WO2024055273A1 - 一种狂犬病mRNA疫苗、其制备及应用 - Google Patents

一种狂犬病mRNA疫苗、其制备及应用 Download PDF

Info

Publication number
WO2024055273A1
WO2024055273A1 PCT/CN2022/119230 CN2022119230W WO2024055273A1 WO 2024055273 A1 WO2024055273 A1 WO 2024055273A1 CN 2022119230 W CN2022119230 W CN 2022119230W WO 2024055273 A1 WO2024055273 A1 WO 2024055273A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
expression vector
rabv
utr
acid expression
Prior art date
Application number
PCT/CN2022/119230
Other languages
English (en)
French (fr)
Inventor
徐建青
张晓燕
白诗梦
Original Assignee
复旦大学附属中山医院
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 复旦大学附属中山医院 filed Critical 复旦大学附属中山医院
Priority to PCT/CN2022/119230 priority Critical patent/WO2024055273A1/zh
Publication of WO2024055273A1 publication Critical patent/WO2024055273A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/20Rubella virus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • C07K14/08RNA viruses
    • C07K14/145Rhabdoviridae, e.g. rabies virus, Duvenhage virus, Mokola virus or vesicular stomatitis virus

Definitions

  • the present disclosure belongs to the field of biomedicine industry, especially involving the manufacturing of genetic engineering drugs and vaccines. Specifically, the present disclosure relates to an mRNA vaccine that efficiently expresses rabies immunogen, its construction, and its application in the prevention and treatment of rabies.
  • Rabies is an acute infectious disease caused by Rabies Virus (RABV) infection.
  • the incubation period is generally 1 to 3 months.
  • RABV Rabies Virus
  • CNS central nervous system
  • the patient's mortality rate is almost 100%.
  • Rabies virus infection causes approximately 59,000 deaths worldwide each year, mainly in developing countries in Asia and Africa.
  • the number of households raising cats, dogs and other pets in my country has gradually increased, and the number of pets is expected to reach 200 million in 2022.
  • the average annual number of rabies cases in my country exceeds 1,000, making it one of the countries with the highest incidence of rabies in the world.
  • rabies virus mainly relies on inactivated vaccines. Due to the poor immunogenicity of inactivated vaccines, they require multiple vaccinations. Generally, 3 vaccinations are required before exposure, and 4 to 5 vaccinations are required to take into account post-exposure prevention and treatment; and, depending on the severity of the exposure, vaccination is sometimes required. globulin. This not only increases the cost of immunization but also reduces patient compliance.
  • the rabies virus glycoprotein (G) is the only protein displayed on the surface of the virus and is the only target that induces the production of neutralizing antibodies (VNA) in the body, providing comprehensive protection against RABV challenge.
  • Vaccines that use in vitro expressed RABV-G as the immunogen are called subunit rabies vaccines.
  • RABV-G currently produced using traditional prokaryotic expression systems, insect expression systems, Drosophila melanogaster S2 cell expression systems, etc. all show certain immunogenicity.
  • the chimeric trimer functional domain expressed by mammalian cell HEK-293T has a certain immunogenicity.
  • RABV-G extracellular domain fusion protein has better immunogenicity and can protect mice from lethal challenge.
  • the main passive immune preparations used for post-exposure prophylaxis of rabies virus are anti-rabies serum and rabies immunoglobulin.
  • Rabishield SII RMab
  • Twinrab Rost (RabiMabs), a mixed mouse-derived monoclonal antibody preparation developed by Zydus Cadila in India, was also approved for Phase I clinical research in India in September 2019, and the clinical trial results have not yet been disclosed.
  • the first anti-rabies virus monoclonal antibody drug independently developed by North China Pharmaceuticals, recombinant human anti-rabies virus monoclonal antibody injection (rhRIG), was applied for listing on July 4, 2020, and is used in combination with human rabies vaccine in order to exert a passive immune effect. .
  • recombinant viral vector-based rabies vaccines are mainly oral rabies vaccines for wild animals.
  • Recombinant poxvirus vector-based vaccines V-RG
  • V-RG canarypox vector-based recombinant rabies vaccines
  • AdHu5 human adenovirus 5
  • the chimpanzee adenovirus vector rabies vaccine can provide protection against a lethal dose of RABV (CVS-11) challenge in beagle dogs.
  • RABV adenovirus vector rabies vaccine
  • the development of viral vector vaccines needs to consider how to overcome the host's "pre-existing immunity" and avoid stimulating the body's response to the vector itself after vaccination and reducing the immune protective effect of the vaccine. Therefore, there is a need in this field to develop new rabies vaccines.
  • the mRNA (messenger ribonucleic acid) vaccine can be transcribed and synthesized in vitro, avoiding problems such as cell culture, production and complex process amplification. It can be mass-produced in a short time, is safe and effective, and has a simple process. It is a good candidate for the development of new and highly effective rabies vaccines. Bring light. Different manufacturers at home and abroad have explored and developed rabies nucleic acid vaccines and entered the clinical trial stage. Among them, the German company CureVac AG developed a vaccine with rabies virus glycoprotein (G) as the immunogen. The rabies vaccine (CV7201) announced the results of its Phase I clinical trial in 2017. However, due to the higher dose and differences in vaccination methods, some patients suffered from muscle pain and other adverse reactions.
  • G rabies virus glycoprotein
  • the low-dose group required 2 injections to induce rabies virus antibodies that meet WHO standards, while the high-dose group had higher The incidence of adverse reactions.
  • Zhuhai Lifanda Biotechnology Co., Ltd. announced the mRNA rabies vaccine it developed in January 2020.
  • 5 ⁇ g was required to be inoculated intramuscularly during the mouse model evaluation, and its serum was evaluated in vitro to produce protective properties.
  • Antibody; Beagle model evaluation requires intramuscular inoculation of 20 ⁇ g, which can induce effective neutralizing antibodies.
  • the vaccine is still in the preclinical stage.
  • This article provides a new rabies immunogen nucleic acid expression vector, a rabies vaccine containing the expression vector, and its preparation method and application.
  • the rabies immunogen nucleic acid expression vector and vaccine herein can efficiently express rabies immunogens (such as RABV-G) in vivo and in vitro, and produce excellent protection and neutralizing effects, thereby achieving effective prevention and/or treatment of rabies.
  • this article provides a rabies immunogen nucleic acid expression vector, which sequentially includes from the 5' end to the 3' end:
  • Poly(A) tail elements with a total length of more than 120 nt which include:
  • each adenylate string independently contains n consecutive adenylates, n is an integer between 10 and 80, and the total number of adenylates in the multiple adenylate strings The number is more than 100;
  • a linker is located between the plurality of adenylate strings, each of the linkers independently contains no adenylate or only contains 1 or 2 adenylate.
  • a rabies mRNA vaccine comprising the nucleic acid expression vector herein, and packaging and/or a delivery system for the nucleic acid expression vector and/or a pharmaceutically or physiologically acceptable carrier .
  • this article also provides the application of the nucleic acid expression vector and/or vaccine of this article in the preparation of products for the prevention and/or treatment of rabies.
  • this article also provides methods for preventing and/or treating rabies, which methods include administering to a subject in need thereof a prophylactically and/or therapeutically effective amount of the nucleic acid expression vector and/or vaccine herein.
  • nucleic acid expression vectors and/or vaccines herein for the prevention and/or treatment of rabies.
  • this article also provides a method for preparing a nucleic acid expression vector or vaccine as described herein, which method includes: providing independent or connected elements; assembling each element into a nucleic acid expression vector.
  • Figure 1 Expression verification of HEK293 cells transfected with in vitro transcribed RABV-G mRNA.
  • FIG. 2A The humoral immune response level of mice after priming mice with different doses of RABV-G mRNA vaccine (the ordinate is the logarithm of log with base 10);
  • Figure 2B The humoral immune response level of mice after boosting immunization with different doses of RABV-G mRNA vaccine (the ordinate is the logarithm of log with base 10);
  • Figure 2C Neutralizing effect of antibodies against rabies virus induced in mice after primary immunization with RABV-G mRNA vaccine at different doses;
  • Figure 2D Neutralizing effect of antibodies against rabies virus induced in mice after booster immunization with RABV-G mRNA vaccine at different doses.
  • ns indicates no significant difference (not significant); * indicates p ⁇ 0.05; ** indicates p ⁇ 0.01; *** indicates p ⁇ 0.001; **** indicates p ⁇ 0.0001.
  • FIG. 3A Antigen-specific CD8 + T cell (IFN ⁇ -positive CD8 + T cell) response induced by different doses of RABV-G mRNA vaccine;
  • Figure 3B Different doses of RABV-G mRNA vaccine induce antigen-specific CD4 + T cell (IFN ⁇ -positive CD4 + T cell) response;
  • Figure 3C Different doses of RABV-G mRNA vaccine induce antigen-specific CD4 + T cell (IL-4 positive CD8 + T cell) responses.
  • ns indicates no significant difference (not significant); * indicates p ⁇ 0.05; ** indicates p ⁇ 0.01.
  • Figure 4A Results of body weight changes of animals after challenge and protection
  • Figure 4B Changes in animal survival rate after challenge and protection.
  • this article provides an mRNA vector encoding RABV-G protein.
  • the mRNA can efficiently express the glycoprotein of rabies virus and can be used for the prevention and treatment of rabies.
  • the RABV-G mRNA rabies vaccine provided in this article adopts a lower dose, single-dose intramuscular injection, and can induce high-titer protective antibodies 14 days after vaccination, and can protect animals that can completely resist a lethal dose of rabies virus. challenge, can also effectively induce T cell responses specific to RABV-G.
  • the RABV-G mRNA vaccine in this article can induce effective protective antibodies that are approximately 2,000 times higher than the WHO detection standard (0.5IU/mL) after two shots of immunization.
  • the mRNA rabies vaccine in this article has good immunogenicity and forms strong immune protection after immunizing the body. It has low production cost and easy preparation technology. It is suitable for rapid, efficient and large-scale vaccine preparation, and has huge development potential. .
  • eukaryote may include humans, primates, rodents (eg, rats, mice, guinea pigs, hamsters), domestic animals, or livestock mammals.
  • a high degree of sequence identity as described herein includes a sequence identity of more than 70%, more than 75%, more than 80%, more preferably more than 85%, such as 85%, 90%, 95%, 98% or even 99% or more. , these high-identity sequences are also within the scope of equivalence that is preferably considered in the present invention. Methods and tools for comparing sequence identities are also well known in the art, such as BLAST.
  • Rabies immunogen nucleic acid expression vector and vaccine containing the vector Rabies immunogen nucleic acid expression vector and vaccine containing the vector
  • This article provides a rabies immunogen nucleic acid expression vector, which contains from the 5' end to the 3' end:
  • Poly(A) tail elements with a total length of more than 120 nt which include:
  • each adenylate string independently contains n consecutive adenylates, n is an integer between 10 and 80, and the total number of adenylates in the multiple adenylate strings The number is more than 100;
  • a linker is located between the plurality of adenylate strings, each of the linkers independently contains no adenylate or only contains 1 or 2 adenylate.
  • the length of the 5'-UTR element used is from 10 to 200 nt, such as from 15 to 100 nt. In some embodiments, the 5'-UTR element used is derived from a highly abundant protein with high translation efficiency within the cell.
  • the 5'-UTR element used is derived from one or more 5'-UTRs from the group consisting of: human alpha-globulin, beta-globulin, ribosomal protein (RP), tubulin beta -2B, complement factor 3 (C3), cytochrome P4502E1 (CYP2E1), apolipoprotein A-II (APOA2), human hemoglobin subunit beta (hHBB), hemoglobin A1 (HBA1), hemoglobin A2 (HBA2), dengue virus (DENV).
  • human alpha-globulin beta-globulin
  • RP ribosomal protein
  • C3 complement factor 3
  • CYP2E1 cytochrome P4502E1
  • APOA2E1 apolipoprotein A-II
  • hHBB human hemoglobin subunit beta
  • HBA1 hemoglobin A1
  • HBA2 hemoglobin A2
  • DEV dengue virus
  • the 5'-UTR element used has the sequence set forth in SEQ ID NO: 2 or has at least 80% sequence identity thereto.
  • the 3'-UTR element used is derived from a highly abundant protein with high translation efficiency within the cell.
  • the 3'-UTR element used is a 3'-UTR derived from a mammal or a virus, for example, a 3'-UTR derived from a source selected from the group consisting of: human alpha-globulin, or a combination thereof (e.g., a tandem sequence) Human beta globin, human albumin, human actin, human hemoglobin subunit alpha 1 (HBA1), cytochrome B-245 alpha chain (CYBA), sequence of eukaryotic mitochondria (Mit), SARAS-Cov-2, dengue fever virus (DENV), turnip wrinkle virus (TCV), tobacco mosaic virus (TMV) and tobacco etch virus (TEV).
  • human alpha-globulin or a combination thereof (e.g., a tandem sequence) Human beta globin, human albumin, human actin, human hemoglobin subunit alpha 1 (HBA1), cytochro
  • the 3'-UTR element used comprises one or more 3'-UTR molecules selected from the group consisting of alpha-globulin 3'-UTR, eukaryotic mitochondrial 3'-UTR, albumin 3'-UTR , ⁇ -globin 3′-UTR or any tandem sequence thereof, preferably ⁇ -globin 3′-UTR, eukaryotic mitochondrial 3′-UTR, or the 3′-UTR formed by their tandem connection.
  • the 3'-UTR used has the sequence shown in SEQ ID NO: 3, or a sequence that has at least 80% sequence identity thereto.
  • the total length of the poly(A) tail element used is 120-400 nt, such as 120-350 nt, 120-320 nt, or any integer therein, such as 120, 304 nt.
  • each adenylate string independently contains 10 to 80, 20 to 70, 25 to 60, 30 to 50, or any integer number of consecutive adenylates therein, such as 20, 30 1, 33, 35, 36, 37, 38, 39, 40, 45, 50, 55, 60, 65, 70 consecutive adenosine nucleotides.
  • a poly(A) tail element further includes a linker at one or both ends of the element.
  • the length of the linker is each independently 3 to 15 nt, such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 nt.
  • the linkers each independently contain no adenosine, or only 1 or 2 adenosine.
  • the linker sequence is each independently selected from: GCTATGACT, GTATGT, GCAAGT, GATTGC, GGCTGC, TACTGC, GGCTTC, GCATATGACT.
  • the poly(A) tail element has the sequence of SEQ ID NO: 4, or has at least 80% sequence identity with any one thereof.
