[go: up one dir, main page]

WO2025110774A1 - Antiviral composition comprising nanoparticle carrier based on gold nanoparticle-nucleic acid conjugate - Google Patents

Antiviral composition comprising nanoparticle carrier based on gold nanoparticle-nucleic acid conjugate Download PDF

Info

Publication number
WO2025110774A1
WO2025110774A1 PCT/KR2024/018565 KR2024018565W WO2025110774A1 WO 2025110774 A1 WO2025110774 A1 WO 2025110774A1 KR 2024018565 W KR2024018565 W KR 2024018565W WO 2025110774 A1 WO2025110774 A1 WO 2025110774A1
Authority
WO
WIPO (PCT)
Prior art keywords
virus
aso
aunp
rnase
rnai
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/KR2024/018565
Other languages
French (fr)
Korean (ko)
Inventor
이강석
배지현
염지현
심세훈
송우석
이수진
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nes Biotechnology Co Ltd
Original Assignee
Nes Biotechnology Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nes Biotechnology Co Ltd filed Critical Nes Biotechnology Co Ltd
Priority claimed from KR1020240167434A external-priority patent/KR20250076427A/en
Publication of WO2025110774A1 publication Critical patent/WO2025110774A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a nanoparticle delivery vehicle comprising a gold nanoparticle and an antisense oligonucleotide complementarily binding to a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA, and an antiviral composition comprising the same.
  • a nanoparticle delivery vehicle comprising a gold nanoparticle and an antisense oligonucleotide complementarily binding to a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA, and an antiviral composition comprising the same.
  • RNase H ribonuclease H
  • coronavirus disease which has spread worldwide, is caused by the highly contagious SARS-CoV-2 virus, causing millions of deaths and causing a global medical crisis and economic loss.
  • SARS-CoV-2 is mainly transmitted through the respiratory route, and enters cells by binding to the ACE2 receptor in the human body through the viral spike protein (S protein).
  • S protein viral spike protein
  • the inventors of the present invention while conducting research to manufacture a genetic material delivery vehicle having antiviral activity, discovered that when an antisense oligonucleotide that complementarily binds to a nucleotide sequence derived from viral RNA, particularly included in a conserved 5'untranslated region, and ribonuclease H (RNase H), which binds to the antisense oligonucleotide and induces cleavage of mRNA, are delivered together with gold nanoparticles, an excellent antiviral effect against the virus can be achieved, thereby completing the present invention.
  • RNase H ribonuclease H
  • the purpose of the present invention is to provide a nanoparticle delivery vehicle comprising a gold nanoparticle and an antisense oligonucleotide (ASO) that complementarily binds to a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA.
  • ASO antisense oligonucleotide
  • RNase H ribonuclease H
  • Another object of the present invention is to provide an antiviral composition comprising the nanoparticle carrier described above.
  • the present invention provides a nanoparticle delivery vehicle comprising a gold nanoparticle; an antisense oligonucleotide (ASO) that complementarily binds to a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA.
  • ASO antisense oligonucleotide
  • RNase H ribonuclease H
  • the present invention provides an antiviral composition comprising the nano particle carrier.
  • the present invention relates to a nanoparticle delivery vehicle comprising a gold nanoparticle and an antisense oligonucleotide (ASO) that complementarily binds to a nucleotide sequence included in a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA.
  • ASO antisense oligonucleotide
  • RNase H ribonuclease H
  • the nanoparticle delivery vehicle of the present invention comprises an antisense oligonucleotide (ASO) that complementarily binds to a nucleotide sequence contained in a genomic RNA sequence located in the 5’untranslated region (UTR) derived from the viral genome.
  • ASO antisense oligonucleotide
  • Antisense oligonucleotides are single-stranded deoxyribonucleotides complementary to specific mRNAs as well as noncoding RNAs such as microRNAs or long noncoding RNAs (Beermann, Piccoli, Viereck, & Thum, 2016; Bennett, 2019). Antisense oligonucleotides specifically bind to target RNA sequences through Watson–Crick base pairing and promote the degradation of bound RNA by endogenous nucleases such as RNaseH (Watts & Corey, 2012). That is, RNaseH can recognize RNA/DNA hybrid duplex structures and induce mRNA cleavage and genome degradation (Bennett, Baker, Pham, Swayze, & Geary, 2017).
  • antisense oligonucleotide in the present invention means an RNA or DNA molecule that binds to another RNA or DNA (target RNA, DNA). For example, if it is an RNA oligonucleotide, it binds to another RNA target by RNA-RNA interaction and modifies the activity of the target RNA. Antisense oligonucleotides can up-regulate or down-regulate the expression and/or function of a specific polynucleotide. It is intended that any foreign RNA or DNA molecule useful from a therapeutic, diagnostic, or other perspective be included in this definition.
  • antisense RNA or DNA molecules interfering RNA (RNAi), micro RNA, bait RNA molecules, siRNA, enzymatic RNA, therapeutic editing RNA, and antisense oligomeric compounds, antisense oligonucleotides, external guide sequence (EGS) oligonucleotides, primers, probes, and other oligomeric compounds that hybridize to at least a portion of a target nucleic acid may be included. Therefore, these compounds can be introduced in the form of single-stranded, double-stranded, partially single-stranded, or circular oligomeric compounds.
  • RNAi interfering RNA
  • micro RNA bait RNA molecules
  • siRNA enzymatic RNA
  • therapeutic editing RNA therapeutic editing RNA
  • antisense oligomeric compounds antisense oligonucleotides
  • antisense oligonucleotides antisense oligonucleotides
  • EGS external guide sequence oligonucleotides
  • primers
  • the RNA-derived sequence of the virus of the present invention may be an antisense oligonucleotide (ASO) that complementarily binds to the 5'untranslated region (UTR) of the viral RNA, which may target a nucleotide sequence included in the 5'UTR of the SARS-CoV-2 genomic RNA.
  • ASO antisense oligonucleotide
  • the antisense oligonucleotide includes a sequence complementary to a nucleotide sequence included in the 5'UTR of the SARS-CoV-2 genomic RNA, and the nucleotide sequence may include a leader sequence, which is an RNaseH binding site, among the 5'UTR region of SARS-CoV-2.
  • the gold nanoparticle carrier of the present invention comprises ribonuclease RNase H, which induces cleavage of mRNA.
  • RNase H is an essential endonuclease involved in various genetic processes such as transcription and replication (Aguilera & Garcia-Muse, 2012; Crossley, Bocek, & Cimprich, 2019).
  • RNA strand can combine with the template DNA to form an RNA/DNA duplex, and the non-template strand is replaced by single-stranded DNA (ssDNA) to form a structure called R-loop (Thomas, White, & Davis, 1976).
  • R-loops can help regulate transcription, but when they are not needed, they can cause double-stranded DNA breaks (DSBs), chromosomal rearrangements, and hypermutations, all causes of genome instability (Richard & Manley, 2017).
  • RNase H increases genome stability by degrading RNA in the RNA/DNA duplex and suppressing unnecessary R-loops (Nguyen et al., 2017). That is, "RNase H enzyme” is an enzyme that hydrolyzes RNA among RNA-DNA hybrids, and RNase H, which was first discovered in calf thymus, was subsequently discovered in various organisms.
  • the RNaseH bound to the gold nanoparticles of the present invention may be derived from various organisms, and is not limited to the derived organisms, but may be isolated from Pyrococcus furiosus, Pyrococcus horikoshi, Thermococcus litoralis, Thermos thermophilus or E. coli.
  • the RNaseH of the present invention may be RNaseH1, and may be an RNaseH having 50%, 60%, 70%, 80%, 90%, 95% or 99% homology to the amino acid sequence of RNaseH1.
  • the antisense oligonucleotide (ASO) and RNaseH of the present invention are bound to the surface of gold nanoparticles.
  • the gold nanoparticles of the present invention are gold particles having a diameter in the nanometer range, preferably 5-500 nm, more preferably 10-200 nm, which are easy to manufacture in the form of stable particles, are easy to control in size, and are harmless to the human body, unlike heavy metals such as manganese, aluminum, cadmium, lead, mercury, cobalt, nickel, and beryllium, and thus have high biocompatibility.
  • the diameter of gold nanoparticles increases to more than 500 nm, not only do their characteristics as nanoparticles disappear, but also the bonding between the gold surface, which does not have the characteristics of a nanomaterial, and functional groups such as thiol groups becomes weak, making it difficult to manufacture a carrier using gold nanoparticles.
  • the gold nanoparticles used in the present invention are not limited to the method, but can be manufactured as follows: HAuCl 4 is used as a gold source, and HAuCl 4 is reduced using sodium citrate as a reducing agent to manufacture gold nanoparticles.
  • the size of the gold nanoparticles can be controlled by varying the amount of citrate added. That is, as the amount of citrate added increases, nucleation increases, and thus the size of the gold nanoparticles decreases.
  • the gold nanoparticles of the present invention include surface modification for binding antisense oligonucleotides and RNaseH to the surface.
  • the antisense oligonucleotides and RNaseH may include one or more functionalities for binding to the surface of the gold nanoparticles.
  • the above functional group may be a thiol group or an amine group and may be included in one or more residues of the antisense oligonucleotide or the RNAI that binds to the antisense oligonucleotide.
  • the antisense oligonucleotide or the RNAI that binds to the antisense oligonucleotide comprises one or more thiolated residues, through which it can directly bind to the surface of the gold nanoparticles.
  • a thiolated RNAI oligo is bound to the surface of a gold nanoparticle, and the thiolated RNAI oligo binds the antisense oligonucleotide to the surface of the gold nanoparticle through specific binding to the antisense oligonucleotide sequence of the present invention.
  • the gold nanoparticles of the present invention may include an aptamer on the surface.
  • the "aptamer” refers to a single-stranded nucleic acid (DNA, RNA or modified nucleic acid) that has a stable tertiary structure in itself and has the characteristic of being able to bind to a target molecule with high affinity and specificity
  • aptamers for various desired target substances proteins, sugars, dyes, DNA, metal ions, cells, etc.
  • SELEX Systematic Evolution of Ligands of Exponential enrichment
  • the aptamer that binds to the gold nanoparticles may use any kind of aptamer of various tags or protein-specific aptamers, particularly for binding RNase H to the surface of the gold nanoparticles, and is not particularly limited.
  • an aptamer including a histidine tag was bound to a gold nanoparticle, and RNaseH was bound to the surface of the gold nanoparticle through the histidine tag aptamer.
  • the gold nanoparticle carrier of the present invention comprises at least one antisense oligonucleotide bound to the surface of a gold nanoparticle and a histidine tag aptamer for RNaseH binding, wherein the antisense oligonucleotide can bind to the surface of the gold nanoparticle directly or through RNAI. That is, the total number of oligos including RNAI, antisense oligonucleotide, and histidine tag aptamer bound to the surface of the gold nanoparticle is not limited thereto, but may be 1 to 100, and preferably 40 to 50.
  • the above gold nanoparticle carrier has an effect of treating viral infection by binding to a leader sequence present in the 5'UTR portion of the mRNA of the target virus through an antisense oligonucleotide sequence bound to the surface, thereby inducing mRNA degradation due to the action of RNaseH.
  • the gold nanoparticle carrier of the present invention can be utilized as a therapeutic agent for infectious diseases caused by viruses.
  • the virus is not limited to a type, but may be selected from the group including coronaviruses including SARS-CoV-2 (COVID-19), SARS-CoV (SARS), and MERS-CoV (MERS); influenza virus; Ebola virus; herpesviruses including HSV-1, HSV-2, VZV, and CMV; retroviruses including HIV, hepatitis viruses including HBV, HCV, and HDV; flaviviruses including dengue and Zika viruses; rotavirus; papillomavirus; measles virus; poliovirus; coxsackievirus; and adenovirus.
  • coronaviruses including SARS-CoV-2 (COVID-19), SARS-CoV (SARS), and MERS-CoV (MERS)
  • influenza virus including Ebola virus; herpesviruses including HSV-1, HSV-2, VZV, and CMV; retroviruses including HIV, hepatitis viruses including HBV, HCV
  • treatment refers to any action that improves symptoms or is beneficial by administering the pharmaceutical composition according to the present invention.
  • the pharmaceutical composition of the present invention can be used by being formulated in the form of oral dosage forms such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc., external preparations, suppositories, and sterile injection solutions according to conventional methods, and may further include carriers or excipients necessary for the formulation.
  • oral dosage forms such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc., external preparations, suppositories, and sterile injection solutions according to conventional methods, and may further include carriers or excipients necessary for the formulation.
  • Pharmaceutically acceptable carriers, excipients, and diluents that may be additionally included in the effective ingredient include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, magnesium stearate, mineral oil, and the like. When formulating, it is usually prepared using diluents or excipients such as fillers, bulking agents, binders, wetting agents, disintegrants, and surfactants.
  • diluents or excipients such as fillers, bulking agents, binders, wetting agents, disintegrants, and surfactants.
  • solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and these solid preparations are prepared by mixing the extract or compound with at least one excipient, at least cotton, starch, calcium carbonate, sucrose or lactose, gelatin, etc.
  • lubricants such as magnesium stearate and talc are also used.
  • Liquid preparations for oral administration include suspensions, oral solutions, emulsions, syrups, etc., and in addition to commonly used simple diluents such as water and liquid paraffin, various excipients such as wetting agents, sweeteners, fragrances, and preservatives may be included.
  • Preparations for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized preparations, and suppositories.
  • Non-aqueous solutions and suspensions may include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate.
  • Suppository bases may include witepsol, macrogol, Tween 61, cacao butter, laurin butter, and glycerogelatin.
  • the pharmaceutical composition of the present invention can be administered orally or parenterally (intravenously, subcutaneously, intraperitoneally, or topically) depending on the intended method, and the dosage varies depending on the patient's condition and weight, the degree of the disease, the drug form, the route of administration, and the time, and can be selected in an appropriate form by a person skilled in the art.
  • the pharmaceutical composition of the present invention is administered in a pharmaceutically effective amount.
  • pharmaceutically effective amount means a reasonable amount applicable to medical treatment, and an amount sufficient to treat a disease, and the standard thereof may be determined according to the patient's disease, severity, drug activity, drug sensitivity, administration time, administration route, and excretion rate, treatment period, concomitantly used ingredients, and other matters.
  • the pharmaceutical composition of the present invention may be administered individually or in combination with other therapeutic agents, and may be administered sequentially or simultaneously with conventional therapeutic agents. The dosage may be determined at a level that minimizes side effects by considering all of the above factors, and this may be easily determined by a person skilled in the art.
  • the dosage of the pharmaceutical composition may vary depending on the patient's age, weight, severity, sex, etc., and may generally be administered in an amount of 0.001 to 150 mg per 1 kg of body weight, more preferably 0.01 to 100 mg, once to three times a day, every day or every other day. However, this is an example and the dosage may be set differently as needed.
  • the present invention relates to an antiviral gene carrier comprising a gold nanoparticle and an antisense oligonucleotide (ASO) that complementarily binds to a nucleotide sequence included in a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA, and an antiviral composition comprising the same, which can have a therapeutic effect on diseases caused by infection with various viruses including SARS-CoV-2 by inhibiting viral mRNA synthesis.
  • ASO antisense oligonucleotide
  • RNase H ribonuclease H
  • Figure 1 is a schematic diagram showing the process of binding of antisense oligonucleotides (ASOs) and RNaseH to the surface of gold nanoparticles through functionalized DNA oligos.
  • ASOs antisense oligonucleotides
  • RNaseH RNaseH
  • Figure 2 shows the results of simply mixing RNase H at the indicated concentrations (0, 1, 2, 4, and 8 ⁇ M) with AuNP- AptHis (5 nM) and reacting for 10 min, and the AuNP- AptHis -RNH (bound RNase H) was analyzed by SDS-polyacrylamide gel electrophoresis.
  • the amount of RNase H bound to AuNP- AptHis was quantified as band intensity and compared to the known amount of RNase H (total RNase H).
  • the results were quantified and displayed in a graph, and the data are expressed as the mean ⁇ standard error of the mean (SEM) of at least three independent experiments.
  • Figure 3 shows the results of evaluating the cytotoxicity of AuNP materials by cell viability analysis, and the colors represent the AuNP untreated group (black), PBS treated group (red), AuNP-DNA oligo (yellow), and AuNP-DNA oligo-RNH (green).
  • the relative cell viability is expressed as the percentage of viable cells compared to the viable cells of cells alone.
  • the data are expressed as the mean ⁇ SEM of three independent experiments. NS, unclear.
  • Figure 4 is a confocal fluorescence microscopy image of VERO-E6 cells treated with PBS, AuNP RNAI/His , ASO, RNase H, ASO loaded on AuNP RNAI/His , and/or RNase H, respectively, wherein each image shows Cy3-labeled ASO (red), Alexa 488-labeled RNase H (green), and nuclei stained with DAPI (blue).
  • the graphs at the bottom are quantified fluorescence intensities of the images, and the fluorescence intensity quantification was expressed as corrected total cell fluorescence (CTCF) using lmage J software (NIH).
  • CTCF integrated density - (area of selected cells ⁇ mean fluorescence of background readings)]. Data are presented as mean ⁇ SEM of three independent experiments, and symbols (*,#) indicate statistical significance. (***, ###p ⁇ 0.001).
  • Figure 5 is a schematic diagram of the RNase H-dependent ASO mechanism.
  • Figure 6 is a real-time qRT-PCR analysis of positive-sense strand SARS-CoV-2 5'UTR mRNA in VERO-E6 cells 24 h after treatment with scrambled DNA, ASO, and/or RNase H loaded onto AuNP RNAI/His (1 nM).
  • the untreated group black
  • the group not treated with RNase H red
  • the group treated with RNase H green
  • Data are presented as the mean ⁇ SEM of three independent experiments.
  • Asterisks indicate statistically significant values (***p ⁇ 0.001).
  • NS unclear.
  • Figure 7A shows the results of real-time qRT-PCR analysis of the expression of 5'UTR mRNA of positive-sense strand SARS-CoV-2 in VERO-E6 cells by injecting AuNP RNAI/His -Scrambled DNA-RNH (black) and AuNP RNAI/His -ASO-RNH (green) at 4-h intervals. Data are presented as the mean ⁇ SEM of three independent experiments. Asterisks indicate statistically significant values (**p ⁇ 0.01; ***p ⁇ 0.001). NS, unclear.
  • Figure 7B shows the qRT-PCR product of Figure 7A separated on a 1.5% agarose gel and visualized using ethidium bromide. GAPDH was used as a loading control.
  • Figure 8 is a real-time qRT-PCR analysis of 5'UTR mRNA of negative-sense strand SARS-CoV-2 in VERO-E6 cells 24 h after treatment with Scrambled DNA, SO, and/or RNase H loaded on AuNP RNAI/His (1 nM) for 24 h.
  • the untreated group black
  • the group not treated with RNase H red
  • the group treated with RNase H yellow
  • Data are presented as the mean ⁇ SEM of three independent experiments. Asterisks indicate statistically significant values.
  • NS unclear.
  • Figure 9 is the real-time qRT-PCR analysis of 5'UTR mRNA of negative-sense strand SARS-CoV-2 in VERO-E6 cells 24 h after treatment with Scrambled DNA, SON, and/or RNase H loaded on AuNP RNAI/His (1 nM) for 24 h.
  • the untreated group black
  • the group not treated with RNase H red
  • the group treated with RNase H purple
  • Data are presented as the mean ⁇ SEM of three independent experiments. Asterisks indicate statistically significant values.
  • NS unclear.
  • Figure 10 shows the expression of the GFP reporter gene fused to the (+) 5'UTR of SARS-CoV-2 by incubating VERO-E6 cells with PBS (control), scrambled DNA, ASO, and AuNP RNAI/His (1 nM) loaded with RNase H for 24 h (A), and the lower graph is its quantification (B).
  • the graph shows the results when the amount of PBS was set to 1, and the data are presented as the mean ⁇ SEM of three independent experiments. Asterisks indicate statistically significant values (**p ⁇ 0.01). NS, unclear.
  • Figure 13 shows the results of measuring the relative GFP fluorescence intensity after treating SARS-CoV-2 infected VERO-E6 cells with PBS (control), AuNP RNAI/His , scrambled DNA, ASO, and AuNP RNAI/His loaded with RNase H.
  • Figure 14 shows the results of confirming the expression of RdRP mRNA in the nasal conchae and lungs after treatment of SARS-CoV-2-infected mice by intranasal injection (A) and retroorbital injection (B) with PBS (control), AuNP RNAI /His (2 nM), or AuNP RNAI /His -ASO-RNH (ASO 0.2 ⁇ M and RNH-loaded AuNP RNAI/His 2 nM), respectively.
  • the plasmids and oligonucleotides used in the present invention are shown in Tables 1 and 2 below.
  • ori origin of replication
  • Km r kanamycin resistance
  • Amp r ampicillin resistance
  • Hyg r hygromycin resistance
  • VERO-E6 African green monkey kidney
  • Calu-3 Human Lung Adenocarcinoma
  • FBS Fetrachloride
  • Plasmids were extracted from E. coli strain DH5 ⁇ using the NucleoBond Xtra Midi preparation kit (Macherey-Nagel, DR, Germany). VERO-E6 cells were seeded on plates 24 h before transfection and transfected with the plasmids using Lipofectamine 3000 (Invitrogen, CA, USA). Plasmid: Lipofectamine 3000 (1 ⁇ g/ ⁇ l): p3000 (1 ⁇ g/ ⁇ l) were mixed in a ratio of 1:2:2.5 and added sequentially to Opti-MEM medium, reacted for 10 minutes, and then added to cells and cultured for 24 h.
  • AuNP-DNA oligo conjugates for intracellular delivery of ASO and RNase H were fabricated by the following method.
  • AuNPs are linked to two types of DNA oligos, RNA I and aptamer (AptHis), which target the 5' UTR of the positive sense strand of SARS-CoV-2 that specifically binds to ASO and the hexahistidine (His) tag of RNase H, respectively.
  • RNA I and aptamer AptHis
  • His hexahistidine
  • citrate-capped AuNPs (diameter 15 nm) were purchased from BBI Life Science (Crumlin, UK).
  • Thiolated RNA I, 6X His-tagged aptamer and ASO were treated with 1 N dithiothreitol (1.545 g of DTT in 0.01 M sodium acetate, pH 5.2) for 1 h at room temperature to cleave the disulfide bond.
  • Excess DTT and unwanted thiol fragments were removed from the free thiol-modified oligonucleotide mixture by extraction with ethyl acetate. The cleaved oligonucleotides were purified using ethanol precipitation.
  • AuNPs, thiolated DNA oligos (RNAI or ASO) and 6X His-tagged aptamer were mixed at a ratio of 1:150:150 in phosphate buffer (100 mM, pH 7.4) and reacted at room temperature for 1 h, and then the mixture was sonicated for 5 min.
  • NaCl (2 M, based on 10 mM pH 7.4 phosphate buffer) was added dropwise while shaking until the final NaCl concentration became 300 mM, three times at 4-h intervals, and incubated at room temperature for 12 h.
  • the AuNP-DNA oligo mixture was transferred to a 15 ml tube and centrifuged at 12,000 ⁇ g for 20 min.
  • the supernatant was removed, and the AuNP-DNA oligo mixture was transferred to a 1.5 ml tube, centrifuged at 20,000 ⁇ g for 20 min, and washed twice with triple-distilled water (TDW).
  • the mixture was redispersed in PB buffer (50 mM phosphate buffer based on 500 mM NaCl) until the final concentration of the fabricated AuNP RNAI/His became 50 nM.
  • AuNP RNAI/His was pre-incubated at 80°C for 5 min to prevent secondary structure formation and cooled to room temperature.
  • 1X PBS, 6X His-tagged RNase H (0.02 ⁇ M) (Prospec, NZ, Israel), ASO (0.1 ⁇ M), and TDW were mixed, and AuNP RNAI/His was added to the mixture.
  • the binding affinity of the above AuNP-Apt His and RNase H was measured as follows.
  • the amount of RNase H in the AuNP-Apt His -RNH (5 nM) complex was analyzed on a 12% SDS-polyacrylamide gel.
  • the dissociation constant (KD) of AuNP-Apt His for RNase H was 1.1 ⁇ M, which shows that RNase H has a high binding affinity for AuNP-Apt His .
  • a sense oligonucleotide (SO) targeting the 5' UTR of the negative sense strand SARS-CoV-2 and a sense oligonucleotide (SON) targeting the N gene of the negative sense strand SARS-CoV-2 were synthesized and loaded onto AuNPs, and the results such as cytotoxicity and binding efficiency were compared.
  • VERO-E6 cells (5 ⁇ 10 3 /well) were seeded in 96-well plates and cultured for 24 h. The cells were then incubated again for an additional 24 h in the medium with 2 nM of ASO, SO, SON, and/or RNase H loaded onto AuNP RNAI/His .
  • Cell viability was measured using the CytoTox 96 ® Non-Radioactive Cytotoxicity Assay (Promega, WI, USA). Colors are represented as cells only (black), PBS (red), AuNP-DNA oligos (yellow), and AuNP-DNA oligo-RNH (green), and relative cell viability is expressed as the percentage of viable cells compared to viable cells in cells only. Data are presented as the mean ⁇ SEM from three independent experiments. (NS: unclear)
  • VERO-E6 cells were transfected with the AuNP RNAI/His -ASO-RNH complex for 24 h, and the 5' UTR mRNA expression level of SARS-CoV-2 was measured by qRT-PCR (Fig. 5).
  • negative-sense strand SARS-CoV-2 (plasmid pMQ131-5'UTR-reversed-EGFP or pcDNA3.1-N gene reversed) was tested in the same manner as above, but AuNP RNAI/His -SO-RNH or AuNP RNAI/His -SON-RNH complexes were not effective in knockdown of the target RNA (Fig. 8, Fig. 9).
  • VERO-E6 cells infected with SARS-CoV-2 were transfected with AuNP RNAI/His or AuNP RNAI/His -ASO-RNH at doses of 0.1 and 1 nM, respectively, and the nucleocapsid protein of SARS-CoV-2 was detected using immunocytochemistry (ICC) analysis.
  • ICC immunocytochemistry
  • the nucleocapsid protein level was reduced by 30-40% in VERO-E6 (Fig. 17) treated with AuNP RNAI/His ( 0.1 and 1 nM) and a lower concentration of AuNP RNAI/His -ASO-RNH (0.1 nM), no significant effect on the N protein level was observed.
  • VERO-E6 Fig. 17
  • AuNP RNAI/His 0.1 and 1 nM
  • AuNP RNAI/His -ASO-RNH 0.1 nM
  • the present invention relates to a nanoparticle delivery vehicle comprising a gold nanoparticle; and an antisense oligonucleotide (ASO) complementary to a virus-derived sequence bound to the surface of the gold nanoparticle and ribonuclease H (RNase H) that induces cleavage of mRNA of the virus.
  • ASO antisense oligonucleotide
  • RNase H ribonuclease H
  • the viral sequence comprises a nucleotide sequence included in the 5’ untranslated region (UTR), and the viral sequence comprises a leader sequence.
  • the virus may be selected from the group comprising coronavirus, influenza virus, ebola virus, herpes virus, retrovirus, hepatitis virus, rotavirus, flavivirus, papillomavirus, measles virus, poliovirus, coxsackievirus and adenovirus.
  • the antisense oligonucleotide (ASO) is bound to the gold nano surface via a moiety comprising a functional group, wherein the moiety comprising a functional group comprises a thiol group or an amine group.
  • the RNaseH can be bound to the gold nano surface via an aptamer, and further, the aptamer can include a histidine tag.
  • the present invention relates to an antiviral composition
  • an antiviral composition comprising the gold nanoparticle carrier.
  • the present invention relates to a gold nanoparticle carrier for delivering into cells gold nanoparticles having an antisense oligonucleotide (ASO) complementary to a virus-derived sequence and ribonuclease H (RNase H) bound to the surface, which induces cleavage of mRNA of the virus, and wherein the antisense oligonucleotide (ASO) complementary to the virus-derived sequence binds to the virus-derived mRNA sequence and degrades the mRNA through RNase H, thereby inhibiting the activity of the virus.
  • ASO antisense oligonucleotide
  • RNase H ribonuclease H

