[go: up one dir, main page]

WO2025126071A1 - Rna molecules - Google Patents

Rna molecules Download PDF

Info

Publication number
WO2025126071A1
WO2025126071A1 PCT/IB2024/062510 IB2024062510W WO2025126071A1 WO 2025126071 A1 WO2025126071 A1 WO 2025126071A1 IB 2024062510 W IB2024062510 W IB 2024062510W WO 2025126071 A1 WO2025126071 A1 WO 2025126071A1
Authority
WO
WIPO (PCT)
Prior art keywords
aspects
rna
rna molecule
rsv
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/IB2024/062510
Other languages
French (fr)
Inventor
Eric Matthew Bennett
Amendra Fernando HEWA DEWAGE
Laura LIN LOHSE
Mohan MADDUR SATHYANARAYANA
Kena Anne SWANSON
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pfizer Corp Belgium
Pfizer Corp SRL
Original Assignee
Pfizer Corp Belgium
Pfizer Corp SRL
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 Pfizer Corp Belgium, Pfizer Corp SRL filed Critical Pfizer Corp Belgium
Publication of WO2025126071A1 publication Critical patent/WO2025126071A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • A61K2039/555Medicinal 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
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • 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

  • RNA genome consists of a single-stranded, negative-sense RNA molecule that encodes 11 proteins, including nine structural proteins (three glycoproteins and six internal proteins) and two non-structural proteins.
  • the structural proteins include three transmembrane surface glycoproteins: the attachment protein G, fusion protein F, and the small hydrophobic SH protein.
  • the mature F glycoprotein has three general domains: ectodomain (ED), transmembrane domain (TM), and a cytoplasmic tail (CT). CT contains a single palmitoylated cysteine residue.
  • the F glycoprotein of human RSV is initially translated from the mRNA as a single 574- amino acid polypeptide precursor (referred to “F0” or “F0 precursor”), which contains a signal peptide sequence (amino acids 1-25) at the N-terminus. Upon translation the signal peptide is removed by a signal peptidase in the endoplasmic reticulum.
  • F0 574- amino acid polypeptide precursor
  • the remaining portion of the F0 precursor may be further cleaved at two polybasic sites (a.a.109/110 and 136/137) by cellular proteases (in particular furin), removing a 27-amino acid intervening sequence designated pep27 (amino acids 110-136) and generating two linked fragments designated F1 (C-terminal portion; amino acids 137-574) and F2 (N-terminal portion; amino acids 26-109).
  • F1 contains a hydrophobic fusion peptide at its N-terminus and two heptad-repeat regions (HRA and HRB). HRA is near the fusion peptide, and HRB is near the TM domain.
  • the 5′ cap moiety is (3′OMe) - m 2 7,3′-O G(5’)ppp (m 1 2′-O )ApG.
  • the present disclosure further provides for RNA molecules and RNA-LNPs that include a 3’ poly-A tail.
  • the poly-A tail comprises a sequence having SEQ ID NO: 22 or 26.
  • the RNA molecule includes a 5’ UTR and a 3’ UTR.
  • the RNA molecule includes a 5’ cap, 5’ UTR, and 3’ UTR.
  • the RNA molecule includes a 5’ cap, 5’ UTR, 3’ UTR, and poly-A tail.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 15, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 16, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 15, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 16, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 15, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 16, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RSV ORF further comprises a stop codon described herein.
  • the poly-A tail length may contain +/-1 A or +/-2 A.
  • each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1 ⁇ ) (e.g., modified RNA; modRNA).
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 633, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 634, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 633, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 634, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 633, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 634, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RSV ORF further comprises a stop codon described herein.
  • the poly-A tail length may contain +/-1 A or +/-2 A.
  • each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1 ⁇ ) (e.g., modified RNA; modRNA).
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 635, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 636, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 635, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 636, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 635, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 636, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RSV ORF further comprises a stop codon described herein.
  • the poly-A tail length may contain +/-1 A or +/-2 A.
  • each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1 ⁇ ) (e.g., modified RNA; modRNA).
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 637, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 638, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 637, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 638, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 637, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 638, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22.
  • the RSV ORF further comprises a stop codon described herein.
  • the poly-A tail length may contain +/-1 A or +/-2 A.
  • each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1 ⁇ ) (e.g., modified RNA; modRNA).
  • m1 ⁇ N1-methylpseudouridine
  • the present disclosure provides for RNA molecules encoding RSV F proteins transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 11-14, 639 and 640.
  • the present disclosure further provides for RNA molecules encoding RSV F proteins comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 17-20, 641 and 642.
  • each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1 ⁇ ) (e.g., modified RNA; modRNA).
  • m1 ⁇ N1-methylpseudouridine
  • modRNA modified RNA
  • the present disclosure further provides for RNA molecules that include at least one open reading frame that was generated from codon-optimized DNA.
  • the open reading frame comprises a G/C content of at least, at most, exactly, or between (inclusive or exclusive) any two of 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, or 75%, e.g., at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, is or is about 50% to 75%, or is or is about 55% to 70%.
  • the G/C content is or is about 58%, is or is about 66%, or is about 62%.
  • the present disclosure further provides for RNA molecules that encode RSV polypeptides that localizes in the cellular membrane, localizes in the Golgi and/or are anchored in the membrane and are secreted.
  • the present disclosure further provides RNA molecules comprising stabilized RNA.
  • the present disclosure further provides for RNA molecules that are messenger-RNA (mRNA) or self- replicating RNA (saRNA).
  • the RNA comprises at least one modified nucleotide (modRNA).
  • modRNA modified nucleotide
  • the present disclosure further provides for RNA molecules that include RNA having at least one modified nucleotide (e.g., modified RNA; modRNA).
  • the modified nucleotide is pseudouridine, N1-methylpseudouridine, N1-ethylpseudouridine, 2-thiouridine, 4′- thiouridine, 5-methylcytosine, 5-methyluridine, 2-thio-1-methyl-1-deaza-pseudouridine, 2-thio-1- methyl-pseudouridine, 2-thio-5-aza-uridine, 2-thio-dihydropseudouridine, 2-thio-dihydrouridine, 2-thio-pseudouridine, 4-methoxy-2-thio-pseudouridine, 4-methoxy-pseudouridine, 4-thio-1- methyl-pseudouridine, 4-thio-pseudouridine, 5-aza-uridine, dihydropseudouridine, 5- methoxyuridine, or 2′-O-methyl uridine.
  • the modified nucleotide is N1- methylpseudouridine (m1 ⁇ ). In some aspects, 1, 2, 3, 4, 5, or more of the foregoing modified nucleotides can be excluded from the RNA molecule.
  • the present disclosure further provides for immunogenic compositions including the RNA molecules described herein.
  • the RNA molecules may be formulated in, encapsulated in, complexed with, bound to or adsorbed on a lipid nanoparticle (LNP) (e.g., RSV RNA-LNPs) in such immunogenic compositions.
  • the immunogenic composition comprises a RSV molecule encoding an F protein as described herein which is of RSV subtype A.
  • the immunogenic composition comprises a RSV molecule encoding an F protein as described herein which is of RSV subtype B.
  • the immunogenic composition is a bivalent immunogenic composition comprising a first RSV molecule encoding an F protein as described herein which is of RSV subtype A and a second RSV molecule encoding an F protein as described herein which is of RSV subtype B.
  • the bivalent immunogenic composition may be a (i) "pre-mix” formulation wherein two or more RNA polynucleotides are combined in equal ratio followed by co-formulation into LNPs, or (ii) a "post-mix” formulation wherein each RNA polynucleotide is individually formulated into LNPs and the two or more LNPs are combined either in equal ratio or not in equal ratio.
  • the ratio of the first RSV molecule to the second RSV molecule in the bivalent immunogenic composition is 1:1.
  • the lipid nanoparticle includes at least one of a cationic lipid, a polymer conjugated lipid (e.g., a PEGylated lipid), and at least one structural lipid (e.g., a neutral lipid and a steroid or steroid analog). In some aspects, 1, 2, 3, or more of the foregoing lipids may be excluded from the lipid nanoparticle. In some aspects, the lipid nanoparticle includes a cationic lipid. In some aspects, the cationic lipid is selected from any cationic lipid described in section B(i) set forth herein.
  • the cationic lipid is (4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate) (ALC-0315).
  • the lipid nanoparticle includes a polymer conjugated lipid.
  • the cationic lipid is selected from any cationic lipid described in section B(ii) set forth herein.
  • the lipid nanoparticle includes a PEGylated lipid, also referred to as a PEG-lipid.
  • the PEGylated lipid is PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramides (e.g., PEG-CerC14 or PEG-CerC20), PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, 2- [(polyethylene glycol)-2000]-N,N-ditetradecylacetamide, glycol-lipids including PEG-c-DOMG, PEG-c-DMA, PEG-s-DMG, N-[(methoxy polyethylene glycol)2000)carbamoyl]-1,2- dimyristyloxlpropyl-3-amine (PEG-c-DMA), and PEG-2000-DMG, PEGylated diacylglycerol (PEG-DAG) such as 1-(monomethoxy-polyethyleneglycol)-2,
  • 1, 2, 3, 4, 5, or more of the foregoing PEGylated lipids can be excluded from the RNA molecule.
  • the PEGylated lipid is 2-[(polyethylene glycol)-2000]-N,N-ditetradecylacetamide (ALC- 0159).
  • the lipid nanoparticle includes at least one structural lipid, such as a neutral lipid.
  • the neutral lipid is 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl-phosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyl-oleoyl-phosphatidylethanolamine (POPE), dioleoyl- phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane-1-carboxylate (DOPE- mal), dipalmitoylphosphatidylethanolamine (DPPE), dimyristoylphosphoethanolamine (DM)
  • the lipid nanoparticle has a mean diameter of about 1 to about 500 nm, e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 1 nm, 10 nm, 20 nm, 30 nm, 40 nm, 50 nm, 60 nm, 70 nm, 80 nm, 90 nm, 100 nm, 110 nm, 120 nm, 130 nm, 140 nm, 150 nm, 160 nm, 170 nm, 180 nm, 190 nm, 200 nm, 210 nm, 220 nm, 230 nm, 240 nm, 250 nm, 260 nm, 270 nm, 280 nm, 290 nm, 300 nm, 310 nm, 320 nm, 330 nm, 340 nm, 350 nm, 360 nm, 370 nm,
  • the lipid nanoparticle has a mean diameter between 40 nm to 120 nm. In another aspect, the lipid nanoparticle has a mean diameter between about 50 nm to about 90 nm. In another aspect, the lipid nanoparticle has a mean diameter between about 55 nm to about 65 nm.
  • the RNA-LNP immunogenic composition is a liquid RNA-LNP composition comprising an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition comprising a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.80, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.90, 0.91, 0.92, 0.93, 0.94, or 0.95 mg/mL), a PEGylated lipid at a concentration of or of about
  • the liquid composition further comprises a buffer composition comprising a first buffer at a concentration of or of about 0.1 to 0.3 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, or 0.30 mg/mL), a second buffer at a concentration of or of about 1.25 to 1.4 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 1.25, 1.26, 1.27, 1.28, 1.29, 1.30, 1.31, 1.32, 1.33, 1.34, 1.35, 1.36, 1.37, 1.38, 1.39, or 1.40 mg/mL), and a stabilizing agent at a concentration of or of about 95 to 110 mg/mL (e.g.,
  • the liquid RNA-LNP immunogenic composition comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition comprising ((4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2- hexyldecanoate) (ALC-0315) at a concentration of or of about 0.8 to 0.95 mg/m
  • the liquid composition further comprises a Tris buffer composition comprising tromethamine at a concentration of or of about 0.1 to 0.3 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, or 0.30 mg/mL) and Tris hydrochloride (HCl) at a concentration of or of about 1.25 to 1.4 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 1.25, 1.26, 1.27, 1.28, 1.29, 1.30, 1.31, 1.32, 1.33, 1.34, 1.35, 1.36, 1.37, 1.38, 1.39, or 1.40 mg/mL), and sucrose at a concentration of or of about 95 to 110 mg/m
  • the liquid RNA-LNP immunogenic composition comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in a LNP, and further comprising of or of about 5 to 15 mM Tris buffer(e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 mM) and of or of about 200 to 400 mM sucrose (e.
  • the RNA-LNP immunogenic composition is a lyophilized (reconstituted) RNA-LNP composition comprising an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition comprising a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.80, 0.
  • the lyophilized composition further comprises a first buffer at a concentration of or of about 0.01 and 0.15 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, or 0.15 mg/mL), a second buffer at a concentration of or of about 0.5 and 0.65 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.50, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.60, 0.61, 0.62, 0.63, 0.64, or 0.65 mg/mL), a stabilizing agent at a concentration of or of about 35 to 50 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 35, 36
  • the lyophilized compositions are reconstituted in or in about 0.6 to 0.75 mL of the salt diluent (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.60, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.70, 0.71, 0.72, 0.73, 0.74, or 0.75 mL).
  • Concentrations in the lyophilized RNA-LNP composition are determined post-reconstitution. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing elements can be excluded from the lyophilized RNA-LNP composition.
  • a lyophilized (reconstituted) RNA-LNP composition comprises an RNA polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition of ALC-0315 at a concentration of or of about 0.8 to 0.95 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.80, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.90, 0.91, 0.92, 0.93, 0.94, or 0.
  • the lyophilized compositions are reconstituted in or in about 0.6 to 0.75 mL of sodium chloride (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.60, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.70, 0.71, 0.72, 0.73, 0.74, or 0.75 mL).
  • Concentrations in the lyophilized RNA-LNP composition are determined post-reconstitution. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing elements can be excluded from the lyophilized RNA-LNP composition.
  • RNA-LNP composition is a liquid composition.
  • the present disclosure provides for RNA molecules, RNA-LNPs and immunogenic compositions that may be administered to a subject at a dose per administration of at least, at most, exactly, or between (inclusive or exclusive) any two of 1 ⁇ g, 15 ⁇ g, 30 ⁇ g, 45 ⁇ g, 60 ⁇ g, 75 ⁇ g, 90 ⁇ g, 100 ⁇ g or higher of RSV RNA encapsulated in LNP.
  • RNA molecules, RNA-LNPs and immunogenic compositions that may be administered in a single dose.
  • RNA molecules, RNA-LNPs and immunogenic compositions that may be administered twice (e.g., Day 0 and on or about Day 7, Day 0 and on or about Day 14, Day 0 and on or about Day 21, Day 0 and on or about Day 28, Day 0 and on or about Day 60, Day 0 and on or about Day 90, Day 0 and on or about Day 120, Day 0 and on or about Day 150, Day 0 and on or about Day 180, Day 0 and on or about 1 month later, Day 0 and on or about 2 months later, Day 0 and on or about 3 months later, Day 0 and on or about 6 months later, Day 0 and on or about 9 months later, Day 0 and on or about 12 months later, Day 0 and on or about 18 months later, Day 0 and on or about 2 years later, Day 0 and on or about 5 years later, or Day 0 and on or about 10 years later).
  • twice e.g., Day 0 and on or about Day 7, Day 0 and on or about Day 14, Day 0 and
  • the present disclosure further provides for RNA molecules, RNA-LNPs and immunogenic compositions that may be administered twice at Day 0 and on or about 2 months later.
  • the present disclosure further provides for RNA molecules, RNA-LNPs and immunogenic compositions that may be administered twice at Day 0 and on or about 6 months later.
  • the present disclosure further provides for RNA molecules, RNA-LNPs and immunogenic compositions that may be administered three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more times.
  • periodic boosters at intervals of 1-5 years may be desirable to maintain protective levels of the antibodies.
  • the present disclosure further provides for administration of at least one booster dose.
  • the present disclosure provides for a method of inducing an immune response against RSV in a subject, including administering to the subject an effective amount of an RNA molecule, RNA- LNP and/or immunogenic composition described herein.
  • the present disclosure further provides for the use of an RNA molecule, RNA-LNP and/or immunogenic composition described herein in the manufacture of a medicament for use in inducing an immune response against RSV in a subject.
  • the present disclosure provides for a method of inducing an immune response against RSV in a subject, including administering to the subject an effective amount of an RNA molecule and/ or RNA-LNP that includes at least one open reading frame encoding a RSV polypeptide or immunogenic composition described herein.
  • the present disclosure further provides for the use of an RNA molecule and/or RNA-LNP that includes at least one open reading frame encoding a RSV polypeptide or immunogenic composition described herein in the manufacture of a medicament for use in inducing an immune response against RSV in a subject.
  • the present disclosure provides for a method of inducing an immune response against RSV in a subject, including administering to the subject an effective amount of an RNA molecule and/or RNA-LNP that includes at least one open reading frame encoding a polypeptide of a gene of interest or composition described herein.
  • the present disclosure further provides for the use of an RNA molecule and/or RNA-LNP that includes at least one open reading frame encoding a polypeptide of a gene of interest or composition described herein in the manufacture of a medicament for use in inducing an immune response against RSV in a subject.
  • the present disclosure provides for a method of preventing, treating, and/or ameliorating an infection, disease, or condition in a subject, including administering to a subject an effective amount of an RNA molecule, RNA-LNP and/or immunogenic composition described herein.
  • the present disclosure further provides for the use of an RNA molecule, RNA-LNP and/or immunogenic composition described herein in the manufacture of a medicament for use in preventing, treating, and/or ameliorating an infection, disease, or condition in a subject.
  • the infection, disease, or condition is associated with RSV.
  • the infection, disease, or condition is acute lower respiratory infection (ALRI), including pneumonia and bronchitis.
  • the infection, disease, or condition is acute lower respiratory infection (ALRI), including pneumonia and bronchitis.
  • ARI acute lower respiratory infection
  • the present disclosure provides for a method of preventing, treating, and/or ameliorating an infection, disease, or condition in a subject, including administering to a subject an effective amount of an RNA molecule and/or RNA-LNP that includes at least one open reading frame encoding a RSV polypeptide or immunogenic composition described herein.
  • the present disclosure further provides for the use of an RNA molecule and/or RNA-LNP that includes at least one open reading frame encoding a RSV polypeptide or immunogenic composition described herein in the manufacture of a medicament for use in preventing, treating, and/or ameliorating an infection, disease, or condition in a subject.
  • the infection, disease, or condition is associated with RSV.
  • the infection, disease, or condition is acute lower respiratory infection (ALRI), including pneumonia and bronchitis.
  • the infection, disease, or condition is acute lower respiratory infection (ALRI), including pneumonia and bronchitis.
  • ARI acute lower respiratory infection
  • the present disclosure further provides for a method of preventing, treating, and/or ameliorating an infection, disease, or condition in a subject, including administering to a subject an effective amount of RNA molecules and/or RNA-LNPs that include at least one open reading frame encoding a polypeptide of a gene of interest or immunogenic compositions described herein.
  • RNA molecules and/or RNA-LNPs that include at least one open reading frame encoding a polypeptide of a gene of interest or immunogenic compositions described herein in the manufacture of a medicament for use in preventing, treating, and/or ameliorating an infection, disease, or condition in a subject.
  • the infection, disease, or condition is associated with the gene of interest.
  • the subject is at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months of age, or 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, or more years of age.
  • RNA molecule comprises at least one open reading frame encoding a respiratory syncytial virus (RSV) fusion protein F (F) polypeptide and a 5’ untranslated region (5’ UTR), wherein the 5’ UTR comprises a nucleic acid sequence at least 90% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336-344, 356-358, 360, 362-367, 370-375, and 378-396.
  • the 5’ UTR comprises a nucleic acid sequence at least 92% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336, 341, 356-358, 372-375, and 378-396.
  • variant includes all mutants, splice variants, post-translationally modified variants, conformations, isoforms, allelic variants, species variants, and species homologs, in particular those which are naturally occurring.
  • variant includes, in particular, fragments of an amino acid or nucleic acid sequence. Changes may be introduced by mutation into a nucleic acid, thereby leading to changes in the amino acid sequence of a polypeptide (e.g., an antigen or antibody or antibody derivative) that it encodes. Mutations may be introduced using any technique known in the art. In one aspect, one or more particular amino acid residues are changed using, for example, a site-directed mutagenesis protocol.
  • postfusion conformation refers to a structural conformation adopted by the RSV F protein that is not specifically bound by D25, AM22, or AM14.
  • Native F protein adopts the postfusion conformation subsequent to the fusion of the virus envelope with the host cellular membrane.
  • RSV F protein may also assume the postfusion conformation outside the context of a fusion event, for example, under stress conditions such as heat and low osmolality, when extracted from a membrane, when expressed as an ectodomain, or upon storage.
  • the term “AM14” refers to an antibody described in WO 2008/147196 A2, which is hereby incorporated by reference in its entirety.
  • the term “AM22” refers to an antibody described in WO 2011/043643 A1, which is hereby incorporated by reference in its entirety.
  • the term “D25” refers to an antibody described in WO 2008/147196 A2, which is hereby incorporated herein by reference in its entirety.
  • the RSV F protein is an RSV F protein of subtype A.
  • the RSV F protein is an RSV F protein of subtype B.
  • the terms “subtype” and “subgroup” are used interchangeably.
  • strain refers to a specific isolate within each subtype or subgroup.
  • the RSV F protein is a mutant of wild type RSV F protein.
  • the RSV F protein is a mutant of wild type RSV F protein of subtype A.
  • the RSV F protein mutant comprises a cavity filling mutation selected from the group consisting of: (1) substitution of S at positions 55, 62, 155, 190, or 290 with I, Y, L, H, or M; (2) substitution of T at position 54, 58, 189, 219, or 397 with I, Y, L, H, or M; (3) substitution of G at position 151 with A or H; (4) substitution of A at position 147 or 298 with I, L, H, or M; (5) substitution of V at position 164, 187, 192, 207, 220, 296, 300, or 495 with I, Y, H; and (6) substitution of R at position 106 with W.
  • the RSV F protein mutant comprises at least one cavity filling mutation selected from the group consisting of: T54H, S190I, and V296I.
  • the RSV F protein mutants comprise electrostatic mutations, which decrease ionic repulsion or increase ionic attraction between resides in a protein that are proximate to each other in the folded structure.
  • the RSV F protein mutant includes an electrostatic substitution that reduces repulsive ionic interactions or increases attractive ionic interactions with acidic residues of Glu487 and Asp489 from another protomer of RSV F trimer.
  • the RSV F protein mutant comprises an electrostatic mutation selected from the group consisting of: (1) substitution of E at position 82, 92, or 487 by D, F, Q, T, S, L, or H; (2) substitution of K at position 315, 394, or 399 by F, M, R, S, L, I, Q, or T; (3) substitution of D at position 392, 486, or 489 by H, S, N, T, or P; and (4) substitution of R at position 106 or 339 by F, Q, N, or W.
  • the RSV F protein mutants comprise a combination of two or more different types of mutations selected from engineered disulfide mutations, cavity filling mutations, and electrostatic mutations.
  • the RSV F protein mutants comprise a combination of mutations relative to the corresponding wild-type RSV F protein, wherein the combination of mutations is selected from the group consisting of: (1) combination of A103C, I148C, S190I, and D486S; (2) combination of T54H S55C L188C D486S; (3) combination of T54H, A103C, I148C, S190I, V296I, and D486S; (4) combination of T54H, S55C, L142C, L188C, V296I, and N371C; (5) combination of S55C, L188C, and D486S; (6) combination of T54H, S55C, L188C, and S190I; (7) combination of S55C, L188C, S190I, and D486S; (8) combination of T54H, S55C, L188C, S190I, and D486S; (9) combination of S155C, S190I, S290C, and D486S;
  • the RSV F protein is of subtype A and comprises the mutations S155C, S190F, S290C, and V207L. In some embodiments, the RSV F protein is of subtype B and comprises the mutations S155C, S190F, S290C, and V207L. In some embodiments, the RSV F protein is of subtype A and comprises the mutations S155C, S190F, and S290C. In some embodiments, the RSV F protein is of subtype B and comprises the mutations S155C, S190F, and S290C. In some embodiments, the RSV F protein is of subtype A and comprises the mutations A103C, I148C, S190I, and D486S.
  • the RSV F protein is of subtype B and comprises the mutations A103C, I148C, S190I, and D486S. In some embodiments, the RSV F protein is of subtype A and comprises the mutations T54H, A103C, I148C, S190I, and D486S. In some embodiments, the RSV F protein is of subtype B and comprises the mutations T54H, A103C, I148C, S190I, and D486S. In some embodiments, the RSV F protein is of subtype A and comprises the mutations T54H, S55C, L188C, and D486S.
  • the RSV F protein is of subtype B and comprises the mutations T54H, S55C, L188C, and D486S.
  • a person of ordinary skill in the art can easily compare amino acid positions between different native RSV F sequences to identify corresponding RSV F amino acid positions between different RSV strains and subtypes. For example, across nearly all identified native RSV F0 precursor proteins, the furin cleavage sites fall in the same amino acid positions.
  • the conservation of native RSV F protein sequences across strains and subtypes allows use of a reference RSV F sequence for comparison of amino acids at particular positions in the RSV F protein.
  • the RSV F protein amino acid positions are given with reference to the amino acid sequence of the full length native F precursor polypeptide of the RSV A2 strain; corresponding to GenInfo Identifier GI 138251 and Swiss Prot identifier P03420 (SEQ ID NO: 1).
  • the RSV F protein is in the mature form of the RSV F protein, which comprises two separate polypeptide chains, namely the F1 polypeptide and F2 polypeptide.
  • the F2 polypeptide is linked to the F1 polypeptide by one or two disulfide bonds to form a F2/F1 heterodimer.
  • the RSV F mutants are in the form a single chain protein, wherein the F2 polypeptide is linked to the F1 polypeptide by a peptide bond or peptide linker.
  • Any suitable peptide linkers for joining two polypeptide chains together may be used. Examples of such linkers include G, GG, GGG, GS, and SAIG linker sequences.
  • the linker may also be the full length pep27 sequence or a fragment thereof, which full length pep27 sequence corresponds to amino acids at positions 110-136 of SEQ ID NO:1.
  • the F1 polypeptide chain of the mutant may be of the same length as the full length F1 polypeptide of the corresponding wild-type RSV F protein; however, it may also have deletions, such as deletions of 1 up to 60 amino acid residues from the C-terminus of the full-length F1 polypeptide.
  • a full-length F1 polypeptide of the RSV F mutants corresponds to amino acid positions 137-574 of the native RSV F0 precursor (SEQ ID NO: 1), and includes (from N- to C- terminus) an extracellular region (residues 137-524), a transmembrane domain (“TM”) (residues 525-550), and a cytoplasmic domain (“CT”) (residues 551-574).
  • amino acid residues 514 onwards in a native F1 polypeptide sequence are optional sequences in a F1 polypeptide of the RSV F protein to be included in the immunogenic composition provided herein, and therefore may be absent from the F1 polypeptide of the mutant.
  • the F1 polypeptide of the RSV F mutants lacks the entire cytoplasmic domain.
  • the F1 polypeptide lacks the cytoplasmic domain and a portion of or all entire transmembrane domain.
  • the mutant comprises a F1 polypeptide wherein the amino acid residues from position 510, 511, 512, 513, 514, 515, 520, 525, or 530 through 574 are absent.
  • amino acids 514 through 574 can be absent.
  • amino acid residues 514 through 574 are absent from the F1 polypeptide of the mutant.
  • the F1 polypeptide of the RSV F mutants comprises or consists of amino acid residues 137-513 of a native F0 polypeptide sequence (SEQ ID NO: 1) or any of alternative F0 precursor sequence such as those disclosed in SEQ ID NOs: 1, 2, 4, 6, and 81-270 of WO2017109629, which is hereby incorporated by reference in its entirety.
  • the F1 polypeptide and F2 polypeptide of the RSV F protein mutants to which one or more mutations are introduced can be from any wild-type RSV F proteins known in the art or discovered in the future, including, without limitations, the F protein amino acid sequence of RSV subtype A, and subtype B strains, including A2 Ontario and wholesome Aires, or any other subtype.
  • the RSV F mutant comprises a F1 and/or a F2 polypeptide from a RSV A virus, for example, a F1 and/or F2 polypeptide from a RSV F0 precursor protein set forth in any one of SEQ ID NOs: 1, 2, 4, 6, and 81-270 of WO2017109629, which sequences are hereby incorporated by reference in their entireties, to which one or more mutations are introduced.
  • the RSV F mutant comprises a F1 and/or a F2 polypeptide from a RSV B virus, for example, a F1 and/or F2 polypeptide from a RSV F0 precursor protein set forth in any one of SEQ ID NOs: 2 and 211-263 of WO2017/109629, which sequences are hereby incorporated by reference in their entireties, to which one or more mutations are introduced.
  • the RSV F mutant comprises a F1 and/or a F2 polypeptide from a RSV bovine virus, for example, a F1 and/or F2 polypeptide from a RSV F0 precursor protein set forth in any one of SEQ ID NOs: 264-270 of WO2017109629, which sequences are hereby incorporated by reference in their entireties, to which one or more mutations are introduced.
  • the term “F0 polypeptide” (F0) refers to the precursor polypeptide of the RSV F protein, which is composed of a signal polypeptide sequence, a F1 polypeptide sequence, a pep27 polypeptide sequence, and a F2 polypeptide sequence.
  • F2 polypeptide refers to the polypeptide chain of a mature RSV F protein. Native F2 includes approximately residues 26-109 of the RSV F0 precursor.
  • the term encompasses both native F2 polypeptides and F2 polypeptides including modifications (e.g., amino acid substitutions, insertions, or deletions) from the native sequence, for example, modifications designed to stabilize an RSV F protein mutant in a prefusion conformation or to enhance the immunogenicity of an RSV F protein mutant.
  • modifications e.g., amino acid substitutions, insertions, or deletions
  • the F2 polypeptide is linked to the F1 polypeptide by two disulfide bonds to form a F2-F1 heterodimer.
  • foldon or “foldon domain” refers to an amino acid sequence that is capable of forming trimers.
  • RNA molecule encodes an RSV F protein mutant as disclosed in WO2009/079796, WO2010/149745, WO2011/008974, WO2014/160463, WO2014/174018, WO2014/202570, WO2015/013551, WO2015/177312, WO2017/005848, WO2017/174564, WO2017/005844 and WO2018/109220.
  • the RSV F proteins disclosed in these references are hereby incorporated by reference in their entirety.
  • the term “respiratory syncytial virus” or “RSV” is not limited to any particular strain or variant.
  • the RNA molecule comprises an open reading frame encoding a RSV antigen.
  • the RSV antigen is a RSV polypeptide.
  • the RSV polypeptide is a RSV glycoprotein or a fragment or a variant thereof.
  • the RNA molecule encodes a RSV F protein.
  • the RSV polypeptide is a full-length RSV polypeptide.
  • the RSV polypeptide is a truncated RSV polypeptide. In some aspects, the RSV polypeptide is a variant of a RSV polypeptide. In some aspects, the RSV polypeptide is a fragment of a RSV polypeptide. In some aspects, the RSV polypeptide is a full-length RSV F protein. In some aspects, the RSV polypeptide is a truncated RSV F protein. In some aspects, the RSV polypeptide is a variant of a RSV F protein. In some aspects, the RSV polypeptide is a fragment of a RSV F protein. In some aspects, the RSV F protein comprises at least one mutation. In some aspects, the RSV F protein comprises at least two mutations.
  • the RSV F protein comprises at least three mutations. In some aspects, the RSV F protein comprises at least four mutations. In some aspects, the RSV F protein comprises 4 mutations. In some aspects, the RSV F protein comprises at least five mutations. In some aspects, the RNA molecule encodes a RSV F protein as set forth in Table 1. In some aspects, the RNA molecule encodes a RSV F protein comprising an amino acid sequence of any of SEQ ID NO: 1 to 6, or fragment or variant thereof.
  • RSV F polypeptide may have at least, at most, exactly, or between any two of 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to any of the amino acid sequences of Table 1, for example, any of SEQ ID NO: 1 to 6.
  • RSV F protein consists of any of the amino acid sequences of Table 1, for example, any of SEQ ID NO: 1 to 6.
  • the RNA molecule sequence is transcribed from a DNA nucleic acid sequence (DNA polynucleotide) of Table 2 and at least one untranslated region (UTR) selected from Table 4, provided that when the ORF is SEQ ID NO: 8 or 10 that the UTR does not consist of only SEQ ID NOs: 111 and 280.
  • the RNA molecule comprises an ORF transcribed from a nucleic acid sequence of any of SEQ ID NO: 7 to 10, or fragment or variant thereof.
  • the RNA molecule is transcribed from a nucleic acid having a sequence that may have at least, at most, exactly, or between any two of 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to any of the nucleic acid sequences of Table 2, for example, any of SEQ ID NO: 7 to 14.
  • the RNA molecule is transcribed from a nucleic acid having a sequence that consists of any of the nucleic acid sequences of Table 2, for example, any of SEQ ID NO:7 to 14.
  • the RNA molecule comprises an ORF encoding RSV F comprising an RNA nucleic acid sequence (RNA polynucleotide) of Table 3 and at least one untranslated region (UTR) selected from Table 5, provided that when the ORF is SEQ ID NO: 15 or 16 that the UTR does not consist of only SEQ ID NOs: 397 and 566.
  • the RNA molecule comprises a nucleic acid having a sequence of any of SEQ ID NO: 15 to 20, or fragment or variant thereof.
  • the RNA molecule comprises a nucleic acid having a sequence that may have at least, at most, exactly, or between any two of 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to any of the RNA nucleic acid sequences of Table 3, for example, any of SEQ ID NO: 15 to 20.
  • the RNA molecule comprises a nucleic acid having a sequence that consists of any of the RNA nucleic acid sequences of Table 3, for example, any of SEQ ID NO: 15 to 20.
  • the RNA molecule comprises stabilized RNA.
  • the RNA molecule comprises a nucleic acid sequence having at least one uridine replaced by N1- methylpseudouridine.
  • the RNA molecule comprises a sequence having all uridines replaced by N1-methylpseudouridine (designated as “ ⁇ ”).
  • the RNA molecule comprises an ORF comprising a nucleic acid sequence of any of SEQ ID NO: 15 or 16, wherein all uridines have been replaced by N1-methylpseudouridine (designated as “ ⁇ ”).
  • the RNA molecule comprises an open reading frame encoding a RSV F protein amino acid sequence that may be at least, at most, exactly, or between any two of 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to any of the RSV F protein sequences of SEQ ID NO: 1 to 6 (Table 1) or other RSV prefusion F proteins described herein.
  • the RNA molecule comprises an open reading frame encoding a RSV F protein amino acid sequence that consists of any of the RSV F protein sequences of SEQ ID NO: 1 to 6 (Table 1) or other RSV prefusion F protein described herein.
  • the RNA molecule comprises an open reading frame transcribed from a DNA nucleic acid sequence that may be at least, at most, exactly, or between any two of 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to any of the nucleic acid sequences of SEQ ID NO: 7 to 14 (Table 2) or other nucleic acid described herein.
  • the RNA molecule comprises an open reading frame transcribed from a DNA nucleic acid sequence that consists of any of the nucleic acid sequences of SEQ ID NO: 7 to 14 (Table 2) or other nucleic acid described herein.
  • the RNA molecule comprises an open reading frame encoding a RSV F, wherein the RNA nucleic acid sequence may be at least, at most, exactly, or between any two of 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to any of the nucleic acid sequences of SEQ ID NO: 15 to 20 (Table 3) or other nucleic acid described herein.
  • the RNA molecule comprises an open reading frame encoding a RSV F, wherein the RNA nucleic acid sequence consists of any of the nucleic acid sequences of SEQ ID NO: 15 to 20 (Table 3) or other nucleic acid described herein.
  • the RNA molecule comprises an ORF encoding a RSV F, wherein the nucleic acid sequence is any of SEQ ID NO: 15 to 20 (Table 3), wherein all uridines have been replaced by N1-methylpseudouridine (designated as “m1 ⁇ ”).
  • m1 ⁇ N1-methylpseudouridine
  • Coding RNA includes a functional RNA molecule that may be translated into a peptide or polypeptide.
  • the coding RNA molecule includes at least one open reading frame (ORF) coding for at least one peptide or polypeptide.
  • An open reading frame comprises a sequence of codons that is translatable into a peptide or protein.
  • the coding RNA molecule may include one (monocistronic), two (bicistronic) or more (multicistronic) ORFs, which may be a sequence of codons that is translatable into a polypeptide or protein of interest.
  • the coding RNA molecule may be a messenger RNA (mRNA) molecule, viral RNA molecule, or self-amplifying RNA molecule (saRNA, also referred to as a replicon).
  • mRNA messenger RNA
  • saRNA self-amplifying RNA molecule
  • the RNA molecule is an mRNA.
  • the RNA molecule of the present disclosure is an mRNA.
  • the RNA molecule is modRNA.
  • the RNA molecule is a saRNA.
  • the saRNA molecule may be a coding RNA molecule.
  • the RNA molecule may encode one polypeptide of interest or more, such as an antigen or more than one antigen, e.g., two, three, four, five, six, seven, eight, nine, ten or more polypeptides.
  • one RNA molecule may also encode more than one polypeptide of interest, such as an antigen, e.g., a bicistronic, or tricistronic RNA molecule that encodes different or identical antigens.
  • the sequence of the RNA molecule may be codon optimized or deoptimized for expression in a desired host, such as a human cell.
  • a gene of interest (e.g., an antigen) described herein is encoded by a coding sequence which is codon-optimized and/or the guanosine/cytidine (G/C) content of which is increased compared to wild type coding sequence.
  • G/C guanosine/cytidine
  • one or more sequence regions of the coding sequence are codon-optimized and/or increased in the G/C content compared to the corresponding sequence regions of the wild type coding sequence.
  • codon-optimization and/or increasing the G/C content does not change the sequence of the encoded amino acid sequence.
  • coding regions are codon-optimized for optimal expression in a subject to be treated using an RNA polynucleotide described herein. Codon-optimization is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNA molecules in cells.
  • the sequence of RNA may be modified such that codons for which frequently occurring tRNA molecules are available are inserted in place of “rare codons.”
  • G/C content of a coding region e.g., of a gene of interest sequence; open reading frame (ORF)
  • ORF open reading frame
  • the amino acid sequence encoded by the RNA is not modified compared to the amino acid sequence encoded by the wild type RNA. This modification of the RNA sequence is based on the fact that the sequence of any RNA region to be translated is important for efficient translation of that mRNA.
  • Sequences having an increased G (guanosine)/C (cytidine) content are more stable than sequences having an increased A (adenosine)/U (uridine) content.
  • the most favorable codons for the stability may be determined (so-called alternative codon usage).
  • alternative codon usage the amino acid to be encoded by the RNA, there are various possibilities for modification of the RNA sequence, compared to its wild type sequence.
  • G/C content of a coding region of an RNA described herein is increased by at least, at most, exactly, or between any two of 10%, 20%, 30%, 40%, 50%, 55%, or even more compared to the G/C content of a coding region of a wild type RNA.
  • the coding region of the RSV RNA described herein comprises a G/C content of at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, or about 80%. In some aspects, the coding region of the RSV RNA described herein comprises a G/C content of about 50% to 75%, about 55% to 70%, about 50% to 60%, about 60% to 70%, about 70% to 80%, about 50% to 55%, about 55% to 60%, about 60% to 65%, about 65% to 70%, about 70% to 75%, or about 75% to 80%.
  • the RNA molecule comprises a nucleic acid sequence having all uridines replaced by N1- methylpseudouridine and/or pseudouridine.
  • a modified nucleobase is a modified adenine.
  • RNA molecules may be further modified on the 2′O position by a 2′-O- methyltransferase, which results in the generation of a Cap 1 structure (m7Gppp [m2′- ⁇ ] N), which may further increase translation efficacy.
  • a Cap 1 structure comprises a guanosine nucleoside methylated at the 7-position of guanine (m7G) and a 2′O methylated first nucleotide in an RNA (2′OMeN 1 ).
  • a m7(3′OMeG)(5′)ppp(5′)(2′OMeN 1 ) Cap 1 structure comprises a second nucleotide, N 2 , which is a cap proximal nucleotide at position 2 and is chosen from A, G, C, or U (m7(3′OMeG)(5′)ppp(5′)(2′OmeN 1 )N 2 ).
  • N 2 is A.
  • N 2 is C.
  • N 2 is G.
  • N 2 is U.
  • 1, 2, 3, 4, 5, or more of the foregoing Cap 1 structures can be excluded from the RNA molecules disclosed herein.
  • a second nucleotide in a Cap 1 structure may comprise one or more modifications, e.g., methylation.
  • an RNA described herein comprises a Cap 2.
  • a Cap 1 structure comprising a second nucleotide comprising a 2′O methylation is a Cap 2 structure.
  • the 5’ cap is m 2 7,3′-O G(5’)ppp (m 1 2′-O )ApG having the structure set forth below: .
  • the RNA molecule may be enzymatically capped at the 5′ end using Vaccinia guanylyltransferase, guanosine triphosphate, and S-adenosyl-L-methionine to yield Cap 0 structure.
  • An inverted 7-methylguanosine cap is added via a 5′ to 5′ triphosphate bridge.
  • use of a 2′O-methyltransferase with Vaccinia guanylyltransferase yields the Cap 1 structure where, in addition to the Cap 0 structure, the 2′OH group is methylated on the penultimate nucleotide.
  • SAM S-adenosyl-L-methionine
  • 5′ cap structures are those which, among other things, have enhanced binding of cap-binding polypeptides, increased half-life, reduced susceptibility to 5′ endonucleases and/or reduced 5′ decapping, as compared to synthetic 5′ cap structures known in the art (or to a wild type, natural or physiological 5′ cap structure).
  • recombinant Vaccinia Virus Capping Enzyme and recombinant 2′ O- methyltransferase enzyme may create a canonical 5′-5′-triphosphate linkage between the 5′- terminal nucleotide of an mRNA and a guanine cap nucleotide wherein the cap guanine includes an N7 methylation and the 5′-terminal nucleotide of the mRNA includes a 2′-O-methyl.
  • Cap 1 structure Such a structure is termed the Cap 1 structure.
  • This cap results in a higher translational-competency and cellular stability and a reduced activation of cellular pro-inflammatory cytokines, as compared, e.g., to other 5′ cap analog structures known in the art.
  • a cap species may include one or more modified nucleosides and/or linker moieties.
  • a cap may include a guanine nucleotide and a guanine (G) nucleotide methylated at the 7 position joined by a triphosphate linkage at their 5′ positions, e.g., m7G(5′)ppp(5′)G, commonly written as m7GpppG.
  • G guanine nucleotide methylated at the 7 position joined by a triphosphate linkage at their 5′ positions, e.g., m7G(5′)ppp(5′)G, commonly written as m7GpppG.
  • a cap species may also be an anti-reverse cap analog.
  • a non- limiting list of possible cap species includes m7GpppG, m7Gpppm7G, m73′dGpppG, m27,O3′GpppG, m27,O3′GppppG, m27,O2′GppppG, m7Gpppm7G, m73′dGpppG, m27,O3′GpppG, m27,O3′GppppG, and m27,O2′GppppG.
  • 1, 2, 3, 4, 5, or more of the foregoing cap species can be excluded from the RNA molecules disclosed herein.
  • the 5′ terminal cap includes a cap analog
  • a 5′ terminal cap may include a guanine analog.
  • Exemplary guanine analogs include, but are not limited to, inosine, N1-methyl-guanosine, 2′-fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino- guanosine, LNA-guanosine, and 2-azido-guanosine.
  • 1, 2, 3, 4, 5, or more of the foregoing guanine analogs can be excluded from the cap structures disclosed herein.
  • the capping region may include a single cap or a series of nucleotides forming the cap.
  • the capping region may be from 1 to 10, e.g., 2-9, 3-8, 4-7, 1-5, 5- 10, or at least 2, or 10 or fewer nucleotides in length. In this aspect, the capping region is at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides in length. In some aspects, the cap is absent. In some aspects, the first and second operational regions may range from 3 to 40, e.g., 5-30, 10-20, 15, or at least 4, or 30 or fewer nucleotides in length and may comprise, in addition to a Start and/or Stop codon, one or more signal and/or restriction sequences.
  • the first and second operational regions are at least, at most, exactly, or between (inclusive or exclusive) any two of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 nucleotides in length and may comprise, in addition to a Start and/or Stop codon, one or more signal and/or restriction sequences.
  • 5′ cap structures include, but are not limited to, glyceryl, inverted deoxy abasic residue (moiety), 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
  • RNA molecules disclosed herein 1, 2, 3, 4, 5, or more of the foregoing 5′ cap structures can be excluded from the RNA molecules disclosed herein.
  • the RNA molecule of the present disclosure comprises at least one 5′ cap structure. In some aspects, the RNA molecule of the present disclosure does not comprise a 5′ cap structure.
  • the 5' UTR contains regulatory elements which regulate protein expression and the 3' UTR contains regulatory regions that post-transcriptionally influence gene expression and affect mRNA half-life.
  • the UTR increases protein synthesis.
  • the UTR may increase protein synthesis by increasing the time that the mRNA remains in translating polysomes (message stability) and/or the rate at which ribosomes initiate translation on the message (message translation efficiency). Accordingly, the UTR sequence may prolong protein synthesis in a tissue-specific manner.
  • the regulatory features of a UTR can be incorporated into the RNAs of the present disclosure to, among other things, enhance the stability of the molecule.
  • the term “altered” as it relates to a UTR sequence means that the UTR has been changed in some way in relation to a reference sequence.
  • 5′ UTRs and/or 3′ UTRs may be altered relative to a wild-type or native UTR by the change in orientation and/or location as taught above and/or may be altered by the inclusion of additional nucleotides, deletion of nucleotides, swapping, and/or transposition of nucleotides. Any of these changes produces an “altered” UTR (whether 5′ and/or 3′) including a variant UTR.
  • a double, triple or quadruple UTR such as a 5′ and/or 3′ UTR may be used.
  • a “double” UTR is one in which two copies of the same UTR are encoded either in series or substantially in series.
  • a double beta-globin 3′ UTR may be used.
  • patterned UTRs are those UTRs which reflect a repeating or alternating pattern, such as AB AB AB or AABBAABBAABB or ABCABCABC or variants thereof repeated once, twice, or more than 3 times. In these patterns, each letter, A, B, or C represent a different UTR at the nucleotide level.
  • RNAs may encode polypeptides of interest belonging to a family of proteins that are expressed in a particular cell, tissue and/or at some time during development.
  • the UTRs from any of these genes may be swapped for any other UTR of the same or different family of proteins to create a new RNA molecule.
  • a “family of proteins” is used in the broadest sense to refer to a group of two or more polypeptides of interest which share at least one function, structure, feature, localization, origin, and/or expression pattern.
  • the 5′ UTR and the 3′ UTR sequences are computationally derived.
