNAi preparations, antisenses, nucleic acid aptamers and ribozymes into cells, and the oligonucleic acid preparation at that time is, for example, A double-stranded RNA comprising at least one base sequence selected from SLC35B1 gene, CHST9 gene and ST8SIA3 gene or a complementary sequence thereof and a base sequence comprising at least 15 consecutive bases as an active ingredient.
  • SLC35B1 gene, CHST9 gene and ST8SIA3 gene expression inhibitor instead of the SLC35B1 gene, CHST9 gene and ST8SIA3 gene expression inhibitor to be administered, at least one sugar chain synthesis-related protein sugar chain selected from SLC35B1, CHST9 and ST8SIA3 A modified activity inhibitor may be used. That is, the present invention can be expressed as follows.
  • the inhibitor of HBV particle formation and secretion is an expression inhibitor of at least one sugar chain gene selected from SLC35B1 gene, CHST9 gene and ST8SIA3 gene, Or a sugar chain modification activity inhibitor of at least one sugar chain synthesis-related protein selected from SLC35B1, CHST9 and ST8SIA3, A method comprising: as an active ingredient.
  • this invention can prevent, suppress or treat HBV infection by inhibiting HBV particle formation and secretion in HBV-infected cells, the following aspects are also included.
  • a method for preventing, suppressing or treating HBV infection Administering a pharmaceutical composition to a patient in need of the method,
  • the pharmaceutical composition comprises an inhibitor of expression of at least one sugar chain gene selected from SLC35B1 gene, CHST9 gene and ST8SIA3 gene, Or a sugar chain modification activity inhibitor of at least one sugar chain synthesis-related protein selected from SLC35B1, CHST9 and ST8SIA3, A method comprising at least one of the above as an active ingredient.
  • the present invention also includes the following aspects.
  • a gene expression inhibitor for use in a method for preventing, suppressing or treating HBV infection wherein the gene expression inhibitor is at least one selected from the group consisting of SLC35B1 gene, CHST9 gene and ST8SIA3 gene A gene expression inhibitor characterized by suppressing the expression of two target sugar chain genes.
  • a nucleic acid for use in a method for preventing, suppressing or treating HBV infection wherein the nucleic acid is at least one sugar chain gene selected from the group consisting of SLC35B1 gene, CHST9 gene and ST8SIA3 gene A nucleic acid characterized by comprising a base sequence comprising at least 15 consecutive bases in the base sequence or its complementary sequence.
  • a double-stranded RNA for use in a method for preventing, suppressing or treating HBV infection wherein the double-stranded RNA is a sugar chain gene selected from the SLC35B1 gene, the CHST9 gene and the ST8SIA3 gene
  • Double-stranded RNA for use in a method for preventing, suppressing or treating HBV infection, wherein the double-stranded RNA is located at positions 102 to 121 of the base sequence shown in SEQ ID NO: 3, From the region 161 to 179, 311 to 330, 477 to 495, 576 to 594 and 627 to 644, region 806 to 824 of SEQ ID NO: 8, and region 178 to 196 of SEQ ID NO: 10
  • a double-stranded RNA comprising a base sequence comprising at least 15 consecutive bases in any selected region or a complementary sequence thereof.
  • a sugar chain modification activity inhibitor for use in a method for preventing, suppressing or treating HBV infection wherein the sugar chain modification activity inhibitor is associated with at least one sugar chain synthesis of SLC35B1, CHST9 and ST8SIA3 A sugar chain-modifying activity inhibitor characterized by specifically binding to a protein.
  • a peptide or antibody for use in a method for the prevention, suppression or treatment of HBV infection wherein the peptide or antibody specifically binds to at least one glycosylation-related protein of SLC35B1, CHST9 and ST8SIA3 A peptide or antibody characterized by binding.
  • the present invention includes the following aspects.
  • [25] Use of an expression inhibitor of at least one target sugar chain gene selected from the group consisting of SLC35B1 gene, CHST9 gene and ST8SIA3 gene in the manufacture of a pharmaceutical composition for prevention, suppression or treatment of HBV infection.
  • a nucleic acid comprising a base sequence comprising at least 15 consecutive bases in the base sequence of at least one target sugar chain gene selected from the group consisting of SLC35B1 gene, CHST9 gene and ST8SIA3 gene or its complementary sequence Use in the manufacture of a pharmaceutical composition for the prevention, suppression or treatment of HBV infection.
  • RNA in the manufacture of a pharmaceutical composition for prevention, suppression or treatment of HBV infection.
  • [26] Use of a sugar chain modification activity inhibitor of at least one sugar chain synthesis-related protein selected from the group consisting of SLC35B1, CHST9 and ST8SIA3 in the manufacture of a pharmaceutical group for prevention, suppression or treatment of HBV infection.
  • a sugar chain modification activity inhibitor of at least one sugar chain synthesis-related protein selected from the group consisting of SLC35B1, CHST9 and ST8SIA3 in the manufacture of a pharmaceutical group for prevention, suppression or treatment of HBV infection.
  • a sugar chain modification activity inhibitor of at least one sugar chain synthesis-related protein selected from the group consisting of SLC35B1, CHST9 and ST8SIA3 in the manufacture of a pharmaceutical group for prevention, suppression or treatment of HBV infection.
  • the present invention targets the SLC35B1 gene, CHST9 gene or ST8SIA3 gene on the human hepatocyte side, or an expressed protein thereof. Unaffected by mutations, there is almost no possibility of emergence of resistant virus strains. It is also advantageous as a target molecule for inhibiting HBV infection that the sugar chain gene involved in sugar chain modification is localized in the endoplasmic reticulum or Golgi apparatus in hepatocytes.
  • the SLC35B1, CHST9, and ST8SIA3 sugar chain synthesis-related proteins and genes thereof of the hepatocytes specified in the present invention are all necessary for HBV particle formation and secretion, but they are inherent to hepatocytes. Since these are proteins and genes that have little effect on synthesis, it is expected that inhibition of their expression will not impair hepatocyte function and have extremely low side effects.
  • a compound that inhibits the expression of SLC35B1, CHST9 or ST8SIA3 gene expressed in human hepatocytes or a compound that suppresses the activity of SLC35B1, CHST9 or ST8SIA3 related to sugar chain synthesis, in the liver Can suppress the growth and secretion of HBV.
  • Typical examples of the former include siRNA preparations or other RNAi preparations or antisense preparations that can hybridize to a specific region of the SLC35B1 gene, and the latter typical compounds include low-level peptides such as peptides that bind to SLC35B1.
  • molecular compounds and anti-SLC35B1 neutralizing antibodies are molecular compounds and anti-SLC35B1 neutralizing antibodies.
  • a pharmaceutical composition for HBV infection using a known liver-specific siRNA transport system eg, Patent Document 16
  • a system for measuring the amount of S-HBs antigen secreted into the culture supernatant by the tagged S-HBs antigen and the sugar chain content of the secreted S-HBs antigen Effects of Glycosynthesis inhibitors on S-HBs antigen secretion Expression profile of glycan genes in hepatocytes and hepatoma cells (NGS) Expression profile of glycan genes in hepatocytes and hepatoma cells (qRT-PCR) Screening method of target glycan gene by analysis of effects on S-HBs antigen secretion using siRNA plate
  • siRNA plate Example of analysis of effects of siRNA on S-HBs antigen secretion
  • No. 15 is CHST9 siRNA
  • No. 19 is ST8SIA3 siRNA
  • No. 79 is SLC35B1 siRNA 2nd screening result using siRNA plate (RT-PCR, ELISA)
  • SiRNA target site on SLC35B1 In the figure, the underlined part is the target position of siRNA, which are called a, b, c, ..., j in order from the first.
  • the white square (c) shows the target site of siSLC35B1 # 1 and the gray square (e and g) ⁇ ⁇ used in the following figures show the target sites of siSLC35B1 # 2 and siSLC35B1 # 3, respectively.