Landscapes

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

Abstract

The present invention relates to a nanoparticle carrier and an antiviral composition comprising same, the nanoparticle carrier comprising: gold nanoparticles; antisense oligonucleotides (ASOs) that complementarily bind to conserved RNA sequences derived from a viral genome; and ribonuclease H (RNase H) that binds to the ASOs to induce the cleavage of mRNA. The nanoparticle carrier and the antiviral composition comprising same can reduce mRNA expression of a virus by RNase H, and in particular, can have the effect of treating infectious diseases caused by a virus.

Description

금 나노 입자-핵산 결합체를 기반으로 하는 나노 입자 전달체를 포함하는 항 바이러스 조성물 Antiviral composition comprising nanoparticle carrier based on gold nanoparticle-nucleic acid conjugate

본 발명은 금 나노 입자 및 이의 표면에 결합된 바이러스 RNA유래 서열에 상보적으로 결합하는 안티센스 올리고뉴클레오타이드; 및 mRNA의 절단을 유발시키는 리보핵산가수분해효소 H(RNase H)를 포함하는 나노 입자 전달체 및 이를 포함하는 항 바이러스 조성물에 대한 것이다. The present invention relates to a nanoparticle delivery vehicle comprising a gold nanoparticle and an antisense oligonucleotide complementarily binding to a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA, and an antiviral composition comprising the same.

전 세계적으로 유행한 코로나바이러스 감염증(COVID-19)은 고도의 전염성을 가진 SARS-CoV-2 바이러스에 의해 발생하며, 수백만 명의 사망자를 초래하고 전 세계적인 의료 위기와 경제적 손실을 불러일으키고 있다. SARS-CoV-2는 주로 호흡기 경로로 전파되며, 바이러스 스파이크 단백질(S protein)을 통해 인체의 ACE2 수용체에 결합하여 세포 내로 침투한다. 이 과정에서 다양한 변이체가 나타나며 전염성과 치명률이 변화하고 있어, 현재 사용되는 백신 및 항바이러스 치료제에 대한 효과를 지속적으로 평가하고 있다.The coronavirus disease (COVID-19), which has spread worldwide, is caused by the highly contagious SARS-CoV-2 virus, causing millions of deaths and causing a global medical crisis and economic loss. SARS-CoV-2 is mainly transmitted through the respiratory route, and enters cells by binding to the ACE2 receptor in the human body through the viral spike protein (S protein). In the process, various variants appear, changing the transmissibility and mortality rate, so the effectiveness of the currently used vaccines and antiviral treatments is continuously being evaluated.

기존 항바이러스제는 주로 단백질 기반 접근법이나 화학적 약물을 통해 바이러스 억제를 시도하지만, 이러한 접근법은 변이 바이러스에 대한 효능 저하와 투여 빈도 증가, 높은 생산 비용 등의 한계가 있다. 이에 비해 바이러스의 유전 물질을 이용한 기술은 인체 내에서 특정 바이러스 항원을 발현시키는 유전자(DNA, RNA 등)를 효과적으로 전달하여 면역 반응을 유도하고, 바이러스 복제를 억제할 수 있어 새로운 가능성을 제시한다. 이러한 기술을 위하여, 다양한 유전 물질 전달체의 개발이 이루어지고 있으며, 이들은 유전 물질의 변이체에 대한 신속한 조정이 가능하고, 다양한 항바이러스 효과를 가지는 특성을 가지고 있어 특히 SARS-CoV-2와 같은 변이 바이러스에 대해 신속하고 광범위한 대응이 가능하다.Existing antiviral agents mainly attempt to suppress viruses through protein-based approaches or chemical drugs, but these approaches have limitations such as reduced efficacy against mutant viruses, increased administration frequency, and high production costs. In contrast, technologies using the genetic material of viruses offer new possibilities by effectively delivering genes (DNA, RNA, etc.) that express specific viral antigens in the human body, inducing an immune response, and inhibiting viral replication. For these technologies, various genetic material delivery vehicles are being developed, and these have the characteristics of rapid adjustment to genetic material variants and various antiviral effects, enabling a rapid and extensive response, especially against mutant viruses such as SARS-CoV-2.

