WO2023227124A1 - Skeleton for constructing mrna in-vitro transcription template - Google Patents

Skeleton for constructing mrna in-vitro transcription template Download PDF

Info

Publication number
WO2023227124A1
WO2023227124A1 PCT/CN2023/096643 CN2023096643W WO2023227124A1 WO 2023227124 A1 WO2023227124 A1 WO 2023227124A1 CN 2023096643 W CN2023096643 W CN 2023096643W WO 2023227124 A1 WO2023227124 A1 WO 2023227124A1
Authority
WO
WIPO (PCT)
Prior art keywords
mrna
utr
universal
sequence
vaccine
Prior art date
Application number
PCT/CN2023/096643
Other languages
French (fr)
Chinese (zh)
Inventor
张丹
邹军
王胜东
张建三
Original Assignee
深圳市三源生生物科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 深圳市三源生生物科技有限公司 filed Critical 深圳市三源生生物科技有限公司
Publication of WO2023227124A1 publication Critical patent/WO2023227124A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • 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
    • C12N15/64General methods for preparing the vector, for introducing it into the cell or for selecting the vector-containing host
    • 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
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • 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
    • C12N15/74Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12RINDEXING SCHEME ASSOCIATED WITH SUBCLASSES C12C - C12Q, RELATING TO MICROORGANISMS
    • C12R2001/00Microorganisms ; Processes using microorganisms
    • C12R2001/01Bacteria or Actinomycetales ; using bacteria or Actinomycetales
    • C12R2001/185Escherichia
    • C12R2001/19Escherichia coli