  • the open reading frame elements used are monocistronic, bicistronic, or polycistronic mRNA.
  • an open reading frame element encodes one or more RABV-Gs of the same or different origin.
  • RABV-G is derived from a rabies virus selected from the group consisting of: Pasteur strain Pitman-Moore (PM) strain, Pasteur (PV) strain, CTN strain, aG strain, Flury-LEP strain , Evelyn-Rokitnicki-Abelseth (ERA) strain, Street-Alabama-Duffering (SAD) strain, KHUV (Khujand lyssavirus), BBLV (Bokeloh bat lyssavirus), ARAV (Aravan lyssavirus), EBLV-1 (European bat 1 lyssavirus), EBLV-2 (European bat 2 lyssavirus), IRKV (Irkutlyssavirus), LBV (Lagos bat lyssavirus), SHIBV (Shimoni bat lyssavirus), MOKV (Mokola lyssavirus), WCBV (West Caucasian bat lyssavirus), I
  • RABV-G is the main immunogen for producing rabies virus neutralizing antibodies and is highly conserved among multiple rabies viruses.
  • RABV-G can be derived from the Pitman-Moore (PM) strain.
  • RABV-G includes one or more molecules selected from the group consisting of unmodified or eukaryotic modified RABV-G, immunogenic fragments or variants thereof, said molecules capable of inducing resistance to Immune neutralization and protective responses to RABV.
  • the element encoding RABV-G is or is not codon optimized, contains or does not contain base modifications and/or nucleoside analogs. In some embodiments, the coding sequence is optimized based on eukaryotic codon preferences.
  • one or more uracils in the element encoding RABV-G are replaced with one or more identical or different modified bases or nucleoside analogues selected from the group consisting of: pseudouridine, 1 -Methyluridine, N1-ethylpseudouridine, 2-thiouridine, 4′-thiouridine, 5-methylcytosine, 5-methyluridine, 2-thio-1- Methyl-1-deaza-pseudouridine, 2-thio T-methyl-pseudouridine, 2-thio-5-aza-uridine, 2-thio-dihydropseudouridine, 2-Thio-dihydrouridine, 2-Thio-pseudouridine, 4-methoxy-2-thio-pseudouridine, 4-methoxy-pseudouridine, 4-thio-1 - Methyl-pseudouridine, 4-thio-pseudouridine, 5-aza-uridine,
  • 50% to 100% of the uracils in the open reading frame element encoding the rabies virus glycoprotein (RABV-G) are substituted. In some embodiments, 50% to 100% of the uracils in the expression vector sequence are substituted. Substitution can improve the stability of mRNA in vivo.
  • the expression vector also includes a 5'-cap element, which can optionally be modified.
  • the 5'-cap element is selected from: m7GpppXpYp, m7GpppXmpYp, m7GpppXmpYmp, or its methylation modification sequence, trans To the binding isomer, anti-reverse cap analog (ARCA), N7-benzyldinucleoside tetraphosphate cap analog.
  • the expression vector further includes a promoter element, such as a T7 promoter, sp6 promoter, or T3 promoter.
  • the expression vector further includes a signal peptide coding element, such as a signal peptide coding element that guides the subcellular localization of the target protein (such as a transmembrane signal peptide, a secretion signal peptide, a nuclear localization signal peptide).
  • the expression vector also includes restriction sites, such as Xbal, EcoRV, BamHI, and XhoI.
  • the expression vector also includes tags, such as molecular tags used for identification, isolation or purification of target molecules, such as Flag tags and HA tags.
  • the expression vector includes from the 5' end to the 3' end: a 5'-UTR element including the sequence shown in SEQ ID NO: 2; an open reading frame element encoding RABV-G; including SEQ ID A 3′-UTR element of the sequence shown in NO: 3; a poly(A) tail element comprising the sequence shown in SEQ ID NO: 4; or an element of a sequence having at least 80% sequence identity with each of the sequences described .
  • the expression vector comprises the sequence set forth in SEQ ID NO: 5 or 6, or a sequence having at least 80% sequence identity thereto.
  • the nucleic acid expression vector is contained in a package alone, or is combined with a carrier in a delivery system, for example, the delivery system is selected from: lipid delivery system, lipid delivery system, polymer delivery The system or its combination delivery system, for example, loaded in lipid nanoparticles, cationic liposomes, polyurethane (PAA), poly ⁇ -aminoester (PBAE), polyethylenimine (PEI), lipid-wrapped polymer micelles.
  • PAA polyurethane
  • PBAE poly ⁇ -aminoester
  • PEI polyethylenimine
  • the nucleic acid expression vector is combined with liposomal nanoparticles.
  • liposomal nanoparticles include a combination of cationic lipids, structural lipids, helper lipids, and stabilizing lipids.
  • the liposome nanoparticles include: 20-50% cationic lipids: 20-50% structural lipids: 5-20% auxiliary lipids: 1-5% stabilizing lipids, calculated as molar percentages The quality is more preferably 50% cationic lipid: 38% structural lipid: 10% auxiliary lipid: 2% stable lipid.
  • the cationic lipid in the liposomal nanoparticles is one or more selected from the group consisting of: (2,3-dioleoyl-propyl)-trimethylammonium chloride (DOTAP ), didecyl adipate (DDA), 3 ⁇ -[N-(N′,N′-dimethylaminoethyl)carbamoyl]cholesterol (DC-Chol), N,N-dimethyl -4-Pyridylamine (DMAP), 1,2-triethanolamine-3-trimethylpropane (DOTMA), N-[1-(2,3-dioleoyl)propyl]-N-(arginine amide)ethyl-N,N-dimethylammonium trifluoroacetate (DOSPA), 4-(N,N-dimethylamino)butyric acid (dilinoleyl)methyl ester (Dlin-MC3-DMA) , KC2, N,N-dimethyla
  • the structural lipids in the liposome nanoparticles include cholesterol, cholesteryl lipids, sterol hormones, sterol vitamins or bile acids, preferably cholesterol.
  • the accessory lipids in liposomal nanoparticles include dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), 1-palmitoyl-2-oleoyl lecithin ( POPC), 1,2 distearic acid-3-phosphatidylethanolamine (DSPE), dioleoyl lecithin (DOPC), dioleoyl phosphatidylserine (DOPS), distearoylphosphatidylcholine (DSPC) , the preferred distearoylphosphatidylcholine (DSPC).
  • DPPC dipalmitoylphosphatidylcholine
  • DOPG dioleoylphosphatidylglycerol
  • POPC 1-palmitoyl-2-oleoyl lecithin
  • DOPC 1,2 distearic acid-3-phosphatidylethanolamine
  • DOPC dioleoyl lecithin
  • the stable lipids in liposomal nanoparticles include polyethylene glycol (PEG)-lipids, such as PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramide, PEG-modified diacylglycerol, preferably PEG-modified diacylglycerol, more preferably long-circulating liposome 1,2-dimyristoyl-rac-glycerol-3-methoxypolyethylene glycol 2000 (PEG 2000-DMG ).
  • PEG polyethylene glycol
  • the mass ratio of cationic liposomes to RABV-G mRNA in liposome nanoparticles is 10-30:1, preferably 15-20:1.
  • the preparation method is as follows: ionizable cationic lipids, structural lipids, auxiliary lipids and stable lipids are mixed according to the formula ratio Dissolve in ethanol to prepare an organic phase. Dissolve the optimized nucleoside-modified RABV-G mRNA in citric acid buffer (pH 4.0) to prepare an aqueous phase. Use a bolus mixer to mix the organic phase and the aqueous phase at a certain ratio to obtain a mixed solution.
  • the ratio of the organic phase to the aqueous phase is 1:2 to 1:5, preferably 3:7.
  • a rabies mRNA vaccine which includes: a rabies immunogen nucleic acid expression vector as described herein, and packaging and/or a delivery system and/or pharmaceutical for the nucleic acid expression vector. or a physiologically acceptable carrier.
  • the nucleic acid expression vector in the vaccine is contained alone in the package or combined with a carrier in a delivery system, for example, the delivery system is selected from: lipid delivery system, lipid delivery system, polymer Delivery systems or combinations thereof, such as lipid nanoparticles, cationic liposomes, polyurethane (PAA), poly ⁇ -aminoester (PBAE), polyethylenimine (PEI), lipid-encapsulated polymer micelles .
  • the delivery system is selected from: lipid delivery system, lipid delivery system, polymer Delivery systems or combinations thereof, such as lipid nanoparticles, cationic liposomes, polyurethane (PAA), poly ⁇ -aminoester (PBAE), polyethylenimine (PEI), lipid-encapsulated polymer micelles .
  • PAA polyurethane
  • PBAE poly ⁇ -aminoester
  • PEI polyethylenimine
  • the nucleic acid expression vector in the vaccine is combined with liposomal nanoparticles.
  • liposomal nanoparticles include a combination of cationic lipids, structural lipids, helper lipids, and stabilizing lipids.
  • the liposome nanoparticles include: 20-50% cationic lipids: 20-50% structural lipids: 5-20% auxiliary lipids: 1-5% stabilizing lipids, calculated as molar percentages.
  • the quality is more preferably 50% cationic lipid: 38% structural lipid: 10% auxiliary lipid: 2% stable lipid.
  • the cationic lipid in the liposomal nanoparticles is one or more selected from the group consisting of: (2,3-dioleoyl-propyl)-trimethylammonium chloride (DOTAP ), didecyl adipate (DDA), 3 ⁇ -[N-(N′,N′-dimethylaminoethyl)carbamoyl]cholesterol (DC-Chol), N,N-dimethyl -4-Pyridylamine (DMAP), 1,2-triethanolamine-3-trimethylpropane (DOTMA), N-[1-(2,3-dioleoyl)propyl]-N-(arginine amide)ethyl-N,N-dimethylammonium trifluoroacetate (DOSPA), 4-(N,N-dimethylamino)butyric acid (dilinoleyl)methyl ester (Dlin-MC3-DMA) , KC2, N,N-dimethyla
  • the structural lipids in the liposome nanoparticles include cholesterol, cholesteryl lipids, sterol hormones, sterol vitamins or bile acids, preferably cholesterol.
  • the accessory lipids in liposomal nanoparticles include dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), 1-palmitoyl-2-oleoyl lecithin ( POPC), 1,2 distearic acid-3-phosphatidylethanolamine (DSPE), dioleoyl lecithin (DOPC), dioleoyl phosphatidylserine (DOPS), distearoylphosphatidylcholine (DSPC) , the preferred distearoylphosphatidylcholine (DSPC).
  • DPPC dipalmitoylphosphatidylcholine
  • DOPG dioleoylphosphatidylglycerol
  • POPC 1-palmitoyl-2-oleoyl lecithin
  • DOPC 1,2 distearic acid-3-phosphatidylethanolamine
  • DOPC dioleoyl lecithin
  • the stable lipids in liposomal nanoparticles include polyethylene glycol (PEG)-lipids, such as PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramide, PEG-modified diacylglycerol, preferably PEG-modified diacylglycerol, more preferably long-circulating liposome 1,2-dimyristoyl-rac-glycerol-3-methoxypolyethylene glycol 2000 (PEG 2000-DMG ).
  • PEG polyethylene glycol
  • the mass ratio of cationic liposomes to RABV-G mRNA in liposome nanoparticles is 10-30:1, preferably 15-20:1.
  • the preparation method is as follows: ionizable cationic lipids, structural lipids, auxiliary lipids and stable lipids are mixed according to the formula ratio Dissolve in ethanol to prepare an organic phase. Dissolve the optimized nucleoside-modified RABV-G mRNA in citric acid buffer (pH 4.0) to prepare an aqueous phase. Use a bolus mixer to mix the organic phase and the aqueous phase at a certain ratio to obtain a mixed solution.
  • the ratio of the organic phase to the aqueous phase is 1:2 to 1:5, preferably 3:7.
  • the vaccine is in a form suitable for one or more administration or delivery methods selected from the group consisting of: respiratory aerosol inhalation, nasal instillation, oral administration, direct injection (e.g., intravenous injection, subcutaneous injection, intradermal injection) injection, intramuscular injection), mucosal administration.
  • respiratory aerosol inhalation e.g., nasal instillation
  • oral administration e.g., direct injection (e.g., intravenous injection, subcutaneous injection, intradermal injection) injection, intramuscular injection), mucosal administration.
  • the vaccine further includes or is used in combination with an adjuvant
  • the adjuvant is selected from: aluminum adjuvant, cholera toxin and its subunits, oligodeoxynucleotides, manganese ion adjuvant, Colloidal manganese adjuvant, Freund's adjuvant, MF59 adjuvant, QS-21 adjuvant, Poly I: C and other TLR ligands, GM-CSF, IL-2, IL-3, IL-7, IL-11, IL-12, IL-18, IL-21.
  • the form of the vaccine is suitable for combined administration of two or more drugs or vaccines, such as combined vaccination or sequential vaccination.
  • a method for preparing a nucleic acid expression vector or vaccine described herein includes: providing individual or connected elements; assembling each element into a nucleic acid expression vector.
  • the method includes employing one or more materials selected from the group consisting of: DNA template (eg, PCR product or linearized plasmid DNA), nuclease, polymerase, capping enzyme, polyadenylation Ulate synthase, DNase, one or more component molecules, linker molecules, natural or modified nucleic acid molecules, buffers, solvents.
  • the method further includes one or more steps selected from the group consisting of: designing, optimizing, transforming and/or modifying each component; and isolating, purifying, and identifying intermediate products and/or final products. , quantification, packaging and/or activity testing; combining the nucleic acid expression vector with a delivery system for the nucleic acid expression vector and/or a pharmaceutically or physiologically acceptable carrier.
  • the rabies is rabies caused by one or more of the following rabies viruses: Pitman-Moore (PM) strain, Pasteur (PV) strain, CTN strain, aG strain, Pasteur strain, Flury-LEP strain, Evelyn-Rokitnicki-Abelseth (ERA) strain, Street-Alabama-Duffering (SAD) strain, KHUV (Khujand lyssavirus), BBLV (Bokeloh bat lyssavirus), ARAV (Aravan lyssavirus), EBLV-1 (European bat 1 lyssavirus), EBLV-2 (European bat 2 lyssavirus), IRKV (Irkutlyssavirus), LBV (Lagos bat lyssavirus), SHIBV
  • the term "pharmaceutically or physiologically acceptable” ingredients are suitable for use in humans and/or animals without undue adverse side effects (e.g., toxicity, irritation, and allergic reactions), i.e., with a reasonable benefit/risk ratio substance.
  • the term “effective amount” refers to an amount that produces a function or activity in humans and/or animals and is acceptable to humans and/or animals.
  • the term "pharmaceutically acceptable carrier” refers to a vehicle for administration of a therapeutic agent, including various excipients and diluents. This term refers to pharmaceutical carriers that do not themselves require the active ingredient and are not unduly toxic upon administration. Suitable carriers are well known to those of ordinary skill in the art. A thorough discussion of pharmaceutically acceptable excipients can be found in Remington's Pharmaceutical Sciences, Mack Pub. Co., N.J. 1991.
  • compositions may contain liquids such as water, saline, glycerin and ethanol.
  • these carriers may also contain auxiliary substances, such as fillers, disintegrants, lubricants, glidants, effervescent agents, wetting agents or emulsifiers, flavoring agents, pH buffering substances, etc.
  • these materials may be formulated in a nontoxic, inert and pharmaceutically acceptable aqueous carrier medium, typically at a pH of about 5-8, preferably at a pH of about 6-8.
  • unit dosage form refers to the preparation of the active substance herein into a dosage form required for a single administration for the convenience of administration, including but not limited to various solid dosage forms (such as tablets), liquid dosage forms, and capsules. agent, sustained-release agent.
