WO2023167487A1 - Recombinant expression vector for prevention of sars-cov-2 infection and application thereof - Google Patents

Recombinant expression vector for prevention of sars-cov-2 infection and application thereof Download PDF

Info

Publication number
WO2023167487A1
WO2023167487A1 PCT/KR2023/002808 KR2023002808W WO2023167487A1 WO 2023167487 A1 WO2023167487 A1 WO 2023167487A1 KR 2023002808 W KR2023002808 W KR 2023002808W WO 2023167487 A1 WO2023167487 A1 WO 2023167487A1
Authority
WO
WIPO (PCT)
Prior art keywords
sars
protein
coronavirus
chimad
amino acid
Prior art date
Application number
PCT/KR2023/002808
Other languages
French (fr)
Korean (ko)
Inventor
김수옥
홍선표
김석준
장선옥
Original Assignee
(주)진매트릭스
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
Priority claimed from KR1020230024283A external-priority patent/KR20230131439A/en
Application filed by (주)진매트릭스 filed Critical (주)진매트릭스
Publication of WO2023167487A1 publication Critical patent/WO2023167487A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/215Coronaviridae, e.g. avian infectious bronchitis virus
    • 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
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors

Definitions

  • the present invention relates to a recombinant expression vector for the prevention of SARS-coronavirus-2 infection and its application, and specifically to the application of the vector to the prevention of SARS-coronavirus-2, etc., and more particularly, to the beta mutant spike protein (S A COVID-19 preventive vaccine containing a recombinant chimeric adenoviral vector encoded by linking a nucleocapsid (N protein) to a nucleocapsid (N protein) and/or a spike protein, as well as SARS-coronavirus-2 strains (beta, delta, Omicron, etc.) suggests the possibility of preventive and therapeutic vaccines against infections.
  • S A COVID-19 preventive vaccine containing a recombinant chimeric adenoviral vector encoded by linking a nucleocapsid (N protein) to a nucleocapsid (N protein) and/or a spike protein, as well as SARS-coronavirus-2 strains
  • Coronaviruses are positive-sense single-stranded RNA viruses that have an envelope on the surface of particles with a diameter of 100-200 mm. Seven types of coronaviruses are known to cause respiratory and digestive infections in humans, and two of the seven types are alpha- genus of coronaviruses, and the remaining five species were identified as genus beta-coronaviruses.
  • Severe acute respiratory syndrome coronavirus 2 (hereinafter referred to as 'SARS-CoV-2'), which causes COVID-19 (novel coronavirus, 2019-nCov), is a beta-coronavirus It is a genus, and after it was first reported in Wuhan, China in December 2019 (Wuhan-Hu-1), confirmed cases are appearing one after another worldwide. (pandemic) is intensifying.
  • the viral envelope of SARS-CoV-2 is composed of spike glycoprotein (S protein) and hemagglutinin-esterase dimer (HE).
  • S protein spike glycoprotein
  • HE hemagglutinin-esterase dimer
  • the S 1 region of the S protein mainly binds to the receptor (Angiotensin-converting enzyme 2, ACE2) through the RBD (Receptor binding domain), and the S 2 region is a direct cell-virus fusion accompanied by a dramatic change in tertiary structure (infection initiation). Therefore, vaccine candidates targeting the S protein as a major antigen have been developed and commercialized.
  • RNA viruses use RNA polymerase to correct errors that occur during genome replication. Since there is no related protein, mutations are more frequent compared to DNA viruses. In the case of the SARS-CoV-2 mutant virus, mutations frequently occurred in the N-terminal domain (NTD) and RBD domain of the S protein, and as a result, a licensed vaccine developed targeting the S protein of the initial state. The antibody-binding ability to the S protein of the mutant virus in which these structural changes occurred was reduced, and the protective efficacy against the mutant virus was lowered.
  • Adenovirus vector vaccine is a vaccine that induces an immune response by expressing an antigen protein in the body by inserting and delivering an antigen gene for a pathogen into an adenovirus template.
  • Human adenoirus serotype 5 (HAdV-5) vector is the most widely used. Although used, there is a problem that vaccine efficacy is reduced due to high serum retention rates in the human population.
  • the chimeric adenovirus (ChimAd) vector is a recombinant key substituted with the projection gene of chimpanzee adenovirus serotype 6 (ChAdV-6), which has a low antiserum retention rate in the human population, based on HAdV-5.
  • ChodV-6 chimpanzee adenovirus serotype 6
  • the present inventors confirmed that the cell infectivity and virus productivity increased up to 2.5 times compared to the conventional HAdV-5 vector.
  • the present inventors conducted a neutralizing antibody test using a polyclonal antibody to HAd-5 and human serum for the corresponding serotype, and found that the chimeric adenoviral vector had an average of 2.6 times lower anti-adenovirus-5 neutralization than the HAdV-5 viral vector.
  • Antibody titer was shown, demonstrating that the ChimAd vector was optimized for transfer of foreign genes into cells by avoiding anti-vector immunity against adenovirus serotype-5.
  • VOC variant of concern
  • the mutant was found to be the first mutant to have all three key mutations (E484K, K417N, N501Y) of VOCs in RBD as well as deletion in NTD, and in the case of AstraZeneca's AZD-1222 vaccine, beta mutant (B.1.351)
  • E484K, K417N, N501Y key mutations
  • NTD deletion in NTD
  • beta mutant B.1.351
  • a rapid decline in efficacy was confirmed for licensed vaccines, such as a decrease in vaccine efficacy by up to 10% compared to the previous initial week.
  • N protein (nucleocapsid protein, NP) is a protein that forms a nucleocapsid of about 50 kDa, and is known to be involved in signal transduction necessary for virus budding, RNA replication, and mRNA transcription.
  • the N protein was predicted to be highly antigenic, as more than 80% of coronavirus-infected patients had neutralizing antibodies against the protein.
  • the present invention was developed in response to the above needs, and an object of the present invention is to provide amino acids containing the S sequence of the beta mutant strain and the N partial sequence of the initial strain in order to prevent infection of SARS-coronavirus-2.
  • Another object of the present invention is to provide a chimeric adenovirus vector containing the S gene sequence of the beta mutant to prevent infection with SARS-coronavirus-2.
  • Another object of the present invention is to provide a chimeric adenovirus vector containing partial sequences of the S gene of the beta mutant strain and the N gene of the initial strain.
  • Another object of the present invention is to provide a vaccine for further mitigating the infection of SARS-coronavirus-2 through the enhancement of T-cell immunity.
  • Another object of the present invention is to provide a vaccine for preventing infection of SARS-coronavirus-2.
  • the present invention includes amino acids 1 to 1270 of the spike protein of the beta mutant strain of SARS-coronavirus-2, wherein, based on the initial week, 614, 682 to 685, 986, It has a mutation at amino acid residue No. 987, where the aspartic acid amino acid of protein No. 614 of the spike protein is substituted with glycine, arginine amino acid No. 682 is glutamine, alanine amino acid No.
  • 683 is glutamine, 684, 685 arginine
  • the amino acids are alanine and glutamine, respectively, at 986 and 987 consecutive
  • amino acid residues from 44 to 180 of the nucleocapsid protein of SARS-coronavirus-2 are linked to the N-terminus of the spike protein of the beta mutant strain of SARS-coronavirus-2, , Contains a gene encoding a fusion polypeptide prepared by linking amino acid residues 247 to 364 of the nucleocapsid protein of SARS-Coronavirus-2 to the C-terminus of the spike protein of the beta mutant SARS-Coronavirus-2 It provides an insert that does.
  • the amino acid residues from positions 44 to 180 of the nucleocapsid protein of the SARS-coronavirus-2 consist of the amino acid sequence of SEQ ID NO: 3, and the nucleocapsid of the SARS-coronavirus-2
  • the amino acid residues at positions 247 to 364 of the capsid protein are preferably composed of the amino acid sequence of SEQ ID NO: 4, but all mutant sequences that achieve the object of the present invention through mutation such as one or more substitutions, deletions, inversions, etc. to the corresponding sequence are also present. included in the scope of the invention.
  • the gene encoding amino acid residues 44 to 180 of the nucleocapsid protein of SARS-coronavirus-2 linked to the N-terminus of the spike protein is the nucleotide sequence shown in SEQ ID NO: 5
  • the gene encoding amino acid residues 247 to 364 of the nucleocapsid protein of SARS-coronavirus-2 linked to the C-terminus of the spike protein is preferably composed of the nucleotide sequence shown in SEQ ID NO: 6, All mutant sequences that achieve the object of the present invention through mutation of one or more substitutions, deletions, inversions, etc. in the sequence are also included in the scope of the present invention.
  • the modified spike protein of the beta mutant of SARS-coronavirus-2 consists of the amino acid sequence shown in SEQ ID NO: 1, and the modified spike protein of the beta mutant of SARS-coronavirus-2
  • the gene encoding the spike protein is preferably composed of the nucleotide sequence shown in SEQ ID NO: 2, but all mutant sequences that achieve the object of the present invention through mutation such as one or more substitutions, deletions, inversions, etc. in the corresponding sequence are also within the scope of the present invention. included in
  • the present invention provides a recombinant expression vector comprising the insert of the present invention.
  • the expression vector is preferably a viral vector or a non-viral vector
  • the viral vector is a group consisting of an adenovirus vector, an adeno-associated virus vector, a lentiviral vector, a retroviral vector, and a herpesvirus vector. It is preferably selected from, and in one embodiment, it is preferably a chimeric adenoviral vector, but is not limited thereto.
  • the non-viral vector is preferably a plasmid, cosmid, phagemid, or bacterial artificial chromosome (BAC), but is not limited thereto.
  • the adenovirus vector replaces the terminal domain of the fiber protein of human adenovirus type 5 with the spine gene of chimpanzee adenovirus serotype 6, and the tail of human adenovirus type 5 and a fibrous protein combined with a Shaft domain and a protrusion domain of chimpanzee adenovirus serotype 6, but is not limited thereto.
  • the protein expressed by the chimpanzee adenovirus serotype 6 protrusion gene preferably includes the amino acid sequence of SEQ ID NO: 7, but is not limited thereto.
  • the present invention provides a vaccine composition for preventing infection of SARS-CoV-2, comprising the recombinant expression vector of the present invention as an active ingredient.
  • the vaccine preferably has an effect on one or more viruses selected from the group consisting of SARS-CoV-2 virus initial strain, delta virus, beta and Omicron virus mutant strains, but is not limited thereto.
  • the vector of the present invention is preferably capable of infecting mammalian cells, but is not limited thereto.
  • the present invention provides a host cell transformed with a chimeric adenovirus vector comprising the S gene derived from the beta mutation of the present invention or a part of the N gene linked to the S gene.
  • the present invention provides a method for producing a recombinant virus using the transformed cell of the present invention.
  • the present invention provides a method of using the two recombinant chimeric adenoviruses obtained by the present invention as a vaccine in an animal model.
  • the present invention provides mouse immunity and virus neutralization efficacy using the recombinant chimeric adenovirus produced by the above method.
  • the present invention provides efficacy for immune response of specific T cells according to the recombinant chimeric adenovirus produced by the above method.
  • the present invention provides the protective efficacy of the initial strain after immunization with the recombinant chimeric adenovirus produced by the above method.
  • the present invention provides a vaccine composition for preventing or treating an infection of SARS-CoV-2, comprising the chimeric adenovirus vector of the present invention as an active ingredient.
  • the present invention is a vaccine composition for preventing SARS-coronavirus-2 infection, preferably a vaccine capable of mitigating or preventing the infection of beta, delta, and omicron mutant strains as well as the initial strain, but is not limited thereto.
  • the present inventors developed a vaccine composition effective against not only the initial strain but also the dominant strain, the delta mutant strain, based on the S protein of the beta mutant strain using the ChimAd vector as a delivery vehicle, and in addition, to maximize the T cell immune response, the form linked to the N protein
  • the present invention was completed through the evaluation of cellular immunogenicity and protective ability against viral infection of the neutralizing antibody against the S protein of .
  • the present invention provides a chimeric adenovirus vector encoding the beta-mutant S protein as a vaccine composition for preventing SARS-coronavirus-2 infection.
  • the present invention provides a chimeric adenoviral vector encoding a protein in which the S protein and the N protein are fused, including a portion of the N protein capable of enhancing T-cell immunity to the beta-mutant S protein.
  • Chimeric protein or “chimeric polypeptide” generally refers to a protein composed of multiple protein components, each of which is a series of polypeptides linked through amino-terminal (N-terminal, NTD) and carboxy-terminal (C-terminal, CTD) bonds.
  • NP chimeric protein or polypeptide
  • CCD chimeric protein or polypeptide
  • the peptide of the present invention is preferably expressed in various delivery vehicles such as recombinant adenovirus vectors, RNA, recombinant proteins, and non-viral vectors, and more preferably inserted into chimeric adenovirus vectors, but is not limited thereto.
  • the chimeric adenoviral vector of the present invention is in the state of patent application (Korean Patent Application No.: 10-2022-0034885), and reference is made to the patent for specific descriptions, and is omitted herein.
  • the chimeric adenovirus containing the polynucleotide S is called ChimAd-CV
  • ChimAd-CVN The chimeric adenovirus containing the NP(NTD)-Spike-NP(CTD) is called ChimAd-CVN.
  • Cells transfected with the chimeric adenovirus vector containing the genes of the present invention are preferably mammalian cells including HEK293, PERC6 or 911 cells, more preferably HEK293 cells, but are not limited thereto.
  • an adenovirus-producing cell suitable for the present invention should be a cell capable of producing a virus by trans-complementation of the E1 and E3 gene deletion sites of a replication defective adenovirus, and the viral early transcription gene A number of cells transformed with .
  • Cells into which the E1 and E3 genes are preferred include A549 (carcinomic human alveolar basal epithelial cell), HeLa (Human cervical cancer cell), CHO (Chinese hamster ovary cell), MRC5 (human lung fibroblast), and PC12 (rat pheochromocytoma cell) etc., but not limited thereto.
  • ChimAd-CV and ChimAd-CVN viruses when producing ChimAd-CV and ChimAd-CVN viruses, it is preferable to infect host cells at a cell density of 1x10 6 cells/ml with a titer of 2 MOI and recover the cells after 48 hours, but is not limited thereto.
  • Tween20 and 20 U/ml benzonase to induce host cell DNA degradation are preferably treated for 4 hours, but are not limited thereto.
  • surfactants capable of eluting viruses include Tween80, Triton X-100, ZwittergentTM3-14, and CHAPS, but are not limited thereto.
  • the present invention provides ChimAd-CV and ChimAd-CVN produced by the above method.
  • the ChimAd-CV and ChimAd-CVN vaccine compositions produced in the present invention provide excellent neutralizing antibody titers against the initial strain, beta mutant strain and delta mutant strain.
  • the ChimAd-CV and ChimAd-CVN vaccine compositions produced according to an embodiment of the present invention provide excellent neutralizing antibody titers against pseudoviruses based on beta mutants.
  • the ChimAd-CV and ChimAd-CVN vaccine compositions produced in the present invention provide efficacy against the immune response of specific T cells.
  • ELISPOT Enzyme-Linked ImmunoSpot, ELISPOT
  • Mouse splenocytes immunized with ChimAd-CV or ChimAd-CVN were isolated to confirm an immune response to S protein or NP peptide.
  • ChimAd-CVN confirmed cellular immunity more than twice as good as ChimAd-CV, it is thought that higher T-cell immunity is induced by linking N protein.
  • a high initial T-cell immune response is known to play a role in mitigating the severity of the coronavirus by itself, and in some cases, anti-N protein antibodies formed by N protein are not related to direct virus neutralization, but N protein specific T -Enhancement of cellular immunity correlates with neutralizing antibody titer.
  • N protein specific T -Enhancement of cellular immunity correlates with neutralizing antibody titer.
  • SARS-CoV-2 responsive T cell numbers and anti-Spike IgG levels are both associated with protection from COVID-19: A prospective cohort study in keyworkers. MedRxiv, 2020-11. Ni, L., Ye, F., Cheng, M. L., Feng, Y., Deng, Y. Q., Zhao, H., ... & Dong, C. (2020). Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals. Immunity, 52(6), 971-977]
  • the ChimAd-CV and ChimAd-CVN vaccine compositions produced by the above method provide protective efficacy against early strains.
  • the present invention can provide excellent efficacy as a preventive vaccine against the SARS-CoV-2 virus.
  • a vaccine composition comprising a beta mutant S sequence or an S sequence fused with N based on a chimeric adenovirus vector was developed. It has excellent cross-immunogenicity with high neutralizing antibodies against the initial strain and mutant strain (beta, delta, omicron), and the vaccine containing the N sequence provided the efficacy of maximizing T-cell immunity.
  • it is considered that it can be applied as a preventive vaccine for SARS-CoV-2 as it has excellent defense against early strains.
  • FIG. 1 is a schematic diagram of cloning ChimAd-CV and ChimAd-CVN using the Gibson assembly gene synthesis method in a chimeric adenovirus (ChimAd) vector obtained through gene synthesis.
  • Figure 2 shows the virus-binding antibody titers induced by ChimAd-CV and ChimAd-CVN using BALB/c female mice as A for the initial strain S protein and as B for the beta mutant S protein.
  • Figure 3 shows the neutralizing antibody titers induced by ChimAd-CV and ChimAd-CVN using BALB/c female mice as neutralizing antibody measurement results using pseudovirus of beta mutant (B.1.351 variant type).
  • A-B SARS-coronavirus-2 (Wuhan-Hu-1) pseudovirus neutralizing antibody test
  • C-D Beta mutant strain (B.1.351) Pseudovirus neutralizing antibody test.
  • Figure 4 shows the results of cellular immunity (A, B) using S peptides of ChimAd-CV and ChimAd-CVN as stimulating antigens using BALB/c female mice and the results of cellular immunization (C, B) using NP peptides as stimulating antigens. D).
  • Figure 5 shows the virus-binding antibody titers induced by ChimAd-CV and ChimAd-CVN using hACE2 transgenic female mice, with A for the initial strain S protein and B for the delta mutant S protein.
  • FIG. 6 shows the results (A, B) of neutralizing antibody measurement using FRNT of the initial strain as a result of confirming the viral neutralizing antibody titer induced by ChimAd-CV and ChimAd-CVN using hACE2 transgenic female mice.
  • C, D neutralizing antibody measurement results using FRNT of the delta mutant strain are shown.
  • A-B SARS-coronavirus-2 (Wuhan-Hu-1) FRNT neutralizing antibody test
  • C-D Delta mutant strain (B.1.617.2) FRNT neutralizing antibody test.
  • Figure 7 shows the results of comparison of body weights and mortality rates between ChimAd-CV and ChimAd-CVN for initial infection in mouse animal models immunized by ChimAd-CV and ChimAd-CVN using hACE2 transgenic female mice.
  • Figure 8 shows the coronavirus titers detected in the lungs after challenge with SARS-coronavirus-2 (Wuhan-Hu-1).
  • Figure 9 shows the antibody titer induced according to the inoculation dose of ChimAd-CV using BALB/c female mice as a result for the S protein of SARS-coronavirus-2.
  • Figure 10 shows the virus neutralizing antibody titer induced by the inoculation dose of ChimAd-CV using BALB/c female mice with various mutant viruses (Wuhan, beta mutant, delta, Omicron (BA.1)) VLP (Virus Like Particle ) is represented by the measurement results (A to D).
  • A is a SARS-coronavirus-2 (Wuhan-Hu-1) VLP neutralizing antibody test
  • B is a beta mutant (B.1.351) VLP neutralizing antibody test
  • C is a delta mutant (B.1.617.2) VLP neutralization.
  • Antibody test is Omicron (BA.1) VLP neutralizing antibody test.
  • FIG. 11 shows the results of cellular immunity induced by the inoculation dose of ChimAd-CV using BALB/c female mice, and S peptide was used as a stimulating antigen.
  • Example 1 Genetic modification encoding SARS-CoV-2 S protein, N protein
  • Example 2 Gene synthesis to be used as SARS-CoV-2 vaccine material
  • ChimAd vector which is a chimeric adenovirus vector
  • PCR amplification was performed excluding part of the gene of the GOI binding region using the plasmid DNA of the ChimAd vector as a template, and the amplified
  • the product (about 33.4 Kb) was purified through a gel extraction process.
  • the inserted gene for cloning was PCR amplified to include the same sequence (homologous region, about 20 bp) as the 5' and 3' ends of the GOI binding region in the ChimAd vector, and the CV of the amplification product obtained using the primers in Table 2 below (about 3.8 Kb) and CVN (about 4.7 Kb), and then combined with the previously purified DNA PCR amplification products through a gibson assembly reaction, respectively, two types of ChimAd-CV and ChimAd-CVN plasmids were finally obtained (Fig. 1).
  • Example 4 Production and purification of chimeric adenovirus-based corona vaccine candidates
  • each plasmid is treated with PacI restriction enzyme (NEB, R0547L) to eliminate unnecessary pBR322 for adenovirus formation.
  • PacI restriction enzyme NEB, R0547L
  • the origin of replication and kanamycin resistance gene sequences were cut and removed, and HEK293 cells were transformed using Lipofectamine 2000 (Invitrogen, 11668027), and maintained for 10-20 days in an incubator at 37°C and a CO2 partial pressure of 5.0%. After culturing, a primary virus stock was obtained.
  • ChimAd-CV and ChimAd-CVN virus production was performed by infecting HEK293 cells at a cell density of about 1x 10 6 cells/ml with a titer of 2 MOI using the primary virus stock obtained above, and after 48 hours, 0.45 ⁇ m CFF microfiltration filter (Cytiva, CFP-4-E-6A) was used to enrich the cells.
  • the recovered cells were treated with 0.5% Tween20 (Sigma-aldrich, P2287-500ML) and 20 U/ml benzonase (Milipore, E1014-25KU) for 4 hours to induce cell lysis and host cell DNA degradation.
  • Virus samples in PBS buffer were subjected to anion exchange chromatography using a HiTrap Capto Q ImpRes column (Cytiva, 17547051) and fast protein liquid chromatography (FPLC) using a Capto core 700 column (Cytiva, 17548115) to detect ChimAd-CV and ChimAd-CVN.
  • the virus was isolated, finally formulated into a vaccine buffer, and finally filtered with a 0.2 ⁇ m ULTA GF filter (Cytiva, KGF-A-9610TT) to secure two vaccine candidates.
  • Mouse immunization was performed using the ChimAd-CV and ChimAd-CVN vaccine candidates obtained by the above preparation method. A total of 4 groups were included in the test group: ChimAd used as buffer, mock-up, ChimAd-CV and ChimAd-CVN.
  • ChimAd used as buffer
  • mock-up ChimAd-CV
  • ChimAd-CVN ChimAd-CVN
  • For the immunity induction test 5-week-old BALB/c female mice were used, and each adenovirus test group was injected at a concentration of 2.5*10 ⁇ 9 vp/dose. Inoculations were administered intramuscularly twice a total of 1st and 2nd vaccinations at 4-week intervals (day 0 and 28), and 4 and 6 weeks after the first vaccination (day 27 and 42), blood was collected and serum was collected. isolated and the spleen was removed.
  • a binding antibody test was performed. To perform the test, 100 ng / well of the coronavirus initial strain and beta mutant S protein were dispensed at a concentration of 1 ug / ml in a microplate and coated for more than 16 hours at 4 degrees. After the reaction, the plate was washed more than 4 times with 1XPBS containing 0.05% Tween20 (PBS-T), and 100ul of 0.5% casein was dispensed and reacted at 37 ° C for 1 hour. After washing with PBS-T, each serum was diluted 1:100 and then serially diluted 3 times to about 1x10 6 .
  • 1XPBS containing 0.05% Tween20 PBS-T
  • the mixture was reacted at 37° C. for 1 hour, washed with PBS-T, diluted with HRP-coupled goat anti-mouse IgG HRP (Thermo) at a ratio of 1:5000, and reacted for 1 hour.
  • 50 ⁇ l of TMB solution (Thermo) was dispensed to proceed with the color development, and the color development reaction proceeded for 15-20 minutes.
  • the reaction was stopped by adding an equal amount of Stop solution (Thermo), and the absorbance was measured at a wavelength of 450 nm. did
  • both the initial strain and the beta mutant showed a high binding antibody titer of 10 4 or more compared to the control group inoculated with ChimAd-CV and ChimAd-CVN.
  • Binding antibody titers for both the initial strain and the beta mutant were measured, but were found to be invalid values (FIG. 2).
  • the neutralizing antibody of the antibody in serum through mouse immunity induction test was tested.
  • the pseudovirus system is a virus that uses the backbone of Lentivirus to express the luminescent gene and Spike protein of the initial strain or beta mutant strain.
  • pseudoviruses containing Spike proteins of the initial strain and the beta mutant strain were produced using Takara's Lenti-XTMSARS-CoV-2 Packaging Mix product.
  • the produced pseudovirus was transformed into 293T cells in which ACE2 protein was overexpressed, and luciferase expression was quantified and measured, and neutralizing antibody tests were performed using a constant RLU (Relative Light Units) value.
  • RLU Relative Light Units
  • DMEM medium containing 10% FBS was used.
  • Serum was diluted 1/8 and then serially diluted 2-fold to about 10 3 .
  • Pseudoviruses having a constant RLU value were mixed in equal amounts with the diluted serum at 37°C and 5% CO 2 .
  • Neutralization was performed in an incubator for 1 hour.
  • the hACE2-293T cells cultured one day before were treated with 100 ⁇ l of the virus and serum mixture, and cultured in a 37°C, 5% CO 2 incubator for about 72 hours.
  • spleens were removed from each group 4 weeks after the first inoculation and 2 weeks after the second inoculation (6 weeks after the first inoculation).
  • Splenocytes were collected using a cell strainer (Falcon), and washed with PBS buffer. After washing, red blood cells were removed using red blood cell lysis buffer (Sigma), and the number of splenocytes was counted and diluted to the cell concentration used for the test.
  • R&D system's mouse IFN-gamma ELISpot kit was used, and the test was performed according to the manufacturer's instructions.
  • splenocytes of 1X10 5 cells and 2X10 4 cells were incubated for 16 hours in a 37°C, 5% CO2 incubator together using a peptide pool or Nucleprotein peptide pool of Genscript's initial stock Spike protein as stimulating antigens. After culturing, it was washed with a washing solution, and 100ul of IFN-gamma detection antibody bound to biotin was added thereto, followed by stirring at room temperature for 2 hours. After the reaction, it was washed 4 times using a washing solution, and 100 ⁇ l of streptavidin-AKP (alkaline phosphate) was added and incubated for 2 hours at room temperature. After the last wash, 100ul of BCIP/NBT substrate was added and reacted at room temperature for about 45 minutes. The number of cells in Unit (SFU) was converted.
  • SFU streptavidin-AKP
  • T-cell immunity was higher after the second vaccination (week 6) than after the first vaccination (week 4).
  • ChimAd-CVN induces a specific immune response about twice as high as ChimAd-CV against Spike protein peptides.
  • ChimAd-CVN induces a specific immune response to the Nucleocapsid peptide.
  • Example 6 ChimAd-CV, ChimAd-CVN antibody titer after animal immunization, neutralizing antibody titer and challenge test
  • mice 6-week-old female human ACE2 transgenic mice (tg(K18-ACE2)2Prlmn) purchased from Jackson Laboratory (ME, USA) were used. All of the above tests were conducted in a BSL-3 grade facility in the International Vaccine Institute. A total of 4 groups were included in the immunity induction test group: ChimAd used as buffer, mock-up, ChimAd-CV and ChimAd-CVN. The inoculum was injected at a concentration of 2.5 x 10 ⁇ 9 vp/dose. Mice were intramuscularly inoculated twice at 4-week intervals, and blood was collected 4 weeks and 6 weeks after the first priming inoculation.
  • mice immunity induction test After mouse immunity induction test, it was performed to measure the antibody titer in serum.
  • the initial strain S protein and the delta mutant strain S protein were dispensed at a concentration of 2ug/ml and 100ng/well each, and coated at 4 degrees for 16 hours or more.
  • 100ul of PBS containing 1% BSA was dispensed and reacted.
  • HRP-coupled Goat anti-mouse IgG HRP (Southern Biotech) was diluted 1:3000 and reacted at 37°C for 1 hour.
  • TMB solution (Millipore) was dispensed to proceed with color development, and then, an equal amount of 0.5 N HCl solution (Merck) was added to stop the reaction, and absorbance was measured at a wavelength of 450 nm.
  • mice immune induction test a test was performed to measure neutralizing antibodies in serum.
  • neutralizing antibody test first, each mouse serum was inactivated at 56° C. for 30 minutes, and serially diluted 2-fold using DMEM medium containing 2% FBS. Diluted serum and 4.5x10 2 PFU/25ul of primary strain coronavirus (NCCP #43326 (BetaCoV/Korea/KCDC03/2020) and 4.0x10 2 PFU/25ul of delta mutant coronavirus (NCCP #43390 (hCoV-19/Korea119861) /KDCA/2021) and reacted for 30 minutes at 37 ° C.
  • NCCP #43326 BetaCoV/Korea/KCDC03/2020
  • 4.0x10 2 PFU/25ul of delta mutant coronavirus NCCP #43390 (hCoV-19/Korea119861) /KDCA/2021
  • Vero cells 1.5x10 4 cells/well cultured one day before were treated with 50 ⁇ l of the virus and serum mixture and kept at 37 ° C, 5% CO 2 Incubated for 4 hours in an incubator After 4 hours of reaction, the supernatant was removed, washed with PBS buffer solution, treated with 300ul of 4% formaldehyde solution (Sigma), and fixed for more than 16 hours in a place without light. After washing with PBS, 100% cold methanol was added, reacted for 10 minutes, and then reacted for 1 hour at room temperature using a blocking solution, then anti-SARS-CoV diluted 1:3000.
  • the neutralizing antibody titer was confirmed as shown in Table 5, which confirmed that both the ChimAd-CV and ChimAd-CVN groups showed more than 100 times higher neutralizing antibody titer than the negative control group after the first inoculation. did After the second inoculation, it was confirmed that the ChimAd-CV inoculation group showed 30-fold higher neutralizing antibody titers and the ChimAd-CVN-inoculated group showed 50-fold higher neutralizing antibody titers.
  • 5x10 5 PFU SARS-CoV-2 virus (initial strain, delta mutant strain) was intranasally infected 4 weeks after boosting inoculation. After inoculation, survival and weight were monitored daily. On the 2nd, 4th and 7th days after infection, blood, lungs, liver, kidneys, and spleens were collected, organ weights were measured, and viral titers in the lungs were measured.
  • Plague assay test was performed to measure the residual virus titer in the lung.
  • Vero cells ATCC, Cat#CCL-81
  • the virus isolated from the lungs collected from all groups was diluted to 10 6 in a 4-fold stepwise manner, and 200 ul of the diluted virus was added to the cultured Vero cells and allowed to react at room temperature for 30 minutes.
  • the virus mixture was removed, covered with DMEM (Overlay media) containing 0.8% Agarose and 2% FBS, reacted at room temperature for 15 minutes, and then cultured for 72 hours in a 37°C, 5% CO 2 incubator. After fixation with 10% formalin solution for 1 hour, overlay media was removed, and plaques were stained for 5 minutes using crystal violet (Sigma). To confirm the virus titer, the number of plaques was counted, and each dilution factor and total amount (ml) were calculated to calculate the final titer.
  • Example 7 Immunogenicity test result through binding antibody/neutralizing antibody/cellular immunity assay according to DRF (Dose range finding) test of ChimAd-CV
  • a dose-response mouse immunization was performed using the ChimAd-CV vaccine candidate.
  • the doses of the ChimAd-CV vaccine candidate were divided into 4 groups as shown in Table 6, and were composed of stabilizer (Vehicle), mock up, and non-immunization groups.
  • the high and low concentrations were set at 2-fold from 5.0.E+09 VP/Dose (1/10 of the mouse standard human body).
  • Inoculations were administered intramuscularly twice a total of 1st and 2nd vaccinations (Day 0, Day 28) at intervals of 4 weeks, and blood was collected 4 weeks and 6 weeks after the first vaccination (Day 27, Day 42) and serum was collected. isolated and the spleen was removed.
  • the isolated serum was measured for binding antibody titers and neutralizing antibody titers to various corona viruses (Wuhan, beta-mutant, delta, and Omicron (BA.1)), and T-cell immune responses were analyzed with isolated spleens.
  • a binding antibody test was performed.
  • the coronavirus SARS-Coronavirus-2 and the beta mutant S protein were dispensed at a concentration of 1ug/ml by 100ng/well in a microplate and coated for more than 16 hours at 4 degrees. After the reaction, the plate was washed more than 4 times with 1XPBS containing 0.05% Tween20 (PBS-T), and 100ul of 0.5% casein was dispensed and reacted at 37 ° C for 1 hour.
  • each serum was diluted 1:100 and then serially diluted 3 times to about 1x10 6 . After dispensing, the mixture was reacted at 37° C. for 1 hour, washed with PBS-T, diluted with HRP-coupled goat anti-mouse IgG HRP (Thermo) at a ratio of 1:5000, and reacted for 1 hour. After the reaction was finished, 50 ⁇ l of TMB solution (Thermo) was dispensed to proceed with the color development, and the color development reaction proceeded for 15-20 minutes. After that, the reaction was stopped by adding an equal amount of Stop solution (Thermo), and the absorbance was measured at a wavelength of 450 nm. did
  • pVNT pseudovirus neutralization test
  • the neutralizing antibody of the antibody in serum through mouse immunity induction test was tested. This experiment was measured using Virongy's Rapid Cell-Based SARS-CoV-2 Neutralizing Antibody Assay Kit using Wuhan virus, beta mutant virus, delta virus, and Omicron (BA.1) virus.
  • the virus used in this test is a hybrid SARS-CoV-2 virus-like particle (VLP) developed by Virongy, which is combined with the 4 structural proteins (S, M, N, E) of SARS-CoV-2 and has luciferase can be expressed.
  • each mouse serum was first inactivated at 56° C. for 30 minutes, and the serum was diluted 1/8 using Virongy infection medium and then serially diluted 2-fold to about 10 3 .
  • a sample diluted in a white plate (cell culture) and 45ul of HA-CoV2(Luc)VLP were mixed and neutralized for 30 minutes in a 37°C 5% CO 2 incubator.
  • HEK293T (ACE2/TMPRSS2) cells were prepared at 2.5 x 10 4 cells/15 ⁇ L, treated with 15 ⁇ L of the virus and serum mixture, and cultured in a 37°C 5% CO 2 incubator for about 18 hours. After the incubation, add 7.5 ul of cell lysis buffer and leave on a shaker for 2 minutes. After adding 25ul of Luciferase solution and reacting for 1 minute, the luminescence value was measured using a luminometer. The measured RLU values were calculated using a statistical program to calculate IC50 values according to serum dilution factors.
  • a neutralizing antibody titer was confirmed (FIG. 10).
  • Target virus pVNTs (IC50) CV1 CV2 CV3 CV4 Wuhan virus 1177 577 311 607 Beta variant 965 416 627 344 Delta variant 517 325 798 115 Omicron (BA.1) 76 117 110 93
  • spleens were removed from each group 4 weeks after the first inoculation and 2 weeks after the second inoculation (6 weeks after the first inoculation).
  • Splenocytes were collected using a cell strainer (Falcon), and washed with PBS buffer. After washing, red blood cells were removed using red blood cell lysis buffer (Sigma), and the number of splenocytes was counted and diluted to the cell concentration used for the test.
  • R&D system's mouse IFN-gamma ELISpot kit was used, and the test was performed according to the manufacturer's instructions.
  • splenocytes of 1X10 5 cells and 2X10 4 cells and Genscript's SARS-coronavirus-2 Spike protein peptide pool were used as stimulating antigens and cultured for 16 hours in a 37°C 5% CO2 incubator. After culturing, it was washed with a washing solution, and 100ul of IFN-gamma detection antibody bound to biotin was added thereto, followed by stirring at room temperature for 2 hours. After the reaction, it was washed 4 times using a washing solution, and 100 ⁇ l of streptavidin-AKP (alkaline phosphate) was added and incubated for 2 hours at room temperature. After the last wash, 100ul of BCIP/NBT substrate was added and reacted at room temperature for about 45 minutes. The number of cells in Unit (SFU) was converted.
  • SFU number of cells in Unit
  • test group parking SPF/10 ⁇ 6 splenocytes (Spike peptide pool) CV1 CV2 CV3 CV4 Week 4 794 934 923 888 Week 6 1575 1691 1878 2491
  • PQGTTLPKGFYAEGSRGGS (SEQ ID NO: 3)
  • SEQ ID NO: 3 is a sequence bound to the N-terminus of the SARS-CoV-2-B.1.1.351 Spike protein
  • SEQ ID NO: 4 is a sequence bound to the C-terminus
  • the underline indicates a linker
  • an arrow indicates a SARS-CoV-2-B.1.1.351 Spike protein.
  • -2-B.1.1.351 Indicates the site where Spike protein is inserted.
  • AGA GGA GGA AGC (SEQ ID NO: 5) GGC GGC GGA GGA AGC GGC GGA GGA
  • AAG GAC CCC AAC TTC AAG GAC CAG GTG ATC CTG CTG AAC AAG CAC
  • ATC GAC GCC TAC AAG ACC TTC CCC TGA (SEQ ID NO: 6)
  • SEQ ID NO: 5 is a gene sequence encoding a polypeptide bound to the N-terminus of the SARS-CoV-2-B.1.1.351 Spike protein
  • SEQ ID NO: 6 is a gene sequence encoding a polypeptide bound to the C-terminus
  • the underline indicates the gene sequence encoding the linker
  • the arrow indicates the insertion site of the SARS-CoV-2-B.1.1.351 Spike protein.
  • SEQ ID NO: 7 Chimpanzee adenovirus serotype 6 Fiber knob domain a.a sequence