Definitions

  • the invention belongs to the field of biomedicine, and specifically relates to a skeleton for constructing an in vitro transcription template of mRNA and its application in the optimized design of mRNA.
  • mRNA vaccine is a new vaccine technology that combines molecular biology and immunology to transduce mRNA into somatic cells to express foreign antigens and activate the host's adaptive immunity.
  • scientists injected mRNA into mouse somatic cells, causing the mouse somatic cells to express fluorescent proteins, ⁇ -galactosidase and chloramphenicol acetyltransferase.
  • Jirikowski et al. injected mRNA encoding oxytocin and vasopressin into diabetic diabetes insipidus mice. As a result, the mice did not develop diabetes insipidus within hours after the injection. Since then, the development of mRNA vaccines has fallen into a trough.
  • mRNA vaccines are mainly to improve the stability and translation activity of mRNA and reduce the self-antigenicity of mRNA.
  • the new coronavirus vaccine which can be launched due to the urgency of research and development, the research and development of other mRNA vaccines have been hindered to some extent. The reason may be that when designing the mRNA vaccine, too much attention is paid to its ability to express antigens, while the risk of toxicity and the risk of apoptosis of surrounding host cells caused by the antigenicity and high expression ability of the mRNA vaccine itself are ignored.
  • the purpose of the present invention is to increase the versatility of the mRNA vaccine by using the UTR of immune-related proteins, while reducing potential side effects.
  • the universal framework can further accelerate the research and development of various infectious diseases.
  • a universal framework for constructing an mRNA transcript wherein the transcript includes an ORF to be expressed and a pair of genes located on both sides of the ORF. 5'-UTR region and 3'-UTR region, wherein one or two of the 5'-UTR region and 3'-UTR region are universal UTRs.
  • the universal UTR is selected from the following group: the UTR conserved sequence of a mutated or optimized antibody gene, the UTR conserved sequence of a mutated or optimized interferon gene, or a combination thereof.
  • the mRNA transcript includes the mRNA transcript in the mRNA vaccine.
  • the universal framework also includes additional UTR regions (ie, other UTR regions besides the universal UTR).
  • nucleotide sequence of the UTR conserved sequence of the antibody gene is shown in SEQ ID NO: 1 or 3.
  • the UTR conserved sequence of the antibody gene includes a 5' antibody conserved sequence and a 3' antibody conserved sequence.
  • nucleotide sequence of the 5' antibody conserved sequence is shown in SEQ ID NO: 1.
  • nucleotide sequence of the 3' antibody conserved sequence is shown in SEQ ID NO: 3.
  • sequence shown in SEQ ID NO:3 is conserved in different IGL genes.
  • nucleotide sequence of the UTR conserved sequence of the interferon gene is shown in SEQ ID NO: 7 or 9.
  • the UTR conserved sequence of the interferon gene includes a 5' conserved interferon sequence and a 3' conserved interferon sequence.
  • nucleotide sequence of the 5' interferon conserved sequence is shown in SEQ ID NO: 7.
  • nucleotide sequence of the 3' interferon conserved sequence is shown in SEQ ID NO: 9.
  • sequence shown in SEQ ID NO: 7 or 9 is conserved among different IFNA subtypes.
  • the universal UTR includes a universal 5'-UTR and a universal 3'-UTR.
  • the universal UTR includes: a 5'-UTR containing a Kozak sequence.
  • the AT-rich sequence in the 3'-UTR of the antibody gene and interferon gene is completely or partially deleted.
  • basically no AT-rich sequences means that in one UTR, the number of AT-rich sequences is ⁇ 2, and more preferably ⁇ 1.
  • the AT-rich sequence refers to a nucleic acid sequence rich in adenine and thymine bases.
  • the GC content in the universal UTR is 44%-64%.
  • nucleotide sequence of the universal 5'-UTR is shown in SEQ ID NO: 2 or 8.
  • nucleotide sequence of the universal 3'-UTR is shown in SEQ ID NO: 4, 5, 6, 10, 11 or 12.
  • any one of the above nucleotide sequences also includes optionally adding, deleting, modifying and/or replacing at least one (such as 1-3) nucleotides and retaining Derived sequences for optimizing mRNA capacity.
  • a general-purpose skeleton is provided, and the general-purpose skeleton has the structure of formula I: Z1-Z2-Z3-Z4-Z5-Z6-Z7 (I)
  • Z1 and Z7 are no or enzyme cutting sites
  • Z2 is none or promoter element
  • Z3 is a 5'-UTR component
  • Z4 is a replaceable ORF region
  • Z5 is a 3'-UTR component
  • Z6 is a polyA tail component.
  • Z1 and Z7 are blunt end enzyme cleavage sites or sticky end enzyme cleavage sites.
  • the universal framework includes an enzyme cleavage site, a promoter, a 5'-UTR, an ORF, a 3'-UTR and polyA.
  • the blunt-end enzyme is selected from the following group: AelI, AatI, AluI, BavAI, BavBI, EcoRV, MlsI, or a combination thereof.
  • the blunt-end enzyme is AleI.
  • the sticky end enzyme is BspQI.
  • the Z2 is selected from the following group: T7 promoter, T3 promoter, SP6 promoter, or a combination thereof.
  • the Z2 is a T7 promoter.
  • one or two of Z3 and Z5 are universal UTRs.
  • the Z3 is selected from the following group: a mutated or optimized 5' antibody conserved sequence, a mutated or optimized 5' interferon conserved sequence, or a combination thereof.
  • the Z3 is selected from the following group: SEQ ID NO: 2, SEQ ID NO: 8, the 5'-UTR of human ⁇ -globin, or a combination thereof.
  • the Z4 can be replaced by a gene selected from the following group: hirudin, rabies virus G protein, dengue virus E protein, Mycobacterium tuberculosis ESAT-6 protein, Ag85A protein, or a combination thereof.
  • Z4 can be replaced by an antigen gene selected from the following group of pathogens: hirudin, cytomegalovirus (CMV), Zika virus (Zika), influenza virus (Influenza), respiratory syncytial virus ( RSV), Chikungunya, Rabies, HIV, Ebola virus, streptococci, malaria, Louping ill virus ), Toxoplasma gondii, dengue fever, plague, yellow fever, tuberculosis, herpes simplex virus, band virus, mycoplasma, chlamydia, foot-and-mouth disease virus, Pseudomonas aeruginosa, or combinations thereof
  • pathogens selected from the following group of pathogens: hirudin, cytomegalovirus (CMV), Zika virus (Zika), influenza virus (Influenza), respiratory syncytial virus ( RSV), Chikungunya, Rabies, HIV, Ebola virus, streptococci, malaria, Louping ill virus
  • Z4 is replaced with hirudin gene or rabies virus G protein gene.
  • the Z4 is codon optimized.
  • the Z4 replacement method includes homologous recombination and enzyme digestion.
  • the stop codon of Z4 is multiple stop codons.
  • the stop codon of Z4 is two stop codons.
  • the Z5 is selected from the following group: a mutated or optimized 3' antibody conserved sequence, a mutated or optimized 3' interferon conserved sequence, or a combination thereof.
  • the Z5 is selected from the following group: SEQ ID NO: 4-6, SEQ ID NO: 10-12, or a combination thereof.
  • the length of Z6 is preferably 100nt-150nt, more preferably 110nt-130nt, more preferably 120nt.
  • a universal UTR element in a third aspect of the present invention, includes:
  • a universal 5'-UTR wherein the sequence of the universal 5'-UTR is selected from the nucleotide sequence shown in SEQ ID NO: 2 or 8 or a derivative sequence thereof;
  • the derivative sequence refers to optionally adding, deleting, modifying and/or substituting at least one (such as 1-3) nuclei to any of the above nucleotide sequences. nucleotides and preserves the derived sequence used to optimize the ability of the mRNA.
  • (a) and (b) can be derived from the same transcript.
  • (a) and (b) can be derived from different transcripts.
  • a carrier which carrier contains the universal scaffold as described in the second aspect of the present invention.
  • the vector is selected from the group consisting of DNA, RNA, viral vectors, plasmids, transposons, other gene transfer systems, or combinations thereof.
  • the vector is a plasmid.
  • the vector is pUC57-Amp vector or pUC-Kana vector.
  • a host cell contains the vector as described in the fourth aspect of the present invention, or the universal framework as described in the second aspect of the present invention is integrated into its genome.
  • the host cells include prokaryotic cells or eukaryotic cells.
  • the host cell is selected from the following group: Escherichia coli, yeast cells, and mammalian cells.
  • an engineered cell contains: the vector as described in the fourth aspect of the present invention, or its genome is integrated with a universal vector as described in the second aspect of the present invention. sexual skeleton and contains the target gene fragment.
  • the engineered cells are stable3 Escherichia coli competent cells.
  • the target gene fragment contains homologous arm sequences.
  • the vector or universal framework contains homologous arm sequences.
  • homologous recombination occurs between the target gene fragment and the vector or universal scaffold.
  • the target gene fragment is connected to the vector or universal scaffold and circularized.
  • the target gene is selected from the following group: hirudin, cytomegalovirus (CMV), Zika virus (Zika), influenza virus (Influenza), respiratory syncytial virus (RSV), chikungunya Chikungunya, Rabies, HIV, Ebola virus, streptococci, malaria, Louping ill virus, Toxoplasma gondii Toxoplasma gondii), dengue fever, plague, yellow fever, tuberculosis vaccine disease, herpes simplex virus, band viruses, mycoplasma, chlamydia, foot-and-mouth disease virus, Pseudomonas aeruginosa, or combinations thereof.
  • a method for producing optimized mRNA for preparing a vaccine comprising the steps:
  • step (d) Optionally, purify and/or modify the optimized mRNA obtained in step (c).
  • a method for preparing an mRNA vaccine includes the steps:
  • kit in a ninth aspect of the present invention, includes:
  • the description also describes a method of using the first plasmid as a template to amplify the first fragment with the homology arm sequence.
  • the description also describes a method of using the second plasmid as a template to amplify a second fragment with a homology arm sequence.
  • the description also describes a method of circularizing the first fragment and the second fragment through homologous recombination and transferring them into a suitable host cell.
  • the description also describes a method for obtaining optimized mRNA from the host cell.
  • the present invention also provides a kit, which includes:
  • an mRNA vaccine composition which vaccine composition contains:
  • the immunogen is selected from the following group: hirudin, cytomegalovirus (CMV), Zika virus (Zika), influenza virus (Influenza), respiratory syncytial virus (RSV), chikungunya Chikungunya, Rabies, HIV, Ebola virus, streptococci, malaria, Louping ill virus, Toxoplasma gondii Toxoplasma gondii), dengue fever, plague, yellow fever, tuberculosis, herpes simplex virus, band viruses, mycoplasma, chlamydia, foot-and-mouth disease virus, Pseudomonas aeruginosa, or combinations thereof.
  • the mRNA itself in the vaccine composition can also serve as an adjuvant.
  • the dosage form of the vaccine composition is selected from the following group: injection and lyophilized agent.
  • the vaccine composition includes 0.01 to 99.99% of the universal framework as described in the second aspect of the present invention and 0.01 to 99.99% of a pharmaceutically acceptable carrier, and the percentage is The mass percentage of the vaccine composition.
  • an mRNA vaccine composition as described in the tenth aspect of the present invention, or the use of engineered cells as described in the sixth aspect of the present invention, which is used to prepare a drug said medicament is used to prevent pathogens selected from the following group: hirudin, cytomegalovirus (CMV), Zika virus (Zika), influenza virus (Influenza), respiratory syncytial virus (RSV), chikungunya disease (Chikungunya), Rabies, HIV, Ebola virus, streptococci, malaria, jumping disease Louping ill virus, Toxoplasma gondii, dengue fever, plague, yellow fever, tuberculosis, herpes simplex virus, band virus, mycoplasma, chlamydia, foot-and-mouth disease virus, Pseudomonas aeruginosa, or combinations thereof.
  • pathogens selected from the following group: hirudin, cytomegalovirus (CMV), Zika virus (Zika), influenza virus
  • Figure 1 is a schematic structural diagram of a carrier in an embodiment of the present invention.
  • Figure 2 shows the sequence IGL-5-O before 5'-UTR optimization, the sequence IGL-5'UTR-F after optimization and their corresponding GC contents.
  • Figure 3 shows the sequence IFN-5-O before 5'-UTR optimization, the sequence IFN-5'UTR-F after optimization and their corresponding GC contents.
  • Figure 4 shows the sequence IGL-3-O before 3'-UTR optimization, the sequence IGL-3'UTR-F after optimization and their corresponding GC contents.
  • Figure 5 shows the sequence INF-3-O before 3'-UTR optimization, the sequence IFN-3'UTR-F after optimization and their corresponding GC contents.
  • Figure 6 is a schematic diagram of inserting two stop codons after the ORF sequence.
  • Figure 7 is a schematic diagram of the hirudin gene synthesis fragment.
  • Figure 8 is a schematic diagram of the backbone amplified fragment.
  • Figure 9 shows the electrophoretic identification results of the insert fragment and the plasmid backbone fragment.
  • Figure 10 shows the results of Hirudin plasmid gel electrophoresis.
  • Figure 11 shows the results of AleI digestion of Hirudin plasmid.
  • Figure 12 shows the changes in plasmid yield during fermentation.
  • Figure 13 shows the expression results of rabies virus G protein after mRNA transfection of cells.
  • Figure 14 shows the agarose gel electrophoresis and enzyme digestion pattern verification results of the universal in vitro transcription template plasmid containing the universal backbone.
  • Figure 15 shows the sequence alignment results of a universal in vitro transcription template plasmid containing a universal scaffold.
  • the inventor developed for the first time a universal UTR and a universal skeleton for constructing mRNA transcripts, thereby obtaining optimized mRNA with improved stability and translation activity. This enables applications in the preparation and/or optimization of mRNA vaccines. Universal use of the present invention The sexual skeleton could speed up the development of mRNA vaccines for a variety of infectious diseases. On this basis, the present invention was completed.
  • general-purpose skeleton of the present invention As used herein, "general-purpose skeleton of the present invention”, “general-purpose skeleton”, “skeleton of the present invention”, “skeleton”, “general-purpose component”, “a series of universal components”, “general-purpose component” “Universal element series” are used interchangeably, and both refer to the framework for constructing mRNA transcripts by replacing the ORF region of the universal UTR element, which is composed of a series of universal element arrangements, typically with the aforementioned formula I The structure shown.
  • UTR conserved sequence of antibody gene and “antibody conserved sequence” can be used interchangeably, both refer to the conserved sequence of the UTR region in the antibody gene, preferably its nucleotide sequence is as shown in SEQ ID NO: 1 or 3 Show.
  • UTR conserved sequence of interferon gene and “interferon conserved sequence” can be used interchangeably, both referring to the conserved sequence of the UTR region in the interferon gene, preferably its nucleotide sequence such as SEQ ID NO:7 Or as shown in 9.
  • the terms "universal skeleton of the invention”, “skeleton of the invention”, “general component”, “general framework”, “a series of universal components”, “general component” “Series” are used interchangeably, and both refer to the universal skeleton composed of a series of universal component arrangements described in the second aspect of the present invention.
  • the general-purpose skeleton of the present invention has the structure of formula I: Z1-Z2-Z3-Z4-Z5-Z6-Z7 (I)
  • Z1 to Z7 are as described above.
  • FIG. 1 A schematic structural diagram of a representative carrier containing the universal scaffold of the present invention is shown in Figure 1.
  • the proteins or polypeptides suitable for expression using the universal framework of the present invention are not particularly limited, including antigenic proteins or antigenic peptides, or other useful proteins.
  • the ORF of the foreign protein can be placed in the universal framework of the present invention, thereby achieving efficient expression.
  • the ORF carries a stop codon.
  • one or more additional stop codons can also be introduced, as shown in Figure 6.
  • mRNA vaccines are divided into self-amplifying RNA (saRNA) and non-amplifying RNA (non-replicating mRNA).
  • Classic non-amplified RNA vaccines include cap, 5'-untranslated regions (5'-UTR), open reading frame (open reading frame, ORF), 3'-untranslated regions (3'- untranslated regions, 3'-UTR) and polyA tail (polyA tail).
  • the ORF region is responsible for encoding antigen expression, but the above five regions Together they determine the stability, expression activity and immunogenicity of mRNA.
  • saRNA The structure of saRNA is derived from the alphavirus genome.
  • the saRNA vaccine utilizes the self-replicating properties of the alphavirus genome to enable the DNA or RNA that enters the body cells to first self-amplify and then transcribe the antigen-encoding mRNA.
  • saRNA vaccines There are currently two types of saRNA vaccines: saRNA based on DNA plasmids and saRNA delivered by virus-like particles.
  • Beissert et al. also developed transgenic amplifying RNA (taRNA), which places the gene encoding the antigen in the alphavirus genome, increasing the safety of the vaccine.
  • taRNA transgenic amplifying RNA
  • non-amplified RNA is smaller, expresses antigens more specifically and does not cause non-specific immunity.
  • a major challenge for mRNA vaccines is reducing the immunogenicity of the exogenous mRNA itself.
  • exogenous mRNA can be recognized by retinoic acid-inducible gene I (RIG-I), activate the innate immune response, and then be degraded.
  • IIG-I retinoic acid-inducible gene I
  • ITT In vitro transcription
  • mRNA can activate immune cells and Toll-like receptor (Toll-like receptor)-mediated inflammatory responses.
  • the U-rich sequence of mRNA is a key factor in activating Toll-like receptors.
  • the immunogenicity of mRNA can be reduced through chemical modification of nucleotides, adding polyA tails, and optimizing the GC content of mRNA.
  • Chemically modified nucleotides include 5-methylcytidine (m5C), 5-methyluridine (m5U), N1-methyladenosine (m1A), N6 -Methyladenosine (N6-methyladenosine, m6A), 2-thiouridine (s2U), 5-oxymethyluridine (5-methoxyuridine, 5moU), pseudouridine (psi) and N1-methylpseudouridine (N1-methylpseudouridine, m1 ⁇ ).
  • adding polyA tail can also reduce the U content and thereby reduce the immunogenicity of mRNA.
  • CureVac and Acuita Therapeutics are trying to transport erythropoietin-encoding mRNA into pigs through lipid nanoparticles.
  • the mRNA has a high GC content and can cause erythropoietin-related reactions without immunogenicity.
  • excessive GC content will inhibit the translation activity of mRNA, which is something that needs to be paid attention to during vaccine development.
  • the purification method of mRNA is also very important in reducing the immunogenicity of mRNA itself.
  • Currently commonly used purification methods include high performance liquid chromatography (HPLC), anion exchange chromatography, affinity chromatography and particle size separation.
  • HPLC high performance liquid chromatography
  • anion exchange chromatography anion exchange chromatography
  • affinity chromatography particle size separation.
  • the purpose of purification is mainly to remove truncated transcripts.
  • Pardi et al. designed to purify m1 ⁇ -modified mRNA encoding anti-HIV-1 antibodies through HPLC through lipid nanoparticles (LNP) to help mice avoid HIV-1 infection.
  • LNP lipid nanoparticles
  • Sequence optimization of mRNA is one of the methods to help stabilize mRNA. Sequence optimization of the 5'-UTR and 3'-UTR of the mRNA can increase the half-life and translation activity of the mRNA. Cap structure using different analogs can To increase the stability of mRNA, the use of enzymes to add a Cap structure to the 5' end of mRNA can have better performance than different forms of Cap analogs. The stabilizing effect of the polyA tail of mRNA is also very important. Some studies have shown that removing the polyA of mRNA makes the mRNA extremely unstable. It also reduces the number of polyribosomes, elongation speed and number of translation rounds of the mRNA. Therefore polyA is crucial for the stable and efficient translation of mRNA.
  • nucleotide modifications and synonymous substitutions of codons can also affect the stability and translation activity of mRNA.
  • sequence optimization may affect the secondary structure and post-translational modification of mRNA.
  • increasing the GC content of mRNA can also increase mRNA stability.
  • 5'-UTR, 3'-UTR, 5'Cap, polyA tail, codon optimization and GC content are all modifiable sites that enhance mRNA stability.
  • LNP lipid nanoparticles
  • mRNA vaccines activate innate and adaptive immunity
  • mRNA vaccines can activate the innate and adaptive immune systems. Direct recognition of mRNA by pattern recognition receptors such as TLRs in somatic cells will lead to the degradation of mRNA and at the same time enhance the IFN pathway.
  • mRNA vaccines play a major role by stimulating adaptive immune responses.
  • the antigen translated from the mRNA is directly presented through MHC-I to activate CD4+ T cells (Helper T cells) or after the antigen is secreted, other cells phagocytose it and present the antigen through the MHC-II pathway to activate CD8+ T cells (cytotoxic T cells).
  • MHC-I CD4+ T cells
  • cytotoxic T cells CD8+ T cells
  • mRNA-containing particles are absorbed by local cells at the injection site, the mRNA is recognized by pattern recognition receptors and also begins to translate the antigen, causing local inflammation at the injection site and promoting the infiltration of immune cells, including neutrophils and monocytes. cells, myeloid dendritic cells (MDCs) and plasmacytoid dendritic cells (PDCs). Neutrophils can efficiently take up LNPs, but monocytes and MDCs translate mRNA more efficiently. The secretion of type I interferon (IFN) is stimulated.
  • IFN type I interferon
  • the mechanism of the mRNA vaccine is to inoculate the mRNA encoding the antigenic protein into the host, and then use the host's genetic material to express and synthesize the antigenic protein in cells in the body.
  • the antigenic protein induces and activates the body's immune system to produce an immune response, thereby achieving prevention. and the purpose of treating disease. Its unique advantages are: 1) Monitoring and quality control of all production processes can be easily realized; 2) The research and development and production cycle of mRNA is short, it is easy to achieve mass production, and the vaccine production capacity is high, which is very important for quickly responding to new emerging diseases worldwide.
  • Infectious diseases are crucial; 3) Due to its own characteristics, mRNA can be degraded quickly after immunization, and the safety risk is low; 4) The immune effect is good, and it can induce both humoral immunity and cellular immunity at the same time, and is effective against infections for which there is currently no better vaccine. There may be potential for more effective vaccines against diseases.
  • the challenges that mRNA vaccines need to face to work include: 1) extending its half-life and enhancing stability; 2) enhancing translation activity; 3) reducing the immunogenicity of mRNA to avoid rapid clearance.
  • the way to achieve these effects is to design special 5'-UTR, 3'-UTR, stop codon, polyA number, etc.
  • 5'-UTR and 3'-UTR there are three effective methods: 1) using the UTR of highly expressed human genes; 2) using the UTR of the antigen protein itself; 3) exponential enrichment ligand system evolution technology ( systematic evolution of ligands by exponential enrichment, SELEX).
  • the first two methods are relatively simple, while the third method is relatively complex and requires continuous trying and optimizing the sequence through in vitro experiments, so it takes a long time. However, the third method is still the best choice when time is sufficient.
  • the 5'-UTR of Pfizer/BioNTech's BNT162b2 vaccine which is currently approved by the FDA, uses the 5'-UTR of human alpha globin and optimizes the Kozak sequence and 5' end sequence, adjusting the second half of the 5'-UTR. hierarchical structure, while the 5'-UTR of Moderna's mRNA-1273 vaccine uses a sequence designed and optimized by its computer.
  • Moderna's mRNA-1273 vaccine uses the 110nt base in the 3'-UTR of human alpha globin (HBA1), while Pfizer's BNT162b2 vaccine uses a SELEX method based on natural genes to select The 3'-UTR of human 12S rRNA (mtRNR1) and AES/TLE5 genes were identified. On this basis, the Pfizer vaccine selected the 136nt sequence of AES 3'-UTR and made two C ⁇ changes, followed by the 139nt mtRNR1 sequence.
  • the truly effective UTR design method is to learn from natural genes and optimize based on experience (PMID: 34358150).
  • Pfizer's BNT162b2 vaccine uses the termination signal UGAUGA, while Moderna's mRNA-1273 vaccine uses UGAUAAUAG.
  • the number of polyA is an important factor affecting the stability of mRNA. 80nt-150n or 100nt-150nt is suitable, and 120nt is better. An appropriate amount of polyA can have higher protein production than an inappropriate amount of mRNA. expression ability and mRNA stability.
  • the framework for constructing mRNA transcripts of the present invention can be used to construct mRNA vaccines against infectious diseases.
  • the vaccine using the skeleton of the mRNA transcript of the present invention can be applied to a variety of different pathogens.
  • Representative pathogens include (but are not limited to): coronavirus (such as new coronavirus), cytomegalovirus (CMV), Zika virus ( Zika), Influenza, Respiratory Syncytial Virus (RSV), Chikungunya, Rabies, HIV, Ebola virus, Streptococci ), malaria, jumping disease virus, Toxoplasma gondii, etc.
  • infectious diseases lack effective inactivated vaccines and recombinant protein vaccines, so it may be hoped that mRNA vaccines can stimulate the body's preventive immunity against these pathogens and then develop effective vaccines.
  • diseases include dengue fever (existing vaccines can only protect one serotype), plague, yellow fever, tuberculosis vaccine, herpes simplex virus, band virus, mycoplasma, chlamydia, foot and mouth disease virus, etc., and can also be used in some anti-tumor drugs of treatment.
  • the inventor used the UTR of the antibody gene as the original UTR.
  • Antibodies are divided into 5 categories (classes), namely IgM, IgD, IgG, IgA and IgE. Their corresponding heavy chains are ⁇ chain, ⁇ chain, ⁇ chain, ⁇ chain and ⁇ chain respectively. Compared with these heavy chains There are two types of coordinated light chains, namely kappa ( ⁇ ) chain and lambda ( ⁇ ) chain. According to the differences in individual amino acids in the constant region of the ⁇ chain, the ⁇ chain can be divided into four subtypes: ⁇ l, ⁇ 2, ⁇ 3 and ⁇ 4. .
  • the recombination rates of the above-mentioned peptide chain genes are very high, suggesting that their 5'-UTR and 3'-UTR sequences may have strong compatibility with changed ORF regions and changed peptide chain expression. Therefore, the 5'-UTR and 3'-UTR of antibody genes may have versatility to support efficient translation of different ORFs.
  • preferred 5'-UTR and 3'-UTR are sequence-optimized UTRs.
  • Figure 2 shows the sequence IGL-5-O before 5'-UTR optimization and the sequence IGL-5'UTR-F after optimization. The GC content increased from 54% to 64% after optimization.
  • Figure 4 shows the sequence IGL-3-O before 3'-UTR optimization and the sequence IGL-3'UTR-F after optimization. The GC content increased from 54% to 56% after optimization.
  • the preferred 5'-UTR based on the sequence optimization of the antibody gene UTR, its nucleotide sequence is shown in SEQ ID NO: 2;
  • the acid sequence is shown in SEQ ID NO: 4, 5 or 6.
  • the inventor In order to improve the versatility of UTR and reduce immunogenicity, the inventor also used the UTR of the gene of immune-related protein interferon, which is widely expressed in the human body, as the original UTR.
  • Interferon is a glycoprotein produced by viruses or other interferon-inducing agents that can be released outside the cells and has broad-spectrum antiviral effects by stimulating intraretinal cells, macrosialocytes, lymphocytes and other cells. It can be stably expressed in a variety of cells, and optimization using its 5'-UTR and 3'-UTR may reduce its immunogenicity and thereby reduce the side effects of mRNA vaccines.
  • preferred 5'-UTR and 3'-UTR are sequence-optimized UTRs.
  • Figure 3 shows the sequence IFN-5-O before 5'-UTR optimization and the sequence IFN-5'UTR-F after optimization. Its GC content increased from 49% to 52% after optimization.
  • Figure 5 shows the sequence INF-3-O before 3'-UTR optimization and the sequence IFN-3'UTR-F after optimization. The GC content increased from 31% to 44% after optimization.
  • the preferred 5'-UTR based on the sequence optimization of the interferon gene UTR its nucleotide sequence is shown in SEQ ID NO: 8; the preferred 3'-UTR based on the sequence optimization of the interferon gene UTR, whose The nucleotide sequence is shown in SEQ ID NO: 10, 11 or 12.
  • the UTR of the antibody and interferon genes as the UTR part of the universal plasmid skeleton, that is, the UTR part of the subsequent generated mRNA, the stability and translation activity of the mRNA are enhanced, mainly to ensure the universality of the plasmid skeleton. and the stability of mRNA vaccines.
  • the inventors designed a plasmid that can replace the ORF, and uses blunt-end or sticky-end restriction sites to facilitate subsequent linearized fragment testing of in vitro transcription.
  • this vector includes a vector backbone, AleI restriction site or BspQI restriction site, 5'-UTR, ORF, 3'-UTR, and polyA.
  • a special competent state was selected and ORF codon optimization was performed.
  • the inventors used high-copy plasmid vector pUC57-Amp or pUC-Kana to amplify the fragments.
  • the high-efficiency blunt-end restriction enzyme AleI or the sticky-end restriction enzyme BspQI is used as an enzyme to cut the inserted fragment from the plasmid.
  • the blunt-end or sticky end it can produce can facilitate subsequent experiments.
  • the promoter selected was T7 promoter.
  • the inventor downloaded the sequences of 10 different antibody peptide chains and different types of interferons from the NCBI database (as shown in Table 1). After comparison, they found out that they are relatively conserved. and a UTR sequence of appropriate length. And based on the natural UTR sequence, the optimized UTR sequence was obtained by optimizing the Kozak sequence and reducing the AT-rich sequence.
  • 5'-UTR Two types of 5'-UTR were selected: antibody conserved sequences and interferon conserved sequences, and Kozak sequence optimization was performed. And the GC content of the 5'-UTR of the antibody and interferon was optimized before vector construction.
  • 3'-UTR Two types of 3'-UTR were selected: antibody conserved sequences and interferon conserved sequences, and AT-rich sequences were eliminated. And the GC content of the 3'-UTR of the antibody and interferon was optimized before vector construction.
  • the translation elongation complex recognizes multiple stop codons at the stop codon, which is beneficial to the depolymerization of the complex and thereby enhances the translation activity of the mRNA, the inventors added two stop codons.
  • the selected polyA length is 120 ⁇ 10nt.
  • the ORF sequence in the plasmid backbone can be easily replaced through homologous recombination or enzyme digestion, which can then be used to construct mRNA vaccines for different antigens.
  • the plasmid backbone fragment was amplified by PCR using the pUC57-Amp vector as a template, as shown in Figure 8. A fragment of the vector backbone of the source arm.
  • the plasmid extracted from the successfully constructed engineering bacteria was subjected to agarose gel electrophoresis.
  • the plasmid extracted from the successfully constructed engineering bacteria was digested with AleI enzyme.
  • the leech plasmid engineered bacteria were passaged for a long time, and part of the bacterial liquid was preserved for several generations to conduct strain identification and detection of bacterial plasmid copy number and bacterial plasmid loss rate.
  • the test results are shown in Table 1-2.
  • the IMVC biochemical test results of the 3rd, 5th, 10th, 15th and 20th generations of bacterial fluids showed that they were typical Escherichia coli; the Gram staining results showed that they were short rod-shaped Gram-negative bacteria without miscellaneous bacterial contamination.
  • bacterial plasmid copy number detection shows that the plasmid copy number of the engineering bacteria is approximately 53.17-240.28 copies/cell.
  • the bacterial plasmid loss rate test showed that no plasmid loss occurred in the engineering bacteria during the passage process.
  • the transcription template plasmid constructed using the universal UTR element of the present invention has a high-level in vitro transcription effect.
  • the gene encoding the rabies virus G protein (GeneBank: GQ918139.1) was inserted into the replaceable ORF plasmid as an ORF to construct recombinant engineering bacteria and plasmids.
  • IVT In vitro transcription
  • the linearized DNA fragment was used as the template for IVT.
  • the amount of synthesized RNA was detected, and the IVT yield was calculated as the multiple of the amount of RNA relative to the amount of DNA template added.
  • the transcription template plasmid constructed by the skeleton in the present invention has high in vitro transcription activity in in vitro transcription.
  • the mRNA constructed using the universal UTR element of the present invention has the ability to express the corresponding protein.
  • Cell transfection experiments were performed using the mRNA prepared in Example 6, which contained a 6 ⁇ His tag at the C terminus of the rabies virus G protein.
  • the amount of mRNA used was 2 ⁇ g/well, and the number of cells in the 6-well plate was 10 6 cells/well.
  • the results show that rabies virus G protein expression exists in the transfected cells, indicating that the mRNA produced by the universal UTR element and skeleton of the present invention has the ability to express the corresponding protein in cells.
  • Example 8 The rabies virus mRNA vaccine constructed using the universal UTR element of the present invention can induce mice to produce extremely high levels of antibodies.
  • the rabies virus G protein encoding gene is used as an ORF to be inserted into the replaceable ORF plasmid (including the universal UTR element of the present invention and the BspQI restriction site), and the high-copy plasmid vector pUC-Kana
  • the replaceable ORF plasmid including the universal UTR element of the present invention and the BspQI restriction site
  • the high-copy plasmid vector pUC-Kana was constructed on the vector skeleton
  • a rabies virus mRNA experimental vaccine was produced based on this recombinant engineered bacterium.
  • lane P1 is the plasmid band
  • lane 1 is the result of ApaLI and PvuII double enzyme digestion.
  • the assay results showed that they were consistent with the expected results of the plasmid design.
  • mice 6-8 weeks old BALB/C mice were used for experiments.
  • the experiment was divided into 4 groups: positive control group, 16 mice (8 males and 8 females), and intramuscular injection of chicken embryo inactivated vaccine (0.6IU/mouse). ); the low-dose group, 16 mice (8 males and 8 females), received a lower dose of mRNA vaccine (5 ⁇ g/mouse) intramuscularly; the high-dose group, 16 mice (8 males and 8 females), received a higher dose intramuscularly. mRNA vaccine (13 ⁇ g/animal); negative control group, no intramuscular injection. Except for the negative control group, two injections were performed on days 0 and 14 to enhance the immune effect, and blood was taken from half of the mice on days 14 and 28 respectively. Serum samples were tested for rabies virus G protein antibody levels by fluorescence focus inhibition method according to the method of "Chinese Pharmacopoeia".
  • the results show that the chicken embryo inactivated vaccine can induce mice to produce antibody levels that are theoretically sufficient to resist rabies virus infection.
  • the rabies virus mRNA vaccine produced more antibodies than the inactivated vaccine under both low-dose and high-dose conditions. High antibody levels.
  • the results indicate that an mRNA vaccine constructed and produced using universal elements can induce a high level of immune response.

