WO2025016031A1 - Self-amplifying nucleic acid molecule and use thereof - Google Patents
Self-amplifying nucleic acid molecule and use thereof Download PDFInfo
- Publication number
- WO2025016031A1 WO2025016031A1 PCT/CN2024/093076 CN2024093076W WO2025016031A1 WO 2025016031 A1 WO2025016031 A1 WO 2025016031A1 CN 2024093076 W CN2024093076 W CN 2024093076W WO 2025016031 A1 WO2025016031 A1 WO 2025016031A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- sequence
- mrna molecule
- seq
- mrna
- rna
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/10—Processes for the isolation, preparation or purification of DNA or RNA
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/10—Cells modified by introduction of foreign genetic material
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/10—Transferases (2.)
Definitions
- the present invention belongs to the field of biomedicine and relates to an alphavirus RNA replicase coding sequence, an optimized self-amplifying nucleic acid molecule, a pharmaceutical composition and uses.
- the optimized self-amplifying nucleic acid molecule is a self-amplifying nucleic acid molecule comprising the alphavirus RNA replicase coding sequence, a tissue-specific self-amplifying nucleic acid molecule or a self-amplifying RNA nucleic acid molecule with enhanced exogenous gene expression level.
- RNA Self-amplifying RNA
- saRNA Self-amplifying RNA
- the in vitro synthesis of mRNA is mainly obtained by in vitro transcription (IVT) using linearized plasmid DNA or PCR amplification products as templates using RNA polymerase.
- IVT in vitro transcription
- linearized plasmid DNA is usually selected as the transcription template to ensure sequence accuracy. Therefore, the plasmid needs to be linearized using a specific DNA endonuclease (such as BspQI or XbaI).
- a specific DNA endonuclease such as BspQI or XbaI
- one of the optimization directions for self-amplified RNA is to further improve its transcription quality.
- mRNA vaccine is a new vaccine technology, which uses synthetic mRNA molecules to encode target non-endogenous antigenic proteins and stimulates immune responses against targets such as pathogens or tumors by injecting them into the human body.
- mRNA vaccines have been successfully used to prevent the new coronavirus and have broad application prospects in research fields such as infectious disease prevention vaccines, tumor treatment, and protein replacement therapy.
- Common mRNA forms include linear mRNA and circular mRNA, among which linear mRNA is divided into non-replicating type and self-amplifying mRNA with self-replication ability.
- Self-amplifying RNA is regarded as the next generation of mRNA molecules. Due to its long expression cycle and self-adjuvant effect, it has shown its advantages in effectively inducing specific immunity in infectious diseases and tumor vaccines. In addition, self-amplifying RNA also has unique potential in protein replacement therapies that expect a long half-life. In 2023, Japan approved a new crown vaccine based on self-amplifying RNA, further proving the safety of self-amplifying RNA as a vaccine.
- an important advantage of self-amplifying RNA is that the expression level of exogenous genes is high and the duration is long, which can significantly reduce the administration dose and reduce side effects. Therefore, further improving the expression efficiency has always been one of the directions of self-amplifying RNA sequence optimization.
- the interferon-stimulated factors induced by this such as protein kinase R (PKR), 2-5-oligoadenylate synthetase (OAS) and interferon induced proteins with tetratricopeptide repeats (IFIT), will perform host translation inhibition and induce cell apoptosis to limit the replication and expression of self-amplifying RNA. Therefore, reducing the immunogenicity of the self-amplifying RNA itself has a positive effect on improving the expression efficiency of the self-amplifying RNA, and is therefore the main idea for optimizing the expression efficiency of the self-amplifying RNA.
- PLR protein kinase R
- OFAS 2-5-oligoadenylate synthetase
- IFIT interferon induced proteins with tetratricopeptide repeats
- the sequence structure of self-amplifying mRNA is relatively complex compared to mRNA, especially the presence of viral replicase and replication-related elements such as RNA promoters.
- Self-amplifying mRNA uses the RNA replicase from alphavirus contained in it to enable mRNA to replicate itself. Therefore, the injected mRNA can continuously produce the target protein in the body, thereby enhancing the durability and intensity of the vaccine immune response, or enhancing the half-life of protein replacement therapy.
- the current self-amplifying mRNA is an mRNA vector modified from the alphavirus genome. It does not have a specific microRNA binding site, and its replication and expression in different tissues are not subject to microRNA-mediated regulation.
- LNPs lipid nanoparticles
- the inherent physical and chemical properties of the lipid nanoparticles may mediate the self-amplifying mRNA to enter the liver and other parenchymal tissues, where it may express a large amount of the target gene encoded by the self-amplifying mRNA, causing potential liver toxicity.
- the present invention obtains an optimized alphavirus RNA replicase coding sequence (translation region), and further obtains an optimized self-amplifying mRNA backbone sequence, which can significantly reduce the production of non-specific enzyme cleavage products of DNA endonuclease BspQI compared with the original sequence, improve the integrity of self-amplifying mRNA in vitro transcription, and maintain the normal replication function and/or expression function of saRNA.
- the following invention is provided:
- One aspect of the present invention relates to a translation region (coding region) of an mRNA molecule, which encodes the alpha virus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C.
- the mRNA molecule translation region is an isolated mRNA molecule translation region.
- the 4509th base in the translation region of the mRNA molecule, is T, C or G.
- the 1672nd base is A
- the 1673rd base is G
- the 1674th base is C.
- the translation region of the mRNA molecule encodes the alphavirus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C and the 4509th base is T.
- the translation region of the mRNA molecule encodes the alphavirus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C, and the 4509th base is C.
- the translation region of the mRNA molecule encodes the alphavirus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C and the 4509th base is G.
- the translation region of the mRNA molecule encodes the alphavirus RNA replicase shown in SEQ ID NO: 20, wherein the 4500th base of the mRNA molecule is C, the 1672nd base is The 1673rd base is A, the 1674th base is G, and the 1674th base is C.
- the translation region of the mRNA molecule encodes the alpha virus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C, the 4509th base is T, the 1672nd base is A, the 1673rd base is G, and the 1674th base is C.
- the translation region of the mRNA molecule encodes the alpha virus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C, the 4509th base is C, the 1672nd base is A, the 1673rd base is G, and the 1674th base is C.
- the translation region of the mRNA molecule encodes the alpha virus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C, the 4509th base is G, the 1672nd base is A, the 1673rd base is G, and the 1674th base is C.
- the mRNA molecule translation region wherein the sequence of the initial mRNA molecule translation region encoding the alphavirus RNA replicase is shown as SEQ ID NO:10.
- the translation region of the mRNA molecule has a sequence as shown in any one of SEQ ID NO:11 to SEQ ID NO:15.
- Another aspect of the present invention relates to an mRNA molecule comprising, in sequence:
- RNA replicase coding region 5' untranslated region, RNA replicase coding region, RNA promoter, optional sequence encoding target protein, 3' untranslated region and polyadenylation sequence;
- RNA replicase coding region is the mRNA molecule translation region described in any one of the present invention.
- the polyadenylic acid is also called PolyA or PolyA tail, which may contain a small amount of non-adenylic acid (A), such as a small amount of T, C and/or G.
- A non-adenylic acid
- the intermediate non-A sequence can increase the stability of DNA template production.
- the mRNA molecule is an isolated mRNA molecule.
- the mRNA molecule wherein
- the 5' untranslated region is derived from an alpha virus; preferably, it is derived from Venezuelan equine encephalomyelitis virus;
- sequence of the 5’ non-translated region is as shown in SEQ ID NO:16.
- the mRNA molecule wherein
- the 3' untranslated region is derived from an alpha virus; preferably, derived from Venezuelan equine encephalomyelitis virus;
- sequence of the 3’ non-translated region is as shown in SEQ ID NO:17.
- the mRNA molecule wherein the RNA promoter is a subgenomic promoter
- the RNA promoter is a subgenomic promoter derived from an alphavirus
- the RNA promoter is a subgenomic promoter derived from Venezuelan equine encephalitis virus;
- the RNA promoter is a 26S promoter
- the sequence of the RNA promoter is as shown in SEQ ID NO:18.
- the mRNA molecule wherein the sequence of the polyadenylic acid sequence is as shown in SEQ ID NO:19.
- the mRNA molecule wherein the sequence encoding the target protein is a sequence encoding a vaccine antigen, a therapeutic protein or an antibody targeting an immune checkpoint.
- the mRNA molecule has a sequence as shown in SEQ ID NO: 3 and any one of SEQ ID NO: 6 to SEQ ID NO: 9.
- the mRNA molecule is a self-amplifying RNA (saRNA).
- Another aspect of the present invention relates to a DNA molecule encoding the translation region of the mRNA molecule described in any one of the present invention or encoding the mRNA molecule described in any one of the present invention.
- Another aspect of the present invention relates to a recombinant vector, which contains the DNA molecule of the present invention; preferably, the recombinant vector is a recombinant prokaryotic expression vector or a recombinant eukaryotic expression vector.
- Another aspect of the present invention relates to a recombinant host cell, which contains the translation region of the mRNA molecule described in any one of the present invention, ... DNA molecule described in any one of the present invention, or the recombinant vector described in the present invention.
- kits which contains the translation region of the mRNA molecule described in any one of the present invention, the mRNA molecule described in any one of the present invention or the DNA molecule described in the present invention, and a liposome delivery system.
- Another aspect of the present invention relates to a pharmaceutical composition, which contains the translation region of the mRNA molecule described in any one of the present invention, the mRNA molecule described in any one of the present invention or the DNA molecule described in the present invention, and one or more pharmaceutically acceptable excipients; preferably, the excipient is a liposome delivery system.
- Another aspect of the present invention relates to a vaccine preparation, which contains the translation region of the mRNA molecule described in any one of the present invention, the mRNA molecule described in any one of the present invention, or the DNA molecule described in the present invention;
- the mRNA molecule or DNA molecule is encapsulated by a liposome-based delivery system
- the vaccine formulation further comprises one or more vaccine adjuvants
- the vaccine preparation is a vaccine preparation for preventing viral infection such as novel coronavirus infection or preventing severe illness caused by novel coronavirus infection.
- Lipid delivery systems include liposomes, lipid nanoparticles (LNP), and lipid polymer nanocarriers (LPP).
- LNP lipid nanoparticles
- LPP lipid polymer nanocarriers
- LPP among lipids is a double-layer structure with polymer-encapsulated mRNA as the core and phospholipids as the shell.
- the double-layer liposome membrane of LPP has a better effect of encapsulating and protecting mRNA, and the core of LPP can gradually release mRNA molecules as the polymer degrades.
- LPP has excellent targeting effect on dendritic cells and can better activate the immune response of T cells through antigen presentation to achieve ideal therapeutic effects.
- mRNA can effectively stimulate cellular immunity and humoral immunity.
- the injected mRNA vaccine is internalized by antigen-presenting cells. After escaping the endosome and entering the cytoplasm, the mRNA is translated into protein by the ribosome.
- the translated antigen protein can stimulate the immune system in a variety of ways, stimulating the body's cellular immunity and humoral immunity.
- nucleic acid vaccines Compared with traditional inactivated vaccines, subunit vaccines and genetically engineered vaccines, nucleic acid vaccines have the following advantages: short R&D cycle; simple production process and easy expansion; no adjuvant is required and the effectiveness is high; it does not enter the cell nucleus and has good safety.
- the mRNA COVID-19 vaccine verifies the applicability of the mRNA technology platform in the vaccine field.
- Another aspect of the present invention relates to the use of the translation region of the mRNA molecule described in any one of the present invention, the mRNA molecule described in any one of the present invention, or the DNA molecule described in any one of the present invention in the preparation of a drug for treating or preventing viral infection, a drug for treating or preventing tumors, or a drug for protein replacement therapy.
- microRNA specifically expressed by a specific tissue binds to and regulates the replication of self-amplifying mRNA molecules. Amplification of mRNA backbone
- the present invention also obtains a self-amplifying mRNA skeleton that can be bound by microRNA specifically expressed in a specific tissue and then regulate the replication of self-amplifying mRNA molecules, thereby achieving the degradation of self-amplifying mRNA molecules in tissues expressing specific microRNAs, while existing normally in tissues that low-express or do not express specific microRNAs, so as to achieve tissue-specific expression of target gene molecules in the form of self-amplifying mRNA.
- the following invention is provided:
- Another aspect of the present invention relates to an mRNA molecule, comprising:
- RNA replicase sequence 5' non-translated sequence, RNA replicase sequence, RNA promoter, and optional sequence encoding target protein column, 3' untranslated sequence and 3' PolyA tail;
- a microRNA binding site sequence is contained between nsp1 and nsp2, between nsp2 and nsp3, between nsp3 and nsp4, and/or between the sequence encoding the target protein and the 3'-end non-translated sequence of the RNA replicase.
- the mRNA molecule wherein
- the RNA replicase is an RNA replicase derived from an alphavirus; preferably, it is an RNA replicase derived from Venezuelan equine encephalomyelitis virus;
- the amino acid sequence of the RNA replicase is as shown in SEQ ID NO:41; preferably, the coding sequence of the RNA replicase is as shown in SEQ ID NO:42.
- the mRNA molecule wherein
- the RNA promoter is a subgenomic promoter
- the RNA promoter is a subgenomic promoter derived from an alphavirus
- the RNA promoter is a subgenomic promoter derived from Venezuelan equine encephalitis virus;
- the RNA promoter is the 26S promoter.
- the mRNA molecule, wherein the 5' non-translated sequence and/or the 3' non-translated sequence is derived from an alpha virus; optimally, it is derived from Venezuelan equine encephalitis virus.
- the mRNA molecule wherein the microRNA binding site sequence is a binding site sequence corresponding to a tissue-specifically expressed microRNA
- the tissue-specifically expressed microRNA is a microRNA that is lowly expressed in tumor tissue
- the microRNA lowly expressed in tumor tissue is selected from miRNA-122, miRNA-143, miRNA-1, miRNA-124, miRNA-217 and miRNA-126.
- the mRNA molecule wherein the microRNA binding site sequence comprises one or more microRNA mature sequences; preferably, the microRNA binding site sequence comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 microRNA mature sequences that are identical or different in sequence.
- the mRNA molecule, wherein the microRNA mature sequence is shown in any one of SEQ ID NO:30 to SEQ ID NO:32.
- the microRNA binding site sequence is theoretically complementary to the microRNA.
- the length of the binding site sequence can be fine-tuned but needs to be greater than or equal to 20 nt.
- the length of MicroRNA-122-5p is 22 nt.
- the length of the binding sequence can be designed to be 20-24 nt, such as 20 nt, 21 nt, 22 nt, 23 nt or 24 nt.
- miRNA binding sites are usually located in the 3' untranslated region (3'UTR) of the target mRNA, and can also be located in the 5'UTR or coding region.
- MicroRNA binds to the target mRNA, thereby inhibiting the translation of the target mRNA or degrading the target mRNA, thereby affecting the expression of the target gene.
- the present invention introduces the binding sites corresponding to the tissue-specifically expressed microRNA into mRNA, and utilizes the microRNA-mediated post-transcriptional regulatory mechanism to regulate the stability and translation rate of different mRNA molecules, thereby achieving the differential expression of mRNA in different tissues, and further achieving the tissue-specific expression of mRNA.
- the mRNA molecule contains one or more identical or different internal spacer sequences between each microRNA mature sequence
- the internal spacer sequence is shown in any one of SEQ ID NO:33 to SEQ ID NO:34.
- the mRNA molecule wherein the 5' end and/or the 3' end of the microRNA binding site sequence contains one or more identical or different external spacer sequences;
- the external spacer sequence at the 5' end is shown in any one of SEQ ID NO:35 to SEQ ID NO:36;
- the external spacer sequence at the 3’ end is as shown in any one of SEQ ID NO:37 to SEQ ID NO:38.
- the mRNA molecule wherein the 5' end and/or 3' end of the microRNA binding site sequence contains one or more restriction site sequences that can be recognized by alphavirus RNA replicase or nsp2;
- nsp1 is a restriction site sequence between nsp1 and nsp2 that can be recognized by the alphavirus replicase or nsp2;
- the restriction site sequence is as shown in SEQ ID NO:53.
- nsp2 in the alphavirus replicase is a part that exerts protease activity, which can recognize a specific amino acid sequence between the four components of the alphavirus replicase, thereby cutting the replicase polyprotein into a single component to exert replication function. Therefore, the restriction site here should be a protease restriction site sequence that can be recognized by the alphavirus replicase or nsp2.
- the protease cleavage site sequence is preferably an amino acid sequence between nsp1 and nsp2 that can be recognized by the alphavirus replicase or nsp2.
- the sequence is: EAGA ⁇ GSVE (SEQ ID NO: 53), where ⁇ represents the cleavage site.
- the mRNA molecule, wherein the microRNA binding site The sequence is shown in SEQ ID NO:22.
- the mRNA molecule further contains a 5' end cap structure.
- the mRNA molecule has a sequence as shown in SEQ ID NO:24 or SEQ ID NO:28.
- the mRNA molecule, wherein the target protein is an antigen.
- the mRNA molecules of the present invention are tissue-specific self-amplifying nucleic acid molecules.
- the mRNA molecule is an isolated mRNA molecule.
- Another aspect of the present invention relates to an mRNA molecule combination, comprising a first mRNA molecule and a second mRNA molecule, wherein:
- the first mRNA molecule comprises, in order:
- the second mRNA molecule comprises, in order:
- the second 5' non-translated sequence the sequence essential for alphavirus replication, the RNA promoter, the sequence encoding the target protein, the second 3' non-translated sequence and the 3' PolyA tail;
- a microRNA binding site sequence is contained between nsp1 and nsp2, between nsp2 and nsp3, between nsp3 and nsp4, and/or between the sequence encoding the target protein and the second 3'-end non-translated sequence of the RNA replicase.
- the mRNA molecule combination wherein,
- the first 5' non-translated sequence is the same as or different from the second 5' non-translated sequence; and/or
- the first 3' non-translated sequence is the same as or different from the second 3' non-translated sequence.
- the mRNA molecule combination wherein the first 5' non-translated sequence and the first 3' non-translated sequence are not derived from alpha virus.
- the mRNA molecule combination wherein the second 5' non-translated sequence and the second 3' non-translated sequence are derived from an alpha virus; preferably, from Venezuelan equine encephalitis virus.
- the mRNA molecule combination wherein,
- the RNA replicase is an RNA replicase derived from an alphavirus; preferably, it is an RNA replicase derived from Venezuelan equine encephalomyelitis virus;
- the amino acid sequence of the RNA replicase is as shown in SEQ ID NO:41; preferably, the coding sequence of the RNA replicase is as shown in SEQ ID NO:42.
- the mRNA molecule combination wherein,
- the RNA promoter is a subgenomic promoter
- the RNA promoter is a subgenomic promoter derived from an alphavirus
- the RNA promoter is a subgenomic promoter derived from Venezuelan equine encephalitis virus;
- the RNA promoter is the 26S promoter.
- the mRNA molecule combination wherein the microRNA binding site sequence is a binding site sequence corresponding to a tissue-specifically expressed microRNA
- the tissue-specifically expressed microRNA is a microRNA that is lowly expressed in tumor tissue
- the microRNA lowly expressed in tumor tissue is selected from miRNA-122, miRNA-143, miRNA-1, miRNA-124, miRNA-217 and miRNA-126.
- the mRNA molecule combination wherein the microRNA binding site sequence comprises one or more microRNA mature sequences; preferably, the microRNA binding site sequence comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 microRNA mature sequences that are identical or different in sequence.
- the mRNA molecule combination wherein the microRNA mature sequence is shown in any one of SEQ ID NO:30 to SEQ ID NO:32.
- the mRNA molecule combination contains one or more identical or different internal spacer sequences between each microRNA mature sequence
- the internal spacer sequence is shown in any one of SEQ ID NO:33 to SEQ ID NO:34.
- the mRNA molecule combination wherein the 5' end and/or 3' end of the microRNA binding site sequence contains one or more identical or different external spacer sequences;
- the external spacer sequence at the 5' end is shown in any one of SEQ ID NO:35 to SEQ ID NO:36;
- the external spacer sequence at the 3’ end is as shown in any one of SEQ ID NO:37 to SEQ ID NO:38.
- the mRNA molecule combination wherein the microRNA
- the 5' end and/or 3' end of the cleavage site sequence contains one or more restriction site sequences that can be recognized by the alphavirus RNA replicase or nsp2;
- nsp1 is a restriction site sequence between nsp1 and nsp2 that can be recognized by the alphavirus replicase or nsp2;
- the restriction site sequence is as shown in SEQ ID NO:53.
- the mRNA molecule combination wherein the microRNA binding site sequence is as shown in SEQ ID NO:22.
- the mRNA molecule combination wherein the first mRNA molecule and/or the second mRNA molecule further contains a 5' end cap structure.
- the mRNA molecule combination wherein the target protein is an antigen.
- the mRNA molecule combination is as shown in Figure 23.
- the first mRNA molecule encodes nsp1 to nsp4, and the second mRNA molecule encodes the target protein.
- the mRNA molecule combination of the present invention is a tissue-specific self-amplifying nucleic acid molecule combination.
- Another aspect of the present invention relates to a DNA molecule encoding the mRNA molecule described in any one of the present invention, or encoding the first mRNA molecule and the second mRNA molecule described in any one of the present invention, and the first mRNA molecule and the second mRNA molecule are on the same DNA molecule.
- the DNA molecule comprises a T7 promoter, an SP6 promoter or a T3 promoter upstream of the sequence encoding the alphavirus RNA replicase.
- the DNA molecule is an isolated DNA molecule.
- Another aspect of the present invention relates to a DNA molecule combination, comprising a first DNA molecule and a second DNA molecule, wherein:
- the first DNA molecule encodes the first mRNA molecule described in any one of the present invention.
- the second DNA molecule encodes the second mRNA molecule described in any one of the present invention.
- the viral infection refers to novel coronavirus infection
- the tumor is selected from liver cancer, rhabdomyosarcoma, glioma, bladder cancer, colorectal cancer, pancreatic cancer, adenocarcinoma and lung cancer;
- the tumor is a tumor with low expression of one microRNA or multiple microRNAs; preferably, the microRNA is one or more selected from miRNA-122, miRNA-143, miRNA-1, miRNA-124, miRNA-217 and miRNA-126.
- MicroRNA is a type of non-coding single-stranded RNA molecule with a length of about 22nt encoded by endogenous genes. They participate in post-transcriptional gene expression regulation in animals and plants. MicroRNA recognizes the mRNA sequence of the target gene and binds to it, resulting in the inhibition of mRNA translation or the degradation of mRNA, thereby regulating the expression of the mRNA encoding gene. This gene expression regulation mechanism is called microRNA-mediated post-transcriptional regulation. MicroRNA has tissue specificity and its expression levels vary in different tissues. For example:
- miRNA-122 is highly expressed in the liver, but is less expressed in non-hepatic cells (e.g., muscle cells) and liver cancer tissues;
- miRNA-143 is highly expressed in colorectal tissues but lowly expressed in colorectal cancer tissues;
- miRNA-1 is highly expressed in skeletal muscle and cardiac muscle tissues, but is lowly expressed in non-muscle tissues and rhabdomyosarcoma tissues;
- miRNA-124 is highly expressed in neuronal tissues, but lowly expressed in glioma and bladder cancer tissues;
- miRNA-217 is highly expressed in pancreatic tissues but lowly expressed in pancreatic cancer tissues;
- miRNA-126 and miRNA-143 were highly expressed in lung tissues, but lowly expressed in lung cancer tissues.
- miRNA a powerful tool for disease diagnosis or treatment.
- any pathogen infection or any tumor type is applicable to this requirement. More specifically, it may also correspond to a tumor type with low expression of a certain type of specific microRNA. For example, most liver cancers have low expression of microRNA122, and the self-amplifying mRNA of the present invention can be specifically expressed in liver cancer tissue.
- Another aspect of the present invention relates to a recombinant vector, which contains the DNA molecule described in any one of the present invention; preferably, the recombinant vector is a recombinant prokaryotic expression vector or a recombinant eukaryotic expression vector.
- Another aspect of the present invention relates to a recombinant host cell, which contains the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the DNA molecule combination described in any one of the present invention.
- kits comprising the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the The DNA molecule combination described above, and the liposome delivery system.
- Another aspect of the present invention relates to a pharmaceutical composition, which contains the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the DNA molecule combination described in any one of the present invention, and one or more pharmaceutically acceptable excipients; preferably, the excipient is a liposome delivery system.
- Another aspect of the present invention relates to a vaccine preparation, which contains the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the DNA molecule combination described in any one of the present invention;
- the mRNA molecule, the combination of mRNA molecules, the DNA molecule or the combination of DNA molecules is encapsulated by a liposome-based delivery system;
- the vaccine formulation further comprises one or more vaccine adjuvants
- the vaccine preparation is a vaccine preparation for preventing viral infection such as novel coronavirus infection or preventing severe illness caused by novel coronavirus infection.
- Lipid delivery systems include liposomes, lipid nanoparticles (LNP), and lipid polymer nanocarriers (LPP).
- LNP lipid nanoparticles
- LPP lipid polymer nanocarriers
- LPP among lipids is a double-layer structure with polymer-encapsulated mRNA as the core and phospholipids as the shell.
- the double-layer liposome membrane of LPP has a better effect of encapsulating and protecting mRNA, and the core of LPP can gradually release mRNA molecules as the polymer degrades.
- LPP has excellent targeting effect on dendritic cells and can better activate the immune response of T cells through antigen presentation to achieve ideal therapeutic effects.
- mRNA can effectively stimulate cellular immunity and humoral immunity.
- the injected mRNA vaccine is internalized by antigen-presenting cells. After escaping the endosome and entering the cytoplasm, the mRNA is translated into protein by the ribosome.
- the translated antigen protein can stimulate the immune system in a variety of ways, stimulating the body's cellular immunity and humoral immunity.
- nucleic acid vaccines Compared with traditional inactivated vaccines, subunit vaccines and genetically engineered vaccines, nucleic acid vaccines have the following advantages: short R&D cycle; simple production process and easy expansion; no adjuvant is required and the effectiveness is high; it does not enter the cell nucleus and has good safety.
- the mRNA COVID-19 vaccine verifies the applicability of the mRNA technology platform in the vaccine field.
- Another aspect of the present invention relates to the use of the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the DNA molecule combination described in any one of the present invention in the preparation of a drug for treating or preventing viral infection, a drug for treating or preventing tumors, or a drug for protein replacement therapy;
- the viral infection refers to novel coronavirus infection
- the tumor is one or more selected from liver cancer, rhabdomyosarcoma, glioma, bladder cancer, colorectal cancer, pancreatic cancer and lung cancer;
- the tumor is a tumor with low expression of one microRNA or multiple microRNAs; preferably, the microRNA is one or more selected from miRNA-122, miRNA-143, miRNA-1, miRNA-124, miRNA-217 and miRNA-126.
- Another aspect of the present invention relates to a method for treating or preventing viral infection or tumor or a protein replacement therapy, comprising the step of administering to a subject in need thereof an effective amount of the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the DNA molecule combination described in any one of the present invention;
- the viral infection refers to novel coronavirus infection
- the tumor is one or more selected from liver cancer, rhabdomyosarcoma, glioma, bladder cancer, colorectal cancer, pancreatic cancer and lung cancer;
- the tumor is a tumor with low expression of one microRNA or multiple microRNAs; preferably, the microRNA is one or more selected from miRNA-122, miRNA-143, miRNA-1, miRNA-124, miRNA-217 and miRNA-126.
- the present invention also provides an improved saRNA construct, and a method for preparing and using the construct.
- the present invention further provides the use of the saRNA to carry a target gene for treatment, prevention and/or diagnosis.
- the present invention has obtained an optimized self-amplified mRNA backbone sequence, which can significantly reduce the intensity of the induced natural immune response, significantly reduce the translation inhibition, and significantly reduce cell apoptosis compared to the original sequence, thereby significantly improving the expression efficiency.
- One aspect of the present invention provides an mRNA molecule encoding an RNA replicase composed of non-structural proteins 1, 2, 3 and 4 derived from an alphavirus, wherein the non-structural protein 3 comprises a macrodomain at its N-terminus as shown in the amino acid sequence of SEQ ID NO: 57 or 59.
- the non-structural proteins 1, 2, 3 and 4 are derived from Venezuelan equine encephalitis virus.
- the mRNA molecule encodes a nonstructural protein 1 having an amino acid sequence as shown in SEQ ID NO: 71 or at least 90% identical to SEQ ID NO: 71, a nonstructural protein 2 having an amino acid sequence as shown in SEQ ID NO: 72 or at least 90% identical to SEQ ID NO: 73.
- the mRNA molecule encodes a nonstructural protein 3 having an amino acid sequence at least 90% identical to SEQ ID NO: 73 or 74 and comprising a macrodomain as shown in the amino acid sequence of SEQ ID NO: 57 or 59 at the N-terminus.
- the mRNA molecule comprises a nucleotide sequence encoding the macro domain as shown in SEQ ID NO:68 or 70.
- the mRNA molecule comprises or consists of the nucleotide sequence of SEQ ID NO:67 or 69.
- the mRNA molecule further comprises a target gene coding sequence and, optionally, an RNA promoter upstream of the target gene coding sequence.
- the mRNA molecule further comprises:
- RNA promoter 5' untranslated region, RNA promoter, target gene coding sequence, 3' untranslated region and polyadenylation sequence,
- the mRNA molecule further comprises a 5' cap sequence, a signal peptide coding sequence, a Kozak sequence and/or a restriction enzyme cleavage site.
- the Kozak sequence can be connected to the 3' end of the RNA promoter.
- the mRNA molecule comprises the following operably linked nucleotide sequences in order from 5' to 3': a 5' untranslated region, coding sequences of nonstructural proteins 1, 2, 3 and 4, an RNA promoter, a target gene coding sequence, a 3' untranslated region and a polyadenylation sequence,
- nucleotide sequences may be connected via a linker sequence.
- the coding sequences of non-structural proteins 1, 2, 3 and 4 comprise a nucleotide sequence encoding non-structural protein 1, a nucleotide sequence encoding non-structural protein 2, a nucleotide sequence encoding non-structural protein 3 and a nucleotide sequence encoding non-structural protein 4 that are operably linked.
- the coding sequences of non-structural proteins 1, 2, 3 and 4 include from 5' to 3': a nucleotide sequence encoding non-structural protein 1, a nucleotide sequence encoding non-structural protein 2, a nucleotide sequence encoding non-structural protein 3 and a nucleotide sequence encoding non-structural protein 4 that are operably linked.
- the 5' untranslated region is derived from an alpha virus; preferably, from a Venezuelan equine encephalitis virus; for example, the 5' untranslated region comprises a nucleotide sequence that is at least 85% identical to SEQ ID NO:63.
- the 3' untranslated region is derived from an alphavirus; preferably, from Venezuela Equine encephalitis virus; for example, the 3' untranslated region comprises a nucleotide sequence that is at least 85% identical to SEQ ID NO:64.
- the RNA promoter is a subgenomic promoter, such as a subgenomic promoter derived from an alphavirus.
- the RNA promoter is a subgenomic promoter derived from Venezuelan equine encephalitis virus.
- the RNA promoter is a 26S promoter.
- the sequence of the RNA promoter is shown in SEQ ID NO:65.
- sequence of the poly(A) sequence is as shown in SEQ ID NO:66.
- the target gene coding sequence is a sequence encoding a therapeutic polypeptide, a preventive polypeptide, a diagnostic polypeptide, a reporter gene, an antigen, or a sequence encoding a regulatory structure (eg, a non-coding gene).
- the mRNA molecule comprises a nucleotide sequence as shown in any one of SEQ ID NO:58, SEQ ID NO:60 to SEQ ID NO:62.
- the invention provides a DNA molecule encoding an mRNA molecule as disclosed herein.
- the present invention provides a recombinant vector comprising a DNA molecule as disclosed herein; preferably, the recombinant vector is a prokaryotic expression vector or a eukaryotic expression vector.
- the present invention provides a recombinant host cell comprising an mRNA molecule, a DNA molecule or a recombinant vector as disclosed herein.
- the present invention provides a pharmaceutical composition
- a pharmaceutical composition comprising an mRNA molecule or a DNA molecule as disclosed herein, and one or more pharmaceutically acceptable carriers; preferably, the carrier is a liposome delivery system.
- the present invention provides a vaccine formulation comprising an mRNA molecule or a DNA molecule as disclosed herein; preferably, the mRNA molecule or the DNA molecule is encapsulated by a liposome delivery system.
- the vaccine formulation may also contain one or more vaccine adjuvants.
- the target gene encoded by the mRNA may encode a vaccine antigen.
- the vaccine formulation is a vaccine formulation for preventing viral infection such as novel coronavirus infection or preventing severe illness caused by novel coronavirus infection.
- the target gene encoded by the mRNA may encode an immunogenic peptide of the novel coronavirus.
- the present invention provides the use of the mRNA molecule or DNA molecule disclosed herein in the preparation of a drug for treating or preventing viral infection, a drug for treating or preventing tumors, or a drug for protein replacement therapy.
- the target gene encoded by the mRNA can be a vaccine antigen gene, a tumor killing gene, a therapeutic protein, or a Protein genes, antibody genes, etc.
- the invention provides a kit comprising an mRNA molecule or a DNA molecule as disclosed herein.
- Figure 1 Capillary electrophoresis results of in vitro transcribed RNA after modification of position 4497-4503 in the saRNA-EGFP template plasmid psaRNA-062.
- FIG. 2 Overlay of RNA capillary electrophoresis results of in vitro transcription of different saRNA templates modified using psaRNA-152 as a template.
- saRNA-062 is the original saRNA
- saRNA-183 to 185 are the modified saRNAs.
- Figure 3 Capillary electrophoresis results of in vitro transcribed RNA after modification of positions 1680-1676 in psaRNA-183 as template.
- the black box indicates the electrophoresis detection peak shape of the short transcript before and after backbone optimization.
- Figure 4 Expression level results of optimized luciferase-encoding saRNA-152 and original saRNA-062 after transfection of BHK-21 cells for 24 and 72 hours.
- Figure 5 Expression level results of saRNA-243 encoding luciferase and original saRNA-062 obtained after detection optimization after transfection of BHK-21 cells for 24 hours.
- Figure 6 Replication results of optimized luciferase-encoding saRNA-152 and original saRNA-062 after transfection of BHK-21 cells for 24 and 72 hours. The data were normalized to the results 2 hours after transfection.
- Figure 7 Capillary electrophoresis results for detecting the integrity of S23H02.
- Figure 8 E7-ELISA test results for detecting the secreted HPV16 antigen expression of S23H02, M22H04 and negative control after transfection into tissue culture containing Lipofectamine 3000.
- Figure 9 Tumor volume data of C57bl/6 mice challenged with TC1 tumor cells and injected with 3 doses of therapeutic vaccine. Tumor volume and mouse body weight were recorded. The dose per mouse per dose was calculated based on the active ingredient.
- Figure 10 Body weight data of C57bl/6 mice after being challenged with TC1 tumor cells and injected with 3 doses of therapeutic vaccine. The dose per mouse per dose was calculated based on the active ingredient.
- Figure 11 Flow cytometry was used to detect the number of E7-specific CD8 T cells in the spleen cells of C57bl/6 mice injected with one dose of therapeutic vaccine. The dose per mouse per dose was calculated based on the active ingredient.
- Figure 12 Schematic diagram of the miRNA122 binding site sequence box. After the inner spacer sequence is separated, an outer spacer sequence is added at both ends, and the outermost portion is a protease cleavage site that can be recognized by the replication enzyme.
- Figure 13 Self-amplifying mRNA structure, the structural components from left to right are cap structure-5-terminal alphavirus non-translated sequence (5'UTR)-alphavirus replicase sequence-26S promoter sequence-arbitrary target protein sequence X-3-terminal alphavirus non-translated sequence (3'UTR)-polyadenine sequence (polyA sequence).
- Figure 14 Schematic diagram of the self-amplified mRNA structure with microRNA122 binding sites inserted in different positions. Arrows represent different insertion positions of microRNA122 binding sites. The microRNA122 binding site is obtained by combining 6 microRNA122 binding sites in series.
- Figure 15 Denaturing agarose gel electrophoresis detection results of in vitro transcription of self-amplified mRNA inserted into the microRNA122 binding site at six different locations.
- Figure 16 qPCR detection results of microRNA-122 in different cells.
- Figure 17 24 hours after transfection of Huh7.5.1 cells and C2C12 cells with self-amplified mRNA expressing Nluc-EGFP and inserted microRNA122 binding site, the expression of fluorescent protein was detected by fluorescence microscopy. The control was self-amplified mRNA without inserted microRNA122 binding site.
- Figure 18 Expression of Nluc luciferase 24 hours after transfection of self-amplified mRNA expressing Nluc-EGFP with microRNA122 binding site inserted into Huh7.5.1 cells and C2C12 cells.
- the control is self-amplified mRNA without microRNA122 binding site inserted.
- the expression value is normalized with firefly luciferase.
- Figure 19 Relative RNA levels of self-amplified mRNA in C2C12 cells 24 hours after transfection of self-amplified mRNA expressing Nluc-EGFP with microRNA122 binding site inserted into cells. Control is self-amplified mRNA without microRNA122 binding site inserted. The relative RNA level value after 24 hours of transfection is normalized with that after 2 hours of transfection. nsp1 represents the relative level of RNA encoding replicase sequence, and EGFP represents the relative level of RNA encoding EGFP sequence.
- Figure 20 Relative RNA level of self-amplified mRNA in Huh7.5.1 cells 24 hours after transfection of self-amplified mRNA expressing Nluc-EGFP with microRNA122 binding site inserted into the cells. Control is self-amplified mRNA without microRNA122 binding site inserted. The relative RNA level value after 24 hours of transfection is normalized with that after 2 hours of transfection. nsp1 represents the relative level of RNA encoding the replicase sequence, and EGFP represents the relative level of RNA encoding the EGFP sequence.
- Figure 21 Schematic diagram of self-amplified mRNA with different copy numbers of liver-specific microRNA-122 binding sequences inserted intention.
- FIG. 22 Expression of Nluc luciferase 24 hours after transfection of self-amplified mRNA expressing Nluc-EGFP and inserted with different copies of microRNA122 binding sites into Huh7.5.1 or C2C12 cells.
- Figure 23 Schematic diagram of mRNA molecule assembly.
- Figure 24 The position of the mutated amino acid in the macrodomain of saRNA and the comparison results of the amino acid sequences of different virus macrodomains.
- Macro is the macrodomain
- AUD is the alphavirus-unique domain
- HVD is the hypervariable domain
- different shapes represent different mutations
- the amino acids with more than 50% homology in the amino acid comparison results are marked with shadows.
- SARS-CoV Severe Acute Respiratory Syndrome Coronavirus
- SARS-CoV-2 Severe Acute Respiratory Syndrome Coronavirus 2
- MERS-CoV Middle East Respiratory Syndrome Coronavirus
- SFV Semliki Forest Virus
- CHIKV Chikungunya Virus
- SINV Sindbis Virus
- MAYV Mayaro virus
- EEEV Eastern Equine Encephalitis Virus
- VEEV Venezuelan Equine Encephalitis Virus.
- Figure 25 Schematic diagram of the protein structure of the saRNA macrodomain. The positions marked by arrows are ADP ribose, glutamine at position 48 (abbreviated Q), and isoleucine at position 113 (abbreviated I) bound to the macrodomain.