  • siRNA siRNA a ⁇ j targeting positions a ⁇ j of SLC35B1 (Fig.
  • SLC35B1 siRNA confirmed SLC35B1 expression suppression effect (qRT-PCR) Comparison of the effects of SLC35B1 siRNA (siSLC35B1 # 1) and sugar chain synthesis inhibitor (Tunicamycin) on AFP secretion Effect of SLC35B1siRNA (siSLC35B1 # 1) on glycan synthesis for host cells:
  • # 1 is siSLC35B1 # 1
  • # 2 is siSLC35B1 # 2
  • siC is a control siRNA (manufactured by SIGMA) Comparison of lectin array analysis between untreated group and siRNA (siSLC35B1) treated group.
  • HBV and HBV infection means a virus capable of causing hepatitis B.
  • HBV currently has a genotype from A to H, but HBV to be treated and suppressed in the pharmaceutical composition for HBV infection of the present invention includes all genotypes.
  • “HBV infection” is typically hepatitis B, and is classified into chronic hepatitis, acute hepatitis, and fulminant hepatitis.
  • liver cancers such as cirrhosis, liver fibrosis, and hepatocellular carcinoma are also included as long as they are caused by infection of living organisms including humans with HBV.
  • HBV infection for example, detection of HBs antigen in blood, detection of HBe antigen in blood, measurement of the amount of HBV-DNA in blood and measurement of the amount of HBV DNA polymerase in blood, and This can be determined by a combination of these.
  • treatment of HBV infection refers to elimination of HBV, reduction of symptoms due to infection with HBV, calming of hepatitis, and prevention and reduction of progression from hepatitis to cirrhosis, liver fibrosis and liver cancer. Means. In addition, it may be expressed as "treatment or suppression of HBV infection” in order to clarify that the symptoms and progression of HBV infection are included even before the remission of HBV infection is not achieved. .
  • prevention of HBV infection means preventing the onset of HBV infection such as hepatitis B before or after infection with HBV.
  • HBs antigens are HBV envelope glycoproteins, which start with a methionine (Met) that differs from one HBs gene, and there are three types of L-HBs, M-HBs, and S-HBs antigens depending on their size.
  • HBs antigen undergoes sugar chain modification in the endoplasmic reticulum and Golgi apparatus after translation by intracellular ribosomes.
  • the core (HBc) encapsulating HBV DNA (RNA) is taken up by the HBs antigen in the endoplasmic reticulum and is modified with a sugar chain in the same manner as the HBs antigen to form infectious HBV particles (Schaedler S et al, Viruses.
  • Non-patent Document 1 the amount of S-HBs antigen in the culture supernatant is used as an index for screening for sugar chain synthesis inhibitors, and a S-HBs antigen with a Flag tag for purification added to the N-terminal is used.
  • Full-length DNA SEQ ID NO: 1 was used.
  • the gene is referred to as “sugar chain gene” or “sugar chain synthesis-related protein gene”, and the protein encoded by the gene is referred to as “sugar chain synthesis-related protein”.
  • GGDB glycan related gene databases
  • CAZy Carbohydrate-active enzymes
  • a group of physiologically active proteins such as enzymes and transporters belonging to these glycosyltransferase-related enzyme groups, and 185 types of sugar chain gene groups in which registration numbers of registered base sequences or amino acid sequences are described. Used as a population. Specifically, 185 types of enzymes and transporter genes belonging to the sugar chain synthesis-related proteins grouped in the present invention are listed below (Tables 1 to 8) (Ito H et al, J Proteome Res. 2009 Mar; 8 (3): 1358-67 Supplemental Table 1) is a glycan gene described above (excluding duplicate genes).
  • sugar chain synthesis-related protein essential for HBV particle formation or secretion in human-infected hepatocytes and a method for identifying the gene In the present invention, it is essential for HBV particle formation or secretion in infected hepatocytes.
  • the sugar chain gene was identified by the following procedure. 1) A cDNA library is prepared from total RNA of human hepatocytes (normal hepatocytes and liver cancer-derived cell lines), comprehensively sequenced, and mapped to human cDNA. 2) Extracted from glycan-related gene database (GGDB, Japan) and mapped to “glycan genes”, and compared and analyzed the expression levels of the 185 types of glycan genes listed above (Tables 1-8) To obtain an expression profile.
  • GGDB glycan-related gene database
  • sugar chain genes that are highly expressed or moderately expressed in hepatocytes are extracted, and corresponding to different regions of each gene sequence 3
  • HBV S-HBs antigen cDNA is transfected, and the amount of S-HBs antigen expressed and the amount of sugar chain added to the culture supernatant Are selected, and those showing a decrease in the expression of S-HBs antigen or a decrease in the addition of sugar chains of S-HBs antigen are selected as compared with the case of introduction of control siRNA.
  • each of the three mixed siRNAs is introduced into HBV-producing hepatoma cells, and the amount of HBV DNA and HBs antigen in the culture supernatant is measured.
  • the amount of HBV DNA and HBs antigen Select a reduced one.
  • a known immunological or biochemical analysis method can be applied to the measurement of the amount of HBV DNA, the amount of HBs antigen expressed, the amount of added sugar chain, etc. in the culture supernatant.
  • the sugar chain gene corresponding to the selected siRNA can be identified as a gene essential for HBV particle formation and / or secretion.
  • the gene and the sugar chain synthesis-related protein encoding the gene are the inhibition target molecules of the present invention.
  • SLC35B1, CHST9 and ST8SIA3 genes, and SLC35B1, CHST9 and ST8SIA3 proteins could be provided as inhibition target molecules.
  • SLC35B1 is a protein belonging to solute carrier (SLC) family 35 and cloned as a gene having homology to nucleic acid-sugar transporters (UDP-Gal transporter related protein, UGTrel1, 322aa) (Non-patent Document 6).
  • the human gene is located on chromosome 17 and there are at least 9 exons.
  • mRNA is ubiquitously expressed, and two proteins, 255aa (NP_001265713) and 359aa (NP_005818), are registered.
  • 322aa (P78383: SEQ ID NOs: 3 and 4) whose activity was measured in (Non-patent Documents 6 and 7) using a partial sequence thereof instead of these full-length sequences is used. So far, SLC35B1 homologs have been reported in yeast and nematodes, but their activity as a nucleic acid-sugar transporter has not been confirmed, and there has been no report on their role in HBV secretion or their intracellular functions.
  • the SLC35B1 protein is constitutively working in hepatocytes, and when it is infected with HBV, a sugar chain that is essential for modifying HBV into particles and secreting it outside the cell It was elucidated to be a synthesis-related protein. In addition, it was confirmed that inhibition of the expression of SLC35B1 and its gene in hepatocytes had almost no effect on the sugar chain synthesis function of hepatocytes themselves and there was no cytotoxicity.
  • CHST9 gene and ST8SIA3 gene have been identified as sugar chain genes that have undergone sugar chain modification essential for HBV particleization and secretion outside the cell.
  • CHST9 protein is a sulfotransferase belonging to the Carbohydrate sulfotransferase family
  • ST8SIA3 protein is one of ⁇ 2,8-sialyltransferase (see Taniguchi N et al., Handbook of Glycosyltransferases and Related Genes 2014).
  • expression inhibition mainly targeting the SLC35B1 gene as a typical sugar chain gene is described, but the same applies to the CHST9 gene and the ST8SIA3 gene.
  • Method for controlling expression level of target sugar chain gene or protein activity related to sugar chain synthesis (4-1) Method for determining target region by RNAi in base sequence of target gene HBV particle formation and / or in (3-2) above
  • a highly effective inhibitory target region in a gene sequence identified as a sugar chain gene essential for secretion can be determined as follows.