Abstract

Provided is a skeleton for constructing an mRNA transcript. Specifically, provided is an application of a universal skeleton for constructing an mRNA transcript in mRNA preparation and/or optimization. The universal skeleton can effectively enhance the stability and translation activity of mRNA, so that the stability of an mRNA vaccine is ensured, and the expression of the mRNA vaccine is optimized.

Description

一种构建mRNA体外转录模板的骨架A scaffold for constructing mRNA in vitro transcription templates 技术领域Technical field
本发明属于生物医学领域,具体涉及一种构建mRNA体外转录模板的骨架及其在mRNA优化设计中的应用。The invention belongs to the field of biomedicine, and specifically relates to a skeleton for constructing an in vitro transcription template of mRNA and its application in the optimized design of mRNA.
背景技术Background technique
mRNA疫苗是一种新的疫苗技术,其通过结合分子生物学和免疫学,将mRNA转导进体细胞中表达外源抗原进而激活宿主的适应性免疫。早在1990年代,科学家们通过注射mRNA进入小鼠体细胞,使小鼠体细胞表达了荧光蛋白、β-半乳糖苷酶和氯霉素乙酰转移酶。1992年,Jirikowski等人向糖尿病尿崩症小鼠注射编码催产素和加压素的mRNA,结果小鼠在注射后的数小时内未发生尿崩症。此后,mRNA疫苗的发展陷入了低谷期。mRNA vaccine is a new vaccine technology that combines molecular biology and immunology to transduce mRNA into somatic cells to express foreign antigens and activate the host's adaptive immunity. As early as the 1990s, scientists injected mRNA into mouse somatic cells, causing the mouse somatic cells to express fluorescent proteins, β-galactosidase and chloramphenicol acetyltransferase. In 1992, Jirikowski et al. injected mRNA encoding oxytocin and vasopressin into diabetic diabetes insipidus mice. As a result, the mice did not develop diabetes insipidus within hours after the injection. Since then, the development of mRNA vaccines has fallen into a trough.
近数十年中,研究者们在实验水平对mRNA疫苗的安全性、表达效率和工业生产等方面进行了研究和优化。这些进步使mRNA疫苗用于肿瘤和病毒感染疾病的预防方面得到优先研发。尤其是近两年新冠病毒的暴发流行使得mRNA疫苗技术的发展和在临床上的应用取得了长足的进步。In recent decades, researchers have studied and optimized the safety, expression efficiency and industrial production of mRNA vaccines at the experimental level. These advances have prioritized the development of mRNA vaccines for the prevention of tumors and viral infections. In particular, the outbreak of the new coronavirus in the past two years has made great progress in the development and clinical application of mRNA vaccine technology.
mRNA疫苗的研发主要是为了提高mRNA的稳定性和翻译活性,并减少mRNA自身抗原性。目前,除新冠病毒疫苗由于研发的紧迫性,而能够上市外,其它mRNA疫苗的研发均受到一定阻碍。究其原因,可能是设计mRNA疫苗时过度关注其表达抗原的能力,而忽视了mRNA疫苗自身的抗原性及高表达能力可能带来的毒性风险和引起周围宿主细胞凋亡的风险。The development of mRNA vaccines is mainly to improve the stability and translation activity of mRNA and reduce the self-antigenicity of mRNA. At present, except for the new coronavirus vaccine, which can be launched due to the urgency of research and development, the research and development of other mRNA vaccines have been hindered to some extent. The reason may be that when designing the mRNA vaccine, too much attention is paid to its ability to express antigens, while the risk of toxicity and the risk of apoptosis of surrounding host cells caused by the antigenicity and high expression ability of the mRNA vaccine itself are ignored.
本领域迫切需要开发一种副作用更低且能加快研发的mRNA疫苗设计方法。There is an urgent need in this field to develop an mRNA vaccine design method with lower side effects and faster development.
发明内容Contents of the invention
本发明的目的在于通过采用免疫相关蛋白的UTR增加mRNA疫苗的泛用性,同时可减少潜在的副作用,借助泛用性的骨架可进一步加快对多种传染性疾病的研发。The purpose of the present invention is to increase the versatility of the mRNA vaccine by using the UTR of immune-related proteins, while reducing potential side effects. The universal framework can further accelerate the research and development of various infectious diseases.
在本发明的第一方面,提供了一种泛用性骨架的用途,所述泛用性骨架用于构建mRNA转录本,其中,所述转录本包括待表达的ORF以及位于所述ORF两侧的5'-UTR区和3'-UTR区,其中所述5'-UTR区和3'-UTR区中1个或2个为泛用性UTR。 In a first aspect of the present invention, there is provided the use of a universal framework for constructing an mRNA transcript, wherein the transcript includes an ORF to be expressed and a pair of genes located on both sides of the ORF. 5'-UTR region and 3'-UTR region, wherein one or two of the 5'-UTR region and 3'-UTR region are universal UTRs.
在另一优选例中,所述的泛用性UTR选自下组:突变的或优化的抗体基因的UTR保守序列、突变的或优化的干扰素基因的UTR保守序列、或其组合。In another preferred embodiment, the universal UTR is selected from the following group: the UTR conserved sequence of a mutated or optimized antibody gene, the UTR conserved sequence of a mutated or optimized interferon gene, or a combination thereof.
在另一优选例中,所述的mRNA转录本包括mRNA疫苗中的mRNA转录本。In another preferred embodiment, the mRNA transcript includes the mRNA transcript in the mRNA vaccine.
在另一优选例中,所述泛用性骨架还包含额外的UTR区(即泛用性UTR之外的其他UTR区)。In another preferred embodiment, the universal framework also includes additional UTR regions (ie, other UTR regions besides the universal UTR).
在另一优选例中,所述抗体基因的UTR保守序列的核苷酸序列如SEQ ID NO:1或3所示。In another preferred embodiment, the nucleotide sequence of the UTR conserved sequence of the antibody gene is shown in SEQ ID NO: 1 or 3.
在另一优选例中,所述抗体基因的UTR保守序列包括5'抗体保守序列和3'抗体保守序列。In another preferred embodiment, the UTR conserved sequence of the antibody gene includes a 5' antibody conserved sequence and a 3' antibody conserved sequence.
在另一优选例中,所述5'抗体保守序列的核苷酸序列如SEQ ID NO:1所示。In another preferred embodiment, the nucleotide sequence of the 5' antibody conserved sequence is shown in SEQ ID NO: 1.
在另一优选例中,所述3'抗体保守序列的核苷酸序列如SEQ ID NO:3所示。In another preferred embodiment, the nucleotide sequence of the 3' antibody conserved sequence is shown in SEQ ID NO: 3.
在另一优选例中,如SEQ ID NO:3所示的序列在不同IGL基因中保守。In another preferred embodiment, the sequence shown in SEQ ID NO:3 is conserved in different IGL genes.
在另一优选例中,所述干扰素基因的UTR保守序列的核苷酸序列如SEQ ID NO:7或9所示。In another preferred embodiment, the nucleotide sequence of the UTR conserved sequence of the interferon gene is shown in SEQ ID NO: 7 or 9.
在另一优选例中,所述干扰素基因的UTR保守序列包括5'干扰素保守序列和3'干扰素保守序列。In another preferred example, the UTR conserved sequence of the interferon gene includes a 5' conserved interferon sequence and a 3' conserved interferon sequence.
在另一优选例中,所述5'干扰素保守序列的核苷酸序列如SEQ ID NO:7所示。In another preferred embodiment, the nucleotide sequence of the 5' interferon conserved sequence is shown in SEQ ID NO: 7.
在另一优选例中,所述3'干扰素保守序列的核苷酸序列如SEQ ID NO:9所示。In another preferred example, the nucleotide sequence of the 3' interferon conserved sequence is shown in SEQ ID NO: 9.
在另一优选例中,如SEQ ID NO:7或9所示的序列在不同IFNA的亚型间保守。In another preferred embodiment, the sequence shown in SEQ ID NO: 7 or 9 is conserved among different IFNA subtypes.
在另一优选例中,所述泛用性UTR包括泛用性5'-UTR,泛用性3'-UTR。In another preferred embodiment, the universal UTR includes a universal 5'-UTR and a universal 3'-UTR.
在另一优选例中,所述泛用性UTR包括:含Kozak序列的5'-UTR。In another preferred embodiment, the universal UTR includes: a 5'-UTR containing a Kozak sequence.
在另一优选例中,所述抗体基因和干扰素基因的3'-UTR中AT-rich序列被全部或部分剔除。In another preferred embodiment, the AT-rich sequence in the 3'-UTR of the antibody gene and interferon gene is completely or partially deleted.
在另一优选例中,所述泛用性UTR中没有或基本没有AT-rich序列。In another preferred embodiment, there is no or substantially no AT-rich sequence in the universal UTR.
在另一优选例中,基本没有AT-rich序列指在一个UTR中,AT-rich序列的数量≤2,更佳地≤1。In another preferred embodiment, basically no AT-rich sequences means that in one UTR, the number of AT-rich sequences is ≤ 2, and more preferably ≤ 1.
在另一优选例中,所述AT-rich序列指富含腺嘌呤和胸腺嘧啶碱基的核酸序列。In another preferred embodiment, the AT-rich sequence refers to a nucleic acid sequence rich in adenine and thymine bases.
在另一优选例中,所述泛用性UTR中的GC含量为44%-64%。In another preferred embodiment, the GC content in the universal UTR is 44%-64%.
在另一优选例中,所述泛用性5'-UTR的核苷酸序列如SEQ ID NO:2或8所示。In another preferred embodiment, the nucleotide sequence of the universal 5'-UTR is shown in SEQ ID NO: 2 or 8.
在另一优选例中,所述泛用性3'-UTR的核苷酸序列如SEQ ID NO:4、5、6、10、11或12所示。In another preferred embodiment, the nucleotide sequence of the universal 3'-UTR is shown in SEQ ID NO: 4, 5, 6, 10, 11 or 12.
在另一优选例中,上述核苷酸序列中任意一种核苷酸序列还包括任选地经过添加、缺失、修饰和/或取代至少一个(如1-3个)核苷酸并能保留用于优化mRNA能力的衍生序列。 In another preferred embodiment, any one of the above nucleotide sequences also includes optionally adding, deleting, modifying and/or replacing at least one (such as 1-3) nucleotides and retaining Derived sequences for optimizing mRNA capacity.
在本发明的第二方面,提供了一种泛用性骨架,所述泛用性骨架具有式I结构:
Z1-Z2-Z3-Z4-Z5-Z6-Z7       (I)
In a second aspect of the present invention, a general-purpose skeleton is provided, and the general-purpose skeleton has the structure of formula I:
Z1-Z2-Z3-Z4-Z5-Z6-Z7 (I)
式中,In the formula,
Z1、Z7为无或酶切位点;Z1 and Z7 are no or enzyme cutting sites;
Z2为无或启动子元件;Z2 is none or promoter element;
Z3为5'-UTR元件;Z3 is a 5'-UTR component;
Z4为可替换的ORF区;Z4 is a replaceable ORF region;
Z5为3'-UTR元件;Z5 is a 3'-UTR component;
Z6为polyA尾部元件。Z6 is a polyA tail component.
在另一优选例中,所述Z1、Z7为平末端酶切位点或粘性末端酶切位点。In another preferred embodiment, Z1 and Z7 are blunt end enzyme cleavage sites or sticky end enzyme cleavage sites.
在另一优选例中,所述泛用性骨架包括酶切位点、启动子、5'-UTR、ORF、3'-UTR和polyA。In another preferred embodiment, the universal framework includes an enzyme cleavage site, a promoter, a 5'-UTR, an ORF, a 3'-UTR and polyA.
在另一优选例中,所述平末端酶选自下组:AleI、AatI、AluI、BavAI、BavBI、EcoRV、MlsI、或其组合。In another preferred embodiment, the blunt-end enzyme is selected from the following group: AelI, AatI, AluI, BavAI, BavBI, EcoRV, MlsI, or a combination thereof.
在另一优选例中,所述平末端酶为AleI。In another preferred embodiment, the blunt-end enzyme is AleI.
在另一优选例中,所述粘性末端酶为BspQI。In another preferred embodiment, the sticky end enzyme is BspQI.
在另一优选例中,所述Z2选自下组:T7启动子、T3启动子、SP6启动子,或其组合。In another preferred embodiment, the Z2 is selected from the following group: T7 promoter, T3 promoter, SP6 promoter, or a combination thereof.
在另一优选例中,所述Z2为T7启动子。In another preferred example, the Z2 is a T7 promoter.
在另一优选例中,所述Z3和Z5中1个或2个为泛用性UTR。In another preferred example, one or two of Z3 and Z5 are universal UTRs.
在另一优选例中,所述Z3选自下组:突变的或优化的5'抗体保守序列、突变的或优化的5'干扰素保守序列、或其组合。In another preferred embodiment, the Z3 is selected from the following group: a mutated or optimized 5' antibody conserved sequence, a mutated or optimized 5' interferon conserved sequence, or a combination thereof.
在另一优选例中,所述Z3选自下组:SEQ ID NO:2、SEQ ID NO:8、人α珠蛋白的5'-UTR、或其组合。In another preferred embodiment, the Z3 is selected from the following group: SEQ ID NO: 2, SEQ ID NO: 8, the 5'-UTR of human α-globin, or a combination thereof.
在另一优选例中,所述Z4可替换为选自下组的基因:水蛭素、狂犬病毒G蛋白、登革热病毒E蛋白、结核分支杆菌ESAT-6蛋白、Ag85A蛋白、或其组合。In another preferred example, the Z4 can be replaced by a gene selected from the following group: hirudin, rabies virus G protein, dengue virus E protein, Mycobacterium tuberculosis ESAT-6 protein, Ag85A protein, or a combination thereof.
在另一优选例中,所述Z4可替换为选自下组病原的抗原基因:水蛭素、巨细胞病毒(CMV)、寨卡病毒(Zika)、流感病毒(Influenza)、呼吸道合胞病毒(RSV)、基孔肯雅病(Chikungunya)、狂犬病(Rabies)、艾滋病毒(HIV)、埃博拉病毒(Ebola virus)、链球菌(streptococci)、疟疾(malaria)、跳跃病病毒(Louping ill virus)、岗地弓形虫(Toxoplasma gondii)、登革热、鼠疫、黄热病、结核病、单纯疱疹病毒、带状病毒、支原体、衣原体、口蹄疫病毒、绿脓杆菌、或其组合In another preferred embodiment, Z4 can be replaced by an antigen gene selected from the following group of pathogens: hirudin, cytomegalovirus (CMV), Zika virus (Zika), influenza virus (Influenza), respiratory syncytial virus ( RSV), Chikungunya, Rabies, HIV, Ebola virus, streptococci, malaria, Louping ill virus ), Toxoplasma gondii, dengue fever, plague, yellow fever, tuberculosis, herpes simplex virus, band virus, mycoplasma, chlamydia, foot-and-mouth disease virus, Pseudomonas aeruginosa, or combinations thereof
在另一优选例中,所述Z4被替换为水蛭素基因或狂犬病毒G蛋白基因。 In another preferred example, Z4 is replaced with hirudin gene or rabies virus G protein gene.
在另一优选例中,所述Z4经过密码子优化。In another preferred example, the Z4 is codon optimized.
在另一优选例中,所述Z4的替换方法包括同源重组和酶切。In another preferred embodiment, the Z4 replacement method includes homologous recombination and enzyme digestion.
在另一优选例中,所述Z4的终止密码子为多个终止密码子。In another preferred example, the stop codon of Z4 is multiple stop codons.
在另一优选例中,所述Z4的终止密码子为2个终止密码子。In another preferred example, the stop codon of Z4 is two stop codons.
在另一优选例中,所述Z5选自下组:突变的或优化的3'抗体保守序列、突变的或优化的3'干扰素保守序列、或其组合。In another preferred embodiment, the Z5 is selected from the following group: a mutated or optimized 3' antibody conserved sequence, a mutated or optimized 3' interferon conserved sequence, or a combination thereof.
在另一优选例中,所述Z5选自下组:SEQ ID NO:4-6、SEQ ID NO:10-12,或其组合。In another preferred example, the Z5 is selected from the following group: SEQ ID NO: 4-6, SEQ ID NO: 10-12, or a combination thereof.
在另一优选例中,所述Z6的长度较佳地为100nt-150nt,更佳地为110nt-130nt,更佳地为120nt。In another preferred example, the length of Z6 is preferably 100nt-150nt, more preferably 110nt-130nt, more preferably 120nt.
在本发明的第三方面,提供了一种泛用性UTR元件,所述泛用性UTR元件包括:In a third aspect of the present invention, a universal UTR element is provided, and the universal UTR element includes:
(a)泛用性5'-UTR,其中所述泛用性5'-UTR的序列选自如SEQ ID NO:2或8所示的核苷酸序列或其衍生序列;和/或(a) a universal 5'-UTR, wherein the sequence of the universal 5'-UTR is selected from the nucleotide sequence shown in SEQ ID NO: 2 or 8 or a derivative sequence thereof; and/or
(b)泛用性3'-UTR,其中所述泛用性3'-UTR的序列选自如SEQ ID NO:4、5、6、10、11或12所示的核苷酸序列或其衍生序列。(b) Universal 3'-UTR, wherein the sequence of the universal 3'-UTR is selected from the nucleotide sequence shown in SEQ ID NO: 4, 5, 6, 10, 11 or 12 or derivatives thereof sequence.
在另一优选例中,所述衍生序列是指对上述核苷酸序列中任意一种核苷酸序列任选地经过添加、缺失、修饰和/或取代至少一个(如1-3个)核苷酸并能保留用于优化mRNA能力的衍生序列。In another preferred embodiment, the derivative sequence refers to optionally adding, deleting, modifying and/or substituting at least one (such as 1-3) nuclei to any of the above nucleotide sequences. nucleotides and preserves the derived sequence used to optimize the ability of the mRNA.
