WO2007043640A1 - Substance for use in treatment or prevention of hcv infection - Google Patents

Substance for use in treatment or prevention of hcv infection Download PDF

Info

Publication number
WO2007043640A1
WO2007043640A1 PCT/JP2006/320440 JP2006320440W WO2007043640A1 WO 2007043640 A1 WO2007043640 A1 WO 2007043640A1 JP 2006320440 W JP2006320440 W JP 2006320440W WO 2007043640 A1 WO2007043640 A1 WO 2007043640A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
hcv
seq
amino acid
acid sequence
Prior art date
Application number
PCT/JP2006/320440
Other languages
French (fr)
Japanese (ja)
Inventor
Masayuki Sudo
Hiroshi Sakamoto
Original Assignee
Chugai Seiyaku Kabushiki Kaisha
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 Chugai Seiyaku Kabushiki Kaisha filed Critical Chugai Seiyaku Kabushiki Kaisha
Priority to JP2007539995A priority Critical patent/JPWO2007043640A1/en
Publication of WO2007043640A1 publication Critical patent/WO2007043640A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • 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
    • A61P31/14Antivirals for RNA viruses
    • 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
    • A61P35/00Antineoplastic agents
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24211Hepacivirus, e.g. hepatitis C virus, hepatitis G virus
    • C12N2770/24222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • G01N2333/08RNA viruses
    • G01N2333/18Togaviridae; Flaviviridae
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • the present invention relates to a drug for treating or preventing HCV infection, which contains a compound that inhibits the binding of sphingomyelin and HCV protein as an active ingredient.
  • the present invention also relates to a method for screening a drug for treating or preventing HCV infection, and a kit for use in the method.
  • HCV was discovered in 1989 as a major causative virus for non-A non-B hepatitis after blood transfusion.
  • HCV is an enveloped RNA virus whose genome consists of single-stranded (+) RNA and is classified into the genus Hepacivirus of the Flaviviridae family.
  • HCV progresses to chronic hepatitis, liver cirrhosis, and liver cancer, where persistent infection is often established even when infected with an adult who has developed an immune system, in order to avoid the host's immune mechanism due to unclear reasons.
  • liver cancer recurrence due to inflammation that continues in non-cancerous areas even after surgery.
  • interferon treatment is known as the only effective treatment for HCV elimination.
  • about 1Z3 of all patients are effective for interferon.
  • the response rate of interferon against HCV genotype lb is very low. Therefore, the development of anti-HCV drugs that can replace or be used in combination with the interface is strongly desired.
  • ribavirin 1— ⁇ -D-ribofuranosyl 1 ⁇ — 1, 2, 4-triazol (Lu-3-Carboxamide) is marketed as a therapeutic agent for hepatitis C in combination with interferon, but there is a need for a new therapeutic agent for hepatitis C whose effectiveness is still low.
  • attempts have been made to eliminate viruses by enhancing the immunity of people such as interferon agonists and interleukin 12 agonists, but no effective drugs have been found yet.
  • Non-Patent Document 1 The mechanism of HCV RNA replication in this system is thought to be identical to the replication of the full-length HCV RNA genome infected with hepatocytes. Therefore, this system can be said to be a cell-based accessory system useful for identifying compounds that inhibit HCV replication.
  • Patent Document 1 International Publication No. W098 / 56755 Pamphlet
  • Patent Document 2 International Publication No. WO04 / 71503 Pamphlet
  • Patent Document 3 International Publication WO05 / 05372 Pamphlet
  • Non-Patent Document 1 Buoy Roman et al., Science, 1999, No. 285, 110-113
  • Patent Document 1 The inventors of the present application disclosed in International Publication No. W098 / 56755 (Patent Document 1), and a series of compounds derived from microorganisms such as the genus Aureobasidium, It was found by the Atsey method that it has high HCV replication inhibitory activity. Permissible literature 2). In addition, the present inventors have found that the compound has minimal in vitro cytotoxicity and is extremely useful as a prophylactic or therapeutic agent for HCV infection, and further constructed a method for synthesizing the compound and derivatives. (Patent Document 3).
  • the inventors of the present invention also found that sphingolipid biosynthesis is involved in HCV infection, and compounds that inhibit the activity and expression of enzymes involved in sphingolipid biosynthesis are extremely useful therapeutic or preventive agents for HCV infection. It was made clear (WO2006 / 16657).
  • Sphingomyelin a sphingolipid
  • rafts on cell membranes, and viruses such as influenza and HIV are replicated via rafts (Takeda M. et al. (2003) P NAS, 100 , 25, Lucero HA, et al. (2004) Archives of Biochemistry and Biophysics, 426, 208, Simons K. (1997) Nature, 387, 569, G.— Z. Leu et al. (2004)) 0 Therefore, the present inventors predicted that HCV is replicated via rafts and attempted to elucidate the detailed mechanism of HCV virus replication via rafts containing sphingolipids.
  • the present invention has been made in view of such a situation, and an object of the present invention is to treat or prevent HCV infectious diseases containing a compound that inhibits the binding of sphingomyelin and HCV protein as an active ingredient. It is to provide a drug to do. Another object of the present invention is to provide a method for screening a drug for treating or preventing HCV infection and a kit for use in the method.
  • the binding site of (sphingomyelin) was identified and the binding ability was examined.
  • the present inventors have succeeded in developing a drug for treating or preventing HCV infection containing a compound that inhibits the binding of sphingomyelin and HCV protein as an active ingredient, thus, the present invention has been completed.
  • the present invention provides the following [1] to [14].
  • a drug for treating or preventing HCV infection comprising as an active ingredient a compound that inhibits the binding of sphingomyelin and HCV protein.
  • [2] Compound ability to inhibit binding of sphingomyelin and HCV protein
  • the drug according to [1] which is a peptide according to the following (a) or (b).
  • [3] Compound ability to inhibit binding of sphingomyelin and HCV protein
  • the drug according to [1] which is an oligonucleotide encoding the peptide according to the following (a) or (b).
  • [4] Compound ability to inhibit binding of sphingomyelin to HCV protein
  • the drug according to [1] which is an antibody that recognizes the peptide according to (a) or (b) below.
  • [6] The drug according to any one of [1] to [5], wherein the HCV infection is hepatitis C, cirrhosis, liver fibrosis, or liver cancer.
  • a screening method for a drug for treating or preventing HCV infection comprising the following steps (A) to (C):
  • a method for screening a drug for treating or preventing HCV infection comprising the following steps (A) to (C):
  • SEQ ID NO: 1 in the amino acid sequence described in any one of 4, 10, or 11 Is a peptide with multiple amino acid substitutions, deletions, additions, and Z or inserted amino acid sequence
  • a method for evaluating the efficacy of a drug for treating or preventing HCV infection comprising the following steps (A) to (C):
  • a method for evaluating the efficacy of a drug for treating or preventing HCV infection comprising the following steps (A) and (B):
  • (B) A step of evaluating the efficacy of the treatment or prevention effect of HCV infection by measuring the binding ability of the peptide described in (a) or (b) above and sphingomyelin.
  • FIG. 1A is a view showing the amino acid sequences of HCV-NS5B sequences E230-G263 and V3 loop.
  • B is a diagram showing the structure of HCV-NS5B.
  • the vertical line is the predicted sphingolipid binding region identified by HCV-NS5B.
  • the diagonal line indicates the sphingolipid binding region of the HIV-1 V3 loop.
  • C shows the helix 'turn' helix motif in HCV-NS5B.
  • FIG. 2 is a diagram showing the results of a binding study of HCV-NS5B sphingo binding domain peptide (NS5B-SBD, SEQ ID NO: 1) and sphingomyelin (SM) using Biacore.
  • A is a diagram showing a sensorgram of NS5B-SB D peptide, and NS5B-SBD peptide was found to bind to sphingomyelin in a concentration-dependent manner.
  • B is a comparison of the binding ability of NS5B-SBD peptide and prion protein (PrP, SEQ ID NO: 2) to sphingomyelin.
  • Sensorgram Shows the RU value at 80 s.
  • FIG. 3 shows the results of measuring HCV replicon inhibitory activity of NS5B sphingo-binding domain peptides and their derivatives.
  • FIG. 4 is a diagram showing a sphingolipid synthetic pathway (synthetic pathway from palmitoyl CoA to sphingomyelin).
  • FIG. 5 is a photograph showing the HCV RNA replication inhibitory activity of myriocin by Northern plot analysis.
  • the horizontal axis represents the concentration of myriocin.
  • FIG. 6 is a photograph showing the HCV RNA replication inhibitory activity of the compound represented by formula (II) by Northern plot analysis.
  • the horizontal axis represents the concentration of the compound represented by the formula ( ⁇ ).
  • FIG. 7 A photograph showing the inhibitory activity of myriocin on HCV protein synthesis by Western plot analysis. The horizontal axis represents the concentration of myriocin.
  • FIG. 8 is a photograph showing the HCV protein synthesis inhibitory activity of the compound represented by formula ( ⁇ ) by Western plot analysis.
  • the horizontal axis represents the concentration of the compound represented by formula (II).
  • FIG. 9 is a graph showing the HCV replicon inhibitory activity of fumosin B1.
  • FIG. 10 is a photograph showing inhibition of protein expression of serine palmitoyltransferase (LCB1) by siRNA.
  • FIG. Ll A graph showing the effects of siRNA on the HCV replicon inhibitory activity and cytotoxicity.
  • FIG. 12 is a graph showing the inhibition of HCV levicon and the toxicity to host cells by the compound represented by formula (II).
  • FIG. 13 is a photograph showing inhibition of HCV-NS3 protein expression by a compound represented by formula (II). After immunostaining, it was observed with a fluorescence microscope. White indicates NS3 protein, gray indicates nuclei stained with hex 33342.
  • FIG. 14 is a photograph showing inhibition of NS3, NS5A, and NS-5B protein expression by a compound represented by formula (II). Each protein was expressed by Western plot analysis.
  • FIG. 15 is a graph showing SPT inhibitory activity of a compound represented by formula (II).
  • FIG. 16 is a photograph showing inhibition of de novo synthesis of ceramide and sphingomyelin by the compound represented by formula (II).
  • FIG. 17 is a photograph showing suppression of HCV replication inhibition of a compound represented by formula (II) by C2-ceramide.
  • FIG. 18 shows inhibition of HCV replication by a raft biosynthesis-related low molecular weight compound.
  • FIG. 19 is a photograph showing the influence of a compound represented by formula (II) on raft protein.
  • FIG. 20 is a diagram showing the influence of a compound represented by formula (II) on raft protein.
  • the present invention relates to a drug for treating or preventing HCV infection, which contains a compound that inhibits the binding of sphingomyelin and HCV protein as an active ingredient.
  • the compound that inhibits the binding of sphingomyelin and HCV protein is any compound that can directly or indirectly inhibit the binding reaction of sphingomyelin and HCV protein. It may be a compound. Further, it may be a compound that generates or increases these inhibitors and indirectly inhibits the binding reaction between sphingomyelin and HCV protein.
  • HCV proteins HCV-NS2, HCV-NS3, HCV-NS4A, HCV-NS4B, HCV-NS5A, HCV-NS5B can be mentioned, more preferably HCV-NS5B. it can.
  • Preferable examples of the compound that inhibits the binding of sphingomyelin and HCV protein of the present invention include the peptides described in the following (a) or (b).
  • These peptides also include peptides functionally equivalent to the peptides comprising the amino acid sequences described in any one of SEQ ID NOs: 1 to 4, 10 or 11, including the entered amino acid sequence.
  • the amino acid residue to be mutated is preferably mutated to another amino acid in which the properties of the amino acid side chain are conserved.
  • amino acid side chain properties include hydrophobic amino acids (A, I, M, F, P, W, Y, V), hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, T), amino acids having aliphatic side chains (G, A, V, L, I, P), amino acids having hydroxyl-containing side chains (S, T, ⁇ ), sulfur atoms
  • Amino acids with side chains C, M
  • amino acids with side chains containing carboxylic acids and amides D, N, E, Q
  • amino acids with side chains R, K, ⁇
  • aromatics Mention may be made of amino acids having a side chain (H, F, Y, W) (in parentheses all represent one letter of the amino acid).
  • SEQ ID NO: 1 to 4, 10 or 11 in the amino acid sequence described in any one or more of the amino acid substitution, deletion, addition and a peptide having an amino acid sequence ability that is Z or inserted, Examples thereof include peptides having amino acid strength described in any one of SEQ ID NOs: 5 to 7.
  • These peptides may specifically bind to the HCV protein binding site in sphingomyelin, and thus may reduce the binding activity of sphingomyelin and HCV protein. As a result, it is considered that replication of HCV virus via rafts is suppressed.
  • Preferred examples of the compound that inhibits the binding of sphingomyelin and HCV protein of the present invention include the oligonucleotides encoding the peptides described in (a) or (b) below. .
  • the oligonucleotide of the present invention can be used by incorporating it into an appropriate vector in a form capable of expressing the peptide.
  • the phrase "comprising an expressible form" of a polynucleotide encoding the peptide means that the polynucleotide is inserted into an expression vector and enters the animal cell. It means that a predetermined peptide can be expressed in a cell. That is, for example, it means that the coding DNA is arranged under the control of a promoter suitable for the species of animal to be administered and the administration site.
  • Preferable examples of the compound that inhibits the binding of sphingomyelin and HCV protein of the present invention include antibodies that recognize the peptides described in (a) or (b) below.
  • the origin of the antibody in the present invention is not particularly limited, but is preferably derived from a mammal, more preferably a human-derived antibody.
  • An antibody recognizing the peptide used in the present invention can be obtained as a polyclonal or monoclonal antibody using a known means.
  • a monoclonal antibody derived from a mammal is particularly preferable.
  • Monoclonal antibodies derived from mammals include those produced by hyperpridoma and those produced by a host transformed with an expression vector containing an antibody gene by genetic engineering techniques. This antibody inhibits the binding of HCV protein to sphingomyelin by binding to HCV protein. As a result, replication of HCV virus via rafts is thought to be suppressed.
  • An antibody-producing hybridoma that recognizes the peptide can be basically produced using a known technique as follows. That is, the peptide is used as a sensitizing antigen, and this is immunized according to a normal immunization method. The obtained immune cells are fused with a known parent cell by a normal cell fusion method, and a normal screening method is used. It can be produced by screening monoclonal antibody-producing cells.
  • an antibody that recognizes the peptide may be prepared as follows!
  • the peptide of interest is purified from the host cell or culture supernatant by a known method.
  • This purified peptide may be used as a sensitizing antigen.
  • a fusion protein of the peptide and another protein may be used as a sensitizing antigen.
  • treatment refers to the administration of the drug of the present invention to a subject to eliminate or reduce HCV, further suppress the spread of HCV, and symptoms caused by HCV infection. Means to reduce.
  • prevention refers to administration to a subject prior to HCV infection to prevent HCV infection or suppress proliferation. means.
  • Symptoms caused by HCV infection preferably include hepatitis C, cirrhosis, liver fibrosis, liver cancer and the like.
  • the compound of the present invention can be used in medicine.
  • the salt is not particularly limited as long as it is pharmacologically acceptable.
  • salts with mineral acids such as hydrochloric acid, sulfuric acid, nitric acid, phosphoric acid, hydrobromic acid; acetic acid, tartaric acid, lactic acid, ken Acid, fumaric acid, maleic acid, succinic acid, methane sulfonic acid, ethane sulfonic acid, benzene sulfonic acid, toluene sulfonic acid, naphthalene sulfonic acid, salt with organic acid such as camphor sulfonic acid; alkali such as sodium, potassium and calcium Examples thereof include salts with metals or alkaline earth metals.
  • the amount of the active ingredient-compound contained in the above-mentioned pharmaceutical preparation is not particularly limited and can be selected as appropriate over a wide range. For example, 0.1 to 99.5% by weight, preferably 0.5 to 90% by weight. .
  • the pharmaceutical preparation technical field such as excipients, binders, disintegrants, lubricants, flavoring agents, solubilizers, suspension agents, coating agents, etc., with the compound of the present invention as the main agent according to conventional methods.
  • it can be formulated using known adjuvants that can be usually used.
  • conventionally known carriers can be widely used as carriers, such as lactose, sucrose, sodium chloride sodium salt, glucose, urea, starch, calcium carbonate, kaolin, crystalline cellulose, and key acid.
  • Excipients such as water, ethanol, propanol, simple syrup, glucose solution, starch solution, gelatin solution, binders such as carboxymethylcellulose, shellac, methylcellulose, potassium phosphate, polybulurpyrrolidone; dry starch, sodium alginate Disintegrants such as lithium, agar powder, laminaran powder, sodium bicarbonate, calcium carbonate, polyoxyethylene sorbitan fatty acid ester, sodium lauryl sulfate, stearic acid monoglyceride, starch, lactose; white sugar, stearin, cocoa butter, hydrogenated oil, etc.
  • binders such as carboxymethylcellulose, shellac, methylcellulose, potassium phosphate, polybulurpyrrolidone
  • dry starch sodium alginate Disintegrants such as lithium, agar powder, laminaran powder, sodium bicarbonate, calcium carbonate, polyoxyethylene sorbitan fatty acid ester, sodium lauryl sulfate, stearic acid
  • Collapse inhibitor 4th Absorption accelerators such as ammonium salts and sodium lauryl sulfate; humectants such as glycerin and starch; adsorbents such as starch, lactose, kaolin, bentonite and colloidal key acid; purified talc, stearate, boric acid powder, Examples include a lubricant such as polyethylene glycol.
  • the tablets can be made into tablets with ordinary coatings, for example, sugar-coated tablets, gelatin-encapsulated tablets, enteric-coated tablets, film-coated tablets or double tablets, and multilayer tablets as necessary.
  • the In molding into the form of a pill those conventionally known in this field can be widely used as a carrier.
  • excipients such as glucose, lactose, cocoa butter, starch, hydrogenated vegetable oil, kaolin, and tar
  • binders such as tragacanth powder, gelatin and ethanol
  • disintegrants such as laminaran agar.
  • conventionally known carriers can be widely used as carriers, such as polyethylene glycol, cocoa butter, higher alcohols, higher alcohol esters, gelatin, semi-synthetic glycerides and the like. it can.
  • solutions and suspensions should be sterilized and used as diluents in the form of these solutions, emulsions and suspensions, which are preferably isotonic with blood. Any of those commonly used in this field can be used, and examples thereof include water, ethanol, propylene glycol, ethoxylated isostearyl alcohol, polyoxylated isostearyl alcohol, and polyoxyethylene sorbitan fatty acid esters.
  • a sufficient amount of sodium chloride, glucose, or glycerin to prepare an isotonic solution may be contained in the pharmaceutical preparation, and a normal solubilizing agent, buffering agent, soothing agent may be used.
  • An agent or the like may be added.
  • it may contain colorants, preservatives, fragrances, flavors, sweeteners, and other medicines as necessary.
  • the pharmaceutical composition is preferably administered in a dosage unit form for oral administration, tissue administration (subcutaneous administration, intramuscular administration, intravenous administration, etc.), topical administration (transdermal administration, etc.) Can be administered rectally.
  • tissue administration subcutaneous administration, intramuscular administration, intravenous administration, etc.
  • topical administration transdermal administration, etc.
  • the pharmaceutical composition is administered in a dosage form suitable for these administration methods.
  • the dose as an antiviral agent is preferably adjusted in consideration of the patient's condition such as age and weight, administration route, nature and degree of disease, etc.
  • the amount of the active ingredient of the present invention for adults is usually in the range of 0.1 to 2000 mg per day. In some cases, doses below the above range may be sufficient. Conversely, doses in excess of the above range may be required.
  • administering a large amount it is desirable to divide it into several times a day.
  • the oral administration can be carried out in solid, powder or liquid dosage units, for example, powders, powders, tablets, dragees, capsules, drops, sublingual agents, other dosage forms, etc. I can.
