WO2024078597A1 - Rsv f protein variants and uses thereof - Google Patents

Rsv f protein variants and uses thereof Download PDF

Info

Publication number
WO2024078597A1
WO2024078597A1 PCT/CN2023/124358 CN2023124358W WO2024078597A1 WO 2024078597 A1 WO2024078597 A1 WO 2024078597A1 CN 2023124358 W CN2023124358 W CN 2023124358W WO 2024078597 A1 WO2024078597 A1 WO 2024078597A1
Authority
WO
WIPO (PCT)
Prior art keywords
polynucleotide
rsv
protein
sequence
polypeptide
Prior art date
Application number
PCT/CN2023/124358
Other languages
French (fr)
Inventor
Jason Jingxin ZHANG
Yue Zhou
Kunlun XIANG
Original Assignee
Rvac Medicines (Us) , Inc.
Shanghai Rvac Biotech Co., Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Rvac Medicines (Us) , Inc., Shanghai Rvac Biotech Co., Ltd. filed Critical Rvac Medicines (Us) , Inc.
Publication of WO2024078597A1 publication Critical patent/WO2024078597A1/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18522New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18534Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the present disclosure relates to variants of RSV F protein, and polynucleotides, vectors, compositions, and kits of these variants.
  • the present disclosure also provides methods for generating an immune response to RSV F protein in a subject and methods for preventing RSV infection in a subject.
  • Respiratory syncytial virus is a common, contagious virus that causes infections of the lungs and the respiratory tract.
  • RSV infection is the leading cause of respiratory hospitalization in infants, and reinfection remains common in later life. It is an important pathogen in all age groups.
  • RSV causes a respiratory tract infection that affects 64 million people per year worldwide. It hospitalizes 3 million children under 5 years old and approximately 336,000 older adults annually.
  • the F glycoprotein plays a pivotal role in the pathogenesis of RSV by mediating the fusion between the viral and the host cell membrane.
  • the F protein presents two different conformations, a lollipop-shaped prefusion form (preF) , present on the virus surface before virus–cell interaction, and a crutch-shaped postfusion form (postF) which is acquired following the fusion between the virus and cell membrane or by unknown mechanisms that spontaneously initiate the rearrangement from the highly metastable preF conformation into the energetically favorable postF conformation (Mejias et al, 2017) .
  • the two forms are antigenically distinct.
  • the preF form has been shown to induce the majority of highly neutralizing antibodies following natural infection or immunization.
  • the two most immunogenic sites on the F protein are only present on the preF conformation, while less immunogenic sites (Site I, Site II, Site III, and Site IV) are shared between the preF and postF forms.
  • F glycoprotein is highly conserved among RSV subtypes, with amino acid sequence identities of 90%or higher. This relative sequence conservation of the F protein combined with its surface location on the virion, its obligatory role in viral entry, and its antigenic sites associated with potent neutralization make the F protein, particularly the preF, an ideal target for neutralizing antibodies and a promising vaccine antigen.
  • Vaccines work by presenting the immune system in a subject with key bits of a pathogen, which induce the production of antibodies and immune cells that can recognize and fight the whole pathogen when the subject is re-exposed to it.
  • the present disclosure provides multiple engineered RSV prefusion F protein variants as well as codon-optimized mRNAs encoding them, which can be useful in vaccine designs.
  • the present disclosure also provides vectors, compositions, kits, and methods related to these engineered preF variants.
  • the present disclosure provides a polypeptide comprising an amino acid sequence of any one of SEQ ID NO: 26-28, 30-50.
  • the present disclosure provides a polypeptide comprising an amino acid sequence with at least 90%identity to any one of SEQ ID NO: 26-28, 30-50.
  • the present disclosure provides a polynucleotide comprising a coding sequence that encodes the polypeptide disclosed herein.
  • the present disclosure provides a polynucleotide comprising a sequence of any one of SEQ ID NOs: 1-25.
  • the polynucleotide is an mRNA sequence.
  • the coding sequence is operably linked to a 5’ untranslated region (5’ UTR) and/or a 3’ untranslated region (3’ UTR) .
  • the polynucleotide disclosed herein further comprises a 5’ cap structure.
  • the polynucleotide disclosed herein further comprises a poly (A) tail at its 3’ end.
  • the polynucleotide disclosed herein comprises at least one modified nucleotide.
  • the polynucleotide disclosed herein comprises from the 5’-end to the 3’-end: a 5’-cap structure, a 5’ UTR, the coding sequence, a 3’ UTR, and a poly (A) sequence.
  • the present disclosure provides a vector comprising the polynucleotide disclosed herein.
  • the vector is a viral vector or a plasmid.
  • the present disclosure provides a composition comprising the polypeptide disclosed herein.
  • the present disclosure provides a composition comprising the polynucleotide disclosed herein.
  • the composition further comprising a carrier selected from the group consisting of lipid nanoparticles, liposomes, cationic nanoemulsions, dendrimer-based lipid nanoparticles, cationic polymers, and polysaccharide particles.
  • a carrier selected from the group consisting of lipid nanoparticles, liposomes, cationic nanoemulsions, dendrimer-based lipid nanoparticles, cationic polymers, and polysaccharide particles.
  • the present disclosure provides a kit comprising the polynucleotide disclosed herein and a carrier selected from the group consisting of lipid nanoparticles, liposomes, cationic nanoemulsions, dendrimer-based lipid nanoparticles, cationic polymers, and polysaccharide particles.
  • the present disclosure provides a method for generating an immune response to RSV F protein in a subject in need thereof, comprising administering to the subject an effective amount of the polypeptide disclosed herein, or the polynucleotide disclosed herein.
  • the present disclosure provides a method for preventing RSV infection in a subject in need thereof, comprising administering to the subject an effective amount of the polypeptide disclosed herein, or the polynucleotide disclosed herein.
  • the present disclosure provides a method for generating an immune response to RSV F protein in a subject in need thereof, comprising administering an effective amount of the composition disclosed herein.
  • the present disclosure provides a method for preventing RSV infection in a subject in need thereof, comprising administering an effective amount of the composition disclosed herein.
  • the RSV is RSV A or RSV B.
  • Fig. 1 shows the results of viral neutralizing titration (PRNT60) against RSV/A2 on day 56, as described in Example 2.
  • Fig. 2 shows the results of viral neutralizing titration (PRNT60) against RSV B on day 56, as described in Example 2.
  • Fig. 3 shows the titration result of RSV lung viral load on day 61, as described in Example 2.
  • Fig. 4 shows the titration result of RSV nose viral load on day 61, as described in Example 2.
  • Fig. 5 shows the results of viral neutralizing titration (PRNT60) against RSV/A2 on day 35, as described in Example 3.
  • Fig. 6 shows the results of viral neutralizing titration (PRNT60) against RSV B on day 35, as described in Example 3.
  • Fig. 7 shows titration result of RSV lung viral load on day 61, as described in Example 3.
  • Fig. 8 shows titration result of RSV nose viral load on day 61, as described in Example 3.
  • nucleic acids are written left to right in the 5' to 3' orientation; and amino acid sequences are written left to right in amino to carboxy orientation, respectively.
  • percent identity and “%identity, ” as applied to nucleic acid or polynucleotide sequences, refer to the percentage of residue matches between at least two nucleic acid or polynucleotide sequences aligned using a standardized algorithm. Such an algorithm may insert, in a standardized and reproducible way, gaps in the sequences being compared in order to optimize alignment between two sequences, and therefore achieve a more meaningful comparison of the two sequences.
  • Percent identity between nucleic acid or polynucleotide sequences may be determined using a suite of commonly used and freely available sequence comparison algorithms provided by the National Center for Biotechnology Information (NCBI) Basic Local Alignment Search Tool (BLAST) (Altschul, S. F. et al. (1990) J. Mol. Biol. 215: 403-410) , which is available from several sources, including the NCBI, Bethesda, Md., and on the Internet at http: //www. ncbi. nlm. nih. gov/BLAST/.
  • NCBI National Center for Biotechnology Information
  • BLAST Basic Local Alignment Search Tool
  • Nucleic acid or polynucleotide sequences that do not show a high degree of identity may nevertheless encode similar amino acid sequences due to the degeneracy of the genetic code. It is understood that changes in a nucleic acid sequence can be made using this degeneracy to produce multiple nucleic acid sequences that all encode substantially the same protein. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al. (1991) Nucleic Acid Res 19: 5081; Ohtsuka et al. (1985) J Biol Chem 260: 2605-2608; Rossolini et al. (1994) Mol Cell Probes 8: 91-98) .
  • nucleic acid refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single-or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides which have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides.
  • nucleic acid is used interchangeably with polynucleotide, and (in appropriate contexts) gene, cDNA, and mRNA encoded by a gene.
  • percent (%) amino acid sequence identity with respect to a peptide, polypeptide or protein sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in another peptide or polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Percent amino acid sequence identity in the current disclosure is measured using BLAST software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • amino acid substitution refers to the replacement of one amino acid in a polypeptide with another amino acid. Exemplary substitutions are shown in Table 1. Amino acid substitutions may be introduced into a protein of interest and the products screened for a desired activity, for example, retained/improved biological activity.
  • Amino acids may be grouped according to common side-chain properties:
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • corresponding to with reference to nucleotide or amino acid positions of a sequence, such as set forth in the Sequence Listing, refers to nucleotides or amino acid positions identified upon alignment with a target sequence based on structural sequence alignment or using a standard alignment algorithm, such as the GAP algorithm.
  • corresponding residues of a similar sequence e.g., a fragment or species variant
  • structural alignment methods By aligning the sequences, one skilled in the art can identify corresponding residues, for example, using conserved and identical amino acid residues as guides.
  • composition refers to any mixture of two or more products, substances, or compounds, including cells.
  • an “effective amount” refers to an amount of a pharmaceutical composition which is sufficient to significantly and positively modify the symptoms and/or conditions to be treated (e.g., provide a positive clinical response) .
  • the effective amount of a pharmaceutical composition will vary with the particular condition being treated, the severity of the condition, the duration of treatment, the nature of concurrent therapy, the particular composition being employed, the particular pharmaceutically-acceptable excipient (s) and/or carrier (s) utilized, and like factors with the knowledge and expertise of the attending physician.
  • the terms “individual” and “subject” are used interchangeably herein to refer to an animal.
  • the animal is a mammal.
  • the animals are humans, rodents, simians, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, or mammalian pets.
  • the animal can be male or female and can be at any suitable age, including infant, juvenile, adolescent, adult, and geriatric.
  • an “individual” or “subject” refers to an animal in need of treatment for a disease or disorder.
  • the animal to receive the treatment can be a “patient, ” designating the fact that the animal has been identified as having a disorder of relevance to the treatment, or being at adequate risk of contracting the disorder.
  • the animal is a human, such as a human patient.
  • coding sequence and “coding region” are used interchangeably. They refer to the portion of a polynucleotide that codes for a protein.
  • engineered in the context of protein and protein variants means that the protein or protein variant is not naturally occurring.
  • Respiratory syncytial virus is a common, contagious virus that causes infections of the lungs and the respiratory tract. RSV infection is the leading cause of respiratory hospitalization in infants, and reinfection remains common in later life. It is an important pathogen in all age groups.
  • RSV is a negative-sense, single-stranded RNA virus. Its genome has 10 genes which encode 11 proteins, including 9 structural proteins (3 glycoproteins and 6 internal proteins) and 2 non-structural proteins.
  • the structural proteins include 3 transmembrane surface glycoproteins: the attachment protein G, fusion protein F, and the small hydrophobic SH protein.
  • F and G glycoproteins are the two major surface proteins that control viral attachment and the initial stages of infection. F and G proteins are also the primary targets for neutralizing antibodies during natural infection.
  • the F glycoprotein plays a pivotal role in the pathogenesis of RSV by mediating the fusion between the viral and the host cell membrane.
  • the F gene encodes a type I integral membrane protein that is synthesized as a 574 amino acid inactive precursor, F 0 .
  • Three F 0 monomers assemble into a trimer and, as the trimer passes through the Golgi, the monomers are activated by a furin-like host protease (Bolt et al. 2000; Collins and Mottet 1991) .
  • the protease cleaves twice, after amino acids 109 and 136 (González-Reyes et al. 2001; Zimmer et al. 2001a) , generating three polypeptides.