Abstract

The present invention relates to a chimeric adenoviral vector containing a spike protein or nucleocapsid (NP) of a beta variant (B.1.351) that prevents infection with SARS-CoV-2 virus. It has been confirmed that the vector of the present invention has a high neutralizing antibody titer and excellent T-cell immune response against not just the initial strain but also the delta variant and beta and omicron variants, and, against the initial strain of the virus, a 100% survival rate is exhibited and protective potency could be confirmed. The vector of the present invention shows a preventive effect against the SARS-CoV-2 virus and can thus be applied as a vaccine composition for prevention and treatment of the SARS-CoV-2 virus.

Description

사스-코로나바이러스-2 감염증 예방용 재조합 발현 벡터 및 그 응용Recombinant expression vector for prevention of SARS-CoV-2 infection and its application
본 발명은 사스-코로나바이러스-2 감염증 예방용 재조합 발현 벡터 및 그 응용 및 구체적으로는 그 벡터의 사스-코로나바이러스-2 예방 등에 대한 응용에 관한 것으로, 보다 상세하게는 베타 변이주의 스파이크 단백질(S 단백질) 및/또는 스파이크 단백질에 뉴클레오캡시드(N 단백질)을 연결하여 인코딩한 재조합 키메릭아데노바이러스 벡터를 포함하는 코로나-19 예방 백신으로, 사스-코로나바이러스-2 뿐만 아니라 변이주(베타, 델타, 오미크론 등) 감염증에 대한 예방 및 치료용 백신의 가능성을 제시한다. The present invention relates to a recombinant expression vector for the prevention of SARS-coronavirus-2 infection and its application, and specifically to the application of the vector to the prevention of SARS-coronavirus-2, etc., and more particularly, to the beta mutant spike protein (S A COVID-19 preventive vaccine containing a recombinant chimeric adenoviral vector encoded by linking a nucleocapsid (N protein) to a nucleocapsid (N protein) and/or a spike protein, as well as SARS-coronavirus-2 strains (beta, delta, Omicron, etc.) suggests the possibility of preventive and therapeutic vaccines against infections.
코로나바이러스는 직경 100-200 mm의 입자표면에 외피를 가지는 positive-sense single stranded RNA 바이러스로, 7종의 코로나바이러스가 사람의 호흡기와 소화기계 감염을 유발한다고 알려져 있고 7종 가운데 2종은 알파-코로나바이러스 속 그리고 나머지 5종은 베타-코로나바이러스 속으로 밝혀졌다. 신종 코로나바이러스 감염증(COVID-19, novel coronavirus, 2019-nCov)을 유발하는 사스-코로나바이러스 -2(Severe acute respiratory syndrome coronavirus 2, 이하, 'SARS-CoV-2'라 함)는 베타-코로나바이러스 속에 해당하며 2019년 12월 중국 우한시에서 최초로 보고된 후(Wuhan-Hu-1), 전세계적으로 확진자가 속출하고 있으며, 발생 이래 다양한 변이(알파 변이주, 베타 변이주, 델타 변이주 등)를 통해 팬데믹(pandemic)이 심화되고 있다.Coronaviruses are positive-sense single-stranded RNA viruses that have an envelope on the surface of particles with a diameter of 100-200 mm. Seven types of coronaviruses are known to cause respiratory and digestive infections in humans, and two of the seven types are alpha- genus of coronaviruses, and the remaining five species were identified as genus beta-coronaviruses. Severe acute respiratory syndrome coronavirus 2 (hereinafter referred to as 'SARS-CoV-2'), which causes COVID-19 (novel coronavirus, 2019-nCov), is a beta-coronavirus It is a genus, and after it was first reported in Wuhan, China in December 2019 (Wuhan-Hu-1), confirmed cases are appearing one after another worldwide. (pandemic) is intensifying.
SARS-CoV-2의 바이러스 외피는 스파이크 당단백질(spike glycoprotein, S 단백질), 헤마글루티닌-에스테라아제 이량체(hemagglutinin esterase dimer, HE) 등으로 이루어져 있다. S 단백질의 S1영역은 주로 RBD (Receptor binding domain)를 통한 수용체 (Angiotensin-converting enzyme 2, ACE2)와의 결합을, S2영역은 3차구조의 극적 변화를 동반한 직접적인 세포-바이러스 융합(감염개시)을 담당한다. 따라서, 주요 항원(antigen)으로서 S 단백질을 표적하는 백신후보물질들이 개발 중 및 상용화되었다.The viral envelope of SARS-CoV-2 is composed of spike glycoprotein (S protein) and hemagglutinin-esterase dimer (HE). The S 1 region of the S protein mainly binds to the receptor (Angiotensin-converting enzyme 2, ACE2) through the RBD (Receptor binding domain), and the S 2 region is a direct cell-virus fusion accompanied by a dramatic change in tertiary structure (infection initiation). Therefore, vaccine candidates targeting the S protein as a major antigen have been developed and commercialized.
RNA 바이러스는 DNA 복제 과정에서 오류를 교정하는 DNA repair pathway(base excision repair, nucleotide excision repair, mismatch repair)가 일어나는 DNA 바이러스와 달리, RNA 중합효소를 이용하므로 게놈복제과정에서 발생한 오류를 교정할 RNA repair 관련 단백질이 없기 때문에 DNA 바이러스 대비 돌연변이 발생이 빈번한 특성이 있다. SARS-CoV-2 변이 바이러스의 경우, S 단백질의 N-말단 도메인(N-terminal domain, NTD)과 RBD 도메인에서 돌연변이가 빈번하게 발생하였고, 이로 인해 초기주의 S단백질을 타겟으로 개발된 기허가 백신들이 구조적 변화가 발생한 변이 바이러스의 S 단백질에 대한 항체 결합능력이 감소되어 변이 바이러스에 대한 방어 효능이 저하되었다.Unlike DNA viruses, which have DNA repair pathways (base excision repair, nucleotide excision repair, mismatch repair) that correct errors during DNA replication, RNA viruses use RNA polymerase to correct errors that occur during genome replication. Since there is no related protein, mutations are more frequent compared to DNA viruses. In the case of the SARS-CoV-2 mutant virus, mutations frequently occurred in the N-terminal domain (NTD) and RBD domain of the S protein, and as a result, a licensed vaccine developed targeting the S protein of the initial state. The antibody-binding ability to the S protein of the mutant virus in which these structural changes occurred was reduced, and the protective efficacy against the mutant virus was lowered.
아데노바이러스 벡터 백신은 병원체에 대한 항원 유전자를 아데노바이러스 주형에 삽입하여 전달하여 체내에서 항원 단백질을 발현해 면역반응을 유도하는 백신으로, 인간 아데노아이러스 혈청형 5(HAdV-5)벡터가 가장 널리 사용되지만 인간 집단내 높은 혈청보유율로 인해 백신 효능이 감소된다는 문제점이 있다. Adenovirus vector vaccine is a vaccine that induces an immune response by expressing an antigen protein in the body by inserting and delivering an antigen gene for a pathogen into an adenovirus template. Human adenoirus serotype 5 (HAdV-5) vector is the most widely used. Although used, there is a problem that vaccine efficacy is reduced due to high serum retention rates in the human population.
이에 반해, 키메릭아데노바이러스(ChimAd) 벡터는 HAdV-5를 기반으로 인간 집단 내 항혈청 보유율이 낮은 침팬지 아데노바이러스 혈청형 6(chimpanzee adenovirus 혈청형 6, ChAdV-6)의 돌기 유전자로 치환한 재조합 키메릭 아데노바이러스 벡터로서 본 발명자들은 기존 HAdV-5 벡터 대비 세포 감염능과 바이러스 생산성이 최대 2.5 배 증가됨을 확인하였다. 또한, 본 발명자들은 HAd-5에 대한 다클론항체와 해당 혈청형에 대한 인간 혈청을 이용한 중화항체 시험을 통해 키메릭 아데노바이러스 벡터가 HAdV-5 바이러스 벡터보다 평균 2.6 배 낮은 항 아데노바이러스-5 중화항체 타이터를 보여주었고 이는 ChimAd 벡터가 아데노바이러스 혈청형-5에 대한 항 벡터 면역을 회피함으로써 세포로의 외래 유전자 전달에 최적화된 벡터임을 증명한 것이다.On the other hand, the chimeric adenovirus (ChimAd) vector is a recombinant key substituted with the projection gene of chimpanzee adenovirus serotype 6 (ChAdV-6), which has a low antiserum retention rate in the human population, based on HAdV-5. As a Meric adenovirus vector, the present inventors confirmed that the cell infectivity and virus productivity increased up to 2.5 times compared to the conventional HAdV-5 vector. In addition, the present inventors conducted a neutralizing antibody test using a polyclonal antibody to HAd-5 and human serum for the corresponding serotype, and found that the chimeric adenoviral vector had an average of 2.6 times lower anti-adenovirus-5 neutralization than the HAdV-5 viral vector. Antibody titer was shown, demonstrating that the ChimAd vector was optimized for transfer of foreign genes into cells by avoiding anti-vector immunity against adenovirus serotype-5.
2020년 12월 남아프리카 공화국에서 사스-코로나바이러스-2의 치명적인 변이가 발생하였고 베타 변이로의 분류 및 우려변이(variant of concern, VOC)로 지정되었다. 해당 변이주는 NTD에서의 deletion 뿐만 아니라 RBD에서 VOC들의 핵심 변이 3종(E484K, K417N, N501Y)을 모두 가지는 최초의 변이로 판명되었으며, 아스트라제네카의 AZD-1222 백신의 경우 베타 변이주(B.1.351)군에서 백신 효력이 기존 초기주 대비 10 %까지 저하되는 등 기허가 백신들에 대한 급격한 효력 저하가 확인되었다. In December 2020, a fatal mutation of SARS-CoV-2 occurred in South Africa and was classified as a beta mutation and designated as a variant of concern (VOC). The mutant was found to be the first mutant to have all three key mutations (E484K, K417N, N501Y) of VOCs in RBD as well as deletion in NTD, and in the case of AstraZeneca's AZD-1222 vaccine, beta mutant (B.1.351) In the group, a rapid decline in efficacy was confirmed for licensed vaccines, such as a decrease in vaccine efficacy by up to 10% compared to the previous initial week.
또한, 베타 변이에 감염되었던 회복기 환자의 혈청에서 초기주 및 다른 변이 바이러스들에 대해 교차반응(cross-neutralization)이 확인됨에 따라 초기주에 대한 S단백질이 아닌 변이 바이러스에 대한 S단백질을 표적으로 한 백신 개발로 방향이 바뀌는 추세이며, 지속적인 변이 바이러스 출현에 맞서 대항 가능한 범용 백신(universal vaccine)에 대한 필요성이 대두되고 있다.In addition, as cross-neutralization was confirmed for the initial strain and other mutant viruses in the serum of convalescent patients who were infected with the beta mutation, the S protein for the mutant virus, not the S protein for the initial strain, was targeted. The direction of vaccine development is changing, and the need for a universal vaccine capable of fighting against the continuous emergence of mutant viruses is emerging.
N 단백질(nucleocapsid protein, NP)은 약 50 kDa의 뉴클레오캡시드를 형성하는 단백질로, 바이러스 출아(budding)에 필요한 신호전달, RNA 복제 및 mRNA 전사에 관여하는 것으로 알려져 있다. N 단백질은 코로나바이러스 감염 환자의 약 80 % 이상이 해당 단백질에 대한 중화항체를 가지고 있어 N 단백질의 항원성이 높은 것으로 예측되었다. N protein (nucleocapsid protein, NP) is a protein that forms a nucleocapsid of about 50 kDa, and is known to be involved in signal transduction necessary for virus budding, RNA replication, and mRNA transcription. The N protein was predicted to be highly antigenic, as more than 80% of coronavirus-infected patients had neutralizing antibodies against the protein.
코로나바이러스에서 변이가 거의 없는 부분으로 알려져 있으나, 해당 영역에 변이는 변이 바이러스의 전염력을 높임이 보고되었다. 뿐만아니라, SARS-CoV-2의 N단백질을 암호화하는 백신을 접종한 C57BL/6 마우스에서 강력한 N 특이적 항체 반응과 세포성 면역반응이 유도되었다. 높은 초기 T-세포성 면역반응은 그 자체로 코로나바이러스에 대한 중증 완화의 역할을 한다고 알려져 있고, 일각에서는 N 단백질에 의해 형성된 항-N단백질 항체가 직접적인 바이러스 중화에는 관련이 없으나 N단백질 특이적인 T-세포 면역의 증대가 중화항체 역가와 상호관계가 있다고 보고되어 백신 항원으로 가치가 더욱 높을 것으로 기대된다.It is known that there is little variation in coronaviruses, but it has been reported that mutations in that area increase the infectivity of the mutant virus. In addition, a strong N-specific antibody response and a cellular immune response were induced in C57BL/6 mice vaccinated with a vaccine encoding the N protein of SARS-CoV-2. A high initial T-cell immune response is known to play a role in mitigating the severity of the coronavirus by itself, and in some cases, anti-N protein antibodies formed by N protein are not related to direct virus neutralization, but N protein specific T - It has been reported that the increase in cellular immunity is correlated with the neutralizing antibody titer, so it is expected to be more valuable as a vaccine antigen.
[선행 특허 문헌][Prior Patent Literature]
대한민국 특허공개번호 제1020210108331호 Korean Patent Publication No. 1020210108331
본 발명은 상기의 필요성에 의하여 개발되었으며, 본 발명의 목적은 사스-코로나바이러스-2의 감염을 예방하기 위해서 베타 변이주의 S 서열과 초기주의 N 일부 서열을 포함하는 아미노산을 제공하는 것이다. The present invention was developed in response to the above needs, and an object of the present invention is to provide amino acids containing the S sequence of the beta mutant strain and the N partial sequence of the initial strain in order to prevent infection of SARS-coronavirus-2.
본 발명의 다른 목적은 사스-코로나바이러스-2의 감염을 예방하기 위해서 베타 변이주의 S 유전자 서열을 포함하는 키메릭아데노바이러스 벡터를 제공하는 것이다.Another object of the present invention is to provide a chimeric adenovirus vector containing the S gene sequence of the beta mutant to prevent infection with SARS-coronavirus-2.
본 발명의 다른 목적은 상기 베타 변이주의 S와 초기주의 N 유전자 일부 서열을 포함하는 키메릭 아데노바이러스 벡터를 제공하는 것이다.Another object of the present invention is to provide a chimeric adenovirus vector containing partial sequences of the S gene of the beta mutant strain and the N gene of the initial strain.
본 발명의 또 다른 목적은 T-세포면역 증대를 통해 사스-코로나바이러스-2의 감염을 더욱 완화하기 위한 백신을 제공하는 것이다. Another object of the present invention is to provide a vaccine for further mitigating the infection of SARS-coronavirus-2 through the enhancement of T-cell immunity.
본 발명의 또 다른 목적은 사스-코로나바이러스-2의 감염을 예방하기 위한 백신을 제공하는 것이다.Another object of the present invention is to provide a vaccine for preventing infection of SARS-coronavirus-2.
상기 목적을 달성하기 위하여, 본 발명은 사스-코로나바이러스-2의 베타변이주의 스파이크 단백질의 아미노산 1번부터 1270번까지를 포함하며, 여기서 초기주 기준 614번, 682번부터 685번, 986번, 987번의 아미노산 잔기에 돌연변이를 가지고 있으며, 여기서 상기 단백질은 스파이크 단백질의 614번 단백질의 아스파르트산 아미노산이 글라이신으로 치환되고, 682번 아르기닌 아미노산은 글루타민, 683번 알라닌 아미노산은 글루타민, 684번, 685번 아르기닌 아미노산은 각각 알라닌, 글루타민으로, 986번 및 987번의 연속된 프롤린 치환 돌연변이로 치환되는 사스-코로나바이러스-2의 베타변이주의 스파이크 단백질을 코딩하는 유전자를 포함하는 삽입체를 제공한다.In order to achieve the above object, the present invention includes amino acids 1 to 1270 of the spike protein of the beta mutant strain of SARS-coronavirus-2, wherein, based on the initial week, 614, 682 to 685, 986, It has a mutation at amino acid residue No. 987, where the aspartic acid amino acid of protein No. 614 of the spike protein is substituted with glycine, arginine amino acid No. 682 is glutamine, alanine amino acid No. 683 is glutamine, 684, 685 arginine The amino acids are alanine and glutamine, respectively, at 986 and 987 consecutive Provided is an insert containing a gene encoding a spike protein of a beta mutant strain of SARS-coronavirus-2 substituted with a proline substitution mutant.
본 발명의 일 구현예에 있어서, 상기 사스-코로나바이러스-2의 베타변이주의 스파이크 단백질의 N말단에 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 44번부터 180번까지의 아미노산 잔기를 연결하고, 사스-코로나바이러스-2의 베타변이주의 스파이크 단백질의 C 말단에 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 247번부터 364번의 아미노산 잔기를 연결하여 제조된 융합 폴리펩타이드를 코딩하는 유전자를 포함하는 삽입체를 제공한다.In one embodiment of the present invention, amino acid residues from 44 to 180 of the nucleocapsid protein of SARS-coronavirus-2 are linked to the N-terminus of the spike protein of the beta mutant strain of SARS-coronavirus-2, , Contains a gene encoding a fusion polypeptide prepared by linking amino acid residues 247 to 364 of the nucleocapsid protein of SARS-Coronavirus-2 to the C-terminus of the spike protein of the beta mutant SARS-Coronavirus-2 It provides an insert that does.
본 발명의 일 구현예에서 상기 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 44번부터 180번까지의 아미노산 잔기는 서열번호 3의 아미노산 서열로 이루어지고, 상기 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 247번부터 364번의 아미노산 잔기는 서열번호 4의 아미노산 서열로 이루어진 것이 바람직하나, 해당 서열에 하나 이상의 치환, 결손, 역위 등의 돌연변이를 통하여 본 발명의 목적을 달성하는 모든 돌연변이 서열도 본 발명의 범위에 포함된다.In one embodiment of the present invention, the amino acid residues from positions 44 to 180 of the nucleocapsid protein of the SARS-coronavirus-2 consist of the amino acid sequence of SEQ ID NO: 3, and the nucleocapsid of the SARS-coronavirus-2 The amino acid residues at positions 247 to 364 of the capsid protein are preferably composed of the amino acid sequence of SEQ ID NO: 4, but all mutant sequences that achieve the object of the present invention through mutation such as one or more substitutions, deletions, inversions, etc. to the corresponding sequence are also present. included in the scope of the invention.
본 발명의 다른 구현예에 있어서, 상기 스파이크 단백질의 N말단에 연결된 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 44번부터 180번까지의 아미노산 잔기를 코딩하는 유전자는 서열번호 5에 기재된 염기서열로 이루어지고, 상기 스파이크 단백질의 C 말단에 연결된 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 247번부터 364번의 아미노산 잔기를 코딩하는 유전자는 서열번호 6에 기재된 염기서열로 이루어진 것이 바람직하나, 해당 서열에 하나 이상의 치환, 결손, 역위 등의 돌연변이를 통하여 본 발명의 목적을 달성하는 모든 돌연변이 서열도 본 발명의 범위에 포함된다.In another embodiment of the present invention, the gene encoding amino acid residues 44 to 180 of the nucleocapsid protein of SARS-coronavirus-2 linked to the N-terminus of the spike protein is the nucleotide sequence shown in SEQ ID NO: 5 The gene encoding amino acid residues 247 to 364 of the nucleocapsid protein of SARS-coronavirus-2 linked to the C-terminus of the spike protein is preferably composed of the nucleotide sequence shown in SEQ ID NO: 6, All mutant sequences that achieve the object of the present invention through mutation of one or more substitutions, deletions, inversions, etc. in the sequence are also included in the scope of the present invention.
본 발명의 또 다른 구현예에 있어서, 상기 사스-코로나바이러스-2의 베타변이주의 변형된 스파이크 단백질은 서열번호 1에 기재된 아미노산 서열로 이루어지고, 상기 사스-코로나바이러스-2의 베타변이주의 변형된 스파이크 단백질을 코딩하는 유전자는 서열번호 2에 기재된 염기서열로 이루어진 것이 바람직하나, 해당 서열에 하나 이상의 치환, 결손, 역위 등의 돌연변이를 통하여 본 발명의 목적을 달성하는 모든 돌연변이 서열도 본 발명의 범위에 포함된다.In another embodiment of the present invention, the modified spike protein of the beta mutant of SARS-coronavirus-2 consists of the amino acid sequence shown in SEQ ID NO: 1, and the modified spike protein of the beta mutant of SARS-coronavirus-2 The gene encoding the spike protein is preferably composed of the nucleotide sequence shown in SEQ ID NO: 2, but all mutant sequences that achieve the object of the present invention through mutation such as one or more substitutions, deletions, inversions, etc. in the corresponding sequence are also within the scope of the present invention. included in
또 본 발명은 상기 본 발명의 삽입체를 포함하는 재조합 발현 벡터를 제공한다.In addition, the present invention provides a recombinant expression vector comprising the insert of the present invention.
본 발명의 일 구현예에 있어서, 발현 벡터는 바이러스 벡터 또는 비바이러스 벡터인 것이 바람직하고, 상기 바이러스 벡터는 아데노바이러스 벡터, 아데노연관바이러스 벡터, 렌티바이러스 벡터, 레트로바이러스 벡터 및 헤르페스바이러스 벡터로 이루어진 그룹으로부터 선택되는 것이 바람직하고, 일 실시예에서 키메릭아데노바이러스 벡터인 것이 바람직하나 이에 한정되지 아니한다.In one embodiment of the present invention, the expression vector is preferably a viral vector or a non-viral vector, and the viral vector is a group consisting of an adenovirus vector, an adeno-associated virus vector, a lentiviral vector, a retroviral vector, and a herpesvirus vector. It is preferably selected from, and in one embodiment, it is preferably a chimeric adenoviral vector, but is not limited thereto.
본 발명의 다른 구현예에서 비바이러스 벡터는 플라스미드, 코스미드, 파지미드, 또는 박테리아 인공 염색체(BAC)인 것이 바람직하나 이에 한정되지 아니한다.In another embodiment of the present invention, the non-viral vector is preferably a plasmid, cosmid, phagemid, or bacterial artificial chromosome (BAC), but is not limited thereto.
본 발명의 다른 구현예에 있어서, 상기 아데노바이러스 벡터는 인간 아데노바이러스 5형의 섬유 단백질 말단의 돌기 도메인을 침팬지 아데노바이러스 혈청형 6의 돌기 유전자로 치환하고, 인간 아데노바이러스 5형의 테일(Tail) 및 사프트(Shaft) 도메인과 침팬지 아데노바이러스 혈청형 6의 돌기 도메인으로 조합된 섬유단백질을 포함하는 것이 바람직하나 이에 한정되지 아니한다.In another embodiment of the present invention, the adenovirus vector replaces the terminal domain of the fiber protein of human adenovirus type 5 with the spine gene of chimpanzee adenovirus serotype 6, and the tail of human adenovirus type 5 and a fibrous protein combined with a Shaft domain and a protrusion domain of chimpanzee adenovirus serotype 6, but is not limited thereto.
본 발명의 또 다른 구현예에 있어서, 상기 침팬지 아데노바이러스 혈청형 6의 돌기 유전자에 의하여 발현된 단백질은 서열번호 7의 아미노산 서열을 포함하는 것이 바람직하나 이에 한정되지 아니한다.In another embodiment of the present invention, the protein expressed by the chimpanzee adenovirus serotype 6 protrusion gene preferably includes the amino acid sequence of SEQ ID NO: 7, but is not limited thereto.