- Figure 26 Western blot test results of the effect of macrodomain amino acid mutations on total cellular protein ADP-ribosylation.
- Mock is a simulated transfection group, in which only transfection reagent was added without transfection of any RNA molecules.
- GAPDH protein was used as a cellular protein internal reference. The bottom value is the ratio of total protein ADP-ribosylation level of each group relative to the control group.
- Figure 27 Comparison of mutant saRNA subgenomic RNA replication capacity in hela cells. The significant differences between the groups were statistically analyzed using two-way ANOVA. **** and ⁇ represent that the P values of WT-saRNA and 190-saRNA or 191-saRNA groups are less than 0.0001, respectively.
- Figure 28 Comparison of the ability of mutant saRNA to form double-stranded RNA during hela cell replication.
- Figure 28a is a schematic diagram of the flow cytometry results 24 hours after transfection, and
- Figure 28b is a statistical bar graph of the proportion of double-stranded RNA.
- Figure 29 Principal component analysis of transcriptome levels 24 hours after Hela cells were transfected with different saRNAs.
- FIG. 30 Venn diagram of the number of genes upregulated or downregulated in different saRNAs relative to the control group.
- Figure 31 Ridge plot of differential gene cluster analysis in wild-type saRNA relative to the control group.
- Figure 31a is a biological function cluster analysis of differential genes
- Figure 31b is a molecular function cluster analysis of differential genes
- the horizontal axis is the log2 value of the change fold
- green represents the adjusted P value
- blue dots represent each down-regulated gene
- red dots represent each up-regulated gene.
- Figure 32 Heat map of genes at the transcriptome level of the interferon pathway after transfection with different saRNAs.
- Figure 33 Fluorescence quantitative PCR verification results of genes related to the innate immune pathway 2, 6, and 24 hours after HeLa cells were transfected with different saRNAs.
- Figure 34 Luciferase expression levels after HeLa cells were transfected with different saRNAs for 2, 6, 24 and 48 hours.
- Figure 35 Western blot detection results of cell translation activity after 2, 6, and 24 hours of transfection with different saRNAs. GAPDH was used as an internal reference.
- Figure 36 Detection of ribosomal RNA integrity after 2, 6, 24 and 48 hours of transfection of Hela cells with different saRNAs.
- Figure 36a is a schematic diagram of the detection of ribosomal RNA integrity 24 hours after transfection of saRNA using capillary electrophoresis, with arrows marking degraded ribosomal RNA.
- Figure 36b is the statistical results of the detection of ribosomal RNA integrity at different times.
- Figure 40 Detection results of activated caspase 8 and caspase 3 proteins that were cleaved 24 hours after transfection with different saRNAs, with the uncleaved full-length caspase as a control. GAPDH was used as an internal reference.
- Figure 41 Detection of cell apoptosis and EGFP expression intensity in Hela cells transfected with wild-type saRNA treated or not with Caspase inhibitors.
- Figure 43 Time curve of luciferase expression level in mice after mRNA-LNP immunization.
- Figure 44 Statistical results of the area under the curve of in vivo expression levels in mice after mRNA-LNP immunization.
- RNA self-amplifying RNA
- saRNA sa-mRNA
- saRNA sa-mRNA
- saRNA is similar to mRNA and generally contains a 5'UTR, a 3'UTR, and a poly(A) tail, but additionally contains a non-structural protein sequence derived from a virus (e.g., an alphavirus) and a subgenomic promoter (sgPr) located upstream of the target protein coding sequence.
- a virus e.g., an alphavirus
- sgPr subgenomic promoter
- saRNA is a positive-strand RNA molecule that is first translated by the ribosome into several non-structural protein components and assembled into RNA replicase after entering the cell.
- RNA replicase first uses the saRNA that first enters the cell to synthesize the saRNA negative strand, and then uses the negative strand as a template to synthesize new saRNA copies, thereby achieving self-amplification of saRNA.
- RNA replicase also recognizes sgPr, and then begins to synthesize subgenomic RNA from its downstream, which accumulates in large quantities in the host cell.
- the target protein encoded by saRNA is an antigen protein (for example, used as a vaccine)
- the antigen gene encoded by the subgenomic RNA will translate a large number of antigen molecules and trigger cellular antigen presentation.
- the saRNA described herein includes both saRNA backbone sequences without a target protein coding sequence and saRNA that operably connects the saRNA backbone sequence to the target protein coding sequence.
- the term "macrodomain” refers to a conserved protein domain encoded by viruses such as alphaviruses, coronaviruses, and hepatitis E virus, which can recognize and remove ADP ribosylation and is therefore considered an ADP ribosyl hydrolase.
- viruses such as alphaviruses, coronaviruses, and hepatitis E virus, which can recognize and remove ADP ribosylation and is therefore considered an ADP ribosyl hydrolase.
- viruses such as alphaviruses, coronaviruses, and hepatitis E virus, which can recognize and remove ADP ribosylation and is therefore considered an ADP ribosyl hydrolase.
- viruses such as alphaviruses, coronaviruses, and hepatitis E virus, which can recognize and remove ADP ribosylation and is therefore considered an ADP ribosyl hydrolase.
- Many studies have shown that the reduction of ADP
- RNA replicase is also called RNA-dependent RNA replicase (RdRp), which is a type of RNA polymerase that can synthesize RNA using RNA as a template. It is present in most RNA viruses and plays a role in replicating viral RNA and synthesizing mRNA. It is an enzyme required for the replication of other RNA viruses and viroids except retroviruses.
- the RNA replicase used for saRNA is usually derived from alphaviruses and contains nonstructural proteins 1, 2, 3, and 4 (nsP1, nsP2, nsP3, and nsP4).
- subgenomic promoter refers to a nucleic acid sequence upstream (5' end) of a nucleic acid sequence (e.g., a coding sequence) that controls the expression of a nucleic acid sequence by providing recognition and structural sites for RNA polymerase, usually RNA-dependent RNA polymerase, and in particular functional alphavirus nonstructural proteins.
- the subgenomic promoter is a genetic element of a positive-strand RNA virus (e.g., an alphavirus).
- the subgenomic promoter of an alphavirus is a nucleic acid sequence contained in the viral genomic RNA.
- RNA-dependent RNA polymerase e.g., a functional alphavirus nonstructural protein
- the RNA (-) strand i.e., the complementary strand of the alphavirus genomic RNA serves as a template for the synthesis of a (+) strand subgenomic transcript, and the synthesis of a (+) strand subgenomic transcript typically begins at or near the subgenomic promoter.
- subgenomic refers to a nucleotide sequence (such as RNA or DNA) that is smaller in length or size than the genomic nucleotide sequence from which it is derived.
- a subgenomic region may be a region encoding a VEEV structural protein, and a subgenomic RNA may be transcribed from a subgenomic region using an internal subgenomic promoter, the sequence of which is located in the genomic viral RNA or its complement. Transcription of the subgenomic region may be mediated by a virally encoded polymerase associated with a host cell-encoded protein (e.g., nsP1-4).
- a host cell-encoded protein e.g., nsP1-4
- Alphavirus structural proteins (core nucleocapsid protein C, envelope protein E2 and envelope protein E1, all components of the virus particle) are usually encoded by a single open reading frame under the control of a subgenomic promoter (Strauss & Strauss, Microbiol. Rev., 1994, vol. 58, pp. 491-562).
- the subgenomic promoter is recognized by cis-acting alphavirus non-structural proteins.
- the alphavirus replicase uses the (-) strand complementary strand of the genomic RNA as a template to synthesize the (+) strand subgenomic transcript.
- the (+) strand subgenomic transcript encodes alphavirus structural proteins (Kim et al., 2004, Virology, vol.
- the subgenomic RNA transcript serves as a template for translation of an open reading frame encoding a structural protein as a polyprotein, and the polyprotein is cleaved to produce the structural protein.
- a packaging signal located within the nsP2 coding sequence ensures the selective packaging of the genomic RNA into budding virions packaged by structural proteins (White et al., 1998, J. Virol., vol. 72, pp. 4320-4326).
- sequence essential for alphavirus replication is a sequence that can be recognized by alphavirus replicase and initiate subsequent RNA transcription and replication, which is intercepted from the alphavirus replicase sequence.
- the sequence essential for alphavirus replication must contain a minimum sequence (shown in SEQ ID NO:54), and the length can be increased or adjusted based on the shortest sequence.
- microRNA mature sequence refers to a microRNA molecule formed after the microRNA precursor (pre-miRNA) that forms a hairpin structure is processed by the Dicer enzyme.
- the two arms of the microRNA precursor each produce a functional mature microRNA, which targets different sites. They are generally named “-5p” (or 5p) and “-3p” (or 3p), such as hsa-miR-21-5p and hsa-miR-21-3p, respectively, indicating that the mature microRNA sequence is derived from hsa-
- the 5' and 3' arms of the mir-21 precursor are processed. From the sequence point of view, most of the sequences of 5p and 3p are complementary.
- the term "integrity" refers to the percentage of the amount of the target RNA product produced after the RNA in vitro transcription reaction is completed (including the target RNA product and the unexpected RNA product) produced; the amount of the target RNA product and the amount of the total RNA product can be determined by electrophoresis. For example, if the amount of the target saRNA product determined by the electrophoresis method accounts for 80% of the amount of all RNA products, the saRNA integrity is 80%.
- the "first” e.g., the first mRNA molecule, the first DNA molecule, etc.
- the "second” e.g., the second mRNA molecule, the second DNA molecule, etc.
- the term "effective amount” refers to the amount of the mRNA molecule of any one of the present invention, the mRNA molecule combination of the present invention, the DNA molecule of any one of the present invention, or the saRNA molecule of any one of the present invention that can prevent or treat the indications or symptoms described in the present invention, or reduce and/or alleviate the indications or symptoms in a subject.
- mRNA can be used to prevent infectious diseases, treat tumors, and for protein replacement therapy.
- mRNA drugs have rich targets and are not restricted by the drugability of the target protein or intracellular/extracellular restrictions.
- the mRNA sequence is easy to design, and the patient's own cells are used to produce molecules, bypassing the challenges of chemical synthesis, and the self-induced molecules have stronger efficacy.
- the mRNA production platform has strong scalability and replicability, and is easy to scale up.
- mRNA vaccines targeting infectious diseases encode antigens of related pathogens. After injection, they can express specific antigens in the body, induce both cellular immunity and humoral immunity, and stimulate the production of corresponding antibodies and immune cells to prevent the corresponding pathogens.
- the mRNA encoding tumor-specific antigen targets is delivered into the body in a specific way, so that these tumor-specific antigens are translated and presented on the cell surface, thereby activating the immune system, enabling it to specifically recognize and kill tumors.
- Tumor cells are delivered into the body in a specific way, so that these tumor-specific antigens are translated and presented on the cell surface, thereby activating the immune system, enabling it to specifically recognize and kill tumors.
- protein replacement therapy is used to treat rare monogenic diseases and aims to restore enzyme function.
- Protein synthesis is difficult, administration presents many challenges and is expensive, and using mRNA to turn the human body into its own protein processing plant is theoretically an economically feasible and efficient way.
- mRNA-based protein replacement therapy mainly focuses on inherited metabolic diseases. mRNA can theoretically synthesize any protein and can be used as a protein supplement or replacement therapy to treat a variety of diseases.
- higher safety requirements are required.
- the present invention constructs an improved saRNA, wherein the saRNA comprises a specific mutation in the macrodomain of the encoded replicase, so that the saRNA has a reduced ADP ribose hydrolysis activity.
- the mutation moderately weakens the replication ability of the replicase encoded by the sgRNA, thereby causing a decrease in the proportion of double-stranded RNA formed in the replication of saRNA.
- double-stranded RNA is a byproduct in the replication process of saRNA, and is often recognized by pattern recognition receptors in cells to stimulate the innate immune response of cells, the reduction of double-stranded RNA reduces the activation of natural immune receptors, thereby appropriately reducing the intensity of the natural immune response induced by saRNA.
- this improved sgRNA may include nucleotide sequences encoding various target proteins (e.g., antigenic proteins), and saRNA that effectively induces specific immunity in vivo can be obtained, and prepared into infectious diseases and tumor vaccines.
- the present application reduces the ADP ribose hydrolysis activity by amino acid mutation of the macro domain in the replicase protein domain encoded by saRNA, thereby reducing the replication efficiency of saRNA, thereby reducing the immunogenicity compared to conventional saRNA, and achieving the improvement of expression efficiency. Therefore, the present invention provides a mutation macro domain and a nucleic acid molecule (such as an mRNA molecule or a DNA molecule) encoding the mutation macro domain.
- the present invention also provides a replicase comprising the mutation macro domain and a nucleic acid molecule (such as an mRNA molecule or a DNA molecule) encoding the replicase.
- the present invention further provides a nucleic acid molecule (such as a saRNA molecule or a DNA molecule) constructed by combining a nucleic acid sequence encoding the replicase with a sequence encoding a target gene.
- the saRNA disclosed herein comprises a nucleotide sequence encoding an RNA replicase, wherein the RNA replicase comprises a mutant macrodomain having a Q48P or I113F substitution compared to the wild-type macrodomain from VEEV (as shown in SEQ ID NO: 55) (numbering according to SEQ ID NO: 56). NO:55).
- the mutant macrodomain encoded by the saRNA may comprise an amino acid sequence as shown in SEQ ID NO:57 or SEQ ID NO:59.
- the saRNA may comprise a nucleotide sequence encoding a mutant macrodomain as shown in SEQ ID NO:68 or SEQ ID NO:70.
- the replicase may be an alphavirus replicase, for example, comprising alphavirus nonstructural proteins 1, 2, 3 and 4.
- the saRNA disclosed herein comprises nucleotide sequences encoding replicase constituent proteins nsP1, nsP2, nsP3 and nsP4 derived from alphavirus, wherein the four replicase constituent proteins are capable of assembling into an RNA replicase complex, wherein the mutant macrodomain is located at the N-terminus of nsP3.
- the four non-structural protein components nsP1, nsP2, nsP3 and nsP4 are assembled into an RNA replicase complex in the form of a polyprotein.
- nsP1-4 all have their unique functions, wherein nsP4 plays the role of an RNA polymerase with RNA as a template.
- the sequence of the replicase constituent protein encoded by the saRNA may be derived from, for example, Venezuelan equine encephalitis virus (VEEV) and forest encephalitis virus (SFV), etc.
- the domain sequence of at least one non-structural replicase comprises a sequence selected from Group IV RNA viruses, including Eastern equine encephalitis virus (EEEV), Venezuelan equine encephalitis virus (VEEV), Everglades virus, Mucambo virus, Pixuna virus, Western equine encephalitis virus (WEE), Sindbis virus, forest encephalitis virus (SFV), etc.
- the plurality of non-structural protein sequences forming the replicase are derived from an alphavirus such as VEEV.
- the saRNA may comprise a nucleotide sequence encoding a replicase constituent protein as shown in SEQ ID NO: 67 or SEQ ID NO: 69.
- the native alphavirus genome encodes structural proteins in addition to the non-structural replicase, in some embodiments, the alphavirus-based saRNA of the present invention does not encode alphavirus structural proteins.
- the 5' untranslated region may be derived from a variety of viruses, such as alphaviruses and coronaviruses. In some embodiments, the 5' untranslated region is from Venezuelan equine encephalitis virus. Specifically, the 5' untranslated region may comprise a nucleotide sequence as shown in SEQ ID NO: 63. The 3' untranslated region may be derived from a variety of viruses, such as alphaviruses and coronaviruses.
- the 3' untranslated region is from Venezuelan equine encephalitis virus.
- the 3' untranslated region may include a nucleotide sequence as shown in SEQ ID NO: 64.
- the poly(A) sequence includes 20-200 adenylic acids.
- the poly(A) sequence may include a restriction enzyme cleavage site inside or at the 3' end. Location.
- the saRNA further comprises a nucleotide sequence encoding a target protein or a target gene (GOI) coding sequence, wherein the target protein is or the target gene encodes any one selected from the following: a therapeutic polypeptide, a preventive polypeptide, a diagnostic polypeptide, a reporter gene, an antigen, or a gene encoding a regulatory structure.
- the target protein can be an infectious disease antigen, an allergic antigen, or a tumor antigen.
- the target gene can encode a non-coding gene, such as siRNA, microRNA, gRNA, etc.
- the saRNA further comprises a nucleotide sequence encoding one or more therapeutic proteins or immunomodulators for use as disease therapeutic agents.
- the immunomodulators can be cytokines, chemokines or other immunostimulants or inhibitors.
- the saRNA of the present disclosure has at least two coding regions, the first coding region encodes multiple non-structural replicase domain sequences; the second coding region encodes a gene of interest operably linked to a subgenomic promoter.
- saRNA disclosed herein may comprise the following 5' to 3' operably linked nucleic acid sequence: 5'UTR-nsP-SGP-GOI-3'UTR-Poly A,
- the saRNA disclosed herein may comprise the following nucleic acid sequence operably linked from 5' to 3': 5'UTR-nsP1-nsP2-nsP3-nsP4-SGP-GOI-3'UTR-Poly A,
- 5'UTR is 5' untranslated region
- nsP (including nsP1, nsP2, nsP3, nsP4) is a plurality of non-structural protein sequences capable of forming replicase
- SGP is a subgenomic promoter
- GOI is one or more target protein encoding genes
- 3'UTR is 3' untranslated region
- Poly-A is 3' polyadenylic acid tail
- nsP comprises a mutated macrodomain.
- each GOI can be operably linked to its respective SGP.
- the SGP is a 26S promoter, for example as shown in SEQ ID NO:65.
- the at least 85% identical can be at least 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identical, and encompasses functional variants that retain the corresponding functions of these sequences.
- 5'UTR, nsP, SGP and 3'UTR sequences are all derived from alphavirus.
- the alphavirus genome encodes 4 nonstructural proteins and 6 structural proteins (capsid, E3, E2, 6K, TF and E1).
- Alphavirus structural proteins core nucleocapsid protein C, envelope protein E2 and envelope protein E1, all components of viral particles
- the subgenomic promoter is recognized by cis-acting alphavirus nonstructural proteins.
- the alphavirus replicase uses the (-) strand complementary strand of the genomic RNA as a template to synthesize the (+) strand subgenomic transcript.
- (+) strand subgenomic transcripts encode alphavirus structural proteins (Kim et al., 2004, Virology, vol. 323, pp. 153-163, Vasiljeva et al., 2003, J. Biol. Chem. vol. 278, pp. 41636-41645).
- Subgenomic RNA transcripts serve as templates for translation of open reading frames encoding structural proteins as a polyprotein, and the polyprotein is cleaved to produce structural proteins.
- the saRNA disclosed herein can linearize the DNA plasmid template before being prepared by in vitro transcription.
- a restriction enzyme site can be added to the 3' end of the saRNA to facilitate cutting.
- the saRNA can further contain other coding sequences, such as a signal peptide coding sequence at the 5' end, other sequences compatible with the replicase coding sequence, a Kozak sequence downstream of the SGP promoter, and additional downstream coding regions, for example, for encoding other desired gene products.
- the saRNA disclosed herein comprises a saRNA backbone sequence as shown in SEQ ID NO: 61 or 62 (i.e., without adding a target protein coding sequence or a target gene sequence).
- the saRNA disclosed herein comprises a nucleic acid sequence as shown in SEQ ID NO: 58 or 60, wherein luciferase and green fluorescent fusion protein coding sequences are used as exemplary target proteins.
- saRNA can be prepared by in vitro transcription of DNA encoding saRNA using a suitable DNA-dependent RNA polymerase, wherein the DNA
- RINA-dependent polymerases include T7 phage RNA polymerase, SP6 phage RNA polymerase, T3 phage RNA polymerase, T5 phage RNA polymerase, RNA polymerase III, RNA polymerase II, Taq polymerase, etc., or mutants of these polymerases.
- the transcription reaction will contain nucleotides as well as other components that support the activity of the selected polymerase, such as a suitable buffer and suitable salts.
- nucleotide analogs can also be incorporated into saRNA to, for example, alter the stability of such RNA molecules, increase resistance to ribonucleases, establish replication after introduction into appropriate host cells, and/or induce or reduce innate and adaptive immune responses.
- mRNA-based therapy is the delivery of in vitro synthesized mRNA to specific cells in the human body, where it is translated into the desired protein in the cytoplasm.
- mRNA can be used to prevent infectious diseases, treat tumors, and for protein replacement therapy.
- mRNA drugs have rich targets and are not restricted by the drugability of the target protein or intracellular/extracellular restrictions.
- the mRNA sequence is easy to design, and the patient's own cells are used to produce molecules, bypassing the challenges of chemical synthesis, and the self-induced molecules have stronger efficacy.
- the mRNA production platform has strong scalability and replicability, and is easy to scale up.
- the present disclosure relates to a pharmaceutical composition, such as a vaccine composition comprising a saRNA of the present disclosure and a pharmaceutically acceptable carrier.
- the pharmaceutically acceptable carrier can be a saRNA delivery system, preferably a nanoparticle composition.
- the nanoparticle composition comprises a cationic lipid, a PEG-modified lipid, a sterol, and a non-cationic lipid.
- the sa-mRNA disclosed herein can be used for vaccination.
- the vaccine can be a preventive vaccine in the field of infectious diseases.
- the target protein encoded in the saRNA is an antigen of a pathogen related to an mRNA vaccine encoding an infectious disease.
- specific antigens can be expressed in the body, which can simultaneously induce cellular immunity and humoral immunity, stimulate the production of corresponding antibodies and immune cells to prevent the corresponding pathogens.
- the vaccine can be a tumor therapeutic vaccine.
- mRNA encoding tumor-specific antigen targets is delivered to the body in a specific manner, so that these tumor-specific antigens are translated and presented on the cell surface, thereby activating the immune system so that it can specifically recognize and kill tumor cells.
- the saRNA disclosed herein can be used to treat diseases, for example, by providing therapeutic proteins or immune regulatory proteins for diseases.
- protein replacement therapy is used to treat rare monogenic diseases, aiming to restore enzyme function.
- Protein synthesis is difficult, administration is difficult and expensive, while the use of mRNA Turning the human body into a processing plant for its own proteins is theoretically an economically feasible and efficient way.
- the saRNA disclosed in the present invention can theoretically synthesize any target protein and can be used as a protein supplement or alternative therapy to treat a variety of diseases.
- the present disclosure provides a method of delivering a protein of interest to a subject, comprising administering to the subject a pharmaceutical composition containing one or more self-amplifying mRNAs of the present disclosure.
- the present invention achieves one or more of the following technical effects (1) to (3):
- the present invention improves the integrity of saRNA in vitro transcription.
- the present invention has no negative impact on the replication function of saRNA.
- the present invention has no negative impact on the expression function of saRNA.
- the current self-amplifying mRNA is an mRNA vector modified from the alphavirus genome, and has no specific microRNA binding site.
- the self-amplifying mRNA may be mediated to enter the liver and other parenchymal tissues for expression in large quantities, which may cause potential liver toxicity in some application examples. Therefore, in another aspect, the technical effect achieved by the present invention also includes that the present invention utilizes the microRNA highly expressed in liver tissue to degrade the self-amplifying mRNA, thereby reducing liver toxicity.
- the saRNA of the present invention has the advantages of known saRNA over traditional mRNA, that is, it can achieve the same protein expression level as traditional mRNA with a very low dose, and prolong the existence time of antigen protein in the body, thereby possibly enhancing the immune response.
- a lower effective dose may reduce the production cost of saRNA.
- the dose and number of injections used in mRNA therapy can be reduced, thereby prolonging the efficacy while reducing the possible toxic side effects of mRNA and delivery vectors.
- the saRNA of the present invention is also improved to have one or more of the following characteristics:
- the saRNA disclosed herein contains a mutated macrodomain, which weakens the ADP ribose hydrolysis activity, thereby causing The ability of the virus to replicate is weakened and the ability to stimulate the innate immune system is reduced. Therefore, the saRNA of the present disclosure may have a reduced cytotoxic effect on host cells or subjects, providing enhanced safety.
- the saRNA of the present disclosure can produce a high expression level of the encoded gene product while reducing the risk of undesirable effects (such as injection site irritation and/or pain).
- the ability of the saRNA of the present disclosure to stimulate the innate immune system is reduced, it is particularly suitable for vaccine preparation to provide appropriate immunity to the host.
- the saRNA of the present invention does not contain nucleotide sequences encoding viral structural proteins and therefore cannot lead to the production of alphavirus particles containing RNA.
- the inability to produce these viral particles means that the saRNA cannot self-perpetuate in an infectious form.
- the alphavirus structural proteins required for perpetuation in wild-type viruses are not present in the saRNA, and their positions are replaced by GOI, so that the saRNA encodes the required gene products, rather than the structural proteins of the alphavirus viral particles.
- the saRNA containing a mutant macrodomain of the present invention leads to a significant reduction in the dsRNA formed during replication.
- pattern recognition receptors There are sensors in human cells that recognize foreign viral invasions, which are called pattern recognition receptors.
- One of the signals they recognize is double-stranded RNA that appears in the cytoplasm, because this may represent the replication of viral RNA in the cell.
- saRNA forms double-stranded RNA, which is very similar to the viral RNA in replication, and therefore may stimulate the innate immune response of the cell. This may further enhance the effect of the vaccine.
- this stimulated immune response may promote the immune response as a vaccine, but on the other hand, as a therapy, the stimulated immune response may cause side effects.
- the double-stranded RNA produced during saRNA replication is the key to activating the interferon or NFKB signaling pathway, thereby inducing a large number of downstream antiviral genes to inhibit saRNA replication. Therefore, the immunogenicity of saRNA requires precise design and adjustment, so as to moderately reduce the replication ability of saRNA and the production of double-stranded RNA, and the sgRNA disclosed in the present invention achieves this purpose.
- the translation inhibition and induced apoptosis of the saRNA containing the mutant macro domain of the present invention are significantly reduced.
- mRNA usually triggers intrinsic receptors in cells, such as toll-like receptors (TLR), which lead to the production of type I interferon and inhibition of translation.
- TLR toll-like receptors
- the translation inhibition of the saRNA of the present invention is reduced due to reduced immunogenicity, so although its replication ability is reduced, the final expression efficiency in the cell is improved compared to the control saRNA encoding the wild-type macro domain.
- Sequence 1 BspQI endonuclease recognition site
- Sequence 2 saRNA-062 sequence
- Sequence 41 Amino acid sequence of alphavirus RNA replicase (SEQ ID NO: 41)
- Sequence 55 Wild-type macrodomain amino acid sequence from VEEV (SEQ ID NO: 55)
- Sequence 61 saRNA-190 backbone sequence
- NNNNNNNNNNNNNNNNNNNNNNN indicates exogenous genes, including but not limited to: vaccine antigen genes, tumor killing genes, therapeutic protein genes, antibody genes; exogenous genes can be adjusted according to different purposes;
- N can be any base among A, T, C, and G, and the number of bases is not particularly limited; the 16 Ns here are only for illustration and are not limited to 16 bases.
- Sequence 63 5' non-translated sequence (SEQ ID NO: 63)
- Sequence 65 Subgenomic promoter sequence (SEQ ID NO: 65)
- Sequence 66 Polyadenylation sequence (SEQ ID NO: 66)
- Sequence 67 saRNA-190 replicase coding sequence (SEQ ID NO: 67)
- Sequence 68 saRNA-190 macrodomain encoding sequence (SEQ ID NO: 68)
- Sequence 69 saRNA-191 replicase coding sequence (SEQ ID NO: 69)
- Sequence 70 saRNA-191 macrodomain encoding sequence (SEQ ID NO: 70)
- Sequence 71 Nonstructural protein 1 amino acid sequence (SEQ ID NO: 71)
- Sequence 72 Nonstructural protein 2 amino acid sequence (SEQ ID NO: 72)
- Sequence 73 Nonstructural protein 3 amino acid sequence (corresponding to saRNA-190, with Q48P mutation introduced) (SEQ ID NO: 73)
- Sequence 74 Nonstructural protein 3 amino acid sequence (corresponding to saRNA-191, with I133F mutation introduced) (SEQ ID NO: 74)
- Sequence 75 Nonstructural protein 4 amino acid sequence (SEQ ID NO: 75)
- Huh7.5.1 cells were purchased from Mingzhou bio, catalog number MZ-2129.
- C2C12 cells were purchased from Mingzhou bio, catalog number MZ-0034.
- Lipofectamine transfection reagents -mRNA Transfection Kit was purchased from Mirus, catalog number MIR 2225.
- Example 1 Modification and completion of positions 4497-4503 in the saRNA-EGFP template plasmid psaRNA-062 sexual testing
- Optimize position Modify the 4497-4503 position (5’-ACTCTTC-3’) in the saRNA template plasmid psaRNA-062 (such as SEQ ID NO: 2) (with the A base in the ATG start codon sequence of the alphavirus replicase as the first position, the same below).
- the T base at the 4500th bp was mutated into the C base or the A base, or the TCA base at the 4501-4503 bp was mutated into the AGC base.
- the three template plasmids obtained were named psaRNA-152 (SEQ ID NO: 3), psaRNA-153 (SEQ ID NO: 4) and psaRNA-154 (SEQ ID NO: 5).
- in vitro transcription template plasmid Specifically, add 8 ⁇ g of in vitro transcription template plasmid (psaRNA-062, psaRNA-152, psaRNA-153 and psaRNA-154), BspQ I 4 ⁇ L, 10 ⁇ Digestion Buffer 4 ⁇ L in sequence, and finally add DEPC water to the system to 40 ⁇ L, and incubate at 50°C for 2 h to linearize the plasmid.
- in vitro transcription template plasmid psaRNA-062, psaRNA-152, psaRNA-153 and psaRNA-154
- BspQ I 4 ⁇ L BspQ I 4 ⁇ L
- 10 ⁇ Digestion Buffer 4 ⁇ L 10 ⁇ Digestion Buffer 4 ⁇ L
- In vitro transcription to obtain modified saRNA Specifically, add 10 ⁇ T7 polymerase buffer, 7.5mM ATP, 7.5mM GTP, 7.5mM UTP, 7.5mM CTP, 1 ⁇ g linearized in vitro transcription template, 400U T7 RNA polymerase, 20U RNase I inhibitor, 6mM cap analog, add water to a total volume of 20 ⁇ L, and react at 37°C for 2 hours. After the reaction, add 2U DNase I enzyme and react at 37°C for 15 minutes.
- the reaction system is replenished to 50 ⁇ L, 25 ⁇ L 7.5M lithium chloride solution is added and mixed, and after standing at -20°C for 30 minutes, centrifuged at 13000g 4°C for 10 minutes, the supernatant is discarded and the precipitate is washed with 70% ethanol, centrifuged at 13000g 4°C for 2 minutes, the supernatant is discarded, and dissolved in 30 ⁇ L water.
- an appropriate volume of RNA was taken and the RNA integrity was tested using the Qsep400 capillary electrophoresis device. The test results are shown in Figure 1. The results show that only the integrity of the optimized saRNA-152 was improved from 76.7% to 81.3%.
- Example 2 Integrity test of different saRNAs constructed with the optimized psaRNA-152 template plasmid as the backbone Test
- the A base at position 4509 was mutated to T, C, and G, respectively, and the template plasmids thus obtained were named psaRNA-183 (SEQ ID NO: 6), psaRNA-184 (SEQ ID NO: 7), and psaRNA-185 (SEQ ID NO: 8).
- the specific site-directed mutation and detection methods were the same as those in Example 1. From the capillary electrophoresis detection results in Figure 2, it can be seen that the integrity of different saRNAs constructed with psaRNA-152 as the backbone is significantly improved compared to the original backbone psaRNA-062.
- Example 3 Modification and integrity detection of positions 1680-1676 in the psaRNA-183 template plasmid
- the 1680-1676 positions (5'-GCTCTTA-3') in the further optimized saRNA template plasmid psaRNA-183 were modified, specifically, the TCT bases at positions 1672-1674 were mutated to AGC bases, and the obtained template plasmid was named psaRNA-243 (SEQ ID NO: 9).
- the specific site-directed mutation and detection method are the same as in Example 1.
- the capillary electrophoresis detection results after optimization are shown in Figure 3. The results show that the proportion of short transcription products in the optimized saRNA is significantly reduced, and the integrity of saRNA-243 is significantly improved.
- Example 4 Verification of the replication and expression efficiency of luciferase gene saRNA after skeleton optimization
- mRNA transfection of Huh7.5.1 cells 0.5 ⁇ g of saRNA-152, saRNA-243 or unoptimized saRNA-062 expressing luciferase gene after backbone optimization was transfected into Huh7.5.1 cells using liposome transfection reagent, and cultured overnight at 5% CO 2 and 37° C. for 72 hours.
- Luciferase detection 24 and 72 hours after transfection, use Nano-Glo Luciferase Assay System detection reagent, transfer 25 ⁇ l Nano-Glo Luciferase Assay Reagent to a 96-well white opaque bottom plate, take 25 ⁇ l supernatant sample to the well and mix evenly, wait for 3 minutes and use a microplate reader to detect the signal.
- the test results are shown in Figures 4 and 5. There is no significant difference in the luciferase expression levels of saRNA-152 and saRNA-243 after backbone optimization and at different time points compared with the unoptimized backbone, indicating that backbone optimization does not affect saRNA expression.
- Treatment of cell samples 2 hours, 24 hours and 72 hours after transfection, discard the supernatant, rinse the cells once with 500 ⁇ l PBS, discard the supernatant, add 500 ⁇ l cell lysis buffer and collect by pipetting into EP tubes, and store in a -80°C refrigerator.
- Reverse transcription fluorescence quantitative PCR was used to detect the replication of saRNA: total RNA was extracted from the cell sample using RNeasy Mini Kit, and RNA was quantified using a UV spectrophotometer. After reverse transcription was completed using HiScript III 1st Strand cDNA Synthesis Kit, fluorescence quantitative PCR experiments were performed using 2 ⁇ ChamQ Universal SYBR qPCR Master Mix.
- the detection primers are as follows, alphavirus replicase sequence (nsp1) upstream primer F: 5‘-GACGGACCGACAAGTCTCTA-3’ (SEQ ID NO:22), alphavirus replicase sequence (nsp1) downstream primer R: 5‘-GGTGGTGTCAAAGCCTATCCA-3’ (SEQ ID NO:23), EGFP sequence upstream primer F: 5‘-AGCTGGAGTACAACTACA-3’ (SEQ ID NO:24), EGFP downstream primer R: 5‘-CTGATCTTGAAGTTCACC-3’ (SEQ ID NO:25), GAPDH sequence upstream primer F: 5‘-GGTATCGTGGAAGGACTC-3’ (SEQ ID NO:26), GAPDH downstream primer R: 5‘-GTAGAGGCAGGGATGATG-3’ (SEQ ID NO:27).
- the reaction cycle conditions were: 95°C 30s-(95°C 10s-60°C 30s) ⁇ 40 cycles, and the test results are shown in Figure 6.
- the results show that there is no significant difference between the saRNA-152 after skeleton optimization and the unoptimized skeleton at different time points, indicating that skeleton optimization also does not affect the replication ability of saRNA.
- Example 5 Preparation of optimized backbone saRNA expressing human papillomavirus antigen gene Ag9.1
- saRNA-243 as a template plasmid, the luciferase coding sequence in it was replaced with the human papillomavirus (HPV) antigen gene Ag9.1 coding sequence through homologous recombination to obtain a self-amplified mRNA in vitro transcription template plasmid expressing Ag9.1, named L23H02.
- HPV human papillomavirus
- In vitro transcription to obtain self-amplified mRNA Specifically, add 10 ⁇ T7 polymerase buffer, 7.5mM ATP, 7.5mM GTP, 7.5mM UTP, 7.5mM CTP, 1 ⁇ g linearized in vitro transcription template, 400U T7RNA polymerase, 20U RNase I inhibitor, 6mM cap analog, add water to a total volume of 20 ⁇ L, and react at 37°C for 2 hours. After the reaction, add 2U DNase I enzyme and react at 37°C for 15 minutes.
- Example 8 Detection of microRNA-122 expression in different cells
- RNA-easy Isolation Reagent After the total RNA was extracted from the cell samples using RNA-easy Isolation Reagent, the RNA was quantified using a UV spectrophotometer. After reverse transcription was completed using the miRNA 1st Strand cDNA Synthesis Kit (by stem-loop), the fluorescence quantitative PCR experiment was performed using miRNA Universal SYBR qPCR Master Mix.
- the detection primers are as follows:
- miRNA122 upstream primer F 5'-CGCGTGGAGTGTGACAATGG-3' (SEQ ID NO: 43),
- miRNA122 downstream primer R 5'-AGTGCAGGGTCCGAGGTATT-3' (SEQ ID NO: 44),
- U6 promoter upstream primer F 5'-CTCGCTTCGGCAGCACAT-3' (SEQ ID NO: 45),
- Example 9 Self-amplification of liver-specific microRNA-122 binding sequences inserted at six different locations In vitro mRNA expression and replication assays
- Huh7.5.1 cells with high expression of microRNA-122 and C2C12 cells with low expression of microRNA-122 were inoculated in 75cm2 cell culture flasks.
- the culture medium was DMEM high glucose medium + 10% fetal bovine serum + 1% double antibody.
- the cells were digested and counted with trypsin.
- An appropriate number of cells were plated in a 24-well cell culture plate and cultured overnight at 37°C in a CO2 incubator.
- Self-amplified mRNA transfection of Huh7.5.1 cells and C2C12 cells 0.5 ⁇ g of six self-amplified mRNAs and a control self-amplified mRNA without microRNA-122 binding sites (SEQ ID NO: 29) were mixed with 50 ng of linear mRNA expressing firefly luciferase and then transfected using liposomes.
- -mRNA Transfection Kit was used to transfect Huh7.5.1 cells and C2C12 cells, and cultured in a CO2 incubator at 37°C 24 hours.
- N21095 self-amplified mRNA showed similar expression to the control self-amplified mRNA, inferring that placing the microRNA-122 binding site at the corresponding site did not affect the normal expression of the self-amplified mRNA, but was not effectively regulated by microRNA.
- Luciferase detection 24 hours after transfection, use Nano-Glo Luciferase Assay System detection reagent, transfer 25 ⁇ l Nano-Glo Luciferase Assay Reagent to a 96-well white opaque bottom plate, take 25 ⁇ l supernatant sample to the well and mix evenly, wait for 3 minutes and use an ELISA reader to detect the signal.
- the detection results are shown in Figure 18, which also show that the expression levels of N21092 and N21096 self-amplified mRNAs in Huh7.5.1 cells with high expression of microRNA-122 and C2C12 cells with low expression of microRNA-122 are significantly different, indicating their good cell expression specificity.
- RNA is quantified using a UV spectrophotometer. After reverse transcription is completed using HiScript III 1st Strand cDNA Synthesis Kit, a fluorescence quantitative PCR experiment is performed using 2 ⁇ ChamQ Universal SYBR qPCR Master Mix.
- the detection primers are as follows:
- Primer F upstream of alphavirus replicase sequence (nsp1): 5'-GACGGACCGACAAGTCTCTA-3' (SEQ ID NO: 47),
- EGFP sequence upstream primer F 5'-AGCTGGAGTACAACTACA-3' (SEQ ID NO: 49),
- EGFP downstream primer R 5'-CTGATCTTGAAGTTCACC-3' (SEQ ID NO: 50),
- GAPDH sequence upstream primer F 5'-GGTATCGTGGAAGGACTC-3' (SEQ ID NO: 51),
- reaction cycle conditions were: 95°C 30s-(95°C 10s-60°C 30s) ⁇ 40 cycles.