  • the N-terminal and C-terminal cleavage products are the F 2 and F 1 subunits, respectively, which are covalently linked to each other by two disulfide bonds (Gruber and Levine 1983; Day et al. 2006) .
  • the intervening 27 amino acid peptide dissociates after cleavage (Begona Ruiz-Arguello et al. 2002) .
  • the F protein presents two different conformations, a lollipop-shaped prefusion form (preF) , present on the virus surface before virus–cell interaction, and a crutch-shaped postfusion form (postF) which is acquired following the fusion between the virus and cell membrane or by unknown mechanisms that spontaneously initiate the rearrangement from the highly metastable preF conformation into the energetically favorable postF conformation (Mejias et al, 2017) .
  • the two forms are antigenically distinct.
  • the preF form has been shown to induce the majority of highly neutralizing antibodies following natural infection or immunization.
  • the two most immunogenic sites on the F protein, named Site and Site V are only present on the preF conformation, while less immunogenic sites (Site I, Site II, Site III, and Site IV) are shared between the preF and postF forms.
  • RSV A and RSV B There are two subtypes of RSV, RSV A and RSV B. They differ primarily in the G glycoprotein, while the F glycoprotein is highly conserved among RSV subtypes, with amino acid sequence identities of 90%or higher. Much of the variability in the F protein (about 25%) is found within an antigenic site at the apex of the prefusion trimer, which is composed of an ⁇ -helix from F1 (aa 196–210) and a strand from F2 (aa 62–69) and may be a site that determines subtype-specific immunity (McLellan et al. 2013) .
  • the present disclosure provides several engineered preF variants and polynucleotide encoding them, which can be useful in vaccine design.
  • the present disclosure also provides vectors, compositions, kits, and methods related to these engineered preF variants.
  • the present disclosure provides engineered RSV prefusion F protein variants such as a polypeptide comprising an amino acid sequence of any one of SEQ ID NO: 26-28, 30-50. (see Table 2)
  • the present disclosure also provides a polypeptide comprising an amino acid sequence that has at least 95%identity to any one of SEQ ID NO: 26-28, 30-50.
  • the polypeptide has an amino acid sequence that has at least 90%identity to any one of SEQ ID NOs: 26-28, 30-50. In some embodiments, the polypeptide has an amino acid sequence that has at least 95%, 96%, 97%, 98%, or 99%identity to any one of SEQ ID NOs: 26-28, 30-50.
  • the polypeptide further comprises a signal peptide at its N terminus or C terminus.
  • a signal peptide refers to the short peptide present at the N terminus or C terminus of the polypeptide when it is initially translated. Signal peptides are usually cleaved off from a protein by a signal peptidase during or immediately after insertion into a cell membrane. Signal peptides function to prompt a cell to translocate the protein, usually to the plasma membrane.
  • the present disclosure provides a polynucleotide comprising a coding sequence that encodes the polypeptide disclosed herein.
  • the polynucleotide is DNA.
  • the mRNA transcribed from the DNA comprises a sequence of any one of SEQ ID NOs: 1-25.
  • the present disclosure provides a polynucleotide comprising a sequence of any one of SEQ ID NOs: 1-25. (Table 3) .
  • the polynucleotide is RNA. In some embodiments, the polynucleotide is mRNA. In some embodiments, the mRNA sequences are codon-optimized for enhanced stability and immunogenicity.
  • the coding sequence is operably linked to a 5’ untranslated region (5’ UTR) and/or a 3’ untranslated region (3’ UTR) .
  • the 5’ UTR comprises a sequence of SEQ ID NO: 51 (Table 3) .
  • the 3’ UTR comprises a sequence of SEQ ID NO: 52 (Table 3) .
  • the untranslated region (UTR) is a regulatory region situated at the 5’ or 3’ end of a coding region.
  • 5’ UTR is directly upstream from the initiation codon of the coding region.
  • the 5’ UTR is further added with a 5’ cap structure.
  • 3’ UTR immediately follows the translation termination codon of the coding region.
  • the 3’ UTR often contains regions for post-transcriptional regulation, such as polyadenylation, localization, and stability of the mRNA.
  • the 3’ UTR is further connected with a poly (A) tail.
  • the mRNA further comprises a 5’ cap structure.
  • the 5’ cap structure is a specially altered nucleotide that “caps” the 5’ end of the mRNA.
  • a 5’ cap structure can regulate nuclear export of the RNA, prevent degradation by exonucleases, and promote translation.
  • the 5’ cap is formed by a derivative of a guanine nucleotide.
  • the 5’ cap is linked to the 5’ terminus via a 5’-5’ triphosphate linkage.
  • the 5’ cap is methylated, e.g., m7GpppN, wherein N is the 5’ terminal nucleotide of the nucleic acid carrying the 5’ cap.
  • the 5’ cap structure is selected from glyceryl, inverted deoxy abasic residue, 4’, 5’-methylene nucleotide, 1- (beta-D-erythrofuranosyl) nucleotide, 4’-thio nucleotide, carbocyclic nucleotide, 1, 5-anhydrohexitol nucleotide, L-nucleotides, alpha-nucleotide, modified base nucleotide, threo-pentofuranosyl nucleotide, acyclic 3’, 4’-seco nucleotide, acyclic 3, 4-dihydroxybutyl nucleotide, acyclic 3, 5 dihydroxypentyl nucleotide, 3’-3’-inverted nucleotide moiety, 3’-3’-inverted abasic moiety, 3’-2’-inverted nucleotide moiety, 3’-2’-inverted abasic residue, 4
  • the mRNA further comprises a poly (A) tail at its 3’ end.
  • the poly (A) tail is a long chain of adenine nucleotides that is added to the 3’ end of a mRNA molecule.
  • the length of the poly (A) tail is at least 80, 90, 100, 150, 200, 250, 300, 350, 400, 450 or 500 nucleotides.
  • the length of the poly (A) tail is adjusted to control the stability of the mRNA molecule disclosed herein.
  • the length of the poly (A) can influence the half-life of the mRNA molecule
  • the length of the poly (A) tail can be adjusted to modify the level of resistance of the mRNA to nucleases and thereby control the time course of protein expression.
  • the polynucleotide comprises at least one modified nucleotide.
  • the modified nucleotide is selected from the group consisting of pseudouridine ( ⁇ ) , 1-methylpseudouridine (m1 ⁇ ) , 5-methyluridine (m5U) , 2-thiouridine (s2U) , and 5-methylcytidine (m5C) .
  • the term “modified nucleotide” includes any synthetic nucleotide and nucleotide analogue, and any naturally existing nucleotide other than adenine, adenine, guanine, thymine, or uracil.
  • Exemplary nucleotide modification includes 2’-O-methylation (Nm) and conversion of uridine to pseudouridine ( ⁇ ) . Both Nm and ⁇ modifications have the potential to stabilize RNA folding domains. Methylation of 2’-OH sites endows a nucleotide with greater hydrophobicity, protects against nucleolytic attack and stabilizes helices, and thus can benefit inter-or intra-molecular interactions. Pseudouridine exerts a significant rigidifying influence on the sugar–phosphate backbone and enhances base stacking. In addition, ⁇ provides an additional donor site for hydrogen-bond formation, which can stabilize RNA–RNA or RNA–protein interactions (Baillieu et al., Nucleic Acids Res. 2009) .
  • the polynucleotide comprises from the 5’-end to the 3’-end: a 5’-cap structure, a 5’ UTR, the coding sequence, a 3’ UTR, and a poly (A) sequence.
  • the polynucleotide further encodes an RNA polymerase.
  • RNA polymerase A self-amplifying RNA (saRNA) encode not only an antigen of interest but also a viral RNA-dependent RNA polymerase to amplify the RNA in the cytoplasm of transfected cells, lead to significantly greater immune responses than conventional RNAs.
  • a polynucleotide encoding the polypeptide disclosed herein can be obtained by methods known in the art.
  • the present disclosure provides a vector comprising the polynucleotide disclosed herein.
  • the vector is a viral vector or a plasmid.
  • Viral vectors can include, but are not limited to, adenoviral vectors, lentiviral vectors, retroviral vectors, and adeno-associated viral vectors.
  • expression vectors contain selection markers such as ampicillin-resistance, hygromycin-resistance, tetracycline resistance, kanamycin resistance, or neomycin resistance to permit detection of those cells transformed with the desired DNA sequences.
  • Suitable vectors, promoter, and enhancer elements are known in the art; many are commercially available for generating subject recombinant constructs.
  • the vector is a polycistronic vector. Any methods known to those skilled in the art for the insertion of polynucleotide fragments into a vector can be used to construct the vector disclosed herein.
  • the polynucleotide disclosed herein can be operably linked to control sequences in the expression vector (s) to ensure the expression of the polypeptide disclosed herein.
  • control sequences may include, but are not limited to, leader or signal sequences, promoters (e.g., naturally associated or heterologous promoters) , ribosomal binding sites, enhancer or activator elements, translational start and termination sequences, and transcription start and termination sequences.
  • the present disclosure provides a composition comprising the polypeptide disclosed herein.
  • the present disclosure provides a composition comprising the polynucleotide disclosed herein.
  • composition includes, but is not limited to, a pharmaceutical composition.
  • a “pharmaceutical composition” refers to an active pharmaceutical agent formulated in pharmaceutically acceptable or physiologically acceptable solutions for administration to a cell or an animal, either alone, or in combination with one or more other modalities of therapy. It will also be understood that, if desired, the compositions of the disclosure may be administered in combination with other agents, such as, e.g., cytokines, growth factors, hormones, small molecules, chemotherapeutics, pro-drugs, drugs, antibodies, or other various pharmaceutically active agents. There is virtually no limit to other components that may also be included in the compositions, provided that the additional agents do not adversely affect the ability of the composition to deliver the intended therapy.
  • phrases “pharmaceutically acceptable” is used herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • compositions may also comprise a pharmaceutically acceptable carrier, diluent, or excipient.
  • pharmaceutically acceptable carrier, diluent, or excipient includes, without limitation, any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, surfactant, or emulsifier which has been approved as being acceptable for use in humans or domestic animals.
  • Exemplary pharmaceutically acceptable carriers include, but are not limited to, to sugars, such as lactose, glucose, and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; tragacanth; malt; gelatin; talc; cocoa butter; waxes; animal and vegetable fats; paraffins; silicones; bentonites; silicic acid; zinc oxide; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; esters, such as ethyl oleate, and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid;
  • the liquid pharmaceutical compositions may include one or more of the following: sterile diluents such as water for injection, saline solution, preferably physiological saline; Ringers solution; isotonic sodium chloride; fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium; polyethylene glycols; glycerin; propylene glycol or other solvents; antibacterial agents, such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates, or phosphates; and agents for the adjustment of tonicity, such as sodium chloride or dextrose.
  • sterile diluents such as water for injection, saline solution, preferably physiological saline
  • Ringers solution isotonic sodium chloride
  • fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspend
  • the parenteral preparation can be enclosed in ampoules, disposable syringes, or multiple dose vials made of glass or plastic.
  • An injectable pharmaceutical composition is preferably sterile.
  • the composition may be suitably developed for intravenous, intratumoral, oral, rectal, vaginal, parenteral, topical, pulmonary, intranasal, buccal, ophthalmic, or another route of administration.
  • the composition further comprises a carrier selected from the group consisting of lipid nanoparticle, liposome, cationic nanoemulsion, dendrimer nanoparticle, cationic polymer, and polysaccharide particle.
  • a carrier selected from the group consisting of lipid nanoparticle, liposome, cationic nanoemulsion, dendrimer nanoparticle, cationic polymer, and polysaccharide particle.
  • carrier refers to compounds or compositions that are used for delivery of the polypeptide or the polynucleotide into a subject.
  • the carrier enhances effectiveness and/or safety of the delivery.
  • the carrier is capable of delivering large nucleic acid sequences (e.g., nucleic acids of at least 1 kDa, 1.5 kDa, 2 kDa, 2.5 kDa, 5 kDa, 10 kDa, 12 kDa, 15 kDa, 20 kDa, 25 kDa, 30 kDa, or more) .
  • the nucleic acids can be formulated with one or more acceptable reagents, which provide a vehicle for delivering such nucleic acids to target cells.
  • Appropriate reagents are generally selected with regards to a number of factors, which include, among other things, the biological or chemical properties of the nucleic acids (e.g., charge) , the intended route of administration, the anticipated biological environment to which such nucleic acids will be exposed and the specific properties of the intended target cells.
  • factors include, among other things, the biological or chemical properties of the nucleic acids (e.g., charge) , the intended route of administration, the anticipated biological environment to which such nucleic acids will be exposed and the specific properties of the intended target cells.
  • Lipid nanoparticles are nanoparticles made of one or more types of lipids.
  • lipid nanoparticles comprise ionizable lipids, which are positively charged at low pH (enabling RNA complexation) and neutral at physiological pH (reducing potential toxic effects, as compared with positively charged lipids, such as liposomes) .
  • lipid nanoparticles are taken up by cells via endocytosis, and the ionizability of the lipids at low pH (likely) enables endosomal escape, which allows release of the cargo into the cytoplasm.