이에, 본 발명자들은 항 바이러스 활성을 가지는 유전 물질 전달체를 제조하기 위하여 연구하던 중, 금 나노 입자와 DNA를 접합한 전달체 기술을 사용하여, 바이러스 RNA로부터 유래된, 특히 보존된 5’비번역 부위에 포함되는 뉴클레오티드 서열에 상보적으로 결합하는 안티센스 올리고뉴클레오타이드와 이에 결합하여 mRNA의 절단을 유발시키는 리보핵산가수분해효소H (RNase H)를 금나노 입자에 함께 결합하여 전달할 경우, 상기 바이러스에 대한 우수한 항 바이러스 효과를 가질 수 있음을 발견하고, 본 발명을 완성하였다. Accordingly, the inventors of the present invention, while conducting research to manufacture a genetic material delivery vehicle having antiviral activity, discovered that when an antisense oligonucleotide that complementarily binds to a nucleotide sequence derived from viral RNA, particularly included in a conserved 5'untranslated region, and ribonuclease H (RNase H), which binds to the antisense oligonucleotide and induces cleavage of mRNA, are delivered together with gold nanoparticles, an excellent antiviral effect against the virus can be achieved, thereby completing the present invention.

따라서 본 발명의 목적은 금 나노 입자 및 상기 금 나노 입자 표면에 결합되는 바이러스 RNA유래 서열에 상보적으로 결합하는 안티센스 올리고뉴클레오타이드(ASO); 및 mRNA의 절단을 유발시키는 리보핵산가수분해효소 H(RNase H)를 포함하는 나노 입자 전달체를 제공하는 것이다. Accordingly, the purpose of the present invention is to provide a nanoparticle delivery vehicle comprising a gold nanoparticle and an antisense oligonucleotide (ASO) that complementarily binds to a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA.

본 발명의 다른 목적은 상기의 나노 입자 전달체를 포함하는 항 바이러스 조성물을 제공하는 것이다.Another object of the present invention is to provide an antiviral composition comprising the nanoparticle carrier described above.

상기와 같은 본 발명의 목적을 달성하기 위하여, 본 발명은 금 나노 입자 및 상기 금 나노 입자 표면에 결합되는 바이러스 RNA유래 서열에 상보적으로 결합하는 안티센스 올리고뉴클레오타이드(ASO); 및 mRNA의 절단을 유발시키는 리보핵산가수분해효소 H(RNase H)를 포함하는 나노 입자 전달체를 제공한다. In order to achieve the above-described purpose of the present invention, the present invention provides a nanoparticle delivery vehicle comprising a gold nanoparticle; an antisense oligonucleotide (ASO) that complementarily binds to a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA.

본 발명의 다른 목적을 달성하기 위하여 본 발명은 상기의 나노 입자 전달체를 포함하는 항 바이러스 조성물을 제공한다. In order to achieve another object of the present invention, the present invention provides an antiviral composition comprising the nano particle carrier.

이하, 본 발명을 상세하게 설명한다. Hereinafter, the present invention will be described in detail.

본 발명은 금 나노 입자 및 상기 금 나노 입자 표면에 결합되는 바이러스 RNA유래 서열에 포함되는 뉴클레오티드 서열에 상보적으로 결합하는 안티센스 올리고뉴클레오타이드(ASO); 및 mRNA의 절단을 유발시키는 리보핵산가수분해효소 H(RNase H)를 포함하는 나노 입자 전달체에 대한 것이다. The present invention relates to a nanoparticle delivery vehicle comprising a gold nanoparticle and an antisense oligonucleotide (ASO) that complementarily binds to a nucleotide sequence included in a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA.

본 발명의 나노 입자 전달체는 바이러스 게놈에 으로부터 유래한 5’비번역 부위(UTR)에 위치하는 게놈 RNA 서열에 포함되는 뉴클레오티드 서열에 상보적으로 결합하는 안티센스 올리고뉴클레오타이드(ASO)를 포함한다. The nanoparticle delivery vehicle of the present invention comprises an antisense oligonucleotide (ASO) that complementarily binds to a nucleotide sequence contained in a genomic RNA sequence located in the 5’untranslated region (UTR) derived from the viral genome.

안티센스 올리고뉴클레오티드(ASO)는 마이크로RNA 또는 긴 비암호화 RNA와 같은 비암호화 RNA뿐만 아니라 특정 mRNA에 상보적인 단일 가닥 데옥시리보뉴클레오티드를 의미한다(Beermann, Piccoli, Viereck, & Thum, 2016; Bennett, 2019). 안티센스 올리고뉴클레오티드는 Watson-Crick 염기쌍을 통해 표적 RNA 서열에 특이적으로 결합하고 RNaseH와 같은 내인성 뉴클레아제에 의해 결합된 RNA의 분해를 촉진한다(Watts & Corey, 2012). 즉, RNaseH는 RNA/DNA 하이브리드 듀플렉스 구조를 인식하고 mRNA 절단 및 게놈의 분해를 유도할 수 있다(Bennett, Baker, Pham, Swayze, & Geary, 2017). Antisense oligonucleotides (ASOs) are single-stranded deoxyribonucleotides complementary to specific mRNAs as well as noncoding RNAs such as microRNAs or long noncoding RNAs (Beermann, Piccoli, Viereck, & Thum, 2016; Bennett, 2019). Antisense oligonucleotides specifically bind to target RNA sequences through Watson–Crick base pairing and promote the degradation of bound RNA by endogenous nucleases such as RNaseH (Watts & Corey, 2012). That is, RNaseH can recognize RNA/DNA hybrid duplex structures and induce mRNA cleavage and genome degradation (Bennett, Baker, Pham, Swayze, & Geary, 2017).

즉, 본 발명에서 용어 "안티센스 올리고뉴클레오티드(ASO)"는 다른 RNA 또는 DNA (표적 RNA, DNA)에 결합하는 RNA 또는 DNA 분자를 의미한다. 가령, RNA 올리고뉴클레오티드이면, 이는 RNA-RNA 상호작용에 의해 다른 RNA 표적에 결합하고 표적 RNA의 활성을 변경시킨다. 안티센스 올리고뉴클레오티드는 특정 폴리뉴클레오티드의 발현 및/또는 기능을 상향 조절 또는 하향 조절할 수 있다. 이러한 정의에는 치료적 관점, 진단적 관점, 또는 다른 관점에 서 유용한 임의의 외래 RNA 또는 DNA 분자가 포함되는 것으로 의도된다. 일 예로써, 안티센스 RNA 또는 DNA 분자, 간섭 RNA (RNAi), 마이크로 RNA, 미끼 RNA 분자, siRNA, 효소 RNA, 치료 편집 RNA 및 안티센스 올리고머 화합물, 안티센스 올리고뉴클레오티드, 외부 가이드 서열 (external guide sequence, EGS) 올리고뉴클레오티드, 프라이머, 프로브, 그리고 표적 핵산의 최소한 일부분에 혼성화되는 다른 올리고머 화합물이 포함될 수 있다. 따라서 이들 화합물은 단일-가닥, 이중-가닥, 부분적으로 단일-가닥, 또는 원형 올리고머 화합물의 형태로 도입될 수 있다.That is, the term "antisense oligonucleotide (ASO)" in the present invention means an RNA or DNA molecule that binds to another RNA or DNA (target RNA, DNA). For example, if it is an RNA oligonucleotide, it binds to another RNA target by RNA-RNA interaction and modifies the activity of the target RNA. Antisense oligonucleotides can up-regulate or down-regulate the expression and/or function of a specific polynucleotide. It is intended that any foreign RNA or DNA molecule useful from a therapeutic, diagnostic, or other perspective be included in this definition. By way of example, antisense RNA or DNA molecules, interfering RNA (RNAi), micro RNA, bait RNA molecules, siRNA, enzymatic RNA, therapeutic editing RNA, and antisense oligomeric compounds, antisense oligonucleotides, external guide sequence (EGS) oligonucleotides, primers, probes, and other oligomeric compounds that hybridize to at least a portion of a target nucleic acid may be included. Therefore, these compounds can be introduced in the form of single-stranded, double-stranded, partially single-stranded, or circular oligomeric compounds.

일 실시예로서, 본 발명의 상기 바이러스의 RNA 유래 서열은, 바이러스 RNA의 5’비번역 부위(UTR)에 상보적으로 결합하는 안티센스 올리고뉴클레오타이드(ASO)는 SARS-CoV-2 게놈 RNA의 5'UTR에 포함되는 뉴클레오티드 서열을 표적으로 하는 것일 수 있다. 상기 안티센스 올리고뉴클레오티드는 상기 SARS-CoV-2 게놈 RNA의 5'UTR에 포함되는 뉴클레오티드 서열에 상보적인 서열을 포함하며, 상기 뉴클레오티드 서열은 SARS-CoV-2의 5'UTR 영역 중, RNaseH 결합 부위인 리더 서열(Leader sequence)을 포함할 수 있다. As an example, the RNA-derived sequence of the virus of the present invention may be an antisense oligonucleotide (ASO) that complementarily binds to the 5'untranslated region (UTR) of the viral RNA, which may target a nucleotide sequence included in the 5'UTR of the SARS-CoV-2 genomic RNA. The antisense oligonucleotide includes a sequence complementary to a nucleotide sequence included in the 5'UTR of the SARS-CoV-2 genomic RNA, and the nucleotide sequence may include a leader sequence, which is an RNaseH binding site, among the 5'UTR region of SARS-CoV-2.

또한, 본 발명의 금 나노 입자 전달체는 mRNA의 절단을 유발시키는 리보핵산가수분해효소 RNase H를 포함한다. Additionally, the gold nanoparticle carrier of the present invention comprises ribonuclease RNase H, which induces cleavage of mRNA.

상기 RNase H는 전사 및 복제와 같은 다양한 유전적 과정에 관여하는 필수 엔도뉴클레아제(Aguilera & Garcia-Muse, 2012; Crossley, Bocek, & Cimprich, 2019)이다. 전사 과정에서 RNA 가닥은 주형 DNA와 결합하여 RNA/DNA 이중 구조를 만들 수 있으며 비주형 가닥은 단일 가닥 DNA(ssDNA)로 대체되어 R-loop라는 구조를 형성한다 (Thomas, White, & 데이비스, 1976). R-루프는 전사 조절에 도움이 될 수 있으나, 필요하지 않은 상황에서는 게놈 불안정성의 모든 원인인 이중 가닥 DNA 절단(DSB), 염색체 재배열 및 초돌연변이를 일으킬 수 있다(Richard & Manley, 2017). RNase H는 RNA/DNA 듀플렉스에서 RNA를 분해하여 불필요한 R-루프의 억제를 통해 게놈 안정성을 증가시킨다(Nguyen et al., 2017). 즉, "RNase H 효소"는 RNA-DNA 하이브리드 중 RNA를 가수분해하는 효소로서, 송아지 흉선에서 처음 발견된 RNase H는 그 이후에 여러 생물에서 개시되었다. The above RNase H is an essential endonuclease involved in various genetic processes such as transcription and replication (Aguilera & Garcia-Muse, 2012; Crossley, Bocek, & Cimprich, 2019). During transcription, the RNA strand can combine with the template DNA to form an RNA/DNA duplex, and the non-template strand is replaced by single-stranded DNA (ssDNA) to form a structure called R-loop (Thomas, White, & Davis, 1976). R-loops can help regulate transcription, but when they are not needed, they can cause double-stranded DNA breaks (DSBs), chromosomal rearrangements, and hypermutations, all causes of genome instability (Richard & Manley, 2017). RNase H increases genome stability by degrading RNA in the RNA/DNA duplex and suppressing unnecessary R-loops (Nguyen et al., 2017). That is, "RNase H enzyme" is an enzyme that hydrolyzes RNA among RNA-DNA hybrids, and RNase H, which was first discovered in calf thymus, was subsequently discovered in various organisms.

본 발명의 금 나노 입자에 결합되는 RNaseH는 다양한 생물로부터 유래된 것일 수 있으며, 유래 생물에 제한되는 것은 아니나, 피로코커스 퓨리오서스 (Pyrococcus furiosus), 피로코커스 호리코시 (Pyrococcus horikoshi), 써모코커스 리토랄리스 (Thermococcus litoralis), 써머스 써모필러스 (Thermus thermophilus) 또는 대장균 (E. coli)으로부터 분리된 것일 수 있다. 일 예로서, 본 발명의 RNaseH는 RNaseH1일 수 있으며, 상기 RNaseH1의 아미노산 서열과 50%, 60%, 70%, 80%, 90%, 95% 또는 99% 상동성을 갖는 RNaseH일 수 있다. The RNaseH bound to the gold nanoparticles of the present invention may be derived from various organisms, and is not limited to the derived organisms, but may be isolated from Pyrococcus furiosus, Pyrococcus horikoshi, Thermococcus litoralis, Thermos thermophilus or E. coli. As an example, the RNaseH of the present invention may be RNaseH1, and may be an RNaseH having 50%, 60%, 70%, 80%, 90%, 95% or 99% homology to the amino acid sequence of RNaseH1.

본 발명의 상기 안티센스 올리고뉴클레오티드(ASO) 및 RNaseH는 금 나노 입자의 표면에 결합된다. The antisense oligonucleotide (ASO) and RNaseH of the present invention are bound to the surface of gold nanoparticles.

본 발명의 상기 금 나노 입자는 나노 단위의 직경, 바람직하게는 5-500 nm, 보다 바람직하게는 10-200 nm의 직경을 가지는 금 입자로서, 안정한 입자의 형태로 제조가 쉬우며, 크기 조절이 용이하고, 망간, 알루미늄, 카드늄, 납, 수은, 코발트, 니켈, 베릴륨 등의 중금속과 달리 인체에 무해하여 높은 생체친화성을 가진다. 금 나노 입자는 직경이 500 nm 이상으로 커질 경우 나노 입자로서의 특성이 소멸될 뿐 아니라, 나노 물질의 특성이 없는 금 표면과 티올기 등의 작용기와의 결합이 약해지기 때문에 금 나노 입자를 이용한 전달체를 제조하기 어려운 단점을 가진다.The gold nanoparticles of the present invention are gold particles having a diameter in the nanometer range, preferably 5-500 nm, more preferably 10-200 nm, which are easy to manufacture in the form of stable particles, are easy to control in size, and are harmless to the human body, unlike heavy metals such as manganese, aluminum, cadmium, lead, mercury, cobalt, nickel, and beryllium, and thus have high biocompatibility. When the diameter of gold nanoparticles increases to more than 500 nm, not only do their characteristics as nanoparticles disappear, but also the bonding between the gold surface, which does not have the characteristics of a nanomaterial, and functional groups such as thiol groups becomes weak, making it difficult to manufacture a carrier using gold nanoparticles.

본 발명에서 이용되는 금 나노 입자는 그 방법에 제한되는 것은 아니나, 예컨대, 다음과 같이 제조될 수 있다: HAuCl4를 금 공급원으로 하고, 소듐 시트레이트를 환원제로 하여 HAuCl4를 환원시켜 금 나노입자를 제조한다. 이 경우, 금 나노입자의 크기는 첨가하는 시트레이트를 달리해 줌으로써 조절이 가능한다. 즉, 시트레이트의 첨가량을 증가시킬수록 핵형성(nucleation)이 많이 되기 때문에 금 나노 입자의 크기는 감소한다.The gold nanoparticles used in the present invention are not limited to the method, but can be manufactured as follows: HAuCl 4 is used as a gold source, and HAuCl 4 is reduced using sodium citrate as a reducing agent to manufacture gold nanoparticles. In this case, the size of the gold nanoparticles can be controlled by varying the amount of citrate added. That is, as the amount of citrate added increases, nucleation increases, and thus the size of the gold nanoparticles decreases.