  • the intra-tissue administration can be performed, for example, by using a liquid dosage unit form for subcutaneous, intramuscular or intravenous injection such as a solution or suspension. These are obtained by suspending or dissolving a certain amount of the compound of the present invention in a non-toxic liquid carrier suitable for injection purposes such as an aqueous or oily medium, and so on. It is manufactured by sterilizing.
  • the topical administration can be performed by using a form of external preparation such as a liquid, a cream, a powder, a paste, a gel, and an ointment.
  • a certain amount of the compound of the present invention is added to a fragrance, a colorant, a filler, a surfactant, a moisturizer, an emollient, a gelling agent, a carrier, a preservative, Manufactured by combining with one or more agents.
  • a certain amount of the compound of the present invention is mixed with a high-melting low-melting solid such as higher esters such as palmitic acid myristyl ester, polyethylene glycol, cocoa butter, and mixtures thereof. It can be performed using a suppository or the like.
  • the administration can be performed, for example, by using a liquid dosage unit form for subcutaneous, intramuscular or intravenous injection such as a solution or suspension.
  • a liquid dosage unit form for subcutaneous, intramuscular or intravenous injection such as a solution or suspension.
  • a liquid dosage unit form for subcutaneous, intramuscular or intravenous injection such as a solution or suspension.
  • a non-toxic liquid carrier suitable for injection purposes such as an aqueous or oily medium, then the suspension or solution. It is manufactured by sterilizing.
  • a peptide having a cell membrane permeation function for example, Pegelin, Penetratin, etc.
  • a cell membrane permeation function for example, Pegelin, Penetratin, etc.
  • Doxorubi cm— peptide conjugates overcome multidrug resistance.
  • Anti-Cancer Drugs 2001, 12, Dcrossi D. et al The third helix of the antennapedia homeodomain translocates th rough biological membranes, J. Biol. Chem. 1994, 269, 10444-10450.
  • a method for introducing the peptide of the present invention into cells (1) one or more copies of a gene encoding a peptide sequence may be incorporated into a viral vector (eg, adenovirus). (2) a method of directly introducing the target peptide by intravenous injection, (3) a target peptide or a DNA encoding the target peptide or DNA encoding it physically.
  • the method for example, particle gun
  • the particle gun is an extremely powerful gene transfer method that introduces nucleic acid attached to gold or tandastene microparticles (microcarriers) into target cells by the pressure of helium gas. This method is much easier to operate than the microinjection method, and can be used in a wide range of applications regardless of the cell type. This is expected to be applied to the introduction of peptides.
  • the present invention relates to a method for screening a drug for treating or preventing HCV infection.
  • a test compound is contacted with the peptide described in the following ( a ) or (b).
  • the state of the peptide used in the first embodiment is not particularly limited, and may be, for example, a purified state, a state expressed in a cell, a state expressed in a cell extract, or the like.
  • Examples of cells expressing the peptide include cells expressing a foreign peptide.
  • a cell expressing the exogenous peptide can be prepared, for example, by introducing a vector containing DNA encoding the peptide into the cell. Introduction of a vector into a cell can be performed by a general method such as calcium phosphate precipitation, electric pulse perforation, ribophetamine, or microinjection.
  • the biological species from which a cell into which such an exogenous peptide is introduced is derived is not limited to mammals, and any biological species that has established a technique for expressing a foreign protein in the cell may be used.
  • the cell extract in which the peptide is expressed is included in, for example, an in vitro transcription / translation system.
  • An example is a cell extract obtained by adding a vector containing DNA encoding a peptide.
  • the in vitro transcription / translation system it is possible to use a commercially available in vitro transcription / translation kit without particular limitation.
  • test compound in the method of the present invention is not particularly limited, for example, natural compounds, organic compounds, inorganic compounds, single compounds such as proteins and peptides, and compound libraries. Gene library expression products, cell extracts, cell culture supernatants, fermentation microorganism products, marine organism extracts, plant extracts, prokaryotic cell extracts, eukaryotic single cell extracts or animal cell extracts, etc. Can be mentioned.
  • the above test compound can be appropriately labeled and used as necessary. Examples of the label include a radiolabel and a fluorescent label.
  • the “plurality of test compounds” is not particularly limited. For example, in addition to the above test compounds, a mixture of a plurality of these test compounds is also included.
  • “contact” is performed depending on the state of the peptide.
  • the peptide if it is in a purified state, it can be carried out by adding a test compound to the purified sample. Moreover, if it is in a state expressed in a cell or in a cell extract, it can be carried out by adding a test compound to the cell culture solution or the cell extract, respectively.
  • the cell in the present invention is not particularly limited, but a cell derived from a mammal including yeast or human is preferable.
  • the test compound is a protein
  • a vector containing DNA encoding the protein is introduced into a cell in which the peptide is expressed! /, Or the vector is expressed in the peptide. It can also be carried out by adding to the cell extract. Further, for example, a two-hybrid method using yeast or animal cells can be used.
  • the binding between the peptide and the test compound is detected.
  • the means for detecting or measuring the binding between proteins can be performed, for example, by using a label attached to the protein.
  • the label type include fluorescent labels and radiolabels.
  • it can also be measured by a known method such as an enzyme two-hybrid method or a measurement method using BIACORE.
  • a test compound bound to the peptide is then selected. Some of the selected test compounds treat HCV infection. Or drugs to prevent. Alternatively, the selected test compound may be used as a test compound for the following screen.
  • test compound is added to sphingomyelin simultaneously with the peptide described in the following (a) or (b).
  • the binding ability of the peptide described in (a) or (b) above and the sphingomyelin is measured next.
  • the binding ability can be measured by the method described above.
  • the test compound is then selected as a pharmaceutical agent for treating or preventing HCV infection when the binding ability is reduced as compared with the case where no test compound is added.
  • the present invention also relates to a method for evaluating the efficacy of a drug for treating or preventing HCV infection.
  • a test compound is brought into contact with the peptide described in the following ( a ) or (b).
  • the binding between the peptide and the test compound is then detected. Detection of the binding between the peptide and the test compound can be performed by the method described above.
  • the efficacy of the therapeutic or prophylactic effect of HCV infection of the test compound bound to the above peptide is evaluated.
  • the test compound can detect HCV infection.
  • the efficacy of the therapeutic or preventive effect can be evaluated. For example, when the test compound shows a significant binding ability to the peptide, it can be evaluated that the test compound shows a significant therapeutic or preventive effect on HCV infection.
  • a test compound is added to sphingomyelin simultaneously with the peptide described in the following (a) or (b).
  • the binding ability of the peptide described in (a) or (b) above and sphingomyelin is measured, and the test compound has an effect of treating or preventing HCV infection.
  • the binding ability can be measured by the method described above.
  • the test compound By detecting the binding ability of the peptide and sphingomyelin (eg, equilibrium binding constant, binding rate constant, dissociation rate constant, etc.), the test compound has the ability to inhibit the binding of the peptide to sphingomyelin. Can be detected.
  • the efficacy of the test compound for treating or preventing HCV infection can be evaluated.
  • test compound when the test compound is added and the binding ability of the peptide and sphingomyelin is significantly reduced compared to the case where the test compound is not added, the test compound is significantly more susceptible to HCV infection. It can be evaluated that it shows an effective therapeutic or preventive effect.
  • the above-described evaluation method of the present invention is not limited to a screening method for finding a new drug for treating or preventing HCV infection, but an evaluation of the efficacy of the drug for the development of a drug candidate compound. It is also useful as an evaluation method when evaluating the efficacy for quality control required in the manufacture and supply of pharmaceuticals.
  • the present invention relates to a kit for use in the screening method or evaluation method described above.
  • a kit may include those used in the detection process and measurement process of the screening method or evaluation method described above.
  • reagents and instruments required for measuring the binding ability of the peptide and sphingomyelin.
  • Other steaming Retention water, salt, buffer solution, protein stabilizer, preservative and the like may be contained. All prior art documents cited in the present specification are incorporated herein by reference.
  • HIV-lgpl20-derived V3 loop (Fig. 1A, SEQ ID NO: 4) and prion-derived PrP protein have sphingolipid binding domains.
  • structures similar to these were searched using the CE program (cl. Sdsc.edu/ce.html).
  • FIG. 1 in HCV-NS5B sequence E230-G263 (FIG. 1A, SEQ ID NO: 3), a similar structure having a helix 'turn' helix motif (FIG. 1) was observed.
  • the full length base sequence of HCV-NS5B is shown in SEQ ID NO: 8 and the amino acid sequence is shown in SEQ ID NO: 9.
  • Figure 1B shows the structure of HCV-NS5B.
  • the vertical line is the putative sphingolipid binding region identified by HCV-NS5B.
  • the diagonal line is the sphingolipid of the HIV-1 V3 loop. Binding area).
  • NS5B-SBD peptide and SM were examined using Biacore S51.
  • SM was dissolved in chloroform and vacuum-dried, and then PBS buffer was added and suspended to 10 mM. The suspension was repeatedly frozen and thawed, passed through a membrane with a pore size of 50 nm, and ribosomes were prepared and immobilized on the sensor chip L1.
  • NS5B DIRVEESIYQCCDL APEARQAIKSLTERLY (SEQ ID NO: 1), synthesized by Sigma genosys) and PrP (KQHT VTTTTKGENFTETDVKMMER (SEQ ID NO: 2), synthesized by Sigma genosys) were used as synthetic peptides.
  • the amount of specific binding was determined by measuring the amount of binding using a sensorgram without a peptide as a blank and subtracting the sensorgram force at each concentration.
  • NS5B-SBD peptide was found to bind to sphingomyelin in a concentration-dependent manner (Fig. 2A, sensorgram of NS5B-SBD peptide). Similar binding was observed in prion protein (PrP) (Fig. 2B, RU value at 80 s sensorgram in Figure 2A).
  • HCV revlikon inhibitory activity of NS5b sphingo binding domain peptide (SEQ ID NO: 1) and its derivative peptides (SEQ ID NO: 10, 11).
  • DMEM was added in O.lmL and further cultured. 20 and 40 hours after peptide addition, replicon with Steady-Glo Luciferase Assay System (Promega, Cat.No. E2520) Activity was measured. A peptide consisting of the amino acid sequence shown in SEQ ID NO: 12 was used as a negative peptide.
  • NS5B sphingo-binding domain peptide and its derivatives showed 20-30% anti-HCV replication inhibition at a final concentration of 0.1 mg / mL (FIG. 3). On the other hand, no inhibition was observed with the negative peptide.
  • Myriocin or a compound represented by the following formula (II) was given to the replicon cell Huh-3-l in the range of ⁇ to 100 ⁇ and cultured at 37 ° C in the presence of 5% CO.
  • the Northern analysis used the following. That is, NorthernMax transfer buffer (Ambion # 8672), transfer film BrightStar-Plus (Ambion # 10100), filter paper (Sigma P-6664), ULTRAhy b (Ambion # 8670).
  • the probe was labeled biotinylated with BiotinStar Psoralen-Biotin kit (Ambion # 9860 G3).
  • High Stringency buffer Amibion # 8674
  • BrightStar BioD etect kit Wash buffer, Ambion # 8650G; Blocking buffer, Ambion # 8651G; Streptavi din-Alkaline Phosphatase, Ambion # 2374G; Assay buffer, Ambion # 8652G; CDP— Starr, (Ambion # 8653G)
  • RNA was run on a 1% agarose gel, and after electrophoresis, RN A was stained and photographed. After decolorization, it was transferred to a transfer film for 2 hours using NorthernMax transfer buffer. In a wet state, RNA was immobilized on the transfer membrane with a UV crosslinker. Using a hybrid rotor, after pre-rotating for 42 minutes at ULTRAhyb for 30 minutes, the pre-treatment solution was discarded, and the neomycin resistance gene and 10 ml of ULTRAhyb solution were added and shaken overnight at 42 degrees.
  • the electrophoresed protein was transferred to a mini-trans blot cell (BIO-RAD # l 70-393 0) was transferred to a membrane (PROTRAN BA85, Nitrocellulose transfer membrane (Shleicher & Sc huell # 10401196)) Western analysis was performed using an anti-NS3 Usagi antibody derived from HCV protein, and an anti-actin Usagi antibody was used as an internal standard.
  • myriocin and the compound represented by formula (II) were found to have an effect of reducing HCV protein expression by 50% at a concentration of 1-lOnM (FIGS. 7 and 8).
  • HCV replicon inhibitory activity of fumonisin B1 which specifically inhibits dihydrosphingosine synthase, which produces dihydrosphingosine, a dihydroceramide, was measured.
  • Thin B1 was diluted three-fold in order, added to a final concentration of 1.37 M to 1000 M, and further cultured for 3 days. Two assembly plates were prepared, one for the white plate and the other for the clear plate. After completion of the culture, the white plate was used for Steady-Glo Luciferase Assay System (Promega cat. No. E2520). That is, 100 ⁇ l of reagent was added per well, mixed 3-4 times with a pipette, allowed to stand for 5 minutes, and then luminescence was measured with 1450 MicroBeta TRILUX (WALLAC).
  • WALLAC MicroBeta TRILUX
  • the IC50 (50% inhibitory concentration) of the drug was calculated by subtracting all values from the value with no added cells as the background, and the value with no drug added as 0% inhibition.
  • Cell's Count Kit 8 (DOJIN Laboratories, cat.No.341-07771) was put in 10 ⁇ l per well, mixed 3-4 times with a pipette, and left at about OD450nm power of about 1.0 after about 30 minutes. It measured when it became.
  • the IC50 (50% inhibitory concentration) of the drug was calculated by subtracting all values from the value with no added cells as the background and the value with no drug added as 0% inhibition.
  • fumosin B1 exhibits HCV replicon inhibitory activity at a concentration of 10-1000 M (FIG. 9).
  • siRNA targeting LCB1 one subunit of SPT heterodimer.
  • siRNAs two specific The siRNAs (si246, si633) were designed based on the LCBlcDNA sequence (GenBank Accession No. Y08685) and synthesized by Qiagen.
  • SEQ ID NO: 15 As a control siRNA (SEQ ID NO: 15), a sequence that does not affect the expression of LCB1 was used.
  • the synthesized siRNA sequence is shown in SEQ ID NO: 13 (si24 6) and SEQ ID NO: 14.
  • Cell lysis buffer 50 mM Tris-HC1 (pH 7.5), 0.5% Triton, 3 mM EDTA, 1 50 mM NaCl, 12 mM glycerophosphate, 50 mM NaF, 1 mM Na ⁇ O ⁇ , 0.5 mM PMSF, Suspended in 0.5 mM aporotinin) and left on ice for 10 minutes. The supernatant was collected by centrifugation at 15,000 X g for 10 minutes.
  • the cells treated with si246 and si633 that suppressed the expression of LCB1 significantly inhibited HCV replicon activity compared to the cells treated with control siRNA.
  • This inhibitory effect was strongly observed with si246, which strongly suppressed the expression of LCB1.
  • the cytotoxicity of siRNA treatment was examined. Almost no effect was observed.
  • the HCV replicon assay and cytotoxicity test were conducted on the compounds represented by formula (I) or their derivatives.
  • HCV-RNA with a firefly-derived luciferase gene introduced as a reporter gene.
  • IRES Internal Ribosome Entry Site
  • a luciferase gene was introduced immediately below the neomycin resistance gene.
  • the RNA was synthesized in vitro, then introduced into Huh7 cells by the electopore method, and isolated as a G418 metaclone.
  • Firefly 'Luciferase HCV replicon cells (Huh-3-l) were suspended in Dulbecco's MEM (Gibco cat. No. 10569-010) containing 5% urine fetal serum (Hyclone cat. No. SH30071.03). Seed the well plate with 5000 cells Zwell, 5% CO
  • Cell counting kit-8 (Dojindo catalog No. CK04) was used for the measurement of cytotoxicity. That is, 10 1 Cell counting kit-8 was added to a clear plate and incubated at 37 degrees for 30-60 minutes. Absorbance was measured with a 96-well plate reader at a wavelength of 450 and a control wavelength of 630 nm. All values were subtracted from the value with no cell added, and the CC (50% cell inhibitory concentration) of the drug was calculated with the value without drug added as 0% inhibition.
  • HCV replicon cells were treated with the compound represented by formula (II) at the concentration shown in FIG. 12, and the replicon replication inhibitory activity and cell survival inhibitory activity were measured.
  • Steady— JLO luciferase assay system (Promega, cat. No. E251 0), cell viability inhibitory activity) by cell count counting kit— 8 (Dojin Laboratories, cat.
  • IC50 2 nM
  • the cytotoxicity of the compound represented by the formula (II) was not observed (IC50> 50 nM).
  • HCV replicon cells were treated with 100 nM of the compound represented by formula (II) for 96 hours, and then the cells were fixed with 3.7% formaldehyde. After blocking with 3% BSA and incubating with NS3 antibody (provided by F. Hoffman Laroche), the washed cells were incubated with Texas-Red labeled Usagi IgG (Molecular probe) and analyzed with a fluorescence microscope (Fig. 13). As a result, as shown in Fig. 13, the HCV-NS3 protein disappeared due to the addition of the compound represented by the force equation (II) existing around the nucleus (the part shown in white is the NS3 protein). The part shown in gray shows the nucleus stained with Hoechst 33342 (Sigma, cat. No. B2261)).
  • Replicon cells were treated with 100 nM of the compound represented by formula (II) for the times shown in FIG. 14 (48 hours and 96 hours).
  • Western plot analysis was performed in the same manner as in Reference Example 5.
  • the non-structural proteins NS3, NS5A and NS5B of HCV were detected with each antibody in a time-dependent manner, and a decrease in the expression level of the viral protein was observed (Fig. 14).
  • LCBl and LCB2 cDNAs were obtained from human liver cDNA library (Clontech, cat. No. 639307) by RT-PCR, and His-tagged pBudCE4.1 vector (Invitogen, cat. No. V532-20) ).
  • the gene was introduced into HEK293 cells (ATCC, cat. No. CRL-1573). After 72 hours, the cells were lysed, and the protein was purified with N ⁇ NTA agarose (Qiagen, cat. No. 1018244).
  • SPT activity is reaction buffer [200 mM HEPES buffer (pH 8.0), 5 mM EDTA, 10 mM DTT, 0.05 mM pyridoxal 5 -phosphate, 0.2 mM palmitoy CoA, 0.1 mM L-serine, and 1 mCi [3 H] Serine (Amer sham, cat. no. TRK308)] was added and the reaction was allowed to proceed at 37 ° C for 15 minutes. After extraction with black mouth form: methanol (1: 2, v / v), the organic layer was re-extracted twice with water, and then the radioactivity of the organic layer was measured with a liquid scintillation counter. As a result, as shown in Figure 15, It was revealed that the compound represented by the formula (II) has an SPT inhibitory activity with an IC50 of about 10 nM.
  • HCV replicon cells were treated with the compound represented by formula (II) at the concentration shown in FIG. 16 for 48 hours, and then labeled with [14C] serine for 3 hours. After extraction with black mouth form: methanol (1: 2, v / v), de novo synthesized ceramide (Fig. 16A) and sphingomyelin (Fig. 16B) were separated by thin layer chromatography. As a result, as shown in FIG. 16, the compound represented by the formula (II) inhibited de novo synthesis of intracellular ceramide and sphingomyelin in a concentration-dependent manner.
  • HCV replicon cells were separated from the known SPT inhibitor myriocin (Sigma, cat. No. M1177), ceramide synthesis inhibitor fumosin Bl (Sigma, cat. No. F1147), and ceramide transport inhibitor H PA-12 [ After treatment with Kobayashi et al., Org.lett. (2002), the levulincon activity and viable cell count were measured 72 hours later. As a result, each compound was found to have an effect of suppressing HCV replication at a concentration where no cytotoxicity was observed (FIG. 18).
  • ImM compound (II) represented by the formula (II) was added to HCV replicon cells for 72 hours, and then the cell extract was treated with 1% NP-40 for 1 hour. Raft proteins (solubilizing agent resistance) and non-raft proteins were separated by sucrose density gradient fractionation, diluted with PBS, concentrated and quantified by ELISA analysis. As a result, the compound represented by the formula (II) was significantly dissociated from the raft in NS5B (FIG. 20).
  • the knowledge of the present invention greatly contributes to the development of anti-HCV agents targeting novel binding of sphingolipids and HCV proteins.