  • the lipid nanoparticles comprise cationic lipids, which have a head group with permanent positive charges.
  • lipid nanoparticles usually contain a helper lipid, for example, phospholipid, to promote cell binding, cholesterol to fill the gaps between the lipids, and a polyethylene glycol (PEG) to reduce opsonization by serum proteins and reticuloendothelial clearance.
  • helper lipid for example, phospholipid
  • PEG polyethylene glycol
  • the relative amounts of ionizable lipid, helper lipid, cholesterol and PEG can vary (See Hou et al., Nature Reviews Materials, 2021) .
  • Liposomes are spherical-shaped vesicles that is composed of one or more phospholipid bilayers. Liposomes are most often composed of phospholipids, especially phosphatidylcholine and cholesterol, but may also include other lipids, such as phosphatidylethanolamine, as long as they are compatible with lipid bilayer structure. The lipid bilayer of liposome can fuse with other bilayers such as the cell membrane, thus delivering the liposome contents. Generally, liposomes are definite as spherical vesicles with particle sizes ranging from 30 nm to several micrometers.
  • lipid bilayers surrounding aqueous units, where the polar head groups are oriented in the pathway of the interior and exterior aqueous phases.
  • self-aggregation of polar lipids is not limited to conventional bilayer structures which rely on molecular shape, temperature, and environmental and preparation conditions but may self-assemble into various types of colloidal particles (See Akbarzadeh, Nanoscale Res Lett., 2013) .
  • Cationic nanoemulsions are mainly composed of two parts: one is the cationic lipid DOTAP (1, 2-dioleoyl-sn-glycero-3-phosphocholine) that can be added to the oil phase to bind the mRNA electrostatically; the other is the emulsion adjuvant MF59 that is an oil-in-water emulsion consisting of squalene and surfactants.
  • CNEs are usually fabricated by the probe sonication method (Brito et al., A cationic nanoemulsion for the delivery of next-generation RNA vaccines, 2014) .
  • Dendrimer-based lipid nanoparticles are nanoparticles made of lipids and dendrimers, which are highly ordered, branched polymeric molecules. Dendrimers are composed of three distinct structural components: (1) the core, (2) the repetitive branching layers (also referred to as “generation” ) , and (3) the abundant terminal groups. These precisely controlled dendritic structures harbor multivalent cooperativity and can exploit membrane-fusion-based endosome release by mimicking lipid vectors, while simultaneously retaining the “proton-sponge” -mediated endosome release of polymer vectors (See Chen et al., Amphiphilic Dendrimer Vectors for RNA Delivery: State-of-the-Art and Future Perspective, 2022) .
  • Cationic polymer is another viable RNA carrier.
  • An exemplary cationic polymer is poly (ethyleneimine) and its derivatives
  • Polyethyleneimine (PEI) is among the earliest and most widely studied cationic polymers for gene delivery, including the delivery of RNA. It has high gene transfection efficiency and is often referred to as the gold standard for non-viral gene transfection (Lungwitz et al., 2005) .
  • PEI can be in either linear or branched structures and its positive charge is conferred by numerous amine groups separated by short alkyl spacers, which lead to very high positive charge density within its structure (Jiang et al., Polymeric nanoparticles for RNA delivery, 2021) .
  • Polysaccharides are a complex collection of biopolymers isolated from plant, animal, microbial and algal sources that are built from monosaccharides linked by O-glycosidic linkages.
  • An exemplary polysaccharide that can be used for RNA delivery is Chitosan, is a polysaccharide contained in the cell walls of fungi and in the shells of arthropods such as crustaceans and consists of a linear chain of 2-acetaylamino-2-deoxy- ⁇ -D-glucopyranose units connected through ⁇ -1, 4 linkages (Bodnar, Hartmann & Borbely, 2005; Barclay et al., Review of polysaccharide particle-based functional drug delivery, 2020) .
  • the present disclosure provides a kit comprising the polynucleotide disclosed herein, and a carrier selected from the group consisting of lipid nanoparticle, liposome, cationic nanoemulsion, dendrimer nanoparticle, cationic polymer, and polysaccharide particle.
  • the present disclosure provides a method for generating an immune response to RSV F protein in a subject in need thereof, comprising administering to the subject an effective amount of the polypeptide disclosed herein, or the polynucleotide disclosed herein.
  • An immune response may typically be a specific reaction of the adaptive immune system to a particular antigen (so called specific or adaptive immune response) or an unspecific reaction of the innate immune system (so called unspecific or innate immune response) , or a combination thereof.
  • specific or adaptive immune response an antigen of the innate immune system
  • innate immune response unspecific or innate immune response
  • it could be a humoral immune response and/or a cellular immune response.
  • Humoral immunity refers typically to antibody production and optionally to accessory processes accompanying antibody production.
  • a humoral immune response may typically be characterized, e.g., by Th2 activation and cytokine production, germinal center formation and isotype switching, affinity maturation and memory cell generation.
  • Humoral immunity also typically may refer to the effector functions of antibodies, which include pathogen and toxin neutralization, classical complement activation, and opsonin promotion of phagocytosis and pathogen elimination.
  • Cellular immunity typically relates to the activation of macrophages, natural killer cells (NK) , antigen-specific cytotoxic T-lymphocytes, and the release of various cytokines in response to an antigen.
  • cellular immunity is not based on antibodies, but on the activation of cells of the immune system.
  • a cellular immune response may be characterized, for example, by activating antigen-specific cytotoxic T-lymphocytes that are able to induce apoptosis in cells, e.g., specific immune cells like dendritic cells or other cells, displaying epitopes of foreign antigens on their surface.
  • Such cells may be virus-infected or infected with intracellular bacteria, or cancer cells displaying tumor antigens.
  • Further characteristics may be activation of macrophages and natural killer cells, enabling them to destroy pathogens, and stimulation of cells to secrete a variety of cytokines that influence the function of other cells involved in adaptive immune responses and innate immune responses.
  • the present disclosure provides a method for preventing RSV infection in a subject in need thereof, comprising administering to the subject an effective amount of the polypeptide disclosed herein, or the polynucleotide disclosed herein.
  • the method prevents RSV infection by generating an immune response to RSV F protein.
  • the method prevents RSV infection by generating an adaptive immune response to RSV F protein in the subject.
  • the present disclosure provides a method for generating an immune response to RSV F protein in a subject in need thereof, comprising administering an effective amount of the composition disclosed herein.
  • the immune response is an adaptive immune response.
  • the present disclosure provides a method for preventing RSV infection in a subject in need thereof, comprising administering an effective amount of the composition disclosed herein.
  • the method prevents RSV infection by generating an adaptive immune response to RSV F protein in the subject.
  • the RSV is RSV A or RSV B.
  • This series of Elisa assays was to confirm RSV prefusion protein expression by the mRNA disclosed herein.
  • the cell culture plates corning 3599 were coated with 1/100 collagen I and the Elisa plates corning 9018 were coated with 100 ⁇ l per well of RSV Pre-F protein at 2 ⁇ g/ml at 4 °C overnight.
  • the HEp2 cells were seeded in coated 96 well plates Corning 3599 with 100 ⁇ l per well cells at the density of 1.2E4 cells/ml. After 24 hours incubation, synthetic mRNA was in vitro transfected by using a TransIT-mRNA Transfection kit (Mirus) .
  • Corning 96 well, flat bottom, high binding plates were coated with 100 ⁇ l per well of RSV Pre-F protein at 2 ⁇ g/ml at 4 °C overnight. The plates were washed in PBS/0.05%Tween 20 and then blocked in PBS/0.05%Tween 20/3%non-fat milk at room temperature for 1 h. After washed with 300 ul PBS/0.05%Tween 20, 100 ⁇ l per well of 3-fold serially titrated mAbs (preF standard protein, 2ug/ml start, 3 fold dilution, 7 points, and the 8 th point was DPBS Mock) and mRNA transfection cell supernatants were then added and incubated at room temperature for 90 min.
  • mAbs preF standard protein, 2ug/ml start, 3 fold dilution, 7 points, and the 8 th point was DPBS Mock
  • D25 mAb dilution and ELISA plate coating Dilute the D25 mAb 4.2 mg/ml to 2 ⁇ g/ml with DPBS and mix gently. Dispense 100 ul 2ug/ml D25 mAb to the ELISA assay plate with 200 ul 12 channel pipette. Place the coated plates in a 4°C refrigerator overnight. Coating the cell culture plate with collagen type I Rat tail. Dilute the collagen type I Rat tail 100-fold with DPBS. Dispense 1/100 diluted collagen I to the 96 well plate with 100 ul per well. Place the coated plate to a 4°C refrigerator overnight.
  • HEp2 Cell seeding Remove HEp2 culture T150 flask from the incubator and check the cell density with a microscope, the cells should reach to 80%-85%confluent. Remove and discard the spent cell culture media from the culture flask. Wash cells using the DPBS solution without calcium and magnesium. Remove and discard the wash solution, add 2.5 ml pre-warmed dissociation TrypLE TM to the side of the flask. Incubate the culture flask at 37 °C cell culture incubator for 5 minutes. When ⁇ 90%of the cells have detached, tilt the vessel for a minimal length of time to allow the cells to drain. Add 10 ml of pre-warmed complete growth medium. Disperse the medium by pipetting over the cell layer surface several times.
  • ELISA plate blocking The coating buffer was removed by flicking the plate over a sink, and the remaining drops were removed by patting the plate on a paper towel.
  • the plates were washed in PBS/0.05%Tween 20 300 ul with three times, and then blocked in PBS/0.05%Tween 20/3%non-fat milk at room temperature for 1 h.
  • the plates were washed in PBS/0.05%Tween 20 300 ul three times, the blocking buffer were removed by flicking the plate over a sink, the remaining drops were removed by patting the plate on a paper towel.
  • the plates were sealed with Self-adhesive Sealing Film for microplate and stored at a 4 °Crefrigerator for future use.
  • TransIT-mRNA Reagent mRNA Boost: RNA complexes. Warm TransIT-mRNA and mRNA Boost reagents to room temperature and vortex gently before using.
  • RNA complex mixture drop-wise to different areas of the well. Gently rock plate for even distribution of complexes. Incubate for 4-48 hours. Harvest cells or supernatant and assay as required and storage in 4 °C refrigerator.
  • Tables 4-6 shows the results of ELISA essays, with 100 ng, 25 ng, 6.25 ng mRNA being transfected into the Hep-2 cells, respectively.
  • mRNAs disclosed herein were administered to cotton rats at 2.5 ⁇ g and 25 ⁇ g.
  • Female cotton rats (6-8 weeks of age) were divided into 12 groups of 5 animals. The cotton rats were immunized according to the schedule shown in Table 7 below.
  • Groups 1-10 were immunized intramuscularly (IM) with 100 ⁇ L dose of the mRNA-LNP composition per animal;
  • Group 11 was infected intranasally (IN) with 100 ⁇ L dose RSV/A2 virus at 10 5 plaque forming units (PFUs) per animal.
  • Groups 12 were immunized intramuscularly with 100 ⁇ L saline per animal.
  • the rats were challenged with an intranasal administration of 0.1 mL of 5.0 log10 RSV/A2.
  • the animals were sacrificed, the lung and nasal tissue was harvested for viral titration measurements.
  • Heat inactivated sera samples were diluted 1: 10 with EMEM and serially diluted further 1: 4. Diluted serum samples were incubated with RSV/A2 or RSV/B (25-50 PFU) for 1 hour at room temperature and inoculated in duplicates onto confluent HEp-2 monolayers in 24 well plates. After one hour incubation at 37 °C in a 5%CO2 incubator, the wells were overlayed with 0.75%Methylcellulose medium. After 4 days of incubation, the overlays were removed, and the cells were fixed and stained with 0.1%crystal violet for one hour and then rinsed and air dried.
  • the corresponding reciprocal neutralizing antibody titers were determined at the 60%reduction end-point of the virus control using the statistics program "plqrd. manual. entry" .
  • the geometric means ⁇ standard error for all animals in a group at a given time were calculated.
  • Lung and nose homogenates were clarified by centrifugation and diluted in EMEM.
  • Confluent HEp-2 monolayers were infected in duplicates with diluted homogenates in 24 well plates. After one hour incubation at 37°C in a 5%CO2 incubator, the wells were overlayed with 0.75%Methylcellulose medium. After 4 days of incubation, the overlay was removed, and the cells were fixed with 0.1%crystal violet stain for one hour and then rinsed and air dried. Plaques were counted and virus titer was expressed as plaque forming units per gram of tissue. Viral titers were calculated as geometric mean + standard error for all animals in a group at a given time.
  • the titration results of RSV lung viral load at day 61 is shown in Fig. 3.
  • the titration results of RSV nose viral load at day 61 is shown in Fig. 4.
  • FI-RSV formalin-inactivated RSV vaccine
  • RSV Neutralizing Antibody Assay (60%PRNT) was carried out according to essentially the same protocol as described in Example 2. The titration results against RSV/A2 and RSV B on day 35 are shown in Fig. 5 and Fig. 6, respectively.
  • RSV/A2 lung and nose titration was carried out according to essentially the same protocol as described in Example 2.
  • the titration results of RSV lung viral load at day 61 is shown in Fig. 7.
  • the titration results of RSV nose viral load on day 61 is shown in Fig. 8.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Provided are polypeptides that are engineered RSV prefusion F protein variants, and polynucleotides, vectors, compositions, kits, and methods related to these polypeptides.