在另一优选例中,所述(a)和(b)可来源于同一转录本。In another preferred embodiment, (a) and (b) can be derived from the same transcript.
在另一优选例中,所述(a)和(b)可来源于不同转录本。In another preferred embodiment, (a) and (b) can be derived from different transcripts.
在本发明的第四方面,提供了一种载体,所述载体含有如本发明第二方面所述的泛用性骨架。In the fourth aspect of the present invention, a carrier is provided, which carrier contains the universal scaffold as described in the second aspect of the present invention.
在另一优选例中,所述载体选自下组:DNA、RNA、病毒载体、质粒、转座子、其他基因转移系统,或其组合。优选地,所述载体为质粒。In another preferred embodiment, the vector is selected from the group consisting of DNA, RNA, viral vectors, plasmids, transposons, other gene transfer systems, or combinations thereof. Preferably, the vector is a plasmid.
在另一优选例中,所述载体为pUC57-Amp载体或pUC-Kana载体。In another preferred embodiment, the vector is pUC57-Amp vector or pUC-Kana vector.
在本发明的第五方面,提供了一种宿主细胞,所述宿主细胞含有如本发明第四方面所述的载体,或其基因组中整合有如本发明第二方面所述的泛用性骨架。In the fifth aspect of the present invention, a host cell is provided, the host cell contains the vector as described in the fourth aspect of the present invention, or the universal framework as described in the second aspect of the present invention is integrated into its genome.
在另一优选例中,所述的宿主细胞包括原核细胞或真核细胞。In another preferred embodiment, the host cells include prokaryotic cells or eukaryotic cells.
在另一优选例中,所述的宿主细胞选自下组:大肠杆菌、酵母细胞、哺乳动物细胞。 In another preferred embodiment, the host cell is selected from the following group: Escherichia coli, yeast cells, and mammalian cells.
在本发明的第六方面,提供了一种工程化细胞,所述工程化细胞含有:如本发明第四方面所述的载体,或其基因组中整合有如本发明第二方面所述的泛用性骨架,并且含有目的基因片段。In the sixth aspect of the present invention, an engineered cell is provided. The engineered cell contains: the vector as described in the fourth aspect of the present invention, or its genome is integrated with a universal vector as described in the second aspect of the present invention. sexual skeleton and contains the target gene fragment.
在另一优选例中,所述工程化细胞为stable3大肠杆菌感受态细胞。In another preferred embodiment, the engineered cells are stable3 Escherichia coli competent cells.
在另一优选例中,所述目的基因片段含有同源臂序列。In another preferred embodiment, the target gene fragment contains homologous arm sequences.
在另一优选例中,所述载体或泛用性骨架含有同源臂序列。In another preferred embodiment, the vector or universal framework contains homologous arm sequences.
在另一优选例中,所述目的基因片段与所述载体或泛用性骨架发生同源重组。In another preferred embodiment, homologous recombination occurs between the target gene fragment and the vector or universal scaffold.
在另一优选例中,所述目的基因片段与所述载体或泛用性骨架连接环化。In another preferred embodiment, the target gene fragment is connected to the vector or universal scaffold and circularized.
在另一优选例中,所述目的基因选自下组:水蛭素、巨细胞病毒(CMV)、寨卡病毒(Zika)、流感病毒(Influenza)、呼吸道合胞病毒(RSV)、基孔肯雅病(Chikungunya)、狂犬病(Rabies)、艾滋病毒(HIV)、埃博拉病毒(Ebola virus)、链球菌(streptococci)、疟疾(malaria)、跳跃病病毒(Louping ill virus)、岗地弓形虫(Toxoplasma gondii)、登革热、鼠疫、黄热病、结核疫苗病、单纯疱疹病毒、带状病毒、支原体、衣原体、口蹄疫病毒、绿脓杆菌、或其组合。In another preferred embodiment, the target gene is selected from the following group: hirudin, cytomegalovirus (CMV), Zika virus (Zika), influenza virus (Influenza), respiratory syncytial virus (RSV), chikungunya Chikungunya, Rabies, HIV, Ebola virus, streptococci, malaria, Louping ill virus, Toxoplasma gondii Toxoplasma gondii), dengue fever, plague, yellow fever, tuberculosis vaccine disease, herpes simplex virus, band viruses, mycoplasma, chlamydia, foot-and-mouth disease virus, Pseudomonas aeruginosa, or combinations thereof.
在本发明的第七方面,提供了一种产生用于制备疫苗的优化mRNA的方法,包括步骤:In a seventh aspect of the present invention, a method for producing optimized mRNA for preparing a vaccine is provided, comprising the steps:
(a)在适合的条件下,培养如本发明第六方面所述的工程化细胞,从而获得含有转录DNA模板的载体的培养物;(a) Under appropriate conditions, culture the engineered cells as described in the sixth aspect of the present invention, thereby obtaining a culture containing a vector for transcribing DNA templates;
(b)从所述培养物中分离和/或回收(a)中所述载体,并酶切线性化为DNA模板;(b) Isolating and/or recovering the vector described in (a) from the culture, and enzymatically digesting and linearizing it into a DNA template;
(c)将(b)中所述DNA模板进行转录,从而获得所述优化mRNA;和(c) transcribing the DNA template described in (b) to obtain the optimized mRNA; and
(d)任选地,对步骤(c)获得的优化mRNA进行纯化和/或修饰。(d) Optionally, purify and/or modify the optimized mRNA obtained in step (c).
在本发明的第八方面,提供了一种mRNA疫苗的制备方法,所述方法包括步骤:In an eighth aspect of the present invention, a method for preparing an mRNA vaccine is provided, which method includes the steps:
(i)通过如本发明第七方面所述的方法获得优化mRNA;(i) Obtain optimized mRNA by the method described in the seventh aspect of the present invention;
(ii)将(i)中获得的优化mRNA与药学上可接受的载体混合,从而获得所述mRNA疫苗。(ii) Mix the optimized mRNA obtained in (i) with a pharmaceutically acceptable carrier to obtain the mRNA vaccine.
在本发明的第九方面,提供了一种试剂盒,所述试剂盒包括:In a ninth aspect of the present invention, a kit is provided, which kit includes:
(a)第一质粒或其线性化DNA片段,所述第一质粒或其线性化DNA片段含有目的基因;和(a) a first plasmid or a linearized DNA fragment thereof, which contains the gene of interest; and
(b)第二质粒,所述第二质粒含有如本发明第二方面所述的泛用性骨架;和(b) a second plasmid containing a universal scaffold according to the second aspect of the invention; and
(c)说明书,所述说明书描述了使用所述第一质粒或其线性化DNA片段和第二质粒生产可用于制备疫苗的优化mRNA的方法。 (c) Instructions describing a method of using said first plasmid or a linearized DNA fragment thereof and a second plasmid to produce optimized mRNA useful in the preparation of a vaccine.
在另一优选例中,所述说明书中还记载了使用第一质粒为模板扩增出带有同源臂序列的第一片段的方法。In another preferred embodiment, the description also describes a method of using the first plasmid as a template to amplify the first fragment with the homology arm sequence.
在另一优选例中,所述说明书中还记载了使用第二质粒为模板扩增出带有同源臂序列的第二片段的方法。In another preferred embodiment, the description also describes a method of using the second plasmid as a template to amplify a second fragment with a homology arm sequence.
在另一优选例中,所述说明书中还记载了将所述第一片段和所述第二片段通过同源重组连接环化并转入一合适的宿主细胞的方法。In another preferred embodiment, the description also describes a method of circularizing the first fragment and the second fragment through homologous recombination and transferring them into a suitable host cell.
在另一优选例中,所述说明书中还记载了从所述宿主细胞获得优化mRNA的方法。In another preferred embodiment, the description also describes a method for obtaining optimized mRNA from the host cell.
在另一优选例中,本发明还提供了一种试剂盒,所述试剂盒包括:In another preferred embodiment, the present invention also provides a kit, which includes:
(Z1)质粒,所述质粒含有如本发明第二方面所述的泛用性骨架;和(Z1) a plasmid containing a universal scaffold as described in the second aspect of the present invention; and
(Z2)说明书,所述说明书描述了使用所述质粒生产可用于制备疫苗的优化mRNA的方法。(Z2) Instructions describing a method of using said plasmid to produce optimized mRNA useful in the preparation of a vaccine.
在本发明的第十方面,提供了一种mRNA疫苗组合物,所述疫苗组合物含有:In a tenth aspect of the present invention, an mRNA vaccine composition is provided, which vaccine composition contains:
(a)用于表达免疫原的mRNA,所述mRNA包含如本发明第二方面所述的泛用性骨架;和(a) mRNA for expressing an immunogen, said mRNA comprising a general scaffold as described in the second aspect of the invention; and
(b)药学上可接受的载体。(b) Pharmaceutically acceptable carrier.
在另一优选例中,所述免疫原选自下组:水蛭素、巨细胞病毒(CMV)、寨卡病毒(Zika)、流感病毒(Influenza)、呼吸道合胞病毒(RSV)、基孔肯雅病(Chikungunya)、狂犬病(Rabies)、艾滋病毒(HIV)、埃博拉病毒(Ebola virus)、链球菌(streptococci)、疟疾(malaria)、跳跃病病毒(Louping ill virus)、岗地弓形虫(Toxoplasma gondii)、登革热、鼠疫、黄热病、结核病、单纯疱疹病毒、带状病毒、支原体、衣原体、口蹄疫病毒、绿脓杆菌,或其组合。In another preferred embodiment, the immunogen is selected from the following group: hirudin, cytomegalovirus (CMV), Zika virus (Zika), influenza virus (Influenza), respiratory syncytial virus (RSV), chikungunya Chikungunya, Rabies, HIV, Ebola virus, streptococci, malaria, Louping ill virus, Toxoplasma gondii Toxoplasma gondii), dengue fever, plague, yellow fever, tuberculosis, herpes simplex virus, band viruses, mycoplasma, chlamydia, foot-and-mouth disease virus, Pseudomonas aeruginosa, or combinations thereof.
在另一优选例中,所述疫苗组合物中mRNA本身还可以充当佐剂。In another preferred embodiment, the mRNA itself in the vaccine composition can also serve as an adjuvant.
在另一优选例中,所述疫苗组合物的剂型选自下组:注射剂、冻干剂。In another preferred embodiment, the dosage form of the vaccine composition is selected from the following group: injection and lyophilized agent.
在另一优选例中,所述疫苗组合物包括0.01~99.99%的如本发明第二方面所述的泛用性骨架和0.01~99.99%的药学上可接受的载体,所述百分比为占所述疫苗组合物的质量百分比。In another preferred embodiment, the vaccine composition includes 0.01 to 99.99% of the universal framework as described in the second aspect of the present invention and 0.01 to 99.99% of a pharmaceutically acceptable carrier, and the percentage is The mass percentage of the vaccine composition.
在本发明的第十一方面,提供了一种如本发明第十方面所述的mRNA疫苗组合物,或如本发明第六方面所述的工程化细胞的用途,其被用于制备一药物,所述药物用于预防选自下组的病原体:水蛭素、巨细胞病毒(CMV)、寨卡病毒(Zika)、流感病毒(Influenza)、呼吸道合胞病毒(RSV)、基孔肯雅病(Chikungunya)、狂犬病(Rabies)、艾滋病毒(HIV)、埃博拉病毒(Ebola virus)、链球菌(streptococci)、疟疾(malaria)、跳跃病 病毒(Louping ill virus)、岗地弓形虫(Toxoplasma gondii)、登革热、鼠疫、黄热病、结核病、单纯疱疹病毒、带状病毒、支原体、衣原体、口蹄疫病毒、绿脓杆菌,或其组合。In the eleventh aspect of the present invention, there is provided an mRNA vaccine composition as described in the tenth aspect of the present invention, or the use of engineered cells as described in the sixth aspect of the present invention, which is used to prepare a drug , said medicament is used to prevent pathogens selected from the following group: hirudin, cytomegalovirus (CMV), Zika virus (Zika), influenza virus (Influenza), respiratory syncytial virus (RSV), chikungunya disease (Chikungunya), Rabies, HIV, Ebola virus, streptococci, malaria, jumping disease Louping ill virus, Toxoplasma gondii, dengue fever, plague, yellow fever, tuberculosis, herpes simplex virus, band virus, mycoplasma, chlamydia, foot-and-mouth disease virus, Pseudomonas aeruginosa, or combinations thereof.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。It should be understood that within the scope of the present invention, the above-mentioned technical features of the present invention and the technical features specifically described below (such as embodiments) can be combined with each other to form new or preferred technical solutions. Due to space limitations, they will not be described one by one here.
附图说明Description of the drawings
图1为本发明实施例中的载体的结构示意图。Figure 1 is a schematic structural diagram of a carrier in an embodiment of the present invention.
图2显示了5'-UTR优化前的序列IGL-5-O、优化后的序列IGL-5'UTR-F及它们对应的GC含量。Figure 2 shows the sequence IGL-5-O before 5'-UTR optimization, the sequence IGL-5'UTR-F after optimization and their corresponding GC contents.
图3显示了5'-UTR优化前的序列IFN-5-O、优化后的序列IFN-5'UTR-F及它们对应的GC含量。Figure 3 shows the sequence IFN-5-O before 5'-UTR optimization, the sequence IFN-5'UTR-F after optimization and their corresponding GC contents.
图4显示了3'-UTR优化前的序列IGL-3-O、优化后的序列IGL-3'UTR-F及它们对应的GC含量。Figure 4 shows the sequence IGL-3-O before 3'-UTR optimization, the sequence IGL-3'UTR-F after optimization and their corresponding GC contents.
图5显示了3'-UTR优化前的序列INF-3-O、优化后的序列IFN-3'UTR-F及它们对应的GC含量。Figure 5 shows the sequence INF-3-O before 3'-UTR optimization, the sequence IFN-3'UTR-F after optimization and their corresponding GC contents.
图6为ORF序列后插入2个终止密码子的示意图。Figure 6 is a schematic diagram of inserting two stop codons after the ORF sequence.
图7为水蛭素基因合成片段示意图。Figure 7 is a schematic diagram of the hirudin gene synthesis fragment.
图8为骨架扩增片段示意图。Figure 8 is a schematic diagram of the backbone amplified fragment.
图9显示了插入片段与质粒骨架片段的电泳鉴定结果。Figure 9 shows the electrophoretic identification results of the insert fragment and the plasmid backbone fragment.
图10显示了Hirudin质粒凝胶电泳结果。Figure 10 shows the results of Hirudin plasmid gel electrophoresis.
图11显示了Hirudin质粒AleI酶切结果。Figure 11 shows the results of AleI digestion of Hirudin plasmid.
图12显示了发酵过程中质粒得率变化。Figure 12 shows the changes in plasmid yield during fermentation.
图13显示了mRNA转染细胞后狂犬病毒G蛋白表达结果。Figure 13 shows the expression results of rabies virus G protein after mRNA transfection of cells.
图14显示了含有泛用性骨架的泛用型体外转录模板质粒的琼脂糖凝胶电泳及酶切图谱检定结果。Figure 14 shows the agarose gel electrophoresis and enzyme digestion pattern verification results of the universal in vitro transcription template plasmid containing the universal backbone.
图15显示了含有泛用性骨架的泛用型体外转录模板质粒的序列比对结果。Figure 15 shows the sequence alignment results of a universal in vitro transcription template plasmid containing a universal scaffold.
具体实施方式Detailed ways
本发明人经过广泛而深入的研究,并经过大量的筛选,首次开发了一种泛用性UTR和泛用性骨架,用于构建mRNA转录本,从而获得稳定性和翻译活性提高的优化mRNA,从而实现在mRNA疫苗制备和/或优化等方面的应用。使用本发明的泛用 性骨架可加快对多种传染性疾病的mRNA疫苗研发。在此基础上完成了本发明。After extensive and in-depth research and extensive screening, the inventor developed for the first time a universal UTR and a universal skeleton for constructing mRNA transcripts, thereby obtaining optimized mRNA with improved stability and translation activity. This enables applications in the preparation and/or optimization of mRNA vaccines. Universal use of the present invention The sexual skeleton could speed up the development of mRNA vaccines for a variety of infectious diseases. On this basis, the present invention was completed.
术语the term
为了可以更容易地理解本公开,首先定义某些术语。如本申请中所使用的,除非本文另有明确规定,否则以下术语中的每一个应具有下面给出的含义。In order that this disclosure may be more easily understood, certain terms are first defined. As used in this application, each of the following terms shall have the meaning given below unless expressly stated otherwise herein.