Abstract

An attempt has been made to specify the HCV protein-binding site on a sphingolipid and to examine the binding capability between an HCV protein and the sphingolipid. As a result, it is found that a sphingomyelin (a constituent of a raft) can strongly bind to HCV-NS5B (one of HCV proteins) at a specific site and that HCV virus can be replicated through this binding. It is also found that the replication of HCV virus can be prevented by inhibiting the binding between a sphingomyelin and an HCV protein.

Description

明 細 書  Specification
HCV感染症を治療または予防するための薬剤  Drugs for treating or preventing HCV infection
技術分野  Technical field
[0001] 本発明は、スフインゴミエリンと HCVタンパク質の結合を阻害する化合物を有効成 分として含有する、 HCV感染症を治療または予防するための薬剤に関する。また、 H CV感染症を治療または予防するための薬剤のスクリーニング方法、およびこれらに 用いるためのキットに関する。  [0001] The present invention relates to a drug for treating or preventing HCV infection, which contains a compound that inhibits the binding of sphingomyelin and HCV protein as an active ingredient. The present invention also relates to a method for screening a drug for treating or preventing HCV infection, and a kit for use in the method.
背景技術  Background art
[0002] HCVの感染者は世界で 1〜2億人、日本国内では 200万人以上と推測されている 。これらの患者の約 50%が慢性肝炎に移行し、そのうち約 20%が感染後 30年以上 たって肝硬変、肝癌となる。肝癌の約 90%の原因が C型肝炎といわれている。 日本 国内では、毎年 3万人以上の患者が HCV感染に伴う肝癌により死亡している。  [0002] It is estimated that there are 100-200 million people infected with HCV worldwide and more than 2 million people in Japan. About 50% of these patients transition to chronic hepatitis, of which about 20% develop cirrhosis and liver cancer more than 30 years after infection. It is said that hepatitis C is responsible for about 90% of liver cancer. In Japan, more than 30,000 patients die from liver cancer associated with HCV infection every year.
[0003] HCVは 1989年に輸血後の非 A非 B型肝炎の主要な原因ウィルスとして発見され た。 HCVはエンベロープを有する RNAウィルスであり、そのゲノムは 1本鎖( + )RN Aからなり、フラビウィルス科のへパチウィルス(Hepacivirus)属に分類される。  [0003] HCV was discovered in 1989 as a major causative virus for non-A non-B hepatitis after blood transfusion. HCV is an enveloped RNA virus whose genome consists of single-stranded (+) RNA and is classified into the genus Hepacivirus of the Flaviviridae family.
[0004] HCVは、いまだ明らかでない原因により宿主の免疫機構を回避するため、免疫機 構の発達した大人に感染した場合でも持続感染が成立することが多ぐ慢性肝炎、 肝硬変、肝癌へと進行し、手術により摘出しても、非癌部で引き続き起こる炎症のた め肝癌が再発する患者が多 、ことも知られて 、る。  [0004] HCV progresses to chronic hepatitis, liver cirrhosis, and liver cancer, where persistent infection is often established even when infected with an adult who has developed an immune system, in order to avoid the host's immune mechanism due to unclear reasons. However, it is also known that many patients with liver cancer recurrence due to inflammation that continues in non-cancerous areas even after surgery.
[0005] よって、 C型肝炎の有効な治療法の確立が望まれており、その中でも、抗炎症剤に より炎症を抑える対処療法とは別に、患部である肝臓において HCVを減らすあるい は根絶させる薬剤の開発が強く望まれている。  [0005] Therefore, establishment of an effective treatment method for hepatitis C is desired. Among them, apart from coping therapy that suppresses inflammation with anti-inflammatory agents, HCV is reduced or eradicated in the affected liver. There is a strong demand for development of drugs that can be used.
[0006] 現在、 HCV排除の唯一の有効な治療法としてインターフェロン治療が知られてい る。しかしインターフェロンが有効な患者は、全患者の 1Z3程度である。特に HCVゲ ノタイプ lbに対するインターフェロンの奏効率は非常に低い。従って、インターフエ口 ンに代わる、もしくはそれと併用し得る抗 HCV薬の開発が強く望まれている。  [0006] Currently, interferon treatment is known as the only effective treatment for HCV elimination. However, about 1Z3 of all patients are effective for interferon. In particular, the response rate of interferon against HCV genotype lb is very low. Therefore, the development of anti-HCV drugs that can replace or be used in combination with the interface is strongly desired.
[0007] 近年、リバビリン(Ribavirin: 1— β— D—リボフラノシル一 1Η— 1, 2, 4—トリァゾー ルー 3—カルボキシアミド)がインターフェロンとの併用による C型肝炎治療薬として巿 販されているが、有効率は依然低ぐ更なる新規な C型肝炎治療薬が望まれている。 また、インターフェロンァゴニスト、インターロイキン 12ァゴニストなど、唐、者の免疫 力を増強させることによってウィルスを排除する手段も試みられているが、いまだ有効 とされる薬剤は見出されていない。 [0007] In recent years, ribavirin: 1—β-D-ribofuranosyl 1 一 — 1, 2, 4-triazol (Lu-3-Carboxamide) is marketed as a therapeutic agent for hepatitis C in combination with interferon, but there is a need for a new therapeutic agent for hepatitis C whose effectiveness is still low. In addition, attempts have been made to eliminate viruses by enhancing the immunity of people such as interferon agonists and interleukin 12 agonists, but no effective drugs have been found yet.
[0008] HCV遺伝子がクローユングされて以来、ウィルス遺伝子の機構と機能、各ウィルス のタンパク質の機能などにっ 、ての分子生物学的解析は急速に進展したが、ホスト 細胞内でのウィルスの複製、持続感染、病原性などのメカニズムは十分に解明され ておらず、信頼できる培養細胞を用いた HCV感染実験系は構築されて 、な力つた。 従って従来、抗 HCV薬の評価をするにあたり他の近縁ウィルスを用いた代替ウィル スアツセィ法を用いなければならな力つた。  [0008] Since the HCV gene has been cloned, molecular biological analysis has progressed rapidly due to the mechanism and function of the viral gene and the function of the protein of each virus. However, mechanisms such as persistent infection and pathogenicity have not been fully elucidated, and a reliable HCV infection experiment system using cultured cells has been established and has been very powerful. Therefore, in the past, when evaluating anti-HCV drugs, it was necessary to use an alternative virus-assy method using other closely related viruses.
[0009] しカゝし近年、 HCVの非構造領域部分を用いてインビトロでの HCV複製を観測する ことが可能になったことにより、レブリコンアツセィ法によって抗 HCV薬を容易に評価 することができるようになった (非特許文献 1)。この系での HCV RNA複製のメカ- ズムは、肝細胞に感染した全長 HCV RNAゲノムの複製と同一であると考えられて いる。従って、この系は、 HCVの複製を阻害する化合物の同定に有用な細胞に基づ くアツセィ系ということができる。  [0009] In recent years, it has become possible to observe HCV replication in vitro using non-structural regions of HCV, so that anti-HCV drugs can be easily evaluated by the levicon assembly assay. (Non-Patent Document 1). The mechanism of HCV RNA replication in this system is thought to be identical to the replication of the full-length HCV RNA genome infected with hepatocytes. Therefore, this system can be said to be a cell-based accessory system useful for identifying compounds that inhibit HCV replication.
[0010] なお、本出願の発明に関連する先行技術文献情報を以下に示す。  [0010] Information on prior art documents related to the invention of the present application is shown below.
特許文献 1:国際公開公報 W098/56755号パンフレット  Patent Document 1: International Publication No. W098 / 56755 Pamphlet
特許文献 2:国際公開公報 WO04/71503号パンフレット  Patent Document 2: International Publication No. WO04 / 71503 Pamphlet
特許文献 3:国際公開公報 WO05/05372号パンフレット  Patent Document 3: International Publication WO05 / 05372 Pamphlet
非特許文献 1 :ブイ'ローマンなど著、サイエンス(Science)、 1999年、第 285卷、第 110 -113頁  Non-Patent Document 1: Buoy Roman et al., Science, 1999, No. 285, 110-113
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0011] 本願発明者らは、国際公開公報 W098/56755号 (特許文献 1)に開示されており、 オーレォバシディウム (Aureobasidium)属などの微生物に由来する一連の化合物が 、上記レブリコンアツセィ法で高 、HCVの複製阻害活性を有することを見出した (特 許文献 2)。また本願発明者らは、該化合物がインビトロの細胞毒性については軽微 であり、 HCV感染症の予防剤または治療剤として極めて有用であることを見出し、さ らに該化合物および誘導体の合成方法を構築した (特許文献 3)。また本願発明者ら は、スフインゴ脂質生合成が HCV感染に関与すること、およびスフインゴ脂質生合成 に関わる酵素の活性や発現を阻害する化合物が、 HCV感染症の極めて有用な治療 剤または予防剤となることを明らかにした (WO2006/16657)。 The inventors of the present application disclosed in International Publication No. W098 / 56755 (Patent Document 1), and a series of compounds derived from microorganisms such as the genus Aureobasidium, It was found by the Atsey method that it has high HCV replication inhibitory activity. Permissible literature 2). In addition, the present inventors have found that the compound has minimal in vitro cytotoxicity and is extremely useful as a prophylactic or therapeutic agent for HCV infection, and further constructed a method for synthesizing the compound and derivatives. (Patent Document 3). The inventors of the present invention also found that sphingolipid biosynthesis is involved in HCV infection, and compounds that inhibit the activity and expression of enzymes involved in sphingolipid biosynthesis are extremely useful therapeutic or preventive agents for HCV infection. It was made clear (WO2006 / 16657).
[0012] スフインゴ脂質であるスフインゴミエリンは細胞膜上のラフトの構成成分であり、イン フルェンザ、 HIV等のウィルスがラフトを介して複製される(Takeda M. et al. (2003) P NAS, 100, 25、 Lucero H. A., et al. (2004) Archives of Biochemistry and Biophysics, 426, 208、 Simons K. (1997) Nature, 387, 569、 G.— Z. Leu et al. (2004)) 0 これらのことから本発明者らは、 HCVがラフトを介して複製されると予想し、スフイン ゴ脂質を含むラフトを介した HCVウィルス複製にっ 、て、詳細な機構の解明を試み た。 [0012] Sphingomyelin, a sphingolipid, is a constituent of rafts on cell membranes, and viruses such as influenza and HIV are replicated via rafts (Takeda M. et al. (2003) P NAS, 100 , 25, Lucero HA, et al. (2004) Archives of Biochemistry and Biophysics, 426, 208, Simons K. (1997) Nature, 387, 569, G.— Z. Leu et al. (2004)) 0 Therefore, the present inventors predicted that HCV is replicated via rafts and attempted to elucidate the detailed mechanism of HCV virus replication via rafts containing sphingolipids.
[0013] 本発明は、このような状況に鑑みてなされたものであり、その目的は、スフインゴミエ リンと HCVタンパク質の結合を阻害する化合物を有効成分として含有する、 HCV感 染症を治療または予防するための薬剤を提供することにある。また、 HCV感染症を治 療または予防するための薬剤のスクリーニング方法、およびこれらに用いるためのキ ットの提供も目的とする。  [0013] The present invention has been made in view of such a situation, and an object of the present invention is to treat or prevent HCV infectious diseases containing a compound that inhibits the binding of sphingomyelin and HCV protein as an active ingredient. It is to provide a drug to do. Another object of the present invention is to provide a method for screening a drug for treating or preventing HCV infection and a kit for use in the method.
課題を解決するための手段  Means for solving the problem
[0014] 上記課題を解決するために、本願発明者らは HCVタンパク質およびスフインゴ脂質 [0014] In order to solve the above problems, the inventors of the present application have developed HCV protein and sphingolipid
(スフインゴミエリン)の結合部位の特定および、結合能について検討を行った。  The binding site of (sphingomyelin) was identified and the binding ability was examined.
まず、 HCVタンパク質の中に、スフインゴ脂質結合領域があるか探索するために、 類似する構造を CEプログラムにより検索した。その結果、 NS5Bの配列 E230-G263に おいて、ヘリックス ·ターン.ヘリックスのモチーフを持った類似構造が認められた(図 1 First, in order to search for a sphingolipid-binding region in the HCV protein, a similar structure was searched by the CE program. As a result, a similar structure with a helix-turn-helix motif was observed in the NS5B sequence E230-G263 (Fig. 1).
) o ) o
[0015] 次に、 Biacore S51を用いて、 NS5Bスフインゴ結合領域(NS5B- SBD)とスフインゴミエ リン (SM)との結合能を確認したところ、 NS5B-SBDペプチドは、濃度依存的にスフイン ゴミエリンと結合することが明ら力となった(図 2A)。また、プリオンタンパク質 (PrP)に ぉ 、ても同様な結合が認められた(図 2B)。 [0015] Next, using Biacore S51, the binding ability of NS5B sphingo binding domain (NS5B-SBD) and sphingomyelin (SM) was confirmed. NS5B-SBD peptide bound to sphingomyelin in a concentration-dependent manner. It became clear that doing this (Figure 2A). In prion protein (PrP) However, similar binding was observed (Figure 2B).
さらに、 NS5Bスフインゴ結合領域ペプチドおよびその誘導体の HCVレプリコン阻害 活性を測定したところ、これらのペプチドは有意な抗 HCV複製阻害活性を示すことが 明らかとなった (図 3)。  Furthermore, when HCV replicon inhibitory activity of NS5B sphingo binding domain peptides and derivatives thereof was measured, it was revealed that these peptides exhibited significant anti-HCV replication inhibitory activity (Fig. 3).
上記の結果よりラフトの構成成分であるスフインゴミエリンと、 HCVタンパク質である HCV-NS5Bが特定の部位において強く結合すること、およびこれらの結合により HCV ウィルスの複製が行われていることが明ら力となった。また、スフインゴミエリンと HCV タンパク質の結合を阻害することにより、 HCVウィルスの複製を抑制出来ることが明ら カゝとなった。  From the above results, it is clear that Sphingomyelin, which is a component of raft, and HCV-NS5B, which is an HCV protein, bind strongly at a specific site, and that the HCV virus is replicated by their binding. It became power. Moreover, it became clear that inhibition of HCV virus replication can be suppressed by inhibiting the binding of sphingomyelin and HCV protein.
[0016] 即ち、本発明者らは、スフインゴミエリンと HCVタンパク質の結合を阻害する化合物 を有効成分として含有する、 HCV感染症を治療または予防するための薬剤を開発す ることに成功し、これにより本発明を完成するに至った。  That is, the present inventors have succeeded in developing a drug for treating or preventing HCV infection containing a compound that inhibits the binding of sphingomyelin and HCV protein as an active ingredient, Thus, the present invention has been completed.
[0017] 本発明は、より具体的には、以下の〔1〕〜〔14〕を提供する。 [0017] More specifically, the present invention provides the following [1] to [14].
〔1〕スフインゴミエリンと HCVタンパク質の結合を阻害する化合物を有効成分として含 有する、 HCV感染症を治療または予防するための薬剤。  [1] A drug for treating or preventing HCV infection, comprising as an active ingredient a compound that inhibits the binding of sphingomyelin and HCV protein.
〔2〕スフインゴミエリンと HCVタンパク質の結合を阻害する化合物力 以下の(a)また は (b)に記載のペプチドである、〔1〕に記載の薬剤。  [2] Compound ability to inhibit binding of sphingomyelin and HCV protein The drug according to [1], which is a peptide according to the following (a) or (b).
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
〔3〕スフインゴミエリンと HCVタンパク質の結合を阻害する化合物力 以下の(a)また は (b)に記載のペプチドをコードするオリゴヌクレオチドである、〔1〕に記載の薬剤。 [3] Compound ability to inhibit binding of sphingomyelin and HCV protein The drug according to [1], which is an oligonucleotide encoding the peptide according to the following (a) or (b).
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド (b) SEQ ID NO: 1 in the amino acid sequence described in any one of 4, 10 or 11, in which one or more amino acids are substituted, deleted, added, and amino acid sequence is not Z or inserted. Peptide
〔4〕スフインゴミエリンと HCVタンパク質の結合を阻害する化合物力 以下の(a)また は (b)に記載のペプチドを認識する抗体である、〔1〕に記載の薬剤。  [4] Compound ability to inhibit binding of sphingomyelin to HCV protein The drug according to [1], which is an antibody that recognizes the peptide according to (a) or (b) below.
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
〔5〕 HCVタンパク質が HCV-NS5Bである〔1〕〜〔4〕の!、ずれかに記載の薬剤。  [5] The drug according to [1] to [4], wherein the HCV protein is HCV-NS5B!
〔6〕HCV感染症が、 C型肝炎、肝硬変、肝繊維化、または肝癌である、〔1〕〜〔5〕の いずれかに記載の薬剤。 [6] The drug according to any one of [1] to [5], wherein the HCV infection is hepatitis C, cirrhosis, liver fibrosis, or liver cancer.
〔7〕以下の (A)〜 (C)の工程を含む、 HCV感染症を治療または予防するための薬剤 のスクリーニング方法。  [7] A screening method for a drug for treating or preventing HCV infection, comprising the following steps (A) to (C):
(A)以下の (a)または (b)に記載のペプチドに被検化合物を接触させる工程  (A) A step of bringing a test compound into contact with the peptide according to the following (a) or (b)
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
(B) (A)に記載のペプチドと被検化合物の結合を検出する工程  (B) a step of detecting the binding between the peptide according to (A) and a test compound
(C) (A)に記載のペプチドと結合する被検化合物を選択する工程  (C) a step of selecting a test compound that binds to the peptide according to (A)
〔8〕以下の (A)〜 (C)の工程を含む、 HCV感染症を治療または予防するための薬剤 のスクリーニング方法。  [8] A method for screening a drug for treating or preventing HCV infection, comprising the following steps (A) to (C):
(A)被検化合物を以下の(a)または (b)に記載のペプチドと同時に、スフインゴミエリ ンに添加する工程  (A) A step of adding the test compound to the sphingomyelin simultaneously with the peptide described in (a) or (b) below
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド (b) SEQ ID NO: 1 in the amino acid sequence described in any one of 4, 10, or 11 Is a peptide with multiple amino acid substitutions, deletions, additions, and Z or inserted amino acid sequence
(B)上記 (a)または (b)に記載のペプチドと、スフインゴミエリンの結合能を測定する 工程  (B) a step of measuring the binding ability of the peptide according to (a) or (b) above and sphingomyelin
(C)被検化合物を添加しな 、場合に比べて、上記結合能を低下させた被検化合物 を選択する工程  (C) a step of selecting a test compound having a reduced binding ability compared to the case without adding a test compound
〔9〕HCV感染症が、 C型肝炎、肝硬変、肝繊維化、または肝癌である、〔7〕または〔8〕 に記載のスクリーニング方法。  [9] The screening method according to [7] or [8], wherein the HCV infection is hepatitis C, cirrhosis, liver fibrosis, or liver cancer.
〔10〕〔7〕〜〔9〕の 、ずれかに記載のスクリーニング方法に用いるためのキット。  [10] A kit for use in the screening method according to any one of [7] to [9].
〔11〕以下の (A)〜 (C)の工程を含む、 HCV感染症を治療または予防するための薬 剤の効力の評価方法。 [11] A method for evaluating the efficacy of a drug for treating or preventing HCV infection, comprising the following steps (A) to (C):
(A)以下の (a)または (b)に記載のペプチドに被検化合物を接触させる工程  (A) A step of bringing a test compound into contact with the peptide according to the following (a) or (b)
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
(B) (A)に記載のペプチドと被検化合物の結合を検出する工程  (B) a step of detecting the binding between the peptide according to (A) and a test compound
(C) (A)に記載のペプチドと結合する被検化合物の、 HCV感染症の治療または予防 効果の効力を評価する工程  (C) A step of evaluating the efficacy of the therapeutic or prophylactic effect of HCV infection of a test compound that binds to the peptide of (A)
[12〕以下の (A)および (B)の工程を含む、 HCV感染症を治療または予防するため の薬剤の効力の評価方法。  [12] A method for evaluating the efficacy of a drug for treating or preventing HCV infection, comprising the following steps (A) and (B):
(A)被検化合物を以下の(a)または (b)に記載のペプチドと同時に、スフインゴミエリ ンに添加する工程  (A) A step of adding the test compound to the sphingomyelin simultaneously with the peptide described in (a) or (b) below
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド (b) SEQ ID NO: 1 in the amino acid sequence described in any one of 4, 10 or 11, in which one or more amino acids are substituted, deleted, added, and amino acid sequence is not Z or inserted. Peptide
(B)上記 (a)または (b)に記載のペプチドと、スフインゴミエリンの結合能を測定するこ とによる、 HCV感染症の治療または予防効果の効力を評価する工程  (B) A step of evaluating the efficacy of the treatment or prevention effect of HCV infection by measuring the binding ability of the peptide described in (a) or (b) above and sphingomyelin.
〔 13〕〔 11〕または〔 12〕の 、ずれかに記載の評価方法に用 、るためのキット。 [13] A kit for use in the evaluation method according to any one of [11] and [12].
〔14〕以下の(a)または (b)に記載のペプチド。 [14] The peptide according to the following (a) or (b).
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
図面の簡単な説明 Brief Description of Drawings
[図 1]Aは、 HCV- NS5Bの配列 E230- G263および V3ループのアミノ酸配列を示す図 である。 Bは、 HCV- NS5Bの構造を示す図である。縦線は HCV- NS5Bの同定した推 定のスフインゴ脂質結合領域。斜線は HIV-1 V3ループのスフインゴ脂質結合領域を 示す。 Cは、 HCV-NS5Bにおけるへリックス 'ターン'ヘリックスのモチーフを示す図で ある。 FIG. 1A is a view showing the amino acid sequences of HCV-NS5B sequences E230-G263 and V3 loop. B is a diagram showing the structure of HCV-NS5B. The vertical line is the predicted sphingolipid binding region identified by HCV-NS5B. The diagonal line indicates the sphingolipid binding region of the HIV-1 V3 loop. C shows the helix 'turn' helix motif in HCV-NS5B.
[図 2]Biacoreを用いた HCV- NS5Bスフインゴ結合領域ペプチド(NS5B- SBD、配列番 号: 1)とスフインゴミエリン(SM)との結合検討の結果を示す図である。 Aは、 NS5B-SB Dペプチドのセンサグラムを示す図であり、 NS5B-SBDペプチドは濃度依存的にスフィ ンゴミエリンとの結合を認めた。 Bは、 NS5B-SBDペプチドとプリオンタンパク質(PrP、 配列番号: 2)の、スフインゴミエリンへの結合能を比較したものである。センサグラム 8 0s時の RU値を示す。  FIG. 2 is a diagram showing the results of a binding study of HCV-NS5B sphingo binding domain peptide (NS5B-SBD, SEQ ID NO: 1) and sphingomyelin (SM) using Biacore. A is a diagram showing a sensorgram of NS5B-SB D peptide, and NS5B-SBD peptide was found to bind to sphingomyelin in a concentration-dependent manner. B is a comparison of the binding ability of NS5B-SBD peptide and prion protein (PrP, SEQ ID NO: 2) to sphingomyelin. Sensorgram Shows the RU value at 80 s.
[図 3]NS5Bスフインゴ結合領域ペプチドおよびその誘導体の HCVレプリコン阻害活性 測定の結果を示す図である。  FIG. 3 shows the results of measuring HCV replicon inhibitory activity of NS5B sphingo-binding domain peptides and their derivatives.
[図 4]スフインゴ脂質合成経路 (パルミトイル CoAからスフインゴミエリンまでの合成経 路)を示す図である。  FIG. 4 is a diagram showing a sphingolipid synthetic pathway (synthetic pathway from palmitoyl CoA to sphingomyelin).