본 발명의 상기 금 나노 입자는 표면에 안티센스 올리고뉴클레오티드 및 RNaseH를 결합하기 위한 표면 개질을 포함한다. 또한, 상기 안티센스 올리고뉴클레오티드 및 RNaseH는 상기 금 나노 입자 표면에 결합하기 위하여 하나 이상의 작용기성(functionality)을 포함할 수 있다. The gold nanoparticles of the present invention include surface modification for binding antisense oligonucleotides and RNaseH to the surface. In addition, the antisense oligonucleotides and RNaseH may include one or more functionalities for binding to the surface of the gold nanoparticles.

상기 작용기성은 티올기 또는 아민기일 수 있으며, 상기 안티센스 올리고뉴클레오티드 또는 상기 안티센스 올리고뉴클레오티드와 결합하는 RNAI의 하나 이상의 잔기에 포함될 수 있다. 이에 제한되는 것은 아니나, 상기 안티센스 올리고뉴클레오티드 또는 상기 안티센스 올리고뉴클레오티드와 결합하는 RNAI는 티올화된 하나 이상의 잔기를 포함하며, 이를 통해 금 나노 입자 표면에 직접 결합할 수 있다. The above functional group may be a thiol group or an amine group and may be included in one or more residues of the antisense oligonucleotide or the RNAI that binds to the antisense oligonucleotide. Although not limited thereto, the antisense oligonucleotide or the RNAI that binds to the antisense oligonucleotide comprises one or more thiolated residues, through which it can directly bind to the surface of the gold nanoparticles.

본 발명의 일 실시예에서는 금 나노 입자 표면에 티올화된 RNAI 올리고(oligo)가 결합되며, 상기 티올화된 RNAI 올리고는 본 발명의 안티센스 올리고뉴클레오티드 서열과 특이적 결합을 통해 금 나노 입자 표면에 안티센스 올리고뉴클레오티드를 결합시켰다. In one embodiment of the present invention, a thiolated RNAI oligo is bound to the surface of a gold nanoparticle, and the thiolated RNAI oligo binds the antisense oligonucleotide to the surface of the gold nanoparticle through specific binding to the antisense oligonucleotide sequence of the present invention.

또한, 본 발명의 상기 금 나노 입자는 표면에 앱타머를 포함할 수 있다. 본 발명에서, 상기 "앱타머 (aptamer)"란 그 자체로 안정된 삼차구조를 가지면서 표적분자에 높은 친화성과 특이성으로 결합할 수 있는 특징을 가진 단일가닥 핵산 (DNA, RNA 또는 변형핵산)을 의미하며, SELEX(Systematic Evolution of Ligands of Exponential enrichment)라는 방법으로 원하는 다양한 목적 물질 (단백질, 당, 염색물질, DNA,금속이온, 세포 등)에 대한 앱타머를 개발할 수 있다. 본 발명에서 금 나노입자와 결합하는 앱타머는 특히 RNase H를 금 나노 입자 표면에 결합하기 위하여, 다양한 tag의 앱타머 또는 단백질 특이적 앱타머의 어떠한 종류도 사용할 수 있으며, 특별히 제한되는 것은 아니다.In addition, the gold nanoparticles of the present invention may include an aptamer on the surface. In the present invention, the "aptamer" refers to a single-stranded nucleic acid (DNA, RNA or modified nucleic acid) that has a stable tertiary structure in itself and has the characteristic of being able to bind to a target molecule with high affinity and specificity, and aptamers for various desired target substances (proteins, sugars, dyes, DNA, metal ions, cells, etc.) can be developed by a method called SELEX (Systematic Evolution of Ligands of Exponential enrichment). In the present invention, the aptamer that binds to the gold nanoparticles may use any kind of aptamer of various tags or protein-specific aptamers, particularly for binding RNase H to the surface of the gold nanoparticles, and is not particularly limited.

본 발명의 일 실시예에서는 히스티딘 tag(His-tag)를 포함하는 앱타머를 금 나노 입자에 결합하여, 상기 히스티딘 tag 앱타머를 통해 RNaseH를 금 나노 입자 표면에 결합시켰다. In one embodiment of the present invention, an aptamer including a histidine tag (His-tag) was bound to a gold nanoparticle, and RNaseH was bound to the surface of the gold nanoparticle through the histidine tag aptamer.

본 발명의 금 나노 입자 전달체는 금 나노 입자 표면에 하나 이상 결합된 안티센스 올리고뉴클레오티드 및 RNaseH 결합을 위한 히스티딘 tag 압타머를 포함하며, 상기 안티센스 올리고뉴클레오티드는 직접 또는 RNAI를 통해 금 나노 입자 표면에 결합할 수 있다. 즉, 금 나노 입자 표면에 결합되는 RNAI, 안티센스 올리고뉴클레오티드 및 히스티딘 tag 압타머를 포함하는 총 올리고(oligo)의 수는 이에 제한되는 것은 아니나, 1 내지 100개일 수 있으며, 바람직하게는 40 내지 50개일 수 있다. The gold nanoparticle carrier of the present invention comprises at least one antisense oligonucleotide bound to the surface of a gold nanoparticle and a histidine tag aptamer for RNaseH binding, wherein the antisense oligonucleotide can bind to the surface of the gold nanoparticle directly or through RNAI. That is, the total number of oligos including RNAI, antisense oligonucleotide, and histidine tag aptamer bound to the surface of the gold nanoparticle is not limited thereto, but may be 1 to 100, and preferably 40 to 50.

상기 금 나노 입자 전달체는 표면에 결합된 안티센스 올리고뉴클레오티드 서열이 표적으로 하는 바이러스의 mRNA의 5'UTR 부분에 존재하는 리더 서열에 결합되어, RNaseH의 작용으로 인한 mRNA 분해를 유도하여, 바이러스에 의한 감염을 치료할 수 있는 효과를 가진다. The above gold nanoparticle carrier has an effect of treating viral infection by binding to a leader sequence present in the 5'UTR portion of the mRNA of the target virus through an antisense oligonucleotide sequence bound to the surface, thereby inducing mRNA degradation due to the action of RNaseH.

즉, 본 발명의 상기 금 나노 입자 전달체는 바이러스에 의한 감염병 치료제의 용도로 활용될 수 있다. That is, the gold nanoparticle carrier of the present invention can be utilized as a therapeutic agent for infectious diseases caused by viruses.

본 발명에서, 상기 바이러스는 그 종류에 제한되는 것은 아니나, SARS-CoV-2 (COVID-19), SARS-CoV (SARS) 및 MERS-CoV (MERS)를 포함하는 코로나바이러스; 인플루엔자바이러스; 에볼라바이러스; HSV-1, HSV-2, VZV 및 CMV를 포함하는 헤르페스바이러스; HIV를 포함하는 레트로바이러스, HBV, HCV, 및 HDV를 포함하는 간염바이러스; 뎅기 및 지카 바이러스를 포함하는 플라비바이러스; 로타바이러스; 파필로마바이러스; 홍역바이러스; 폴리오바이러스; 콕사키바이러스; 및 아데노바이러스를 포함하는 군으로부터 선택될 수 있다. In the present invention, the virus is not limited to a type, but may be selected from the group including coronaviruses including SARS-CoV-2 (COVID-19), SARS-CoV (SARS), and MERS-CoV (MERS); influenza virus; Ebola virus; herpesviruses including HSV-1, HSV-2, VZV, and CMV; retroviruses including HIV, hepatitis viruses including HBV, HCV, and HDV; flaviviruses including dengue and Zika viruses; rotavirus; papillomavirus; measles virus; poliovirus; coxsackievirus; and adenovirus.

본 명세서에서 용어 "치료"는 본 발명에 따른 상기 약학적 조성물의 투여에 의해 증세가 호전되거나 이롭게 되는 모든 행위를 말한다. The term “treatment” as used herein refers to any action that improves symptoms or is beneficial by administering the pharmaceutical composition according to the present invention.

본 발명의 상기 약학적 조성물은 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 현탁액, 에멀젼, 시럽, 에어로졸 등의 경구형 제형, 외용제, 좌제 및 멸균 주사용액의 형태로 제형화하여 사용될 수 있으며, 상기 제형화에 필요한 담체 혹은 부형제 등을 추가로 포함할 수 있다. 상기 유효 성분에 추가로 포함될 수 있는 약학적으로 허용 가능한 담체, 부형제 및 희석제로는 락토즈, 덱스트로즈, 수크로스, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로즈, 메틸 셀룰로즈, 미정질 셀룰로스, 폴리비닐 피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 마그네슘 스테아레이트 및 광물유 등이 포함된다. 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제된다. The pharmaceutical composition of the present invention can be used by being formulated in the form of oral dosage forms such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc., external preparations, suppositories, and sterile injection solutions according to conventional methods, and may further include carriers or excipients necessary for the formulation. Pharmaceutically acceptable carriers, excipients, and diluents that may be additionally included in the effective ingredient include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, magnesium stearate, mineral oil, and the like. When formulating, it is usually prepared using diluents or excipients such as fillers, bulking agents, binders, wetting agents, disintegrants, and surfactants.

예를 들어, 경구 투여를 위한 고형 제제에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 이러한 고형제제는 상기 추출물 또는 화합물에 적어도 하나 이상의 부형제 적어도 면, 전분, 칼슘카보네이트 (calcium carbonate), 수크로스 (sucrose) 또는 락토오스 (lactose), 젤라틴 등을 섞어 조제된다. 또한 단순한 부형제 이외에 마그네슘 스티레이트 탈크 같은 윤활제들도 사용된다. 경구 투여를 위한 액상제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데 흔히 사용되는 단순희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. For example, solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and these solid preparations are prepared by mixing the extract or compound with at least one excipient, at least cotton, starch, calcium carbonate, sucrose or lactose, gelatin, etc. In addition to simple excipients, lubricants such as magnesium stearate and talc are also used. Liquid preparations for oral administration include suspensions, oral solutions, emulsions, syrups, etc., and in addition to commonly used simple diluents such as water and liquid paraffin, various excipients such as wetting agents, sweeteners, fragrances, and preservatives may be included.

비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조제제, 좌제가 포함된다. 비수성용제, 현탁제로는 프로필렌글리콜 (propylene glycol), 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔 (witepsol), 마크로골, 트윈 (tween) 61, 카카오지, 라우린지, 글리세로제라틴 등이 사용될 수 있다.Preparations for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized preparations, and suppositories. Non-aqueous solutions and suspensions may include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate. Suppository bases may include witepsol, macrogol, Tween 61, cacao butter, laurin butter, and glycerogelatin.

본 발명의 약학적 조성물은 목적하는 방법에 따라 경구 또는 비경구 투여(정맥주사, 피하, 복강 내 또는 국소에 적용)될 수 있으며, 투여량은 환자의 상태 및 체중, 질병의 정도 및 약물 형태, 투여 경로 및 시간에 따라 다르며, 당업자에 의해 적절한 형태로 선택될 수 있다. The pharmaceutical composition of the present invention can be administered orally or parenterally (intravenously, subcutaneously, intraperitoneally, or topically) depending on the intended method, and the dosage varies depending on the patient's condition and weight, the degree of the disease, the drug form, the route of administration, and the time, and can be selected in an appropriate form by a person skilled in the art.

본 발명의 상기 약학적 조성물은, 약학적으로 유효한 양으로 투여한다. 본 발명에서 "약학적으로 유효한 양"이란 의학적 치료에 적용 가능한 합리적인 양으로, 질병을 치료하기에 충분한 양을 의미하며, 그 기준은 환자의 질환, 중증도, 약물 활성, 약물에 대한 민감도, 투여 시간, 투여 경로 및 배출 비율, 치료 기간, 병용되는 성분 및 기타 사항에 따라 결정될 수 있다. 본 발명의 상기 약학적 조성물은, 개별 치료제 혹은 다른 치료제와 병용하여 투여될 수 있고, 종래의 치료제와는 순차적 또는 동시에 투여될 수 있다. 상기 요소들을 모두 고려하여 부작용을 최소화할 수 있는 수준으로 투여량을 결정할 수 있고, 이는 당업자에 의해 용이한 수준으로 결정될 수 있다. 구체적으로 상기 약학적 조성물의 투여량은 환자의 연령, 체중, 중증도, 성별 등에 따라 달라질 수 있으며, 일반적으로 체중 1kg 당 0.001 내지 150 mg, 보다 바람직하게는 0.01 내지 100mg의 양을 매일 또는 격일, 1일 1 내지 3회 투여할 수 있다. 다만, 이는 예시적인 것으로, 상기 투여량은 필요에 달리 설정할 수 있다.The pharmaceutical composition of the present invention is administered in a pharmaceutically effective amount. In the present invention, the term "pharmaceutically effective amount" means a reasonable amount applicable to medical treatment, and an amount sufficient to treat a disease, and the standard thereof may be determined according to the patient's disease, severity, drug activity, drug sensitivity, administration time, administration route, and excretion rate, treatment period, concomitantly used ingredients, and other matters. The pharmaceutical composition of the present invention may be administered individually or in combination with other therapeutic agents, and may be administered sequentially or simultaneously with conventional therapeutic agents. The dosage may be determined at a level that minimizes side effects by considering all of the above factors, and this may be easily determined by a person skilled in the art. Specifically, the dosage of the pharmaceutical composition may vary depending on the patient's age, weight, severity, sex, etc., and may generally be administered in an amount of 0.001 to 150 mg per 1 kg of body weight, more preferably 0.01 to 100 mg, once to three times a day, every day or every other day. However, this is an example and the dosage may be set differently as needed.

본 발명은 금 나노 입자 및 상기 금 나노 입자 표면에 결합되는 바이러스 RNA 유래 서열에 포함되는 뉴클레오티드 서열에 상보적으로 결합하는 안티센스 올리고뉴클레오타이드(ASO); 및 mRNA의 절단을 유발시키는 리보핵산가수분해효소 H(RNase H)를 포함하는 항 바이러스용 유전자 운반체 및 이를 포함하는 항 바이러스 조성물에 대한 것으로, 바이러스의 mRNA 합성을 저해하여 SARS-CoV-2를 포함하는 다양한 바이러스의 감염에 의해 유발되는 질병에 대한 치료 효과를 가질 수 있다. The present invention relates to an antiviral gene carrier comprising a gold nanoparticle and an antisense oligonucleotide (ASO) that complementarily binds to a nucleotide sequence included in a viral RNA-derived sequence bound to the surface of the gold nanoparticle; and ribonuclease H (RNase H) that induces cleavage of mRNA, and an antiviral composition comprising the same, which can have a therapeutic effect on diseases caused by infection with various viruses including SARS-CoV-2 by inhibiting viral mRNA synthesis.

도 1은 기능화된 DNA 올리고를 통해 금 나노 입자 표면에 안티센스 올리고뉴클레오타이드(ASO) 및 RNaseH가 결합하는 과정을 나타낸 모식도이다. Figure 1 is a schematic diagram showing the process of binding of antisense oligonucleotides (ASOs) and RNaseH to the surface of gold nanoparticles through functionalized DNA oligos.

도 2는 표시된 농도(0, 1, 2, 4 및 8μM)의 RNase H를 AuNP-AptHis(5nM)과 단순 혼합하여 10분 동안 반응한 결과로, AuNP-AptHis-RNH(결합 RNase H)를 SDS-폴리아크릴아미드 겔 전기영동으로 분석한 것이다. AuNP-AptHis에 결합된 RNase H의 양을 밴드 강도의 정량화하고 알려진 양의 RNase H(total RNase H)와 비교하였다. 결과는 정량화되어 그래프로 표시되었으며, 데이터는 최소 세 번의 독립적인 실험의 평균 ± 평균의 표준 오차(SEM)로 표시되었다. Figure 2 shows the results of simply mixing RNase H at the indicated concentrations (0, 1, 2, 4, and 8 μM) with AuNP- AptHis (5 nM) and reacting for 10 min, and the AuNP- AptHis -RNH (bound RNase H) was analyzed by SDS-polyacrylamide gel electrophoresis. The amount of RNase H bound to AuNP- AptHis was quantified as band intensity and compared to the known amount of RNase H (total RNase H). The results were quantified and displayed in a graph, and the data are expressed as the mean ± standard error of the mean (SEM) of at least three independent experiments.