  • the siRNA target sequence can be determined and searched and designed using commercially available software or a website (such as GENETYX or siDirect http://sidirect2.rnai.jp manufactured by Genetics). In this study, we used 3 types of mixed siRNAs combining siRNAs corresponding to 3 different positions on the base sequence of each glycan gene to determine the inhibition target gene.
  • siRNA was introduced into HBV-infected cells, and the amount of HBV DNA and HBs antigen in the culture supernatant were compared. Identify the region on the gene sequence. This region becomes a target region for inhibiting the expression of the sugar chain gene. Furthermore, by preparing siRNA having a base sequence in the vicinity region and comparing the inhibition ability of each HBV secretion function, a base sequence of a more effective target region can be determined.
  • RNAi preparations that induce RNAi such as shRNA, antisense RNA or DNA that hybridizes to the region, or antisense preparations or ribozyme preparations that prepare ribozymes
  • HBV in hepatocytes infected with HBV Particleization and secretion can be inhibited. Since these oligonucleic acid preparations can directly act on HBV-infected cells in the liver using a liver-specific transport system (Patent Document 16, etc.) described later, treatment of HBV infection and It is effective as a pharmaceutical composition for prevention.
  • RNAi preparation as an oligonucleic acid useful as a pharmaceutical composition for the treatment and prevention of HBV infection of the present invention is the SLC35B1 gene (SEQ ID NO: 3), CHST9 gene (SEQ ID NO: 8) or ST8SIA3 gene (SEQ ID NO: 10) siRNA or shRNA comprising a double-stranded RNA in which at least 15, preferably 17, and more preferably 18 or more consecutive base sequences correspond to a complementary strand corresponding to a specific region in the base sequence of 10) RNAi preparation.
  • RNAi preparations act as SLC35B1 gene, CHST9 gene or ST8SIA3 gene expression inhibitors in HBV-infected cells, so they have excellent pharmaceutical composition for treatment, suppression or prevention of HBV infection It becomes an active ingredient of things.
  • SEQ ID NO: 3 eg
  • CHST9 the amount of HBV DNA among CHST9 # 1 (positions 1188 to 1206), CHST9 # 2 (positions 806 to 824) and CHST9 # 3 (positions 588 to 606) used in this example was determined.
  • ST8SIA3 # 1 (positions 178 to 196) used in this Example ST8SIA3 # 2 (SEQ ID NO: 10) of ST8SIA3 # 1 that significantly reduced both HBs antigen level and HBV DNA level among ST8SIA3 # 2 (positions 354-372) and ST8SIA3 # 3 (positions 208-226)
  • Double-stranded RNA targeting a base sequence containing at least 15, preferably 17, and more preferably 18 consecutive bases in the region from position 178 to 196 corresponding to the position is the same as in the SLC35B1 gene. .
  • RNAi preparations not only RNAi preparations, but also antisense preparations containing the above-mentioned specific base sequences, oligonucleic acid preparations such as nucleic acid aptamers or ribozymes also act as inhibitors of the expression of each sugar chain gene. It becomes an active ingredient of a pharmaceutical composition for prevention.
  • peptides or low molecular weight compounds that specifically bind with the specific region of each sugar chain gene as a target are also active ingredients of a pharmaceutical composition for the treatment and prevention of HBV infection, like double-stranded RNA.
  • the gene is disrupted by genome editing technology (Shibata T, Aburatani H. Nat Rev Gastroenterol Hepatol. 2014 Jun; 11 (6): 340-9.). It is also possible to suppress the function. Furthermore, using the sequence of the promoter region of the target sugar chain gene, the binding peptide to the sequence can be searched by the ChIP method or the like, and the transcription and expression of the gene can be performed using the binding peptide or a transcription factor inhibitor. Can be inhibited.
  • peptides or low molecular weight compounds that specifically bind to the gene expression control region such as the promoter sequence of the SLC35B1 gene, CHST9 gene or ST8SIA3 gene identified in this way also express the target sugar chain gene in HBV-infected cells. Since it inhibits, it becomes an active ingredient of a pharmaceutical composition for the treatment and prevention of HBV infection.
  • neutralizing antibodies, peptides, or other low molecular weight compounds that specifically bind to SLC35B1, CHST9 or ST8SIA3 protein act as activity inhibitors of SLC35B1, CHST9 or ST8SIA3 protein in HBV-infected cells. It becomes an active ingredient of the pharmaceutical composition for treatment and prevention.
  • the peptide sequence may be determined by an evolutionary molecular engineering display technique or the like.
  • random phage display Smith, GP, et al, Chem. Rev. 1997 Apr 1; 97 (2): 391-410.
  • ribosome display mRNA display, cDNA display, etc.
  • Cell-free translation system display technology Lihse PA et al., Curr Opin Drug Discov Devel., 2001 Mar; 4 (2): 198-204, Bradbury AR et al., Nat Biotechnol. 2011 Mar; 29 (3) : 245-54 etc.
  • SLC35B1, CHST9 or ST8SIA3 protein-specific antibodies can be obtained from commercially available products such as Abcam, and can also be obtained by known immunoengineering techniques. Can do.
  • the antibody may be a polyclonal antibody, not a monoclonal antibody, and may be a full-length monoclonal antibody, in addition to an antibody fragment such as a Fab fragment in which the antigen recognition site is conserved, a humanized antibody, Single chain antibodies and the like can also be used.
  • Such antibody production methods are also known to those skilled in the art.
  • a screening system (alpha screen method: Demeulemeester J et al., J Biomol Screen. 2012 Jun; 17 (5): 618-28 that searches for inhibitors that bind to SLC35B1 using regions other than the ER transmembrane domain of SLC35B1. .) And a screening system based on activity (HTS method: Trubetskoy O et al., J Pharm Pharmacol. 2008 Aug; 60 (8): 1061-7.) In addition, low molecular weight compounds that can inhibit the activity can be screened.
  • composition for preventing, suppressing or treating HBV infection of the present invention (5-1) Method for delivering drug to hepatocytes or their endoplasmic reticulum Sugar chain synthesis-related protein selected from SLC35B1, CHST9 and ST8SIA3 of the present invention
  • a sugar chain gene expression inhibitor such as an oligonucleic acid inhibitor or an oligonucleic acid (hereinafter also referred to as a sugar chain modification inhibitor)
  • the sugar chain modification inhibitor is made into HBV particles and secreted. It is efficient to directly deliver to and inhibit the endoplasmic reticulum (ER) or Golgi apparatus where proteins essential for sugar chain synthesis are localized.
  • an intracellular delivery system (WO2008 / 088581) for an antiviral agent administered by encapsulating in a pH-sensitive liposome containing phosphatidylinositol (PI) lipid can be applied.
  • a liver-specific transport system (Patent Document 16, etc.), or a cationic composed of a glycerol derivative and a phospholipid.
  • Techniques such as WO99 / 48531 that are complexed with liposomes and accumulated in the liver region by setting the average particle size to 100 nm or more can also be used.
  • a method of complexing with a hydrophobically modified HBV preS-derived peptide (WO2009 / 092612) can also be applied.
  • gene therapy approaches including the use of conventionally used non-pathogenic viruses, modified viral vectors such as AAV, and delivery by nanoparticles or liposomes can also be used.
  • modified viral vectors such as AAV
  • delivery by nanoparticles or liposomes can also be used.
  • oligonucleic acid preparations such as siRNA
  • the delivery system for oligonucleic acid preparations has been actively researched and developed for each target organ to be delivered (Kanasty R et al. Nat Mater. 2013 Nov; 12 (11): 967 -77., Haynes M, et al. Drug Deliv Transl Res. 2014 Feb; 4 (1): 61-73.), And clinical trials in which siRNA preparations were actually administered to the liver were also reported ( Coelho T et al. N Engl J Med. 2013 Aug 29; 369 (9): 819-29.), These methods can be applied as appropriate.
  • the dosage of oligonucleic acid by intravenous injection is 0.01 to 1.0 mg / kg.