Description

RSV F Protein Variants and Uses Thereof
FIELD OF DISCLOSURE
The present disclosure relates to variants of RSV F protein, and polynucleotides, vectors, compositions, and kits of these variants. The present disclosure also provides methods for generating an immune response to RSV F protein in a subject and methods for preventing RSV infection in a subject.
SEQUENCE LISTING
This application contains a Sequence Listing electronically submitted as an XML file entitled “CIE23B0119PCT-sequence listing” having a size of 97 Kbytes and created on 2023-10-12. The information contained in the Sequence Listing is incorporated by reference herein.
BACKGROUND
Respiratory syncytial virus (RSV) is a common, contagious virus that causes infections of the lungs and the respiratory tract. RSV infection is the leading cause of respiratory hospitalization in infants, and reinfection remains common in later life. It is an important pathogen in all age groups. RSV causes a respiratory tract infection that affects 64 million people per year worldwide. It hospitalizes 3 million children under 5 years old and approximately 336,000 older adults annually.
The F glycoprotein plays a pivotal role in the pathogenesis of RSV by mediating the fusion between the viral and the host cell membrane. The F protein presents two different conformations, a lollipop-shaped prefusion form (preF) , present on the virus surface before virus–cell interaction, and a crutch-shaped postfusion form (postF) which is acquired following the fusion between the virus and cell membrane or by unknown mechanisms that spontaneously initiate the rearrangement from the highly metastable preF conformation into the energetically favorable postF conformation (Mejias et al, 2017) . The two forms are antigenically distinct. The preF form has been shown to induce the majority of highly neutralizing antibodies following natural infection or immunization. The two most immunogenic sites on the F protein, named Siteand Site V, are only present on the preF conformation, while less immunogenic sites (Site I, Site II, Site III, and Site IV) are shared between the preF and postF forms. F glycoprotein is highly conserved among RSV subtypes, with amino acid sequence identities of 90%or higher. This relative sequence conservation of the F protein combined with its surface location on the virion, its obligatory role in viral entry,  and its antigenic sites associated with potent neutralization make the F protein, particularly the preF, an ideal target for neutralizing antibodies and a promising vaccine antigen.
Vaccines work by presenting the immune system in a subject with key bits of a pathogen, which induce the production of antibodies and immune cells that can recognize and fight the whole pathogen when the subject is re-exposed to it. As the structure of RSV prefusion F protein being revealed in the art, there is a need to identify RSV prefusion F protein variants that are suitable for vaccine designs.
SUMMARY
The present disclosure provides multiple engineered RSV prefusion F protein variants as well as codon-optimized mRNAs encoding them, which can be useful in vaccine designs. The present disclosure also provides vectors, compositions, kits, and methods related to these engineered preF variants.
In an aspect, the present disclosure provides a polypeptide comprising an amino acid sequence of any one of SEQ ID NO: 26-28, 30-50.
In an aspect, the present disclosure provides a polypeptide comprising an amino acid sequence with at least 90%identity to any one of SEQ ID NO: 26-28, 30-50.
In an aspect, the present disclosure provides a polynucleotide comprising a coding sequence that encodes the polypeptide disclosed herein.
In an aspect, the present disclosure provides a polynucleotide comprising a sequence of any one of SEQ ID NOs: 1-25.
In some embodiments, the polynucleotide is an mRNA sequence.
In some embodiments, the coding sequence is operably linked to a 5’ untranslated region (5’ UTR) and/or a 3’ untranslated region (3’ UTR) .
In some embodiments, the polynucleotide disclosed herein further comprises a 5’ cap structure.
In some embodiments, the polynucleotide disclosed herein further comprises a poly (A) tail at its 3’ end.
In some embodiments, the polynucleotide disclosed herein comprises at least one modified nucleotide.
In some embodiments, the polynucleotide disclosed herein comprises from the 5’-end to the 3’-end: a 5’-cap structure, a 5’ UTR, the coding sequence, a 3’ UTR, and a poly (A) sequence.
In another aspect, the present disclosure provides a vector comprising the polynucleotide disclosed herein.
In some embodiments, the vector is a viral vector or a plasmid.
In another aspect, the present disclosure provides a composition comprising the polypeptide disclosed herein.
In another aspect, the present disclosure provides a composition comprising the polynucleotide disclosed herein.
In some embodiments, the composition further comprising a carrier selected from the group consisting of lipid nanoparticles, liposomes, cationic nanoemulsions, dendrimer-based lipid nanoparticles, cationic polymers, and polysaccharide particles.
In another aspect, the present disclosure provides a kit comprising the polynucleotide disclosed herein and a carrier selected from the group consisting of lipid nanoparticles, liposomes, cationic nanoemulsions, dendrimer-based lipid nanoparticles, cationic polymers, and polysaccharide particles.
In another aspect, the present disclosure provides a method for generating an immune response to RSV F protein in a subject in need thereof, comprising administering to the subject an effective amount of the polypeptide disclosed herein, or the polynucleotide disclosed herein.
In another aspect, the present disclosure provides a method for preventing RSV infection in a subject in need thereof, comprising administering to the subject an effective amount of the polypeptide disclosed herein, or the polynucleotide disclosed herein.
In another aspect, the present disclosure provides a method for generating an immune response to RSV F protein in a subject in need thereof, comprising administering an effective amount of the composition disclosed herein.
In another aspect, the present disclosure provides a method for preventing RSV infection in a subject in need thereof, comprising administering an effective amount of the composition disclosed herein.
In some embodiments, the RSV is RSV A or RSV B.
BRIEF DESCRIPTION OF THE FIGURES
Fig. 1 shows the results of viral neutralizing titration (PRNT60) against RSV/A2 on day 56, as described in Example 2.
Fig. 2 shows the results of viral neutralizing titration (PRNT60) against RSV B on day 56, as described in Example 2.
Fig. 3 shows the titration result of RSV lung viral load on day 61, as described in Example 2.
Fig. 4 shows the titration result of RSV nose viral load on day 61, as described in Example 2.
Fig. 5 shows the results of viral neutralizing titration (PRNT60) against RSV/A2 on day 35, as described in Example 3.
Fig. 6 shows the results of viral neutralizing titration (PRNT60) against RSV B on day 35, as described in Example 3.
Fig. 7 shows titration result of RSV lung viral load on day 61, as described in Example 3.
Fig. 8 shows titration result of RSV nose viral load on day 61, as described in Example 3.
DETAILED DESCRIPTION
Definition
All publications, patents, and patent applications referred to herein are incorporated by reference in their entirety to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference in its entirety.
In the present disclosure, unless otherwise specified, the scientific and technical terms used herein have the meanings generally understood by a person skilled in the art. Although any methods and materials similar or equivalent to those described herein find use in the practice of the present disclosure, the preferred methods and materials are described herein. Accordingly, the terms defined herein are more fully described by reference to the Specification as a whole.
As used herein, the singular terms “a, ” “an, ” and “the” include the plural reference unless the context clearly indicates otherwise.
As used herein, “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations when interpreted in the alternative ( “or” ) . Moreover, the present invention also contemplates that in some embodiments of the invention, any feature or combination of features set forth herein can be excluded or omitted.
The term “about, ” as used herein when referring to a measurable value such as a sequence length and the like, is meant to encompass variations of 5%, 1 %, 0.5%, or even 0.1 %of the specified amount.
Unless the context requires otherwise, the terms “comprise, ” “comprises, ” and “comprising, ” or similar terms are intended to mean a non-exclusive inclusion, such that a  recited list of elements or features does not include those stated or listed elements solely, but may include other elements or features that are not listed or stated.
Unless otherwise indicated, nucleic acids are written left to right in the 5' to 3' orientation; and amino acid sequences are written left to right in amino to carboxy orientation, respectively.
It is to be understood that this disclosure is not limited to the particular methodology, protocols, and reagents described, as these may vary, depending upon the context in which they are used by those skilled in the art.
As used herein, the terms “percent identity” and “%identity, ” as applied to nucleic acid or polynucleotide sequences, refer to the percentage of residue matches between at least two nucleic acid or polynucleotide sequences aligned using a standardized algorithm. Such an algorithm may insert, in a standardized and reproducible way, gaps in the sequences being compared in order to optimize alignment between two sequences, and therefore achieve a more meaningful comparison of the two sequences.
Percent identity between nucleic acid or polynucleotide sequences may be determined using a suite of commonly used and freely available sequence comparison algorithms provided by the National Center for Biotechnology Information (NCBI) Basic Local Alignment Search Tool (BLAST) (Altschul, S. F. et al. (1990) J. Mol. Biol. 215: 403-410) , which is available from several sources, including the NCBI, Bethesda, Md., and on the Internet at http: //www. ncbi. nlm. nih. gov/BLAST/.
Nucleic acid or polynucleotide sequences that do not show a high degree of identity may nevertheless encode similar amino acid sequences due to the degeneracy of the genetic code. It is understood that changes in a nucleic acid sequence can be made using this degeneracy to produce multiple nucleic acid sequences that all encode substantially the same protein. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al. (1991) Nucleic Acid Res 19: 5081; Ohtsuka et al. (1985) J Biol Chem 260: 2605-2608; Rossolini et al. (1994) Mol Cell Probes 8: 91-98) .
The term “nucleic acid” refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single-or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides which have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. The term nucleic acid is used interchangeably with polynucleotide, and (in appropriate contexts) gene, cDNA, and mRNA encoded by a gene.
As used herein, “percent (%) amino acid sequence identity” with respect to a peptide, polypeptide or protein sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in another peptide or  polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Percent amino acid sequence identity in the current disclosure is measured using BLAST software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
An amino acid substitution refers to the replacement of one amino acid in a polypeptide with another amino acid. Exemplary substitutions are shown in Table 1. Amino acid substitutions may be introduced into a protein of interest and the products screened for a desired activity, for example, retained/improved biological activity.
Table 1
Amino acids may be grouped according to common side-chain properties:
(1) hydrophobic: Norleucine, Met, Ala, Val, Leu, Ile;
(2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gln;
(3) acidic: Asp, Glu;
(4) basic: His, Lys, Arg;
(5) residues that influence chain orientation: Gly, Pro;
(6) aromatic: Trp, Tyr, Phe.
Non-conservative substitutions will entail exchanging a member of one of these classes for another class. The term, “corresponding to” with reference to nucleotide or amino acid positions of a sequence, such as set forth in the Sequence Listing, refers to nucleotides or amino acid positions identified upon alignment with a target sequence based on structural sequence alignment or using a standard alignment algorithm, such as the GAP algorithm. For example, corresponding residues of a similar sequence (e.g., a fragment or species variant) can be determined by alignment to a reference sequence by structural alignment methods. By aligning the sequences, one skilled in the art can identify corresponding residues, for example, using conserved and identical amino acid residues as guides.
As used herein, a composition refers to any mixture of two or more products, substances, or compounds, including cells.
As used herein, an “effective amount” refers to an amount of a pharmaceutical composition which is sufficient to significantly and positively modify the symptoms and/or conditions to be treated (e.g., provide a positive clinical response) . The effective amount of a pharmaceutical composition will vary with the particular condition being treated, the severity of the condition, the duration of treatment, the nature of concurrent therapy, the particular composition being employed, the particular pharmaceutically-acceptable excipient (s) and/or carrier (s) utilized, and like factors with the knowledge and expertise of the attending physician.
As used herein, the terms “individual” and “subject” are used interchangeably herein to refer to an animal. For example, in some embodiments, the animal is a mammal. In some embodiments, the animals are humans, rodents, simians, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, or mammalian pets. The animal can be male or female and can be at any suitable age, including infant, juvenile, adolescent, adult, and geriatric. In some examples, an “individual” or “subject” refers to an animal in need of treatment for a disease or disorder. In some embodiments, the animal to receive the treatment can be a “patient, ” designating the fact that the animal has been identified as having a disorder of relevance to the treatment, or being at adequate risk of contracting the disorder. In particular embodiments, the animal is a human, such as a human patient.
As used herein, the term “coding sequence” and “coding region” are used interchangeably. They refer to the portion of a polynucleotide that codes for a protein.
As used herein, the term “engineered” in the context of protein and protein variants means that the protein or protein variant is not naturally occurring.
RSV F Protein
Respiratory syncytial virus (RSV) is a common, contagious virus that causes infections of the lungs and the respiratory tract. RSV infection is the leading cause of respiratory hospitalization in infants, and reinfection remains common in later life. It is an important pathogen in all age groups.
RSV is a negative-sense, single-stranded RNA virus. Its genome has 10 genes which encode 11 proteins, including 9 structural proteins (3 glycoproteins and 6 internal proteins) and 2 non-structural proteins. The structural proteins include 3 transmembrane surface glycoproteins: the attachment protein G, fusion protein F, and the small hydrophobic SH protein. F and G glycoproteins are the two major surface proteins that control viral attachment and the initial stages of infection. F and G proteins are also the primary targets for neutralizing antibodies during natural infection.
The F glycoprotein plays a pivotal role in the pathogenesis of RSV by mediating the fusion between the viral and the host cell membrane. The F gene encodes a type I integral membrane protein that is synthesized as a 574 amino acid inactive precursor, F0. Three F0 monomers assemble into a trimer and, as the trimer passes through the Golgi, the monomers are activated by a furin-like host protease (Bolt et al. 2000; Collins and Mottet 1991) . The protease cleaves twice, after amino acids 109 and 136 (González-Reyes et al. 2001; Zimmer et al. 2001a) , generating three polypeptides. The N-terminal and C-terminal cleavage products are the F2 and F1 subunits, respectively, which are covalently linked to each other by two disulfide bonds (Gruber and Levine 1983; Day et al. 2006) . The intervening 27 amino acid peptide dissociates after cleavage (Begona Ruiz-Arguello et al. 2002) .
The F protein presents two different conformations, a lollipop-shaped prefusion form (preF) , present on the virus surface before virus–cell interaction, and a crutch-shaped postfusion form (postF) which is acquired following the fusion between the virus and cell membrane or by unknown mechanisms that spontaneously initiate the rearrangement from the highly metastable preF conformation into the energetically favorable postF conformation (Mejias et al, 2017) . The two forms are antigenically distinct. The preF form has been shown to induce the majority of highly neutralizing antibodies following natural infection or immunization. The two most immunogenic sites on the F protein, named Siteand Site V, are only present on the preF conformation, while less immunogenic sites (Site I, Site II, Site III, and Site IV) are shared between the preF and postF forms.
There are two subtypes of RSV, RSV A and RSV B. They differ primarily in the G glycoprotein, while the F glycoprotein is highly conserved among RSV subtypes, with  amino acid sequence identities of 90%or higher. Much of the variability in the F protein (about 25%) is found within an antigenic siteat the apex of the prefusion trimer, which is composed of an α-helix from F1 (aa 196–210) and a strand from F2 (aa 62–69) and may be a site that determines subtype-specific immunity (McLellan et al. 2013) . This relative sequence conservation of the F protein combined with its surface location on the virion, its obligatory role in viral entry, and its antigenic sites associated with potent neutralization make the F protein, particularly the preF, an ideal target for neutralizing antibodies and a promising vaccine antigen (Anderson et al. 1988; Walsh and Hruska 1983; Costello et al. 2012) .
The present disclosure provides several engineered preF variants and polynucleotide encoding them, which can be useful in vaccine design. The present disclosure also provides vectors, compositions, kits, and methods related to these engineered preF variants.
Polypeptides
In an aspect, the present disclosure provides engineered RSV prefusion F protein variants such as a polypeptide comprising an amino acid sequence of any one of SEQ ID NO: 26-28, 30-50. (see Table 2)
Table 2 Polypeptide Sequences