  • the 5′ UTR and the 3′ UTRs are derived from a naturally abundant mRNA in a tissue.
  • the tissue may be, for example, liver, a stem cell and/or lymphoid tissue.
  • the lymphoid tissue may include, for example, any one of a lymphocyte (e.g., a B-lymphocyte, a helper T- lymphocyte, a cytotoxic T-lymphocyte, a regulatory T-lymphocyte, and/or a natural killer cell), a macrophage, a monocyte, a dendritic cell, a neutrophil, an eosinophil and a reticulocyte.
  • a lymphocyte e.g., a B-lymphocyte, a helper T- lymphocyte, a cytotoxic T-lymphocyte, a regulatory T-lymphocyte, and/or a natural killer cell
  • a macrophage e.g., a monocyte, a dendritic cell, a neutrophil, an eosinophil and a reticulocyte.
  • the 5′ UTR and the 3′ UTR are derived
  • untranslated regions may also include translation enhancer elements (TEE).
  • TEE translation enhancer elements
  • the TEE may include those described in US Application No. 20090226470, herein incorporated by reference in its entirety, and those known in the art.
  • Those of ordinary skill in the art will understand that 5′ UTRs that are heterologous and/or synthetic may be used with any desired 3′ UTR sequence, and vice versa.
  • a heterologous 5′ UTR may be used with a synthetic and/or heterologous 3′ UTR.
  • an RNA molecule disclosed herein comprises a 5′ UTR selected from Table 5.
  • a 5′ UTR if present, is located at the 5′ end and starts with the transcriptional start site upstream of the start codon of a protein encoding region.
  • a 5′ UTR is downstream of the 5′ cap (if present), e.g. directly adjacent to the 5′ cap.
  • the 5′ UTR may contain various regulatory elements, e.g., 5′ cap structure, stem-loop structure, and an internal ribosome entry site (IRES), which may play a role in the control of translation initiation.
  • the 5′ UTR may harbor signatures like Kozak sequences, which are also involved in the process by which the ribosome initiates translation of many genes.5′ UTRs may also form secondary structures involved in elongation factor binding.
  • a 5′ UTR disclosed herein comprises a cap proximal sequence, e.g., as disclosed herein.
  • a cap proximal sequence comprises a sequence adjacent to a 5′ cap.
  • a cap proximal sequence comprises nucleotides in positions +1, +2, +3, +4, and/or +5 of an RNA polynucleotide.
  • a Cap structure comprises one or more polynucleotides of a cap proximal sequence.
  • a Cap structure comprises an m7 Guanosine cap and nucleotide +1 (N 1 ) of an RNA polynucleotide.
  • a Cap structure comprises an m7 Guanosine cap and nucleotide +2 (N 2 ) of an RNA polynucleotide. In some aspects, a Cap structure comprises an m7 Guanosine cap and nucleotides +1 and +2 (N 1 and N 2 ) of an RNA polynucleotide.
  • one or more residues of a cap proximal sequence may be included in an RNA by virtue of having been included in a cap entity that (e.g., a Cap 1 structure, etc); alternatively, in some aspects, at least some of the residues in a cap proximal sequence may be enzymatically added (e.g., by a polymerase such as a T7 polymerase).
  • a cap proximal sequence comprises N 1 and/or N 2 of a Cap structure, wherein N 1 and N 2 are any nucleotide, e.g., A, C, G or U.
  • N 1 is A.
  • N 1 is C.
  • N 1 is G.
  • N 1 is U.
  • N 2 is A. In some aspects, N 2 is C. In some aspects,N 2 is G. In some aspects, N 2 is U. In some aspects, a cap proximal sequence comprisesN 1 and N 2 of a Cap structure and N 3 , N 4 and N 5 , wherein N 1 to N 5 correspond to positions +1, +2, +3, +4, and/or +5 of an RNA polynucleotide. In some aspects, N 1 , N 2 , N 3 , N 4 , or N 5 are any nucleotide, e.g., A, C, G or U.
  • N 1 N 2 comprises any one of the following: AA, AC, AG, AU, CA, CC, CG, CU, GA, GC, GG, GU, UA, UC, UG, or UU.
  • N1N2 comprises AG and N3N4N5 comprises any one of the following: AAA, ACA, AGA, AUA, AAG, AGG, ACG, AUG, AAC, ACC, AGC, AUC, , AAU, ACU, AGU, AUU, CAA, CCA, CGA, CUA, CAG, CGG, CCG, CUG, CAC, CCC, CGC, CUC, , CAU, CCU, CGU, CUU, , GAA, GCA, GGA, GUA, , GAG, GGG, GCG, GUG, , GAC, GCC, GGC, GUC, , GAU, GCU, GGU, GUU, UAA, UCA, UGA, UUA, UCA, UGA,
  • a cap proximal sequence comprises N 1 and N 2 of a Cap structure, and a sequence comprising: A 3 A 4 X 5 (SEQ ID NO: 44; wherein X 5 is A, G, C, or U), where N 1 and N 2 are each independently chosen from: A, C, G, or U.
  • N 1 is A and N 2 is G.
  • X 5 is chosen from A, C, G or U.
  • X 5 is A.
  • X 5 is C.
  • X 5 is G.
  • X 5 is U.
  • a cap proximal sequence comprises N 1 and N 2 of a Cap structure, and a sequence comprising: C 3 A 4 X 5 (SEQ ID NO: 45; wherein X 5 is A, G, C, or U), where N 1 and N 2 are each independently chosen from: A, C, G, or U.
  • N 1 is A and N 2 is G.
  • X 5 is chosen from A, C, G or U.
  • X 5 is A.
  • X 5 is C.
  • X 5 is G.
  • X 5 is U.
  • a cap proximal sequence comprises N 1 and N 2 of a Cap structure, and a sequence comprising X 3 Y 4 X 5 (SEQ ID NO: 46; wherein X 3 or X 5 are each independently chosen from A, G, C, or U; and Y 4 is not C).
  • N 1 and N 2 are each independently chosen from: A, C, G, or U.
  • N 1 is A and N 2 is G.
  • X 3 and X 5 is each independently chosen from A, C, G or U.
  • X 3 and/or X 5 is A.
  • X 3 and/or X 5 is C.
  • X 3 and/or X 5 is G.
  • X 3 and/or X 5 is U.
  • Y 4 is C. In other aspects, Y 4 is not C. In some aspects, Y 4 is A. In some aspects, Y 4 is G. In other aspects, Y 4 is not G. In some aspects, Y 4 is U.
  • a cap proximal sequence comprises N 1 and N 2 of a Cap structure, and a sequence comprising A 3 C 4 A 5 (SEQ ID NO: 47).
  • N 1 and N 2 are each independently chosen from: A, C, G, or U. In some aspects, N 1 is A and N 2 is G.
  • a cap proximal sequence comprises N 1 and N 2 of a Cap structure, and a sequence comprising A 3 U 4 G 5 (SEQ ID NO: 48).
  • N 1 and N 2 are each independently chosen from: A, C, G, or U.
  • N 1 is A and N 2 is G.
  • 1, 2, 3, 4, 5, or more of the foregoing cap proximal sequences can be excluded from the 5′ UTR of the RNA molecules disclosed herein.
  • a 5′ UTR is a heterologous UTR, e.g., is a UTR found in nature associated with a different ORF.
  • a 5′ UTR is a synthetic UTR, e.g., does not occur in nature.
  • Synthetic UTRs include UTRs that have been mutated or synthesized to improve their properties, e.g., to increase gene expression.
  • the 5′ UTR is functionally linked to the ORF, e.g., associated with the ORF such that it may exert a function, e.g., increasing, enhancing, stabilizing, and/or prolonging protein production from an RNA molecule and/or increasing protein expression and/or total protein production from an RNA molecule, compared to a reference RNA molecule comprising a reference 5′ UTR or an RNA molecule lacking a 5′ UTR.
  • Transfection experiments can be conducted in relevant cell lines, using nucleic acids of the disclosure and protein production can be assayed at various time points post-transfection.
  • cells can be transfected with different ARE-engineering molecules and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hour, 12 hour, 24 hour, 48 hour, and 7 days post-transfection.
  • a 3′ UTR may have one or more AU-rich sequences removed. Alternatively the AU-rich sequences may remain in the 3′ UTR.
  • a 3′ UTR may also comprise elements, which are not encoded in the template, from which an RNA is transcribed, but which are added after transcription during maturation, e.g. a poly-A tail.
  • An open reading frame may terminate with at least one stop codon, including but not limited to TAA, TAG, TGA or UAA, UAG or UGA, or any combination thereof.
  • an open reading frame may terminate with one, two, three, four or more stop codons, including but not limited to TAATAA (SEQ ID NO: 25), TAATAG (SEQ ID NO: 26), TAATGA (SEQ ID NO: 27), TAGTGA (SEQ ID NO: 28), TAGTAA (SEQ ID NO: 29), TAGTAG (SEQ ID NO: 30), TGATGA (SEQ ID NO: 31), TGATAG (SEQ ID NO: 32), TGATAA (SEQ ID NO: 33) or UAAUAA (SEQ ID NO: 34), UAAUAG (SEQ ID NO: 35), UAAUGA (SEQ ID NO: 36), UAGUGA (SEQ ID NO: 37), UAGUAA (SEQ ID NO:38), UAGUAG (SEQ ID NO: 39), UG
  • antigen may refer to a substance, which is capable of being recognized by the immune system, e.g., by the adaptive immune system, and which is capable of eliciting an antigen-specific immune response, e.g., by formation of antibodies and/or antigen-specific T cells as part of an adaptive immune response.
  • An antigen may be or may comprise a peptide or protein, which may be presented by the MHC to T cells.
  • An antigen may be the product of translation of a provided nucleic acid molecule, e.g., an RNA molecule comprising at least one coding sequence as described herein.
  • a DNA template used to transcribe an RNA polynucleotide described herein comprises a sequence complementary to an RNA polynucleotide.
  • a gene of interest described herein is encoded by an RNA polynucleotide described herein comprising a nucleotide sequence disclosed herein.
  • an RNA polynucleotide encodes a polypeptide having at least 80% identity to a polypeptide sequence disclosed herein.
  • a polypeptide described herein is encoded by an RNA polynucleotide transcribed by a DNA template comprising a sequence complementary to an RNA polynucleotide.
  • the RNA molecule encodes a RSV preF protein comprising the sequence of any one of SEQ ID NOs: 1-6 or 621-626, or a fragment or variant thereof. In a preferred aspect, the RNA molecule encodes a RSV preF protein comprising the sequence of any one of SEQ ID NOs: 4 or 6, or a fragment or variant thereof. In some aspects, the RNA molecule encodes a RSV preF protein wherein the RNA molecule is synthesized from the nucleic acid having the sequence comprising any one of SEQ ID NOs: 7-10, 627-632, 639 or 640, or fragment or variant thereof.
  • the RNA molecule encodes a RSV preF protein wherein the RNA molecule is synthesized from the nucleic acid having the sequence of any one of SEQ ID NOs: 8, 10-14, 639 or 640, or a fragment or variant thereof.
  • the RNA molecule encodes a RSV preF protein wherein the RNA molecule comprises the nucleic acid sequence set forth in any one of SEQ ID NOs: 15-20, 633- 638, 641 or 642, or fragment or variant thereof.
  • the RNA molecule encodes a RSV preF protein wherein the RNA molecule comprises the nucleic acid sequence set forth in any one of SEQ ID NOs: 15-20, 641 or 642, or a fragment or variant thereof.
  • RNA molecules disclosed herein comprise a poly-adenylate (poly-A) sequence, e.g., as described herein.
  • a poly-A sequence is situated downstream of a 3′ UTR, e.g., adjacent to a 3′ UTR.
  • the poly-A tail may increase the stability, half-life, and/or translational efficiency of the RNA molecule.
  • 3′-end processing is a nuclear co-transcriptional process that promotes transport of mRNAs from the nucleus to the cytoplasm and affects the stability and the translation of mRNAs.
  • this 3′ end occurs in a two-step reaction directed by the cleavage/polyadenylation machinery and depends on the presence of two sequence elements in mRNA precursors (pre-mRNAs); a hexanucleotide polyadenylation signal and a downstream G/U-rich sequence.
  • pre-mRNAs mRNA precursors
  • a hexanucleotide polyadenylation signal a downstream G/U-rich sequence.
  • a first step pre-mRNAs are cleaved between these two elements to a free 3′ hydroxyl.
  • the newly formed 3′ end is extended by polyadenylation or addition of a poly-A sequence.
  • Polyadenylation refers to the addition of a poly-A sequence to an RNA molecule, e.g., to a premature mRNA.
  • Polyadenylation may be induced by a so-called polyadenylation signal. This signal may be located within a stretch of nucleotides close to or at the 3′-end of an RNA molecule to be polyadenylated. A polyadenylation signal may also be comprised by the 3′ UTR of the artificial nucleic acid molecule.
  • a polyadenylation signal typically comprises a hexamer consisting of adenine and uracil/thymine nucleotides, preferably the hexamer sequence AAUAAA, though other sequences, preferably hexamer sequences, are also conceivable. Polyadenylation typically occurs during processing of a pre-mRNA (also called premature-mRNA).
  • RNA maturation comprises the step of polyadenylation.
  • Poly-A tailing of in vitro transcribed mRNA can be achieved using various approaches including, but not limited to, cloning of a poly-T tract into the DNA template or by post-transcriptional addition using poly-A polymerase.
  • the term may relate to polyadenylation of RNA as a cellular process or to polyadenylation carried out by enzymatic reaction in vitro with a suitable enzyme, such as E. coli poly-A polymerase, or by chemical synthesis.
  • RNA molecules disclosed herein may have a poly-A sequence attached to the free 3′-end of the RNA by a template-independent RNA polymerase after transcription or a poly-A sequence encoded by DNA and transcribed by a template-dependent RNA polymerase.
  • a poly-A sequence is attached during RNA transcription, e.g., during preparation of in vitro transcribed RNA, based on a DNA template comprising repeated dT nucleotides (deoxythymidylate) in the strand complementary to the coding strand.
  • the DNA sequence encoding a poly-A sequence (coding strand) is referred to as poly-A cassette.
  • the poly-A cassette present in the coding strand of DNA essentially consists of dA nucleotides, but is interrupted by a random sequence of the four nucleotides (dA, dC, dG, and dT).
  • a random sequence may be at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleotides in length.
  • a poly-A cassette that essentially consists of dA nucleotides, but is interrupted by a random sequence having an equal distribution of the four nucleotides (dA, dC, dG, dT) and having a length of e.g., 5 to 50 nucleotides, shows, on a DNA level, constant propagation of plasmid DNA in E. coli and is still associated, on an RNA level, with the beneficial properties with respect to supporting RNA stability and translational efficiency.
  • the poly-A sequence contained in an RNA polynucleotide described herein consists essentially of adenosine nucleotides, but is interrupted by a random sequence of the four nucleotides (A, C, G, U).
  • a random sequence may be at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleotides in length.
  • the poly-A sequence may be located at any position within the 3′ UTR.
  • no nucleotides other than adenosine nucleotides flank a poly-A sequence at its 3′-end, e.g., the poly-A sequence is not masked or followed at its 3′-end by a nucleotide other than adenosine.
  • the poly-A sequence may be located at the 3′ terminus of the 3′ UTR, e.g., the 3′ UTR does not contain more than 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 nucleotides located 3′ of the poly-A sequence; more preferably the 3′ UTR does not contain further elements located 3′ to the poly-A sequence.
  • poly-A sequence is located at the 3′ terminus of the RNA molecule, e.g., the artificial nucleic acid molecule does not contain more than 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 nucleotides located 3′ of the poly-A sequence.
  • the poly-A sequence may be located at the 5′ terminus of the 3′ UTR, e.g., immediately 3′ of the ORF of the artificial nucleic acid molecule, or located within the 3′ UTR, e.g., flanked on the 5′ and on the 3′ side by other 3′ UTR elements.
  • the poly-A sequence is flanked on the 3′ side by a poly- C sequence and/or a histone stem-loop sequence.
  • the poly-A sequence can be flanked on the 5′ side by a 3′ UTR element derived from, e.g., a human albumin or globin gene.
  • the RNA molecule may further include an endonuclease recognition site sequence immediately downstream of the poly-A tail sequence.
  • the RNA molecule may further include a poly-A polymerase recognition sequence (e.g., a polyadenylation signal) (e.g., AAUAAA) near its 3′ end.
  • the polyadenylation signal is located 3′ of the poly-A sequence comprised in the 3′ UTR.
  • the poly-A sequence is separated from the polyadenylation signal by a nucleotide sequence comprising or consisting of at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, or 150 nucleotides, wherein the nucleotide sequence does preferably not comprise more than 10, 9, 8, 7, 6, 5, 4, 3, or 2 consecutive adenine nucleotides.
  • the nucleotide sequence that separates the poly-A sequence and the polyadenylation signal comprises from or from about 1 to about 200 nucleotides, e.g., from 10 to 90, from 20 to 85, from 30 to 80, from 40 to 80, from 50 to 75 or from 55 to 85 nucleotides, more preferably from 55 to 80 nucleotides, and the nucleotide sequence does not comprise more than 10, 9, 8, 7, 6, 5, 4, 3, or 2 consecutive adenine nucleotides.
  • Such a consensus sequence may be recognized by most animal and bacterial cell-systems, for example, by the polyadenylation-factors, such as cleavage/polyadenylation specificity factor (CPSF) cooperating with CstF, PAP, PAB2, CFI and/or CFII.
  • CPSF cleavage/polyadenylation specificity factor
  • the polyadenylation signal (e.g., the consensus sequence NNUANA) is located less than or less than about 50 nucleotides, e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides, downstream of the 3′-end of the 3′ UTR element as defined herein such that transcription of an RNA molecule will result in a premature-RNA containing the polyadenylation signal downstream of its 3′ UTR and subsequent attachment of a poly-A sequence to the premature-RNA.
  • the polyadenylation signal e.g., the consensus sequence NNUANA
  • a resulting RNA may comprise a 3′ UTR, which comprises at least one poly-A sequence, and wherein the 3′ UTR is followed by an additional poly-A sequence.
  • the poly-A sequence may be of any length.
  • the poly-A tail may be 5 to 300 nucleotides in length.
  • the RNA molecule includes a poly-A tail that comprises, consists essentially of, or consists of a sequence of or of about 25 to about 400 adenosine nucleotides, a sequence of or of about 50 to about 400 adenosine nucleotides, a sequence of or of about 50 to about 300 adenosine nucleotides, a sequence of or of about 50 to about 250 adenosine nucleotides, a sequence of or of about 60 to about 250 adenosine nucleotides, or a sequence of or of about 40 to about 100 adenosine nucleotides.
  • the poly-A tail comprises, consists essentially of, or consists of at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 250, 255, 260, 265, 270, 275, 280, 285, 290, 295, 300, 305, 310, 315, 320, 325, 330, 335, 340, 345, 350, 355, 360, 365, 370, 375, 380, 385, 390, 395,
  • “consists essentially of” means that most nucleotides in the poly-A sequence, typically at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% by number of nucleotides in the poly-A sequence are adenosine nucleotides, but permits remaining nucleotides to be nucleotides other than adenosine nucleotides, such as uridine, guanosine, and/or cytosine.
  • RNA molecule includes a poly-A tail that includes a sequence of greater than 30 adenosine nucleotides. In some aspects, the RNA molecule includes a poly-A tail that includes or includes about 40 adenosine nucleotides. In some aspects, the RNA molecule includes a poly-A tail that includes or includes about 80 adenosine nucleotides.
  • Purification can be performed using a variety of commercially available kits including, but not limited to SV Total Isolation System (Promega) and In vitro Transcription Cleanup and Concentration Kit (Norgen Biotek). In some aspects, 1, 2, 3, 4, 5, or more of the foregoing purification may be excluded.
  • the term “purified” when used in relation to a nucleic acid such as a “purified nucleic acid” refers to one that is separated from at least one contaminant.
  • a “contaminant” is any substance that makes another unfit, impure or inferior.
  • the RNA is at a concentration of at least or at least about 1 mg/mL. In another aspect, the RNA concentration is from or from about 0.05 mg/mL to about 0.5 mg/mL. In another aspect, the RNA is at a concentration of at least 10 mg/mL. In another aspect, the RNA is at a concentration of at least 50 mg/mL.
  • the RNA is or is not at a concentration of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 0.05 mg/mL, 0.06 mg/mL, 0.07 mg/mL, 0.08 mg/mL, 0.09 mg/mL, 0.10 mg/mL, 0.20 mg/mL, 0.30 mg/mL, 0.40 mg/mL, 0.5 mg/mL, 1 mg/mL, 10 mg/mL, 50 mg/mL, 75 mg/mL, 100 mg/mL, 150 mg/mL, 200 mg/mL, 250 mg/mL, 300 mg/mL, 400 mg/mL, or more.
  • the LNPs comprise at least one cationic (e.g., ionizable) lipid, at least one neutral (e.g., non-cationic) lipid, at least one structural lipid (e.g., a steroid), and/or at least one polymer conjugated lipid (e.g., a polyethylene glycol (PEG)-modified lipid).
  • a polyethylene glycol (PEG)-modified lipid e.g., 1, 2, 3, or more of the foregoing excipients may be excluded from the LNPs.
  • the LNPs comprise 20-60 mol% cationic (e.g., ionizable) lipid(s).
  • the LNPs may comprise 20-50 mol%, 20-40 mol%, 20-30 mol%, 30-60 mol%, 30-50 mol%, 30-40 mol%, 40-60 mol%, 40-50 mol%, or 50-60 mol% cationic (e.g., ionizable) lipid(s).
  • the LNPs comprise or do not comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 20 mol%, 30 mol%, 40 mol%, 50, or 60 mol% cationic (e.g., ionizable) lipid(s).
  • the LNPs comprise 45 to 55 mole percent (mol%) cationic (e.g., ionizable) lipid(s).
  • LNPs may comprise or not comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, or 55 mol% cationic (e.g., ionizable) lipid(s).
  • the LNPs comprise 5-25 mol% neutral (e.g., non-cationic) lipid(s).
  • LNPs may comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 mol% neutral (e.g., non-cationic) lipid(s).
  • the LNPs comprise 25-55 mol% structural lipid(s) (e.g., a steroid).
  • the LNPs may comprise 25-50 mol%, 25-45 mol%, 25-40 mol%, 25-35 mol%, 25-30 mol%, 30-55 mol%, 30-50 mol%, 30-45 mol%, 30-40 mol%, 30-35 mol%, 35-55 mol%, 35-50 mol%, 35-45 mol%, 35-40 mol%, 40-55 mol%, 40-50 mol%, 40-45 mol%, 45-55 mol%, 45-50 mol%, or 50-55 mol% structural lipid(s) (e.g., a steroid).
  • structural lipid(s) e.g., a steroid
  • the LNPs are or are not at least, at most, exactly, or between (inclusive or exclusive) any two of 25 mol%, 30 mol%, 35 mol%, 40 mol%, 45 mol%, 50 mol%, or 55 mol% structural lipid(s) (e.g., a steroid).
  • the LNPs comprise 35 to 40 mol% structural lipid(s) (e.g., a steroid).
  • LNPs may comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 35, 36, 37, 38, 39, or 40 mol% structural lipid(s) (e.g., a steroid).
  • the LNPs comprise 0.5-15 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)-modified lipid).
  • the lipid nanoparticle may comprise 0.5- 10 mol%, 0.5-5 mol%, 1-15 mol%, 1-10 mol%, 1-5 mol%, 2-15 mol%, 2-10 mol%, 2-5 mol%, 5- 15 mol%, 5-10 mol%, or 10-15 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)-modified lipid).
  • the lipid LNPs are or are not at least, at most, exactly, or between (inclusive or exclusive) any two of 0.5 mol%, 1 mol%, 2 mol%, 3 mol%, 4 mol%, 5 mol%, 6 mol%, 7 mol%, 8 mol%, 9 mol%, 10 mol%, 11 mol%, 12 mol%, 13 mol%, 14 mol%, or 15 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)-modified lipid).
  • the LNPs comprise 1 to 2 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)- modified lipid).
  • LNPs may comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 1.5, or 2 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)-modified lipid).
  • PEG polyethylene glycol
  • the LNPs comprise 20-75 mol% cationic (e.g., ionizable) lipid(s) (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, and 75%), 0.5-25 mol% neutral (e.g., non-cationic) lipid(s) (e.g., at least, at most, exactly, or between (inclusive or exclusive) of 0.5%, 2.25%, 4%, 5.75%, 7.5%, 9.25%, 11%, 12.75%, 14.5%, 16.25%, 18%, 19.75%, 21.5%, 23.25%, and 25%), 5-55 mol% structural lipid(s) (e.g., a sterol) e.g., non-cationic) lipid(s) (e.g., at least, at most, exactly, or between (inclusive or exclusive) of 5%, 10%
  • the molar lipid ratio is 50/10/38.5/1.5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 60/7.5/31/1.5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 57.5/7.5/31.5/3.5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 57.2/7.1/34.3/1.4 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 40/15/40/5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 50/10/35/4.5/0.5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 50/10/35/5 (mol% cationic lipid/35/5 (mol% cationic lipid/cationic lipid/neutral lipid/structural lipid/polymer
  • the active agent or therapeutic agent such as a nucleic acid (e.g., mRNA)
  • a nucleic acid e.g., mRNA
  • the active agent or therapeutic agent may be encapsulated in the lipid portion of the lipid nanoparticle and/or an aqueous space enveloped by some or all of the lipid portion of the lipid nanoparticle, thereby protecting it from enzymatic degradation or other undesirable effects induced by the mechanisms of the host organism or cells, e.g., an adverse immune response.
  • the nucleic acid (e.g., mRNA) or a portion thereof may also be associated and complexed with the lipid nanoparticle.
  • a lipid nanoparticle may comprise any lipid capable of forming a particle to which the nucleic acids are attached, and/or in which the one or more nucleic acids are encapsulated.
  • provided RNA molecules e.g., mRNA
  • LNPs LNPs
  • the lipid nanoparticles may or may not have a mean diameter of or of about 1 to 500 nm (e.g., at least, at most, exactly, or between (inclusive or exclusive) of 1, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, or 500 nm).
  • 1 to 500 nm e.g., at least, at most, exactly, or between (inclusive or exclusive) of 1, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220,
  • the lipid nanoparticles have a mean diameter of or of from about 30 nm to about 150 nm, about 40 nm to about 150 nm, about 50 nm to about 150 nm, about 60 nm to about 130 nm, about 70 nm to about 110 nm, about 70 nm to about 100 nm, about 80 nm to about 100 nm, about 90 nm to about 100 nm, about 70 to about 90 nm, about 80 nm to about 90 nm, about 70 nm to about 80 nm, or at least, at most, exactly, or between (inclusive or exclusive) of 30 nm, 35 nm, 40 nm, 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, 80 nm, 85 nm, 90 nm, 95 nm, 100 nm, 105 nm, 110 nm, 115
  • mean diameter refers to the mean hydrodynamic diameter of particles as measured by dynamic laser light scattering (DLS) with data analysis using the so-called cumulant algorithm, which provides as results the so-called Z-average with the dimension of a length, and the polydispersity index (PI), which is dimensionless (Koppel, D., J. Chem. Phys. 57, 1972, pp 4814-4820, ISO 13321).
  • mean diameter “diameter,” or “size” for particles is used synonymously with the value of the Z-average.
  • LNPs described herein may exhibit a polydispersity index less than or less than about 0.5, 0.4, 0.3, or 0.2 or less.
  • the LNPs may or may not exhibit a polydispersity index of at least, at most, exactly, or between (inclusive or exclusive) of 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, or 0.5.
  • the polydispersity index is, in some aspects, calculated based on dynamic light scattering measurements by the so-called cumulant analysis referred to in the definition of “average diameter.” Under certain prerequisites, it may be taken as a measure of the size distribution of an ensemble of nanoparticles.
  • an LNP of the disclosure comprises or does not comprise an N:P ratio of or of from about 2:1 to about 30:1, e.g., at least, at most, exactly, or between (inclusive or exclusive) of 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, 21:1, 22:1, 23:1, 24:1, 25:1, 26:1, 27:1, 28:1, 29:1, or 30:1.
  • an LNP of the disclosure comprises an N:P ratio of or of about 6:1.
  • an LNP of the disclosure comprises an N:P ratio of or of about 3:1.
  • N:P is the ratio of cationic lipid to RNA.
  • an LNP of the disclosure comprises or does not comprise a wt/wt ratio of the cationic lipid component to the RNA of or of from about 5:1 to about 100:1, e.g., at least, at most, exactly, or between (inclusive or exclusive) of 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, 21:1, 22:1, 23:1, 24:1, 25:1, 26:1, 27:1, 28:1, 29:1, 30:1, 31:1, 32:1, 33:1, 34:1, 35:1, 36:1, 37:1, 38:1, 39:1, 40:1, 41:1, 42:1, 43:1, 44:1, 45:1, 46:1, 47:1, 48:1, 49:1, 50:1, 51:1, 52:1, 53:1, 54:1, 55:1, 56:1, 57:1,
  • the term refers to molecules, which comprise hydrophilic and hydrophobic moieties with different structural organization, which may or may not be similar to that of lipids.
  • the RNA solution and lipid preparation mixture or compositions thereof may comprise cationic lipids, neutral lipids, cholesterol, and/or polymer (e.g., polyethylene glycol) conjugated lipids which form lipid nanoparticles that encompass the RNA molecules.
  • the LNP may comprise a cationic lipid and one or more excipients, e.g., one or more neutral lipids, charged lipids, steroids or steroid analogs (e.g., cholesterol), polymer conjugated lipids (e.g.
  • a cationic lipid may comprise from or from about 10 mol % to about 100 mol %, about 20 mol % to about 100 mol %, about 30 mol % to about 100 mol %, about 40 mol % to about 100 mol %, or about 50 mol % to about 100 mol % of the total lipid present in the particle.
  • a cationic lipid may or may not be at least, at most, exactly, or between (inclusive or exclusive) of 10 mol %, 20 mol %, 30 mol %, 40 mol %, 50 mol %, 60 mol %, 70 mol %, 80 mol %, 90 mol %, or 100 mol %, or any range or value derivable therein, of the total lipid present in the particle.
  • the cationic lipid comprises 47.5 mol% of the total lipid present in the LNP.
  • cationic lipids include, but are not limited to: ((4- hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate), 1,2-dioleoyl-3- trimethylammonium propane (DOTAP), N,N-dimethyl-2,3-dioleyloxypropylamine (DODMA), 1,2- di-O-octadecenyl-3-trimethylammonium propane (DOTMA), 3-(N-( N′,N′-dimethylaminoethane)- carbamoyl)cholesterol (DC-Chol), dimethyldioctadecylammonium (DDAB), 1,2-dioleoyl-3- dimethylammonium-propane (DODAP), 1,2-diacyloxy-3-dimethylammonium propanes, 1,2- dialkyloxy-3-dimethylammonium propanes; dioct
  • a cationic lipid of the disclosure is a compound having Formula (II): or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein: L 1 and L 2 are each independently —O(C ⁇ O)—, —(C ⁇ O)O—, —C( ⁇ O)—, —O—, —S— S—, —C( ⁇ O)S—, —SC( ⁇ O)—, —NR a C( ⁇ O)—, —C( ⁇ O)NR a —, —NR a C( ⁇ O)NR a —, —OC( ⁇ O)NR a —, —NR a C( ⁇ O)O—, or a direct bond; G 1 is C 1 -C 2 alkylene, —(C ⁇ O)—, —O(C ⁇ O)—, —SC( ⁇ O)—, —NR a C( ⁇ O)— or a direct bond; G 2 is —C( ⁇ O)—,
  • the cationic lipid is selected from any one of Compound Nos.1- 12 as specifically exemplified in Table 1 of PCT Publication No. WO2019/036030, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes.
  • the cationic lipid is described in PCT Publication No. WO2019/036008, the contents of which is hereby incorporated herein by reference in its entirety for all purposes.
  • a cationic lipid of the disclosure is a compound having the following structures (IA), wherein y and z are each independently integers ranging from 2 to 12.
  • a cationic lipid of the disclosure is a compound having any one of the following structures (IB), (IC), (ID) or (IE):
  • a cationic lipid of the disclosure is a compound having any one of the following structures (IF), (IG), (IH) or (IJ):
  • the cationic lipid is selected from any one of Compound Nos.1- 18 as specifically exemplified in Table 1 of PCT Publication No. WO2019/036008, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes.
  • the cationic lipid is described in PCT Publication No. WO2020/081938, the contents of which is hereby incorporated herein by reference in its entirety for all purposes.
  • an ionizable cationic lipid of the disclosure comprises a compound of structure (I): or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: G 1 is —N(R 3 )R 4 or —OR′; R 1 is optionally substituted branched, saturated or unsaturated C 12 -C 36 alkyl; R 2 is optionally substituted branched or unbranched, saturated or unsaturated C 12 - C 36 alkyl when L is —C( ⁇ O)—; or R 2 is optionally substituted branched or unbranched, saturated or unsaturated C 4 -C 36 alkyl when L is C 6 -C 12 alkylene, C 6 -C 12 alkenylene, or C 2 -C 6 alkynylene; R 3 and R 4 are each independently H, optionally substituted branched or unbranched, saturated or unsaturated C 1 -C 6 alkyl; or R 3 and R 4 are each independently optionally substituted branched,
  • a cationic lipid of the disclosure is a compound having the following structure (IA): wherein R 8 and R 9 are each independently H or optionally substituted branched or unbranched, saturated or unsaturated C 2 -C 12 alkyl, provided that R 8 and R 9 are each independently selected such that R 1 is optionally substituted branched, saturated or unsaturated C 12 -C 36 alkyl; and R 10 and R 11 are each independently H or optionally substituted branched or unbranched, saturated or unsaturated C 2 -C 12 alkyl, provided that R 10 and R 11 are each independently selected such that: R 2 is optionally substituted branched or unbranched, saturated or unsaturated C 12 - C 36 alkyl when L is —C( ⁇ O)—; and R 2 is optionally substituted branched or unbranched, saturated or unsaturated C 4 -C 36 alkyl when L is C 6 -C 12 alkylene, C 6 -C 12 alkenylene, or C 2
  • a cationic lipid of the disclosure is a compound having the following structure (IA): .
  • a cationic lipid of the disclosure is a compound having the following structure (IB):
  • the cationic lipid is selected from any one of Compound Nos. I- 1 to I-40 as specifically exemplified in Table 1 of PCT Publication No. WO2020/146805, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes.
  • the cationic lipid is described in PCT Publication No. WO2022/016070, the contents of which is hereby incorporated herein by reference in its entirety for all purposes.
  • an ionizable cationic lipid of the disclosure comprises a compound of structure (I): or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein: G 1 and G 2 are each independently C 1 -C 6 alkylene; L 1 and L 2 are each independently —O(C ⁇ O)— or —(C ⁇ O)O—; R 1a and R 1b are, at each occurrence, independently either: (a) H or C 1 -C 12 alkyl; or (b) R 1a is H or C 1 -C 12 alkyl, and R 1b together with the carbon atom to which it is bound is taken together with an adjacent R 1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R 2a and R 2b are, at each occurrence, independently either: (a) H or C 1 -C 12 alkyl; or (b) R 2a is H or C 1 -C 12 alkyl, and R 2b together with the carbon
  • the cationic lipid is selected from any one of Compound Nos. I- 1 to I-23 as specifically exemplified in Table 1 of PCT Publication No. WO2022/016070, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes.
  • the cationic lipid is described in PCT Publication No. WO2023/114944, the contents of which is hereby incorporated herein by reference in its entirety for all purposes.
  • a cationic lipid of the disclosure is a compound having any one of the following structures (IA) or (IB): .
  • the cationic lipid is selected from any one of Compound Nos. I- 1 to I-25 as specifically exemplified in Table 1 of PCT Publication No. WO2023/114944, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes.
  • the cationic lipid is described in PCT Publication No. WO2023/114937, the contents of which is hereby incorporated herein by reference in its entirety for all purposes.
  • a cationic lipid of the disclosure is a compound having the following structure (IA): wherein: R 6 is, at each occurrence, independently H, F, OH or C 1 -C 24 alkyl; and n is an integer ranging from 1 to 15.
  • a cationic lipid of the disclosure is a compound having the following structure (IB): wherein: y and z are each independently integers ranging from 1 to 12; and R 7 is, at each occurrence, independently H or F.
  • a cationic lipid of the disclosure is a compound having any one of the following structures (IC) or (ID): In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IE) or (IF): (IE) (IF) In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IG) or (IH): (IG) (IH) wherein R 11 and R 12 are each independently C 1 -C 12 alkyl; or R 11 and R 12 , together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring comprising one nitrogen atom.
  • the cationic lipid is selected from any one of Compound Nos. I- 1 to I-10 as specifically exemplified in Table 1 of PCT Publication No. WO2023/114937, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes.
  • the cationic lipid is described in PCT Publication No. WO2023/114943, the contents of which is hereby incorporated herein by reference in its entirety for all purposes.
  • a cationic lipid of the disclosure is a compound having the following structure (IA): wherein y and z are each independently integers ranging from 2 to 12.
  • a cationic lipid of the disclosure is a compound having any one of the following structures (IB) or (IC): (IB) (IC)
  • the cationic lipid is selected from any one of Compound Nos. I- 1 to I-85 as specifically exemplified in Table 1 of PCT Publication No. WO2023/114943, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes.
  • the ionizable cationic lipid is described in PCT Publication No.
  • R 4b is C 8 -C 24 -alkyl. In some embodiments, R 4b is C 10 -C 18 -alkyl. In certain embodiments, R 4b is C 11 -C 16 -alkyl. In some embodiments, R 4b is C 11 -alkyl. In certain embodiments, R 4b is C 15 -alkyl. In some embodiments, R 4b is C 16 -alkyl. In certain embodiments, R 4a is branched. In some embodiments, R 4b is branched. In certain embodiments, R 4a is unsubstituted. In some embodiments, R 4b is unsubstituted.
  • R 6 is C 1 -C 6 alkyl. In some embodiments, R 6 is C 1 -C 10 alkyl. In certain embodiments, R 6 is C 1 -C 4 -alkyl. In some embodiments, R 6 is C 1 -, C 2 -, C 3 -, C 6 -, C 8 -, or C 10 -alkyl. In certain embodiments, methyl, ethyl, n-butyl, n-hexyl, n-octyl, or n-decyl. In some embodiments, R 6 is unbranched. In certain embodiments, R 6 is methyl or n-butyl. In some embodiments, R 6 is unsubstituted.
  • R 7 is unsubstituted. In certain embodiments, R 7 is -CH3 or has the following structure: In certain embodiments, R 7 has the following structure: In some embodiments, R 3 is optionally substituted C 1 -C 6 alkyl. In certain embodiments, R 3 is optionally substituted methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-hexyl. In some embodiments, R 3 is optionally substituted methyl. In some embodiments, R 3 is C 1 -C 6 alkyl substituted with one or more hydroxyl.
  • R 3 is C 2 - or C 4 - alkyl substituted with one or more hydroxyl. In certain embodiments, R 3 is unsubstituted. In some embodiments, R 3 is hydrogen.
  • X is
  • the compound has the following Structure (II): or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof.
  • n2 is 3, 4, or 5.
  • X is In some such embodiments, the compound has the following Structure (III): or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof.
  • n3 is 0 or 1.
  • n4 is 2 or 3.
  • n5 is 3. In some embodiments, n1 is 3, 4, or 5. In certain embodiments, n1 is 2. In some embodiments, the compound has one of the structures set forth in the Table below or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof:
  • the cationic lipid is selected from any one of Compound Nos. I- 1 to I-63 as specifically exemplified in Table 1 of PCT Publication No. WO2024/233387, including any racemic mixtures of enantiomer pairs of such compounds as described therein, which Table, compounds and racemic mixtures thereof listed therein are hereby incorporated herein by reference for all purposes.
  • 1, 2, 3, 4, 5, or more of the foregoing cationic lipids may be excluded from the LNPs of the present disclosure.
  • the RNA-LNPs comprise a cationic lipid, a RNA molecule as described herein and one or more of neutral lipids, steroids, pegylated lipids, or combinations thereof.
  • the cationic lipid is present in the LNP in an amount such as at least, at most, exactly, or between (inclusive or exclusive) of, or about 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59 or 60 mole percent (mol%).
  • two or more cationic lipids are incorporated within the LNP. If more than one cationic lipid is incorporated within the LNP, the foregoing percentages apply to the combined cationic lipids.
  • the LNP comprises a combination or mixture of any the lipids described above. ii. POLYMER CONJUGATED LIPID
  • the LNPs comprise a polymer conjugated lipid.
  • the term “polymer conjugated lipid” refers to a molecule comprising both a lipid portion and a polymer portion.
  • An example of a polymer conjugated lipid is a pegylated lipid (e.g., polyethylene glycol-lipid, PEG- lipid).
  • the LNP comprises an additional, stabilizing lipid that is a pegylated lipid.
  • pegylated lipid refers to a molecule comprising both a lipid portion and a polyethylene glycol portion.
  • Pegylated lipids include, but are not limited to, PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramides (e.g., PEG-CerC14 or PEG-CerC20), PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, 2-[(polyethylene glycol)-2000]-N,N-ditetradecylacetamide, and mixtures thereof.
  • PEG-modified phosphatidylethanolamine PEG-modified phosphatidic acid
  • PEG-modified ceramides e.g., PEG-CerC14 or PEG-CerC20
  • PEG-modified dialkylamines e.g., PEG-modified diacylglycerols
  • PEG-modified dialkylglycerols 2-[(polyethylene glycol)
  • polyethylene glycol-lipids include PEG-c-DOMG, PEG-c-DMA, PEG-DSG, PEG-DPG, and PEG-s-DMG (1-(monomethoxy-polyethyleneglycol)-2,3- dimyristoylglycerol).
  • the polyethylene glycol-lipid is N-[(methoxy polyethylene glycol) 2 000)carbamoyl]-1,2-dimyristyloxlpropyl-3-amine (PEG-c-DMA).
  • the polyethylene glycol-lipid is PEG-2000-DMG.
  • the polyethylene glycol-lipid is PEG- c-DOMG.
  • the LNPs comprise a PEGylated diacylglycerol (PEG-DAG) such as 1-(monomethoxy-polyethyleneglycol)-2,3-dimyristoylglycerol (PEG-DMG), a PEGylated phosphatidylethanolamine (PEG-PE), a PEG succinate diacylglycerol (PEG-S-DAG) such as 4- O-(2′,3′-di(tetradecanoyloxy)propyl-1-O-((O-methoxy(polyethoxy)ethyl)butanedioate (PEG-S- DMG), a PEGylated ceramide (PEG-cer), or a PEG dialkoxypropylcarbamate such as co- methoxy(polyethoxy)ethyl-N-(2,3di(tetradecanoxy)propyl)carbamate or 2,3- di(tetrade
  • PEG-lipids are disclosed in, e.g., U.S.9,737,619, the full disclosures of which is herein incorporated by reference in its entirety for all purposes. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing pegylated lipids may be excluded from the LNPs of the present disclosure.
  • the composition comprises a pegylated lipid having the following structure: or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: R 8 and R 9 are each independently a straight or branched, saturated or unsaturated alkyl chain containing from 10 to 30 carbon atoms, wherein the alkyl chain is optionally interrupted by one or more ester bonds; and w has a mean value ranging from 30 to 60. In some aspects, R 8 and R 9 are each independently straight, saturated alkyl chains containing from 12 to 16 carbon atoms. In some aspects, w has a mean value ranging from 43 to 53. In other aspects, the average w is or is about 45. In other different embodiments, the average w is or is about 49.
  • the lipid nanoparticles comprise a polymer conjugated lipid.
  • the lipid nanoparticle comprises 2-[(polyethylene glycol)-2000]-N,N- ditetradecylacetamide (ALC-0159), having the formula:
  • the molar ratio of the cationic lipid to the pegylated lipid ranges from or from about 100:1 to about 20:1, e.g., 20:1, 25:1, 30:1, 35:1, 40:1, 45:1, 50:1, 55:1, 60:1, 65:1, 70:1, 75:1, 80:1, 85:1, 90:1, 95:1, or 100:1, or any range or value derivable therein.
  • the PEG-lipid is or is not present in the LNP in an amount from or from about 1 to about 10 mole percent (mol %) (e.g., at least, at most, exactly, or between (inclusive or exclusive) of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 mol %), relative to the total lipid content of the nanoparticle.
  • the ratio of PEG in the lipid nanoparticle formulations may be increased or decreased and/or the carbon chain length of the PEG lipid may be modified to alter the pharmacokinetics and/or biodistribution of the lipid nanoparticle formulations.
  • the LNP comprises one or more additional lipids or lipid-like materials that stabilize particles during their formation.
  • Suitable stabilizing or structural lipids include non- cationic lipids, e.g., neutral lipids and anionic lipids.
  • optimizing the formulation of LNPs by addition of other hydrophobic moieties, such as cholesterol and lipids, in addition to an ionizable/cationic lipid or lipid-like material may enhance particle stability and efficacy of nucleic acid delivery.
  • an “anionic lipid” refers to any lipid that is negatively charged at a selected pH.
  • neutral lipid refers to any one of a number of lipid species that exist in either an uncharged or neutral zwitterionic form at physiological pH.
  • additional lipids comprise one of the following neutral lipid components: (1) a phospholipid, (2) cholesterol or a derivative thereof; or (3) a mixture of a phospholipid and cholesterol or a derivative thereof.
  • Representative neutral lipids include phosphatidylcholines, phosphatidylethanolamines, phosphatidylglycerols, phosphatidic acids, phosphatidylserines, ceramides, sphingomyelins, dihydro-sphingomyelins, cephalins, and cerebrosides.
  • Exemplary phospholipids include, for example, phosphatidylcholines, e.g., diacylphosphatidylcholines, such as distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dimyristoylphosphatidylcholine (DMPC), dipentadecanoylphosphatidylcholine, dilauroylphosphatidylcholine, dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), diarachidoylphosphatidylcholine (DAPC), dibehenoylphosphatidylcholine (DBPC), ditricosanoylphosphatidylcholine (DTPC), dilignoceroylphatidylcholine (DLPC), palmitoyloleoy
  • 1, 2, 3, 4, 5, or more of the foregoing neutral lipids may be excluded from the LNPs of the present disclosure.
  • the neutral lipid is 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), having the formula:
  • the LNPs comprise a neutral lipid, and the neutral lipid comprises one or more of DSPC, DPPC, DMPC, DOPC, POPC, DOPE, and/or SM.
  • 1, 2, 3, 4, 5, or more of the foregoing neutral lipids may be excluded from the LNPs of the present disclosure.
  • the LNPs further comprise a steroid or steroid analogue.