또 본 발명은 사기 본 발명의 재조합 발현 벡터를 유효성분으로 포함하는 사스-코로나바이러스-2(SARS-CoV-2)의 감염증 예방 백신 조성물을 제공한다. In addition, the present invention provides a vaccine composition for preventing infection of SARS-CoV-2, comprising the recombinant expression vector of the present invention as an active ingredient.
본 발명의 일 구현예에 있어서, 상기 백신은 SARS-CoV-2 바이러스 초기주, 델타 바이러스, 베타 및 오미크론 바이러스 변이주로 구성된 군으로부터 선택된 하나 이상의 바이러스에 효과를 가지는 것이 바람직하나 이에 한정되지 아니한다.In one embodiment of the present invention, the vaccine preferably has an effect on one or more viruses selected from the group consisting of SARS-CoV-2 virus initial strain, delta virus, beta and Omicron virus mutant strains, but is not limited thereto.
또한, 본 발명의 상기 벡터는 포유동물 세포를 감염시킬 수 있는 것이 바람직하나 이에 한정되지 아니한다.In addition, the vector of the present invention is preferably capable of infecting mammalian cells, but is not limited thereto.
또한 본 발명은 상기 본 발명의 베타 변이 유래의 S 유전자 또는 S유전자에 N 유전자 일부를 연결하여 포함하는 키메릭아데노바이러스 벡터로 형질전환된 숙주세포를 제공한다.In addition, the present invention provides a host cell transformed with a chimeric adenovirus vector comprising the S gene derived from the beta mutation of the present invention or a part of the N gene linked to the S gene.
또한 본 발명은 상기 본 발명의 형질전환 세포를 이용하여 재조합 바이러스를 생산하는 방법을 제공한다.In addition, the present invention provides a method for producing a recombinant virus using the transformed cell of the present invention.
또한, 본 발명은 상기 본 발명에 의해 얻어진 2종의 재조합 키메릭아데노바이러스를 동물모델에서 백신으로 사용하는 방법을 제공한다.In addition, the present invention provides a method of using the two recombinant chimeric adenoviruses obtained by the present invention as a vaccine in an animal model.
본 발명은 상기 방법에 의해 생산된 재조합 키메릭아데노바이러스를 사용하여 마우스 면역 및 바이러스 중화효능을 제공한다.The present invention provides mouse immunity and virus neutralization efficacy using the recombinant chimeric adenovirus produced by the above method.
본 발명은 상기 방법에 의해 생산된 재조합 키메릭아데노바이러스에 따른 특이적 T 세포의 면역반응에 대한 효능을 제공한다.The present invention provides efficacy for immune response of specific T cells according to the recombinant chimeric adenovirus produced by the above method.
본 발명은 상기 방법에 의해 생산된 재조합 키메릭아데노바이러스를 면역 후 초기주의 방어 효능을 제공한다.The present invention provides the protective efficacy of the initial strain after immunization with the recombinant chimeric adenovirus produced by the above method.
또 본 발명은 상기 본 발명의 키메릭 아데노바이러스 벡터를 유효성분으로 포함하는 사스-코로나바이러스-2(SARS-CoV-2)의 감염증 예방 또는 치료용 백신 조성물을 제공한다. In addition, the present invention provides a vaccine composition for preventing or treating an infection of SARS-CoV-2, comprising the chimeric adenovirus vector of the present invention as an active ingredient.
본 발명은 사스-코로나바이러스-2 감염증 예방 백신 조성물로써, 초기주뿐만 아니라 베타, 델타, 오미크론 변이주감염에 대한 중증완화 또는 예방할 수 있는 백신인 것이 바람직하나 이에 한정되지 아니한다.The present invention is a vaccine composition for preventing SARS-coronavirus-2 infection, preferably a vaccine capable of mitigating or preventing the infection of beta, delta, and omicron mutant strains as well as the initial strain, but is not limited thereto.
이하 본 발명을 설명한다. The present invention will be described below.
본 발명자들은 ChimAd 벡터를 전달체로 하여 베타 변이주의 S 단백질을 기반으로 초기주 뿐만 아니라 우세 변종인 델타 변이주에 대해 유효한 백신조성물을 개발하였고, 이와 더불어 T 세포 면역반응을 극대화 하기위해 N 단백질과 연결된 형태의 S단백질에 대해 중화항체가, 세포성 면역원성 및 바이러스 감염에 대한 방어능 평가를 통해 본 발명을 완성하였다.The present inventors developed a vaccine composition effective against not only the initial strain but also the dominant strain, the delta mutant strain, based on the S protein of the beta mutant strain using the ChimAd vector as a delivery vehicle, and in addition, to maximize the T cell immune response, the form linked to the N protein The present invention was completed through the evaluation of cellular immunogenicity and protective ability against viral infection of the neutralizing antibody against the S protein of .
본 발명은 사스-코로나바이러스-2 감염증 예방 백신 조성물로써, 베타 변이주의 S 단백질이 인코딩된 키메릭아데노바이러스 벡터를 제공한다.The present invention provides a chimeric adenovirus vector encoding the beta-mutant S protein as a vaccine composition for preventing SARS-coronavirus-2 infection.
S단백질의 융합 후(Post-fusion) 형태로의 구조변화를 저해하기 위해 초기 주 기준 614번의 글라이신, 682~685번 잔기의 산성 아미노산으로의 치환 및 986, 987번의 연속된 프롤린 치환 돌연변이가 도입되었다. [Pallesen, Jesper, et al. "Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen." Proceedings of the National Academy of Sciences 114.35 (2017): E7348-E7357. Bangaru, Sandhya, et al. "Structural analysis of full-length SARS-CoV-2 spike protein from an advanced vaccine candidate." Science 370.6520 (2020): 1089-1094. Plante, Jessica A., et al. "Spike mutation D614G alters SARS-CoV-2 fitness." Nature 592.7852 (2021): 116-121.]In order to inhibit the post-fusion structural change of the S protein, substitution of glycine at 614, residues 682 to 685 with acidic amino acids, and consecutive proline substitution mutations at 986 and 987 were introduced based on the initial week. . [Pallesen, Jesper, et al. "Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen." Proceedings of the National Academy of Sciences 114.35 (2017): E7348-E7357. Bangaru, Sandhya, et al. "Structural analysis of full-length SARS-CoV-2 spike protein from an advanced vaccine candidate." Science 370.6520 (2020): 1089-1094. Plante, Jessica A., et al. "Spike mutation D614G alters SARS-CoV-2 fitness." Nature 592.7852 (2021): 116-121.]
또한, 본 발명은 상기 베타 변이 S 단백질에 T-세포 면역을 증대할 수 있는 N 단백질 일부를 포함하여 S 단백질과 N 단백질이 융합된 단백질이 인코딩된 키메릭아데노바이러스 벡터를 제공한다. In addition, the present invention provides a chimeric adenoviral vector encoding a protein in which the S protein and the N protein are fused, including a portion of the N protein capable of enhancing T-cell immunity to the beta-mutant S protein.
변이주 S 단백질과의 융합에 있어 구조적으로 불안정한 영역 및 과인산화 도메인을 제외하였고, T cell epitope을 포함하는 NTD영역과, T cell epitope을 가짐과 동시에 이합체(Dimer) 형성을 통해 구조적 안정성을 가지는 CTD를 연결자(linker)를 통해 S단백질에 연결하였다 [Peng, Y., Mentzer, A. J., Liu, G., Yao, X., Yin, Z., Dong, D., ... & Dong, T. (2020). Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nature immunology, 21(11), 1336-1345, Zinzula, L., Basquin, J., Bohn, S., Beck, F., Klumpe, S., Pfeifer, G., ... & Baumeister, W. (2021). High-resolution structure and biophysical characterization of the nucleocapsid phosphoprotein dimerization domain from the Covid-19 severe acute respiratory syndrome coronavirus 2. Biochemical and biophysical research communications, 538, 54-62.] 최종적으로, NP의 NTD (44-180) 및 CTD(247-364)가 (GGGGS)3 linker를 통해 S단백질의 양 말단에 연결하였다.In fusion with the mutant S protein, structurally unstable regions and hyperphosphorylation domains were excluded, and the NTD region containing the T cell epitope and the CTD having structural stability through dimer formation while having the T cell epitope Linked to S protein via a linker [Peng, Y., Mentzer, A. J., Liu, G., Yao, X., Yin, Z., Dong, D., ... & Dong, T. ( 2020). Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nature immunology, 21(11), 1336-1345, Zinzula, L., Basquin, J., Bohn, S., Beck, F., Klumpe, S., Pfeifer, G., ... & Baumeister, W. (2021). High-resolution structure and biophysical characterization of the nucleocapsid phosphoprotein dimerization domain from the Covid-19 severe acute respiratory syndrome coronavirus 2. Biochemical and biophysical research communications, 538, 54-62.] Finally, the NTD of NP (44-180) and CTD(247-364) was linked to both ends of the S protein via the (GGGGS)3 linker.
“키메라 단백질” 또는 "키메라 폴리펩타이드"란 일반적으로 복수의 단백질 성분으로 구성된 단백질을 말하며, 각각 아미노 말단 (N 말단, NTD) 및 카르복시 말단 (C 말단, CTD)의 결합을 통해 연결된 연속의 폴리펩타이드를 의미한다.“Chimeric protein” or “chimeric polypeptide” generally refers to a protein composed of multiple protein components, each of which is a series of polypeptides linked through amino-terminal (N-terminal, NTD) and carboxy-terminal (C-terminal, CTD) bonds. means
본 발명에서는 아미노 말단에 N 폴리펩타이드(44번부터 180번)와 카르복시 말단 N 폴리펩타이드(247번부터 364번) 사이에 베타변이주의 S 폴리펩타이드가 포함된 것을 키메라 단백질 또는 폴리펩타이드(NP(NTD)-Spike-NP(CTD))라 한다.In the present invention, a chimeric protein or polypeptide (NP (NTD )-Spike-NP (CTD)).
본 발명의 상기 펩타이드는 재조합 아데노바이러스 벡터, RNA, 재조합 단백질, 비바이러스벡터 등의 다양한 전달체에서 발현되는 것이 바람직하고, 키메릭아데노바이러스 벡터에 삽입되는 것이 더욱 바람직하나, 이에 한정되지 않는다. The peptide of the present invention is preferably expressed in various delivery vehicles such as recombinant adenovirus vectors, RNA, recombinant proteins, and non-viral vectors, and more preferably inserted into chimeric adenovirus vectors, but is not limited thereto.
본 발명의 상기 키메릭아데노바이러스 벡터는 특허 출원 상태이며(대한민국 특허출원번호:10-2022-0034885) 구체적인 설명은 해당 특허를 참고하고, 본 명세서에서는 생략한다.The chimeric adenoviral vector of the present invention is in the state of patent application (Korean Patent Application No.: 10-2022-0034885), and reference is made to the patent for specific descriptions, and is omitted herein.
상기 폴리뉴클레오타이드 S를 포함하는 키메릭 아데노바이러스는 ChimAd-CV, 상기 NP(NTD)-Spike-NP(CTD)를 포함하는 키메릭 아데노바이러스는 ChimAd-CVN 라고 명한다. The chimeric adenovirus containing the polynucleotide S is called ChimAd-CV, and the chimeric adenovirus containing the NP(NTD)-Spike-NP(CTD) is called ChimAd-CVN.
본 발명의 상기 유전자들을 포함된 키메릭아데노바이러스 벡터가 형질전환된 세포는 HEK293, PERC6 또는 911 세포를 포함하는 포유 동물 세포인 것이 바람직하고, HEK293 세포가 더욱 바람직하나, 이에 한정되지 않는다.Cells transfected with the chimeric adenovirus vector containing the genes of the present invention are preferably mammalian cells including HEK293, PERC6 or 911 cells, more preferably HEK293 cells, but are not limited thereto.
예컨대, 본 발명을 위해 적합한 아데노바이러스 생산 세포는 복제불능 아데노바이러스(replication defective adenovirus)의 E1 및 E3 유전자 결손 부위를 상보성 전환(trans-complementation)하여 바이러스 생산이 가능한 세포여야 하며, 상기 바이러스 초기전사유전자들로 형질 전환시킨 세포들이 많이 개발되고 있다. E1 및 E3 유전자가 도입되기 바람직한 세포에는 A549 (carcinomic human alveolar basal epithelial cell), HeLa(Human Cervical cancer cell), CHO(Chinese hamster ovary cell), MRC5 (human lung fibroblast), 그리고 PC12(rat pheochromocytoma cell)등이나, 이에 한정되지 아니한다.For example, an adenovirus-producing cell suitable for the present invention should be a cell capable of producing a virus by trans-complementation of the E1 and E3 gene deletion sites of a replication defective adenovirus, and the viral early transcription gene A number of cells transformed with . Cells into which the E1 and E3 genes are preferred include A549 (carcinomic human alveolar basal epithelial cell), HeLa (Human cervical cancer cell), CHO (Chinese hamster ovary cell), MRC5 (human lung fibroblast), and PC12 (rat pheochromocytoma cell) etc., but not limited thereto.
본 발명은 ChimAd-CV 및 ChimAd-CVN 바이러스 생산 시 1x 106 cells/ml cell density의 숙주세포에 2 MOI의 역가로 감염시켜 48시간 후 세포를 회수하는 것이 바람직하나 이에 한정되지 아니한다.In the present invention, when producing ChimAd-CV and ChimAd-CVN viruses, it is preferable to infect host cells at a cell density of 1x10 6 cells/ml with a titer of 2 MOI and recover the cells after 48 hours, but is not limited thereto.
상기 회수한 세포로부터 바이러스를 용출하기 위해 0.5 % Tween20과 숙주세포 DNA 분해를 유도하고 자 20 U/ml benzonase를 4시간 처리함이 바람직하나 이에 한정되지 아니한다.In order to elute the virus from the recovered cells, 0.5% Tween20 and 20 U/ml benzonase to induce host cell DNA degradation are preferably treated for 4 hours, but are not limited thereto.
바이러스 용출 가능한 계면활성제로는 Tween20 이외에도 Tween80, Triton X-100, Zwittergent™3-14 그리고 CHAPS등이 있으나, 이에 한정되지 아니한다.In addition to Tween20, surfactants capable of eluting viruses include Tween80, Triton X-100, Zwittergent™3-14, and CHAPS, but are not limited thereto.
본 발명은 상기 방법으로 생산된 ChimAd-CV 및 ChimAd-CVN을 제공한다.The present invention provides ChimAd-CV and ChimAd-CVN produced by the above method.
본 발명에서 생산된 ChimAd-CV 및 ChimAd-CVN 백신 조성물은 초기주와 베타변이주, 델타변이주 에 대한 우수한 중화항체 역가를 제공한다.The ChimAd-CV and ChimAd-CVN vaccine compositions produced in the present invention provide excellent neutralizing antibody titers against the initial strain, beta mutant strain and delta mutant strain.
본 발명의 구현예에 따라 생산된 ChimAd-CV 및 ChimAd-CVN 백신 조성물은 베타 변이주 기반 슈도바이러스에 대한 우수한 중화항체 역가를 제공한다.The ChimAd-CV and ChimAd-CVN vaccine compositions produced according to an embodiment of the present invention provide excellent neutralizing antibody titers against pseudoviruses based on beta mutants.
본 발명은 생산된 ChimAd-CV 및 ChimAd-CVN 백신 조성물은 특이적 T 세포의 면역반응에 대한 효능을 제공한다.The ChimAd-CV and ChimAd-CVN vaccine compositions produced in the present invention provide efficacy against the immune response of specific T cells.
본 발명의 구현예에 따라 생산된 ChimAd-CV와 ChimAd-CVN의 특이적 T 세포의 면역반응을 확인하고자 ELISPOT (Enzyme-Linked ImmunoSpot, ELISPOT) 실험 기법을 사용하였다. ChimAd-CV 또는 ChimAd-CVN의 면역이 유도된 마우스 비장세포를 분리하여 S 단백질 또는 NP의 펩타이드에 대한 면역반응을 확인하였다. 그 결과, ChimAd-CVN이 ChimAd-CV 보다 2배이상의 우수한 세포성 면역능을 확인함에 따라 N 단백질을 연결함에 따라, 보다 높은 T-cell 면역이 유도되는 것으로 사료된다.[문헌 참조 시, 아데노바이러스 벡터 전달체 또는 DNA를 이용한 COVID-19 백신의 T-세포 면역 반응 결과, ChimAd-CV와 ChimAd-CVN 의 prime 4주차 결과와 유사함을 확인하였다(Graham, Simon P., et al. "Evaluation of the immunogenicity of prime-boost vaccination with the replication-deficient viral vectored COVID-19 vaccine candidate ChAdOx1 nCoV-19." npj Vaccines 5.1 (2020): 1-6. Seo, Yong Bok, et al. "Soluble spike DNA vaccine provides long-term protective immunity against SARS-CoV-2 in mice and nonhuman primates." Vaccines 9.4 (2021): 307.). In order to confirm the immune response of ChimAd-CV and ChimAd-CVN-specific T cells produced according to an embodiment of the present invention, ELISPOT (Enzyme-Linked ImmunoSpot, ELISPOT) experimental technique was used. Mouse splenocytes immunized with ChimAd-CV or ChimAd-CVN were isolated to confirm an immune response to S protein or NP peptide. As a result, as ChimAd-CVN confirmed cellular immunity more than twice as good as ChimAd-CV, it is thought that higher T-cell immunity is induced by linking N protein. [Referring to the literature, adenoviral vector As a result of the T-cell immune response of the COVID-19 vaccine using the delivery vehicle or DNA, it was confirmed that the prime 4-week results of ChimAd-CV and ChimAd-CVN were similar (Graham, Simon P., et al. "Evaluation of the immunogenicity of prime-boost vaccination with the replication-deficient viral vectored COVID-19 vaccine candidate ChAdOx1 nCoV-19." npj Vaccines 5.1 (2020): 1-6. Seo, Yong Bok, et al. "Soluble spike DNA vaccine provides long- term protective immunity against SARS-CoV-2 in mice and nonhuman primates." Vaccines 9.4 (2021): 307.).
높은 초기 T-세포성 면역반응은 그 자체로 코로나바이러스에 대한 중증 완화의 역할을 한다고 알려져 있고, 일각에서는 N 단백질에 의해 형성된 항-N단백질 항체가 직접적인 바이러스 중화에는 관련이 없으나 N단백질 특이적인 T-세포 면역의 증대가 중화항체 역가와 상호관계가 있다. [Tan, A. T., Linster, M., Tan, C. W., Le Bert, N., Chia, W. N., Kunasegaran, K., ... & Bertoletti, A. (2021). Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients. Cell reports, 34(6), 108728. Wyllie, D., Jones, H. E., Mulchandani, R., Trickey, A., Taylor-Phillips, S., Brooks, T., ... & Oliver, I. (2021). SARS-CoV-2 responsive T cell numbers and anti-Spike IgG levels are both associated with protection from COVID-19: A prospective cohort study in keyworkers. MedRxiv, 2020-11. Ni, L., Ye, F., Cheng, M. L., Feng, Y., Deng, Y. Q., Zhao, H., ... & Dong, C. (2020). Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals. Immunity, 52(6), 971-977]A high initial T-cell immune response is known to play a role in mitigating the severity of the coronavirus by itself, and in some cases, anti-N protein antibodies formed by N protein are not related to direct virus neutralization, but N protein specific T -Enhancement of cellular immunity correlates with neutralizing antibody titer. [Tan, A. T., Linster, M., Tan, C. W., Le Bert, N., Chia, W. N., Kunasegaran, K., ... & Bertoletti, A. (2021). Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients. Cell reports,  34(6), 108728. Wyllie, D., Jones, H. E., Mulchandani, R., Trickey, A., Taylor-Phillips, S., Brooks, T., ... & Oliver, I. ( 2021). SARS-CoV-2 responsive T cell numbers and anti-Spike IgG levels are both associated with protection from COVID-19: A prospective cohort study in keyworkers. MedRxiv, 2020-11. Ni, L., Ye, F., Cheng, M. L., Feng, Y., Deng, Y. Q., Zhao, H., ... & Dong, C. (2020). Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals. Immunity, 52(6), 971-977]
본 발명은 상기 방법으로 생산된 ChimAd-CV 및 ChimAd-CVN 백신 조성물은 초기주에 대한 방어 효능을 제공한다. In the present invention, the ChimAd-CV and ChimAd-CVN vaccine compositions produced by the above method provide protective efficacy against early strains.
본 발명의 구현예에 따라 생산된 ChimAd-CV 와 ChimAd-CVN로 면역 및 바이러스 감염 후 몸무게 변화와 생존율을 확인하였다. 초기주 감염에 대해 ChimAd-CV 와 ChimAd-CVN 접종한 실험군 모두 100% 생존율을 보이는 것으로 보아 우수한 방어능을 확인하였다. Body weight change and survival rate after immunization and viral infection with ChimAd-CV and ChimAd-CVN produced according to an embodiment of the present invention were confirmed. Both the experimental groups inoculated with ChimAd-CV and ChimAd-CVN showed a 100% survival rate against the initial strain infection, confirming the excellent defense ability.
본 발명을 통하여 알 수 있는 바와 같이, 본 발명은 SARS-CoV-2 바이러스에 대한 예방백신으로써 우수한 효능을 제공할 수 있다. 본 발명에서 키메릭 아데노바이러스 벡터 기반의 베타변이주 S 서열 또는 N과 융합된 S 서열을 포함하는 백신 조성물을 개발하였다. 초기주 및 변이주(베타, 델타, 오미크론)에 대한 중화항체가가 높아 우수한 교차면역원성을 가지고 있으며, N서열이 포함된 백신은 T-세포면역능이 극대화되는 효능을 제공하였다. 또한, 초기주에 대한 방어능이 우수함에 따라 SARS-CoV-2 예방 백신으로 적용 가능할 것으로 사료된다. As can be seen through the present invention, the present invention can provide excellent efficacy as a preventive vaccine against the SARS-CoV-2 virus. In the present invention, a vaccine composition comprising a beta mutant S sequence or an S sequence fused with N based on a chimeric adenovirus vector was developed. It has excellent cross-immunogenicity with high neutralizing antibodies against the initial strain and mutant strain (beta, delta, omicron), and the vaccine containing the N sequence provided the efficacy of maximizing T-cell immunity. In addition, it is considered that it can be applied as a preventive vaccine for SARS-CoV-2 as it has excellent defense against early strains.
도 1은 유전자 합성을 통해 확보한 코로나바이러스 유전자를 키메릭아데노바이러스(ChimAd) 벡터에 Gibson assembly 유전자 합성 방법을 이용하여 ChimAd-CV, ChimAd-CVN을 클로닝하는 모식도이다.1 is a schematic diagram of cloning ChimAd-CV and ChimAd-CVN using the Gibson assembly gene synthesis method in a chimeric adenovirus (ChimAd) vector obtained through gene synthesis.
도 2은 BALB/c female 마우스를 이용하여 ChimAd-CV, ChimAd-CVN에 의해 유도된 바이러스 결합 항체 역가를 초기주 S단백질에 대한 결과는 A로, 베타변이주 S단백질 결과는 B로 나타낸다.Figure 2 shows the virus-binding antibody titers induced by ChimAd-CV and ChimAd-CVN using BALB/c female mice as A for the initial strain S protein and as B for the beta mutant S protein.