The present disclosure also provides a polypeptide comprising an amino acid sequence that has at least 95%identity to any one of SEQ ID NO: 26-28, 30-50.
In some embodiments, the polypeptide has an amino acid sequence that has at least 90%identity to any one of SEQ ID NOs: 26-28, 30-50. In some embodiments, the polypeptide has an amino acid sequence that has at least 95%, 96%, 97%, 98%, or 99%identity to any one of SEQ ID NOs: 26-28, 30-50.
In some embodiments, the polypeptide further comprises a signal peptide at its N terminus or C terminus. As used herein, a signal peptide refers to the short peptide present at the N terminus or C terminus of the polypeptide when it is initially translated. Signal peptides are usually cleaved off from a protein by a signal peptidase during or immediately after insertion into a cell membrane. Signal peptides function to prompt a cell to translocate the protein, usually to the plasma membrane.
Polynucleotides
In another aspect, the present disclosure provides a polynucleotide comprising a coding sequence that encodes the polypeptide disclosed herein.
In some embodiments, the polynucleotide is DNA. In some embodiments, the mRNA transcribed from the DNA comprises a sequence of any one of SEQ ID NOs: 1-25.
In an aspect, the present disclosure provides a polynucleotide comprising a sequence of any one of SEQ ID NOs: 1-25. (Table 3) .
In some embodiments, the polynucleotide is RNA. In some embodiments, the polynucleotide is mRNA. In some embodiments, the mRNA sequences are codon-optimized for enhanced stability and immunogenicity.
Table 3 RNA Sequences
