  • compositions of the present invention comprises an oligonucleic acid such as siRNA or shRNA as an active ingredient, and optionally a pharmaceutically acceptable carrier and / or excipient.
  • oligonucleic acid such as siRNA or shRNA
  • a pharmaceutically acceptable carrier and / or excipient for the treatment and prevention of HBV infection.
  • the oligonucleic acid as the active ingredient may be incorporated into a known liver-specific deliverable vector.
  • a liposome delivery method such as a pH-sensitive liposome (WO2008 / 088581) that can be delivered specifically in the endoplasmic reticulum is also preferably used.
  • formulation of oligonucleic acid such as siRNA (dsRNA) and specific administration methods, dosages, etc. will be mainly described.
  • the present invention is not limited to oligonucleic acid, and as a target sugar chain synthesis-related protein activity inhibitor. It can also be applied to low molecular compounds such as working peptides, antibodies, and the
  • the route of administration may be either oral or parenteral, and in the case of parenteral administration, it is preferably subcutaneous or intravenous injection, by nasal administration such as nasal spray, transdermal administration, inhalation, suppository, etc. There may be.
  • the siRNA can be administered to a patient by intravenous injection, subcutaneous injection, oral delivery, liposome delivery or intranasal delivery and then accumulate in the patient's whole body, liver, or endoplasmic reticulum in hepatocytes.
  • “Pharmaceutically acceptable carriers and / or excipients” are known to those skilled in the art, and include any type of encapsulating material such as non-toxic solid, semi-solid or liquid fillers, diluents, liposomes or Contains formulation aids.
  • compositions for parenteral injection include pharmaceutically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, as well as sterile injectable solutions just before use. Or it may contain a sterile powder for reconstitution into a dispersion.
  • suitable aqueous and non-aqueous carriers, diluents, solvents or vehicles are water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, etc.), carboxymethylcellulose and suitable mixtures thereof, vegetable oils (such as olive oil) A), as well as injectable organic esters such as ethyl oleate.
  • Appropriate fluidity can be maintained by blending a coating material such as lecithin and a surfactant.
  • adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents, antibacterial and antifungal agents, isotonic agents such as sugars and sodium chloride may be included.
  • a depot injectable formulation is prepared by forming a microcapsule matrix with a biodegradable polymer such as polylactide-polyglycolide or encapsulating the pharmaceutical composition in liposomes or microemulsions that are compatible with body tissue be able to.
  • Injectable formulations are made sterile by, for example, filtration through a bacteria-retaining filter or by incorporating a sterilizing agent in the form of a sterile solid composition that can be dissolved or dispersed in sterile water or other sterile injectable medium immediately before use. Can be done.
  • Solid dosage forms for oral administration include, but are not limited to, capsules, tablets, pills, powders and granules.
  • the active compound comprises at least one component pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and / or (a) starch, lactose, sucrose Fillers or extenders such as glucose, mannitol and silicic acid, (b) binders such as, for example, carboxymethylcellulose, alginate, polyvinylpyrrolidone, sucrose and gum arabic, (c) wetting agents such as glycerol, (d ) Disintegrants such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate, (e) solution retarders such as paraffin, and (f) absorption enhancers such as quaternary ammonium compounds.
  • a component pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and / or
  • starch
  • G wetting agents such as acetyl alcohol and glycerol monostearate, (h) Mixed with absorbents such as phosphorus and bentonite clays and (i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate and mixtures thereof.
  • absorbents such as phosphorus and bentonite clays
  • lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate and mixtures thereof.
  • the dosage forms may also contain buffering agents.
  • Solid dosage forms of tablets, dragees, capsules, pills and granules are prepared with coatings such as enteric coatings and other coatings well known in the pharmaceutical formulating art. Lactose and high molecular weight polyethylene glycol may be used to form soft and hard filled gelatin capsules.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs.
  • liquid dosage forms can solubilize the agent, eg, inert diluents commonly used in the art such as water or other solvents, as well as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate , Benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oil (especially cottonseed, peanut, corn, germ, olive, castor and sesame oil), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycol And emulsifiers such as fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as water or other solvents
  • ethyl alcohol isopropyl
  • oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents, the suspension containing the active compound.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents, the suspension containing the active compound.
  • suspensions may be included such as, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sulfitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar and tragacanth, and mixtures thereof.
  • Compositions for oral administration include compressed gases such as nitrogen or liquefied gas propellants containing liquid or solid nonionic surfactants to the extent that the active ingredient does not dissolve, or solid anionic surfactants. But you can.
  • liposome delivery methods such as pH-sensitive liposomes (WO2008 / 088581) containing phosphatidylinositol (PI) lipids and cationic liposomes (WO99 / 48531 etc.) that can be delivered specifically to the liver are applied.
  • PI phosphatidylinositol
  • cationic liposomes WO99 / 48531 etc.
  • liposomes are generally drawn from phospholipids or other lipid substances. Liposomes are formed by mono- or multi-lamellar hydrated liquid crystals that are dispersed in an aqueous medium.
  • any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used.
  • the present composition in liposome form may contain stabilizers, preservatives, excipients and the like in addition to the compound of the present invention.
  • Preferred lipids are both natural and synthetic phospholipids and phosphatidylcholines (lecithins). Methods for forming liposomes are known in the art (see, eg, Prescott, Meth. Cell Biol. 14: 33-34 (1976)).
  • the “therapeutically effective” amount of the pharmaceutical composition for preventing, suppressing or treating HBV infection of the present invention is used to prevent and / or treat liver diseases or liver disorders caused by various HBV infections such as hepatitis B.
  • the agent can be administered to a patient in need of treatment for HBV infection as a pharmaceutical composition in combination with one or more pharmaceutically acceptable excipients.
  • the therapeutically effective dose level varies depending on the age, weight, gender, etc. of the patient as well as the subtype of HBV, the degree of infection, the degree of symptoms, and the dosage form.
  • oligonucleic acid it is preferably in the range of 0.01 to 1.0 mg. In the case of other compounds such as antibodies, the dose is preferably 2.5 to 10 mg / kg. In some cases, a dose lower than the above range may be sufficient, and conversely, a dose exceeding the above range may be required. When administering a large amount, it is desirable to divide the dose into several times a day.