- the fluorescent protein and luciferase detection method is the same as in Example 8.
- the relative expression results of the self-amplified mRNA to be tested and the control self-amplified mRNA without the microRNA-122 binding sequence in different cells are shown in FIG. 22 .
- the relative expression level of self-amplifying mRNA bound to microRNA-122 was inversely proportional to the number of copies of the inserted microRNA-122 binding sequence.
- the expression level of self-amplifying RNA with 1, 2, 3 or 6 copies of the microRNA-122 binding sequence inserted gradually decreased, among which the expression level of self-amplifying RNA with 6 copies of the microRNA-122 binding sequence inserted decreased the most, which was reduced by more than 90% compared with the wild type.
- the relative expression level of self-amplifying mRNA with 6 microRNA-122 binding sequences placed in the 3'UTR in C2C12 cells with low expression of microRNA-122 was still more than 70%.
- the regulation of self-amplified mRNA expression is based on microRNA-122 in a dose-dependent manner, and the 3’UTR is the optimal microRNA binding sequence placement site.
- the present invention selected saRNA derived from Venezuelan Equine Encephalitis Virus (VEEV) for subsequent design.
- VEEV Venezuelan Equine Encephalitis Virus
- the macrodomain is present at the N-terminus of nonstructural protein 3, as shown in Figure 24.
- the amino acid sequence of the macrodomain is relatively conservative between different viruses, with a homology of more than 50%. Therefore, we selected the 113th amino acid close to the ADP ribose hydrolysis active center and the 48th amino acid away from the ADP ribose hydrolysis active center for amino acid mutation, and the specific positions are shown in Figure 25.
- the 113th position encodes isoleucine (I) or valine (V), which is highly conservative.
- saRNA-190, saRNA-191 and wild-type saRNA were transfected into Hela cells. After 24 hours, the level of ADP-ribosylation of total protein in the cells was detected with an anti-mono-ADP-ribosylation antibody. As shown in Figure 26, 24 hours after transfection, the ADP-ribosylation of 190/191-saRNA cell proteins was significantly stronger than that of the wild-type transfection group, indicating that the ADP ribosyl hydrolase activity of saRNA-190 and saRNA-191 was significantly reduced relative to that of wild-type saRNA. The data show that the design of amino acid mutations at positions 113 and 48 of the macrodomain successfully reduced the ADP ribosyl hydrolase activity.
- Example 12 Detection of replication levels of mutant saRNAs with impaired ADP ribose hydrolase activity
- 190/191-saRNA significantly reduced the level of RNA encoding EGFP compared to wild-type saRNA.
- saRNA-190 and saRNA-191 compared with wild-type saRNA, saRNA-190 and saRNA-191 had a lower proportion of double-stranded RNA formed during replication 6 hours after transfection, and this difference was more significant 24 hours after transfection.
- Example 13 Detection of differences in natural immunity induced by mutant saRNA
- RNA sequencing After transfecting Hela cells with 190/191-saRNA and wild-type saRNA for 24 hours, we extracted total cell RNA for RNA sequencing and analyzed the differences in transcriptome levels.
- the transcriptome difference of the transfected 190-saRNA was the smallest, while the wild-type saRNA group induced very significant transcriptome changes (see Figure 29). Further, we counted the number of genes that were differentially expressed in different saRNA groups relative to the control group. We found that in all saRNA groups, the number of upregulated genes was far greater than the downregulated genes, and most of the upregulated genes in the 190 and 191 groups were almost the same as the wild-type saRNA group.
- the mRNA levels of interferon pathway and cytokines were verified by fluorescence quantitative PCR 2, 6 and 24 hours after transfection. It was found that within 2 hours, the RNA transcription levels of each group did not change compared with the control group, while at 6 hours, the IFN-beta, CXCL10, CCL5 and IFIT2 RNA levels in the wild-type saRNA group were significantly upregulated compared with the control group, and CXCL10, IFIT2 and PKR were also upregulated in the 190 and 191 groups, but there was no difference between 190 and 191. 24 hours after transfection, all RNAs including NF-KB and interferon pathways were most significantly upregulated in the wild-type saRNA group, followed by 191-saRNA, and the lowest in 190-saRNA.
- Example 14 Detection of protein translation activity of mutant saRNA in in vitro cell models
- ribosomal RNA of cells transfected with wild-type saRNA was still relatively intact within 2 and 6 hours, but severely degraded at 24 and 48 hours, while the integrity of ribosomal RNA transfected with 190/191saRNA was not significantly reduced relative to the control group.
- the phosphorylation level of eIF2 ⁇ was significantly reduced, and the phosphorylation of eIF2 ⁇ induced by wild-type saRNA was even weaker than that of the control group (i.e., the mock group: only the transfection reagent was added, and no RNA molecules were transfected), while the phosphorylation degree of eIF2 ⁇ of the mutant saRNA was similar to that of the control group (see Figure 37).
- Example 15 Apoptosis induction detection of mutant saRNA in in vitro cell models
- Example 16 Detection of in vivo expression level of 190-saRNA
- 190-saRNA, wild-type saRNA and non-replicating mRNA (nrmRNA) expressing the firefly luciferase gene were encapsulated with nanolipid particles (LNP).
- LNP nanolipid particles
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- Zoology (AREA)
- Biomedical Technology (AREA)
- Biotechnology (AREA)
- General Health & Medical Sciences (AREA)
- Microbiology (AREA)
- General Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Biochemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Molecular Biology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Immunology (AREA)
- Crystallography & Structural Chemistry (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Cell Biology (AREA)
- Mycology (AREA)
- Epidemiology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Physics & Mathematics (AREA)
- Biophysics (AREA)
- Plant Pathology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Description
相关申请的交叉引用CROSS-REFERENCE TO RELATED APPLICATIONS
本发明要求于2023年7月19日提交的申请号为202310890565.0、于2023年10月10日提交的申请号为202311309636.X以及于2024年3月8日提交的申请号为202410268968.6的中国专利申请的优先权,其全部内容通过引用并入本文。The present invention claims priority to Chinese patent applications with application numbers 202310890565.0 filed on July 19, 2023, 202311309636.X filed on October 10, 2023, and 202410268968.6 filed on March 8, 2024, the entire contents of which are incorporated herein by reference.
本发明属于生物医药领域,涉及甲病毒RNA复制酶编码序列、经优化的自扩增核酸分子、药物组合物及用途,所述经优化的自扩增核酸分子为包含该甲病毒RNA复制酶编码序列的自扩增核酸分子、组织特异性自扩增核酸分子或具有增强的外源基因表达水平的自扩增RNA核酸分子。The present invention belongs to the field of biomedicine and relates to an alphavirus RNA replicase coding sequence, an optimized self-amplifying nucleic acid molecule, a pharmaceutical composition and uses. The optimized self-amplifying nucleic acid molecule is a self-amplifying nucleic acid molecule comprising the alphavirus RNA replicase coding sequence, a tissue-specific self-amplifying nucleic acid molecule or a self-amplifying RNA nucleic acid molecule with enhanced exogenous gene expression level.
自扩增RNA(saRNA)是一种能够在体内进行自我扩增的RNA,用于疫苗可以从而产生更多的抗原,并且由于复制带来的免疫刺激作用,可以激发更强的免疫反应。Self-amplifying RNA (saRNA) is a type of RNA that can self-amplify in vivo. It can be used in vaccines to produce more antigens and stimulate a stronger immune response due to the immunostimulatory effect brought about by replication.
通常,mRNA的体外合成主要是以线性化质粒DNA或PCR扩增产物为模板,利用RNA聚合酶进行体外转录(In vitro transcription,IVT)得到。由于自扩增RNA长度较长,为保证序列准确性,转录模板通常选择线性化质粒DNA,因此需要利用特定DNA内切酶(例如BspQI或XbaI)对质粒进行线性化。但是,可能由于或者部分由于DNA内切酶的非特异消化,产生非预期的线性化产物,导致非目标体外转录产物,最终降低自扩增RNA产品完整性以及表达效率。因此,对自扩增RNA的优化方向之一是进一步提高其转录质量。Generally, the in vitro synthesis of mRNA is mainly obtained by in vitro transcription (IVT) using linearized plasmid DNA or PCR amplification products as templates using RNA polymerase. Due to the long length of self-amplified RNA, linearized plasmid DNA is usually selected as the transcription template to ensure sequence accuracy. Therefore, the plasmid needs to be linearized using a specific DNA endonuclease (such as BspQI or XbaI). However, due to or in part due to the non-specific digestion of the DNA endonuclease, unexpected linearization products are produced, resulting in non-target in vitro transcription products, which ultimately reduce the integrity and expression efficiency of the self-amplified RNA product. Therefore, one of the optimization directions for self-amplified RNA is to further improve its transcription quality.
mRNA疫苗是一种新型的疫苗技术,其原理是利用合成的mRNA分子来编码目标非内源性抗原蛋白,通过注射到人体中来激发针对病原体或肿瘤等靶点的免疫反应。目前,mRNA疫苗已被成功应用于新冠病毒的预防,并在传染病预防疫苗、肿瘤治疗以及蛋白替代疗法等研究领域具有广泛的应用前景。mRNA vaccine is a new vaccine technology, which uses synthetic mRNA molecules to encode target non-endogenous antigenic proteins and stimulates immune responses against targets such as pathogens or tumors by injecting them into the human body. Currently, mRNA vaccines have been successfully used to prevent the new coronavirus and have broad application prospects in research fields such as infectious disease prevention vaccines, tumor treatment, and protein replacement therapy.
常见的mRNA形式包括线性mRNA和环状mRNA,其中线性mRNA分为非复制型和具有自行复制能力的自扩增mRNA。自扩增RNA被视作下一代mRNA分子, 由于其长表达周期及自佐剂效应等特点,在传染病及肿瘤疫苗中都显示了其有效诱导特异性免疫的优势,此外,在期望长半衰期的蛋白替代疗法中,自扩增RNA也具有独特潜力。2023年日本批准了基于自扩增RNA的新冠疫苗,进一步证明了自扩增RNA作为疫苗应用的安全性。相比于非复制mRNA,自扩增RNA的一个重要优势是外源基因表达水平高,持续时间长,可以显著降低施用剂量,减少副作用。因此,进一步提升表达效率一直是自扩增RNA序列优化的方向之一。Common mRNA forms include linear mRNA and circular mRNA, among which linear mRNA is divided into non-replicating type and self-amplifying mRNA with self-replication ability. Self-amplifying RNA is regarded as the next generation of mRNA molecules. Due to its long expression cycle and self-adjuvant effect, it has shown its advantages in effectively inducing specific immunity in infectious diseases and tumor vaccines. In addition, self-amplifying RNA also has unique potential in protein replacement therapies that expect a long half-life. In 2023, Japan approved a new crown vaccine based on self-amplifying RNA, further proving the safety of self-amplifying RNA as a vaccine. Compared with non-replicating mRNA, an important advantage of self-amplifying RNA is that the expression level of exogenous genes is high and the duration is long, which can significantly reduce the administration dose and reduce side effects. Therefore, further improving the expression efficiency has always been one of the directions of self-amplifying RNA sequence optimization.
对于非复制mRNA的优化,利用核苷酸修饰来规避模式识别受体对mRNA的识别已被证明是一种极为成功的策略,旨在通过降低天然免疫反应来提高翻译效率。然而,由于自扩增RNA本质上是模拟病毒复制表达机制,因此相对于非复制mRNA更为复杂。除了存在未修饰的核苷酸外,自扩增RNA复制产物包括双链RNA、带有Cap0结构的子代RNA和未加帽的5'-三磷酸子代RNA都能够被模式识别受体(pattern recognition receptor,PRR)识别并启动包括但不限于干扰素或NF-KB信号通路在内的抗病毒免疫反应,由此诱导的干扰素刺激因子如蛋白激酶R(protein kinase R,PKR)、2-5-寡腺苷酸合成酶(oligoadenylate synthetase,OAS)和具有四三肽重复的干扰素诱导蛋白(interferon induced proteins with tetratricopeptide repeats,IFIT)会执行宿主翻译抑制和诱导细胞凋亡等功能来限制自扩增RNA复制及表达。所以,通过降低自扩增RNA本身的免疫原性对于提升自扩增RNA表达效率有正向作用,因此是优化自扩增RNA表达效率的主要思路。For the optimization of non-replicating mRNA, the use of nucleotide modification to circumvent the recognition of mRNA by pattern recognition receptors has been proven to be a very successful strategy aimed at improving translation efficiency by reducing the innate immune response. However, since self-amplifying RNA essentially mimics the replication and expression mechanism of viruses, it is more complicated than non-replicating mRNA. In addition to the presence of unmodified nucleotides, self-amplifying RNA replication products including double-stranded RNA, daughter RNA with Cap0 structure and uncapped 5'-triphosphate daughter RNA can be recognized by pattern recognition receptors (PRR) and initiate antiviral immune responses including but not limited to interferon or NF-KB signaling pathways. The interferon-stimulated factors induced by this, such as protein kinase R (PKR), 2-5-oligoadenylate synthetase (OAS) and interferon induced proteins with tetratricopeptide repeats (IFIT), will perform host translation inhibition and induce cell apoptosis to limit the replication and expression of self-amplifying RNA. Therefore, reducing the immunogenicity of the self-amplifying RNA itself has a positive effect on improving the expression efficiency of the self-amplifying RNA, and is therefore the main idea for optimizing the expression efficiency of the self-amplifying RNA.
自扩增mRNA的序列结构较mRNA相对复杂,尤其是存在病毒复制酶及复制相关元件如RNA启动子等。自扩增mRNA利用自身包含的来源于甲病毒的RNA复制酶,使mRNA能够自我复制。因此,注射的mRNA可以在体内持续产生靶蛋白,从而增强疫苗免疫反应的持久性和强度,或者增强蛋白替代疗法的半衰期。The sequence structure of self-amplifying mRNA is relatively complex compared to mRNA, especially the presence of viral replicase and replication-related elements such as RNA promoters. Self-amplifying mRNA uses the RNA replicase from alphavirus contained in it to enable mRNA to replicate itself. Therefore, the injected mRNA can continuously produce the target protein in the body, thereby enhancing the durability and intensity of the vaccine immune response, or enhancing the half-life of protein replacement therapy.
目前的自扩增mRNA为甲病毒基因组改造而来的mRNA载体,本身无特定microRNA结合位点,其在不同的组织中的复制和表达并不会受到microRNA介导的调控。The current self-amplifying mRNA is an mRNA vector modified from the alphavirus genome. It does not have a specific microRNA binding site, and its replication and expression in different tissues are not subject to microRNA-mediated regulation.
此外,自扩增mRNA经由目前常用于核酸分子递送的纳米脂质颗粒(lipid nanoparticles,LNP)包封并递送到体内后,由于纳米脂质颗粒固有的理化性质,有可能介导自扩增mRNA进入肝脏等实质组织后大量表达自扩增mRNA编码的靶基因,引起潜在的肝脏毒性。 In addition, after the self-amplifying mRNA is encapsulated and delivered into the body via lipid nanoparticles (LNPs), which are currently commonly used for nucleic acid molecule delivery, the inherent physical and chemical properties of the lipid nanoparticles may mediate the self-amplifying mRNA to enter the liver and other parenchymal tissues, where it may express a large amount of the target gene encoded by the self-amplifying mRNA, causing potential liver toxicity.
因此,为了在各个相关领域更好地应用自扩增mRNA,尚需要开发新的转录质量提高的自扩增mRNA,新的可调控的和/或低毒性的自扩增mRNA,以及新的具有增强的外源基因表达水平的自扩增mRNA。Therefore, in order to better apply self-amplifying mRNA in various related fields, it is still necessary to develop new self-amplifying mRNA with improved transcription quality, new regulatable and/or low-toxicity self-amplifying mRNA, and new self-amplifying mRNA with enhanced exogenous gene expression level.
发明内容Summary of the invention
优化的甲病毒RNA复制酶编码序列(翻译区)以及自扩增mRNA骨架序列Optimized alphavirus RNA replicase coding sequence (translation region) and self-amplifying mRNA backbone sequence
本发明得到了一种优化的甲病毒RNA复制酶编码序列(翻译区),并进一步得到了一种优化的自扩增mRNA骨架序列,与原始序列相比,能够显著降低DNA内切酶BspQI的非特异酶切产物的产生,提高了自扩增mRNA体外转录的完整性,同时保持saRNA正常的复制功能和/或表达功能。由此提供了下述发明:The present invention obtains an optimized alphavirus RNA replicase coding sequence (translation region), and further obtains an optimized self-amplifying mRNA backbone sequence, which can significantly reduce the production of non-specific enzyme cleavage products of DNA endonuclease BspQI compared with the original sequence, improve the integrity of self-amplifying mRNA in vitro transcription, and maintain the normal replication function and/or expression function of saRNA. The following invention is provided:
本发明的一个方面涉及一种mRNA分子翻译区(编码区),其编码SEQ ID NO:20所示的甲病毒RNA复制酶,其中,所述mRNA分子的第4500位碱基为C。One aspect of the present invention relates to a translation region (coding region) of an mRNA molecule, which encodes the alpha virus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C.
在本发明的一些实施方式中,所述的mRNA分子翻译区是一种分离的mRNA分子翻译区。In some embodiments of the present invention, the mRNA molecule translation region is an isolated mRNA molecule translation region.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其中,第4509位碱基为T、C或G。In some embodiments of the present invention, in the translation region of the mRNA molecule, the 4509th base is T, C or G.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其中,第1672位碱基为A,第1673位碱基为G,并且第1674位碱基为C。In some embodiments of the present invention, in the translation region of the mRNA molecule, the 1672nd base is A, the 1673rd base is G, and the 1674th base is C.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其编码SEQ ID NO:20所示的甲病毒RNA复制酶,其中,所述mRNA分子的第4500位碱基为C,并且第4509位碱基为T。In some embodiments of the present invention, the translation region of the mRNA molecule encodes the alphavirus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C and the 4509th base is T.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其编码SEQ ID NO:20所示的甲病毒RNA复制酶,其中,所述mRNA分子的第4500位碱基为C,并且第4509位碱基为C。In some embodiments of the present invention, the translation region of the mRNA molecule encodes the alphavirus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C, and the 4509th base is C.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其编码SEQ ID NO:20所示的甲病毒RNA复制酶,其中,所述mRNA分子的第4500位碱基为C,并且第4509位碱基为G。In some embodiments of the present invention, the translation region of the mRNA molecule encodes the alphavirus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C and the 4509th base is G.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其编码SEQ ID NO:20所示的甲病毒RNA复制酶,其中,所述mRNA分子的第4500位碱基为C,第1672 位碱基为A,第1673位碱基为G,并且第1674位碱基为C。In some embodiments of the present invention, the translation region of the mRNA molecule encodes the alphavirus RNA replicase shown in SEQ ID NO: 20, wherein the 4500th base of the mRNA molecule is C, the 1672nd base is The 1673rd base is A, the 1674th base is G, and the 1674th base is C.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其编码SEQ ID NO:20所示的甲病毒RNA复制酶,其中,所述mRNA分子的第4500位碱基为C,第4509位碱基为T,第1672位碱基为A,第1673位碱基为G,并且第1674位碱基为C。In some embodiments of the present invention, the translation region of the mRNA molecule encodes the alpha virus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C, the 4509th base is T, the 1672nd base is A, the 1673rd base is G, and the 1674th base is C.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其编码SEQ ID NO:20所示的甲病毒RNA复制酶,其中,所述mRNA分子的第4500位碱基为C,第4509位碱基为C,第1672位碱基为A,第1673位碱基为G,并且第1674位碱基为C。In some embodiments of the present invention, the translation region of the mRNA molecule encodes the alpha virus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C, the 4509th base is C, the 1672nd base is A, the 1673rd base is G, and the 1674th base is C.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其编码SEQ ID NO:20所示的甲病毒RNA复制酶,其中,所述mRNA分子的第4500位碱基为C,第4509位碱基为G,第1672位碱基为A,第1673位碱基为G,并且第1674位碱基为C。In some embodiments of the present invention, the translation region of the mRNA molecule encodes the alpha virus RNA replicase shown in SEQ ID NO:20, wherein the 4500th base of the mRNA molecule is C, the 4509th base is G, the 1672nd base is A, the 1673rd base is G, and the 1674th base is C.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其中,编码甲病毒RNA复制酶的初始mRNA分子翻译区的序列如SEQ ID NO:10所示。In some embodiments of the present invention, the mRNA molecule translation region, wherein the sequence of the initial mRNA molecule translation region encoding the alphavirus RNA replicase is shown as SEQ ID NO:10.
在本发明的一些实施方式中,所述的mRNA分子翻译区,其序列如SEQ ID NO:11至SEQ ID NO:15中的任一序列所示。In some embodiments of the present invention, the translation region of the mRNA molecule has a sequence as shown in any one of SEQ ID NO:11 to SEQ ID NO:15.
本发明的另一方面涉及一种mRNA分子,依次包括:Another aspect of the present invention relates to an mRNA molecule comprising, in sequence:
5’端非翻译区、RNA复制酶编码区、RNA启动子、可选的编码目标蛋白的序列、3’端非翻译区和多聚腺苷酸序列;5' untranslated region, RNA replicase coding region, RNA promoter, optional sequence encoding target protein, 3' untranslated region and polyadenylation sequence;
其中,所述RNA复制酶编码区为本发明中任一项所述的mRNA分子翻译区。Wherein, the RNA replicase coding region is the mRNA molecule translation region described in any one of the present invention.
所述多聚腺苷酸也称为PolyA或PolyA尾巴;其中可以含有少量的非腺苷酸(A),例如少量的T、C和/或G。不拘于理论的限制,中间非A序列可增加DNA模板生产的稳定性。The polyadenylic acid is also called PolyA or PolyA tail, which may contain a small amount of non-adenylic acid (A), such as a small amount of T, C and/or G. Without being bound by theory, the intermediate non-A sequence can increase the stability of DNA template production.
在本发明的一些实施方式中,所述的mRNA分子,其为分离的mRNA分子。In some embodiments of the present invention, the mRNA molecule is an isolated mRNA molecule.
在本发明的一些实施方式中,所述的mRNA分子,其中,In some embodiments of the present invention, the mRNA molecule, wherein
其中,所述5’端非翻译区来源于甲病毒;优选地,来源于委内瑞拉马脑脊髓炎病毒;Wherein, the 5' untranslated region is derived from an alpha virus; preferably, it is derived from Venezuelan equine encephalomyelitis virus;
优选地,所述5’端非翻译区的序列如SEQ ID NO:16所示。Preferably, the sequence of the 5’ non-translated region is as shown in SEQ ID NO:16.
在本发明的一些实施方式中,所述的mRNA分子,其中,In some embodiments of the present invention, the mRNA molecule, wherein
其中,所述3’端非翻译区来源于甲病毒;优选地,来源于委内瑞拉马脑脊髓炎病毒; Wherein, the 3' untranslated region is derived from an alpha virus; preferably, derived from Venezuelan equine encephalomyelitis virus;
优选地,所述3’端非翻译区的序列如SEQ ID NO:17所示。Preferably, the sequence of the 3’ non-translated region is as shown in SEQ ID NO:17.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述RNA启动子所是亚基因组启动子;In some embodiments of the present invention, the mRNA molecule, wherein the RNA promoter is a subgenomic promoter;
优选地,所述RNA启动子是来源于甲病毒的亚基因组启动子;Preferably, the RNA promoter is a subgenomic promoter derived from an alphavirus;
优选地,所述RNA启动子是来源于委内瑞拉马脑脊髓炎病毒的亚基因组启动子;Preferably, the RNA promoter is a subgenomic promoter derived from Venezuelan equine encephalitis virus;
优选地,所述RNA启动子是26S启动子;Preferably, the RNA promoter is a 26S promoter;
优选地,所述RNA启动子的序列如SEQ ID NO:18所示。Preferably, the sequence of the RNA promoter is as shown in SEQ ID NO:18.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述多聚腺苷酸序列的序列如SEQ ID NO:19所示。In some embodiments of the present invention, the mRNA molecule, wherein the sequence of the polyadenylic acid sequence is as shown in SEQ ID NO:19.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述编码目标蛋白的序列为编码疫苗抗原、治疗性蛋白或靶向免疫检查点的抗体的序列。In some embodiments of the present invention, the mRNA molecule, wherein the sequence encoding the target protein is a sequence encoding a vaccine antigen, a therapeutic protein or an antibody targeting an immune checkpoint.
在本发明的一些实施方式中,所述的mRNA分子,其序列如SEQ ID NO:3以及SEQ ID NO:6至SEQ ID NO:9中的任一序列所示。In some embodiments of the present invention, the mRNA molecule has a sequence as shown in SEQ ID NO: 3 and any one of SEQ ID NO: 6 to SEQ ID NO: 9.
在本发明的一些实施方式中,所述的mRNA分子,其为自扩增RNA(saRNA)。In some embodiments of the invention, the mRNA molecule is a self-amplifying RNA (saRNA).
本发明的再一方面涉及一种DNA分子,其编码本发明中任一项所述的mRNA分子翻译区或者编码本发明中任一项所述的mRNA分子。Another aspect of the present invention relates to a DNA molecule encoding the translation region of the mRNA molecule described in any one of the present invention or encoding the mRNA molecule described in any one of the present invention.
本发明的再一方面涉及一种重组载体,其含有本发明的DNA分子;优选地,所述重组载体为重组的原核表达载体或重组的真核表达载体。Another aspect of the present invention relates to a recombinant vector, which contains the DNA molecule of the present invention; preferably, the recombinant vector is a recombinant prokaryotic expression vector or a recombinant eukaryotic expression vector.
本发明的再一方面涉及一种重组宿主细胞,其含有本发明中任一项所述的mRNA分子翻译区、本发明中任一项所述的mRNA分子、本发明的DNA分子或者本发明的重组载体。Another aspect of the present invention relates to a recombinant host cell, which contains the translation region of the mRNA molecule described in any one of the present invention, ... DNA molecule described in any one of the present invention, or the recombinant vector described in the present invention.
本发明的再一方面涉及一种试剂盒,其含有本发明中任一项所述的mRNA分子翻译区、本发明中任一项所述的mRNA分子或者本发明的DNA分子,以及脂质体类递送系统。Another aspect of the present invention relates to a kit, which contains the translation region of the mRNA molecule described in any one of the present invention, the mRNA molecule described in any one of the present invention or the DNA molecule described in the present invention, and a liposome delivery system.
本发明的再一方面涉及一种药物组合物,其含有本发明中任一项所述的mRNA分子翻译区、权本发明中任一项所述的mRNA分子或者本发明的DNA分子,以及一种或多种药学上可接受的辅料;优选地,所述辅料为脂质体类递送系统。Another aspect of the present invention relates to a pharmaceutical composition, which contains the translation region of the mRNA molecule described in any one of the present invention, the mRNA molecule described in any one of the present invention or the DNA molecule described in the present invention, and one or more pharmaceutically acceptable excipients; preferably, the excipient is a liposome delivery system.
本发明的再一方面涉及一种疫苗制剂,其含有本发明中任一项所述的mRNA分子翻译区、本发明中任一项所述的mRNA分子或者本发明的DNA分子; Another aspect of the present invention relates to a vaccine preparation, which contains the translation region of the mRNA molecule described in any one of the present invention, the mRNA molecule described in any one of the present invention, or the DNA molecule described in the present invention;
优选地,所述mRNA分子或者DNA分子被脂质体类递送系统包裹;Preferably, the mRNA molecule or DNA molecule is encapsulated by a liposome-based delivery system;
可选地,所述疫苗制剂还包含一种或多种疫苗用佐剂;Optionally, the vaccine formulation further comprises one or more vaccine adjuvants;
优选地,所述疫苗制剂为预防病毒感染例如新冠病毒感染或预防新冠病毒感染所致重症的疫苗制剂。Preferably, the vaccine preparation is a vaccine preparation for preventing viral infection such as novel coronavirus infection or preventing severe illness caused by novel coronavirus infection.
脂质类递送系统包括脂质体(liposome)、脂质纳米粒(lipid Nanoparticle,LNP)、脂质多聚物纳米载体(Lipopolyplex,LPP)。Lipid delivery systems include liposomes, lipid nanoparticles (LNP), and lipid polymer nanocarriers (LPP).
脂质类中的LPP是一种以聚合物包载mRNA为内核、磷脂包裹为外壳的双层结构。LPP的双层脂质体膜具有更好的包载、保护mRNA的效果,LPP的内核能够随聚合物的降解逐步释放mRNA分子。LPP靶向树突状细胞效果优异,能够更好的通过抗原递呈激活T细胞的免疫反应,达到理想的治疗效果。LPP among lipids is a double-layer structure with polymer-encapsulated mRNA as the core and phospholipids as the shell. The double-layer liposome membrane of LPP has a better effect of encapsulating and protecting mRNA, and the core of LPP can gradually release mRNA molecules as the polymer degrades. LPP has excellent targeting effect on dendritic cells and can better activate the immune response of T cells through antigen presentation to achieve ideal therapeutic effects.
不拘于理论的限制,mRNA能有效激起细胞免疫和体液免疫。注射的mRNA疫苗被抗原呈递细胞内吞。在逃离内质体进入胞浆后,mRNA被核糖体翻译成蛋白质。翻译后的抗原蛋白可以通过多种方式刺激免疫系统,激起人体的细胞免疫和体液免疫。与传统的灭活疫苗、亚单位疫苗和基因工程疫苗相比,核酸疫苗具有如下优点:研发周期短;生产工艺简单、扩产容易;无需佐剂、有效性高;不进入细胞核、安全性较好等。mRNA新冠疫苗验证了mRNA技术平台在疫苗领域的适用性。Without being bound by theory, mRNA can effectively stimulate cellular immunity and humoral immunity. The injected mRNA vaccine is internalized by antigen-presenting cells. After escaping the endosome and entering the cytoplasm, the mRNA is translated into protein by the ribosome. The translated antigen protein can stimulate the immune system in a variety of ways, stimulating the body's cellular immunity and humoral immunity. Compared with traditional inactivated vaccines, subunit vaccines and genetically engineered vaccines, nucleic acid vaccines have the following advantages: short R&D cycle; simple production process and easy expansion; no adjuvant is required and the effectiveness is high; it does not enter the cell nucleus and has good safety. The mRNA COVID-19 vaccine verifies the applicability of the mRNA technology platform in the vaccine field.
本发明的再一方面涉及本发明中任一项所述的mRNA分子翻译区、本发明中任一项所述的mRNA分子或者本发明的DNA分子在制备治疗或预防病毒感染的药物、治疗或预防肿瘤的药物、或者用于蛋白替代疗法的药物中的用途。Another aspect of the present invention relates to the use of the translation region of the mRNA molecule described in any one of the present invention, the mRNA molecule described in any one of the present invention, or the DNA molecule described in any one of the present invention in the preparation of a drug for treating or preventing viral infection, a drug for treating or preventing tumors, or a drug for protein replacement therapy.
被特定组织特异性表达的microRNA结合进而调控自扩增mRNA分子复制的自The microRNA specifically expressed by a specific tissue binds to and regulates the replication of self-amplifying mRNA molecules. 扩增mRNA骨架Amplification of mRNA backbone
本发明还得到了一种能够被特定组织特异性表达的microRNA结合进而调控自扩增mRNA分子复制的自扩增mRNA骨架,从而实现了自扩增mRNA分子在表达特定microRNA的组织中降解,而在低表达或不表达特定microRNA的组织中正常存在,以实现靶基因分子以自扩增mRNA的形式进行组织特异性表达。由此提供了下述发明:The present invention also obtains a self-amplifying mRNA skeleton that can be bound by microRNA specifically expressed in a specific tissue and then regulate the replication of self-amplifying mRNA molecules, thereby achieving the degradation of self-amplifying mRNA molecules in tissues expressing specific microRNAs, while existing normally in tissues that low-express or do not express specific microRNAs, so as to achieve tissue-specific expression of target gene molecules in the form of self-amplifying mRNA. The following invention is provided:
本发明的又一个方面涉及一种mRNA分子,依次包括:Another aspect of the present invention relates to an mRNA molecule, comprising:
5’端非翻译序列、RNA复制酶的序列、RNA启动子、可选的编码目标蛋白的序 列、3’端非翻译序列和3’端PolyA尾巴;5' non-translated sequence, RNA replicase sequence, RNA promoter, and optional sequence encoding target protein column, 3' untranslated sequence and 3' PolyA tail;
其中,in,
在所述RNA复制酶的nsp1和nsp2之间、nsp2和nsp3之间、nsp3和nsp4之间、和/或编码目标蛋白的序列和3’端非翻译序列之间含有microRNA结合位点序列。A microRNA binding site sequence is contained between nsp1 and nsp2, between nsp2 and nsp3, between nsp3 and nsp4, and/or between the sequence encoding the target protein and the 3'-end non-translated sequence of the RNA replicase.
在本发明的一些实施方式中,所述的mRNA分子,其中,In some embodiments of the present invention, the mRNA molecule, wherein
所述RNA复制酶是来源于甲病毒的RNA复制酶;优选地,是来源于委内瑞拉马脑脊髓炎病毒的RNA复制酶;The RNA replicase is an RNA replicase derived from an alphavirus; preferably, it is an RNA replicase derived from Venezuelan equine encephalomyelitis virus;
优选地,所述RNA复制酶的氨基酸序列如SEQ ID NO:41所示;优选地,所述RNA复制酶的编码序列如SEQ ID NO:42所示。Preferably, the amino acid sequence of the RNA replicase is as shown in SEQ ID NO:41; preferably, the coding sequence of the RNA replicase is as shown in SEQ ID NO:42.
在本发明的一些实施方式中,所述的mRNA分子,其中,In some embodiments of the present invention, the mRNA molecule, wherein
所述RNA启动子是亚基因组启动子;The RNA promoter is a subgenomic promoter;
优选地,所述RNA启动子是来源于甲病毒的亚基因组启动子;Preferably, the RNA promoter is a subgenomic promoter derived from an alphavirus;
优选地,所述RNA启动子是来源于委内瑞拉马脑脊髓炎病毒的亚基因组启动子;Preferably, the RNA promoter is a subgenomic promoter derived from Venezuelan equine encephalitis virus;
优选地,所述RNA启动子是26S启动子。Preferably, the RNA promoter is the 26S promoter.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述5’端非翻译序列和/或所述3’端非翻译序列来源于甲病毒;优化地,来源于委内瑞拉马脑脊髓炎病毒。In some embodiments of the present invention, the mRNA molecule, wherein the 5' non-translated sequence and/or the 3' non-translated sequence is derived from an alpha virus; optimally, it is derived from Venezuelan equine encephalitis virus.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述microRNA结合位点序列为组织特异性表达的microRNA对应的结合位点序列;In some embodiments of the present invention, the mRNA molecule, wherein the microRNA binding site sequence is a binding site sequence corresponding to a tissue-specifically expressed microRNA;
优选地,所述组织特异性表达的microRNA为在肿瘤组织中低表达的microRNA;Preferably, the tissue-specifically expressed microRNA is a microRNA that is lowly expressed in tumor tissue;
优选地,所述在肿瘤组织中低表达的microRNA选自miRNA-122、miRNA-143、miRNA-1、miRNA-124、miRNA-217和miRNA-126。Preferably, the microRNA lowly expressed in tumor tissue is selected from miRNA-122, miRNA-143, miRNA-1, miRNA-124, miRNA-217 and miRNA-126.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述microRNA结合位点序列包含一个或多个microRNA成熟序列;优选地,所述microRNA结合位点序列包含2、3、4、5、6、7、8、9、10、11或12个序列相同或不同的microRNA成熟序列。In some embodiments of the present invention, the mRNA molecule, wherein the microRNA binding site sequence comprises one or more microRNA mature sequences; preferably, the microRNA binding site sequence comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 microRNA mature sequences that are identical or different in sequence.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述microRNA成熟序列如SEQ ID NO:30至SEQ ID NO:32中的任一序列所示。In some embodiments of the present invention, the mRNA molecule, wherein the microRNA mature sequence is shown in any one of SEQ ID NO:30 to SEQ ID NO:32.
不拘于理论的限制,microRNA结合位点序列理论上是与microRNA互补配对,结合位点序列长度可微调但需要大于等于20nt,如MicroRNA-122-5p长度为22nt, 其结合序列长度则可设计为20-24nt,例如20nt、21nt、22nt、23nt或24nt。Without being bound by theory, the microRNA binding site sequence is theoretically complementary to the microRNA. The length of the binding site sequence can be fine-tuned but needs to be greater than or equal to 20 nt. For example, the length of MicroRNA-122-5p is 22 nt. The length of the binding sequence can be designed to be 20-24 nt, such as 20 nt, 21 nt, 22 nt, 23 nt or 24 nt.
不拘于理论的限制,miRNA结合位点通常位于靶mRNA的3’非翻译区域(3’UTR),也可以位于5’UTR或编码区域。microRNA通过与靶mRNA结合,从而抑制靶mRNA的翻译或降解靶mRNA,从而影响靶基因的表达。Without being limited by theory, miRNA binding sites are usually located in the 3' untranslated region (3'UTR) of the target mRNA, and can also be located in the 5'UTR or coding region. MicroRNA binds to the target mRNA, thereby inhibiting the translation of the target mRNA or degrading the target mRNA, thereby affecting the expression of the target gene.
本发明将组织特异性表达的microRNA对应的结合位点引入mRNA,利用microRNA介导的转录后调控机制,对不同mRNA分子的稳定性和翻译速率进行调节,从而实现mRNA在不同组织的差异化表达,进而实现mRNA的组织特异性表达。The present invention introduces the binding sites corresponding to the tissue-specifically expressed microRNA into mRNA, and utilizes the microRNA-mediated post-transcriptional regulatory mechanism to regulate the stability and translation rate of different mRNA molecules, thereby achieving the differential expression of mRNA in different tissues, and further achieving the tissue-specific expression of mRNA.
在本发明的一些实施方式中,所述的mRNA分子,其中,各microRNA成熟序列之间含有一个或多个相同或不同的内部间隔序列(inner spacer);In some embodiments of the present invention, the mRNA molecule contains one or more identical or different internal spacer sequences between each microRNA mature sequence;
优选地,所述内部间隔序列如SEQ ID NO:33至SEQ ID NO:34中的任一序列所示。Preferably, the internal spacer sequence is shown in any one of SEQ ID NO:33 to SEQ ID NO:34.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述microRNA结合位点序列的5’端和/或3’端含有一个或多个相同或不同的外部间隔序列(outer spacer);In some embodiments of the present invention, the mRNA molecule, wherein the 5' end and/or the 3' end of the microRNA binding site sequence contains one or more identical or different external spacer sequences;
优选地,5’端的外部间隔序列如SEQ ID NO:35至SEQ ID NO:36中的任一序列所示;Preferably, the external spacer sequence at the 5' end is shown in any one of SEQ ID NO:35 to SEQ ID NO:36;
优选地,3’端的外部间隔序列如SEQ ID NO:37至SEQ ID NO:38中的任一序列所示。Preferably, the external spacer sequence at the 3’ end is as shown in any one of SEQ ID NO:37 to SEQ ID NO:38.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述microRNA结合位点序列的5’端和/或3’端含有一个或多个甲病毒RNA复制酶或nsp2能够识别的酶切位点序列;In some embodiments of the present invention, the mRNA molecule, wherein the 5' end and/or 3' end of the microRNA binding site sequence contains one or more restriction site sequences that can be recognized by alphavirus RNA replicase or nsp2;
优选地,为nsp1和nsp2之间的能够被甲病毒复制酶或nsp2识别的酶切位点序列;Preferably, it is a restriction site sequence between nsp1 and nsp2 that can be recognized by the alphavirus replicase or nsp2;
优选地,所述酶切位点序列如SEQ ID NO:53所示。Preferably, the restriction site sequence is as shown in SEQ ID NO:53.