  • cholesterol derivatives include, but are not limited to, cholestanol, cholestanone, cholestenone, coprostanol, cholesteryl-2′-hydroxyethyl ether, cholesteryl-4′-hydroxybutyl ether, tocopherol and derivatives thereof, and mixtures thereof.
  • 1, 2, 3, 4, 5, or more of the foregoing cholesterol derivatives may be excluded from the LNPs of the present disclosure.
  • the cholesterol has the formula: Without being bound by any theory, the amount of the at least one cationic lipid compared to the amount of the at least one additional lipid may affect important nucleic acid particle characteristics, such as charge, particle size, stability, tissue selectivity, and bioactivity of the nucleic acid.
  • the non-cationic lipid may comprise from or from about 0 mol % to about 90 mol %, from or from about 0 mol % to about 80 mol %, from or from about 0 mol % to about 70 mol %, from or from about 0 mol % to about 60 mol %, or from or from about 0 mol % to about 50 mol %, of the total lipid present in the particle.
  • neutral lipid e.g., one or more phospholipids and/or cholesterol
  • the non-cationic lipid e.g., neutral lipid (e.g., one or more phospholipids and/or cholesterol)
  • VI. CHARACTERIZATION AND ANALYSIS OF RNA MOLECULE The RNA molecule described herein may be analyzed and characterized using various methods. Analysis may be performed before and/or after capping.
  • analysis may be performed before and/or after poly-A capture-based affinity purification.
  • analysis may be performed before and/or after additional purification steps, e.g., anion exchange chromatography and the like.
  • RNA template quality may be determined using a Bioanalyzer chip-based electrophoresis system.
  • RNA template purity is analyzed using analytical reverse phase HPLC.
  • Capping efficiency may be analyzed using, e.g., total nuclease digestion followed by MS/MS quantitation of the dinucleotide cap species vs. uncapped GTP species.
  • In vitro efficacy may be analyzed by, e.g., transfecting an RNA molecule into a human cell line.
  • Protein expression of the polypeptide of interest may be quantified using methods such as ELISA and/or flow cytometry.
  • Immunogenicity may be analyzed by, e.g., transfecting RNA molecules into cell lines that indicate innate immune stimulation, e.g., PBMCs.
  • Cytokine induction may be analyzed using, e.g., methods such as ELISA to quantify a cytokine, e.g., Interferon- ⁇ .
  • Biodistribution may be analyzed by, e.g., bioluminescence measurements. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing analytic methods may be excluded.
  • an RNA polynucleotide disclosed herein is characterized in that, when assessed in an organism administered a composition or medical preparation comprising an RNA polynucleotide, elevated expression of a gene of interest (e.g., an antigen); increased duration of expression (e.g., prolonged expression) of a gene of interest (e.g., an antigen); elevated expression and increased duration of expression (e.g., prolonged expression) of a gene of interest (e.g., an antigen); decreased interaction with IFIT1 of an RNA polynucleotide; and/or increased translation of an RNA polynucleotide; is observed relative to an appropriate reference.
  • a gene of interest e.g., an antigen
  • increased duration of expression e.g., prolonged expression
  • elevated expression and increased duration of expression e.g., prolonged expression
  • decreased interaction with IFIT1 of an RNA polynucleotide e.g., an antigen
  • a reference comprises an organism administered an otherwise similar RNA polynucleotide without a m7(3′OMeG)(5′)ppp(5′)(2′OMeAi)pG 2 cap.
  • a reference comprises an organism administered an otherwise similar RNA polynucleotide without a cap proximal sequence disclosed herein.
  • a reference comprises an organism administered an otherwise similar RNA polynucleotide with a self-hybridizing sequence.
  • elevated expression is determined at least 24 hours, at least 48 hours, at least 72 hours, at least 96 hours, or at least 120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at least 24 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at least 48 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at least 72 hours after administration of a composition or medical preparation comprising an RNA polynucleotide.
  • elevated expression is determined at least 96 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at least 120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at or at about 24-120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide.
  • elevated expression is determined at or at about 24-110 hours, 24-100 hours, 24- 90 hours, 24-80 hours, 24-70 hours, 24-60 hours, 24-50 hours, 24-40 hours, 24-30 hours, 30-120 hours, 40-120 hours, 50-120 hours, 60-120 hours, 70-120 hours, 80-120 hours, 90-120 hours, 100-120 hours, or 110-120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide.
  • expression of a gene of interest e.g., an antigen
  • expression of a gene of interest is or is not elevated at least 2-fold to at least 10-fold.
  • expression of a gene of interest e.g., an antigen is elevated at least 2-fold.
  • expression of a gene of interest is elevated at least 3-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated at least 4-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated at least 6-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated at least 8-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated at least 10-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated or elevated about 2-fold to about 50-fold.
  • expression of a gene of interest is elevated or elevated about 2-fold to about 45-fold, about 2-fold to about 40-fold, about 2-fold to about 30-fold, about 2-fold to about 25-fold, about 2-fold to about 20-fold, about 2-fold to about 15-fold, about 2-fold to about 10-fold, about 2-fold to about 8-fold, about 2-fold to about 5-fold, about 5-fold to about 50-fold, about 10-fold to about 50-fold, about 15-fold to about 50-fold, about 20-fold to about 50-fold, about 25-fold to about 50-fold, about 30-fold to about 50-fold, about 40- fold to about 50-fold, or about 45-fold to about 50-fold.
  • a gene of interest e.g., an antigen
  • expression of a gene of interest is or is not elevated at least, at most, exactly, or between (inclusive or exclusive) of 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 11-fold, 12-fold, 13- fold, 14-fold, 15-fold, 16-fold, 17-fold, 18-fold, 19-fold, 20-fold, 21-fold, 22-fold, 23-fold, 24-fold, 25-fold, 26-fold, 27-fold, 28-fold, 29-fold, 30-fold, 31-fold, 32-fold, 33-fold, 34-fold, 35-fold, 36- fold, 37-fold, 38-fold, 39-fold, 40-fold, 41-fold, 42-fold, 43-fold, 44-fold, 45-fold, 46-fold, 47-fold, 48-fold, 49-fold, or 50-fold, or any range or value derivable therein.
  • elevated expression (e.g., increased duration of expression) of a gene of interest persists for at least, at most, exactly, or between (inclusive or exclusive) of 24 hours, 48 hours, 72 hours, 96 hours, or 120 hours after administration of a composition or a medical preparation comprising an RNA polynucleotide.
  • elevated expression of a gene of interest persists for at least 24 hours after administration.
  • elevated expression of a gene of interest persists for at least 48 hours after administration.
  • elevated expression of a gene of interest (e.g., an antigen) persists for at least 72 hours after administration.
  • elevated expression of a gene of interest persists for at least 96 hours after administration. In some aspects, elevated expression of a gene of interest (e.g., an antigen) persists for at least 120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression of a gene of interest (e.g., an antigen) persists for or for about 24-120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide.
  • elevated expression persists for or for about 24-110 hours, 24-100 hours, 24-90 hours, 24-80 hours, 24-70 hours, 24-60 hours, 24-50 hours, 24-40 hours, 24-30 hours, 30-120 hours, 40-120 hours, 50-120 hours, 60-120 hours, 70-120 hours, 80- 120 hours, 90-120 hours, 100-120 hours, or 110-120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide.
  • the immunogenic compositions of the disclosure are to prevent RSV infections by inoculating or vaccination of a subject.
  • the immunogenic compositions immunize the subject against RSV up to 1 year (e.g., for a single RSV season).
  • the immunogenic compositions immunize the subject against RSV for up to 2 years.
  • the immunogenic compositions immunize the subject against RSV for more than 2 years.
  • the immunogenic compositions immunize the subject against RSV for more than 3 years.
  • the immunogenic compositions immunize the subject against RSV for more than 4 years.
  • the immunogenic compositions immunize the subject against RSV for 5-10 years. A.
  • IMMUNOASSAYS The present disclosure includes the implementation of serological assays to evaluate whether and to what extent an immune response is induced or evoked by compositions of the disclosure.
  • immunoassays encompassed by the present disclosure include, but are not limited to, those described in U.S. Patent 4,367,110 (double monoclonal antibody sandwich assay) and U.S. Patent 4,452,901 (western blot).
  • Other assays include immunoprecipitation of labeled ligands and immunocytochemistry, both in vitro and in vivo.
  • Immunoassays generally are binding assays.
  • the immunoassays are the various types of enzyme linked immunosorbent assays (ELISAs) and radioimmunoassays (RIA) known in the art. Immunohistochemical detection using tissue sections is also particularly useful.
  • ELISAs enzyme linked immunosorbent assays
  • RIA radioimmunoassays
  • Immunohistochemical detection using tissue sections is also particularly useful.
  • antibodies or antigens are immobilized on a selected surface, such as a well in a polystyrene microtiter plate, dipstick, or column support. Then, a test composition suspected of containing the desired antigen or antibody, such as a clinical sample, is added to the wells. After binding and washing to remove non-specifically bound immune complexes, the bound antigen or antibody may be detected.
  • Detection is generally achieved by the addition of another antibody, specific for the desired antigen or antibody, that is linked to a detectable label. This type of ELISA is known as a “sandwich ELISA.” Detection also may be achieved by the addition of a second antibody specific for the desired antigen, followed by the addition of a third antibody that has binding affinity for the second antibody, with the third antibody being linked to a detectable label. Competition ELISAs are also possible implementations in which test samples compete for binding with known amounts of labeled antigens or antibodies. The amount of reactive species in the unknown sample is determined by mixing the sample with the known labeled species before or during incubation with coated wells.
  • ELISAs have certain features in common, such as coating, incubating or binding, washing to remove non-specifically bound species, and detecting the bound immune complexes.
  • Antigen or antibodies may also be linked to a solid support, such as in the form of plate, beads, dipstick, membrane, or column matrix, and the sample to be analyzed is applied to the immobilized antigen or antibody. In coating a plate with either antigen or antibody, one will generally incubate the wells of the plate with a solution of the antigen or antibody, either overnight or for a specified period.
  • the wells of the plate will then be washed to remove incompletely- adsorbed material. Any remaining available surfaces of the wells are then “coated” with a nonspecific protein that is antigenically neutral with regard to the test antisera. These include bovine serum albumin (BSA), casein, and solutions of milk powder. The coating allows for blocking of nonspecific adsorption sites on the immobilizing surface and thus reduces the background caused by nonspecific binding of antisera onto the surface.
  • BSA bovine serum albumin
  • casein casein
  • solutions of milk powder solutions of milk powder.
  • the coating allows for blocking of nonspecific adsorption sites on the immobilizing surface and thus reduces the background caused by nonspecific binding of antisera onto the surface.
  • a method of detecting the presence of infections involves the steps of obtaining a sample suspected of being infected by one or more RSV strains, such as a sample taken from an individual, for example, from one’s blood, saliva, tissues, bone, muscle, cartilage, or skin.
  • diagnostic assays utilizing the polypeptides, proteins, and/or peptides of the present disclosure may be carried out to detect the presence of RSV, and such assay techniques for determining such presence in a sample are well known to those skilled in the art and include methods such as radioimmunoassay, western blot analysis and ELISA assays.
  • a method of diagnosing an infection wherein a sample suspected of being infected with RSV has added to it the polypeptide, protein, or peptide, in accordance with the present disclosure, and RSV is indicated by antibody binding to the polypeptides, proteins, and/or peptides, or polypeptides, proteins, and/or peptides binding to the antibodies in the sample.
  • RNA molecules encoding RSV polypeptides, proteins, and/or peptides in accordance with the disclosure may be used for to treat, prevent, or reduce the severity of illness due to RSV infection (e.g., active or passive immunization) or for use as research tools.
  • RNA molecules encoding RSV preF polypeptides, RNA-LNPs and compositions thereof, confer protective immunity to a subject.
  • Protective immunity refers to a body’s ability to mount a specific immune response that protects the subject from developing a particular disease or condition that involves the agent against which there is an immune response.
  • An immunogenically effective amount is capable of conferring protective immunity to the subject.
  • the RNA molecules encoding RSV polypeptides, RNA- LNPs and compositions thereof of the present disclosure may be used to induce a balanced immune response against RSV comprising both cellular and humoral immunity, without many of the risks associated with attenuated virus vaccination.
  • a subject thus treated would donate plasma from which hyperimmune globulin would then be obtained, via conventional plasma-fractionation methodology, and administered to another subject in order to impart resistance against or to treat RSV infection.
  • epitopes and antigenic determinant are used interchangeably to refer to a site on an antigen to which B and/or T cells respond or recognize.
  • B-cell epitopes may be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • Methods of determining spatial conformation of epitopes include, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols (1996).
  • Antibodies that recognize the same epitope may be identified in a simple immunoassay showing the ability of one antibody to block the binding of another antibody to a target antigen.
  • T-cells recognize continuous epitopes of about nine amino acids for CD8 cells or about 13-15 amino acids for CD4 cells.
  • T cells that recognize the epitope may be identified by in vitro assays that measure antigen-dependent proliferation, as determined by 3H- thymidine incorporation by primed T cells in response to an epitope (Burke et al., 1994), by antigen-dependent killing (cytotoxic T lymphocyte assay, Tigges et al., 1996) or by cytokine secretion.
  • the presence of a cell-mediated immunological response may be determined by proliferation assays (CD4 (+) T cells) or CTL (cytotoxic T lymphocyte) assays.
  • the relative contributions of humoral and cellular responses to the protective or therapeutic effect of an immunogenic composition may be distinguished by separately isolating IgG and T-cells from an immunized syngeneic animal and measuring protective or therapeutic effect in a second subject.
  • antibody or “immunoglobulin” are used interchangeably and refer to any of several classes of structurally related proteins that function as part of the immune response of an animal or recipient, which proteins include IgG, IgD, IgE, IgA, IgM and related proteins. Under normal physiological conditions antibodies are found in plasma and other body fluids and in the membrane of certain cells and are produced by lymphocytes of the type denoted B cells or their functional equivalent.
  • RNA molecules and/or RNA-LNPs disclosed herein may be administered in a pharmaceutical composition or a medicament and may be administered in the form of any suitable pharmaceutical composition.
  • a pharmaceutical composition is for therapeutic and/or prophylactic treatment.
  • the disclosure relates to a composition for administration to a host.
  • the host is a human.
  • the host is a non-human.
  • Formulations of the compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology.
  • preparatory methods include the step of bringing an active ingredient (e.g., RNA molecules and/or RNA-LNPs) into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi-dose unit.
  • an active ingredient e.g., RNA molecules and/or RNA-LNPs
  • a pharmaceutical composition or formulation in accordance with the present disclosure can be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a composition in accordance with the disclosure will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 100%, e.g., between 0.5 and 50%, between 1-30%, between 5-80%, at least 80% (w/w), or at least, at most, exactly, or between (inclusive or exclusive) any two of 0.1%, 0.15%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.55%, 0.6%, 0.65%, 0.7%, 0.75%, 0.8%, 0.85%, 0.9%, 0.95%, 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% (w/w) active ingredient.
  • 0.1% and 100% e.g., between 0.5 and 50%, between 1-30%, between 5-80%, at least 80% (w/w), or at least, at most, exactly, or between (inclusive or exclusive) any two of 0.1%, 0.15%, 0.2%, 0.25%, 0.3%,
  • RNA molecules and/or RNA-LNPs disclosed herein may be administered in a pharmaceutical composition which may be formulated into preparations in solid, semi-solid, liquid, lyophilized, frozen, and/or gaseous forms.
  • an RNA molecule and/or RNA-LNPs disclosed herein may be administered in a pharmaceutical composition which may comprise a pharmaceutically acceptable carrier and may optionally comprise one or more adjuvants, stabilizers, salts, buffers, preservatives, and optionally other therapeutic agents.
  • a pharmaceutical composition disclosed herein comprises one or more pharmaceutically acceptable carriers, diluents and/or excipients.
  • pharmaceutical compositions do not include an adjuvant (e.g., they are adjuvant free).
  • excipient refers to a substance which may be present in a pharmaceutical composition of the present disclosure but is not an active ingredient.
  • excipients include without limitation, carriers, diluents (e.g., solvents, dispersion media, and/or other liquid vehicles, dispersion or suspension aids), granulating and/or dispersing agents, surface active agents, isotonic agents, thickening and/or emulsifying agents, preservatives, binders, lubricants and/or oil, coloring, sweetening and/or flavoring agents, stabilizers, antioxidants, antimicrobial and/or antifungal agents, osmolality adjusting agents, pH adjusting agents, buffers, chelants, cryoprotectants, and/or bulking agents.
  • diluents e.g., solvents, dispersion media, and/or other liquid vehicles, dispersion or suspension aids
  • granulating and/or dispersing agents e.g., surface active agents, isotonic agents, thickening and/or emulsifying agents, preservatives, binders, lubricants and/
  • carrier refers to a component which may be natural, synthetic, organic, or inorganic, in which the active component is combined in order to facilitate, enhance and/or enable administration of the pharmaceutical composition.
  • a carrier as used herein may be one or more compatible solid or liquid fillers, diluents or encapsulating substances, which are suitable for administration to subject.
  • Suitable carriers include, without limitation, sterile water, Ringer’s solution, Ringer’s lactate solution, sterile sodium chloride solution, isotonic saline, polyalkylene glycols, hydrogenated naphthalenes and, in particular, biocompatible lactide polymers, lactide/glycolide copolymers or polyoxyethylene/polyoxy-propylene copolymers.
  • the pharmaceutical composition of the present disclosure includes sodium chloride.
  • 1, 2, 3, 4, 5, or more of the foregoing carriers may be excluded from the pharmaceutical compositions disclosed herein.
  • the term “diluent” relates a diluting and/or thinning agent.
  • diluent includes any one or more of fluid, liquid or solid suspension and/or mixing media.
  • suitable diluents for use in a pharmaceutical compositions of the present disclosure include, without limitation, ethanol, glycerol, saline, water, calcium or sodium carbonate, calcium phosphate, calcium hydrogen phosphate, sodium phosphate, lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, etc., and/or combinations thereof.
  • 1, 2, 3, 4, 5, or more of the foregoing diluents may be excluded from the pharmaceutical compositions disclosed herein.
  • suitable granulating and/or dispersing agents include, without limitation, starches, pregelatinized starches, or microcrystalline starch, alginic acid, guar gum, agar, poly(vinyl-pyrrolidone), (providone), cross-linked poly(vinyl-pyrrolidone) (crospovidone), cellulose, methylcellulose, carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), magnesium aluminum silicate (VEEGUM®), sodium lauryl sulfate, etc., and/or combinations thereof.
  • 1, 2, 3, 4, 5, or more of the foregoing granulating and/or dispersing agents may be excluded from the pharmaceutical compositions disclosed herein.
  • Suitable surface active agents for use in a pharmaceutical compositions of the present disclosure include, without limitation, natural emulsifiers (e.g., acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), sorbitan fatty acid esters (e.g., polyoxyethylene sorbitan monooleate [TWEEN®80], sorbitan monopalmitate [SPAN®40], glyceryl monooleate, polyoxyethylene esters, polyethylene glycol fatty acid esters (e.g., CREMOPHOR®), polyoxyethylene ethers (e.g., polyoxyethylene lauryl ether [BRIJ®30]), PLUORINC®F 68, POLOXAMER®188, etc.
  • natural emulsifiers e.g., acacia, a
  • 1, 2, 3, 4, 5, or more of the foregoing preservatives may be excluded from the pharmaceutical compositions disclosed herein.
  • suitable antimicrobial and/or antifungal agents for use in a pharmaceutical compositions of the present disclosure include, without limitation, benzalkonium chloride, benzethonium chloride, methyl paraben, ethyl paraben, propyl paraben, butyl paraben, benzoic acid, hydroxybenzoic acid, potassium or sodium benzoate, potassium or sodium sorbate, sodium propionate, sorbic acid, etc., and combinations thereof.
  • 1, 2, 3, 4, 5, or more of the foregoing antimicrobial and/or antifungal agents may be excluded from the pharmaceutical compositions disclosed herein.
  • Suitable binders for use in a pharmaceutical compositions of the present disclosure include, without limitation, starch, gelatin, sugars (e.g., sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol), amino acids (e.g., glycine), natural and synthetic gums (e.g., acacia, sodium alginate), ethylcellulose, hydroxyethylcellulose, hydroxypropyl methylcellulose, etc., and combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing binders may be excluded from the pharmaceutical compositions disclosed herein.
  • cryoprotectants may be excluded from the pharmaceutical compositions disclosed herein.
  • suitable bulking agents include, without limitation, sucrose, trehalose, mannitol, glycine, lactose, raffinose, and combinations thereof.
  • 1, 2, 3, 4, 5, or more of the foregoing bulking agents may be excluded from the pharmaceutical compositions disclosed herein.
  • Compositions can be formulated using one or more excipients (e.g., one or more carriers and/or diluents) to, e.g.: (1) increase stability; (2) increase cell transfection; (3) permit the sustained and/or delayed release (e.g., from a depot formulation); (4) alter the biodistribution (e.g., target to specific tissues and/or cell types); (5) increase the translation of encoded protein in vivo; and/or (6) alter the release profile of encoded protein (antigen) in vivo.
  • excipients e.g., one or more carriers and/or diluents
  • compositions comprises an RNA molecule comprising an open reading frame encoding an immunogenic polypeptide.
  • immunogenic polypeptide comprises a RSV antigen.
  • RSV antigen is a RSV F protein or a fragment or variant thereof.
  • the composition comprises an RNA molecule comprising an open reading frame encoding a full-length RSV F protein.
  • the encoded immunogenic polypeptide is a truncated RSV F protein.
  • the encoded immunogenic polypeptide is a variant of a RSV F protein.
  • the encoded immunogenic polypeptide is a fragment of a RSV F protein.
  • a pharmaceutical composition comprises an RNA molecule (e.g., polynucleotide) disclosed herein formulated with a lipid-based delivery system.
  • the composition includes a lipid-based delivery system (e.g., LNPs) (e.g., a lipid-based vaccine), which delivers a nucleic acid molecule to the interior of a cell, where it may then replicate, inhibit protein expression of interest, and/or express the encoded polypeptide of interest.
  • the delivery system may have adjuvant effects which enhance the immunogenicity of an encoded antigen.
  • the composition comprises at least one RNA molecule encoding a RSV polypeptide complexed with, encapsulated in, and/or formulated with one or more lipids, and forming lipid nanoparticles (LNPs), liposomes, lipoplexes and/or nanoliposomes.
  • LNPs lipid nanoparticles
  • the composition comprises a lipid nanoparticle.
  • the present disclosure concerns compositions comprising one or more lipids associated with a nucleic acid or a polypeptide/peptide (e.g., RSV RNA-LNPs).
  • an immunogenic composition may be multivalent, wherein the immunogenic composition comprises (i) a first ribonucleic acid (RNA) polynucleotide having an open reading frame encoding a first antigen, said antigen including at least one RSV F protein polypeptide or an immunogenic fragment thereof, and (ii) a second RNA polynucleotide having an open reading frame encoding a second antigen, said second antigen including at least one RSV F protein polypeptide or an immunogenic fragment thereof, wherein the first and second RNA polynucleotides are formulated in a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • the first and second antigens include RSV F protein (F) as described herein, or an immunogenic fragment or variant thereof.
  • the first antigen includes an RSV F protein from a different subtype of RSV to the RSV antigenic polypeptide or an immunogenic fragment thereof of the second antigen.
  • the first and second RNA polynucleotides are formulated in a lipid nanoparticle.
  • the immunogenic composition is a bivalent immunogenic composition comprising a first RSV molecule encoding an F protein as described herein which is RSV subtype A and a second RSV molecule encoding an F protein as described herein which is RSV subtype B.
  • the ratio of the first RSV molecule to the second RSV molecule in the immunogenic composition is 1:1.
  • the multivalent immunogenic composition may be a (i) "pre-mix” formulation wherein two or more RNA polynucleotides are combined in equal ratio followed by co-formulation into LNPs, or (ii) a "post- mix” formulation wherein each RNA polynucleotide is individually formulated into LNPs and the two or more LNPs are combined either in equal ratio or not in equal ratio.
  • the immunogenic composition including a lipid-based delivery system may further include one or more salts and/or one or more pharmaceutically acceptable surfactants, preservatives, carriers, diluents, and/or excipients, in some cases.
  • the immunogenic composition including a lipid-based delivery system further includes a pharmaceutically acceptable vehicle.
  • each of a buffer, stabilizing agent, and optionally a salt may be included in the immunogenic composition including a lipid-based delivery system.
  • any one or more of a buffer, stabilizing agent, salt, surfactant, preservative, and excipient may be excluded from the immunogenic composition including a lipid-based delivery system.
  • the immunogenic composition including a lipid-based delivery system further comprises a stabilizing agent.
  • the stabilizing agent comprises sucrose, mannose, sorbitol, raffinose, trehalose, mannitol, inositol, sodium chloride, arginine, lactose, hydroxyethyl starch, dextran, polyvinylpyrolidone, glycine, or a combination thereof.
  • 1, 2, 3, 4, 5, or more of the foregoing stabilizing agents may be excluded from the immunogenic compositions disclosed herein.
  • the stabilizing agent is a disaccharide, or sugar.
  • the stabilizing agent is sucrose.
  • the stabilizing agent is trehalose. In a further aspect, the stabilizing agent is a combination of sucrose and trehalose. In some aspects, the total concentration of the stabilizing agent(s) in the composition is or is about 5% to about 10% w/v. For example, the total concentration of the stabilizing agent may or may not be equal to at least, at most, exactly, or between (inclusive or exclusive) of 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, or 20% w/v or any range or value derivable therein.
  • the stabilizing agent concentration includes, but is not limited to, a concentration of or of about 10 mg/mL to about 400 mg/mL, about 100 mg/mL to about 200 mg/mL, about 100 mg/mL to about 150 mg/mL, about 100 mg/mL to about 140 mg/mL, about 100 mg/mL to about 130 mg/mL, about 100 mg/mL to about 120 mg/mL, about 100 mg/mL to about 110 mg/mL, or about 100 mg/mL to about 105 mg/mL.
  • the concentration of the stabilizing agent is or is not equal to at least, at most, exactly, or between (inclusive or exclusive) of 10 mg/mL, 20 mg/mL, 50 mg/mL, 100 mg/mL, 101 mg/mL, 102 mg/mL, 103 mg/mL, 104 mg/mL, 105 mg/mL, 106 mg/mL, 107 mg/mL, 108 mg/mL, 109 mg/mL, 110 mg/mL, 150 mg/mL, 200 mg/mL, 300 mg/mL, 400 mg/mL, or more.
  • the mass amount of the stabilizing agent and the mass amount of the RNA are in a specific ratio.
  • the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 5000. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 2000. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 1000. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 500. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 100. In another aspect, the ratio of the mass amount of the stabilizing agent and the pharmaceutical substance is no greater than 50. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 10.
  • the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 1. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 0.5. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 0.1. In another aspect, the stabilizing agent and RNA comprise a mass ratio of or of about 200 – 2000 of the stabilizing agent : 1 of the RNA. In some aspects, the immunogenic composition including a lipid-based delivery system further comprises a buffer.
  • buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, d-gluconic acid, calcium glycerophosphate, calcium lactate, calcium lactobionate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, Tris hydrochloride (HCl), amino-sulfonate buffers (HC
  • the buffer is a HEPES buffer, a Tris buffer, and/or a PBS buffer.
  • the buffer is Tris buffer.
  • the buffer is a HEPES buffer.
  • the buffer is a PBS buffer.
  • the liquid RNA-LNP immunogenic composition comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably of or of about 0.06 mg/mL, encapsulated in a LNP, and further comprising or comprising about 10 mM Tris buffer and 300 mM sucrose at a pH of or of about 7.4.
  • the RNA-LNP immunogenic composition (pre-lyophilized) comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in a LNP, and further comprising or comprising about 5 to 15 mM Tris buffer and 200 to 400 mM sucrose at a pH of or of about 7.0 to 8.0, and reconstituted with 0.9% sodium chloride diluent.
  • the RNA-LNP immunogenic composition (pre-lyophilized) comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably 0.15 mg/mL, encapsulated in a LNP, and further comprising or comprising about 10 mM Tris buffer and 300 mM sucrose at a pH of or of about 7.4, and reconstituted with 0.9% sodium chloride diluent.
  • the RNA-LNP immunogenic composition (pre- lyophilized) comprises a first RNA molecule/polynucleotide encoding a RSV subtype A polypeptide as disclosed herein and a second RNA molecule/polynucleotide encoding a RSV subtype B as disclosed herein, each RNA molecule/polynucleotide at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably 0.09 mg/mL, encapsulated in LNPs with a lipid composition of a cationic lipid at a concentration of or of about 1.0 to 3.0 mg/mL, a PEGylated
  • the RNA-LNP immunogenic composition (pre-lyophilized) comprises a first RNA molecule/polynucleotide encoding a first RSV subtype A polypeptide as disclosed herein and a second RNA molecule/polynucleotide encoding a RSV subtype B as disclosed herein, each RNA molecule/polynucleotide at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably 0.09 mg/mL, encapsulated in LNPs with a lipid composition of ALC-0315 at a concentration of or of about 1.0 to 3.0 mg/mL, preferably about 2.56 mg/mL, ALC-05
  • a pharmaceutical composition described herein is an immunogenic composition for inducing an immune response.
  • an immunogenic composition is a vaccine.
  • the compositions described herein include at least one isolated nucleic acid or polypeptide molecule as described herein.
  • the immunogenic compositions comprise nucleic acids, and the immunogenic compositions are nucleic acid vaccines.
  • the immunogenic compositions comprise RNA (e.g., mRNA, saRNA), and vaccines are RNA vaccines.
  • the immunogenic compositions comprise DNA, and vaccines are DNA vaccines.
  • the immunogenic compositions comprise a polypeptide
  • vaccines are polypeptide vaccines.
  • Conditions and/or diseases that may be treated with the nucleic acid and/or peptide or polypeptide compositions include, but are not limited to, those caused and/or impacted by infection, cancer, rare diseases, and other diseases or conditions caused by overproduction, underproduction, and/or improper production of protein or nucleic acids.
  • the composition is substantially free of one or more impurities or contaminants and, for instance, includes nucleic acid or polypeptide molecules that are equal to at least, at most, exactly, or between (inclusive or exclusive) of 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% pure; at least 98% pure, or at least 99% pure.
  • the present disclosure includes methods for preventing, treating and/or ameliorating an infection, disease or condition in a subject, including administering to a subject an effective amount of an RNA molecule that includes at least one open reading frame encoding a polypeptide or composition described herein. As such, the disclosure contemplates vaccines for use in both active and passive immunization aspects.
  • Immunogenic compositions may be prepared from RNA molecules encoding polypeptide(s), such as RSV preF polypeptides.
  • immunogenic compositions are lyophilized for more ready formulation into a desired vehicle.
  • the preparation of vaccines that contain nucleic acid and/or peptide or polypeptide as active ingredients is generally well understood in the art, as exemplified by U.S. Patents 4,608,251; 4,601,903; 4,599,231; 4,599,230; 4,596,792; and 4,578,770, all of which are incorporated herein by reference.
  • suppositories may be formed from mixtures containing the active ingredient in the range of or of about 1% to about 2%.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing excipients may be excluded from an oral formulation. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and contain or contain about 10% to about 95% of active ingredient.
  • the polypeptide-encoding nucleic acid constructs and polypeptides may be formulated into a vaccine as neutral or salt forms.
  • Optically active (+) and (-), (R)- and (5)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization.
  • Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC).
  • HPLC high pressure liquid chromatography
  • an effective amount of a RSV RNA-LNP composition comprising at least one RSV RNA molecule having an open reading frame encoding at least one RSV antigenic polypeptide is a dose equivalent to an at least 2-fold reduction in a standard of care dose of a traditional composition against RSV .
  • an effective amount of a RSV RNA-LNP composition may or may not be a dose equivalent to a 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, 10-, 20-, 50-, 100-, 250-, 500-, or 1000-fold reduction in a standard of care dose of a traditional composition against RSV .
  • an anti-RSV antigenic polypeptide antibody titer produced in the subject is equivalent to an anti-RSV antigenic polypeptide antibody titer produced in a control subject administered the standard of care dose of a traditional composition against RSV .
  • a traditional composition against RSV refers to a composition other than the RNA molecules, RNA-LNPs and/or compositions described herein.
  • a traditional composition includes, but is not limited, to live microorganism vaccines, killed microorganism vaccines, attenuated vaccines, subunit vaccines, protein antigen vaccines containing recombinant protein produced in a heterologous expression system or purified from large amounts of the pathogenic organism, DNA vaccines, virus-like particle (VLP) vaccines containing viral capsid proteins (e.g., pre- and/or post-fusion F proteins) but lacking viral genome, etc.
  • a traditional vaccine is a vaccine that has achieved regulatory approval and/or is registered by a national drug regulatory body, for example the Food and Drug Administration (FDA) in the United States or the European Medicines Agency (EMA).
  • FDA Food and Drug Administration
  • EMA European Medicines Agency
  • an RNA molecule, RNA-LNP and/or composition described herein e.g., a RSV RNA-LNP composition comprising at least one RSV RNA molecule having an open reading frame encoding at least one RSV antigenic polypeptide
  • a RSV RNA-LNP composition comprising at least one RSV RNA molecule having an open reading frame encoding at least one RSV antigenic polypeptide
  • the term antibody titer refers to the amount of antigen-specific antibody produces in a subject, e.g., a human subject.
  • antibody titer is expressed as the inverse of the greatest dilution (in a serial dilution) that still gives a positive result.
  • antibody titer is determined or measured by enzyme-linked immunosorbent assay (ELISA). In exemplary embodiments, antibody titer is determined or measured by neutralization assay, e.g., by microneutralization assay. In certain aspects, antibody titer measurement is expressed as a ratio, such as 1:40, 1:100, etc.
  • ELISA enzyme-linked immunosorbent assay
  • neutralization assay e.g., by microneutralization assay.
  • antibody titer measurement is expressed as a ratio, such as 1:40, 1:100, etc.
  • an efficacious an RNA molecule, RNA-LNP and/or composition described herein produces an antibody titer of greater than 1:10, greater that 1:100, greater than 1:400, greater than 1:1000, greater than 1:2000, greater than 1:3000, greater than 1:4000, greater than 1:5000, greater than 1:6000, greater than 1:7500, or greater than 1:10000.
  • the antibody titer is produced or reached by 10 days following vaccination, by 20 days following vaccination, by 30 days following vaccination, by 40 days following vaccination, or by 50 or more days following vaccination.
  • the titer is produced or reached following a single dose of vaccine administered to the subject.
  • the titer is produced or reached following multiple doses, e.g., following a first and a second dose (e.g., a booster dose).
  • the methods disclosed herein may involve administering to the subject a RSV RNA-LNP composition comprising at least one RSV RNA molecule having an open reading frame encoding at least one RSV antigenic polypeptide, thereby inducing in the subject an immune response specific to RSV antigenic polypeptide, wherein the immune response in the subject is equivalent to an immune response in a subject administered with a traditional composition against the RSV that is or is not at least, at most, in between (inclusive or exclusive) any two of, or exactly 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, or 100 times the dosage level relative to the RNA composition.
  • the RNA molecule, RNA-LNP and/or composition is administered in a single dose. In some aspects, a second, third or fourth dose may be given. In some aspects, the RNA molecule, RNA-LNP and/or composition is administered in multiple doses. In some aspects, the RNA molecule, RNA-LNP and/or composition is administered intramuscularly (IM) or intradermally (ID).
  • IM intramuscularly
  • ID intradermally
  • the present disclosure further provides a kit comprising the RNA molecule, RNA-LNP, and/or composition.
  • the RNA molecule, RNA-LNP and/or composition described herein is administered to a subject that is less than about 1 years old, or about 1 years old to about 10 years old, or about 10 years old to about 20 years old, or about 20 years old to about 50 years old, or about 60 years old to about 70 years old, or older.
  • the subject is at least, at most, exactly, or between any two of less than 1 year of age, greater than 1 year of age, greater than 5 years of age, greater than 10 years of age, greater than 20 years of age, greater than 30 years of age, greater than 40 years of age, greater than 50 years of age, greater than 60 years of age, greater than 70 years of age, or older.
  • the subject is greater than 50 years of age.
  • the subject is at least, at most, exactly, or between (inclusive or exclusive) any two of about 4 months of age or older, about 1 year of age or older, about 5 years of age or older, about 10 years of age or older, about 20 years of age or older, about 30 years of age or older, about 40 years of age or older, about 50 years of age or older, about 60 years of age or older, about 70 years of age or older, or older.
  • the subject may be about 50 years of age or older.
  • the composition of the present invention further comprises a human metapneumovirus subtype A (hMPV A) F protein mutant or a nucleic acid encoding a hMPV A F protein mutant.
  • hMPV A human metapneumovirus subtype A
  • the composition of the present invention further comprises a human metapneumovirus subtype B (hMPV B) F protein mutant or a nucleic acid encoding a hMPV B F protein mutant.
  • the composition of the present invention further comprises both hMPV A and hMPV B F protein mutants or a nucleic acids encoding a hMPV A F protein mutant and a hMPV B F protein mutant.
  • hMPV F protein mutants are described in any of PCT Publication Nos.
  • the composition of the present invention further comprises a parainfluenza virus type 1 (PIV1) F protein mutant or a nucleic acid encoding a PIV1 F protein mutant.
  • the composition of the present invention further comprises a parainfluenza virus type 3 (PIV3) F protein mutant or a nucleic acid encoding a PIV3 F protein mutant.
  • the composition of the present invention further comprises both PIV1 and PIV3 F protein mutants or a nucleic acids encoding a PIV1 F protein mutant and a PIV3 F protein mutant.
  • PIV1 and PIV3 F protein mutants are described in any of PCT Publication Nos. WO2018081289, and WO2022207839, the full disclosures of which are herein incorporated by reference in their entirety for all purposes.
  • the vaccines provided by the present disclosure may be used together with one or more other vaccines. For example, in adults they may be used together with an influenza vaccine, Prevnar, tetanus vaccine, diphtheria vaccine, COVID19 vaccine and pertussis vaccine.
  • the RSV RNA-LNP vaccines of the present disclosure comprise nucleoside-modified mRNA encoding prefusion F (preF) polypeptide from RSV (modified RNA; modRNA).
  • the RSV RNA-LNP vaccines may comprise RNA comprising a single-stranded, 5′-capped and polyadenylated modified RNA that is translated after entering the cell.
  • the RNA comprises an open reading frame (ORF) that encodes variations of the RSV preF polypeptide.
  • the RNA may comprise structural elements, such as untranslated regions (UTRs), optimized for high efficacy of the RNA.
  • the RSV RNA-LNPs may comprise RNA as provided in Table 7 of Example 1 disclosed herein.
  • the RSV RNA-LNPs may comprise RNA as provided in Tables 1 to 3 of Example 6 disclosed herein.
  • the RNA may also comprise a substitution of 1-methyl-pseudouridine for uridine to decrease recognition of the vaccine RNA by innate immune sensors, such as toll-like receptors (TLRs) 7 and 8, resulting in decreased innate immune activation and increased protein translation.
  • TLRs toll-like receptors
  • the RNA molecules described herein are formulated/encapsulated into lipid nanoparticles (LNPs) to enable delivery of the RNA into host cells after, e.g., intramuscular (IM), intradermal (ID), or intranasal (IN) injection.
  • IM intramuscular
  • ID intradermal
  • IN intranasal
  • the LNP formulation may comprise two functional lipids, ALC- 0315 and ALC-0159, and two structural lipids, DSPC (1,2-distearoyl-sn-glycero-3- phosphocholine) and cholesterol. In some aspects, 1, 2, 3, or more of the forgoing lipids may be excluded from the LNP formulation.
  • the potency of RNA vaccines is optimized by LNP encapsulation, which protects the RNA from degradation by extracellular RNases and facilitates delivery in the cell. After IM injection of RSV RNA-LNP vaccines, the LNPs are taken up by the cells, and the RNA is released into the cytosol. In the cytosol, the RNA is translated, and the encoded viral antigen is produced.
  • the Examples herein demonstrate the RSV RNA-LNP vaccines of the present disclosure are immunogenic in mice and induce both humoral and cell mediated immune responses in mice.
  • Clinical studies of the present disclosure evaluate the safety, tolerability, and immunogenicity of RSV RNA-LNP vaccines against RSV.
  • the RSV RNA-LNPs vaccines may be indicated for active immunization for the prevention of acute lower respiratory infection (ALRI), including pneumonia and bronchitis caused by RSV for adults (e.g., ⁇ 45, ⁇ 50, ⁇ 55, ⁇ 60, ⁇ 70...etc.
  • RSV RNA-LNP vaccines may or may not be dosed in the range of or of about 15 ⁇ g, 30 ⁇ g, 45 ⁇ g, 60 ⁇ g, 75 ⁇ g, 90 ⁇ g, 100 ⁇ g or higher per dose with an injection volume of or of about 0.25 to 1 mL (e.g., of or of about 0.25, 0.5, 1 mL). Dilution with sterile 0.9% sodium chloride (normal saline) may be required.
  • the objectives of RSV RNA-LNP clinical studies may include, but are not limited to: -To describe the safety and tolerability profile of RSV RNA-LNP vaccines administered at selected dose levels and schedules in participants.
  • the efficacy (or effectiveness) against RSV of the RNA molecules encoding RSV polypeptides, RNA-LNPs and compositions thereof disclosed herein is or is not greater than 50% (e.g., at least, at most, exactly, or between any two of 50%, 60%, 70%, 80%, 90%, or more).
  • Vaccine efficacy may be assessed using standard analyses (see, e.g., Weinberg et al, J Infect Dis.2010 Jun 1;201(11): 1607-10).
  • vaccine efficacy may be measured by double-blind, randomized, clinical controlled trials such as those described herein.
  • AR disease attack rate
  • RR relative risk
  • Efficacy (ARU ⁇ ARV)/ARU x 100
  • Vaccine effectiveness is an assessment of how a vaccine (which may have already proven to have high vaccine efficacy) reduces disease in a population. This measure can assess the net balance of benefits and adverse effects of a vaccination program, not just the vaccine itself, under natural field conditions rather than in a controlled clinical trial. Vaccine effectiveness is proportional to vaccine efficacy (potency) but is also affected by how well target groups in the population are immunized, as well as by other non-vaccine-related factors that influence the real-world outcomes of hospitalizations, ambulatory visits, and/or costs. For example, a retrospective case control analysis may be used, in which the rates of vaccination among a set of infected cases and appropriate controls are compared.
  • the DNA was purified, spectrophotometrically quantified and in vitro-transcribed by T7 RNA polymerase in the presence of a trinucleotide cap1 analogue ((m 2 7,3′-O )Gppp(m 2’-O )ApG) (TriLink) and with N1-methylpseudouridine (m1 ⁇ ) replacing uridine (modified RNA (modRNA)).
  • TriLink trinucleotide cap1 analogue
  • m1 ⁇ N1-methylpseudouridine
  • modRNA modified RNA
  • the RSV RNA was generated from codon-optimized (CO) DNA for stabilization and superior protein expression.
  • a representative dataset of 64 constructs were tested for RSV-F expression (FIG.2) in both HEK and hDCs.
  • a majority of the UTR constructs yielded higher expression compared to the WHO UTRs (5UTR_1_RSV_3UTR_1) (5’UTR sequence SEQ ID NO: 111 and 3’UTR sequence SEQ ID NO: 280) and the human hemoglobin (5UTR_2_RSV_3UTR_2) (5’UTR sequence SEQ ID NO: 73 and 3’UTR sequence SEQ ID NO: 281) benchmarks.
  • 5UTR_562 SEQ ID NO:86
  • 5UTR_563 SEQ ID NO:87
  • this parent UTR showed robust immunogenicity in mice using RSV-F as the antigen.
  • 5UTR_563 SEQ ID NO:87
  • 5UTR_563 showed comparable immunogenicity to its parent 5UTR_15 (SEQ ID NO:50) (FIG. 5A)
  • 5UTR_562 was the second-best performer in an additional follow-up immunogenicity study in mice (FIG.5B, below).
  • UTR variants based on 5UTR_105 and 5UTR_273 were designed, in which these upstream start codons were removed.
  • FIG.5B examined additional UTR variants (including 5UTR_576 based on 5UTR_105, and 5UTR_581/582/583/599 based on 5UTR_273).
  • RSV-F modRNA-LNP based on 5UTR_582 induced robust neutralizing responses higher than the 5UTR_1/3UTR_1 benchmark as well as other UTRs.
  • the supernatant is aspirated, and cells resuspended with 250 uL wash buffer, the washing of the cell pellet is repeated twice.
  • the cells are pelleted, supernatant aspirated, and resuspended in 50 uL of secondary antibody solution.
  • the plates are sealed and incubated for 45 minutes at 2-8° C, protected from light. Once the incubation is completed the cells are pelleted using a swinging bucket centrifuge at 500 rcf for 5 min at RT. The supernatant is removed and cells resuspended with 250 uL wash buffer, this is repeated twice.
  • the RSV microneutralization assay is a 3-day assay using A549 cells (human alveolar basal epithelial cells) to measure functional antibodies in serum that neutralize RSV activity, preventing infection of a host cell monolayer.
  • A549 cells human alveolar basal epithelial cells; ATCC, cat # CCL-185
  • ATCC human alveolar basal epithelial cells
  • ATCC human alveolar basal epithelial cells
  • a 50% neutralization titer is calculated as the last reciprocal serum dilution at which 50% of the virus is neutralized compared to wells containing virus only.
  • a titer is reported as the geometric mean titer (GMT) of the two replicate titers of each sample.
  • the assay titer range is 20 to 43,740. Any samples with a titer >43,740 are prediluted and repeated to extend the upper titer limit. Any samples below the lower limit of detection (LLOD) are reported at LLOD of 20.
  • UTR_563, 5UTR_582 and 5UTR_599 were assessed using RSV A preF carrying 847 stabilizing mutations as the GOI.
  • a 5'/3'-UTR pair (5UTR_1 (SEQ ID NO: 397)/ 3UTR_1 (SEQ ID NO: 566), named “WHO”) was selected as a benchmark comparison (Demongeot & Fougere, 2023). The in vivo study was performed similarly to what was described in Example 3.
  • mice were immunized on day 0 and 21 with 0.05 ⁇ g or 0.2 ⁇ g of RSV modRNAs. Serum was collected on day 35 (2 weeks post dose 2, PD2) for RSV neutralizing response and T cell responses. Neutralization assays were performed as described in Example 3. The detailed T cell response evaluation by intracellular cytokine staining is described in this example below.
  • RSV F-specific T cell responses were measured from splenocytes of selected groups by a flow cytometry-based intracellular cytokine staining assay (ICS). Splenocytes were cultured under unstimulated or RSV F peptide pool stimulated conditions in the presence of anti-CD107a APC antibody and protein transport inhibitors.