  • the effective dose of active substance employed may vary depending on the severity of the subject to be administered or treated. The specific situation is determined based on the individual situation of the subject (such as the subject's weight, age, physical condition, and desired effects), which is within the scope of a skilled physician's judgment.
  • the products herein may be in solid state (such as granules, tablets, freeze-dried powder, suppositories, capsules, sublingual tablets) or liquid state (such as oral liquid) or other suitable shapes.
  • the route of administration can be: (1) direct naked nucleic acid injection; (2) connecting the mRNA expression vector to the transferrin/poly-L-lysine complex to enhance its biological effect; (3) expressing the mRNA The carrier forms a complex with positively charged lipids to overcome the difficulty in crossing the cell membrane caused by the negative charge of the phosphate backbone; (4) wrapping the mRNA expression vector with liposomes and mediating its entry into cells is beneficial to the smooth movement of macromolecules Enter and avoid hydrolysis by various extracellular enzymes; (5) Combine the mRNA expression vector with cholesterol to increase its retention time; (6) Use immunoliposomes to transport the mRNA to specifically transport it to the target tissue and target cells; (7) transfecting the mRNA expression vector into the transfected cells in vitro; (8) electropor
  • a method for preventing and/or treating rabies virus infection and/or symptoms thereof which includes: administering at least once a prophylactically and/or therapeutically effective amount of one or more vaccines of the present disclosure.
  • the vaccination methods that can be used include but are not limited to: systemic immunization methods, such as intramuscular injection, subcutaneous injection and intradermal injection, etc., intramuscular injection and subcutaneous injection are preferred, and intramuscular injection is more preferred due to the difficulty of administration; respiratory tract Internal immunization methods, such as atomization, intranasal drip, etc.
  • the primary immunization is systemic vaccination or intrarespiratory vaccination, with systemic vaccination being preferred.
  • the interval between each two vaccinations is at least 1 week, such as 2 weeks, 4 weeks, 2 months, 3 months, 6 months or longer.
  • an mRNA vaccine is used for primary immunization, and one or more booster immunizations are performed with the same or other types of vaccines.
  • the immunization method of the present disclosure can adopt the method of "primary immunization-boost” or “primary immunization-boost-reboost", and can adopt a single systemic immunization or local respiratory tract immunization method, or a combination of the two immunization methods.
  • a combination product herein is provided in the form of a pharmaceutical package or kit, for example, one or more vaccine compositions herein or one or more components thereof may be packaged in one or more containers, For example, packaged in a sealed container such as an ampoule or sachet indicating the amount of composition.
  • Vaccine compositions may be provided in the form of liquids, sterile lyophilized powders, or anhydrous concentrates, which may be diluted, reconstituted, and/or formulated with appropriate liquids (e.g., water, saline, etc.) prior to use for administration. to the appropriate concentration and form of the object.
  • RABV-G mRNA preparation cationic liposome nanoparticle preparation, animal immunization scheme and detection method involved in the experiments in the examples are as follows:
  • a suitable DNA template use pCDNA3.1+ as a template, transform it, and include from the N-terminus to the C-terminus after the T7 promoter: 5'-UTR sequence (SEQ ID NO: 2), coding sequence (SEQ ID NO : 1), 3′-UTR sequence (SEQ ID NO: 3), poly(A) tail sequence (SEQ ID NO: 4) containing 250 consecutive adenosine interspersed with linker sequences, linearized enzyme digestion
  • the modified rabies Pitman-Moore (PM) vaccine strain glycoprotein (RABV-G) coding sequence is used as the immunogen coding sequence, inserted into the coding sequence, and the restriction sites EcoRV and Flag are inserted at the C-terminal of the coding sequence.
  • Tag add Flag tag for subsequent gene expression identification.
  • the constructed DNA template sequence (SEQ ID NO: 6) was optimized according to the preference of eukaryotic codons and used for subsequent in vitro transcription experiments.
  • the transcription kit was purchased from Nearshore Protein Technology Co., Ltd., using the plasmid described in SEQ ID NO: 6 after single enzyme digestion as a template, adding the NTP required for transcription according to the appropriate ratio, and using 1- Methyl uracil nucleoside ( ⁇ ) replaces uracil nucleoside (U), and T7 polymerase is used to transcribe the corresponding RABV-G mRNA (SEQ ID NO: 5, 5′UTR+RABV-G mRNA+( ⁇ -globin +Mit)3′UTR+250A, in which U is replaced by ⁇ and contains codon optimization), and then capped by enzymatic capping method using both vaccinia virus capping enzyme and 2′-O-methyltransferase in one step , add a 7-methylguanosine cap structure to the purified RABV-G mRNA, and use lithium chloride to purify the capped mRNA for the preparation of lipid nanoparticles.
  • Maianna (Shanghai) Instrument Technology Co., Ltd. was commissioned to prepare rabies mRNA liposome nanoparticles: combine the cationic lipid Dlin-MC3-DMA, structural lipid cholesterol, auxiliary lipid DSPC and stable lipid DMG-PEG2000, according to 50: The molar ratio of 38:10:2 was dissolved in ethanol, and the ethanol concentration was 30% (v/v) to obtain an oil phase mixture. Then, the oil phase mixture was added to 50mM pH 4.0 citrate buffer at room temperature to obtain a lipid mixture.
  • lipid mixture to a liposome extruder, first squeeze and filter it with a 200nm filter membrane, and then filter it with a 100nm filter membrane to change the solution from milky white to clear, and obtain cationic liposome nanoparticles.
  • RABV-G mRNA cationic lipid nanoparticles Dissolve the optimized nucleoside-modified RABV-G mRNA in citrate buffer (pH 4.0), and add it dropwise to the cationic lipid nanoparticles at a ratio of cationic lipid nanoparticles to mRNA mass ratio of 20:1. , use a Vortex shaker to mix to obtain a mixed solution. After thorough mixing, the mixture was heated and incubated at 42°C for 1 hour. The mixture was then dialyzed into sterile PBS and filtered through a 0.22 ⁇ m sterile filter to obtain RABV-G mRNA cationic lipid nanoparticles, which is the rabies virus nucleic acid vaccine.
  • mice in each group 6 of which are used for humoral response detection and 6 for (for detecting cellular immune response), namely negative control group, RABV-G high-dose group, RABV-G medium-dose group, and RABV-G low-dose group.
  • Each group was administered intramuscular injection, with initial immunization on the 1st day of the trial and booster immunization on the 21st day of the trial.
  • the doses of each immunization were the same, as shown in Table 1.
  • the second immunization plan is a one-shot immunization, and the RABV-G low-dose group is selected, with 8 animals in each group.
  • the volume of a single immunization is 100 ⁇ l.
  • the immunization doses of the RABV-G high-dose group, medium-dose group, and low-dose group all refer to the immunization dose of each mRNA.
  • FAVN Fluorescent antibody virus neutralization test
  • Serum titer is the dilution of 50% of the wells and 100% of the virus (log D50). 0.5IU/ml is the neutralization standard recommended by WHO. Under the same experimental conditions, the neutralization dilution ratio of the test serum and the canine-derived International Office for Animal Epidemics (OIE) reference serum to the half of the quantified virus is calculated. Comparing the two, multiplied by the standard serum titer (0.5IU/ml) is the rabies neutralizing titer of the serum to be tested, expressed as the geometric mean (GMT) of the neutralizing titer titers, and is calculated using Graphpad Prism8 software. picture.
  • GTT geometric mean
  • 1E6 cells per well were plated in 96 wells, and rabies virus glycoprotein peptide library with a final concentration of 5 ⁇ g/mL was added for stimulation.
  • the negative control group was the DMSO group, and the positive control group was PMA with a final concentration of 50 ng/mL and 5 ⁇ g/mL.
  • Ionomycin incubate for 1 hour at 37°C and 5% CO 2 , add BD GolgiStop TM Protein Transport Inhibitor (1:1000), and incubate in a 37°C incubator for 5 hours. The stimulation is completed.
  • BD Cytofix/Cytoperm
  • Permeabilize cells Dilute 10x BD Perm/Wash TM buffer with ultrapure water to 1 ⁇ working solution, resuspend the cells in 1x BD Perm/Wash TM buffer, and incubate at room temperature in the dark for 15 minutes. Centrifuge at 800g for 5 minutes and carefully remove the supernatant. Dilute mouse IFN-GMA FITC and mouse IL-4APC with 1x BD Perm/Wash TM buffer and add to the cell suspension for staining. All antibodies were purchased from BD Biosciences. Data acquisition was performed on a BD LSRFortessa flow cytometer; at least 30,000 cells were collected per sample, followed by data analysis using FlowJo software.
  • Example 1 Verification of expression of RABV-G mRNA transfected into HEK293T cells
  • HEK293T cells were transfected with RABV-G mRNA prepared in Experimental Method I above for expression verification. 24 hours before transfection, HEK293 cells were seeded into a 12-well plate at a density of 200,000 cells/well. The medium was DMEM complete medium (10% FBS and 1% P.S.). The transfection reagent was Lipofectamine 3000. Each well plate was transfected. 2 ⁇ g RABV-G mRNA, cultured in 37°C incubator for 40-48h. Collect cells into pre-cooled EP tubes, add RIPA lysis buffer to lyse cells. Perform SDS-polyacrylic acid amine gel electrophoresis (SDS-PAGE).
  • SDS-PAGE SDS-polyacrylic acid amine gel electrophoresis
  • RABV-G mRNA is expressed in large amounts after transfection into HEK293T cells and can be specifically recognized by anti-rabies virus antibodies.
  • the size of the target band is at the 60kDa position and is the correct size.
  • RABV-G mRNA vaccine induces humoral immune response in mice
  • the mRNA selected in this example is the RABV-G mRNA whose correct expression was verified in Example 1.
  • the cationic lipid nanoparticles encapsulating RABV-G mRNA were prepared according to the above experimental method II.
  • RABV-G mRNA lipid nanoparticle vaccine was used to immunize BALB/c mice to evaluate its immunogenicity. Three different doses of 10 ⁇ g (high, group 4), 3 ⁇ g (medium, group 3) and 1 ⁇ g (low, group 2) were selected. The dose was 6 mice per group, intramuscular injection, each injection volume was 100 ⁇ l, and the negative control was an equal volume of cationic lipid nanoparticles without nucleic acid encapsulation. Three different doses of vaccines and the negative control group were administered once on the 1st day, and then received a booster injection on the 21st day. Blood samples were collected on days 14, 21, 28, and 35 respectively. The RABV-G specific binding antibody titer was detected by enzyme-linked immunosorbent assay, and the serum true virus neutralizing antibody titer was detected by fluorescent antibody virus neutralization test (FAVN).
  • FAVN fluorescent antibody virus neutralization test
  • FIGS. 2A and 2B The results of enzyme-linked immunosorbent assay to detect RABV-G specific binding antibody titers are shown in Figures 2A and 2B.
  • the serum RABV-G antibody titers of the 10 ⁇ g group immunized mice on the 14th and 21st days after the first administration were 12800 and 22807 respectively, and the serum RABV-G IgG antibody titers of the 3 ⁇ g group immunized mice were The titers of serum RABV-G antibodies in the 1 ⁇ g group of immunized mice were 1600 and 2015.9 respectively. Then they received a booster injection on the 21st day.
  • the results are shown in Figure 2B.
  • the RABV-G antibody titers of the mice immunized in the 10 ⁇ g group were 645080 and 705035 respectively.
  • the RABV-G antibody titers of mice were 256000 and 516064 respectively, and the RABV-G antibody titers of mice immunized in the 1 ⁇ g group were 128000 and 409600 respectively.
  • the neutralizing antibody titers in the serum of mice immunized in the 10 ⁇ g group were 1018.5 and 1166 IU/ml respectively on the 7th and 14th days after the booster immunization. and antibody titers were 966.3 and 1255IU/ml respectively, and the neutralizing antibody titers in the serum of mice immunized in the 1 ⁇ g group were 557.9 and 1001.7IU/ml respectively.
  • the results of this experiment show that whether it is a single dose or a booster shot, the serum antibodies induced by the vaccination doses of 10 ⁇ g, 3 ⁇ g, and 1 ⁇ g can completely neutralize virus-infected cells.
  • the neutralizing antibody titer after one shot of vaccination is WHO It is 16 to 30 times the standard (0.5IU/mL), and after strengthening, it is 1000 to 2000 times the WHO standard (0.5IU/mL), and its binding antibody has the same trend as the neutralizing antibody.
  • the rabies mRNA vaccine of the present application can achieve extremely significant neutralizing and protective effects at a low dose (microgram level) and a small number of times (two vaccinations, or even a single vaccination).
  • RABV-G antigen-specific T cells were analyzed by intracellular cytokine staining (ICS) according to the above experimental method III.
  • ICS intracellular cytokine staining
  • the spleen cells were isolated from the 3 ⁇ g and 1 ⁇ g vaccination dose groups and the liposome nanoparticles group without nucleic acid encapsulation (i.e., the mice in the negative control group).
  • RABV-G mRNA vaccine can effectively induce RABV-G-specific CD4 + and/or CD8 + T cells, and the effect can be further improved as the dose increases.
  • Example 4 Protective effect of a single dose of low-dose RABV-G mRNA vaccine on mouse challenge
  • this example uses BALB/c mice as a model, and selects a single intramuscular injection of 1 ⁇ g low-dose group.
  • the negative control is an equal volume of lipids without nucleic acid encapsulation. Nanoparticles. There were 8 mice in each group, and the injection volume per mouse was 100 ⁇ l.
  • CVS-11 fixed virus was challenged via intramuscular injection, and the challenge dose was 20 LD50, 50 ⁇ l/animal. Put the mice into the IVC cage, weigh the mice every day for 14 consecutive days, and observe the survival and living status of the mice.
  • mice in the lipid nanoparticles group without nucleic acid encapsulation began to lose weight suddenly on the 4th day after infection (Figure 4A), began to die on the 7th day after infection, and the mortality rate reached 60% on the 8th day. All died on the 9th day ( Figure 4B).
  • the RABV-G mRNA vaccine provided in this application can form strong immune protection when vaccinated at a low dose, and can not only induce high-titer neutralizing antibodies, but also activate T Cellular immune response has broad application prospects in the field of rabies virus prevention and treatment.
  • SEQ ID NO sequence information 1 DNA sequence of RABV-G immunogen 2 5′-UTR 3 3′-UTR sequence (tandem 3′UTR sequence derived from human ⁇ -globulin and eukaryotic mitochondria) 4 3′-poly(A) tail sequence (250A) 5 Modified RABV-G mRNA sequence (codon optimization + uracil substitution) 6 Preparation of DNA template sequence for RABV-G mRNA