[図 5]ノザンプロット解析によるミリオシンの HCV RNA複製阻害活性を示す写真であ る。横軸はミリオシンの濃度を表す。 [図 6]ノザンプロット解析による式 (II)で表される化合物の HCV RNA複製阻害活性を 示す写真である。横軸は式 (Π)で表される化合物の濃度を表す。 FIG. 5 is a photograph showing the HCV RNA replication inhibitory activity of myriocin by Northern plot analysis. The horizontal axis represents the concentration of myriocin. FIG. 6 is a photograph showing the HCV RNA replication inhibitory activity of the compound represented by formula (II) by Northern plot analysis. The horizontal axis represents the concentration of the compound represented by the formula (Π).
[図 7]ウェスタンプロット解析によるミリオシンの HCVタンパク合成阻害活性を示す写 真である。横軸はミリオシンの濃度を表す。 [Fig. 7] A photograph showing the inhibitory activity of myriocin on HCV protein synthesis by Western plot analysis. The horizontal axis represents the concentration of myriocin.
[図 8]ウェスタンプロット解析による式 (Π)で表される化合物の HCVタンパク合成阻害 活性を示す写真である。横軸は式 (II)で表される化合物の濃度を表す。  FIG. 8 is a photograph showing the HCV protein synthesis inhibitory activity of the compound represented by formula (Π) by Western plot analysis. The horizontal axis represents the concentration of the compound represented by formula (II).
[図 9]フモ-シン B1の HCVレプリコン阻害活性を示す図である。  FIG. 9 is a graph showing the HCV replicon inhibitory activity of fumosin B1.
[図 10]siRNAによるセリンパルミトイル転移酵素(LCB1)のタンパク質発現阻害を示す 写真である。  FIG. 10 is a photograph showing inhibition of protein expression of serine palmitoyltransferase (LCB1) by siRNA.
[図 ll]siRNAによる HCVレブリコン阻害活性および細胞毒性の影響を示す図である。  [Fig. Ll] A graph showing the effects of siRNA on the HCV replicon inhibitory activity and cytotoxicity.
[図 12]式 (II)で表される化合物による HCVレブリコンの阻害及び宿主細胞への毒性 を示す図である。 FIG. 12 is a graph showing the inhibition of HCV levicon and the toxicity to host cells by the compound represented by formula (II).
[図 13]式 (II)で表される化合物による HCV-NS3タンパク質の発現阻害を示す写真で ある。免疫染色の後、蛍光顕微鏡にて観察した。白色は NS3タンパク質、灰色はへキ スト 33342で染色された核を示す。  FIG. 13 is a photograph showing inhibition of HCV-NS3 protein expression by a compound represented by formula (II). After immunostaining, it was observed with a fluorescence microscope. White indicates NS3 protein, gray indicates nuclei stained with hex 33342.
[図 14]式(II)で表される化合物による NS3、 NS5A、および NS-5Bタンパク質の発現阻 害を示す写真である。各タンパク質の発現は、ウェスタンプロット解析により行った。  FIG. 14 is a photograph showing inhibition of NS3, NS5A, and NS-5B protein expression by a compound represented by formula (II). Each protein was expressed by Western plot analysis.
[図 15]式 (II)で表される化合物による SPT阻害活性を示す図である。 FIG. 15 is a graph showing SPT inhibitory activity of a compound represented by formula (II).
[図 16]式 (II)で表される化合物によるセラミド、スフインゴミエリンの de novo合成阻害 を示す写真である。 FIG. 16 is a photograph showing inhibition of de novo synthesis of ceramide and sphingomyelin by the compound represented by formula (II).
[図 17]C2—セラミドによる式 (II)で表される化合物の HCV複製阻害の抑制を示す写 真である。  FIG. 17 is a photograph showing suppression of HCV replication inhibition of a compound represented by formula (II) by C2-ceramide.
[図 18]ラフト生合成関連低分子化合物による HCV複製阻害を示す図である。  FIG. 18 shows inhibition of HCV replication by a raft biosynthesis-related low molecular weight compound.
[図 19]式 (II)で表される化合物によるラフトタンパク質への影響を示す写真である。  FIG. 19 is a photograph showing the influence of a compound represented by formula (II) on raft protein.
[図 20]式 (II)で表される化合物によるラフトタンパク質への影響を示す図である。 発明を実施するための最良の形態  FIG. 20 is a diagram showing the influence of a compound represented by formula (II) on raft protein. BEST MODE FOR CARRYING OUT THE INVENTION
本発明は、スフインゴミエリンと HCVタンパク質の結合を阻害する化合物を有効成 分として含有する、 HCV感染症を治療または予防するための薬剤に関する。 [0020] 本発明にお 、て、スフインゴミエリンと HCVタンパク質の結合を阻害する化合物とは 、スフインゴミエリンと HCVタンパク質の結合反応を、直接的もしくは間接的に阻害す るものであれば如何なる化合物であってもよい。また、これらの阻害剤を生成または 増加させる化合物であって、スフインゴミエリンと HCVタンパク質の結合反応を間接的 に阻害する化合物であってもよ 、。 The present invention relates to a drug for treating or preventing HCV infection, which contains a compound that inhibits the binding of sphingomyelin and HCV protein as an active ingredient. [0020] In the present invention, the compound that inhibits the binding of sphingomyelin and HCV protein is any compound that can directly or indirectly inhibit the binding reaction of sphingomyelin and HCV protein. It may be a compound. Further, it may be a compound that generates or increases these inhibitors and indirectly inhibits the binding reaction between sphingomyelin and HCV protein.
[0021] 本発明にお!/、て、 HCVタンパク質としては、 HCV- NS2、 HCV- NS3、 HCV- NS4A、 H CV- NS4B、 HCV- NS5A、 HCV- NS5Bを挙げることが出来る力 より好ましくは、 HCV- NS5Bを挙げることができる。  [0021] In the present invention! As HCV proteins, HCV-NS2, HCV-NS3, HCV-NS4A, HCV-NS4B, HCV-NS5A, HCV-NS5B can be mentioned, more preferably HCV-NS5B. it can.
[0022] 本発明のスフインゴミエリンと HCVタンパク質の結合を阻害する化合物の好ま ヽ例 としては、以下の(a)または (b)に記載のペプチドを挙げることができる。  [0022] Preferable examples of the compound that inhibits the binding of sphingomyelin and HCV protein of the present invention include the peptides described in the following (a) or (b).
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
[0023] 配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列において例えば 1〜10個、好ましくは 1〜5個のアミノ酸残基力 置換、欠失、付加、および Zまたは挿 入されたアミノ酸配列を含み、配列番号: 1から 4、 10または 11のいずれかに記載の アミノ酸配列からなるペプチドと機能的に同等なペプチドもこれらのペプチドに含ま れる。変異するアミノ酸残基においては、アミノ酸側鎖の性質が保存されている別の アミノ酸に変異されることが望ましい。例えばアミノ酸側鎖の性質としては、疎水性アミ ノ酸 (A、 I、し、 M、 F、 P、 W、 Y、 V)、親水性アミノ酸(R、 D、 N、 C、 E、 Q、 G、 H、 K、 S、 T )、脂肪族側鎖を有するアミノ酸 (G、 A、 V、 L、 I、 P)、水酸基含有側鎖を有するァミノ 酸 (S、 T、 Υ)、硫黄原子含有側鎖を有するアミノ酸 (C、 M)、カルボン酸及びアミド含 有側鎖を有するアミノ酸 (D、 N、 E、 Q)、塩基含有側鎖を有するアミノ酸 (R、 K、 Η)、 芳香族含有側鎖を有するアミノ酸 (H、 F、 Y、 W)を挙げることができる (括弧内はいず れもアミノ酸の一文字標記を表す)。あるアミノ酸配列に対する 1又は複数個のァミノ 酸残基の欠失、付加及び Z又は他のアミノ酸による置換により修飾されたアミノ酸配 列を有するポリペプチドがその生物学的活性を維持することはすでに知られている。 [0023] SEQ ID NO: 1 to 4, 10 or 11, for example, 1 to 10, preferably 1 to 5 amino acid residues in the amino acid sequence substitution, deletion, addition, and Z or insertion These peptides also include peptides functionally equivalent to the peptides comprising the amino acid sequences described in any one of SEQ ID NOs: 1 to 4, 10 or 11, including the entered amino acid sequence. The amino acid residue to be mutated is preferably mutated to another amino acid in which the properties of the amino acid side chain are conserved. For example, amino acid side chain properties include hydrophobic amino acids (A, I, M, F, P, W, Y, V), hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, T), amino acids having aliphatic side chains (G, A, V, L, I, P), amino acids having hydroxyl-containing side chains (S, T, Υ), sulfur atoms Amino acids with side chains (C, M), amino acids with side chains containing carboxylic acids and amides (D, N, E, Q), amino acids with side chains (R, K, Η), aromatics Mention may be made of amino acids having a side chain (H, F, Y, W) (in parentheses all represent one letter of the amino acid). An amino acid sequence modified by deletion or addition of one or more amino acid residues to a certain amino acid sequence, and substitution by Z or another amino acid. It is already known that a polypeptide having a sequence maintains its biological activity.
[0024] 配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチドとしては、配列番号: 5〜7のいずれかに記載のアミノ酸力 なるペプチド を挙げることが出来る。  [0024] SEQ ID NO: 1 to 4, 10 or 11, in the amino acid sequence described in any one or more of the amino acid substitution, deletion, addition and a peptide having an amino acid sequence ability that is Z or inserted, Examples thereof include peptides having amino acid strength described in any one of SEQ ID NOs: 5 to 7.
これらのペプチドは、スフインゴミエリンにおける HCVタンパク質結合部位に特異的 に結合し、そのためスフインゴミエリンと HCVタンパク質の結合活性を低下させること が考えられる。その結果として、ラフトを介した HCVウィルスの複製が抑制されるもの と考えられる。  These peptides may specifically bind to the HCV protein binding site in sphingomyelin, and thus may reduce the binding activity of sphingomyelin and HCV protein. As a result, it is considered that replication of HCV virus via rafts is suppressed.
[0025] 本発明のスフインゴミエリンと HCVタンパク質の結合を阻害する化合物の好ま ヽ例 としては、以下の(a)または (b)に記載のペプチドをコードするオリゴヌクレオチドを挙 げることが出来る。  [0025] Preferred examples of the compound that inhibits the binding of sphingomyelin and HCV protein of the present invention include the oligonucleotides encoding the peptides described in (a) or (b) below. .
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
本発明のオリゴヌクレオチドは、該ペプチドを発現しうる形で適当なベクターに組み 込んで用いることもできる。  The oligonucleotide of the present invention can be used by incorporating it into an appropriate vector in a form capable of expressing the peptide.
[0026] 本明細書中、該ペプチドをコードするポリヌクレオチドを「発現しうる形で含む」とは、 該ポリヌクレオチドが発現ベクターに挿入されており、動物の細胞内に侵入した場合 に、該細胞内で所定のペプチドを発現させうる形態で含むことをいう。すなわち、例え ば、投与される動物種および投与部位に適したプロモータの制御下に該コード DNA が配置されていることを指す。 [0026] In the present specification, the phrase "comprising an expressible form" of a polynucleotide encoding the peptide means that the polynucleotide is inserted into an expression vector and enters the animal cell. It means that a predetermined peptide can be expressed in a cell. That is, for example, it means that the coding DNA is arranged under the control of a promoter suitable for the species of animal to be administered and the administration site.
[0027] 本発明のスフインゴミエリンと HCVタンパク質の結合を阻害する化合物の好ま ヽ例 としては、以下の(a)または (b)に記載のペプチドを認識する抗体を挙げることが出来 る。 [0027] Preferable examples of the compound that inhibits the binding of sphingomyelin and HCV protein of the present invention include antibodies that recognize the peptides described in (a) or (b) below.
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド (a) SEQ ID NO: 1 to 4, 10, or 11 Chido
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
本発明における抗体の由来は特に限定されるものではな 、が、好ましくは哺乳動 物由来であり、より好ましくはヒト由来の抗体を挙げることが出来る。  The origin of the antibody in the present invention is not particularly limited, but is preferably derived from a mammal, more preferably a human-derived antibody.
[0028] 本発明で使用される該ペプチドを認識する抗体は、公知の手段を用いてポリクロー ナル又はモノクローナル抗体として得ることができる。本発明で使用される該ペプチド を認識する抗体として、特に哺乳動物由来のモノクローナル抗体が好ましい。哺乳動 物由来のモノクローナル抗体としては、ハイプリドーマに産生されるもの、および遺伝 子工学的手法により抗体遺伝子を含む発現ベクターで形質転換した宿主に産生さ れるものがある。この抗体は HCVタンパク質と結合することにより、 HCVタンパク質とス フインゴミエリンの結合を阻害する。その結果として、ラフトを介した HCVウィルスの複 製が抑制されるものと考えられる。  [0028] An antibody recognizing the peptide used in the present invention can be obtained as a polyclonal or monoclonal antibody using a known means. As the antibody recognizing the peptide used in the present invention, a monoclonal antibody derived from a mammal is particularly preferable. Monoclonal antibodies derived from mammals include those produced by hyperpridoma and those produced by a host transformed with an expression vector containing an antibody gene by genetic engineering techniques. This antibody inhibits the binding of HCV protein to sphingomyelin by binding to HCV protein. As a result, replication of HCV virus via rafts is thought to be suppressed.
[0029] 該ペプチドを認識する抗体産生ハイブリドーマは、基本的には公知技術を使用し、 以下のようにして作製できる。すなわち、該ペプチドを感作抗原として使用して、これ を通常の免疫方法にしたがって免疫し、得られる免疫細胞を通常の細胞融合法によ つて公知の親細胞と融合させ、通常のスクリーニング法により、モノクローナルな抗体 産生細胞をスクリーニングすることによって作製できる。  [0029] An antibody-producing hybridoma that recognizes the peptide can be basically produced using a known technique as follows. That is, the peptide is used as a sensitizing antigen, and this is immunized according to a normal immunization method. The obtained immune cells are fused with a known parent cell by a normal cell fusion method, and a normal screening method is used. It can be produced by screening monoclonal antibody-producing cells.
[0030] 具体的には、該ペプチドを認識する抗体を作製するには次のようにすればよ!、。  [0030] Specifically, an antibody that recognizes the peptide may be prepared as follows!
該ペプチドをコードする塩基配列を公知の発現ベクター系に挿入して適当な宿主 細胞を形質転換させた後、その宿主細胞中又は、培養上清中から目的の該ペプチド を公知の方法で精製し、この精製ペプチドを感作抗原として用いればよい。また、該 ペプチドと他の蛋白質との融合蛋白質を感作抗原として用 、てもよ 、。  After inserting a base sequence encoding the peptide into a known expression vector system and transforming an appropriate host cell, the peptide of interest is purified from the host cell or culture supernatant by a known method. This purified peptide may be used as a sensitizing antigen. In addition, a fusion protein of the peptide and another protein may be used as a sensitizing antigen.
[0031] 本明細書において「治療」という記載は、本発明の薬物を被験者に投与することに よって、 HCVを消滅あるいは軽減させること、さらなる HCVの広がりを抑制すること、 HCVの感染による症状を軽減することを意味する。また「予防」という記載は、 HCVが 感染する前に、被験者に投与され、 HCVの感染を防いだり、増殖を抑制させることを 意味する。 HCVの感染による症状としては、好ましくは C型肝炎、肝硬変、肝繊維化 、肝癌などが挙げられる。 [0031] In the present specification, the term "treatment" refers to the administration of the drug of the present invention to a subject to eliminate or reduce HCV, further suppress the spread of HCV, and symptoms caused by HCV infection. Means to reduce. The term “prevention” refers to administration to a subject prior to HCV infection to prevent HCV infection or suppress proliferation. means. Symptoms caused by HCV infection preferably include hepatitis C, cirrhosis, liver fibrosis, liver cancer and the like.
[0032] 本発明の化合物は、医薬に使用することができる。上記塩としては薬理学的に許容 されるものであれば特に限定されず、例えば、塩酸、硫酸、硝酸、リン酸、臭化水素 酸などの鉱酸との塩;酢酸、酒石酸、乳酸、クェン酸、フマル酸、マレイン酸、コハク 酸、メタンスルホン酸、エタンスルホン酸、ベンゼンスルホン酸、トルエンスルホン酸、 ナフタレンスルホン酸、ショウノウスルホン酸などの有機酸との塩;ナトリウム、カリウム 、カルシウムなどのアルカリ金属又はアルカリ土類金属などとの塩などを挙げることが できる。 [0032] The compound of the present invention can be used in medicine. The salt is not particularly limited as long as it is pharmacologically acceptable. For example, salts with mineral acids such as hydrochloric acid, sulfuric acid, nitric acid, phosphoric acid, hydrobromic acid; acetic acid, tartaric acid, lactic acid, ken Acid, fumaric acid, maleic acid, succinic acid, methane sulfonic acid, ethane sulfonic acid, benzene sulfonic acid, toluene sulfonic acid, naphthalene sulfonic acid, salt with organic acid such as camphor sulfonic acid; alkali such as sodium, potassium and calcium Examples thereof include salts with metals or alkaline earth metals.
上記医薬製剤に含まれる有効成分ィ匕合物の量は、特に限定されず広範囲に適宜 選択される力 例えば、 0. 1-99. 5重量%、好ましくは 0. 5〜90重量%である。  The amount of the active ingredient-compound contained in the above-mentioned pharmaceutical preparation is not particularly limited and can be selected as appropriate over a wide range. For example, 0.1 to 99.5% by weight, preferably 0.5 to 90% by weight. .
[0033] 本発明の化合物を、常法に従って主薬として、賦形剤、結合剤、崩壊剤、滑沢剤、 矯味矯臭剤、溶解補助剤、懸濁剤、コーティング剤などの医薬の製剤技術分野にお いて通常使用し得る既知の補助剤を用いて製剤化することができる。錠剤の形態に 成形するに際しては、担体としてこの分野で従来公知のものを広く使用でき、例えば 乳糖、白糖、塩ィ匕ナトリウム、グルコース、尿素、澱粉、炭酸カルシウム、カオリン、結 晶セルロース、ケィ酸などの賦形剤;水、エタノール、プロパノール、単シロップ、グル コース液、澱粉液、ゼラチン溶液、カルボキシメチルセルロース、セラック、メチルセル ロース、リン酸カリウム、ポリビュルピロリドンなどの結合剤;乾燥澱粉、アルギン酸ナト リウム、寒天末、ラミナラン末、炭酸水素ナトリウム、炭酸カルシウム、ポリオキシェチレ ンソルビタン脂肪酸エステル、ラウリル硫酸ナトリウム、ステアリン酸モノグリセリド、澱 粉、乳糖などの崩壊剤;白糖、ステアリン、カカオバター、水素添加油などの崩壊抑 制剤;第 4級アンモ-ゥム塩類、ラウリル硫酸ナトリウムなどの吸収促進剤;グリセリン、 澱粉などの保湿剤;澱粉、乳糖、カオリン、ベントナイト、コロイド状ケィ酸などの吸着 剤;精製タルク、ステアリン酸塩、硼酸末、ポリエチレングリコールなどの潤沢剤などが 例示できる。 [0033] The pharmaceutical preparation technical field such as excipients, binders, disintegrants, lubricants, flavoring agents, solubilizers, suspension agents, coating agents, etc., with the compound of the present invention as the main agent according to conventional methods. In this case, it can be formulated using known adjuvants that can be usually used. In the case of forming into a tablet form, conventionally known carriers can be widely used as carriers, such as lactose, sucrose, sodium chloride sodium salt, glucose, urea, starch, calcium carbonate, kaolin, crystalline cellulose, and key acid. Excipients such as water, ethanol, propanol, simple syrup, glucose solution, starch solution, gelatin solution, binders such as carboxymethylcellulose, shellac, methylcellulose, potassium phosphate, polybulurpyrrolidone; dry starch, sodium alginate Disintegrants such as lithium, agar powder, laminaran powder, sodium bicarbonate, calcium carbonate, polyoxyethylene sorbitan fatty acid ester, sodium lauryl sulfate, stearic acid monoglyceride, starch, lactose; white sugar, stearin, cocoa butter, hydrogenated oil, etc. Collapse inhibitor; 4th Absorption accelerators such as ammonium salts and sodium lauryl sulfate; humectants such as glycerin and starch; adsorbents such as starch, lactose, kaolin, bentonite and colloidal key acid; purified talc, stearate, boric acid powder, Examples include a lubricant such as polyethylene glycol.
[0034] さらに錠剤は、必要に応じ、通常の剤皮を施した錠剤、例えば、糖衣錠、ゼラチン 被包錠、腸溶被錠、フィルムコーティング錠あるいは二重錠、多層錠とすることができ る。丸剤の形態に成形するに際しては、担体としてこの分野で従来公知のものを広く 使用でき、例えばグルコース、乳糖、カカオバター、澱粉、硬化植物油、カオリン、タ ルクなどの賦形剤;アラビアゴム末、トラガント末、ゼラチン、エタノールなどの結合剤; ラミナラン寒天などの崩壊剤などが例示できる。坐剤の形態に成形するに際しては、 担体としてこの分野で従来公知のものを広く使用でき、例えばポリエチレングリコール 、カカオバター、高級アルコール、高級アルコールのエステル類、ゼラチン、半合成 グリセリドなどを挙げることができる。注射剤として調製される場合には、液剤および 懸濁剤は殺菌され、かっ血液と等張であるのが好ましぐこれら液剤、乳剤および懸 濁剤の形態に成形するに際しては、希釈剤としてこの分野で慣用されているものをす ベて使用でき、例えば、水、エタノール、プロピレングリコール、エトキシ化イソステアリ ルアルコール、ポリオキシ化イソステアリルアルコール、ポリオキシエチレンソルビタン 脂肪酸エステル類などを挙げることができる。なお、この場合、等張性の溶液を調製 するのに充分な量の食塩、グルコース、あるいはグリセリンを医薬製剤中に含有せし めてもよく、また通常の溶解補助剤、緩衝剤、無痛化剤などを添加してもよい。さらに 必要に応じて着色剤、保存剤、香料、風味剤、甘味剤などや他の医薬品を含有する ことちでさる。 [0034] Furthermore, the tablets can be made into tablets with ordinary coatings, for example, sugar-coated tablets, gelatin-encapsulated tablets, enteric-coated tablets, film-coated tablets or double tablets, and multilayer tablets as necessary. The In molding into the form of a pill, those conventionally known in this field can be widely used as a carrier. For example, excipients such as glucose, lactose, cocoa butter, starch, hydrogenated vegetable oil, kaolin, and tar; And binders such as tragacanth powder, gelatin and ethanol; and disintegrants such as laminaran agar. In the case of forming into a suppository, conventionally known carriers can be widely used as carriers, such as polyethylene glycol, cocoa butter, higher alcohols, higher alcohol esters, gelatin, semi-synthetic glycerides and the like. it can. When prepared as injections, solutions and suspensions should be sterilized and used as diluents in the form of these solutions, emulsions and suspensions, which are preferably isotonic with blood. Any of those commonly used in this field can be used, and examples thereof include water, ethanol, propylene glycol, ethoxylated isostearyl alcohol, polyoxylated isostearyl alcohol, and polyoxyethylene sorbitan fatty acid esters. In this case, a sufficient amount of sodium chloride, glucose, or glycerin to prepare an isotonic solution may be contained in the pharmaceutical preparation, and a normal solubilizing agent, buffering agent, soothing agent may be used. An agent or the like may be added. In addition, it may contain colorants, preservatives, fragrances, flavors, sweeteners, and other medicines as necessary.
[0035] 上記医薬組成物は、投与単位形態で投与することが好ましぐ経口投与、組織内 投与 (皮下投与、筋肉内投与、静脈内投与など)、局所投与 (経皮投与など)又は経 直腸的に投与することができる。上記医薬組成物は、これらの投与方法に適した剤 型で投与されることは当然である。  [0035] The pharmaceutical composition is preferably administered in a dosage unit form for oral administration, tissue administration (subcutaneous administration, intramuscular administration, intravenous administration, etc.), topical administration (transdermal administration, etc.) Can be administered rectally. Of course, the pharmaceutical composition is administered in a dosage form suitable for these administration methods.
[0036] 本発明の化合物を医薬として投与する場合、抗ウィルス剤としての用量は、年齢、 体重などの患者の状態、投与経路、病気の性質と程度などを考慮した上で調整する ことが望ましいが、通常は、ヒトについては、成人に対して本発明の有効成分量として 、一日当たり、 0.1〜2000mgの範囲である。上記範囲未満の用量で足りる場合もある 力 逆に上記範囲を超える用量を必要とする場合もある。多量に投与するときは、一 日数回に分割して投与することが望ましい。  [0036] When the compound of the present invention is administered as a medicine, the dose as an antiviral agent is preferably adjusted in consideration of the patient's condition such as age and weight, administration route, nature and degree of disease, etc. However, for humans, the amount of the active ingredient of the present invention for adults is usually in the range of 0.1 to 2000 mg per day. In some cases, doses below the above range may be sufficient. Conversely, doses in excess of the above range may be required. When administering a large amount, it is desirable to divide it into several times a day.