  • the target sugar chain synthesis-related protein activity inhibitor and target sugar chain gene expression inhibitor of the present invention may be used in combination with or in combination with other anti-HBV agents. That is, the present invention includes an activity inhibitor of a target sugar chain synthesis-related protein selected from SLC35B1, CHST9, and ST8SIA3 or an expression inhibitor of a target sugar chain gene selected from SLC35B1, CHST9 gene, and ST8SIA3 gene and another 1 Also provided is a pharmaceutical composition for treating or preventing HBV infection, comprising a combination of two or more anti-HBV agents. When other anti-HBV agents are combined, they can be administered at the same time, separately or sequentially, so they may be included in the same pharmaceutical composition, and other anti-HBV agents may be used separately.
  • the dosage forms of two or more separate preparations may be the same or different.
  • any one or more may be a parenteral preparation, an injection, an infusion, or an intravenous infusion.
  • interferon interferon
  • nucleic acid analogs acting that has a nucleic acid partial structure in the molecule and inhibits HBV DNA synthesis
  • the generic name of compounds having Examples of interferon (IFN) include IFN- ⁇ or IFN- ⁇ preparations, and examples of nucleic acid analogs include lamivudine [LMV], adefovir [ADV], entecavir [ETV], and the like.
  • the interferon preparation is preferably a PEGylated preparation.
  • HBV secretion inhibitor screening method and kit for the same The present invention also provides a screening method for searching for HBV particle formation and / or HBV secretion inhibitor as a therapeutic agent for HBV infection.
  • the search target of the present invention is sometimes simply referred to as “HBV secretion inhibitor”.
  • One of the methods for screening for the HBV secretion inhibitor of the present invention is a method for searching for a substance that inhibits the transcription / expression of SLC35B1 gene, CHST9 gene or ST8SIA3 gene expressed in cells.
  • a test substance may be administered intracellularly or in its medium, and the presence or absence of a decrease in the expression level of the SLC35B1, CHST9, or ST8SIA3 gene in the hepatocytes may be observed. At that time, changes in the amount of mRNA can also be observed by RT-PCR, etc., but generally the amount of expression product SLC35B1, CHST9 or ST8SIA3 is measured by an immunological technique such as ELISA or immunoblot, and the expression level is determined. Select the test substance to be reduced.
  • At least one gene selected from SLC35B1, CHST9 and ST8SIA3 genes was introduced and overexpressed It is preferable to use transformed cells.
  • more efficient measurement is possible by tagging the SLC35B1 to be expressed and the like to obtain a tagged target sugar chain synthesis-related protein (eg, Flag-SLC35B1).
  • a tagged target sugar chain synthesis-related protein eg, Flag-SLC35B1
  • the inhibitory effect of the constructed siRNA can be measured, and a more effective target region can be determined.
  • tag sequences such as a His tag, Myc tag, GFP tag, GST tag, Maltose tag, and S tag can be used.
  • Antibodies that recognize each of these tag sequences are known and are sold as commercial kits, so the amount of expressed target sugar chain synthesis-related protein can be measured as the amount of anti-tag antibodies. is there.
  • An enzyme label includes a conjugate of an enzyme and a molecule of interest, such as a polypeptide, and can be detected by either colorimetric, spectrophotometric or fluorescent spectrophotometric methods.
  • the expression level of the target sugar chain synthesis-related protein can also be changed by using a transformed cell with a fusion of the gene encoding the labeled protein and the target sugar chain gene. It can be measured efficiently.
  • the method for screening for an HBV secretion inhibitor in the present invention can be described as follows, for example. Using a transformed cell that expresses at least one labeled sugar chain synthesis-related protein selected from SLC35B1, CHST9 and ST8SIA3 directly or indirectly labeled on the surface of the endoplasmic reticulum or Golgi membrane, Measuring the amount of labeling on the surface of the intracellular membrane, and administering a test compound in the cell or in the medium, Selecting a compound that reduces the amount of labeling, A method of screening for an HBV secretion inhibitor.
  • a transformed cell that expresses at least one labeled sugar chain synthesis-related protein selected from SLC35B1, CHST9 and ST8SIA3 directly or indirectly labeled on the surface of the endoplasmic reticulum or Golgi membrane Measuring the amount of labeling on the surface of the intracellular membrane, and administering a test compound in the cell or in the medium, Selecting a compound that reduces the
  • the transformed cell is a cell expressing a sugar chain synthesis-related protein tagged with a Flag-tag or the like, and the intracellular membrane surface is fluorescently labeled with an anti-tag antibody such as a fluorescently labeled anti-Flag antibody.
  • the labeling amount of the intracellular membrane surface to be measured is a fluorescence amount because it can be visualized as a decrease in the fluorescence amount.
  • the test substance at that time may be a nucleic acid such as an oligonucleic acid or a low molecular weight compound such as a peptide. Therefore, the administration method may be introduced into a transformed cell or may be administered in a medium.
  • the kit for screening the HBV secretion inhibitor in that case can be expressed as a kit containing the following (1) and (2).
  • the second method of screening for the HBV secretion inhibitor of the present invention is a screening method focusing on the inhibition of the activity of at least one sugar chain synthesis-related protein selected from SLC35B1, CHST9 and ST8SIA3.
  • the method comprises the step of measuring the activity of at least one sugar chain synthesis-related protein selected from SLC35B1, CHST9 and ST8SIA3 in cultured hepatocytes, and introducing a test oligonucleic acid or test compound into the cell or You may select the test substance which reduces the said active amount by administering in a culture medium.
  • the activities of SLC35B1, CHST9, and ST8SIA3 that can be used as indices in this case are the activity of transporting a sugar chain-nucleotide such as UDP-Gal, the activity of transferring a sulfate group to the 4-position of GalNAc, and the activity of sialic acid 8 The activity of transferring sialic acid to the position.
  • a reaction solution containing UDP-Gal which is a sugar chain-nucleotide having UDP or labeled UDP, or measuring the concentration of UDP-Gal in the free or endoplasmic reticulum.
  • the released UDP concentration may be measured with a UDP detection reagent or by the methods described in Non-Patent Documents 6 and 7.
  • the HTS method (Trubetskoy O et al., J Pharm Pharmacol. 2008 Aug; 60 (8): 1061-7.), which is a known screening system using activity as an index, can also be applied.
  • a peptide sequence that binds to a target glycosylation-related protein such as SLC35B1 is determined by an evolutionary molecular engineering display technique. Typically, random phage display (Smith, GP, et al, Chem. Rev. 1997 Apr 1; 97 (2): 391-410.) Can be applied, but ribosome display, mRNA display, cDNA display, etc. Cell-free translation system display technology (Lohse PA et al., Curr Opin Drug Discov Devel., 2001 Mar; 4 (2): 198-204, Bradbury AR et al., Nat Biotechnol. 2011 Mar; 29 (3) : 245-54 etc.) can also be applied.
  • Kits for this purpose include: (1) Beads on which either SLC35B1, CHST9, or ST8SIA3 sugar chain synthesis-related proteins are immobilized, substrates such as plates, and random phage display for application to random phage displays.
  • Peptide libraries containing nucleic acids containing sequences or corresponding proteins can be used.
  • the parent base sequence used as a template in this case is preferably a synthetic library of M13 or T7 phage, and the length of the random base sequence is usually 21 to 32 bases (7 to 12 amino acids).
  • anti-SLC35B1 neutralizing antibodies can be obtained by known immunoengineering techniques.
  • An inhibitor that binds using a region other than the ER transmembrane domain of SLC35B1 (Fig. 27) can be screened for low molecular weight compounds that can bind to and inhibit the activity of glycosylation-related proteins.
  • an inhibitor that inhibits binding to a probe such as an antibody or an inhibitor that inhibits binding to a substrate can be obtained.