In some embodiments, the coding sequence is operably linked to a 5’ untranslated region (5’ UTR) and/or a 3’ untranslated region (3’ UTR) . In some embodiments, the 5’ UTR comprises a sequence of SEQ ID NO: 51 (Table 3) . In some embodiments, the 3’ UTR comprises a sequence of SEQ ID NO: 52 (Table 3) . The untranslated region (UTR) is a regulatory region situated at the 5’ or 3’ end of a coding region. 5’ UTR is directly upstream from the initiation codon of the coding region. In some embodiments, the 5’ UTR is further added with a 5’ cap structure. 3’ UTR immediately follows the translation termination codon of the coding region. The 3’ UTR often contains regions for post-transcriptional regulation, such as polyadenylation, localization, and stability of the mRNA. In some embodiments, the 3’ UTR is further connected with a poly (A) tail.
In some embodiments, the mRNA further comprises a 5’ cap structure. The 5’ cap structure is a specially altered nucleotide that “caps” the 5’ end of the mRNA. A 5’ cap structure can regulate nuclear export of the RNA, prevent degradation by exonucleases, and promote translation. In some embodiments, the 5’ cap is formed by a derivative of a guanine nucleotide. In some embodiments, the 5’ cap is linked to the 5’ terminus via a 5’-5’ triphosphate linkage. In some embodiments, the 5’ cap is methylated, e.g., m7GpppN, wherein N is the 5’ terminal nucleotide of the nucleic acid carrying the 5’ cap. In some embodiments, the 5’ cap structure is selected from glyceryl, inverted deoxy abasic residue, 4’, 5’-methylene nucleotide, 1- (beta-D-erythrofuranosyl) nucleotide, 4’-thio nucleotide, carbocyclic nucleotide, 1, 5-anhydrohexitol nucleotide, L-nucleotides, alpha-nucleotide, modified base nucleotide, threo-pentofuranosyl nucleotide, acyclic 3’, 4’-seco nucleotide, acyclic 3, 4-dihydroxybutyl nucleotide, acyclic 3, 5 dihydroxypentyl nucleotide, 3’-3’-inverted nucleotide moiety, 3’-3’-inverted abasic moiety, 3’-2’-inverted nucleotide moiety, 3’-2’-inverted abasic moiety, 1, 4-butanediol phosphate, 3’-phosphoramidate, hexylphosphate, aminohexyl phosphate, 3’-phosphate, 3’-phosphorothioate, phosphorodithioate, bridging methylphosphonate moiety, and non-bridging methylphosphonate moiety.
In some embodiments, the mRNA further comprises a poly (A) tail at its 3’ end. The poly (A) tail is a long chain of adenine nucleotides that is added to the 3’ end of a mRNA molecule. In some embodiments, the length of the poly (A) tail is at least 80, 90, 100, 150, 200, 250, 300, 350, 400, 450 or 500 nucleotides. In some embodiments, the length of the poly (A) tail is adjusted to control the stability of the mRNA molecule disclosed herein. For example, since the length of the poly (A) can influence the half-life of the mRNA molecule, the length of the poly (A) tail can be adjusted to modify the level of resistance of the mRNA to nucleases and thereby control the time course of protein expression.
In some embodiments, the polynucleotide comprises at least one modified nucleotide. In some embodiments, the modified nucleotide is selected from the group consisting of pseudouridine (Ψ) , 1-methylpseudouridine (m1Ψ) , 5-methyluridine (m5U) , 2-thiouridine (s2U) , and 5-methylcytidine (m5C) . As used herein, the term “modified nucleotide” includes any synthetic nucleotide and nucleotide analogue, and any naturally existing nucleotide other than adenine, adenine, guanine, thymine, or uracil. Exemplary nucleotide modification includes 2’-O-methylation (Nm) and conversion of uridine to pseudouridine (Ψ) . Both Nm and Ψ modifications have the potential to stabilize RNA folding domains. Methylation of 2’-OH sites endows a nucleotide with greater hydrophobicity, protects against nucleolytic attack and stabilizes helices, and thus can benefit inter-or intra-molecular interactions. Pseudouridine exerts a significant rigidifying influence on the sugar–phosphate backbone and enhances base stacking. In addition, Ψ provides an additional donor site for hydrogen-bond formation, which can stabilize RNA–RNA or RNA–protein interactions (Baillieu et al., Nucleic Acids Res. 2009) .
In some embodiments, the polynucleotide comprises from the 5’-end to the 3’-end: a 5’-cap structure, a 5’ UTR, the coding sequence, a 3’ UTR, and a poly (A) sequence.
In some embodiments, the polynucleotide further encodes an RNA polymerase. A self-amplifying RNA (saRNA) encode not only an antigen of interest but also a viral RNA-dependent RNA polymerase to amplify the RNA in the cytoplasm of transfected cells, lead to significantly greater immune responses than conventional RNAs.
A polynucleotide encoding the polypeptide disclosed herein can be obtained by methods known in the art.
Vector
In another aspect, the present disclosure provides a vector comprising the polynucleotide disclosed herein. In some embodiments, the vector is a viral vector or a plasmid. Viral vectors can include, but are not limited to, adenoviral vectors, lentiviral vectors, retroviral vectors, and adeno-associated viral vectors. Commonly, expression vectors contain selection markers such as ampicillin-resistance, hygromycin-resistance, tetracycline resistance, kanamycin resistance, or neomycin resistance to permit detection of those cells transformed with the desired DNA sequences. Suitable vectors, promoter, and enhancer elements are known in the art; many are commercially available for generating subject recombinant constructs. In some embodiments, the vector is a polycistronic vector. Any methods known to those skilled in the art for the insertion of polynucleotide fragments into a vector can be used to construct the vector disclosed herein. The polynucleotide disclosed herein can be operably linked to control sequences in the expression vector (s) to ensure the expression of the polypeptide disclosed herein. Such control sequences may include, but are not limited to, leader or signal sequences, promoters (e.g., naturally associated or heterologous promoters) , ribosomal binding sites, enhancer or activator elements, translational start and termination sequences, and transcription start and termination sequences.
Composition
In another aspect, the present disclosure provides a composition comprising the polypeptide disclosed herein.
In another aspect, the present disclosure provides a composition comprising the polynucleotide disclosed herein.
As used herein, the term “composition” includes, but is not limited to, a pharmaceutical composition. A “pharmaceutical composition” refers to an active pharmaceutical agent formulated in pharmaceutically acceptable or physiologically acceptable solutions for administration to a cell or an animal, either alone, or in combination with one or more other modalities of therapy. It will also be understood that, if desired, the compositions of the disclosure may be administered in combination with other agents, such as,  e.g., cytokines, growth factors, hormones, small molecules, chemotherapeutics, pro-drugs, drugs, antibodies, or other various pharmaceutically active agents. There is virtually no limit to other components that may also be included in the compositions, provided that the additional agents do not adversely affect the ability of the composition to deliver the intended therapy. The phrase “pharmaceutically acceptable” is used herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The compositions may also comprise a pharmaceutically acceptable carrier, diluent, or excipient. As used herein “pharmaceutically acceptable carrier, diluent, or excipient” includes, without limitation, any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, surfactant, or emulsifier which has been approved as being acceptable for use in humans or domestic animals. Exemplary pharmaceutically acceptable carriers include, but are not limited to, to sugars, such as lactose, glucose, and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; tragacanth; malt; gelatin; talc; cocoa butter; waxes; animal and vegetable fats; paraffins; silicones; bentonites; silicic acid; zinc oxide; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; esters, such as ethyl oleate, and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and any other compatible substances employed in pharmaceutical formulations.
The liquid pharmaceutical compositions, whether they be solutions, suspensions or other like form, may include one or more of the following: sterile diluents such as water for injection, saline solution, preferably physiological saline; Ringers solution; isotonic sodium chloride; fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium; polyethylene glycols; glycerin; propylene glycol or other solvents; antibacterial agents, such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates, or phosphates; and agents for the adjustment of tonicity, such as sodium chloride or dextrose. The parenteral preparation can be enclosed in ampoules, disposable syringes, or multiple dose vials made of glass or plastic. An injectable pharmaceutical composition is preferably sterile. The composition may be suitably developed for intravenous, intratumoral, oral, rectal, vaginal, parenteral, topical, pulmonary, intranasal, buccal, ophthalmic, or another route of administration.
In some embodiments, the composition further comprises a carrier selected from the group consisting of lipid nanoparticle, liposome, cationic nanoemulsion, dendrimer nanoparticle, cationic polymer, and polysaccharide particle. As used herein, the term “carrier” refers to compounds or compositions that are used for delivery of the polypeptide or the polynucleotide into a subject. Preferably, the carrier enhances effectiveness and/or safety of the delivery. In some embodiments, the carrier is capable of delivering large nucleic acid sequences (e.g., nucleic acids of at least 1 kDa, 1.5 kDa, 2 kDa, 2.5 kDa, 5 kDa, 10 kDa, 12 kDa, 15 kDa, 20 kDa, 25 kDa, 30 kDa, or more) . The nucleic acids can be formulated with one or more acceptable reagents, which provide a vehicle for delivering such nucleic acids to target cells. Appropriate reagents are generally selected with regards to a number of factors, which include, among other things, the biological or chemical properties of the nucleic acids (e.g., charge) , the intended route of administration, the anticipated biological environment to which such nucleic acids will be exposed and the specific properties of the intended target cells.
Lipid nanoparticles (LNPs) are nanoparticles made of one or more types of lipids. In some embodiments, lipid nanoparticles comprise ionizable lipids, which are positively charged at low pH (enabling RNA complexation) and neutral at physiological pH (reducing potential toxic effects, as compared with positively charged lipids, such as liposomes) . Owing to their size and properties, lipid nanoparticles are taken up by cells via endocytosis, and the ionizability of the lipids at low pH (likely) enables endosomal escape, which allows release of the cargo into the cytoplasm. In some embodiments, the lipid nanoparticles comprise cationic lipids, which have a head group with permanent positive charges. In addition, lipid nanoparticles usually contain a helper lipid, for example, phospholipid, to promote cell binding, cholesterol to fill the gaps between the lipids, and a polyethylene glycol (PEG) to reduce opsonization by serum proteins and reticuloendothelial clearance. The relative amounts of ionizable lipid, helper lipid, cholesterol and PEG can vary (See Hou et al., Nature Reviews Materials, 2021) .
Liposomes are spherical-shaped vesicles that is composed of one or more phospholipid bilayers. Liposomes are most often composed of phospholipids, especially phosphatidylcholine and cholesterol, but may also include other lipids, such as phosphatidylethanolamine, as long as they are compatible with lipid bilayer structure. The lipid bilayer of liposome can fuse with other bilayers such as the cell membrane, thus delivering the liposome contents. Generally, liposomes are definite as spherical vesicles with particle sizes ranging from 30 nm to several micrometers. They consist of one or more lipid bilayers surrounding aqueous units, where the polar head groups are oriented in the pathway of the interior and exterior aqueous phases. On the other hand, self-aggregation of polar lipids is not limited to conventional bilayer structures which rely on molecular shape, temperature, and environmental and preparation conditions but may self-assemble into various types of colloidal particles (See Akbarzadeh, Nanoscale Res Lett., 2013) .
Cationic nanoemulsions (CNE) are mainly composed of two parts: one is the cationic lipid DOTAP (1, 2-dioleoyl-sn-glycero-3-phosphocholine) that can be added to the oil phase to bind the mRNA electrostatically; the other is the emulsion adjuvant MF59 that is an oil-in-water emulsion consisting of squalene and surfactants. CNEs are usually fabricated by the probe sonication method (Brito et al., A cationic nanoemulsion for the delivery of next-generation RNA vaccines, 2014) .
Dendrimer-based lipid nanoparticles are nanoparticles made of lipids and dendrimers, which are highly ordered, branched polymeric molecules. Dendrimers are composed of three distinct structural components: (1) the core, (2) the repetitive branching layers (also referred to as “generation” ) , and (3) the abundant terminal groups. These precisely controlled dendritic structures harbor multivalent cooperativity and can exploit membrane-fusion-based endosome release by mimicking lipid vectors, while simultaneously retaining the “proton-sponge” -mediated endosome release of polymer vectors (See Chen et al., Amphiphilic Dendrimer Vectors for RNA Delivery: State-of-the-Art and Future Perspective, 2022) .
Cationic polymer is another viable RNA carrier. An exemplary cationic polymer is poly (ethyleneimine) and its derivatives Polyethyleneimine (PEI) is among the earliest and most widely studied cationic polymers for gene delivery, including the delivery of RNA. It has high gene transfection efficiency and is often referred to as the gold standard for non-viral gene transfection (Lungwitz et al., 2005) . PEI can be in either linear or branched structures and its positive charge is conferred by numerous amine groups separated by short alkyl spacers, which lead to very high positive charge density within its structure (Jiang et al., Polymeric nanoparticles for RNA delivery, 2021) .
Polysaccharides are a complex collection of biopolymers isolated from plant, animal, microbial and algal sources that are built from monosaccharides linked by O-glycosidic linkages. An exemplary polysaccharide that can be used for RNA delivery is Chitosan, is a polysaccharide contained in the cell walls of fungi and in the shells of arthropods such as crustaceans and consists of a linear chain of 2-acetaylamino-2-deoxy-β-D-glucopyranose units connected through β-1, 4 linkages (Bodnar, Hartmann & Borbely, 2005; Barclay et al., Review of polysaccharide particle-based functional drug delivery, 2020) .
In another aspect, the present disclosure provides a kit comprising the polynucleotide disclosed herein, and a carrier selected from the group consisting of lipid nanoparticle, liposome, cationic nanoemulsion, dendrimer nanoparticle, cationic polymer, and polysaccharide particle.
Methods
In another aspect, the present disclosure provides a method for generating an immune response to RSV F protein in a subject in need thereof, comprising administering to the subject an effective amount of the polypeptide disclosed herein, or the polynucleotide  disclosed herein. An immune response may typically be a specific reaction of the adaptive immune system to a particular antigen (so called specific or adaptive immune response) or an unspecific reaction of the innate immune system (so called unspecific or innate immune response) , or a combination thereof. For adaptive immune response, it could be a humoral immune response and/or a cellular immune response.
Humoral immunity refers typically to antibody production and optionally to accessory processes accompanying antibody production. A humoral immune response may typically be characterized, e.g., by Th2 activation and cytokine production, germinal center formation and isotype switching, affinity maturation and memory cell generation. Humoral immunity also typically may refer to the effector functions of antibodies, which include pathogen and toxin neutralization, classical complement activation, and opsonin promotion of phagocytosis and pathogen elimination.
Cellular immunity typically relates to the activation of macrophages, natural killer cells (NK) , antigen-specific cytotoxic T-lymphocytes, and the release of various cytokines in response to an antigen. In more general terms, cellular immunity is not based on antibodies, but on the activation of cells of the immune system. Typically, a cellular immune response may be characterized, for example, by activating antigen-specific cytotoxic T-lymphocytes that are able to induce apoptosis in cells, e.g., specific immune cells like dendritic cells or other cells, displaying epitopes of foreign antigens on their surface. Such cells may be virus-infected or infected with intracellular bacteria, or cancer cells displaying tumor antigens. Further characteristics may be activation of macrophages and natural killer cells, enabling them to destroy pathogens, and stimulation of cells to secrete a variety of cytokines that influence the function of other cells involved in adaptive immune responses and innate immune responses.
In another aspect, the present disclosure provides a method for preventing RSV infection in a subject in need thereof, comprising administering to the subject an effective amount of the polypeptide disclosed herein, or the polynucleotide disclosed herein. In some embodiments, the method prevents RSV infection by generating an immune response to RSV F protein. In some embodiments, the method prevents RSV infection by generating an adaptive immune response to RSV F protein in the subject.
In another aspect, the present disclosure provides a method for generating an immune response to RSV F protein in a subject in need thereof, comprising administering an effective amount of the composition disclosed herein. In some embodiments, the immune response is an adaptive immune response.
In another aspect, the present disclosure provides a method for preventing RSV infection in a subject in need thereof, comprising administering an effective amount of the composition disclosed herein. In some embodiments, the method prevents RSV infection by generating an adaptive immune response to RSV F protein in the subject.
In some embodiments, the RSV is RSV A or RSV B.
EXAMPLES
Example 1 In vitro screening assay
This series of Elisa assays was to confirm RSV prefusion protein expression by the mRNA disclosed herein. The cell culture plates corning 3599 were coated with 1/100 collagen I and the Elisa plates corning 9018 were coated with 100μl per well of RSV Pre-F protein at 2 μg/ml at 4 ℃ overnight. The HEp2 cells were seeded in coated 96 well plates Corning 3599 with 100 μl per well cells at the density of 1.2E4 cells/ml. After 24 hours incubation, synthetic mRNA was in vitro transfected by using a TransIT-mRNA Transfection kit (Mirus) . After 48 hours incubation, a competitive antibody binding assay was used to determine the levels of antibodies in transfected cell supernatant that competed the binding of RSV F site-specific monoclonal antibodies D25 (site ) , the goat anti RSV HRP secondary antibody (1: 500) was used for detection of the prefusion protein expression.
Sandwich ELISA
Corning 96 well, flat bottom, high binding plates were coated with 100 μl per well of RSV Pre-F protein at 2 μg/ml at 4 ℃ overnight. The plates were washed in PBS/0.05%Tween 20 and then blocked in PBS/0.05%Tween 20/3%non-fat milk at room temperature for 1 h. After washed with 300 ul PBS/0.05%Tween 20, 100 μl per well of 3-fold serially titrated mAbs (preF standard protein, 2ug/ml start, 3 fold dilution, 7 points, and the 8th point was DPBS Mock) and mRNA transfection cell supernatants were then added and incubated at room temperature for 90 min. Plates were washed with PBS/0.05%Tween 20 three times, and 100 μl per well of HRP-conjugated goat anti-RSV (1: 500) (Meridian, catalog#: B65940G, Lot#: 3K30721) ) was added. After 60 min incubation at room temperature, plates were washed and 100 μl per well of TMB Solution (Thermo: 002023) was added. Plates were incubated at room temperature for 15 min and the reaction was stopped by adding 100 μl per well of Stop Solution for TMB ELISA. Plates were then read on (Tecan/spark) at 450 nm.
Materials used are listed below.
Reagents:
-Collagen Type-I Rat Tail: Corning #354236
-D25 mAb: BIOINTRON #B21266501-CHO
-DPBS, 1X: Corning #21-031-CVC
-DMEM/F12 (1: 1) (1X) : Gibco #11330-032
-Fetal bovine serum: Corning #35-081-CV
-TrypLETM Express: Gibco #12605028
-Penicillin Streptomycin: Gibco #15070-063
-Gluta MAXTM-I: Gibco #35050-061
-Tween-20: Sangon #A600560-0500
-DifcoTM Skim Milk: BD#232100
-AO/PI staining solution: Count star #RE010212
-Opti-MEMTM I: Gibco #31985062
-TransIT-mRNA Tranfection Kit: MIR #2250
-UltraPureTM DNase/RNase free distilled water: Invitrogen #10977015
-Goat anti RSV, Polyclonal, HRP: Meridian life science #B65840G
-0.5N Sulfuric acid: 14ml to 1L ddH2O
-TMB single solution: #002023
Consumables:
-Costar Assay plate, 96well clear, flat Bottom, High binding: Corning #9018
-96 well cell culture plate: Corning #3599
-50ml Reagent Reservior: Corning #4870
-50ml centrifuge tubes: Corning #430291
-Self-adhesive Sealing Film for microplate, clear, Axygen: Beyotime #FSF007
Equipment:
-Count Star, Fluorescence Cell Analyzer: Conutstar Rigel S2 #RY071L2102
-Water Bath: SAP12 #UE2134006
-Centrifuge: Multifuge X4R Pro #75009915
-Inverted microscope: #CKX53
Procedures
Coating ELISA plate with capture antibody D25 mAb and coating the cell plate with Collagen Type I Rat Tail (day 1) .
D25 mAb dilution and ELISA plate coating: Dilute the D25 mAb 4.2 mg/ml to 2 μg/ml with DPBS and mix gently. Dispense 100 ul 2ug/ml D25 mAb to the ELISA assay plate with 200 ul 12 channel pipette. Place the coated plates in a 4℃ refrigerator overnight. Coating the cell culture plate with collagen type I Rat tail. Dilute the collagen type I Rat tail 100-fold with DPBS. Dispense 1/100 diluted collagen I to the 96 well plate with 100 ul per well. Place the coated plate to a 4℃ refrigerator overnight.
ELISA assay plate blocking and cell plating (day 2) 
HEp2 Cell seeding: Remove HEp2 culture T150 flask from the incubator and check the cell density with a microscope, the cells should reach to 80%-85%confluent. Remove and discard the spent cell culture media from the culture flask. Wash cells using the DPBS solution without calcium and magnesium. Remove and discard the wash solution, add 2.5 ml pre-warmed dissociation TrypLETM to the side of the flask. Incubate the culture flask at 37 ℃ cell culture incubator for 5 minutes. When ≥ 90%of the cells have detached, tilt the vessel for a minimal length of time to allow the cells to drain. Add 10 ml of pre-warmed complete growth medium. Disperse the medium by pipetting over the cell layer surface several times. Transfer the cells to a 50 mL conical tube and centrifuge then at 1000 rpm for 5 minutes. Resuspend the cell pellet in 2 ml of pre-warmed complete growth medium, dissociate cell clumps by gently pipetting up and down. Determine the total number of cells and percent viability using the Counterstar Automated Cell Counter. Adjust the cell density to 1.2×105 cells/ml, dispense the diluted cells to the collagen-I coated 96 well plate. Incubate the 96 well plates at 37℃, 5%CO2 overnight.
ELISA plate blocking: The coating buffer was removed by flicking the plate over a sink, and the remaining drops were removed by patting the plate on a paper towel. The plates were washed in PBS/0.05%Tween 20 300 ul with three times, and then blocked in PBS/0.05%Tween 20/3%non-fat milk at room temperature for 1 h. The plates were washed in PBS/0.05%Tween 20 300 ul three times, the blocking buffer were removed by flicking the plate over a sink, the remaining drops were removed by patting the plate on a paper towel. The plates were sealed with Self-adhesive Sealing Film for microplate and stored at a 4 ℃refrigerator for future use.
In vitro mRNA transfection (Day 3)
Prepare TransIT-mRNA Reagent: mRNA Boost: RNA complexes. Warm TransIT-mRNA and mRNA Boost reagents to room temperature and vortex gently before using.
Distribute complexes to cells: AddReagent: mRNA Boost: RNA complex mixture drop-wise to different areas of the well. Gently rock plate for even distribution of complexes. Incubate for 4-48 hours. Harvest cells or supernatant and assay as required and storage in 4 ℃ refrigerator.
Binding of the prefusion RSV F proteins to prefusion-specific neutralizing antibodies tested in an ELISA (Day 4)
60 μl per well of titrated cell transfected supernatant and standard (RSV F protein, 2ug/ml start, 3-fold dilution, 6 points) were then added to the blocked ELISA plates and incubated at room temperature for 90 min. Plates were washed and 100 μl per well of Goat anti RSV, Polyclonal, HRP (1: 500) was added. After 60 min incubation at room temperature, plates were washed and 100 μl per well of TMB Solution was added. Plates were incubated at  room temperature for 5-15 min and the reaction was stopped by adding 100 μl per well of Stop Solution (0.5N sulfuric acid) for TMB ELISA. Plates were then read on microplate reader (Tecan/Spark) at 450 nm.
Tables 4-6 shows the results of ELISA essays, with 100 ng, 25 ng, 6.25 ng mRNA being transfected into the Hep-2 cells, respectively.
Table 4 D25 ELISA readout (100 ng mRNA transfected into Hep-2 cells)
Table 5 D25 ELISA readout (25 ng mRNA transfected into Hep-2 cells)