如本文所用,“本发明的泛用性骨架”、“泛用性骨架”、“本发明的骨架”、“骨架”、“泛用型元件”、“一系列泛用型元件”、“泛用型元件系列”可互换使用,均指包含泛用性UTR元件的ORF区可替换的用于构建mRNA转录本的骨架,其由一系列泛用型元件排列组成,典型地具有前述式I所示的结构。As used herein, "general-purpose skeleton of the present invention", "general-purpose skeleton", "skeleton of the present invention", "skeleton", "general-purpose component", "a series of universal components", "general-purpose component" "Universal element series" are used interchangeably, and both refer to the framework for constructing mRNA transcripts by replacing the ORF region of the universal UTR element, which is composed of a series of universal element arrangements, typically with the aforementioned formula I The structure shown.
如本文所用,“抗体基因的UTR保守序列”、“抗体保守序列”可互换使用,均指抗体基因中UTR区的保守序列,优选地其核苷酸序列如SEQ ID NO:1或3所示。As used herein, "UTR conserved sequence of antibody gene" and "antibody conserved sequence" can be used interchangeably, both refer to the conserved sequence of the UTR region in the antibody gene, preferably its nucleotide sequence is as shown in SEQ ID NO: 1 or 3 Show.
如本文所用,“干扰素基因的UTR保守序列”、“干扰素保守序列”可互换使用,均指干扰素基因中UTR区的保守序列,优选地其核苷酸序列如SEQ ID NO:7或9所示。As used herein, "UTR conserved sequence of interferon gene" and "interferon conserved sequence" can be used interchangeably, both referring to the conserved sequence of the UTR region in the interferon gene, preferably its nucleotide sequence such as SEQ ID NO:7 Or as shown in 9.
泛用性骨架及表达载体Universal scaffolds and expression vectors
如本文所用,术语“本发明的泛用性骨架”、“本发明的骨架”、“泛用型元件”、“泛用性骨架”、“一系列泛用型元件”、“泛用型元件系列”等可互换使用,均指本发明第二方面中所述的由一系列泛用型元件排列组成的泛用性骨架。As used herein, the terms "universal skeleton of the invention", "skeleton of the invention", "general component", "general framework", "a series of universal components", "general component" "Series" are used interchangeably, and both refer to the universal skeleton composed of a series of universal component arrangements described in the second aspect of the present invention.
典型地,本发明的泛用性骨架具有式I结构:
Z1-Z2-Z3-Z4-Z5-Z6-Z7       (I)
Typically, the general-purpose skeleton of the present invention has the structure of formula I:
Z1-Z2-Z3-Z4-Z5-Z6-Z7 (I)
其中,Z1~Z7如上所述。Among them, Z1 to Z7 are as described above.
一种代表性的含有本发明泛用性骨架的载体的结构示意图如图1所示。A schematic structural diagram of a representative carrier containing the universal scaffold of the present invention is shown in Figure 1.
应理解,适合用本发明的泛用性骨架表达的蛋白或多肽没有特别限制,包括抗原蛋白或抗原肽,或其他有用的蛋白。在本发明中,可将外源蛋白的ORF置于本发明泛用性骨架之中,从而实现高效的表达。通常,ORF带有一个终止密码子。然而,如果需要,也可以引入一个或多个额外的终止密码子,如图6所示。It should be understood that the proteins or polypeptides suitable for expression using the universal framework of the present invention are not particularly limited, including antigenic proteins or antigenic peptides, or other useful proteins. In the present invention, the ORF of the foreign protein can be placed in the universal framework of the present invention, thereby achieving efficient expression. Typically, the ORF carries a stop codon. However, if desired, one or more additional stop codons can also be introduced, as shown in Figure 6.
mRNA疫苗类型mRNA vaccine types
mRNA疫苗分为自我扩增RNA(self-amplifying RNA,saRNA)和非扩增RNA(non-replicating mRNA)。经典的非扩增RNA疫苗,包括cap帽子、5'非编码区(5'-untranslated regions,5'-UTR)、开放阅读框(open reading frame,ORF)、3'非编码区(3'-untranslated regions,3'-UTR)和多聚A尾(polyA tail)。ORF区负责编码抗原表达,但以上5个区域 共同决定mRNA的稳定性、表达活性和免疫原性。mRNA vaccines are divided into self-amplifying RNA (saRNA) and non-amplifying RNA (non-replicating mRNA). Classic non-amplified RNA vaccines include cap, 5'-untranslated regions (5'-UTR), open reading frame (open reading frame, ORF), 3'-untranslated regions (3'- untranslated regions, 3'-UTR) and polyA tail (polyA tail). The ORF region is responsible for encoding antigen expression, but the above five regions Together they determine the stability, expression activity and immunogenicity of mRNA.
而saRNA的结构来源于α病毒基因组。saRNA疫苗利用α病毒的基因组能够自我复制的特性使进入体细胞的DNA或RNA先自我扩增,再转录出抗原编码mRNA。saRNA疫苗目前有以DNA质粒为基础的saRNA和病毒样颗粒递送的saRNA两种。基于saRNA,Beissert等人还发展了转基因扩增RNA(trans-amplifying RNA,taRNA),其将编码抗原的基因放在α病毒基因组中,增加了疫苗的安全性。与自我扩增RNA相比,非扩增RNA具有更小,表达抗原更特异不引起非特异性免疫的特点。The structure of saRNA is derived from the alphavirus genome. The saRNA vaccine utilizes the self-replicating properties of the alphavirus genome to enable the DNA or RNA that enters the body cells to first self-amplify and then transcribe the antigen-encoding mRNA. There are currently two types of saRNA vaccines: saRNA based on DNA plasmids and saRNA delivered by virus-like particles. Based on saRNA, Beissert et al. also developed transgenic amplifying RNA (taRNA), which places the gene encoding the antigen in the alphavirus genome, increasing the safety of the vaccine. Compared with self-amplifying RNA, non-amplified RNA is smaller, expresses antigens more specifically and does not cause non-specific immunity.
mRNA疫苗免疫原性mRNA vaccine immunogenicity
mRNA疫苗的一大挑战是减少外源mRNA本身的免疫原性。自然情况下,外源mRNA进入细胞后,能够被维甲酸诱导基因I(retinoic acid-iinducible gene I,RIG-I)识别,激活固有免疫反应,进而被降解。体外转录(in vitro transcription,IVT)mRNA能够激活免疫细胞和Toll样受体(Toll-like receptor)介导的炎症反应。mRNA的U富集(U-rich)序列是激活Toll样受体的关键因素。通过核苷酸化学修饰、添加polyA尾和优化mRNA GC含量等均能够减少mRNA的免疫原性。A major challenge for mRNA vaccines is reducing the immunogenicity of the exogenous mRNA itself. Under natural circumstances, after exogenous mRNA enters cells, it can be recognized by retinoic acid-inducible gene I (RIG-I), activate the innate immune response, and then be degraded. In vitro transcription (IVT) mRNA can activate immune cells and Toll-like receptor (Toll-like receptor)-mediated inflammatory responses. The U-rich sequence of mRNA is a key factor in activating Toll-like receptors. The immunogenicity of mRNA can be reduced through chemical modification of nucleotides, adding polyA tails, and optimizing the GC content of mRNA.
化学修饰的核苷酸包括5-甲基胞苷(5-methylcytidine,m5C)、5-甲基尿苷(5-methyluridine,m5U)、N1-甲基腺苷(N1-methyladenosine,m1A)、N6-甲基腺苷(N6-methyladenosine,m6A)、2-硫尿核苷(2-thiouridine,s2U)、5-氧甲基尿苷(5-methoxyuridine,5moU)、假尿苷(pseudouridine,ψ)和N1-甲基假尿苷(N1-methylpseudouridine,m1ψ)。Chemically modified nucleotides include 5-methylcytidine (m5C), 5-methyluridine (m5U), N1-methyladenosine (m1A), N6 -Methyladenosine (N6-methyladenosine, m6A), 2-thiouridine (s2U), 5-oxymethyluridine (5-methoxyuridine, 5moU), pseudouridine (psi) and N1-methylpseudouridine (N1-methylpseudouridine, m1ψ).
此外,添加polyA尾也能减少U含量进而减小mRNA的免疫原性。CureVac和Acuita Therapeutics公司尝试通过脂质纳米颗粒运输红细胞生成素编码mRNA进入猪体内,该mRNA具有较高GC含量,结果能够引起红细胞生成素相关反应而没有免疫原性。然而,过高的GC含量会抑制mRNA的翻译活性,这也是疫苗研发过程中需要注意的。In addition, adding polyA tail can also reduce the U content and thereby reduce the immunogenicity of mRNA. CureVac and Acuita Therapeutics are trying to transport erythropoietin-encoding mRNA into pigs through lipid nanoparticles. The mRNA has a high GC content and can cause erythropoietin-related reactions without immunogenicity. However, excessive GC content will inhibit the translation activity of mRNA, which is something that needs to be paid attention to during vaccine development.
mRNA的纯化方式在减少mRNA自身的免疫原性中也相当重要。目前常用的纯化方法包括高效液相色谱法(high performance liquid chromatography,HPLC)、阴离子交换色谱法、亲和色谱法和粒子大小分离法。纯化的目的主要是去除截短的转录本。一个好的例子是Pardi等人设计的通过HPLC纯化m1ψ修饰的编码抗HIV-1抗体的mRNA通过脂质纳米颗粒(lipid nanoparticles,LNP)帮助小鼠避免了HIV-1的感染。The purification method of mRNA is also very important in reducing the immunogenicity of mRNA itself. Currently commonly used purification methods include high performance liquid chromatography (HPLC), anion exchange chromatography, affinity chromatography and particle size separation. The purpose of purification is mainly to remove truncated transcripts. A good example is that Pardi et al. designed to purify m1ψ-modified mRNA encoding anti-HIV-1 antibodies through HPLC through lipid nanoparticles (LNP) to help mice avoid HIV-1 infection.
mRNA的序列优化Sequence optimization of mRNA
mRNA的序列优化是帮助mRNA稳定的方法之一。mRNA的5'-UTR和3'-UTR的序列优化能够增加mRNA的半衰期和翻译活性。Cap结构采用不同的类似物能够 增加mRNA的稳定性,利用酶使mRNA的5'端加上Cap结构能够比不同形式的Cap类似物有更好的效能。mRNA的polyA尾的稳定mRNA效果也是非常重要的,有研究去除了mRNA的polyA使得mRNA极度不稳定,同时也降低了mRNA的多聚核糖体数目、延伸速度和翻译轮数。因而polyA对mRNA的稳定和有效翻译至关重要。另外,核苷酸的修饰和密码子的同义替换也能影响mRNA的稳定性和翻译活性。同时序列的优化可能影响mRNA的二级结构和翻译后修饰。此外,增加mRNA的GC含量也能增加mRNA稳定性。综上所述,5'-UTR、3'-UTR、5'Cap、polyA尾、密码子优化和GC含量是增强mRNA稳定性的所有可调节位点。Sequence optimization of mRNA is one of the methods to help stabilize mRNA. Sequence optimization of the 5'-UTR and 3'-UTR of the mRNA can increase the half-life and translation activity of the mRNA. Cap structure using different analogs can To increase the stability of mRNA, the use of enzymes to add a Cap structure to the 5' end of mRNA can have better performance than different forms of Cap analogs. The stabilizing effect of the polyA tail of mRNA is also very important. Some studies have shown that removing the polyA of mRNA makes the mRNA extremely unstable. It also reduces the number of polyribosomes, elongation speed and number of translation rounds of the mRNA. Therefore polyA is crucial for the stable and efficient translation of mRNA. In addition, nucleotide modifications and synonymous substitutions of codons can also affect the stability and translation activity of mRNA. At the same time, sequence optimization may affect the secondary structure and post-translational modification of mRNA. In addition, increasing the GC content of mRNA can also increase mRNA stability. In summary, 5'-UTR, 3'-UTR, 5'Cap, polyA tail, codon optimization and GC content are all modifiable sites that enhance mRNA stability.
mRNA递送mRNA delivery
mRNA目前的递送方法有很多,科学家们建立了脂质体运输、聚合物运输、肽链运输、病毒样复制子颗粒运输和阳离子纳米乳化剂运输等方法,此外裸mRNA也能够被直接注射进细胞。在研的mRNA疫苗最常用的递送方法是脂质纳米颗粒(lipid nanoparticles,LNP)运输。该方法具有毒性小、递送效率高等优势。There are currently many delivery methods for mRNA. Scientists have established methods such as liposome transport, polymer transport, peptide chain transport, virus-like replicon particle transport, and cationic nanoemulsifier transport. In addition, naked mRNA can also be injected directly into cells. . The most commonly used delivery method for mRNA vaccines under development is lipid nanoparticles (LNP) delivery. This method has the advantages of low toxicity and high delivery efficiency.
mRNA疫苗激活固有免疫和适应性免疫mRNA vaccines activate innate and adaptive immunity
mRNA疫苗通过在体细胞内表达抗原,并通过抗原呈递系统呈递,能够激活固有免疫系统和适应性免疫系统。mRNA直接被体细胞内的模式识别受体如TLR识别会导致mRNA的降解,同时能够增强IFN通路。By expressing antigens in somatic cells and presenting them through the antigen presentation system, mRNA vaccines can activate the innate and adaptive immune systems. Direct recognition of mRNA by pattern recognition receptors such as TLRs in somatic cells will lead to the degradation of mRNA and at the same time enhance the IFN pathway.
mRNA疫苗通过激发适应性免疫反应发挥主要作用。mRNA翻译出的抗原,通过MHC-I直接呈递激活CD4+T细胞(Helper T细胞)或抗原分泌后,其他细胞吞噬后以MHC-II通路呈递抗原激活CD8+T细胞(细胞毒性T细胞)。抗原表达于细胞表面后也能够被B细胞受体识别激活B细胞的抗体表达和记忆B细胞形成,进而引起感染细胞凋亡和病原体的中和。mRNA vaccines play a major role by stimulating adaptive immune responses. The antigen translated from the mRNA is directly presented through MHC-I to activate CD4+ T cells (Helper T cells) or after the antigen is secreted, other cells phagocytose it and present the antigen through the MHC-II pathway to activate CD8+ T cells (cytotoxic T cells). After the antigen is expressed on the cell surface, it can also be recognized by B cell receptors and activate the antibody expression of B cells and the formation of memory B cells, thereby causing the apoptosis of infected cells and the neutralization of pathogens.
mRNA接种后产生适应性免疫反应的事件Events that generate adaptive immune responses after mRNA vaccination
a)疫苗的含mRNA颗粒被注射部位局部细胞吸收后,mRNA被模式识别受体识别,同时也开始翻译抗原,导致注射部位的局部炎症,促进免疫细胞的浸润,包括中性粒细胞、单核细胞、髓系树突状细胞(myeloid dendritic cells,MDCs)和浆细胞样树突状细胞(plasmacytoid dendritic cells,PDCs)。中性粒细胞可以有效地吸收LNPs,但单核细胞和MDCs更有效地翻译mRNA。I型干扰素(interferon,IFN)的分泌受到刺激。a) After the vaccine’s mRNA-containing particles are absorbed by local cells at the injection site, the mRNA is recognized by pattern recognition receptors and also begins to translate the antigen, causing local inflammation at the injection site and promoting the infiltration of immune cells, including neutrophils and monocytes. cells, myeloid dendritic cells (MDCs) and plasmacytoid dendritic cells (PDCs). Neutrophils can efficiently take up LNPs, but monocytes and MDCs translate mRNA more efficiently. The secretion of type I interferon (IFN) is stimulated.
b)mRNA/LNP和蛋白质抗原将传播,细胞将迁移到接种疫苗的淋巴结。b) The mRNA/LNP and protein antigens will spread and the cells will migrate to the vaccinated lymph nodes.
c)抗原呈递到T细胞和抗原与B细胞的相互作用发生的导致生发中心的形成,记忆B细胞和生产抗体的浆细胞在此产生,这些细胞驻留在骨髓中。 c) Antigen presentation to T cells and interaction of antigen with B cells occurs leading to the formation of germinal centers where memory B cells and antibody-producing plasma cells are produced. These cells reside in the bone marrow.
mRNA疫苗面对传染性疾病的优势Advantages of mRNA vaccines against infectious diseases
由于mRNA疫苗的机制是通过将编码抗原蛋白的mRNA接种到宿主,然后在体内细胞中利用宿主的遗传物质进行表达合成抗原蛋白,通过抗原蛋白诱导和激活机体的免疫系统产生免疫反应,从而达到预防和治疗疾病的目的。其独特的优势为:1)所有生产过程的监测和质控可以轻松实现;2)mRNA的研发和生产周期短,容易实现量产,疫苗的产能较高,这对快速应对全球范围的新发传染病至关重要;3)mRNA由于自身的特性,免疫后能很快降解,安全风险较低;4)免疫效力好,能够同时诱导产生体液免疫和细胞免疫,对目前没有较好疫苗的传染性疾病而言可能存在研发出更有效疫苗的潜力。The mechanism of the mRNA vaccine is to inoculate the mRNA encoding the antigenic protein into the host, and then use the host's genetic material to express and synthesize the antigenic protein in cells in the body. The antigenic protein induces and activates the body's immune system to produce an immune response, thereby achieving prevention. and the purpose of treating disease. Its unique advantages are: 1) Monitoring and quality control of all production processes can be easily realized; 2) The research and development and production cycle of mRNA is short, it is easy to achieve mass production, and the vaccine production capacity is high, which is very important for quickly responding to new emerging diseases worldwide. Infectious diseases are crucial; 3) Due to its own characteristics, mRNA can be degraded quickly after immunization, and the safety risk is low; 4) The immune effect is good, and it can induce both humoral immunity and cellular immunity at the same time, and is effective against infections for which there is currently no better vaccine. There may be potential for more effective vaccines against diseases.