不拘于理论的限制,甲病毒复制酶中的nsp2是发挥蛋白酶活性的部分,其能够识别甲病毒复制酶四组分之间的特定氨基酸序列,从而将复制酶多聚蛋白切割为单组分发挥复制功能。因此,此处的酶切位点应该为甲病毒复制酶或nsp2能够识别的蛋白酶酶切位点序列。Without being bound by theory, nsp2 in the alphavirus replicase is a part that exerts protease activity, which can recognize a specific amino acid sequence between the four components of the alphavirus replicase, thereby cutting the replicase polyprotein into a single component to exert replication function. Therefore, the restriction site here should be a protease restriction site sequence that can be recognized by the alphavirus replicase or nsp2.
所述蛋白酶酶切位点序列优选为nsp1和nsp2之间的,可被甲病毒复制酶或nsp2识别的氨基酸序列。序列为:EAGA↓GSVE(SEQ ID NO:53),↓代表切割位点。The protease cleavage site sequence is preferably an amino acid sequence between nsp1 and nsp2 that can be recognized by the alphavirus replicase or nsp2. The sequence is: EAGA↓GSVE (SEQ ID NO: 53), where ↓ represents the cleavage site.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述microRNA结合位点 序列如SEQ ID NO:22所示。In some embodiments of the present invention, the mRNA molecule, wherein the microRNA binding site The sequence is shown in SEQ ID NO:22.
在本发明的一些实施方式中,所述的mRNA分子,其还含有5’端帽子结构。In some embodiments of the present invention, the mRNA molecule further contains a 5' end cap structure.
在本发明的一些实施方式中,所述的mRNA分子,其序列如SEQ ID NO:24或SEQ ID NO:28所示。In some embodiments of the present invention, the mRNA molecule has a sequence as shown in SEQ ID NO:24 or SEQ ID NO:28.
在本发明的一些实施方式中,所述的mRNA分子,其中,所述目标蛋白是抗原。In some embodiments of the present invention, the mRNA molecule, wherein the target protein is an antigen.
本发明的mRNA分子是组织特异性自扩增的核酸分子。The mRNA molecules of the present invention are tissue-specific self-amplifying nucleic acid molecules.
在本发明的一些实施方式中,所述的mRNA分子是分离的mRNA分子。In some embodiments of the invention, the mRNA molecule is an isolated mRNA molecule.
本发明的另一方面涉及一种mRNA分子组合,包含第一mRNA分子和第二mRNA分子,其中:Another aspect of the present invention relates to an mRNA molecule combination, comprising a first mRNA molecule and a second mRNA molecule, wherein:
第一mRNA分子,依次包括:The first mRNA molecule comprises, in order:
第一5’端非翻译序列、RNA复制酶的序列、第一3’端非翻译序列和3’端PolyA尾巴;The first 5' non-translated sequence, the RNA replicase sequence, the first 3' non-translated sequence and the 3' PolyA tail;
第二mRNA分子,依次包括:The second mRNA molecule comprises, in order:
第二5’端非翻译序列、甲病毒复制必需序列、RNA启动子、编码目标蛋白的序列、第二3’端非翻译序列和3’端PolyA尾巴;The second 5' non-translated sequence, the sequence essential for alphavirus replication, the RNA promoter, the sequence encoding the target protein, the second 3' non-translated sequence and the 3' PolyA tail;
其中,在所述RNA复制酶的nsp1和nsp2之间、nsp2和nsp3之间、nsp3和nsp4之间、和/或编码目标蛋白的序列和第二3’端非翻译序列之间含有microRNA结合位点序列。Wherein, a microRNA binding site sequence is contained between nsp1 and nsp2, between nsp2 and nsp3, between nsp3 and nsp4, and/or between the sequence encoding the target protein and the second 3'-end non-translated sequence of the RNA replicase.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,In some embodiments of the present invention, the mRNA molecule combination, wherein,
第一5’端非翻译序列与第二5’端非翻译序列相同或不同;和/或The first 5' non-translated sequence is the same as or different from the second 5' non-translated sequence; and/or
第一3’端非翻译序列与第二3’端非翻译序列相同或不同。The first 3' non-translated sequence is the same as or different from the second 3' non-translated sequence.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,所述第一5’端非翻译序列和第一3’端非翻译序列不来源于甲病毒。In some embodiments of the present invention, the mRNA molecule combination, wherein the first 5' non-translated sequence and the first 3' non-translated sequence are not derived from alpha virus.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,所述第二5’端非翻译序列和第二3’端非翻译序列来源于甲病毒;优选地,来源于委内瑞拉马脑脊髓炎病毒。In some embodiments of the present invention, the mRNA molecule combination, wherein the second 5' non-translated sequence and the second 3' non-translated sequence are derived from an alpha virus; preferably, from Venezuelan equine encephalitis virus.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,In some embodiments of the present invention, the mRNA molecule combination, wherein,
所述RNA复制酶是来源于甲病毒的RNA复制酶;优选地,是来源于委内瑞拉马脑脊髓炎病毒的RNA复制酶; The RNA replicase is an RNA replicase derived from an alphavirus; preferably, it is an RNA replicase derived from Venezuelan equine encephalomyelitis virus;
优选地,所述RNA复制酶的氨基酸序列如SEQ ID NO:41所示;优选地,所述RNA复制酶的编码序列如SEQ ID NO:42所示。Preferably, the amino acid sequence of the RNA replicase is as shown in SEQ ID NO:41; preferably, the coding sequence of the RNA replicase is as shown in SEQ ID NO:42.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,In some embodiments of the present invention, the mRNA molecule combination, wherein,
所述RNA启动子是亚基因组启动子;The RNA promoter is a subgenomic promoter;
优选地,所述RNA启动子是来源于甲病毒的亚基因组启动子;Preferably, the RNA promoter is a subgenomic promoter derived from an alphavirus;
优选地,所述RNA启动子是来源于委内瑞拉马脑脊髓炎病毒的亚基因组启动子;Preferably, the RNA promoter is a subgenomic promoter derived from Venezuelan equine encephalitis virus;
优选地,所述RNA启动子是26S启动子。Preferably, the RNA promoter is the 26S promoter.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,所述microRNA结合位点序列为组织特异性表达的microRNA对应的结合位点序列;In some embodiments of the present invention, the mRNA molecule combination, wherein the microRNA binding site sequence is a binding site sequence corresponding to a tissue-specifically expressed microRNA;
优选地,所述组织特异性表达的microRNA为在肿瘤组织中低表达的microRNA;Preferably, the tissue-specifically expressed microRNA is a microRNA that is lowly expressed in tumor tissue;
优选地,所述在肿瘤组织中低表达的microRNA选自miRNA-122、miRNA-143、miRNA-1、miRNA-124、miRNA-217和miRNA-126。Preferably, the microRNA lowly expressed in tumor tissue is selected from miRNA-122, miRNA-143, miRNA-1, miRNA-124, miRNA-217 and miRNA-126.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,所述microRNA结合位点序列包含一个或多个microRNA成熟序列;优选地,所述microRNA结合位点序列包含2、3、4、5、6、7、8、9、10、11或12个序列相同或不同的microRNA成熟序列。In some embodiments of the present invention, the mRNA molecule combination, wherein the microRNA binding site sequence comprises one or more microRNA mature sequences; preferably, the microRNA binding site sequence comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 microRNA mature sequences that are identical or different in sequence.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,所述microRNA成熟序列如SEQ ID NO:30至SEQ ID NO:32中的任一序列所示。In some embodiments of the present invention, the mRNA molecule combination, wherein the microRNA mature sequence is shown in any one of SEQ ID NO:30 to SEQ ID NO:32.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,各microRNA成熟序列之间含有一个或多个相同或不同的内部间隔序列;In some embodiments of the present invention, the mRNA molecule combination contains one or more identical or different internal spacer sequences between each microRNA mature sequence;
优选地,所述内部间隔序列如SEQ ID NO:33至SEQ ID NO:34中的任一序列所示。Preferably, the internal spacer sequence is shown in any one of SEQ ID NO:33 to SEQ ID NO:34.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,所述microRNA结合位点序列的5’端和/或3’端含有一个或多个相同或不同的外部间隔序列;In some embodiments of the present invention, the mRNA molecule combination, wherein the 5' end and/or 3' end of the microRNA binding site sequence contains one or more identical or different external spacer sequences;
优选地,5’端的外部间隔序列如SEQ ID NO:35至SEQ ID NO:36中的任一序列所示;Preferably, the external spacer sequence at the 5' end is shown in any one of SEQ ID NO:35 to SEQ ID NO:36;
优选地,3’端的外部间隔序列如SEQ ID NO:37至SEQ ID NO:38中的任一序列所示。Preferably, the external spacer sequence at the 3’ end is as shown in any one of SEQ ID NO:37 to SEQ ID NO:38.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,所述microRNA结 合位点序列的5’端和/或3’端含有一个或多个甲病毒RNA复制酶或nsp2能够识别的酶切位点序列;In some embodiments of the present invention, the mRNA molecule combination, wherein the microRNA The 5' end and/or 3' end of the cleavage site sequence contains one or more restriction site sequences that can be recognized by the alphavirus RNA replicase or nsp2;
优选地,为nsp1和nsp2之间的能够被甲病毒复制酶或nsp2识别的酶切位点序列;Preferably, it is a restriction site sequence between nsp1 and nsp2 that can be recognized by the alphavirus replicase or nsp2;
优选地,所述酶切位点序列如SEQ ID NO:53所示。Preferably, the restriction site sequence is as shown in SEQ ID NO:53.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,所述microRNA结合位点序列如SEQ ID NO:22所示。In some embodiments of the present invention, the mRNA molecule combination, wherein the microRNA binding site sequence is as shown in SEQ ID NO:22.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,所述第一mRNA分子和/或第二mRNA分子还含有5’端帽子结构。In some embodiments of the present invention, the mRNA molecule combination, wherein the first mRNA molecule and/or the second mRNA molecule further contains a 5' end cap structure.
在本发明的一些实施方式中,所述的mRNA分子组合,其中,所述目标蛋白是抗原。In some embodiments of the present invention, the mRNA molecule combination, wherein the target protein is an antigen.
在本发明的一些实施方式中,所述的mRNA分子组合,其如图23所示。In some embodiments of the present invention, the mRNA molecule combination is as shown in Figure 23.
不拘于理论的限制,第一mRNA分子编码nsp1至nsp4,第二mRNA分子编码目标蛋白。Without being bound by theory, the first mRNA molecule encodes nsp1 to nsp4, and the second mRNA molecule encodes the target protein.
本发明的mRNA分子组合是组织特异性自扩增的核酸分子组合。The mRNA molecule combination of the present invention is a tissue-specific self-amplifying nucleic acid molecule combination.
本发明的另一方面涉及一种DNA分子,其编码本发明中任一项所述的mRNA分子,或者编码本发明中任一项所述的第一mRNA分子和第二mRNA分子,并且第一mRNA分子和第二mRNA分子在同一个DNA分子上。Another aspect of the present invention relates to a DNA molecule encoding the mRNA molecule described in any one of the present invention, or encoding the first mRNA molecule and the second mRNA molecule described in any one of the present invention, and the first mRNA molecule and the second mRNA molecule are on the same DNA molecule.
在本发明的一些实施方式中,所述的DNA分子,其在编码甲病毒RNA复制酶的序列的上游包含T7启动子、SP6启动子或T3启动子。In some embodiments of the present invention, the DNA molecule comprises a T7 promoter, an SP6 promoter or a T3 promoter upstream of the sequence encoding the alphavirus RNA replicase.
在本发明的一些实施方式中,所述的DNA分子是分离的DNA分子。In some embodiments of the invention, the DNA molecule is an isolated DNA molecule.
本发明的另一方面涉及一种DNA分子组合,包含第一DNA分子和第二DNA分子,其中:Another aspect of the present invention relates to a DNA molecule combination, comprising a first DNA molecule and a second DNA molecule, wherein:
第一DNA分子编码本发明中任一项所述的第一mRNA分子;和The first DNA molecule encodes the first mRNA molecule described in any one of the present invention; and
第二DNA分子编码本发明中任一项所述的第二mRNA分子。The second DNA molecule encodes the second mRNA molecule described in any one of the present invention.
根据本发明中任一项所述的mRNA分子、本发明中任一项所述的mRNA分子组合、本发明任一项所述的DNA分子或本发明任一项所述的DNA分子组合,其用于治疗或预防病毒感染、治疗或预防肿瘤、或者蛋白替代疗法;The mRNA molecule according to any one of the present invention, the mRNA molecule combination according to any one of the present invention, the DNA molecule according to any one of the present invention, or the DNA molecule combination according to any one of the present invention, for use in treating or preventing viral infection, treating or preventing tumors, or protein replacement therapy;
优选地,所述病毒感染是指新冠病毒感染;Preferably, the viral infection refers to novel coronavirus infection;
优选地,所述肿瘤选自肝癌、横纹肌肉瘤、神经胶质瘤、膀胱癌、结直肠癌、胰 腺癌和肺癌;Preferably, the tumor is selected from liver cancer, rhabdomyosarcoma, glioma, bladder cancer, colorectal cancer, pancreatic cancer, adenocarcinoma and lung cancer;
优选地,所述肿瘤为具有一种microRNA低表达或多种microRNA低表达的肿瘤;优选地,所述microRNA为选自miRNA-122、miRNA-143、miRNA-1、miRNA-124、miRNA-217和miRNA-126中的一种或多种。Preferably, the tumor is a tumor with low expression of one microRNA or multiple microRNAs; preferably, the microRNA is one or more selected from miRNA-122, miRNA-143, miRNA-1, miRNA-124, miRNA-217 and miRNA-126.
微小RNA(microRNA)是一类由内源基因编码的长度约为22nt的非编码单链RNA分子,它们在动植物中参与转录后基因表达调控。microRNA通过识别目标基因的mRNA序列并与其结合,导致mRNA的翻译被抑制或者mRNA的降解,从而调控mRNA编码基因的表达量。这种基因表达调控机制被称为microRNA介导的转录后调控(miRNA-mediated post-transcriptional regulation)。microRNA具有组织特异性,其在不同的组织中表达量不同。例如:MicroRNA is a type of non-coding single-stranded RNA molecule with a length of about 22nt encoded by endogenous genes. They participate in post-transcriptional gene expression regulation in animals and plants. MicroRNA recognizes the mRNA sequence of the target gene and binds to it, resulting in the inhibition of mRNA translation or the degradation of mRNA, thereby regulating the expression of the mRNA encoding gene. This gene expression regulation mechanism is called microRNA-mediated post-transcriptional regulation. MicroRNA has tissue specificity and its expression levels vary in different tissues. For example:
miRNA-122在肝脏中高水平表达,而在非肝细胞(例如肌肉细胞等)和肝癌组织中表达量较低;miRNA-122 is highly expressed in the liver, but is less expressed in non-hepatic cells (e.g., muscle cells) and liver cancer tissues;
miRNA-143在结直肠组织中高表达,而在结直肠癌组织中低表达;miRNA-143 is highly expressed in colorectal tissues but lowly expressed in colorectal cancer tissues;
miRNA-1在骨骼肌、心肌组织中高表达,而在非肌组织和横纹肌肉瘤组织中低表达;miRNA-1 is highly expressed in skeletal muscle and cardiac muscle tissues, but is lowly expressed in non-muscle tissues and rhabdomyosarcoma tissues;
miRNA-124在神经元组织中高表达,而在神经胶质瘤和膀胱癌组织中低表达;miRNA-124 is highly expressed in neuronal tissues, but lowly expressed in glioma and bladder cancer tissues;
miRNA-217在胰腺组织中高表达,而在胰腺癌组织中低表达;miRNA-217 is highly expressed in pancreatic tissues but lowly expressed in pancreatic cancer tissues;
miRNA-126和miRNA-143在肺组织中高表达,而在肺癌组织中低表达。miRNA-126 and miRNA-143 were highly expressed in lung tissues, but lowly expressed in lung cancer tissues.
上述特异性使得miRNA成为疾病诊断或治疗的有力工具。The above specificity makes miRNA a powerful tool for disease diagnosis or treatment.
不拘于理论的限制,任何病原体感染或任何肿瘤类型都适用于该条要求,更具体地可以也对应某类特定microRNA低表达的肿瘤类型,如大多数肝癌microRNA122低表达,则本发明的自扩增mRNA可特异地在肝癌组织中表达。Without being limited by theory, any pathogen infection or any tumor type is applicable to this requirement. More specifically, it may also correspond to a tumor type with low expression of a certain type of specific microRNA. For example, most liver cancers have low expression of microRNA122, and the self-amplifying mRNA of the present invention can be specifically expressed in liver cancer tissue.
本发明的另一方面涉及一种重组载体,其含有本发明中任一项所述的DNA分子;优选地,所述重组载体为重组的原核表达载体或重组的真核表达载体。Another aspect of the present invention relates to a recombinant vector, which contains the DNA molecule described in any one of the present invention; preferably, the recombinant vector is a recombinant prokaryotic expression vector or a recombinant eukaryotic expression vector.
本发明的另一方面涉及一种重组宿主细胞,其含有本发明中任一项所述的mRNA分子、本发明中任一项所述的mRNA分子组合、本发明中任一项所述的DNA分子或者本发明中任一项所述的DNA分子组合。Another aspect of the present invention relates to a recombinant host cell, which contains the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the DNA molecule combination described in any one of the present invention.
本发明的另一方面涉及一种试剂盒,其含有本发明中任一项所述的mRNA分子、本发明中任一项所述的mRNA分子组合、本发明中任一项所述的DNA分子或者本发明 中任一项所述的DNA分子组合,以及脂质体类递送系统。Another aspect of the present invention relates to a kit comprising the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the The DNA molecule combination described above, and the liposome delivery system.
本发明的另一方面涉及一种药物组合物,其含有本发明中任一项所述的mRNA分子、本发明中任一项所述的mRNA分子组合、本发明中任一项所述的DNA分子或者本发明中任一项所述的DNA分子组合,以及一种或多种药学上可接受的辅料;优选地,所述辅料为脂质体类递送系统。Another aspect of the present invention relates to a pharmaceutical composition, which contains the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the DNA molecule combination described in any one of the present invention, and one or more pharmaceutically acceptable excipients; preferably, the excipient is a liposome delivery system.
本发明的另一方面涉及一种疫苗制剂,其含有本发明中任一项所述的mRNA分子、本发明中任一项所述的mRNA分子组合、本发明中任一项所述的DNA分子或者本发明中任一项所述的DNA分子组合;Another aspect of the present invention relates to a vaccine preparation, which contains the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the DNA molecule combination described in any one of the present invention;
优选地,所述mRNA分子、mRNA分子组合、DNA分子或DNA分子组合物被脂质体类递送系统包裹;Preferably, the mRNA molecule, the combination of mRNA molecules, the DNA molecule or the combination of DNA molecules is encapsulated by a liposome-based delivery system;
可选地,所述疫苗制剂还包含一种或多种疫苗用佐剂;Optionally, the vaccine formulation further comprises one or more vaccine adjuvants;
优选地,所述疫苗制剂为预防病毒感染例如新冠病毒感染或预防新冠病毒感染所致重症的疫苗制剂。Preferably, the vaccine preparation is a vaccine preparation for preventing viral infection such as novel coronavirus infection or preventing severe illness caused by novel coronavirus infection.
脂质类递送系统包括脂质体(liposome)、脂质纳米粒(lipid Nanoparticle,LNP)、脂质多聚物纳米载体(Lipopolyplex,LPP)。Lipid delivery systems include liposomes, lipid nanoparticles (LNP), and lipid polymer nanocarriers (LPP).
脂质类中的LPP是一种以聚合物包载mRNA为内核、磷脂包裹为外壳的双层结构。LPP的双层脂质体膜具有更好的包载、保护mRNA的效果,LPP的内核能够随聚合物的降解逐步释放mRNA分子。LPP靶向树突状细胞效果优异,能够更好的通过抗原递呈激活T细胞的免疫反应,达到理想的治疗效果。LPP among lipids is a double-layer structure with polymer-encapsulated mRNA as the core and phospholipids as the shell. The double-layer liposome membrane of LPP has a better effect of encapsulating and protecting mRNA, and the core of LPP can gradually release mRNA molecules as the polymer degrades. LPP has excellent targeting effect on dendritic cells and can better activate the immune response of T cells through antigen presentation to achieve ideal therapeutic effects.
不拘于理论的限制,mRNA能有效激起细胞免疫和体液免疫。注射的mRNA疫苗被抗原呈递细胞内吞。在逃离内质体进入胞浆后,mRNA被核糖体翻译成蛋白质。翻译后的抗原蛋白可以通过多种方式刺激免疫系统,激起人体的细胞免疫和体液免疫。与传统的灭活疫苗、亚单位疫苗和基因工程疫苗相比,核酸疫苗具有如下优点:研发周期短;生产工艺简单、扩产容易;无需佐剂、有效性高;不进入细胞核、安全性较好等。mRNA新冠疫苗验证了mRNA技术平台在疫苗领域的适用性。Without being bound by theory, mRNA can effectively stimulate cellular immunity and humoral immunity. The injected mRNA vaccine is internalized by antigen-presenting cells. After escaping the endosome and entering the cytoplasm, the mRNA is translated into protein by the ribosome. The translated antigen protein can stimulate the immune system in a variety of ways, stimulating the body's cellular immunity and humoral immunity. Compared with traditional inactivated vaccines, subunit vaccines and genetically engineered vaccines, nucleic acid vaccines have the following advantages: short R&D cycle; simple production process and easy expansion; no adjuvant is required and the effectiveness is high; it does not enter the cell nucleus and has good safety. The mRNA COVID-19 vaccine verifies the applicability of the mRNA technology platform in the vaccine field.
本发明的另一方面涉及本发明中任一项所述的mRNA分子、本发明中任一项所述的mRNA分子组合、本发明中任一项所述的DNA分子或者本发明中任一项所述的DNA分子组合在制备治疗或预防病毒感染的药物、治疗或预防肿瘤的药物、或者用于蛋白替代疗法的药物中的用途; Another aspect of the present invention relates to the use of the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the DNA molecule combination described in any one of the present invention in the preparation of a drug for treating or preventing viral infection, a drug for treating or preventing tumors, or a drug for protein replacement therapy;
优选地,所述病毒感染是指新冠病毒感染;Preferably, the viral infection refers to novel coronavirus infection;
优选地,所述肿瘤为选自肝癌、横纹肌肉瘤、神经胶质瘤、膀胱癌、结直肠癌、胰腺癌和肺癌中的一种或多种;Preferably, the tumor is one or more selected from liver cancer, rhabdomyosarcoma, glioma, bladder cancer, colorectal cancer, pancreatic cancer and lung cancer;
优选地,所述肿瘤为具有一种microRNA低表达或多种microRNA低表达的肿瘤;优选地,所述microRNA为选自miRNA-122、miRNA-143、miRNA-1、miRNA-124、miRNA-217和miRNA-126中的一种或多种。Preferably, the tumor is a tumor with low expression of one microRNA or multiple microRNAs; preferably, the microRNA is one or more selected from miRNA-122, miRNA-143, miRNA-1, miRNA-124, miRNA-217 and miRNA-126.
本发明的再一方面涉及一种治疗或预防病毒感染或者肿瘤的方法或者一种蛋白替代疗法,包括给予有需求的受试者以有效量的本发明中任一项所述的mRNA分子、本发明中任一项所述的mRNA分子组合、本发明中任一项所述的DNA分子或者本发明中任一项所述的DNA分子组合的步骤;Another aspect of the present invention relates to a method for treating or preventing viral infection or tumor or a protein replacement therapy, comprising the step of administering to a subject in need thereof an effective amount of the mRNA molecule described in any one of the present invention, the mRNA molecule combination described in any one of the present invention, the DNA molecule described in any one of the present invention, or the DNA molecule combination described in any one of the present invention;
优选地,所述病毒感染是指新冠病毒感染;Preferably, the viral infection refers to novel coronavirus infection;
优选地,所述肿瘤为选自肝癌、横纹肌肉瘤、神经胶质瘤、膀胱癌、结直肠癌、胰腺癌和肺癌中的一种或多种;Preferably, the tumor is one or more selected from liver cancer, rhabdomyosarcoma, glioma, bladder cancer, colorectal cancer, pancreatic cancer and lung cancer;
优选地,所述肿瘤为具有一种microRNA低表达或多种microRNA低表达的肿瘤;优选地,所述microRNA为选自miRNA-122、miRNA-143、miRNA-1、miRNA-124、miRNA-217和miRNA-126中的一种或多种。Preferably, the tumor is a tumor with low expression of one microRNA or multiple microRNAs; preferably, the microRNA is one or more selected from miRNA-122, miRNA-143, miRNA-1, miRNA-124, miRNA-217 and miRNA-126.
具有增强的外源基因表达水平的自扩增mRNA核酸序列Self-amplifying mRNA nucleic acid sequences with enhanced expression levels of foreign genes
本发明还提供了改进的saRNA构建体,以及制备和使用该构建体的方法。本发明进一步提供了将该saRNA用于携带目的基因以用于治疗、预防和/或诊断用途。具体地,本发明经过深入的研究和创造性劳动,得到了一种优化的自扩增mRNA骨架序列,与原始序列相比,能够显著降低诱导的天然免疫反应强度,显著减弱受到的翻译抑制,以及明显减少细胞凋亡,进而显著提升表达效率。由此提供了下述发明:The present invention also provides an improved saRNA construct, and a method for preparing and using the construct. The present invention further provides the use of the saRNA to carry a target gene for treatment, prevention and/or diagnosis. Specifically, after in-depth research and creative work, the present invention has obtained an optimized self-amplified mRNA backbone sequence, which can significantly reduce the intensity of the induced natural immune response, significantly reduce the translation inhibition, and significantly reduce cell apoptosis compared to the original sequence, thereby significantly improving the expression efficiency. The following invention is thus provided:
本发明的一个方面提供了一种mRNA分子,其编码来源于甲病毒的由非结构性蛋白1、2、3和4组成的RNA复制酶,其中所述非结构性蛋白3在其N末端包含如SEQ ID NO:57或59的氨基酸序列所示的宏结构域。在一些实施方案中,所述非结构性蛋白1、2、3和4来源于委内瑞拉马脑脊髓炎病毒。One aspect of the present invention provides an mRNA molecule encoding an RNA replicase composed of non-structural proteins 1, 2, 3 and 4 derived from an alphavirus, wherein the non-structural protein 3 comprises a macrodomain at its N-terminus as shown in the amino acid sequence of SEQ ID NO: 57 or 59. In some embodiments, the non-structural proteins 1, 2, 3 and 4 are derived from Venezuelan equine encephalitis virus.
在一些实施方案中,所述mRNA分子编码具有如SEQ ID NO:71所示或与SEQ ID NO:71至少90%相同的氨基酸序列的非结构性蛋白1、具有如SEQ ID NO:72所示或与 SEQ ID NO:72至少90%相同的氨基酸序列的非结构性蛋白2、和具有如SEQ ID NO:75所示或与SEQ ID NO:75至少90%相同的氨基酸序列的非结构性蛋白4。进一步地,所述mRNA分子编码具有与SEQ ID NO:73或74至少90%相同的氨基酸序列且在N末端包含如SEQ ID NO:57或59的氨基酸序列所示的宏结构域的非结构性蛋白3。In some embodiments, the mRNA molecule encodes a nonstructural protein 1 having an amino acid sequence as shown in SEQ ID NO: 71 or at least 90% identical to SEQ ID NO: 71, a nonstructural protein 2 having an amino acid sequence as shown in SEQ ID NO: 72 or at least 90% identical to SEQ ID NO: 73. The mRNA molecule encodes a nonstructural protein 3 having an amino acid sequence at least 90% identical to SEQ ID NO: 73 or 74 and comprising a macrodomain as shown in the amino acid sequence of SEQ ID NO: 57 or 59 at the N-terminus.
在一些实施方案中,所述mRNA分子包含如SEQ ID NO:68或70所示的编码所述宏结构域的核苷酸序列。In some embodiments, the mRNA molecule comprises a nucleotide sequence encoding the macro domain as shown in SEQ ID NO:68 or 70.
在一些实施方案中,所述mRNA分子包含SEQ ID NO:67或69的核苷酸序列或由其组成。In some embodiments, the mRNA molecule comprises or consists of the nucleotide sequence of SEQ ID NO:67 or 69.
在一些实施方案中,所述mRNA分子还包含目的基因编码序列以及任选地,在目的基因编码序列上游的RNA启动子。In some embodiments, the mRNA molecule further comprises a target gene coding sequence and, optionally, an RNA promoter upstream of the target gene coding sequence.
在一些实施方案中,所述mRNA分子还包含:In some embodiments, the mRNA molecule further comprises:
5’端非翻译区、RNA启动子、目的基因编码序列、3’端非翻译区和多聚腺苷酸序列,5' untranslated region, RNA promoter, target gene coding sequence, 3' untranslated region and polyadenylation sequence,
任选地,所述mRNA分子还包含5’帽序列、信号肽编码序列、Kozak序列和/或限制酶切割位点。所述Kozak序列可连接于RNA启动子的3’端。Optionally, the mRNA molecule further comprises a 5' cap sequence, a signal peptide coding sequence, a Kozak sequence and/or a restriction enzyme cleavage site. The Kozak sequence can be connected to the 3' end of the RNA promoter.
在一些实施方案中,所述mRNA分子从5’至3’依次包含以下可操作地连接的核苷酸序列:5’端非翻译区、非结构性蛋白1、2、3和4的编码序列、RNA启动子、目的基因编码序列、3’端非翻译区和多聚腺苷酸序列,In some embodiments, the mRNA molecule comprises the following operably linked nucleotide sequences in order from 5' to 3': a 5' untranslated region, coding sequences of nonstructural proteins 1, 2, 3 and 4, an RNA promoter, a target gene coding sequence, a 3' untranslated region and a polyadenylation sequence,
任选地,所述核苷酸序列之间可通过接头序列连接。Optionally, the nucleotide sequences may be connected via a linker sequence.
在一些实施方案中,所述非结构性蛋白1、2、3和4的编码序列包含可操作地连接的编码非结构性蛋白1的核苷酸序列、编码非结构性蛋白2的核苷酸序列、编码非结构性蛋白3的核苷酸序列和编码非结构性蛋白4的核苷酸序列。In some embodiments, the coding sequences of non-structural proteins 1, 2, 3 and 4 comprise a nucleotide sequence encoding non-structural protein 1, a nucleotide sequence encoding non-structural protein 2, a nucleotide sequence encoding non-structural protein 3 and a nucleotide sequence encoding non-structural protein 4 that are operably linked.
在一些实施方案中,所述非结构性蛋白1、2、3和4的编码序列从5’至3’包含:可操作地连接的编码非结构性蛋白1的核苷酸序列、编码非结构性蛋白2的核苷酸序列、编码非结构性蛋白3的核苷酸序列和编码非结构性蛋白4的核苷酸序列。In some embodiments, the coding sequences of non-structural proteins 1, 2, 3 and 4 include from 5' to 3': a nucleotide sequence encoding non-structural protein 1, a nucleotide sequence encoding non-structural protein 2, a nucleotide sequence encoding non-structural protein 3 and a nucleotide sequence encoding non-structural protein 4 that are operably linked.
在一些实施方案中,所述5’端非翻译区来源于甲病毒;优选地,来源于委内瑞拉马脑脊髓炎病毒;例如,所述5’端非翻译区包含与SEQ ID NO:63至少85%相同的核苷酸序列。In some embodiments, the 5' untranslated region is derived from an alpha virus; preferably, from a Venezuelan equine encephalitis virus; for example, the 5' untranslated region comprises a nucleotide sequence that is at least 85% identical to SEQ ID NO:63.
在一些实施方案中,所述3’端非翻译区来源于甲病毒;优选地,来源于委内瑞拉 马脑脊髓炎病毒;例如,所述3’端非翻译区包含与SEQ ID NO:64至少85%相同的核苷酸序列。In some embodiments, the 3' untranslated region is derived from an alphavirus; preferably, from Venezuela Equine encephalitis virus; for example, the 3' untranslated region comprises a nucleotide sequence that is at least 85% identical to SEQ ID NO:64.
在一些实施方案中,所述RNA启动子是亚基因组启动子,例如来源于甲病毒的亚基因组启动子。优选地,所述RNA启动子是来源于委内瑞拉马脑脊髓炎病毒的亚基因组启动子。In some embodiments, the RNA promoter is a subgenomic promoter, such as a subgenomic promoter derived from an alphavirus. Preferably, the RNA promoter is a subgenomic promoter derived from Venezuelan equine encephalitis virus.
在一些实施方案中,所述RNA启动子是26S启动子。例如,所述RNA启动子的序列如SEQ ID NO:65所示。In some embodiments, the RNA promoter is a 26S promoter. For example, the sequence of the RNA promoter is shown in SEQ ID NO:65.
在一些实施方案中,所述多聚腺苷酸序列的序列如SEQ ID NO:66所示。In some embodiments, the sequence of the poly(A) sequence is as shown in SEQ ID NO:66.
在一些实施方案中,所述目的基因编码序列为编码治疗多肽、预防多肽、诊断多肽、报告基因、抗原或编码调节结构(例如非编码基因)的序列。In some embodiments, the target gene coding sequence is a sequence encoding a therapeutic polypeptide, a preventive polypeptide, a diagnostic polypeptide, a reporter gene, an antigen, or a sequence encoding a regulatory structure (eg, a non-coding gene).
在一些实施方案中,所述mRNA分子包含如SEQ ID NO:58、SEQ ID NO:60至SEQ ID NO:62中的任一序列所示的核苷酸序列。In some embodiments, the mRNA molecule comprises a nucleotide sequence as shown in any one of SEQ ID NO:58, SEQ ID NO:60 to SEQ ID NO:62.
在一方面,本发明提供了一种DNA分子,其编码如本文公开的mRNA分子。In one aspect, the invention provides a DNA molecule encoding an mRNA molecule as disclosed herein.
在一方面,本发明提供了一种重组载体,其含有如本文公开的DNA分子;优选地,所述重组载体为原核表达载体或真核表达载体。In one aspect, the present invention provides a recombinant vector comprising a DNA molecule as disclosed herein; preferably, the recombinant vector is a prokaryotic expression vector or a eukaryotic expression vector.
在一方面,本发明提供了一种重组宿主细胞,其含有如本文公开的mRNA分子、DNA分子或者重组载体。In one aspect, the present invention provides a recombinant host cell comprising an mRNA molecule, a DNA molecule or a recombinant vector as disclosed herein.
在一方面,本发明提供了一种药物组合物,其含有如本文公开的mRNA分子或者DNA分子,以及一种或多种药学上可接受的载剂;优选地,所述载剂为脂质体类递送系统。In one aspect, the present invention provides a pharmaceutical composition comprising an mRNA molecule or a DNA molecule as disclosed herein, and one or more pharmaceutically acceptable carriers; preferably, the carrier is a liposome delivery system.
在一方面,本发明提供了一种疫苗制剂,其含有如本文公开的mRNA分子或者DNA分子;优选地,所述mRNA分子或者DNA分子被脂质体类递送系统包裹。所述疫苗制剂还可包含一种或多种疫苗用佐剂。相应地,所述mRNA编码的目的基因可以编码疫苗抗原。例如,所述疫苗制剂为预防病毒感染例如新冠病毒感染或预防新冠病毒感染所致重症的疫苗制剂。相应地,所述mRNA编码的目的基因可以编码新冠病毒的免疫原性肽。In one aspect, the present invention provides a vaccine formulation comprising an mRNA molecule or a DNA molecule as disclosed herein; preferably, the mRNA molecule or the DNA molecule is encapsulated by a liposome delivery system. The vaccine formulation may also contain one or more vaccine adjuvants. Accordingly, the target gene encoded by the mRNA may encode a vaccine antigen. For example, the vaccine formulation is a vaccine formulation for preventing viral infection such as novel coronavirus infection or preventing severe illness caused by novel coronavirus infection. Accordingly, the target gene encoded by the mRNA may encode an immunogenic peptide of the novel coronavirus.
在一方面,本发明提供了如本文公开的mRNA分子或者DNA分子在制备治疗或预防病毒感染的药物、治疗或预防肿瘤的药物、或者用于蛋白替代疗法的药物中的用途。相应地,所述mRNA编码的目的基因可以是疫苗抗原基因、肿瘤杀伤基因、治疗性蛋 白基因、抗体基因等。In one aspect, the present invention provides the use of the mRNA molecule or DNA molecule disclosed herein in the preparation of a drug for treating or preventing viral infection, a drug for treating or preventing tumors, or a drug for protein replacement therapy. Accordingly, the target gene encoded by the mRNA can be a vaccine antigen gene, a tumor killing gene, a therapeutic protein, or a Protein genes, antibody genes, etc.
在一方面,本发明提供了一种试剂盒,其含有如本文公开的mRNA分子或者DNA分子。In one aspect, the invention provides a kit comprising an mRNA molecule or a DNA molecule as disclosed herein.
图1:对saRNA-EGFP模板质粒psaRNA-062中4497-4503位改造后后的体外转录RNA毛细管电泳结果。Figure 1: Capillary electrophoresis results of in vitro transcribed RNA after modification of position 4497-4503 in the saRNA-EGFP template plasmid psaRNA-062.
图2:以psaRNA-152为模板改造得到的不同saRNA模板进行体外转录的RNA毛细管电泳叠加结果图。saRNA-062为原始saRNA,saRNA-183至185为改造后得到的saRNA。Figure 2: Overlay of RNA capillary electrophoresis results of in vitro transcription of different saRNA templates modified using psaRNA-152 as a template. saRNA-062 is the original saRNA, and saRNA-183 to 185 are the modified saRNAs.
图3:对psaRNA-183为模板中1680-1676位改造后的体外转录RNA毛细管电泳结果。黑框表示骨架优化前后短转录产物的电泳检测峰形。Figure 3: Capillary electrophoresis results of in vitro transcribed RNA after modification of positions 1680-1676 in psaRNA-183 as template. The black box indicates the electrophoresis detection peak shape of the short transcript before and after backbone optimization.
图4:检测优化后得到的编码荧光素酶的saRNA-152及原始saRNA-062在转染BHK-21细胞24和72小时后的表达水平结果。Figure 4: Expression level results of optimized luciferase-encoding saRNA-152 and original saRNA-062 after transfection of BHK-21 cells for 24 and 72 hours.
图5:检测优化后得到的编码荧光素酶的saRNA-243及原始saRNA-062在转染BHK-21细胞24小时后的表达水平结果。Figure 5: Expression level results of saRNA-243 encoding luciferase and original saRNA-062 obtained after detection optimization after transfection of BHK-21 cells for 24 hours.
图6:检测优化后得到的编码荧光素酶的saRNA-152及原始saRNA-062在转染BHK-21细胞24和72小时后的复制结果,数据和转染后2小时结果进行了归一化处理。Figure 6: Replication results of optimized luciferase-encoding saRNA-152 and original saRNA-062 after transfection of BHK-21 cells for 24 and 72 hours. The data were normalized to the results 2 hours after transfection.