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

本文提供了一种狂犬病mRNA疫苗、其制备及应用。本文具体提供了一种狂犬病免疫原核酸表达载体,其从5'端至3'端依次包含:(a)5'-UTR元件;(b)编码狂犬病病毒糖蛋白(RABV-G)的开放阅读框元件;(c)3'-UTR元件;(d)总长度为120nt以上的多聚腺苷酸尾元件;还提供了包含该狂犬病免疫原核酸表达载体狂犬病疫苗;以及所述表达载体和疫苗的制备和应用。

Description

一种狂犬病mRNA疫苗、其制备及应用 技术领域
本公开属于生物医药产业领域,尤其涉及基因工程药物和疫苗制造。具体而言,本公开涉及一种高效表达狂犬病免疫原的mRNA疫苗、其构建以及在狂犬病防治中的应用。
背景技术
狂犬病是由狂犬病病毒(Rabies Virus,RABV)感染引起的急性传染病,潜伏期一般为1至3个月。多数情况下携带狂犬病病毒的犬、猫等动物唾液中的RABV会沿着被其咬伤者的周围神经系统播散并最终感染中枢神经系统(CNS)。一旦出现明显的临床症状,患者的死亡率几乎达100%。即便感染RABV后存活的患者亦通常伴随有严重的神经元损伤。全球每年因狂犬病病毒感染导致约59,000人死亡,主要发生在亚洲和非洲的发展中国家。近年来,我国饲养猫、狗等宠物的家庭逐渐增加,2022年预计宠物数量达2亿只。同时,人被宠物意外咬伤的案例也不断增加,我国年均狂犬病病例超过1000例,属全球高发国家之一。
目前针对狂犬病病毒的预防和治疗,主要以灭活疫苗为主。由于灭活疫苗免疫原性较差,其需要多次接种,一般暴露前预防接种需要3次,暴露后防治兼顾则需接种4~5次;并且,根据暴露的严重程度,有时还需注射免疫球蛋白。这不仅增加了免疫接种的成本,也降低了患者的依从性。
狂犬病病毒糖蛋白(G)是唯一展示在病毒表面的蛋白,是诱导机体产生中和抗体(VNA)的唯一靶标,可提供针对RABV攻击的全面保护。以体外表达的RABV-G作为免疫原的疫苗称为亚单位狂犬病疫苗。目前采用传统的原核表达系统、昆虫表达系统、黑腹果蝇S2细胞表达系统等生产的RABV-G均显示一定的免疫原性,哺乳动物细胞HEK-293T表达的嵌合三聚体功能域的RABV-G胞外域融合蛋白免疫原性更好,可保护小鼠免受致死性攻击。然而,体外表达的重组蛋白因生产周期长、工艺复杂、产量较低等因素,目前国内还处于临床 前研究阶段,国外也只有美国Novavax开发的狂犬病G蛋白候选疫苗处于I/II期临床试验阶段。
用于狂犬病病毒暴露后预防的被动免疫制剂主要是抗狂犬病血清和狂犬病人免疫球蛋白。2016年12月,印度血清研究所研发的重组抗狂犬病病毒单抗注射液Rabishield(SII RMab)被批准上市。印度Zydus Cadila开发的鼠源单抗混合制剂Twinrab(RabiMabs)也于2019年9月在印度获得批准I期临床研究,尚未披露临床试验结果。国内华北制药自主研发的首个抗狂犬病病毒单抗药物重组人源抗狂犬病病毒单抗注射液(rhRIG),于2020年07月04日申报上市,与人用狂犬病疫苗联用以期发挥被动免疫作用。
目前,重组病毒载体类狂犬病疫苗主要为野生动物口服用狂犬病疫苗。基于复制型重组痘病毒载体疫苗(V-RG)已在北美洲使用多年,但其在部分野生动物中保护效果不是很理想;金丝雀痘病毒(canarypox)载体重组狂犬病疫苗(
Figure PCTCN2022119230-appb-000001
Feline Rabies和
Figure PCTCN2022119230-appb-000002
Rabies,Merial)在猫上可提供3年的免疫保护;基于人5型腺病毒(human adenovirus 5,AdHu5)载体开发的
Figure PCTCN2022119230-appb-000003
也具有较好的免疫效果。近年来非人灵长类腺病毒载体疫苗发展迅速,如黑猩猩腺病毒载体狂犬病疫苗(ChAd68-Gp)能为比格犬提供致死剂量的RABV(CVS-11)攻击保护。然而,病毒载体类疫苗的开发需要考虑如何克服宿主的“预存免疫”,避免疫苗接种后激起机体对载体本身的应答而降低疫苗的免疫保护效果。因此,本领域中有必要开发新型狂犬病疫苗。
mRNA(信使核糖核酸)疫苗可在体外进行转录合成,避免了细胞培养、生产和复杂的工艺放大等问题,并且可在短时间内大量生产,安全有效,工艺简单,为新型高效狂犬病疫苗的研发带来曙光。国内外先后有不同厂家探索开发狂犬病核酸疫苗并进入临床试验阶段,其中,德国CureVac AG公司开发的以狂犬病病毒糖蛋白(G)为免疫原的
Figure PCTCN2022119230-appb-000004
狂犬病疫苗(CV7201)在2017年公布了其临床I期试验结果,但因接种剂量较高及接种方式的差异导致部分患者出现了肌肉疼痛等较多的不良反应。2021年,该公司公布了其新型的mRNA-LNP制剂CV7202的临床I期实验结果,但低剂量组需要接种2针才可诱导出符合WHO标准的狂犬病病毒抗体,而高剂量组则有较高的不良反应发 生率。此外,珠海丽凡达生物技术有限公司于2020年1月份公布其开发的mRNA狂犬病疫苗,在该疫苗的动物模型评价中,小鼠模型评价时需肌肉接种5μg,体外评价其血清可产生保护性抗体;比格犬模型评价需肌肉接种20μg,可诱导出有效的中和抗体,目前该疫苗还处于临床前阶段。
在保证安全性的前提下,如何获得一种免疫原性更好、低剂量即可达到足够保护效果,临床规模生产时具有更低生产成本的狂犬病疫苗是目前本领域中狂犬病疫苗开发设计的重要目标。
发明内容
本文中提供了一种新型的狂犬病免疫原核酸表达载体、包含该表达载体的狂犬病疫苗及其制备方法和应用。本文的狂犬病免疫原核酸表达载体和疫苗能够在体内外高效表达狂犬病免疫原(例如RABV-G),并产生优异的保护和中和效果,从而实现对狂犬病的有效预防和/或治疗。
在一些方面中,本文提供了一种狂犬病免疫原核酸表达载体,其从5′端至3′端依次包含:
(a)5′-UTR元件;
(b)编码狂犬病病毒糖蛋白(RABV-G)的开放阅读框元件;
(c)3′-UTR元件;
(d)总长度为120nt以上的多聚腺苷酸尾元件,其包含:
多个腺苷酸串,每个腺苷酸串各自独立地包含n个连续的腺苷酸,n为10~80之间的整数,且所述多个腺苷酸串的腺苷酸总个数为100个以上;
位于所述多个腺苷酸串之间的连接子,所述连接子各自独立地不包含腺苷酸或仅包含1个或2个腺苷酸。
在一些方面中,本文提供了一种狂犬病mRNA疫苗,其包含本文的核酸表达载体,以及包装物和/或用于该核酸表达载体的递送系统和/或药学上或生理学上可接受的运载体。
在一些方面中,本文还提供了本文核酸表达载体和/或疫苗在制备狂犬病预防和/或治疗的产品中的应用。
在一些方面中,本文还提供了狂犬病预防和/或治疗狂犬病的方法,所述方法包括给予有需要的对象预防和/或治疗有效量的本文核酸表达载体和/或疫苗。
在一些方面中,本文还提供了用于狂犬病预防和/或治疗的本文核酸表达载体和/或疫苗。
在一些方面中,本文还提供了制备如本文所述核酸表达载体或疫苗的方法,所述方法包括:提供独立或连接的各元件;将各元件组装成核酸表达载体。
本领域的技术人员可对前述的技术方案和技术特征进行任意组合而不脱离本发明的发明构思和保护范围。本发明的其它方面由于本文的公开内容,对本领域的技术人员而言是显而易见的。
附图说明
下面结合附图对本公开作进一步说明。这些附图仅为了图示说明本公开的实施方案,而不是为了局限本公开的范围。
图1:体外转录的RABV-G mRNA转染HEK293细胞的表达验证。
图2:用RABV-G mRNA疫苗免疫小鼠后的抗体滴度以及中和效果:
图2A:用不同剂量的RABV-G mRNA疫苗初免小鼠后,小鼠的体液免疫应答水平(纵坐标为log以10为底的对数);
图2B:不同剂量的RABV-G mRNA疫苗加强免疫小鼠后,小鼠的体液免疫应答水平(纵坐标为log以10为底的对数);
图2C:不同剂量的RABV-G mRNA疫苗初免后小鼠体内诱导的抗体针对狂犬病病毒的中和效果;
图2D:不同剂量的RABV-G mRNA疫苗加强免疫后小鼠体内诱导的抗体针对狂犬病病毒的中和效果。
ns:表示无显著性差异(not significant);*表示p<0.05;**表示p<0.01;***表示p<0.001;****表示p<0.0001。
图3:RABV-G mRNA疫苗诱导的抗原特异性T细胞应答:
图3A:不同剂量的RABV-G mRNA疫苗诱导的抗原特异性的CD8 +T细胞(IFNγ阳性CD8 +T细胞)应答;
图3B:不同剂量的RABV-G mRNA疫苗诱导抗原特异性的CD4 +T细胞(IFNγ阳性CD4 +T细胞)应答;
图3C:不同剂量的RABV-G mRNA疫苗诱导抗原特异性的CD4 +T细胞(IL-4阳性CD8 +T细胞)应答。
ns:表示无显著性差异(not significant);*表示p<0.05;**表示p<0.01。
图4:单剂量1μg组的RABV-G mRNA疫苗攻毒保护效果:
图4A:攻毒保护后动物的体重变化结果;
图4B:攻毒保护后动物的存活率变化。
具体实施方式
针对现有技术中存在的问题,本文中提供一种可编码RABV-G蛋白的mRNA载体,该mRNA可高效的表达狂犬病病毒的糖蛋白,可用于狂犬病的预防和治疗。本文中提供的RABV-G mRNA狂犬病疫苗,采用较低剂量,单剂量肌肉注射,接种后14天即可诱导高滴度具有保护效果的抗体,并可保护动物能够完全抵抗致死性剂量狂犬病病毒的攻击,也可有效诱导针对RABV-G特异性T细胞应答。本文的RABV-G mRNA疫苗经2针免疫后可诱导高于WHO检测标准(0.5IU/mL)约2000倍的有效保护性抗体。本文的mRNA狂犬病疫苗具有良好的免疫原性,并在免疫机体后形成强效的免疫保护,且生产成本低、制备技术操作简便,适用于快速高效、规模化的疫苗制备,具有巨大的开发潜力。
本文中提供的所有数值范围旨在清楚地包括落在范围端点之间的所有数值及它们之间的数值范围。可对本发明提到的特征或实施例提到的特征进行组合。本说明书所揭示的所有特征可与任何组合物形式并用,说明书中所揭示的各个特征,可以任何可提供相同、均等或相似目的的替代性特征取代。因此除有特别说明,所揭示的特征仅为均等或相似特征的一般性例子。
如本文所用,“含有”、“具有”或“包括”包括了“包含”、“主要由……构成”、“基本上由……构成”、和“由……构成”;“主要由……构成”、“基本上由……构成”和“由……构成”属于“含有”、“具有”或“包括”的下位概念。
如本文所用,“真核生物”可包括人、灵长类动物、啮齿动物(例如大鼠、小鼠、豚鼠、仓鼠)、驯养动物或畜牧哺乳动物。
本文所述的高度序列同一性,包括具有70%以上、75%以上、80%以上,更优选85%以上,如85%、90%、95%、98%甚至99%或以上的序列同一性,这些高同一性序列也在本发明优选考虑的等同范围之内。比对序列同一性的方法和工具也是本领域周知的,如BLAST。
狂犬病免疫原核酸表达载体及包含该载体的疫苗
本文中提供了一种狂犬病免疫原核酸表达载体,其从5′端至3′端依次包含:
(a)5′-UTR元件;
(b)编码狂犬病病毒糖蛋白(RABV-G)的开放阅读框元件;
(c)3′-UTR元件;
(d)总长度为120nt以上的多聚腺苷酸尾元件,其包含:
多个腺苷酸串,每个腺苷酸串各自独立地包含n个连续的腺苷酸,n为10~80之间的整数,且所述多个腺苷酸串的腺苷酸总个数为100个以上;
位于所述多个腺苷酸串之间的连接子,所述连接子各自独立地不包含腺苷酸或仅包含1个或2个腺苷酸。
在一些实施方式中,所用5′-UTR元件的长度为10~200nt,例如15~100nt。在一些实施方式中,所用5′-UTR元件为来自细胞内高翻译效率丰度高的蛋白质。
在一些实施方式中,所用5′-UTR元件源自下组中的一种或多种5′-UTR:人α-球蛋白、β-球蛋白、核糖体蛋白(RP)、微管蛋白β-2B、补体因子3(C3)、细胞色素P4502E1(CYP2E1)、载脂蛋白A-II(APOA2)、人类血红蛋白亚基β(hHBB)、血红蛋白A1(HBA1)、血红蛋白A2(HBA2)、登革热病毒(DENV)。
在一些实施方式中,所用5′-UTR元件具有如SEQ ID NO:2所示的序列或与其具有至少80%的序列同一性。