도 3는 BALB/c female 마우스를 이용하여 ChimAd-CV, ChimAd-CVN의에 의해 유도된 바이러스 중화항체 역가를 베타 변이주(B.1.351 variant type)의 슈도바이러스을 이용한 중화항체 측정 결과로 나타낸다. A-B. 사스-코로나바이러스-2 (Wuhan-Hu-1) 슈도바이러스 중화항체 시험, C-D. 베타변이주 (B.1.351) 슈도바이러스 중화항체 시험.Figure 3 shows the neutralizing antibody titers induced by ChimAd-CV and ChimAd-CVN using BALB/c female mice as neutralizing antibody measurement results using pseudovirus of beta mutant (B.1.351 variant type). A-B. SARS-coronavirus-2 (Wuhan-Hu-1) pseudovirus neutralizing antibody test, C-D. Beta mutant strain (B.1.351) Pseudovirus neutralizing antibody test.
도 4는 BALB/c female 마우스를 이용하여 ChimAd-CV, ChimAd-CVN의 S 펩타이드를 자극항원으로 사용한 세포성 면역결과(A, B)와 NP 펩타이드를 자극항원으로 사용한 세포성 면역결과(C, D)를 나타낸다.Figure 4 shows the results of cellular immunity (A, B) using S peptides of ChimAd-CV and ChimAd-CVN as stimulating antigens using BALB/c female mice and the results of cellular immunization (C, B) using NP peptides as stimulating antigens. D).
도 5는 hACE2 transgenic female 마우스를 이용하여 ChimAd-CV, ChimAd-CVN애 의해 유도된 바이러스 결합 항체 역가를 초기주 S단백질에 대한 결과는 A로, 델타변이주 S단백질 결과는 B로 나타낸다.Figure 5 shows the virus-binding antibody titers induced by ChimAd-CV and ChimAd-CVN using hACE2 transgenic female mice, with A for the initial strain S protein and B for the delta mutant S protein.
도 6은 hACE2 transgenic female 마우스를 이용하여 ChimAd-CV, ChimAd-CVN에 의해 유도된 바이러스 중화항체 역가를 확인한 결과로 초기주의 FRNT를 이용한 중화항체 측정 결과(A, B) 로 나타낸다. 또한, 델타 변이주의 FRNT를 이용한 중화항체 측정 결과(C, D) 로 나타낸다. A-B. 사스-코로나바이러스-2(Wuhan-Hu-1) FRNT 중화항체 시험, C-D. 델타변이주 (B.1.617.2) FRNT 중화항체 시험.6 shows the results (A, B) of neutralizing antibody measurement using FRNT of the initial strain as a result of confirming the viral neutralizing antibody titer induced by ChimAd-CV and ChimAd-CVN using hACE2 transgenic female mice. In addition, the neutralizing antibody measurement results (C, D) using FRNT of the delta mutant strain are shown. A-B. SARS-coronavirus-2 (Wuhan-Hu-1) FRNT neutralizing antibody test, C-D. Delta mutant strain (B.1.617.2) FRNT neutralizing antibody test.
도 7은 hACE2 transgenic female 마우스를 이용하여 ChimAd-CV, ChimAd-CVN에 의해 면역된 마우스 동물모델에서 초기주 감염에 대해 ChimAd-CV는 ChimAd-CVN의 몸무게 비교결과와 사망률을 나타낸다. Figure 7 shows the results of comparison of body weights and mortality rates between ChimAd-CV and ChimAd-CVN for initial infection in mouse animal models immunized by ChimAd-CV and ChimAd-CVN using hACE2 transgenic female mice.
도 8은 사스-코로나바이러스-2(Wuhan-Hu-1)로 공격시험 후 폐에서 검출된 코로나바이러스 역가를 나타낸다. Figure 8 shows the coronavirus titers detected in the lungs after challenge with SARS-coronavirus-2 (Wuhan-Hu-1).
도 9은 BALB/c female 마우스를 이용하여 ChimAd-CV의 접종 용량에 따라 유도된 결합항체 역가를 사스-코로나바이러스-2의 S단백질에 대한 결과로 나타낸다.Figure 9 shows the antibody titer induced according to the inoculation dose of ChimAd-CV using BALB/c female mice as a result for the S protein of SARS-coronavirus-2.
도 10은 BALB/c female 마우스를 이용하여 ChimAd-CV의 접종용량에 의해 유도된 바이러스 중화항체 역가를 다양한 변이바이러스(우한, 베타변이, 델타, 오미크론(BA.1)) VLP(Virus Like particle)에 대한 측정 결과(A~D) 로 나타낸다.Figure 10 shows the virus neutralizing antibody titer induced by the inoculation dose of ChimAd-CV using BALB/c female mice with various mutant viruses (Wuhan, beta mutant, delta, Omicron (BA.1)) VLP (Virus Like Particle ) is represented by the measurement results (A to D).
도 10에서 A는 사스-코로나바이러스-2 (Wuhan-Hu-1) VLP 중화항체 시험, B는 베타변이주 (B.1.351) VLP 중화항체 시험, C는 델타변이주(B.1.617.2) VLP 중화항체 시험, D는 오미크론(BA.1) VLP 중화항체 시험.In FIG. 10, A is a SARS-coronavirus-2 (Wuhan-Hu-1) VLP neutralizing antibody test, B is a beta mutant (B.1.351) VLP neutralizing antibody test, and C is a delta mutant (B.1.617.2) VLP neutralization. Antibody test, D is Omicron (BA.1) VLP neutralizing antibody test.
도 11은 BALB/c female 마우스를 이용하여 ChimAd-CV의 접종용량에 의해 유도된 세포성면역 결과로 S 펩타이드를 자극항원으로 사용하였다. 11 shows the results of cellular immunity induced by the inoculation dose of ChimAd-CV using BALB/c female mice, and S peptide was used as a stimulating antigen.
이하, 실시예를 통하여 본 발명을 더욱 상세하게 설명한다. 단 하기 실시예는 발명을 보다 구체적으로 설명하기 위한 것으로, 본 발명의 범위는 하기 실시예에 의해 제한되지 아니한다.Hereinafter, the present invention will be described in more detail through examples. However, the following examples are for explaining the invention in more detail, and the scope of the present invention is not limited by the following examples.
실시예 1: SARS-CoV-2 S 단백질, N 단백질을 암호화하는 유전자 변형Example 1: Genetic modification encoding SARS-CoV-2 S protein, N protein
코로나바이러스 구조단백질을 이용하여 백신을 개발하기 위하여, 아미노산 치환 등의 변형을 포함한 Spike 단백질 및 N단백질의 NTD 및 CTD를 포함하는 S 폴리펩타이드 합성 유전자를 2종을 확보하였다. (표 1 참조) In order to develop a vaccine using the coronavirus structural protein, two S polypeptide synthesis genes including NTD and CTD of Spike protein and N protein including modifications such as amino acid substitution were secured. (See Table 1)
구체적으로, CV유전자의 경우, 베타변이 S단백질의 초기 주 기준 614번의 글라이신, 682~685번 잔기의 산성 아미노산으로의 치환 및 986, 987번의 연속된 프롤린 치환 돌연변이가 도입되었다. [Pallesen, Jesper, et al. "Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen." Proceedings of the National Academy of Sciences 114.35 (2017): E7348-E7357. Bangaru, Sandhya, et al. "Structural analysis of full-length SARS-CoV-2 spike protein from an advanced vaccine candidate." Science 370.6520 (2020): 1089-1094. Plante, Jessica A., et al. "Spike mutation D614G alters SARS-CoV-2 fitness." Nature 592.7852 (2021): 116-121.]. Specifically, in the case of the CV gene, substitution of glycine at 614, residues 682 to 685 with acidic amino acids, and consecutive proline substitution mutations at 986 and 987 of the initial strain of the beta-mutant S protein were introduced. [Pallesen, Jesper, et al. "Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen." Proceedings of the National Academy of Sciences 114.35 (2017): E7348-E7357. Bangaru, Sandhya, et al. "Structural analysis of full-length SARS-CoV-2 spike protein from an advanced vaccine candidate." Science 370.6520 (2020): 1089-1094. Plante, Jessica A., et al. "Spike mutation D614G alters SARS-CoV-2 fitness." Nature 592.7852 (2021): 116-121.].
CVN유전자의 경우 N단백질의 44번부터 180번, 247번부터 364번이 각각 Spike 단백질의 N말단 및 C말단에 (GGGGS)3 linker를 통해 연결되었으며, 이 때 trRosetta, Alphafold 와 같은 prediction tool을 통해 추가적인 구조 예측을 진행하였다. [Du, Zongyang, et al. "The trRosetta server for fast and accurate protein structure prediction." Nature Protocols (2021): 1-18. Jumper, John, et al. "Highly accurate protein structure prediction with AlphaFold." Nature 596.7873 (2021): 583-589.] In the case of the CVN gene, positions 44 to 180 and 247 to 364 of the N protein were linked to the N-terminus and C-terminus (GGGGS) of the Spike protein through 3 linkers, respectively, and at this time, through prediction tools such as trRosetta and Alphafold. Further structural predictions were made. [Du, Zongyang, et al. "The trRosetta server for fast and accurate protein structure prediction." Nature Protocols (2021): 1-18. Jumper, John, et al. "Highly accurate protein structure prediction with AlphaFold." Nature 596.7873 (2021): 583-589.]
실시예 2: SARS-CoV-2 백신물질로 사용될 유전자 합성 Example 2: Gene synthesis to be used as SARS-CoV-2 vaccine material
코로나바이러스 구조단백질을 이용하여 백신을 개발하기 위하여, 구조 단백질 중 spike 단백질과 Nucleocapsid 단백질을 암호화하는 합성 유전자를 확보하였다. 각각의 백신후보는 표 1과 같이 코로나바이러스의 B.1.351 유래 S 단백질, Wuhan-Hu-1 유래 N 단백질을 선정하여 아데노바이러스에서 발현이 용이하도록 codon 최적화를 하였다. S 단백질을 포함하는 region을 CV, N 단백질 일부와 S 단백질을 융합하는 키메라 형태를 CVN으로 명명함.In order to develop a vaccine using coronavirus structural proteins, synthetic genes encoding spike and nucleocapsid proteins among structural proteins were secured. As shown in Table 1, for each vaccine candidate, S protein derived from coronavirus B.1.351 and N protein derived from Wuhan-Hu-1 were selected and codon optimized for easy expression in adenovirus. The region containing the S protein is called CV, and the chimera form in which a part of the N protein is fused with the S protein is called CVN.
Region Region Accession no.Accession no.
CVCV Spike surface glycoprotein Spike surface glycoprotein QUT64557(B.1.351)QUT64557 (B.1.351)
CVNCVN NP(NTD)-Spike-NP(CTD)NP(NTD)-Spike-NP(CTD) QUT64557(B.1.351), MN908947(Wuhan-Hu-1)QUT64557 (B.1.351), MN908947 (Wuhan-Hu-1)
실시예 3: SARS-CoV-2 유전자를 포함한 키메릭 아데노바이러스 클로닝Example 3: Cloning of Chimeric Adenovirus Containing the SARS-CoV-2 Gene
키메릭 아데노바이러스 벡터인 ChimAd벡터에 상기 합성한 SARS-CoV-2 백신 항원 2종을 클로닝하기 위하여, ChimAd 벡터의 plasmid DNA를 주형으로 하여 GOI binding region의 유전자 일부를 제외하고 PCR 증폭하여 그 증폭된 산물(약 33.4 Kb)을 gel extraction 과정을 거쳐 정제하였다. 클로닝을 위한 삽입 유전자는 ChimAd 벡터에서 GOI binding region 5' 및 3' 말단과 동일한 서열(homologous region, 약 20bp)을 포함하도록 PCR 증폭하기위해 하기 표2의 프라이머를 이용하여 얻은 증폭산물 CV (약 3.8 Kb)와 CVN (약 4.7 Kb)를 확보한 후 앞서 DNA 정제한 벡터 PCR 증폭산물과 각각 gibson assembly 반응을 통해 결합하여 최종적으로 ChimAd-CV, ChimAd-CVN plasmid 2종을 확보하였다(도 1).In order to clone the two SARS-CoV-2 vaccine antigens synthesized above into ChimAd vector, which is a chimeric adenovirus vector, PCR amplification was performed excluding part of the gene of the GOI binding region using the plasmid DNA of the ChimAd vector as a template, and the amplified The product (about 33.4 Kb) was purified through a gel extraction process. The inserted gene for cloning was PCR amplified to include the same sequence (homologous region, about 20 bp) as the 5' and 3' ends of the GOI binding region in the ChimAd vector, and the CV of the amplification product obtained using the primers in Table 2 below (about 3.8 Kb) and CVN (about 4.7 Kb), and then combined with the previously purified DNA PCR amplification products through a gibson assembly reaction, respectively, two types of ChimAd-CV and ChimAd-CVN plasmids were finally obtained (Fig. 1).
Primer namePrimer name Sequence (5' to 3' direction)Sequence (5' to 3' direction) PCR productPCR products
ChimAd vectorChimAd vector F: CTAAGCTTCTAGATAAGATATCCGATCCAC (서열번호 8)F: CTAAGCTTCTAGATAAGATATCCGATCCAC (SEQ ID NO: 8) 33.4 Kb33.4 Kb
R: CGCGTCGACGGTACCAGATCTCTAGCGGATC (서열번호 9)R: CGCGTCGACGGTACCAGATCTCTAGCGGATC (SEQ ID NO: 9)
CV regionCV region F: GATCTGGTACCGTCGACGCGATGTTTGTGTTTCTGGTGCTG (서열번호 10)F: GATCTGGTACCGTCGACGCGATGTTTGTGTTTCTGGTGCTG (SEQ ID NO: 10) 3.8 Kb3.8 Kb
R: TATCTTATCTAGAAGCTTAGTCAGGTATAGTGCAGTTTCAC (서열번호 11)R: TATCTTATCTAGAAGCTTAGTCAGGTATAGTGCAGTTTCAC (SEQ ID NO: 11)
CVN regionCVN region F: GATCTGGTACCGTCGACGCGATGGGCCTGCCCAACAACAC (서열번호 12)F: GATCTGGTACCGTCGACGCGATGGGCCTGCCCAACAACAC (SEQ ID NO: 12) 4.7 Kb4.7 Kb
R: TATCTTATCTAGAAGCTTAGTCAGGGGAAGGTCTTGTAGGC (서열번호 13)R: TATCTTATCTAGAAGCTTAGTCAGGGGAAGGTCTTGTAGGC (SEQ ID NO: 13)
실시예 4: 키메릭아데노바이러스 기반 코로나 백신후보물질의 생산 및 정제Example 4: Production and purification of chimeric adenovirus-based corona vaccine candidates
본 발명에서 제작한 ChimAd-CV, ChimAd-CVN plasmid를 이용하여 키메릭아데노바이러스 기반 코로나 백신후보물질을 생산하기 위하여, 각 plasmid를 PacⅠ 제한효소(NEB사, R0547L)를 처리해 아데노바이러스 형성에 불필요한 pBR322 복제 개시점 및 카나마이신 저항성 유전자 서열 등을 절단하여 제거하고, HEK293 세포에 Lipofectamine 2000 (Invitrogen사, 11668027)을 사용하여 형질 전환하여 온도 37℃, CO2 분압 5.0 %가 유지되는 배양기에서 10-20일간 배양한 후 primary virus stock을 확보하였다.In order to produce a chimeric adenovirus-based coronavirus vaccine candidate using the ChimAd-CV and ChimAd-CVN plasmids produced in the present invention, each plasmid is treated with PacI restriction enzyme (NEB, R0547L) to eliminate unnecessary pBR322 for adenovirus formation. The origin of replication and kanamycin resistance gene sequences were cut and removed, and HEK293 cells were transformed using Lipofectamine 2000 (Invitrogen, 11668027), and maintained for 10-20 days in an incubator at 37°C and a CO2 partial pressure of 5.0%. After culturing, a primary virus stock was obtained.
ChimAd-CV 및 ChimAd-CVN 바이러스 생산은 상기 확보한 primary virus stock을 이용하여 약 1x 106 cells/ml cell density의 HEK293 세포에 2 MOI의 역가로 감염시켜 48시간 후 0.45 ㎛ CFF microfiltration filter(Cytiva, CFP-4-E-6A)를 이용하여 세포를 농축하였다. 회수한 세포에 0.5 % Tween20(sigma-aldrich, P2287-500ML)과 20 U/ml benzonase(Milipore, E1014-25KU)를 4시간 처리하여 cell lysis와 host cell DNA degradation을 유도하였다. 이후 2 ㎛ 및 0.6 ㎛ ULTA GF filter (Cytiva, KGF-A-0210TT, KGF-A-9606TT)를 이용해 cell debris를 제거하고, tangential flow filtration (TFF)을 통해 바이러스 농축과 buffer exchange를 진행하였다. PBS 완충액 상태의 바이러스 시료는 HiTrap Capto Q ImpRes 컬럼(Cytiva, 17547051)을 이용한 anion exchange chromatography와 Capto core 700 컬럼(Cytiva, 17548115)을 이용한 fast protein liquid chromatography(FPLC)를 통해 ChimAd-CV, ChimAd-CVN 바이러스를 분리하여 최종적으로 백신 완충액으로 제형화 단계를 거쳐 0.2 ㎛ ULTA GF filter (Cytiva, KGF-A-9610TT)로 최종 여과하여 2개의 백신후보물질을 확보하였다.ChimAd-CV and ChimAd-CVN virus production was performed by infecting HEK293 cells at a cell density of about 1x 10 6 cells/ml with a titer of 2 MOI using the primary virus stock obtained above, and after 48 hours, 0.45 ㎛ CFF microfiltration filter (Cytiva, CFP-4-E-6A) was used to enrich the cells. The recovered cells were treated with 0.5% Tween20 (Sigma-aldrich, P2287-500ML) and 20 U/ml benzonase (Milipore, E1014-25KU) for 4 hours to induce cell lysis and host cell DNA degradation. Thereafter, cell debris was removed using 2 ㎛ and 0.6 ㎛ ULTA GF filters (Cytiva, KGF-A-0210TT, KGF-A-9606TT), and virus concentration and buffer exchange were performed through tangential flow filtration (TFF). Virus samples in PBS buffer were subjected to anion exchange chromatography using a HiTrap Capto Q ImpRes column (Cytiva, 17547051) and fast protein liquid chromatography (FPLC) using a Capto core 700 column (Cytiva, 17548115) to detect ChimAd-CV and ChimAd-CVN. The virus was isolated, finally formulated into a vaccine buffer, and finally filtered with a 0.2 μm ULTA GF filter (Cytiva, KGF-A-9610TT) to secure two vaccine candidates.
실시예 5 : 결합항체가/ 중화항체가/ 세포성면역 분석을 통한 면역원성 실험 결과Example 5: Immunogenicity test results through binding antibody/neutralizing antibody/cellular immunity assay
5-1) BALB/c female을 이용한 면역 유도5-1) Induction of immunity using BALB/c female
상기 제조방법으로 확보한 ChimAd-CV 및 ChimAd-CVN 백신후보물질을 이용하여 마우스 면역을 실시하였다. 시험 그룹에는 완충제, Mock-up으로 사용된 ChimAd, 그리고 ChimAd-CV, ChimAd-CVN으로 총 4그룹을 포함한다. 면역 유도 시험에는 5주령 BALB/c 암컷 마우스를 사용하였으며, 각 아데노바이러스 시험군은 2.5*10^9 vp/dose의 농도로 주사하였다. 4주 간격으로 접종은 1차접종 과 2차접종으로 총 2회 (0일차, 28일차) 근육주사 하였으며, 최초 접종 이후 4주, 6주 후에 (27일차, 42일차) 혈액을 채취하여 혈청을 분리하고 비장을 적출하였다.Mouse immunization was performed using the ChimAd-CV and ChimAd-CVN vaccine candidates obtained by the above preparation method. A total of 4 groups were included in the test group: ChimAd used as buffer, mock-up, ChimAd-CV and ChimAd-CVN. For the immunity induction test, 5-week-old BALB/c female mice were used, and each adenovirus test group was injected at a concentration of 2.5*10^9 vp/dose. Inoculations were administered intramuscularly twice a total of 1st and 2nd vaccinations at 4-week intervals (day 0 and 28), and 4 and 6 weeks after the first vaccination (day 27 and 42), blood was collected and serum was collected. isolated and the spleen was removed.
5-2) ELISA; ChimAd-CV, ChimAd-CVN에 의한 마우스 항-혈청의 결합항체가 5-2) ELISA; Mouse anti-serum binding antibodies by ChimAd-CV and ChimAd-CVN
마우스 면역 유도 시험을 통한 혈청 내의 항체여부를 확인하기 위하여 결합항체가 테스트를 수행하였다. 테스트를 수행하기 위해, Microplate에 코로나바이러스 초기주와 베타변이주 S 단백질을 1ug/ml의 농도로 100ng/well씩 분주하고 4도에서 16시간 이상 코팅하였다. 반응 후 plate는 0.05% Tween20이 포함된 1XPBS로 (PBS-T) 4번 이상 세척하였으며, 0.5% casein을 100ul 분주하여 37℃에서 1시간 반응시켜주었다. PBS-T로 세척을 진행하고 각각의 혈청을 1:100으로 희석한 후 3배씩 연속적으로 희석하여 약 1x106까지 희석하였다. 분주 후 37℃에서 1시간 반응시킨 뒤, PBS-T로 세척 후 HRP가 결합된 Goat anti-mouse IgG HRP (Thermo)를 1:5000으로 희석하여 1시간 반응하였다. 반응이 끝나고 발색을 진행하기 위하여 TMB 용액 (Thermo)을 50ul 분주하여 15-20분간 발색 반응을 진행하였으며, 이후 Stop 용액 (Thermo)을 동량 첨가하여 반응을 정지시켜준 뒤, 450nm 파장에서 흡광도를 측정하였다.In order to confirm the presence of antibodies in the serum through the mouse immunity induction test, a binding antibody test was performed. To perform the test, 100 ng / well of the coronavirus initial strain and beta mutant S protein were dispensed at a concentration of 1 ug / ml in a microplate and coated for more than 16 hours at 4 degrees. After the reaction, the plate was washed more than 4 times with 1XPBS containing 0.05% Tween20 (PBS-T), and 100ul of 0.5% casein was dispensed and reacted at 37 ° C for 1 hour. After washing with PBS-T, each serum was diluted 1:100 and then serially diluted 3 times to about 1x10 6 . After dispensing, the mixture was reacted at 37° C. for 1 hour, washed with PBS-T, diluted with HRP-coupled goat anti-mouse IgG HRP (Thermo) at a ratio of 1:5000, and reacted for 1 hour. After the reaction was finished, 50 μl of TMB solution (Thermo) was dispensed to proceed with the color development, and the color development reaction proceeded for 15-20 minutes. After that, the reaction was stopped by adding an equal amount of Stop solution (Thermo), and the absorbance was measured at a wavelength of 450 nm. did
결합 항체 역가 결과, ChimAd-CV, ChimAd-CVN 접종한 실험 군 대조군 대비하여 모두 초기주와 베타변이주에 104이상의 높은 결합 항체 역가를 나타내는 것을 확인하였다. 대조군을 접종한 실험군에서는 초기주와 베타변이주에 대한 결합 항체 역가가 모두 측정되었으나, 유효하지 않은 값으로 확인되었다(도 2). As a result of the binding antibody titer, it was confirmed that both the initial strain and the beta mutant showed a high binding antibody titer of 10 4 or more compared to the control group inoculated with ChimAd-CV and ChimAd-CVN. In the experimental group inoculated with the control group, Binding antibody titers for both the initial strain and the beta mutant were measured, but were found to be invalid values (FIG. 2).
5-3) pVNT (pseudovirus neutralization test) 5-3) pVNT (pseudovirus neutralization test)
; ChimAd-CV, ChimAd-CVN에 의한 마우스 항-혈청의 슈도바이러스에 대한 중화항체가 측정 ; Measurement of neutralizing antibody against pseudovirus in mouse anti-serum by ChimAd-CV and ChimAd-CVN