도 3은 AuNP 물질의 세포 독성을 세포생존율 분석으로 평가한 결과로서, 색상은 AuNP 무처리군(검은색), PBS 처리군(빨간색), AuNP-DNA 올리고(노란색) 및 AuNP-DNA 올리고-RNH(녹색)로 나타내었다. 상대적인 세포 생존율은 세포만의 생존 세포와 비교하여 생존 세포의 백분율로 표시되었다. 데이터는 세 번의 독립적인 실험에서 평균 ± SEM으로 표시하였다. NS, 불명확.Figure 3 shows the results of evaluating the cytotoxicity of AuNP materials by cell viability analysis, and the colors represent the AuNP untreated group (black), PBS treated group (red), AuNP-DNA oligo (yellow), and AuNP-DNA oligo-RNH (green). The relative cell viability is expressed as the percentage of viable cells compared to the viable cells of cells alone. The data are expressed as the mean ± SEM of three independent experiments. NS, unclear.

도 4는 PBS, AuNPRNAI/His, ASO, RNase H, AuNPRNAI/His에 로드된 ASO 및/또는 RNase H로 각각 처리된 VERO-E6 세포의 공초점 형광 현미경 이미지이며, 각 이미지는 Cy3 표지 ASO(빨간색), Alexa 488 표지 RNase H(녹색) 및 핵이 DAPI(파란색)로 염색되었음을 나타낸다. 하단의 그래프는 상기 이미지의 형광 강도를 정량화한 것으로, 형광 강도의 정량화는 lmage J 소프트웨어(NIH)를 사용하여 보정된 총 세포 형광(CTCF)으로 표현하였다. [CTCF = 통합 밀도 - (선택한 셀의 면적 × 배경 판독값의 평균 형광)]. 데이터는 세 번의 독립적인 실험에서 평균 ± SEM으로 표시되며, 기호(*,#)는 통계적 유의차를 나타낸다. (***, ###p < 0.001).Figure 4 is a confocal fluorescence microscopy image of VERO-E6 cells treated with PBS, AuNP RNAI/His , ASO, RNase H, ASO loaded on AuNP RNAI/His , and/or RNase H, respectively, wherein each image shows Cy3-labeled ASO (red), Alexa 488-labeled RNase H (green), and nuclei stained with DAPI (blue). The graphs at the bottom are quantified fluorescence intensities of the images, and the fluorescence intensity quantification was expressed as corrected total cell fluorescence (CTCF) using lmage J software (NIH). [CTCF = integrated density - (area of selected cells × mean fluorescence of background readings)]. Data are presented as mean ± SEM of three independent experiments, and symbols (*,#) indicate statistical significance. (***, ###p < 0.001).

도 5는 RNase H 의존적 ASO 메커니즘을 모식화한 것이다. Figure 5 is a schematic diagram of the RNase H-dependent ASO mechanism.

도 6은 AuNPRNAI/His(1nM)에 로드된 Scrambled DNA, ASO 및/또는 RNase H 처리 후, 24시간 이후 VERO-E6 세포에서 포지티브 센스 가닥 SARS-CoV-2의 5'UTR mRNA에 대한 실시간 qRT-PCR 분석 결과이다. 각각 무처리군(검은색), RNase H를 처리하지 않은 군(빨간색) 및 RNase H를 처리한 군(녹색)을 나타낸다. 데이터는 세 번의 독립적인 실험에서 평균 ± SEM으로 표시된다. 별표는 통계적으로 유의한 값을 나타낸다(***p < 0.001). NS, 불명확.Figure 6 is a real-time qRT-PCR analysis of positive-sense strand SARS-CoV-2 5'UTR mRNA in VERO-E6 cells 24 h after treatment with scrambled DNA, ASO, and/or RNase H loaded onto AuNP RNAI/His (1 nM). The untreated group (black), the group not treated with RNase H (red), and the group treated with RNase H (green) are shown, respectively. Data are presented as the mean ± SEM of three independent experiments. Asterisks indicate statistically significant values (***p < 0.001). NS, unclear.

도 7A는 VERO-E6 세포에서 포지티브 센스 가닥 SARS-CoV-2의 5'UTR mRNA 발현을 AuNPRNAI/His-Scrambled DNA-RNH(검은색) 및 AuNPRNAI/His-ASO-RNH(녹색)를 주입하여 4시간 간격으로 실시간 qRT-PCR 분석으로 확인한 결과이다. 데이터는 세 번의 독립적인 실험에서 평균 ± SEM으로 표시된다. 별표는 통계적으로 유의한 값을 나타낸다(**p < 0.01; ***p < 0.001). NS, 불명확.Figure 7A shows the results of real-time qRT-PCR analysis of the expression of 5'UTR mRNA of positive-sense strand SARS-CoV-2 in VERO-E6 cells by injecting AuNP RNAI/His -Scrambled DNA-RNH (black) and AuNP RNAI/His -ASO-RNH (green) at 4-h intervals. Data are presented as the mean ± SEM of three independent experiments. Asterisks indicate statistically significant values (**p <0.01; ***p < 0.001). NS, unclear.

도 7B는 상기 도 7A의 qRT-PCR 산물을 1.5% 아가로즈 겔에서 분리하고 에티디움 브로마이드를 사용하여 시각화한 것이다. GAPDH는 로딩 컨트롤로 사용되었다.Figure 7B shows the qRT-PCR product of Figure 7A separated on a 1.5% agarose gel and visualized using ethidium bromide. GAPDH was used as a loading control.

도 8은 24시간 동안 AuNPRNAI/His(1nM)에 로드된 Scrambled DNA, SO 및/또는 RNase H 처리 후, 24시간 이후 VERO-E6 세포에서 네거티브 센스 가닥 SARS-CoV-2의 5'UTR mRNA에 대한 실시간 qRT-PCR 분석 결과이다. 각각 무처리군(검은색), RNase H를 처리하지 않은 군(빨간색) 및 RNase H를 처리한 군(노란색)을 나타낸다. 데이터는 세 번의 독립적인 실험에서 평균 ± SEM으로 표시된다. 별표는 통계적으로 유의한 값을 나타낸다. NS, 불명확.Figure 8 is a real-time qRT-PCR analysis of 5'UTR mRNA of negative-sense strand SARS-CoV-2 in VERO-E6 cells 24 h after treatment with Scrambled DNA, SO, and/or RNase H loaded on AuNP RNAI/His (1 nM) for 24 h. The untreated group (black), the group not treated with RNase H (red), and the group treated with RNase H (yellow) are shown, respectively. Data are presented as the mean ± SEM of three independent experiments. Asterisks indicate statistically significant values. NS, unclear.

도 9는 24시간 동안 AuNPRNAI/His(1nM)에 로드된 Scrambled DNA, SON 및/또는 RNase H 처리 후, 24시간 이후 VERO-E6 세포에서 네거티브 센스 가닥 SARS-CoV-2의 5'UTR mRNA에 대한 실시간 qRT-PCR 분석 결과이다. 각각 무처리군(검은색), RNase H를 처리하지 않은 군(빨간색) 및 RNase H를 처리한 군(보라색)을 나타낸다. 데이터는 세 번의 독립적인 실험에서 평균 ± SEM으로 표시된다. 별표는 통계적으로 유의한 값을 나타낸다. NS, 불명확.Figure 9 is the real-time qRT-PCR analysis of 5'UTR mRNA of negative-sense strand SARS-CoV-2 in VERO-E6 cells 24 h after treatment with Scrambled DNA, SON, and/or RNase H loaded on AuNP RNAI/His (1 nM) for 24 h. The untreated group (black), the group not treated with RNase H (red), and the group treated with RNase H (purple) are shown, respectively. Data are presented as the mean ± SEM of three independent experiments. Asterisks indicate statistically significant values. NS, unclear.

도 10은 SARS-CoV-2의 (+) 5'UTR에 융합된 GFP 리포터 유전자의 발현을 PBS (control), scrambled DNA, ASO, 및 RNase H 가 로드된 AuNPRNAI/His(1nM)을 VERO-E6 세포와 함께 24시간 동안 배양하여 확인한 것(A)이며, 하단의 그래프는 이를 정량화한 것(B)이다. 그래프는 PBS의 양을 1로 설정하여 표시한 결과이며, 데이터는 세 번의 독립적인 실험에서 평균 ± SEM으로 표시된다. 별표는 통계적으로 유의한 값을 나타낸다 (**p < 0.01). NS, 불명확.Figure 10 shows the expression of the GFP reporter gene fused to the (+) 5'UTR of SARS-CoV-2 by incubating VERO-E6 cells with PBS (control), scrambled DNA, ASO, and AuNP RNAI/His (1 nM) loaded with RNase H for 24 h (A), and the lower graph is its quantification (B). The graph shows the results when the amount of PBS was set to 1, and the data are presented as the mean ± SEM of three independent experiments. Asterisks indicate statistically significant values (**p < 0.01). NS, unclear.

도 11은 SARS-CoV-2의 (+) 5'UTR에 융합된 GFP 리포터 유전자의 발현을 PBS (control), scrambled DNA, ASO, 및 RNase H 가 로드된 AuNPRNAI/His(1nM)을 VERO-E6 세포와 함께 24시간 동안 배양하여 FACS 분석에 의해 확인한 것이다. 세포는 40 Х 물 침지 대물렌즈(Scale bar = 20 μm)를 사용하여 관찰되었다. 유세포 분석 플롯을 통해 GFP-발현 세포의 수를 측정하였다. Figure 11. Expression of the GFP reporter gene fused to the (+) 5'UTR of SARS-CoV-2 was determined by FACS analysis in VERO-E6 cells cultured with PBS (control), scrambled DNA, ASO, and AuNP RNAI/His (1 nM) loaded with RNase H for 24 h. Cells were observed using a 40 X water-immersion objective (Scale bar = 20 μm). The number of GFP-expressing cells was quantified using a flow cytometry plot.

도 12는 PBS (control), AuNPRNAI/His, ASO 및 RNase H 가 로드된 AuNPRNAI/His로 처리한 SARS-CoV-2 감염 VERO-E6 세포의 대표적인 공초점 형광 현미경 이미지(A)이며, 녹색은 단백질, 핵은 파란색으로 염색되었다. 세포는 40 Х 물 침지 대물렌즈(Scale bar = 20 μm)를 사용하여 관찰되었다. 형광 강도의 정량화는 lmage J 소프트웨어(NIH)를 사용하여 보정된 총 세포 형광(CTCF)으로 표현하였다(B). [CTCF = 통합 밀도 - (선택한 셀의 면적 × 배경 판독값의 평균 형광)]. 데이터는 세 번의 독립적인 실험에서 평균 ± SEM으로 표시되며, 기호(*)는 통계적 유의차를 나타낸다(*p<0.05, ***p < 0.001).Figure 12 shows representative confocal fluorescence microscopy images (A) of SARS-CoV-2-infected VERO-E6 cells treated with PBS (control), AuNP RNAI/His , ASO, and AuNP RNAI/His loaded with RNase H, with proteins stained in green and nuclei in blue. Cells were observed using a 40 °C water-immersion objective (scale bar = 20 μm). Quantification of fluorescence intensity was expressed as corrected total cell fluorescence (CTCF) using lmage J software (NIH) (B). [CTCF = integrated density - (area of selected cells × mean fluorescence of background readings)]. Data are presented as mean ± SEM of three independent experiments, and symbols (*) indicate statistical significance (*p < 0.05, ***p < 0.001).

도 13은 SARS-CoV-2 감염 VERO-E6 세포를 PBS (control), AuNPRNAI/His, scrambled DNA, ASO 및 RNase H 가 로드된 AuNPRNAI/His로 처리한 후, 상대적 GFP 형광 강도를 측정한 결과이다. Figure 13 shows the results of measuring the relative GFP fluorescence intensity after treating SARS-CoV-2 infected VERO-E6 cells with PBS (control), AuNP RNAI/His , scrambled DNA, ASO, and AuNP RNAI/His loaded with RNase H.

도 14는 SARS-CoV-2 감염 마우스에 PBS (control), AuNPRNAI/His (2nM) 또는 AuNPRNAI/His-ASO-RNH(ASO 0.2 μM 및 RNH 로드된 AuNPRNAI/His 2 nM)를 각각 비강 내(A), 안와 후방(B)에 주입하여 처리한 후, RdRP mRNA의 발현을 비갑개와 폐에서 확인한 결과이다.Figure 14 shows the results of confirming the expression of RdRP mRNA in the nasal conchae and lungs after treatment of SARS-CoV-2-infected mice by intranasal injection (A) and retroorbital injection (B) with PBS (control), AuNP RNAI /His (2 nM), or AuNP RNAI /His -ASO-RNH (ASO 0.2 μM and RNH-loaded AuNP RNAI/His 2 nM), respectively.

이하, 본 명세서를 구체적으로 설명하기 위해 실시예를 들어 상세히 설명한다. 그러나, 본 명세서에 따른 실시예들은 여러 가지 다른 형태로 변형될 수 있으며, 본 명세서의 범위가 아래에서 상술하는 실시예들에 한정되는 것으로 해석되지는 않는다. 본 명세서의 실시예들은 당업계에서 평균적인 지식을 가진 자에게 본 명세서를 보다 완전하게 설명하기 위해 제공되는 것이다.Hereinafter, examples will be described in detail to specifically explain the present specification. However, the embodiments according to the present specification may be modified in various different forms, and the scope of the present specification is not construed as being limited to the embodiments described below. The embodiments of the present specification are provided to more completely explain the present specification to a person having average knowledge in the art.

<실험 방법 및 재료><Experimental methods and materials>

1. 플라스미드 및 올리고뉴클레오티드1. Plasmids and oligonucleotides

본 발명에서 사용된 플라스미드 및 올리고뉴클레오티드는 하기의 표 1 및 2에 나타내었다. The plasmids and oligonucleotides used in the present invention are shown in Tables 1 and 2 below.

플라스미드Plasmid 설명explanation pMQ131-5´ UTR-EGFPpMQ131-5´UTR-EGFP pBBR1 ori, Kmr, CEN/ARS, URA3, (+) SARS-CoV-2-5´ UTR, EGFPpBBR1 ori , Km r , CEN/ARS, URA3, (+) SARS-CoV-2-5´ UTR, EGFP pcDNA3.1-N genepcDNA3.1-N gene SV40 ori, f1 ori, Ampr, Hygr, T7, CMV, (+) SARS-CoV-2-N gene SV40 ori , f1 ori, Amp r , Hyg r , T7, CMV, (+) SARS-CoV-2-N gene pMQ131-5´ UTR-reversed-EGFPpMQ131-5´UTR-reversed-EGFP pBBR1 ori, Kmr, CEN/ARS, URA3, (-) SARS-CoV-2-5´ UTR, EGFPpBBR1 ori , Km r , CEN/ARS, URA3, (-) SARS-CoV-2-5´ UTR, EGFP pcDNA3.1-N gene-reversedpcDNA3.1-N gene-reversed SV40 ori, f1 ori, Ampr, Hygr, T7, CMV, (-) SARS-CoV-2-N gene SV40 ori , f1 ori, Amp r , Hyg r , T7, CMV, (-) SARS-CoV-2-N gene

*ori: 복제원점, Kmr: 가나마이신 저항성, Ampr: 암피실린 저항성, Hygr: 하이그로마이신 저항성, (+): positive-sense strand, (-): negative-sense strand.* ori : origin of replication, Km r : kanamycin resistance, Amp r : ampicillin resistance, Hyg r : hygromycin resistance, (+): positive-sense strand, (-): negative-sense strand.