  • ICS flow cytometry-based intracellular cytokine staining assay
  • bivalent modRNA we evaluated immunogenicity of two different LNP- formulation approaches: 1) a "pre-mix” approach where both mRNAs are combined in equal ratio followed by co-formulation into LNPs, or 2) a "post-mix” approach where each mRNA is individually formulated into LNPs and the two LNP preparations combined in equal ratio.
  • RSV A and RSV B neutralizing responses for pre-mix and post-mix groups were similar or higher compared to the monovalent control groups.
  • Table 13 RSV Neutralization Responses with 563-UTR Table 14.
  • RSV F Antigens Sequences of the RSV antigens/polypeptides, RSV DNA and RSV RNA of the present invention are provided in Tables 1-3. The sequences may comprise any stop codon, including but not limited to the stop codons provided in the Tables below. Table 1.
  • RNA molecule comprising at least one open reading frame encoding a respiratory syncytial virus (RSV) polypeptide and at least one untranslated region (UTR) selected from Table 5, provided that the UTR does not consist of only SEQ ID NOs: 397 and 566.
  • RSV respiratory syncytial virus
  • UTR untranslated region
  • RNA molecule of any one of paragraphs 1-4 wherein the 5’ UTR comprises a nucleic acid sequence at least 98% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 357, 372-374, 378, 380, 384 and 385.
  • the 5’ UTR comprises a nucleic acid sequence at least 98% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 357, 373, 378, 380, 384 and 385. 7.
  • RNA molecule of any one of paragraphs 1-6 wherein the 5’ UTR comprises a nucleic acid sequence selected from the group consisting of: SEQ ID NO: 373 (5UTR_563); SEQ ID NO: 380 (5UTR_582); and SEQ ID NO: 385 (5UTR_599).
  • the UTR is a 3’ untranslated region (3’ UTR).
  • the 3’ UTR comprises nucleotides having sequence SEQ ID NO: 567 (3UTR_2 (hHBB)).
  • RSV polypeptide is an RSV glycoprotein.
  • RNA molecule of paragraphs 1-10 wherein the RSV glycoprotein is RSV F protein.
  • RNA molecule of paragraph 20 wherein the open reading frame is transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 8, 10, and 627-632. 22.
  • the RNA molecule of paragraph 21 wherein the open reading frame is transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 8 and 10. 23.
  • the open reading frame comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 15-16 and 633-638.
  • the RNA molecule of paragraph 25 wherein the open reading frame comprises a nucleic acid sequence selected from the group consisting of SEQ ID Nos: 15 and 16. 27.
  • the poly-A tail comprises a sequence having SEQ ID NO: 22.
  • the poly-A tail comprises a sequence having SEQ ID NO: 26, optionally comprising +/-1 or +/-2 adenosine (A).
  • RNA is from: (i) RSV subtype A and (ii) RSV subtype B. 57.
  • the composition of any one of paragraphs 45 to 56 comprising an RNA molecule at a concentration of or of about 0.01 to 0.09 mg/mL formulated in a lipid nanoparticle (LNP) comprising a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL, a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL, a neutral lipid at a concentration of or of about 0.1 to 0.25 mg/mL and a steroid or steroid analog at a concentration of or of about 0.3 to 0.45 mg/mL.
  • LNP lipid nanoparticle

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Zoology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Mycology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Plant Pathology (AREA)
  • Communicable Diseases (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure relates to RNA molecules encoding a respiratory syncytial virus (RSV) polypeptide. The present disclosure further relates to compositions comprising the RNA molecules formulated in a lipid nanoparticle (RNA-LNP). The present disclosure further relates to the use of the RNA molecules, RNA-LNPs and compositions for the treatment and/or prevention of RSV infection-induced acute respiratory tract illness, including pneumonia and bronchitis.

Description

RNA MOLECULES RELATED APPLICATIONS This application claims priority to U.S. Provisional Application No. 63/610,042 filed December 14, 2023, U.S. Provisional Application No.63/553,480 filed February 14, 2024, U.S. Provisional Application No. 63/565,428 filed March 14, 2024, U.S. Provisional Application No. 63/567,866 filed March 20, 2024, U.S. Provisional Application No.63/639,366 filed April 26, 2024 and U.S. Provisional Application No.63/725,096 filed November 26, 2024. The entire content of each of the foregoing applications is hereby incorporated by reference herein in its entirety. REFERENCE TO SEQUENCE LISTING This application is being filed electronically via EFS-Web and includes an electronically submitted sequence listing in .xml format. The .xml file contains a sequence listing entitled "PC073061A Sequence Listing.xml” created on December 4, 2024 and having a size of 644 KB. The sequence listing contained in this .xml file is part of the specification and is incorporated herein by reference in its entirety. BACKGROUND Respiratory syncytial virus (RSV) is a respiratory virus that infects the lungs and breathing passages. RSV is the leading cause of serious viral lower respiratory tract illness in infants worldwide and an important cause of respiratory illness in the elderly. RSV is a member of the Pneumoviridae family. Its genome consists of a single-stranded, negative-sense RNA molecule that encodes 11 proteins, including nine structural proteins (three glycoproteins and six internal proteins) and two non-structural proteins. The structural proteins include three transmembrane surface glycoproteins: the attachment protein G, fusion protein F, and the small hydrophobic SH protein. There are two subtypes of RSV, A and B. They differ primarily in the G glycoprotein, while the sequence of the F glycoprotein is more conserved between the two subtypes. The mature F glycoprotein has three general domains: ectodomain (ED), transmembrane domain (TM), and a cytoplasmic tail (CT). CT contains a single palmitoylated cysteine residue. The F glycoprotein of human RSV is initially translated from the mRNA as a single 574- amino acid polypeptide precursor (referred to “F0” or “F0 precursor”), which contains a signal peptide sequence (amino acids 1-25) at the N-terminus. Upon translation the signal peptide is removed by a signal peptidase in the endoplasmic reticulum. The remaining portion of the F0 precursor (e.g., residues 26-574) may be further cleaved at two polybasic sites (a.a.109/110 and 136/137) by cellular proteases (in particular furin), removing a 27-amino acid intervening sequence designated pep27 (amino acids 110-136) and generating two linked fragments designated F1 (C-terminal portion; amino acids 137-574) and F2 (N-terminal portion; amino acids 26-109). F1 contains a hydrophobic fusion peptide at its N-terminus and two heptad-repeat regions (HRA and HRB). HRA is near the fusion peptide, and HRB is near the TM domain. The F1 and F2 fragments are linked together through two disulfide bonds. Either the uncleaved F0 protein without the signal peptide sequence or a F1-F2 heterodimer can form a RSV F protomer. Three such protomers assemble to form the final RSV F protein complex, which is a homotrimer of the three protomers. The F proteins of subtypes A and B are about 90 percent identical in amino acid sequence. An example sequence of the F0 precursor polypeptide for the A subtype is provided in SEQ ID NO: 1 (A2 strain; GenBank GI: 138251; Swiss Prot P03420), and for the B subtype is provided in SEQ ID NO: 2 (18537 strain; GenBank GI: 138250; Swiss Prot P13843). SEQ ID NO: 1 and SEQ ID NO: 2 are both 574 amino acid sequences. The signal peptide sequence for SEQ ID NO: 1 and SEQ ID NO: 2 has also been reported as amino acids 1-25 (GenBank and UniProt). In both sequences the TM domain is from approximately amino acids 530 to 550 but has alternatively been reported as 525-548. The cytoplasmic tail begins at either amino acid 548 or 550 and ends at amino acid 574, with the palmitoylated cysteine residue located at amino acid 550. RSV F protein is a primary antigen explored for RSV vaccines. The RSV F protein trimer mediates fusion between the virion membrane and the host cellular membrane and also promotes the formation of syncytia. In the virion prior to fusion with the membrane of the host cell, the largest population of F molecules forms a lollipop-shaped structure, with the TM domain anchored in the viral envelope [Dormitzer, P.R., Grandi, G., Rappuoli, R., Nature Reviews Microbiol, 10, 807, 2012.]. This conformation is referred to as the pre-fusion conformation. Pre-fusion RSV F is recognized by monoclonal antibodies (mAbs) D25, AM22, and MPE8, without discrimination between oligomeric states. Pre-fusion F trimers are specifically recognized by mAb AM14 [Gilman MS, Moin SM, Mas V et al., PLoS Pathogens,11(7), 2015]. During RSV entry into cells, the F protein rearranges from the pre-fusion state (which may be referred to herein as “pre-F”), through an intermediate extended structure, to a post-fusion state (“post-F”). During this rearrangement, the C-terminal coiled-coil of the pre-fusion molecule dissociates into its three constituent strands, which then wrap around the globular head and join three additional helices to form the post-fusion six helix bundle. If a pre-fusion RSV F trimer is subjected to increasingly harsh chemical or physical conditions, such as elevated temperature, it undergoes structural changes. Initially, there is loss of trimeric structure (at least locally within the molecule), and then rearrangement to the post-fusion form, and then denaturation of the domains. To prevent viral entry, F-specific neutralizing antibodies presumably must bind the pre- fusion conformation of F on the virion, or potentially the extended intermediate, before the viral envelope fuses with a cellular membrane. Thus, the pre-fusion form of the F protein is considered the preferred conformation as the desired vaccine antigen [Ngwuta, J.O., Chen, M., Modjarrad, K., Joyce, M.G., Kanekiyo, M., Kumar, A., Yassine, H.M., Moin, S.M., Killikelly, A.M., Chuang, G.Y., Druz, A., Georgiev, I.S., Rundlet, E.J., Sastry, M., Stewart-Jones, G.B., Yang. Y., Zhang, B., Nason, M.C., Capella, C., Peeples, M., Ledgerwood, J. E., Mclellan, J.S., Kwong, P.D., Graham, B.S., Science Translat. Med., 14, 7, 309 (2015)]. Upon extraction from a membrane with surfactants such as Triton X-100, Triton X-114, NP-40, Brij-35, Brij-58, Tween 20, Tween 80, Octyl glucoside, Octyl thioglucoside, SDS, CHAPS, CHAPSO, or expression as an ectodomain, physical or chemical stress, or storage, the F glycoprotein readily converts to the post-fusion form [McLellan JS, Chen M, Leung S et al. Structure of RSV fusion glycoprotein trimer bound to a pre- fusion-specific neutralizing antibody. Science 340, 1113–1117 (2013); Chaiwatpongsakorn, S., Epand, R.F., Collins, P.L., Epand R.M., Peeples, M.E., J Virol. 85(8):3968-77 (2011); Yunus, A.S., Jackson T.P., Crisafi, K., Burimski, I., Kilgore, N.R., Zoumplis, D., Allaway, G.P., Wild, C.T., Salzwedel, K. Virology.2010 Jan 20;396(2):226-37]. Therefore, the preparation of prefusion F as a vaccine antigen has remained a challenge. Since the neutralizing and protective antibodies function by interfering with virus entry, it is postulated that an F antigen that does not elicit pre- fusion specific antibodies is not expected to be as effective as an F antigen that elicits pre-fusion specific antibodies. Therefore, it is considered more desirable to utilize an F protein vaccine that contains a F protein immunogen in the pre-fusion form. Mutants of the RSV F protein have been provided to increase pre-fusion stability (see for example PCT application No WO2017/109629) and are promising vaccine candidates. RSV vaccines that incorporate F protein antigen have been under development. Currently licensed RSV vaccines for protection against RSV disease for older adults by active immunization (AREXVY®, MRESVIA®), and for both older adults and infants through 6 months of age by maternal immunization (ABRYSVO®) are based on the prefusion conformation of RSV F. RNA technology, especially mRNA technology, is particularly advantageous as a vaccine or therapeutic platform. For an effective RNA vaccine or therapeutic, it is important to maximize protein expression such that amounts of desired proteins or antigens are generated from minimal amounts of RNAs. However, mRNA-based therapies can suffer from challenges including low manufacturing efficiency, short half-life of administered mRNA in circulation, and low translation efficiency. As such, there is a need for RNA compositions with improved stability and translation efficiency, including methods to improve protein expression by optimizing the sequence and structure of the 5' untranslated regions of the mRNA and enable high levels of expression. SUMMARY The present disclosure provides improved immunogenic compositions against RSV infection comprising RSV preF RNA molecules and methods for treating a subject comprising administration of such compositions, as provided herein. In one aspect, the present disclosure provides immunogenic compositions and methods for preventing, treating or ameliorating an infection, disease or condition in a subject comprising the administration of RNA molecules, e.g., immunogenic RNA polynucleotide encoding an amino acid sequence, e.g., an immunogenic antigen, comprising a Respiratory syncytial virus (RSV) F protein in a prefusion conformation, an immunogenic variant thereof, or an immunogenic fragment of the RSV protein or the immunogenic variant thereof, e.g., an antigenic peptide or protein. Thus, the immunogenic antigen comprises an epitope of a RSV protein for inducing an immune response against RSV, in the subject. RNA polynucleotide encoding an immunogenic antigen is administered to provide (following expression of the polynucleotide by appropriate target cells) antigen for induction, e.g., stimulation, priming, and/or expansion, of an immune response, e.g., antibodies and/or immune effector cells. In one aspect, the immune response to be induced according to the present disclosure is both B cell-mediated immune response, e.g., an antibody-mediated immune response as well as T-cell-mediated immune response. In one aspect, the immune response is an anti-RSV immune response. The immunogenic compositions described herein comprise RNA molecules (as the active principle) that may be translated into one or more proteins in a recipient’s cells. In addition to wild type, codon-optimized or mutant sequences encoding the antigen sequence, the RNA molecules may contain one or more structural elements optimized for maximal efficacy of the RNA with respect to stability and translational efficiency (5 cap, 5 UTR, 3 UTR, poly-A-tail). In one aspect, the RNA molecules contain all of these elements. In some aspects, each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1Ψ) (e.g., modified RNA; modRNA). The RNA molecules described herein may be formulated with, encapsulated in, or complexed with lipids and/or proteins to generate RNA-particles (e.g., lipid nanoparticles (LNPs)) for administration to a subject. In one aspect, the RNA molecules described herein are formulated with, encapsulated in, or complexed with lipids to generate RNA-lipid nanoparticles (e.g. RNA- LNPs) for administration to a subject. In one aspect, the RNA molecules described herein are formulated with, encapsulated in, or complexed with proteins for administration. In one aspect, the RNA molecules described herein are formulated with, encapsulated in, or complexed with lipids and proteins for administration. If a combination of different RNA molecules is used, the RNA molecules may be formulated together or formulated separately with lipids and/or proteins to generate RNA-particles for administration. The present disclosure provides for RNA molecules and RNA-LNPs that include at least one open reading frame (ORF) encoding a RSV antigen. In some aspects, the RSV antigen is a RSV polypeptide. In some aspects, the RSV polypeptide is RSV F protein. In some aspects, the RSV F protein is a full-length, truncated, fragment or variant thereof. In some aspects, the RSV F protein comprises at least one mutation. In a preferred aspect, the RSV F protein is in a prefusion conformation (preF). In another aspect, the RSV F polypeptide has, has at least, or has at most 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or higher identity to any of the amino acid sequences of Table 1, for example, any of SEQ ID NO: 1-6 or 621-626. In another aspect, the RSV F polypeptide has an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 6 and 621-626. In another aspect, the RSV F polypeptide has an amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 6. In another aspect, the RSV F polypeptide has the amino acid sequence set forth in SEQ ID NO: 4. In another aspect, the RSV F polypeptide has the amino acid sequence set forth in SEQ ID NO: 6. The present disclosure provides for RNA molecules and RNA-LNPs that include at least one ORF encoding an RSV polypeptide of Table 1 and at least one untranslated region (UTR) selected from Table 5, provided that the UTR does not consist of the combination of only SEQ ID NOs: 397 and 566. In some aspects, the RNA molecule comprises at least one open reading frame encoding a respiratory syncytial virus (RSV) fusion protein F (F) polypeptide and a 5’ untranslated region (5’ UTR), wherein the 5’ UTR comprises a nucleic acid sequence that has, has at least, or has at most 90% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336-344, 356-358, 360, 362-367, 370-375, and 378-396. In another aspect, the 5’ UTR comprises a nucleic acid sequence that has, has at least, or has at most 92% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336, 341, 356-358, 372-375, and 378-396. In another aspect, the 5’ UTR comprises a nucleic acid sequence that has, has at least, or has at most 95% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336, 357, 372-374, 378, 380, 384 and 385. In another aspect, the 5’ UTR comprises a nucleic acid sequence that has, has at least, or has at most 98% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 357, 372- 374, 378, 380, 384 and 385. In another aspect, the 5’ UTR comprises a nucleic acid sequence that has, has at least, or has at most 98% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 357, 373, 378, 380, 384 and 385. In another aspect, the 5’ UTR comprises a nucleic acid sequence selected from the group consisting of: SEQ ID NO: 373 (5UTR_563); SEQ ID NO: 380 (5UTR_582); and SEQ ID NO: 385 (5UTR_599). The present disclosure provides for RNA molecules and RNA-LNPs comprising at least one ORF transcribed from at least one DNA nucleic acid of Table 2 and at least one untranslated region (UTR) selected from Table 4, provided that when the ORF is SEQ ID NO: 8 or 9 that the UTR does not consist of the combination of only SEQ ID NOs: 111 and 280. In some aspects, the ORF is transcribed from a nucleic acid comprising a nucleotide sequence that has, has at least, or has at most 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or higher identity to any one of the sequences of SEQ ID NOs: 7-10 or 627-632. In some aspects, the ORF is transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 8, 10, and 627-632. In another aspect, the ORF is transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 8 and 10. In another aspect, the ORF is transcribed from a nucleic acid has the sequence set forth in SEQ ID NO: 8. In another aspect, the ORF is transcribed from a nucleic acid has the sequence set forth in SEQ ID NO: 10. The present disclosure further provides for RNA molecules and RNA-LNPs comprising at least one ORF comprising an RNA nucleic acid sequence of Table 3 and at least one untranslated region (UTR) selected from Table 5, provided that when the ORF is SEQ ID NO: 15 or 16 that the UTR does not consist of the combination of only SEQ ID NOs: 397 and 566. In another aspect, the ORF comprises a nucleic acid sequence that has, has at least, or has at most 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or higher identity to any one of the sequences of SEQ ID NOs: 15-16 or 633-638. In another aspect, the ORF comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 15-16 and 633-638. In a preferred aspect, the ORF comprises a nucleic acid sequence selected from the group consisting of SEQ ID Nos: 15 and 16. In another aspect, the ORF comprises a nucleic acid sequence set forth in SEQ ID NO: 15. In another aspect, the ORF comprises a nucleic acid sequence set forth in SEQ ID NO: 16. In some aspects, each uridine of any of SEQ ID NO: 15-16 and 633-638 is replaced by N1-methylpseudouridine (m1Ψ) (e.g., modified RNA; modRNA). The present disclosure further provides for RNA molecules and RNA-LNPs that include a 3’ untranslated region (3’-UTR). In one aspect, the 3’UTR is selected from Table 5. In another aspect, the 3’ UTR comprises nucleotides having a sequence set forth in SEQ ID NO: 567 (3UTR_2 (hHBB)). The present disclosure further provides for RNA molecules and RNA-LNPs that comprise a 5’ cap moiety. In some aspects, the 5′ cap moiety is (3′OMe) - m2 7,3′-OG(5’)ppp (m1 2′-O)ApG. The present disclosure further provides for RNA molecules and RNA-LNPs that include a 3’ poly-A tail. In some aspects, the poly-A tail comprises a sequence having SEQ ID NO: 22 or 26. In some aspects, the RNA molecule includes a 5’ UTR and a 3’ UTR. In some aspects, the RNA molecule includes a 5’ cap, 5’ UTR, and 3’ UTR. In some aspects, the RNA molecule includes a 5’ cap, 5’ UTR, 3’ UTR, and poly-A tail. In some aspects, the RNA molecule includes a 5’ UTR, 3’ UTR, and poly-A tail. In some aspects, 1, 2, 3, or more of the foregoing elements can be excluded from the RNA molecule. In some aspects, each uridine of any of the 5′ UTR, 3′ UTR, and poly-A tail is replaced by N1-methylpseudouridine (m1Ψ) (e.g., modified RNA; modRNA). In some aspects, the poly-A tail length may contain +/-1 adenosine (A) or +/-2 adenosine (A). The present disclosure provides for RNA molecules as described in Table 7. In some aspects, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 15, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 16, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 15, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 16, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 15, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 16, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In some aspects, the RSV ORF further comprises a stop codon described herein. In some aspects, the poly-A tail length may contain +/-1 A or +/-2 A. In some aspects, each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1Ψ) (e.g., modified RNA; modRNA). In other aspects, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 633, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 634, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 633, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 634, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 633, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 634, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In some aspects, the RSV ORF further comprises a stop codon described herein. In some aspects, the poly-A tail length may contain +/-1 A or +/-2 A. In some aspects, each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1Ψ) (e.g., modified RNA; modRNA). In other aspects, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 635, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 636, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 635, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 636, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 635, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 636, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In some aspects, the RSV ORF further comprises a stop codon described herein. In some aspects, the poly-A tail length may contain +/-1 A or +/-2 A. In some aspects, each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1Ψ) (e.g., modified RNA; modRNA). In further aspects, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 637, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 373, a RSV ORF of SEQ ID NO: 638, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 637, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 380, a RSV ORF of SEQ ID NO: 638, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 637, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In another aspect, the RNA molecule comprises a 5′ UTR of SEQ ID NO: 385, a RSV ORF of SEQ ID NO: 638, a 3′ UTR of SEQ ID NO: 567 and/or a poly-A tail of SEQ ID NO: 22. In some aspects, the RSV ORF further comprises a stop codon described herein. In some aspects, the poly-A tail length may contain +/-1 A or +/-2 A. In some aspects, each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1Ψ) (e.g., modified RNA; modRNA). The present disclosure provides for RNA molecules encoding RSV F proteins transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 11-14, 639 and 640. The present disclosure further provides for RNA molecules encoding RSV F proteins comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 17-20, 641 and 642. In some aspects, each uridine of the RNA molecule is replaced by N1-methylpseudouridine (m1Ψ) (e.g., modified RNA; modRNA). The present disclosure further provides for RNA molecules that include at least one open reading frame that was generated from codon-optimized DNA. In some aspects, the open reading frame comprises a G/C content of at least, at most, exactly, or between (inclusive or exclusive) any two of 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, or 75%, e.g., at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, is or is about 50% to 75%, or is or is about 55% to 70%. In some aspects, the G/C content is or is about 58%, is or is about 66%, or is or is about 62%. The present disclosure further provides for RNA molecules that encode RSV polypeptides that localizes in the cellular membrane, localizes in the Golgi and/or are anchored in the membrane and are secreted. The present disclosure further provides RNA molecules comprising stabilized RNA. The present disclosure further provides for RNA molecules that are messenger-RNA (mRNA) or self- replicating RNA (saRNA). In some aspects, the RNA comprises at least one modified nucleotide (modRNA).The present disclosure further provides for RNA molecules that include RNA having at least one modified nucleotide (e.g., modified RNA; modRNA). In some aspects, the modified nucleotide is pseudouridine, N1-methylpseudouridine, N1-ethylpseudouridine, 2-thiouridine, 4′- thiouridine, 5-methylcytosine, 5-methyluridine, 2-thio-1-methyl-1-deaza-pseudouridine, 2-thio-1- methyl-pseudouridine, 2-thio-5-aza-uridine, 2-thio-dihydropseudouridine, 2-thio-dihydrouridine, 2-thio-pseudouridine, 4-methoxy-2-thio-pseudouridine, 4-methoxy-pseudouridine, 4-thio-1- methyl-pseudouridine, 4-thio-pseudouridine, 5-aza-uridine, dihydropseudouridine, 5- methoxyuridine, or 2′-O-methyl uridine. In some aspects, the modified nucleotide is N1- methylpseudouridine (m1Ψ). In some aspects, 1, 2, 3, 4, 5, or more of the foregoing modified nucleotides can be excluded from the RNA molecule. The present disclosure further provides for immunogenic compositions including the RNA molecules described herein. The RNA molecules may be formulated in, encapsulated in, complexed with, bound to or adsorbed on a lipid nanoparticle (LNP) (e.g., RSV RNA-LNPs) in such immunogenic compositions. In one aspect, the immunogenic composition comprises a RSV molecule encoding an F protein as described herein which is of RSV subtype A. In another aspect, the immunogenic composition comprises a RSV molecule encoding an F protein as described herein which is of RSV subtype B. In a further aspect, the immunogenic composition is a bivalent immunogenic composition comprising a first RSV molecule encoding an F protein as described herein which is of RSV subtype A and a second RSV molecule encoding an F protein as described herein which is of RSV subtype B. In one aspect, the bivalent immunogenic composition may be a (i) "pre-mix" formulation wherein two or more RNA polynucleotides are combined in equal ratio followed by co-formulation into LNPs, or (ii) a "post-mix" formulation wherein each RNA polynucleotide is individually formulated into LNPs and the two or more LNPs are combined either in equal ratio or not in equal ratio. In another aspect, the ratio of the first RSV molecule to the second RSV molecule in the bivalent immunogenic composition is 1:1. In some aspects, the lipid nanoparticle includes at least one of a cationic lipid, a polymer conjugated lipid (e.g., a PEGylated lipid), and at least one structural lipid (e.g., a neutral lipid and a steroid or steroid analog). In some aspects, 1, 2, 3, or more of the foregoing lipids may be excluded from the lipid nanoparticle. In some aspects, the lipid nanoparticle includes a cationic lipid. In some aspects, the cationic lipid is selected from any cationic lipid described in section B(i) set forth herein. In some aspects, the cationic lipid is (4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate) (ALC-0315). In some aspects, the lipid nanoparticle includes a polymer conjugated lipid. In some aspects, the cationic lipid is selected from any cationic lipid described in section B(ii) set forth herein. In some aspects, the lipid nanoparticle includes a PEGylated lipid, also referred to as a PEG-lipid. In some aspects, the PEGylated lipid is PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramides (e.g., PEG-CerC14 or PEG-CerC20), PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, 2- [(polyethylene glycol)-2000]-N,N-ditetradecylacetamide, glycol-lipids including PEG-c-DOMG, PEG-c-DMA, PEG-s-DMG, N-[(methoxy polyethylene glycol)2000)carbamoyl]-1,2- dimyristyloxlpropyl-3-amine (PEG-c-DMA), and PEG-2000-DMG, PEGylated diacylglycerol (PEG-DAG) such as 1-(monomethoxy-polyethyleneglycol)-2,3-dimyristoylglycerol (PEG-DMG), a PEGylated phosphatidylethanolamine (PEG-PE), a PEG succinate diacylglycerol (PEG-S-DAG) such as 4-O-(2’,3′- di(tetradecanoyloxy)propyl-1-O-((o-methoxy(polyethoxy)ethyl)butanedioate (PEG-S-DMG), a PEGylated ceramide (PEG-cer), or a PEG dialkoxypropylcarbamate such as co-methoxy(polyethoxy)ethyl-N-(2,3di(tetradecanoxy)propyl)carbamate or 2,3- di(tetradecanoxy)propyl-N-(u>-methoxy(polyethoxy)ethyl)carbamate. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing PEGylated lipids can be excluded from the RNA molecule. In some aspects, the PEGylated lipid is 2-[(polyethylene glycol)-2000]-N,N-ditetradecylacetamide (ALC- 0159). In some aspects, the lipid nanoparticle includes at least one structural lipid, such as a neutral lipid. In some aspects, the neutral lipid is 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl-phosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyl-oleoyl-phosphatidylethanolamine (POPE), dioleoyl- phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane-1-carboxylate (DOPE- mal), dipalmitoylphosphatidylethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoylphosphatidylethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1- trans PE, 1-stearoyl-2-oleoylphosphatidyethanolamine (SOPE), and/or 1,2-dielaidoyl-sn-glycero- 3-phosphoethanolamine (transDOPE). In some aspects, 1, 2, 3, 4, 5, or more of the foregoing structural lipids can be excluded from the RNA molecule. In some aspects, the neutral lipid is 1,2- distearoyl-sn-glycero-3-phosphocholine (DSPC). In some aspects, the lipid nanoparticle includes a second structural lipid, such as a steroid or steroid analog. In some aspects, the steroid or steroid analog is cholesterol. In some aspects, the lipid nanoparticle has a mean diameter of about 1 to about 500 nm, e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 1 nm, 10 nm, 20 nm, 30 nm, 40 nm, 50 nm, 60 nm, 70 nm, 80 nm, 90 nm, 100 nm, 110 nm, 120 nm, 130 nm, 140 nm, 150 nm, 160 nm, 170 nm, 180 nm, 190 nm, 200 nm, 210 nm, 220 nm, 230 nm, 240 nm, 250 nm, 260 nm, 270 nm, 280 nm, 290 nm, 300 nm, 310 nm, 320 nm, 330 nm, 340 nm, 350 nm, 360 nm, 370 nm, 380 nm, 390 nm, 400 nm, 410 nm, 420 nm, 430 nm, 440 nm, 450 nm, 460 nm, 470 nm, 480 nm, 490 nm, or 500 nm. In one aspect, the lipid nanoparticle has a mean diameter between 40 nm to 120 nm. In another aspect, the lipid nanoparticle has a mean diameter between about 50 nm to about 90 nm. In another aspect, the lipid nanoparticle has a mean diameter between about 55 nm to about 65 nm. In some aspects, the RNA-LNP immunogenic composition is a liquid RNA-LNP composition comprising an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition comprising a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.80, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.90, 0.91, 0.92, 0.93, 0.94, or 0.95 mg/mL), a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, or 0.15 mg/mL), a first structural lipid at a concentration of or of about 0.1 to 0.25 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, or 0.25 mg/mL), and a second structural lipid at a concentration of or of about 0.3 to 0.45 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.30, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.40, 0.41, 0.42, 0.43, 0.44, or 0.45 mg/mL). In some aspects, the liquid composition further comprises a buffer composition comprising a first buffer at a concentration of or of about 0.1 to 0.3 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, or 0.30 mg/mL), a second buffer at a concentration of or of about 1.25 to 1.4 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 1.25, 1.26, 1.27, 1.28, 1.29, 1.30, 1.31, 1.32, 1.33, 1.34, 1.35, 1.36, 1.37, 1.38, 1.39, or 1.40 mg/mL), and a stabilizing agent at a concentration of or of about 95 to 110 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, or 110 mg/mL). In some aspects, 1, 2, 3, 4, 5, or more of the foregoing elements can be excluded from the liquid RNA-LNP composition. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing element concentrations can be excluded from the liquid RNA-LNP composition. In specific aspects, the liquid RNA-LNP immunogenic composition comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition comprising ((4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2- hexyldecanoate) (ALC-0315) at a concentration of or of about 0.8 to 0.95 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.80, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.90, 0.91, 0.92, 0.93, 0.94, or 0.95 mg/mL), 2-[(polyethylene glycol)- 2000]-N,N-ditetradecylacetamide (ALC-0159) at a concentration of or of about 0.05 to 0.15 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, or 0.15 mg/mL), 1,2-distearoyl-sn-glycero-3- phosphocholine (DSPC) at a concentration of or of about 0.1 to 0.25 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, or 0.25 mg/mL), and cholesterol at a concentration of or of about 0.3 to 0.45 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.30, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.40, 0.41, 0.42, 0.43, 0.44, or 0.45 mg/mL). In some aspects, the liquid composition further comprises a Tris buffer composition comprising tromethamine at a concentration of or of about 0.1 to 0.3 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, or 0.30 mg/mL) and Tris hydrochloride (HCl) at a concentration of or of about 1.25 to 1.4 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 1.25, 1.26, 1.27, 1.28, 1.29, 1.30, 1.31, 1.32, 1.33, 1.34, 1.35, 1.36, 1.37, 1.38, 1.39, or 1.40 mg/mL), and sucrose at a concentration of or of about 95 to 110 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, or 110 mg/mL). In some aspects, 1, 2, 3, 4, 5, or more of the foregoing elements can be excluded from the liquid RNA-LNP composition. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing element concentrations can be excluded from the liquid RNA-LNP composition. In some aspects, the liquid RNA-LNP immunogenic composition comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in a LNP, and further comprising of or of about 5 to 15 mM Tris buffer(e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 mM) and of or of about 200 to 400 mM sucrose (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, or 400 mM) at a pH of or of about 7.0 to 8.0 (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0). In some aspects, 1, 2, 3, or more of the foregoing elements can be excluded from the liquid RNA-LNP composition. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing element concentrations can be excluded from the liquid RNA-LNP composition. In some aspects, the RNA-LNP immunogenic composition is a lyophilized (reconstituted) RNA-LNP composition comprising an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition comprising a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.80, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.90, 0.91, 0.92, 0.93, 0.94, or 0.95 mg/mL), a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, or 0.15 mg/mL), a first structural lipid at a concentration of or of about 0.1 to 0.25 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, or 0.25 mg/mL), and a second structural lipid at a concentration of or of about 0.3 to 0.45 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.30, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.40, 0.41, 0.42, 0.43, 0.44, or 0.45 mg/mL). In some aspects, the lyophilized composition further comprises a first buffer at a concentration of or of about 0.01 and 0.15 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, or 0.15 mg/mL), a second buffer at a concentration of or of about 0.5 and 0.65 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.50, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.60, 0.61, 0.62, 0.63, 0.64, or 0.65 mg/mL), a stabilizing agent at a concentration of or of about 35 to 50 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 mg/mL), and a salt diluent at a concentration of or of about 5 to 15 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 mg/mL) for reconstitution. In specific aspects, the lyophilized compositions are reconstituted in or in about 0.6 to 0.75 mL of the salt diluent (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.60, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.70, 0.71, 0.72, 0.73, 0.74, or 0.75 mL). Concentrations in the lyophilized RNA-LNP composition are determined post-reconstitution. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing elements can be excluded from the lyophilized RNA-LNP composition. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing element concentrations can be excluded from the lyophilized RNA-LNP composition. In specific aspects, a lyophilized (reconstituted) RNA-LNP composition comprises an RNA polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition of ALC-0315 at a concentration of or of about 0.8 to 0.95 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.80, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.90, 0.91, 0.92, 0.93, 0.94, or 0.95 mg/mL), ALC-0159 at a concentration of or of about 0.05 to 0.15 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, or 0.15 mg/mL), DSPC at a concentration of or of about 0.1 to 0.25 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, or 0.25 mg/mL), and cholesterol at a concentration of or of about 0.3 to 0.45 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.30, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.40, 0.41, 0.42, 0.43, 0.44, or 0.45 mg/mL), and further comprises a Tris buffer composition comprising tromethamine at a concentration of or of about 0.01 to 0.15 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, or 0.15 mg/mL) and Tris HCl at a concentration of or of about 0.5 to 0.65 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.50, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.60, 0.61, 0.62, 0.63, 0.64, or 0.65 mg/mL), sucrose at a concentration of or of about 35 to 50 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 mg/mL), and sodium chloride (NaCl) diluent at a concentration of or of about 5 to 15 mg/mL (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 mg/mL) for reconstitution. In specific aspects, the lyophilized compositions are reconstituted in or in about 0.6 to 0.75 mL of sodium chloride (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 0.60, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.70, 0.71, 0.72, 0.73, 0.74, or 0.75 mL). Concentrations in the lyophilized RNA-LNP composition are determined post-reconstitution. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing elements can be excluded from the lyophilized RNA-LNP composition. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing element concentrations can be excluded from the lyophilized RNA-LNP composition. In one aspect, the RNA-LNP composition is a liquid composition. The present disclosure provides for RNA molecules, RNA-LNPs and immunogenic compositions that may be administered to a subject at a dose per administration of at least, at most, exactly, or between (inclusive or exclusive) any two of 1 µg, 15 µg, 30 µg, 45 µg, 60 µg, 75 µg, 90 µg, 100 µg or higher of RSV RNA encapsulated in LNP. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing concentrations of RSV RNA encapsulated in LNP can be excluded. The present disclosure provides for RNA molecules, RNA-LNPs and immunogenic compositions that may be administered in a single dose. The present disclosure further provides for RNA molecules, RNA-LNPs and immunogenic compositions that may be administered twice (e.g., Day 0 and on or about Day 7, Day 0 and on or about Day 14, Day 0 and on or about Day 21, Day 0 and on or about Day 28, Day 0 and on or about Day 60, Day 0 and on or about Day 90, Day 0 and on or about Day 120, Day 0 and on or about Day 150, Day 0 and on or about Day 180, Day 0 and on or about 1 month later, Day 0 and on or about 2 months later, Day 0 and on or about 3 months later, Day 0 and on or about 6 months later, Day 0 and on or about 9 months later, Day 0 and on or about 12 months later, Day 0 and on or about 18 months later, Day 0 and on or about 2 years later, Day 0 and on or about 5 years later, or Day 0 and on or about 10 years later). The present disclosure further provides for RNA molecules, RNA-LNPs and immunogenic compositions that may be administered twice at Day 0 and on or about 2 months later. The present disclosure further provides for RNA molecules, RNA-LNPs and immunogenic compositions that may be administered twice at Day 0 and on or about 6 months later. The present disclosure further provides for RNA molecules, RNA-LNPs and immunogenic compositions that may be administered three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more times. In some aspects, periodic boosters at intervals of 1-5 years may be desirable to maintain protective levels of the antibodies. The present disclosure further provides for administration of at least one booster dose. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing dosing regimens can be excluded. The present disclosure provides for a method of inducing an immune response against RSV in a subject, including administering to the subject an effective amount of an RNA molecule, RNA- LNP and/or immunogenic composition described herein. The present disclosure further provides for the use of an RNA molecule, RNA-LNP and/or immunogenic composition described herein in the manufacture of a medicament for use in inducing an immune response against RSV in a subject. The present disclosure provides for a method of inducing an immune response against RSV in a subject, including administering to the subject an effective amount of an RNA molecule and/ or RNA-LNP that includes at least one open reading frame encoding a RSV polypeptide or immunogenic composition described herein. The present disclosure further provides for the use of an RNA molecule and/or RNA-LNP that includes at least one open reading frame encoding a RSV polypeptide or immunogenic composition described herein in the manufacture of a medicament for use in inducing an immune response against RSV in a subject. The present disclosure provides for a method of inducing an immune response against RSV in a subject, including administering to the subject an effective amount of an RNA molecule and/or RNA-LNP that includes at least one open reading frame encoding a polypeptide of a gene of interest or composition described herein. The present disclosure further provides for the use of an RNA molecule and/or RNA-LNP that includes at least one open reading frame encoding a polypeptide of a gene of interest or composition described herein in the manufacture of a medicament for use in inducing an immune response against RSV in a subject. The present disclosure provides for a method of preventing, treating, and/or ameliorating an infection, disease, or condition in a subject, including administering to a subject an effective amount of an RNA molecule, RNA-LNP and/or immunogenic composition described herein. The present disclosure further provides for the use of an RNA molecule, RNA-LNP and/or immunogenic composition described herein in the manufacture of a medicament for use in preventing, treating, and/or ameliorating an infection, disease, or condition in a subject. In some aspects, the infection, disease, or condition is associated with RSV. In some aspects, the infection, disease, or condition is acute lower respiratory infection (ALRI), including pneumonia and bronchitis. In some aspects, the infection, disease, or condition is acute lower respiratory infection (ALRI), including pneumonia and bronchitis. The present disclosure provides for a method of preventing, treating, and/or ameliorating an infection, disease, or condition in a subject, including administering to a subject an effective amount of an RNA molecule and/or RNA-LNP that includes at least one open reading frame encoding a RSV polypeptide or immunogenic composition described herein. The present disclosure further provides for the use of an RNA molecule and/or RNA-LNP that includes at least one open reading frame encoding a RSV polypeptide or immunogenic composition described herein in the manufacture of a medicament for use in preventing, treating, and/or ameliorating an infection, disease, or condition in a subject. In some aspects, the infection, disease, or condition is associated with RSV. In some aspects, the infection, disease, or condition is acute lower respiratory infection (ALRI), including pneumonia and bronchitis. In some aspects, the infection, disease, or condition is acute lower respiratory infection (ALRI), including pneumonia and bronchitis. The present disclosure further provides for a method of preventing, treating, and/or ameliorating an infection, disease, or condition in a subject, including administering to a subject an effective amount of RNA molecules and/or RNA-LNPs that include at least one open reading frame encoding a polypeptide of a gene of interest or immunogenic compositions described herein. The present disclosure further provides for the use of RNA molecules and/or RNA-LNPs that include at least one open reading frame encoding a polypeptide of a gene of interest or immunogenic compositions described herein in the manufacture of a medicament for use in preventing, treating, and/or ameliorating an infection, disease, or condition in a subject. In some aspects, the infection, disease, or condition is associated with the gene of interest. In some aspects, the subject is at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months of age, or 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, or more years of age. In some aspects, the subject is, is at least, is at most, or is about less than 1 year of age, 1 year of age or older, 5 years of age or older, 10 years of age or older, 20 years of age or older, 30 years of age or older, 40 years of age or older, 50 years of age or older, 60 years of age or older, 70 years of age or older, or older. In some aspects, the subject the subject is or is about 50 years of age or older. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing age groups are not administered the RNA molecules and/or RNA- LNPs. In some aspects, the subject is immunocompetent. In some aspects, the subject is immunocompromised. The present disclosure provides for a method or use described herein, wherein the RNA molecule, RNA-LNP and/or immunogenic composition is administered as a vaccine. In one aspect, the subject is administered a single dose, two doses, three doses, or more, and optionally, a booster dose of the RNA molecule, composition or vaccine. The present disclosure provides a method or use described herein, wherein the RNA molecule, RNA-LNP and/or immunogenic composition is administered by intradermal, intramuscular, or intranasal injection. It is contemplated that any aspect discussed in this specification may be implemented with respect to any method or composition of the disclosure, and vice versa. Furthermore, compositions of the disclosure may be used to achieve methods of the disclosure. Any method in the context of a therapeutic, diagnostic, or physiologic purpose or effect may also be described in “use” claim language such as “use of” any compound, composition, or agent discussed herein for achieving or implementing a described therapeutic, diagnostic, or physiologic purpose or effect. Use of the one or more compositions may be employed based on any of the methods described herein. Other objects, features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific aspects of the disclosure, are given by way of illustration only, since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description. BRIEF DESCRIPTION OF THE DRAWINGS FIG.1 schematically illustrates the wild-type (WT) RSV F protein (RSV WT) and variant RSV F protein constructs, where “SP” refers to a signal peptide sequence (amino acid residues 1-25 of each construct), “TM” refers to a transmembrane peptide sequence corresponding to the portion of the protein that spans the cell membrane, “CT” refers to a cytoplasmic tail peptide sequence corresponding to the portion of the protein that extends into the cell cytoplasm, “ectodomain” comprises amino acid residues 1-513 (without TM and CT, denoted by “ΔTM & CT”) and localizes to the extracellular space. The amino acid positions of each portion (e.g. SP, F2, pep27, F1) or mutation for each construct are indicated therein, e.g. SP of each construct spans from amino acid residues 1-25 of each construct. Each RSV protein construct depicted is applicable to both RSV subtype A and subtype B constructs of the WT or variant thereof as shown. FIG.2 depicts RSV-F expression by candidate UTR constructs with hDC expression along the X-axis and HEK expression along the Y-axis. Labeled constructs (also denoted by darker gray plot points) were nominated for in vivo testing in mice. WHO benchmark is denoted by the intersection of the lines. Additional benchmarks are indicated by asterisks (*). FIG.3A - 3B depicts RSV-F neutralizing titers from mice injected with modRNA LNPs expressing RSV-F driven by different UTR combinations after 21 days (21 days post dose 1) (FIG. 3A) and after 35 days (14 days post dose 2) (FIG. 3B). The two WHO benchmark constructs with 5'/3' UTRs with different polyA tails are denoted as 5UTR_1/3UTR_1 constructs. Female BALB/c mice (10/group) were immunized intramuscularly at day 0 and 21 with RSV-F subgroup A modRNA-LNPs with novel UTR candidates at 0.5 µg dose. On day 21 (3 weeks post dose 1, 3W PD1) and day 35 (2 weeks post dose 2, 2W PD2), serum was collected for RSV neutralizing assay. Neutralization assay results for RSV A on day 21 (FIG.3A) and day 35 (FIG. 3B) expressed as 50% neutralizing titers. Each symbol represents a titer from an individual animal. Horizontal line represents geometric mean titer (GMT) of the group. FIG.4 is a diagram of the organization of the translation machinery. The 5' cap and part of the 5' UTR interact with the 43S pre-initiation complex. The 43S complex then scans the 5' UTR towards the AUG start site, where it assembles with the 60S complex. Following the release of the elongation factors, the ribosome starts translation (adapted from Leppek et al., Nat Rev Mol Cell Biol 2018). FIG. 5A - 5B depict RSV-F neutralizing titers from mice injected with modRNA LNPs expressing RSV-F driven by different UTR combinations after 35 days (2 weeks post dose 2) in an initial (FIG.5A) and a follow-on (FIG.5B) experiment. Benchmark constructs with 5'/3' UTRs with different polyA tails are denoted. Female BALB/c mice (10/group) were immunized intramuscularly at day 0 and 21 with RSV-F subgroup A modRNA-LNPs with novel UTR candidates at 0.2 µg dose. On day 35 (2 weeks post dose 2, 2W PD2), serum was collected for RSV neutralizing assay. Neutralization assay results for RSV A on day 35 expressed as 50% neutralizing titers. Each “○” symbol represents a titer from an individual animal. Horizontal line represents the geometric mean titer (GMT) of the group. FIG.6A – 6D show the immunogenicity of modRNAs carrying different novel 5’-UTRs in mice. Female BALB/c mice (N=10 per group) were immunized IM with 0.05, 0.2 or 0.5 μg of LNP- formulated modRNA encoding RSV A 847 preF design carrying different 5'-UTRs, namely 5UTR_1 (SEQ ID NO: 397) named “WHO” was selected as a benchmark comparison, 5UTR_563 (also known as “BMD563”), 5UTR_582 (also known as “BMD582”), and 5UTR_599 (also known as “BMD599”). FIG.6A shows virus neutralization responses against RSV A strain, M37, which were measured by MNT assay on Day 35 (2 weeks post-dose 2). LOD is defined at 50% neutralizing titer of 20. Error bars represent the 95% confidence interval of the geometric means. FIG.6B shows Total % of RSV F antigen-specific IFN-γ+ CD4+ T-cells and FIG.6C shows Total % of RSV F antigen-specific IL-4+ CD4+ T-cells across selected groups. FIG.6D shows Total % of RSV F antigen-specific IFN-γ+ CD8+ T-cells across selected groups as measured by ICS assays in freshly isolated splenocytes (5 out of 10 mice) from 0.2 μg group. Error bars represent the standard deviation of the means in T-cell analysis. FIG. 7A - 7D show the immunogenicity of monovalent versus bivalent RSV preF modRNAs with different LNP-formulations in mice. Female BALB/c mice (N=10 per group) were immunized IM with 0.25 or 0.50 μg of monovalent LNP-formulated modRNA encoding RSV A or B 847 preF , or bivalent modRNA (0.25 μg RSV A + 0.25 μg RSV B) prepared using post-mix or pre-mixed LNP-formulation approaches. Virus neutralization responses against RSV A strain, M37 (FIG.7A and FIG.7C), or RSV B strain, B18537 (FIG.7B and FIG.7D), were measured by MNT assay on Day 35 (2 weeks post-dose 2). LOD is defined at 50% neutralizing titer of 20. Error bars represent the 95% confidence interval of the geometric means. Statistical analyses were performed on log 10 transformed neutralization titers. The final results were transformed back to the original scale. Analysis of variance (ANOVA) was applied to compare means among the six groups. p-values were adjusted with Tukey method (* indicates p < 0.05). DETAILED DESCRIPTION The present disclosure provides for an RNA molecule (e.g., RNA polynucleotide) comprising at least one open reading frame (ORF) encoding a respiratory syncytial virus (RSV) antigen and at least one untranslated region (UTR) selected from Table 5, provided that the UTR does not consist of only SEQ ID NOs: 397 and 566. In some aspects, the RSV antigen is a RSV polypeptide. In some aspects, the RSV polypeptide is a RSV F polypeptide. In some aspects, the RSV polypeptide comprises an amino acid sequence set forth in Table 1. In some aspects, the RNA molecules comprise an ORF transcribed from at least one DNA nucleic acid sequence of Table 2. In some aspects, the RNA molecules comprise an ORF comprising an RNA nucleic acid sequence of Table 3. In some aspects the RNA molecule comprises at least one of a 5’ cap, 5’ UTR, 3’ UTR and poly-A tail. In other aspects the RNA molecule comprises at least one of a 5’ cap, 3’ UTR and poly-A tail. The present disclosure provides for an RNA molecule comprising modified nucleotides (e.g., modified RNA; modRNA). In some aspects, RNA molecule comprises at least one open reading frame encoding a respiratory syncytial virus (RSV) fusion protein F (F) polypeptide and a 5’ untranslated region (5’ UTR), wherein the 5’ UTR comprises a nucleic acid sequence at least 90% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336-344, 356-358, 360, 362-367, 370-375, and 378-396. In another aspect, the 5’ UTR comprises a nucleic acid sequence at least 92% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336, 341, 356-358, 372-375, and 378-396. In another aspect, the 5’ UTR comprises a nucleic acid sequence selected from the group consisting of: SEQ ID NO: 373 (5UTR_563); SEQ ID NO: 380 (5UTR_582); and SEQ ID NO: 385 (5UTR_599). The present disclosure provides for an immunogenic composition comprising any one of the RNA molecules encoding a RSV polypeptide described herein complexed with, encapsulated in, or formulated with one or more lipids, and forming lipid nanoparticles (RNA-LNPs). The present disclosure further provides for an immunogenic composition comprising any one of the RNA molecules comprising at least one RNA nucleic acid described herein complexed with, encapsulated in, or formulated with one or more lipids, and forming RNA-LNPs. The present disclosure further provides for a method of preventing, treating or ameliorating an infection, disease or condition (e.g., RSV infection-related Respiratory tract illness, including pneumonia and bronchitis) in a subject via administering to a subject an effective amount of an RNA molecule, RNA-LNP or an immunogenic composition described herein. The present disclosure further provides for the use of the RNA molecule, RNA-LNP and/or an immunogenic composition described herein as a vaccine. The present invention may be understood more readily by reference to the following detailed description of the embodiments of the invention and the Examples included herein. It is to be understood that this invention is not limited to specific methods of making that may of course vary. It is to be also understood that the terminology used herein is for the purpose of describing specific embodiments only and is not intended to be limiting. The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All references cited herein, including patent applications, patent publications, UniProtKB accession numbers are herein incorporated by reference, as if each individual reference were specifically and individually indicated to be incorporated by reference in its entirety. I. EXAMPLES OF DEFINITIONS Unless otherwise defined herein, scientific and technical terms used in connection with the present invention have the meanings that are commonly understood by those of ordinary skill in the art. Throughout this application, the terms “about” and “approximately” and “substantially” are used according to their plain and ordinary meaning in the area of cell and molecular biology to indicate a deviation of ±10% of the value(s) to which it is attached. Therefore, in any disclosed aspect, the terms may be substituted with “within [a percentage] of” what is specified. In one non- limiting aspect, the percentage includes 0.1, 0.5, 1, 5, and 10 percent. Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it was individually recited herein. The use of the word “a” or “an” when used in conjunction with the term “comprising” may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.” The phrase “and/or” means “and” or “or.” To illustrate, A, B, and/or C includes: A alone, B alone, C alone, a combination of A and B, a combination of A and C, a combination of B and C, or a combination of A, B, and C. In other words, “and/or” operates as an inclusive or. The phrase “essentially all” is defined as “at least 95%”; if essentially all members of a group have a certain property, then at least 95% of members of the group have that property. In some aspects, essentially all means equal to any one of, at least any one of, or between any two of 95, 96, 97, 98, 99, or 100% of members of the group have that property. The compositions and methods for their use may “comprise,” “consist essentially of,” or “consist of” any of the ingredients or steps disclosed throughout the specification. Throughout this specification, unless the context requires otherwise, the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open- ended and will be understood to imply the inclusion of a stated step or element or group of steps or elements but not the exclusion of any other step or element or group of steps or elements. It is contemplated that aspects described herein in the context of the term “comprising” may also be implemented in the context of the term “consisting of” or “consisting essentially of.” Compositions and methods “consisting essentially of” any of the ingredients or steps disclosed limits the scope of the claim to the specified materials or steps which do not materially affect the basic and novel characteristic of the claimed disclosure. The words “consisting of” (and any form of consisting of, such as “consist of” and “consists of”) means including, and limited to, whatever follows the phrase “consisting of.” Thus, the phrase “consisting of” indicates that the listed elements are required or mandatory, and that no other elements may be present. Reference throughout this specification to “one aspect,” “an aspect,” “a particular aspect,” “a related aspect,” “a certain aspect,” “an additional aspect,” or “a further aspect” or combinations thereof means that a particular feature, structure or characteristic described in connection with the aspect is included in at least one aspect of the present disclosure. Thus, the appearances of the foregoing phrases in various places throughout this specification are not necessarily all referring to the same aspect. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more aspects. The terms “inhibiting,” “decreasing,” or “reducing” or any variation of these terms includes any measurable decrease (e.g., a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% decrease) or complete inhibition to achieve a desired result. The terms “improve,” “promote,” or “increase” or any variation of these terms includes any measurable increase (e.g., a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% increase) to achieve a desired result or production of a protein or molecule. As used herein, the terms “reference,” “standard,” or “control” describe a value relative to which a comparison is performed. For example, an agent, subject, population, sample, or value of interest is compared with a reference, standard, or control agent, subject, population, sample, or value of interest. A reference, standard, or control may be tested and/or determined substantially simultaneously and/or with the testing or determination of interest for an agent, subject, population, sample, or value of interest and/or may be determined or characterized under comparable conditions or circumstances to the agent, subject, population, sample, or value of interest under assessment. The term “isolated” may refer to a nucleic acid or polypeptide that is substantially free of cellular material, bacterial material, viral material, or culture medium (when produced by recombinant DNA techniques) of their source of origin, or chemical precursors or other chemicals (when chemically synthesized). Moreover, an isolated compound refers to one that may be administered to a subject as an isolated compound; in other words, the compound may not simply be considered “isolated” if it is adhered to a column or embedded in an agarose gel. Moreover, an “isolated nucleic acid fragment” or “isolated peptide” is a nucleic acid or protein fragment that is not naturally occurring as a fragment and/or is not typically in the functional state and/or that is altered or removed from the natural state through human intervention. For example, a DNA naturally present in a living animal is not “isolated,” but a synthetic DNA, or a DNA partially or completely separated from the coexisting materials of its natural state is “isolated.” An isolated nucleic acid may exist in substantially purified form, or may exist in a non-native environment such as, for example, a cell into which the nucleic acid has been delivered. A “nucleic acid,” as used herein, is a molecule comprising nucleic acid components and refers to DNA or RNA molecules. It may be used interchangeably with the term “polynucleotide.” A nucleic acid molecule is a polymer comprising or consisting of nucleotide monomers, which are covalently linked to each other by phosphodiester-bonds of a sugar/phosphate-backbone. Nucleic acids may also encompass modified nucleic acid molecules, such as base-modified, sugar-modified or backbone-modified etc. DNA or RNA molecules. Nucleic acids may exist in a variety of forms such as: isolated segments and recombinant vectors of incorporated sequences or recombinant polynucleotides encoding polypeptides, such as antigens or one or both chains of an antibody, or a fragment, derivative, mutein, or variant thereof, polynucleotides sufficient for use as hybridization probes, PCR primers or sequencing primers for identifying, analyzing, mutating or amplifying a polynucleotide encoding a polypeptide, anti-sense nucleic acids for inhibiting expression of a polynucleotide, mRNA, saRNA, modRNA and complementary sequences of the foregoing described herein. Nucleic acids may encode an epitope to which antibodies may bind. The term “epitope” refers to a moiety that is specifically recognized by an immunoglobulin (e.g., antibody or receptor) binding component. In some aspects, an epitope is comprised of a plurality of chemical atoms or groups on an antigen. In some aspects, such chemical atoms or groups are surface-exposed when the antigen adopts a relevant three-dimensional conformation. In some aspects, such chemical atoms or groups are physically near to each other in space when the antigen adopts such a conformation. In some aspects, at least some such chemical atoms are groups are physically separated from one another when the antigen adopts an alternative conformation (e.g., is linearized). Nucleic acids may be single-stranded or double-stranded and may comprise RNA and/or DNA nucleotides and artificial variants thereof (e.g., peptide nucleic acids). In some cases, a nucleic acid sequence may encode a polypeptide sequence with additional heterologous coding sequences, for example to allow for purification of the polypeptide, transport, secretion, post- translational modification, or for therapeutic benefits such as targeting or efficacy. A tag or other heterologous polypeptide may be added to the modified polypeptide-encoding sequence, wherein “heterologous” refers to a polypeptide that is not the same as the modified polypeptide. The term “polynucleotide” refers to a nucleic acid molecule that may be recombinant or has been isolated from total genomic nucleic acid. Included within the term “polynucleotide” are oligonucleotides (nucleic acids 100 residues or less in length), recombinant vectors, including, for example, plasmids, cosmids, phage, viruses, and the like. Polynucleotides include, in certain aspects, regulatory sequences, isolated substantially away from their naturally occurring genes or protein encoding sequences. Polynucleotides may be single-stranded (coding or antisense) or double-stranded, and may be RNA, DNA (genomic, cDNA, or synthetic), analogs thereof, or a combination thereof. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide. In certain aspects, there are polynucleotide variants having substantial identity to the sequences disclosed herein; those comprising equal to any one of, at least any one of, at most any one of, or between any two of 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% or higher sequence identity, compared to a polynucleotide sequence provided herein using the methods described herein (e.g., BLAST analysis using standard parameters). In certain aspects, the isolated polynucleotide will comprise a nucleotide sequence encoding a polypeptide that has at least 90% identity to an amino acid sequence described herein, over the entire length of the sequence; or a nucleotide sequence complementary to said isolated polynucleotide. In some aspects, the isolated polynucleotide will comprise a nucleotide sequence encoding a polypeptide that has at least 95% identity to an amino acid sequence described herein, over the entire length of the sequence; or a nucleotide sequence complementary to said isolated polynucleotide. The nucleic acid segments, regardless of the length of the coding sequence itself, may be combined with other nucleic acid sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably. The nucleic acids may be any length. They may be, for example, equal to any one of, at least any one of, at most any one of, or between any two of 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, 125, 175, 200, 250, 300, 350, 400, 450, 500, 750, 1000, 1500, 3000, 5000, 6000, 7000, 8000, 9000, 10000, 11000, 12000, 13000, 14000, 15000 or more nucleotides in length, and/or may comprise one or more additional sequences, for example, regulatory sequences, and/or be a part of a larger nucleic acid, for example, a vector. It is therefore contemplated that a nucleic acid fragment of almost any length may be employed, with the total length being limited by the ease of preparation and use in the intended recombinant nucleic acid protocol. In this respect, the term “gene” is used to refer to a nucleic acid that encodes a protein, polypeptide, or peptide (including any sequences required for proper transcription, post- translational modification, or localization). As will be understood by those in the art, this term encompasses genomic sequences, expression cassettes, cDNA sequences, and smaller engineered nucleic acid segments that express, or may be adapted to express, proteins, polypeptides, domains, peptides, fusion proteins, and mutants. A nucleic acid encoding all or part of a polypeptide may contain a contiguous nucleic acid sequence encoding all or a portion of such a polypeptide. It also is contemplated that a particular polypeptide may be encoded by nucleic acids containing variations having slightly different nucleic acid sequences but, nonetheless, encode the same or substantially similar polypeptide. As used herein, the term “expression” of a nucleic acid sequence refers to the generation of any gene product from the nucleic acid sequence. In some aspects, a gene product may be a transcript. In some aspects, a gene product may be a polypeptide. In some aspects, expression of a nucleic acid sequence involves one or more of the following: (1) production of an RNA template from a DNA sequence (e.g., by transcription); (2) processing of an RNA transcript (e.g., by splicing, editing, etc.); (3) translation of an RNA into a polypeptide or protein; and/or (4) post- translational modification of a polypeptide or protein. In general, the term “engineered” refers to the aspect of having been manipulated by the hand of man. For example, a polynucleotide is considered to be “engineered” when two or more sequences that are not linked together in that order in nature are manipulated by the hand of man to be directly linked to one another in the engineered polynucleotide and/or when a particular residue in a polynucleotide is non-naturally occurring and/or is caused through action of the hand of man to be linked with an entity or moiety with which it is not linked in nature. The term “DNA,” as used herein, means a nucleic acid molecule comprising nucleotides such as deoxy-adenosine-monophosphate, deoxy-thymidine-monophosphate, deoxy- guanosine-monophosphate and deoxy-cytidine-monophosphate monomers which are composed of a sugar moiety (deoxyribose), a base moiety and a phosphate moiety, and polymerize by a characteristic backbone structure. The backbone structure is, typically, formed by phosphodiester bonds between the sugar moiety of the nucleotide, e.g., deoxyribose, of a first and a phosphate moiety of a second, adjacent monomer. The specific order of the monomers, e.g., the order of the bases linked to the sugar/phosphate-backbone, is called the DNA sequence. DNA may be single stranded or double stranded. In the double stranded form, the nucleotides of the first strand typically hybridize with the nucleotides of the second strand, e.g. by A/T-base-pairing and G/C- base-pairing. DNA may contain all, or a majority of, deoxyribonucleotide residues. As used herein, the term “deoxyribonucleotide” means a nucleotide lacking a hydroxyl group at the 2′ position of a β-D-ribofuranosyl group. Without any limitation, DNA may encompass double stranded DNA, antisense DNA, single stranded DNA, isolated DNA, synthetic DNA, DNA that is recombinantly produced, and modified DNA. The term “RNA,” as used herein, means a nucleic acid molecule comprising nucleotides such as adenosine-monophosphate, uridine-monophosphate, guanosine-monophosphate and cytidine-monophosphate monomers which are connected to each other along a so-called backbone. The backbone is formed by phosphodiester bonds between the sugar, e.g., ribose, of a first and a phosphate moiety of a second, adjacent monomer. RNA may be obtainable by transcription of a DNA-sequence, e.g., inside a cell. In eukaryotic cells, transcription is typically performed inside the nucleus or the mitochondria. In vivo, transcription of DNA may result in premature RNA which is processed into messenger-RNA (mRNA). Processing of the premature RNA, e.g. in eukaryotic organisms, comprises various posttranscriptional modifications such as splicing, 5′ capping, polyadenylation, export from the nucleus or the mitochondria. Mature messenger RNA is processed and provides the nucleotide sequence that may be translated into an amino acid sequence of a peptide or protein. A mature mRNA may comprise a 5′ cap, a 5′ UTR, an open reading frame, a 3′ UTR and a poly-A tail sequence. RNA may contain all, or a majority of, ribonucleotide residues. As used herein, the term “ribonucleotide” means a nucleotide with a hydroxyl group at the 2′ position of a β-D-ribofuranosyl group. In one aspect, RNA may be messenger RNA (mRNA) that relates to a RNA transcript which encodes a peptide or protein. As known to those of skill in the art, mRNA generally contains a 5′ untranslated region (5′ UTR), a polypeptide coding region, and a 3′ untranslated region (3′ UTR). Without any limitation, RNA may encompass double stranded RNA, antisense RNA, single stranded RNA, isolated RNA, synthetic RNA, RNA that is recombinantly produced, and modified RNA (modRNA). An “isolated RNA” is defined as an RNA molecule that may be recombinant or has been isolated from total genomic nucleic acid. An isolated RNA molecule or protein may exist in substantially purified form, or may exist in a non-native environment such as, for example, a host cell. A “modified RNA” or “modRNA” refers to an RNA molecule having at least one addition, deletion, substitution, and/or alteration of one or more nucleotides as compared to naturally occurring RNA. Such alterations may refer to the addition of non-nucleotide material to internal RNA nucleotides, or to the 5′ and/or 3′ end(s) of RNA. In one aspect, such modRNA contains at least one modified nucleotide, such as an alteration to the base of the nucleotide. For example, a modified nucleotide may replace one or more uridine and/or cytidine nucleotides. For example, these replacements may occur for every instance of uridine and/or cytidine in the RNA sequence, or may occur for only select uridine and/or cytidine nucleotides. Such alterations to the standard nucleotides in RNA may include non-standard nucleotides, such as chemically synthesized nucleotides or deoxynucleotides. For example, at least one uridine nucleotide may be replaced with N1-methylpseudouridine in an RNA sequence. Other such altered nucleotides are known to those of skill in the art. Such altered RNA molecules are considered analogs of naturally-occurring RNA. In some aspects, the RNA is produced by in vitro transcription using a DNA template, where DNA refers to a nucleic acid that contains deoxyribonucleotides. In some aspects, the RNA may be replicon RNA (replicon), in particular self-replicating RNA, or self-amplifying RNA (saRNA). As contemplated herein, without any limitations, RNA may be used as a therapeutic modality to treat and/or prevent a number of conditions in mammals, including humans. Methods described herein comprise administration of the RNA described herein to a mammal, such as a human. For example, in one aspect such methods of use for RNA include an antigen-coding RNA vaccine to induce robust neutralizing antibodies and accompanying/concomitant T-cell response to achieve protective immunization. In some aspects, minimal vaccine doses are administered to induce robust neutralizing antibodies and accompanying/concomitant T-cell response to achieve protective immunization. In one aspect, the RNA administered is in vitro transcribed RNA. For example, such RNA may be used to encode at least one antigen intended to generate an immune response in said mammal. Pathogenic antigens are peptide or protein antigens derived from a pathogen associated with infectious disease. In specific aspects, the pathogenic are peptide or protein antigens derived from RSV. Conditions and/or diseases that may be treated with RNA disclosed herein include, but are not limited to, those caused and/or impacted by viral infection. Such viruses include, but are not limited to, RSV. “Prevent” or “prevention,” as used herein when used in connection with the occurrence of a disease, disorder, and/or condition, refers to reducing the risk of developing the disease, disorder and/or condition and/or to delaying onset of one or more characteristics or symptoms of the disease, disorder or condition. Prevention may be considered complete when onset of a disease, disorder, or condition has been delayed for a predefined period of time. As will be understood from context, “risk” of a disease, disorder, and/or condition refers to a likelihood that a particular individual will develop the disease, disorder, and/or condition. In some aspects, risk is expressed as a percentage. In some aspects, risk is, is at least, or is at most from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90 up to 100%. In some aspects risk is expressed as a risk relative to a risk associated with a reference sample or group of reference samples. In some aspects, a reference sample or group of reference samples have a known risk of a disease, disorder, condition and/or event. In some aspects a reference sample or group of reference samples are from individuals comparable to a particular individual. In some aspects, risk may reflect one or more genetic attributes, e.g., which may predispose an individual toward development (or not) of a particular disease, disorder and/or condition. In some aspects, risk may reflect one or more epigenetic events or attributes and/or one or more lifestyle or environmental events or attributes. Susceptible to: An individual who is “susceptible to” a disease, disorder, and/or condition is one who has a higher risk of developing the disease, disorder, and/or condition than does a member of the general public. In some aspects, an individual who is susceptible to a disease, disorder and/or condition may not have been diagnosed with the disease, disorder, and/or condition. In some aspects, an individual who is susceptible to a disease, disorder, and/or condition may exhibit symptoms of the disease, disorder, and/or condition. In some aspects, an individual who is susceptible to a disease, disorder, and/or condition may not exhibit symptoms of the disease, disorder, and/or condition. In some aspects, an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some aspects, an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition. The terms “protein,” “polypeptide,” or “peptide” are used herein as synonyms and refer to a polymer of amino acid monomers, e.g., a molecule comprising at least two amino acid residues. Polypeptides may include gene products, naturally occurring polypeptides, synthetic polypeptides, homologs, orthologs, paralogs, fragments and other equivalents, variants, and analogs of the foregoing. Polypeptides may be a single molecule or may be a multi-molecular complex such as a dimer, trimer or tetramer. A protein comprises one or more peptides or polypeptides, and may be folded into a 3-dimensional form, which may be required for the protein to exert its biological function. As used herein, the term “wild type” or ”WT” or “native” refers to the endogenous version of a molecule that occurs naturally in an organism. In some aspects, wild type versions of a protein or polypeptide are employed, however, in other aspects of the disclosure, a modified protein or polypeptide is employed to generate an immune response. The terms described above may be used interchangeably. A “modified protein” or “modified polypeptide” or a “variant” refers to a protein or polypeptide whose chemical structure, particularly its amino acid sequence, is altered with respect to the wild type protein or polypeptide. In some aspects, a modified/variant protein or polypeptide has at least one modified activity or function (recognizing that proteins or polypeptides may have multiple activities or functions). It is specifically contemplated that a modified/variant protein or polypeptide may be altered with respect to one activity or function yet retain a wild type activity or function in other respects, such as immunogenicity. Where a protein is specifically mentioned herein, it is in general a reference to a native (wild type) or recombinant (modified) protein. The protein may be isolated directly from the organism of which it is native, produced by recombinant DNA/exogenous expression methods, produced by solid-phase peptide synthesis (SPPS), or other in vitro methods. In particular aspects, there are isolated nucleic acid segments and recombinant vectors incorporating nucleic acid sequences that encode a polypeptide (e.g., an antigen or fragment thereof). The term “recombinant” may be used in conjunction with a polypeptide or the name of a specific polypeptide, and this generally refers to a polypeptide produced from a nucleic acid molecule that has been manipulated in vitro or that is a replication product of such a molecule. The term “fragment,” with reference to an amino acid sequence (peptide or protein), relates to a part of an amino acid sequence, e.g., a sequence which represents the amino acid sequence shortened at the N-terminus and/or C-terminus. A fragment shortened at the C- terminus (N-terminal fragment) is obtainable, e.g., by translation of a truncated open reading frame that lacks the 3′-end of the open reading frame. A fragment shortened at the N-terminus (C-terminal fragment) is obtainable, e.g., by translation of a truncated open reading frame that lacks the 5′-end of the open reading frame, as long as the truncated open reading frame comprises a start codon that serves to initiate translation. A fragment of an amino acid sequence comprises, e.g., at least 50 %, at least 60 %, at least 70 %, at least 80%, at least 90%, or at least 99% of the amino acid residues from an amino acid sequence. In the present disclosure, a fragment of a polypeptide, DNA nucleic acid or RNA nucleic acid sequence refers to a sequence having sequence identity of at least, at most, exactly, or between any two of 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% with a polypeptide, DNA nucleic acid or RNA nucleic acid sequence, from which it is derived. In one aspect, a fragment of a polypeptide, DNA nucleic acid or RNA nucleic acid sequence refers to a sequence having sequence identity of at least 70% with a polypeptide, DNA nucleic acid or RNA nucleic acid sequence, from which it is derived. In one aspect, a fragment of a polypeptide, DNA nucleic acid or RNA nucleic acid sequence refers to a sequence having sequence identity of at least 80% with a polypeptide, DNA nucleic acid or RNA nucleic acid sequence, from which it is derived. In one aspect, a fragment of a polypeptide, DNA nucleic acid or RNA nucleic acid sequence refers to a sequence having sequence identity of at least 85% with a polypeptide, DNA nucleic acid or RNA nucleic acid sequence, from which it is derived. In one aspect, a fragment of a polypeptide, DNA nucleic acid or RNA nucleic acid sequence refers to a sequence having sequence identity of at least 90% with a polypeptide, DNA nucleic acid or RNA nucleic acid sequence, from which it is derived. In one aspect, a fragment of a polypeptide, DNA nucleic acid or RNA nucleic acid sequence refers to a sequence having sequence identity of at least 95% with a polypeptide, DNA nucleic acid or RNA nucleic acid sequence, from which it is derived. In one aspect, a fragment of a polypeptide, DNA nucleic acid or RNA nucleic acid sequence refers to a sequence having sequence identity of at least 97% with a polypeptide, DNA nucleic acid or RNA nucleic acid sequence, from which it is derived. In one aspect, a fragment of a polypeptide, DNA nucleic acid or RNA nucleic acid sequence refers to a sequence having sequence identity of at least 99% with a polypeptide, DNA nucleic acid or RNA nucleic acid sequence, from which it is derived. As used herein in the context of molecules, e.g., nucleic acids, proteins, or small molecules, the term “variant” refers to a molecule that shows significant structural identity with a reference molecule but differs structurally from the reference molecule, e.g., in the presence or absence or in the level of one or more chemical moieties as compared to the reference entity. In some aspects, a variant also differs functionally from its reference molecule. In general, whether a particular molecule is properly considered to be a “variant” of a reference molecule is based on its degree of structural identity with the reference molecule. As will be appreciated by those skilled in the art, any biological or chemical reference molecule has certain characteristic structural elements. A variant, by definition, is a distinct molecule that shares one or more such characteristic structural elements but differs in at least one aspect from the reference molecule. In some aspects, a variant polypeptide or nucleic acid may differ from a reference polypeptide or nucleic acid as a result of one or more differences in amino acid or nucleotide sequence and/or one or more differences in chemical moieties (e.g., carbohydrates, lipids, phosphate groups) that are covalently components of the polypeptide or nucleic acid (e.g., that are attached to the polypeptide or nucleic acid backbone). In some aspects, a variant polypeptide or nucleic acid shows an overall sequence identity with a reference polypeptide or nucleic acid that is at least, at most, exactly, or between any two of 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99%. In some aspects, a variant polypeptide or nucleic acid does not share at least one characteristic sequence element with a reference polypeptide or nucleic acid. In some aspects, a reference polypeptide or nucleic acid has one or more biological activities. In some aspects, a variant polypeptide or nucleic acid shares one or more of the biological activities of the reference polypeptide or nucleic acid. In some aspects, a variant polypeptide or nucleic acid lacks one or more of the biological activities of the reference polypeptide or nucleic acid. In some aspects, a variant polypeptide or nucleic acid shows a reduced level of one or more biological activities as compared to the reference polypeptide or nucleic acid. In some aspects, a polypeptide or nucleic acid of interest is considered to be a “variant” of a reference polypeptide or nucleic acid if it has an amino acid or nucleotide sequence that is identical to that of the reference but for a small number of sequence alterations at particular positions. Preferably, the variant polypeptide or nucleic acid sequence has at least one modification compared to the reference polypeptide or nucleic acid sequence, e.g., from 1 to about 20 modifications. In one aspect, the variant polypeptide or nucleic acid sequence has from 1 to about 10 modifications compared to the reference polypeptide or nucleic acid sequence. In one aspect, the variant polypeptide or nucleic acid sequence has from 1 to about 5 modifications compared to the reference polypeptide or nucleic acid sequence. In one aspect, the variant polypeptide or nucleic acid sequence has from 1 to about 4 modifications compared to the reference polypeptide or nucleic acid sequence. Typically, fewer than about 20%, about 15%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, or about 2% of the residues in a variant are substituted, inserted, or deleted, as compared to the reference. Often, a variant polypeptide or nucleic acid comprises a very small number (e.g., fewer than about 5, about 4, about 3, about 2, or about 1) number of substituted, inserted, or deleted, functional residues (e.g., residues that participate in a particular biological activity) relative to the reference. In some aspects, a variant polypeptide or nucleic acid comprises about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2, or about 1 substituted residues as compared to a reference. In some aspects, a variant polypeptide or nucleic acid comprises fewer than about 25, about 20, about 19, about 18, about 17, about 16, about 15, about 14, about 13, about 10, about 9, about 8, about 7, about 6, and commonly fewer than about 5, about 4, about 3, or about 2 additions or deletions as compared to the reference. In some aspects, a variant polypeptide or nucleic acid comprises not more than about 5, about 4, about 3, about 2, or about 1 addition or deletion, and, in some aspects, comprises no additions or deletions, as compared to the reference. In some aspects, a reference polypeptide or nucleic acid is a “wild type” or “WT” or “native” sequence found in nature, including allelic variations. A wild type polypeptide or nucleic acid sequence has a sequence that has not been intentionally modified. For the purposes of the present disclosure, “variants” of an amino acid sequence (peptide, protein, or polypeptide) comprise amino acid insertion variants, amino acid addition variants, amino acid deletion variants and/or amino acid substitution variants. “Variants” of a nucleotide sequence comprise nucleotide insertion variants, nucleotide addition variants, nucleotide deletion variants and/or nucleotide substitution variants. The term “variant” includes all mutants, splice variants, post-translationally modified variants, conformations, isoforms, allelic variants, species variants, and species homologs, in particular those which are naturally occurring. The term “variant” includes, in particular, fragments of an amino acid or nucleic acid sequence. Changes may be introduced by mutation into a nucleic acid, thereby leading to changes in the amino acid sequence of a polypeptide (e.g., an antigen or antibody or antibody derivative) that it encodes. Mutations may be introduced using any technique known in the art. In one aspect, one or more particular amino acid residues are changed using, for example, a site-directed mutagenesis protocol. In another aspect, one or more randomly selected residues are changed using, for example, a random mutagenesis protocol. In some aspects, however it is made, a mutant polypeptide may be expressed and screened for a desired property. Mutations may be introduced into a nucleic acid without significantly altering the biological activity of a polypeptide that it encodes. For example, one may make nucleotide substitutions leading to amino acid substitutions at non-essential amino acid residues. Alternatively, one or more mutations may be introduced into a nucleic acid that selectively changes the biological activity of a polypeptide that it encodes. For example, the mutation may quantitatively or qualitatively change the biological activity. Examples of quantitative changes include increasing, reducing or eliminating the activity. Examples of qualitative changes include altering the antigen specificity of an antibody. “Sequence similarity” indicates the percentage of amino acids that either are identical or that represent conservative amino acid substitutions. “Sequence identity” between two amino acid sequences indicates the percentage of amino acids that are identical between the sequences. “Sequence identity” between two nucleic acid sequences indicates the percentage of nucleotides that are identical between the sequences. The terms “% identical,” “% identity,” or similar terms are intended to refer, in particular, to the percentage of nucleotides or amino acids which are identical in an optimal alignment between the sequences to be compared. Said percentage is purely statistical, and the differences between the two sequences may be but are not necessarily randomly distributed over the entire length of the sequences to be compared. Comparisons of two sequences are usually carried out by comparing the sequences, after optimal alignment, with respect to a segment or “window of comparison,” in order to identify local regions of corresponding sequences. The optimal alignment for a comparison may be carried out manually or with the aid of the local homology algorithm by Smith and Waterman, 1981, Ads App. Math.2, 482, with the aid of the local homology algorithm by Neddleman and Wunsch, 1970, J. Mol. Biol. 48, 443, with the aid of the similarity search algorithm by Pearson and Lipman, 1988, Proc. Natl Acad. Sci. USA 88, 2444, or with the aid of computer programs using said algorithms (GAP, BESTFIT, FASTA, BLAST P, BLAST N, and TFASTA in Wisconsin Genetics Software Package, Genetics Computer Group). In some aspects, percent identity of two sequences is determined using the BLASTN or BLASTP algorithm, as available on the United States National Center for Biotechnology Information (NCBI) website. Percentage identity is obtained by determining the number of identical positions at which the sequences to be compared correspond, dividing this number by the number of positions compared (e.g., the number of positions in the reference sequence) and multiplying this result by 100. In some aspects, the degree of similarity or identity is given for a region that is at least, at most, exactly, or between any two of about 50%, about 60%, about 70%, about 80%, about 90%, or about 100% of the entire length of the reference sequence. For example, if the reference nucleic acid sequence consists of 200 nucleotides, the degree of identity is given for at least, at most, exactly, or between any two of about 100, about 120, about 140, about 160, about 180, or about 200 nucleotides, in some aspects, continuous nucleotides. In some aspects, the degree of similarity or identity is given for the entire length of the reference sequence. Homologous amino acid sequences may exhibit at least, at most, exactly, or between any two of 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 99% identity of the amino acid residues. In one aspect, homologous amino acid sequences exhibit at least 95% identity of the amino acid residues. In one aspect, homologous amino acid sequences exhibit at least 98% identity of the amino acid residues. In one aspect, homologous amino acid sequences exhibit at least 99% identity of the amino acid residues. A fragment or variant of an amino acid sequence (peptide or protein) may be a “functional fragment” or “functional variant.” The term “functional fragment” or “functional variant” of an amino acid sequence relates to any fragment or variant exhibiting one or more functional properties identical or similar to those of the amino acid sequence from which it is derived, e.g., it is functionally equivalent. With respect to antigens or antigenic sequences, one particular function is one or more immunogenic activities displayed by the amino acid sequence from which the fragment or variant is derived. The term “functional fragment” or “functional variant,” as used herein, in particular refers to a variant molecule or sequence that comprises an amino acid sequence that is altered by one or more amino acids compared to the amino acid sequence of the parent molecule or sequence and that is still capable of fulfilling one or more of the functions of the parent molecule or sequence, e.g., inducing an immune response. In one aspect, the modifications in the amino acid sequence of the parent molecule or sequence do not significantly affect or alter the characteristics of the molecule or sequence. The term “mutant” of a wild-type RSV F protein, “mutant” of a RSV F protein, “RSV F protein mutant,” or “modified RSV F protein” refers to a polypeptide that displays introduced mutations relative to a wild-type F protein and is immunogenic against the wild-type F protein. An amino acid sequence (peptide, protein, or polypeptide) “derived from” a designated amino acid sequence (peptide, protein, or polypeptide) refers to the origin of the first amino acid sequence. Preferably, the amino acid sequence which is derived from a particular amino acid sequence has an amino acid sequence that is identical, essentially identical, or homologous to that particular sequence or a fragment thereof. Amino acid sequences derived from a particular amino acid sequence may be variants of that particular sequence or a fragment thereof. For example, it will be understood by one of ordinary skill in the art that the antigens suitable for use herein may be altered such that they vary in sequence from the naturally occurring or native sequences from which they were derived, while retaining the desirable activity of the native sequences. In the present disclosure, a vector refers to a nucleic acid molecule, such as an artificial nucleic acid molecule. A vector may be used to incorporate a nucleic acid sequence, such as a nucleic acid sequence comprising an open reading frame. Vectors include, but are not limited to, storage vectors, expression vectors, cloning vectors, transfer vectors. A vector may be an RNA vector or a DNA vector. In some aspects the vector is a DNA molecule. In some aspects, the vector is a plasmid vector. In some aspects, the vector is a viral vector. Typically, an expression vector will contain a desired coding sequence and appropriate other sequences necessary for the expression of the operably linked coding sequence in a particular host organism (e.g., bacteria, yeast, plant, insect, or mammal) or in in vitro expression systems. Cloning vectors are generally used to engineer and amplify a certain desired fragment (typically a DNA fragment), and may lack functional sequences needed for expression of the desired fragment(s). As used herein, the term “pharmaceutical composition” refers to an active agent, formulated together with one or more pharmaceutically acceptable carriers. Pharmaceutical compositions may be immunogenic compositions. In some aspects, active agent is present in unit dose amount appropriate for administration in a therapeutic regimen that shows a statistically significant probability of achieving a predetermined therapeutic effect when administered to a relevant population. In some aspects, pharmaceutical compositions may be specially formulated for parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation. As used herein, the term “vaccination” refers to the administration of an immunogenic composition intended to generate an immune response, for example to a disease-associated (e.g., disease-causing) agent (e.g., a virus). In some aspects, vaccination may be administered before, during, and/or after exposure to a disease-associated agent, and in certain aspects, before, during, and/or shortly after exposure to the agent. In some aspects, vaccination includes multiple administrations, appropriately spaced in time, of a vaccine composition. In some aspects, vaccination generates an immune response to an infectious agent. In some aspects, vaccination generates an immune response to a tumor; in some such aspects, vaccination is “personalized” in that it is partly or wholly directed to epitope(s) (e.g., which may be or include one or more neoepitopes) determined to be present in a particular individual’s tumors. An immune response refers to a humoral response, a cellular response, or both a humoral and cellular response in an organism. An immune response may be measured by assays that include, but are not limited to, assays measuring the presence or amount of antibodies that specifically recognize a protein or cell surface protein, assays measuring T-cell activation or proliferation, and/or assays that measure modulation in terms of activity or expression of one or more cytokines. As used herein, the term “combination therapy” refers to those situations in which a subject is simultaneously exposed to two or more therapeutic regimens (e.g., two or more therapeutic agents). In some aspects, the two or more regimens may be administered simultaneously; in some aspects, such regimens may be administered sequentially (e.g., all “doses” of a first regimen are administered prior to administration of any doses of a second regimen); in some aspects, such agents are administered in overlapping dosing regimens. In some aspects, “administration” of combination therapy may involve administration of one or more agent(s) or modality(ies) to a subject receiving the other agent(s) or modality(ies) in the combination. For clarity, combination therapy does not require that individual agents be administered together in a single composition (or even necessarily at the same time), although in some aspects, two or more agents, or active moieties thereof, may be administered together in a combination composition, or even in a combination compound (e.g., as part of a single chemical complex or covalent entity). Those skilled in the art will appreciate that the term “dosing regimen” may be used to refer to a set of unit doses (typically more than one) that are administered individually to a subject, typically separated by periods of time. In some aspects, a given therapeutic agent has a recommended dosing regimen, which may involve one or more doses. In some aspects, a dosing regimen comprises a plurality of doses each of which is separated in time from other doses. In some aspects, individual doses are separated from one another by a time period of the same length; in some aspects, a dosing regimen comprises a plurality of doses and at least two different time periods separating individual doses. In some aspects, all doses within a dosing regimen are of the same unit dose amount. In some aspects, different doses within a dosing regimen are of different amounts. In some aspects, a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount different from the first dose amount. In some aspects, a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount same as the first dose amount. In some aspects, a dosing regimen is correlated with a desired or beneficial outcome when administered across a relevant population (e.g., is a therapeutic dosing regimen). II. RESPIRATORY SYNCYTIAL VIRUS (RSV) The present disclosure provides for RNA molecules (e.g., RNA polynucleotides) comprising at least one open reading frame encoding a respiratory syncytial virus (RSV) polypeptide. The present disclosure further provides for an immunogenic composition comprising at least one RNA molecule encoding an RSV polypeptide complexed with, encapsulated in, or formulated with one or more lipids, and forming lipid nanoparticles (LNPs). The RSV polypeptide to be included in the immunogenic composition disclosed herein can be any RSV F protein in the prefusion conformation. The term “prefusion conformation” refers to a structural conformation adopted by an RSV F protein or mutant thereof that can be specifically bound by (i) antibody D25 or AM22 when the RSV F protein or mutant is in the form of a monomer or trimer, or (ii) by antibody AM14 when the RSV F protein mutant is in the form of a trimer. The prefusion trimer conformation is a subset of prefusion conformations. As used herein, an RSV F protein or polypeptide or mutant thereof in prefusion conformation may be denoted as “RSV preF”. The term “postfusion conformation” refers to a structural conformation adopted by the RSV F protein that is not specifically bound by D25, AM22, or AM14. Native F protein adopts the postfusion conformation subsequent to the fusion of the virus envelope with the host cellular membrane. RSV F protein may also assume the postfusion conformation outside the context of a fusion event, for example, under stress conditions such as heat and low osmolality, when extracted from a membrane, when expressed as an ectodomain, or upon storage. The term “AM14” refers to an antibody described in WO 2008/147196 A2, which is hereby incorporated by reference in its entirety. The term “AM22” refers to an antibody described in WO 2011/043643 A1, which is hereby incorporated by reference in its entirety. The term “D25” refers to an antibody described in WO 2008/147196 A2, which is hereby incorporated herein by reference in its entirety. In some embodiments, the RSV F protein is an RSV F protein of subtype A. In some embodiments, the RSV F protein is an RSV F protein of subtype B. As used herein the terms “subtype” and “subgroup” are used interchangeably. As used herein the term “strain” refers to a specific isolate within each subtype or subgroup. In some embodiments, the RSV F protein is a mutant of wild type RSV F protein. In some embodiments, the RSV F protein is a mutant of wild type RSV F protein of subtype A. In some embodiments, the RSV F protein is a mutant of wild type RSV F protein of subtype B. In some embodiments, the mutants display introduced mutations in the amino acid sequence relative to the amino acid sequence of the corresponding wild-type RSV F protein and are immunogenic against the wild-type RSV F protein in the prefusion conformation or against a virus comprising the wild-type F protein. The amino acid mutations in the mutants include amino acid substitutions, deletions, or additions relative to a wild-type RSV F protein. In some embodiments, the RSV F protein is an RSV protein mutant as described in WO2017/109629, which is hereby incorporated by reference in its entirety. In some embodiments, the RSV F protein is a mutant of a wild-type RSV F protein, wherein the introduced amino acid mutations are mutation of a pair of amino acid residues in a wild-type RSV F protein to a pair of cysteines (”engineered disulfide mutation”). The introduced pair of cysteine residues allows for formation of a disulfide bond between the cysteine residues that stabilize the protein’s conformation or oligomeric state, such as the prefusion conformation. Examples of specific pairs of such mutations include: 55C and 188C; 155C and 290C; 103C and 148C; and 142C and 371C, such as S55C and L188C; S155C and S290C; A103C and I148C; and L142C and N371C. In still other embodiments, the RSV F protein mutants comprise amino acid mutations that are one or more cavity filling mutations. Examples of amino acids that may be replaced with the goal of cavity filling include small aliphatic (e.g. Gly, Ala, and Val) or small polar amino acids (e.g. Ser and Thr) and amino acids that are buried in the prefusion conformation, but exposed to solvent in the postfusion conformation. Examples of the replacement amino acids include large aliphatic amino acids (Ile, Leu and Met) or large aromatic amino acids (His, Phe, Tyr and Trp). In some specific embodiments, the RSV F protein mutant comprises a cavity filling mutation selected from the group consisting of: (1) substitution of S at positions 55, 62, 155, 190, or 290 with I, Y, L, H, or M; (2) substitution of T at position 54, 58, 189, 219, or 397 with I, Y, L, H, or M; (3) substitution of G at position 151 with A or H; (4) substitution of A at position 147 or 298 with I, L, H, or M; (5) substitution of V at position 164, 187, 192, 207, 220, 296, 300, or 495 with I, Y, H; and (6) substitution of R at position 106 with W. In some particular embodiments, the RSV F protein mutant comprises at least one cavity filling mutation selected from the group consisting of: T54H, S190I, and V296I. In still other embodiments, the RSV F protein mutants comprise electrostatic mutations, which decrease ionic repulsion or increase ionic attraction between resides in a protein that are proximate to each other in the folded structure. In several embodiments, the RSV F protein mutant includes an electrostatic substitution that reduces repulsive ionic interactions or increases attractive ionic interactions with acidic residues of Glu487 and Asp489 from another protomer of RSV F trimer. In some specific embodiments, the RSV F protein mutant comprises an electrostatic mutation selected from the group consisting of: (1) substitution of E at position 82, 92, or 487 by D, F, Q, T, S, L, or H; (2) substitution of K at position 315, 394, or 399 by F, M, R, S, L, I, Q, or T; (3) substitution of D at position 392, 486, or 489 by H, S, N, T, or P; and (4) substitution of R at position 106 or 339 by F, Q, N, or W. In still other embodiments, the RSV F protein mutants comprise a combination of two or more different types of mutations selected from engineered disulfide mutations, cavity filling mutations, and electrostatic mutations. In some particular embodiments, the RSV F protein mutants comprise a combination of mutations relative to the corresponding wild-type RSV F protein, wherein the combination of mutations is selected from the group consisting of: (1) combination of A103C, I148C, S190I, and D486S; (2) combination of T54H S55C L188C D486S; (3) combination of T54H, A103C, I148C, S190I, V296I, and D486S; (4) combination of T54H, S55C, L142C, L188C, V296I, and N371C; (5) combination of S55C, L188C, and D486S; (6) combination of T54H, S55C, L188C, and S190I; (7) combination of S55C, L188C, S190I, and D486S; (8) combination of T54H, S55C, L188C, S190I, and D486S; (9) combination of S155C, S190I, S290C, and D486S; (10) combination of T54H, S55C, L142C, L188C, V296I, N371C, D486S, E487Q, and D489S; (11) combination of T54H, S155C, S190I, S290C, and V296I, and, (12) combination of S155C, S190F, S290C, and V207L. In some embodiments, the RSV F protein is of subtype A and comprises the mutations S155C, S190F, S290C, and V207L. In some embodiments, the RSV F protein is of subtype B and comprises the mutations S155C, S190F, S290C, and V207L. In some embodiments, the RSV F protein is of subtype A and comprises the mutations S155C, S190F, and S290C. In some embodiments, the RSV F protein is of subtype B and comprises the mutations S155C, S190F, and S290C. In some embodiments, the RSV F protein is of subtype A and comprises the mutations A103C, I148C, S190I, and D486S. In some embodiments, the RSV F protein is of subtype B and comprises the mutations A103C, I148C, S190I, and D486S. In some embodiments, the RSV F protein is of subtype A and comprises the mutations T54H, A103C, I148C, S190I, and D486S. In some embodiments, the RSV F protein is of subtype B and comprises the mutations T54H, A103C, I148C, S190I, and D486S. In some embodiments, the RSV F protein is of subtype A and comprises the mutations T54H, S55C, L188C, and D486S. In some embodiments, the RSV F protein is of subtype B and comprises the mutations T54H, S55C, L188C, and D486S. In view of the substantial conservation of RSV F sequences, a person of ordinary skill in the art can easily compare amino acid positions between different native RSV F sequences to identify corresponding RSV F amino acid positions between different RSV strains and subtypes. For example, across nearly all identified native RSV F0 precursor proteins, the furin cleavage sites fall in the same amino acid positions. Thus, the conservation of native RSV F protein sequences across strains and subtypes allows use of a reference RSV F sequence for comparison of amino acids at particular positions in the RSV F protein. For the purposes of this disclosure (unless context indicates otherwise), the RSV F protein amino acid positions are given with reference to the amino acid sequence of the full length native F precursor polypeptide of the RSV A2 strain; corresponding to GenInfo Identifier GI 138251 and Swiss Prot identifier P03420 (SEQ ID NO: 1). In some embodiments, the RSV F protein is in the mature form of the RSV F protein, which comprises two separate polypeptide chains, namely the F1 polypeptide and F2 polypeptide. In some other embodiments, the F2 polypeptide is linked to the F1 polypeptide by one or two disulfide bonds to form a F2/F1 heterodimer. In still other embodiments, the RSV F mutants are in the form a single chain protein, wherein the F2 polypeptide is linked to the F1 polypeptide by a peptide bond or peptide linker. Any suitable peptide linkers for joining two polypeptide chains together may be used. Examples of such linkers include G, GG, GGG, GS, and SAIG linker sequences. The linker may also be the full length pep27 sequence or a fragment thereof, which full length pep27 sequence corresponds to amino acids at positions 110-136 of SEQ ID NO:1. The F1 polypeptide chain of the mutant may be of the same length as the full length F1 polypeptide of the corresponding wild-type RSV F protein; however, it may also have deletions, such as deletions of 1 up to 60 amino acid residues from the C-terminus of the full-length F1 polypeptide. A full-length F1 polypeptide of the RSV F mutants corresponds to amino acid positions 137-574 of the native RSV F0 precursor (SEQ ID NO: 1), and includes (from N- to C- terminus) an extracellular region (residues 137-524), a transmembrane domain (“TM”) (residues 525-550), and a cytoplasmic domain (“CT”) (residues 551-574). It should be noted that amino acid residues 514 onwards in a native F1 polypeptide sequence are optional sequences in a F1 polypeptide of the RSV F protein to be included in the immunogenic composition provided herein, and therefore may be absent from the F1 polypeptide of the mutant. In some embodiments, the F1 polypeptide of the RSV F mutants lacks the entire cytoplasmic domain. In other embodiments, the F1 polypeptide lacks the cytoplasmic domain and a portion of or all entire transmembrane domain. In some specific embodiments, the mutant comprises a F1 polypeptide wherein the amino acid residues from position 510, 511, 512, 513, 514, 515, 520, 525, or 530 through 574 are absent. Typically, for mutants that are linked to trimerization domain, such as a foldon, amino acids 514 through 574 can be absent. Thus, in some specific embodiment, amino acid residues 514 through 574 are absent from the F1 polypeptide of the mutant. In still other specific embodiments, the F1 polypeptide of the RSV F mutants comprises or consists of amino acid residues 137-513 of a native F0 polypeptide sequence (SEQ ID NO: 1) or any of alternative F0 precursor sequence such as those disclosed in SEQ ID NOs: 1, 2, 4, 6, and 81-270 of WO2017109629, which is hereby incorporated by reference in its entirety. The F1 polypeptide and F2 polypeptide of the RSV F protein mutants to which one or more mutations are introduced can be from any wild-type RSV F proteins known in the art or discovered in the future, including, without limitations, the F protein amino acid sequence of RSV subtype A, and subtype B strains, including A2 Ontario and Buenos Aires, or any other subtype. In some embodiments, the RSV F mutant comprises a F1 and/or a F2 polypeptide from a RSV A virus, for example, a F1 and/or F2 polypeptide from a RSV F0 precursor protein set forth in any one of SEQ ID NOs: 1, 2, 4, 6, and 81-270 of WO2017109629, which sequences are hereby incorporated by reference in their entireties, to which one or more mutations are introduced. In some other embodiments, the RSV F mutant comprises a F1 and/or a F2 polypeptide from a RSV B virus, for example, a F1 and/or F2 polypeptide from a RSV F0 precursor protein set forth in any one of SEQ ID NOs: 2 and 211-263 of WO2017/109629, which sequences are hereby incorporated by reference in their entireties, to which one or more mutations are introduced. In still other embodiments, the RSV F mutant comprises a F1 and/or a F2 polypeptide from a RSV bovine virus, for example, a F1 and/or F2 polypeptide from a RSV F0 precursor protein set forth in any one of SEQ ID NOs: 264-270 of WO2017109629, which sequences are hereby incorporated by reference in their entireties, to which one or more mutations are introduced. The term “F0 polypeptide” (F0) refers to the precursor polypeptide of the RSV F protein, which is composed of a signal polypeptide sequence, a F1 polypeptide sequence, a pep27 polypeptide sequence, and a F2 polypeptide sequence. With rare exceptions the F0 polypeptides of the known RSV strains consist of 574 amino acids. The term “F1 polypeptide” (F1) refers to a polypeptide chain of a mature RSV F protein. Native F1 includes approximately residues 137-574 of the RSV F0 precursor and is composed of (from N- to C-terminus) an extracellular region (approximately residues 137-524), a transmembrane domain (“TM”) (approximately residues 525-550), and a cytoplasmic tail (“CT”) (approximately residues 551-574). As used herein, the term encompasses both native F1 polypeptides and F1 polypeptides including modifications (e.g., amino acid substitutions, insertions, or deletions) from the native sequence, for example, modifications designed to stabilize an RSV F protein mutant or to enhance the immunogenicity of an RSV F protein mutant. The term “F2 polypeptide” (F2) refers to the polypeptide chain of a mature RSV F protein. Native F2 includes approximately residues 26-109 of the RSV F0 precursor. As used herein, the term encompasses both native F2 polypeptides and F2 polypeptides including modifications (e.g., amino acid substitutions, insertions, or deletions) from the native sequence, for example, modifications designed to stabilize an RSV F protein mutant in a prefusion conformation or to enhance the immunogenicity of an RSV F protein mutant. In native RSV F protein, the F2 polypeptide is linked to the F1 polypeptide by two disulfide bonds to form a F2-F1 heterodimer. The term “foldon” or “foldon domain” refers to an amino acid sequence that is capable of forming trimers. One example of such foldon domains is the peptide sequence derived from bacteriophage T4 fibritin, which has the sequence of GYIPEAPRDGQAYVRKDGEWVLLSTFL (SEQ ID NO: 43). In some aspects, the RNA molecule encodes an RSV F protein mutant as disclosed in WO2009/079796, WO2010/149745, WO2011/008974, WO2014/160463, WO2014/174018, WO2014/202570, WO2015/013551, WO2015/177312, WO2017/005848, WO2017/174564, WO2017/005844 and WO2018/109220. The RSV F proteins disclosed in these references are hereby incorporated by reference in their entirety. Antibodies to RSV F protein are prevalent after natural infection and following vaccination and have been shown to neutralize viral activity in vitro. As used herein, the term “respiratory syncytial virus” or “RSV” is not limited to any particular strain or variant. In some aspects, the RNA molecule comprises an open reading frame encoding a RSV antigen. In some aspects, the RSV antigen is a RSV polypeptide. In some aspects, the RSV polypeptide is a RSV glycoprotein or a fragment or a variant thereof. In some aspects, the RNA molecule encodes a RSV F protein. In some aspects, the RSV polypeptide is a full-length RSV polypeptide. In some aspects, the RSV polypeptide is a truncated RSV polypeptide. In some aspects, the RSV polypeptide is a variant of a RSV polypeptide. In some aspects, the RSV polypeptide is a fragment of a RSV polypeptide. In some aspects, the RSV polypeptide is a full-length RSV F protein. In some aspects, the RSV polypeptide is a truncated RSV F protein. In some aspects, the RSV polypeptide is a variant of a RSV F protein. In some aspects, the RSV polypeptide is a fragment of a RSV F protein. In some aspects, the RSV F protein comprises at least one mutation. In some aspects, the RSV F protein comprises at least two mutations. In some aspects, the RSV F protein comprises at least three mutations. In some aspects, the RSV F protein comprises at least four mutations. In some aspects, the RSV F protein comprises 4 mutations. In some aspects, the RSV F protein comprises at least five mutations. In some aspects, the RNA molecule encodes a RSV F protein as set forth in Table 1. In some aspects, the RNA molecule encodes a RSV F protein comprising an amino acid sequence of any of SEQ ID NO: 1 to 6, or fragment or variant thereof. In some aspects, RSV F polypeptide may have at least, at most, exactly, or between any two of 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to any of the amino acid sequences of Table 1, for example, any of SEQ ID NO: 1 to 6. In some aspects, RSV F protein consists of any of the amino acid sequences of Table 1, for example, any of SEQ ID NO: 1 to 6. In some aspects, the RNA molecule sequence is transcribed from a DNA nucleic acid sequence (DNA polynucleotide) of Table 2 and at least one untranslated region (UTR) selected from Table 4, provided that when the ORF is SEQ ID NO: 8 or 10 that the UTR does not consist of only SEQ ID NOs: 111 and 280. In some aspects, the RNA molecule comprises an ORF transcribed from a nucleic acid sequence of any of SEQ ID NO: 7 to 10, or fragment or variant thereof. In some aspects, the RNA molecule is transcribed from a nucleic acid having a sequence that may have at least, at most, exactly, or between any two of 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to any of the nucleic acid sequences of Table 2, for example, any of SEQ ID NO: 7 to 14. In some aspects, the RNA molecule is transcribed from a nucleic acid having a sequence that consists of any of the nucleic acid sequences of Table 2, for example, any of SEQ ID NO:7 to 14. In some aspects, the RNA molecule comprises an ORF encoding RSV F comprising an RNA nucleic acid sequence (RNA polynucleotide) of Table 3 and at least one untranslated region (UTR) selected from Table 5, provided that when the ORF is SEQ ID NO: 15 or 16 that the UTR does not consist of only SEQ ID NOs: 397 and 566. In some aspects, the RNA molecule comprises a nucleic acid having a sequence of any of SEQ ID NO: 15 to 20, or fragment or variant thereof. In some aspects, the RNA molecule comprises a nucleic acid having a sequence that may have at least, at most, exactly, or between any two of 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to any of the RNA nucleic acid sequences of Table 3, for example, any of SEQ ID NO: 15 to 20. In some aspects, the RNA molecule comprises a nucleic acid having a sequence that consists of any of the RNA nucleic acid sequences of Table 3, for example, any of SEQ ID NO: 15 to 20. In some aspects, the RNA molecule comprises stabilized RNA. In some aspects, the RNA molecule comprises a nucleic acid sequence having at least one uridine replaced by N1- methylpseudouridine. In some aspects, the RNA molecule comprises a sequence having all uridines replaced by N1-methylpseudouridine (designated as “Ψ”). In some aspects, the RNA molecule comprises an ORF comprising a nucleic acid sequence of any of SEQ ID NO: 15 or 16, wherein all uridines have been replaced by N1-methylpseudouridine (designated as “Ψ”). In some aspects, the RNA molecule comprises an open reading frame encoding a RSV F protein amino acid sequence that may be at least, at most, exactly, or between any two of 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to any of the RSV F protein sequences of SEQ ID NO: 1 to 6 (Table 1) or other RSV prefusion F proteins described herein. In some aspects, the RNA molecule comprises an open reading frame encoding a RSV F protein amino acid sequence that consists of any of the RSV F protein sequences of SEQ ID NO: 1 to 6 (Table 1) or other RSV prefusion F protein described herein. In some aspects, the RNA molecule comprises an open reading frame transcribed from a DNA nucleic acid sequence that may be at least, at most, exactly, or between any two of 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to any of the nucleic acid sequences of SEQ ID NO: 7 to 14 (Table 2) or other nucleic acid described herein. In some aspects, the RNA molecule comprises an open reading frame transcribed from a DNA nucleic acid sequence that consists of any of the nucleic acid sequences of SEQ ID NO: 7 to 14 (Table 2) or other nucleic acid described herein. In some aspects, the RNA molecule comprises an open reading frame encoding a RSV F, wherein the RNA nucleic acid sequence may be at least, at most, exactly, or between any two of 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to any of the nucleic acid sequences of SEQ ID NO: 15 to 20 (Table 3) or other nucleic acid described herein. In some aspects, the RNA molecule comprises an open reading frame encoding a RSV F, wherein the RNA nucleic acid sequence consists of any of the nucleic acid sequences of SEQ ID NO: 15 to 20 (Table 3) or other nucleic acid described herein. In some aspects, the RNA molecule comprises an ORF encoding a RSV F, wherein the nucleic acid sequence is any of SEQ ID NO: 15 to 20 (Table 3), wherein all uridines have been replaced by N1-methylpseudouridine (designated as “m1Ψ”). III. RNA MOLECULE In some aspects, the RNA molecule described herein is a coding RNA molecule. Coding RNA includes a functional RNA molecule that may be translated into a peptide or polypeptide. In some aspects, the coding RNA molecule includes at least one open reading frame (ORF) coding for at least one peptide or polypeptide. An open reading frame comprises a sequence of codons that is translatable into a peptide or protein. The coding RNA molecule may include one (monocistronic), two (bicistronic) or more (multicistronic) ORFs, which may be a sequence of codons that is translatable into a polypeptide or protein of interest. The coding RNA molecule may be a messenger RNA (mRNA) molecule, viral RNA molecule, or self-amplifying RNA molecule (saRNA, also referred to as a replicon). In some aspects, the RNA molecule is an mRNA. Preferably, the RNA molecule of the present disclosure is an mRNA. In some aspects, the RNA molecule is modRNA. In some aspects, the RNA molecule is a saRNA. In some aspects, the saRNA molecule may be a coding RNA molecule. The RNA molecule may encode one polypeptide of interest or more, such as an antigen or more than one antigen, e.g., two, three, four, five, six, seven, eight, nine, ten or more polypeptides. Alternatively, or in addition, one RNA molecule may also encode more than one polypeptide of interest, such as an antigen, e.g., a bicistronic, or tricistronic RNA molecule that encodes different or identical antigens. The sequence of the RNA molecule may be codon optimized or deoptimized for expression in a desired host, such as a human cell. In some aspects, a gene of interest (e.g., an antigen) described herein is encoded by a coding sequence which is codon-optimized and/or the guanosine/cytidine (G/C) content of which is increased compared to wild type coding sequence. In some aspects, one or more sequence regions of the coding sequence are codon-optimized and/or increased in the G/C content compared to the corresponding sequence regions of the wild type coding sequence. In some aspects, codon-optimization and/or increasing the G/C content does not change the sequence of the encoded amino acid sequence. The term “codon-optimized” is understood by those in the art to refer to alteration of codons in the coding region of a nucleic acid molecule to reflect the typical codon usage of a host organism without altering the amino acid sequence encoded by the nucleic acid molecule. Within the context of the present disclosure, in some aspects, coding regions are codon-optimized for optimal expression in a subject to be treated using an RNA polynucleotide described herein. Codon-optimization is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNA molecules in cells. Thus, the sequence of RNA may be modified such that codons for which frequently occurring tRNA molecules are available are inserted in place of “rare codons.” In some aspects, G/C content of a coding region (e.g., of a gene of interest sequence; open reading frame (ORF)) of an RNA is increased compared to the G/C content of the corresponding coding sequence of a wild type RNA encoding the gene of interest, wherein in some aspects, the amino acid sequence encoded by the RNA is not modified compared to the amino acid sequence encoded by the wild type RNA. This modification of the RNA sequence is based on the fact that the sequence of any RNA region to be translated is important for efficient translation of that mRNA. Sequences having an increased G (guanosine)/C (cytidine) content are more stable than sequences having an increased A (adenosine)/U (uridine) content. In respect to the fact that several codons code for one and the same amino acid (so-called degeneration of the genetic code), the most favorable codons for the stability may be determined (so-called alternative codon usage). Depending on the amino acid to be encoded by the RNA, there are various possibilities for modification of the RNA sequence, compared to its wild type sequence. In particular, codons which contain A and/or U nucleosides may be modified by substituting these codons by other codons, which code for the same amino acids but contain no A and/or U or contain a lower content of A and/or U nucleosides. Thus, in some aspects, G/C content of a coding region of an RNA described herein is increased by at least, at most, exactly, or between any two of 10%, 20%, 30%, 40%, 50%, 55%, or even more compared to the G/C content of a coding region of a wild type RNA. In some aspects, the coding region of the RSV RNA described herein comprises a G/C content of at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, or about 80%. In some aspects, the coding region of the RSV RNA described herein comprises a G/C content of about 50% to 75%, about 55% to 70%, about 50% to 60%, about 60% to 70%, about 70% to 80%, about 50% to 55%, about 55% to 60%, about 60% to 65%, about 65% to 70%, about 70% to 75%, or about 75% to 80%. In some aspects, the coding region of the RSV RNA described herein comprises a G/C content of about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, about 60%, about 61%, about 62%, about 63%, about 64%, about 65%, about 66%, about 67%, about 68%, about 69%, about 70%, about 71%, about 72%, about 73%, about 74%, or about 75%. In some aspects, the coding region of the RSV RNA described herein comprises a G/C content of about 58%, about 66% or about 62%. In some aspects, the RNA molecule includes from about 20 to about 100,000 nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to 500, from 30 to 1,000, from 30 to 1,500, from 30 to 3,000, from 30 to 5,000, from 30 to 7,000, from 30 to 10,000, from 30 to 25,000, from 30 to 50,000, from 30 to 70,000, from 100 to 250, from 100 to 500, from 100 to 1,000, from 100 to 1,500, from 100 to 3,000, from 100 to 5,000, from 100 to 7,000, from 100 to 10,000, from 100 to 25,000, from 100 to 50,000, from 100 to 70,000, from 100 to 100,000, from 500 to 1,000, from 500 to 1,500, from 500 to 2,000, from 500 to 3,000, from 500 to 5,000, from 500 to 7,000, from 500 to 10,000, from 500 to 25,000, from 500 to 50,000, from 500 to 70,000, from 500 to 100,000, from 1,000 to 1,500, from 1,000 to 2,000, from 1,000 to 3,000, from 1,000 to 5,000, from 1,000 to 7,000, from 1,000 to 10,000, from 1,000 to 25,000, from 1,000 to 50,000, from 1,000 to 70,000, from 1,000 to 100,000, from 1,500 to 3,000, from 1,500 to 5,000, from 1,500 to 7,000, from 1,500 to 10,000, from 1,500 to 25,000, from 1,500 to 50,000, from 1,500 to 70,000, from 1,500 to 100,000, from 2,000 to 3,000, from 2,000 to 5,000, from 2,000 to 7,000, from 2,000 to 10,000, from 2,000 to 25,000, from 2,000 to 50,000, from 2,000 to 70,000, and from 2,000 to 100,000 nucleotides). In some aspects, the RNA molecule has at least, at most, exactly, or between any two of about 20, 40, 60, 80, 100, 120, 140, 160, 180, 200, 220, 240, 260, 280, 300, 320, 340, 360, 380, 400, 420, 440, 460, 480, 500, 520, 540, 560, 580, 600, 620, 640, 660, 680, 700, 720, 740, 760, 780, 800, 820, 840, 860, 880, 900, 920, 940, 960, 980, 1000, 1200, 1400, 1600, 1800, 2000, 2200, 2400, 2600, 2800, 3000, 3200, 3400, 3600, 3800, 4000, 4200, 4400, 4600, 4800, 5000, 5200, 5400, 5600, 5800, 6000, 6200, 6400, 6600, 6800, 7000, 7200, 7400, 7600, 7800, 8000, 8200, 8400, 8600, 8800, 9000, 9200, 9400, 9600, 9800, 10000, 12000, 14000, 16000, 18000, 20000, 22000, 24000, 26000, 28000, 30000, 32000, 34000, 36000, 38000, 40000, 42000, 44000, 46000, 48000, 50000, 52000, 54000, 56000, 58000, 60000, 62000, 64000, 66000, 68000, 70000, 72000, 74000, 76000, 78000, 80000, 82000, 84000, 86000, 88000, 90000, 92000, 94000, 96000, 98000, or 100000 nucleotides. In some aspects, the RNA molecule includes at least 100 nucleotides. For example, in some aspects, the RNA has a length between 100 and 15,000 nucleotides; between 7,000 and 16,000 nucleotides; between 8,000 and 15,000 nucleotides; between 9,000 and 12,500 nucleotides; between 11,000 and 15,000 nucleotides; between 13,000 and 16,000 nucleotides; between 7,000 and 25,000 nucleotides. In some aspects, the RNA molecule has at least, at most, exactly, or between any two of about 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350, 1400, 1450, 1500, 1550, 1600, 1650, 1700, 1750, 1800, 1850, 1900, 1950, 2000, 2050, 2100, 2150, 2200, 2250, 2300, 2350, 2400, 2450, 2500, 2550, 2600, 2650, 2700, 2750, 2800, 2850, 2900, 2950, 3000, 3050, 3100, 3150, 3200, 3250, 3300, 3350, 3400, 3450, 3500, 3550, 3600, 3650, 3700, 3750, 3800, 3850, 3900, 3950, 4000, 4050, 4100, 4150, 4200, 4250, 4300, 4350, 4400, 4450, 4500, 4550, 4600, 4650, 4700, 4750, 4800, 4850, 4900, 4950, 5000, 5050, 5100, 5150, 5200, 5250, 5300, 5350, 5400, 5450, 5500, 5550, 5600, 5650, 5700, 5750, 5800, 5850, 5900, 5950, 6000, 6050, 6100, 6150, 6200, 6250, 6300, 6350, 6400, 6450, 6500, 6550, 6600, 6650, 6700, 6750, 6800, 6850, 6900, 6950, 7000, 7050, 7100, 7150, 7200, 7250, 7300, 7350, 7400, 7450, 7500, 7550, 7600, 7650, 7700, 7750, 7800, 7850, 7900, 7950, 8000, 8050, 8100, 8150, 8200, 8250, 8300, 8350, 8400, 8450, 8500, 8550, 8600, 8650, 8700, 8750, 8800, 8850, 8900, 8950, 9000, 9050, 9100, 9150, 9200, 9250, 9300, 9350, 9400, 9450, 9500, 9550, 9600, 9650, 9700, 9750, 9800, 9850, 9900, 9950, 10000, 10050, 10100, 10150, 10200, 10250, 10300, 10350, 10400, 10450, 10500, 10550, 10600, 10650, 10700, 10750, 10800, 10850, 10900, 10950, 11000, 11050, 11100, 11150, 11200, 11250, 11300, 11350, 11400, 11450, 11500, 11550, 11600, 11650, 11700, 11750, 11800, 11850, 11900, 11950, 12000, 12050, 12100, 12150, 12200, 12250, 12300, 12350, 12400, 12450, 12500, 12550, 12600, 12650, 12700, 12750, 12800, 12850, 12900, 12950, 13000, 13050, 13100, 13150, 13200, 13250, 13300, 13350, 13400, 13450, 13500, 13550, 13600, 13650, 13700, 13750, 13800, 13850, 13900, 13950, 14000, 14050, 14100, 14150, 14200, 14250, 14300, 14350, 14400, 14450, 14500, 14550, 14600, 14650, 14700, 14750, 14800, 14850, 14900, 14950, or 15000 nucleotides. The RNA molecules of the present disclosure may be prepared by any method known in the art, including chemical synthesis and in vitro methods, such as RNA in vitro transcription. In some of the aspects, the RNA of the present disclosure is prepared using in vitro transcription. In some aspects, the RNA molecule of the present disclosure is purified, e.g., such as by filtration that may occur via, e.g., ultrafiltration, diafiltration, or, e.g., tangential flow ultrafiltration/diafiltration. In some aspects, the RNA molecule of the present disclosure is lyophilized to be temperature stable. In some aspects of the present disclosure, an RNA is or comprises messenger RNA (mRNA) that relates to an RNA transcript which encodes a polypeptide. In some aspects, an RNA disclosed herein comprises: a 5′ cap comprising a 5′ cap disclosed herein; a 5′ untranslated region comprising a cap proximal sequence (5′ UTR), a sequence encoding a protein (e.g. polypeptide) (e.g., a RSV prefusion F protein); a 3′ untranslated region (3′ UTR); and/or a polyadenylate (poly- A) sequence. In some aspects, an RNA disclosed herein comprises the following components in 5′ to 3′ orientation: a 5′ cap comprising a 5′ cap disclosed herein; a 5′ untranslated region comprising a cap proximal sequence (5′ UTR), a sequence encoding a protein (e.g. polypeptide) (e.g., a RSV prefusion F protein); a 3′ untranslated region (3′ UTR); and a poly-A sequence. In some aspects, an RNA disclosed herein further comprises a signal peptide. Non- limiting examples of signal peptides and amino acid and nucleic acid sequences encoding such peptides can be found in, e.g., WO2017/109629, the disclosure of which is incorporated by reference herein in its entirety. In some aspects, an RNA disclosed herein encodes an antigenic fusion protein. Thus, the encoded antigen or antigens may include two or more proteins (e.g., protein and/or protein fragment) joined together. Alternatively, the protein to which a protein antigen is fused does not promote a strong immune response to itself, but rather to an antigen. Antigenic fusion proteins, in some aspects, retain the functional property from each original protein. In some aspects, an RNA disclosed herein encodes fusion proteins that comprise an antigen linked to a scaffold moieties. In some aspects, the RNA further encodes a linker located between at least one or each domain of the fusion protein. Non-limiting examples of such scaffold moieties and linkers can be found in, e.g., WO 2022/067010, the disclosure of which is incorporated by reference herein in its entirety. A. MODIFIED NUCLEOBASES In some aspects of the present disclosure, the RNA molecules are not chemically modified and comprise the standard ribonucleotides consisting of adenosine, guanosine, cytosine and uridine. In some aspects, nucleotides and nucleosides of the present disclosure comprise standard nucleoside residues such as those present in transcribed RNA (e.g., A, G, C, and/or U). In some aspects, nucleotides and nucleosides of the present disclosure comprise standard deoxyribonucleosides such as those present in DNA (e.g., dA, dG, dC, and/or dT). In other aspects of the present disclosure the RNA molecules may comprise modified nucleobases which may be incorporated into modified nucleosides and nucleotides. In some aspects, the RNA molecule may include one or more modified nucleotides. In some aspects, the RNA molecule may include one or more modified nucleotides. Naturally occurring nucleotide modifications are known in the art. In some aspects, the RNA molecule may include a modified nucleotide. Non-limiting examples of modified nucleotides that may be included in the RNA molecule include pseudouridine, N1-methylpseudouridine, 5-methyluridine, 3-methyl-uridine, 5- methoxy-uridine, 5-aza-uridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2-thio-uridine, 4-thio-uridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-uridine, 5-aminoallyl-uridine, 5-halo- uridine (e.g., 5-iodo-uridine or 5-bromo-uridine), uridine 5-oxyacetic acid, uridine 5-oxyacetic acid methyl ester, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5-carboxy hydroxymethyl-uridine, 5-carboxy hydroxy methyl-uridine methyl ester, 5- methoxycarbonylmethyl-uridine, 5-methoxycarbonylmethyl-2-thio-uridine, 5-aminomethyl-2-thio- uridine, 5-methylaminomethyl-uridine, 1-ethyl-pseudouridine, 5-methylaminomethyl-2-thio- uridine, 5-methylaminomethyl-2-seleno-uridine, 5-carbamoylmethyl-uridine, 5- carboxymethylaminomethyl-uridine, 5-carboxymethylaminomethyl-2-thio-uridine, 5-propynyl- uridine, 1-propynyl-pseudouridine, 5-taurinomethyl-uridine, 1-taurinomethyl-pseudouridine, 5- taurinomethyl-2-thio-uridine, 1-taurinomethyl-4-thio-pseudouridine, 5-methyl-2-thio-uridine, 1- methyl-4-thio-pseudouridine, 4-thio-1-methyl-pseudouridine, 3-methyl-1-pseudouridine, 2-thio-1- methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-deaza- pseudouridine, dihydrouridine, dihydropseudouridine, 5,6-dihydrouridine, 5-methyl- dihydrouridine, 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-methoxy-uridine, 2- methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4-methoxy-2-thio-pseudouridine, N1-methyl- pseudouridine, 3-(3-amino-3-carboxypropyl)uridine, 1-methyl-3-(3-amino-3- carboxypropyl)pseudouridine, 5-(isopentenylaminomethyl)uridine, 5-(isopentenylaminomethyl)- 2-thio-uridine, a-thio-uridine, 2′-O-methyl-uridine, 5,2′-O-dimethyl-uridine, 2′-O-methyl- pseudouridine, 2-thio-2′-O-methyl-uridine, 5-methoxycarbonylmethyl-2′-O-methyl-uridine, 5- carbamoylmethyl-2′-O-methyl-uridine, 5-carboxymethylaminomethyl-2′-O-methyl-uridine, 3,2′-O- dimethyl-uridine, 5-(isopentenylaminomethyl)-2′-O-methyl-uridine, 1-thio-uridine, deoxythymidine, 2′-F-ara-uridine, 2′-F-uridine, 2′-OH-ara-uridine, 5-(2-carbomethoxyvinyl) uridine, 5-[3-(1-E-propenylamino)uridine, any other modified uridine known in the art, or combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing modified nucleotides can be excluded from the RNA molecules disclosed herein. Modifications that may be present in the RNA molecules further include, but are not limited to, e.g., the following: ms2io6A (2-methylthio-(N6-(cis-hydroxyisopentenyl)adenosine); ms2m6A (2-methylthio-N6-methyladenosine); ms2t6A 2-methylthio-N6-threonylcarbamoyladenosine; g6A (N6-glycinylcarbamoyladenosine); i6A (N6-isopentenyladenosine); m6A (N6-methyladenosine); t6A (N6-threonylcarbamoyladenosine); m′Am (1,2′-O-dimethyladenosine); m1A (1- methyladenosine); 2′-O-methyladenosine; Ar(p) (2′-O-ribosyladenosine (phosphate)); 2-m ethyl adenosine; 2-methylthio-N6 isopentenyladenosine; ms2hn6A (2-methylthio-N6- hydroxynorvalylcarbamoyladenosine); 2-O-methyladenosine; Am (2-1-O-methyladenosine); 2′- O-ribosyladenosine (phosphate); Isopentenyladenosine; io6A N6-(cis- hydroxyisopentenyl)adenosine; m6Am (N6,2′-O-dimethyladenosine); m62Am (N6,N6,2′-O- trimethyladenosine); m62A (N6,N6-dimethyladenosine); ac6A (N6-acetyladenosine); hn6A (N6- hydroxynorvalylcarbamoyladenosine); m6t6A (N6-methyl-N6-threonylcarbamoyladenosine); m2A (2-methyladenosine); ms2i6A (2-methylthio-N6-isopentenyladenosine); 7-deaza- adenosine; N1-methyl-adenosine; N6,N6 (dimethyl)adenine; N6-cis-hydroxy-isopentenyl- adenosine; a-thio-adenosine; 2 (amino)adenine; 2 (aminopropyl)adenine; 2 (methylthio) N6 (isopentenyl)adenine; 2-(alkyl)adenine; 2-(aminoalkyl)adenine; 2-(aminopropyl)adenine; 2- (halo)adenine; 2-(halo)adenine; 2-(propyl)adenine; 2′-amino-2′-deoxy-ATP; 2′-azido-2′-deoxy- ATP; 2′-deoxy-2′-a-aminoadenosine TP; 2′-deoxy-2′-a-azidoadenosine TP; 6 (alkyl)adenine; 6 (methyl)adenine; 6-(alkyl)adenine; 6-(methyl)adenine; 7 (deaza)adenine; 8 (alkenyl)adenine; 8 (alkynyl)adenine; 8 (amino)adenine; 8 (thioalkyl)adenine; 8-(alkenyl)adenine; 8-(alkyl)adenine; 8- (alkynyl)adenine; 8-(amino)adenine; 8-(halo)adenine; 8-(hydroxyl)adenine; 8-(thioalkyl)adenine; 8-(thiol)adenine; 8-azido-adenosine; 8-oxo-adenine; aza adenine; deaza adenine; N6 (methyl)adenine; N6-(isopentyl)adenine; 7-deaza-8-aza-adenosine; 7-methyladenine; 1- deazaadenosine TP; 2′fluoro-N6-Bz-deoxyadenosine TP; 2′-OMe-2-amino-ATP; 2′O-methyl-N6- Bz-deoxyadenosine TP; 2′-a-ethynyladenosine TP; 2-aminoadenine; 2-aminoadenosine TP; 2- amino-ATP; 2′-a-trifluoromethyladenosine TP; 2-azidoadenosine TP; 2′-b-Ethynyladenosine TP; 2-bromoadenosine TP; 2′-b-trifluoromethyladenosine TP; 2-chloroadenosine TP; 2′-deoxy-2′,2′- difluoroadenosine TP; 2′-deoxy-2′-a-mercaptoadenosine TP; 2′-deoxy-2′-a- thiomethoxyadenosine TP; 2′-deoxy-2′-b-aminoadenosine TP; 2′-deoxy-2′-b-azidoadenosine TP; 2′-deoxy-2′-b-bromoadenosine TP; 2′-deoxy-2′-b-chloroadenosine TP; 2′-deoxy-2′-b- fluoroadenosine TP; 2′-deoxy-2′-b-iodoadenosine TP; 2′-deoxy-2′-b-mercaptoadenosine TP; 2′- deoxy-2′-b-thiomethoxyadenosine TP; 2-fluoroadenosine TP; 2-iodoadenosine TP; 2- mercaptoadenosine TP; 2-methoxy-adenine; 2-methylthio-adenine; 2-Trifluoromethyladenosine TP; 3-deaza-3-bromoadenosine TP; 3-deaza-3-chloroadenosine TP; 3-deaza-3-fluoroadenosine TP; 3-deaza-3-iodoadenosine TP; 3-deazaadenosine