마우스 면역 유도 시험을 통한 혈청 내의 항체의 중화항체가 테스트를 수행하였다. 슈도바이러스 시스템은 Lentivirus의 back bone을 사용하여 발광유전자와 초기주 또는 베타변이주의 Spike 단백질을 발현하고 있는 바이러스이다. 본 실험은 Takara사의 Lenti-X™SARS-CoV-2 Packaging Mix 제품을 사용하여 초기주와 베타변이주의 Spike 단백질을 포함한 슈도바이러스를 생산하였다. 생산된 슈도바이러스는 ACE2 단백질이 과 발현된 293T 세포에 형질전환 시켜 luciferase 발현을 정량화 하여 측정한 뒤 일정 RLU (Relative Light Units) 값을 사용하여 중화항체 시험을 수행하였다. 중화항체 시험은 먼저 각각의 마우스 혈청을 56℃에서 30분간 불활화 하였고, 10% FBS가 포함된 DMEM 배지를 사용하였다. 혈청은 1/8로 희석한 뒤 2-fold로 연속적으로 희석하여 약 103까지 수행하였으며, 각각의 일정한 RLU값을 가진 슈도바이러스를 희석된 혈청과 동일한 양으로 혼합하여 37℃, 5% CO2 배양기에서 1시간 중화반응을 시켜주었다. 하루 전에 배양한 hACE2-293T 세포에 바이러스와 혈청 혼합물을 100ul 처리하여 약 72시간 37℃, 5% CO2 배양기에서 배양하였다. 배양 종료 후 약 100ul 배양액을 제거하고, One-Glo luciferase reagent (Promega)를 30ul 추가하여 3-5분 반응시켜준 뒤, 60ul의 혼합물을 white plate로 옮겨주어 luminometer를 이용하여 발광 값을 측정하였다. 측정된 RLU 값을 통계 프로그램을 사용하여 혈청의 희석 배율에 따른 IC50 값을 계산하였다. The neutralizing antibody of the antibody in serum through mouse immunity induction test was tested. The pseudovirus system is a virus that uses the backbone of Lentivirus to express the luminescent gene and Spike protein of the initial strain or beta mutant strain. In this experiment, pseudoviruses containing Spike proteins of the initial strain and the beta mutant strain were produced using Takara's Lenti-X™SARS-CoV-2 Packaging Mix product. The produced pseudovirus was transformed into 293T cells in which ACE2 protein was overexpressed, and luciferase expression was quantified and measured, and neutralizing antibody tests were performed using a constant RLU (Relative Light Units) value. For the neutralizing antibody test, each mouse serum was first inactivated at 56° C. for 30 minutes, and DMEM medium containing 10% FBS was used. Serum was diluted 1/8 and then serially diluted 2-fold to about 10 3 . Pseudoviruses having a constant RLU value were mixed in equal amounts with the diluted serum at 37°C and 5% CO 2 . Neutralization was performed in an incubator for 1 hour. The hACE2-293T cells cultured one day before were treated with 100 μl of the virus and serum mixture, and cultured in a 37°C, 5% CO 2 incubator for about 72 hours. After the incubation was completed, about 100 ul culture medium was removed, 30 ul of One-Glo luciferase reagent (Promega) was added, reacted for 3-5 minutes, and then 60 ul of the mixture was transferred to a white plate and the luminescence value was measured using a luminometer. The measured RLU value was calculated using a statistical program to calculate the IC50 value according to the serum dilution factor.
베타변이 바이러스에 대한 중화항체 결과, 표 3과 같이 ChimAd-CV 접종군은 IC50=1253, ChimAd-CVN 접종 그룹은 IC50=1131으로 모두 103 이상으로 음성대조군 대비 높은 중화항체가를 확인하였다. As a result of the neutralizing antibody against the beta-mutant virus, as shown in Table 3, the IC 50 =1253 for the ChimAd-CV vaccinated group and the IC 50 =1131 for the ChimAd-CVN vaccinated group were all higher than 10 3 , confirming a higher neutralizing antibody titer than the negative control group. .
NoNo GroupGroup pVNT(IC50)pVNTs (IC50)
4wk4wk 6wk6wk
1One PBS PBS 77 4242
22 ChimAdChimAd 99 00
33 ChimAd-CVChimAd-CV 244244 12531253
44 ChimAd-CVNChimAd-CVN 409409 11311131
5-4) ELISpot; ChimAd-CV, ChimAd-CVN 아데노바이러스에 의한 특이적 T 세포 면역반응5-4) ELISpot; ChimAd-CV, ChimAd-CVN specific T cell immune response by adenovirus
마우스 면역 유도 시험에서 1차접종 4주후, 2차접종 2주후 (1차접종 6주후)에 각각의 그룹에서 비장을 적출하였다. 비장은 Cell strainer (Falcon) 제품을 사용하여 비장세포를 회수하였고, PBS 완충용액으로 세척하였다. 세척 후 적혈구 용해 버퍼 (Sigma)를 이용하여 적혈구를 제거하였고, 비장 세포의 수를 세어 시험에 사용되는 세포의 농도로 희석하였다. 본 실험은 R&D system의 mouse IFN-gamma ELISpot kit를 사용하였으며, 제조사의 사용법에 따라 시험을 수행하였다. 각 시험에는 1X105 cells, 2X104 cells의 비장세포와 Genscript사의 초기주의 Spike 단백질의 펩타이드 풀 또는 Nucleprotein 펩타이드 풀을 자극 항원으로 사용하여 함께 37℃, 5% CO2 배양기에서 16시간 배양하였다. 배양 후에 세척 용액으로 세척을 하였으며, 비오틴이 결합되어 있는 IFN-gamma 검출 항체를 100ul씩 넣어주고 상온에서 2시간 교반 하여 반응하였다. 반응 후에도 세척용액을 이용하여 4번의 세척하였고, streptavidin-AKP(alkaline phosphate)을 100ul씩 넣어주고 상온에서 2시간 배양하였다. 마지막 세척을 한 뒤, 100ul의 BCIP/NBT 기질을 첨가하여 상온에서 약 45분 동안 반응시켜주었으며, 반응에 의한 발색이 나타나게 되면 멸균된 물로 세척한 후 발색 된 spot의 개수를 세어 준 뒤, Spot Forming Unit (SFU)의 세포 수를 환산하였다. In the mouse immunity induction test, spleens were removed from each group 4 weeks after the first inoculation and 2 weeks after the second inoculation (6 weeks after the first inoculation). Splenocytes were collected using a cell strainer (Falcon), and washed with PBS buffer. After washing, red blood cells were removed using red blood cell lysis buffer (Sigma), and the number of splenocytes was counted and diluted to the cell concentration used for the test. In this experiment, R&D system's mouse IFN-gamma ELISpot kit was used, and the test was performed according to the manufacturer's instructions. In each test, splenocytes of 1X10 5 cells and 2X10 4 cells were incubated for 16 hours in a 37°C, 5% CO2 incubator together using a peptide pool or Nucleprotein peptide pool of Genscript's initial stock Spike protein as stimulating antigens. After culturing, it was washed with a washing solution, and 100ul of IFN-gamma detection antibody bound to biotin was added thereto, followed by stirring at room temperature for 2 hours. After the reaction, it was washed 4 times using a washing solution, and 100 μl of streptavidin-AKP (alkaline phosphate) was added and incubated for 2 hours at room temperature. After the last wash, 100ul of BCIP/NBT substrate was added and reacted at room temperature for about 45 minutes. The number of cells in Unit (SFU) was converted.
S 단백질, N 단백질 각각의 자극 항원에 대한 특이적 T-세포 면역 유도를 ELISpot 시험을 통해 측정한 결과, 1차접종 후 (4주차) 보다 2차접종 후 (6주차)에 T-세포 면역이 증가하였으며, 특히, Spike 단백질의 펩타이드에 대하여 ChimAd-CV보다 ChimAd-CVN에서 약 2배 높은 특이적 면역반응을 유도하는 것을 확인할 수 있었다. 또한, ChimAd-CVN은 Nucleocapsid peptide에 대하여 특이적 면역반응이 유도됨을 확인하였다. As a result of measuring the induction of specific T-cell immunity to each stimulatory antigen of S protein and N protein through the ELISpot test, T-cell immunity was higher after the second vaccination (week 6) than after the first vaccination (week 4). In particular, it was confirmed that ChimAd-CVN induces a specific immune response about twice as high as ChimAd-CV against Spike protein peptides. In addition, it was confirmed that ChimAd-CVN induces a specific immune response to the Nucleocapsid peptide.
NoNo GroupGroup SPF/10^6 splenocytes (Spike peptide pool)SPF/10^6 splenocytes (Spike peptide pool)
4wk4wk 6wk6wk
1One PBSPBS 3434 1717
22 ChimAdChimAd 6161 4242
33 ChimAd-CVChimAd-CV 23672367 38243824
44 ChimAd-CVNChimAd-CVN 30203020 69496949
실시예 6 : ChimAd-CV, ChimAd-CVN 동물면역 후 항체역가, 중화항체가 및 공격시험Example 6: ChimAd-CV, ChimAd-CVN antibody titer after animal immunization, neutralizing antibody titer and challenge test
6-1) hACE2 transgenic female mouse를 이용한 면역 유도 및 공격시험6-1) Immunity induction and challenge test using hACE2 transgenic female mice
IVI에서 실시한 면역 유도 시험 및 공격 시험은 Jackson Laboratory (ME, USA)에서 구입한 6주령의 암컷 human ACE2 transgenic mice (tg(K18-ACE2)2Prlmn)을 사용하였다. 위 시험은 모두 국제백신연구소 내 BSL-3등급 시설에서 실시되었다. 면역 유도 시험 그룹에는 완충제, Mock-up으로 사용된 ChimAd, 그리고 ChimAd-CV, ChimAd-CVN으로 총 4그룹을 포함한다. 접종농도는 2.5 x 10^9 vp/dose의 농도로 주사하였다. 마우스는 4주간격으로 2회 근육내 접종하였으며, 1차 프라이밍 접종 4주, 6주 후에 혈액을 채취하였다. For the immune induction test and challenge test conducted at IVI, 6-week-old female human ACE2 transgenic mice (tg(K18-ACE2)2Prlmn) purchased from Jackson Laboratory (ME, USA) were used. All of the above tests were conducted in a BSL-3 grade facility in the International Vaccine Institute. A total of 4 groups were included in the immunity induction test group: ChimAd used as buffer, mock-up, ChimAd-CV and ChimAd-CVN. The inoculum was injected at a concentration of 2.5 x 10^9 vp/dose. Mice were intramuscularly inoculated twice at 4-week intervals, and blood was collected 4 weeks and 6 weeks after the first priming inoculation.
6-2) ELISA ; ChimAd-CV, ChimAd-CVN에 의한 마우스 항-혈청의 항체 역가 측정6-2) ELISA; Antibody titer measurement of mouse anti-serum by ChimAd-CV, ChimAd-CVN
마우스 면역 유도 시험 후 혈청 내의 항체 역가를 측정하기 위하여 수행되었다. 96-well plate에 초기주 S 단백질과 델타변이주 S 단백질을 2ug/ml의 농도로 100ng/well씩 분주하고 4도에서 16시간 이상 코팅하였다. 반응 후 1% BSA가 포함된 PBS를 100ul 분주하여 반응시켜 주었다. 각각의 혈청을 1:100으로 희석한 뒤, 5-fold로 연속적으로 희석하여 4도에서 16시간 이상 반응시켜 주었다. HRP가 결합된 Goat anti-mouse IgG HRP (Southern Biotech)를 1:3000으로 희석하여 37℃에서 1시간 반응하였다. 반응이 끝나고 발색을 진행하기 위하여 TMB 용액 (Millipore)을 분주하여 발색 반응을 진행하였으며, 이후 0.5 N HCl 용액(Merck)을 동량 첨가하여 반응을 정지시켜준 뒤, 450nm 파장에서 흡광도를 측정하였다. After mouse immunity induction test, it was performed to measure the antibody titer in serum. In a 96-well plate, the initial strain S protein and the delta mutant strain S protein were dispensed at a concentration of 2ug/ml and 100ng/well each, and coated at 4 degrees for 16 hours or more. After the reaction, 100ul of PBS containing 1% BSA was dispensed and reacted. After diluting each serum 1:100, it was serially diluted 5-fold and reacted at 4 degrees for more than 16 hours. HRP-coupled Goat anti-mouse IgG HRP (Southern Biotech) was diluted 1:3000 and reacted at 37°C for 1 hour. After the reaction was completed, TMB solution (Millipore) was dispensed to proceed with color development, and then, an equal amount of 0.5 N HCl solution (Merck) was added to stop the reaction, and absorbance was measured at a wavelength of 450 nm.
대조군에 비교하여 ChimAd-CV을 접종한 실험군과 ChimAd-CVN를 접종한 실험군 모두 초기주와 델타 변이주에 대하여 105 이상의 높은 결합 항체 역가를 나타내는 것을 확인하였다(도 5). Compared to the control group, it was confirmed that both the experimental group inoculated with ChimAd-CV and the experimental group inoculated with ChimAd-CVN showed a high binding antibody titer of 10 5 or more to the initial strain and the delta mutant strain (FIG. 5).
6-3) FRNT (Focus Reduction Neutralization Test); 6-3) FRNT (Focus Reduction Neutralization Test);
ChimAd-CV, ChimAd-CVN 마우스 항-혈청의 코로나바이러스에 대한 중화항체가 측정 Measurement of neutralizing antibodies against coronavirus in ChimAd-CV and ChimAd-CVN mouse anti-serum
마우스 면역 유도 시험 후 혈청 내의 중화항체를 측정하기 위하여 시험을 수행하였다. 중화항체 시험을 위해 먼저 각각의 마우스 혈청을 56℃에서 30분간 불활화 반응을 하였고, 2% FBS가 포함된 DMEM 배지를 사용하여 2-fold로 연속적으로 희석하였다. 희석된 혈청과 4.5x102PFU/25ul의 초기주 코로나바이러스 (NCCP #43326 (BetaCoV/Korea/KCDC03/2020)와 4.0x102PFU/25ul의 델타변이주 코로나바이러스 (NCCP #43390 (hCoV-19/Korea119861/KDCA/2021)와 혼합하여 37℃에서 30분간 반응시켜주었다. 반응 후, 하루 전에 배양한 Vero cells (1.5x104 cells/well)에 바이러스와 혈청 혼합물을 50ul 처리하여 37℃,5% CO2 배양기에서 4시간 배양하였다. 4시간 반응 후에 상층액을 제거하고 PBS 완충 용액을 이용하여 세척하였으며, 4% formaldehyde 용액 (Sigma)를 300ul 처리하여 빛이 없는 곳에서 16시간 이상 고정시켜주었다. 반응 후 PBS를 이용하여 세척을 진행한 뒤, 100% cold methanol을 추가하여 10분간 반응시켜준 뒤, Blocking 용액을 이용하여 상온에서 1시간 반응시켜주었다. 이후, 1:3000으로 희석된 anti-SARS-CoV-2 NP rabbit monoclonal antibody (Sino Biological)를 처리하여 37도에서 1시간 배양하였고, 배양 후 0.1% Tween20이 포함된 PBS 용액 (PBS-T)으로 세척한 뒤, 1:2000로 희석한 HRP-conjugated goat anti-rabbit IgG (Bio-Rad)를 처리하여 37℃에서 1시간 배양하였다. PBS-T로 세척 진행 후, TMB 용액을 빛이 없는 조건에서 상온에서 30분간 반응시켜주었고, 물로 반응을 정지시킨 후 건조시켰다. FRNT50 분석을 위해 Spot reader (Cytation 7 Cell Imaging Multi-Mode Reader)를 사용하였으며, 혈청의 희석 배율에 따라 값을 계산하였다. FRNT법은 바이러스에 감염된 세포에 바이러스 유래 단클론 항체를 부착시킨 후 HRP(horedish peroxidase)로 표지된 2차항체를 붙여 기질로 발색시켜 바이러스에 감염된 세포를 확인하여 중화항체를 측정하는 방법이다. After the mouse immune induction test, a test was performed to measure neutralizing antibodies in serum. For the neutralizing antibody test, first, each mouse serum was inactivated at 56° C. for 30 minutes, and serially diluted 2-fold using DMEM medium containing 2% FBS. Diluted serum and 4.5x10 2 PFU/25ul of primary strain coronavirus (NCCP #43326 (BetaCoV/Korea/KCDC03/2020) and 4.0x10 2 PFU/25ul of delta mutant coronavirus (NCCP #43390 (hCoV-19/Korea119861) /KDCA/2021) and reacted for 30 minutes at 37 ° C. After the reaction, Vero cells (1.5x10 4 cells/well) cultured one day before were treated with 50 μl of the virus and serum mixture and kept at 37 ° C, 5% CO 2 Incubated for 4 hours in an incubator After 4 hours of reaction, the supernatant was removed, washed with PBS buffer solution, treated with 300ul of 4% formaldehyde solution (Sigma), and fixed for more than 16 hours in a place without light. After washing with PBS, 100% cold methanol was added, reacted for 10 minutes, and then reacted for 1 hour at room temperature using a blocking solution, then anti-SARS-CoV diluted 1:3000. -2 NP rabbit monoclonal antibody (Sino Biological) was treated and incubated at 37 degrees for 1 hour, washed with PBS solution (PBS-T) containing 0.1% Tween20 after incubation, and then diluted 1:2000 HRP-conjugated It was treated with goat anti-rabbit IgG (Bio-Rad) and incubated for 1 hour at 37 ° C. After washing with PBS-T, the TMB solution was reacted for 30 minutes at room temperature in the absence of light, and the reaction was stopped with water. For FRNT50 analysis, a Spot reader (Cytation 7 Cell Imaging Multi-Mode Reader) was used, and the value was calculated according to the dilution factor of serum. After that, a secondary antibody labeled with HRP (horedish peroxidase) is attached to the substrate to develop color, and virus-infected cells are identified to measure neutralizing antibodies.
초기주 바이러스에 대한 중화항체 결과, 표 5와 같이 중화항체가를 확인할 수 있으며, 이는 ChimAd-CV와 ChimAd-CVN 그룹 모두 음성 대조군에 비해 1차 접종 이후 100배 이상 높은 중화항체 역가를 나타낸 것을 확인하였다. 2차 접종 이후에는 ChimAd-CV 접종군은 30배 이상, ChimAd-CVN 접종군은 50배 이상 높은 중화 항체 역가를 나타내는 것을 확인하였다. As a result of the neutralizing antibody against the initial strain virus, the neutralizing antibody titer was confirmed as shown in Table 5, which confirmed that both the ChimAd-CV and ChimAd-CVN groups showed more than 100 times higher neutralizing antibody titer than the negative control group after the first inoculation. did After the second inoculation, it was confirmed that the ChimAd-CV inoculation group showed 30-fold higher neutralizing antibody titers and the ChimAd-CVN-inoculated group showed 50-fold higher neutralizing antibody titers.
또한, 델타변이주 바이러스에 대한 중화항체 결과, 2차 접종 이후 ChimAd-CV 접종군과 ChimAd-CVN 접종군 모두 음성 대조군에 비해 40배 이상 높은 중화 항체 역가를 나타내는 것을 확인하였다. In addition, as a result of the neutralizing antibody to the delta mutant virus, it was confirmed that both the ChimAd-CV and ChimAd-CVN inoculated groups showed neutralizing antibody titers 40 times higher than the negative control group after the second inoculation.
NoNo GroupGroup 사스-코로나바이러스-2(FRNT50)SARS-coronavirus-2 (FRNT50) 델타변이주(FRNT50)Delta mutant strain (FRNT50)
4wk4wk 6wk6wk 4wk4wk 6wk6wk
1One PBS PBS 1010 1010 1010 1010
22 ChimAd ChimAd 1010 5050 1010 2626
33 ChimAd-CVChimAd-CV 12881288 16601660 646646 12021202
44 ChimAd-CVNChimAd-CVN 10961096 30203020 513513 10961096
6-4) ChimAd-CV, ChimAd-CVN 면역 후 바이러스 공격 시험6-4) Virus challenge test after immunization with ChimAd-CV and ChimAd-CVN
공격 시험은 부스팅 접종 4주 후에 5x105 PFU SARS-CoV-2 바이러스 (초기주, 델타변이주)를 비강 내로 감염시켰다. 접종 후에는 매일 생존과 체중을 모니터링 하였으며, 감염 후 2일, 4일 7일차에는 혈액, 폐, 간, 신장, 비장을 채취하여 장기 무게를 측정하고 폐에서의 바이러스 역가를 측정하였다. In the challenge test, 5x10 5 PFU SARS-CoV-2 virus (initial strain, delta mutant strain) was intranasally infected 4 weeks after boosting inoculation. After inoculation, survival and weight were monitored daily. On the 2nd, 4th and 7th days after infection, blood, lungs, liver, kidneys, and spleens were collected, organ weights were measured, and viral titers in the lungs were measured.
초기주 (Wuhan-Hu-1) 공격에 대해 ChimAd-CV와 ChimAd-CVN의 백신 방어능을 확인 결과, 모두생존율 100%로 나타났으며, ChimAd-CVN 접종군은 몸무게가 감소하지 않고 유지되는 것을 확인하였으며, ChimAd-CV 접종군은 한 마리를 제외하고 모두 몸무게가 감소하지 않고 유지되었으며, 몸무게가 10% 이상 감소했다가 회복한 개체가 있는 것을 확인하였다.As a result of confirming the vaccine defense ability of ChimAd-CV and ChimAd-CVN against the initial strain (Wuhan-Hu-1) attack, both showed 100% survival rate, and the ChimAd-CVN inoculation group showed that body weight was maintained without loss. It was confirmed that all but one of the ChimAd-CV inoculated groups did not lose weight and maintained, and it was confirmed that there was an individual whose body weight decreased by more than 10% and then recovered.
6-5) ChimAd-CV, ChimAd-CVN 면역 유도와 공격 시험에 대한 폐 바이러스 역가 (Plaque assay)6-5) ChimAd-CV, ChimAd-CVN immunity induction and lung virus titer for challenge test (plaque assay)
마우스 면역유도 및 공격시험 진행 후 폐에서의 잔존 바이러스 역가 측정을 위하여 Plague assay 테스트를 진행하였다. 역가 측정을 위해서 Vero cells (ATCC, Cat#CCL-81)을 사용하였으며, 37℃, 5% CO2 배양기에서 16시간 이상 배양하였다. 모든 그룹에서 채취한 폐에서 분리한 바이러스는 4-fold로 단계적으로 106까지 희석하여, 배양한 Vero cell에 희석한 바이러스를 200ul씩 추가하여 상온에서 30분 반응시켜주었다. 반응 후 바이러스 혼합물을 제거해주고 0.8% Agarose와 2% FBS가 포함된 DMEM (Overlay media)을 덮어주고 15분간 상온에서 반응시킨 후, 37℃, 5% CO2 배양기에서 72시간 배양하였다. 10% 포르말린 용액으로 1시간 동안 고정한 뒤, Overlay media를 제거하고, crystal violet (Sigma)을 사용하여 plaque를 5분 동안 염색하였다. 바이러스 역가를 확인하기 위하여, plaque의 개수를 세어 준 뒤, 각각의 희석배수와 총 양(ml)을 계산하여 최종적인 역가를 계산하였다. After induction of mouse immunity and challenge test, a Plague assay test was performed to measure the residual virus titer in the lung. For titer measurement, Vero cells (ATCC, Cat#CCL-81) were used and cultured for more than 16 hours in a 37°C, 5% CO 2 incubator. The virus isolated from the lungs collected from all groups was diluted to 10 6 in a 4-fold stepwise manner, and 200 ul of the diluted virus was added to the cultured Vero cells and allowed to react at room temperature for 30 minutes. After the reaction, the virus mixture was removed, covered with DMEM (Overlay media) containing 0.8% Agarose and 2% FBS, reacted at room temperature for 15 minutes, and then cultured for 72 hours in a 37°C, 5% CO 2 incubator. After fixation with 10% formalin solution for 1 hour, overlay media was removed, and plaques were stained for 5 minutes using crystal violet (Sigma). To confirm the virus titer, the number of plaques was counted, and each dilution factor and total amount (ml) were calculated to calculate the final titer.
공격시험 이후 폐에서의 잔존 바이러스 역가 측정 결과, 초기주 공격에 대해 ChimAd-CV 접종군에서는 바이러스 공격 후 대조군에 비해 공격 2일 후에 lung virus titer가 20,000배 이상 감소하는 것을 확인하였으며, 공격 4일 후와 7일 후에는 virus clearance되어 측정되지 않는 것을 확인하였다. ChimAd-CVN 접종군은 virus clearance되어 2일 후, 4일 후, 7일 후까지 모두 lung virus titer가 측정되지 않는 것을 확인하였다. As a result of measuring the residual virus titer in the lungs after the challenge test, it was confirmed that the lung virus titer decreased more than 20,000 times 2 days after the challenge compared to the control group after the virus challenge in the ChimAd-CV vaccinated group, and 4 days after the challenge. and 7 days later, it was confirmed that virus clearance was not measured. In the ChimAd-CVN inoculation group, it was confirmed that no lung virus titer was measured until 2, 4, and 7 days after virus clearance.
실시예 7 : ChimAd-CV의 DRF(Dose range finding) 시험에 따른 결합항체가/ 중화항체가/ 세포성면역 분석을 통한 면역원성 실험 결과Example 7: Immunogenicity test result through binding antibody/neutralizing antibody/cellular immunity assay according to DRF (Dose range finding) test of ChimAd-CV
7-1) BALB/c female을 이용한 ChimAd-CV의 용량반응시험7-1) Dose response test of ChimAd-CV using BALB/c female
ChimAd-CV 백신후보물질을 이용하여 용량반응별 마우스 면역을 실시하였다. 면역 유도 시험에는 5주령 BALB/c 암컷 마우스를 사용하였으며, ChimAd-CV 백신후보물질의 용량은 표 6과 같이 4그룹으로 나누었으며 안정화제(Vehicle), Mock up, Non-immunization 그룹으로 구성되었다. 기허가 백신 접종 농도를 고려하여 5.0.E+09 VP/Dose (마우스 기준 인체 대비 1/10)로부터 2-fold로 고농도와 저농도로 설정하였다. 4주 간격으로 접종은 1차접종 과 2차접종으로 총 2회 (0일차, 28일차) 근육주사 하였으며, 최초 접종 이후 4주, 6주 후에 (27일차, 42일차) 혈액을 채취하여 혈청을 분리하고 비장을 적출하였다.A dose-response mouse immunization was performed using the ChimAd-CV vaccine candidate. For the immunity induction test, 5-week-old BALB/c female mice were used, and the doses of the ChimAd-CV vaccine candidate were divided into 4 groups as shown in Table 6, and were composed of stabilizer (Vehicle), mock up, and non-immunization groups. Considering the previously licensed vaccination concentration, the high and low concentrations were set at 2-fold from 5.0.E+09 VP/Dose (1/10 of the mouse standard human body). Inoculations were administered intramuscularly twice a total of 1st and 2nd vaccinations (Day 0, Day 28) at intervals of 4 weeks, and blood was collected 4 weeks and 6 weeks after the first vaccination (Day 27, Day 42) and serum was collected. isolated and the spleen was removed.