图7:检测S23H02完整性的毛细管电泳检测结果。Figure 7: Capillary electrophoresis results for detecting the integrity of S23H02.
图8:将S23H02、M22H04和阴性对照分别转染至含有脂质体3000的组织培养物后,检测其分泌的HPV16抗原表达的E7-ELISA试验结果。Figure 8: E7-ELISA test results for detecting the secreted HPV16 antigen expression of S23H02, M22H04 and negative control after transfection into tissue culture containing Lipofectamine 3000.
图9:将C57bl/6小鼠用TC1肿瘤细胞进行攻毒,并注射3剂治疗性疫苗后的肿瘤体积数据。记录肿瘤体积和小鼠体重。每只小鼠每剂的剂量为按照活性成分计算。Figure 9: Tumor volume data of C57bl/6 mice challenged with TC1 tumor cells and injected with 3 doses of therapeutic vaccine. Tumor volume and mouse body weight were recorded. The dose per mouse per dose was calculated based on the active ingredient.
图10:将C57bl/6小鼠用TC1肿瘤细胞进行攻毒,并注射3剂治疗性疫苗后的小鼠体重数据。每只小鼠每剂的剂量为按照活性成分计算。Figure 10: Body weight data of C57bl/6 mice after being challenged with TC1 tumor cells and injected with 3 doses of therapeutic vaccine. The dose per mouse per dose was calculated based on the active ingredient.
图11:用流式细胞仪检测注射了1剂治疗性疫苗的C57bl/6小鼠脾细胞中E7特异性的CD8T细胞的数量。每只小鼠每剂的剂量为按照活性成分计算。Figure 11: Flow cytometry was used to detect the number of E7-specific CD8 T cells in the spleen cells of C57bl/6 mice injected with one dose of therapeutic vaccine. The dose per mouse per dose was calculated based on the active ingredient.
图12:miRNA122结合位点序列盒的示意图。通过将6个microRNA-122-5p以 内部间隔序列(inner spacer)分隔后两端加入外部间隔序列(outer sapcer),最外侧为复制酶可识别的蛋白酶酶切位点。Figure 12: Schematic diagram of the miRNA122 binding site sequence box. After the inner spacer sequence is separated, an outer spacer sequence is added at both ends, and the outermost portion is a protease cleavage site that can be recognized by the replication enzyme.
图13:自扩增mRNA结构,结构组成从左至右分别为帽结构-5端甲病毒非翻译序列(5’UTR)-甲病毒复制酶序列-26S启动子序列-任意靶蛋白序列X-3端甲病毒非翻译序列(3’UTR)-多聚腺嘌呤序列(polyA序列)。Figure 13: Self-amplifying mRNA structure, the structural components from left to right are cap structure-5-terminal alphavirus non-translated sequence (5'UTR)-alphavirus replicase sequence-26S promoter sequence-arbitrary target protein sequence X-3-terminal alphavirus non-translated sequence (3'UTR)-polyadenine sequence (polyA sequence).
图14:不同部位分别插入microRNA122结合位点的自扩增mRNA结构的示意图。箭头代表microRNA122结合位点的不同置入位置。microRNA122结合位点由6个microRNA122结合位点串联组合得到。Figure 14: Schematic diagram of the self-amplified mRNA structure with microRNA122 binding sites inserted in different positions. Arrows represent different insertion positions of microRNA122 binding sites. The microRNA122 binding site is obtained by combining 6 microRNA122 binding sites in series.
图15:六个不同部位分别插入microRNA122结合位点的自扩增mRNA体外转录变性琼脂糖凝胶电泳检测结果。Figure 15: Denaturing agarose gel electrophoresis detection results of in vitro transcription of self-amplified mRNA inserted into the microRNA122 binding site at six different locations.
图16:不同细胞中microRNA-122的qPCR检测结果。Figure 16: qPCR detection results of microRNA-122 in different cells.
图17:转染表达Nluc-EGFP的插入microRNA122结合位点的自扩增mRNA至Huh7.5.1细胞和C2C12细胞24小时后,通过荧光显微镜检测荧光蛋白的表达情况,control为未插入microRNA122结合位点的自扩增mRNA。Figure 17: 24 hours after transfection of Huh7.5.1 cells and C2C12 cells with self-amplified mRNA expressing Nluc-EGFP and inserted microRNA122 binding site, the expression of fluorescent protein was detected by fluorescence microscopy. The control was self-amplified mRNA without inserted microRNA122 binding site.
图18:转染表达Nluc-EGFP的插入microRNA122结合位点的自扩增mRNA至Huh7.5.1细胞和C2C12细胞24小时后,Nluc荧光酶素的表达情况,control为未插入microRNA122结合位点的自扩增mRNA,表达值通过与萤火虫荧光酶素进行归一化处理。Figure 18: Expression of Nluc luciferase 24 hours after transfection of self-amplified mRNA expressing Nluc-EGFP with microRNA122 binding site inserted into Huh7.5.1 cells and C2C12 cells. The control is self-amplified mRNA without microRNA122 binding site inserted. The expression value is normalized with firefly luciferase.
图19:转染表达Nluc-EGFP的插入microRNA122结合位点的自扩增mRNA至C2C12细胞24小时后,细胞中自扩增mRNA的RNA相对水平结果,control为未插入microRNA122结合位点的自扩增mRNA,转染24小时后的RNA相对水平值与转染2小时进行归一化处理,nsp1代表编码复制酶序列的RNA相对水平,EGFP代表编码EGFP序列的RNA相对水平。Figure 19: Relative RNA levels of self-amplified mRNA in C2C12 cells 24 hours after transfection of self-amplified mRNA expressing Nluc-EGFP with microRNA122 binding site inserted into cells. Control is self-amplified mRNA without microRNA122 binding site inserted. The relative RNA level value after 24 hours of transfection is normalized with that after 2 hours of transfection. nsp1 represents the relative level of RNA encoding replicase sequence, and EGFP represents the relative level of RNA encoding EGFP sequence.
图20:转染表达Nluc-EGFP的插入microRNA122结合位点的自扩增mRNA至Huh7.5.1细胞24小时后,细胞中自扩增mRNA的RNA相对水平结果,control为未插入microRNA122结合位点的自扩增mRNA,转染24小时后的RNA相对水平值与转染2小时进行归一化处理,nsp1代表编码复制酶序列的RNA相对水平,EGFP代表编码EGFP序列的RNA相对水平。Figure 20: Relative RNA level of self-amplified mRNA in Huh7.5.1 cells 24 hours after transfection of self-amplified mRNA expressing Nluc-EGFP with microRNA122 binding site inserted into the cells. Control is self-amplified mRNA without microRNA122 binding site inserted. The relative RNA level value after 24 hours of transfection is normalized with that after 2 hours of transfection. nsp1 represents the relative level of RNA encoding the replicase sequence, and EGFP represents the relative level of RNA encoding the EGFP sequence.
图21:插入不同拷贝数肝特异性microRNA-122结合序列的自扩增mRNA的示 意图。Figure 21: Schematic diagram of self-amplified mRNA with different copy numbers of liver-specific microRNA-122 binding sequences inserted intention.
图22:转染表达Nluc-EGFP的插入不同拷贝数microRNA122结合位点的自扩增mRNA至Huh7.5.1或C2C12细胞24小时后,Nluc荧光酶素的表达情况。FIG. 22 : Expression of Nluc luciferase 24 hours after transfection of self-amplified mRNA expressing Nluc-EGFP and inserted with different copies of microRNA122 binding sites into Huh7.5.1 or C2C12 cells.
图23:mRNA分子组合的示意图。Figure 23: Schematic diagram of mRNA molecule assembly.
图24:saRNA中宏结构域(Macrodomain)的突变氨基酸位置及不同病毒宏结构域氨基酸序列比对结果。其中Macro为宏结构域,AUD为甲病毒独具的结构域(alphavirus-unique domain),HVD为高变结构域(hypervariabledomain),不同形状代表不同突变,氨基酸比对结果中同源性超过50%的氨基酸用阴影标记。图中所列病毒的全称如下:SARS-CoV:严重急性呼吸综合征冠状病毒(Severe Acute Respiratory Syndrome Coronavirus);SARS-CoV-2:严重急性呼吸综合征冠状病毒2型(Severe Acute Respiratory Syndrome Coronavirus 2);MERS-CoV:中东呼吸综合征冠状病毒(Middle East Respiratory Syndrome Coronavirus);SFV:塞姆利基森林病毒(Semliki Forest Virus);CHIKV:奇昆古尼亚病毒(Chikungunya Virus);SINV:辛德比斯病毒(Sindbis Virus);MAYV:马雅罗病毒(Mayaro virus);EEEV:东方马脑炎病毒(Eastern Equine Encephalitis Virus);VEEV:委内瑞拉马脑炎病毒(Venezuelan Equine Encephalitis Virus)。Figure 24: The position of the mutated amino acid in the macrodomain of saRNA and the comparison results of the amino acid sequences of different virus macrodomains. Macro is the macrodomain, AUD is the alphavirus-unique domain, HVD is the hypervariable domain, different shapes represent different mutations, and the amino acids with more than 50% homology in the amino acid comparison results are marked with shadows. The full names of the viruses listed in the figure are as follows: SARS-CoV: Severe Acute Respiratory Syndrome Coronavirus; SARS-CoV-2: Severe Acute Respiratory Syndrome Coronavirus 2; MERS-CoV: Middle East Respiratory Syndrome Coronavirus; SFV: Semliki Forest Virus; CHIKV: Chikungunya Virus; SINV: Sindbis Virus; MAYV: Mayaro virus; EEEV: Eastern Equine Encephalitis Virus; VEEV: Venezuelan Equine Encephalitis Virus.
图25:saRNA宏结构域的蛋白结构示意图。箭头标记的位置分别为与宏结构域结合的ADP核糖、第48位谷氨酰胺(缩写Q)和第113位异亮氨酸(缩写I)。Figure 25: Schematic diagram of the protein structure of the saRNA macrodomain. The positions marked by arrows are ADP ribose, glutamine at position 48 (abbreviated Q), and isoleucine at position 113 (abbreviated I) bound to the macrodomain.
图26:宏结构域氨基酸突变对细胞总蛋白ADP-核糖基化影响的免疫印迹检测结果。Mock为模拟转染组,仅加入转染试剂,并未转染任何RNA分子。GAPDH蛋白作为细胞蛋白内参。最下方数值为各组相对于对照组的总蛋白ADP-核糖基化水平比例。Figure 26: Western blot test results of the effect of macrodomain amino acid mutations on total cellular protein ADP-ribosylation. Mock is a simulated transfection group, in which only transfection reagent was added without transfection of any RNA molecules. GAPDH protein was used as a cellular protein internal reference. The bottom value is the ratio of total protein ADP-ribosylation level of each group relative to the control group.
图27:突变saRNA在hela细胞中亚基因组RNA复制能力的比较。各组之间的显著性差异利用双因素方差进行统计分析。****和^^^^分别代表WT-saRNA与190-saRNA或191-saRNA组的P值小于0.0001。Figure 27: Comparison of mutant saRNA subgenomic RNA replication capacity in hela cells. The significant differences between the groups were statistically analyzed using two-way ANOVA. **** and ^^^^ represent that the P values of WT-saRNA and 190-saRNA or 191-saRNA groups are less than 0.0001, respectively.
图28:突变saRNA在hela细胞复制过程中双链RNA形成能力的比较。图28a为转染后24小时的流式细胞检测结果示意图,图28b为双链RNA的比例统计柱状图。Figure 28: Comparison of the ability of mutant saRNA to form double-stranded RNA during hela cell replication. Figure 28a is a schematic diagram of the flow cytometry results 24 hours after transfection, and Figure 28b is a statistical bar graph of the proportion of double-stranded RNA.
图29:不同saRNA转染hela细胞24小时后转录组水平的主成分分析图。 Figure 29: Principal component analysis of transcriptome levels 24 hours after Hela cells were transfected with different saRNAs.
图30:不同saRNA相对于对照组中上调或下调基因数的维恩统计图。FIG. 30 : Venn diagram of the number of genes upregulated or downregulated in different saRNAs relative to the control group.
图31:野生型saRNA相对于对照组中,差异基因聚类分析的山脊图。图31a为对差异基因的生物学功能聚类分析;图31b为对差异基因的分子功能聚类分析;横坐标为变化倍数的log2值,绿色代表调整后的P值,蓝色点代表每一个下调基因,红色点代表每一个上调基因。Figure 31: Ridge plot of differential gene cluster analysis in wild-type saRNA relative to the control group. Figure 31a is a biological function cluster analysis of differential genes; Figure 31b is a molecular function cluster analysis of differential genes; the horizontal axis is the log2 value of the change fold, green represents the adjusted P value, blue dots represent each down-regulated gene, and red dots represent each up-regulated gene.
图32:不同saRNA转染后,干扰素通路转录组水平的基因热图。Figure 32: Heat map of genes at the transcriptome level of the interferon pathway after transfection with different saRNAs.
图33:不同saRNA转染hela细胞2、6、24小时后,天然免疫通路相关基因的荧光定量PCR验证结果。Figure 33: Fluorescence quantitative PCR verification results of genes related to the innate immune pathway 2, 6, and 24 hours after HeLa cells were transfected with different saRNAs.
图34:不同saRNA转染hela细胞2、6、24和48小时后的荧光酶素表达水平。Figure 34: Luciferase expression levels after HeLa cells were transfected with different saRNAs for 2, 6, 24 and 48 hours.
图35:不同saRNA转染2、6、24小时后细胞翻译活性免疫印迹检测结果。GAPDH作为内参。Figure 35: Western blot detection results of cell translation activity after 2, 6, and 24 hours of transfection with different saRNAs. GAPDH was used as an internal reference.
图36:不同saRNA转染hela细胞2、6、24和48小时后核糖体RNA的完整性检测。图36a为利用毛细管电泳检测转染saRNA 24小时后的核糖体RNA完整性示意图,箭头标记发生降解的核糖体RNA。图36b为不同时间核糖体RNA完整性的检测统计结果。Figure 36: Detection of ribosomal RNA integrity after 2, 6, 24 and 48 hours of transfection of Hela cells with different saRNAs. Figure 36a is a schematic diagram of the detection of ribosomal RNA integrity 24 hours after transfection of saRNA using capillary electrophoresis, with arrows marking degraded ribosomal RNA. Figure 36b is the statistical results of the detection of ribosomal RNA integrity at different times.
图37:不同saRNA转染6和24小时后的eIF2α磷酸化水平检测。peIF2α为发生磷酸化的eIF2α,总eIF2α蛋白及GAPDH作为内参。Figure 37: Detection of eIF2α phosphorylation level after 6 and 24 hours of transfection with different saRNAs. peIF2α is phosphorylated eIF2α, and total eIF2α protein and GAPDH are used as internal references.
图38:不同saRNA转染后24小时的荧光显微图像,红色箭头标记了发生核皱缩的细胞及其EGFP表达。DAPI表示经染色的细胞核,EGFP代表表达EGFP的细胞。Figure 38: Fluorescence microscopy images 24 hours after transfection with different saRNAs, red arrows mark cells with nuclear shrinkage and their EGFP expression. DAPI represents stained cell nuclei, and EGFP represents cells expressing EGFP.
图39:不同saRNA转染24和48小时后,未发生凋亡的活细胞比例统计结果。Figure 39: Statistical results of the proportion of live cells that did not undergo apoptosis 24 and 48 hours after transfection with different saRNAs.
图40:不同saRNA转染24小时后,发生切割的活化caspase 8和caspase 3蛋白检测结果,未切割的全长caspase作为对照。GAPDH作为内参。Figure 40: Detection results of activated caspase 8 and caspase 3 proteins that were cleaved 24 hours after transfection with different saRNAs, with the uncleaved full-length caspase as a control. GAPDH was used as an internal reference.
图41:Caspase抑制剂处理或未处理的野生型saRNA转染的hela细胞,其细胞凋亡及EGFP表达强度检测。Figure 41: Detection of cell apoptosis and EGFP expression intensity in Hela cells transfected with wild-type saRNA treated or not with Caspase inhibitors.
图42:不同mRNA-LNP体内表达水平检测示意图。图42a为检测流程示意图,D0为免疫时间。图42b为免疫后第1、3、7天小鼠活体成像荧光图。Figure 42: Schematic diagram of in vivo expression level detection of different mRNA-LNPs. Figure 42a is a schematic diagram of the detection process, D0 is the immunization time. Figure 42b is a fluorescent image of in vivo imaging of mice on days 1, 3, and 7 after immunization.
图43:mRNA-LNP免疫后小鼠体内荧光酶素的表达水平时间曲线。Figure 43: Time curve of luciferase expression level in mice after mRNA-LNP immunization.
图44:mRNA-LNP免疫后小鼠后体内表达水平的曲线下面积统计结果。 Figure 44: Statistical results of the area under the curve of in vivo expression levels in mice after mRNA-LNP immunization.
发明详述Detailed description of the invention
如本文所用,术语“自扩增RNA”或“saRNA”或“sa-mRNA”可交换使用,是指一种编码有RNA复制酶结构域序列从而在进入细胞后具备自我复制扩增的能力的mRNA。saRNA与mRNA类似,通常包含5’UTR、3’UTR和poly(A)尾巴,但另外还包含源于病毒(例如甲病毒)的非结构蛋白序列和位于目的蛋白编码序列上游的亚基因组启动子(Subgenomic promoter,sgPr)。saRNA是正链RNA分子,进入细胞后首先被核糖体翻译出若干个非结构蛋白组分并组装成RNA复制酶。RNA复制酶首先利用初次进入细胞的saRNA合成saRNA负链,再以负链为模版合成新的saRNA拷贝,从而实现saRNA的自扩增。同时,RNA复制酶也识别sgPr,然后自其下游开始合成亚基因组RNA,这些亚基因组RNA在宿主细胞中大量累积。在saRNA编码的目的蛋白是抗原蛋白时(例如用作疫苗),亚基因组RNA编码的抗原基因将翻译出大量的抗原分子并触发细胞抗原呈递。本文中所述的saRNA既包括不含目的蛋白编码序列的saRNA骨架序列,也包括将saRNA骨架序列与目的蛋白编码序列可操作性连接的saRNA。As used herein, the terms "self-amplifying RNA" or "saRNA" or "sa-mRNA" are used interchangeably and refer to an mRNA that encodes an RNA replicase domain sequence and thus has the ability to self-replicate and amplify after entering a cell. saRNA is similar to mRNA and generally contains a 5'UTR, a 3'UTR, and a poly(A) tail, but additionally contains a non-structural protein sequence derived from a virus (e.g., an alphavirus) and a subgenomic promoter (sgPr) located upstream of the target protein coding sequence. saRNA is a positive-strand RNA molecule that is first translated by the ribosome into several non-structural protein components and assembled into RNA replicase after entering the cell. RNA replicase first uses the saRNA that first enters the cell to synthesize the saRNA negative strand, and then uses the negative strand as a template to synthesize new saRNA copies, thereby achieving self-amplification of saRNA. At the same time, RNA replicase also recognizes sgPr, and then begins to synthesize subgenomic RNA from its downstream, which accumulates in large quantities in the host cell. When the target protein encoded by saRNA is an antigen protein (for example, used as a vaccine), the antigen gene encoded by the subgenomic RNA will translate a large number of antigen molecules and trigger cellular antigen presentation. The saRNA described herein includes both saRNA backbone sequences without a target protein coding sequence and saRNA that operably connects the saRNA backbone sequence to the target protein coding sequence.
如本文所用,术语“宏结构域(macrodomain)”是指由甲病毒、冠状病毒、戊肝病毒等病毒编码的一种保守蛋白质结构域,其能够识别并去除ADP核糖基化,因此又被视为ADP核糖基水解酶。多项研究表明,病毒宏结构域中ADP核糖水解活性的减弱将导致病毒复制能力减弱,因此宏结构域是近期抗病毒药物开发的潜在热点之一。在以VEEV为代表的甲病毒家族中,宏结构域存在于非结构蛋白3(nonstructural protein 3)的N端,如图24所示。宏结构域氨基酸序列在不同病毒之间都较为保守,同源性超过50%。As used herein, the term "macrodomain" refers to a conserved protein domain encoded by viruses such as alphaviruses, coronaviruses, and hepatitis E virus, which can recognize and remove ADP ribosylation and is therefore considered an ADP ribosyl hydrolase. Many studies have shown that the reduction of ADP ribose hydrolysis activity in the viral macrodomain will lead to a reduction in the ability of the virus to replicate, so the macrodomain is one of the potential hotspots for the development of antiviral drugs in the near future. In the alphavirus family represented by VEEV, the macrodomain is present at the N-terminus of nonstructural protein 3, as shown in Figure 24. The amino acid sequence of the macrodomain is relatively conserved among different viruses, with a homology of more than 50%.
如本文所用,术语“RNA复制酶”又称为依赖于RNA的RNA复制酶(RdRp),是RNA聚合酶的一种,其能够以RNA为模板合成RNA。它存在于大部分RNA病毒中,起到复制病毒RNA以及合成mRNA的作用,是除逆转录病毒外的其他RNA病毒和类病毒复制所必需的酶。应用于saRNA的RNA复制酶通常来源于甲病毒且包含非结构蛋白1、2、3和4(nsP1、nsP2、nsP3和nsP4)。As used herein, the term "RNA replicase" is also called RNA-dependent RNA replicase (RdRp), which is a type of RNA polymerase that can synthesize RNA using RNA as a template. It is present in most RNA viruses and plays a role in replicating viral RNA and synthesizing mRNA. It is an enzyme required for the replication of other RNA viruses and viroids except retroviruses. The RNA replicase used for saRNA is usually derived from alphaviruses and contains nonstructural proteins 1, 2, 3, and 4 (nsP1, nsP2, nsP3, and nsP4).
术语“亚基因组启动子”(Subgenomic Promoter,简称为SGP)是指核酸序列(例如编码序列)上游(5’端)的核酸序列,其通过提供对于RNA聚合酶,通常是RNA依赖性RNA聚合酶,特别是功能性甲病毒非结构蛋白的识别和结构位点来控制所述 核酸序列的转录。SGP可包含对于另外的因子的另外的识别或结合位点。亚基因组启动子通常是正链RNA病毒(例如甲病毒)的遗传元件。甲病毒的亚基因组启动子是病毒基因组RNA中包含的核酸序列。亚基因组启动子的一般特征在于,其允许在RNA依赖性RNA聚合酶(例如功能性甲病毒非结构蛋白)的存在下开始转录(RNA合成)。RNA(-)链(即甲病毒基因组RNA的互补链)充当用于合成(+)链亚基因组转录物的模板,并且(+)链亚基因组转录物的合成通常在亚基因组启动子处或附近开始。The term "subgenomic promoter" (SGP) refers to a nucleic acid sequence upstream (5' end) of a nucleic acid sequence (e.g., a coding sequence) that controls the expression of a nucleic acid sequence by providing recognition and structural sites for RNA polymerase, usually RNA-dependent RNA polymerase, and in particular functional alphavirus nonstructural proteins. The subgenomic promoter is a genetic element of a positive-strand RNA virus (e.g., an alphavirus). The subgenomic promoter of an alphavirus is a nucleic acid sequence contained in the viral genomic RNA. The general feature of a subgenomic promoter is that it allows transcription (RNA synthesis) to begin in the presence of an RNA-dependent RNA polymerase (e.g., a functional alphavirus nonstructural protein). The RNA (-) strand (i.e., the complementary strand of the alphavirus genomic RNA) serves as a template for the synthesis of a (+) strand subgenomic transcript, and the synthesis of a (+) strand subgenomic transcript typically begins at or near the subgenomic promoter.
如本文所用,术语“亚基因组”是指长度或大小小于其来源的基因组核苷酸序列的核苷酸序列(如RNA或DNA)。例如,亚基因组可以是编码VEEV结构蛋白的区域,亚基因组RNA可以使用内部亚基因组启动子从亚基因组转录,其序列位于基因组病毒RNA或其补体中。亚基因组的转录可以由与宿主细胞编码的蛋白质(例如nsP1-4)相关的病毒编码聚合酶介导。As used herein, the term "subgenomic" refers to a nucleotide sequence (such as RNA or DNA) that is smaller in length or size than the genomic nucleotide sequence from which it is derived. For example, a subgenomic region may be a region encoding a VEEV structural protein, and a subgenomic RNA may be transcribed from a subgenomic region using an internal subgenomic promoter, the sequence of which is located in the genomic viral RNA or its complement. Transcription of the subgenomic region may be mediated by a virally encoded polymerase associated with a host cell-encoded protein (e.g., nsP1-4).
甲病毒结构蛋白(核心核衣壳蛋白C、包膜蛋白E2和包膜蛋白E1,病毒颗粒的所有成分)通常由处于亚基因组启动子控制下的一个单一开放阅读框编码(Strauss&Strauss,Microbiol.Rev.,1994,vol.58,pp.491-562)。亚基因组启动子被顺式作用的甲病毒非结构蛋白识别。特别地,甲病毒复制酶使用基因组RNA的(-)链互补链作为模板来合成(+)链亚基因组转录物。(+)链亚基因组转录物编码甲病毒结构蛋白(Kim et al.,2004,Virology,vol.323,pp.153-163,Vasiljeva et al.,2003,J.Biol.Chem.vol.278,pp.41636-41645)。亚基因组RNA转录物充当模板,用于翻译编码作为一种多蛋白的结构蛋白的开放阅读框,并且多蛋白(poly-protein)被切割以产生结构蛋白。在宿主细胞中甲病毒感染的后期,位于nsP2编码序列内的包装信号确保将基因组RNA选择性包装到由结构蛋白包装的出芽病毒体(budding viron)中(White et al.,1998,J.Virol.,vol.72,pp.4320-4326)。Alphavirus structural proteins (core nucleocapsid protein C, envelope protein E2 and envelope protein E1, all components of the virus particle) are usually encoded by a single open reading frame under the control of a subgenomic promoter (Strauss & Strauss, Microbiol. Rev., 1994, vol. 58, pp. 491-562). The subgenomic promoter is recognized by cis-acting alphavirus non-structural proteins. In particular, the alphavirus replicase uses the (-) strand complementary strand of the genomic RNA as a template to synthesize the (+) strand subgenomic transcript. The (+) strand subgenomic transcript encodes alphavirus structural proteins (Kim et al., 2004, Virology, vol. 323, pp. 153-163, Vasiljeva et al., 2003, J. Biol. Chem. vol. 278, pp. 41636-41645). The subgenomic RNA transcript serves as a template for translation of an open reading frame encoding a structural protein as a polyprotein, and the polyprotein is cleaved to produce the structural protein. At the late stage of alphavirus infection in host cells, a packaging signal located within the nsP2 coding sequence ensures the selective packaging of the genomic RNA into budding virions packaged by structural proteins (White et al., 1998, J. Virol., vol. 72, pp. 4320-4326).
本发明中,术语“甲病毒复制必需序列”是可被甲病毒复制酶识别并启动后续RNA转录及复制的序列,其截取于甲病毒复制酶序列。甲病毒复制必需序列必须含有一段最短序列(SEQ ID NO:54所示),可以在最短序列的基础上增加或调整长度。In the present invention, the term "sequence essential for alphavirus replication" is a sequence that can be recognized by alphavirus replicase and initiate subsequent RNA transcription and replication, which is intercepted from the alphavirus replicase sequence. The sequence essential for alphavirus replication must contain a minimum sequence (shown in SEQ ID NO:54), and the length can be increased or adjusted based on the shortest sequence.
本发明中,术语“microRNA成熟序列”是指会形成发夹结构的microRNA前体(pre-miRNA)经过Dicer酶加工后形成的microRNA分子,microRNA前体两个臂分别产生一条有功能的成熟microRNA,它们分别靶向不同的位点。一般以“-5p”(或5p)和“-3p”(或3p)分别命名,如hsa-miR-21-5p和hsa-miR-21-3p,分别表明从hsa- mir-21前体的5’端臂和3’端臂加工而来的,从序列上来看,5p和3p大部分序列互补配对。In the present invention, the term "microRNA mature sequence" refers to a microRNA molecule formed after the microRNA precursor (pre-miRNA) that forms a hairpin structure is processed by the Dicer enzyme. The two arms of the microRNA precursor each produce a functional mature microRNA, which targets different sites. They are generally named "-5p" (or 5p) and "-3p" (or 3p), such as hsa-miR-21-5p and hsa-miR-21-3p, respectively, indicating that the mature microRNA sequence is derived from hsa- The 5' and 3' arms of the mir-21 precursor are processed. From the sequence point of view, most of the sequences of 5p and 3p are complementary.
本发明中,如果没有特别说明,术语“完整性”是指RNA体外转录反应完成后,所产生的目标RNA产物的量占所产生的总RNA产物(包含目标RNA产物及非预期RNA产物)的量的百分比;目标RNA产物的量和总RNA产物的量可以通过电泳方法测定。例如,如果通过电泳方法测定的目标saRNA产物的量占所有RNA产物的量的80%,则saRNA完整性为80%。In the present invention, if not otherwise specified, the term "integrity" refers to the percentage of the amount of the target RNA product produced after the RNA in vitro transcription reaction is completed (including the target RNA product and the unexpected RNA product) produced; the amount of the target RNA product and the amount of the total RNA product can be determined by electrophoresis. For example, if the amount of the target saRNA product determined by the electrophoresis method accounts for 80% of the amount of all RNA products, the saRNA integrity is 80%.
本发明中,如果没有特别说明,所述“第一”(例如第一mRNA分子、第一DNA分子等)和“第二”(例如第二mRNA分子、第二DNA分子等)仅仅是为了指代上的区分,并不没有特别的次序上的含义。In the present invention, unless otherwise specified, the "first" (e.g., the first mRNA molecule, the first DNA molecule, etc.) and the "second" (e.g., the second mRNA molecule, the second DNA molecule, etc.) are merely for reference distinction and do not have any particular order meaning.
本发明,术语“有效量”是指可在受试者中实现预防或治疗本发明所述的适应症或者症状,或减轻和/或缓解所述适应症或者症状的本发明中任一项所述的mRNA分子、本发明的mRNA分子组合、本发明中任一项所述的DNA分子或者本发明中任一项所述的saRNA分子的量。In the present invention, the term "effective amount" refers to the amount of the mRNA molecule of any one of the present invention, the mRNA molecule combination of the present invention, the DNA molecule of any one of the present invention, or the saRNA molecule of any one of the present invention that can prevent or treat the indications or symptoms described in the present invention, or reduce and/or alleviate the indications or symptoms in a subject.
mRNA疗法mRNA Therapeutics
基于mRNA技术的疗法,是将体外合成的mRNA递送到人体中的特定细胞,mRNA在细胞质中被翻译成所需的蛋白质。mRNA作为疫苗或药物,可应用于预防传染病、治疗肿瘤和蛋白替代疗法。mRNA药物靶点丰富,不受靶点蛋白成药性,胞内/外限制。mRNA序列设计简便,利用患者自身细胞产生分子,绕过化学合成中的挑战,且自身诱导产生的分子药效更强。mRNA生产平台具有较强的延展性和可复制性,易于规模化生产。Therapies based on mRNA technology deliver in vitro synthesized mRNA to specific cells in the human body, where it is translated into the desired protein in the cytoplasm. As a vaccine or drug, mRNA can be used to prevent infectious diseases, treat tumors, and for protein replacement therapy. mRNA drugs have rich targets and are not restricted by the drugability of the target protein or intracellular/extracellular restrictions. The mRNA sequence is easy to design, and the patient's own cells are used to produce molecules, bypassing the challenges of chemical synthesis, and the self-induced molecules have stronger efficacy. The mRNA production platform has strong scalability and replicability, and is easy to scale up.
(1)传染病领域预防性疫苗(1) Preventive vaccines for infectious diseases
针对传染性疾病的mRNA疫苗编码相关病原体的抗原,注射后可在体内表达出特异性的抗原,可同时诱导细胞免疫和体液免疫,刺激产生相应抗体和免疫细胞,用以预防相应病原体。mRNA vaccines targeting infectious diseases encode antigens of related pathogens. After injection, they can express specific antigens in the body, induce both cellular immunity and humoral immunity, and stimulate the production of corresponding antibodies and immune cells to prevent the corresponding pathogens.
(2)肿瘤治疗性疫苗/药物(2) Tumor therapeutic vaccines/drugs
将编码肿瘤特异性抗原靶点的mRNA通过特定的方式递送到体内,使这些肿瘤特异性抗原翻译并呈递到细胞表面,进而激活免疫系统,使其能特异性识别并杀伤肿 瘤细胞。The mRNA encoding tumor-specific antigen targets is delivered into the body in a specific way, so that these tumor-specific antigens are translated and presented on the cell surface, thereby activating the immune system, enabling it to specifically recognize and kill tumors. Tumor cells.
(3)蛋白替代疗法(3) Protein replacement therapy
通常情况下,蛋白质替代疗法用于治疗罕见的单基因疾病,旨在恢复酶的功能。蛋白质合成困难,给药存在诸多难题且价格昂贵,而利用mRNA将人体变成自身蛋白质的加工厂,理论上是一种经济可行并且高效的方式。目前基于mRNA的蛋白替代疗法主要聚焦于遗传性代谢疾病。mRNA理论上可以合成任意一种蛋白,可作为蛋白质补充或替代疗法治疗多种疾病。但由于需要mRNA靶向表达和重复给药,甚至是全身给药,对安全性要求更高。Typically, protein replacement therapy is used to treat rare monogenic diseases and aims to restore enzyme function. Protein synthesis is difficult, administration presents many challenges and is expensive, and using mRNA to turn the human body into its own protein processing plant is theoretically an economically feasible and efficient way. Currently, mRNA-based protein replacement therapy mainly focuses on inherited metabolic diseases. mRNA can theoretically synthesize any protein and can be used as a protein supplement or replacement therapy to treat a variety of diseases. However, due to the need for targeted expression of mRNA and repeated administration, or even systemic administration, higher safety requirements are required.
自扩增RNASelf-amplifying RNA
本发明构建了一种改进的saRNA,所述saRNA在所编码的复制酶的宏结构域中包含特定突变,从而使得该saRNA具有降低的ADP核糖水解活性。同时,该突变使得所述sgRNA编码的复制酶的复制能力适度减弱,从而导致saRNA复制中所形成的双链RNA的比例下降。由于双链RNA是saRNA复制过程中的副产物,且经常被细胞中的的模式识别受体识别而激发细胞的先天免疫反应,因此双链RNA的减少会减少对天然免疫受体的激活,从而使得saRNA诱导的天然免疫反应强度适当地降低。如实施例中验证的,相比于原始的野生型saRNA,包含突变的sgRNA受到的翻译抑制显著减弱,同时细胞凋亡明显减少,因此突变后saRNA表达效率明显提升。相应地,这种改进的sgRNA可包含编码各种目的蛋白(例如抗原蛋白)的核苷酸序列,可获得在体内有效诱导特异性免疫的saRNA,并制备成传染病及肿瘤疫苗。The present invention constructs an improved saRNA, wherein the saRNA comprises a specific mutation in the macrodomain of the encoded replicase, so that the saRNA has a reduced ADP ribose hydrolysis activity. At the same time, the mutation moderately weakens the replication ability of the replicase encoded by the sgRNA, thereby causing a decrease in the proportion of double-stranded RNA formed in the replication of saRNA. Since double-stranded RNA is a byproduct in the replication process of saRNA, and is often recognized by pattern recognition receptors in cells to stimulate the innate immune response of cells, the reduction of double-stranded RNA reduces the activation of natural immune receptors, thereby appropriately reducing the intensity of the natural immune response induced by saRNA. As verified in the embodiment, compared to the original wild-type saRNA, the translation inhibition of the sgRNA containing mutations is significantly weakened, and cell apoptosis is significantly reduced, so the expression efficiency of saRNA after mutation is significantly improved. Accordingly, this improved sgRNA may include nucleotide sequences encoding various target proteins (e.g., antigenic proteins), and saRNA that effectively induces specific immunity in vivo can be obtained, and prepared into infectious diseases and tumor vaccines.
在又一个方面,本申请通过对saRNA编码的复制酶蛋白结构域中的宏结构域进行氨基酸突变,减弱了ADP核糖水解活性,从而降低saRNA的复制效率,进而相比于常规的saRNA降低了免疫原性,并实现表达效率的提升。因此,本发明提供了一种突变宏结构域以及编码该突变宏结构域的核酸分子(如mRNA分子或DNA分子)。本发明还提供了包含该突变宏结构域的复制酶以及编码该复制酶的核酸分子(如mRNA分子或DNA分子)。本发明还进一步提供了编码该复制酶的核酸序列与编码目的基因的序列组合构建的核酸分子(如saRNA分子或DNA分子)。In another aspect, the present application reduces the ADP ribose hydrolysis activity by amino acid mutation of the macro domain in the replicase protein domain encoded by saRNA, thereby reducing the replication efficiency of saRNA, thereby reducing the immunogenicity compared to conventional saRNA, and achieving the improvement of expression efficiency. Therefore, the present invention provides a mutation macro domain and a nucleic acid molecule (such as an mRNA molecule or a DNA molecule) encoding the mutation macro domain. The present invention also provides a replicase comprising the mutation macro domain and a nucleic acid molecule (such as an mRNA molecule or a DNA molecule) encoding the replicase. The present invention further provides a nucleic acid molecule (such as a saRNA molecule or a DNA molecule) constructed by combining a nucleic acid sequence encoding the replicase with a sequence encoding a target gene.
在一些实施方案中,本文公开的saRNA包含编码RNA复制酶的核苷酸序列,其中所述RNA复制酶包含突变宏结构域,相比于来自VEEV的野生型宏结构域(如SEQ ID NO:55所示),所述突变宏结构域具有Q48P或I113F取代(编号按照SEQ ID NO:55)。具体地,所述saRNA编码的突变宏结构域可包含如SEQ ID NO:57或SEQ ID NO:59所示的氨基酸序列。更具体地,所述saRNA可包含如SEQ ID NO:68或SEQ ID NO:70所示的编码突变宏结构域的核苷酸序列。所述复制酶可以是甲病毒复制酶,例如包含甲病毒非结构蛋白1、2、3和4。In some embodiments, the saRNA disclosed herein comprises a nucleotide sequence encoding an RNA replicase, wherein the RNA replicase comprises a mutant macrodomain having a Q48P or I113F substitution compared to the wild-type macrodomain from VEEV (as shown in SEQ ID NO: 55) (numbering according to SEQ ID NO: 56). NO:55). Specifically, the mutant macrodomain encoded by the saRNA may comprise an amino acid sequence as shown in SEQ ID NO:57 or SEQ ID NO:59. More specifically, the saRNA may comprise a nucleotide sequence encoding a mutant macrodomain as shown in SEQ ID NO:68 or SEQ ID NO:70. The replicase may be an alphavirus replicase, for example, comprising alphavirus nonstructural proteins 1, 2, 3 and 4.