종류type 올리고 뉴클레오티드Oligonucleotides 서열 (5’ to 3’)Sequence (5’ to 3’) 서열번호Sequence number For cloning pMQ131-5´ UTR-reversedFor cloning pMQ131-5´ UTR-reversed 5´UTR-P1 (F)5´UTR-P1 (F) ACGCGTCGACCGGTCCGGGCCATTACGCGTCGACCGGTCCGGGCCATT 11 5´UTR-P1 (R)5´UTR-P1 (R) GTAGGCGCCGGTCACAGCTTGTAGGCGCCGGTCACAGCTT 22 5´UTR-P2 (F)5´UTR-P2 (F) AGTCGTCTCTCGCGAATTAAAGGTTTATACCTTCCAGTCGTCTCTCGCGAATTAAAGGTTTAATACCTTCC 33 5´UTR-Rev-P2-Sma I-F2 (R)5´UTR-Rev-P2-Sma I-F2 (R) TCCCCCGGGCGATCCAAGCTGTGACCGGCGCCTACACCTGTAAAACAGGCAAACTTCCCCCGGGCGATCCAAGCTGTGACCGGCGCCTACACCTGTAAAACAGGCAAACT 44 For cloning pcDNA3.1-N gene-reversedFor cloning pcDNA3.1-N gene-reversed N-BamH-Rev (F)N-BamH-Rev (F) CGGATCCTGCAGGGGCCTGAGTTGAGTCAGCACCGGATCCTGCAGGGGCCTGAGTTGAGTCAGCAC 55 N-ApaI-Rev (R)N-ApaI-Rev (R) TCGAGGGGCCCTTATCTGATAATGGACCCCAAAATCGAGGGGCCCTTATCTGATAATGGACCCCCAAAA 66 For qRT-PCRFor qRT-PCR 5´UTR-qR-F (F)5´UTR-qR-F (F) TTTATACCTTCCCAGGTAACTTTATACCTTCCCAGGTAAC 77 5´UTR-qR-R (R)5´UTR-qR-R (R) TAATTATACTGCGTGAGTGCTAATTATACTGCGTGAGTGC 88 N-qR-F (F)N-qR-F (F) GGTTTACCCAATAATACTGGGTTTACCCAATAATACTG 99 N-qR-R (R)N-qR-R (R) CTTCGGTAGTAGCCAATTTGCTTCGGTAGTAGCCAATTTG 1010 hGAPDH-F (F)hGAPDH-F (F) AGGGGCCATCCACAGTCTTAGGGGCCATCCACAGTCTT 1111 hGAPDH-R (R)hGAPDH-R (R) AGCCAAAAGGGTCATCTCTAGCCAAAAGGGTCATCTCT 1212 SARS-CoV-2 RdRP gene-F (F)SARS-CoV-2 RdRP gene-F (F) GTGAAATGGTCATGTGTGGCGGGTGAAATGGTCATGTGTGGCGG 1313 SARS-CoV-2 RdRP gene-R (R)SARS-CoV-2 RdRP gene-R (R) CAAATGTTAAAAACACTATTAGCATACAAATGTTAAAAACACTATTAGCATA 1414 For Antisense oligonucleotidesFor Antisense oligonucleotides ASO-RNA I-leaderASO-RNA I-leader CAGAGCCGAGATACAAGAGATCGAAAGTTGGTTCAGAGCCGAGATACAAGAGATCGAAAGTTGGTT 1515 SO-RNA I-leaderSO-RNA I-leader CAGAGCCGAGATAACCAACTTTCGATCTCTTGTCAGAGCCGAGATAACCAACTTTCGATCTCTTGT 1616 SON-RNA I-leaderSON-RNA I-leader CAGAGCCGAGATACCGCTCTCACTCAACATGGCAGAGCCGAGATACCGCTCTCACTCAACATGG 1717 For Scrambled DNAFor Scrambled DNA N-BamH-Rev (F)N-BamH-Rev (F) CGGATCCTGCAGGGGCCTGAGTTGAGTCAGCACCGGATCCTGCAGGGGCCTGAGTTGAGTCAGCAC 1818 N-ApaI-Rev (R)N-ApaI-Rev (R) TCGAGGGGCCCTTATCTGATAATGGACCCCAAAATCGAGGGGCCCTTATCTGATAATGGACCCCCAAAA 1919 Rnh-Nde I (F)Rnh-Nde I (F) GGAATTCCATATGCTTAAACAGGTAGAAATTGGAATTCCATATGCTTAAACAGGTAGAAATT 2020 For AuNP conjugates oligonucleotidesFor AuNP conjugates oligonucleotides RNA I oligosRNA I oligos TCTCGGCTCTGCTAGCG-A10-ThiolTCTCGGCTCTGCTAGCG-A10-Thiol 2121 His-tagged aptamerHis-tagged aptamer GCTATGGGTGGTCTGGTTGGGATTGGCCCCGGGAGCTGGC-A10-ThiolGCTATGGGTGGTCTGGTTGGGATTGGCCCCGGGAGCTGGC-A10-Thiol 2222 ASO-leaderASO-leader ACAAGAGATCGAAAGTTGGTT-A10-ThiolACAAGAGATCGAAAGTTGGTT-A10-Thiol 2323 SO-leaderSO-leader AACCAACTTTCGATCTCTTGT-A10-ThiolAACCAACTTTCGATCTCTTGT-A10-Thiol 2424 SON-leaderSON leader ACCGCTCTCACTCAACATGG-A10-ThiolACCGCTCTCACTCAACATGG-A10-Thiol 2525

2. 세포 배양 2. Cell culture

VERO-E6(아프리카 녹색 원숭이 신장) 및 Calu-3(Human Lung Adenocarcinoma) 세포를 10%(v/v) FBS(Welgene) 및 1%(v/v) 페니실린-스트렙토마이신(Welgene)이 함유된 Dulbecco's modified Eagle's medium(Welgene, Korea)에서, 37°C, 5%(v/v) 이산화탄소(CO2)를 포함하는 가습된 환경에서 배양하였다.VERO-E6 (African green monkey kidney) and Calu-3 (Human Lung Adenocarcinoma) cells were cultured in Dulbecco's modified Eagle's medium (Welgene, Korea) containing 10% (v/v) FBS (Welgene) and 1% (v/v) penicillin-streptomycin (Welgene) at 37°C in a humidified atmosphere containing 5% (v/v) carbon dioxide (CO 2 ).

3. 플라스미드 형질 감염 3. Plasmid transfection

E. coli 균주 DH5α에서 NucleoBond Xtra Midi 준비 키트(Macherey-Nagel, DR, Germany)를 사용하여 플라스미드를 추출하였다. VERO-E6 세포를 형질 감염 24시간 전에 플레이트에 시딩하고, Lipofectamine 3000(Invitrogen, CA, USA)을 사용하여 플라스미드로 형질 감염시켰다. 플라스미드: Lipofectamine 3000(1μg/μl): p3000(1μg/μl)을 1:2:2.5의 비율로 혼합하고, Opti-MEM 배지에 순서대로 넣고 10분간 반응 후 세포에 투여하여 24시간 동안 배양하였다.Plasmids were extracted from E. coli strain DH5α using the NucleoBond Xtra Midi preparation kit (Macherey-Nagel, DR, Germany). VERO-E6 cells were seeded on plates 24 h before transfection and transfected with the plasmids using Lipofectamine 3000 (Invitrogen, CA, USA). Plasmid: Lipofectamine 3000 (1 μg/μl): p3000 (1 μg/μl) were mixed in a ratio of 1:2:2.5 and added sequentially to Opti-MEM medium, reacted for 10 minutes, and then added to cells and cultured for 24 h.

4. AuNP-DNA 올리고 접합체 및 AuNP RNAI/His-ASO-RNH 복합체 제작 4. Preparation of AuNP-DNA oligo conjugates and AuNP RNAI/His - ASO-RNH complexes

ASO와 RNase H의 세포 내 전달을 위한 AuNP-DNA 올리고 접합체를 하기의 방법으로 제작하였다. AuNP는 ASO에 특이적으로 결합하는 SARS-CoV-2의 포지티브 센스 가닥의 5' UTR과 RNase H의 헥사히스티딘(His) 태그를 각각 표적으로 하는 두 가지 유형의 DNA 올리고인 RNA I 및 앱타머(AptHis)와 연결된다. (도 1) 제조 과정은 아래와 같다. AuNP-DNA oligo conjugates for intracellular delivery of ASO and RNase H were fabricated by the following method. AuNPs are linked to two types of DNA oligos, RNA I and aptamer (AptHis), which target the 5' UTR of the positive sense strand of SARS-CoV-2 that specifically binds to ASO and the hexahistidine (His) tag of RNase H, respectively. (Fig. 1) The fabrication process is as follows.

먼저, Citrate-capped AuNP(직경 15nm)는 BBI Life Science(Crumlin, UK)에서 구입하였다. Thiolated RNA I, 6X His-tagged aptamer 및 ASO를 1N dithiothreitol(0.01M pH 5.2 아세트산나트륨 기준 DTT 1.545g)로 실온에서 1시간 동안 처리하여 이황화 결합을 절단하였다. 에틸 아세테이트로 추출하여 유리 티올-변형 올리고뉴클레오티드 혼합물로부터 과량의 DTT 및 원치 않는 티올 단편을 제거하였다. 절단된 올리고뉴클레오티드를 에탄올 침전을 사용하여 정제하였다. AuNP, 티올화된 DNA 올리고(RNAI 또는 ASO) 및 6X His-태그가 붙은 압타머를 인산염 완충액(100mM, pH 7.4)에서 각각 1:150:150의 비율로 혼합하여, 실온에서 1시간 동안 반응한 후, 혼합물을 5분 동안 초음파 처리하였다. 다음으로 NaCl(2M, 10mM pH 7.4 인산염 완충액 기준)을 동시에 진탕하면서 적가하여 최종 NaCl 농도가 300mM이 되도록 4시간 간격으로 3회, 실온에서 12시간 동안 배양하였다. 다음으로, AuNP-DNA 올리고 혼합물을 15 ml 튜브로 옮기고 12,000 x g에서 20분 동안 원심 분리하였다. 상청액을 제거하고 AuNP-DNA 올리고 혼합물을 1.5ml 튜브로 옮기고 20,000 x g에서 20분 동안 원심 분리하고, 3개의 증류수(T.D.W.)로 2회 세척하였다. 혼합물을 PB 완충액(500mM NaCl 기반 50mM 인산염 완충액)에 재분산시켜, 제작된 AuNPRNAI/His 의 최종 농도 50nM 가 되도록 하였다. First, citrate-capped AuNPs (diameter 15 nm) were purchased from BBI Life Science (Crumlin, UK). Thiolated RNA I, 6X His-tagged aptamer and ASO were treated with 1 N dithiothreitol (1.545 g of DTT in 0.01 M sodium acetate, pH 5.2) for 1 h at room temperature to cleave the disulfide bond. Excess DTT and unwanted thiol fragments were removed from the free thiol-modified oligonucleotide mixture by extraction with ethyl acetate. The cleaved oligonucleotides were purified using ethanol precipitation. AuNPs, thiolated DNA oligos (RNAI or ASO) and 6X His-tagged aptamer were mixed at a ratio of 1:150:150 in phosphate buffer (100 mM, pH 7.4) and reacted at room temperature for 1 h, and then the mixture was sonicated for 5 min. Next, NaCl (2 M, based on 10 mM pH 7.4 phosphate buffer) was added dropwise while shaking until the final NaCl concentration became 300 mM, three times at 4-h intervals, and incubated at room temperature for 12 h. Next, the AuNP-DNA oligo mixture was transferred to a 15 ml tube and centrifuged at 12,000 ×g for 20 min. The supernatant was removed, and the AuNP-DNA oligo mixture was transferred to a 1.5 ml tube, centrifuged at 20,000 ×g for 20 min, and washed twice with triple-distilled water (TDW). The mixture was redispersed in PB buffer (50 mM phosphate buffer based on 500 mM NaCl) until the final concentration of the fabricated AuNP RNAI/His became 50 nM.

AuNPRNAI/His는 2차 구조 형성을 방지하기 위해 80°C에서 5분 동안 사전 배양하고 실온에서 냉각시켰다. 1X PBS, 6X His-tagged RNase H (0.02 μM) (Prospec, NZ, Israel), ASO(0.1 μM), T.D.W를 혼합한 다음, AuNPRNAI/His를 혼합물에 첨가하였다. MgCl2(0.05mM)를 첨가하고 혼합물을 실온에서 10분 동안 인큐베이션하였다.AuNP RNAI/His was pre-incubated at 80°C for 5 min to prevent secondary structure formation and cooled to room temperature. 1X PBS, 6X His-tagged RNase H (0.02 μM) (Prospec, NZ, Israel), ASO (0.1 μM), and TDW were mixed, and AuNP RNAI/His was added to the mixture. MgCl 2 (0.05 mM) was added and the mixture was incubated at room temperature for 10 min.

(RNaseH 서열 : MGSSHHHHHHSSGLVPRGSHMGSMLKQVEIFTDGSCLGNP GPGGYGAILRYRGREKTFSAGYTRTTNNRMELMAAIVALEALKEHCEVILSTDSQYVRQGITQWIHNWKKRGWKTADKKPVKNVDLWQRLDAALGQHQIKWEWVKGHAGHPENERCDELARAAAMNPTLEDTGYQVEV : 서열번호 26)(RNaseH sequence: MGSSHHHHHHSSGLVPRGSHMGSMLKQVEIFTDGSCLGNP GPGGYGAILRYRGREKTFSAGYTRTTNNRMELMAAIVALEALKEHCEVILSTDSQYVRQGITQWIHNWKKRGWKTADKKPVKNVDLWQRLDAALGQHQIKWEWVKGHAGHPENERCDELARAAAMNPTLEDTGYQVEV: SEQ ID NO: 26)

상기 AuNP-AptHis와 RNase H(RNH)의 결합 친화도를 하기와 같이 측정하였다. AuNP-AptHis-RNH(5nM) 복합체의 RNase H 양을 12% SDS-폴리아크릴아미드 겔에서 분석한 결과, RNase H에 대한 AuNP-AptHis의 해리상수(KD)는 1.1 μM이었으며, 이는 RNase H가 AuNP-AptHis에 대한 높은 결합 친화도를 가지는 것을 보여주는 것이다. (도 2)The binding affinity of the above AuNP-Apt His and RNase H (RNH) was measured as follows. The amount of RNase H in the AuNP-Apt His -RNH (5 nM) complex was analyzed on a 12% SDS-polyacrylamide gel. The dissociation constant (KD) of AuNP-Apt His for RNase H was 1.1 μM, which shows that RNase H has a high binding affinity for AuNP-Apt His . (Figure 2)

또한, 네거티브 센스 가닥 SARS-CoV-2의 5' UTR을 표적으로 하는 센스 올리고뉴클레오티드(SO)와 네거티브 센스 가닥 SARS-CoV-2의 N 유전자를 표적으로 하는 센스 올리고뉴클레오티드(SON)를 합성하고 AuNP에 로드하여, 세포 독성 및 결합 효율성 등의 결과를 비교하였다. In addition, a sense oligonucleotide (SO) targeting the 5' UTR of the negative sense strand SARS-CoV-2 and a sense oligonucleotide (SON) targeting the N gene of the negative sense strand SARS-CoV-2 were synthesized and loaded onto AuNPs, and the results such as cytotoxicity and binding efficiency were compared.