분리된 혈청은 결합 항체가와 다양한 코로나 바이러스(우한, 베타변이, 델타, 오미크론(BA.1))에 대한 중화항체가를 측정하였고, 적출된 비장으로 T-세포 면역반응을 분석하였다. The isolated serum was measured for binding antibody titers and neutralizing antibody titers to various corona viruses (Wuhan, beta-mutant, delta, and Omicron (BA.1)), and T-cell immune responses were analyzed with isolated spleens.
NoNo VaccineVaccine Dose(VP/dose)Dose (VP/dose) MouseMouse
1One Non-immunizationNon-immunization -- 55
22 안정화제 stabilizer 100ul100ul 55
33 ChimAd(Mock up)ChimAd (Mock up) 100ul100ul 55
44 ChimAd-CV1ChimAd-CV1 1.00.E+101.00.E+10 1010
55 ChimAd-CV2ChimAd-CV2 5.00.E+095.00.E+09 1010
66 ChimAd-CV3ChimAd-CV3 2.50.E+092.50.E+09 1010
77 ChimAd-CV4ChimAd-CV4 1.25.E+091.25.E+09 1010
7-2) ELISA; ChimAd-CV 백신후보물질의 용량반응별 마우스 항-혈청의 결합항체가 7-2) ELISA; Mouse anti-serum binding antibody by dose response of ChimAd-CV vaccine candidate
ChimAd-CV 백신후보물질의 용량반응별 마우스 면역 유도 시험을 통한 혈청 내의 항체여부를 확인하기 위하여 결합항체가 테스트를 수행하였다. 테스트를 수행하기 위해, Microplate에 코로나바이러스 사스-코로나바이러스-2와 베타변이주 S 단백질을 1ug/ml의 농도로 100ng/well씩 분주하고 4도에서 16시간 이상 코팅하였다. 반응 후 plate는 0.05% Tween20이 포함된 1XPBS로 (PBS-T) 4번 이상 세척하였으며, 0.5% casein을 100ul 분주하여 37℃에서 1시간 반응시켜주었다. PBS-T로 세척을 진행하고 각각의 혈청을 1:100으로 희석한 후 3배씩 연속적으로 희석하여 약 1x106까지 희석하였다. 분주 후 37℃에서 1시간 반응시킨 뒤, PBS-T로 세척 후 HRP가 결합된 Goat anti-mouse IgG HRP (Thermo)를 1:5000으로 희석하여 1시간 반응하였다. 반응이 끝나고 발색을 진행하기 위하여 TMB 용액 (Thermo)을 50ul 분주하여 15-20분간 발색 반응을 진행하였으며, 이후 Stop 용액 (Thermo)을 동량 첨가하여 반응을 정지시켜준 뒤, 450nm 파장에서 흡광도를 측정하였다.In order to confirm the presence of antibodies in the serum through a mouse immunity induction test for each dose response of the ChimAd-CV vaccine candidate, a binding antibody test was performed. To perform the test, the coronavirus SARS-Coronavirus-2 and the beta mutant S protein were dispensed at a concentration of 1ug/ml by 100ng/well in a microplate and coated for more than 16 hours at 4 degrees. After the reaction, the plate was washed more than 4 times with 1XPBS containing 0.05% Tween20 (PBS-T), and 100ul of 0.5% casein was dispensed and reacted at 37 ° C for 1 hour. After washing with PBS-T, each serum was diluted 1:100 and then serially diluted 3 times to about 1x10 6 . After dispensing, the mixture was reacted at 37° C. for 1 hour, washed with PBS-T, diluted with HRP-coupled goat anti-mouse IgG HRP (Thermo) at a ratio of 1:5000, and reacted for 1 hour. After the reaction was finished, 50 μl of TMB solution (Thermo) was dispensed to proceed with the color development, and the color development reaction proceeded for 15-20 minutes. After that, the reaction was stopped by adding an equal amount of Stop solution (Thermo), and the absorbance was measured at a wavelength of 450 nm. did
결합 항체 역가 결과, 접종용량에 상관없이 실험 군 대조군 대비하여 모두 사스-코로나바이러스-2와 베타변이주에 104이상의 높은 결합 항체 역가를 나타내는 것을 확인하였다(도 9). As a result of the binding antibody titer, regardless of the inoculated dose, it was confirmed that all SARS-Coronavirus-2 and beta mutants exhibited a high binding antibody titer of 10 4 or more compared to the experimental group control group (FIG. 9).
7-3) pVNT (pseudovirus neutralization test); ChimAd-CV에 의한 마우스 항-혈청의 슈도바이러스에 대한 중화항체가 측정 7-3) pVNT (pseudovirus neutralization test); Measurement of neutralizing antibody against pseudovirus in mouse anti-serum by ChimAd-CV
마우스 면역 유도 시험을 통한 혈청 내의 항체의 중화항체가 테스트를 수행하였다. 본 실험은 Virongy사의 Rapid Cell-Based SARS-CoV-2 Neutralizing Antibody Assay Kit를 사용하여 우한바이러스, 베타변이바이러스, 델타바이러스, 오미크론(BA.1)바이러스를 이용하여 측정하였다. 본 시험에 사용하는 바이러스는 Virongy사에서 개발한 hybrid SARS-CoV-2 virus-like particle (VLP)로 SARS-CoV-2의 4가지 구조단백질(S,M,N,E)과 결합되어 있으며 luciferase를 발현할 수 있다. The neutralizing antibody of the antibody in serum through mouse immunity induction test was tested. This experiment was measured using Virongy's Rapid Cell-Based SARS-CoV-2 Neutralizing Antibody Assay Kit using Wuhan virus, beta mutant virus, delta virus, and Omicron (BA.1) virus. The virus used in this test is a hybrid SARS-CoV-2 virus-like particle (VLP) developed by Virongy, which is combined with the 4 structural proteins (S, M, N, E) of SARS-CoV-2 and has luciferase can be expressed.
중화항체 시험은 먼저 각각의 마우스 혈청을 56℃에서 30분간 불활화 하였고, 혈청은 Virongy infection 배지를 이용하여 1/8로 희석한 뒤 2-fold로 연속적으로 희석하여 약 103까지 수행하였다. White plate(cell culture)에 희석된 샘플과 HA-CoV2(Luc)VLP 45ul 혼합하여 37℃5% CO2 배양기에서 30분 중화반응을 시켜주었다. In the neutralizing antibody test, each mouse serum was first inactivated at 56° C. for 30 minutes, and the serum was diluted 1/8 using Virongy infection medium and then serially diluted 2-fold to about 10 3 . A sample diluted in a white plate (cell culture) and 45ul of HA-CoV2(Luc)VLP were mixed and neutralized for 30 minutes in a 37°C 5% CO 2 incubator.
HEK293T(ACE2/TMPRSS2) 세포를 2.5 x 104 cells/15 μL 준비하여 바이러스와 혈청 혼합물에 15 μL 처리하여 약 18시간 37℃5% CO2 배양기에서 배양하였다. 배양 종료 후 Cell lysis buffer 7.5 ul를 넣고 Shaker에서 2min 방치한다. Luciferase solution를 25ul 추가하여 1분 반응시켜준 뒤, luminometer를 이용하여 발광 값을 측정하였다. 측정된 RLU 값을 통계 프로그램을 사용하여 혈청의 희석 배율에 따른 IC50 값을 계산하였다.HEK293T (ACE2/TMPRSS2) cells were prepared at 2.5 x 10 4 cells/15 μL, treated with 15 μL of the virus and serum mixture, and cultured in a 37°C 5% CO 2 incubator for about 18 hours. After the incubation, add 7.5 ul of cell lysis buffer and leave on a shaker for 2 minutes. After adding 25ul of Luciferase solution and reacting for 1 minute, the luminescence value was measured using a luminometer. The measured RLU values were calculated using a statistical program to calculate IC50 values according to serum dilution factors.
표 7과 같이 Boosting 후(6주차) 접종용량별 코로나바이러스에 대한 중화항체가 확인하였다. 우한바이러스에 대한 중화항체가 확인결과, CV1은 IC50=1177, CV2은 IC50=577, CV3은 IC50=311, CV4은 IC50=607 중화항체로 CV1에서 가장 높은 중화항체가 확인하였다. 베타바이러스에 대한 중화항체가 확인결과, CV1은 IC50=965, CV2은 IC50=416, CV3은 IC50=627, CV4은 IC50=304 중화항체가 확인됨에 따라, CV1에서 가장 높은 중화항체가로 나타나며, dose dependent한 중화항체가를 확인하였다.As shown in Table 7, after boosting (week 6), neutralizing antibodies to coronavirus by inoculation dose were confirmed. As a result of confirming neutralizing antibodies against the Wuhan virus, CV1 had IC50=1177, CV2 had IC50=577, CV3 had IC50=311, and CV4 had IC50=607 neutralizing antibodies. The highest level of neutralizing antibodies was confirmed in CV1. As a result of confirming neutralizing antibody against beta virus, CV1 has IC50=965, CV2 has IC50=416, CV3 has IC50=627, and CV4 has IC50=304. A dose dependent neutralizing antibody titer was confirmed.
델타바이러스에 대한 중화항체가 확인결과, CV1은 IC50=517, CV2은 IC50=139, CV3은 IC50=798, CV4은 IC50=115 중화항체가로 델타바이러스에 대해 CV3을 제외한 dose dependent한 중화항체가를 확인하였다. 오미크론바이러스에 대한 중화항체가 확인 결과, CV1은 IC50=76, CV2은 IC50=117, CV3은 IC50=110, CV4은 IC50=93 중화항체가 확인되었고, CV2, CV3에서 100이상의 오미크론바이러스에 대한 중화항체가를 확인하였다(도 10). As a result of confirming neutralizing antibodies against delta virus, CV1 had IC50=517, CV2 had IC50=139, CV3 had IC50=798, and CV4 had IC50=115 neutralizing antibodies. confirmed. As a result of confirming neutralizing antibodies against Omicron virus, CV1 had IC50=76, CV2 had IC50=117, CV3 had IC50=110, and CV4 had IC50=93. A neutralizing antibody titer was confirmed (FIG. 10).
Target virusTarget virus pVNT(IC50)pVNTs (IC50)
CV1CV1 CV2CV2 CV3CV3 CV4 CV4
Wuhan virus Wuhan virus 11771177 577577 311311 607607
Beta variant Beta variant 965965 416416 627627 344344
Delta variant Delta variant 517517 325325 798798 115115
Omicron(BA.1)Omicron (BA.1) 7676 117117 110110 9393
7-4) ELISpot; ChimAd-CV 아데노바이러스에 의한 특이적 T 세포 면역반응7-4) ELISpot; Specific T cell immune response by ChimAd-CV adenovirus
마우스 면역 유도 시험에서 1차접종 4주후, 2차접종 2주후 (1차접종 6주후)에 각각의 그룹에서 비장을 적출하였다. 비장은 Cell strainer (Falcon) 제품을 사용하여 비장세포를 회수하였고, PBS 완충용액으로 세척하였다. 세척 후 적혈구 용해 버퍼 (Sigma)를 이용하여 적혈구를 제거하였고, 비장 세포의 수를 세어 시험에 사용되는 세포의 농도로 희석하였다. 본 실험은 R&D system의 mouse IFN-gamma ELISpot kit를 사용하였으며, 제조사의 사용법에 따라 시험을 수행하였다. 각 시험에는 1X105 cells, 2X104 cells의 비장세포와 Genscript사의 사스-코로나바이러스-2의 Spike 단백질의 펩타이드 풀을 자극 항원으로 사용하여 함께 37℃5% CO2 배양기에서 16시간 배양하였다. 배양 후에 세척 용액으로 세척을 하였으며, 비오틴이 결합되어 있는 IFN-gamma 검출 항체를 100ul씩 넣어주고 상온에서 2시간 교반 하여 반응하였다. 반응 후에도 세척용액을 이용하여 4번의 세척하였고, streptavidin-AKP(alkaline phosphate)을 100ul씩 넣어주고 상온에서 2시간 배양하였다. 마지막 세척을 한 뒤, 100ul의 BCIP/NBT 기질을 첨가하여 상온에서 약 45분 동안 반응시켜주었으며, 반응에 의한 발색이 나타나게 되면 멸균된 물로 세척한 후 발색 된 spot의 개수를 세어 준 뒤, Spot Forming Unit (SFU)의 세포 수를 환산하였다. In the mouse immunity induction test, spleens were removed from each group 4 weeks after the first inoculation and 2 weeks after the second inoculation (6 weeks after the first inoculation). Splenocytes were collected using a cell strainer (Falcon), and washed with PBS buffer. After washing, red blood cells were removed using red blood cell lysis buffer (Sigma), and the number of splenocytes was counted and diluted to the cell concentration used for the test. In this experiment, R&D system's mouse IFN-gamma ELISpot kit was used, and the test was performed according to the manufacturer's instructions. In each test, splenocytes of 1X10 5 cells and 2X10 4 cells and Genscript's SARS-coronavirus-2 Spike protein peptide pool were used as stimulating antigens and cultured for 16 hours in a 37°C 5% CO2 incubator. After culturing, it was washed with a washing solution, and 100ul of IFN-gamma detection antibody bound to biotin was added thereto, followed by stirring at room temperature for 2 hours. After the reaction, it was washed 4 times using a washing solution, and 100 μl of streptavidin-AKP (alkaline phosphate) was added and incubated for 2 hours at room temperature. After the last wash, 100ul of BCIP/NBT substrate was added and reacted at room temperature for about 45 minutes. The number of cells in Unit (SFU) was converted.
표 8과 같이 S 단백질 자극 항원에 대한 특이적 T-세포 면역 유도를 ELISpot 시험을 통해 측정한 결과, ChimAd-CV는 접종 용량별 4주차 대비 접종 6주차에서 모두 높은 T-세포 면역반응 유도 확인되었다. 접종 용량에 상관없이 모든 접종 범위에서 6주차에 우수한 세포성 면역유도능이 확인하였다 (도 11).As shown in Table 8, as a result of measuring the induction of specific T-cell immunity to the S protein stimulating antigen through the ELISpot test, ChimAd-CV induces a high T-cell immune response at the 6th week compared to the 4th week by inoculation dose. It was confirmed. . Regardless of the inoculation dose, excellent cellular immunity induction ability was confirmed at 6 weeks in all inoculation ranges (FIG. 11).
시험군
주차
test group
parking
SPF/10^6 splenocytes (Spike peptide pool)SPF/10^6 splenocytes (Spike peptide pool)
CV1CV1 CV2CV2 CV3 CV3 CV4CV4
4주차Week 4 794794 934934 923923 888888
6주차 Week 6 15751575 16911691 18781878 24912491
[서열 정보][Sequence information]
SEQ ID NO: 1 - SARS-CoV-2_B.1.351(beta) spike modified amino acid sequenceSEQ ID NO: 1 - SARS-CoV-2_B.1.351(beta) spike modified amino acid sequence
MFVFLVLLPLVSSQCVNFTTRTQLPPAYTNSFTRGVYYPDKVFRSSVLHSTQDLFLPFFSMFVFLVLLPLVSSQCVNFTTRTQLPPAYTNSFTRGVYYPDKVFRSSVLHSTQDLFLPFFS
NVTWFHAIHVSGTNGTKRFANPVLPFNDGVYFASTEKSNIIRGWIFGTTLDSKTQSLLIVNVTWFHAIHVSGTNGTKRFANPVLPFNDGVYFASTEKSNIIRGWIFGTTLDSKTQSLLIV
NNATNVVIKVCEFQFCNDPFLGVYYHKNNKSWMESEFRVYSSANNCTFEYVSQPFLMDLNNATNVVIKVCEFQFCNDPFLGVYYHKNNKSWMESEFRVYSSANNCTFEYVSQPFLMDL
EGKQGNFKNLREFVFKNIDGYFKIYSKHTPINLVRGLPQGFSALEPLVDLPIGINITRFQTEGKQGNFKNLREFVFKNIDGYFKIYSKHTPINLVRGLPQGFSALEPLVDLPIGINITRFQT
LHRSYLTPGDSSSGWTAGAAAYYVGYLQPRTFLLKYNENGTITDAVDCALDPLSETKLHRSYLTPGDSSSGWTAGAAAYYVGYLQPRTFLLKYNENGTITDAVDCALDPLSETK
CTLKSFTVEKGIYQTSNFRVQPTESIVRFPNITNLCPFGEVFNATRFASVYAWNRKRISNCTLKSFTVEKGIYQTSNFRVQPTESIVRFPNITNLCPFGEVFNATRFASVYAWNRKRISN
CVADYSVLYNSASFSTFKCYGVSPTKLNDLCFTNVYADSFVIRGDEVRQIAPGQTGNIADCVADYSVLYNSASFSTFKCYGVSPTKLNDLCFTNVYADSFVIRGDEVRQIAPGQTGNIAD
YNYKLPDDFTGCVIAWNSNNLDSKVGGNYNYLYRLFRKSNLKPFERDISTEIYQAGSTPCYNYKLPDDFTGCVIAWNSNNLDSKVGGNYNYLYRLFRKSNLKPFERDISTEIYQAGSTPC
NGVKGFNCYFPLQSYGFQPTYGVGYQPYRVVVLSFELLHAPATVCGPKKSTNLVKNKCVNGVKGFNCYFPLQSYGFQPTYGVGYQPYRVVVLSFELLHAPATVCGPKKSTNLVKNKCV
NFNFNGLTGTGVLTESNKKFLPFQQFGRDIADTTDAVRDPQTLEILDITPCSFGGVSVITPNFNFNGLTGTGVLTESNKKFLPFQQFGRDIADTTDAVRDPQTLEILDITPCSFGGVSVITP
GTNTSNQVAVLYQGVNCTEVPVAIHADQLTPTWRVYSTGSNVFQTRAGCLIGAEHVNNSGTNTSNQVAVLYQGVNCTEVPVAIHADQLTPTWRVYSTGSNVFQTRAGCLIGAEHVNNS
YECDIPIGAGICASYQTQTNSPqqaqSVASQSIIAYTMSLGVENSVAYSNNSIAIPTNFTIYECDIPIGAGICASYQTQTNSPqqaqSVASQSIIAYTMSLGVENSVAYSNNSIAIPTNFTI
SVTTEILPVSMTKTSVDCTMYICGDSTECSNLLLQYGSFCTQLNRALTGIAVEQDKNTQESVTTEILPVSMTKTSVDCTMYICGDSTECSNLLLQYGSFCTQLNRALTGIAVEQDKNTQE
VFAQVKQIYKTPPIKDFGGFNFSQILPDPSKPSKRSFIEDLLFNKVTLADAGFIKQYGDCVFAQVKQIYKTPPIKDFGGFNFSQILPDPSKPSKRSFIEDLLFNKVTLADAGFIKQYGDC
LGDIAARDLICAQKFNGLTVLPPLLTDEMIAQYTSALLAGTITSGWTFGAGAALQIPFAMLGDIAARDLICAQKFNGLTVLPPLLTDEMIAQYTSALLAGTITSGWTFGAGAALQIPFAM
QMAYRFNGIGVTQNVLYENQKLIANQFNSAIGKIQDSLSSTASALGKLQDVVNQNAQALQMAYRFNGIGVTQNVLYENQKLIANQFNSAIGKIQDSLSSTASALGKLQDVVNQNAQAL
NTLVKQLSSNFGAISSVLNDILSRLDPPEAEVQIDRLITGRLQSLQTYVTQQLIRAAEIRANTLVKQLSSNFGAISSVLNDILSRLDPPEAEVQIDRLITGRLQSLQTYVTQQLIRAAEIRA
SANLAATKMSECVLGQSKRVDFCGKGYHLMSFPQSAPHGVVFLHVTYVPAQEKNFTTAPSANLAATKMSECVLGQSKRVDFCGKGYHLMSFPQSAPHGVVFLHVTYVPAQEKNFTTAP
AICHDGKAHFPREGVFVSNGTHWFVTQRNFYEPQIITTDNTFVSGNCDVVIGIVNNTVYDPAICHDGKAHFPREGVFVSNGTHWFVTQRNFYEPQIITTDNTFVSGNCDVVIGIVNNTVYDP
LQPELDSFKEELDKYFKNHTSPDVDLGDISGINASVVNIQKEIDRLNEVAKNLNESLIDLLQPELDSFKEELDKYFKNHTSPDVDLGDISGINASVVNIQKEIDRLNEVAKNLNESLIDL
QELGKYEQYIKWPWYIWLGFIAGLIAIVMVTIMLCCMTSCCSCLKGCCSCGSCCKFDEDDQELGKYEQYIKWPWYIWLGFIAGLIAIVMVTIMLCCMTSCCSCLKGCCSCGSCCKFDEDD
SEPVLKGVKLHYT*SEPVLKGVKLHYT*
ID NO: 2 - SARS-CoV-2_B.1.351(beta) spike modified nucleic acid sequence ID NO: 2 - SARS-CoV-2_B.1.351(beta) spike modified nucleic acid sequence
하기는 본 발명의 융합 폴리펩타이드 삽입체의 서열을 기재한다 The following describes the sequences of fusion polypeptide inserts of the invention
MGLPNNTASWFTALTQHGKEDLKFPRGQGVPINTNSSPDDQIGYYRRATRRIRGGDGKMMGLPNNTASWFTALTQHGKEDLKFPRGQGVPINTNSSPDDQIGYYRRATRRIRGGDGKM
KDLSPRWYFYYLGTGPEAGLPYGANKDGIIWVATEGALNTPKDHIGTRNPANNAAIVLQLKDLSPRWYFYYLGTGPEAGLPYGANKDGIIWVATEGALNTPKDHIGTRNPANNAAIVLQL
PQGTTLPKGFYAEGSRGGS(서열번호 3)PQGTTLPKGFYAEGSRGGS (SEQ ID NO: 3)
GGGGSGGGGSGGGGSGGGGSGGGGSGGGGS GGGGSGGGGSGGGGSGGGGSGGGGSGGGGS
TKKSAAEASKKPRQKRTATKAYNVTQAFGRRGPEQTQGNFGDQELIRQGTDYKHWPQIATKKSAAEASKKPRQKRTATKAYNVTQAFGRRGPEQTQGNFGDQELIRQGTDYKHWPQIA
QFAPSASAFFGMSRIGMEVTPSGTWLTYTGAIKLDDKDPNFKDQVILLNKHIDAYKTFP*(서열번호 4)QFAPSASAFFGMSRIGMEVTPSGTWLTYTGAIKLDDKDPNFKDQVILLNKHIDAYKTFP* (SEQ ID NO: 4)
상기 서열에서 서열번호 3은 SARS-CoV-2-B.1.1.351 Spike 단백질의 N말단에 결합한 서열이고, 서열4는 C말단에 결합한 서열이며, 밑줄은 링커를 의미하고, 화살표는 SARS-CoV-2-B.1.1.351 Spike 단백질이 삽입되는 위치를 나타낸다.In the above sequences, SEQ ID NO: 3 is a sequence bound to the N-terminus of the SARS-CoV-2-B.1.1.351 Spike protein, SEQ ID NO: 4 is a sequence bound to the C-terminus, the underline indicates a linker, and an arrow indicates a SARS-CoV-2-B.1.1.351 Spike protein. -2-B.1.1.351 Indicates the site where Spike protein is inserted.
ATG GGC CTG CCC AAC AAC ACC GCC AGC TGG TTT ACC GCC CTG ACCATG GGC CTG CCC AAC AAC ACC GCC AGC TGG TTT ACC GCC CTG ACC
CAG CAC GGC AAG GAG GAC CTG AAG TTC CCC AGA GGC CAG GGC GTG CAG CAC GGC AAG GAG GAC CTG AAG TTC CCC AGA GGC CAG GGC GTG
CCC ATT AAC ACC AAC AGC AGC CCC GAC GAC CAG ATC GGC TAC CCC ATT AAC ACC AAC AGC AGC CCC GAC GAC CAG ATC GGC TAC
TAC AGA AGA GCC ACC AGA AGA ATC AGA GGC GGC GAC GGC AAG ATGTAC AGA AGA GCC ACC AGA AGA ATC AGA GGC GGC GAC GGC AAG ATG
AAG GAC CTG AGC CCC AGA TGG TAC TTC TAC TAT CTG GGA ACC GGCAAG GAC CTG AGC CCC AGA TGG TAC TTC TAC TAT CTG GGA ACC GGC
CCC GAA GCC GGC CTG CCC TAT GGC GCC AAC AAA GAT GGC ATCCCC GAA GCC GGC CTG CCC TAT GGC GCC AAC AAA GAT GGC ATC
ATC TGG GTG GCC ACC GAA GGC GCC CTG AAC ACC CCC AAG GAT CAC ATCATC TGG GTG GCC ACC GAA GGC GCC CTG AAC ACC CCC AAG GAT CAC ATC
GGA ACC AGA AAT CCC GCC AAC AAT GCC GCC ATC GTG CTG GGA ACC AGA AAT CCC GCC AAC AAT GCC GCC ATC GTG CTG
CAG CTG CCC CAG GGA ACC ACC CTG CCT AAA GGC TTT TAT GCC GAA GGA AGCCAG CTG CCC CAG GGA ACC ACC CTG CCT AAA GGC TTT TAT GCC GAA GGA AGC
AGA GGA GGA AGC (서열번호 5) GGC GGC GGA GGA AGC GGC GGA GGA AGA GGA GGA AGC (SEQ ID NO: 5) GGC GGC GGA GGA AGC GGC GGA GGA
GGC AGC GGC GGA GGC GGA AGC GGC GGA GGA GGA AGC GGC GGA GGC AGC GGC GGA GGC GGA AGCGGC GGA GGA GGA AGC GGC GGA
GGC GGA AGC GGC GGC GGA GGA AGC ACC AAA AAG AGC GCC GCC GAG GCC GGC GGA AGC GGC GGC GGA GGA AGC ACC AAA AAG AGC GCC GCC GAG GCC
AGC AAA AAG CCC AGA CAG AAA AGA ACC GCC ACC AAGAGC AAA AAG CCC AGA CAG AAA AGA ACC GCC ACC AAG
GCC TAC AAT GTG ACC CAG GCC TTT GGC AGA AGA GGA CCC GAG CAGGCC TAC AAT GTG ACC CAG GCC TTT GGC AGA AGA GGA CCC GAG CAG
ACC CAG GGC AAT TTT GGC GAC CAG GAG CTG ATC AGA CAG GGC ACCACC CAG GGC AAT TTT GGC GAC CAG GAG CTG ATC AGA CAG GGC ACC
GAC TAC AAA CAC TGG CCC CAG ATC GCC CAG TTC GCC CCC AGC GCCGAC TAC AAA CAC TGG CCC CAG ATC GCC CAG TTC GCC CCC AGC GCC
AGC GCC TTC TTC GGC ATG AGC AGA ATC GGC ATG GAG GTG ACC CCCAGC GCC TTC TTC GGC ATG AGC AGA ATC GGC ATG GAG GTG ACC CCC
AGC GGC ACC TGG CTG ACC TAC ACC GGC GCC ATC AAG CTG GAC GACAGC GGC ACC TGG CTG ACC TAC ACC GGC GCC ATC AAG CTG GAC GAC
AAG GAC CCC AAC TTC AAG GAC CAG GTG ATC CTG CTG AAC AAG CACAAG GAC CCC AAC TTC AAG GAC CAG GTG ATC CTG CTG AAC AAG CAC
ATC GAC GCC TAC AAG ACC TTC CCC TGA (서열번호 6) ATC GAC GCC TAC AAG ACC TTC CCC TGA (SEQ ID NO: 6)
상기 서열에서 서열번호 5는 SARS-CoV-2-B.1.1.351 Spike 단백질의 N말단에 결합한 폴리펩타이드를 코딩하는 유전자 서열이고, 서열6은 C말단에 결합한 폴리펩타이드를 코딩하는 유전자 서열이고, 밑줄은 링커를 코딩하는 유전자 서열을 의미하고, 화살표는 SARS-CoV-2-B.1.1.351 Spike 단백질이 삽입되는 위치를 나타낸다.In the above sequences, SEQ ID NO: 5 is a gene sequence encoding a polypeptide bound to the N-terminus of the SARS-CoV-2-B.1.1.351 Spike protein, SEQ ID NO: 6 is a gene sequence encoding a polypeptide bound to the C-terminus, The underline indicates the gene sequence encoding the linker, and the arrow indicates the insertion site of the SARS-CoV-2-B.1.1.351 Spike protein.
서열번호 7 : Chimpanzee adenovirus serotype 6 Fiber knob domain a.a sequenceSEQ ID NO: 7: Chimpanzee adenovirus serotype 6 Fiber knob domain a.a sequence
PDPSPNCQLLSDRDAKFTLCLTKCGSQILGTVAVAAVTVGSALNPINDTVKSAIVFLRFDSPDPSPNCQLLSDRDAKFTLCLTKCGSQILGTVAVAAVTVGSALNPINDTVKSAIVFLRFDS
DGVLMSNSSMVGDYWNFREGQTTQSVAYTNAVGFMPNIGAYPKTQSKTPKNSIVSQVYDGVLMSNSSMVGDYWNFREGQTTQSVAYTNAVGFMPNIGAYPKTQSKTPKNSIVSQVY
LTGETTMPMTLTITFNGTDEKDTTPVSTYSMTFTWQWTGDYKDKNITFATNSFSFSYIAQELTGETTMPMTLTITFNGTDEKDTTPVSTYSMTFTWQWTGDYKDKNITFATNSFSFSYIAQE