mRNA疫苗优化策略Optimization strategy for mRNA vaccines
mRNA疫苗发挥作用需要面对的挑战包括:1)延长自身半衰期,增强稳定性;2)增强翻译活性;3)减少mRNA的免疫原性,避免被快速清除。The challenges that mRNA vaccines need to face to work include: 1) extending its half-life and enhancing stability; 2) enhancing translation activity; 3) reducing the immunogenicity of mRNA to avoid rapid clearance.
达到这些作用的方法就是设计特殊的5'-UTR、3'-UTR、终止密码子和polyA数量等。对于5'-UTR和3'-UTR的设计,有三种有效的手段:1)采用高表达的人类基因的UTR;2)使用抗原蛋白自身的UTR;3)指数富集配体系统进化技术(systematic evolution of ligands by exponential enrichment,SELEX)。前两种方法均较为简单,第三种方法相对复杂,要求通过体外实验不断尝试并优化序列,因此需要很长时间,但第三种方法仍是时间充裕时的最佳选择。The way to achieve these effects is to design special 5'-UTR, 3'-UTR, stop codon, polyA number, etc. For the design of 5'-UTR and 3'-UTR, there are three effective methods: 1) using the UTR of highly expressed human genes; 2) using the UTR of the antigen protein itself; 3) exponential enrichment ligand system evolution technology ( systematic evolution of ligands by exponential enrichment, SELEX). The first two methods are relatively simple, while the third method is relatively complex and requires continuous trying and optimizing the sequence through in vitro experiments, so it takes a long time. However, the third method is still the best choice when time is sufficient.
目前已经FDA批准上市的辉瑞(Pfizer/BioNTech)的BNT162b2疫苗的5'-UTR采用了人α珠蛋白的5'-UTR并优化了Kozak序列和5'端序列,调整了5'-UTR的二级结构,而莫得那(Moderna)的mRNA-1273疫苗的5'-UTR则采用了其通过计算机设计并优化的序列。对于3'-UTR,莫得那的mRNA-1273疫苗采用了人α珠蛋白(HBA1)的3'-UTR中的110nt碱基,而辉瑞的BNT162b2疫苗采用了基于自然基因进行SELEX的方法,挑选出了人12S rRNA(mtRNR1)和AES/TLE5基因的3'-UTR。辉瑞疫苗在此基础上选用了AES 3'-UTR的136nt序列并进行了两个C→Ψ的改变,随后接续139nt的mtRNR1序列。目前真正有效的UTR设计方法还是借鉴自然基因并基于经验进行优化(PMID:34358150)。The 5'-UTR of Pfizer/BioNTech's BNT162b2 vaccine, which is currently approved by the FDA, uses the 5'-UTR of human alpha globin and optimizes the Kozak sequence and 5' end sequence, adjusting the second half of the 5'-UTR. hierarchical structure, while the 5'-UTR of Moderna's mRNA-1273 vaccine uses a sequence designed and optimized by its computer. For the 3'-UTR, Moderna's mRNA-1273 vaccine uses the 110nt base in the 3'-UTR of human alpha globin (HBA1), while Pfizer's BNT162b2 vaccine uses a SELEX method based on natural genes to select The 3'-UTR of human 12S rRNA (mtRNR1) and AES/TLE5 genes were identified. On this basis, the Pfizer vaccine selected the 136nt sequence of AES 3'-UTR and made two C→Ψ changes, followed by the 139nt mtRNR1 sequence. At present, the truly effective UTR design method is to learn from natural genes and optimize based on experience (PMID: 34358150).
此外,在抗原蛋白的终止密码子后加入1-2个终止密码子的方法也被采用,以更有效的翻译延伸复合物解聚,利于mRNA的翻译和稳定。辉瑞的BNT162b2疫苗采用了终止信号UGAUGA,而莫得那的mRNA-1273疫苗采用了UGAUAAUAG。In addition, the method of adding 1-2 stop codons after the stop codon of the antigen protein has also been adopted to more effectively depolymerize the translation elongation complex and facilitate the translation and stability of mRNA. Pfizer's BNT162b2 vaccine uses the termination signal UGAUGA, while Moderna's mRNA-1273 vaccine uses UGAUAAUAG.
polyA的数量是影响mRNA稳定性的重要因素,80nt-150n或100nt-150nt为宜,更佳地120nt。适宜数量的polyA数量能够比不适宜数量的mRNA具有更高的蛋 白表达能力和mRNA稳定性。The number of polyA is an important factor affecting the stability of mRNA. 80nt-150n or 100nt-150nt is suitable, and 120nt is better. An appropriate amount of polyA can have higher protein production than an inappropriate amount of mRNA. expression ability and mRNA stability.
感染性疾病mRNA疫苗Infectious disease mRNA vaccines
本发明的构建mRNA转录本的骨架可用于构建针对感染性疾病的mRNA疫苗。The framework for constructing mRNA transcripts of the present invention can be used to construct mRNA vaccines against infectious diseases.
采用本发明mRNA转录本的骨架的疫苗可适用于各种不同的病原体,代表性的病原体包括(但并不限于):冠状病毒(如新冠病毒)、巨细胞病毒(CMV)、寨卡病毒(Zika)、流感病毒(Influenza)、呼吸道合胞病毒(RSV)、基孔肯雅病(Chikungunya)、狂犬病(Rabies)、艾滋病毒(HIV)、埃博拉病毒(Ebola virus)、链球菌(streptococci)、疟疾(malaria)、跳跃病病毒(Louping ill virus)、岗地弓形虫(Toxoplasma gondii)等等。The vaccine using the skeleton of the mRNA transcript of the present invention can be applied to a variety of different pathogens. Representative pathogens include (but are not limited to): coronavirus (such as new coronavirus), cytomegalovirus (CMV), Zika virus ( Zika), Influenza, Respiratory Syncytial Virus (RSV), Chikungunya, Rabies, HIV, Ebola virus, Streptococci ), malaria, jumping disease virus, Toxoplasma gondii, etc.
此外,很多感染性疾病缺乏有效的灭活疫苗和重组蛋白疫苗,因而可能寄希望于mRNA疫苗能否激发人体对这些病原体的预防性免疫,进而研发出有效的疫苗。这些疾病包括登革热(现有疫苗只能保护一种血清型)、鼠疫、黄热病、结核疫苗、单纯疱疹病毒、带状病毒、支原体、衣原体、口蹄疫病毒等,还可以用于一些抗肿瘤药物的治疗中。In addition, many infectious diseases lack effective inactivated vaccines and recombinant protein vaccines, so it may be hoped that mRNA vaccines can stimulate the body's preventive immunity against these pathogens and then develop effective vaccines. These diseases include dengue fever (existing vaccines can only protect one serotype), plague, yellow fever, tuberculosis vaccine, herpes simplex virus, band virus, mycoplasma, chlamydia, foot and mouth disease virus, etc., and can also be used in some anti-tumor drugs of treatment.
抗体基因UTRAntibody gene UTR
为提高UTR的泛用性并减少免疫原性,发明人采用了抗体基因的UTR作为原始UTR。抗体(antibody)分为5类(class),即IgM、IgD、IgG、IgA和IgE,其相应的重链分别为μ链、δ链、γ链、α链和ε链,与这些重链相配合的轻链有两种,分别为kappa(κ)链和lambda(λ)链,根据λ链恒定区个别氨基酸的差异,又可将λ链分为λl、λ2、λ3和λ4四个亚型。上述这些肽链的基因的重组率均很高,提示它们的5'-UTR和3'-UTR序列可能对变化的ORF区及变化的肽链表达具有很强的兼容性。因此,抗体基因的5'-UTR和3'-UTR可能具有支持不同ORF高效翻译的泛用性。In order to improve the versatility of UTR and reduce immunogenicity, the inventor used the UTR of the antibody gene as the original UTR. Antibodies are divided into 5 categories (classes), namely IgM, IgD, IgG, IgA and IgE. Their corresponding heavy chains are μ chain, δ chain, γ chain, α chain and ε chain respectively. Compared with these heavy chains There are two types of coordinated light chains, namely kappa (κ) chain and lambda (λ) chain. According to the differences in individual amino acids in the constant region of the λ chain, the λ chain can be divided into four subtypes: λl, λ2, λ3 and λ4. . The recombination rates of the above-mentioned peptide chain genes are very high, suggesting that their 5'-UTR and 3'-UTR sequences may have strong compatibility with changed ORF regions and changed peptide chain expression. Therefore, the 5'-UTR and 3'-UTR of antibody genes may have versatility to support efficient translation of different ORFs.
在本发明中,优选的5'-UTR和3'-UTR是经序列优化的UTR。图2中显示了5'-UTR优化前的序列IGL-5-O和优化后的序列IGL-5'UTR-F,其GC含量从54%提升至优化后的64%。图4中显示了3'-UTR优化前的序列IGL-3-O和优化后的序列IGL-3'UTR-F,其GC含量从54%提升至优化后的56%。In the present invention, preferred 5'-UTR and 3'-UTR are sequence-optimized UTRs. Figure 2 shows the sequence IGL-5-O before 5'-UTR optimization and the sequence IGL-5'UTR-F after optimization. The GC content increased from 54% to 64% after optimization. Figure 4 shows the sequence IGL-3-O before 3'-UTR optimization and the sequence IGL-3'UTR-F after optimization. The GC content increased from 54% to 56% after optimization.
在本发明中,基于抗体基因UTR经序列优化的优选5'-UTR,其核苷酸序列如SEQ ID NO:2所示;基于抗体基因UTR经序列优化的优选3'-UTR,其核苷酸序列如SEQ ID NO:4、5或6所示。In the present invention, the preferred 5'-UTR based on the sequence optimization of the antibody gene UTR, its nucleotide sequence is shown in SEQ ID NO: 2; the preferred 3'-UTR based on the sequence optimization of the antibody gene UTR, its nucleoside The acid sequence is shown in SEQ ID NO: 4, 5 or 6.
干扰素基因UTRinterferon gene UTR
为提高UTR的泛用性并减少免疫原性,发明人还采用了人体广泛表达的免疫相关蛋白干扰素的基因的UTR作为原始UTR。 In order to improve the versatility of UTR and reduce immunogenicity, the inventor also used the UTR of the gene of immune-related protein interferon, which is widely expressed in the human body, as the original UTR.
干扰素是病毒或其它干扰素诱生剂,刺激网内细胞、巨唾细胞、淋巴细胞等多种细胞所产生的一种可游离到细胞外的、具有广谱抗病毒作用的糖蛋白。其能在多种细胞内稳定表达,使用其5'-UTR和3'-UTR进行优化,可能减少其免疫原性,并进而减少mRNA疫苗的副作用。Interferon is a glycoprotein produced by viruses or other interferon-inducing agents that can be released outside the cells and has broad-spectrum antiviral effects by stimulating intraretinal cells, macrosialocytes, lymphocytes and other cells. It can be stably expressed in a variety of cells, and optimization using its 5'-UTR and 3'-UTR may reduce its immunogenicity and thereby reduce the side effects of mRNA vaccines.
在本发明中,优选的5'-UTR和3'-UTR是经序列优化的UTR。图3中显示了5'-UTR优化前的序列IFN-5-O和优化后的序列IFN-5'UTR-F,其GC含量从49%提升至优化后的52%。图5中显示了3'-UTR优化前的序列INF-3-O和优化后的序列IFN-3'UTR-F,其GC含量从31%提升至优化后的44%。In the present invention, preferred 5'-UTR and 3'-UTR are sequence-optimized UTRs. Figure 3 shows the sequence IFN-5-O before 5'-UTR optimization and the sequence IFN-5'UTR-F after optimization. Its GC content increased from 49% to 52% after optimization. Figure 5 shows the sequence INF-3-O before 3'-UTR optimization and the sequence IFN-3'UTR-F after optimization. The GC content increased from 31% to 44% after optimization.
在本发明中,基于干扰素基因UTR经序列优化的优选5'-UTR,其核苷酸序列如SEQ ID NO:8所示;基于干扰素基因UTR经序列优化的优选3'-UTR,其核苷酸序列如SEQ ID NO:10、11或12所示。In the present invention, the preferred 5'-UTR based on the sequence optimization of the interferon gene UTR, its nucleotide sequence is shown in SEQ ID NO: 8; the preferred 3'-UTR based on the sequence optimization of the interferon gene UTR, whose The nucleotide sequence is shown in SEQ ID NO: 10, 11 or 12.
本发明的主要优点包括:The main advantages of the present invention include:
(1)通过构建带有可替换ORF模板骨架的质粒,能够加快mRNA疫苗的研发速度。(1) By constructing a plasmid with a replaceable ORF template backbone, the development of mRNA vaccines can be accelerated.
(2)通过选择抗体和干扰素基因的UTR作为泛用质粒骨架的UTR部分,即后续产生mRNA的UTR部分,增强了mRNA的稳定性和翻译活性,主要是保证了该质粒骨架的泛用性和mRNA疫苗的稳定性。(2) By selecting the UTR of the antibody and interferon genes as the UTR part of the universal plasmid skeleton, that is, the UTR part of the subsequent generated mRNA, the stability and translation activity of the mRNA are enhanced, mainly to ensure the universality of the plasmid skeleton. and the stability of mRNA vaccines.
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,例如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数是重量百分比和重量份数。The present invention will be further described below in conjunction with specific embodiments. It should be understood that these examples are only used to illustrate the invention and are not intended to limit the scope of the invention. Experimental methods without specifying specific conditions in the following examples usually follow conventional conditions, such as the conditions described in Sambrook et al., Molecular Cloning: Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to manufacturing Conditions recommended by the manufacturer. Unless otherwise stated, percentages and parts are by weight.
实施例1.UTR序列的筛选与优化方法Example 1. Screening and optimization method of UTR sequences
本发明人设计了一种可替换ORF的质粒,并且使用平末端或粘性末端酶切位点使其可以方便后续体外转录的线性化片段试验。The inventors designed a plasmid that can replace the ORF, and uses blunt-end or sticky-end restriction sites to facilitate subsequent linearized fragment testing of in vitro transcription.
典型地,本载体包括载体骨架、AleI酶切位点或BspQI酶切位点、5'-UTR、ORF、3'-UTR、polyA。此外选用了特殊感受态,并进行了ORF密码子优化。Typically, this vector includes a vector backbone, AleI restriction site or BspQI restriction site, 5'-UTR, ORF, 3'-UTR, and polyA. In addition, a special competent state was selected and ORF codon optimization was performed.
本发明人使用了高拷贝质粒载体pUC57-Amp或pUC-Kana进行扩增片段。利用高效平末端限制性内切酶AleI或粘性末端限制性内切酶BspQI作为从质粒上剪切下插入片段的酶,其能够产生的平末端或粘性末端可方便以后实验。选用的启动子为T7启动子。 The inventors used high-copy plasmid vector pUC57-Amp or pUC-Kana to amplify the fragments. The high-efficiency blunt-end restriction enzyme AleI or the sticky-end restriction enzyme BspQI is used as an enzyme to cut the inserted fragment from the plasmid. The blunt-end or sticky end it can produce can facilitate subsequent experiments. The promoter selected was T7 promoter.
基于抗体和干扰素表达的特点,本发明人从NCBI数据库中下载了10条不同抗体肽链、不同型干扰素的序列(如表1所示),经过比对后,找出了相对保守,且长度合适的UTR序列。并且基于自然UTR序列,通过优化Kozak序列,减少AT-rich序列等步骤,得到了优化后的UTR序列。Based on the expression characteristics of antibodies and interferons, the inventor downloaded the sequences of 10 different antibody peptide chains and different types of interferons from the NCBI database (as shown in Table 1). After comparison, they found out that they are relatively conserved. and a UTR sequence of appropriate length. And based on the natural UTR sequence, the optimized UTR sequence was obtained by optimizing the Kozak sequence and reducing the AT-rich sequence.
表1参与对比序列及其编号
Table 1 Participating comparison sequences and their numbers
实施例2. 5'-UTR及3'-UTR的选择与优化Example 2. Selection and optimization of 5'-UTR and 3'-UTR
5'-UTR分别选择了抗体保守序列和干扰素保守序列两种,并进行了Kozak序列优化。并且在载体构建之前分别将抗体和干扰素的5'-UTR进行了GC含量的优化。Two types of 5'-UTR were selected: antibody conserved sequences and interferon conserved sequences, and Kozak sequence optimization was performed. And the GC content of the 5'-UTR of the antibody and interferon was optimized before vector construction.
3'-UTR分别选择了抗体保守序列和干扰素保守序列两种,并进行了AT-rich序列的剔除。并且在载体构建之前分别将抗体和干扰素的3'-UTR进行了GC含量的优化。Two types of 3'-UTR were selected: antibody conserved sequences and interferon conserved sequences, and AT-rich sequences were eliminated. And the GC content of the 3'-UTR of the antibody and interferon was optimized before vector construction.
上述序列信息如表2所示:The above sequence information is shown in Table 2:
表2野生型序列与优化序列