TP; 4′-Azidoadenosine TP; 4′-Carbocyclic adenosine TP; 4′-Ethynyladenosine TP; 5′-Homo-adenosine TP; 8-Aza-ATP; 8-bromo-adenosine TP; 8-Trifluoromethyladenosine TP; 9-deazaadenosine TP; 2-aminopurine; substituted 7- deazapurine; 7-deaza-7-substituted purine; 7-deaza-8-substituted purine; 7-deaza-2,6- diaminopurine; 7-deaza-8-aza-2,6-diaminopurine; 7-deaza-8-aza-2-aminopurine; 2,4- diaminopurine; 2,6-diaminopurine; 7-deaza-8-aza-adenine; 7-deaza-2-aminopurine; 8- azapurine; s2C (2-thiocytidine); m3C (3 -methylcytidine); f5C (5-formylcytidine); hm5C (5- hydroxymethylcytidine); m5C (5-methylcytidine); ac4C (N4-acetylcytidine); Cm (2′-O- methylcytidine); m5Cm (5,2′-O-dimethylcytidine); f5Cm (5-formyl-2′-O-methylcytidine); k2C (Lysidine); m4Cm (N4,2′-O-dimethylcytidine); ac4Cm (N4-acetyl-2′-O-methylcytidine); m4C (N4- methylcytidine); N4,N4-dimethyl-2′-OMe-Cytidine TP; 4-methylcytidine; 5-aza-cytidine; Pseudo- iso-cytidine; pyrrolo-cytidine; a-thio-cytidine; 2-(thio)cytosine; 2′-amino-2′-deoxy-CTP; 2′-azido-2′- deoxy-CTP; 2′-deoxy-2′-a-aminocytidine TP; 2′-deoxy-2′-a-azidocytidine TP; 3 (deaza) 5 (aza)cytosine; 3 (methyl)cytosine; 3-(alkyl)cytosine; 3 -(deaza) 5 (aza)cytosine; 3- (methyl)cytidine; 4,2′-O-dimethylcytidine; 5 (halo)cytosine; 5 (methyl)cytosine; 5 (propynyl)cytosine; 5 (trifluoromethyl)cytosine; 5-chlorocytosine; 5-fluorocytosine; 5- bromocytosine; 5-hydroxycytosine; 5-methylcytosine; 5-(alkyl)cytosine; 5-(alkenyl)cytosine; 5- (alkynyl)cytosine; 5-(halo)cytosine; 5-(propynyl)cytosine; 5-(trifluoromethyl)cytosine; 5-bromo- cytidine; 5-iodo-cytidine; 5-propynyl cytosine; 6-(azo)cytosine; 6-aza-cytidine; aza cytosine; deaza cytosine; N4 (acetyl)cytosine; 1-methyl-1-deaza-pseudoisocytidine; 1-methyl- pseudoisocytidine; 2-methoxy-5-methyl-cytidine; 2-methoxy-cytidine; 2-thio-5-methyl-cytidine; 4- methoxy-1-methyl-pseudoisocytidine; 4-methoxy-pseudoisocytidine; 4-thio-1-methyl-1-deaza- pseudoisocytidine; 4-thio-1-methyl-pseudoisocytidine; 4-thio-pseudoisocytidine; 5-aza- zebularine; 5-methyl-zebularine; pyrrolo-pseudoisocytidine; Zebularine; (E)-5-(2-Bromo- vinyl)cytidine TP; 2,2′-anhydro-cytidine TP hydrochloride; 2′fluor-N4-Bz-cytidine TP; 2′fluoro-N4- Acetyl-cytidine TP; 2′-O-methyl-N4-Acetyl-cytidine TP; 2′O-methyl-N4-Bz-cytidine TP; 2′-a- ethynylcytidine TP; 2′-a-trifluoromethylcytidine TP; 2′-b-Ethynylcytidine TP; 2′-b- Trifluoromethylcytidine TP; 2′-deoxy-2′,2′-difluorocytidine TP; 2′-deoxy-2′-a-mercaptocytidine TP; 2′-deoxy-2′-a-thiomethoxycytidine TP; 2′-deoxy-2′-b-aminocytidine TP; 2′-deoxy-2′-b- azidocytidine TP; 2′-deoxy-2′-b-bromocytidine TP; 2′-deoxy-2′-b-chlorocytidine TP; 2′-deoxy-2′-b- fluorocytidine TP; 2′-deoxy-2′-b-iodocytidine TP; 2′-deoxy-2′-b-mercaptocytidine TP; 2′-deoxy-2′- b-thiomethoxycytidine TP; 2′-O-methyl-5-(1-propynyl)cytidine TP; 3′-ethynylcytidine TP; 4′- azidocytidine TP; 4′-carbocyclic cytidine TP; 4′-ethynyl cytidine TP; 5-(1-propynyl)ara-cytidine TP; 5-(2-chloro-phenyl)-2-thiocytidine TP; 5-(4-Amino-phenyl)-2-thiocytidine TP; 5-Aminoallyl-CTP; 5-cyanocytidine TP; 5-ethynylara-cytidine TP; 5-Ethynylcytidine TP; 5′-Homo-cytidine TP; 5- methoxycytidine TP; 5-Trifluoromethyl-Cytidine TP; N4-Amino-cytidine TP; N4-Benzoyl-cytidine TP; Pseudoisocytidine; mimG (methylguanosine); m7G (7-methylguanosine); m2Gm (N2,2′-O- dimethylguanosine); m2G (N2-methylguanosine); imG (Wyosine); m1Gm (1,2′-O- dimethylguanosine); m1G (1-methylguanosine); 2′-O-methylguanosine; 2′-O-ribosylguanosine (phosphate); Gm (2′-O-methylguanosine); Gr(p) (2′-O-ribosyl guanosine (phosphate)); preQi (7- aminomethyl-7-deazaguanosine); preQo (7-cyano-7-deazaguanosine); G* (Archaeosine); methylwyosine; m2′7G (N2,7-dimethylguanosine); m22Gm (N2,N2,2′-O-trimethylguanosine); m2′2′7G (N2,N2,7-trimethylguanosine); m22G (N2,N2-dimethylguanosine); N2,7,2′-O- trimethylguanosine; 6-thio-guanosine; 7-deaza-guanosine; 8-oxo-guanosine; N1-methyl- guanosine; a-thio-guanosine; 2 (propyl)guanine; 2-(alkyl)guanine; 2′-amino-2′-deoxy-GTP; 2′- azido-2′-deoxy-GTP; 2′-deoxy-2′-a-aminoguanosine TP; 2′-deoxy-2′-a-azidoguanosine TP; N2- dimethylguanine; 6-(methyl)guanine; 6-(alkyl)guanine; 6-(methyl)guanine; 6-methyl-guanosine; 6-thioguanine; 7 (alkyl)guanine; 7-deaza-7-substituted guanine; 7-deaza-7-(C2- c6)alkynylguanine; 7-deaza-8-substituted guanine; 7 (methyl)guanine; 7-(alkyl)guanine; 7- (deaza)guanine; 7-(methyl)guanine; 8-azaguanine; 8-hydroxyguanine; 8-oxoguanine; 8 (alkyl)guanine; 8 (alkynyl)guanine; 8 (halo)guanine; 8 (thioalkyl)guanine; 8-(alkenyl)guanine; 8- (alkyl)guanine; 8-(alkynyl)guanine; 8-(amino)guanine; 8-(halo)guanine; 8-(hydroxyl)guanine; 8- (thioalkyl)guanine; 8-(thiol)guanine; aza guanine; deaza guanine; N (methyl)guanine; N- (methyl)guanine; 1-methyl-6-thio-guanosine; 6-methoxy-guanosine; 6-thio-7-deaza-8-aza- guanosine; 6-thio-7-deaza-guanosine; 6-thio-7-methyl-guanosine; 7-deaza-8-aza-guanosine; 7- methyl-8-oxo-guanosine; N2,N2-dimethyl-6-thio-guanosine; N2-methyl-6-thio-guanosine; 1-me- GTP; 2′fluoro-N2-isobutyl-guanosine TP; 2′0-methyl-N2-isobutyl-guanosine TP; 2′-a- ethynylguanosine TP; 2′-a-trifluoromethylguanosine TP; 2′-b-ethynylguanosine TP; 2′-b- trifluoromethylguanosine TP; 2′-deoxy-2′,2′-difluoroguanosine TP; 2′-deoxy-2′-a- mercaptoguanosine TP; 2′-deoxy-2′-a-thiomethoxyguanosine TP; 2′-deoxy-2′-b-aminoguanosine TP; 2′-deoxy-2′-b-azidoguanosine TP; 2′-deoxy-2′-b-bromoguanosine TP; 2′-deoxy-2′-b- chloroguanosine TP; 2′-deoxy-2′-b-fluoroguanosine TP; 2′-deoxy-2′-b-iodoguanosine TP; 2′- deoxy-2′-b-mercaptoguanosine TP; 2′-deoxy-2′-b-thiomethoxyguanosine TP; 4′-Azidoguanosine TP; 4′-Carbocyclic guanosine TP; 4′-Ethynylguanosine TP; 5′-Homo-guanosine TP; 8-bromo- guanosine TP; 9-deazaguanosine TP; N2-isobutyl-guanosine TP; miI (1-methylinosine); I (Inosine); m′lm (1,2′-O-dimethylinosine); 2′-O-methylinosine; 7-methylinosine; Tm (2′-O- methylinosine); oQ (Epoxyqueuosine); galQ (galactosyl-queuosine); manQ (mannosylqueuosine); Q (Queuosine); allyamino-thymidine; aza thymidine; deaza thymidine; deoxy-thymidine; Um (2′-O-methyluridine); s2U (2-thiouridine); m3U (3-methyluridine); cm5U (5- carboxymethyluridine); ho5U (5-hydroxyuridine); m5U (5-methyluridine); tm5s2U (5- taurinomethyl-2-thiouridine); 5-taurinomethyluridine; D (dihydrouridine); pseudouridine; acp3U (3-(3-amino-3-carboxypropyl)uridine); 1-methyl-3-(3-amino-5-carboxypropyl)pseudouridine; 1- methylpseudouridine; 1-ethyl-pseudouridine; 2′-O-methyluridine; 2′-O-methylpseudouridine; 2′- O-methyluridine; s2Um (2-thio-2′-O-methyluridine); 3-(3-amino-3-carboxypropyl)uridine; m3Um (3,2′-O-dimethyluridine); 3-methyl-pseudo-Uridine TP; s4U (4-thiouridine); chm5U (5- (carboxyhydroxymethyl)uridine); mchm5U (5-(carboxyhydroxymethyl)uridine methyl ester); m5Um (5,2′-O-dimethyluridine); 5,6-dihydro-uridine; nm5s2U (5-aminomethyl-2-thiouridine); ncm5Um (5-carbamoylmethyl-2′-O-methyluridine); ncm5U (5-carbamoylmethyluridine); 5- carboxyhydroxymethyluridine; 5-carboxyhydroxymethyluridine methyl ester; cnmm5Um (5- carboxymethylaminomethyl-2′-O-methyluridine); cmnm5s2U (5-carboxymethylaminomethyl-2- thiouridine); 5-carboxymethylaminomethyluridine; cmnm5U (5- carboxymethylaminomethyluridine); 5-Carbamoylmethyluridine TP; mcm5Um (5- methoxycarbonylmethyl-2′-O-methyluridine); mcm5s2U (5-methoxycarbonylmethyl-2- thiouridine); mcm5U (5-methoxycarbonylmethyluridine); mo5U (5-methoxyuridine); m5s2U (5- methyl-2-thiouridine); mnm5se2U (5-methylaminomethyl-2-selenouridine); mnm5s2U (5- methylaminomethyl-2-thiouridine); mnm5U (5-methylaminomethyluridine); m5D (5- methyldihydrouridine); 5-Oxyacetic acid-Uridine TP; 5-Oxyacetic acid-methyl ester-Uridine TP; dihydrouracil; pseudouracil; N1-methyl-pseudo-uracil; N1-ethyl-pseudo-uracil; cmo5U (uridine 5- oxyacetic acid); mcmo5U (uridine 5-oxyacetic acid methyl ester); 3-(3-Amino-3-carboxypropyl)- Uridine TP; 5-(iso-pentenylaminomethyl)-2-thiouridine TP; 5-(iso-pentenylaminomethyl)-2′-O- methyluridine TP; 5-(iso-pentenylaminomethyl)uridine TP; 5-propynyl uracil; a-thio-uridine; 1 (aminoalkylamino-carbonylethylenyl)-2(thio)-pseudouracil; 1 (aminoalkylamino- carbonylethylenyl)-2,4-(dithio)pseudouracil; 1 (aminoalkylamino-carbonylethylenyl)-4 (thio)pseudouracil; 1 (aminoalkylamino-carbonylethylenyl)-pseudouracil; 1 (aminocarbonylethylenyl)-2(thio)-pseudouracil; 1 (aminocarbonylethylenyl)-2,4- (dithio)pseudouracil; 1 (aminocarbonylethylenyl)-4 (thio)pseudouracil; 1 (aminocarbonylethylenyl)-pseudouracil; 1 substituted 2(thio)-pseudouracil; 1 substituted 2,4- (dithio)pseudouracil; 1 substituted 4 (thio)pseudouracil; 1 substituted pseudouracil; 1- (aminoalkylamino-carbonylethylenyl)-2-(thio)-pseudouracil; 1-methyl-3-(3-amino-3- carboxypropyl) pseudouridine TP; 1-methyl-3-(3-amino-3-carboxypropyl)pseudo-UTP; 1-methyl- pseudo-UTP; 1-ethyl-pseudo-UTP; 2 (thio)pseudouracil; 2′ deoxy uridine; 2′ fluorouridine; 2- (thio)uracil; 2,4-(dithio)pseudouracil; 2′methyl, 2′amino, 2′azido, 2′fluoro-guanosine; 2′-amino-2′- deoxy-UTP; 2′-azido-2′-deoxy-UTP; 2′-azido-deoxyuridine TP; 2′-O-methylpseudouridine; 2′ deoxyuridine; 2′ fluorouridine; 2′-deoxy-2′-a-aminouridine TP; 2′-deoxy-2′-a-azidouridine TP; 2- methylpseudouridine; 3 (3 amino-3 carboxypropyl)uracil; 4 (thio)pseudouracil; 4- (thio)pseudouracil; 4-(thio)uracil; 4-thiouracil; 5-aminouracil; 5 (1,3-diazole-1-alkyl)uracil; 5 (2- aminopropyl)uracil; 5 (aminoalkyl)uracil; 5 (dimethylaminoalkyl)uracil; 5 (guanidiniumalkyl)uracil; 5 (methoxycarbonylmethyl)-2-(thio)uracil; 5 (methoxycarbonyl-methyl)uracil; 5 (methyl) 2 (thio)uracil; 5 (methyl) 2,4 (dithio)uracil; 5 (methyl) 4 (thio)uracil; 5 (methylaminomethyl)-2 (thio)uracil; 5 (methylaminomethyl)-2,4 (dithio)uracil; 5 (methylaminomethyl)-4 (thio)uracil; 5 (propynyl)uracil; 5 (trifluoromethyl)uracil; 5-(2-aminopropyl)uracil; 5-(alkyl)-2-(thio)pseudouracil; 5-(alkyl)-2,4 (dithio)pseudouracil; 5-(alkyl)-4 (thio)pseudouracil; 5-(alkyl)pseudouracil; 5- (alkyl)uracil; 5-(alkenyl)uracil; 5-(alkynyl)uracil; 5-(allylamino)uracil; 5-(cyanoalkyl)uracil; 5- (dialkylaminoalkyl)uracil; 5-(dimethylaminoalkyl)uracil; 5-(guanidiniumalkyl)uracil; 5-(halo)uracil; 5-(1,3-diazole-1-alkyl)uracil; 5-(methoxy)uracil; 5-(methoxycarbonylmethyl)-2-(thio)uracil; 5- (methoxycarbonyl-methyl)uracil; 5-(methyl) 2(thio)uracil; 5-(methyl) 2,4 (dithio)uracil; 5-(methyl) 4 (thio)uracil; 5-(methyl)-2-(thio)pseudouracil; 5-(methyl)-2,4 (dithio)pseudouracil; 5-(methyl)-4 (thio)pseudouracil; 5-(methyl)pseudouracil; 5-(methylaminomethyl)-2 (thio)uracil; 5- (methylaminomethyl)-2,4(dithio)uracil; 5-(methylaminomethyl)-4-(thio)uracil; 5-(propynyl)uracil; 5-(trifluoromethyl)uracil; 5-aminoallyl-uridine; 5-bromo-uridine; 5-iodo-uridine; 5-uracil; 6 (azo)uracil; 6-(azo)uracil; 6-aza-uridine; allyamino-uracil; aza uracil; deaza uracil; 5-methyluracil; 5-(hydroxymethyl)uracil; 5-chlorouracil; 5-fluorouracil; 5-bromouracil; N3 (methyl)uracil; pseudo- UTP-1-2-ethanoic acid; Pseudouracil; 4-Thio-pseudo-UTP; 1-carboxymethyl-pseudouridine; 1- methyl-1-deaza-pseudouridine; 1-propynyl-uridine; 1-taurinomethyl-1-methyl-uridine; 1- taurinomethyl-4-thio-uridine; 1-taurinomethyl-pseudouridine; 2-methoxy-4-thio-pseudouridine; 2- thio-1-methyl-1-deaza-pseudouridine; 2-thio-1-methyl-pseudouridine; 2-thio-5-aza-uridine; 2- thio-dihydropseudouridine; 2-thio-dihydrouridine; 2-thio-pseudouridine; 4-methoxy-2-thio- pseudouridine; 4-methoxy-pseudouridine; 4-thio-1-methyl-pseudouridine; 4-thio-pseudouridine; 5-aza-uridine; Dihydropseudouridine; (±)1-(2-Hydroxypropyl)pseudouridine TP; (2R)-1-(2- Hydroxypropyl)pseudouridine TP; (2S)-1-(2-Hydroxypropyl)pseudouridine TP; (E)-5-(2-Bromo- vinyl)ara-uridine TP; (E)-5-(2-Bromo-vinyl)uridine TP; (Z)-5-(2-Bromo-vinyl)ara-uridine TP; (Z)-5- (2-Bromo-vinyl)uridine TP; 1-(2,2,2-trifluoroethyl)-pseudo-UTP; 1-(2,2,3,3,3- pentafluoropropyl)pseudouridine TP; 1-(2,2-diethoxyethyl)pseudouridine TP; 1-(2,4,6- trimethylbenzyl)pseudouridine TP; 1-(2,4,6-Trimethyl-benzyl)pseudo-UTP; 1-(2,4,6-trimethyl- phenyl )pseudo-UTP; 1-(2-amino-2-carboxyethyl)pseudo-UTP; 1-(2-amino-ethyl)pseudo-UTP; 1- (2-Hydroxyethyl)pseudouridine TP; 1-(2-methoxyethyl)pseudouridine TP; 1-(3,4-Bis- trifluoromethoxybenzyl)pseudouridine TP; 1-(3,4-dimethoxybenzyl)pseudouridine TP; 1-(3- Amino-3-carboxypropyl)pseudo-UTP; 1-(3-Amino-propyl)pseudo-UTP; 1-(3-Cyclopropyl-prop-2- ynyl)pseudouridine TP; 1-(4-Amino-4-carboxybutyl)pseudo-UTP; 1-(4-Amino-benzyl)pseudo- UTP; 1-(4-Amino-butyl)pseudo-UTP; 11(4-Amino-phenyl)pseudo-UTP; 1-(4- azidobenzyl)pseudouridine TP; 1-(4-Bromobenzyl)pseudouridine TP; 1-(4- Chlorobenzyl)pseudouridine TP; 1-(4-Fluorobenzyl)pseudouridine TP; 1-(4- iodobenzyl)pseudouridine TP; 1-(4-methanesulfonylbenzyl)pseudouridine TP; 1-(4- methoxybenzyl)pseudouridine TP; 1-(4-methoxy-benzyl)pseudo-UTP; 1 -(4-methoxy- phenyl)pseudo-UTP; 1 -(4-methylbenzyl)pseudouridine TP; 1-(4-methyl-benzyl)pseudo-UTP; 1- (4-nitrobenzyl)pseudouridine TP; 1-(4-Nitro-benzyl)pseudo-UTP; 1(4-Nitro-phenyl)pseudo-UTP; 1-(4-thiomethoxybenzyl)pseudouridine TP; 1-(4-Trifluoromethoxybenzyl)pseudouridine TP; 1-(4- trifluoromethylbenzyl)pseudouridine TP; 1-(5-Amino-pentyl)pseudo-UTP; 1-(6-Amino- hexyl)pseudo-UTP; 1,6-dimethyl-pseudo-UTP; 1-[3-(2-{2-[2-(2-aminoethoxy)-ethoxy]-ethoxy}- ethoxy)-propionyl]pseudouridine TP; 1-{3-[2-(2-aminoethoxy)-ethoxy]-propionyl} pseudouridine TP; 1-acetylpseudouridine TP; 1-Alkyl-6-(1-propynyl)-pseudo-UTP; 1-Alkyl-6-(2-propynyl)- pseudo-UTP; 1-Alkyl-6-allyl-pseudo-UTP; 1-Alkyl-6-ethynyl-pseudo-UTP; 1-Alkyl-6-homoallyl- pseudo-UTP; 1-Alkyl-6-vinyl-pseudo-UTP; 1-allylpseudouridine TP; 1-Aminomethyl-pseudo- UTP; 1-benzoylpseudouridine TP; 1-benzyloxymethylpseudouridine TP; 1-benzyl-pseudo-UTP; 1-biotinyl-PEG2-pseudouridine TP; 1-biotinylpseudouridine TP; 1-butyl-pseudo-UTP; 1- cyanomethylpseudouridine TP; 1-cyclobutylmethyl-pseudo-UTP; 1-cyclobutyl-pseudo-UTP; 1- cycloheptylmethyl-pseudo-UTP; 1-cycloheptyl-pseudo-UTP; 1-cyclohexylmethyl-pseudo-UTP; 1- cyclohexyl-pseudo-UTP; 1-cyclooctylmethyl-pseudo-UTP; 1-cyclooctyl-pseudo-UTP; 1- cyclopentylmethyl-pseudo-UTP; 1-cyclopentyl-pseudo-UTP; 1-cyclopropylmethyl-pseudo-UTP; 1-cyclopropyl-pseudo-UTP; 1-Ethyl-pseudo-UTP; 1-Hexyl-pseudo-UTP; 1- homoallylpseudouridine TP; 1-hydroxymethylpseudouridine TP; 1-iso-propyl-pseudo-UTP; 1-me- 2-thio-pseudo-UTP; 1-me-4-thio-pseudo-UTP; 1-me-alpha-thio-pseudo-UTP; 1- methanesulfonylmethylpseudouridine TP; 1-methoxymethylpseudouridine TP; 1-methyl-6-(2,2,2- Trifluoroethyl)pseudo-UTP; 1-methyl-6-(4-morpholino)-pseudo-UTP; 1-methyl-6-(4- thiomorpholino)-pseudo-UTP; 1-methyl-6-(substituted phenyl)pseudo-UTP; 1-methyl-6-amino- pseudo-UTP; 1-methyl-6-azido-pseudo-UTP; 1-methyl-6-bromo-pseudo-UTP; 1-methyl-6-butyl- pseudo-UTP; 1-methyl-6-chloro-pseudo-UTP; 1-methyl-6-cyano-pseudo-UTP; 1-methyl-6- dimethylamino-pseudo-UTP; 1-methyl-6-ethoxy-pseudo-UTP; 1-methyl-6-ethylcarboxylate- pseudo-UTP; 1-methyl-6-ethyl-pseudo-UTP; 1-methyl-6-fluoro-pseudo-UTP; 1-methyl-6-formyl- pseudo-UTP; 1-methyl-6-hydroxyamino-pseudo-UTP; 1-methyl-6-hydroxy-pseudo-UTP; 1- methyl-6-iodo-pseudo-UTP; 1-methyl-6-iso-propyl-pseudo-UTP; 1-methyl-6-methoxy-pseudo- UTP; 1-methyl-6-methylamino-pseudo-UTP; 1-methyl-6-phenyl-pseudo-UTP; 1-methyl-6-propyl- pseudo-UTP; 1-methyl-6-tert-butyl-pseudo-UTP; 1-methyl-6-trifluoromethoxy-pseudo-UTP; 1- methyl-6-trifluoromethyl-pseudo-UTP; 1-morpholinomethylpseudouridine TP; 1-Pentyl-pseudo- UTP; 1-Phenyl-pseudo-UTP; 1-pivaloylpseudouridine TP; 1-propargylpseudouridine TP; 1- propyl-pseudo-UTP; 1-propynyl-pseudouridine; 1-p-tolyl-pseudo-UTP; 1-tert-butyl-pseudo-UTP; 1-thiomethoxymethylpseudouridine TP; 1-thiomorpholinomethylpseudouridine TP; 1- trifluoroacetylpseudouridine TP; 1-trifluoromethyl-pseudo-UTP; 1-vinylpseudouridine TP; 2,2′- anhydro-uridine TP; 2′-bromo-deoxyuridine TP; 2′-F-5 -methyl-2′-deoxy-UTP; 2′-OMe-5-me-UTP; 2′-OMe-pseudo-UTP; 2′-a-ethynyluridine TP; 2′-a-trifluoromethyluridine TP; 2′-b-ethynyluridine TP; 2′-b-trifluoromethyluridine TP; 2′-deoxy-2′,2′-difluorouridine TP; 2′-deoxy-2′-a- mercaptouridine TP; 2′-deoxy-2′-a-thiomethoxyuridine TP; 2′-deoxy-2′-b-aminouridine TP; 2′- deoxy-2′-b-azidouridine TP; 2′-deoxy-2′-b-bromouridine TP; 2′-deoxy-2′-b-chlorouridine TP; 2′- deoxy-2′-b-fluorouridine TP; 2′-deoxy-2′-b-iodouridine TP; 2′-deoxy-2′-b-mercaptouridine TP; 2′- deoxy-2′-b-thiomethoxyuridine TP; 2-methoxy-4-thio-uridine; 2-methoxyuridine; 2′-O-methyl-5-(1- propynyl)uridine TP; 3-Alkyl-pseudo-UTP; 4′-Azidouridine TP; 4′-Carbocyclic uridine TP; 4′- Ethynyluridine TP; 5-(1-propynyl)ara-uridine TP; 5-(2-furanyl)uridine TP; 5-cyanouridine TP; 5- dimethylaminouridine TP; 5′-homo-uridine TP; 5-iodo-2′-fluoro-deoxyuridine TP; 5- phenylethynyluridine TP; 5-trideuteromethyl-6-deuterouridine TP; 5-Trifluoromethyl-Uridine TP; 5-vinylarauridine TP; 6-(2,2,2-Trifluoroethyl)-pseudo-UTP; 6-(4-morpholino)-pseudo-UTP; 6-(4- Thiomorpholino)-pseudo-UTP; 6-(Substituted-Phenyl)-pseudo-UTP; 6-Amino-pseudo-UTP; 6- Azido-pseudo-UTP; 6-Bromo-pseudo-UTP; 6-Butyl-pseudo-UTP; 6-Chloro-pseudo-UTP; 6- Cyano-pseudo-UTP; 6-Dimethylamino-pseudo-UTP; 6-Ethoxy-pseudo-UTP; 6-Ethylcarboxylate- pseudo-UTP; 6-Ethyl-pseudo-UTP; 6-Fluoro-pseudo-UTP; 6-Formyl-pseudo-UTP; 6- Hydroxyamino-pseudo-UTP; 6-Hydroxy-pseudo-UTP; 6-Iodo-pseudo-UTP; 6-iso-Propyl-pseudo- UTP; 6-methoxy-pseudo-UTP; 6-methylamino-pseudo-UTP; 6-methyl-pseudo-UTP; 6-Phenyl- pseudo-UTP; 6-Phenyl-pseudo-UTP; 6-Propyl-pseudo-UTP; 6-tert-Butyl-pseudo-UTP; 6- Trifluoromethoxy-pseudo-UTP; 6-Trifluoromethyl-pseudo-UTP; Alpha-thio-pseudo-UTP; Pseudouridine 1-(4-methylbenzenesulfonic acid) TP; Pseudouridine 1-(4-methylbenzoic acid) TP; Pseudouridine TP 1-[3-(2-ethoxy)]propionic acid; Pseudouridine TP 1-[3-{2-(2-[2-(2-ethoxy)- ethoxy]-ethoxy)-ethoxy}]propionic acid; Pseudouridine TP 1-[3-{2-(2-[2-{2(2-ethoxy)-ethoxy}- ethoxy]-ethoxy)-ethoxy}]propionic acid; Pseudouridine TP 1-[3-{2-(2-[2-ethoxy ]-ethoxy)- ethoxy}]propionic acid; Pseudouridine TP 1-[3-{2-(2-ethoxy)-ethoxy}] propionic acid; Pseudouridine TP 1-methylphosphonic acid; Pseudouridine TP 1-methylphosphonic acid diethyl ester; Pseudo-UTP-N1-3-propionic acid; Pseudo-UTP-N1-4-butanoic acid; Pseudo-UTP-N1-5- pentanoic acid; Pseudo-UTP-N1-6-hexanoic acid; Pseudo-UTP-N1-7-heptanoic acid; Pseudo- UTP-N1-methyl-p-benzoic acid; Pseudo-UTP-N1-p-benzoic acid; yW (Wybutosine); OHyW (Hydroxywybutosine); imG2 (isowyosine); o2yW (Peroxywybutosine); OHyW* (undermodified hydroxywybutosine); imG-14 (4-demethylwyosine); 2,6-(diamino)purine; 1-(aza)-2-(thio)-3-(aza)- phenoxazin-1-yl: 1,3-(diaza)-2-(oxo)-phenthiazin-1-yl; 1,3-(diaza)-2-(oxo)-phenoxazin-1-yl; 1,3,5-(triaza)-2,6-(dioxa)-naphthalene; 2 (amino)purine; 2,4,5-(trimethyl)phenyl; 2′ methyl, 2′amino, 2′azido, 2′fluoro-cytidine; 2′ methyl, 2′amino, 2′azido, 2′fluoro-adenine; 2′methyl, 2′amino, 2′azido, 2′fluoro-uridine; 2′-amino-2′-deoxyribose; 2-amino-6-Chloro-purine; 2-aza- inosinyl; 2′-azido-2′-deoxyribose; 2′fluoro-2′-deoxyribose; 2′-fluoro-modified bases; 2′-O-methyl- ribose; 2-oxo-7-aminopyridopyrimidin-3-yl; 2-oxo-pyridopyrimidine-3-yl; 2-pyridinone; 3 nitropyrrole; 3-(methyl)-7-(propynyl)isocarbostyrilyl; 3-(methyl)isocarbostyrilyl; 4-(fluoro)-6- (methyl)benzimidazole; 4-(methyl)benzimidazole; 4-(m ethyl )indolyl; 4,6-(dimethyl)indolyl; 5 nitroindole; 5 substituted pyrimidines; 5-(methyl)isocarbostyrilyl; 5-nitroindole; 6-(aza)pyrimidine; 6-(azo)thymine; 6-(methyl)-7-(aza)indolyl; 6-chloro-purine; 6-phenyl-pyrrolo-pyrimidin-2-on-3-yl; 7-(aminoalkylhydroxy)-1-(aza)-2-(thio)-3-(aza)-phenthiazin-1-yl; 7-(aminoalkylhydroxy)-1-(aza)- 2-(thio)-3-(aza)-phenoxazin-1-yl; 7-(aminoalkylhydroxy)-1,3-(diaza)-2-(oxo)-phenoxazin-1-yl; 7- (aminoalkylhydroxy)-1,3-(diaza)-2-(oxo)-phenthiazin-1-yl; 7-(aminoalkylhydroxy)-1,3-(diaza)-2- (oxo)-phenoxazin-1-yl; 7-(aza)indolyl; 7-(guanidiniumalkylhydroxy)-1-(aza)-2-(thio)-3-(aza)- phenoxazinl-yl; 7-(guanidiniumalkylhydroxy)-1-(aza)-2-(thio)-3-(aza)-phenthiazin-1-yl; 7- (guanidiniumalkylhydroxy)-1-(aza)-2-(thio)-3-(aza)-phenoxazin-1-yl; 7- (guanidiniumalkylhydroxy)-1,3-(diaza)-2-(oxo)-phenoxazin-1-yl; 7-(guanidiniumalkyl-hydroxy)- 1,3-(diaza)-2-(oxo)-phenthiazin-1-yl; 7-(guanidiniumalkylhydroxy)-1,3-(diaza)-2-(oxo)- phenoxazin-1-yl; 7-(propynyl)isocarbostyrilyl; 7-(propynyl)isocarbostyrilyl; propynyl-7- (aza)indolyl; 7-deaza-inosinyl; 7-substituted 1-(aza)-2-(thio)-3-(aza)-phenoxazin-1-yl; 7- substituted 1,3-(diaza)-2-(oxo)-phenoxazin-1-yl; 9-(methyl)-imidizopyridinyl; aminoindolyl; Anthracenyl; bis-ortho-(aminoalkylhydroxy)-6-phenyl-pyrrolo-pyrimidin-2-on-3-yl; bis-ortho- substituted-6-phenyl-pyrrolo-pyrimidin-2-on-3-yl; difluorotolyl; Hypoxanthine; Imidizopyridinyl; Inosinyl; Isocarbostyrilyl; isoguanisine; N2-substituted purines; N6-methyl-2-amino-purine; N6- substituted purines; N-alkylated derivative; napthalenyl; nitrobenzimidazolyl; nitroimidazolyl; nitroindazolyl; nitropyrazolyl; nubularine; O6-substituted purines; O-alkylated derivative; ortho- (aminoalkylhydroxy)-6-phenyl-pyrrolo-pyrimidin-2-on-3-yl; ortho-substituted-6-phenyl-pyrrolo- pyrimidin-2-on-3-yl; oxoformycin TP; para-(aminoalkylhydroxy)-6-phenyl-pyrrolo-pyrimidin-2-on- 3-yl; para-substituted-6-phenyl-pyrrolo-pyrimidin-2-on-3-yl; pentacenyl; phenanthracenyl; phenyl; propynyl-7-(aza)indolyl; pyrenyl; pyridopyrimidin-3-yl; pyridopyrimidin-3-yl; 2-oxo-7-amino- pyridopyrimidin-3-yl; pyrrolo-pyrimidin-2-on-3-yl; pyrrolopyrimidinyl; pyrrolopyrizinyl; stilbenzyl; substituted 1,2,4-triazoles; tetracenyl; tubercidine; xanthine; xanthosine-5′-TP; 2-thio-zebularine; 5-aza-2-thio-zebularine; 7-deaza-2-amino-purine; pyridin-4-one ribonucleoside; 2-amino- riboside-TP; formycin A TP; formycin B TP; pyrrolosine TP; 2′-OH-ara-adenosine TP; 2′-OH-ara- cytidine TP; 2′-OH-ara-uridine TP; 2′-OH-ara-guanosine TP; 5-(2-carbomethoxyvinyl)uridine TP; N6-(19-Amino-pentaoxanonadecyl)adenosine TP; hydrogen (abasic residue); and 2′-O-methyl- U. In some aspects, RNA molecules include a combination of at least two (e.g., 2, 3, 4, or more) of the aforementioned modified nucleobases. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing modifications can be excluded from the RNA molecules disclosed herein. In some aspects, modified nucleobases in RNA molecules comprise pseudouridine (ψ), 2-thiouridine (s2U), 4′-thiouridine, 5-methylcytosine, 2-thio-1-methyl-1-deaza-pseudouridine, 2- thio-1-methyl-pseudouridine, 2-thio-5-aza-uridine, 2-thio-dihydropseudouridine, 2-thio- dihydrouridine, 2-thio-pseudouridine, 4-methoxy-2-thio-pseudouridine, 4-methoxy- pseudouridine, 4-thio-1-methyl-pseudouridine, 4-thio-pseudouridine, 5-aza-uridine, dihydropseudouridine, 5-methyluridine, 5-methoxyuridine, 2′-O-methyl uridine, 1-methyl- pseudouridine (m1ψ), 1-ethyl-pseudouridine (e1ψ), 5-methoxy-uridine (mo5U), 5-methyl-cytidine (m5C), a-thio-guanosine, a-thio-adenosine, 5-cyanouridine, 4′-thio uridine 7-deaza-adenine, 1- methyl-adenosine (m1A), 2-methyl-adenine (m2A), N6-methyl-adenosine (m6A), 2,6- Diaminopurine, inosine (I), 1-methyl-inosine (m1I), wyosine (imG), methylwyosine (mimG), 7- deaza-guanosine, 7-cyano-7-deaza-guanosine (preQO), 7-aminomethyl-7-deaza-guanosine (preQl), 7-methyl-guanosine (m7G), 1-methyl-guanosine (m1G), 8-oxo-guanosine, 7-methyl-8- oxo-guanosine, 2,8-dimethyladenosine, 2-geranylthiouridine, 2-lysidine, 2-selenouridine, 3-(3- amino-3-carboxypropyl)-5,6-dihydrouridine, 3-(3-amino-3-carboxypropyl)pseudouridine, 3- methylpseudouridine, 5-(carboxyhydroxymethyl)-2′-O-methyluridine methyl ester, 5- aminomethyl-2-geranylthiouridine, 5-aminomethyl-2-selenouridine, 5-aminomethyluridine, 5- carbamoylhydroxymethyluridine, 5-carbamoylmethyl-2-thiouridine, 5-carboxymethyl-2- thiouridine, 5-carboxymethylaminomethyl-2-geranylthiouridine, 5-carboxymethylaminomethyl-2- selenouridine, 5-cyanomethyluridine, 5-hydroxycytidine, 5-methylaminomethyl-2- geranylthiouridine, 7-aminocarboxypropyl-demethylwyosine, 7-aminocarboxypropylwyosine, 7- aminocarboxypropylwyosine methyl ester, 8-methyladenosine, N4,N4-dimethylcytidine, N6- formyl adenosine, N6-hydroxymethyladenosine, agmatidine, cyclic N6- threonylcarbamoyladenosine, glutamyl-queuosine, methylated undermodified hydroxywybutosine, N4,N4,2′-O-trimethylcytidine, geranylated 5-methylaminomethyl-2- thiouridine, geranylated 5-carboxymethylaminomethyl-2-thiouridine, Qbase, preQObase, preQlbase, and combinations of two or more thereof. In some aspects, the RNA molecule includes a combination of at least two (e.g., 2, 3, 4, or more) of the aforementioned modified nucleobases, including but not limited to chemical modifications. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing modified nucleobases can be excluded from the RNA molecules disclosed herein. Exemplary nucleobases and nucleosides having a modified cytosine include 5-aza- cytidine, 6-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine (m3C), N4-acetyl-cytidine (ac4C), 5-formyl-cytidine (f5C), N4-methyl-cytidine (m4C), 5-methyl-cytidine (m5C), 5-halo-cytidine (e.g., 5-iodo-cytidine), 5-hydroxymethyl-cytidine (hm5C), 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine (s2C), 2-thio-5-methyl-cytidine, 4-thio- pseudoisocytidine, 4-thio-1-methyl-pseudoisocytidine, 4-thio-1-methyl-1-deaza- pseudoisocytidine, 1-methyl-1-deaza-pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl- zebularine, 5-aza-2-thio-zebularine, 2-thio-zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl- cytidine, 4-methoxy-pseudoisocytidine, 4-methoxy-1-methyl-pseudoisocytidine, lysidine (k2C), a- thio-cytidine, 2′-O-methyl-cytidine (Cm), 5,2′-O-dimethyl-cytidine (m5Cm), N4-acetyl-2′-O-methyl- cytidine (ac4Cm), N4,2′-O-dimethyl-cytidine (m4Cm), 5-formyl-2′-O-methyl-cytidine (f5Cm), N4,N4,2′-O-trimethyl-cytidine (m42Cm), 1-thio-cytidine, 2′-F-ara-cytidine, 2′-F-cytidine, and 2′- OH-ara-cytidine. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing modified cytosines can be excluded from the RNA molecules disclosed herein. In some aspects, a modified nucleobase is a modified uridine. Exemplary nucleobases and nucleosides having a modified uracil include pseudouridine (ψ), pyridin-4-one ribonucleoside, 5-aza-uridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2-thio-uridine (s2U), 4-thio-uridine (s4U), 4- thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-uridine (ho5U), 5-aminoallyl-uridine, 5-halo- uridine (e.g., 5-iodo-uridine or 5-bromo-uridine), 5-cyanouridine, 3-methyl-uridine (m3U), 5- methoxy-uridine (mo5U), uridine 5-oxyacetic acid (cmo5U), uridine 5-oxyacetic acid methyl ester (mcmo5U), 5-carboxymethyl-uridine (cm5U), 1-carboxymethyl-pseudouridine, 5- carboxyhydroxymethyl-uridine (chm5U), 5-carboxyhydroxymethyl-uridine methyl ester (mchm5U), 5-methoxycarbonylmethyl-uridine (mcm5U), 5-methoxycarbonylmethyl-2-thio-uridine (mcm5s2U), 5-aminomethyl-2-thio-uridine (nm5s2U), 5-methylaminomethyl-uridine (mnm5U), 5- methylaminomethyl-2-thio-uridine (mnm5s2U), 5-methylaminomethyl-2-seleno-uridine (mnm5se2U), 5-carbamoylmethyl -uridine (ncm5U), 5-carboxymethylaminomethyl-uridine (cmnm5U), 5-carboxymethylaminomethyl-2-thio-uridine (cmnmVU), 5-propynyl-uridine, 1- propynyl-pseudouridine, 5-taurinomethyl-uridine (xm5U), 1-taurinomethyl-pseudouridine, 5- taurinomethyl-2-thio-uridine (xmVu), 1-taurinomethyl-4-thio-pseudouridine, 5-methyl-uridine (m5U, e.g., having the nucleobase deoxythymine), 1-methyl-pseudouridine (m1Ψ), 1-ethyl- pseudouridine (e1ψ), 5-methyl-2-thio-uridine (m5s2U), 1-methyl-4-thio-pseudouridine (m1s4Ψ), 4-thio-1-methyl-pseudouridine, 3-methyl-pseudouridine (m3Ψ), 2-thio-1-methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-deaza-pseudouridine, dihydrouridine (D), dihydropseudouridine, 5,6-dihydrouridine, 5-methyl-dihydrouridine (m5D), 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-methoxy-uridine, 2-methoxy-4-thio-uridine, 4-methoxy- pseudouridine, 4-methoxy-2-thio-pseudouridine, N1-methyl-pseudouridine, 3-(3-amino-3- carboxypropyl)uridine (acp3U), 1-methyl-3-(3-amino-3-carboxypropyl)pseudouridine (acp3 ψ), 5- (isopentenylaminomethyl)uridine (inm5U), 5-(isopentenylaminomethyl)-2-thio-uridine (inm5s2U), a-thio-uridine, 2′-O-methyl-uridine (Um), 5,2′-O-dimethyl-uridine (m5Um), 2′-O-methyl- pseudouridine (Ψm), 2-thio-2′-O-methyl-uridine (s2Um), 5-methoxycarbonylmethyl-2′-O-methyl- uridine (mcm5Um), 5-carbamoylmethyl-2′-O-methyl-uridine (ncm5Um), 5- carboxymethylaminomethyl-2′-O-methyl -uridine (cmnm5Um), 3,2′-O-dimethyl-uridine (m3Um), and 5-(isopentenylaminomethyl)-2′-O-methyl-uridine (inm5Um), 1-thio-uridine, deoxythymidine, 2′-F-ara-uridine, 2′-F-uridine, 2′-OH-ara-uridine, 5-(2-carbomethoxyvinyl) uridine, and 5-[3-(l-E- propenylamino)]uridine. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing modified uridines can be excluded from the RNA molecules disclosed herein. In some aspects of the present disclosure, modified nucleotides include any one of N1- methylpseudouridine and/or pseudouridine. In some aspects, the RNA molecule comprises nucleotides that are N1- methylpseudouridine modified. In some aspects, the RNA molecule comprises nucleotides that are pseudouridine modified. In some aspects, an RNA comprises a modified nucleoside in place of at least one uridine. In some aspects, an RNA comprises a modified nucleoside in place of each uridine. In some aspects, the RNA molecule comprises a sequence having at least one uridine replaced by N1- methylpseudouridine. In some aspects, the RNA molecule comprises a sequence having all uridines replaced by N1-methylpseudouridine. N1-methylpseudouridine is designated in sequences as “m1Ψ”. The term “uracil,” as used herein, describes one of the nucleobases that may occur in the nucleic acid of RNA. The term “uridine,” as used herein, describes one of the nucleosides that may occur in RNA. “Pseudouridine” is one example of a modified nucleoside that is an isomer of uridine, where the uracil is attached to the pentose ring via a carbon-carbon bond instead of a nitrogen-carbon glycosidic bond. Pseudouridine is designated in sequences as “Ψ”. In some aspects, the RNA molecule comprises a nucleic acid sequence having at least one uridine replaced by N1-methylpseudouridine and/or pseudouridine. In some aspects, the RNA molecule comprises a nucleic acid sequence having at least, at most, exactly, or between (inclusive or exclusive) any two of 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% of uridines replaced by N1-methylpseudouridine and/or pseudouridine. In some aspects, the RNA molecule comprises a nucleic acid sequence having all uridines replaced by N1- methylpseudouridine and/or pseudouridine. In some aspects, a modified nucleobase is a modified adenine. Exemplary nucleobases and nucleosides having a modified adenine include 2-amino-purine, 2, 6-diaminopurine, 2-amino- 6-halo-purine (e.g., 2-amino-6-chloro-purine), 6-halo-purine (e.g., 6-chloro-purine), 2-amino-6- methyl-purine, 8-azido-adenosine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-amino- purine, 7-deaza-8-aza-2-amino-purine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6- diaminopurine, 1-methyl-adenosine (m1A), 2-methyl-adenine (m2A), N6-methyl -adenosine (m6A), 2-methylthio-N6-methyl-adenosine (ms2m6A), N6-isopentenyl-adenosine (i6A), 2- methylthio-N6-isopentenyl-adenosine (ms2i6A), N6-(cis-hydroxyisopentenyl)adenosine (io6A), 2-methylthio-N6-(cis-hydroxyisopentenyl)adenosine (ms2io6A), N6-glycinylcarbamoyl- adenosine (g6A), N6-threonylcarbamoyl-adenosine (t6A), N6-methyl-N6-threonylcarbamoyl- adenosine (m6t6A), 2-methylthio-N6-threonylcarbamoyl-adenosine (ms2g6A), N6,N6-dimethyl- adenosine (m62A), N6-hydroxynorvalylcarbamoyl-adenosine (hn6A), 2-methylthio-N6- hydroxynorvalylcarbamoyl-adenosine (ms2hn6A), N6-acetyl-adenosine (ac6A), 7-methyl- adenine, 2-methylthio-adenine, 2-methoxy-adenine, a-thio-adenosine, 2′-O-methyl-adenosine (Am), N6,2′-O-dimethyl -adenosine (m6Am), N6,N6,2′-O-trimethyl-adenosine (m62Am), 1,2′-O- dimethyl-adenosine (m1Am), 2′-O-ribosyladenosine (phosphate) (Ar(p)), 2-amino-N6-methyl- purine, 1-thio-adenosine, 8-azido-adenosine, 2′-F-ara-adenosine, 2′-F-adenosine, 2′-OH-ara- adenosine, and N6-(19-amino-pentaoxanonadecyl)-adenosine. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing modified adenines can be excluded from the RNA molecules disclosed herein. In some aspects, a modified nucleobase is a modified guanine. Exemplary nucleobases and nucleosides having a modified guanine include inosine (I), 1-methyl-inosine (m1I), wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG-14), isowyosine (imG2), wybutosine (yW), peroxywybutosine (o2yW), hydroxywybutosine (OhyW), undermodified hydroxywybutosine (OhyW*), 7-deaza-guanosine, queuosine (Q), epoxyqueuosine (oQ), galactosyl-queuosine (galQ), mannosyl-queuosine (manQ), 7-cyano-7-deaza-guanosine (preQo), 7-aminomethyl-7- deaza-guanosine (preQ1), archaeosine (G+), 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6- thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine (m7G), 6-thio-7- methyl-guanosine, 7-methyl-inosine, 6-methoxy-guanosine, 1-methyl-guanosine (m1G), N2- methyl-guanosine (m2G), N2,N2-dimethyl-guanosine (m22G), N2,7-dimethyl-guanosine (m2′7G), N2, N2,7-dimethyl-guanosine (m2′2′7G), 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, 1-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, N2,N2-dimethyl-6-thio-guanosine, a- thio-guanosine, 2′-O-methyl-guanosine (Gm), N2-methyl-2′-O-methyl-guanosine (m2Gm), N2,N2-dimethyl-2′-O-methyl-guanosine (m22Gm), 1-methyl-2′-O-methyl-guanosine, N2,7- dimethyl-2′-O-methyl-guanosine (m2′7Gm), 2′-O-methyl-inosine (Im), 1,2′-O-dimethyl-inosine (m1Im), 2′-O-ribosylguanosine (phosphate) (Gr(p)), 1-thio-guanosine, O6-methyl-guanosine, 2′- F-ara-guanosine, and 2′-F-guanosine. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing modified guanines can be excluded from the RNA molecules disclosed herein. In some aspects, RNA molecules are uniformly modified (e.g., fully modified, modified throughout the entire sequence) for a particular modification. In some aspects, the RNA molecules may be partially or fully (e.g., uniformly) modified along the entire length of the molecule. For example, one or more or all or a given type of nucleotide (e.g., purine and/or pyrimidine, or any one or more or all of A, G, U, C) may be uniformly modified in a polynucleotide of the disclosure, or in a given predetermined sequence region thereof. In some aspects, all nucleotides X in a polynucleotide of the present disclosure (or in a given sequence region thereof) are modified nucleotides, wherein X may be any one of nucleotides A, G, U, C, and/or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C and/or A+G+C. For example, a polynucleotide can be uniformly modified with pseudouridine, meaning that all uridine residues in the RNA sequence are replaced with pseudouridine. Similarly, a polynucleotide can be uniformly modified for any type of nucleoside residue present in the sequence by replacement with a modified residue such as those set forth above. The modified nucleotide can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4, or more unique structures). The RNA molecules may contain from or from about 1% to 100% modified nucleotides (either in relation to overall nucleotide content, or in relation to one or more types of nucleotide, e.g., any one or more of A, G, U and/or C) (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100%) or any intervening percentage (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90%) to 100%), and from 95% to 100%). It will be understood that any remaining percentage is accounted for by the presence of unmodified A, G, U, and/or C. In some aspects, the RNA molecule may include phosphoramidate, phosphorothioate, and/or methylphosphonate linkages. In some aspects, the RNA molecules may include one or more structural and/or chemical modifications and/or alterations which impart useful properties to the polynucleotide including, in some aspects, reduced degradation in the cell or organism and/or lack of a substantial induction of the innate immune response of a cell into which the RNA molecule is introduced. As used herein, a “structural” feature or modification is one in which two or more linked nucleotides are inserted, deleted, duplicated, inverted and/or randomized in an RNA molecule without significant chemical modification to the nucleotides themselves. Because chemical bonds will necessarily be broken and reformed to affect a structural modification, structural modifications are of a chemical nature and hence are chemical modifications. However, structural modifications will result in a different sequence of nucleotides. For example, the polynucleotide “ATCG” may be chemically modified to “AT-5meC-G”. The same polynucleotide may be structurally modified from “ATCG” to “ATCCCG”. Here, the dinucleotide “CC” has been inserted, resulting in a structural modification to the polynucleotide. In some aspects, a modified RNA molecule, introduced to a cell or organism, exhibits reduced degradation in the cell or organism, respectively, relative to an unmodified nucleic acid comprising standard nucleotides and nucleosides. In some aspects, a modified RNA molecule, introduced into a cell or organism, may exhibit reduced immunogenicity in the cell or organism, respectively (e.g., a reduced innate response) relative to an unmodified nucleic acid comprising standard nucleotides and nucleosides. In some aspects, the RNA molecule may include one or more modified nucleotides in addition to any 5′ cap structure. In some aspects, the RNA molecule does not include modified nucleotides, e.g., does not include modified nucleobases, and all of the nucleotides in the RNA molecule are conventional standard ribonucleotides A, U, G and C, with the exception of an optional 5′ cap that may include, for example, 7-methylguanosine, which is further described below. In some aspects, the RNA may include a 5′ cap comprising a 7’-methylguanosine, and the first 1, 2, or 35′ ribonucleotides may be methylated at the 2’ position of the ribose. B. 5′ CAP In some aspects, the RNA molecule described herein includes a 5′ cap which generally “caps” the 5′ end of the RNA and stabilizes the RNA molecule. In some aspects, the 5′ cap moiety is a natural 5′ cap. A “natural 5′ cap” is defined as a cap that includes 7-methylguanosine connected to the 5′ end of an mRNA molecule through a 5′ to 5′ triphosphate linkage. In some aspects, a guanosine nucleoside included in a 5′ cap may be modified, for example, by methylation at one or more positions (e.g., at the 7-position) on a base (guanine), and/or by methylation at one or more positions of a ribose. In some aspects, a guanosine nucleoside included in a 5′ cap comprises a 3′O methylation at a ribose (3′OMeG). In some aspects, a guanosine nucleoside included in a 5′ cap comprises methylation at the 7- position of guanine (m7G). In some aspects, a guanosine nucleoside included in a 5′ cap comprises methylation at the 7-position of guanine and a 3′O methylation at a ribose (m7(3′OMeG)). The 5′ cap may be incorporated during RNA synthesis (e.g., co-transcriptional capping) or may be enzymatically engineered after RNA transcription (e.g., post-transcriptional capping). In some aspects, co-transcriptional capping with a cap disclosed herein improves the capping efficiency of an RNA compared to co-transcriptional capping with an appropriate reference comparator. In some aspects, improving capping efficiency may increase translation efficiency and/or translation rate of an RNA, and/or increase expression of an encoded polypeptide. In some aspects, capping is performed after purification, e.g., tangential flow filtration, of the RNA molecule. In some aspects, an RNA described herein comprises a 5′ cap or a 5′ cap analog, e.g., a Cap 0, a Cap 1 or a Cap 2. In some aspects, a provided RNA does not have uncapped 5′- triphosphates. In some aspects, the 5′ end of the RNA is capped with a modified ribonucleotide. In some aspects, the 5′ cap moiety is a 5′ cap analog. In some aspects, an RNA may be capped with a 5′ cap analog. Cap structures include, but are not limited to, 7mG(5′)ppp(5′)N1pN2p (Cap 0), 7mG(5′)ppp(5′)N1 mpNp (Cap 1), and 7mG(5′)ppp(5′)N1 mpN2 mp (Cap 2), wherein “N” is any ribonucleotide. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing cap structures can be excluded from the RNA molecules disclosed herein. In some aspects, an RNA described herein comprises a Cap 0. In some aspects, Cap 0 is a N7-methyl guanosine, and a Cap 0 structure comprises a guanosine nucleoside methylated at the 7-position of guanine (m7G). In some aspects, a Cap 0 structure is connected to an RNA via a 5′ to 5′-triphosphate linkage and is also referred to herein as m7G, m7Gppp, and/or m7G(5′)ppp(5′).·A 5′ cap may be methylated with the structure 7mG(5′)ppp(5′)N1pN2p (Cap 0) or a derivative thereof, wherein N is the terminal 5′ nucleotide of the nucleic acid carrying the 5′ cap, typically the 5′-end of an mRNA. An exemplary enzymatic reaction for capping may include use of Vaccinia Virus Capping Enzyme (VCE) that includes mRNA triphosphatase, guanylyl- transferase and guanine-7-methytransferase, which catalyzes the construction of N7- monomethylated Cap 0 structures. Cap 0 structures play an important role in maintaining the stability and translational efficacy of the RNA molecule. In the cell, the Cap 0 structure is essential for efficient translation of the mRNA that carries the cap. In some aspects, an RNA described herein comprises a Cap 1, e.g., as described herein. The 5′ cap of the RNA molecule may be further modified on the 2′O position by a 2′-O- methyltransferase, which results in the generation of a Cap 1 structure (m7Gppp [m2′-Ο] N), which may further increase translation efficacy. In some aspects, a Cap 1 structure comprises a guanosine nucleoside methylated at the 7-position of guanine (m7G) and a 2′O methylated first nucleotide in an RNA (2′OMeN1). In some aspects, a Cap 1 structure is connected to an RNA via a 5′- to 5′-triphosphate linkage and is also referred to herein as m7GpppNm, wherein Nm denotes any nucleotide with a 2′O methylation, 7mG(5′)ppp(5′)N1 mpNp, m7Gppp(2′OMeN1), and/or m7G(5′)ppp(5′)(2′OMeN1). In some aspects, N1 is chosen from A, C, G, or U. In some aspects, N1 is A. In some aspects, N1 is C. In some aspects, N1 is G. In some aspects, N1 is U. In some aspects, a m7G(5′)ppp(5′)(2′OMeN1) Cap 1 structure comprises a second nucleotide, N2, which is a cap proximal nucleotide at position 2 and is chosen from A, G, C, or U (m7G(5′)ppp(5′)(2′OMeN1)N2). In some aspects, N2 is A. In some aspects, N2 is C. In some aspects, N2 is G. In some aspects, N2 is U. In some aspects, a Cap 1 structure comprises a guanosine nucleoside methylated at the 7-position of guanine (m7G) and one or more additional modifications, e.g., methylation on a ribose, and a 2′O methylated first nucleotide in an RNA. In some aspects, a Cap 1 structure comprises a guanosine nucleoside methylated at the 7-position of guanine, a 3′O methylation at a ribose (m7(3′OMeG)), and a 2′O methylated first nucleotide in an RNA (2′OMeN1). In some aspects, a Cap 1 structure is connected to an RNA via a 5′- to 5′-triphosphate linkage and is also referred to herein as m7(3′OMeG)ppp(2′OMeN1) and/or m7(3′OMeG)(5′)ppp(5′)(2′OMeN1). In some aspects, N1 is chosen from A, C, G, or U. In some aspects, N1 is A. In some aspects, N1 is C. In some aspects, N1 is G. In some aspects, N1 is U. In some aspects, a m7(3′OMeG)(5′)ppp(5′)(2′OMeN1) Cap 1 structure comprises a second nucleotide, N2, which is a cap proximal nucleotide at position 2 and is chosen from A, G, C, or U (m7(3′OMeG)(5′)ppp(5′)(2′OmeN1)N2). In some aspects, N2 is A. In some aspects, N2 is C. In some aspects, N2 is G. In some aspects, N2 is U. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing Cap 1 structures can be excluded from the RNA molecules disclosed herein. In some aspects, a second nucleotide in a Cap 1 structure may comprise one or more modifications, e.g., methylation. In some aspects, an RNA described herein comprises a Cap 2. In some aspects, a Cap 1 structure comprising a second nucleotide comprising a 2′O methylation is a Cap 2 structure. In some embodiments, the 5’ cap is m2 7,3′-OG(5’)ppp (m1 2′-O)ApG having the structure set forth below:
Figure imgf000063_0001