  • Example 1 Establishment of HBs antigen-secreting hepatic cell line (1-1) Preparation of plasmid for secreting HBs antigen (membrane protein on HBV) L-HBs antigen (389aa), an HBV antigen derived from three types of HBV envelopes ), M-HBs antigen (281 aa), S-HBs antigen (226 aa) DNA (Accession # is AB246345) was amplified by PCR and incorporated into pcDNA3.1 (Life Technologies).
  • the DNA encoding the full length of the S-HBs antigen is used for the following experiments, and for easy detection, a secretory expression vector in which a signal peptide and a purification Flag tag are added to the N-terminus. Recombined into pFLAG-CMV3 (manufactured by SIGMA).
  • the DNA and amino acid sequences of the tagged S-HBs antigen are shown as SEQ ID NOs: 1 and 2.
  • (1-2) Expression of HBs antigen in hepatoma cells Using the secretory expression vector prepared in (1-1) above, S-HBs were obtained from hepatoma cell-derived HuH7 cells (obtained from RIKEN BioResource Center). The antigen was expressed and purified. Specifically, the expression vector (2 ⁇ g) containing the S-HBs antigen DNA was transfected into HuH7 cells (about 2 ⁇ 10 6 cells) using Lipofectamine LTX (manufactured by Life Technologies), and 48 hours later. The culture supernatant was collected, and the recombinant HBs antigen was adsorbed using anti-FLAG antibody beads (manufactured by SIGMA).
  • Elution from the anti-FLAG antibody was performed in two steps. First, competitive elution was performed with the FLAG peptide, and then SDS-PAGE sample buffer was added, followed by elution and recovery by heating at 100 ° C. The recovered recombinant HBs antigen was developed by SDS-PAGE and Western blotted with an anti-FLAG antibody. As a result, by adding a secretory signal sequence to the N-terminal side of the S-HBs antigen, approximately 10 times the amount of purified S-HBs compared to when expressing only the S-HBs antigen without a secretory signal. It became clear that the antigen could be recovered.
  • Example 2 Effect on secretion of HBs antigen due to inhibition of sugar chain synthesis system
  • Example 3 Extraction of sugar chain genes expressed in hepatocytes and hepatoma cells Normal cells derived from human liver (primary culture, Clonetics TM Human Hepatocyte, hNHEPS TM Cells: LONZA Inc.) and hepatoma cells Total RNA was prepared from HuH7 cells and HepG2 cells (obtained from RIKEN BioResource Center), and the expression level of glycan gene in each cell was analyzed by qRT-PCR (glycan gene qPCR array system) and transcript Measured by tome analysis (next-generation sequencer).
  • RNA libraries and cDNA libraries were prepared using Ion Total RNA Seq Kit v2 (Life Technologies) and sequenced using IonPGM (Life Technologies). The sequence of millions of reads obtained by one-time sequencing was mapped to human cDNA, and only those mapped to sugar chain genes were extracted and the expression levels were compared and analyzed (FIG. 3A).
  • RNA was purified from primary cultured normal hepatocytes, HepG2 cells, and HuH7 cells using RNeasy Plus Mini Kit (QIAGEN). Using 4 ⁇ g of total RNA, cDNA was prepared with QuantiTect Reverse Transcription Kit (manufactured by QIAGEN), and qRT-PCR array system (Ito H et al) containing the 185 types of sugar chain genes described above (Tables 1 to 8). , J Proteome Res. 2009 Mar; 8 (3): 1358-67.), The expression level of the sugar chain gene expressed in hepatocytes was measured. As a result of sugar chain gene expression analysis, expression profiles of about 185 kinds of sugar chain genes in two types of liver cancer cell lines (HuH7 cells and HepG2 cells) were obtained (FIG. 3B).
  • Example 4 Screening of glycan genes affecting the glycan modification of S-HBs antigen (4-1) Preparation of siRNA plate Based on the expression profiles of the 185 types of glycan genes obtained in Example 3, The glycan genes in hepatocytes were divided into two groups, a “high expression-medium expression” group and a “low expression-no expression” group. Subsequently, 3 types of siRNA (manufactured by AB / Life Technologies) were obtained for 86 sugar chain genes included in the “high expression to medium expression” group, and 3 types of mixed siRNAs were prepared for each gene.
  • siRNA manufactured by AB / Life Technologies
  • 10-well is coated with 100 pmol / well of each siRNA, and the other wells are made with 100 pmol of control siRNA (manufactured by SIGMA) and blank, and then air-dried. Stored at -80 ° C until use.
  • S-HBs antigen cDNA was transfected with Lipofectamine LTX, and 48 hours later, S-HBs antigen was recovered from the supernatant with anti-Flag antibody beads (manufactured by SIGMA) and S-HBs was obtained by Western blotting.
  • the amount of antigen expression and the addition of sugar chains were confirmed (FIG. 4A).
  • 10 or more genes were found to have reduced S-HBs antigen expression or reduced sugar chain addition compared to control siRNA. The typical case is illustrated in FIG. 4B.
  • siRNA screening in HepG2.2.15.7 cells 5-1st screening Among the sugar chain genes expressed in hepatocytes, screening was performed for genes that affect the secretion of HBV particles.
  • HepG2.2.15.7 cells distributed by the National Institute of Infectious Diseases
  • the culture medium was changed the next day. Three days later, the amount of HBV DNA secreted into the supernatant was measured by RT-PCR (Roche LightCycler 96 real-time PCR system) (FIG. 5A).
  • RT-PCR conditions used for measurement of the amount of HBV DNA are as follows.
  • Primer 1 5'-CTTCATCCTGCTGCTATGCCT-3 '(SEQ ID NO: 5)
  • Primer 2 5'-AAAGCCCAGGATGATGGGAT-3 '(SEQ ID NO: 6)
  • Probe FAM-ATGTTGCCCGTTTGTCCTCTAATTCCAG-TAMRA (SEQ ID NO: 7)
  • Reagent Eagle Taq Master Mix (Roche)
  • the amount of HBs antigen was quantified by ELISA using a specific antibody (manufactured by Special Immunology Institute) (FIG. 5A).
  • an MTT assay (Roche) was performed to confirm the influence on cell proliferation.
  • SLC35B1 (SEQ ID NOs: 3 and 4) is one of nucleic acid-sugar transporter-related genes, and homologs have been reported in yeast and nematodes, but the nucleic acid-sugar transporter activity has not been confirmed. Also, the role in HBV secretion and the function in the cell are unknown.
  • CHST9 (SEQ ID NOs: 8 and 9) is a sulfotransferase belonging to the Carbohydrate sulfotransferase family, and adds a sulfate group at the 4-position of GalNAc.
  • ST8SIA3 (SEQ ID NOs: 10 and 11) is one of ⁇ 2,8-sialyltransferase (sialyltransferase), and further adds sialic acid to the position of the 8-position of sugar chain sialic acid (NeuAc) on glycoprotein. . Subsequently, 2nd screening was performed for these sugar chain genes.
  • siRNA-SLC35B1 # 2 also confirmed the effect of decreasing the amount of HBV DNA
  • siRNA-SLC35B1 # 3 confirmed the effect of decreasing both the amount of HBs antigen and the amount of HBV DNA.
  • SLC35B1 # 1 corresponds to the position of c (positions 311 to 330 of SEQ ID NO.
  • SLC35B1 # 2 corresponds to e (positions 477 to 495)
  • SLC35B1 # 3 corresponds to position g (positions 576 to 594).
  • the regions b, d, f, and h to j are also candidate target regions. As will be described later, it can be said that the regions a to c, e, g, and i are particularly effective.
  • CHST9 # 1 positions 1188 to 1206 of SEQ ID NO: 8
  • CHST9 # 2 positions 806 to 824
  • CHST9 # 3 positions 588 to 606
  • HBV secretion inhibitory effect of siRNA-SLC35B1 (6-1) Comparison between siRNA-SLC35B1 # 1 and entecavir The HBV secretion inhibitory effect of siRNA-SLC35B1 # 1 selected in the above (5-2) It was measured using HepG2.2.15.7 cells and compared with nucleic acid analogs (reverse transcriptase inhibitors) entecavir (Abcam) and lamivudine (SIGMA), which are known to be effective as HBV therapeutics.
  • nucleic acid analogs reverse transcriptase inhibitors
  • entecavir Abcam
  • SIGMA lamivudine
  • Entecavir and lamivudine were added to 10 ⁇ M and 100 ⁇ M, and SLC35B1 siRNA-SLC35B1 # 1 to # 3 were added to 100 nM to HepG2.2.15.7 cells, respectively.
  • FIG. 8 shows that both entecavir and lamivudine have the effect of reducing the amount of HBV DNA, they did not significantly affect the amount of HBs antigen secreted into the culture supernatant.