Table 2 Wild-type sequence and optimized sequence

由于翻译延伸复合物在终止密码子处识别多个终止密码子有利于复合物的解聚,进而增强mRNA的翻译活性,因此本发明人增加了2个终止密码子。此外,选用的polyA长度为120±10nt。Since the translation elongation complex recognizes multiple stop codons at the stop codon, which is beneficial to the depolymerization of the complex and thereby enhances the translation activity of the mRNA, the inventors added two stop codons. In addition, the selected polyA length is 120±10nt.
通过同源重组或酶切的方法可以方便的替换质粒骨架中的ORF序列,进而能够用于不同抗原的mRNA疫苗的构建。The ORF sequence in the plasmid backbone can be easily replaced through homologous recombination or enzyme digestion, which can then be used to construct mRNA vaccines for different antigens.
实施例3.水蛭素mRNA疫苗工程菌株构建Example 3. Construction of hirudin mRNA vaccine engineering strain
(1)使用带有水蛭素基因的质粒为模板,从其上PCR扩增出水蛭素基因(Hirudin)片段,如图7所示,通过引物引入同源臂序列、T7启动子、AleI酶切位点、GFP和6×His标签。(1) Use the plasmid containing the hirudin gene as a template, and PCR amplify the hirudin gene (Hirudin) fragment from it, as shown in Figure 7, introduce the homology arm sequence, T7 promoter, and AleI digestion through primers locus, GFP and 6×His tags.
质粒骨架片段以pUC57-Amp载体为模板,如图8所示,通过PCR扩增出带有同 源臂的载体骨架的片段。The plasmid backbone fragment was amplified by PCR using the pUC57-Amp vector as a template, as shown in Figure 8. A fragment of the vector backbone of the source arm.
如图9所示,基因合成片段与PCR产物分别在琼脂糖凝胶中进行电泳鉴定。As shown in Figure 9, gene synthesis fragments and PCR products were electrophoresed in agarose gels for identification.
(2)通过胶回收获得(1)中得到的两个片段,进而采用同源重组的方法连接环化并转入stable3大肠杆菌感受态细胞中构建工程菌,挑取单克隆培养成菌液,进行sanger测序鉴定工程菌。(2) Obtain the two fragments obtained in (1) through gel recovery, then use homologous recombination to connect, circularize and transfer them into stable3 Escherichia coli competent cells to construct engineering bacteria, select single clones and culture them into bacterial liquid. Sanger sequencing was performed to identify the engineering bacteria.
如图10所示,构建成功的工程菌提取的质粒进行琼脂糖凝胶电泳。As shown in Figure 10, the plasmid extracted from the successfully constructed engineering bacteria was subjected to agarose gel electrophoresis.
如图11所示,构建成功的工程菌提取的质粒进行AleI酶单酶切。As shown in Figure 11, the plasmid extracted from the successfully constructed engineering bacteria was digested with AleI enzyme.
实施例4.水蛭素质粒工程菌鉴定Example 4. Identification of leech plasmid engineering bacteria
将水蛭素质粒工程菌进行长期传代,并对其中部分代数保存菌液,进行菌株鉴定和细菌质粒拷贝数、细菌质粒丢失率检测。检测结果如表1-2所示。The leech plasmid engineered bacteria were passaged for a long time, and part of the bacterial liquid was preserved for several generations to conduct strain identification and detection of bacterial plasmid copy number and bacterial plasmid loss rate. The test results are shown in Table 1-2.
3、5、10、15、20代菌液的IMVC生化实验结果显示为典型大肠埃希氏菌;革兰氏染色结果显示为短棒状革兰氏阴性菌,无杂菌污染。The IMVC biochemical test results of the 3rd, 5th, 10th, 15th and 20th generations of bacterial fluids showed that they were typical Escherichia coli; the Gram staining results showed that they were short rod-shaped Gram-negative bacteria without miscellaneous bacterial contamination.
表3水蛭素工程菌细菌质粒拷贝数
Table 3 Bacterial plasmid copy number of hirudin engineering bacteria
如表3所示,细菌质粒拷贝数检测显示工程菌的质粒拷贝数约为53.17-240.28copies/cell。As shown in Table 3, bacterial plasmid copy number detection shows that the plasmid copy number of the engineering bacteria is approximately 53.17-240.28 copies/cell.
表4水蛭素质粒工程菌细菌质粒丢失率检定结果

Table 4 Test results of bacterial plasmid loss rate of leech plasmid engineered bacteria

如表4所示,细菌质粒丢失率检测显示工程菌在传代过程中未出现质粒丢失。As shown in Table 4, the bacterial plasmid loss rate test showed that no plasmid loss occurred in the engineering bacteria during the passage process.
实施例5.水蛭素工程菌发酵质粒得率Example 5. Fermentation plasmid yield of hirudin engineering bacteria
在5L发酵罐中,加入3L LB培养基,在罐压0.05±0.02MPa、通风2-10L/min、搅拌速度100-250rpm、温度37±0.5℃、pH 7.0±0.1条件下进行菌液发酵。发酵10小时过程中每小时取2支5mL菌液,用于提取质粒。质粒浓度结果如表5、图12所示。In the 5L fermentation tank, add 3L LB culture medium, and perform bacterial liquid fermentation under the conditions of tank pressure 0.05±0.02MPa, ventilation 2-10L/min, stirring speed 100-250rpm, temperature 37±0.5℃, and pH 7.0±0.1. During the 10-hour fermentation process, take 2 tubes of 5mL bacterial liquid every hour for plasmid extraction. The plasmid concentration results are shown in Table 5 and Figure 12.
表5水蛭素工程菌发酵过程质粒得率
Table 5 Plasmid yield during fermentation process of hirudin engineering bacteria
如图12的实验结果显示种子菌发酵4h,可得到最高的质粒得率。The experimental results shown in Figure 12 show that the highest plasmid yield can be obtained by fermenting the seed bacteria for 4 hours.
实施例6.体外转录效率的验证Example 6. Verification of in vitro transcription efficiency
利用本发明的泛用性UTR元件构建获得的转录模板质粒具有高水平体外转录效果。参照实施例3的构建方法,使用狂犬病毒G蛋白的编码基因(GeneBank:GQ918139.1)作为ORF插入到可替换ORF质粒上构建重组工程菌和质粒。基于该质粒进行体外转录(IVT)实验。The transcription template plasmid constructed using the universal UTR element of the present invention has a high-level in vitro transcription effect. Referring to the construction method of Example 3, the gene encoding the rabies virus G protein (GeneBank: GQ918139.1) was inserted into the replaceable ORF plasmid as an ORF to construct recombinant engineering bacteria and plasmids. In vitro transcription (IVT) experiments were performed based on this plasmid.
使用BspQI对质粒进行酶切线性化后,利用线性化DNA片段作为IVT的模板。体外转录体系中加入T7Polymerase、NTPs、RNase inhibitor、无机焦磷酸酶PPasse及 相应的缓冲体系。37℃恒温反应3小时后,采用LiCl沉淀方法纯化后,检测合成的RNA的量,以RNA的量相对加入DNA模板的量的倍数计算IVT的产率。After linearizing the plasmid using BspQI, the linearized DNA fragment was used as the template for IVT. T7 Polymerase, NTPs, RNase inhibitor, inorganic pyrophosphatase PPasse and Corresponding buffer system. After reacting at a constant temperature of 37°C for 3 hours, and purifying using the LiCl precipitation method, the amount of synthesized RNA was detected, and the IVT yield was calculated as the multiple of the amount of RNA relative to the amount of DNA template added.
共进行8批次IVT反应及纯化,反应中模板DNA投入量均为2.20μg,RNA产量均在0.27-0.40mg,RNA产量/DNA模板量之比(转录效率)在约122-182倍。本发明中的骨架构建的转录模板质粒,在体外转录中具有较高的体外转录活性。A total of 8 batches of IVT reactions and purification were carried out. The input amount of template DNA in the reaction was 2.20 μg, the RNA yield was 0.27-0.40 mg, and the ratio of RNA yield/DNA template amount (transcription efficiency) was about 122-182 times. The transcription template plasmid constructed by the skeleton in the present invention has high in vitro transcription activity in in vitro transcription.
表6泛用骨架构建转录模板进行IVT mRNA产率(DNA投入量为2.20μg)
Table 6 IVT mRNA yield using universal scaffolds to construct transcription templates (DNA input amount is 2.20 μg)
实施例7.构建获得的mRNA表达能力的验证Example 7. Verification of expression ability of mRNA obtained by construction
利用本发明的泛用性UTR元件构建获得的mRNA具有表达相应蛋白的能力。使用实施例6中制备的mRNA进行细胞转染实验,其中在狂犬病毒G蛋白的C端带有6×His标签。同时采用两种转染试剂,转染试剂A(LipofectamineTM MessengerMAXTM)和转染试剂B(YEASEN-Hieff Trans mRNA),进行mRNA转染293T细胞实验。mRNA用量为2μg/孔,6孔板细胞数为106细胞/孔。The mRNA constructed using the universal UTR element of the present invention has the ability to express the corresponding protein. Cell transfection experiments were performed using the mRNA prepared in Example 6, which contained a 6×His tag at the C terminus of the rabies virus G protein. Two transfection reagents, transfection reagent A (Lipofectamine TM MessengerMAX TM ) and transfection reagent B (YEASEN-Hieff Trans mRNA), were used at the same time to conduct the mRNA transfection experiment on 293T cells. The amount of mRNA used was 2 μg/well, and the number of cells in the 6-well plate was 10 6 cells/well.
转染2天后,收集细胞沉淀于1.5mL离心管中,加入裂解液(RIPA:PMSF=99:1),冰上裂解5min,收集蛋白裂解液。使用BCA法检测蛋白浓度后,加入6×Protein Loading buffer(全式金,DL101-02),沸水中加热变性10min。Two days after transfection, collect the cell pellet in a 1.5 mL centrifuge tube, add lysis buffer (RIPA:PMSF=99:1), lyse on ice for 5 minutes, and collect the protein lysate. After using the BCA method to detect the protein concentration, add 6×Protein Loading buffer (full gold, DL101-02), and heat and denature in boiling water for 10 minutes.
已变性蛋白样品进行WB实验,7.5%SDS-PAGE凝胶进行电泳,每孔加入蛋白总量约20μg。使用Anti-His小鼠单克隆抗体(北京全式金,HL102-01)和HRP标记的Goat Anti-Mouse IgG(H+L)为二抗(1:5000,PBS稀释)标记狂犬病毒G蛋白。孵育完成后使用超敏ECL发光液(雅酶,SQ201)进行显影。Denatured protein samples were subjected to WB experiments, and 7.5% SDS-PAGE gel was electrophoresed. A total of approximately 20 μg of protein was added to each well. Anti-His mouse monoclonal antibody (Beijing Quanshijin, HL102-01) and HRP-labeled Goat Anti-Mouse IgG (H+L) were used as secondary antibodies (1:5000, diluted in PBS) to label rabies virus G protein. After the incubation is completed, use ultra-sensitive ECL luminescent solution (Yase, SQ201) for development.
如图13所示,结果显示,在经转染的细胞中存在狂犬病毒G蛋白表达,表明本发明的泛用性UTR元件及骨架生产出的mRNA具有在细胞中表达相应蛋白的能力。As shown in Figure 13, the results show that rabies virus G protein expression exists in the transfected cells, indicating that the mRNA produced by the universal UTR element and skeleton of the present invention has the ability to express the corresponding protein in cells.
实施例8.利用本发明的泛用性UTR元件构建获得的狂犬病毒mRNA疫苗可诱导小鼠产生极高水平抗体Example 8. The rabies virus mRNA vaccine constructed using the universal UTR element of the present invention can induce mice to produce extremely high levels of antibodies.
参照实施例3的构建方法,使用狂犬病毒G蛋白的编码基因作为ORF插入到可替换ORF质粒(包括本发明的泛用性UTR元件及BspQI酶切位点,并以高拷贝质粒载体pUC-Kana为载体骨架)上构建重组工程菌,并基于此重组工程菌生产出狂犬病毒mRNA实验用疫苗。Referring to the construction method of Example 3, the rabies virus G protein encoding gene is used as an ORF to be inserted into the replaceable ORF plasmid (including the universal UTR element of the present invention and the BspQI restriction site), and the high-copy plasmid vector pUC-Kana A recombinant engineered bacterium was constructed on the vector skeleton), and a rabies virus mRNA experimental vaccine was produced based on this recombinant engineered bacterium.
如图14所示,显示了构建成功的工程菌提取的质粒进行琼脂糖凝胶电泳和酶切图谱检定结果。其中,泳道P1为质粒条带,泳道1为ApaLI和PvuII双酶切结果。 检定结果表明符合该质粒设计的预期结果。As shown in Figure 14, the results of agarose gel electrophoresis and enzyme digestion map verification of the plasmid extracted from the successfully constructed engineering bacteria are shown. Among them, lane P1 is the plasmid band, and lane 1 is the result of ApaLI and PvuII double enzyme digestion. The assay results showed that they were consistent with the expected results of the plasmid design.
如图15所示,显示了构建成功的工程菌提取的质粒序列与设计序列比对结果。比对结果表明测序结果与设计序列完全一致。As shown in Figure 15, the comparison results between the plasmid sequence extracted from the successfully constructed engineering bacteria and the designed sequence are shown. The comparison results showed that the sequencing results were completely consistent with the designed sequence.
以上结果表明,已成功构建狂犬病毒mRNA疫苗工程菌。The above results show that the rabies virus mRNA vaccine engineering strain has been successfully constructed.
接下来,使用6-8周龄BALB/C小鼠进行实验,实验分为4组:阳性对照组,16只小鼠(8雄8雌),肌肉注射鸡胚灭活疫苗(0.6IU/只);低剂量组,16只小鼠(8雄8雌),肌肉注射较低剂量mRNA疫苗(5μg/只);高剂量组,16只小鼠(8雄8雌),肌肉注射较高剂量mRNA疫苗(13μg/只);阴性对照组,不进行肌肉注射。除阴性对照组外,均在第0天和第14天,进行两次注射达到加强免疫效果,在第14天和第28天分别对半数小鼠取血。血清样本依照《中国药典》方法进行荧光灶抑制法检测狂犬病毒G蛋白抗体水平。Next, 6-8 weeks old BALB/C mice were used for experiments. The experiment was divided into 4 groups: positive control group, 16 mice (8 males and 8 females), and intramuscular injection of chicken embryo inactivated vaccine (0.6IU/mouse). ); the low-dose group, 16 mice (8 males and 8 females), received a lower dose of mRNA vaccine (5 μg/mouse) intramuscularly; the high-dose group, 16 mice (8 males and 8 females), received a higher dose intramuscularly. mRNA vaccine (13 μg/animal); negative control group, no intramuscular injection. Except for the negative control group, two injections were performed on days 0 and 14 to enhance the immune effect, and blood was taken from half of the mice on days 14 and 28 respectively. Serum samples were tested for rabies virus G protein antibody levels by fluorescence focus inhibition method according to the method of "Chinese Pharmacopoeia".
如表7所示,结果显示鸡胚灭活疫苗能够诱导小鼠产生理论上足以抵抗狂犬病毒感染的抗体水平,狂犬病毒mRNA疫苗无论低剂量条件或高剂量条件下均产生了较灭活疫苗更高的抗体水平。该结果表明使用泛用型元件构建并生产的mRNA疫苗能够诱导产生高水平的免疫反应。As shown in Table 7, the results show that the chicken embryo inactivated vaccine can induce mice to produce antibody levels that are theoretically sufficient to resist rabies virus infection. The rabies virus mRNA vaccine produced more antibodies than the inactivated vaccine under both low-dose and high-dose conditions. High antibody levels. The results indicate that an mRNA vaccine constructed and produced using universal elements can induce a high level of immune response.
表7小鼠血清抗狂犬病毒中和抗体水平
Table 7 Mouse serum anti-rabies virus neutralizing antibody levels
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。 All documents mentioned in this application are incorporated by reference in this application to the same extent as if each individual document was individually incorporated by reference. In addition, it should be understood that after reading the above teaching content of the present invention, those skilled in the art can make various changes or modifications to the present invention, and these equivalent forms also fall within the scope defined by the appended claims of this application.