[0037] 上記経口投与は、固形、粉末又は液状の用量単位で行うことができ、例えば、末剤 、散剤、錠剤、糖衣剤、カプセル剤、ドロップ剤、舌下剤、その他の剤型などにより行 うことができる。 [0037] The oral administration can be carried out in solid, powder or liquid dosage units, for example, powders, powders, tablets, dragees, capsules, drops, sublingual agents, other dosage forms, etc. I can.
[0038] 上記組織内投与は、例えば、溶液や懸濁剤などの皮下、筋肉内又は静脈内注射 用の液状用量単位形態を用いることによって行うことができる。これらのものは、本発 明の化合物の一定量を、例えば、水性や油性の媒体などの注射目的に適合する非 毒性の液状担体に懸濁又は溶解し、っ 、で上記懸濁液又は溶液を滅菌することに より製造される。  [0038] The intra-tissue administration can be performed, for example, by using a liquid dosage unit form for subcutaneous, intramuscular or intravenous injection such as a solution or suspension. These are obtained by suspending or dissolving a certain amount of the compound of the present invention in a non-toxic liquid carrier suitable for injection purposes such as an aqueous or oily medium, and so on. It is manufactured by sterilizing.
[0039] 上記局所投与 (経皮投与など)は、例えば、液剤、クリーム剤、粉末剤、ペースト剤、 ゲル剤、軟膏剤などの外用製剤の形態を用いることによって行うことができる。これら のものは、本発明の化合物の一定量を、外用製剤の目的に適合する香料、着色料、 充填剤、界面活性剤、保湿剤、皮膚軟化剤、ゲル化剤、担体、保存剤、安定剤など のうちの一種以上と組み合わせることにより製造される。  [0039] The topical administration (eg, transdermal administration) can be performed by using a form of external preparation such as a liquid, a cream, a powder, a paste, a gel, and an ointment. For these, a certain amount of the compound of the present invention is added to a fragrance, a colorant, a filler, a surfactant, a moisturizer, an emollient, a gelling agent, a carrier, a preservative, Manufactured by combining with one or more agents.
[0040] 上記経直腸的投与は、本発明の化合物の一定量を、例えば、パルミチン酸ミリスチ ルエステルなどの高級エステル類、ポリエチレングリコール、カカオ脂、これらの混合 物など力もなる低融点固体に混入した座剤などを用いて行うことができる。  [0040] In the above-mentioned rectal administration, a certain amount of the compound of the present invention is mixed with a high-melting low-melting solid such as higher esters such as palmitic acid myristyl ester, polyethylene glycol, cocoa butter, and mixtures thereof. It can be performed using a suppository or the like.
[0041] 上記投与は、例えば、溶液や懸濁剤などの皮下、筋肉内又は静脈内注射用の液 状用量単位形態を用いることによって行うことができる。これらのものは、本発明の化 合物の一定量を、例えば、水性や油性の媒体などの注射の目的に適合する非毒性 の液状担体に懸濁又は溶解し、ついで上記懸濁液又は溶液を滅菌することにより製 造される。 [0041] The administration can be performed, for example, by using a liquid dosage unit form for subcutaneous, intramuscular or intravenous injection such as a solution or suspension. These consist of suspending or dissolving a certain amount of a compound of the invention in a non-toxic liquid carrier suitable for injection purposes, such as an aqueous or oily medium, then the suspension or solution. It is manufactured by sterilizing.
本発明のペプチド或いは抗体などのタンパク質を細胞内で作用させるために細胞 内に当該タンパク質を輸送する手法として、例えば、細胞膜透過機能を有するぺプ チド(例えば Pegelinや Penetratinなど)を付カ卩すること(Martine Mazel etal, Doxorubi cm— peptide conjugates overcome multidrug resistance. Anti-Cancer Drugs 2001, 12 、 Dcrossi D. et al" The third helix of the antennapedia homeodomain translocates th rough biological membranes, J. Biol. Chem. 1994, 269, 10444-10450.)により本発明 のタンパク質を細胞内に輸送させることが可能である。  As a method for transporting a protein such as the peptide or antibody of the present invention into a cell, for example, a peptide having a cell membrane permeation function (for example, Pegelin, Penetratin, etc.) is attached. (Martine Mazel etal, Doxorubi cm— peptide conjugates overcome multidrug resistance. Anti-Cancer Drugs 2001, 12, Dcrossi D. et al "The third helix of the antennapedia homeodomain translocates th rough biological membranes, J. Biol. Chem. 1994, 269, 10444-10450.), It is possible to transport the protein of the present invention into cells.
[0042] また、本発明のペプチドを細胞内に導入する方法としては、(1)ウィルスベクター( たとえば、アデノウイルス)にペプチド配列をコードする遺伝子を 1コピー又は複数コ ピー連結し、このウィルスを感染させることによって導入する方法、(2)目的のぺプチ ドを静脈注射によって直接導入する方法、(3)目的のペプチド又はそれをコードする DNAを物理的に細胞や組織に導入する方法 (たとえば、パーティクルガン)等を挙げ ることが出来る。パーティクルガンは、極めて強力な遺伝子導入法で、金またはタンダ ステンの微粒子 (マイクロキャリア)に付着させた核酸をヘリウムガスの圧力によって標 的細胞に導入する方法である。この方法は、マイクロインジェクション法に比べ、操作 が非常に簡単であり、細胞種に関わらず広範囲な応用が期待できる。このことを利用 して、ペプチドの導入にも応用することが期待される。 [0042] In addition, as a method for introducing the peptide of the present invention into cells, (1) one or more copies of a gene encoding a peptide sequence may be incorporated into a viral vector (eg, adenovirus). (2) a method of directly introducing the target peptide by intravenous injection, (3) a target peptide or a DNA encoding the target peptide or DNA encoding it physically. The method (for example, particle gun) introduced into the organization can be mentioned. The particle gun is an extremely powerful gene transfer method that introduces nucleic acid attached to gold or tandastene microparticles (microcarriers) into target cells by the pressure of helium gas. This method is much easier to operate than the microinjection method, and can be used in a wide range of applications regardless of the cell type. This is expected to be applied to the introduction of peptides.
[0043] 本発明は、 HCV感染症を治療または予防するための薬剤のスクリーニング方法に 関する。 [0043] The present invention relates to a method for screening a drug for treating or preventing HCV infection.
本発明のスクリーニング方法の第一の態様としては、まず、以下の(a)または (b)に 記載のペプチドに被検化合物を接触させる。 In the first embodiment of the screening method of the present invention, first, a test compound is contacted with the peptide described in the following ( a ) or (b).
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
[0044] 第一の態様に用いられるペプチドの状態としては、特に制限はなぐ例えば、精製 された状態、細胞内に発現した状態、細胞抽出液内に発現した状態などであっても よい。該ペプチドが発現している細胞としては、外来性のペプチドを発現している細 胞が挙げられる。上記外来性のペプチドを発現している細胞は、例えば、該ペプチド をコードする DNAを含むベクターを細胞に導入することで作製できる。ベクターの細 胞への導入は、一般的な方法、例えば、リン酸カルシウム沈殿法、電気パルス穿孔 法、リボフエタミン法、マイクロインジェクション法等によって実施することができる。こ のような外来性のペプチドが導入される細胞が由来する生物種としては、哺乳類に 限定されず、外来タンパク質を細胞内に発現させる技術が確立されている生物種で あればよい。  [0044] The state of the peptide used in the first embodiment is not particularly limited, and may be, for example, a purified state, a state expressed in a cell, a state expressed in a cell extract, or the like. Examples of cells expressing the peptide include cells expressing a foreign peptide. A cell expressing the exogenous peptide can be prepared, for example, by introducing a vector containing DNA encoding the peptide into the cell. Introduction of a vector into a cell can be performed by a general method such as calcium phosphate precipitation, electric pulse perforation, ribophetamine, or microinjection. The biological species from which a cell into which such an exogenous peptide is introduced is derived is not limited to mammals, and any biological species that has established a technique for expressing a foreign protein in the cell may be used.
[0045] ペプチドが発現して 、る細胞抽出液は、例えば、試験管内転写翻訳系に含まれる 細胞抽出液に、ペプチドをコードする DNAを含むベクターを添カ卩したものを挙げるこ とができる。該試験管内転写翻訳系としては、特に制限はなぐ市販の試験管内転写 翻訳キットなどを使用することが可能である。 [0045] The cell extract in which the peptide is expressed is included in, for example, an in vitro transcription / translation system. An example is a cell extract obtained by adding a vector containing DNA encoding a peptide. As the in vitro transcription / translation system, it is possible to use a commercially available in vitro transcription / translation kit without particular limitation.
[0046] 本発明の方法における「被検化合物」としては、特に制限はなぐ例えば、天然ィ匕 合物、有機化合物、無機化合物、タンパク質、ペプチド等の単一化合物、並びに、化 合物ライブラリー、遺伝子ライブラリーの発現産物、細胞抽出物、細胞培養上清、発 酵微生物産生物、海洋生物抽出物、植物抽出物、原核細胞抽出物、真核単細胞抽 出物もしくは動物細胞抽出物等を挙げることができる。上記被検化合物は必要に応 じて適宜標識して用いることができる。標識としては、例えば、放射標識、蛍光標識等 を挙げることができる。また、「複数の被検化合物」としては、特に制限はなぐ例えば 、上記被検化合物に加えて、これらの被検化合物を複数種混合した混合物も含まれ る。 [0046] The "test compound" in the method of the present invention is not particularly limited, for example, natural compounds, organic compounds, inorganic compounds, single compounds such as proteins and peptides, and compound libraries. Gene library expression products, cell extracts, cell culture supernatants, fermentation microorganism products, marine organism extracts, plant extracts, prokaryotic cell extracts, eukaryotic single cell extracts or animal cell extracts, etc. Can be mentioned. The above test compound can be appropriately labeled and used as necessary. Examples of the label include a radiolabel and a fluorescent label. The “plurality of test compounds” is not particularly limited. For example, in addition to the above test compounds, a mixture of a plurality of these test compounds is also included.
[0047] 本発明にお 、て「接触」は、ペプチドの状態に応じて行う。例えば、ペプチドが精製 された状態であれば、精製標品に被検化合物を添加することにより行うことができる。 また、細胞内に発現した状態または細胞抽出液内に発現した状態であれば、それぞ れ、細胞の培養液または該細胞抽出液に被検化合物を添加することにより行うことが できる。本発明における細胞としては、特に制限されないが、酵母またはヒトを含む哺 乳動物由来の細胞が好ましい。被検化合物がタンパク質の場合には、例えば、該タ ンパク質をコードする DNAを含むベクターを、該ペプチドが発現して!/、る細胞へ導入 する、または該ベクターをペプチドが発現して 、る細胞抽出液に添加することで行う ことも可能である。また、例えば、酵母または動物細胞等を用いた 2ハイブリッド法を 利用することも可能である。  In the present invention, “contact” is performed depending on the state of the peptide. For example, if the peptide is in a purified state, it can be carried out by adding a test compound to the purified sample. Moreover, if it is in a state expressed in a cell or in a cell extract, it can be carried out by adding a test compound to the cell culture solution or the cell extract, respectively. The cell in the present invention is not particularly limited, but a cell derived from a mammal including yeast or human is preferable. When the test compound is a protein, for example, a vector containing DNA encoding the protein is introduced into a cell in which the peptide is expressed! /, Or the vector is expressed in the peptide. It can also be carried out by adding to the cell extract. Further, for example, a two-hybrid method using yeast or animal cells can be used.
[0048] 第一の態様では、次 、で、上記ペプチドと被検化合物の結合を検出する。タンパク 質間の結合を検出または測定する手段は、例えばタンパク質に付した標識を利用す ることにより行うことができる。標識の種類は、例えば、蛍光標識、放射標識等が挙げ られる。また、酵素ツーハイブリット法や、 BIACOREを用いた測定方法等、公知の方 法によって測定することもできる。本方法においては、ついで、上記ペプチドと結合し た被検化合物を選択する。選択された被検化合物の中には、 HCV感染症を治療ま たは予防するための薬剤が含まれる。また、選択された被検化合物を、以下のスクリ 一ユングの被検化合物として用いてもよ 、。 [0048] In the first embodiment, next, the binding between the peptide and the test compound is detected. The means for detecting or measuring the binding between proteins can be performed, for example, by using a label attached to the protein. Examples of the label type include fluorescent labels and radiolabels. Moreover, it can also be measured by a known method such as an enzyme two-hybrid method or a measurement method using BIACORE. In this method, a test compound bound to the peptide is then selected. Some of the selected test compounds treat HCV infection. Or drugs to prevent. Alternatively, the selected test compound may be used as a test compound for the following screen.
[0049] 本発明のスクリーニング方法の第二の態様としては、まず、被検化合物を以下の (a )または (b)に記載のペプチドと同時に、スフインゴミエリンに添加する。  [0049] In a second embodiment of the screening method of the present invention, first, a test compound is added to sphingomyelin simultaneously with the peptide described in the following (a) or (b).
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
[0050] 第二の態様では、次 、で、上記 (a)または (b)に記載のペプチドと、スフインゴミエリ ンの結合能を測定する。結合能の測定は上記に記載の方法で行うことが出来る。 本方法においては、ついで、被検化合物を添加しない場合と比較して、上記結合 能を低下させた場合に、被検化合物を、 HCV感染症を治療または予防するための薬 剤として選択する。  [0050] In the second embodiment, the binding ability of the peptide described in (a) or (b) above and the sphingomyelin is measured next. The binding ability can be measured by the method described above. In this method, the test compound is then selected as a pharmaceutical agent for treating or preventing HCV infection when the binding ability is reduced as compared with the case where no test compound is added.
[0051] また、本発明は、 HCV感染症を治療または予防するための薬剤の効力の評価方法 に関する。  [0051] The present invention also relates to a method for evaluating the efficacy of a drug for treating or preventing HCV infection.
本発明の薬剤の評価方法の第一の態様としては、まず、以下の (a)または (b)に記 載のペプチドに被検化合物を接触させる。 In the first aspect of the method for evaluating a drug of the present invention, first, a test compound is brought into contact with the peptide described in the following ( a ) or (b).
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
第一の態様では、ついで、上記ペプチドと被検化合物の結合を検出する。ペプチド と被検化合物の結合の検出は、上記に記載の方法により行うことが出来る。ついで、 上記ペプチドと結合した被検化合物の、 HCV感染症の治療または予防効果の効力 を評価する。上記ペプチドに対する被検化合物の結合能 (例えば、平衡結合定数、 結合速度定数、解離速度定数、等)を検出することで、該被検化合物の HCV感染症 の治療または予防効果の効力を評価することが出来る。例えば、被検化合物が上記 ペプチドに対し有意な結合能を示す場合には、該被検化合物は HCV感染症の有意 な治療または予防効果を示すと評価することが出来る。 In the first embodiment, the binding between the peptide and the test compound is then detected. Detection of the binding between the peptide and the test compound can be performed by the method described above. Next, the efficacy of the therapeutic or prophylactic effect of HCV infection of the test compound bound to the above peptide is evaluated. By detecting the binding ability of the test compound to the peptide (for example, equilibrium binding constant, binding rate constant, dissociation rate constant, etc.), the test compound can detect HCV infection. The efficacy of the therapeutic or preventive effect can be evaluated. For example, when the test compound shows a significant binding ability to the peptide, it can be evaluated that the test compound shows a significant therapeutic or preventive effect on HCV infection.
[0052] 本発明の薬剤の評価方法の第二の態様としては、まず、被検化合物を以下の(a)ま たは (b)に記載のペプチドと同時に、スフインゴミエリンに添加する。 [0052] In a second embodiment of the method for evaluating a drug of the present invention, first, a test compound is added to sphingomyelin simultaneously with the peptide described in the following (a) or (b).
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
[0053] 第二の態様では、次 、で、上記 (a)または (b)に記載のペプチドと、スフインゴミエリ ンの結合能を測定し、該被検化合物の HCV感染症の治療または予防効果の効力を 評価する。結合能の測定は上記に記載の方法で行うことが出来る。上記ペプチドとス フインゴミエリンの結合能 (例えば、平衡結合定数、結合速度定数、解離速度定数、 等)を検出することで、該被検化合物の、上記ペプチドとスフインゴミエリンの結合阻 害能を検出することができる。さらに、これらの値を基にして、該被検化合物の HCV 感染症の治療または予防効果の効力を評価することが出来る。例えば、被検化合物 を添加することにより、被検化合物を添加しない場合に比べて、上記ペプチドとスフィ ンゴミエリンの結合能を有意に低下させた場合には、該被検化合物は HCV感染症の 有意な治療または予防効果を示すと評価することができる。  [0053] In the second aspect, in the following, the binding ability of the peptide described in (a) or (b) above and sphingomyelin is measured, and the test compound has an effect of treating or preventing HCV infection. Evaluate efficacy. The binding ability can be measured by the method described above. By detecting the binding ability of the peptide and sphingomyelin (eg, equilibrium binding constant, binding rate constant, dissociation rate constant, etc.), the test compound has the ability to inhibit the binding of the peptide to sphingomyelin. Can be detected. Furthermore, based on these values, the efficacy of the test compound for treating or preventing HCV infection can be evaluated. For example, when the test compound is added and the binding ability of the peptide and sphingomyelin is significantly reduced compared to the case where the test compound is not added, the test compound is significantly more susceptible to HCV infection. It can be evaluated that it shows an effective therapeutic or preventive effect.
[0054] 本発明の上記記載の評価方法は、 HCV感染症を治療又は予防するための新規薬 剤を見出すためのスクリーニング方法のみに限らず、医薬候補化合物の開発のため の薬効の効力の評価や医薬品の製造供給の際に必要な品質管理のための効力の 評価を行う場合の評価方法としても有用である。 [0054] The above-described evaluation method of the present invention is not limited to a screening method for finding a new drug for treating or preventing HCV infection, but an evaluation of the efficacy of the drug for the development of a drug candidate compound. It is also useful as an evaluation method when evaluating the efficacy for quality control required in the manufacture and supply of pharmaceuticals.
本発明は、上記に記載のスクリーニング方法または評価方法に用いるためのキットに 関する。このようなキットには、上記に記載のスクリーニング方法または評価方法の検 出工程や測定工程に使用されるものを含みうる。例えば、該ペプチドと、スフインゴミ エリンの結合能測定に必要とされる試薬、機器等を挙げることができる。その他、蒸 留水、塩、緩衝液、タンパク質安定剤、保存剤等が含まれていてもよい。 なお本明細書において引用された全ての先行技術文献は、参照として本明細書に 組み入れられる。 The present invention relates to a kit for use in the screening method or evaluation method described above. Such a kit may include those used in the detection process and measurement process of the screening method or evaluation method described above. For example, there can be mentioned reagents and instruments required for measuring the binding ability of the peptide and sphingomyelin. Other steaming Retention water, salt, buffer solution, protein stabilizer, preservative and the like may be contained. All prior art documents cited in the present specification are incorporated herein by reference.
実施例  Example
[0055] 以下、本発明を実施例によりさらに具体的に説明するが本発明はこれら実施例に 制限されるものではない。  [0055] Hereinafter, the present invention will be described more specifically with reference to Examples, but the present invention is not limited to these Examples.
[0056] 〔実施例 1〕 HCV-NS5Bのスフインゴ脂質結合モチーフの探索  [Example 1] Search for sphingolipid-binding motif of HCV-NS5B
HIV-lgpl20由来の V3ループ(図 1A、配列番号: 4)やプリオン由来の PrPタンパク 質は、スフインゴ脂質結合ドメインをもっている。 HCVタンパク質の中に、スフインゴ脂 質結合領域があるかどうか探索するために、これらと類似する構造を CEプログラム (cl. sdsc.edu/ce.html)を用いて検索した。その結果、図 1に示すように HCV-NS5Bの配列 E230- G263 (図 1A、配列番号: 3)において、ヘリックス'ターン'ヘリックスのモチーフ (図 1)をもった類似構造が認められた。 HCV-NS5B全長の塩基配列を配列番号 : 8 に、アミノ酸配列を配列番号: 9に示す。この構造ドメインのサイズは V3ループとほぼ 一致した(図 1Bは、 HCV-NS5Bの構造。縦線は HCV-NS5Bの同定した推定のスフィ ンゴ脂質結合領域。斜線は HIV-1 V3ループのスフインゴ脂質結合領域)。  HIV-lgpl20-derived V3 loop (Fig. 1A, SEQ ID NO: 4) and prion-derived PrP protein have sphingolipid binding domains. In order to search for the presence of a sphingolipid binding region in HCV proteins, structures similar to these were searched using the CE program (cl. Sdsc.edu/ce.html). As a result, as shown in FIG. 1, in HCV-NS5B sequence E230-G263 (FIG. 1A, SEQ ID NO: 3), a similar structure having a helix 'turn' helix motif (FIG. 1) was observed. The full length base sequence of HCV-NS5B is shown in SEQ ID NO: 8 and the amino acid sequence is shown in SEQ ID NO: 9. The size of this structural domain closely matched that of the V3 loop (Figure 1B shows the structure of HCV-NS5B. The vertical line is the putative sphingolipid binding region identified by HCV-NS5B. The diagonal line is the sphingolipid of the HIV-1 V3 loop. Binding area).
[0057] 〔実施例 2〕推定の NS5Bスフインゴ結合領域ペプチド(NS5B-SBD)とスフインゴミエリ ン (SM)との結合  [0057] [Example 2] Binding of putative NS5B sphingo binding domain peptide (NS5B-SBD) to sphingomyelin (SM)
Biacore S51を使用して、 NS5B-SBDペプチドと SMとの結合能を検討した。 SMをクロ 口ホルムに溶解し、真空乾固したあと、 PBS緩衝液を加え、 10mMとなるよう懸濁した 。懸濁液は凍結融解を繰り返し、ポアサイズ 50 nmのメンブレンに通し、リボソームを 調製し、センサチップ L1に固定化した。合成ペプチドは、 NS5B(DIRVEESIYQCCDL APEARQAIKSLTERLY (配列番号: 1) , Sigma genosysにより合成)、及び PrP(KQHT VTTTTKGENFTETDVKMMER (配列番号: 2) , Sigma genosysにより合成)を使用した 。特異的結合量は、ペプチドをカ卩えていないセンサグラムをブランクとし、各濃度のセ ンサグラム力も差し引いたものを結合量の測定に用いた。その結果、 NS5B-SBDぺプ チドは、濃度依存的にスフインゴミエリンとの結合を認めた(図 2A、 NS5B-SBDぺプチ ドのセンサグラム)。プリオンタンパク質 (PrP)にお 、ても同様な結合が認められた(図 2B、図 2Aのセンサグラム 80s時の RU値)。 The binding ability of NS5B-SBD peptide and SM was examined using Biacore S51. SM was dissolved in chloroform and vacuum-dried, and then PBS buffer was added and suspended to 10 mM. The suspension was repeatedly frozen and thawed, passed through a membrane with a pore size of 50 nm, and ribosomes were prepared and immobilized on the sensor chip L1. NS5B (DIRVEESIYQCCDL APEARQAIKSLTERLY (SEQ ID NO: 1), synthesized by Sigma genosys) and PrP (KQHT VTTTTKGENFTETDVKMMER (SEQ ID NO: 2), synthesized by Sigma genosys) were used as synthetic peptides. The amount of specific binding was determined by measuring the amount of binding using a sensorgram without a peptide as a blank and subtracting the sensorgram force at each concentration. As a result, NS5B-SBD peptide was found to bind to sphingomyelin in a concentration-dependent manner (Fig. 2A, sensorgram of NS5B-SBD peptide). Similar binding was observed in prion protein (PrP) (Fig. 2B, RU value at 80 s sensorgram in Figure 2A).