在一些实施方案中,本文公开的saRNA包含编码来源于甲病毒的复制酶组成蛋白nsP1、nsP2、nsP3和nsP4的核苷酸序列,所述四种复制酶组成蛋白能够组装成RNA复制酶复合物,其中所述突变宏结构域位于nsP3的N端。四个非结构蛋白组分nsP1、nsP2、nsP3和nsP4以多聚蛋白的形式组装成一个RNA复制酶复合物。nsP1-4均具有其特有的功能,其中nsP4扮演着以RNA为模版的RNA聚合酶的角色。saRNA编码的复制酶组成蛋白的序列可来源于例如委内瑞拉马脑炎病毒(VEEV)和森林脑炎病毒(SFV)等。在一些实施方案中,至少一种非结构复制酶的结构域序列包含选自第IV组RNA病毒的序列,所述病毒包括东部马脑炎病毒(EEEV)、委内瑞拉马脑炎病毒(VEEV)、大沼泽地病毒、Mucambo病毒、Pixuna病毒、西部马脑炎病毒(WEE)、Sindbis病毒、森林脑炎病毒(SFV)等。在一些实施方案中,所述形成复制酶的多个非结构蛋白序列来源于甲病毒例如VEEV。具体地,所述saRNA可包含如SEQ ID NO:67或SEQ ID NO:69所示的编码复制酶组成蛋白的核苷酸序列。In some embodiments, the saRNA disclosed herein comprises nucleotide sequences encoding replicase constituent proteins nsP1, nsP2, nsP3 and nsP4 derived from alphavirus, wherein the four replicase constituent proteins are capable of assembling into an RNA replicase complex, wherein the mutant macrodomain is located at the N-terminus of nsP3. The four non-structural protein components nsP1, nsP2, nsP3 and nsP4 are assembled into an RNA replicase complex in the form of a polyprotein. nsP1-4 all have their unique functions, wherein nsP4 plays the role of an RNA polymerase with RNA as a template. The sequence of the replicase constituent protein encoded by the saRNA may be derived from, for example, Venezuelan equine encephalitis virus (VEEV) and forest encephalitis virus (SFV), etc. In some embodiments, the domain sequence of at least one non-structural replicase comprises a sequence selected from Group IV RNA viruses, including Eastern equine encephalitis virus (EEEV), Venezuelan equine encephalitis virus (VEEV), Everglades virus, Mucambo virus, Pixuna virus, Western equine encephalitis virus (WEE), Sindbis virus, forest encephalitis virus (SFV), etc. In some embodiments, the plurality of non-structural protein sequences forming the replicase are derived from an alphavirus such as VEEV. Specifically, the saRNA may comprise a nucleotide sequence encoding a replicase constituent protein as shown in SEQ ID NO: 67 or SEQ ID NO: 69.
尽管天然甲病毒基因组除了非结构复制酶外还编码结构蛋白,但在一些实施方案中,本发明的基于甲病毒的saRNA不编码甲病毒结构蛋白。Although the native alphavirus genome encodes structural proteins in addition to the non-structural replicase, in some embodiments, the alphavirus-based saRNA of the present invention does not encode alphavirus structural proteins.
在一些实施方案中,除了编码四种复制酶蛋白nsP1、nsP2、nsP3和nsP4的核苷酸序列以外,所述saRNA还包含5’端非翻译区、3’端非翻译区和poly(A)尾,其中所述编码四种复制酶蛋白nsP1、nsP2、nsP3和nsP4的核苷酸序列位于5’端非翻译区和3’端非翻译区之间。本领域技术人员将容易地理解,5’UTR、3’UTR和poly(A)序列不限于本文示例的那些,常规用于mRNA构建的序列均可用于本发明的saRNA。所述5’端非翻译区可来源于多种病毒,例如甲病毒、冠状病毒。在一些实施方案中,所述5’端非翻译区来自委内瑞拉马脑脊髓炎病毒。具体地,所述5’端非翻译区可包含如SEQ ID NO:63所示的核苷酸序列。所述3’端非翻译区可来源于多种病毒,例如甲病毒、冠状病毒。在一些实施方案中,所述3’端非翻译区来自委内瑞拉马脑脊髓炎病毒。具体地,所述3’端非翻译区可包含如SEQ ID NO:64所示的核苷酸序列。在一些实施方案中,poly(A)序列包含20-200个腺苷酸。poly(A)序列内部或3’端可包含限制性酶切 位点。In some embodiments, in addition to the nucleotide sequences encoding the four replicase proteins nsP1, nsP2, nsP3 and nsP4, the saRNA further comprises a 5' untranslated region, a 3' untranslated region and a poly (A) tail, wherein the nucleotide sequences encoding the four replicase proteins nsP1, nsP2, nsP3 and nsP4 are located between the 5' untranslated region and the 3' untranslated region. It will be readily understood by those skilled in the art that the 5'UTR, 3'UTR and poly (A) sequences are not limited to those exemplified herein, and sequences conventionally used for mRNA construction can be used for the saRNA of the present invention. The 5' untranslated region may be derived from a variety of viruses, such as alphaviruses and coronaviruses. In some embodiments, the 5' untranslated region is from Venezuelan equine encephalitis virus. Specifically, the 5' untranslated region may comprise a nucleotide sequence as shown in SEQ ID NO: 63. The 3' untranslated region may be derived from a variety of viruses, such as alphaviruses and coronaviruses. In some embodiments, the 3' untranslated region is from Venezuelan equine encephalitis virus. Specifically, the 3' untranslated region may include a nucleotide sequence as shown in SEQ ID NO: 64. In some embodiments, the poly(A) sequence includes 20-200 adenylic acids. The poly(A) sequence may include a restriction enzyme cleavage site inside or at the 3' end. Location.
在一些实施方案中,所述saRNA还包含编码目的蛋白的核苷酸序列或包含目的基因(GOI)编码序列,所述目的蛋白是或者目的基因编码的是选自以下的任一种:治疗多肽、预防多肽、诊断多肽、报告基因、抗原或编码调节结构的基因。例如,目的蛋白可以是传染病抗原、过敏性抗原或肿瘤抗原。或者,目的基因可以编码一种非编码基因,如siRNA、microRNA、gRNA等。In some embodiments, the saRNA further comprises a nucleotide sequence encoding a target protein or a target gene (GOI) coding sequence, wherein the target protein is or the target gene encodes any one selected from the following: a therapeutic polypeptide, a preventive polypeptide, a diagnostic polypeptide, a reporter gene, an antigen, or a gene encoding a regulatory structure. For example, the target protein can be an infectious disease antigen, an allergic antigen, or a tumor antigen. Alternatively, the target gene can encode a non-coding gene, such as siRNA, microRNA, gRNA, etc.
在一些实施方案中,所述saRNA进一步包含编码一种或多种抗原蛋白的核苷酸序列以用作疫苗。所述抗原蛋白可以是病毒抗原蛋白。In some embodiments, the saRNA further comprises a nucleotide sequence encoding one or more antigenic proteins for use as a vaccine. The antigenic protein can be a viral antigenic protein.
在一些实施方案中,所述saRNA进一步包含编码一种或多种治疗蛋白或免疫调控剂的核苷酸序列以用作疾病治疗剂。所述免疫调控剂可以是细胞因子、趋化因子或其他免疫刺激剂或抑制剂。In some embodiments, the saRNA further comprises a nucleotide sequence encoding one or more therapeutic proteins or immunomodulators for use as disease therapeutic agents. The immunomodulators can be cytokines, chemokines or other immunostimulants or inhibitors.
在一些实施方案中,本文公开的saRNA包含两个表达单元,其中第一表达单元编码RNA复制酶的多个非结构结构域序列,第二表达单元编码与亚基因组启动子可操作性地连接的目的蛋白,第一和第二表达单元可操作性地连接并且第一表达单元位于第二表达单元的5’端。在一些实施方案中,所述亚基因组启动子是26S启动子,例如如SEQ ID NO:65所示。In some embodiments, the saRNA disclosed herein comprises two expression units, wherein the first expression unit encodes multiple nonstructural domain sequences of an RNA replicase, the second expression unit encodes a protein of interest operably linked to a subgenomic promoter, the first and second expression units are operably linked and the first expression unit is located at the 5' end of the second expression unit. In some embodiments, the subgenomic promoter is a 26S promoter, such as shown in SEQ ID NO: 65.
因此,本公开的saRNA具有至少两个编码区,第一编码区编码多个非结构复制酶结构域序列;第二个编码区编码与亚基因组启动子可操作连接的目的基因。Therefore, the saRNA of the present disclosure has at least two coding regions, the first coding region encodes multiple non-structural replicase domain sequences; the second coding region encodes a gene of interest operably linked to a subgenomic promoter.
更具体地,本文公开的saRNA可包含以下5’至3’的可操作连接的核酸序列:5’UTR-nsP-SGP-GOI-3’UTR-Poly A,More specifically, the saRNA disclosed herein may comprise the following 5' to 3' operably linked nucleic acid sequence: 5'UTR-nsP-SGP-GOI-3'UTR-Poly A,
更具体地,本文公开的saRNA可包含以下从5’至3’可操作连接的核酸序列:5’UTR-nsP1-nsP2-nsP3-nsP4-SGP-GOI-3’UTR-Poly A,More specifically, the saRNA disclosed herein may comprise the following nucleic acid sequence operably linked from 5' to 3': 5'UTR-nsP1-nsP2-nsP3-nsP4-SGP-GOI-3'UTR-Poly A,
其中,5’UTR是5’非翻译区,nsP(包括nsP1、nsP2、nsP3、nsP4)是能形成复制酶的多个非结构蛋白序列,SGP是亚基因组启动子,GOI是一个或多个目的蛋白编码基因,3’UTR为3’非翻译区域,Poly-A是3’多腺苷酸尾部,并且其中nsP包含突变的宏结构域。当存在多个GOI时,每个GOI可以可操作地连接于其各自的SGP。在一些实施方案中,所述SGP是26S启动子,例如如SEQ ID NO:65所示。Wherein, 5'UTR is 5' untranslated region, nsP (including nsP1, nsP2, nsP3, nsP4) is a plurality of non-structural protein sequences capable of forming replicase, SGP is a subgenomic promoter, GOI is one or more target protein encoding genes, 3'UTR is 3' untranslated region, Poly-A is 3' polyadenylic acid tail, and wherein nsP comprises a mutated macrodomain. When multiple GOIs are present, each GOI can be operably linked to its respective SGP. In some embodiments, the SGP is a 26S promoter, for example as shown in SEQ ID NO:65.
在一些实施方案中,本文公开的saRNA包含与SEQ ID NO:63至少85%相同的5’非翻译序列,与SEQ ID NO:68或70至少85%相同且包含编码如SEQ ID NO:57 或59所示的宏结构域的核苷酸序列的非结构蛋白编码序列,和/或与SEQ ID NO:64至少85%相同的3’非翻译序列。所述至少85%相同可以是至少86%、87%、88%、89%、90%、91%、92%、93%、94%、95%、96%、97%、98%、99%相同,且涵盖保留这些序列相应功能的功能性变体。In some embodiments, the saRNA disclosed herein comprises a 5' untranslated sequence that is at least 85% identical to SEQ ID NO: 63, at least 85% identical to SEQ ID NO: 68 or 70 and comprises a sequence encoding a sequence such as SEQ ID NO: 57. or a nonstructural protein coding sequence of the nucleotide sequence of the macrodomain shown in 59, and/or a 3' non-translated sequence that is at least 85% identical to SEQ ID NO: 64. The at least 85% identical can be at least 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identical, and encompasses functional variants that retain the corresponding functions of these sequences.
在一些实施方案中,5’UTR、nsP、SGP和3’UTR序列均来源于甲病毒。甲病毒基因组编码4个非结构蛋白和6个结构蛋白(衣壳、E3、E2、6K、TF和E1)。甲病毒结构蛋白(核心核衣壳蛋白C、包膜蛋白E2和包膜蛋白E1,病毒颗粒的所有成分)通常由处于亚基因组启动子控制下的一个单一开放阅读框编码(Strauss&Strauss,Microbiol.Rev.,1994,vol.58,pp.491-562)。亚基因组启动子被顺式作用的甲病毒非结构蛋白识别。特别地,甲病毒复制酶使用基因组RNA的(-)链互补链作为模板来合成(+)链亚基因组转录物。(+)链亚基因组转录物编码甲病毒结构蛋白(Kim et al.,2004,Virology,vol.323,pp.153-163,Vasiljeva et al.,2003,J.Biol.Chem.vol.278,pp.41636-41645)。亚基因组RNA转录物充当模板,用于翻译编码作为一种多蛋白的结构蛋白的开放阅读框,并且多蛋白(poly-protein)被切割以产生结构蛋白。In some embodiments, 5'UTR, nsP, SGP and 3'UTR sequences are all derived from alphavirus. The alphavirus genome encodes 4 nonstructural proteins and 6 structural proteins (capsid, E3, E2, 6K, TF and E1). Alphavirus structural proteins (core nucleocapsid protein C, envelope protein E2 and envelope protein E1, all components of viral particles) are usually encoded by a single open reading frame under the control of a subgenomic promoter (Strauss & Strauss, Microbiol. Rev., 1994, vol. 58, pp. 491-562). The subgenomic promoter is recognized by cis-acting alphavirus nonstructural proteins. In particular, the alphavirus replicase uses the (-) strand complementary strand of the genomic RNA as a template to synthesize the (+) strand subgenomic transcript. (+) strand subgenomic transcripts encode alphavirus structural proteins (Kim et al., 2004, Virology, vol. 323, pp. 153-163, Vasiljeva et al., 2003, J. Biol. Chem. vol. 278, pp. 41636-41645). Subgenomic RNA transcripts serve as templates for translation of open reading frames encoding structural proteins as a polyprotein, and the polyprotein is cleaved to produce structural proteins.
本文公开的saRNA在利用体外转录制备前可将DNA质粒模版进行线性化。为此,可以在saRNA 3’端加入限制性酶切位点以便于切割。除此之外,所述saRNA可以进一步包含其他编码序列,例如在5’端包含信号肽编码序列、与复制酶编码序列相容的其他序列、在SGP启动子下游包含Kozak序列、以及另外的下游编码区例如用于编码其他所需的基因产物。The saRNA disclosed herein can linearize the DNA plasmid template before being prepared by in vitro transcription. To this end, a restriction enzyme site can be added to the 3' end of the saRNA to facilitate cutting. In addition, the saRNA can further contain other coding sequences, such as a signal peptide coding sequence at the 5' end, other sequences compatible with the replicase coding sequence, a Kozak sequence downstream of the SGP promoter, and additional downstream coding regions, for example, for encoding other desired gene products.
在一些具体的实施方案中,本文公开的saRNA包含如SEQ ID NO:61或62所示的saRNA骨架序列(即未加入目的蛋白编码序列或目的基因序列)。在一些具体的实施方案中,本文公开的saRNA包含如SEQ ID NO:58或60所示的核酸序列,其中以荧光酶素及绿色荧光融合蛋白编码序列作为示例目的蛋白。In some specific embodiments, the saRNA disclosed herein comprises a saRNA backbone sequence as shown in SEQ ID NO: 61 or 62 (i.e., without adding a target protein coding sequence or a target gene sequence). In some specific embodiments, the saRNA disclosed herein comprises a nucleic acid sequence as shown in SEQ ID NO: 58 or 60, wherein luciferase and green fluorescent fusion protein coding sequences are used as exemplary target proteins.
制备saRNA的方法Methods for preparing saRNA
saRNA通常通过RNA聚合酶进行体外转录(IVT)得到。在一个方面,本公开涉及获得自扩增mRNA的方法,包括:使用编码本公开的saRNA的DNA分子(例如质粒)作为模板和RNA聚合酶通过体外转录产生saRNA。例如,可以通过使用合适的DNA依赖性RNA聚合酶体外转录编码saRNA的DNA来制备saRNA,所述DNA 依赖性RINA聚合酶例如:T7噬菌体RNA聚合酶、SP6噬菌体RNA聚合酶、T3噬菌体RNA聚合酶、T5噬菌体RNA聚合酶、RNA聚合酶III、RNA聚合酶II、Taq聚合酶等,或这些聚合酶的突变体。saRNA is generally obtained by in vitro transcription (IVT) by RNA polymerase. In one aspect, the present disclosure relates to a method for obtaining self-amplified mRNA, comprising: using a DNA molecule (e.g., a plasmid) encoding the saRNA of the present disclosure as a template and RNA polymerase to produce saRNA by in vitro transcription. For example, saRNA can be prepared by in vitro transcription of DNA encoding saRNA using a suitable DNA-dependent RNA polymerase, wherein the DNA Examples of the RINA-dependent polymerases include T7 phage RNA polymerase, SP6 phage RNA polymerase, T3 phage RNA polymerase, T5 phage RNA polymerase, RNA polymerase III, RNA polymerase II, Taq polymerase, etc., or mutants of these polymerases.
转录反应将包含核苷酸以及支持所选聚合酶活性的其他成分,例如合适的缓冲液和合适的盐。此外,还可以将核苷酸类似物掺入saRNA中,以例如改变这种RNA分子的稳定性,增加对核糖核酸酶的抵抗力,在引入适当的宿主细胞后建立复制,和/或诱导或减少先天和适应性免疫反应。The transcription reaction will contain nucleotides as well as other components that support the activity of the selected polymerase, such as a suitable buffer and suitable salts. In addition, nucleotide analogs can also be incorporated into saRNA to, for example, alter the stability of such RNA molecules, increase resistance to ribonucleases, establish replication after introduction into appropriate host cells, and/or induce or reduce innate and adaptive immune responses.
saRNA的应用Applications of saRNA
基于mRNA的疗法,是将体外合成的mRNA递送到人体中的特定细胞,mRNA在细胞质中被翻译成所需的蛋白质。mRNA作为疫苗或药物,可应用于预防传染病、治疗肿瘤和蛋白替代疗法。mRNA药物靶点丰富,不受靶点蛋白成药性,胞内/外限制。mRNA序列设计简便,利用患者自身细胞产生分子,绕过化学合成中的挑战,且自身诱导产生的分子药效更强。mRNA生产平台具有较强的延展性和可复制性,易于规模化生产。mRNA-based therapy is the delivery of in vitro synthesized mRNA to specific cells in the human body, where it is translated into the desired protein in the cytoplasm. As a vaccine or drug, mRNA can be used to prevent infectious diseases, treat tumors, and for protein replacement therapy. mRNA drugs have rich targets and are not restricted by the drugability of the target protein or intracellular/extracellular restrictions. The mRNA sequence is easy to design, and the patient's own cells are used to produce molecules, bypassing the challenges of chemical synthesis, and the self-induced molecules have stronger efficacy. The mRNA production platform has strong scalability and replicability, and is easy to scale up.
在一个方面,本公开涉及一种药物组合物,例如包含本公开的saRNA和药学上可接受的载剂的疫苗组合物。所述药学上可接受的载剂可以是saRNA递送系统,优选纳米粒子组合物。在一些方面,所述纳米粒子组合物包含阳离子脂质、PEG修饰的脂质、甾醇和非阳离子脂质。In one aspect, the present disclosure relates to a pharmaceutical composition, such as a vaccine composition comprising a saRNA of the present disclosure and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier can be a saRNA delivery system, preferably a nanoparticle composition. In some aspects, the nanoparticle composition comprises a cationic lipid, a PEG-modified lipid, a sterol, and a non-cationic lipid.
在一个方面,本公开的sa-mRNA可用于疫苗接种。所述疫苗可以是传染病领域预防性疫苗。例如,saRNA中编码的目的蛋白是针对传染性疾病的mRNA疫苗编码相关病原体的抗原,注射后可在体内表达出特异性的抗原,可同时诱导细胞免疫和体液免疫,刺激产生相应抗体和免疫细胞,用以预防相应病原体。所述疫苗可以是肿瘤治疗性疫苗。例如,将编码肿瘤特异性抗原靶点的mRNA通过特定的方式递送到体内,使这些肿瘤特异性抗原翻译并呈递到细胞表面,进而激活免疫系统,使其能特异性识别并杀伤肿瘤细胞。In one aspect, the sa-mRNA disclosed herein can be used for vaccination. The vaccine can be a preventive vaccine in the field of infectious diseases. For example, the target protein encoded in the saRNA is an antigen of a pathogen related to an mRNA vaccine encoding an infectious disease. After injection, specific antigens can be expressed in the body, which can simultaneously induce cellular immunity and humoral immunity, stimulate the production of corresponding antibodies and immune cells to prevent the corresponding pathogens. The vaccine can be a tumor therapeutic vaccine. For example, mRNA encoding tumor-specific antigen targets is delivered to the body in a specific manner, so that these tumor-specific antigens are translated and presented on the cell surface, thereby activating the immune system so that it can specifically recognize and kill tumor cells.
在一个方面,本公开的saRNA可用于疾病的治疗,例如通过提供针对疾病的治疗性蛋白或免疫调控蛋白。通常情况下,蛋白质替代疗法用于治疗罕见的单基因疾病,旨在恢复酶的功能。蛋白质合成困难,给药存在诸多难题且价格昂贵,而利用mRNA 将人体变成自身蛋白质的加工厂,理论上是一种经济可行并且高效的方式。本公开的saRNA理论上可以合成任意一种目的蛋白,可作为蛋白质补充或替代疗法治疗多种疾病。In one aspect, the saRNA disclosed herein can be used to treat diseases, for example, by providing therapeutic proteins or immune regulatory proteins for diseases. Typically, protein replacement therapy is used to treat rare monogenic diseases, aiming to restore enzyme function. Protein synthesis is difficult, administration is difficult and expensive, while the use of mRNA Turning the human body into a processing plant for its own proteins is theoretically an economically feasible and efficient way. The saRNA disclosed in the present invention can theoretically synthesize any target protein and can be used as a protein supplement or alternative therapy to treat a variety of diseases.
在一个方面,本公开提供了一种将目的蛋白递送至受试者的方法,包括向受试者施用含有一种或多种本公开的自扩增mRNA的药物组合物。In one aspect, the present disclosure provides a method of delivering a protein of interest to a subject, comprising administering to the subject a pharmaceutical composition containing one or more self-amplifying mRNAs of the present disclosure.
发明的有益效果Advantageous Effects of the Invention
在一个方面,本发明取得了如下技术效果(1)至(3)中的一项或者多项:In one aspect, the present invention achieves one or more of the following technical effects (1) to (3):
(1)本发明提高了saRNA体外转录完整性。(1) The present invention improves the integrity of saRNA in vitro transcription.
(2)本发明对saRNA的复制功能无负面影响。(2) The present invention has no negative impact on the replication function of saRNA.
(3)本发明对saRNA的表达功能无负面影响。(3) The present invention has no negative impact on the expression function of saRNA.
此外,目前的自扩增mRNA为甲病毒基因组改造而来的mRNA载体,无特定microRNA结合位点。经目前常用的纳米脂质颗粒(lipid nanoparticles,LNP)包封并递送到体内后,由于纳米脂质颗粒固有理化性质,可能介导自扩增mRNA大量进入肝脏等实质组织表达,在某些应用实例中会引起潜在的肝脏毒性。因此,在另一个方面,本发明取得的技术效果还包括,本发明利用肝脏组织特异高表达的microRNA来降解自扩增mRNA,从而实现降低肝脏毒性。In addition, the current self-amplifying mRNA is an mRNA vector modified from the alphavirus genome, and has no specific microRNA binding site. After being encapsulated and delivered into the body by the currently commonly used nanolipid particles (lipid nanoparticles, LNP), due to the inherent physical and chemical properties of the nanolipid particles, the self-amplifying mRNA may be mediated to enter the liver and other parenchymal tissues for expression in large quantities, which may cause potential liver toxicity in some application examples. Therefore, in another aspect, the technical effect achieved by the present invention also includes that the present invention utilizes the microRNA highly expressed in liver tissue to degrade the self-amplifying mRNA, thereby reducing liver toxicity.
在又一个方面,本发明的saRNA具有已知的saRNA相对于传统mRNA的优点,即它可以用很低的剂量达到与传统mRNA相同的蛋白表达水平,并延长抗原蛋白在体内存在的时间,从而可能增强免疫反应,在生产方面,更低有效剂量可能降低saRNA的生产成本。作为疗法,可减少mRNA疗法使用的剂量和注射次数,从而在延长疗效的同时,降低mRNA和递送载体可能产生的毒副作用。In another aspect, the saRNA of the present invention has the advantages of known saRNA over traditional mRNA, that is, it can achieve the same protein expression level as traditional mRNA with a very low dose, and prolong the existence time of antigen protein in the body, thereby possibly enhancing the immune response. In terms of production, a lower effective dose may reduce the production cost of saRNA. As a therapy, the dose and number of injections used in mRNA therapy can be reduced, thereby prolonging the efficacy while reducing the possible toxic side effects of mRNA and delivery vectors.
进一步地,本发明的saRNA还经过改进,从而具有以下一种或多种特性:Furthermore, the saRNA of the present invention is also improved to have one or more of the following characteristics:
(1)降低了复制效率,saRNA复制过程中形成的dsRNA明显减少;(1) The replication efficiency is reduced, and the dsRNA formed during saRNA replication is significantly reduced;
(2)降低了saRNA诱导的天然免疫反应强度;(2) Reduced the intensity of the natural immune response induced by saRNA;
(3)减弱了saRNA受到的翻译抑制;(3) Reduced translational inhibition of saRNA;
(4)减少了saRNA诱导的细胞调亡;(4) Reduced saRNA-induced cell apoptosis;
(5)提高了saRNA的表达效率。(5) Improved the expression efficiency of saRNA.
本公开的saRNA含有突变的宏结构域,使得ADP核糖水解活性减弱,从而导致 病毒复制能力减弱以及刺激先天免疫系统的能力降低。因此,本公开的saRNA可能对宿主细胞或受试者具有降低的细胞毒性作用,提供了增强的安全性。例如,本公开的saRNA能够产生高表达水平的编码基因产物,同时降低不期望的影响(如注射部位刺激和/或疼痛)的风险。此外,由于本公开的saRNA刺激先天免疫系统的能力降低,其特别适用于疫苗制备以提供对宿主的适当的免疫。The saRNA disclosed herein contains a mutated macrodomain, which weakens the ADP ribose hydrolysis activity, thereby causing The ability of the virus to replicate is weakened and the ability to stimulate the innate immune system is reduced. Therefore, the saRNA of the present disclosure may have a reduced cytotoxic effect on host cells or subjects, providing enhanced safety. For example, the saRNA of the present disclosure can produce a high expression level of the encoded gene product while reducing the risk of undesirable effects (such as injection site irritation and/or pain). In addition, since the ability of the saRNA of the present disclosure to stimulate the innate immune system is reduced, it is particularly suitable for vaccine preparation to provide appropriate immunity to the host.
另外,本发明的saRNA不包含编码病毒结构蛋白的核苷酸序列,因此不能导致含有RNA的甲病毒颗粒的产生。无法产生这些病毒颗粒意味着saRNA不能以感染形式自我延续。在野生型病毒中延续所需的甲病毒结构蛋白不存在于saRNA中,它们的位置由GOI取代,使得saRNA编码所需的基因产物,而不是甲病毒病毒颗粒的结构蛋白。In addition, the saRNA of the present invention does not contain nucleotide sequences encoding viral structural proteins and therefore cannot lead to the production of alphavirus particles containing RNA. The inability to produce these viral particles means that the saRNA cannot self-perpetuate in an infectious form. The alphavirus structural proteins required for perpetuation in wild-type viruses are not present in the saRNA, and their positions are replaced by GOI, so that the saRNA encodes the required gene products, rather than the structural proteins of the alphavirus viral particles.
更具体而言,本发明的包含突变宏结构域的saRNA导致复制过程中形成的dsRNA明显减少。人体细胞中存在着识别外来病毒入侵的传感器,它们称为模式识别受体。它们识别的信号之一是在细胞质中出现的双链RNA,因为这可能代表着病毒RNA在细胞中进行复制。而saRNA在复制的过程中会形成双链RNA,它们与复制中的病毒RNA很像,因此可能激发细胞的先天免疫反应。这有可能进一步增强疫苗的作用。然而,这种激发的免疫反应一方面作为疫苗,它可能达到促进免疫应答的作用,但是另一方面,作为疗法时,激发的免疫反应可能造成副作用。如果激发的先天免疫反应过强,可能导致mRNA的表达受到抑制,反而影响疗法和疫苗的疗效。此外,saRNA复制中产生的双链RNA是激活干扰素或NFKB信号通路的关键,从而诱导下游大量抗病毒基因以抑制saRNA复制。因此,对saRNA的免疫原性需要精准的设计和调整,从而适度降低saRNA的复制能力和双链RNA的产生,本发明公开的sgRNA实现了这一目的。More specifically, the saRNA containing a mutant macrodomain of the present invention leads to a significant reduction in the dsRNA formed during replication. There are sensors in human cells that recognize foreign viral invasions, which are called pattern recognition receptors. One of the signals they recognize is double-stranded RNA that appears in the cytoplasm, because this may represent the replication of viral RNA in the cell. During the replication process, saRNA forms double-stranded RNA, which is very similar to the viral RNA in replication, and therefore may stimulate the innate immune response of the cell. This may further enhance the effect of the vaccine. However, this stimulated immune response may promote the immune response as a vaccine, but on the other hand, as a therapy, the stimulated immune response may cause side effects. If the stimulated innate immune response is too strong, the expression of mRNA may be inhibited, which in turn affects the efficacy of therapy and vaccines. In addition, the double-stranded RNA produced during saRNA replication is the key to activating the interferon or NFKB signaling pathway, thereby inducing a large number of downstream antiviral genes to inhibit saRNA replication. Therefore, the immunogenicity of saRNA requires precise design and adjustment, so as to moderately reduce the replication ability of saRNA and the production of double-stranded RNA, and the sgRNA disclosed in the present invention achieves this purpose.
同时,本发明的包含突变宏结构域的saRNA受到的翻译抑制和诱导的细胞凋亡都显著减少。mRNA通常会触发细胞内固有受体,如toll样受体(toll like recepotor,TLR),其导致I型干扰素的产生和翻译的抑制。然而,本发明的saRNA由于免疫原性降低,受到的翻译抑制是降低的,因此虽然其复制能力有所下降,但最终在细胞中的表达效率相比于编码野生型宏结构域的对照saRNA是提高的。At the same time, the translation inhibition and induced apoptosis of the saRNA containing the mutant macro domain of the present invention are significantly reduced. mRNA usually triggers intrinsic receptors in cells, such as toll-like receptors (TLR), which lead to the production of type I interferon and inhibition of translation. However, the translation inhibition of the saRNA of the present invention is reduced due to reduced immunogenicity, so although its replication ability is reduced, the final expression efficiency in the cell is improved compared to the control saRNA encoding the wild-type macro domain.
本发明涉及的序列如下汇总(当为RNA序列时T为U):
序列1:BspQI内切酶识别位点
The sequences involved in the present invention are summarized as follows (when it is an RNA sequence, T is U): Sequence 1: BspQI endonuclease recognition site
序列2:saRNA-062序列
Sequence 2: saRNA-062 sequence
序列3:saRNA-152序列
Sequence 3: saRNA-152 sequence
序列4:saRNA-153序列
Sequence 4: saRNA-153 sequence
序列5:saRNA-154序列
Sequence 5: saRNA-154 sequence
序列6:saRNA-183序列
Sequence 6: saRNA-183 sequence
序列7:saRNA-184序列
Sequence 7: saRNA-184 sequence
序列8:saRNA-185序列
Sequence 8: saRNA-185 sequence
序列9:saRNA-243序列
Sequence 9: saRNA-243 sequence
上面的序列2-9中:In the above sequence 2-9:
“NNNNNNNNNNNNNNNN”表示外源基因,包括但不限于:疫苗抗原基因、肿瘤杀伤基因、治疗性蛋白基因、抗体基因;可以依据不同目的调整外源基因;“NNNNNNNNNNNNNNNN” indicates exogenous genes, including but not limited to: vaccine antigen genes, tumor killing genes, therapeutic protein genes, antibody genes; exogenous genes can be adjusted according to different purposes;
其中“N”可以为A、T、C、G中的任何一种碱基,碱基数目并不特别限定;此处16个N仅为示意,并不是限定为16个碱基。Here, "N" can be any base among A, T, C, and G, and the number of bases is not particularly limited; the 16 Ns here are only for illustration and are not limited to 16 bases.
序列10:062复制酶编码序列(SEQ ID NO:10)Sequence 10:062 Replicase coding sequence (SEQ ID NO:10)
序列11:152复制酶编码序列(SEQ ID NO:11)Sequence 11: 152 replicase coding sequence (SEQ ID NO: 11)
序列12:183复制酶编码序列(SEQ ID NO:12)Sequence 12: 183 replicase coding sequence (SEQ ID NO: 12)
序列13:184复制酶编码序列(SEQ ID NO:13)Sequence 13: 184 replicase coding sequence (SEQ ID NO: 13)
序列14:185复制酶编码序列(SEQ ID NO:14)Sequence 14: 185 replicase coding sequence (SEQ ID NO: 14)
序列15:243复制酶编码序列(SEQ ID NO:15)Sequence 15: 243 replicase coding sequence (SEQ ID NO: 15)
序列16:5’端非翻译序列(SEQ ID NO:16)Sequence 16: 5' non-translated sequence (SEQ ID NO: 16)
序列17:3’端非翻译序列(SEQ ID NO:17)Sequence 17: 3’ non-translated sequence (SEQ ID NO: 17)
序列18:亚基因组启动子序列(SEQ ID NO:18)Sequence 18: Subgenomic promoter sequence (SEQ ID NO: 18)
序列19:多聚腺苷酸序列(SEQ ID NO:19)Sequence 19: Polyadenylation sequence (SEQ ID NO: 19)
序列20:甲病毒RNA复制酶的氨基酸序列(SEQ ID NO:20)Sequence 20: Amino acid sequence of alphavirus RNA replicase (SEQ ID NO: 20)
序列21:S23H02序列
Sequence 21: S23H02 sequence
序列22:6个microRNA-122结合位点串联盒序列
Sequence 22: Six microRNA-122 binding site tandem cassette sequences
序列23:saRNA-EGFP-miRNA-1序列(SEQ ID NO:23)Sequence 23: saRNA-EGFP-miRNA-1 sequence (SEQ ID NO: 23)
序列24:saRNA-EGFP-miRNA-2(SEQ ID NO:24)Sequence 24: saRNA-EGFP-miRNA-2 (SEQ ID NO:24)
序列25:saRNA-EGFP-miRNA-3(SEQ ID NO:25)Sequence 25: saRNA-EGFP-miRNA-3 (SEQ ID NO:25)
序列26:saRNA-EGFP-miRNA-4(SEQ ID NO:26)Sequence 26: saRNA-EGFP-miRNA-4 (SEQ ID NO:26)
序列27:saRNA-EGFP-miRNA-5(SEQ ID NO:27)Sequence 27: saRNA-EGFP-miRNA-5 (SEQ ID NO:27)
序列28:saRNA-EGFP-miRNA-6(SEQ ID NO:28)Sequence 28: saRNA-EGFP-miRNA-6 (SEQ ID NO:28)
序列29:saRNA-EGFP序列(SEQ ID NO:29)Sequence 29: saRNA-EGFP sequence (SEQ ID NO: 29)
序列30:microRNA-122-5p结合位点序列(SEQ ID NO:30)Sequence 30: microRNA-122-5p binding site sequence (SEQ ID NO: 30)
序列31:microRNA-1-5p结合位点序列(SEQ ID NO:31) Sequence 31: microRNA-1-5p binding site sequence (SEQ ID NO: 31)
序列32:microRNA-124-5p结合位点序列(SEQ ID NO:32)Sequence 32: microRNA-124-5p binding site sequence (SEQ ID NO: 32)
序列33:Inner spacer序列1Sequence 33: Inner spacer sequence 1
AGCGCGA(SEQ ID NO:33)AGCGCGA (SEQ ID NO:33)
序列34:Inner spacer序列2Sequence 34: Inner spacer sequence 2
ACGCGAA(SEQ ID NO:34)ACGCGAA (SEQ ID NO:34)
序列35:5’outer spacer序列1(SEQ ID NO:35)Sequence 35: 5’outer spacer sequence 1 (SEQ ID NO: 35)
序列36:5’outer spacer序列2(SEQ ID NO:36)Sequence 36: 5’outer spacer sequence 2 (SEQ ID NO: 36)
序列37:3’outer spacer序列1(SEQ ID NO:37)Sequence 37: 3’outer spacer sequence 1 (SEQ ID NO: 37)
序列38:3’outer spacer序列2(SEQ ID NO:38)Sequence 38: 3’outer spacer sequence 2 (SEQ ID NO: 38)
序列39:5’复制酶识别蛋白酶切位点1(SEQ ID NO:39)Sequence 39: 5' replicase recognition protease cleavage site 1 (SEQ ID NO: 39)
序列40:3’复制酶识别蛋白酶切位点1(SEQ ID NO:40)Sequence 40: 3’ replicase recognition protease cleavage site 1 (SEQ ID NO: 40)
序列41:甲病毒RNA复制酶的氨基酸序列(SEQ ID NO:41)Sequence 41: Amino acid sequence of alphavirus RNA replicase (SEQ ID NO: 41)
序列42:甲病毒RNA复制酶的编码序列(SEQ ID NO:42)Sequence 42: Coding sequence of alphavirus RNA replicase (SEQ ID NO: 42)
序列54:甲病毒复制必需序列含有的最短序列:(SEQ ID NO:54)Sequence 54: The shortest sequence contained in the essential sequence for virus replication: (SEQ ID NO: 54)
序列55:来自VEEV的野生型宏结构域氨基酸序列(SEQ ID NO:55)Sequence 55: Wild-type macrodomain amino acid sequence from VEEV (SEQ ID NO: 55)
序列57:saRNA-190对应的宏结构域氨基酸序列(引入Q48P突变,相应RNA编码序列由CAG变为CCG)(SEQ ID NO:57)Sequence 57: amino acid sequence of the macrodomain corresponding to saRNA-190 (introducing Q48P mutation, the corresponding RNA coding sequence changes from CAG to CCG) (SEQ ID NO: 57)
序列59:saRNA-191对应的宏结构域氨基酸序列(引入I113F突变,相应RNA编码序列由ATC变为TTC)(SEQ ID NO:59)Sequence 59: amino acid sequence of the macrodomain corresponding to saRNA-191 (introducing the I113F mutation, the corresponding RNA coding sequence changed from ATC to TTC) (SEQ ID NO: 59)
序列56:野生型saRNA序列
Sequence 56: Wild-type saRNA sequence
单下划线:5’和3’UTR;双下划线:复制酶结构域序列(阴影部分为宏结构域编码序列);方框:26S启动子和Kozak序列;斜体:荧光酶素及绿色荧光融合蛋白编码序列,但可更换为任意外源基因序列;虚线下划线:BsQI酶切序列Single underline: 5' and 3' UTR; Double underline: replicase domain sequence (shaded part is macro domain coding sequence); Box: 26S promoter and Kozak sequence; Italic: luciferase and green fluorescent fusion protein coding sequence, but can be replaced by any exogenous gene sequence; Dotted underline: BsQI restriction enzyme sequence
序列58:saRNA-190序列(与序列57不同仅在于引入宏结构域中的Q48P突变)(SEQ ID NO:58)Sequence 58: saRNA-190 sequence (different from sequence 57 only by the introduction of the Q48P mutation in the macrodomain) (SEQ ID NO: 58)
序列60:saRNA-191序列(与序列57不同仅在于引入宏结构域中的I113F突变)(SEQ ID NO:60)Sequence 60: saRNA-191 sequence (different from sequence 57 only by the introduction of the I113F mutation in the macrodomain) (SEQ ID NO: 60)
序列61:saRNA-190骨架序列
Sequence 61: saRNA-190 backbone sequence
序列62:saRNA-191骨架序列
Sequence 62: saRNA-191 backbone sequence
上面的序列61-62中:In the sequence 61-62 above:
“NNNNNNNNNNNNNNNN”表示外源基因,包括但不限于:疫苗抗原基因、肿瘤杀伤基因、治疗性蛋白基因、抗体基因;可以依据不同目的调整外源基因;“NNNNNNNNNNNNNNNN” indicates exogenous genes, including but not limited to: vaccine antigen genes, tumor killing genes, therapeutic protein genes, antibody genes; exogenous genes can be adjusted according to different purposes;
其中“N“可以为A、T、C、G中的任何一种碱基,碱基数目并不特别限定;此处16个N仅为示意,并不是限定为16个碱基。Here, "N" can be any base among A, T, C, and G, and the number of bases is not particularly limited; the 16 Ns here are only for illustration and are not limited to 16 bases.