Table 6 D25 ELISA readout (6.25 ng mRNA transfected into Hep-2 cells)

Example 2 Immunogenicity and viral protection in cotton rats
mRNAs disclosed herein were administered to cotton rats at 2.5 μg and 25 μg. Female cotton rats (6-8 weeks of age) were divided into 12 groups of 5 animals. The cotton rats were immunized according to the schedule shown in Table 7 below. Groups 1-10 were immunized intramuscularly (IM) with 100 μL dose of the mRNA-LNP composition per animal; Group 11 was infected intranasally (IN) with 100 μL dose RSV/A2 virus at 105 plaque forming units (PFUs) per animal. Groups 12 were immunized intramuscularly with 100 μL saline per animal. On Day 56, the rats were challenged with an intranasal administration of 0.1 mL of 5.0 log10 RSV/A2. On day 61, the animals were sacrificed, the lung and nasal tissue was harvested for viral titration measurements.
Table 7 In vivo Immunization Schedule
RSV Neutralizing Antibody Assay (60%PRNT)
Heat inactivated sera samples were diluted 1: 10 with EMEM and serially diluted further 1: 4. Diluted serum samples were incubated with RSV/A2 or RSV/B (25-50 PFU) for 1 hour at room temperature and inoculated in duplicates onto confluent HEp-2 monolayers in 24 well plates. After one hour incubation at 37 ℃ in a 5%CO2 incubator, the wells were overlayed with 0.75%Methylcellulose medium. After 4 days of incubation, the overlays were removed, and the cells were fixed and stained with 0.1%crystal violet for one hour and then rinsed and air dried. The corresponding reciprocal neutralizing antibody titers were determined at the 60%reduction end-point of the virus control using the statistics program "plqrd. manual. entry" . The geometric means ± standard error for all animals in a group at a given time were calculated.
The titration results against RSV/A2 and RSV B on day 56 are shown in Fig. 1 and Fig. 2 respectively.
RSV/A2 Lung and nose viral titration
Lung and nose homogenates were clarified by centrifugation and diluted in EMEM. Confluent HEp-2 monolayers were infected in duplicates with diluted homogenates in 24 well plates. After one hour incubation at 37℃ in a 5%CO2 incubator, the wells were overlayed with 0.75%Methylcellulose medium. After 4 days of incubation, the overlay was removed, and the cells were fixed with 0.1%crystal violet stain for one hour and then rinsed and air dried. Plaques were counted and virus titer was expressed as plaque forming units per gram of tissue. Viral titers were calculated as geometric mean + standard error for all animals in a group at a given time.
The titration results of RSV lung viral load at day 61 is shown in Fig. 3. The titration results of RSV nose viral load at day 61 is shown in Fig. 4.
Example 3 Immunogenicity and viral protection in cotton rats
Certain mRNAs disclosed herein were administered to cotton rats at 2.5 μg and 0.25 μg. Cotton rats were immunized with essentially the same protocol as described in Example 2 according to the schedule shown in Table 8 below. FI-RSV refers to formalin-inactivated RSV vaccine.
Table 8 In vivo Immunization Schedule