[0058] 上記の結果よりラフトの構成成分であるスフインゴミエリンと、 HCVタンパク質である HCV-NS5Bが特定の部位において強く結合すること、およびこれらの結合により HCV ウィルスの複製が行われていることが明ら力となった。これらの知見により、スフインゴ ミエリンと HCVタンパク質の結合を阻害することにより、 HCVウィルスの複製を抑制す ることが出来るものと示唆された。  [0058] From the above results, Sphingomyelin, which is a component of raft, and HCV protein HCV-NS5B are strongly bound at a specific site, and the HCV virus is replicated by their binding. Clearly became a force. These findings suggest that replication of HCV virus can be suppressed by inhibiting the binding of sphingomyelin and HCV protein.
[0059] 〔実施例 3〕 NS5Bスフインゴ結合領域ペプチドおよびその誘導体の HCVレプリコン阻 害活性測定  [Example 3] Measurement of HCV replicon inhibitory activity of NS5B sphingo binding domain peptide and its derivatives
次に、 HCVレブリコン細胞を用いて、 NS5bスフインゴ結合領域ペプチド (配列番号: 1)およびその誘導体ペプチド (配列番号: 10、 11)の HCVレブリコン阻害活性を測 し 7こ。  Next, using HCV revlikon cells, measure the HCV revlikon inhibitory activity of NS5b sphingo binding domain peptide (SEQ ID NO: 1) and its derivative peptides (SEQ ID NO: 10, 11).
[表 1] ぺプチドの種類 N末→C末の配列 配列番号 [Table 1] Types of peptides N-terminal → C-terminal sequence SEQ ID NO:
Pep-1 誘導体 1 D 1 RVEES 1 YQCCALAPAAAQA 1 KSLTERLY 10Pep-1 derivative 1 D 1 RVEES 1 YQCCALAPAAAQA 1 KSLTERLY 10
Pep- 2 誘導体 2 D 1 RVEES IAQCCDAAPEARQAAKSLTERLY 11Pep-2 derivative 2 D 1 RVEES IAQCCDAAPEARQAAKSLTERLY 11
Pep- 3 NS5bタンパク D 1 RVEES 1 YQCCDLAPEARQA 1 KSLTERLY 1Pep-3 NS5b protein D 1 RVEES 1 YQCCDLAPEARQA 1 KSLTERLY 1
Pep-4 ネガティブペプチド YLRETLSK 1 AQRAEPALDCCQY 1 SEEVR 1 D 12 Pep-4 negative peptide YLRETLSK 1 AQRAEPALDCCQY 1 SEEVR 1 D 12
HCVレプリコン細胞を 96穴プレートの 1穴当たり 10,000細胞(O.lmL)入れ、 5%FetalPlace HCV replicon cells in a 96-well plate at 10,000 cells per well (O.lmL), 5% Fetal
Bovine Serum (J¾ BS) (Hv lone , Cat. No. SH30071)を含む Dulbecco' s ModifiedDulbecco 's Modified including Bovine Serum (J¾ BS) (Hv lone, Cat.No. SH30071)
Eagle Medium (以下 DMEM) (Invitrogen, Cat. No. 10569- 010)中で 37。C、 5%CO存 37 in Eagle Medium (DMEM) (Invitrogen, Cat. No. 10569-010). C, 5% CO
2 在下 20時間培養した。 96穴プレートの培地を捨て、 FBSを含まない DMEMを O.lmLカロ えた。 BioPORTER (Gene Therapy System, Cat. No.BP502401)をクロ口ホルムで溶解 し、風乾にてフィルム化した。これを Phosphate Buffered Saline (Sigma, Cat. No. D853 7)に溶解したペプチド(2mg/mL)で再溶解した。 5分後、 96穴プレートに O.OlmLの上 記各ペプチドを添加し、 37°C、 5%CO存在下で 3〜4時間培養した。 10%FBSを含む  2 Cultured for 20 hours. The medium in the 96-well plate was discarded, and O.lmL of DMEM without FBS was added. BioPORTER (Gene Therapy System, Cat. No. BP502401) was dissolved in black mouth form and filmed by air drying. This was redissolved with a peptide (2 mg / mL) dissolved in Phosphate Buffered Saline (Sigma, Cat. No. D8537). After 5 minutes, each peptide of O.OlmL was added to a 96-well plate and cultured at 37 ° C. in the presence of 5% CO for 3 to 4 hours. Includes 10% FBS
2  2
DMEMを O.lmLカ卩え、更に培養を行なった。ペプチド添加から 20時間および 40時間 後に、 Steady- Glo Luciferase Assay System (Promega, Cat. No. E2520)でレプリコン 活性を測定した。ネガティブペプチドとして配列番号: 12に記載のアミノ酸配列から なるペプチドを使用した。 DMEM was added in O.lmL and further cultured. 20 and 40 hours after peptide addition, replicon with Steady-Glo Luciferase Assay System (Promega, Cat.No. E2520) Activity was measured. A peptide consisting of the amino acid sequence shown in SEQ ID NO: 12 was used as a negative peptide.
その結果、 NS5Bスフインゴ結合領域ペプチドおよびその誘導体は終濃度 0. lmg/m Lにおいて、 20〜30%の抗 HCV複製阻害を示すことが明ら力となった(図 3)。一方、 ネガティブペプチドでは阻害が観測されなかった。  As a result, it became clear that NS5B sphingo-binding domain peptide and its derivatives showed 20-30% anti-HCV replication inhibition at a final concentration of 0.1 mg / mL (FIG. 3). On the other hand, no inhibition was observed with the negative peptide.
[0060] 以下に参考例として、 WO2006/16657に記載された実施例を例示する。 [0060] Examples described in WO2006 / 16657 are given below as reference examples.
〔参考例 1〕ノザンブロット解析によるミリオシンまたはオーレォバシディウム (Aureobasi dium)属などの微生物に由来する式 (II)で表される化合物の HCV RNA複製阻害活 性の測定  [Reference Example 1] Measurement of HCV RNA replication inhibitory activity of compounds represented by formula (II) derived from microorganisms such as myriocin or Aureobasidium by Northern blot analysis
ミリオシン又は以下の式(II)で表される化合物を ΙρΜから 100 μ Μの範囲でレプリコ ン細胞 Huh-3-lに与え、 5%CO存在下、 37度にて培養した。  Myriocin or a compound represented by the following formula (II) was given to the replicon cell Huh-3-l in the range of ΙρΜ to 100 μΜ and cultured at 37 ° C in the presence of 5% CO.
2  2
式 (Π) :  Formula (Π):
Figure imgf000022_0001
Figure imgf000022_0001
[0062] 72時間後に細胞を回収し、全 RNAを抽出した後で、 Ambion社のノザンマックスキッ トの方法に従いネオマイシン耐性遺伝子をプローブとしてノザン解析を行った。  [0062] After 72 hours, cells were collected and total RNA was extracted, and then Northern analysis was performed using a neomycin resistance gene as a probe in accordance with the Northern Max Kit method of Ambion.
ノザン解析は以下のものを用いた。すなわち、 NorthernMax transfer buffer (Ambion #8672),転写膜 BrightStar- Plus (Ambion #10100),ろ紙(Sigma P-6664) , ULTRAhy b (Ambion #8670)。プローブの標識は BiotinStar Psoralen- Biotin kit (Ambion #9860 G3)でビォチン化標識した。 High Stringency buffer (Amibion #8674); BrightStar BioD etect kit (Wash buffer, Ambion #8650G; Blocking buffer, Ambion #8651G; Streptavi din-Alkaline Phosphatase, Ambion #2374G; Assay buffer, Ambion #8652G;CDP— Sta r, Ambion #8653G)  The Northern analysis used the following. That is, NorthernMax transfer buffer (Ambion # 8672), transfer film BrightStar-Plus (Ambion # 10100), filter paper (Sigma P-6664), ULTRAhy b (Ambion # 8670). The probe was labeled biotinylated with BiotinStar Psoralen-Biotin kit (Ambion # 9860 G3). High Stringency buffer (Amibion # 8674); BrightStar BioD etect kit (Wash buffer, Ambion # 8650G; Blocking buffer, Ambion # 8651G; Streptavi din-Alkaline Phosphatase, Ambion # 2374G; Assay buffer, Ambion # 8652G; CDP— Starr, (Ambion # 8653G)
[0063] 1%ァガロースゲルで 1 μ gのトータル RNAを泳動し、泳動後ェチジゥムブ口ミドで RN Aを染色して写真をとり、脱色後 NorthernMax transfer bufferを用いて転写膜に 2時間 転写した。湿ったままの状態で UVクロスリンカ一にて RNAを転写膜に固定ィ匕した。ハ イブリローターを用いて、 ULTRAhybにて 42度、 30分間の回転前処理の後、前処理 液を捨て、ピオチン化したネオマイシン耐性遺伝子と 10mlの ULTRAhyb液を加えて 42 度一夜振とう処理した。 [0063] 1 μg of total RNA was run on a 1% agarose gel, and after electrophoresis, RN A was stained and photographed. After decolorization, it was transferred to a transfer film for 2 hours using NorthernMax transfer buffer. In a wet state, RNA was immobilized on the transfer membrane with a UV crosslinker. Using a hybrid rotor, after pre-rotating for 42 minutes at ULTRAhyb for 30 minutes, the pre-treatment solution was discarded, and the neomycin resistance gene and 10 ml of ULTRAhyb solution were added and shaken overnight at 42 degrees.
[0064] ULTRAhyb液を捨て、 42度に保温した High Stringency bufferを 15ml加え 42度で 30 分間振とうした。同様の操作をもう一度繰り返した。浸る程度の Wash bufferで転写膜 を洗浄した。転写膜を Blocking bufferで 30分間振とうした。液を捨て、 10mlの Blocking bufferに 2 μ 1の Streptavidin- Alkaline Phosphataseをカ卩えたもので室温 30分間振とうし た。転写膜を浸る程度の Blocking bufferで 10分間振とうした。 Wash bufferで 5分間洗 浄し、 Wash bufferで転写膜を洗浄した後、 CDP-Starで転写膜を覆い、 5分後に余分 な液を除き、 1時間後に X線フィルムに感光させ、バンドの濃さから RNA量を比較した 。その結果、ミリオシン及び式 (II)で表される化合物に 1-lOnMの濃度でレプリコン RN Aを 50%減少させる効果が認められた(図 5、 6)。  [0064] The ULTRAhyb solution was discarded, 15 ml of High Stringency buffer kept at 42 ° C was added, and the mixture was shaken at 42 ° C for 30 minutes. The same operation was repeated once more. The transfer film was washed with a wash buffer that was soaked. The transfer membrane was shaken with Blocking buffer for 30 minutes. The solution was discarded, and 2 ml of Streptavidin-Alkaline Phosphatase in 10 ml Blocking buffer was added and shaken for 30 minutes at room temperature. Shake for 10 minutes with Blocking buffer soaking the transfer membrane. Wash with Wash buffer for 5 minutes, wash the transfer film with Wash buffer, cover the transfer film with CDP-Star, remove excess liquid after 5 minutes, expose to X-ray film after 1 hour, and concentrate the band. The amount of RNA was compared. As a result, the effect of reducing replicon RNA by 50% at a concentration of 1-lOnM was recognized for myriocin and the compound represented by formula (II) (FIGS. 5 and 6).
[0065] 〔参考例 2〕ウェスタンプロット解析によるミリオシンまたは式 (II)で表される化合物の H CVタンパク合成阻害の測定  [0065] [Reference Example 2] Measurement of inhibition of HCV protein synthesis by myriocin or a compound represented by formula (II) by Western plot analysis
ウェスタン解析は以下の方法でおこなった。ミリオシン又は式 (II)で表される化合物 を ΙρΜから 100 /z Mの範囲でレプリコン細胞 Huh-3-lに与え、 5%CO存在下、 37度  Western analysis was performed by the following method. Myriocin or a compound represented by the formula (II) is given to the replicon cell Huh-3-l in the range of ΙρΜ to 100 / z M, 37 ° C in the presence of 5% CO.
2  2
にて培養した。 72時間後に培地を捨て、 PBS(Phosphate buffered saline)をカ卩え、ピぺ ッティングにより細胞をはがし、遠心により細胞を回収した。 Phosphate用溶解液 (50 mM Tris— HCl(pH7.5),0.5% Triton, 3 mM EDTA, 150 mM NaCl, 12mM glycerophosp hate, 50 mM NaF, 1 mM Na VO , 0.5 mM PMSF, 0.5 mM aporotinin)をカ卩えてピぺッ  Incubated in After 72 hours, the medium was discarded, PBS (Phosphate buffered saline) was added, the cells were removed by pipetting, and the cells were collected by centrifugation. Phosphate solution (50 mM Tris—HCl (pH 7.5), 0.5% Triton, 3 mM EDTA, 150 mM NaCl, 12 mM glycerophosp hate, 50 mM NaF, 1 mM Na VO, 0.5 mM PMSF, 0.5 mM aporotinin) Pipette
3 4  3 4
ティングにより細胞を破壊し、高速遠心により上清を回収した。 Dye Reagent (Nacalai tesque #074-27)にてタンパク定量をおこなった(ゥシ γグロブリン標準液、 BIO- RAD# 500-0005)。得られたタンパク質 5 gを 9 11%グラジェントゲル(第一化学薬品、 # 3 17552)でトリス-グリシン- SDS緩衝液(BIO-RAD # 161-0772)で電気泳動した。分子 量サイズマ ~~力' ~~ i Rain。ow molecular weight markers (AmershamBioscience#RPN7 56)を用いた。電気泳動したタンパク質をミニトランスブロットセル(BIO-RAD#l 70-393 0)にてメンブレン(PROTRAN BA85, Nitrocellulose transfer membrane(Shleicher&Sc huell #10401196)に転写した。 HCVタンパク由来の抗 NS3ゥサギ抗体を用いてウェス タン解析を行った。内部標準として抗ァクチンゥサギ抗体を用いた。その結果、 ミリオ シン及び式 (II)で表される化合物に 1-lOnMの濃度で HCVタンパク質の発現を 50%減 少させる効果が認められた(図 7、 8)。 Cells were disrupted by ting and the supernatant was collected by high speed centrifugation. Protein quantification was performed with Dye Reagent (Nacalai tesque # 074-27) (USH γ globulin standard solution, BIO-RAD # 500-0005). 5 g of the obtained protein was electrophoresed with Tris-Glycine-SDS buffer (BIO-RAD # 161-0772) on 911% gradient gel (Daiichi Kagaku, # 3 17552). Molecular weight size ~~ force '~~ i Rain. ow molecular weight markers (AmershamBioscience # RPN7 56) were used. The electrophoresed protein was transferred to a mini-trans blot cell (BIO-RAD # l 70-393 0) was transferred to a membrane (PROTRAN BA85, Nitrocellulose transfer membrane (Shleicher & Sc huell # 10401196)) Western analysis was performed using an anti-NS3 Usagi antibody derived from HCV protein, and an anti-actin Usagi antibody was used as an internal standard. As a result, myriocin and the compound represented by formula (II) were found to have an effect of reducing HCV protein expression by 50% at a concentration of 1-lOnM (FIGS. 7 and 8).
[0066] 〔参考例 3〕フモ-シン B1の HCVレプリコン阻害活性の測定 [0066] [Reference Example 3] Measurement of HCV replicon inhibitory activity of fumosin B1
スフインゴ脂質生合成の途中段階において、ジヒドロスフインゴシン力 ジヒドロセラ ミドを生成するジヒドロスフインゴシン N ァシル転移酵素を特異的に阻害するフモ 二シン B1の HCVレプリコン阻害活性を測定した。  In the middle stage of sphingolipid biosynthesis, HCV replicon inhibitory activity of fumonisin B1, which specifically inhibits dihydrosphingosine synthase, which produces dihydrosphingosine, a dihydroceramide, was measured.
ホタル.ルシフェラーゼ HCVレプリコン細胞 (Huh-3-l)を 5%ゥシ胎児血清(Hyclone c at. no. SH30071.03)を含むダルベッコ MEM (G¾co cat. no. 10569)に懸濁し 96穴プ レートに 5000細胞 ZWellで蒔き、 5%CO、 37度で一夜培養した。約 20時間後、フモ- Suspension of firefly luciferase HCV replicon cells (Huh-3-l) in Dulbecco's MEM (G¾co cat. No. 10569) containing 5% urine fetal serum (Hyclone cat. No. SH30071.03) The cells were seeded with 5000 cells ZWell and cultured overnight at 37% with 5% CO. About 20 hours later
2 2
シン B1を順次 3倍希釈し、終濃度 1.37 Mから 1000 Mになるように加え、さらに 3日 間培養した。アツセィプレートは 2系統用意し、 1つは白色プレート、他はクリアプレー トでアツセィを行った。培養終了後、白色プレートは Steady- Glo Luciferase Assay Sys tem (Promega cat. no. E2520)に用いた。すなわち、 Wellあたり 100 μ 1の試薬を入れ、 3〜4回ピペットで混ぜ、 5分間放置後に 1450 MicroBeta TRILUX (WALLAC)にてル ミネッセンスを測定した。細胞未添加の値をバックグランドとして全ての値力も差し引 き、薬剤未添加の値を阻害 0%として薬剤の IC50 (50%阻害濃度)を算出した。一方、細 胞毒性試験はセル'カウントキット 8 (DOJIN Laboratories, cat. No.341- 07761)を W ellあたり 10 μ 1入れ、 3〜4回ピペットで混ぜ、約 30分間放置後に OD450nm力 1.0程度 になった時点で測定した。細胞未添加の値をバックグランドとして全ての値力も差し 引き、薬剤未添加の値を阻害 0%として薬剤の IC50 (50%阻害濃度)を算出した。  Thin B1 was diluted three-fold in order, added to a final concentration of 1.37 M to 1000 M, and further cultured for 3 days. Two assembly plates were prepared, one for the white plate and the other for the clear plate. After completion of the culture, the white plate was used for Steady-Glo Luciferase Assay System (Promega cat. No. E2520). That is, 100 μl of reagent was added per well, mixed 3-4 times with a pipette, allowed to stand for 5 minutes, and then luminescence was measured with 1450 MicroBeta TRILUX (WALLAC). The IC50 (50% inhibitory concentration) of the drug was calculated by subtracting all values from the value with no added cells as the background, and the value with no drug added as 0% inhibition. On the other hand, in the cell toxicity test, Cell's Count Kit 8 (DOJIN Laboratories, cat.No.341-07771) was put in 10 μl per well, mixed 3-4 times with a pipette, and left at about OD450nm power of about 1.0 after about 30 minutes. It measured when it became. The IC50 (50% inhibitory concentration) of the drug was calculated by subtracting all values from the value with no added cells as the background and the value with no drug added as 0% inhibition.
その結果、フモ-シン B1は 10-1000 Mの濃度で、 HCVレプリコン阻害活性を示す ことがわかった(図 9)。  As a result, it was found that fumosin B1 exhibits HCV replicon inhibitory activity at a concentration of 10-1000 M (FIG. 9).
[0067] 〔参考例 4〕セリンパルミトイル転移酵素の siRNA合成 [0067] [Reference Example 4] siRNA synthesis of serine palmitoyltransferase
SPTの阻害が HCVレプリコン活性を阻害しているかを確認する目的で、 LCB1 (SPT のへテロダイマーのうち 1サブユニット)を標的とした siRNAを合成した。 2種の特異的 な siRNA (si246、 si633)は、 LCBlcDNAの配列(GenBank Accession No. Y08685)をも とにデザインし、 Qiagen社により合成された。コントロール siRNA (配列番号: 15)は、 L CB1の発現に影響しない配列を使用した。合成した siRNA配列を配列番号: 13 (si24 6)および配列番号: 14に示した。 In order to confirm whether inhibition of SPT inhibited HCV replicon activity, we synthesized siRNA targeting LCB1 (one subunit of SPT heterodimer). Two specific The siRNAs (si246, si633) were designed based on the LCBlcDNA sequence (GenBank Accession No. Y08685) and synthesized by Qiagen. As a control siRNA (SEQ ID NO: 15), a sequence that does not affect the expression of LCB1 was used. The synthesized siRNA sequence is shown in SEQ ID NO: 13 (si24 6) and SEQ ID NO: 14.
〔参考例 5〕ウェスタンプロット解析を用いた siRNAによる LCB1のタンパク質発現阻害 6穴プレートに 1.2 X 105個の Huh-3-l細胞をまき、 37°C、 5%でー晚培養した。 2.3 μ Lの 20 μ M siRNAを 100 μ Lの ECR buffer(RNAiFect Kit中に含まれる Buffer)に加 え、激しく攪拌し、さらに 4 /z Lの RNAiFectトランスフエクシヨン試薬(Qiagen cat. No. 3 01605)を加え、緩やかに攪拌して室温にて 10分間放置した。 siRNAは、最終濃度が 35 nMになるようにカロえ、 4日間培養した。 [Reference Example 5] Inhibition of protein expression of LCB1 by siRNA using Western plot analysis 1.2 × 10 5 Huh-3-l cells were seeded in a 6-well plate and cultured at 37 ° C. and 5%. Add 2.3 μL of 20 μM siRNA to 100 μL of ECR buffer (Buffer included in RNAiFect Kit), mix vigorously, and then add 4 / z L of RNAiFect transfection reagent (Qiagen cat. No. 3). 01605) was added and the mixture was gently stirred and allowed to stand at room temperature for 10 minutes. The siRNA was culled to a final concentration of 35 nM and cultured for 4 days.
細胞を細胞溶解用緩衝液(50 mM Tris- HC1 (pH 7.5), 0.5% Triton, 3 mM EDTA, 1 50 mM NaCl, 12 mM glycerophosphate, 50 mM NaF, 1 mM Na^O^, 0.5 mM PMSF, 0.5 mM aporotinin)に懸濁し、氷上で 10分間放置した。 15,000 X gで 10分間遠心し、 上清を回収した。タンパク質の定量後、各サンプルの蛋白量を 10 gに調製し、 1/4 量の 5 X SDSサンプル緩衝液(125 mM Tris- HC1 (pH 6.5), 25 % Glycerol, 5% SDS, 0. 25 % BPB, 5% 2-Mercaptoethanol)〖こカロえ、 98°C5分処理する。ポリアクリルアミドゲル PAGミニ 9/11 (第一化学、 cat. No. 317552)で電気泳動後、ゲルを-トロセルロースメ ンブレン PROTRAN BA85 (Schleicher&Schuell cat. No. 10404496)にブロッテイング( 70V、 3時間)する。メンブレンを Blocking Buffer(10% skim milk, 0.1% Tween 20/PBS) で Blockingを行う。 1000倍希釈した抗 LCB1抗体(Transduction cat. No. L89820)及 び 250倍希釈した抗ァクチン (20-33)抗体(Sigma cat. No. A5060)で 2時間室温にて 反応させる。メンブレンを 0.1%Tween 20/PBSにて洗浄後、二次抗体として、 1000倍希 釈した抗マウス IgG- HRP (Cell Signaling cat. No. A7076)及び抗ゥサギ IgG- HRP (Cel 1 Signaling cat. No. A7074)を 1時間室温にてそれぞれ反応させる。メンブレンを 0.1%T ween 20/PBSにて洗浄後、 ECLで 1分間反応させ、オートラジオグラフィ一にてシグナ ルを検出した。  Cell lysis buffer (50 mM Tris-HC1 (pH 7.5), 0.5% Triton, 3 mM EDTA, 1 50 mM NaCl, 12 mM glycerophosphate, 50 mM NaF, 1 mM Na ^ O ^, 0.5 mM PMSF, Suspended in 0.5 mM aporotinin) and left on ice for 10 minutes. The supernatant was collected by centrifugation at 15,000 X g for 10 minutes. After protein quantification, the amount of protein in each sample was adjusted to 10 g, and 1/4 volume of 5 X SDS sample buffer (125 mM Tris-HC1 (pH 6.5), 25% Glycerol, 5% SDS, 0.25) % BPB, 5% 2-Mercaptoethanol) Treat at 98 ° C for 5 minutes. After electrophoresis on polyacrylamide gel PAG mini 9/11 (Daiichi Kagaku, cat. No. 317552), the gel was blotted onto -trocellulose membrane PROTRAN BA85 (Schleicher & Schuell cat. No. 10404496) (70V, 3 hours) To do. Block the membrane with Blocking Buffer (10% skim milk, 0.1% Tween 20 / PBS). Incubate with anti-LCB1 antibody (Transduction cat. No. L89820) diluted 1000-fold and anti-actin (20-33) antibody (Sigma cat. No. A5060) diluted 250-fold at room temperature for 2 hours. After washing the membrane with 0.1% Tween 20 / PBS, the secondary antibodies were diluted 1000-fold with anti-mouse IgG-HRP (Cell Signaling cat. No. A7076) and anti-rabbit IgG-HRP (Cel 1 Signaling cat. No. A7074) is reacted for 1 hour at room temperature. The membrane was washed with 0.1% Tween 20 / PBS, reacted with ECL for 1 minute, and the signal was detected by autoradiography.
その結果、図 10に示すように、コントロール siRNAと比較して、 si246、 si633はともに LCB1のタンパク質発現量の減少が認められた。特に、 si246では強く発現阻害が認 められた。 As a result, as shown in FIG. 10, a decrease in the protein expression level of LCB1 was observed for both si246 and si633 as compared to the control siRNA. In particular, si246 strongly inhibited inhibition of expression. I was cut off.
[0069] 〔参考例 6〕 LCB1のノックダウンによる HCVレブリコン阻害活性効果  [0069] [Reference Example 6] Effect of LCB1 knockdown on HCV levicon control activity