Claims (12)

  1. 사스-코로나바이러스-2의 베타변이주의 스파이크 단백질의 아미노산 1번부터 1270번까지를 포함하며, 여기서 초기주 기준 614번, 682번부터 685번, 986번, 987번의 아미노산 잔기에 돌연변이를 가지고 있으며, 여기서 상기 단백질은 스파이크 단백질의 614번 단백질의 아스파르트산 아미노산이 글라이신으로 치환되고, 682번 아르기닌 아미노산은 글루타민, 683번 알라닌 아미노산은 글루타민, 684번, 685번 아르기닌 아미노산은 각각 알라닌, 글루타민으로, 986번 및 987번의 연속된 프롤린 치환 돌연변이로 치환되는 사스-코로나바이러스-2의 베타변이주의 스파이크 단백질을 코딩하는 유전자를 포함하는 삽입체.It includes amino acids 1 to 1270 of the spike protein of the beta mutant strain of SARS-coronavirus-2, where it has mutations at amino acid residues 614, 682 to 685, 986, and 987 based on the initial week, Here, in the protein, the aspartic acid amino acid of protein No. 614 of the spike protein is substituted with glycine, arginine amino acid No. 682 is glutamine, alanine amino acid No. 683 is glutamine, and arginine amino acids No. 684 and 685 are alanine and glutamine, respectively, and No. 986 and 987 consecutive An insert containing a gene encoding a spike protein of a beta mutant strain of SARS-coronavirus-2 substituted with a proline substitution mutation.
  2. 제1항에 있어서, 상기 삽입체는 상기 사스-코로나바이러스-2의 베타변이주의 스파이크 단백질의 N말단에 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 44번부터 180번까지의 아미노산 잔기를 연결하고, 사스-코로나바이러스-2의 베타변이주의 스파이크 단백질의 C 말단에 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 247번부터 364번의 아미노산 잔기를 연결하여 제조된 융합 폴리펩타이드를 코딩하는 유전자를 포함하는 삽입체.The method of claim 1, wherein the insert connects amino acid residues 44 to 180 of the nucleocapsid protein of SARS-coronavirus-2 to the N-terminus of the spike protein of the beta mutant strain of SARS-coronavirus-2. and a gene encoding a fusion polypeptide prepared by linking amino acid residues 247 to 364 of the nucleocapsid protein of SARS-Coronavirus-2 to the C-terminus of the spike protein of the beta mutant SARS-Coronavirus-2. containing inserts.
  3. 제2항에 있어서, 상기 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 44번부터 180번까지의 아미노산 잔기는 서열번호 3의 아미노산 서열로 이루어지고, 상기 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 247번부터 364번의 아미노산 잔기는 서열번호 4의 아미노산 서열로 이루어진 삽입체.The method of claim 2, wherein amino acid residues from positions 44 to 180 of the nucleocapsid protein of SARS-coronavirus-2 consist of the amino acid sequence of SEQ ID NO: 3, and the nucleocapsid of SARS-coronavirus-2 Amino acid residues at positions 247 to 364 of the protein are an insert consisting of the amino acid sequence of SEQ ID NO: 4.
  4. 제2항에 있어서, 상기 스파이크 단백질의 N말단에 연결된 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 44번부터 180번까지의 아미노산 잔기를 코딩하는 유전자는 서열번호 5에 기재된 염기서열로 이루어지고, 상기 스파이크 단백질의 C 말단에 연결된 사스-코로나바이러스-2의 뉴클레오캡시드 단백질의 247번부터 364번의 아미노산 잔기를 코딩하는 유전자는 서열번호 6에 기재된 염기서열로 이루어진 것을 특징으로 하는 삽입체.The method of claim 2, wherein the gene encoding amino acid residues 44 to 180 of the nucleocapsid protein of SARS-coronavirus-2 linked to the N-terminus of the spike protein consists of the nucleotide sequence shown in SEQ ID NO: 5, , The insert, characterized in that the gene encoding amino acid residues 247 to 364 of the nucleocapsid protein of SARS-coronavirus-2 linked to the C-terminus of the spike protein consists of the nucleotide sequence shown in SEQ ID NO: 6.
  5. 제1항에 있어서, 상기 사스-코로나바이러스-2의 베타변이주의 변형된 스파이크 단백질은 서열번호 1에 기재된 아미노산 서열로 이루어지고, 상기 사스-코로나바이러스-2의 베타변이주의 변형된 스파이크 단백질을 코딩하는 유전자는 서열번호 2에 기재된 염기서열로 이루어진 것을 특징으로 하는 삽입체.The method of claim 1, wherein the modified spike protein of the beta mutant strain of SARS-coronavirus-2 consists of the amino acid sequence shown in SEQ ID NO: 1, and encodes the modified spike protein of the beta-mutant strain of SARS-coronavirus-2. The insert characterized in that the gene consists of the nucleotide sequence shown in SEQ ID NO: 2.
  6. 제1항 내지 제5항 중 어느 한 항의 삽입체를 포함하는 재조합 발현 벡터. A recombinant expression vector comprising the insert of any one of claims 1 to 5.
  7. 제6항에 있어서, 발현 벡터는 바이러스 벡터 또는 비바이러스 벡터인 것을 특징으로 하는 재조합 발현 벡터.7. The recombinant expression vector according to claim 6, wherein the expression vector is a viral vector or a non-viral vector.
  8. 제7항에 있어서, 상기 바이러스 벡터는 아데노바이러스 벡터, 아데노연관바이러스 벡터, 렌티바이러스 벡터, 레트로바이러스 벡터 및 헤르페스바이러스 벡터로 이루어진 그룹으로부터 선택되는 것을 특징으로 하는 재조합 발현 벡터.8. The recombinant expression vector according to claim 7, wherein the viral vector is selected from the group consisting of adenovirus vectors, adeno-associated virus vectors, lentiviral vectors, retroviral vectors and herpesvirus vectors.
  9. 제8항에 있어서, 상기 아데노바이러스 벡터는 인간 아데노바이러스 5형의 섬유 단백질 말단의 돌기 도메인을 침팬지 아데노바이러스 혈청형 6의 돌기 유전자로 치환하고, 인간 아데노바이러스 5형의 테일(Tail) 및 사프트(Shaft) 도메인과 침팬지 아데노바이러스 혈청형 6의 돌기 도메인으로 조합된 섬유단백질을 포함하는 재조합 발현 벡터.The method according to claim 8, wherein the adenovirus vector replaces the terminal domain of the fiber protein of human adenovirus type 5 with the spine gene of chimpanzee adenovirus serotype 6, and the tail and shaft of human adenovirus type 5 ( Shaft) domain and a protrusion domain of chimpanzee adenovirus serotype 6. A recombinant expression vector containing a fiber protein combined.
  10. 제9항에 있어서, 상기 침팬지 아데노바이러스 혈청형 6의 돌기 유전자에 의하여 발현된 단백질은 서열번호 7의 아미노산 서열을 포함하는 재조합 발현 벡터.The recombinant expression vector according to claim 9, wherein the protein expressed by the chimpanzee adenovirus serotype 6 protrusion gene comprises the amino acid sequence of SEQ ID NO: 7.
  11. 제 6항 내지 제10항 중 어느 한 항의 재조합 발현 벡터를 유효성분으로 포함하는 사스-코로나바이러스-2(SARS-CoV-2)의 감염증 예방 또는 치료용 백신 조성물.A vaccine composition for preventing or treating infections of SARS-CoV-2, comprising the recombinant expression vector according to any one of claims 6 to 10 as an active ingredient.
  12. 제11항에 있어서, 상기 백신은 SARS-CoV-2 바이러스 초기주, 델타 바이러스, 베타 및 오미크론 바이러스 변이주로 구성된 군으로부터 선택된 하나 이상의 바이러스에 효과를 가지는 것을 특징으로 하는 사스-코로나바이러스-2(SARS-CoV-2)의 감염증 예방 또는 치료용 백신 조성물.The method of claim 11, wherein the vaccine is SARS-CoV-2, characterized in that it has an effect on one or more viruses selected from the group consisting of virus initial strain, delta virus, beta and Omicron virus mutant strains (SARS-coronavirus-2 ( A vaccine composition for preventing or treating an infection of SARS-CoV-2).
PCT/KR2023/002808 2022-03-03 2023-02-28 Recombinant expression vector for prevention of sars-cov-2 infection and application thereof WO2023167487A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20220027718 2022-03-03
KR10-2022-0027718 2022-03-03
KR10-2023-0024283 2023-02-23
KR1020230024283A KR20230131439A (en) 2022-03-03 2023-02-23 Recombinant expression vector for preventing SARS-coronavirus-2 infection and its application