  • siRNA-SLC35B1 # 1 showed the effect of reducing both the amount of HBV DNA and the amount of HBs antigen to 50% or less.
  • Primer 3 5'-GGGAGCTCTGGGAGTTCTTGA-3 '(SEQ ID NO: 13)
  • Primer 4 5'-GCCCAAAGAGCAGGATGTTATAGA-3 '(SEQ ID NO: 14)
  • Reagent Taq Man Mix (Roche)
  • HuH7 cells were transfected with siRNA (siRNA aj) corresponding to knockout target candidate regions aj (Fig. 7) of SLC35B1 gene calculated using various software, and each SLC35B1 expression level was measured did.
  • the efficiency of siRNA at that time was calculated with the measured value when transfection of siRNA-negative control (siC, SIGMA) was taken as 100%.
  • the amount of ⁇ -actin in each cDNA was used as an endogenous control (FIG. 9).
  • the amount of SLC35B1 mRNA was reduced to about 10% by siRNA-SLC35B1 # 1 transfection compared to cells transfected with siRNA-negative control. Similarly, it decreased to about 27% in HuH7 cells.
  • Example 7 Toxicity test of siRNA-SLC35B1 (7-1) Effect of siRNA-SLC35B1 on cells According to the MTT assay in the screening process of (Example 5), even if siRNA-SLC35B1 is used in a mixture of 3 types, although it has already been confirmed that the cytotoxicity to cells is low, in this example, individual siRNA preparations were further used, and the effects on the overall sugar chain synthesis function in hepatocytes were mainly observed.
  • HuH7 cells (approximately 2 ⁇ 10 5 cells) were transfected with 100 nM siRNA-SLC35B1 # 1 at 12-well, and 48 hours later, the cells were treated with RIPA buffer (50 mM Tris-HCl pH 8.0, 150 mM NaCl, 0.5% Sodium). The protein was recovered by dissolving with deoxycholate, 0.1% SDS, 1% NP40). A soluble fraction was prepared after centrifugation, and the protein was developed by SDS-PAGE and blotted on a PVDF membrane.
  • RIPA buffer 50 mM Tris-HCl pH 8.0, 150 mM NaCl, 0.5% Sodium
  • HuH7 cells were treated with tunicamycin (manufactured by Wako Pure Chemical Industries, Ltd.) for 48 hours to detect ⁇ -fetoprotein (AFP), a glycoprotein serving as a liver cancer marker.
  • AFP ⁇ -fetoprotein
  • the decrease in AFP expression as observed with tunicamycin was not observed when siRNA-SLC35B1 # 1 was used (FIG. 11).
  • siRNA-SLC35B1 # 1 was transfected into HepG2 cells and HuH7 cells to prepare membrane fractions, and lectin array analysis was performed. As a result of statistical analysis based on the three measurement results, it was confirmed that no significant change was observed in the sugar chain profile (FIGS. 13A and 13B).
  • siRNA-SLC35B1 # 1 treatment In order to examine the effect of siRNA-SLC35B1 # 1 treatment on ER stress and cytotoxicity, we compared HuH7 cells with siRNA transfected with those treated with Tunicamycin (TM) and Brefeldin A (BreA) by Western analysis. In the case of siRNA-SLC35B1 # 1 treatment, increased expression of BiP, which is an ER stress marker, was not observed compared with TM and BreA, which are sugar chain synthesis inhibitors. (Fig.
  • siRNA-SLC35B1 # 1 was transfected into HepG2.2.15.7 cells and cytotoxicity assay was performed using LDH cytotoxicity detection kit (manufactured by Takara Bio Inc.) to confirm that siRNA-SLC35B1 # 1 did not adversely affect the cells. (FIG. 15).
  • siRNA-SLC35B1 # 1 To verify the effect of siRNA-SLC35B1 # 1, off-target searches were analyzed from gene expression from three different experiments. HuH7 cells were transfected with siRNA-SLC35B1 # 1 at 100 nM (Experiment 1), 10 nM (Experiment 2), and HepG2.2.15.7 cells were transfected with 10 nM (Experiment 3), and total RNA was prepared using the RNeasy Plus Mini Kit. Transcriptome analysis was performed. The genes whose expression levels were changed 3 times or more in all experiments were listed as off-targets in comparison with the control (-siRNA) (FIG. 18A).
  • PLC / PRL / 5 cells secreting HBs antigen were transfected with siRNA-SLC35B1 # 1 at 1, 10, 50 nM, and the expression levels of SLC35B1 gene and HBV (HBs antigen gene) were analyzed by qRT-PCR (Fig. 19). It was confirmed that the number of siRNA-SLC35B1 # 1 off-target genes was relatively small and the expression level of the HBs antigen gene was also decreased. From the above results, it was confirmed from the viewpoint of gene expression that there were few side effects when targeting SLC35B1.
  • Example 8 Confirmation of effective siRNA-SLC35B1 concentration
  • HuH7 cells were transfected with siRNA-SLC35B1 # 1 at concentrations of 100, 10, 1, 0.1 nM, and after 48 hours, total RNA was prepared ( QRT-PCR was performed in the same manner as in 6-2) to measure the amount of SLC35B1 mRNA, and the siRNA concentration having a knockdown effect was analyzed. The knockdown efficiency of siRNA was compared with the measured value when siRNA-negative control was transfected as 100%. The amount of ⁇ -actin in each cDNA was corrected as an endogenous control (FIG. 20).
  • siRNA-SLC35B1 when HepG2.2.15.7 cells were immunostained with HBs antigen using anti-HBs antigen antibody (Abcam) and Alexa Fluor (R) 488-anti-horse IgG goat antibody (Jackson ImmunoResearch Laboratories), siRNA-SLC35B1 It was confirmed that the amount of HBs antigen was decreased by # 1 treatment (FIG. 23). As a result, it was found that the HBs antigen amount was suppressed to 1/8 or less at an siRNA concentration of 10 nM and decreased to 1/4 or less at 5 nM. The above results indicate that 1-10 nM siRNA-SLC35B1 # 1 is sufficient to reduce the amount of SLC35B1 mRNA, and inhibits HBV secretion in a concentration-dependent manner at 5 nM or more. Yes.
  • HBV secretion inhibitory effect of siRNA-SLC35B1 (PHH cells) (9-1) Comparison between siRNA-SLC35B1 # 1 and Entecavir The inhibitory effect of siRNA-SLC35B1 # 1 on HBV secretion was measured using human chimeric liver cells (PHH, manufactured by PhoenixBio), and the nucleic acid analog agent entecavir (1 ⁇ M) (FIG. 24).
  • siRNA-SLC35B1 # 1 reduced the amount of HBV DNA as did entecavir.
  • siRNA-SLC35B1 # 1 is considered to be able to be used in combination with a nucleic acid analog agent because it is reduced more than entecavir alone (FIG.
  • Example 10 Effect of siRNA-SLC35B1 on Entecavir-Resistant Strain (10-1) Assay with Entecavir-Resistant Strain It is known that a resistant strain is produced in HBV by using a nucleic acid analog agent. It was measured whether siRNA-SLC35B1 had an inhibitory effect on secretion of entecavir-resistant HBV. The S202G mutant strain was used as an entecavir resistant strain. HuH7 cells were transfected with HBV DNA (genotype C or S202G, obtained from Nagoya City University), and treated with siRNA-SLC35B1 and entecavir two days later.
  • HBs antigen secreted into the supernatant was measured by ELISA and the amount of HBV DNA was measured by RT-PCR (FIG. 25).
  • the amount of HBs antigen was not affected by genotype and decreased in the presence of siRNA-SLC35B1 (FIG. 26A).
  • the amount of HBV DNA was also suppressed when siRNA-SLC35B1 was present (FIG. 26B). That is, the effect of siRNA-SLC35B1 to inhibit secretion of entecavir-resistant HBV was confirmed.
  • the SLC35B1 gene is The effectiveness of drug expression inhibitors or SLC35B1 protein activity inhibitors such as RNAi preparations including siRNA as a target was confirmed.
  • Example 11 Confirmation of intracellular localization of SLC35B1
  • the transmembrane domain is shown in bold, and the signal for localization in the ER is underlined.
  • the inhibitory effect of the constructed siRNA can be visualized as a decrease in the amount of fluorescence corresponding to the expression level of SLC35B1-Flag, and quantitative measurement can be performed, so that a more effective target region can be determined. .
  • it can be used for screening of low molecular weight compounds such as peptides that bind to SLC35B1 and other inhibitors that inhibit SLC35B1 activity (FIG. 28).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Wood Science & Technology (AREA)
  • Cell Biology (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • Hematology (AREA)
  • General Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • Zoology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pathology (AREA)
  • Communicable Diseases (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Food Science & Technology (AREA)
  • Analytical Chemistry (AREA)