参考例 5の結果にもとづいて、 Huh-3-l細胞が LCB1発現を低下する条件下での細 胞毒性及び HCVレブリコン活性の影響を以下の方法にて測定した。すなわち、 96穴 プレートに 1ゥエルあたり 3500個の細胞をまき、 37°C、 5%でー晚培養した。 1.75 L の 2 M 3«^ を23.3 μ Lの ECR緩衝液 (RNAiFect Kit中に含まれる緩衝液)に加 え、激しく攪拌し、さらに 0.31 μ Lの RNAiFectトランスフエクシヨン試薬をカ卩え、緩やか に攪拌して室温にて 10分間放置した。 siRNAは、最終濃度が 35 nMになるように加え 、 4日間培養した。細胞毒性による影響は、培地に 10 μ Lの Cell counting kit-8 (DOJI N Laboratories cat. No. 341-07761)をカ卩えて 3— 4回ピペットで混和し、 37°C、 1時 間放置後、マイクロプレートリーダー Emax (Molecular devices)を用いて 450 nmの吸 光度で測定した。 HCVレブリコン活性は、培養終了後、新しい培地に交換し、 Steady -Glo Luciferase Assay System (Promega cat. no. E2520)を用い、 Wellあたり 100 μ 1の 試薬を入れ、 3〜4回ピペットで混ぜ、 5分間放置後に 1450 MicroBeta TRILUX (WAL LAC)にてルミネッセンスを測定した。細胞未添カロの値をバックグランドとして全ての 値力も差し引き、薬剤未添加の値を阻害 0%として薬剤の IC50 (50%阻害濃度)を算 出した。  Based on the results of Reference Example 5, the effects of cell toxicity and HCV revlikon activity under conditions where Huh-3-l cells decrease LCB1 expression were measured by the following methods. That is, 3500 cells per well were seeded in a 96-well plate and cultured at 37 ° C and 5%. Add 1.75 L of 2 M 3 «^ to 23.3 μL of ECR buffer (buffered in RNAiFect Kit), mix vigorously, and add 0.31 μL of RNAiFect transfection reagent gently. And allowed to stand at room temperature for 10 minutes. siRNA was added to a final concentration of 35 nM and cultured for 4 days. To determine the effects of cytotoxicity, add 10 μL of Cell counting kit-8 (DOJI N Laboratories cat. No. 341-07761) to the medium and mix 3-4 times with a pipette, and leave at 37 ° C for 1 hour. Thereafter, the absorbance was measured at 450 nm using a microplate reader Emax (Molecular devices). HCV levulincon activity is replaced with fresh medium after completion of culture, Steady-Glo Luciferase Assay System (Promega cat.no.E2520) is used, 100 μl of reagent is added per well, and mixed by pipetting 3-4 times. After standing for 5 minutes, luminescence was measured with 1450 MicroBeta TRILUX (WAL LAC). The IC50 (50% inhibitory concentration) of the drug was calculated by subtracting all values from the value of the non-cell-added caro as the background, and the value with no drug added as 0% inhibition.
その結果、図 11に示すように、 LCB1の発現を抑制した si246、 si633処理した細胞で は、コントロール siRNA処理した細胞に対し、有意に HCVレプリコン活性を阻害した。 この阻害効果は、 LCB1の発現を強く抑制した si246で強く認められた。また、同条件 下にお 、て siRNA処理による細胞毒性を調べたところ、ほとんど影響が認められなか つた o  As a result, as shown in FIG. 11, the cells treated with si246 and si633 that suppressed the expression of LCB1 significantly inhibited HCV replicon activity compared to the cells treated with control siRNA. This inhibitory effect was strongly observed with si246, which strongly suppressed the expression of LCB1. Under the same conditions, the cytotoxicity of siRNA treatment was examined. Almost no effect was observed.
[0070] 〔参考例 7〕 HCVレブリコンアツセィおよび細胞毒性試験  [0070] [Reference Example 7] HCV rebricon assembly and cytotoxicity test
式 (I)で表される化合物またはそれらの誘導体につ!、て、 HCVレプリコンアツセィぉ よび細胞毒性試験を行った。  The HCV replicon assay and cytotoxicity test were conducted on the compounds represented by formula (I) or their derivatives.
まず、 HCV—RNAのコピー数を定量するために HCV—RNAの中にレポーター遺伝 子としてホタル由来のルシフェラーゼ遺伝子を導入したものを構築した。 Kriegerら(J. Virol.75:4614)の方法に従い、 HCV遺伝子の IRES (Internal Ribosome Entry Site)の 直下にネオマイシン耐性遺伝子と融合する形でルシフェラーゼ遺伝子を導入した。ィ ンビトロで当該 RNAを合成後、エレクト口ポレーシヨン法で Huh7細胞に導入し、 G418 而性クローンとして単離した。ホタル 'ルシフェラーゼ HCVレプリコン細胞(Huh-3-l) を 5%ゥシ胎児血清(Hyclone cat. no. SH30071.03)を含むダルベッコ MEM (Gibco c at. no. 10569-010)に懸濁し、 96穴プレートに 5000細胞 Zゥエルで播種し、 5%CO First, in order to quantify the number of HCV-RNA copies, we constructed HCV-RNA with a firefly-derived luciferase gene introduced as a reporter gene. According to the method of Krieger et al. (J. Virol. 75: 4614), the HCV gene IRES (Internal Ribosome Entry Site) A luciferase gene was introduced immediately below the neomycin resistance gene. The RNA was synthesized in vitro, then introduced into Huh7 cells by the electopore method, and isolated as a G418 metaclone. Firefly 'Luciferase HCV replicon cells (Huh-3-l) were suspended in Dulbecco's MEM (Gibco cat. No. 10569-010) containing 5% urine fetal serum (Hyclone cat. No. SH30071.03). Seed the well plate with 5000 cells Zwell, 5% CO
2 2
37°Cで一夜培養した。約 20時間後、希釈した試験化合物をゥエルあたり 10 1加え、 さらに 3日間培養した。アツセィプレートを 2系統用意し、 1つは白色プレート、他はタリ ァープレートでアツセィを行った。培養終了後、白色プレートは Steady- Glo Luciferas e Assay System (Promega cat. no. E2520)に用いた。すなわち、ゥエルあたり 100 μ 1の 試薬を入れ、 3〜4回ピペットで混ぜ、 5分間放置後に 1450 MicroBeta TRILUX (WAL LAC)にてルミネッセンスを測定した。細胞未添カロの値をバックグランドとして全ての 値力も差し引き、試験化合物未添加の値を阻害 0%として薬剤の IC (50%阻害濃度 Incubated overnight at 37 ° C. After about 20 hours, 10 1 of diluted test compound was added per well, and further cultured for 3 days. Two assembly plates were prepared, one for the white plate and the other for the plate. After completion of the culture, the white plate was used for Steady-Glo Luciferas e Assay System (Promega cat. No. E2520). That is, 100 μl of reagent was added per well, mixed 3-4 times with a pipette, allowed to stand for 5 minutes, and luminescence was measured with 1450 MicroBeta TRILUX (WAL LAC). Subtract all the values from the value of un-added caroline as a background, and set the IC (50% inhibitory concentration) of the drug to 0% as the value without the test compound added.
50  50
)を算出した。  ) Was calculated.
[0071] また、細胞毒性の測定には Cell counting kit- 8 (同人堂カタログ No. CK04)を用い た。すなわち、 10 1の Cell counting kit- 8をクリア一プレートに添カ卩し、 37度で 30〜60 分間保温した。 96穴プレートリーダーにて波長 450應、対照波長 630nmで吸光度を測 定した。細胞未添加の値をバックグランドとして全ての値力も差し引き、薬剤未添加の 値を阻害 0%として薬剤の CC (50%細胞阻害濃度)を算出した。  [0071] Cell counting kit-8 (Dojindo catalog No. CK04) was used for the measurement of cytotoxicity. That is, 10 1 Cell counting kit-8 was added to a clear plate and incubated at 37 degrees for 30-60 minutes. Absorbance was measured with a 96-well plate reader at a wavelength of 450 and a control wavelength of 630 nm. All values were subtracted from the value with no cell added, and the CC (50% cell inhibitory concentration) of the drug was calculated with the value without drug added as 0% inhibition.
50  50
上記の HCVレプリコンアツセィおよび細胞毒性試験の結果を表 2および 3に示す。  The results of the above HCV replicon assay and cytotoxicity test are shown in Tables 2 and 3.
[0072] [表 2] [0072] [Table 2]
レブリコンアツセィ 細胞毒性 化合物番号 I C50 [ M] CC50 Ifl M]Lebricon Atsey Cytotoxic Compound No. I C50 [M] CC50 Ifl M]
(1 ) 0. 002 >5(1) 0. 002> 5
(2) 0. 01 0 >5(2) 0. 01 0> 5
(3) < 0. 001 >5(3) <0. 001> 5
(4) 0. 001 >5(4) 0. 001> 5
(5) 0. 002 >5(5) 0. 002> 5
(6) 0. 007 >5(6) 0. 007> 5
(7) 0. 004 > 1(7) 0. 004> 1
(8) 0. 01 4 > 1(8) 0. 01 4> 1
(9) 0. 01 7 > 1(9) 0. 01 7> 1
( 1 0) 0. 01 1 > 1(1 0) 0. 01 1> 1
(1 1) 0. 009 > 1(1 1) 0. 009> 1
( 1 2) 0. 01 7 > 1(1 2) 0. 01 7> 1
( 1 3) 0. 01 0 > 1(1 3) 0. 01 0> 1
( 1 4) 0. 009 > 1(1 4) 0. 009> 1
( 1 5) 0. 006 > 1(1 5) 0. 006> 1
( 1 6) 0. 008 > 1(1 6) 0. 008> 1
(1 7) 0. 01 2 > 1(1 7) 0. 01 2> 1
( 1 8) 0. 068 > 1(1 8) 0. 068> 1
( 1 9) 0. 01 2 > 1(1 9) 0. 01 2> 1
(20) 0. 055 > 1(20) 0. 055> 1
(21) 0. 080 > 1(21) 0. 080> 1
(22) 0. 500 > 1(22) 0. 500> 1
(23) 0. 21 0 > 1(23) 0. 21 0> 1
(24) 0. 024 > 1(24) 0. 024> 1
(25) 0. 020 > 1(25) 0. 020> 1
(26) 0. 001 > 1(26) 0. 001> 1
(27) 0. 002 > 1 (27) 0. 002> 1
レブリコンアツセィ 細胞毒性 Lebricon Atsey Cytotoxicity
化合物番号 CC50 [jU M] Compound number CC50 [jU M]
(28) 0. 001 > 1 (28) 0. 001> 1
(29) 0. 003 > 1  (29) 0. 003> 1
(30) 0. 001 > 1  (30) 0. 001> 1
(31 ) 0. 005 > 1  (31) 0. 005> 1
(32) 0. 800 >5  (32) 0.800> 5
(33) 0. 250 > 1  (33) 0. 250> 1
(34) 0. 003 > 1  (34) 0. 003> 1
(35) 0. 004 > 1  (35) 0. 004> 1
(36) 0. 004 > 1  (36) 0. 004> 1
(37) 0. 01 7 > 1  (37) 0. 01 7> 1
(38) o 0. 024 > 1  (38) o 0. 024> 1
(39) 0. 002 > 1  (39) 0. 002> 1
(40) 0. 002 >5  (40) 0. 002> 5
(41 ) 0. 1 28 > 1  (41) 0. 1 28> 1
(42) 0. 076 > 1  (42) 0. 076> 1
(43) 0. 1 03 > 1  (43) 0. 1 03> 1
(44) 0. 082 > 1  (44) 0. 082> 1
(45) 0. 007 > 1  (45) 0. 007> 1
(46) 0. 002 >5  (46) 0. 002> 5
(47) 0. 005 >5  (47) 0. 005> 5
(48) 0. 020 >5  (48) 0. 020> 5
(49) 0. 245 >50 (49) 0. 245> 50
(50) 0. 262 >5 (50) 0. 262> 5
(51 ) 0. 072 >5  (51) 0. 072> 5
(52) 0. 1 > 50 (52) 0. 1> 50
(53) 0. 020 22(53) 0. 020 22
(54) 0. 020 > 50 (54) 0. 020> 50
[0074] 〔参考例 8〕式 (II)で表される化合物による HCVレブリコンの阻害及び宿主細胞への 毒性 [0074] [Reference Example 8] Inhibition of HCV rebricon by compound represented by formula (II) and toxicity to host cells
HCVレプリコン細胞に、図 12に示された濃度の式 (II)で表される化合物で処理を 行い、レブリコン複製阻害活性及び細胞生存阻害活性を測定した。 レブリコンによる ノレシフェフ ~~セ 7舌'性 ίま Steady— (JLO luciferase assay system (Promega, cat. no. E251 0)、細胞生存阻害活'性は、 Cell counting counting kit— 8 (Dojin Laboratories, cat. no. 341-07761)を用いて測定したその結果図 12に示すように、式 (II)で表される化合物 による処理により濃度依存的に HCVレブリコン阻害活性を認めた (IC50 = 2 nM)。ま た、式(II)で表される化合物の細胞毒性は、認められなかった(IC50〉50nM)。  HCV replicon cells were treated with the compound represented by formula (II) at the concentration shown in FIG. 12, and the replicon replication inhibitory activity and cell survival inhibitory activity were measured. Steady— (JLO luciferase assay system (Promega, cat. No. E251 0), cell viability inhibitory activity) by cell count counting kit— 8 (Dojin Laboratories, cat. As a result of measurement using no. 341-07761), as shown in FIG. 12, HCV levulincon inhibitory activity was observed in a concentration-dependent manner by treatment with the compound represented by formula (II) (IC50 = 2 nM). In addition, the cytotoxicity of the compound represented by the formula (II) was not observed (IC50> 50 nM).
[0075] 〔参考例 9〕免疫染色法による式 (II)で表される化合物の HCV-NS3タンパク質発現阻 害の確認 [0075] [Reference Example 9] Inhibition of HCV-NS3 protein expression by a compound represented by formula (II) by immunostaining Harm confirmation
HCVレプリコン細胞を 100 nMの式(II)で表される化合物で 96時間処理した後、細 胞を 3.7%ホルムアルデヒドで固定した。 3% BSAでブロッキング後、 NS3抗体(Fホフ マン ·ラロシュより供与)でインキュベートした後、洗浄した細胞を Texas- Redラベルし たゥサギ IgG (Molecular probe )でインキュベートし、蛍光顕微鏡で解析した (図 13)。 その結果、図 13に示すように、 HCV-NS3タンパク質は核周辺に存在した力 式 (II) で表される化合物の添カ卩によりそれが消失した (白色で示された部分は NS3タンパク 質、灰色で示された部分はへキスト 33342 (Sigma, cat. no. B2261)で染色された核を 示す)。  HCV replicon cells were treated with 100 nM of the compound represented by formula (II) for 96 hours, and then the cells were fixed with 3.7% formaldehyde. After blocking with 3% BSA and incubating with NS3 antibody (provided by F. Hoffman Laroche), the washed cells were incubated with Texas-Red labeled Usagi IgG (Molecular probe) and analyzed with a fluorescence microscope (Fig. 13). As a result, as shown in Fig. 13, the HCV-NS3 protein disappeared due to the addition of the compound represented by the force equation (II) existing around the nucleus (the part shown in white is the NS3 protein). The part shown in gray shows the nucleus stained with Hoechst 33342 (Sigma, cat. No. B2261)).
[0076] 〔参考例 10〕ウェスタンブロット解析による式(II)で表される化合物の NS3、 NS5A及び NS5Bタンパク質の発現阻害  [Reference Example 10] Inhibition of NS3, NS5A and NS5B protein expression of compound represented by formula (II) by Western blot analysis
レプリコン細胞を 100 nMの式(II)で表される化合物で図 14に示された時間(48時 間および 96時間)処理した。ウェスタンプロット解析は、参考例 5と同様の方法にてお こなった。その結果、時間依存的に HCVの非構造タンパク質 NS3、 NS5A及び NS5Bを 各抗体で検出した結果、ウィルスタンパク質の発現レベルの低下が認められた(図 1 4)。  Replicon cells were treated with 100 nM of the compound represented by formula (II) for the times shown in FIG. 14 (48 hours and 96 hours). Western plot analysis was performed in the same manner as in Reference Example 5. As a result, the non-structural proteins NS3, NS5A and NS5B of HCV were detected with each antibody in a time-dependent manner, and a decrease in the expression level of the viral protein was observed (Fig. 14).
[0077] 〔参考例 11〕式 (II)で表される化合物による SPT阻害活性  [0077] [Reference Example 11] SPT inhibitory activity of the compound represented by formula (II)
in vitr。における SPT阻害活性を測定するために、ヒト組み換え型 SPT (ヘテロダイマ 一 LCB1及び LCB2)タンパク質を調製した。ヒト肝臓の cDNAライブラリー(Clontech, c at. no. 639307)から、 LCBl及び LCB2の cDNAを RT- PCRによって取得し、 Hisタグ付 きの pBudCE4.1ベクター (Invitogen, cat. no. V532- 20)に組み込んだ。 HEK293細胞( ATCC, cat. no. CRL- 1573)に遺伝子導入し、 72時間後、細胞を溶解し、 N卜 NTAァ ガロース(Qiagen, cat. no. 1018244)にてタンパク質を精製した。 SPT活性は反応緩衝 液 [200 mM HEPES buffer (pH 8.0), 5 mM EDTA, 10 mM DTT, 0.05 mM pyridoxal 5 -phosphate, 0.2 mM palmitoyト CoA, 0.1 mM L- serine, and 1 mCi [3 H] serine (Amer sham, cat. no. TRK308)]に精製した SPTを加え、 15分 37°Cにて反応した。クロ口ホル ム:メタノール(1 : 2, v/v)で抽出後有機層を水で 2回再抽出した後、有機層の放射活 性を液体シンチレーシヨンカウンタ一にて測定した。その結果、図 15に示すように、 式 (II)で表される化合物は IC50約 10nMで SPT阻害活性を有していることが明らかと なった。 in vitr. In order to measure the SPT inhibitory activity in, human recombinant SPT (heterodimer LCB1 and LCB2) proteins were prepared. LCBl and LCB2 cDNAs were obtained from human liver cDNA library (Clontech, cat. No. 639307) by RT-PCR, and His-tagged pBudCE4.1 vector (Invitogen, cat. No. V532-20) ). The gene was introduced into HEK293 cells (ATCC, cat. No. CRL-1573). After 72 hours, the cells were lysed, and the protein was purified with N 卜 NTA agarose (Qiagen, cat. No. 1018244). SPT activity is reaction buffer [200 mM HEPES buffer (pH 8.0), 5 mM EDTA, 10 mM DTT, 0.05 mM pyridoxal 5 -phosphate, 0.2 mM palmitoy CoA, 0.1 mM L-serine, and 1 mCi [3 H] Serine (Amer sham, cat. no. TRK308)] was added and the reaction was allowed to proceed at 37 ° C for 15 minutes. After extraction with black mouth form: methanol (1: 2, v / v), the organic layer was re-extracted twice with water, and then the radioactivity of the organic layer was measured with a liquid scintillation counter. As a result, as shown in Figure 15, It was revealed that the compound represented by the formula (II) has an SPT inhibitory activity with an IC50 of about 10 nM.
[0078] 〔参考例 12〕式 (II)で表される化合物によるセラミド、スフインゴミエリンの de novo合成 阻害  [0078] [Reference Example 12] Inhibition of de novo synthesis of ceramide and sphingomyelin by a compound represented by formula (II)
HCVレプリコン細胞を、図 16に示された濃度の式 (II)で表される化合物で 48時間 処理した後、 [14C]セリンで 3時間標識した。クロ口ホルム:メタノール(1 : 2, v/v)で抽 出後、 de novo合成されたセラミド(図 16A)、スフインゴミエリン(図 16B)を薄層クロマ トグラフィ一により分離した。その結果、図 16に示すように、式 (II)で表される化合物 は濃度依存的に細胞内のセラミド及びスフインゴミエリンの de novo合成を阻害した。  HCV replicon cells were treated with the compound represented by formula (II) at the concentration shown in FIG. 16 for 48 hours, and then labeled with [14C] serine for 3 hours. After extraction with black mouth form: methanol (1: 2, v / v), de novo synthesized ceramide (Fig. 16A) and sphingomyelin (Fig. 16B) were separated by thin layer chromatography. As a result, as shown in FIG. 16, the compound represented by the formula (II) inhibited de novo synthesis of intracellular ceramide and sphingomyelin in a concentration-dependent manner.
[0079] 〔参考例 13〕C2—セラミドによる式 (II)で表される化合物の複製阻害の回復 [0079] [Reference Example 13] Recovery of replication inhibition of compound represented by formula (II) by C2-ceramide
式 (II)で表される化合物による HCVレブリコン阻害活性が、スフインゴ脂質生合成 経路に依存する力否かを明らかにするために、細胞性浸透性セラミド: C2—セラミド (S igma, cat. no. A7191)を式(II)で表される化合物と同時に HCVレプリコン細胞に添カロ し、 96時間培養した。細胞抽出物を用いて、実施例 5と同様の方法にてウェスタンブ ロット解析を行った。その結果 (図 17)、式 (II)で表される化合物による HCV複製阻害 は C2セラミドの濃度に依存して抑制されることが明らかとなった。  To elucidate whether the HCV revlikon inhibitory activity of the compound represented by the formula (II) depends on the sphingolipid biosynthesis pathway, a cellular permeable ceramide: C2-ceramide (S igma, cat. No A7191) was added to the HCV replicon cells simultaneously with the compound represented by formula (II) and cultured for 96 hours. Using the cell extract, Western blot analysis was performed in the same manner as in Example 5. As a result (FIG. 17), it was revealed that inhibition of HCV replication by the compound represented by the formula (II) is suppressed depending on the concentration of C2 ceramide.
[0080] 〔参考例 14〕ラフト生合成関連低分子化合物による HCV複製阻害 [0080] [Reference Example 14] Inhibition of HCV replication by low molecular weight compounds related to raft biosynthesis
HCVレプリコン細胞を、既知の SPT阻害剤ミリオシン(Sigma, cat. no. M1177)、セラ ミド合成阻害剤フモ-シン Bl(Sigma, cat. no. F1147)、およびセラミド輸送阻害剤 H PA-12 [Kobayashiら Org.lett.(2002)の合成方法に準じて合成)で処理した後、 72時間 後レブリコン活性及び生細胞数を測定した。その結果、いずれの化合物も細胞毒性 を認めない濃度で HCVの複製を抑制する効果が認められた (図 18)。  HCV replicon cells were separated from the known SPT inhibitor myriocin (Sigma, cat. No. M1177), ceramide synthesis inhibitor fumosin Bl (Sigma, cat. No. F1147), and ceramide transport inhibitor H PA-12 [ After treatment with Kobayashi et al., Org.lett. (2002), the levulincon activity and viable cell count were measured 72 hours later. As a result, each compound was found to have an effect of suppressing HCV replication at a concentration where no cytotoxicity was observed (FIG. 18).
[0081] 〔参考例 15〕式 (II)で表される化合物によるラフトタンパク質の影響(1) [0081] [Reference Example 15] Effect of raft protein by the compound represented by formula (II) (1)
HCVレプリコン細胞に ImMの式 (II)で表される化合物を 72時間添加した後、細胞抽 出液を可溶化剤 1% Nonidet P- 40 (Nacalai tesque, cat. no. 252- 23)にて 1時間処理し た。ショ糖密度勾配分画法により、フラクション 1—9まで分離し、ウェスタンプロット解 析は、参考例 5と同様な方法にて行った。その結果、図 19に示すように式 (II)で表さ れる化合物は可溶化剤耐性画分の NS5Bの発現レベルを低下させた。し力しながら、 NS5Aや宿主のラフト結合タンパク質力べオリン -2にお 、て式 (II)で表される化合物に よる影響は認められなかった。力べォリン- 2の発現は力べォリン- 2抗体(BD Transduc tion, cat. no. 610684)により確認した。 After adding the compound represented by formula (II) of ImM to HCV replicon cells for 72 hours, the cell extract was solubilized with 1% Nonidet P-40 (Nacalai tesque, cat. No. 252-23). Treated for 1 hour. Fractions 1-9 were separated by sucrose density gradient fractionation, and Western plot analysis was performed in the same manner as in Reference Example 5. As a result, as shown in FIG. 19, the compound represented by the formula (II) decreased the expression level of NS5B in the solubilizer-resistant fraction. While doing NS5A and the host raft-binding protein strength beolin-2 were not affected by the compound represented by formula (II). The expression of strength veolin-2 was confirmed with the strength veolin-2 antibody (BD Transduction, cat. No. 610684).
[0082] 〔参考例 16〕式 (II)で表される化合物によるラフトタンパク質の影響 (2) [0082] [Reference Example 16] Influence of raft protein by compound represented by formula (II) (2)
HCVレプリコン細胞に ImMの式 (II)で表される化合物を 72時間添加した後、細胞抽 出液を 1% NP-40で 1時間処理した。ショ糖密度勾配分画法により、ラフトタンパク質( 可溶化剤耐性)、非ラフトタンパク質を分離し、 PBSで希釈し濃縮後 ELISA解析により 定量した。その結果、式 (II)で表される化合物は、 NS5Bにおいて有意にラフト上から 解離が認められた (図 20)。  ImM compound (II) represented by the formula (II) was added to HCV replicon cells for 72 hours, and then the cell extract was treated with 1% NP-40 for 1 hour. Raft proteins (solubilizing agent resistance) and non-raft proteins were separated by sucrose density gradient fractionation, diluted with PBS, concentrated and quantified by ELISA analysis. As a result, the compound represented by the formula (II) was significantly dissociated from the raft in NS5B (FIG. 20).
産業上の利用可能性  Industrial applicability
[0083] これまで、 HCV遺伝子の機構と機能の解明が行われ、 HCVの非構造タンパク質を 標的とした HCV治療剤の開発が進められて 、たが、非構造タンパク質自体が複製の 過程で変異を起こす可能性があるため、効果的な治療剤の開発が困難であった。 [0083] So far, the mechanism and function of the HCV gene has been elucidated, and the development of HCV therapeutics targeting non-structural proteins of HCV has progressed. However, the non-structural proteins themselves mutated during the replication process. Therefore, it was difficult to develop an effective therapeutic agent.
[0084] 本発明により、スフインゴ脂質カ¾じ¥タンパク質の特定の部位において強く結合す ることがわかり、ホスト細胞内でのウィルスの複製のメカニズムが明ら力となった。これ らの結合を阻害する化合物が、 HCV感染症の極めて有用な治療剤または予防剤と なるものと考えられる。 [0084] According to the present invention, it was found that strong binding was performed at a specific site of the sphingolipid protein, and the mechanism of viral replication in the host cell became clear. Compounds that inhibit these bindings are considered to be extremely useful therapeutic or preventive agents for HCV infection.
本発明の知見は、スフインゴ脂質および HCVタンパク質の結合を新規なターゲット とした抗 HCV剤の開発に大きく貢献するものである。  The knowledge of the present invention greatly contributes to the development of anti-HCV agents targeting novel binding of sphingolipids and HCV proteins.
[0085] これまでの抗 HCV剤は、標的となるカスケードが不明確であり副作用が心配されて いたが、本発明の薬剤は標的力スフインゴ脂質および HCVタンパク質の結合作用で あり、結合部位もより明確であるため、副作用の排除、薬剤の効果の調節等が容易に 行えるものと考えられる。 [0085] Conventional anti-HCV agents have unclear target cascades and have been worried about side effects. However, the agents of the present invention have a binding action of targeting sphingolipids and HCV proteins, and the binding sites are also more It is clear that side effects can be easily eliminated and the effects of drugs can be easily adjusted.