5. 세포 독성 실험 5. Cytotoxicity test

VERO-E6 세포(5×103/웰)를 96-웰 플레이트에 시딩하고 24시간 동안 배양하였다. 세포를 추가로 24시간 동안 배지에서 AuNPRNAI/His에 로드된 ASO, SO, SON 및/또는 RNase H 를 2nM씩 추가하여 다시 인큐베이션하였다. 세포 생존율은 CytoTox 96® Non-Radioactive Cytotoxicity Assay(Promega, WI, USA)를 사용하여 측정되었다. 색상은 세포만(검은색), PBS(빨간색), AuNP-DNA 올리고(노란색) 및 AuNP-DNA 올리고-RNH(녹색)으로 나타내고, 상대적인 세포 생존율은 세포만의 생존 세포와 비교하여 생존 세포의 백분율로 표시하였다. 데이터는 세 번의 독립적인 실험에서 평균 ± SEM으로 표시하였다. (NS : 불명확함) VERO-E6 cells (5 × 10 3 /well) were seeded in 96-well plates and cultured for 24 h. The cells were then incubated again for an additional 24 h in the medium with 2 nM of ASO, SO, SON, and/or RNase H loaded onto AuNP RNAI/His . Cell viability was measured using the CytoTox 96 ® Non-Radioactive Cytotoxicity Assay (Promega, WI, USA). Colors are represented as cells only (black), PBS (red), AuNP-DNA oligos (yellow), and AuNP-DNA oligo-RNH (green), and relative cell viability is expressed as the percentage of viable cells compared to viable cells in cells only. Data are presented as the mean ± SEM from three independent experiments. (NS: unclear)

그 결과, 도 3에서 보듯이, 지시된 농도를 갖는 모든 물질은 VERO-E6 세포에 대해 세포 독성을 나타내지 않았다. As a result, as shown in Fig. 3, all substances with the indicated concentrations did not exhibit cytotoxicity against VERO-E6 cells.

6. AuNP6. AuNP RNAI/His RNAI/His 에 의한 ASO와 RNase H의 세포 내 전달 Intracellular delivery of ASO and RNase H by

동물 세포에서, AuNPRNAI/His에 의한 ASO와 RNase H의 전달 효율성을 조사하기 위해 공초점 현미경 분석과 형광 신호 강도 분석(Corrected Total Cell Fluorescence, CTCF)을 수행하였다. 가시화를 위해 ASO의 3' 말단을 시아닌 3(Cy3)으로 표지하고 RNase H의 1차 아민을 Alexa-488로 표지하여 관찰한 결과, 도 4와 같이 AuNPRNAI/His-ASO, AuNPRNAI/His-RNH 및 AuNPRNAI/His-ASO-RNH를 각각 처리한 VERO-E6 세포에서 높은 형광 신호가 검출되었으며, 이와 대조적으로, PBS, AuNPRNAI/His, ASO 또는 RNH만으로 처리된 VERO-E6 세포에서는 유의미한 형광이 검출되지 않았다. 이러한 결과는 AuNPRNAI/His에 ASO와 RNase H가 로딩되어 세포 내로 효율적으로 전달되었음을 보여주는 것이다. To investigate the delivery efficiency of ASO and RNase H by AuNP RNAI/His in animal cells, confocal microscopy and fluorescence signal intensity analysis (Corrected Total Cell Fluorescence, CTCF) were performed. For visualization, the 3' end of ASO was labeled with cyanine 3 (Cy3) and the primary amine of RNase H was labeled with Alexa-488. As shown in Fig. 4, high fluorescence signals were detected in VERO-E6 cells treated with AuNP RNAI/His -ASO, AuNP RNAI/His -RNH, and AuNP RNAI/His -ASO-RNH, respectively. In contrast, no significant fluorescence was detected in VERO-E6 cells treated with PBS, AuNP RNAI/ His , ASO, or RNH alone. These results demonstrate that ASO and RNase H were loaded onto AuNP RNAI/His and efficiently delivered into cells.

7. AuNP RNAI/His-ASO-RNH에 의한 바이러스 mRNA 발현 억제 7. Inhibition of viral mRNA expression by AuNP RNAI/His -ASO-RNH

AuNPRNAI/His-ASO-RNH 복합체가 바이러스 mRNA 발현 수준에 영향을 미칠 수 있는지 여부를 조사하기 위해 VERO-E6 세포를 AuNPRNAI/His-ASO-RNH 복합체로 24시간 동안 형질 감염시켰고, SARS-CoV-2의 5' UTR mRNA 발현 수준을 qRT-PCR에 의해 측정하였다(도 5). To investigate whether the AuNP RNAI/His -ASO-RNH complex could affect the level of viral mRNA expression, VERO-E6 cells were transfected with the AuNP RNAI/His -ASO-RNH complex for 24 h, and the 5' UTR mRNA expression level of SARS-CoV-2 was measured by qRT-PCR (Fig. 5).

그 결과, 도 6에서 확인되는 바와 같이, 포지티브 센스 가닥 SARS-CoV-2(플라스미드 pMQ131-5' UTR-EGFP로 사전 형질 감염된 VERO-E6 세포)의 5'UTR mRNA를 발현하는 VERO-E6 세포를 AuNPRNAI/His-ASO-RNH 복합체로 처리한 결과(레인 8) 완충액 또는 다른 AuNPRNAI/His 복합체(AuNPRNAI/His, AuNPRNAI/His-ASO, AuNPRNAI/His-RNH 및 AuNPRNAI/His-scrambled DNA-RNH)로 처리된 대조군 세포에 비해 약 30-40% mRNA 발현 수준이 감소됨을 확인하였다. 또한, RNAI와의 혼성화 없이 AuNP에 ASO가 직접 결합된 AuNPHis-ASO의 경우에는 RNase H가 5'UTR mRNA의 발현 수준에 영향을 미치지 않았으며(레인 4 또는 6 비교), AuNPRNAI/His-ASO 와 비교하여 발현 억제 효율이 낮음을 확인하였다. As a result, as confirmed in Fig. 6, when VERO-E6 cells expressing 5'UTR mRNA of positive-sense strand SARS-CoV-2 (VERO-E6 cells pre-transfected with plasmid pMQ131-5' UTR-EGFP) were treated with AuNP RNAI/His -ASO-RNH complex (lane 8), it was confirmed that the mRNA expression level was reduced by about 30-40% compared to the control cells treated with buffer or other AuNP RNAI/His complexes (AuNP RNAI /His , AuNP RNAI/His -ASO, AuNP RNAI/His -RNH, and AuNP RNAI/His -scrambled DNA-RNH). In addition, in the case of AuNP His -ASO, in which ASO was directly bound to AuNP without hybridization with RNAI, RNase H did not affect the expression level of 5'UTR mRNA (compare lane 4 or 6), and it was confirmed that the expression inhibition efficiency was lower compared to AuNP RNAI/His -ASO.

또한, 도 7과 같이 AuNPRNAI/His-ASO-RNH 복합체의 형질 감염 후, 시간에 따른 발현 억제 효율을 4시간 마다 관찰하여 qRT-PCR로 분석하였다. 그 결과, 배양 시간이 경과됨에 따라 5' UTR mRNA 의 발현 수준이 점차 감소되었으며, qRT-PCR 산물을 1.5% 아가로즈 겔에서 분리하고 에티디움 브로마이드를 사용하여 시각화한 결과에서도 이와 같음을 확인하였다. In addition, as shown in Fig. 7, after transfection of the AuNP RNAI/His -ASO-RNH complex, the expression inhibition efficiency according to time was observed every 4 hours and analyzed by qRT-PCR. As a result, the expression level of 5' UTR mRNA gradually decreased as the incubation time elapsed, and this was also confirmed by separating the qRT-PCR product on a 1.5% agarose gel and visualizing it using ethidium bromide.

아울러, 네거티브 센스 가닥 SARS-CoV-2(플라스미드 pMQ131-5'UTR-reversed-EGFP 또는 pcDNA3.1-N 유전자 반전)을 상기와 동일한 방식으로 테스트했지만, AuNPRNAI/His-SO-RNH 또는 AuNPRNAI/His-SON-RNH 복합체는 표적 RNA에 대한 녹다운에 효과적이지 않았다(도 8, 도 9).In addition, negative-sense strand SARS-CoV-2 (plasmid pMQ131-5'UTR-reversed-EGFP or pcDNA3.1-N gene reversed) was tested in the same manner as above, but AuNP RNAI/His -SO-RNH or AuNP RNAI/His -SON-RNH complexes were not effective in knockdown of the target RNA (Fig. 8, Fig. 9).

종합하면, 이러한 결과는 AuNPRNAI/His-ASO-RNH 복합체가 VERO-E6 세포에서 SARS-CoV-2의 표적 mRNA 수준을 효율적으로 감소시킬 수 있음을 보여주는 것이다. Taken together, these results demonstrate that the AuNP RNAI/His -ASO-RNH complex can efficiently reduce the target mRNA levels of SARS-CoV-2 in VERO-E6 cells.

8. AuNP RNAI/His-ASO-RNH 복합체의 타겟 단백질 발현 억제 효과 8. Target protein expression inhibition effect of AuNP RNAI/His -ASO-RNH complex

본 발명의 AuNPRNAI/His-ASO-RNH 복합체가 실제 타겟 단백질 발현을 억제하는지 여부를 확인하기 위하여, SARS-CoV-2의 (+) 5'UTR에 융합된 GFP mRNA를 발현하는 VERO-E6 세포의 처리가 GFP의 발현 수준을 감소시키는지 여부를 추가로 조사하였다. 그 결과, 도 10에 나타낸 바와 같이, 약 35%의 GFP 발현량 감소가 관찰되었다. 또한, mRNA 발현 수준과 일치하게, AuNPRNAI/His, AuNPRNAI/His-RNH 또는 AuNPRNAI/His-스크램블된DNA-RNH로 세포를 처리한 경우에는 유의미한 효과가 관찰되지 않았다. 또한, GFP-발현 세포의 FACS 분석으로부터 유사한 결과를 확인하였다(도 11). 이러한 결과는 AuNPRNAI/His-ASO-RNH가 SARS-CoV-2의 포지티브 센스 가닥에 융합된 GFP 리포터 유전자의 발현을 녹다운할 수 있음을 보여주는 것이다.To determine whether the AuNP RNAI/His -ASO-RNH complex of the present invention actually inhibits target protein expression, we further investigated whether treatment of VERO-E6 cells expressing GFP mRNA fused to the (+) 5'UTR of SARS-CoV-2 reduces the expression level of GFP. As a result, as shown in Fig. 10, a decrease in GFP expression of about 35% was observed. In addition, consistent with the mRNA expression level, no significant effect was observed when cells were treated with AuNP RNAI/His , AuNP RNAI/His -RNH, or AuNP RNAI/ His -scrambledDNA-RNH. In addition, similar results were confirmed from FACS analysis of GFP-expressing cells (Fig. 11). These results demonstrate that AuNP RNAI/His -ASO-RNH can knockdown the expression of the GFP reporter gene fused to the positive sense strand of SARS-CoV-2.

9. AuNP RNAI/His-ASO-RNH 복합체의 SARS-CoV-2 감염 동물세포에 대한 효과 9. Effect of AuNP RNAI/His -ASO-RNH complex on SARS-CoV-2 infected animal cells

SARS-CoV-2에 감염된 포유류 세포에서 AuNPRNAI/His-ASO-RNH의 효과를 추가로 테스트하기 위해, SARS-CoV-2에 감염된 VERO-E6 세포에 AuNPRNAI/His 또는 AuNPRNAI/His-ASO-RNH를 각각 0.1 및 1nM의 용량으로 주입하여 세포를 형질 감염시키고, SARS-CoV-2의 뉴클레오캡시드 단백질을 면역세포화학(ICC) 분석을 사용하여 검출하였다. To further test the effect of AuNP RNAI/His -ASO-RNH in mammalian cells infected with SARS-CoV-2, VERO-E6 cells infected with SARS-CoV-2 were transfected with AuNP RNAI/His or AuNP RNAI/His -ASO-RNH at doses of 0.1 and 1 nM, respectively, and the nucleocapsid protein of SARS-CoV-2 was detected using immunocytochemistry (ICC) analysis.

그 결과, 도 12에서 보듯이, 뉴클레오캡시드 단백질 수준은 AuNPRNAI/His-ASO-RNH(1nM) 처리된 VERO-E6(도 17)에서 30-40% 감소하였다. 상기 세포를 AuNPRNAI/His(0.1 및 1nM) 및 더 낮은 농도의 AuNPRNAI/His-ASO-RNH(0.1nM)로 처리한 경우에는 N 단백질 수준에 유의한 영향을 미치지 않았다. 이러한 결과는 1nM의 AuNPRNAI/His-ASO-RNH가 포유류 세포에서 SARS-CoV-2 N 단백질의 발현을 효과적으로 녹다운할 수 있음을 보여주는 것이다. As a result, as shown in Fig. 12, the nucleocapsid protein level was reduced by 30-40% in VERO-E6 (Fig. 17) treated with AuNP RNAI/His ( 0.1 and 1 nM) and a lower concentration of AuNP RNAI/His -ASO-RNH (0.1 nM), no significant effect on the N protein level was observed. These results demonstrate that 1 nM of AuNP RNAI/His -ASO-RNH can effectively knockdown the expression of SARS-CoV-2 N protein in mammalian cells.

또한 GFP(SARS-CoV-2-mNeon)를 발현할 수 있는 재조합 SARS-CoV-2로 감염된 VERO-E6 세포를 사용하여 바이러스 복제를 억제하는 AuNPRNAI/His-ASO-RNH의 능력을 확인하였다. 그 결과, 도 13에 도시된 바와 같이, 바이러스 단독, AuNPRNAI/His, AuNPRNAI/His-RNH, 또는 AuNPRNAI/His-Scrambled DNA-RNH로 처리된 세포와 비교하여 AuNPRNAI/His-ASO-RNH로 처리된 세포에서 30-40%의 GFP 강도의 감소가 관찰되었다. 이러한 결과는 AuNPRNAI/His-ASO-RNH가 바이러스 mRNA의 절단을 통해 GFP 번역을 억제함을 보여주는 것이다. Furthermore, the ability of AuNP RNAI/His -ASO-RNH to inhibit viral replication was confirmed using VERO-E6 cells infected with recombinant SARS-CoV-2 capable of expressing GFP (SARS-CoV-2-mNeon). As a result, as shown in Fig. 13, a 30-40% decrease in GFP intensity was observed in cells treated with AuNP RNAI / His -ASO-RNH compared to cells treated with virus alone, AuNP RNAI /His , AuNP RNAI/His -RNH, or AuNP RNAI/His -Scrambled DNA-RNH. These results demonstrate that AuNP RNAI/His -ASO-RNH inhibits GFP translation through the cleavage of viral mRNA.

10. AuNP RNAI/His-ASO-RNH 복합체의 SARS-CoV-2 감염 동물세포에 대한 효과 10. Effect of AuNP RNAI/His -ASO-RNH complex on SARS-CoV-2 infected animal cells

AuNPRNAI/His-ASO-RNH가 바이러스 유발 호흡기 질환 치료제로 사용될 수 있는지 여부를 평가하기 위해 SARS-CoV-2 감염의 확립된 동물 모델에서 AuNPRNAI/His-ASO-RNH의 생체 내 효능을 테스트하였다. K18-hACE2 마우스를 1 x 105 PFU SARS-CoV-2 오미크론 변이체로 감염시키고 PBS, AuNPRNAI/His 또는 AuNPRNAI/His-ASO-RNH를 비강내(IN) 또는 안와후면(RO)으로, 24시간 간격으로 3회 주사하였다. 바이러스에 감염된 마우스를 5일째에 안락사 시키고 폐 및 비갑개 조직을 수집하였다. 이들 기관으로부터 qRT-PCR을 사용하여 바이러스 RNA-의존성 RNA 중합효소(RdRP) mRNA를 측정하였다. To evaluate whether AuNP RNAI/His -ASO-RNH could be used as a therapeutic agent for virus-induced respiratory diseases, the in vivo efficacy of AuNP RNAI/His -ASO-RNH was tested in an established animal model of SARS-CoV-2 infection. K18-hACE2 mice were infected with 1 × 10 5 PFU of the SARS-CoV-2 omicron variant and injected intranasally (IN) or retroorbitally (RO) with PBS, AuNP RNAI/His , or AuNP RNAI/His -ASO-RNH for three times at 24-h intervals. Mice infected with the viruses were euthanized on day 5, and lung and nasal conchae tissues were collected. Viral RNA-dependent RNA polymerase (RdRP) mRNA was measured from these organs using qRT-PCR.