Abstract

L'invention concerne : un procédé pour supprimer la formation de particules et/ou la sécrétion de HBV dans des hépatocytes infectés ; un procédé pour traiter ou supprimer une infection par HBV par ce dernier; et une composition médicinale pour ce dernier. La présente invention est caractérisée par le fait qu'un inhibiteur de l'expression d'au moins un gène de chaîne de sucre choisi parmi le gène SLC35B1, le gène CHST9 et le gène ST8SIA3, ou un inhibiteur d'activité de modification de chaîne de sucre d'au moins une protéine liée à la synthèse de chaîne de sucre choisi parmi SLC35B1, CHST9 et ST8SIA3, est utilisé comme suppresseur de sécrétion de HBV à partir d'hépatocytes infectés par le HBV ou un inhibiteur de formation de particules de HBV. Le suppresseur mentionné ci-dessus d'expression de gène de chaîne de sucre ou l'inhibiteur d'activité de modification de chaîne de sucre d'une protéine liée à la synthèse de chaîne de sucre peut être utilisé, seul ou en combinaison, comme composition médicinale pour prévenir et traiter une infection par HBV.
PCT/JP2016/062225 2015-04-16 2016-04-18 Inhibiteur de sécrétion de virus d'hépatite b WO2016167369A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2017512609A JP6566368B2 (ja) 2015-04-16 2016-04-18 B型肝炎ウイルス分泌阻害剤

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2015084520 2015-04-16
JP2015-084520 2015-04-16

Publications (1)

Publication Number Publication Date
WO2016167369A1 true WO2016167369A1 (fr) 2016-10-20

Family

ID=57126591

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2016/062225 WO2016167369A1 (fr) 2015-04-16 2016-04-18 Inhibiteur de sécrétion de virus d'hépatite b

Country Status (2)

Country Link
JP (1) JP6566368B2 (fr)
WO (1) WO2016167369A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019235584A1 (fr) 2018-06-06 2019-12-12 国立研究開発法人産業技術総合研究所 Procédé permettant d'induire de manière efficace un anticorps contre le virus de l'hépatite, anticorps et système de détection
CN113476607A (zh) * 2021-08-04 2021-10-08 中南大学湘雅二医院 Slc12a5及其抑制剂的应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120009202A1 (en) * 2010-07-07 2012-01-12 University Of Manitoba Target host factors for treating viral infection
JP2013501038A (ja) * 2009-08-07 2013-01-10 トランジェーヌ、ソシエテ、アノニム Hbv感染を治療するための組成物
JP2013507933A (ja) * 2009-10-16 2013-03-07 グラクソ グループ リミテッド Hbvアンチセンス阻害剤
JP2014527404A (ja) * 2011-07-12 2014-10-16 トランジェーヌ、ソシエテ、アノニムTransgene S.A. Hbvポリメラーゼ変異体

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013501038A (ja) * 2009-08-07 2013-01-10 トランジェーヌ、ソシエテ、アノニム Hbv感染を治療するための組成物
JP2013507933A (ja) * 2009-10-16 2013-03-07 グラクソ グループ リミテッド Hbvアンチセンス阻害剤
US20120009202A1 (en) * 2010-07-07 2012-01-12 University Of Manitoba Target host factors for treating viral infection
JP2014527404A (ja) * 2011-07-12 2014-10-16 トランジェーヌ、ソシエテ、アノニムTransgene S.A. Hbvポリメラーゼ変異体

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BLOCK, T. M. ET AL.: "Secretion of human hepatitis B virus is inhibited by the imino sugar N-butyldeoxynojirimycin", PNAS, vol. 91, no. 6, 1994, pages 2235 - 2239, XP000952542 *
ITO, H. ET AL.: "Strategy for Glycoproteomics: Identification of Glyco-Alteration Using Multiple Glycan Profiling Tools", JOURNAL OF PROTEOME RESEARCH, vol. 8, no. 3, 2009, pages 1358 - 1367, XP008151008 *
MASASHI YONEDA ET AL.: "Tosa Hen'i no HBV no Zoshoku Kansenno eno Eikyo", KOSEI RODO KAGAKU KENKYUHI HOJOKIN B-GATA KAN'EN SOYAKU JITSUYOKA TO KENKYU JIGYO 'B-GATA KAN'EN VIRUS NI OKERU TOSA NO KINO KAISEKI TO IYO OYO GIJUTSU NO JITSUYOKA E', HEISEI 25 NENDO SOKATSU BUNTAN KENKYU HOKOKUSHO, May 2014 (2014-05-01), pages 45 - 47 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019235584A1 (fr) 2018-06-06 2019-12-12 国立研究開発法人産業技術総合研究所 Procédé permettant d'induire de manière efficace un anticorps contre le virus de l'hépatite, anticorps et système de détection
US11571473B2 (en) 2018-06-06 2023-02-07 National Institute Of Advanced Industrial Method for efficiently inducing antibody, antibody and detection system for hepatitis virus
CN113476607A (zh) * 2021-08-04 2021-10-08 中南大学湘雅二医院 Slc12a5及其抑制剂的应用

Also Published As

Publication number Publication date
JP6566368B2 (ja) 2019-08-28
JPWO2016167369A1 (ja) 2018-03-29

Similar Documents

Publication Publication Date Title
Sureau et al. The hepatitis delta virus: Replication and pathogenesis
Yan et al. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus
Hosmillo et al. Noroviruses subvert the core stress granule component G3BP1 to promote viral VPg-dependent translation
Lubyova et al. PRMT5: A novel regulator of Hepatitis B virus replication and an arginine methylase of HBV core
Yaron et al. The FDA-approved drug Alectinib compromises SARS-CoV-2 nucleocapsid phosphorylation and inhibits viral infection in vitro
Florian et al. Characterization of the anti‐HBV activity of HLP1–23, a human lactoferrin‐derived peptide
JP2024076385A (ja) B型肝炎ウイルスタンパク質の産生を阻害する医薬組成物およびスクリーニング方法
US8580759B2 (en) Anti-hepatitis C virus composition
JP6566368B2 (ja) B型肝炎ウイルス分泌阻害剤
KR20220097463A (ko) 바이러스 분자 수준을 감소시키는 방법
Shirasaki et al. Nonlytic quasi-enveloped hepatovirus release is facilitated by pX protein interaction with the E3 ubiquitin ligase ITCH
JP6986263B2 (ja) 抗ウイルス薬
CN113631192B (zh) 抑制乙型肝炎病毒蛋白产生的医药组合物、用于处置乙型肝炎的医药组合物及筛选方法
JP7545148B2 (ja) ウイルス感染及び活性抑制方法
JP2022529481A (ja) B型肝炎ウイルスの増殖を抑制する組成物及びその方法
Xu et al. FUSE binding protein FUBP3 is a potent regulator in Japanese encephalitis virus infection
CN113262293A (zh) 一组具有抗病毒活性的肽类化合物奥米克欣及其应用
WO2018030534A1 (fr) Composition pour le traitement de l'hépatite b et procédé d'évaluation de l'activité de réplication du virus de l'hépatite b
Chen et al. Hepatitis C virus NS5A protein interacts with lysine methyltransferase SET and MYND domain‐containing 3 and induces activator protein 1 activation
KR102085635B1 (ko) B형 간염 바이러스에서의 c-FLIP 단백질의 용도
Sheng et al. Hepatitis E virus ORF3 protein hijacking thioredoxin domain-containing protein 5 (TXNDC5) for its stability to promote viral particle release
KR101221777B1 (ko) Hsp90 억제제를 포함하는 간질환 치료용 조성물 및 Hsp90과 HBV 코어 단백질간의 상호작용 억제를 통한 간질환 치료제 스크리닝 방법
CN112367993A (zh) 治疗乙型肝炎病毒(hbv)感染的方法
Tang et al. miR-342-5p targets CTNNBIP1 to promote enterovirus 71 replication
Sureau Hepatitis Delta Virus: Virology and Replication

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16780160

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017512609

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16780160

Country of ref document: EP

Kind code of ref document: A1