在一些实施方式中,所用3′-UTR元件为来自细胞内高翻译效率丰度高的蛋白质。在一些实施方式中,所用3′-UTR元件为来源于哺乳动物或病毒的 3′-UTR,例如来源选自下组的3′-UTR或其组合(如串联序列):人α球蛋白、人β球蛋白、人白蛋白、人肌动蛋白、人血红蛋白亚基α1(HBA1)、细胞色素B-245α链(CYBA)、真核生物线粒体(Mit)的序列、SARAS-Cov-2、登革热病毒(DENV)、萝卜皱病毒(TCV)、烟草花叶病毒(TMV)和烟草蚀刻病毒(TEV)。
在一些实施方式中,所用3′-UTR元件包含选自下组一个或多个3′-UTR分子:α球蛋白3′-UTR、真核生物线粒体3′-UTR、白蛋白3′-UTR、β球蛋白3′-UTR或其任意串联序列,优选α球蛋白3′-UTR、真核生物线粒体3′-UTR、或它们串联形成的3′-UTR。
在一些实施方式中,所用3′-UTR具有如SEQ ID NO:3所示的序列,或与其具有至少80%的序列同一性的序列。
在一些实施方式中,所用多聚腺苷酸尾元件的总长度为120~400nt,例如120~350nt,120~320nt,或其中的任意整数,例如120、304nt。在一些实施方式中,各腺苷酸串各自独立地包含10~80个、20~70个、25~60个、30~50个或其中任意整数个连续的腺苷酸,如20个、30个、33个、35个、36个、37个、38个、39个、40个、45个、50个、55个、60个、65个、70个连续腺苷酸。
在一些实施方式中,多聚腺苷酸尾元件还进一步包含位于所述元件一个或两个端部的连接子。在一些实施方式中,连接子的长度各自独立地为3~15nt,例如3、4、5、6、7、8、9、10、11、12、13、14或15nt。在一些实施方式中,连接子各自独立地不包含腺苷酸,或仅包含1个腺苷酸或2个腺苷酸。
在一些实施方式中,连接子的序列各自独立地选自:GCTATGACT、GTATGT、GCAAGT、GATTGC、GGCTGC、TACTGC、GGCTTC、GCATATGACT。
在一些实施方式中,多聚腺苷酸尾元件具有SEQ ID NO:4的序列,或与其中任何一者具有至少80%的序列同一性。
在一些实施方式中,所用开放阅读框元件为单顺反子、双顺反子或多顺反子mRNA。在一些实施方式中,开放阅读框元件编码一个或多个来源相同或不同的RABV-G。
在一些实施方式中,RABV-G来源于选自下组的狂犬病病毒:巴斯德毒株 中的Pitman-Moore(PM)株、Pasteur(PV)株、CTN株、aG株、Flury-LEP株、Evelyn-Rokitnicki-Abelseth(ERA)株、Street-Alabama-Duffering(SAD)株、KHUV(Khujand lyssavirus)、BBLV(Bokeloh bat lyssavirus)、ARAV(Aravan lyssavirus)、EBLV-1(European bat 1 lyssavirus)、EBLV-2(European bat 2 lyssavirus)、IRKV(Irkutlyssavirus)、LBV(Lagos bat lyssavirus)、SHIBV(Shimoni bat lyssavirus)、MOKV(Mokola lyssavirus)、WCBV(West Caucasian bat lyssavirus)、IKOV(Ikomalyssavirus)、DUVV(Duvenhagelyssavirus)、ABLV(Australian bat lyssavirus)、GBLV(Gannoruwa bat lyssavirus)或LLEBV(Lleida bat lyssavirus)。
在一些实施方式中,RABV-G为产生狂犬病病毒中和抗体的主要免疫原,在多种狂犬病病毒间具有高度保守性。例如,RABV-G可来源于Pitman-Moore(PM)株。
在一些实施方式中,RABV-G包括选自下组的一种或多种分子:未经修饰或经真核修饰的RABV-G、其免疫原性片段或变体,所述分子能够诱导针对RABV的免疫中和和保护反应。
在一些实施方式中,编码RABV-G的元件经过或未经密码子优化、包含或不包含碱基修饰和/或核苷类似物。在一些实施方式中,编码序列根据真核生物密码子的偏好性进行优化。
在一些实施方式中,编码RABV-G的元件中的一个或多个尿嘧啶被选自下组中的一个或多个相同或不同的修饰碱基或核苷类似物置换:假尿苷、1-甲基尿嘧啶核苷、N1-乙基假尿苷、2-硫尿苷、4′-硫尿苷、5-甲基胞嘧啶、5-甲基尿苷、2-硫基-1-甲基-1-去氮杂-假尿苷、2-硫基T-甲基-假尿苷、2-硫基-5-氮杂-尿苷、2-硫基-二氢假尿苷、2-硫基-二氢尿苷、2-硫基-假尿苷、4-甲氧基-2-硫基-假尿苷、4-甲氧基-假尿苷、4-硫基-1-甲基-假尿苷、4-硫基-假尿苷、5-氮杂-尿苷、二氢假尿苷或5-甲氧基尿苷和2′-O-甲基尿苷,优选假尿苷或N1-甲基假尿苷或N1-乙基假尿苷,进一步优选为N1-甲基假尿苷。在一些实施方式中,编码狂犬病病毒糖蛋白(RABV-G)的开放阅读框元件中50%~100%的尿嘧啶被置换。在一些实施方式中,表达载体序列中的50%~100%尿嘧啶被置换。通过置换能够提高mRNA在生物体内的稳定性。
在一些实施方式中,所述表达载体还包括5′-帽元件,其可选被修饰,例如所述5′-帽元件选自:m7GpppXpYp、m7GpppXmpYp、m7GpppXmpYmp、或其甲基化修饰序列、反向结合异构体、抗-反转帽类似物(ARCA)、N7-苄基二核苷四磷酸帽类似物。
在一些实施方式中,所述表达载体还包括启动子元件,例如T7启动子、sp6启动子或T3启动子。在一些实施方式中,所述表达载体还包括信号肽编码元件,例如指导目的蛋白亚细胞定位的信号肽(如跨膜信号肽、分泌信号肽、核定位信号肽)编码元件。在一些实施方式中,所述表达载体还包括酶切位点,例如XbaI、EcoRV、BamHI、XhoI。在一些实施方式中,所述表达载体还包括标签,例如用于目的分子鉴定、分离或纯化的分子标签,如Flag标签、HA标签。
在一些实施方式中,所述表达载体从5′端到3′端包含:包含SEQ ID NO:2所示序列的5′-UTR元件;编码RABV-G的开放阅读框元件;包含如SEQ ID NO:3所示序列的3′-UTR元件;包含如SEQ ID NO:4所示序列的多聚腺苷酸尾元件;或与所述各序列各自具有至少80%序列同一性的序列的元件。
在一些实施方式中,所述表达载体包含如SEQ ID NO:5或6所示的序列、或与其具有至少80%序列同一性的序列。
在一些实施方式中,所述核酸表达载体单独包含于包装物中,或与递送系统中的运载体组合,例如,所述递送系统选自:脂质递送系统、类脂递送系统、聚合物递送系统或其组合递送系统,例如加载于脂质纳米颗粒、阳离子脂质体、聚氨酯(PAA)、聚β氨基酯(PBAE)、聚乙烯亚胺(PEI)、脂质包裹的聚合物胶束。
在一些实施方式中,所述核酸表达载体与脂质体纳米颗粒组合。在一些实施方式中,脂质体纳米颗粒包括:阳离子脂质、结构脂质、辅助脂质和稳定脂质的组合。
在一些实施方式中,按摩尔百分比计算,所述脂质体纳米颗粒包括:20~50%阳离子脂质:20~50%结构脂质:5~20%辅助脂质:1~5%稳定脂质,更优选为50%阳离子脂质:38%结构脂质:10%辅助脂质:2%稳定脂质。
在一些实施方式中,脂质体纳米颗粒中的阳离子脂质为选自下组中的一种 或多种:(2,3-二油酰基-丙基)-三甲基氯化铵(DOTAP)、己二酸二癸酯(DDA)、3β-[N-(N′,N′-二甲基胺乙基)胺基甲酰基]胆固醇(DC-Chol)、N,N-二甲基-4-吡啶胺(DMAP),1,2-三乙醇胺-3-三甲基丙烷(DOTMA)、N-[1-(2,3-二油酰基)丙基]-N-(精氨酸基酰胺)乙基-N,N-二甲基三氟乙酸铵(DOSPA)、4-(N,N-二甲基氨基)丁酸(二亚油基)甲酯(Dlin-MC3-DMA)、KC2、N,N-二甲基-2,2-二-(9Z,12Z)-9,12-十八碳二烯-1-基-1,3-二氧′戊环-4-乙′胺(Dlin-KC2-DMA),优选为4-(N,N-二甲基氨基)丁酸(二亚油基)甲酯(Dlin-MC3-DMA)。
在一些实施方式中,脂质体纳米颗粒中的结构脂质包括胆固醇、胆固醇脂、固醇类激素、固醇类维生素或胆汁酸,优选为胆固醇。
在一些实施方式中,脂质体纳米颗粒中的辅助脂质包括二棕榈酰磷脂酰胆碱(DPPC)、二油酰磷脂酰甘油(DOPG)、1-棕榈酰基-2-油酰基卵磷脂(POPC)、1,2二硬脂酸-3-磷脂酰乙醇胺(DSPE)、二油酰基卵磷脂(DOPC)、二油酰基磷脂酰丝氨酸(DOPS)、二硬脂酰磷脂酰胆碱(DSPC),优选的二硬脂酰磷脂酰胆碱(DSPC)。
在一些实施方式中,脂质体纳米颗粒中的稳定脂质包括聚乙二醇(PEG)-脂质,如PEG修饰的磷脂酰乙醇胺、PEG修饰的磷脂酸、PEG修饰的神经酰胺、PEG修饰的二酰基甘油,优选为PEG修饰的二酰基甘油,更优选为长循环脂质体1,2-二肉豆蔻酰-rac-甘油-3-甲氧基聚乙二醇2000(PEG 2000-DMG)。
在一些实施方式中,脂质体纳米颗粒中的阳离子脂质体与RABV-G mRNA质量比为10~30∶1,优选为15~20∶1。
在一些实施方式中,阳离子脂质纳米颗粒与优化后核苷修饰的RABV-G mRNA混合后,制备方法如下:将可电离阳离子脂质,结构脂质,辅助脂质和稳定脂质按配方比例溶解于乙醇中,制备得到有机相。将优化后核苷修饰的RABV-G mRNA溶于柠檬酸缓冲液(pH4.0)中,制备得到水相。使用推注混合器将有机相和水相以一定比例混合得到混合液,通过透析至PBS替换外部缓冲液,0.22μm无菌过滤器过滤混合液得到狂犬病毒核酸疫苗。所述的有机相和水相的比例1∶2至1∶5,优选为3∶7。
在本文的一些方面中,提供了一种狂犬病mRNA疫苗,其包含:如本文 所述的狂犬病免疫原核酸表达载体,以及包装物和/或用于该核酸表达载体的递送系统和/或药学上或生理学上可接受的运载体。
在一些实施方式中,疫苗中的核酸表达载体单独包含于包装物中,或与递送系统中的运载体组合,例如,所述递送系统选自:脂质递送系统、类脂递送系统、聚合物递送系统或其组合递送系统,例如加载于脂质纳米颗粒、阳离子脂质体、聚氨酯(PAA)、聚β氨基酯(PBAE)、聚乙烯亚胺(PEI)、脂质包裹的聚合物胶束。
在一些实施方式中,疫苗中的核酸表达载体与脂质体纳米颗粒组合。在一些实施方式中,脂质体纳米颗粒包括:阳离子脂质、结构脂质、辅助脂质和稳定脂质的组合。
在一些实施方式中,按摩尔百分比计算,所述脂质体纳米颗粒包括:20~50%阳离子脂质∶20~50%结构脂质∶5~20%辅助脂质∶1~5%稳定脂质,更优选为50%阳离子脂质∶38%结构脂质∶10%辅助脂质∶2%稳定脂质。
在一些实施方式中,脂质体纳米颗粒中的阳离子脂质为选自下组中的一种或多种:(2,3-二油酰基-丙基)-三甲基氯化铵(DOTAP)、己二酸二癸酯(DDA)、3β-[N-(N′,N′-二甲基胺乙基)胺基甲酰基]胆固醇(DC-Chol)、N,N-二甲基-4-吡啶胺(DMAP),1,2-三乙醇胺-3-三甲基丙烷(DOTMA)、N-[1-(2,3-二油酰基)丙基]-N-(精氨酸基酰胺)乙基-N,N-二甲基三氟乙酸铵(DOSPA)、4-(N,N-二甲基氨基)丁酸(二亚油基)甲酯(Dlin-MC3-DMA)、KC2、N,N-二甲基-2,2-二-(9Z,12Z)-9,12-十八碳二烯-1-基-1,3-二氧′戊环-4-乙′胺(Dlin-KC2-DMA),优选为4-(N,N-二甲基氨基)丁酸(二亚油基)甲酯(Dlin-MC3-DMA)。
在一些实施方式中,脂质体纳米颗粒中的结构脂质包括胆固醇、胆固醇脂、固醇类激素、固醇类维生素或胆汁酸,优选为胆固醇。
在一些实施方式中,脂质体纳米颗粒中的辅助脂质包括二棕榈酰磷脂酰胆碱(DPPC)、二油酰磷脂酰甘油(DOPG)、1-棕榈酰基-2-油酰基卵磷脂(POPC)、1,2二硬脂酸-3-磷脂酰乙醇胺(DSPE)、二油酰基卵磷脂(DOPC)、二油酰基磷脂酰丝氨酸(DOPS)、二硬脂酰磷脂酰胆碱(DSPC),优选的二硬脂酰磷脂酰胆碱(DSPC)。
在一些实施方式中,脂质体纳米颗粒中的稳定脂质包括聚乙二醇(PEG)-脂质,如PEG修饰的磷脂酰乙醇胺、PEG修饰的磷脂酸、PEG修饰的神经酰胺、PEG修饰的二酰基甘油,优选为PEG修饰的二酰基甘油,更优选为长循环脂质体1,2-二肉豆蔻酰-rac-甘油-3-甲氧基聚乙二醇2000(PEG 2000-DMG)。