Publications (1)

Publication Number Publication Date
WO2023167487A1 true WO2023167487A1 (en) 2023-09-07

Family

ID=87883969

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/002808 WO2023167487A1 (en) 2022-03-03 2023-02-28 Recombinant expression vector for prevention of sars-cov-2 infection and application thereof

Country Status (1)

Country Link
WO (1) WO2023167487A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210355170A1 (en) * 2020-05-18 2021-11-18 The Regents Of The University Of Colorado, A Body Corporate Stabilizing Mutants of Prefusion Sars-Cov-2 (Covid-19) Spike Protein And Improved Yeast Surface Display Engineering Platform For The Same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210355170A1 (en) * 2020-05-18 2021-11-18 The Regents Of The University Of Colorado, A Body Corporate Stabilizing Mutants of Prefusion Sars-Cov-2 (Covid-19) Spike Protein And Improved Yeast Surface Display Engineering Platform For The Same

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE Protein 27 August 2021 (2021-08-27), ANONYMOUS : "Chain A, Spike glycoprotein", XP093087933, retrieved from NCBI Database accession no. 7N1Q_A *
TIAN JING-HUI, PATEL NITA, HAUPT ROBERT, ZHOU HAIXIA, WESTON STUART, HAMMOND HOLLY, LOGUE JAMES, PORTNOFF ALYSE D., NORTON JAMES, : "SARS-CoV-2 spike glycoprotein vaccine candidate NVX-CoV2373 immunogenicity in baboons and protection in mice", NATURE COMMUNICATIONS, vol. 12, no. 1, 14 January 2021 (2021-01-14), pages 372, XP055844876, DOI: 10.1038/s41467-020-20653-8 *
XU KUN, AN YALING, LI QUNLONG, HUANG WEIJIN, HAN YUXUAN, ZHENG TIANYI, FANG FANG, LIU HUI, LIU CHUANYU, GAO PING, XU SENYU, LIU XU: "Recombinant chimpanzee adenovirus AdC7 expressing dimeric tandem-repeat spike protein RBD protects mice against COVID-19", EMERGING MICROBES & INFECTIONS, vol. 10, no. 1, 1 January 2021 (2021-01-01), pages 1574 - 1588, XP093011508, DOI: 10.1080/22221751.2021.1959270 *
YANG TZU-JING, YU PEI-YU, CHANG YUAN-CHIH, HSU SHANG-TE DANNY: "D614G mutation in the SARS-CoV-2 spike protein enhances viral fitness by desensitizing it to temperature-dependent denaturation", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY FOR BIOCHEMISTRY AND MOLECULAR BIOLOGY, US, vol. 297, no. 4, 1 October 2021 (2021-10-01), US , pages 101238, XP093087928, ISSN: 0021-9258, DOI: 10.1016/j.jbc.2021.101238 *

Similar Documents

Publication Publication Date Title
WO2021221486A1 (en) Vaccine composition for preventing or treating infection of sars-cov-2
WO2019209079A1 (en) Nucleic acid molecules inserted expression regulation sequences, expression vector comprising nucleic acid moleclues and pharmaceutical use thereof
WO2021000969A2 (en) Nucleic acid sequence expressing sars-cov-2 virus antigen peptide and use thereof
WO2009088256A9 (en) Baculovirus-based vaccines
WO2019225962A1 (en) Antigen variant of varicella zoster virus and use thereof
WO2020005028A1 (en) Vaccine composition for preventing or treating diseases caused by severe fever with thrombocytopenia syndrome (sfts) viral infection
WO2020091529A1 (en) Respiratory syncytial virus recombinant f protein and vaccine composition containing same
WO2019182417A1 (en) Ns1 protein derived from yellow fever virus, monoclonal antibody specifically binding thereto, and use thereof
WO2016085284A1 (en) Epitope of hepatitis b virus surface antigen and binding molecule specifically binding to same for neutralizing hepatitis b virus
WO2023167487A1 (en) Recombinant expression vector for prevention of sars-cov-2 infection and application thereof
WO2019212312A1 (en) Chimeric zika virus vaccine
WO2022119380A1 (en) Novel ace2 variant and use thereof
WO2021034101A1 (en) Novel interferon lambda variant and production method therefor
WO2019194393A1 (en) Nanoparticles in which antigen peptide and adjuvant are bound to ferritin self assembly, and use thereof
WO2022211373A1 (en) Recombinant chimeric adenoviral vector substituted by knob gene of chimpanzee adenovirus serotype 6, and application thereof
KR20230131439A (en) Recombinant expression vector for preventing SARS-coronavirus-2 infection and its application
WO2010076932A1 (en) Tnf-α antagonists containing igfbp5
WO2022216025A1 (en) Recombinant mycobacterium strain expressing sars-cov-2 antigen, and vaccine composition including same
WO2020076141A1 (en) Recombinant rsv live vaccine strain and production method therefor
WO2022050520A1 (en) Fusion protein comprising coronavirus-derived receptor binding domain and nucleocapsid protein, and use thereof
WO2023003332A1 (en) Plant-based covid-19 variant recombinant spike protein expression vector and recombinant protein using same
WO2024014943A1 (en) Sars-cov-2 vaccine booster composition
WO2023229330A1 (en) Pentameric toxin protein-based target protein fusion pentamer producing platform using viral nucleocapsid
WO2022216028A1 (en) Vaccine composition for preventing severe acute respiratory syndrome coronavirus type 2 infectious disease, having improved neutralization potency
WO2022065889A1 (en) Vaccine composition comprising recombinant protein for prevention or treatment of sars-corona virus-2 infection

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: 23763686

Country of ref document: EP

Kind code of ref document: A1