Claims (10)

  1. 一种泛用性骨架的用途,其特征在于,所述泛用性骨架用于构建mRNA转录本,其中,所述转录本包括待表达的ORF以及位于所述ORF两侧的5'-UTR区和3'-UTR区,其中所述5'-UTR区和3'-UTR区中1个或2个为泛用性UTR。The use of a universal framework, characterized in that the universal framework is used to construct mRNA transcripts, wherein the transcripts include the ORF to be expressed and the 5'-UTR regions located on both sides of the ORF and a 3'-UTR region, wherein one or both of the 5'-UTR region and the 3'-UTR region are universal UTRs.
  2. 一种泛用性骨架,其特征在于,所述泛用性骨架具有式I结构:
    Z1-Z2-Z3-Z4-Z5-Z6-Z7   (I)
    A general-purpose skeleton, characterized in that the general-purpose skeleton has the structure of formula I:
    Z1-Z2-Z3-Z4-Z5-Z6-Z7 (I)
    式中,In the formula,
    Z1、Z7为无或酶切位点;Z1 and Z7 are no or enzyme cutting sites;
    Z2为无或启动子元件;Z2 is none or promoter element;
    Z3为5'-UTR元件;Z3 is a 5'-UTR component;
    Z4为可替换的ORF区;Z4 is a replaceable ORF region;
    Z5为3'-UTR元件;Z5 is a 3'-UTR component;
    Z6为polyA尾部元件。Z6 is a polyA tail component.
  3. 一种泛用性UTR元件,其特征在于,所述泛用性UTR元件包括:A universal UTR element, characterized in that the universal UTR element includes:
    (a)泛用性5'-UTR,其中所述泛用性5'-UTR的序列选自如SEQ ID NO:2或8所示的核苷酸序列或其衍生序列;和/或(a) a universal 5'-UTR, wherein the sequence of the universal 5'-UTR is selected from the nucleotide sequence shown in SEQ ID NO: 2 or 8 or a derivative sequence thereof; and/or
    (b)泛用性3'-UTR,其中所述泛用性3'-UTR的序列选自如SEQ ID NO:4、5、6、10、11或12所示的核苷酸序列或其衍生序列。(b) Universal 3'-UTR, wherein the sequence of the universal 3'-UTR is selected from the nucleotide sequence shown in SEQ ID NO: 4, 5, 6, 10, 11 or 12 or derivatives thereof sequence.
  4. 一种载体,其特征在于,所述载体含有如权利要求2所述的泛用性骨架。A carrier, characterized in that the carrier contains the universal skeleton as claimed in claim 2.
  5. 一种宿主细胞,其特征在于,所述宿主细胞含有如权利要求4所述的载体,或其基因组中整合有如权利要求2所述的泛用性骨架。A host cell, characterized in that the host cell contains the vector according to claim 4, or the universal framework according to claim 2 is integrated into its genome.
  6. 一种工程化细胞,其特征在于,所述工程化细胞含有:如权利要求4所述的载体,或其基因组中整合有如权利要求2所述的泛用性骨架,并且含有目的基因片段。An engineered cell, characterized in that the engineered cell contains: the vector according to claim 4, or the universal scaffold according to claim 2 integrated into its genome, and contains a target gene fragment.
  7. 一种产生用于制备疫苗的优化mRNA的方法,包括步骤:A method of producing optimized mRNA for use in preparing a vaccine, comprising the steps of:
    (a)在适合的条件下,培养如权利要求6所述的工程化细胞,从而获得含有转录DNA模板的载体的培养物;(a) Under suitable conditions, culture the engineered cells as claimed in claim 6 to obtain a culture containing a vector that transcribes the DNA template;
    (b)从所述培养物中分离和/或回收(a)中所述载体,并酶切线性化为DNA模板;(b) Isolating and/or recovering the vector described in (a) from the culture, and enzymatically digesting and linearizing it into a DNA template;
    (c)将(b)中所述DNA模板进行转录,从而获得所述优化mRNA;和(c) transcribing the DNA template described in (b) to obtain the optimized mRNA; and
    (d)任选地,对步骤(c)获得的优化mRNA进行纯化和/或修饰。(d) Optionally, purify and/or modify the optimized mRNA obtained in step (c).
  8. 一种mRNA疫苗的制备方法,其特征在于,所述方法包括步骤:A method for preparing an mRNA vaccine, characterized in that the method includes the steps:
    (i)通过如权利要求7所述的方法获得优化mRNA; (i) Obtain optimized mRNA by the method as claimed in claim 7;
    (ii)将(i)中获得的优化mRNA与药学上可接受的载体混合,从而获得所述mRNA疫苗。(ii) Mix the optimized mRNA obtained in (i) with a pharmaceutically acceptable carrier to obtain the mRNA vaccine.
  9. 一种试剂盒,其特征在于,所述试剂盒包括:A test kit, characterized in that the test kit includes:
    (a)第一质粒或其线性化DNA片段,所述第一质粒或其线性化DNA片段含有目的基因;和(a) a first plasmid or a linearized DNA fragment thereof, which contains the gene of interest; and
    (b)第二质粒,所述第二质粒含有如权利要求2所述的泛用性骨架;和(b) a second plasmid containing the universal scaffold of claim 2; and
    (c)说明书,所述说明书描述了使用所述第一质粒或其线性化DNA片段和第二质粒生产可用于制备疫苗的优化mRNA的方法。(c) Instructions describing a method of using said first plasmid or a linearized DNA fragment thereof and a second plasmid to produce optimized mRNA useful in the preparation of a vaccine.
  10. 一种mRNA疫苗组合物,其特征在于,所述疫苗组合物含有:An mRNA vaccine composition, characterized in that the vaccine composition contains:
    (a)用于表达免疫原的mRNA,所述mRNA包含如权利要求2所述的泛用性骨架;和(a) mRNA for expressing an immunogen, said mRNA comprising a universal scaffold as claimed in claim 2; and
    (b)药学上可接受的载体。 (b) Pharmaceutically acceptable carrier.
PCT/CN2023/096643 2022-05-27 2023-05-26 Skeleton for constructing mrna in-vitro transcription template WO2023227124A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210594377.9A CN117165611A (en) 2022-05-27 2022-05-27 Framework for constructing mRNA in-vitro transcription template
CN202210594377.9 2022-05-27

Publications (1)

Publication Number Publication Date
WO2023227124A1 true WO2023227124A1 (en) 2023-11-30

Family

ID=88918516

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/096643 WO2023227124A1 (en) 2022-05-27 2023-05-26 Skeleton for constructing mrna in-vitro transcription template

Country Status (2)

Country Link
CN (1) CN117165611A (en)
WO (1) WO2023227124A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117487809B (en) * 2023-12-29 2024-03-19 艾斯拓康医药科技(北京)有限公司 Optimized 5' -UTR sequence and application thereof
CN117535295A (en) * 2024-01-09 2024-02-09 艾斯拓康医药科技(北京)有限公司 Optimized 3' -UTR sequence and application thereof
CN117568338A (en) * 2024-01-17 2024-02-20 艾斯拓康医药科技(北京)有限公司 Optimized polyA sequence and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5807707A (en) * 1995-02-10 1998-09-15 Mcmaster University High efficiency translation of mRNA molecules
CN111607612A (en) * 2020-05-19 2020-09-01 深圳市新合生物医疗科技有限公司 Plasmid vector for expressing humoral immunity vaccine mRNA and construction method and application thereof
CN111607613A (en) * 2020-05-19 2020-09-01 深圳市新合生物医疗科技有限公司 Plasmid vector for expressing mRNA of cellular immune vaccine and construction method and application thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5807707A (en) * 1995-02-10 1998-09-15 Mcmaster University High efficiency translation of mRNA molecules
CN111607612A (en) * 2020-05-19 2020-09-01 深圳市新合生物医疗科技有限公司 Plasmid vector for expressing humoral immunity vaccine mRNA and construction method and application thereof
CN111607613A (en) * 2020-05-19 2020-09-01 深圳市新合生物医疗科技有限公司 Plasmid vector for expressing mRNA of cellular immune vaccine and construction method and application thereof

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE Nucleotide 27 February 2022 (2022-02-27), ANONYMOUS : "Homo sapiens interferon alpha 1 (IFNA1), mRNA", XP093111804, retrieved from NCBI Database accession no. NM_024013 *
DATABASE Nucleotide 7 November 2006 (2006-11-07), ANONYMOUS : "Homo sapiens immunoglobulin lambda locus, mRNA (cDNA clone MGC:111580 IMAGE:30325463), complete cds", XP093111800, retrieved from NCBI Database accession no. BC093098.1 *
PAN YU-TING, XU QI-LIN, KANG BO: "Reviews Eukaryotic mRNA UTR: Its Structural Characteristics and mRNA Translation ", CHINESE JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY, vol. 32, no. 9, 1 September 2016 (2016-09-01), pages 990 - 993, XP093111817, DOI: 10.13865/j.cnki.cjbmb.2016.09.04 *
ZARGHAMPOOR FARZANEH; AZARPIRA NEGAR; KHATAMI SAEED REZA; BEHZAD-BEHBAHANI ABBAS; FOROUGHMAND ALI MOHAMMAD: "Improved translation efficiency of therapeutic mRNA", GENE, ELSEVIER AMSTERDAM, NL, vol. 707, 1 January 1900 (1900-01-01), NL , pages 231 - 238, XP085700944, ISSN: 0378-1119, DOI: 10.1016/j.gene.2019.05.008 *

Also Published As

Publication number Publication date
CN117165611A (en) 2023-12-05

Similar Documents

Publication Publication Date Title
WO2023227124A1 (en) Skeleton for constructing mrna in-vitro transcription template
AU2017249424B2 (en) Recombinant arterivirus replicon systems and uses thereof
JP2021501572A (en) New artificial nucleic acid molecule
KR20160102024A (en) A method of making adenovirus and corresponding plasmids
WO2023064907A1 (en) Compositions and methods for vaccination against pathogenic coronavirus species and variants
ES2902787T3 (en) DNAi vaccines and procedures for using the same
US9125845B2 (en) DNA vaccines, uses for unprocessed rolling circle amplification product and methods for making the same
JP2024515300A (en) Alphavirus vectors containing universal cloning adaptors
CN115698295A (en) Vaccine reagent and inoculation method
CN116904489B (en) Duck tembusu virus nucleic acid vaccine and application thereof
Liu et al. Development and application of an uncapped mRNA platform
CN117205309B (en) Influenza immunogen composition, preparation method and application thereof
EP4123029A1 (en) In-vitro transcript mrna and pharmaceutical composition comprising same
US20240091343A1 (en) Technology platform of uncapped-linear mrna with unmodified uridine
CN116159133B (en) Pig A-type foot-and-mouth disease mRNA vaccine and preparation method thereof
WO2023208118A1 (en) Nucleic acid construct and use thereof
WO2024051266A1 (en) Mrna for expressing varicella-zoster virus antigen protein and use thereof
WO2024055272A1 (en) Mrna vector system capable of efficiently expressing target gene and construction and use thereof
US20230235337A1 (en) Circular rna platforms, uses thereof, and their manufacturing processes from engineered dna
CN117568337A (en) Self-circularization RNA based on humanized tRNA
CN114107176A (en) CHO cell line for stably expressing African swine fever CD2v protein and construction method and application thereof
WO2023242425A1 (en) Compositions and methods for circular rna affinity purification
CN117257929A (en) Rabies virus mRNA vaccine and application thereof
CN116024237A (en) mRNA vaccine for preventing heat of rift grains and preparation method thereof
CN114317572A (en) CircRNA vaccine for resisting infectious spleen and kidney necrosis virus and construction method and application thereof

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: 23811194

Country of ref document: EP

Kind code of ref document: A1