在一些实施方式中,脂质体纳米颗粒中的阳离子脂质体与RABV-G mRNA质量比为10~30∶1,优选为15~20∶1。
在一些实施方式中,阳离子脂质纳米颗粒与优化后核苷修饰的RABV-G mRNA混合后,制备方法如下:将可电离阳离子脂质,结构脂质,辅助脂质和稳定脂质按配方比例溶解于乙醇中,制备得到有机相。将优化后核苷修饰的RABV-G mRNA溶于柠檬酸缓冲液(pH4.0)中,制备得到水相。使用推注混合器将有机相和水相以一定比例混合得到混合液,通过透析至PBS替换外部缓冲液,0.22μm无菌过滤器过滤混合液得到狂犬病毒核酸疫苗。所述的有机相和水相的比例1∶2至1∶5,优选为3∶7。
在一些实施方式中,所述疫苗的形式适于选自下组的一种或多种给予或递送方式:呼吸道雾化吸入、滴鼻、口服、直接注射(例如静脉注射、皮下注射、皮内注射、肌肉注射)、黏膜给药。
在一些实施方式中,所述疫苗还包含佐剂或与佐剂联合使用,例如所述佐剂选自:铝佐剂、霍乱毒素及其亚单位、寡脱氧核苷酸、锰离子佐剂、胶体锰佐剂、弗氏佐剂、MF59佐剂、QS-21佐剂、Poly I:C及其他TLR配体、GM-CSF、IL-2、IL-3、IL-7、IL-11、IL-12、IL-18、IL-21。
在一些实施方式中,疫苗的形式适于进行2种或以上药物或疫苗的组合给予,例如联合接种或序贯接种。
制备方法、相关产品及其应用
在本文的一些方面中,提供了制备本文所述的核酸表达载体或疫苗的方法,所述方法包括:提供独立或连接的各元件;将各元件组装成核酸表达载体。
在一些实施方式中,所述方法包括采用选自下组的一种或多种材料:DNA模板(例如PCR产物或已线性化的质粒DNA)、核酸酶、聚合酶、加帽酶、聚 腺苷酸合酶、DNA酶、一个或多个元件分子、接头分子、天然或修饰的核酸分子、缓冲液、溶剂。
在一些实施方式中,所述方法还包括选自下组的一个或多个步骤:对各元件进行设计、优化、改造和/或修饰;对中间产物和/或最终产物进行分离、纯化、鉴定、定量、包装和/或活性测试;将核酸表达载体与用于该核酸表达载体的递送系统和/或药学上或生理学上可接受的运载体组合。
在本公开的一些方面中,提供了本文的核酸表达载体和/或疫苗在制备用于预防和/或治疗狂犬病的产品中的应用。在一些实施方式中,所述狂犬病为由以下一种或多种狂犬病病毒引起的狂犬病:巴斯德毒株中的Pitman-Moore(PM)株、Pasteur(PV)株、CTN株、aG株、Flury-LEP株、Evelyn-Rokitnicki-Abelseth(ERA)株、Street-Alabama-Duffering(SAD)株、KHUV(Khujand lyssavirus)、BBLV(Bokeloh bat lyssavirus)、ARAV(Aravan lyssavirus)、EBLV-1(European bat 1 lyssavirus)、EBLV-2(European bat 2 lyssavirus)、IRKV(Irkutlyssavirus)、LBV(Lagos bat lyssavirus)、SHIBV(Shimoni bat lyssavirus)、MOKV(Mokola lyssavirus)、WCBV(West Caucasian bat lyssavirus)、IKOV(Ikomalyssavirus)、DUVV(Duvenhagelyssavirus)、ABLV(Australian bat lyssavirus)、GBLV(Gannoruwa bat lyssavirus)或LLEBV(Lleida bat lyssavirus)。
如本文所用,术语“药学上或生理上可接受的”成分是适用于人和/或动物而无过度不良副反应(如毒性、刺激和变态反应)的,即有合理的效益/风险比的物质。如本文所用,术语“有效量”是指可对人和/或动物产生功能或活性的且可被人和/或动物所接受的量。
如本文所用,术语“药学上可接受的运载体”指用于治疗剂给药的载体,包括各种赋形剂和稀释剂。该术语指这样一些药剂载体:它们本身并不是必要的活性成分,且施用后没有过分的毒性。合适的载体是本领域普通技术人员所熟知的。在《雷明顿药物科学》(Remington′s Pharmaceutical Sciences,Mack Pub.Co.,N.J.1991)中可找到关于药学上可接受的赋形剂的充分讨论。
在组合物中药学上可接受的运载体可含有液体,如水、盐水、甘油和乙醇。另外,这些载体中还可能存在辅助性的物质,如填充剂、崩解剂、润滑剂、 助流剂、泡腾剂、润湿剂或乳化剂、矫味剂、pH缓冲物质等。通常,可将这些物质配制于无毒的、惰性的和药学上可接受的水性载体介质中,其中pH通常约为5-8,较佳地,pH约为6-8。
如本文所用,术语“单位剂型”是指为了给药方便,将本文的活性物质制备成单次给药所需的剂型,包括但不限于各种固体剂(如片剂)、液体剂、胶囊剂、缓释剂。
应理解,所用活性物质的有效剂量可随待施用或治疗的对象的严重程度而变化。具体情况根据对象的个体情况(例如对象体重、年龄、身体状况、所需达到的效果)来决定,这在熟练医师可以判断的范围内。
本文的产品可以为固态(如颗粒剂、片剂、冻干粉、栓剂、胶囊、舌下含片)或液态(如口服液)或其它合适的形状。给药途径可采用:(1)直接裸核酸注射法;(2)将mRNA表达载体与转铁蛋白/多聚L-赖氨酸复合物连接,以增强其生物效应;(3)使mRNA表达载体与带正电荷的脂类形成复合物,以克服磷酸骨架负电荷所致的穿越细胞膜的困难;(4)用脂质体包裹mRNA表达载体后介导进入细胞,既有利于大分子的顺利进入又免受细胞外各种酶的水解作用;(5)使mRNA表达载体与胆固醇结合使其保持时间增加;(6)用免疫脂质体转运mRNA以使其特异性转运至靶组织和靶细胞;(7)将mRNA表达载体体外转染给转载细胞;(8)电打孔(electroporation),即借助于电流将mRNA载体导入靶细胞。
本文还提供了一种用于预防和/或治疗狂犬病病毒感染和/或其症状的方法,其包括:至少一次给予预防和/或治疗有效量的本公开的一种或多种疫苗。可采用的接种方式包括但不限于:系统性免疫接种方式,如肌肉注射、皮下注射和皮内注射等,优选为肌肉注射和皮下注射,从给药程度的难易更优选为肌肉注射;呼吸道内免疫接种方式,如雾化、滴鼻等。在一些实施方式中,初次免疫采用系统性接种或呼吸道内接种,优选系统性接种。
在本公开的一些实施方式中,每两次接种之间的间隔至少为1周,例如2周、4周、2个月、3个月、6个月或更长间隔。
在一些实施方式中,采用mRNA疫苗进行初次免疫,并采用同种或其他 类型的疫苗进行一次或多次加强免疫。本公开的免疫方法可采用“初免-加强”或“初免-加强-再加强”的方式,可采用单一的全身系统免疫或呼吸道局部免疫方式,或采用两种免疫方式的组合。
在一些实施方式中,以药物包或试剂盒的形式提供本文的组合产品,例如可将本文的一种或多种疫苗组合物或其一种或多种成分包装在一个或多个容器中,例如包装在指明组合物的量的密封容器诸如安瓿或小药囊中。可以液体、无菌冻干粉或无水浓缩物等形式提供疫苗组合物,可在临用前用适当液体(例如水、盐水等)对其进行稀释、复原和/或配制以获得用于给予至对象的适当浓度和形式。
实施例
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。本领域技术人员可对本发明做出适当的修改、变动,这些修改和变动都在本发明的范围之内。
下列实施例中未注明具体条件的实验方法,可采用本领域中的常规方法,例如参考《分子克隆实验指南》(第三版,纽约,冷泉港实验室出版社,NewYork:Cold Spring Harbor Laboratory Press,1989)或按照供应商所建议的条件。DNA的测序方法为本领域常规的方法,也可由商业公司提供测试。
除非另外说明,否则百分比和份数按重量计算。除非另行定义,文中所使用的所有专业与科学用语与本领域熟练人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本发明方法中。文中所述的较佳实施方法与材料仅作示范之用。
材料、方法与动物
实施例中的实验中所涉及到的RABV-G mRNA制备、阳离子脂质体纳米颗粒制备、动物免疫方案及检测方法如下:
I.RABV-G mRNA的制备
制备合适的DNA模板:以pCDNA3.1+为模板,进行改造,在T7启动子 后从N端-C端依次包括:5′-UTR序列(SEQ ID NO:2)、编码序列(SEQ ID NO:1)、3′-UTR序列(SEQ ID NO:3)、含250个间插有连接子序列的连续腺苷酸的聚腺苷酸尾序列(SEQ ID NO:4)、线性化酶切位点XbaI,将修饰的狂犬病Pitman-Moore(PM)疫苗株糖蛋白(RABV-G)编码序列作为免疫原编码序列,插入到编码序列,并在编码序列C端插入酶切位点EcoRV和Flag标签,加入Flag标签以便后续的基因表达鉴定。其中,所构建的DNA模板序列(SEQ ID NO:6)根据真核生物密码子的偏好性进行优化,用于后续的体外转录实验。
体外转录、优化与修饰:转录试剂盒购自近岸蛋白质科技有限公司,以SEQ ID NO:6所述的质粒单酶切后为模板,按照合适的比例加入转录所需的NTP,并用1-甲基尿嘧啶核苷(ψ)替代尿嘧啶核苷(U),使用T7聚合酶,转录得到相应的RABV-G mRNA(SEQ ID NO:5,5′UTR+RABV-G mRNA+(α-globin+Mit)3′UTR+250A,其中U被ψ取代,且含密码子优化),然后通过酶加帽法同时使用牛痘病毒加帽酶和2′-O-甲基转移酶一步法进行加帽,将纯化后的RABV-G mRNA加上7-甲基鸟苷帽结构,用氯化锂纯化加帽后的mRNA,用于脂质纳米颗粒的制备。
修饰后的RABV-G mRNA序列
(转录后的产物序列,其中尿嘧啶核苷(U)替换为1-甲基尿嘧啶核苷(Ψ))
Figure PCTCN2022119230-appb-000005
Figure PCTCN2022119230-appb-000006
II.包裹RABV-G mRNA的阳离子脂质纳米颗粒的制备
委托迈安纳(上海)仪器科技有限公司制备狂犬病mRNA脂质体纳米颗粒:将阳离子脂质Dlin-MC3-DMA、结构脂质胆固醇、辅助脂质DSPC和稳定脂质DMG-PEG2000,按50∶38∶10∶2的摩尔比例溶解于乙醇中,乙醇浓度为30%(v/v),得到油相混合液。然后,将该油相混合液于室温加入到50mM pH 4.0的柠檬酸盐缓冲液中得到脂质混合物。将该脂质混合物加入脂质体挤出器,先用200nm滤膜挤出过滤,然后再用100nm滤膜过滤,使溶液从乳白变为澄清,得到阳离子脂质体纳米颗粒。
将优化后核苷修饰的RABV-G mRNA溶于柠檬酸盐缓冲液(pH 4.0)中,按照阳离子脂质纳米颗粒与mRNA质量比为20∶1的比例,逐滴加入至阳离子脂质纳米颗粒中,使用振荡仪Vortex混匀得到混合液。彻底混匀后42℃金属浴加热孵育1小时,随后将混合液透析至无菌PBS,0.22μm无菌过滤器过滤后得到RABV-G mRNA阳离子脂质纳米颗粒,即狂犬病病毒核酸疫苗。
III.动物免疫方案
小鼠免疫实验分为2种方案,第一种方案将6~8周龄雌性BALB/c小鼠,随机分为4组,每组12只(其中6只用于体液应答检测,6只用于检测细胞免疫应答),即阴性对照组、RABV-G高剂量组、RABV-G中剂量组、RABV-G 低剂量组。各组均采用肌肉注射,试验第1天进行初次免疫,试验第21天进行加强免疫,每次免疫剂量相同,见表1。第二种免疫方案为一针免疫接种,选择RABV-G低剂量组,每组8只。单次免疫体积均为100μl。
表1
Figure PCTCN2022119230-appb-000007
其中,RABV-G高剂量组、中剂量组、低剂量组的免疫剂量均指的是每次mRNA的免疫剂量。
IV.检测方法
A.米血:
小鼠在第1天进行初次免疫前采血,随后分别在第14天、第21天、第28天、第35天眼眶静脉丛采血,并于最后一次免疫后4周脱颈椎处死。将采集的小鼠全血收集于1.5mL无菌EP管,37℃静置使其自然凝血,将凝固后的小鼠血清于7000g,离心15min。随后将血清在56℃灭活30min,去除其补体活性。一针免疫接种组免疫4周后直接攻毒,不需要再采血。
B.酶联免疫吸附(ELISA)检测RABV-G结合抗体滴度:
用包被缓溶液(50mM碳酸盐缓冲液,pH=9.6)将RABV-G蛋白(武汉普建生物,ATAPl0593)稀释到1ng/μL,100μl/孔,4℃孵育过夜。用PBST(含0.05%Tween-20)洗板1次,加入5%脱脂牛奶200μl/孔,37℃封闭2h。以5%脱脂牛奶为样品稀释液,将血清样品首孔稀释100倍,进行2倍倍比梯度稀释,加入至反应孔中,37℃反应1h。用PBST(含0.05%Tween-20)洗板5次后,拍干,加入HRP标记的山羊抗小鼠IgG抗体(1∶5000),100μl/孔,37℃下反应1h。用PBST(含0.05%Tween-20)洗涤6次,拍干后避光加入TMB底物缓冲液反应10min,加入2M硫酸终止液中止反应。在酶标仪450nm处读值(Biotek公司, 酶标仪)。
分析数据,以阴性对照组读值的2.1倍为cut-off值,计算RABV-G特异性结合抗体稀释终点对应的滴度,以滴度的几何平均值(GMT)表示,利用Graphpad Prism8软件作图。
C.荧光抗体病毒中和试验(FAVN)检测血清真病毒中和抗体滴度:
参考英国犬源国际兽疫局(OIE)狂犬病实验室的标准操作程序。各孔内首先加入100μL DMEM完全培养基,首列孔加入50μL预先灭活后的血清样品,以多道移液器混匀,吸取50μL进入第2列,同法稀释至最后一列,混匀后吸弃50μL。取另一96孔板为对照板,按上述方式对标准阳性血清(军事医学科学院兽医研究所)(0.5IU/mL)、标准阴性血清(非免疫小鼠血清)进行稀释,同时设病毒重复滴定区,以及病毒对照、BHK-21细胞对照、培养基对照区。然后在每孔中加入50μL的100TCID50狂犬病病毒(病毒标准株CVS-11株),细胞及培养基对照孔不加病毒,只加DMEM完全培养基。在37℃和5%CO 2条件下孵育1h。随后每孔中加入50μL的BHK-21细胞悬液,细胞密度为4E5个/mL,培养基对照孔不加细胞,在37℃和5%CO 2条件下孵育48h。孵育结束后弃细胞上清,进行固定和染色。先加80%丙酮溶液固定30min,弃固定液,晾干。加入抗狂犬病病毒核衣壳蛋白标记FITC的荧光抗体,每孔加入50μL1%的伊文思蓝染色液,37℃避光反应45~60min。荧光显微镜下观察是否存在一个或多个荧光细胞,将孔定性地分为“阳性”(有荧光为感染)或“阴性”(无荧光)。
血清滴度是50%孔中和100%病毒的稀释量(log D50)。0.5IU/ml为WHO推荐中和标准,在相同的实验条件下,具体计算待测血清和犬源国际兽疫局(OIE)参比血清的中和稀释液对定量病毒的半数中和稀释倍数,二者相比,再乘以标准血清效价(0.5IU/ml)即为待测血清狂犬病中和效价,以中和效价滴度的几何平均值(GMT)表示,用Graphpad Prism8软件作图。
D.胞内细胞因子染色法(ICS)分析RABV-G抗原特异性T细胞:
分离免疫后的小鼠脾脏细胞,制成单细胞悬液。每孔1E6个细胞铺于96孔中,加入终浓度为5μg/mL的狂犬病病毒糖蛋白肽库刺激,阴性对照组为 DMSO组,阳性对照组为终浓度50ng/mL的PMA和5μg/mL的Ionomycin,37℃和5%CO 2条件下孵育1小时,加入BD GolgiStop TM Protein Transport Inhibitor(1∶1000),37℃培养箱孵育5h,刺激结束。500g离心5min,PBS洗一次,然后进行荧光抗体染色(染色体系均为100μL,下同)。先用Zombie AquaTM,小鼠CD8a-PB,小鼠CD4PerCP-Cy5.5表染,室温下避光染色20min。PBS洗一次,500g离心5min。然后用Cytofix/Cytoperm(BD)固定,室温下避光固定20min,PBS洗一次,800g离心5min。通透细胞:将10x BD Perm/Wash TM缓冲液用超纯水稀释为1×工作液使用,用1x BD Perm/Wash TM缓冲液重悬细胞后,室温下避光孵育15min。800g离心5min,小心去除上清。用1x BD Perm/Wash TM缓冲液稀释小鼠IFN-GMA FITC和小鼠IL-4APC,加入细胞悬液中染色。所有抗体均购自BD Biosciences。BD LSRFortessa流式细胞分析仪进行数据采集;每个样本至少收集30,000个细胞,随后使用FlowJo软件进行数据分析。
下面结合附图和具体实验进一步详细说明本申请的内容。除非特别说明,所用试剂、仪器、设备和方法均为本技术领域的常规市购试剂、仪器、设备和方法。
实施例1.RABV-G mRNA转染HEK293T细胞表达验证
用按上述实验方法I中制备的RABV-G mRNA转染HEK293T细胞进行表达验证。转染前24h将HEK293细胞以200000个细胞/孔的密度接种到12孔板,培养基为DMEM完全培养基(10%FBS和1%P.S.),转染试剂为Lipofectamine 3000,每孔板转染2μg RABV-G mRNA,37℃孵箱培养40~48h。收集细胞到预冷的EP管,加入RIPA裂解缓冲液裂解细胞。进行SDS-聚丙烯酸胺凝胶电泳(SDS-PAGE),转膜完毕后,置于5%的脱脂奶粉中室温封闭1小时。加入一抗(Rab-50,Santa Cruz,1∶2000),置于摇床4℃孵育过夜。PBST(含0.05%Tween-20)洗膜后加入羊抗鼠二抗(翊圣,33201ES60,1∶5000),室温摇床孵育1小时,PBST(含0.05%Tween-20)洗5次,每次洗3min。对膜进行显色, 胶片采用定量分析仪曝光2分钟,记录并分析显色结果。
结果如图1所示,RABV-G mRNA转染HEK293T细胞后大量表达,可被抗狂犬病病毒抗体特异性识别,目的条带大小在60kDa位置,大小正确。
以上结果表明所构建的RABV-G mRNA可高效转染细胞,并有效表达目的蛋白。
实施例2.RABV-G mRNA疫苗诱导小鼠的体液免疫应答
本实施例中选用的mRNA为实施例一中验证表达正确的RABV-G mRNA,按上述实验方法II制备包裹RABV-G mRNA的阳离子脂质纳米颗粒。
用RABV-G mRNA脂质纳米颗粒疫苗免疫BALB/c小鼠评价其免疫原性,分别选取10μg(高,组4)、3μg(中,组3)和1μg(低,组2)三种不同的剂量,每组6只小鼠,肌肉注射,每只注射体积为100μl,阴性对照为等体积的未包裹核酸的阳离子脂质纳米颗粒。三种不同的剂量的疫苗及阴性对照组第1天1次给药后,在21天接受加强注射。分别在第14天、21天、28天、35天收集血液样品。通过酶联免疫吸附检测RABV-G特异性结合抗体滴度,荧光抗体病毒中和试验检(FAVN)检测血清真病毒中和抗体滴度。
酶联免疫吸附检测RABV-G特异性结合抗体滴度的结果如图2A和2B所示。如图2A所示,第1次给药后的第14天、第21天10μg组免疫小鼠血清RABV-G抗体滴度分别为12800、22807,3μg组免疫小鼠血清RABV-G IgG抗体滴度分别为3591.9、5701.5,1μg组免疫小鼠血清RABV-G抗体滴度分别为1600、2015.9。然后在第21天接受加强注射,结果如图2B所示,在加强注射后的第7天、第14天,10μg组免疫小鼠RABV-G抗体滴度分别为645080、705035,3μg组免疫小鼠RABV-G抗体滴度分别为256000、516064,1μg组免疫小鼠RABV-G抗体滴度分别为128000、409600。
真病毒中和实验检测该核酸疫苗对宿主的预防性保护作用的结果如图2C和图2D所示。如图2C所示,第1次给药后第14天、第21天10μg组免疫小鼠血清中和抗体滴度分别为5.5、13.7IU/ml,3μg组免疫小鼠血清中和抗体滴度分别为7.1、17.3IU/ml,1μg组免疫小鼠血清中和抗体滴度分别为4.2、8.6 IU/ml。如图2D所示,在21天接受加强注射,加强免疫后的第7天、14天,10μg组免疫小鼠血清中和抗体滴度分别为1018.5、1166IU/ml,3μg组免疫小鼠血清中和抗体滴度分别为966.3、1255IU/ml,1μg组免疫小鼠血清中和抗体滴度分别为557.9、1001.7IU/ml。
该实验结果显示,无论是1次给药、还是接种加强针后,10μg、3μg、1μg的接种剂量诱导的血清抗体完全可以中和病毒感染的细胞,1针接种后中和抗体滴度为WHO标准(0.5IU/mL)的16~30倍,加强后为WHO标准(0.5IU/mL)的1000~2000倍,并且其结合抗体与中和抗体的趋势一致。
以上结果证明了本申请的狂犬病mRNA疫苗能够以低剂量(微克级别)、少次数(2次接种,或甚至单次接种),实现极显著的中和保护效果。
实施例3.RABV-G mRNA疫苗诱导小鼠的细胞免疫应答
T细胞虽不能阻止病毒感染宿主细胞,但T细胞对机体中狂犬病病毒的清除有重要作用。本实施例按上述实验方法III通过胞内细胞因子染色法(ICS)分析RABV-G抗原特异性T细胞。分别在加强注射后的第7天取3μg、1μg接种剂量组及未包裹核酸的脂质体纳米颗粒组(即阴性对照组的小鼠),分离其脾脏细胞。
结果如图3A、3B和3C所示,3μg RABV-G mRNA疫苗组的RABV-G特异性IFN-γ阳性CD8 +T细胞明显高于1μg接种剂量组,而1μg接种剂量组与阴性对照组的CD8 +T细胞比例相当(3μg RABV-G mRNA疫苗组IFN-γ阳性CD8 +T平均占比为0.33%,1μg接种剂量组平均占比为0.14%,阴性对照组IFN-γ阳性CD4 +T细胞平均占比为0.12%)(图3A)。相比之下,RABV-G特异性IFN-γ阳性CD4 +T细胞3μg接种剂量组和1μg接种剂量组均明显高于阴性对照组(3μg剂量组IFN-γ阳性CD4 +T细胞平均占比0.60%,1μg剂量组IFN-γ阳性CD4 +T细胞平均占比0.34%,阴性对照组IFN-γ阳性CD4 +T细胞平均占比为0.2%)(图3B)。另外,RABV-G特异性IL-4阳性CD4 +T细胞,3μg,1μg接种剂量组占比相当,且都显著高于阴性对照组(RABV-G特异性IL-4阳性CD4 +T细胞接种3μg剂量组平均占比0.16%,1μg接种剂量组平均占比0.16%, 阴性对照组平均占比0.05%)(图3C)。
该结果证明了RABV-G mRNA疫苗能够有效诱导RABV-G特异性CD4 +和/或CD8 +T细胞,并且该随着剂量增加,效果可有进一步的提高。
实施例4.单剂低剂量RABV-G mRNA疫苗的小鼠攻毒保护效果
为进一步评估本发明RABV-G mRNA核酸疫苗的体内保护效果,本实施例以BALB/c小鼠为模型,选取1μg低剂量组单次肌肉注射,阴性对照为等体积的未包裹核酸的脂质纳米颗粒。每组8只小鼠,每只注射体积为100μl。初次免疫后第4周通过肌肉注射,进行CVS-11固定毒攻毒,攻毒剂量为20LD50,50μl/只。将小鼠放入IVC笼中,连续14天每天称量小鼠体重,观察小鼠存活情况及生存状态。
结果如图4A和4B所示,1μg RABV-G mRNA疫苗接种组,小鼠在感染后的前2天体重下降了3~7%,从第3天开始体重逐渐恢复,第7天开始体重恢复到感染前水平,第10天开始体重逐渐增加(图4A);并且,1μg的RABV-G mRNA疫苗可完全保护小鼠免受致死性剂量的CVS-11病毒的攻击,小鼠能可持续生存2周以上(图4B)。而与疫苗接种组相反,未包裹核酸的脂质纳米颗粒组小鼠在感染后第4天开始体重骤降(图4A),感染后第7天开始死亡,第8天死亡率达到60%,第9天全部死亡(图4B)。
以上结果证明了本申请的狂犬病mRNA疫苗即便在低剂量下,也可实现优异的攻毒保护效果。
综合前述实施例可看出,本申请中所提供的RABV-G mRNA疫苗,在低剂量下接种就可形成强效的免疫保护,既可诱导出高滴度的中和抗体,也可激活T细胞免疫应答,在狂犬病病毒预防和治疗领域具有广阔的应用前景。
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申 请所附权利要求书所限定的范围。
附:序列表对应信息
SEQ ID NO 序列信息
1 RABV-G免疫原的DNA序列
2 5′-UTR
3 3′-UTR序列(来源于人α球蛋白和真核生物线粒体的串联3′UTR序列)
4 3′-多聚腺苷酸尾序列(250A)
5 修饰后的RABV-G mRNA序列(密码子优化+尿嘧啶替换)
6 制备RABV-G mRNA的DNA模板序列