그 결과, 도 14A에 도시된 바와 같이, RdRP mRNA의 발현 수준은 PBS 또는 AuNP-DNA 올리고만으로 처리된 것과 비교하여 AuNPRNAI/His-ASO-RNH를 비강내로 전달했을 때 폐에서 ~30%까지 감소하였다. 마찬가지로, 도 14B와 같이, AuNPRNAI/His-ASO-RNH를 안와후방으로 전달했을 때 PBS 또는 AuNP-DNA 올리고만으로 처리한 경우와 비교하여 비갑개와 폐에서 바이러스 mRNA가 ~20% 감소하였다. 이러한 데이터는 AuNPRNAI/His-ASO-RNH가 SARS-CoV-2바이러스를 치료하기 위해 투여되는 경우, 감염된 동물의 기도로 효과적으로 전달되어 바이러스 역가를 감소시킬 수 있다는 것을 보여주는 결과이다. As a result, as shown in Fig. 14A, the expression level of RdRP mRNA was reduced by ~30% in the lung when AuNP RNAI/His -ASO-RNH was delivered intranasally compared to treatment with PBS or only AuNP-DNA oligos. Similarly, as shown in Fig. 14B, when AuNP RNAI/His -ASO-RNH was delivered retroorbitally, viral mRNA was reduced by ~20% in the nasal conchae and lung compared to treatment with PBS or only AuNP-DNA oligos. These data demonstrate that AuNP RNAI/His -ASO-RNH can be effectively delivered to the airways of infected animals and reduce the viral titer when administered to treat the SARS-CoV-2 virus.

이제까지 본 발명에 대하여 그 바람직한 실시예들을 중심으로 살펴보았다. 본 발명이 속하는 기술 분야에서 통상의 지식을 가진 자는 본 발명이 본 발명의 본질적인 특성에서 벗어나지 않는 범위에서 변형된 형태로 구현될 수 있음을 이해할 수 있을 것이다. 그러므로 개시된 실시예들은 한정적인 관점이 아니라 설명적인 관점에서 고려되어야 한다. 본 발명의 범위는 전술한 설명이 아니라 청구범위에 나타나 있으며, 그와 동등한 범위 내에 있는 모든 차이점은 본 발명에 포함된 것으로 해석되어야 할 것이다.The present invention has been described with reference to preferred embodiments thereof. Those skilled in the art will appreciate that the present invention may be implemented in modified forms without departing from the essential characteristics of the present invention. Therefore, the disclosed embodiments should be considered in an illustrative rather than a restrictive sense. The scope of the present invention is set forth in the claims, not in the foregoing description, and all differences within the scope equivalent thereto should be construed as being included in the present invention.

일 양태로서, 본 발명은 금 나노 입자; 및 상기 금 나노 입자 표면에 결합된 바이러스 유래 서열에 상보적인 안티센스 올리고뉴클레오타이드(ASO) 및 상기 바이러스의 mRNA의 절단을 유발시키는 리보핵산가수분해효소 H(RNase H)를 포함하는 나노 입자 전달체에 대한 것이다. In one aspect, the present invention relates to a nanoparticle delivery vehicle comprising a gold nanoparticle; and an antisense oligonucleotide (ASO) complementary to a virus-derived sequence bound to the surface of the gold nanoparticle and ribonuclease H (RNase H) that induces cleavage of mRNA of the virus.

일 실시예로서, 상기 바이러스 유래 서열은 5’비번역 부위(UTR)에 포함된 뉴클레오티드 서열을 포함하고, 상기 바이러스 유래의 서열은 리더 서열(Leader sequence)을 포함한다. As an example, the viral sequence comprises a nucleotide sequence included in the 5’ untranslated region (UTR), and the viral sequence comprises a leader sequence.

일 실시예로서, 상기 바이러스는 코로나바이러스, 인플루엔자바이러스, 에볼라바이러스, 헤르페스바이러스, 레트로바이러스, 간염바이러스, 로타바이러스, 플라비바이러스, 파필로마바이러스, 홍역바이러스, 폴리오바이러스, 콕사키바이러스 및 아데노바이러스를 포함하는 군으로부터 선택될 수 있다. As an example, the virus may be selected from the group comprising coronavirus, influenza virus, ebola virus, herpes virus, retrovirus, hepatitis virus, rotavirus, flavivirus, papillomavirus, measles virus, poliovirus, coxsackievirus and adenovirus.

일 실시예로서, 상기 안티센스 올리고뉴클레오타이드(ASO)는 작용기성을 포함하는 잔기를 통해 상기 금 나노 표면에 결합되며, 상기 작용기성을 포함하는 잔기는 티올기 또는 아민기를 포함한다. As an example, the antisense oligonucleotide (ASO) is bound to the gold nano surface via a moiety comprising a functional group, wherein the moiety comprising a functional group comprises a thiol group or an amine group.

일 실시예로서, 상기 RNaseH는 앱타머를 통해 상기 금 나노 표면에 결합될 수 있으며, 또한, 상기 앱타머는 히스티딘 tag를 포함할 수 있다. As an example, the RNaseH can be bound to the gold nano surface via an aptamer, and further, the aptamer can include a histidine tag.

다른 양태로서, 본 발명은 상기 금 나노 입자 전달체를 포함하는 항 바이러스용 조성물에 대한 것이다. In another aspect, the present invention relates to an antiviral composition comprising the gold nanoparticle carrier.

또 다른 양태로서, 본 발명은 바이러스 유래 서열에 상보적인 안티센스 올리고뉴클레오타이드(ASO) 및 상기 바이러스의 mRNA의 절단을 유발시키는 리보핵산가수분해효소 H(RNase H)를 표면에 결합한 금 나노 입자를 세포 내로 전달하고, 상기 바이러스 유래 서열에 상보적인 안티센스 올리고뉴클레오타이드(ASO)가 바이러스 유래 mRNA 서열에 결합하여 RNase H를 통해 상기 mRNA를 분해하여 바이러스의 활성을 억제하기 위한 금 나노 입자 운반체의 용에 대한 것이다. In another aspect, the present invention relates to a gold nanoparticle carrier for delivering into cells gold nanoparticles having an antisense oligonucleotide (ASO) complementary to a virus-derived sequence and ribonuclease H (RNase H) bound to the surface, which induces cleavage of mRNA of the virus, and wherein the antisense oligonucleotide (ASO) complementary to the virus-derived sequence binds to the virus-derived mRNA sequence and degrades the mRNA through RNase H, thereby inhibiting the activity of the virus.

Claims (10)

금 나노 입자; 및 gold nanoparticles; and 상기 금 나노 입자 표면에 결합된 바이러스 유래 서열에 상보적인 안티센스 올리고뉴클레오타이드(ASO) 및 상기 바이러스의 mRNA의 절단을 유발시키는 리보핵산가수분해효소 H(RNase H)를 포함하는 나노 입자 전달체. A nanoparticle delivery vehicle comprising an antisense oligonucleotide (ASO) complementary to a virus-derived sequence bound to the surface of the gold nanoparticle and ribonuclease H (RNase H) that induces cleavage of mRNA of the virus. 제1항에 있어서, In the first paragraph, 상기 바이러스 유래 서열은 5’비번역 부위(UTR)에 포함된 뉴클레오티드 서열인, 나노 입자 전달체. A nanoparticle carrier wherein the above viral-derived sequence is a nucleotide sequence contained in the 5’untranslated region (UTR). 제1항에 있어서, In the first paragraph, 상기 바이러스 유래의 서열은 리더 서열(Leader sequence)을 포함하는 것인 나노 입자 전달체. A nanoparticle carrier wherein the above virus-derived sequence comprises a leader sequence. 제1항에 있어서, In the first paragraph, 상기 바이러스는 코로나바이러스, 인플루엔자바이러스, 에볼라바이러스, 헤르페스바이러스, 레트로바이러스, 간염바이러스, 로타바이러스, 플라비바이러스, 파필로마바이러스, 홍역바이러스, 폴리오바이러스, 콕사키바이러스 및 아데노바이러스를 포함하는 군으로부터 선택된 것인, 나노 입자 전달체. A nanoparticle carrier, wherein the virus is selected from the group consisting of coronavirus, influenza virus, ebola virus, herpes virus, retrovirus, hepatitis virus, rotavirus, flavivirus, papillomavirus, measles virus, poliovirus, coxsackievirus and adenovirus. 제1항에 있어서, In the first paragraph, 상기 안티센스 올리고뉴클레오타이드(ASO)는 작용기성을 포함하는 잔기를 통해 상기 금 나노 표면에 결합되는 것인 나노 입자 전달체. A nanoparticle carrier wherein the antisense oligonucleotide (ASO) is bound to the gold nano surface via a moiety containing a functional group. 제5항에 있어서, In paragraph 5, 상기 작용기성을 포함하는 잔기는 티올기 또는 아민기를 포함하는 것인 나노 입자 전달체. A nanoparticle carrier wherein the residue containing the above functional group contains a thiol group or an amine group. 제1항에 있어서, In the first paragraph, 상기 RNaseH는 앱타머를 통해 상기 금 나노 표면에 결합되는 것인 금 나노 입자 전달체. A gold nanoparticle carrier wherein the above RNaseH is bound to the surface of the gold nano via an aptamer. 제7항에 있어서, In Article 7, 상기 앱타머는 히스티딘 tag를 포함하는 것인 금 나노 입자 전달체. The above aptamer is a gold nanoparticle carrier containing a histidine tag. 제1항의 나노 입자 전달체를 포함하는 항 바이러스용 조성물.An antiviral composition comprising the nano particle carrier of claim 1. 바이러스 유래 서열에 상보적인 안티센스 올리고뉴클레오타이드(ASO) 및 상기 바이러스의 mRNA의 절단을 유발시키는 리보핵산가수분해효소 H(RNase H)를 표면에 결합한 금 나노 입자를 세포 내로 전달하고, 상기 바이러스 유래 서열에 상보적인 안티센스 올리고뉴클레오타이드(ASO)가 바이러스 유래 mRNA 서열에 결합하여 RNase H를 통해 상기 mRNA를 분해하여 바이러스의 활성을 억제하기 위한 금 나노 입자 운반체의 용도. Use of a gold nanoparticle carrier to deliver into cells gold nanoparticles having an antisense oligonucleotide (ASO) complementary to a virus-derived sequence and ribonuclease H (RNase H) bound to the surface that induces cleavage of mRNA of the virus, and to inhibit the activity of the virus by causing the antisense oligonucleotide (ASO) complementary to the virus-derived sequence to bind to the virus-derived mRNA sequence and degrade the mRNA through RNase H.
PCT/KR2024/018565 2023-11-21 2024-11-21 Antiviral composition comprising nanoparticle carrier based on gold nanoparticle-nucleic acid conjugate Pending WO2025110774A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2023-0162636 2023-11-21
KR20230162636 2023-11-21
KR10-2024-0167434 2024-11-21
KR1020240167434A KR20250076427A (en) 2023-11-21 2024-11-21 Antiviral composition comprising nanoparticle carrier based on gold nanoparticle-nucleic acid conjugate

Publications (1)

Publication Number Publication Date
WO2025110774A1 true WO2025110774A1 (en) 2025-05-30

Family

ID=95827189

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2024/018565 Pending WO2025110774A1 (en) 2023-11-21 2024-11-21 Antiviral composition comprising nanoparticle carrier based on gold nanoparticle-nucleic acid conjugate

Country Status (1)

Country Link
WO (1) WO2025110774A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20170140377A (en) * 2015-04-29 2017-12-20 로도스 바이오타겟 게엠베하 Targeted nanocarriers for targeted drug delivery of gene therapy agents
CN114272391A (en) * 2021-12-28 2022-04-05 深圳湾实验室坪山生物医药研发转化中心 Nucleic acid-guided target RNA degradation nano-composite and preparation method thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20170140377A (en) * 2015-04-29 2017-12-20 로도스 바이오타겟 게엠베하 Targeted nanocarriers for targeted drug delivery of gene therapy agents
CN114272391A (en) * 2021-12-28 2022-04-05 深圳湾实验室坪山生物医药研发转化中心 Nucleic acid-guided target RNA degradation nano-composite and preparation method thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BARREY, E.: "Think Different with RNA Therapy: Can Antisense Oligonucleotides Be Used to Inhibit Replication and Transcription of SARS-Cov-2?. ", HAL OPEN SCIENCE, 23 April 2020 (2020-04-23), XP093317273, Retrieved from the Internet <URL:https://hal.inrae.fr/hal-04106554v1/document> *
DEANASA RAEHAN SATYA, AFLADHANTI PUTRI MAHIRAH AFLADHANTI, SYAFIRA FARA: "The Potential Of Antisense Oligonucleotides (ASO) Through Inhalation Based On Gold Nanoparticle (AuNP) Delivery System In Inhibiting SARS-CoV-2 Replication And Transcription : English", SCRIPTA SCORE SCIENTIFIC MEDICAL JOURNAL, vol. 4, no. 1, pages 82 - 91, XP093317256, ISSN: 2088-8686, DOI: 10.32734/scripta.v4i1.8384 *
MOITRA PARIKSHIT, ALAFEEF MAHA, DIGHE KETAN, FRIEMAN MATTHEW B., PAN DIPANJAN: "Selective Naked-Eye Detection of SARS-CoV-2 Mediated by N Gene Targeted Antisense Oligonucleotide Capped Plasmonic Nanoparticles", ACS NANO, AMERICAN CHEMICAL SOCIETY, US, vol. 14, no. 6, 23 June 2020 (2020-06-23), US , pages 7617 - 7627, XP055796333, ISSN: 1936-0851, DOI: 10.1021/acsnano.0c03822 *

Similar Documents

Publication Publication Date Title
US11359198B2 (en) Extended dicer substrate agents and methods for the specific inhibition of gene expression
JP7526195B2 (en) Compositions, methods, and kits for delivery of polyribonucleotides
JP5986928B2 (en) Dicer substrate agents and methods for specific inhibition of gene expression
CN112567036A (en) Modified guide RNA for gene editing
JP2009524430A (en) RNA interference agents for therapeutic use
JP2021532812A (en) Alternative splicing regulation and treatment of gene expression
JPH08500481A (en) Methods and agents for inhibiting viral replication
CN109312338A (en) CRISPR/CAS related method and composition for treating herpes simplex virus
CA3151965A1 (en) Extended dicer substrate agents and methods for the specific inhibition of gene expression
JP2024515348A (en) Therapeutic interfering particles against coronaviruses
US10155946B2 (en) Particle-nucleic acid conjugates and therapeutic uses related thereto
JP2021529525A (en) Oligonucleotide for TAU expression regulation
CN102238967A (en) Telomerase inhibitors and methods of use thereof
CN113528516A (en) Nucleic acid, pharmaceutical composition and siRNA conjugate containing the nucleic acid, and preparation method and use
JP2021522178A (en) MRNA-driven expression of RNA editor for pathological treatment
KR20150095763A (en) Methods and compositions for the specific inhibition of ckap5 by double-stranded rna
US20220290159A1 (en) miRNAS FOR REDUCING VENTRICLE ENLARGEMENT
WO2025110774A1 (en) Antiviral composition comprising nanoparticle carrier based on gold nanoparticle-nucleic acid conjugate
KR20250076427A (en) Antiviral composition comprising nanoparticle carrier based on gold nanoparticle-nucleic acid conjugate
US10036020B2 (en) Compositions and methods for inhibiting JC virus (JCV)
ES2753355T3 (en) Small interference RNA (siRNA) for therapy of autosomal dominant osteopetrosis type 2 (ADO2) caused by mutation of the CLCN7 gene (ADO2 dependent on CLCN7)
KR101917287B1 (en) self-assembled ribonucleoprotein nanoparticles
EP3954773A1 (en) Compositions and methods for the treatment of diseases by enhancing arginase 2 in macrophages
WO2020154594A2 (en) Compositions and methods for treatment of human papillomavirus
CN119662640A (en) A small nucleic acid inhibiting the replication of the new coronavirus, its conjugate and application

Legal Events

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

Ref document number: 24894665

Country of ref document: EP

Kind code of ref document: A1