Claims

請求の範囲 The scope of the claims
[1] スフインゴミエリンと HCVタンパク質の結合を阻害する化合物を有効成分として含有 する、 HCV感染症を治療または予防するための薬剤。  [1] A drug for treating or preventing HCV infection, comprising as an active ingredient a compound that inhibits the binding of sphingomyelin and HCV protein.
[2] スフインゴミエリンと HCVタンパク質の結合を阻害する化合物力 以下の(a)または( b)に記載のペプチドである、請求項 1に記載の薬剤。 [2] The drug according to claim 1, which is a compound having the ability to inhibit the binding of sphingomyelin and HCV protein, which is the peptide according to the following (a) or (b).
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
[3] スフインゴミエリンと HCVタンパク質の結合を阻害する化合物力 以下の(a)または( b)に記載のペプチドをコードするオリゴヌクレオチドである、請求項 1に記載の薬剤。 [3] The drug according to claim 1, which is an oligonucleotide encoding the peptide according to (a) or (b) below, which is a compound ability to inhibit the binding of sphingomyelin and HCV protein.
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
[4] スフインゴミエリンと HCVタンパク質の結合を阻害する化合物力 以下の(a)または( b)に記載のペプチドを認識する抗体である、請求項 1に記載の薬剤。  [4] The drug according to claim 1, which is an antibody that recognizes the peptide according to (a) or (b) below, which has a compound ability to inhibit the binding of sphingomyelin and HCV protein.
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
[5] HCVタンパク質が HCV-NS5Bである請求項 1〜4の!、ずれかに記載の薬剤。  [5] The drug according to any one of claims 1 to 4, wherein the HCV protein is HCV-NS5B!
[6] HCV感染症が、 C型肝炎、肝硬変、肝繊維化、または肝癌である、請求項 1〜5の いずれかに記載の薬剤。  [6] The drug according to any one of claims 1 to 5, wherein the HCV infection is hepatitis C, cirrhosis, liver fibrosis, or liver cancer.
[7] 以下の (A)〜(C)の工程を含む、 HCV感染症を治療または予防するための薬剤の スクリーニング方法。 [7] An agent for treating or preventing HCV infection comprising the following steps (A) to (C): Screening method.
(A)以下の (a)または (b)に記載のペプチドに被検化合物を接触させる工程  (A) A step of bringing a test compound into contact with the peptide according to the following (a) or (b)
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
(B) (A)に記載のペプチドと被検化合物の結合を検出する工程  (B) a step of detecting the binding between the peptide according to (A) and a test compound
(C) (A)に記載のペプチドと結合する被検化合物を選択する工程  (C) a step of selecting a test compound that binds to the peptide according to (A)
[8] 以下の (A)〜(C)の工程を含む、 HCV感染症を治療または予防するための薬剤の スクリーニング方法。  [8] A screening method for a drug for treating or preventing HCV infection, comprising the following steps (A) to (C):
(A)被検化合物を以下の(a)または (b)に記載のペプチドと同時に、スフインゴミエリ ンに添加する工程  (A) A step of adding the test compound to the sphingomyelin simultaneously with the peptide described in (a) or (b) below
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
(B)上記 (a)または (b)に記載のペプチドと、スフインゴミエリンの結合能を測定する 工程  (B) a step of measuring the binding ability of the peptide according to (a) or (b) above and sphingomyelin
(C)被検化合物を添加しな 、場合に比べて、上記結合能を低下させた被検化合物 を選択する工程  (C) a step of selecting a test compound having a reduced binding ability compared to the case without adding a test compound
[9] HCV感染症が、 C型肝炎、肝硬変、肝繊維化、または肝癌である、請求項 7または [9] The HCV infection is hepatitis C, cirrhosis, liver fibrosis, or liver cancer, or 7
8に記載のスクリーニング方法。 The screening method according to 8.
[10] 請求項 7〜9のいずれかに記載のスクリーニング方法に用いるためのキット。 [10] A kit for use in the screening method according to any one of claims 7 to 9.
[11] 以下の (A)〜(C)の工程を含む、 HCV感染症を治療または予防するための薬剤の 効力の評価方法。 [11] A method for evaluating the efficacy of a drug for treating or preventing HCV infection, comprising the following steps (A) to (C):
(A)以下の (a)または (b)に記載のペプチドに被検化合物を接触させる工程 (a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド (A) A step of bringing a test compound into contact with the peptide according to the following (a) or (b) (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
(B) (A)に記載のペプチドと被検化合物の結合を検出する工程  (B) a step of detecting the binding between the peptide according to (A) and a test compound
(C) (A)に記載のペプチドと結合する被検化合物の、 HCV感染症の治療または予防 効果の効力を評価する工程  (C) A step of evaluating the efficacy of the therapeutic or prophylactic effect of HCV infection of a test compound that binds to the peptide of (A)
[12] 以下の (A)および (B)の工程を含む、 HCV感染症を治療または予防するための薬 剤の効力の評価方法。  [12] A method for evaluating the efficacy of a drug for treating or preventing HCV infection, comprising the following steps (A) and (B):
(A)被検化合物を以下の(a)または (b)に記載のペプチドと同時に、スフインゴミエリ ンに添加する工程  (A) A step of adding the test compound to the sphingomyelin simultaneously with the peptide described in (a) or (b) below
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
(B)上記 (a)または (b)に記載のペプチドと、スフインゴミエリンの結合能を測定するこ とによる、 HCV感染症の治療または予防効果の効力を評価する工程  (B) A step of evaluating the efficacy of the treatment or prevention effect of HCV infection by measuring the binding ability of the peptide described in (a) or (b) above and sphingomyelin.
[13] 請求項 11または 12の 、ずれかに記載の評価方法に用いるためのキット。  [13] A kit for use in the evaluation method according to any one of claims 11 and 12.
[14] 以下の(a)または (b)に記載のペプチド。 [14] The peptide according to the following (a) or (b).
(a)配列番号: 1から 4、 10または 11のいずれかに記載のアミノ酸配列力 なるぺプ チド  (a) SEQ ID NO: 1 to 4, 10 or 11 peptide having amino acid sequence ability
(b)配列番号: 1力 4、 10または 11のいずれかに記載のアミノ酸配列において 1また は複数のアミノ酸が置換、欠失、付加および Zもしくは挿入されたアミノ酸配列力もな るペプチド  (b) SEQ ID NO: 1 peptide in which one or more amino acids are substituted, deleted, added, and Z or inserted in the amino acid sequence described in any one of 4, 10, or 11
PCT/JP2006/320440 2005-10-14 2006-10-13 Substance for use in treatment or prevention of hcv infection WO2007043640A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2007539995A JPWO2007043640A1 (en) 2005-10-14 2006-10-13 Drugs for treating or preventing HCV infection

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2005-301059 2005-10-14
JP2005301059 2005-10-14

Publications (1)

Publication Number Publication Date
WO2007043640A1 true WO2007043640A1 (en) 2007-04-19

Family

ID=37942861

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2006/320440 WO2007043640A1 (en) 2005-10-14 2006-10-13 Substance for use in treatment or prevention of hcv infection

Country Status (2)

Country Link
JP (1) JPWO2007043640A1 (en)
WO (1) WO2007043640A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8957199B2 (en) 2008-11-26 2015-02-17 Chugai Seiyaku Kabushiki Kaisha Oligoribonucleotide or peptide nucleic acid capable of inhibiting activity of hepatitis C virus
US8981123B2 (en) 2011-12-12 2015-03-17 Microbial Chemistry Research Foundation Compound and asymmetric synthesis reaction

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
AIZAKI H. ET AL.: "Characterization of the hepatitis C virus RNA replication complex associated with lipid rafts", VIROLOGY, vol. 324, 2004, pages 450 - 461, XP004519196 *
MAHFOUD R. ET AL.: "Identification of a common sphingolipid-binding domain in Alzheimer, Prion, and HIV-1 proteins", J. BIOL. CHEM., vol. 277, no. 13, 2002, pages 11292 - 11296, XP003010459 *
NEHETE P. ET AL.: "A post-CD4-binding step involving interaction of the V3 region of viral gp120 with host cell surface glycosphingolipids is common to entry and infection by diverse HIV-1 strains", ANTIVIRAL RESEARCH, vol. 56, 2002, pages 233 - 251, XP003010462 *
SAKAMOTO H. ET AL.: "Host sphingolipid biosynthesis as a target for hepatitis C virus therapy", NATURE CHEMICAL BIOLOGY, vol. 1, no. 6, November 2005 (2005-11-01), pages 333 - 337, XP003010463 *
SAKAMOTO H. ET AL.: "Identification of a novel small molecule Hepatitis C virus replication inhibitor that targets host sphingolipid biosynthesis", HEPATOLOGY, vol. 42, no. 4, SUPPL. 1, 5 October 2005 (2005-10-05), pages 535A, XP003010461 *
SHI S.T. ET AL.: "Hepatitis C virus RNA replication occurs on a detergent-resistant membrane that confractionates with Cvaeolin-2", J. VIROL., vol. 77, no. 7, 2003, pages 4160 - 4168, XP003010460 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8957199B2 (en) 2008-11-26 2015-02-17 Chugai Seiyaku Kabushiki Kaisha Oligoribonucleotide or peptide nucleic acid capable of inhibiting activity of hepatitis C virus
US8981123B2 (en) 2011-12-12 2015-03-17 Microbial Chemistry Research Foundation Compound and asymmetric synthesis reaction
US9187498B2 (en) 2011-12-12 2015-11-17 Microbial Chemistry Research Foundation Compound and asymmetric synthesis reaction

Also Published As

Publication number Publication date
JPWO2007043640A1 (en) 2009-04-16

Similar Documents

Publication Publication Date Title
CN107245502B (en) CD 2-binding protein (CD2AP) and its interacting protein
Wang et al. Identification of FBL2 as a geranylgeranylated cellular protein required for hepatitis C virus RNA replication
Gao et al. Caspase-dependent cytosolic release of cytochrome c and membrane translocation of Bax in p53-induced apoptosis
Lee et al. The hepatitis C virus NS5A inhibitor (BMS-790052) alters the subcellular localization of the NS5A non-structural viral protein
Gerold et al. Quantitative proteomics identifies serum response factor binding protein 1 as a host factor for hepatitis C virus entry
WO2007058384A1 (en) Method of suppressing replication of hepatitis c virus, inhibitor of replication of the virus and method of screening for the same
Wang et al. Hepatitis C virus non-structural protein NS5A interacts with FKBP38 and inhibits apoptosis in Huh7 hepatoma cells
Lee et al. Identification of a resveratrol tetramer as a potent inhibitor of hepatitis C virus helicase
Tang et al. Classic swine fever virus NS2 protein leads to the induction of cell cycle arrest at S-phase and endoplasmic reticulum stress
Ming et al. Inhibition of USP14 influences alphaherpesvirus proliferation by degrading viral VP16 protein via ER stress-triggered selective autophagy
JP4849622B2 (en) Drugs for treating or preventing HCV infection
Lin et al. Rab5 enhances classical swine fever virus proliferation and interacts with viral NS4B protein to facilitate formation of NS4B related complex
EP3738603A1 (en) Ntcp inhibitor
Liu et al. A new LKB1 activator, piericidin analogue S14, retards renal fibrosis through promoting autophagy and mitochondrial homeostasis in renal tubular epithelial cells
Georgopoulou et al. The protein phosphatase 2A represents a novel cellular target for hepatitis C virus NS5A protein
Nitahara-Kasahara et al. Cellular vimentin content regulates the protein level of hepatitis C virus core protein and the hepatitis C virus production in cultured cells
WO2007043640A1 (en) Substance for use in treatment or prevention of hcv infection
WO2005032329A2 (en) Methods and compositions for identifying anti-hcv agents
Udawatte et al. Dengue virus downregulates TNFR1-and TLR3-stimulated NF-κB activation by targeting RIPK1
KR101871199B1 (en) Pharmaceutical composition for prevention and treatment for Hepatitis C containing Rab32 expression or activity inhibitors
Bolder et al. Heat stress prevents impairment of bile acid transport in endotoxemic rats by a posttranscriptional mechanism
US11002729B2 (en) CD2-associated protein (CD2AP) and its interactive proteins
EP2561885A2 (en) Use of hades as tumor suppressor target
KR101701764B1 (en) Pharmaceutical composition for prevention and treatment for Hepatitis C containing AAM-B expression or activity inhibitor
Stoess et al. Pyroptosis and gasdermins—Emerging insights and therapeutic opportunities in metabolic dysfunction-associated steatohepatitis

Legal Events

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

Ref document number: 2007539995

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: 06811728

Country of ref document: EP

Kind code of ref document: A1