Claims (15)

  1. 一种狂犬病免疫原核酸表达载体,其从5′端至3′端依次包含:
    (a)5′-UTR元件;
    (b)编码狂犬病病毒糖蛋白(RABV-G)的开放阅读框元件;
    (c)3′-UTR元件;
    (d)总长度为120nt以上的多聚腺苷酸尾元件,其包含:
    多个腺苷酸串,每个腺苷酸串各自独立地包含n个连续的腺苷酸,n为10~80之间的整数,且所述多个腺苷酸串的腺苷酸总个数为100个以上;
    位于所述多个腺苷酸串之间的连接子,所述连接子各自独立地不包含腺苷酸或仅包含1个或2个腺苷酸。
  2. 如权利要求1所述的核酸表达载体,其中,所述5′-UTR元件的长度为10~200nt,例如15~100nt;和/或
    所述5′-UTR元件源自下组中的一种或多种5′-UTR:人α-球蛋白、β-球蛋白、核糖体蛋白(RP)、微管蛋白β-2B、补体因子3(C3)、细胞色素P4502E1(CYP2E1)、载脂蛋白A-II(APOA2)、人类血红蛋白亚基β(hHBB)、血红蛋白A1(HBA1)、血红蛋白A2(HBA2)、登革热病毒(DENV);和/或
    所述5′-UTR元件具有如SEQ ID NO:2所示的序列或与其具有至少80%的序列同一性。
  3. 如权利要求1所述的核酸表达载体,其中,所述3′-UTR元件为来源于哺乳动物或病毒的3′-UTR,例如来源选自下组的3′-UTR或其组合(如串联序列):人α球蛋白、人β球蛋白、人白蛋白、人肌动蛋白、人血红蛋白亚基α1(HBA1)、细胞色素B-245α链(CYBA)、真核生物线粒体(Mit)的序列、SARAS-Cov-2、登革热病毒(DENV)、萝卜皱病毒(TCV)、烟草花叶病毒(TMV)和烟草蚀刻病毒(TEV);和/或
    所述3′-UTR元件包含选自下组一个或多个3′-UTR分子:α球蛋白3′-UTR、真核生物线粒体3′-UTR、白蛋白3′-UTR、β球蛋白3′-UTR或其任意串联序列,优选α球蛋白3′-UTR、真核生物线粒体3′-UTR、或它们串联形成的3′-UTR; 和/或
    所述3′-UTR具有如SEQ ID NO:3所示的序列,或与其具有至少80%的序列同一性的序列。
  4. 如权利要求1所述的核酸表达载体,其中,所述多聚腺苷酸尾元件的总长度为120~400nt,例如120~350nt,120~320nt,或其中的任意整数,例如120、304nt;和/或
    各腺苷酸串各自独立地包含10~80个、20~70个、25~60个、30~50个或其中任意整数个连续的腺苷酸,如20个、30个、33个、35个、36个、37个、38个、39个、40个、45个、50个、55个、60个、65个、70个连续腺苷酸;和/或
    所述多聚腺苷酸尾元件还进一步包含位于所述元件一个或两个端部的连接子;和/或
    所述连接子的长度各自独立地为3~15nt,例如3、4、5、6、7、8、9、10、11、12、13、14或15nt;和/或
    例如,所述连接子的序列各自独立地选自:GCTATGACT、GTATGT、GCAAGT、GATTGC、GGCTGC、TACTGC、GGCTTC;和/或
    所述多聚腺苷酸尾元件具有SEQ ID NO:4的序列,或与其中任何一者具有至少80%的序列同一性。
  5. 如权利要求1所述的核酸表达载体,其中,
    所述开放阅读框元件为单顺反子、双顺反子或多顺反子mRNA;和/或
    所述开放阅读框元件编码一个或多个来源相同或不同的RABV-G;和/或
    所述RABV-G来源于选自下组的狂犬病病毒:巴斯德毒株中的Pitman-Moore(PM)株、Pasteur(PV)株、CTN株、aG株、Flury-LEP株、Evelyn-Rokitnicki-Abelseth(ERA)株、Street-Alabama-Duffering(SAD)株、KHUV(Khujand lyssavirus)、BBLV(Bokeloh bat lyssavirus)、ARAV(Aravan lyssavirus)、EBLV-1(European bat 1 lyssavirus)、EBLV-2(European bat 2 lyssavirus)、IRKV(Irkutlyssavirus)、LBV(Lagos bat lyssavirus)、SHIBV(Shimoni bat lyssavirus)、MOKV(Mokola lyssavirus)、WCBV(West Caucasian bat lyssavirus)、 IKOV(Ikomalyssavirus)、DUVV(Duvenhagelyssavirus)、ABLV(Australian bat lyssavirus)、GBLV(Gannoruwa bat lyssavirus)或LLEBV(Lleida bat lyssavirus);和/或
    所述RABV-G包括选自下组的一种或多种分子:未经修饰或经真核修饰的RABV-G、其免疫原性片段或变体,所述分子能够诱导针对RABV的免疫中和和保护反应。
  6. 如权利要求1所述的核酸表达载体,其中,编码RABV-G的元件经过或未经密码子优化、包含或不包含碱基修饰和/或核苷类似物;和/或
    例如所述编码RABV-G的元件中的一个或多个尿嘧啶被选自下组中的一个或多个相同或不同的修饰碱基或核苷类似物置换:假尿苷、1-甲基尿嘧啶核苷、N1-乙基假尿苷、2-硫尿苷、4′-硫尿苷、5-甲基胞嘧啶、5-甲基尿苷、2-硫基-1-甲基-1-去氮杂-假尿苷、2-硫基T-甲基-假尿苷、2-硫基-5-氮杂-尿苷、2-硫基-二氢假尿苷、2-硫基-二氢尿苷、2-硫基-假尿苷、4-甲氧基-2-硫基-假尿苷、4-甲氧基-假尿苷、4-硫基-1-甲基-假尿苷、4-硫基-假尿苷、5-氮杂-尿苷、二氢假尿苷或5-甲氧基尿苷和2′-O-甲基尿苷,优选假尿苷或N1-甲基假尿苷或N1-乙基假尿苷,进一步优选为N1-甲基假尿苷。
  7. 如权利要求1所述的核酸表达载体,其中,所述表达载体还包括选自下组的一种或多种元件:
    5′-帽元件,其可选被修饰,例如所述5′-帽元件选自:m7GpppXpYp、m7GpppXmpYp、m7GpppXmpYmp、或其甲基化修饰序列、反向结合异构体、抗-反转帽类似物(ARCA)、N7-苄基二核苷四磷酸帽类似物;
    启动子元件,例如T7启动子、sp6启动子或T3启动子;
    信号肽编码元件,例如指导目的蛋白亚细胞定位的信号肽(如跨膜信号肽、分泌信号肽、核定位信号肽)编码元件;
    酶切位点,例如XbaI、EcoRV、BamHI、XhoI;
    标签,例如用于目的分子鉴定、分离或纯化的分子标签,如Flag标签、HA标签。
  8. 如权利要求1所述的核酸表达载体,其中,从5′端到3′端包含:
    包含SEQ ID NO:2所示序列的5′-UTR元件;编码RABV-G的开放阅读框元件;包含如SEQ ID NO:3所示序列的3′-UTR元件;包含如SEQ ID NO:4所示序列的多聚腺苷酸尾元件;或与所述各序列各自具有至少80%序列同一性的序列的元件;和/或
    所述核酸表达载体包含如SEQ ID NO:5或6所示的序列、或与其具有至少80%序列同一性的序列。
  9. 如权利要求1所述的核酸表达载体,其单独包含于包装物中,或与递送系统中的运载体组合,例如,所述递送系统选自:脂质递送系统、类脂递送系统、聚合物递送系统或其组合递送系统,例如加载于脂质纳米颗粒、聚氨酯(PAA)、聚β氨基酯(PBAE)、聚乙烯亚胺(PEI)、脂质包裹的聚合物胶束。
  10. 如权利要求1所述的核酸表达载体,其中,核酸表达载体与脂质体纳米颗粒组合,例如:
    所述脂质体纳米颗粒包括:阳离子脂质、结构脂质、辅助脂质和稳定脂质的组合;和/或
    所述的阳离子脂质体与RABV-G mRNA质量比为10~30∶1,优选为15~20∶1。
  11. 一种狂犬病mRNA疫苗,其包含:如权利要求1~10中任一项所述的狂犬病免疫原核酸表达载体,以及包装物和/或用于该核酸表达载体的递送系统和/或药学上或生理学上可接受的运载体。
  12. 如权利要求11所述的mRNA疫苗,其中,所述疫苗的形式适于选自下组的一种或多种给予或递送方式:呼吸道雾化吸入、滴鼻、口服、直接注射(例如静脉注射、皮下注射、皮内注射、肌肉注射)、黏膜给药;和/或
    所述疫苗还包含佐剂或与佐剂联合使用,例如所述佐剂选自:铝佐剂、霍乱毒素及其亚单位、寡脱氧核苷酸、锰离子佐剂、胶体锰佐剂、弗氏佐剂、MF59佐剂、QS-21佐剂、Poly I:C及其他TLR配体、GM-CSF、IL-2、IL-3、IL-7、IL-11、IL-12、IL-18、IL-21;和/或
    所述疫苗的形式适于进行2种或以上药物或疫苗的组合给予,例如联合接种或序贯接种。
  13. 如权利要求1~10中任一项所述的核酸表达载体和/或如权利要求11~12中任一项所述的疫苗在制备用于狂犬病预防和/或治疗的产品中的应用。
  14. 制备如权利要求1~10中任一项所述的核酸表达载体或如权利要求11~12中任一项所述的疫苗的方法,所述方法包括:提供独立或连接的各元件;将各元件组装成核酸表达载体。
  15. 如权利要求14所述的方法,其中,所述方法包括采用选自下组的一种或多种材料:DNA模板(例如PCR产物或已线性化的质粒DNA)、核酸酶、聚合酶、加帽酶、聚腺苷酸合酶、DNA酶、一个或多个元件分子、接头分子、天然或修饰的核酸分子、缓冲液、溶剂;和/或
    所述方法还包括选自下组的一个或多个步骤:对各元件进行设计、优化、改造和/或修饰;对中间产物和/或最终产物进行分离、纯化、鉴定、定量、包装和/或活性测试;将核酸表达载体与用于该核酸表达载体的递送系统和/或药学上或生理学上可接受的运载体组合。
PCT/CN2022/119230 2022-09-16 2022-09-16 一种狂犬病mRNA疫苗、其制备及应用 WO2024055273A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/CN2022/119230 WO2024055273A1 (zh) 2022-09-16 2022-09-16 一种狂犬病mRNA疫苗、其制备及应用

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2022/119230 WO2024055273A1 (zh) 2022-09-16 2022-09-16 一种狂犬病mRNA疫苗、其制备及应用

Publications (1)

Publication Number Publication Date
WO2024055273A1 true WO2024055273A1 (zh) 2024-03-21

Family

ID=90273915

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/119230 WO2024055273A1 (zh) 2022-09-16 2022-09-16 一种狂犬病mRNA疫苗、其制备及应用

Country Status (1)

Country Link
WO (1) WO2024055273A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH08269092A (ja) * 1995-03-31 1996-10-15 Chemo Sero Therapeut Res Inst 大腸菌組換え狂犬病ワクチン
CN105517569A (zh) * 2013-08-21 2016-04-20 库瑞瓦格股份公司 狂犬病疫苗
CN110714015A (zh) * 2019-10-29 2020-01-21 珠海丽凡达生物技术有限公司 一种mRNA狂犬病疫苗
CN114729373A (zh) * 2022-01-27 2022-07-08 深圳市瑞吉生物科技有限公司 一种新型冠状病毒mRNA疫苗及其制备方法与应用
CN114921481A (zh) * 2022-02-25 2022-08-19 上海赛伦生物技术股份有限公司 一种狂犬病病毒修饰性mRNA疫苗及其制备方法

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH08269092A (ja) * 1995-03-31 1996-10-15 Chemo Sero Therapeut Res Inst 大腸菌組換え狂犬病ワクチン
CN105517569A (zh) * 2013-08-21 2016-04-20 库瑞瓦格股份公司 狂犬病疫苗
CN110714015A (zh) * 2019-10-29 2020-01-21 珠海丽凡达生物技术有限公司 一种mRNA狂犬病疫苗
CN114729373A (zh) * 2022-01-27 2022-07-08 深圳市瑞吉生物科技有限公司 一种新型冠状病毒mRNA疫苗及其制备方法与应用
CN114921481A (zh) * 2022-02-25 2022-08-19 上海赛伦生物技术股份有限公司 一种狂犬病病毒修饰性mRNA疫苗及其制备方法

Similar Documents

Publication Publication Date Title
WO2021254327A1 (zh) 一种包膜替换型病毒载体疫苗及其构建方法
CA2817005C (en) Rabies glycoprotein virus-like particles (vlps)
CN112076315A (zh) 新冠病毒s蛋白和铁蛋白亚基融合的纳米抗原颗粒、新冠疫苗及其制备方法和应用
CN105934441A (zh) 新型sars免疫原性组合物
US9732122B2 (en) Attenuated recombinant vesicular stomatitis viruses comprising modified matrix proteins
US11931410B1 (en) SARS-CoV-2 mRNA vaccine and preparation method and use thereof
WO2023051701A1 (zh) 抗SARS-CoV-2感染的mRNA、蛋白以及抗SARS-CoV-2感染的疫苗
US20230174588A1 (en) A vaccine against sars-cov-2 and preparation thereof
US9060972B2 (en) Recombinant hemagglutinin protein of influenza virus and vaccine containing the same
CN116036259B (zh) 猴痘病毒多抗原mRNA疫苗及其应用
CN116410992A (zh) 预防和/或治疗新型冠状病毒的mRNA、疫苗及其制备方法和应用
WO2024055273A1 (zh) 一种狂犬病mRNA疫苗、其制备及应用
CN116024237A (zh) 一种用于预防裂谷热的mRNA疫苗及其制备方法
US11111275B2 (en) Compositions and methods for making and using virus-like particles (VLPs)
CN115322247A (zh) 一种新型冠状病毒受体结合区电荷突变体抗原及应用
CN117721150A (zh) 一种狂犬病mRNA疫苗、其制备及应用
CN117205309B (zh) 一种流感免疫原组合物和制备方法及其用途
WO2023202711A1 (zh) 一种基于新型冠状病毒的mRNA疫苗
WO2013011942A1 (ja) 変異狂犬病ウイルス及びワクチン
CN116904489B (zh) 一种鸭坦布苏病毒核酸疫苗及应用
US20230338512A1 (en) Coronavirus vaccine
WO2023142283A1 (zh) 一种新型冠状病毒mRNA疫苗及其制备方法与应用
CN112316127B (zh) 基于自组装铁蛋白纳米抗原颗粒以及由其制备的猪圆环疫苗和应用
MXPA01010481A (es) Acidos nucleicos y polipeptidos de lisavirus quimerico.
CN117838848A (zh) 一种口蹄疫二价mRNA疫苗及其制备方法和应用

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: 22958464

Country of ref document: EP

Kind code of ref document: A1