RSV Neutralizing Antibody Assay (60%PRNT) was carried out according to essentially the same protocol as described in Example 2. The titration results against RSV/A2 and RSV B on day 35 are shown in Fig. 5 and Fig. 6, respectively.
RSV/A2 lung and nose titration was carried out according to essentially the same protocol as described in Example 2. The titration results of RSV lung viral load at day 61 is shown in Fig. 7. The titration results of RSV nose viral load on day 61 is shown in Fig. 8.
References
Altschul, Stephen F., et al. "Basic local alignment search tool. " Journal of molecular biology 215.3 (1990) : 403-410.
Batzer, Mark A., James E. Carlton, and Prescott L. Deininger. "Enhanced evolutionary PCR using oligonucleotides with inosine at the 3'-terminus. " Nucleic Acids Research 19.18 (1991) : 5081.
Ohtsuka, Eiko, et al. "An alternative approach to deoxyoligonucleotides as hybridization probes by insertion of deoxyinosine at ambiguous codon positions. " Journal of Biological Chemistry 260.5 (1985) : 2605-2608.
Rossolini, Gian Maria, et al. "Use of deoxyinosine-containing primers vs degenerate primers for polymerase chain reaction based on ambiguous sequence information. " Molecular and Cellular Probes 8.2 (1994) : 91-98.
McLellan JS, Ray WC, Peeples ME. Structure and function of respiratory syncytial virus surface glycoproteins. Curr Top Microbiol Immunol. 2013; 372: 83-104. doi: 10.1007/978-3-642-38919-1_4. PMID: 24362685; PMCID: PMC4211642.
Bolt G, PedersenBirkeslund HH. Cleavage of the respiratory syncytial virus fusion protein is required for its surface expression: role of furin. Virus Res. 2000; 68 (1) : 25–33.
Collins PL, Mottet G. Post-translational processing and oligomerization of the fusion glycoprotein of human respiratory syncytial virus. J Gen Virol. 1991; 72 (12) : 3095–3101.
Gonzalez-Reyes L, Ruiz-Arguello MB, Garcia-Barreno B, Calder L, Lopez JA, Albar JP, Skehel JJ, Wiley DC, Melero JA. Cleavage of the human respiratory syncytial virus fusion protein at two distinct sites is required for activation of membrane fusion. Proc Natl Acad Sci U S A. 2001; 98 (17) : 9859–9864.
Zimmer G, Budz L, Herrler G. Proteolytic activation of respiratory syncytial virus fusion protein. Cleavage at two furin consensus sequences. J Biol Chem. 2001a; 276 (34) : 31642–31650.
Gruber C, Levine S. Respiratory syncytial virus polypeptides. III. The envelope-associated proteins. J Gen Virol. 1983; 64 (4) : 825–832.
Day ND, Branigan PJ, Liu C, Gutshall LL, Luo J, Melero JA, Sarisky RT, Del Vecchio AM. Contribution of cysteine residues in the extracellular domain of the F protein of human respiratory syncytial virus to its function. Virol J. 2006; 3: 34.
Begona Ruiz-Arguello M, Gonzalez-Reyes L, Calder LJ, Palomo C, Martin D, Saiz MJ, Garcia-Barreno B, Skehel JJ, Melero JA. Effect of proteolytic processing at two distinct sites on shape and aggregation of an anchorless fusion protein of human respiratory syncytial virus and fate of the intervening segment. Virology. 2002; 298 (2) : 317–326.
Mejias A, Garcia-Maurino C, Rodriguez-Fernandez R, Peeples ME, Ramilo O (2017) Development and clinical applications of novel antibodies for prevention and treatment of respiratory syncytial virus infection. Vaccine 35: 496–502
Graham BS (2019) Immunological goals for respiratory syncytial virus vaccine development. Curr Opin Immunol 59: 57–64
McLellan JS, Chen M, Leung S, Graepel KW, Du X, Yang Y, Zhou T, Baxa U, Yasuda E, Beaumont T et al (2013) Structure of RSV fusion glycoprotein trimer bound to a prefusion-specific neutralizing antibody. Science 340: 1113–1117
Anderson LJ, Bingham P, Hierholzer JC. Neutralization of respiratory syncytial virus by individual and mixtures of F and G protein monoclonal antibodies. J Virol. 1988; 62 (11) : 4232–4238.
Walsh EE, Hruska J. Monoclonal antibodies to respiratory syncytial virus proteins: identification of the fusion protein. J Virol. 1983; 47 (1) : 171–177.
Costello HM, Ray WC, Chaiwatpongsakorn S, Peeples ME. Targeting RSV with vaccines and small molecule drugs. Infect Disord Drug Targets. 2012; 12 (2) : 110–128.
McLellan JS, Ray WC, Peeples ME. Structure and function of respiratory syncytial virus surface glycoproteins. Curr Top Microbiol Immunol. 2013; 372: 83-104. doi: 10.1007/978-3-642-38919-1_4. PMID: 24362685; PMCID: PMC4211642.
Decroly, E., Ferron, F., Lescar, J. et al. Conventional and unconventional mechanisms for capping viral mRNA. Nat Rev Microbiol 10, 51–65 (2012) . https: //doi. org/10.1038/nrmicro2675
Hou, X., Zaks, T., Langer, R. et al. Lipid nanoparticles for mRNA delivery. Nat Rev Mater 6, 1078–1094 (2021) . https: //doi. org/10.1038/s41578-021-00358-0.
Akbarzadeh A, Rezaei-Sadabady R, Davaran S, Joo SW, Zarghami N, Hanifehpour Y, Samiei M, Kouhi M, Nejati-Koshki K. Liposome: classification, preparation, and applications. Nanoscale Res Lett. 2013 Feb 22; 8 (1) : 102. doi: 10.1186/1556-276X-8-102. PMID: 23432972; PMCID: PMC3599573.
Chen, Jiaxuan, et al. "Amphiphilic Dendrimer Vectors for RNA Delivery: State-of-the-Art and Future Perspective. " Accounts of Materials Research (2022) .
Brito LA, Chan M, Shaw CA, Hekele A, Carsillo T, Schaefer M, Archer J, Seubert A, Otten GR, Beard CW, Dey AK, Lilja A, Valiante NM, Mason PW, Mandl CW, Barnett SW, Dormitzer PR, Ulmer JB, Singh M, O'Hagan DT, Geall AJ. A cationic nanoemulsion for the delivery of next-generation RNA vaccines. Mol Ther. 2014 Dec; 22 (12) : 2118-2129. doi: 10.1038/mt. 2014.133. Epub 2014 Jul 16. PMID: 25027661; PMCID: PMC4429691.
Jiang X, Abedi K, Shi J. Polymeric nanoparticles for RNA delivery. Reference Module in Materials Science and Materials Engineering. 2021: B978-0-12-822425-0.00017-8. doi: 10.1016/B978-0-12-822425-0.00017-8. Epub 2021 Nov 4. PMCID: PMC8568333.
Barclay TG, Day CM, Petrovsky N, Garg S. Review of polysaccharide particle-based functional drug delivery. Carbohydr Polym. 2019 Oct 1; 221: 94-112. doi: 10.1016/j. carbpol. 2019.05.067. Epub 2019 May 26. PMID: 31227171; PMCID: PMC6626612.
Baudin-Baillieu, Agnes, et al. "Nucleotide modifications in three functionally important regions of the Saccharomyces cerevisiae ribosome affect translation accuracy. " Nucleic acids research 37.22 (2009) : 7665-7677.
Lungwitz U., Breunig M., Blunk T., A. Polyethylenimine-based non-viral gene delivery systems. European Journal of Pharmaceutics and Biopharmaceutics. 2005; 60: 247–266.
Bodnar M, Hartmann JF, & Borbely J (2005) . Preparation and characterization of chitosan-based nanoparticles. Biomacromolecules, 6 (5) , 2521–2527.

Claims (21)

  1. A polypeptide comprising an amino acid sequence of any one of SEQ ID NOs: 26-28, 30-50.
  2. A polypeptide comprising an amino acid sequence with at least 90%identity to any one of SEQ ID NOs: 26-28, 30-50.
  3. A polynucleotide comprising a coding sequence that encodes the polypeptide of claim 1 or claim 2.
  4. A polynucleotide comprising a sequence of any one of SEQ ID NOs: 1-25.
  5. The polynucleotide of claim 3 or claim 4, wherein the polynucleotide is an mRNA sequence.
  6. The polynucleotide of any one of claims 3 to 5, wherein the coding sequence is operably linked to a 5’ untranslated region (5’ UTR) and/or a 3’ untranslated region (3’ UTR) .
  7. The polynucleotide of any one of claims 3-6, further comprising a 5’ cap structure.
  8. The polynucleotide of any one of claims 3-7 further comprising a poly (A) tail at its 3’ end.
  9. The polynucleotide of any one of claims 3-8, wherein the polynucleotide comprises at least one modified nucleotide.
  10. The polynucleotide of any one of claims 3-9, wherein the polynucleotide comprises, from the 5’-end to the 3’-end, a 5’-cap structure, a 5’ UTR, the coding sequence, a 3’ UTR, and a poly (A) sequence.
  11. A vector comprising the polynucleotide in any one of claims 3-10.
  12. The vector of claim 11, wherein the vector is a viral vector or a plasmid.
  13. A composition comprising the polypeptide of claim 1 or claim 2.
  14. A composition comprising the polynucleotide of any one of claims 3-10.
  15. The composition of claim 14 further comprising a carrier selected from the group consisting of lipid nanoparticles, liposomes, cationic nanoemulsions, dendrimer-based lipid nanoparticles, cationic polymers, and polysaccharide particles.
  16. A kit comprising the polynucleotide of any one of claims 3-10 and a carrier selected from the group consisting of lipid nanoparticles, liposomes, cationic nanoemulsions, dendrimer-based lipid nanoparticles, cationic polymers, and polysaccharide particles.
  17. A method for generating an immune response to RSV F protein in a subject in need thereof, comprising administering to the subject an effective amount of the polypeptide of claim 1, or the polynucleotide of any one of claims 3-10.
  18. A method for preventing RSV infection in a subject in need thereof, comprising administering to the subject an effective amount of the polypeptide of claim 1, or the polynucleotide of any one of claims 3-10.
  19. A method for generating an immune response to RSV F protein in a subject in need thereof, comprising administering an effective amount of the composition of any one of claims 13-15.
  20. A method for preventing RSV infection in a subject in need thereof, comprising administering an effective amount of the composition of any one of claims 13-15.
  21. The method of any one of claims 17-20, wherein the RSV is RSV A or RSV B.
PCT/CN2023/124358 2022-10-13 2023-10-12 Rsv f protein variants and uses thereof WO2024078597A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNPCT/CN2022/125192 2022-10-13
CN2022125192 2022-10-13

Publications (1)

Publication Number Publication Date
WO2024078597A1 true WO2024078597A1 (en) 2024-04-18

Family

ID=90668873

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/124358 WO2024078597A1 (en) 2022-10-13 2023-10-12 Rsv f protein variants and uses thereof

Country Status (1)

Country Link
WO (1) WO2024078597A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110305727A1 (en) * 2009-07-15 2011-12-15 Novartis Ag Rsv f protein compositions and methods for making same
CN108738312A (en) * 2015-12-23 2018-11-02 辉瑞公司 Rsv f protein mutant
CN111405907A (en) * 2017-08-07 2020-07-10 考尔德生物科技有限公司 Conformationally stabilized RSV pre-fusion F proteins
CN111655715A (en) * 2018-01-29 2020-09-11 默沙东公司 Stabilized RSV F proteins and uses thereof
CN112512567A (en) * 2018-04-03 2021-03-16 赛诺菲 Antigenic respiratory syncytial virus polypeptides

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110305727A1 (en) * 2009-07-15 2011-12-15 Novartis Ag Rsv f protein compositions and methods for making same
CN105214080A (en) * 2009-07-15 2016-01-06 诺华股份有限公司 RSV F protein compositions and its manufacture method
CN108738312A (en) * 2015-12-23 2018-11-02 辉瑞公司 Rsv f protein mutant
CN111405907A (en) * 2017-08-07 2020-07-10 考尔德生物科技有限公司 Conformationally stabilized RSV pre-fusion F proteins
CN111655715A (en) * 2018-01-29 2020-09-11 默沙东公司 Stabilized RSV F proteins and uses thereof
US20210300971A1 (en) * 2018-01-29 2021-09-30 Merck Sharp & Dohme Corp. Stabilized rsv f proteins and uses thereof
CN112512567A (en) * 2018-04-03 2021-03-16 赛诺菲 Antigenic respiratory syncytial virus polypeptides

Similar Documents

Publication Publication Date Title
US20220047521A1 (en) Drug Delivery Particle and Method for Producing the Same
RU2723328C2 (en) Vaccine against respiratory syncytial virus (rsv)
CN109310751A (en) Wide spectrum influenza virus vaccine
TW202233232A (en) Lipid nanoparticles for delivering mrna vaccines
US20230108926A1 (en) COMPOSITION AND METHOD OF mRNA VACCINES AGAINST NOVEL CORONAVIRUS INFECTION
WO2017044507A2 (en) Sirna/nanoparticle formulations for treatment of middle-east respiratory syndrome coronaviral infection
Sawaengsak et al. Intranasal chitosan-DNA vaccines that protect across influenza virus subtypes
CN113186173A (en) Novel coronavirus pneumonia vaccine based on attenuated influenza virus vector
Rodriguez-Gascon et al. Vaginal gene therapy
US20100068226A1 (en) Polynucleotides and Uses Thereof
US20230043128A1 (en) Multivalent influenza vaccines
CA3194325A1 (en) Lipid nanoparticles for delivering mrna vaccines
CN115725612A (en) RNA, RNA combination and application thereof, and multivalent monkeypox vaccine
CN117098551A (en) Influenza virus encoding truncated NS1 protein and SARS-COV receptor binding domain
JP2024509938A (en) Vaccine composition for preventing SARS-CoV-2
EP4138908A1 (en) A vaccine against sars-cov-2 and preparation thereof
WO2024078597A1 (en) Rsv f protein variants and uses thereof
CN116410992A (en) mRNA and vaccine for preventing and/or treating novel coronavirus, and preparation method and application thereof
CN108431214A (en) Human rotavirus G9P [6] strains and the purposes as vaccine
TW202330922A (en) Compositions and methods of ribonucleic acid respiratory syncytial virus (rsv) vaccines
CN112424367A (en) Nucleic acid molecules and bifunctional peptides having antiviral and delivery activities, compositions and methods thereof
WO2023044505A2 (en) Piv5-based coronavirus vaccines and methods of use thereof
CN115960180A (en) 2019-nCoV S protein mutant and genetically engineered mRNA and vaccine composition thereof
WO2024008192A2 (en) Modified self-replicating mrna
US12029786B2 (en) Composition and method of mRNA vaccines against novel coronavirus infection

Legal Events

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

Ref document number: 23876796

Country of ref document: EP

Kind code of ref document: A1