序列63:5’端非翻译序列(SEQ ID NO:63)Sequence 63: 5' non-translated sequence (SEQ ID NO: 63)
序列64:3’端非翻译序列(SEQ ID NO:64)Sequence 64: 3' non-translated sequence (SEQ ID NO: 64)
序列65:亚基因组启动子序列(SEQ ID NO:65)Sequence 65: Subgenomic promoter sequence (SEQ ID NO: 65)
序列66:多聚腺苷酸序列(SEQ ID NO:66)Sequence 66: Polyadenylation sequence (SEQ ID NO: 66)
序列67:saRNA-190复制酶编码序列(SEQ ID NO:67)Sequence 67: saRNA-190 replicase coding sequence (SEQ ID NO: 67)
序列68:saRNA-190宏结构域编码序列(SEQ ID NO:68)Sequence 68: saRNA-190 macrodomain encoding sequence (SEQ ID NO: 68)
序列69:saRNA-191复制酶编码序列(SEQ ID NO:69)Sequence 69: saRNA-191 replicase coding sequence (SEQ ID NO: 69)
序列70:saRNA-191宏结构域编码序列(SEQ ID NO:70)Sequence 70: saRNA-191 macrodomain encoding sequence (SEQ ID NO: 70)
序列71:非结构性蛋白1氨基酸序列(SEQ ID NO:71)Sequence 71: Nonstructural protein 1 amino acid sequence (SEQ ID NO: 71)
序列72:非结构性蛋白2氨基酸序列(SEQ ID NO:72)Sequence 72: Nonstructural protein 2 amino acid sequence (SEQ ID NO: 72)
序列73:非结构性蛋白3氨基酸序列(对应saRNA-190,引入Q48P突变)(SEQ ID NO:73)Sequence 73: Nonstructural protein 3 amino acid sequence (corresponding to saRNA-190, with Q48P mutation introduced) (SEQ ID NO: 73)
序列74:非结构性蛋白3氨基酸序列(对应saRNA-191,引入I133F突变)(SEQ ID NO:74)Sequence 74: Nonstructural protein 3 amino acid sequence (corresponding to saRNA-191, with I133F mutation introduced) (SEQ ID NO: 74)
序列75:非结构性蛋白4氨基酸序列(SEQ ID NO:75)Sequence 75: Nonstructural protein 4 amino acid sequence (SEQ ID NO: 75)
下面将结合实施例对本发明的实施方案进行详细描述,但是本领域技术人员将会理解,下列实施例仅用于说明本发明,而不应视为限定本发明的范围。实施例中未注 明具体条件者,按照常规条件或制造商建议的条件进行。所用试剂或仪器未注明生产厂商者,均为可以通过市购获得的常规产品。The embodiments of the present invention will be described in detail below with reference to the examples, but those skilled in the art will appreciate that the following examples are only used to illustrate the present invention and should not be construed as limiting the scope of the present invention. If the specific conditions are not specified, the experiments were carried out under conventional conditions or the conditions recommended by the manufacturer. If the manufacturer of the reagents or instruments is not specified, they are all conventional products that can be purchased from the market.
Huh7.5.1细胞购自Mingzhou bio,货号MZ-2129。Huh7.5.1 cells were purchased from Mingzhou bio, catalog number MZ-2129.
C2C12细胞购自Mingzhou bio,货号MZ-0034。C2C12 cells were purchased from Mingzhou bio, catalog number MZ-0034.
脂质体转染试剂-mRNA Transfection Kit购自Mirus,货号MIR 2225。Lipofectamine transfection reagents -mRNA Transfection Kit was purchased from Mirus, catalog number MIR 2225.
实施例1:对saRNA-EGFP模板质粒psaRNA-062中4497-4503位的改造和完整Example 1: Modification and completion of positions 4497-4503 in the saRNA-EGFP template plasmid psaRNA-062 性检测Sexual testing
1.优化位置:对saRNA模板质粒psaRNA-062(如SEQ ID NO:2)中4497-4503位(5’-ACTCTTC-3’)进行改造(以甲病毒复制酶起始密码子序列ATG中A碱基记为第1位,下同)。1. Optimize position: Modify the 4497-4503 position (5’-ACTCTTC-3’) in the saRNA template plasmid psaRNA-062 (such as SEQ ID NO: 2) (with the A base in the ATG start codon sequence of the alphavirus replicase as the first position, the same below).
2.优化方法:通过序列定点突变方法,将第4500bp所在T碱基分别突变为C碱基或A碱基,或者将第4501~4503bp的TCA碱基突变为AGC碱基,由此获得3个模板质粒分别命名为psaRNA-152(SEQ ID NO:3)、psaRNA-153(SEQ ID NO:4)和psaRNA-154(SEQ ID NO:5)。2. Optimization method: Through the sequence-directed mutagenesis method, the T base at the 4500th bp was mutated into the C base or the A base, or the TCA base at the 4501-4503 bp was mutated into the AGC base. The three template plasmids obtained were named psaRNA-152 (SEQ ID NO: 3), psaRNA-153 (SEQ ID NO: 4) and psaRNA-154 (SEQ ID NO: 5).
3.改造后的saRNA体外转录制备:3. Preparation of modified saRNA by in vitro transcription:
a.体外转录模板质粒线性化:具体为,依次加入8μg体外转录模板质粒质粒(psaRNA-062、psaRNA-152、psaRNA-153和psaRNA-154),BspQ I 4μL,10×Digestion Buffer 4μL,最后用DEPC水将体系补足40μL,50℃孵育2h线性化质粒。a. Linearization of in vitro transcription template plasmid: Specifically, add 8 μg of in vitro transcription template plasmid (psaRNA-062, psaRNA-152, psaRNA-153 and psaRNA-154), BspQ I 4 μL, 10×Digestion Buffer 4 μL in sequence, and finally add DEPC water to the system to 40 μL, and incubate at 50°C for 2 h to linearize the plasmid.
b.体外转录获得改造后的saRNA:具体为,依次加入10×T7聚合酶缓冲液,7.5mM ATP,7.5mM GTP,7.5mM UTP,7.5mM CTP,1μg线性化体外转录模板,400U T7 RNA聚合酶,20U RNase I抑制剂,6mM帽类似物,补水至总体积20μL,37℃反应2小时。反应结束后加入2U DNase I酶,37℃反应15分钟。反应体系补水至50μL,加入25μL7.5M氯化锂溶液混匀,-20℃静置处理30分钟后,13000g 4℃离心10分钟,弃上清后用70%乙醇清洗沉淀,13000g 4℃离心2分钟,弃上清,溶解在30μL水中。用分光光度计测定浓度后,取适量体积RNA使用Qsep400毛细管电泳设备检测RNA完整性,检测结果如图1,结果显示,只有经过优化后的saRNA-152完整性得到提高,从76.7%提升至81.3%。 b. In vitro transcription to obtain modified saRNA: Specifically, add 10×T7 polymerase buffer, 7.5mM ATP, 7.5mM GTP, 7.5mM UTP, 7.5mM CTP, 1μg linearized in vitro transcription template, 400U T7 RNA polymerase, 20U RNase I inhibitor, 6mM cap analog, add water to a total volume of 20μL, and react at 37°C for 2 hours. After the reaction, add 2U DNase I enzyme and react at 37°C for 15 minutes. The reaction system is replenished to 50μL, 25μL 7.5M lithium chloride solution is added and mixed, and after standing at -20°C for 30 minutes, centrifuged at 13000g 4°C for 10 minutes, the supernatant is discarded and the precipitate is washed with 70% ethanol, centrifuged at 13000g 4°C for 2 minutes, the supernatant is discarded, and dissolved in 30μL water. After measuring the concentration with a spectrophotometer, an appropriate volume of RNA was taken and the RNA integrity was tested using the Qsep400 capillary electrophoresis device. The test results are shown in Figure 1. The results show that only the integrity of the optimized saRNA-152 was improved from 76.7% to 81.3%.
实施例2:以优化后psaRNA-152模板质粒为骨架所构建不同saRNA的完整性检Example 2: Integrity test of different saRNAs constructed with the optimized psaRNA-152 template plasmid as the backbone 测Test
以psaRNA-152质粒为模板,对其4509位A碱基分别突变为T、C、G,由此获得模板质粒命名为psaRNA-183(SEQ ID NO:6)、psaRNA-184(SEQ ID NO:7)、psaRNA-185(SEQ ID NO:8)。具体定点突变及检测方法同实施例1,从图2中毛细管电泳检测结果可知,以psaRNA-152为骨架所构建的不同saRNA的完整性相比原始骨架psaRNA-062均有明显提升。Using the psaRNA-152 plasmid as a template, the A base at position 4509 was mutated to T, C, and G, respectively, and the template plasmids thus obtained were named psaRNA-183 (SEQ ID NO: 6), psaRNA-184 (SEQ ID NO: 7), and psaRNA-185 (SEQ ID NO: 8). The specific site-directed mutation and detection methods were the same as those in Example 1. From the capillary electrophoresis detection results in Figure 2, it can be seen that the integrity of different saRNAs constructed with psaRNA-152 as the backbone is significantly improved compared to the original backbone psaRNA-062.
实施例3:对psaRNA-183模板质粒中1680-1676位改造和完整性检测Example 3: Modification and integrity detection of positions 1680-1676 in the psaRNA-183 template plasmid
对进一步优化后saRNA模板质粒psaRNA-183中1680-1676位(5’-GCTCTTA-3’)进行改造,具体为将1672-1674位TCT碱基突变为AGC碱基,由此获得模板质粒命名为psaRNA-243(SEQ ID NO:9)。具体定点突变及检测方法同实施例1,优化后毛细管电泳检测结果如图3,结果显示经过优化后的saRNA中明显的短转录产物比例降低,saRNA-243完整性得到明显提高。The 1680-1676 positions (5'-GCTCTTA-3') in the further optimized saRNA template plasmid psaRNA-183 were modified, specifically, the TCT bases at positions 1672-1674 were mutated to AGC bases, and the obtained template plasmid was named psaRNA-243 (SEQ ID NO: 9). The specific site-directed mutation and detection method are the same as in Example 1. The capillary electrophoresis detection results after optimization are shown in Figure 3. The results show that the proportion of short transcription products in the optimized saRNA is significantly reduced, and the integrity of saRNA-243 is significantly improved.
实施例4:骨架优化后的表达荧光素酶基因saRNA复制表达效率验证Example 4: Verification of the replication and expression efficiency of luciferase gene saRNA after skeleton optimization
1.荧光素酶检测1. Luciferase Assay
a.细胞接种:将适宜数量Huh7.5.1细胞接种至24孔细胞培养板,在5% CO2 37℃过夜培养。a. Cell inoculation: An appropriate number of Huh7.5.1 cells were inoculated into a 24-well cell culture plate and cultured overnight at 37°C with 5% CO 2 .
b.自扩增mRNA转染Huh7.5.1细胞:分别将0.5μg骨架优化后的表达荧光素酶基因saRNA-152、saRNA-243或未优化的saRNA-062使用脂质体转染试剂转染至Huh7.5.1细胞,在5% CO2 37℃过夜培养至72小时。b. Self-amplified mRNA transfection of Huh7.5.1 cells: 0.5 μg of saRNA-152, saRNA-243 or unoptimized saRNA-062 expressing luciferase gene after backbone optimization was transfected into Huh7.5.1 cells using liposome transfection reagent, and cultured overnight at 5% CO 2 and 37° C. for 72 hours.
c.荧光素酶的检测:在转染24和72小时后,使用Nano-Glo Luciferase Assay System检测试剂,转移25μl Nano-Glo Luciferase Assay Reagent至96孔白色不透明底孔板,取25μl上清样品至孔中混合均匀,等待3分钟后使用酶标仪检测信号,检测结果如图4和图5,骨架优化后的saRNA-152及saRNA-243中荧光素酶表达量在不同时间点与未优化骨架无显著性差异,说明骨架优化并不影响saRNA表达。c. Luciferase detection: 24 and 72 hours after transfection, use Nano-Glo Luciferase Assay System detection reagent, transfer 25μl Nano-Glo Luciferase Assay Reagent to a 96-well white opaque bottom plate, take 25μl supernatant sample to the well and mix evenly, wait for 3 minutes and use a microplate reader to detect the signal. The test results are shown in Figures 4 and 5. There is no significant difference in the luciferase expression levels of saRNA-152 and saRNA-243 after backbone optimization and at different time points compared with the unoptimized backbone, indicating that backbone optimization does not affect saRNA expression.
2.自扩增mRNA复制效率检测2. Self-amplification mRNA replication efficiency detection
细胞接种及自扩增mRNA转染:同荧光蛋白及荧光素酶检测。 Cell seeding and self-amplified mRNA transfection: Same as fluorescent protein and luciferase detection.
细胞样品的处理:在转染2小时、24小时和72小时后,吸弃上清,使用500μlPBS润洗细胞一次,弃上清,加入500μl细胞裂解液并吹打收集至EP管,保存于-80℃冰箱。Treatment of cell samples: 2 hours, 24 hours and 72 hours after transfection, discard the supernatant, rinse the cells once with 500 μl PBS, discard the supernatant, add 500 μl cell lysis buffer and collect by pipetting into EP tubes, and store in a -80°C refrigerator.
逆转录荧光定量PCR检测saRNA的复制:细胞样品使用RNeasy Mini Kit提取总RNA后,紫外分光光度计进行RNA定量,使用HiScript III 1st Strand cDNA Synthesis Kit完成逆转录后,利用2×ChamQ Universal SYBR qPCR Master Mix进行荧光定量PCR实验。检测引物如下,甲病毒复制酶序列(nsp1)上游引物F:5‘-GACGGACCGACAAGTCTCTA-3’(SEQ ID NO:22),甲病毒复制酶序列(nsp1)下游引物R:5‘-GGTGGTGTCAAAGCCTATCCA-3’(SEQ ID NO:23),EGFP序列上游引物F:5‘-AGCTGGAGTACAACTACA-3’(SEQ ID NO:24),EGFP下游引物R:5‘-CTGATCTTGAAGTTCACC-3’(SEQ ID NO:25),GAPDH序列上游引物F:5‘-GGTATCGTGGAAGGACTC-3’(SEQ ID NO:26),GAPDH下游引物R:5‘-GTAGAGGCAGGGATGATG-3’(SEQ ID NO:27)。反应循环条件为:95℃30s-(95℃10s-60℃30s)×40个循环,检测结果如图6。该结果显示,骨架优化后的saRNA-152在不同时间点与未优化骨架无显著性差异,说明骨架优化也同样不影响saRNA的复制能力。Reverse transcription fluorescence quantitative PCR was used to detect the replication of saRNA: total RNA was extracted from the cell sample using RNeasy Mini Kit, and RNA was quantified using a UV spectrophotometer. After reverse transcription was completed using HiScript III 1st Strand cDNA Synthesis Kit, fluorescence quantitative PCR experiments were performed using 2×ChamQ Universal SYBR qPCR Master Mix. The detection primers are as follows, alphavirus replicase sequence (nsp1) upstream primer F: 5‘-GACGGACCGACAAGTCTCTA-3’ (SEQ ID NO:22), alphavirus replicase sequence (nsp1) downstream primer R: 5‘-GGTGGTGTCAAAGCCTATCCA-3’ (SEQ ID NO:23), EGFP sequence upstream primer F: 5‘-AGCTGGAGTACAACTACA-3’ (SEQ ID NO:24), EGFP downstream primer R: 5‘-CTGATCTTGAAGTTCACC-3’ (SEQ ID NO:25), GAPDH sequence upstream primer F: 5‘-GGTATCGTGGAAGGACTC-3’ (SEQ ID NO:26), GAPDH downstream primer R: 5‘-GTAGAGGCAGGGATGATG-3’ (SEQ ID NO:27). The reaction cycle conditions were: 95°C 30s-(95°C 10s-60°C 30s)×40 cycles, and the test results are shown in Figure 6. The results show that there is no significant difference between the saRNA-152 after skeleton optimization and the unoptimized skeleton at different time points, indicating that skeleton optimization also does not affect the replication ability of saRNA.
实施例5:制备表达人乳头瘤病毒抗原基因Ag9.1的优化骨架saRNAExample 5: Preparation of optimized backbone saRNA expressing human papillomavirus antigen gene Ag9.1
1.体外转录模板质粒构建1. Construction of in vitro transcription template plasmid
以saRNA-243为模板质粒,将其中的荧光素酶编码序列,通过同源重组方法更换为人乳头瘤病毒(HPV)抗原基因Ag9.1编码序列,得到表达Ag9.1的自扩增mRNA体外转录模板质粒,命名为L23H02。Using saRNA-243 as a template plasmid, the luciferase coding sequence in it was replaced with the human papillomavirus (HPV) antigen gene Ag9.1 coding sequence through homologous recombination to obtain a self-amplified mRNA in vitro transcription template plasmid expressing Ag9.1, named L23H02.
2.表达Ag9.1的saRNA体外转录制备:2. Preparation of in vitro transcription of saRNA expressing Ag9.1:
a.体外转录模板质粒线性化a. Linearization of in vitro transcription template plasmid
依次加入8μg体外转录模板质粒质粒L23H04,BspQI 4μL,10×Digestion Buffer4μL,最后用双重蒸馏水将体系补足40μL,50℃孵育2小时线性化质粒,琼脂糖凝胶电泳检测质粒线性化完全后不纯化回收直接作为体外转录模板。Add 8 μg of in vitro transcription template plasmid L23H04, 4 μL of BspQI, and 4 μL of 10×Digestion Buffer in sequence, and finally make up the system to 40 μL with double distilled water. Incubate at 50°C for 2 hours to linearize the plasmid. After agarose gel electrophoresis to detect that the plasmid is completely linearized, it can be directly used as an in vitro transcription template without purification.
b.体外转录获得L23H02自扩增mRNAb. Obtaining L23H02 self-amplified mRNA by in vitro transcription
依次加入10×T7聚合酶缓冲液,7.5mM ATP,7.5mM GTP,7.5mM UTP,7.5mM CTP,1μg线性化体外转录模板,400U T7 RNA聚合酶,20U RNase I抑制剂,6mM帽类似物,补水至总体积20μL,37℃反应2小时。反应结束后加入2U DNase I酶,37℃反应15分钟。反应体系补水至50μL,加入25μL 7.5M氯化锂溶液混匀,-20℃静置处理30分钟后,13000g 4℃离心10分钟,弃上清后用70%乙醇清洗沉淀,13000g4℃离心2分钟,弃上清,重复清洗一次,吸去上清后溶解在30μL水中,得到表达Ag9.1的saRNA,命名为S23H02(序列如SEQ ID NO:21所示)。用分光光度计测定浓度后,使用Qsep400检测完整性,毛细管电泳检测结果如图7,结果显示体外转录mRNA大小符合预期,完整性为82%,无明显的短转录产物。Add 10×T7 polymerase buffer, 7.5mM ATP, 7.5mM GTP, 7.5mM UTP, 7.5mM CTP, 1 μg linearized in vitro transcription template, 400U T7 RNA polymerase, 20U RNase I inhibitor, 6mM cap analog, add water to a total volume of 20 μL, and react at 37°C for 2 hours. After the reaction, add 2U DNase I enzyme and react at 37°C for 15 minutes. The reaction system was added with water to 50 μL, 25 μL 7.5M lithium chloride solution was added and mixed, and after standing at -20°C for 30 minutes, centrifuged at 13000g 4°C for 10 minutes, the supernatant was discarded and the precipitate was washed with 70% ethanol, centrifuged at 13000g4°C for 2 minutes, the supernatant was discarded, and the washing was repeated once, the supernatant was removed and dissolved in 30 μL water to obtain saRNA expressing Ag9.1, named S23H02 (sequence as shown in SEQ ID NO: 21). After the concentration was determined by a spectrophotometer, the integrity was detected using Qsep400. The capillary electrophoresis detection results are shown in Figure 7, which showed that the size of the in vitro transcribed mRNA was consistent with expectations, the integrity was 82%, and there was no obvious short transcription product.
实施例6:表达Ag9.1的saRNA脂质纳米颗粒制剂的制备Example 6: Preparation of saRNA lipid nanoparticle formulations expressing Ag9.1
将实施例5制得的S23H02封装在四种组分脂质中以形成mRNA脂质纳米颗粒,具体为:使用商业化微流体装置NanoAssembler将溶解于乙醇中的中的脂质混合物(可电离脂质ALC-0315:二硬脂酰磷脂酰胆碱:胆固醇:PEG脂质ALC-0159)和溶解于柠檬酸盐缓冲液中的S23H02混合。混合完毕后,将脂质纳米颗粒与磷酸盐缓冲盐混合,并通过超滤除去残余乙醇。最后,将脂质纳米颗粒与冷冻保护剂一起储存在-80℃下获得脂质纳米颗粒制剂S23H02-LNP,用于下面的实验。The S23H02 prepared in Example 5 was encapsulated in four component lipids to form mRNA lipid nanoparticles, specifically: a lipid mixture (ionizable lipid ALC-0315: distearoylphosphatidylcholine: cholesterol: PEG lipid ALC-0159) dissolved in ethanol and S23H02 dissolved in citrate buffer were mixed using a commercial microfluidic device NanoAssembler. After mixing, the lipid nanoparticles were mixed with phosphate buffered saline and residual ethanol was removed by ultrafiltration. Finally, the lipid nanoparticles were stored at -80°C with a cryoprotectant to obtain a lipid nanoparticle preparation S23H02-LNP for the following experiments.
实施例7:表达HPV抗原的优化骨架saRNA与表达HPV抗原的mRNA的肿瘤Example 7: Tumors expressing optimized backbone saRNA of HPV antigens and mRNA expressing HPV antigens 治疗效果比较Comparison of treatment effects
本实施例所用药品见下面的表1。The drugs used in this example are shown in Table 1 below.
表1
Table 1
将S23H02、M22H04和阴性对照分别转染至含有脂质体3000的组织培养物。在图8中所标注的时间点收集上清液,使用检测E7域的ELISA试验来检测分泌的HPV16抗原的表达,结果如图8所示。S23H02针对特异性抗原显示出强于M22H04的体液免疫。S23H02, M22H04 and negative control were transfected into tissue culture containing lipofectamine 3000. Supernatants were collected at the time points marked in Figure 8, and the expression of secreted HPV16 antigens was detected using an ELISA test for detecting the E7 domain, and the results are shown in Figure 8. S23H02 showed stronger humoral immunity than M22H04 against specific antigens.
将C57bl/6小鼠用TC1肿瘤细胞进行攻毒,设为第0天;随后分别在第5、8、11天注射3剂治疗性疫苗;后腿肌肉注射。C57bl/6 mice were challenged with TC1 tumor cells, set as day 0; then three doses of therapeutic vaccine were injected on days 5, 8, and 11, respectively; injected into the hind leg muscle.
记录肿瘤体积和小鼠体重,结果分别如图9和图10所示。The tumor volume and mouse body weight were recorded, and the results are shown in Figures 9 and 10, respectively.
收集注射了1剂治疗性疫苗的C57bl/6小鼠的脾细胞。标记后用流式细胞仪检测脾脏中E7特异性的CD8T细胞的数量。结果如图11所示。S23H02针对特异性抗原显示出显著强于M22H04的细胞免疫。The spleen cells of C57bl/6 mice injected with one dose of therapeutic vaccine were collected. After labeling, the number of E7-specific CD8 T cells in the spleen was detected by flow cytometry. The results are shown in Figure 11. S23H02 showed significantly stronger cellular immunity than M22H04 against specific antigens.
以上实验结果显示,优化的saRNA疫苗与传统mRNA疫苗相比,可以以更低的剂量获取更好的治疗效果,显示出更高的抗原表达水平、更好的持久性和安全性。The above experimental results show that compared with traditional mRNA vaccines, the optimized saRNA vaccine can achieve better therapeutic effects at a lower dose, showing higher antigen expression levels, better durability and safety.
制备例1:六个不同位置分别插入肝特异性microRNA-122结合序列的自扩增Preparation Example 1: Self-amplification of liver-specific microRNA-122 binding sequences inserted at six different locations mRNA的制备Preparation of mRNA
1.MicroRNA结合位点设计:通过将不同数量的相同或不同microRNA成熟序列(5p或3p)以内部间隔序列(inner spacer)分隔后两端加入外部间隔序列(outer sapcer),最外侧为复制酶可识别的蛋白酶酶切位点,具体如图12。1. MicroRNA binding site design: Different numbers of identical or different microRNA mature sequences (5p or 3p) are separated by an inner spacer sequence and then external spacers are added to both ends. The outermost site is a protease cleavage site that can be recognized by the replicase, as shown in Figure 12.
2.自扩增mRNA结构设计:由甲病毒家族中委内瑞拉马脑脊髓炎病毒TC-83株基因组改造而来,具体为保留非结构蛋白即病毒复制酶序列及非翻译序列,包括病毒基因组5端、3端非翻译序列和非结构蛋白后的26S promoter,将甲病毒结构蛋白编码序列替换为任意靶蛋白序列X。同时,在5端甲病毒非翻译序列前加入T7聚合酶启动子位点,在3端甲病毒非翻译序列后加入多聚腺嘌呤序列(polyA序列),具体结构如图13。2. Design of self-amplifying mRNA structure: It is modified from the genome of Venezuelan equine encephalitis virus TC-83 strain in the alphavirus family. Specifically, the non-structural protein, namely the viral replicase sequence and the non-translated sequence, including the non-translated sequence at the 5-end and 3-end of the viral genome and the 26S promoter after the non-structural protein, is retained, and the alphavirus structural protein coding sequence is replaced with any target protein sequence X. At the same time, a T7 polymerase promoter site is added before the alphavirus non-translated sequence at the 5-end, and a polyadenine sequence (polyA sequence) is added after the alphavirus non-translated sequence at the 3-end. The specific structure is shown in Figure 13.
3.六个不同部位分别插入microRNA122序列的自扩增mRNA体外转录模板制 备:通过基因合成将T7聚合酶启动子位点、5端甲病毒非翻译序列、甲病毒复制酶序列、26S启动子序列、Nluc-EGFP报告基因序列、3端甲病毒非翻译序列、多聚腺嘌呤序列及限制性内切酶BspQI酶切位点序列构建到pUC57-Kan质粒中,得到自扩增mRNA质粒psaRNA-Nluc(荧光霉素)-EGFP(绿色荧光蛋白),命名为saRNA-EGFP(SEQ ID NO:29);之后通过无缝克隆技术将6个microRNA-122结合序列串联盒(SEQ ID NO:22)分别插入5端甲病毒非翻译序列和甲病毒复制酶序列nsP1之间、甲病毒复制酶序列nsP1和nsP2之间、甲病毒复制酶序列nsP2和nsP3之间、甲病毒复制酶序列nsP3和nsP4之间、26S启动子序列和EGFP报告基因序列之间以及EGFP报告基因序列和3端甲病毒非翻译序列之间,得到六个microRNA不同插入位置的自扩增mRNA体外转录模板质粒,分别命名为N21091、N21092、N21093、N21094、N21095和N21096,对应自扩增mRNA结构如图14,对应序列如SEQ ID NO:23至SEQ ID NO:28。3. Preparation of self-amplified mRNA in vitro transcription templates with microRNA122 sequences inserted in six different locations Preparation: The T7 polymerase promoter site, the 5-terminal alphavirus non-translated sequence, the alphavirus replicase sequence, the 26S promoter sequence, the Nluc-EGFP reporter gene sequence, the 3-terminal alphavirus non-translated sequence, the polyadenylation sequence and the restriction endonuclease BspQI restriction site sequence were constructed into the pUC57-Kan plasmid by gene synthesis to obtain the self-amplifying mRNA plasmid psaRNA-Nluc (fluorescein)-EGFP (green fluorescent protein), named saRNA-EGFP (SEQ ID NO: 29); then the 6 microRNA-122 binding sequence tandem cassettes (SEQ ID NO: 30) were inserted into the pUC57-Kan plasmid by seamless cloning technology. NO:22) were respectively inserted between the 5-terminal alphavirus non-translated sequence and the alphavirus replicase sequence nsP1, between the alphavirus replicase sequence nsP1 and nsP2, between the alphavirus replicase sequence nsP2 and nsP3, between the alphavirus replicase sequence nsP3 and nsP4, between the 26S promoter sequence and the EGFP reporter gene sequence, and between the EGFP reporter gene sequence and the 3-terminal alphavirus non-translated sequence to obtain six self-amplified mRNA in vitro transcription template plasmids with different microRNA insertion positions, which were named N21091, N21092, N21093, N21094, N21095 and N21096, respectively. The corresponding self-amplified mRNA structures are shown in Figure 14, and the corresponding sequences are shown in SEQ ID NO:23 to SEQ ID NO:28.
4.六个不同部位分别插入microRNA-122结合序列串联盒的自扩增mRNA体外转录制备:4. Preparation of self-amplified mRNA in vitro transcription with microRNA-122 binding sequence cassettes inserted in six different locations:
a.体外转录模板质粒线性化:具体为,依次加入8μg体外转录模板质粒质粒psaRNA-EGFP-microRNA,BspQI 4μL,10×Digestion Buffer 4μL,最后用ddH2O将体系补足40μL,50℃孵育2h线性化质粒。完成后,将酶切混合物体系用ddH2O补足至100μl,使用大量琼脂糖凝胶DNA回收试剂盒纯化回收线性化体外转录模板。a. Linearization of in vitro transcription template plasmid: Specifically, add 8 μg of in vitro transcription template plasmid psaRNA-EGFP-microRNA, 4 μL of BspQI, and 4 μL of 10×Digestion Buffer in sequence, and finally add ddH 2 O to the system to 40 μL, and incubate at 50°C for 2 hours to linearize the plasmid. After completion, add ddH 2 O to the enzyme digestion mixture system to 100 μl, and use a large amount of agarose gel DNA recovery kit to purify and recover the linearized in vitro transcription template.
b.体外转录获得自扩增mRNA:具体为,依次加入10×T7聚合酶缓冲液,7.5mM ATP,7.5mM GTP,7.5mM UTP,7.5mM CTP,1μg线性化体外转录模板,400U T7RNA聚合酶,20U RNase I抑制剂,6mM帽类似物,补水至总体积20μL,37℃反应2小时。反应结束后加入2U DNase I酶,37℃反应15分钟。反应体系补水至50μL,加入25μL 7.5M氯化锂溶液混匀,-20℃静置处理30分钟后,13000g 4℃离心10分钟,弃上清后用70%乙醇清洗沉淀,13000g 4℃离心2分钟,弃上清,重复清洗一次,吸去上清后溶解在30μL水中。用分光光度计测定浓后,取500ng体外转录得到的自扩增mRNA稀释至2μL后和2μL 2×RNA Loading Dye混合,于70℃孵育10分钟后,立即放置冰上冷却2min,进行变性琼脂糖凝胶电泳检测,体外转录mRNA大小符合预期,完成六个不同部位分别插入microRNA122序列的自扩增mRNA体外转录制备,电泳检测结果如图15,结果显示,成功制备了自扩增mRNA。 b. In vitro transcription to obtain self-amplified mRNA: Specifically, add 10×T7 polymerase buffer, 7.5mM ATP, 7.5mM GTP, 7.5mM UTP, 7.5mM CTP, 1μg linearized in vitro transcription template, 400U T7RNA polymerase, 20U RNase I inhibitor, 6mM cap analog, add water to a total volume of 20μL, and react at 37℃ for 2 hours. After the reaction, add 2U DNase I enzyme and react at 37℃ for 15 minutes. Add water to the reaction system to 50μL, add 25μL 7.5M lithium chloride solution and mix well, stand at -20℃ for 30 minutes, centrifuge at 13000g at 4℃ for 10 minutes, discard the supernatant and wash the precipitate with 70% ethanol, centrifuge at 13000g at 4℃ for 2 minutes, discard the supernatant, repeat the washing once, remove the supernatant and dissolve in 30μL water. After measuring the concentration with a spectrophotometer, 500 ng of the self-amplified mRNA obtained by in vitro transcription was diluted to 2 μL and mixed with 2 μL of 2× RNA Loading Dye. After incubation at 70°C for 10 minutes, it was immediately placed on ice for 2 minutes and detected by denaturing agarose gel electrophoresis. The size of the in vitro transcribed mRNA was consistent with expectations, and the in vitro transcription preparation of self-amplified mRNA with microRNA122 sequences inserted in six different locations was completed. The electrophoresis detection results are shown in Figure 15, and the results show that the self-amplified mRNA was successfully prepared.
实施例8:不同细胞中microRNA-122表达量检测Example 8: Detection of microRNA-122 expression in different cells
检测方法:逆转录荧光定量PCR检测C2C12和huh7.5.1细胞中miRNA122表达量:细胞样品使用RNA-easy Isolation Reagent提取总RNA后,紫外分光光度计进行RNA定量,使用miRNA 1st Strand cDNA Synthesis Kit(by stem-loop)完成逆转录后,利用miRNA Universal SYBR qPCR Master Mix进行荧光定量PCR实验。检测引物如下:Detection method: Reverse transcription fluorescence quantitative PCR was used to detect the expression of miRNA122 in C2C12 and huh7.5.1 cells: After the total RNA was extracted from the cell samples using RNA-easy Isolation Reagent, the RNA was quantified using a UV spectrophotometer. After reverse transcription was completed using the miRNA 1st Strand cDNA Synthesis Kit (by stem-loop), the fluorescence quantitative PCR experiment was performed using miRNA Universal SYBR qPCR Master Mix. The detection primers are as follows:
miRNA122上游引物F:5‘-CGCGTGGAGTGTGACAATGG-3’(SEQ ID NO:43),miRNA122 upstream primer F: 5'-CGCGTGGAGTGTGACAATGG-3' (SEQ ID NO: 43),
miRNA122下游引物R:5‘-AGTGCAGGGTCCGAGGTATT-3’(SEQ ID NO:44),miRNA122 downstream primer R: 5'-AGTGCAGGGTCCGAGGTATT-3' (SEQ ID NO: 44),
U6启动子上游引物F:5‘-CTCGCTTCGGCAGCACAT-3’(SEQ ID NO:45),U6 promoter upstream primer F: 5'-CTCGCTTCGGCAGCACAT-3' (SEQ ID NO: 45),
U6启动子下游引物R:5‘-TTTGCGTGTCATCCTTGCG-3’(SEQ ID NO:46)。U6 promoter downstream primer R: 5’-TTTGCGTGTCATCCTTGCG-3’ (SEQ ID NO:46).
将C2C12和huh7.5.1细胞的microRNA-122CT值与细胞RNA内参U6 CT值进行均一化后,不同细胞中microRNA-122的deltaCT检测结果如图16所示,Huh7.5.1细胞的deltaCT值比C2C12细胞高约15,说明microRNA-122在Huh7.5.1表达量远远高于C2C12细胞。After normalizing the microRNA-122 CT values of C2C12 and huh7.5.1 cells with the CT values of the cellular RNA internal reference U6, the deltaCT detection results of microRNA-122 in different cells are shown in Figure 16. The deltaCT value of Huh7.5.1 cells is about 15 higher than that of C2C12 cells, indicating that the expression level of microRNA-122 in Huh7.5.1 cells is much higher than that in C2C12 cells.
实施例9:六个不同位置分别插入肝特异性microRNA-122结合序列的自扩增Example 9: Self-amplification of liver-specific microRNA-122 binding sequences inserted at six different locations mRNA的体外表达及复制检测In vitro mRNA expression and replication assays
1.荧光蛋白及荧光素酶检测1. Fluorescent protein and luciferase detection
a.细胞接种:将microRNA-122高表达的Huh7.5.1细胞和microRNA-122低表达的C2C12细胞接种在75cm2细胞培养瓶中,培养基为DMEM高糖培养基+10%胎牛血清+1%双抗,待细胞在培养瓶中汇合度达到80%以上,用胰酶将细胞消化、计数。在24孔细胞培养板中铺入适宜数量的细胞,CO2培养箱37℃过夜培养。a. Cell inoculation: Huh7.5.1 cells with high expression of microRNA-122 and C2C12 cells with low expression of microRNA-122 were inoculated in 75cm2 cell culture flasks. The culture medium was DMEM high glucose medium + 10% fetal bovine serum + 1% double antibody. When the confluence of cells in the culture flask reached more than 80%, the cells were digested and counted with trypsin. An appropriate number of cells were plated in a 24-well cell culture plate and cultured overnight at 37°C in a CO2 incubator.
b.自扩增mRNA转染Huh7.5.1细胞和C2C12细胞:分别将0.5μg六个自扩增mRNA和无microRNA-122结合位点的对照组自扩增mRNA(SEQ ID NO:29)与50ng的表达萤火虫荧光酶素的线性mRNA混合后使用脂质体转染试剂-mRNA Transfection Kit转染至Huh7.5.1细胞和C2C12细胞,CO2培养箱37℃培养 24小时。b. Self-amplified mRNA transfection of Huh7.5.1 cells and C2C12 cells: 0.5 μg of six self-amplified mRNAs and a control self-amplified mRNA without microRNA-122 binding sites (SEQ ID NO: 29) were mixed with 50 ng of linear mRNA expressing firefly luciferase and then transfected using liposomes. -mRNA Transfection Kit was used to transfect Huh7.5.1 cells and C2C12 cells, and cultured in a CO2 incubator at 37°C 24 hours.
c.荧光蛋白表达情况验证:将转染24小时后的细胞培养板放置在荧光显微镜下进行荧光成像,结果如图17。c. Verification of fluorescent protein expression: 24 hours after transfection, the cell culture plate was placed under a fluorescence microscope for fluorescence imaging. The results are shown in Figure 17.
结果显示:在转染相同时间内,在microRNA-122低表达的C2C12细胞中N21091、N21093、N21094号自扩增mRNA都无明显表达,而N21092号及N21096号则正常表达,水平与对照自扩增mRNA相当。而在microRNA-122高表达的Huh7.5.1细胞中N21092号和N21096号都有明显的表达降低,说明在N21092、N21096号自扩增mRNA受到了细胞内microRNA的预期调控,有较好的细胞表达特异性。N21095号自扩增mRNA显示出与对照自扩增mRNA相似的表达情况,推断将microRNA-122结合位点置于相应位点并不影响自扩增mRNA的正常表达,但也未有效被microRNA调控。The results showed that: in the same transfection time, N21091, N21093, and N21094 self-amplified mRNAs were not significantly expressed in C2C12 cells with low expression of microRNA-122, while N21092 and N21096 were expressed normally, with the same level as the control self-amplified mRNA. In Huh7.5.1 cells with high expression of microRNA-122, the expression of N21092 and N21096 was significantly reduced, indicating that N21092 and N21096 self-amplified mRNAs were regulated by intracellular microRNAs as expected and had good cell expression specificity. N21095 self-amplified mRNA showed similar expression to the control self-amplified mRNA, inferring that placing the microRNA-122 binding site at the corresponding site did not affect the normal expression of the self-amplified mRNA, but was not effectively regulated by microRNA.
d.荧光素酶的检测:在转染24小时后,使用Nano-Glo Luciferase Assay System检测试剂,转移25μl Nano-Glo Luciferase Assay Reagent至96孔白色不透明底孔板,取25μl上清样品至孔中混合均匀,等待3分钟后使用酶标仪检测信号,检测结果如图18,该结果同样显示N21092、N21096号自扩增mRNA在microRNA-122高表达的Huh7.5.1细胞和microRNA-122低表达的C2C12细胞中表达量有明显的差异,说明其较好的细胞表达特异性。d. Luciferase detection: 24 hours after transfection, use Nano-Glo Luciferase Assay System detection reagent, transfer 25μl Nano-Glo Luciferase Assay Reagent to a 96-well white opaque bottom plate, take 25μl supernatant sample to the well and mix evenly, wait for 3 minutes and use an ELISA reader to detect the signal. The detection results are shown in Figure 18, which also show that the expression levels of N21092 and N21096 self-amplified mRNAs in Huh7.5.1 cells with high expression of microRNA-122 and C2C12 cells with low expression of microRNA-122 are significantly different, indicating their good cell expression specificity.
2.自扩增mRNA复制效率检测2. Self-amplification mRNA replication efficiency detection
a.细胞接种及自扩增mRNA转染:同荧光蛋白及荧光素酶检测。a. Cell seeding and self-amplified mRNA transfection: Same as fluorescent protein and luciferase detection.
b.细胞样品的处理:在转染2小时和24小时后,吸弃上清,使用500μl PBS润洗细胞一次,弃上清,加入500μl细胞裂解液并吹打收集至EP管,保存于-80℃冰箱。b. Processing of cell samples: 2 hours and 24 hours after transfection, discard the supernatant, rinse the cells once with 500μl PBS, discard the supernatant, add 500μl cell lysis buffer and collect by pipetting into EP tubes, and store in a -80℃ refrigerator.
c.逆转录荧光定量PCR检测saRNA的复制:细胞样品使用RNeasy Mini Kit提取总RNA后,紫外分光光度计进行RNA定量,使用HiScript III 1st Strand cDNA Synthesis Kit完成逆转录后,利用2×ChamQ Universal SYBR qPCR Master Mix进行荧光定量PCR实验。检测引物如下:c. Reverse transcription fluorescence quantitative PCR detection of saRNA replication: After the total RNA of the cell sample is extracted using RNeasy Mini Kit, RNA is quantified using a UV spectrophotometer. After reverse transcription is completed using HiScript III 1st Strand cDNA Synthesis Kit, a fluorescence quantitative PCR experiment is performed using 2×ChamQ Universal SYBR qPCR Master Mix. The detection primers are as follows:
甲病毒复制酶序列(nsp1)上游引物F:5‘-GACGGACCGACAAGTCTCTA-3’(SEQ ID NO:47),Primer F upstream of alphavirus replicase sequence (nsp1): 5'-GACGGACCGACAAGTCTCTA-3' (SEQ ID NO: 47),
甲病毒复制酶序列(nsp1)下游引物R:5‘-GGTGGTGTCAAAGCCTATCCA-3’(SEQ ID NO:48),Alphavirus replicase sequence (nsp1) downstream primer R: 5'-GGTGGTGTCAAAGCCTATCCA-3' (SEQ ID NO: 48),
EGFP序列上游引物F:5‘-AGCTGGAGTACAACTACA-3’(SEQ ID NO:49), EGFP sequence upstream primer F: 5'-AGCTGGAGTACAACTACA-3' (SEQ ID NO: 49),
EGFP下游引物R:5‘-CTGATCTTGAAGTTCACC-3’(SEQ ID NO:50),EGFP downstream primer R: 5'-CTGATCTTGAAGTTCACC-3' (SEQ ID NO: 50),
GAPDH序列上游引物F:5‘-GGTATCGTGGAAGGACTC-3’(SEQ ID NO:51),GAPDH sequence upstream primer F: 5'-GGTATCGTGGAAGGACTC-3' (SEQ ID NO: 51),
GAPDH下游引物R:5‘-GTAGAGGCAGGGATGATG-3’(SEQ ID NO:52)。GAPDH downstream primer R: 5’-GTAGAGGCAGGGATGATG-3’ (SEQ ID NO:52).
反应循环条件为:95℃30s-(95℃10s-60℃30s)×40个循环。The reaction cycle conditions were: 95°C 30s-(95°C 10s-60°C 30s)×40 cycles.
检测结果如图19和图20所示。The detection results are shown in Figures 19 and 20.
该结果显示,N21092号和N21096号自扩增mRNA在microRNA-122高表达的Huh7.5.1细胞和microRNA-122低表达的C2C12细胞中的RNA相对表达量均有明显的差异。在Huh7.5.1细胞中,N21092号和N21096号自扩增RNA的相对表达量较C2C12细胞均有明显下调,说明microRNA-122能够通过有效降解自扩增RNA水平来调控基因表达。The results showed that the relative expression of N21092 and N21096 self-amplified mRNAs in Huh7.5.1 cells with high expression of microRNA-122 and C2C12 cells with low expression of microRNA-122 were significantly different. In Huh7.5.1 cells, the relative expression of N21092 and N21096 self-amplified RNAs was significantly downregulated compared with C2C12 cells, indicating that microRNA-122 can regulate gene expression by effectively degrading the level of self-amplified RNA.
实施例10:插入不同拷贝数肝特异性microRNA-122结合序列的自扩增mRNAExample 10: Self-amplifying mRNA with different copy numbers of liver-specific microRNA-122 binding sequences inserted 的体外表达及复制检测In vitro expression and replication assay
1.自扩增mRNA结构如图21,分别将1、2、3或6个拷贝的microRNA-122结合序列分别置入Nsp1/2或3’UTR位点,得到插入不同拷贝数肝特异性microRNA-122结合序列的自扩增mRNA,制备方法参照制备例1。1. The structure of self-amplifying mRNA is shown in Figure 21. 1, 2, 3 or 6 copies of the microRNA-122 binding sequence are respectively placed into the Nsp1/2 or 3'UTR site to obtain self-amplifying mRNA with different copy numbers of liver-specific microRNA-122 binding sequences inserted. The preparation method refers to Preparation Example 1.
2.荧光蛋白及荧光素酶检测方法同实施例8。在不同细胞中,待测试自扩增mRNA与未置入microRNA-122结合序列的对照自扩增mRNA的相对表达量结果如图22所示。2. The fluorescent protein and luciferase detection method is the same as in Example 8. The relative expression results of the self-amplified mRNA to be tested and the control self-amplified mRNA without the microRNA-122 binding sequence in different cells are shown in FIG. 22 .
结果显示,在microRNA-122高表达的Huh7.5.1细胞中,置入microRNA-122结合的自扩增mRNA其相对表达量和所插入的microRNA-122结合序列拷贝数成反比,相同插入位置下,分别插入1、2、3或6个拷贝microRNA-122结合序列的自扩增RNA表达量逐渐降低,其中插入6个拷贝microRNA-122结合序列的自扩增RNA表达量下降比例最高,相比野生型均下降了超过90%,并且将6个microRNA-122结合序列置于3’UTR的自扩增mRNA在microRNA-122低表达的C2C12细胞中的相对表达量则仍超过70%。The results showed that in Huh7.5.1 cells with high expression of microRNA-122, the relative expression level of self-amplifying mRNA bound to microRNA-122 was inversely proportional to the number of copies of the inserted microRNA-122 binding sequence. At the same insertion position, the expression level of self-amplifying RNA with 1, 2, 3 or 6 copies of the microRNA-122 binding sequence inserted gradually decreased, among which the expression level of self-amplifying RNA with 6 copies of the microRNA-122 binding sequence inserted decreased the most, which was reduced by more than 90% compared with the wild type. In addition, the relative expression level of self-amplifying mRNA with 6 microRNA-122 binding sequences placed in the 3'UTR in C2C12 cells with low expression of microRNA-122 was still more than 70%.
结果还显示,分别插入1、2或3个拷贝microRNA-122结合序列的自扩增RNA,其在Huh7.5.1细胞中的表达量相比野生型也有一定程度的下降,且在C2C12细胞中保持了较高的表达量。 The results also showed that the expression levels of self-amplified RNA with 1, 2 or 3 copies of microRNA-122 binding sequence inserted in Huh7.5.1 cells were also reduced to a certain extent compared with the wild type, and maintained a high expression level in C2C12 cells.
综上可见,自扩增mRNA表达量的调控以剂量依赖的方式基于microRNA-122,且3’UTR为较优的microRNA结合序列放置位点。In summary, the regulation of self-amplified mRNA expression is based on microRNA-122 in a dose-dependent manner, and the 3’UTR is the optimal microRNA binding sequence placement site.
实施例11:saRNA编码的宏结构域突变设计及ADP核糖水解酶活性的检测Example 11: Design of macrodomain mutations encoded by saRNA and detection of ADP ribose hydrolase activity
本发明选择了由委内瑞拉马脑炎病毒(Venezuelan Equine Encephalitis Virus,VEEV)衍生的saRNA进行后续设计,在VEEV为代表的甲病毒家族中,宏结构域存在于非结构蛋白3(nonstructural protein 3)的N端,如图24。宏结构域氨基酸序列在不同病毒之间都较为保守,同源性超过50%,因此我们分别选择了靠近ADP核糖水解活性中心的113位氨基酸及或远离ADP核糖水解活性中心的48位氨基酸进行氨基酸突变,具体位置见图25。在冠状病毒和甲病毒中第113位编码异亮氨酸(I)或缬氨酸(V),保守性高,我们推断其涉及ADP核糖水解功能。而48位则差异较大,仅在VEEV中编码为谷氨酰胺(Q),因此可能并不参与ADP核糖水解功能。由于苯丙氨酸和脯氨酰胺侧链都为环状结构,与侧链仅带有氨基基团的谷氨酰胺或带有一个甲基和一个乙基基团的异亮氨酸有明显结构差异,因此我们通过同源重组及体外转录,分别构建了以下三种saRNA:
The present invention selected saRNA derived from Venezuelan Equine Encephalitis Virus (VEEV) for subsequent design. In the alphavirus family represented by VEEV, the macrodomain is present at the N-terminus of nonstructural protein 3, as shown in Figure 24. The amino acid sequence of the macrodomain is relatively conservative between different viruses, with a homology of more than 50%. Therefore, we selected the 113th amino acid close to the ADP ribose hydrolysis active center and the 48th amino acid away from the ADP ribose hydrolysis active center for amino acid mutation, and the specific positions are shown in Figure 25. In coronaviruses and alphaviruses, the 113th position encodes isoleucine (I) or valine (V), which is highly conservative. We infer that it involves the ADP ribose hydrolysis function. The 48th position is quite different, and is only encoded as glutamine (Q) in VEEV, so it may not be involved in the ADP ribose hydrolysis function. Since the side chains of phenylalanine and prolineamide are both cyclic structures, they have obvious structural differences from glutamine with only an amino group on the side chain or isoleucine with a methyl group and an ethyl group. Therefore, we constructed the following three saRNAs through homologous recombination and in vitro transcription:
接下来将saRNA-190、saRNA-191及野生型saRNA各200ng转染至hela细胞中,24小时后,用抗mono-ADP-核糖基化的抗体检测细胞内总蛋白ADP-核糖基化的水平。如图26结果显示,在转染24小时后,相比野生型转染组,190/191-saRNA的细胞蛋白ADP-核糖基化明显更强,说明saRNA-190和saRNA-191的ADP核糖基水解酶活性相对于野生型saRNA明显降低。该数据说明,对宏结构域的113位及48位氨基酸突变设计均成功降低了ADP核糖基水解酶活性。 Next, 200 ng of saRNA-190, saRNA-191 and wild-type saRNA were transfected into Hela cells. After 24 hours, the level of ADP-ribosylation of total protein in the cells was detected with an anti-mono-ADP-ribosylation antibody. As shown in Figure 26, 24 hours after transfection, the ADP-ribosylation of 190/191-saRNA cell proteins was significantly stronger than that of the wild-type transfection group, indicating that the ADP ribosyl hydrolase activity of saRNA-190 and saRNA-191 was significantly reduced relative to that of wild-type saRNA. The data show that the design of amino acid mutations at positions 113 and 48 of the macrodomain successfully reduced the ADP ribosyl hydrolase activity.
实施例12:ADP核糖水解酶活性受损的突变saRNA的复制水平检测Example 12: Detection of replication levels of mutant saRNAs with impaired ADP ribose hydrolase activity
在验证了两种突变saRNA的ADP核糖水解酶活性降低后,我们检测了突变saRNA的复制能力是否会发生变化。将saRNA-190、saRNA-191及野生型saRNA转染至hela细胞后2,6,24和48小时,分别提取RNA,对saRNA所编码的EGFP基因进行荧光定量PCR。After verifying that the ADP ribose hydrolase activity of the two mutant saRNAs was reduced, we tested whether the replication ability of the mutant saRNAs would change. After transfecting saRNA-190, saRNA-191 and wild-type saRNA into Hela cells for 2, 6, 24 and 48 hours, RNA was extracted and the EGFP gene encoded by saRNA was subjected to fluorescence quantitative PCR.
如图27结果显示,190/191-saRNA相比于野生型saRNA,在编码EGFP的RNA水平方面明显降低。同时,我们用抗双链RNA抗体对野生型saRNA、saRNA-190和saRNA-191转染后6小时和24小时后hela细胞中的双链RNA进行了流式细胞检测。如图28结果显示,相对于野生型saRNA,saRNA-190和saRNA-191在转染后6小时后,复制过程中形成的双链RNA比例均较低,并且这种差异在转染后24小时则更为显著,在转染后24小时有超过半数的转染了野生型saRNA的细胞中都检测到了dsRNA,远高于另外两组。而190-saRNA和191-saRNA之间则无显著性差异。总之,以上结果表明,带有宏结构域氨基酸突变的190/191-saRNA在体外细胞上的复制能力减弱。As shown in Figure 27, 190/191-saRNA significantly reduced the level of RNA encoding EGFP compared to wild-type saRNA. At the same time, we used anti-double-stranded RNA antibodies to perform flow cytometry on double-stranded RNA in hela cells 6 hours and 24 hours after transfection of wild-type saRNA, saRNA-190 and saRNA-191. As shown in Figure 28, compared with wild-type saRNA, saRNA-190 and saRNA-191 had a lower proportion of double-stranded RNA formed during replication 6 hours after transfection, and this difference was more significant 24 hours after transfection. More than half of the cells transfected with wild-type saRNA were detected with dsRNA 24 hours after transfection, which was much higher than the other two groups. There was no significant difference between 190-saRNA and 191-saRNA. In summary, the above results show that the replication ability of 190/191-saRNA with macrodomain amino acid mutations in in vitro cells is weakened.
实施例13:突变saRNA所诱导的天然免疫差异检测Example 13: Detection of differences in natural immunity induced by mutant saRNA
考虑到双链RNA能够显著触发天然免疫反应,因此需要检查突变saRNA与野生型saRNA诱导的天然免疫水平变化。我们将190/191-saRNA及野生型saRNA转染hela细胞24小时后,提取细胞总RNA进行RNA测序,分析转录组水平差异。Considering that double-stranded RNA can significantly trigger the innate immune response, it is necessary to examine the changes in the innate immune level induced by mutant saRNA and wild-type saRNA. After transfecting Hela cells with 190/191-saRNA and wild-type saRNA for 24 hours, we extracted total cell RNA for RNA sequencing and analyzed the differences in transcriptome levels.
通过主成分分析结果看,相比对照组,转染了190-saRNA的转录组差异最小,而野生型saRNA组诱导了非常显著的转录组变化(见图29)。进一步地,我们统计了不同saRNA组相对于对照组中出现差异表达的基因数量。我们发现在所有saRNA组中,上调的基因数量均远多于下调基因,而190和191组大部分上调的基因与野生型saRNA组几乎一致,然而我们发现不同组间共同下调的基因则数量较少(见图30),提示190/191-saRNA与野生型saRNA上调基因所参与的生物学功能可能相似,因此对野生型saRNA组相对于对照组的差异基因进行了聚类分析,结果显示在生物学过程(biological process,GO:BP)中差异最显著的10组几乎都与免疫反应相关,而且其中几乎所有基因都高度上调(见图31a),说明野生型saRNA诱导了细胞强烈的抗病毒天然免疫反应。进一步对差异表达基因的分子功能(molecular function,GO:MF)进行分析发现,上调基因的功 能与细胞因子/趋化因子/生长因子/受体结合以及双链RNA/单链RNA结合(cytokine/chemokine/growth factor/receptor binding and dsRNA/ssRNA binding)相关(见图31b)。已知190/191-saRNA转染后形成双链RNA的细胞比例明显低于野生型RNA,因此不难联想到不同saRNA复制时形成双链RNA的比例差异可能会影响天然免疫反应的诱导。According to the results of principal component analysis, compared with the control group, the transcriptome difference of the transfected 190-saRNA was the smallest, while the wild-type saRNA group induced very significant transcriptome changes (see Figure 29). Further, we counted the number of genes that were differentially expressed in different saRNA groups relative to the control group. We found that in all saRNA groups, the number of upregulated genes was far greater than the downregulated genes, and most of the upregulated genes in the 190 and 191 groups were almost the same as the wild-type saRNA group. However, we found that the number of genes that were downregulated in common between different groups was small (see Figure 30), suggesting that the biological functions involved in the upregulated genes of 190/191-saRNA and wild-type saRNA may be similar. Therefore, the differential genes of the wild-type saRNA group relative to the control group were clustered. The results showed that the 10 groups with the most significant differences in biological processes (GO:BP) were almost all related to immune response, and almost all of them were highly upregulated (see Figure 31a), indicating that wild-type saRNA induced a strong antiviral innate immune response in cells. Further analysis of the molecular function (GO: MF) of the differentially expressed genes revealed that the function of the up-regulated genes It can be related to cytokine/chemokine/growth factor/receptor binding and dsRNA/ssRNA binding (see Figure 31b). It is known that the proportion of cells that form double-stranded RNA after 190/191-saRNA transfection is significantly lower than that of wild-type RNA. Therefore, it is not difficult to associate the difference in the proportion of double-stranded RNA formed during the replication of different saRNAs may affect the induction of natural immune response.
接下来我们检查了双链RNA/单链RNA结合蛋白中上调的基因,我们看到包括胞内RNA传感器RIG-I、MDA-5及内体RNA传感器TLR3以及IFN通路下游干扰素刺激基因如OAS家族、EIF2AK2的高度上调(见图32),说明saRNA复制中产生的双链RNA是激活干扰素或NFKB信号通路的关键,从而诱导下游大量抗病毒基因以抑制saRNA复制。从对不同组之间干扰素通路的基因热图中可以看到,相对于对照组,所有转染了saRNA的细胞中干扰素通路基因的转录水平都有明显上调,除提到的RNA结合蛋白以外,还有干扰素beta,转录因子IRF7/9,STAT1以及众多的下游的干扰素刺激基因。另外也看到了细胞因子的上调,提示了NF-kb通路也被有效激活。与预期一致,不同组saRNA所诱导的干扰素信号通路基因转录水平明显不同,相比对照组,190诱导了最弱的IFN免疫反应,191稍强,而野生型则最强。如图33,通过荧光定量PCR对转染后2、6及24小时后,干扰素通路及细胞因子的mRNA水平进行了验证,发现2小时内,各组的RNA转录水平相比对照组都没有发生变化,而在6小时,相比对照组,野生saRNA组IFN-beta,CXCL10,CCL5以及IFIT2 RNA水平显著上调,并且CXCL10,IFIT2和PKR在190和191组也发生上调,但190和191间并无差异。在转染后24小时后,包括NF-KB和干扰素通路的所有RNA在野生型saRNA组上调最显著,191-saRNA次之,190-saRNA最低。Next, we checked the upregulated genes in the double-stranded RNA/single-stranded RNA binding protein. We saw that the intracellular RNA sensor RIG-I, MDA-5 and the endosomal RNA sensor TLR3 as well as the interferon-stimulated genes downstream of the IFN pathway, such as the OAS family and EIF2AK2, were highly upregulated (see Figure 32), indicating that the double-stranded RNA produced in saRNA replication is the key to activating the interferon or NFKB signaling pathway, thereby inducing a large number of downstream antiviral genes to inhibit saRNA replication. From the gene heat map of the interferon pathway between different groups, it can be seen that compared with the control group, the transcription levels of interferon pathway genes in all cells transfected with saRNA were significantly upregulated. In addition to the RNA binding proteins mentioned, there were interferon beta, transcription factors IRF7/9, STAT1 and many downstream interferon-stimulated genes. In addition, the upregulation of cytokines was also seen, suggesting that the NF-kb pathway was also effectively activated. As expected, the transcription levels of interferon signaling pathway genes induced by saRNA in different groups were significantly different. Compared with the control group, 190 induced the weakest IFN immune response, 191 was slightly stronger, and the wild type was the strongest. As shown in Figure 33, the mRNA levels of interferon pathway and cytokines were verified by fluorescence quantitative PCR 2, 6 and 24 hours after transfection. It was found that within 2 hours, the RNA transcription levels of each group did not change compared with the control group, while at 6 hours, the IFN-beta, CXCL10, CCL5 and IFIT2 RNA levels in the wild-type saRNA group were significantly upregulated compared with the control group, and CXCL10, IFIT2 and PKR were also upregulated in the 190 and 191 groups, but there was no difference between 190 and 191. 24 hours after transfection, all RNAs including NF-KB and interferon pathways were most significantly upregulated in the wild-type saRNA group, followed by 191-saRNA, and the lowest in 190-saRNA.
总之,检测到了不同saRNA所诱导的天然免疫通路基因转录水平与其复制能力正相关,由于190/191-saRNA复制及形成双链RNA的能力减弱,因此也就诱导了较弱的天然免疫反应。In summary, it was detected that the transcription levels of genes in the innate immune pathway induced by different saRNAs were positively correlated with their replication ability. Since the ability of 190/191-saRNA to replicate and form double-stranded RNA was weakened, it induced a weaker innate immune response.
实施例14:突变saRNA在体外细胞模型的蛋白翻译活性检测Example 14: Detection of protein translation activity of mutant saRNA in in vitro cell models
为了检测不同saRNA表达外源基因的活性,将编码荧光酶素的不同saRNA转染hela细胞。结果显示,在2小时及6小时野生型saRNA的表达量都要高于突变saRNA,但在24小时,190/191-saRNA的表达量反而略高,而在48小时,190-saRNA的表达量 比野生型saRNA高6倍(见图34)。推测saRNA所诱导的天然免疫反应会通过宿主蛋白翻译抑制从而限制病毒劫持细胞翻译机制进行复制,因此检测了不同saRNA转染后细胞翻译活性。收集细胞前将嘌呤霉素加入培养基至终浓度5ug/mL处理5分钟使其掺入多肽链,用抗嘌呤霉素抗体检测胞内被标记的蛋白含量。结果显示在转染2小时后,细胞内翻译活性并没有明显区别,而在6小时及24小时,野生型saRNA组翻译活性均弱于突变saRNA组,而对于190-saRNA蛋白翻译活性在6小时稍微弱于对照,但在24h与对照组相似,相比之下转染191-saRNA组细胞翻译活力在24h稍弱(见图35)。该结果证明了saRNA转染后,细胞内的蛋白翻译发生了抑制,结合之前观察到dsRNA结合蛋白中PKR及OAS基因在saRNA转染后显著上调,推测可能OAS/RNase L及PKR/eIF2α途径的激活参与了宿主蛋白翻译抑制,从而限制了saRNA的表达。因此检测了转染saRNA后不同时间点核糖体RNA的完整性及eIF2α磷酸化(见图36、37)。结果显示,转染了野生型saRNA的细胞核糖体RNA在2、6小时内仍然较为完整,但在24小时及48小时则发生了严重的降解,而转染了190/191saRNA的核糖体RNA完整性相对于对照组未显著性降低。该结果提示野生型saRNA转染后核糖体RNA发生了降解,这是OAS/RNase L途径的激活的结果(见图36)。同时,eIF2α磷酸化检测结果显示,转染6小时后各组saRNA均诱导了eIF2α磷酸化,且与6h的亚基因组水平成正相关,野生型saRNA最强,191-saRNA最弱。出于意料的是,在转染后24小时,eIF2α磷酸化水平均明显降低,野生型saRNA诱导的eIF2α磷酸化甚至弱于对照组(即mock组:仅加入转染试剂,并未转染任何RNA分子),而突变saRNA的eIF2α磷酸化程度与对照组相似(见图37)。综合以上结果分析发现,在转染2小时宿主翻译并未受到抑制,而在6小时出现了由PKR/eIF2α介导的宿主翻译,但此时由于野生型saRNA亚基因组RNA扩增水平远高于突变saRNA,因此此时表达水平仍较其他两组更高。在24小时,野生型eIF2α磷酸化水平降低,因此宿主翻译抑制很大程度由OAS/RNase L激活导致的rRNA降解促进,因此受到抑制最显著的野生型saRNA的表达水平最低。In order to detect the activity of exogenous genes expressed by different saRNAs, different saRNAs encoding luciferase were transfected into HeLa cells. The results showed that the expression level of wild-type saRNA was higher than that of mutant saRNA at 2 hours and 6 hours, but the expression level of 190/191-saRNA was slightly higher at 24 hours, and the expression level of 190-saRNA was higher at 48 hours. 6 times higher than wild-type saRNA (see Figure 34). It is speculated that the natural immune response induced by saRNA will limit the virus hijacking the cell translation mechanism for replication through host protein translation inhibition, so the cell translation activity after different saRNA transfections was detected. Before collecting cells, puromycin was added to the culture medium to a final concentration of 5ug/mL and treated for 5 minutes to incorporate it into the polypeptide chain, and the intracellular labeled protein content was detected with anti-puromycin antibodies. The results showed that there was no significant difference in intracellular translation activity 2 hours after transfection, while at 6 hours and 24 hours, the translation activity of the wild-type saRNA group was weaker than that of the mutant saRNA group, and the 190-saRNA protein translation activity was slightly weaker than the control at 6 hours, but similar to the control group at 24h. In contrast, the translation activity of cells in the transfected 191-saRNA group was slightly weaker at 24h (see Figure 35). The results prove that after saRNA transfection, protein translation in cells is inhibited. Combined with the previous observation that PKR and OAS genes in dsRNA binding proteins are significantly upregulated after saRNA transfection, it is speculated that the activation of OAS/RNase L and PKR/eIF2α pathways may be involved in the inhibition of host protein translation, thereby limiting the expression of saRNA. Therefore, the integrity of ribosomal RNA and eIF2α phosphorylation at different time points after saRNA transfection were detected (see Figures 36 and 37). The results showed that the ribosomal RNA of cells transfected with wild-type saRNA was still relatively intact within 2 and 6 hours, but severely degraded at 24 and 48 hours, while the integrity of ribosomal RNA transfected with 190/191saRNA was not significantly reduced relative to the control group. The results suggest that ribosomal RNA is degraded after wild-type saRNA transfection, which is the result of activation of the OAS/RNase L pathway (see Figure 36). At the same time, the results of eIF2α phosphorylation detection showed that after 6 hours of transfection, each group of saRNA induced eIF2α phosphorylation, and was positively correlated with the subgenomic level of 6h, with wild-type saRNA being the strongest and 191-saRNA being the weakest. Unexpectedly, 24 hours after transfection, the phosphorylation level of eIF2α was significantly reduced, and the phosphorylation of eIF2α induced by wild-type saRNA was even weaker than that of the control group (i.e., the mock group: only the transfection reagent was added, and no RNA molecules were transfected), while the phosphorylation degree of eIF2α of the mutant saRNA was similar to that of the control group (see Figure 37). Based on the above results, it was found that host translation was not inhibited at 2 hours of transfection, and host translation mediated by PKR/eIF2α appeared at 6 hours, but at this time, because the subgenomic RNA amplification level of wild-type saRNA was much higher than that of mutant saRNA, the expression level was still higher than that of the other two groups. At 24 hours, wild-type eIF2α phosphorylation levels were reduced, and thus host translational repression was largely promoted by rRNA degradation caused by OAS/RNase L activation, so the wild-type saRNA that was most significantly inhibited had the lowest expression level.
以上结果证明saRNA转染细胞后激活了PKR/eIF2α和OAS/RNase L介导的宿主翻译抑制,但相对于野生型saRNA,突变saRNA翻译抑制程度较低,因此展现了更高的表达水平。The above results prove that saRNA transfection of cells activated PKR/eIF2α and OAS/RNase L-mediated host translation inhibition, but compared with wild-type saRNA, the mutant saRNA had a lower degree of translation inhibition and therefore showed a higher expression level.
实施例15:突变saRNA在体外细胞模型的凋亡诱导检测Example 15: Apoptosis induction detection of mutant saRNA in in vitro cell models
观察到在转染了野生型saRNA 24小时后,细胞核发生了明显的崩解,这提示hela细胞可能发生了凋亡(见图38)。对转染24小时及48小时的细胞进行了annexin-V及PI染色,如图39所示,转染了saRNA的细胞都发生细胞凋亡,但转染了190-saRNA组的死亡细胞比例明显低于其他组。通过对凋亡起始分子caspase 8及效应分子caspase 3的蛋白表达水平以及活化状态进行western blot检测,结果显示所有caspase在转染了saRNA的细胞中都发生了明显的活化,但野生型saRNA组的caspase活化明显强于其他组(见图40)。此外,如图41,用caspase抑制剂处理转染了野生型saRNA的细胞,发现经处理后细胞凋亡被强烈抑制,并且saRNA所表达的EGFP荧光强度提升了3倍。这些结果说明,saRNA转染hela细胞后导致凋亡,而抑制凋亡可以提高saRNA的表达效率。It was observed that 24 hours after the wild-type saRNA was transfected, the cell nucleus collapsed significantly, suggesting that Hela cells may have undergone apoptosis (see Figure 38). Annexin-V and PI staining were performed on cells transfected for 24 hours and 48 hours. As shown in Figure 39, all cells transfected with saRNA underwent apoptosis, but the proportion of dead cells in the 190-saRNA transfected group was significantly lower than that in other groups. Western blot was performed to detect the protein expression level and activation state of the apoptosis initiator molecule caspase 8 and the effector molecule caspase 3. The results showed that all caspases were significantly activated in cells transfected with saRNA, but the caspase activation in the wild-type saRNA group was significantly stronger than that in other groups (see Figure 40). In addition, as shown in Figure 41, cells transfected with wild-type saRNA were treated with caspase inhibitors, and it was found that cell apoptosis was strongly inhibited after treatment, and the fluorescence intensity of EGFP expressed by saRNA increased by 3 times. These results show that saRNA transfection of Hela cells leads to apoptosis, and inhibiting apoptosis can improve the expression efficiency of saRNA.
实施例16:190-saRNA的体内表达水平检测Example 16: Detection of in vivo expression level of 190-saRNA
为了验证了突变saRNA是否在体内也有较高的表达水平,用纳米脂质颗粒(lipid nanoparticles,LNP)包封了表达萤火虫荧光酶素基因的190-saRNA、野生型saRNA及非复制mRNA(nrmRNA),如图42a所示,分别用5μg表达萤火虫荧光酶素基因的不同mRNA-LNP肌内免疫4-6周龄的balb/c小鼠,在第1、3、7、14、21天进行活体成像。结果发现,非复制mRNA在免疫后第1天表达量均略高于saRNA,但在之后开始衰减,在第7天后几乎检测不到表达(见图42b、43)。而190-saRNA在至少7天内表达水平持续上升,且高于野生型saRNA,虽然在14天后与野生型saRNA维持一致,但总表达量均高于野生型saRNA及非复制mRNA(见图44)。与在体外观察到的一致,190-saRNA在体内也体现了更高的表达水平。In order to verify whether the mutant saRNA also has a high expression level in vivo, 190-saRNA, wild-type saRNA and non-replicating mRNA (nrmRNA) expressing the firefly luciferase gene were encapsulated with nanolipid particles (LNP). As shown in Figure 42a, 4-6 week-old balb/c mice were intramuscularly immunized with 5 μg of different mRNA-LNP expressing the firefly luciferase gene, and live imaging was performed on days 1, 3, 7, 14, and 21. The results showed that the expression level of non-replicating mRNA was slightly higher than that of saRNA on the first day after immunization, but it began to decay afterwards, and almost no expression was detected after the 7th day (see Figures 42b and 43). The expression level of 190-saRNA continued to rise for at least 7 days, and was higher than that of wild-type saRNA. Although it remained consistent with wild-type saRNA after 14 days, the total expression level was higher than that of wild-type saRNA and non-replicating mRNA (see Figure 44). Consistent with what was observed in vitro, 190-saRNA also showed higher expression levels in vivo.
尽管本发明的具体实施方式已经得到详细的描述,本领域技术人员将会理解。根据已经公开的所有教导,可以对那些细节进行各种修改和替换,这些改变均在本发明的保护范围之内。本发明的全部范围由所附权利要求及其任何等同物给出。 Although the specific embodiments of the present invention have been described in detail, it will be understood by those skilled in the art. According to all the teachings disclosed, various modifications and replacements can be made to those details, and these changes are all within the protection scope of the present invention. The full scope of the present invention is given by the attached claims and any equivalents thereof.
Claims (75)
Applications Claiming Priority (6)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN202310890565.0 | 2023-07-19 | ||
| CN202310890565 | 2023-07-19 | ||
| CN202311309636.X | 2023-10-10 | ||
| CN202311309636 | 2023-10-10 | ||
| CN202410268968.6 | 2024-03-08 | ||
| CN202410268968.6A CN118147171B (en) | 2024-03-08 | 2024-03-08 | Self-amplifying mRNA nucleic acid sequences with enhanced expression levels of exogenous genes |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025016031A1 true WO2025016031A1 (en) | 2025-01-23 |
Family
ID=94281087
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2024/093076 Pending WO2025016031A1 (en) | 2023-07-19 | 2024-05-14 | Self-amplifying nucleic acid molecule and use thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025016031A1 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN120555511A (en) * | 2025-05-29 | 2025-08-29 | 杭州依思康医药科技有限公司 | An mRNA transcription backbone vector and its application in preparing self-amplifying mRNA vaccines |
Citations (7)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN110612116A (en) * | 2017-05-08 | 2019-12-24 | 磨石肿瘤生物技术公司 | Alphavirus neoantigen vector |
| CN110643632A (en) * | 2019-09-26 | 2020-01-03 | 中国科学院武汉病毒研究所 | Rabies virus infectious clone based on alphavirus replicon vector and preparation method and application thereof |
| CN113227122A (en) * | 2018-10-08 | 2021-08-06 | 杨森制药公司 | Alphavirus-based replicons for administration of biotherapeutic agents |
| WO2021245090A1 (en) * | 2020-06-04 | 2021-12-09 | BioNTech SE | Rna replicon for versatile and efficient gene expression |
| CN113846113A (en) * | 2021-09-09 | 2021-12-28 | 臻赫医药(杭州)有限公司 | Limited self-replicating mRNA molecular system, preparation method and application |
| US20220265807A1 (en) * | 2019-06-18 | 2022-08-25 | Imperial College Innovations Limited | Rna construct |
| CN116390752A (en) * | 2020-06-19 | 2023-07-04 | 纪费亚斯疫苗公司 | Self-amplifying SARS-COV-2 RNA vaccine |
-
2024
- 2024-05-14 WO PCT/CN2024/093076 patent/WO2025016031A1/en active Pending
Patent Citations (7)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN110612116A (en) * | 2017-05-08 | 2019-12-24 | 磨石肿瘤生物技术公司 | Alphavirus neoantigen vector |
| CN113227122A (en) * | 2018-10-08 | 2021-08-06 | 杨森制药公司 | Alphavirus-based replicons for administration of biotherapeutic agents |
| US20220265807A1 (en) * | 2019-06-18 | 2022-08-25 | Imperial College Innovations Limited | Rna construct |
| CN110643632A (en) * | 2019-09-26 | 2020-01-03 | 中国科学院武汉病毒研究所 | Rabies virus infectious clone based on alphavirus replicon vector and preparation method and application thereof |
| WO2021245090A1 (en) * | 2020-06-04 | 2021-12-09 | BioNTech SE | Rna replicon for versatile and efficient gene expression |
| CN116390752A (en) * | 2020-06-19 | 2023-07-04 | 纪费亚斯疫苗公司 | Self-amplifying SARS-COV-2 RNA vaccine |
| CN113846113A (en) * | 2021-09-09 | 2021-12-28 | 臻赫医药(杭州)有限公司 | Limited self-replicating mRNA molecular system, preparation method and application |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN120555511A (en) * | 2025-05-29 | 2025-08-29 | 杭州依思康医药科技有限公司 | An mRNA transcription backbone vector and its application in preparing self-amplifying mRNA vaccines |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Fang et al. | Advances in COVID-19 mRNA vaccine development | |
| Le et al. | Nucleic acid-based technologies targeting coronaviruses | |
| US9737620B2 (en) | Vectors and methods for genetic immunization | |
| EP4289956A1 (en) | Cis-replicon construct | |
| US20250249053A1 (en) | Using infectious nucleic acid to treat cancer | |
| WO2023056980A1 (en) | Design of self-replicating rna molecules and application thereof | |
| Wei et al. | Technological breakthroughs and advancements in the application of mRNA vaccines: a comprehensive exploration and future prospects | |
| WO2025016031A1 (en) | Self-amplifying nucleic acid molecule and use thereof | |
| Koppu et al. | Current perspectives and future prospects of mRNA vaccines against viral diseases: a brief review | |
| CN118147171B (en) | Self-amplifying mRNA nucleic acid sequences with enhanced expression levels of exogenous genes | |
| Penza et al. | The long-lasting enigma of polycytidine (polyC) tract | |
| WO2023109975A2 (en) | Rna replicon for improving gene expression and use thereof | |
| CN110088273B (en) | Vaccines against infectious diseases caused by positive-strand RNA viruses | |
| WO2022026885A2 (en) | Modified chikungunya viruses and sindbis viruses and uses thereof | |
| Khorshid Sokhangouy et al. | mRNA Vaccines: Design Principles, Mechanisms, and Manufacturing—Insights From COVID‐19 as a Model for Combating Infectious Diseases | |
| CN120379688A (en) | Self-replicating RNA and application thereof | |
| CN119679932A (en) | Prolonged efficacy self-replicating nucleic acid vaccine for the new coronavirus | |
| CN117247954A (en) | Circular RNA vaccine, novel circular RNA and preparation method thereof | |
| Zhang et al. | Research progress of mRNA vaccines for infectious diseases | |
| US20250281602A1 (en) | Replicon Compositions and Methods of Using Same for the Treatment of Diseases | |
| TW202526021A (en) | Systems and compositions comprising highly active trans-amplifying replicases | |
| WO2025157249A1 (en) | Self-replicating rna and use thereof | |
| KR20250145021A (en) | nucleic acid molecules | |
| WO2024242637A1 (en) | A novel self-amplifying rna | |
| WO2024231810A1 (en) | Vaccine compositions |
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: 24842031 Country of ref document: EP Kind code of ref document: A1 |