. In some aspects, the RNA molecule may be enzymatically capped at the 5′ end using Vaccinia guanylyltransferase, guanosine triphosphate, and S-adenosyl-L-methionine to yield Cap 0 structure. An inverted 7-methylguanosine cap is added via a 5′ to 5′ triphosphate bridge. Alternatively, use of a 2′O-methyltransferase with Vaccinia guanylyltransferase yields the Cap 1 structure where, in addition to the Cap 0 structure, the 2′OH group is methylated on the penultimate nucleotide. S-adenosyl-L-methionine (SAM) is a cofactor utilized as a methyl transfer reagent. Non-limiting examples of 5′ cap structures are those which, among other things, have enhanced binding of cap-binding polypeptides, increased half-life, reduced susceptibility to 5′ endonucleases and/or reduced 5′ decapping, as compared to synthetic 5′ cap structures known in the art (or to a wild type, natural or physiological 5′ cap structure). For example, recombinant Vaccinia Virus Capping Enzyme and recombinant 2′ O- methyltransferase enzyme may create a canonical 5′-5′-triphosphate linkage between the 5′- terminal nucleotide of an mRNA and a guanine cap nucleotide wherein the cap guanine includes an N7 methylation and the 5′-terminal nucleotide of the mRNA includes a 2′-O-methyl. Such a structure is termed the Cap 1 structure. This cap results in a higher translational-competency and cellular stability and a reduced activation of cellular pro-inflammatory cytokines, as compared, e.g., to other 5′ cap analog structures known in the art. A cap species may include one or more modified nucleosides and/or linker moieties. For example, a cap may include a guanine nucleotide and a guanine (G) nucleotide methylated at the 7 position joined by a triphosphate linkage at their 5′ positions, e.g., m7G(5′)ppp(5′)G, commonly written as m7GpppG. A cap species may also be an anti-reverse cap analog. A non- limiting list of possible cap species includes m7GpppG, m7Gpppm7G, m73′dGpppG, m27,O3′GpppG, m27,O3′GppppG, m27,O2′GppppG, m7Gpppm7G, m73′dGpppG, m27,O3′GpppG, m27,O3′GppppG, and m27,O2′GppppG. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing cap species can be excluded from the RNA molecules disclosed herein. In some aspects, the 5′ terminal cap includes a cap analog, for example, a 5′ terminal cap may include a guanine analog. Exemplary guanine analogs include, but are not limited to, inosine, N1-methyl-guanosine, 2′-fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino- guanosine, LNA-guanosine, and 2-azido-guanosine. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing guanine analogs can be excluded from the cap structures disclosed herein. In some aspects, the capping region may include a single cap or a series of nucleotides forming the cap. In this aspect the capping region may be from 1 to 10, e.g., 2-9, 3-8, 4-7, 1-5, 5- 10, or at least 2, or 10 or fewer nucleotides in length. In this aspect, the capping region is at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides in length. In some aspects, the cap is absent. In some aspects, the first and second operational regions may range from 3 to 40, e.g., 5-30, 10-20, 15, or at least 4, or 30 or fewer nucleotides in length and may comprise, in addition to a Start and/or Stop codon, one or more signal and/or restriction sequences. In some aspects, the first and second operational regions are at least, at most, exactly, or between (inclusive or exclusive) any two of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 nucleotides in length and may comprise, in addition to a Start and/or Stop codon, one or more signal and/or restriction sequences. Further examples of 5′ cap structures include, but are not limited to, glyceryl, inverted deoxy abasic residue (moiety), 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, and/or bridging or non-bridging methylphosphonate moiety. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing 5′ cap structures can be excluded from the RNA molecules disclosed herein. In some aspects, the RNA molecule of the present disclosure comprises at least one 5′ cap structure. In some aspects, the RNA molecule of the present disclosure does not comprise a 5′ cap structure. Numerous synthetic 5' cap analogs have been developed and are known in the art to enhance mRNA stability and translatability (see, e.g., Grudzien-Nogalska, E., Kowalska, J., Su, W., Kuhn, A.N., Slepenkov, S.V., Darynkiewicz, E., Sahin, U., Jemielity, J., and Rhoads, R.E., Synthetic mRNAs with superior translation and stability properties in Synthetic Messenger RNA and Cell Metabolism Modulation in Methods in Molecular Biology 69 (Rabinovich, P.H. Ed), 2013). In one aspect, the 5′ capping structure comprises a modified 5′ Cap 1 structure (m7G+m3′-5′-ppp- 5′-Am). In one aspect, the 5′ capping structure comprises is (3′OMe)-m2 7,3′-OGppp(m1 2’-O)ApG (TriLink BioTechnologies). This molecule is identical to the natural RNA cap structure in that it starts with a guanosine methylated at N7, and is linked by a 5′ to 5′ triphosphate linkage to the first coded nucleotide of the transcribed RNA (in this case, an adenosine). This guanosine is also methylated at the 3′ hydroxyl of the ribose to mitigate possible reverse incorporation of the cap molecule. The 2’ hydroxyl of the ribose on the adenosine is methylated, conferring a Cap 1 structure. C. UNTRANSLATED REGIONS (UTRs) An untranslated region (UTR) is located on either side of a coding sequence on an mRNA molecule and is involved in many regulatory aspects of gene expression. A 5' UTR (or leader sequence) is present upstream from the initiation codon on the 5' side of the coding sequence and a 3' UTR is present immediately following the translation termination codon on the 3' side of the coding sequence. The 5' UTR contains regulatory elements which regulate protein expression and the 3' UTR contains regulatory regions that post-transcriptionally influence gene expression and affect mRNA half-life. In some aspects, the UTR increases protein synthesis. Without being bound by mechanism or theory, the UTR may increase protein synthesis by increasing the time that the mRNA remains in translating polysomes (message stability) and/or the rate at which ribosomes initiate translation on the message (message translation efficiency). Accordingly, the UTR sequence may prolong protein synthesis in a tissue-specific manner. In some aspects, the regulatory features of a UTR can be incorporated into the RNAs of the present disclosure to, among other things, enhance the stability of the molecule. The specific features can also be incorporated to ensure controlled down-regulation of the transcript in case they are misdirected to undesired organs sites It should be understood that any UTR from any gene may be incorporated into the regions of the RNAs of the present disclosure. Furthermore, multiple wild-type UTRs of any known gene may be utilized. It is also within the scope of the present disclosure to provide artificial UTRs which are not variants of wild type regions. These UTRs or portions thereof may be placed in the same orientation as in the transcript from which they were selected or may be altered in orientation and/or location. Hence a 5′ and/or 3′ UTR may be inverted, shortened, lengthened, and/or made with one or more other 5′ UTRs or 3′ UTRs. As used herein, the term “altered” as it relates to a UTR sequence, means that the UTR has been changed in some way in relation to a reference sequence. For example, 5′ UTRs and/or 3′ UTRs may be altered relative to a wild-type or native UTR by the change in orientation and/or location as taught above and/or may be altered by the inclusion of additional nucleotides, deletion of nucleotides, swapping, and/or transposition of nucleotides. Any of these changes produces an “altered” UTR (whether 5′ and/or 3′) including a variant UTR. In some embodiments, a double, triple or quadruple UTR such as a 5′ and/or 3′ UTR may be used. As used herein, a “double” UTR is one in which two copies of the same UTR are encoded either in series or substantially in series. For example, a double beta-globin 3′ UTR may be used. It is also within the scope of the present disclosure to have patterned UTRs. As used herein “patterned UTRs” are those UTRs which reflect a repeating or alternating pattern, such as AB AB AB or AABBAABBAABB or ABCABCABC or variants thereof repeated once, twice, or more than 3 times. In these patterns, each letter, A, B, or C represent a different UTR at the nucleotide level. RNAs may encode polypeptides of interest belonging to a family of proteins that are expressed in a particular cell, tissue and/or at some time during development. In some aspects, the UTRs from any of these genes may be swapped for any other UTR of the same or different family of proteins to create a new RNA molecule. As used herein, a “family of proteins” is used in the broadest sense to refer to a group of two or more polypeptides of interest which share at least one function, structure, feature, localization, origin, and/or expression pattern. In some aspects, the 5′ UTR and the 3′ UTR sequences are computationally derived. In some aspects, the 5′ UTR and the 3′ UTRs are derived from a naturally abundant mRNA in a tissue. The tissue may be, for example, liver, a stem cell and/or lymphoid tissue. The lymphoid tissue may include, for example, any one of a lymphocyte (e.g., a B-lymphocyte, a helper T- lymphocyte, a cytotoxic T-lymphocyte, a regulatory T-lymphocyte, and/or a natural killer cell), a macrophage, a monocyte, a dendritic cell, a neutrophil, an eosinophil and a reticulocyte. In some aspects, the 5′ UTR and the 3′ UTR are derived from an alphavirus. In some aspects, the 5′ UTR and the 3′ UTR are from a wild type alphavirus. In some aspects, untranslated regions may also include translation enhancer elements (TEE). As a non- limiting example, the TEE may include those described in US Application No. 20090226470, herein incorporated by reference in its entirety, and those known in the art. Those of ordinary skill in the art will understand that 5′ UTRs that are heterologous and/or synthetic may be used with any desired 3′ UTR sequence, and vice versa. For example, a heterologous 5′ UTR may be used with a synthetic and/or heterologous 3′ UTR. In some aspects, an RNA molecule disclosed herein comprises a 5′ UTR selected from Table 5. A 5′ UTR, if present, is located at the 5′ end and starts with the transcriptional start site upstream of the start codon of a protein encoding region. A 5′ UTR is downstream of the 5′ cap (if present), e.g. directly adjacent to the 5′ cap. The 5′ UTR may contain various regulatory elements, e.g., 5′ cap structure, stem-loop structure, and an internal ribosome entry site (IRES), which may play a role in the control of translation initiation. The 5′ UTR may harbor signatures like Kozak sequences, which are also involved in the process by which the ribosome initiates translation of many genes.5′ UTRs may also form secondary structures involved in elongation factor binding. In some aspects, a 5′ UTR disclosed herein comprises a cap proximal sequence, e.g., as disclosed herein. In some aspects, a cap proximal sequence comprises a sequence adjacent to a 5′ cap. In some aspects, a cap proximal sequence comprises nucleotides in positions +1, +2, +3, +4, and/or +5 of an RNA polynucleotide. In some aspects, a Cap structure comprises one or more polynucleotides of a cap proximal sequence. In some aspects, a Cap structure comprises an m7 Guanosine cap and nucleotide +1 (N1) of an RNA polynucleotide. In some aspects, a Cap structure comprises an m7 Guanosine cap and nucleotide +2 (N2) of an RNA polynucleotide. In some aspects, a Cap structure comprises an m7 Guanosine cap and nucleotides +1 and +2 (N1 and N2) of an RNA polynucleotide. Those skilled in the art, reading the present disclosure, will appreciate that, in some aspects, one or more residues of a cap proximal sequence (e.g., one or more of residues +1, +2, +3, +4, and/or +5) may be included in an RNA by virtue of having been included in a cap entity that (e.g., a Cap 1 structure, etc); alternatively, in some aspects, at least some of the residues in a cap proximal sequence may be enzymatically added (e.g., by a polymerase such as a T7 polymerase). For example, in certain exemplified aspects where a (m2 7,3′-O)Gppp(m2’-O)ApG cap is utilized, +1 and +2 residues are the (m2 7,3′-O) A and G residues of the cap, and +3, +4, and +5 residues are added by polymerase (e.g., T7 polymerase). In some aspects, a cap proximal sequence comprises N1 and/or N2 of a Cap structure, wherein N1 and N2 are any nucleotide, e.g., A, C, G or U. In some aspects, N1 is A. In some aspects, N1 is C. In some aspects, N1 is G. In some aspects, N1 is U. In some aspects, N2 is A. In some aspects, N2 is C. In some aspects,N2 is G. In some aspects, N2 is U. In some aspects, a cap proximal sequence comprisesN1 and N2 of a Cap structure and N3, N4 and N5, wherein N1 to N5 correspond to positions +1, +2, +3, +4, and/or +5 of an RNA polynucleotide. In some aspects, N1, N2, N3, N4, or N5 are any nucleotide, e.g., A, C, G or U. In some aspects, N1N2 comprises any one of the following: AA, AC, AG, AU, CA, CC, CG, CU, GA, GC, GG, GU, UA, UC, UG, or UU. In some aspects, N1N2 comprises AG and N3N4N5 comprises any one of the following: AAA, ACA, AGA, AUA, AAG, AGG, ACG, AUG, AAC, ACC, AGC, AUC, , AAU, ACU, AGU, AUU, CAA, CCA, CGA, CUA, CAG, CGG, CCG, CUG, CAC, CCC, CGC, CUC, , CAU, CCU, CGU, CUU, , GAA, GCA, GGA, GUA, , GAG, GGG, GCG, GUG, , GAC, GCC, GGC, GUC, , GAU, GCU, GGU, GUU, UAA, UCA, UGA, UUA, UAG, UGG, UCG, UUG, UAC, UCC, UGC, UUC, UAU, UCU, UGU, or UUU. In some aspects, a cap proximal sequence comprises N1 and N2 of a Cap structure, and a sequence comprising: A3A4X5 (SEQ ID NO: 44; wherein X5 is A, G, C, or U), where N1 and N2 are each independently chosen from: A, C, G, or U. In some aspects, N1 is A and N2 is G. In some aspects, X5 is chosen from A, C, G or U. In some aspects, X5 is A. In some aspects, X5 is C. In some aspects, X5 is G. In some aspects, X5 is U. In some aspects, a cap proximal sequence comprises N1 and N2 of a Cap structure, and a sequence comprising: C3A4X5 (SEQ ID NO: 45; wherein X5 is A, G, C, or U), where N1 and N2 are each independently chosen from: A, C, G, or U. In some aspects, N1 is A and N2 is G. In some aspects, X5 is chosen from A, C, G or U. In some aspects, X5 is A. In some aspects, X5 is C. In some aspects, X5 is G. In some aspects, X5 is U. In some aspects, a cap proximal sequence comprises N1 and N2 of a Cap structure, and a sequence comprising X3Y4X5 (SEQ ID NO: 46; wherein X3 or X5 are each independently chosen from A, G, C, or U; and Y4 is not C). In some aspects, N1 and N2 are each independently chosen from: A, C, G, or U. In some aspects, N1 is A and N2 is G. In some aspects, X3 and X5 is each independently chosen from A, C, G or U. In some aspects, X3 and/or X5 is A. In some aspects, X3 and/or X5 is C. In some aspects, X3 and/or X5 is G. In some aspects, X3 and/or X5 is U. In some aspects, Y4 is C. In other aspects, Y4 is not C. In some aspects, Y4 is A. In some aspects, Y4 is G. In other aspects, Y4 is not G. In some aspects, Y4 is U. In some aspects, a cap proximal sequence comprises N1 and N2 of a Cap structure, and a sequence comprising A3C4A5 (SEQ ID NO: 47). In some aspects, N1 and N2 are each independently chosen from: A, C, G, or U. In some aspects, N1 is A and N2 is G. In some aspects, a cap proximal sequence comprises N1 and N2 of a Cap structure, and a sequence comprising A3U4G5 (SEQ ID NO: 48). In some aspects, N1 and N2 are each independently chosen from: A, C, G, or U. In some aspects, N1 is A and N2 is G. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing cap proximal sequences can be excluded from the 5′ UTR of the RNA molecules disclosed herein. In some aspects of the disclosure, a 5′ UTR is a heterologous UTR, e.g., is a UTR found in nature associated with a different ORF. In another aspect, a 5′ UTR is a synthetic UTR, e.g., does not occur in nature. Synthetic UTRs include UTRs that have been mutated or synthesized to improve their properties, e.g., to increase gene expression. In some aspects, the 5′ UTR is functionally linked to the ORF, e.g., associated with the ORF such that it may exert a function, e.g., increasing, enhancing, stabilizing, and/or prolonging protein production from an RNA molecule and/or increasing protein expression and/or total protein production from an RNA molecule, compared to a reference RNA molecule comprising a reference 5′ UTR or an RNA molecule lacking a 5′ UTR. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing 5′ UTR functions can be excluded. Exemplary 5′ UTRs include 5′ UTRs set forth in Tables 4 and 5. In some aspects, the RNA molecules described herein include one or more 5’UTRs. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing 5′ UTR sequences may be excluded from the RNA molecules disclosed herein. In one aspect, a DNA encoding a 5’ UTR disclosed herein comprises a sequence having at least, at most, exactly, or between any two of 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to any nucleic acid sequence set forth in Table 4. In one aspect, the DNA encoding the 5’ UTR comprises a sequence having at least, at most, exactly, or between any two of 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to any nucleic acid sequence selected from the group consisting of SEQ ID NO: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence having at least 80% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence having at least 85% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence having at least 90% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence having at least 92% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence having at least 94% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence having at least 95% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence having at least 96% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence having at least 97% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence having at least 98% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence having at least 99% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In some aspects, the DNA encoding the 5’ UTR described herein comprises a sequence selected from the group consisting of SEQ ID NOs: 49-279 and 333. In another aspect, an RNA disclosed herein comprises a 5’ UTR having a sequence with at least, at most, exactly, or between any two of 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to any nucleic acid sequence set forth in Table 5. In one aspect, the 5’ UTR comprises a sequence having at least, at most, exactly, or between any two of 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 335-565 and 619. In some aspects, the 5’ UTR comprises a sequence having at least 80% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 335- 565 and 619. In some aspects, the 5’ UTR comprises a sequence having at least 85% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 335-565 and 619. In some aspects, the 5’ UTR comprises a sequence having at least 90% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 335-565 and 619. In some aspects, the 5’ UTR comprises a sequence having at least 92% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 335-565 and 619. In some aspects, the 5’ UTR comprises a sequence having at least 94% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 335-565 and 619. In some aspects, the 5’ UTR comprises a sequence having at least 95% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 335-565 and 619. In some aspects, the 5’ UTR described herein comprises a sequence having at least 96% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 335-565 and 619. In some aspects, the 5’ UTR comprises a sequence having at least 97% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 335-565 and 619. In some aspects, the 5’ UTR comprises a sequence having at least 98% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 335-565 and 619. In some aspects, the 5’ UTR comprises a sequence having at least 99% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 335-565 and 619. In some aspects, the 5’ UTR comprises a sequence selected from the group consisting of SEQ ID NOs: 335-565 and 619. In one aspect, the 5′ UTR comprises a sequence of any of SEQ ID NO: 49-279, 333, 335- 565 or 619, in which the 5′ cap structure is underlined. In some aspects, 1, 2, 3, or more of the foregoing 5′ UTR sequences may be excluded from the RNA molecules disclosed herein. In some aspects, an RNA disclosed herein comprises a 3′ UTR. A 3′ UTR, if present, is situated downstream of a protein coding sequence open reading frame, e.g., downstream of the termination codon of a protein-encoding region. A 3′ UTR is typically the part of an mRNA which is located between the protein coding sequence and the poly-A tail of the mRNA. Thus, in some aspects, the 3′ UTR is upstream of the poly-A sequence (if present), e.g. directly adjacent to the poly-A sequence. The 3′ UTR may be involved in regulatory processes including transcript cleavage, stability and polyadenylation, translation, and mRNA localization. Natural or wild type 3′ UTRs comprise stretches of adenosines and uridines. These AU rich signatures are particularly prevalent in genes with high rates of turnover. Based on their sequence features and functional properties, the AU rich elements (AREs) can be separated into three classes: Class I AREs contain several dispersed copies of an AUUUA motif within U-rich regions. Class II AREs possess two or more overlapping UUAUUUA(U/A)(U/A) nonamers. Class III ARES do not contain an AUUUA motif. Most proteins binding to AREs are known to destabilize the molecule. Accordingly, introduction, removal and/or modification of 3′ UTR AREs can be used to modulate the stability of nucleic acids (e.g., RNA) of the disclosure. When engineering specific nucleic acids, in some aspects, one or more copies of an ARE can be introduced to make RNAs less stable and thereby curtail translation and decrease production of the resultant protein. Likewise, in some aspects, AREs can be identified and removed and/or mutated to increase the intracellular stability and thus increase translation and production of the resultant protein. Transfection experiments can be conducted in relevant cell lines, using nucleic acids of the disclosure and protein production can be assayed at various time points post-transfection. For example, cells can be transfected with different ARE-engineering molecules and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hour, 12 hour, 24 hour, 48 hour, and 7 days post-transfection. In some aspects, a 3′ UTR may have one or more AU-rich sequences removed. Alternatively the AU-rich sequences may remain in the 3′ UTR. A 3′ UTR may also comprise elements, which are not encoded in the template, from which an RNA is transcribed, but which are added after transcription during maturation, e.g. a poly-A tail. A 3′ UTR of the mRNA is not translated into an amino acid sequence. In some aspects, an RNA disclosed herein comprises a 3′ UTR comprising an F element and/or an I element. In some aspects, a 3′ UTR or a proximal sequence thereto comprises a restriction site. In some aspects, a restriction site is a BamHI site. In some aspects, a restriction site is a Xhol site. In some aspects of the disclosure, a 3′ UTR is a heterologous UTR, e.g., is a UTR found in nature associated with a different ORF. In another aspect, a 3′ UTR is a synthetic UTR, e.g., does not occur in nature. In some aspects, the 3′ UTR is functionally linked to the ORF, e.g., associated with the ORF such that it may exert a function, e.g., increasing, enhancing, stabilizing, and/or prolonging protein production from an RNA molecule and/or increasing protein expression and/or total protein production from an RNA molecule, compared to a reference RNA molecule comprising a reference 3′ UTR or an RNA molecule lacking a 3′ UTR. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing 3′ UTR functions may be excluded. Exemplary 3′ UTRs include 3′ UTRs set forth in Tables 4 and 5. In some aspects, the RNA molecules described herein include one or more 3’UTRs. In some aspects, the sequence UUUGAAUU is used. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing 3′ UTR sequences may be excluded from the RNA molecules disclosed herein. In one aspect, a DNA encoding a 3’ UTR disclosed herein comprises a sequence having at least, at most, exactly, or between any two of 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to any nucleic acid sequence set forth in Table 4. In one aspect, the DNA encoding the 3’ UTR comprises a sequence having at least, at most, exactly, or between any two of 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to any nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence having at least 80% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence having at least 85% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence having at least 90% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence having at least 92% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence having at least 94% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence having at least 95% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence having at least 96% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence having at least 97% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence having at least 98% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence having at least 99% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In some aspects, the DNA encoding the 3’ UTR described herein comprises a sequence selected from the group consisting of SEQ ID NO: 280-332 and 334. In another aspect, an RNA disclosed herein comprises a 3’ UTR having a sequence with at least, at most, exactly, or between any two of 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to any nucleic acid sequence set forth in Table 5. In one aspect, the 3’ UTR comprises a sequence having at least, at most, exactly, or between any two of 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 566-618 and 620. In some aspects, the 3’ UTR comprises a sequence having at least 80% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 566- 618 and 620. In some aspects, the 3’ UTR comprises a sequence having at least 85% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 566-618 and 620. In some aspects, the 3’ UTR comprises a sequence having at least 90% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 566-618 and 620. In some aspects, the 3’ UTR comprises a sequence having at least 92% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 566-618 and 620. In some aspects, the 3’ UTR comprises a sequence having at least 94% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 566-618 and 620. In some aspects, the 3’ UTR comprises a sequence having at least 95% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 566-618 and 620. In some aspects, the 3’ UTR described herein comprises a sequence having at least 96% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 566-618 and 620. In some aspects, the 3’ UTR comprises a sequence having at least 97% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 566-618 and 620. In some aspects, the 3’ UTR comprises a sequence having at least 98% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 566-618 and 620. In some aspects, the 3’ UTR comprises a sequence having at least 99% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 566-618 and 620. In some aspects, the 3’ UTR comprises a sequence selected from the group consisting of SEQ ID NOs: 566-618 and 620. In some aspects, the third nucleotide of any one of SEQ ID NOs: 49-620 is replaced with an adenine (A). For example, 5UTR_14 (SEQ ID NO:49) would begin “AGA” instead of “AGG.” In some aspects, the third nucleotide of any one of SEQ ID NOs: 49-620 is replaced with a cytosine (C). For example, 5UTR_14 (SEQ ID NO:49) would begin “AGC” instead of “AGG. In some aspects, the RNA molecules described herein do not consist of SEQ ID NO: 63 and SEQ ID NO: 232. Table 4: DNA Sequences for UTRs described herein
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Table 5: RNA Sequences for UTRs described herein
Figure imgf000089_0002
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
D. OPEN READING FRAME (ORF) The 5′ and 3′ UTRs may be operably linked to an open reading frame (ORF), which may be a sequence of codons that is capable of being translated into a polypeptide of interest. An open reading frame may be a sequence of several DNA or RNA nucleotide triplets, which may be translated into a peptide or protein. An ORF may begin with a start codon, e.g., a combination of three subsequent nucleotides coding usually for the amino acid methionine (ATG or AUG), at its 5’ end and a subsequent region, which usually exhibits a length which is a multiple of 3 nucleotides. An open reading frame may terminate with at least one stop codon, including but not limited to TAA, TAG, TGA or UAA, UAG or UGA, or any combination thereof. In some aspects, an open reading frame may terminate with one, two, three, four or more stop codons, including but not limited to TAATAA (SEQ ID NO: 25), TAATAG (SEQ ID NO: 26), TAATGA (SEQ ID NO: 27), TAGTGA (SEQ ID NO: 28), TAGTAA (SEQ ID NO: 29), TAGTAG (SEQ ID NO: 30), TGATGA (SEQ ID NO: 31), TGATAG (SEQ ID NO: 32), TGATAA (SEQ ID NO: 33) or UAAUAA (SEQ ID NO: 34), UAAUAG (SEQ ID NO: 35), UAAUGA (SEQ ID NO: 36), UAGUGA (SEQ ID NO: 37), UAGUAA (SEQ ID NO:38), UAGUAG (SEQ ID NO: 39), UGAUGA (SEQ ID NO: 40), UGAUAG (SEQ ID NO: 41), UGAUAA (SEQ ID NO: 42), or any combination thereof. An open reading frame may be isolated or it may be incorporated in a longer nucleic acid sequence, e.g. in a vector or an mRNA. An open reading frame may also be termed “(protein) coding region” or “coding sequence”. As stated herein, the RNA molecule may include one (monocistronic), two (bicistronic) or more (multicistronic) open reading frames. In some aspects, the ORF encodes a non-structural viral gene. In some aspects, the ORF further includes one or more subgenomic promoters. In some aspects, the RNA molecule includes a subgenomic promoter operably linked to the ORF. In some aspects, a first RNA molecule does not include an ORF encoding any polypeptide of interest, whereas a second RNA molecule includes an ORF encoding a polypeptide of interest. In some aspects, the first RNA molecule does not include a subgenomic promoter. The present disclosure provides for an RNA molecule comprising at least one open reading frame encoding a respiratory syncytial virus (RSV) polypeptide. In some aspects, an RNA molecule comprising at least one open reading frame encoding a RSV F protein. In a preferred aspect, an RNA molecule comprising at least one open reading frame encoding a respiratory syncytial virus (RSV) prefusion F protein (preF) polypeptide. E. GENES OF INTEREST The RNA molecules described herein may include a gene of interest. The gene of interest encodes a polypeptide of interest. Non-limiting examples of polypeptides of interest include, e.g., biologics, antibodies, vaccines, therapeutic polypeptides or peptides, cell penetrating peptides, secreted polypeptides, plasma membrane polypeptides, cytoplasmic or cytoskeletal polypeptides, intracellular membrane bound polypeptides, nuclear polypeptides, polypeptides associated with human disease, targeting moieties, those polypeptides encoded by the human genome for which no therapeutic indication has been identified but which nonetheless have utility in areas of research and discovery, or combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing polypeptides of interest may be excluded. The sequence for a particular gene of interest is readily identified by one of skill in the art using public and private databases, e.g., GENBANK®. In some aspects, the RNA molecules include a coding region for a gene of interest. In some aspects, a gene of interest is or comprises an antigenic polypeptide or an immunogenic variant or an immunogenic fragment thereof. In some aspects, an antigenic polypeptide comprises one epitope from an antigen. In some aspects, an antigenic polypeptide comprises a plurality of distinct epitopes from an antigen. In some aspects, an antigenic polypeptide comprising a plurality of distinct epitopes from an antigen is polyepitopic. In some aspects, an antigenic polypeptide comprises: an antigenic polypeptide from an allergen, a viral antigenic polypeptide, a bacterial antigenic polypeptide, a fungal antigenic polypeptide, a parasitic antigenic polypeptide, an antigenic polypeptide from an infectious agent, an antigenic polypeptide from a pathogen, a tumor antigenic polypeptide, or a self-antigenic polypeptide. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing antigenic polypeptides may be excluded. The term “antigen” may refer to a substance, which is capable of being recognized by the immune system, e.g., by the adaptive immune system, and which is capable of eliciting an antigen-specific immune response, e.g., by formation of antibodies and/or antigen-specific T cells as part of an adaptive immune response. An antigen may be or may comprise a peptide or protein, which may be presented by the MHC to T cells. An antigen may be the product of translation of a provided nucleic acid molecule, e.g., an RNA molecule comprising at least one coding sequence as described herein. In addition, fragments, variants and derivatives of an antigen, such as a peptide or a protein, comprising at least one epitope are understood as antigens. In some aspects, an RNA encoding a gene of interest, e.g., an antigen, is expressed in cells of a subject treated to provide a gene of interest, e.g., an antigen. In some aspects, the RNA is transiently expressed in cells of the subject. In some aspects, expression of a gene of interest, e.g., an antigen, is at the cell surface. In some aspects, a gene of interest, e.g., an antigen, is expressed and presented in the context of MHC. In some aspects, expression of a gene of interest, e.g., an antigen, is into the extracellular space, e.g., the antigen is secreted. In some aspects, the RNA molecules include a coding region for a gene of interest, e.g., an antigen. In some aspects, the RNA molecules include a coding region for a gene of interest, e.g., an antigen, that is derived from a pathogen associated with an infectious disease. In some aspects, the RNA molecules include a coding region for a gene of interest, e.g., an antigen, that is derived from RSV. In some aspects, the RNA molecule encodes a RSV preF protein or a fragment or a variant thereof. In some aspects, an RNA polynucleotide described herein or a composition or medical preparation comprising the same comprises a nucleotide sequence disclosed herein. In some aspects, an RNA polynucleotide comprises a sequence having at least 80% identity to a nucleotide sequence disclosed herein. In some aspects, an RNA polynucleotide comprises a sequence encoding a polypeptide having at least 80% identity to a polypeptide sequence disclosed herein. In some aspects, an RNA polynucleotide described herein or a composition or medical preparation comprising the same is transcribed by a DNA template. In some aspects, a DNA template used to transcribe an RNA polynucleotide described herein comprises a sequence complementary to an RNA polynucleotide. In some aspects, a gene of interest described herein is encoded by an RNA polynucleotide described herein comprising a nucleotide sequence disclosed herein. In some aspects, an RNA polynucleotide encodes a polypeptide having at least 80% identity to a polypeptide sequence disclosed herein. In some aspects, a polypeptide described herein is encoded by an RNA polynucleotide transcribed by a DNA template comprising a sequence complementary to an RNA polynucleotide. In some aspects, the RNA molecule encodes a RSV preF protein comprising the sequence of any one of SEQ ID NOs: 1-6 or 621-626, or a fragment or variant thereof. In a preferred aspect, the RNA molecule encodes a RSV preF protein comprising the sequence of any one of SEQ ID NOs: 4 or 6, or a fragment or variant thereof. In some aspects, the RNA molecule encodes a RSV preF protein wherein the RNA molecule is synthesized from the nucleic acid having the sequence comprising any one of SEQ ID NOs: 7-10, 627-632, 639 or 640, or fragment or variant thereof. In a preferred aspect, the RNA molecule encodes a RSV preF protein wherein the RNA molecule is synthesized from the nucleic acid having the sequence of any one of SEQ ID NOs: 8, 10-14, 639 or 640, or a fragment or variant thereof. In further aspects, the RNA molecule encodes a RSV preF protein wherein the RNA molecule comprises the nucleic acid sequence set forth in any one of SEQ ID NOs: 15-20, 633- 638, 641 or 642, or fragment or variant thereof. In a preferred aspect, the RNA molecule encodes a RSV preF protein wherein the RNA molecule comprises the nucleic acid sequence set forth in any one of SEQ ID NOs: 15-20, 641 or 642, or a fragment or variant thereof. F. POLY-A TAIL In some aspects, RNA molecules disclosed herein comprise a poly-adenylate (poly-A) sequence, e.g., as described herein. In some aspects, a poly-A sequence is situated downstream of a 3′ UTR, e.g., adjacent to a 3′ UTR. A “poly-A tail” or “poly-A sequence” refers to a stretch of consecutive adenine residues, e.g., of up to or up to about 400 adenosine nucleotides, e.g., from or from about 20 to about 400, preferably from or from about 50 to about 400, more preferably from or from about 50 to about 300, even more preferably from or from about 50 to about 250, most preferably from or from about 60 to about 250 adenosine nucleotides, which may be attached to the 3′ end of the RNA molecule. Poly-A sequences are known to those of skill in the art and may follow the 3′ UTR in the RNA molecules described herein. The poly-A tail may increase the stability, half-life, and/or translational efficiency of the RNA molecule. After cleavage, most pre-mRNAs, with exceptions that include replication-dependent histone transcripts that terminate with a histone stem-loop instead of a poly-A sequence, acquire a polyadenylated tail. In this context, 3′-end processing is a nuclear co-transcriptional process that promotes transport of mRNAs from the nucleus to the cytoplasm and affects the stability and the translation of mRNAs. Formation of this 3′ end occurs in a two-step reaction directed by the cleavage/polyadenylation machinery and depends on the presence of two sequence elements in mRNA precursors (pre-mRNAs); a hexanucleotide polyadenylation signal and a downstream G/U-rich sequence. In a first step, pre-mRNAs are cleaved between these two elements to a free 3′ hydroxyl. In a second step, the newly formed 3′ end is extended by polyadenylation or addition of a poly-A sequence. Polyadenylation refers to the addition of a poly-A sequence to an RNA molecule, e.g., to a premature mRNA. Polyadenylation may be induced by a so-called polyadenylation signal. This signal may be located within a stretch of nucleotides close to or at the 3′-end of an RNA molecule to be polyadenylated. A polyadenylation signal may also be comprised by the 3′ UTR of the artificial nucleic acid molecule. A polyadenylation signal typically comprises a hexamer consisting of adenine and uracil/thymine nucleotides, preferably the hexamer sequence AAUAAA, though other sequences, preferably hexamer sequences, are also conceivable. Polyadenylation typically occurs during processing of a pre-mRNA (also called premature-mRNA). Typically, RNA maturation (from pre-mRNA to mature mRNA) comprises the step of polyadenylation. Poly-A tailing of in vitro transcribed mRNA can be achieved using various approaches including, but not limited to, cloning of a poly-T tract into the DNA template or by post-transcriptional addition using poly-A polymerase. The term may relate to polyadenylation of RNA as a cellular process or to polyadenylation carried out by enzymatic reaction in vitro with a suitable enzyme, such as E. coli poly-A polymerase, or by chemical synthesis. RNA molecules disclosed herein may have a poly-A sequence attached to the free 3′-end of the RNA by a template-independent RNA polymerase after transcription or a poly-A sequence encoded by DNA and transcribed by a template-dependent RNA polymerase. In some aspects, a poly-A sequence is attached during RNA transcription, e.g., during preparation of in vitro transcribed RNA, based on a DNA template comprising repeated dT nucleotides (deoxythymidylate) in the strand complementary to the coding strand. The DNA sequence encoding a poly-A sequence (coding strand) is referred to as poly-A cassette. In some aspects, the poly-A cassette present in the coding strand of DNA essentially consists of dA nucleotides, but is interrupted by a random sequence of the four nucleotides (dA, dC, dG, and dT). Such a random sequence may be at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleotides in length. Such a cassette is disclosed in, e.g., WO 2016/005324 A1, hereby incorporated by reference. Any poly-A cassette disclosed in WO 2016/005324 A1 may be used in the present disclosure. A poly-A cassette that essentially consists of dA nucleotides, but is interrupted by a random sequence having an equal distribution of the four nucleotides (dA, dC, dG, dT) and having a length of e.g., 5 to 50 nucleotides, shows, on a DNA level, constant propagation of plasmid DNA in E. coli and is still associated, on an RNA level, with the beneficial properties with respect to supporting RNA stability and translational efficiency. In some aspects, the poly-A sequence contained in an RNA polynucleotide described herein consists essentially of adenosine nucleotides, but is interrupted by a random sequence of the four nucleotides (A, C, G, U). Such a random sequence may be at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleotides in length. The poly-A sequence may be located at any position within the 3′ UTR. In some aspects, no nucleotides other than adenosine nucleotides flank a poly-A sequence at its 3′-end, e.g., the poly-A sequence is not masked or followed at its 3′-end by a nucleotide other than adenosine. In some aspects, the poly-A sequence may be located at the 3′ terminus of the 3′ UTR, e.g., the 3′ UTR does not contain more than 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 nucleotides located 3′ of the poly-A sequence; more preferably the 3′ UTR does not contain further elements located 3′ to the poly-A sequence. In some aspects, poly-A sequence is located at the 3′ terminus of the RNA molecule, e.g., the artificial nucleic acid molecule does not contain more than 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 nucleotides located 3′ of the poly-A sequence. Alternatively, the poly-A sequence may be located at the 5′ terminus of the 3′ UTR, e.g., immediately 3′ of the ORF of the artificial nucleic acid molecule, or located within the 3′ UTR, e.g., flanked on the 5′ and on the 3′ side by other 3′ UTR elements. In some aspects, the poly-A sequence is flanked on the 3′ side by a poly- C sequence and/or a histone stem-loop sequence. In addition or alternatively, the poly-A sequence can be flanked on the 5′ side by a 3′ UTR element derived from, e.g., a human albumin or globin gene. In some aspects, the RNA molecule may further include an endonuclease recognition site sequence immediately downstream of the poly-A tail sequence. The RNA molecule may further include a poly-A polymerase recognition sequence (e.g., a polyadenylation signal) (e.g., AAUAAA) near its 3′ end. In some aspects, the polyadenylation signal is located 3′ of the poly-A sequence comprised in the 3′ UTR. In some aspects, the poly-A sequence is separated from the polyadenylation signal by a nucleotide sequence comprising or consisting of at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, or 150 nucleotides, wherein the nucleotide sequence does preferably not comprise more than 10, 9, 8, 7, 6, 5, 4, 3, or 2 consecutive adenine nucleotides. In some aspects, the nucleotide sequence that separates the poly-A sequence and the polyadenylation signal comprises from or from about 1 to about 200 nucleotides, e.g., from 10 to 90, from 20 to 85, from 30 to 80, from 40 to 80, from 50 to 75 or from 55 to 85 nucleotides, more preferably from 55 to 80 nucleotides, and the nucleotide sequence does not comprise more than 10, 9, 8, 7, 6, 5, 4, 3, or 2 consecutive adenine nucleotides. In some aspects, the polyadenylation signal comprises the consensus sequence NN(U/T)ANA, with N = A or U, preferably AA(U/T)AAA or A(U/T)(U/T)AAA. Such a consensus sequence may be recognized by most animal and bacterial cell-systems, for example, by the polyadenylation-factors, such as cleavage/polyadenylation specificity factor (CPSF) cooperating with CstF, PAP, PAB2, CFI and/or CFII. In some aspects, the polyadenylation signal (e.g., the consensus sequence NNUANA) is located less than or less than about 50 nucleotides, e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides, downstream of the 3′-end of the 3′ UTR element as defined herein such that transcription of an RNA molecule will result in a premature-RNA containing the polyadenylation signal downstream of its 3′ UTR and subsequent attachment of a poly-A sequence to the premature-RNA. Accordingly, a resulting RNA may comprise a 3′ UTR, which comprises at least one poly-A sequence, and wherein the 3′ UTR is followed by an additional poly-A sequence. The poly-A sequence may be of any length. In some aspects, the poly-A tail may be 5 to 300 nucleotides in length. In some aspects, the RNA molecule includes a poly-A tail that comprises, consists essentially of, or consists of a sequence of or of about 25 to about 400 adenosine nucleotides, a sequence of or of about 50 to about 400 adenosine nucleotides, a sequence of or of about 50 to about 300 adenosine nucleotides, a sequence of or of about 50 to about 250 adenosine nucleotides, a sequence of or of about 60 to about 250 adenosine nucleotides, or a sequence of or of about 40 to about 100 adenosine nucleotides. In some aspects, the poly-A tail comprises, consists essentially of, or consists of at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 250, 255, 260, 265, 270, 275, 280, 285, 290, 295, 300, 305, 310, 315, 320, 325, 330, 335, 340, 345, 350, 355, 360, 365, 370, 375, 380, 385, 390, 395, 400, 405, 410, 415, 420, 425, 430, 435, 440, 445, 450, 455, 460, 465, 470, 475, 480, 485, 490, 495, 500, 505, 510, 515, 520, 525, 530, 535, 540, 545, 550, 555, 560, 565, 570, 575, 580, 585, 590, 595, 600, 605, 610, 615, 620, 625, 630, 635, 640, 645, 650, 655, 660, 665, 670, 675, 680, 685, 690, 695, 700, 705, 710, 715, 720, 725, 730, 735, 740, 745, 750, 755, 760, 765, 770, 775, 780, 785, 790, 795, 800, 805, 810, 815, 820, 825, 830, 835, 840, 845, 850, 855, 860, 865, 870, 875, 880, 885, 890, 895, 900, 905, 910, 915, 920, 925, 930, 935, 940, 945, 950, 955, 960, 965, 970, 975, 980, 985, 990, 995, or 1000 adenosine nucleotides. In this context, “consists essentially of” means that most nucleotides in the poly-A sequence, typically at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% by number of nucleotides in the poly-A sequence are adenosine nucleotides, but permits remaining nucleotides to be nucleotides other than adenosine nucleotides, such as uridine, guanosine, and/or cytosine. In this context, “consists of” means that all nucleotides in the poly-A sequence, e.g., 100% by number of nucleotides in the poly-A sequence, are adenosine nucleotides. In some aspects, the RNA molecule includes a poly-A tail that includes a sequence of greater than 30 adenosine nucleotides. In some aspects, the RNA molecule includes a poly-A tail that includes or includes about 40 adenosine nucleotides. In some aspects, the RNA molecule includes a poly-A tail that includes or includes about 80 adenosine nucleotides. In some aspects, the poly-A tail may have contiguous A residues or interrupted A residues comprising a linker (L). In some aspects, the 3′ poly-A tail has a stretch of at least 10 consecutive adenosine residues and at most 300 consecutive adenosine residues. In some specific aspects, the RNA molecule includes or includes about 40 consecutive adenosine residues. In some aspects, the RNA molecule includes or includes about 80 consecutive adenosine residues. Poly-A tails may play key regulatory roles in enhancing translation efficiency and regulating the efficiency of mRNA quality control and degradation. Short sequences or hyperpolyadenylation may signal for RNA degradation. In some aspects, a poly-A tail may be located within an RNA molecule or other nucleic acid molecule, such as, e.g., in a vector, for example, in a vector serving as template for the generation of an RNA, e.g., an mRNA, e.g., by transcription of the vector. In some aspects, the RNA molecule may not include a poly-A tail. In some aspects, a poly-A tail may be located within an RNA molecule or other nucleic acid molecule, such as, e.g., in a vector, for example, in a vector serving as template for the generation of an RNA, e.g. an mRNA, e.g., by transcription of the vector. In some aspects, the RNA molecule may not include a poly-A tail. In one aspect, a DNA encoding a poly-A tail disclosed herein comprises a sequence having at least, at most, exactly, or between (inclusive or exclusive) any two of 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to SEQ ID NO: 21 or 22. In one aspect, the DNA encoding the poly-A tail comprises a sequence of SEQ ID NO: 21 or 22. In one aspect, an RNA disclosed herein comprises a poly-A tail comprising a sequence having at least, at most, exactly, or between (inclusive or exclusive) any two of 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to SEQ ID NO: 21 or 22. In one aspects, the poly-A tail comprises a sequence of SEQ ID NO: 21 or 22. In one aspect, the poly-A tail comprises a sequence of SEQ ID NO: 21 or 22+/- 2 adenosine (A) nucleotides. In one aspect, the poly-A tail comprises a sequence of SEQ ID NO: 21 or 22+/- 1 adenosine (A) nucleotides. In one aspect, the poly-A tail comprises a sequence of SEQ ID NO: 21 or 22. In one aspect, the poly-A tail comprises a sequence of SEQ ID NO: 21 or 22+/- 2 adenosine (A) nucleotides. In one aspect, the poly-A tail comprises a sequence of SEQ ID NO: 21 or 22+/- 1 adenosine (A) nucleotides. In some aspects, the poly-A tail comprises a sequence of SEQ ID NO: 21 or 22. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing poly-A sequences may be excluded from the RNA molecules disclosed herein. SEQ ID NO: 21 (DNA) AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA AAAAAAAAAAAAAAAAA SEQ ID NO: 22 (RNA) AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA AAAAAAAAAAAAAAAAA SEQ ID NO: 23 (polyA linker (L); DNA) GCATATGACT SEQ ID NO: 24 (polyA linker (L); RNA) GCAUAUGACU G. OTHER ELEMENTS In some aspects of the present disclosure, the RNA molecules additionally include a chain terminating nucleoside. For example, a chain terminating nucleoside may include those nucleosides deoxygenated at the 2’ and/or 3′ positions of their sugar group. Such species may include 3′ deoxyadenosine (cordycepin), 3′ deoxyuridine, 3′ deoxycytosine, 3′ deoxyguanosine, 3′ deoxythymine, and 2',3′ dideoxynucleosides, such as 2',3′ dideoxyadenosine, 2',3′ dideoxyuridine, 2',3′ dideoxycytosine, 2',3′ dideoxyguanosine, and 2',3′ dideoxythymine. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing chain terminating nucleosides may be excluded from the RNA molecules disclosed herein. In some aspects, incorporation of a chain terminating nucleotide into an mRNA, for example at the 3′-terminus, may result in stabilization of the mRNA, as described, for example, in International Patent Publication No. WO 2013/103659. In some aspects of the present disclosure, the RNA molecules additionally include a stem loop, such as a histone stem loop. A stem loop may include 2, 3, 4, 5, 6, 7, 8, or more nucleotide base pairs. For example, a stem loop may include 4, 5, 6, 7, or 8 nucleotide base pairs. A stem loop may be located in any region of an mRNA. For example, a stem loop may be located in, before, or after an untranslated region (a 5′ UTR or a 3′ UTR), a coding region, or a poly-A sequence or tail. In some aspects, a stem loop may affect one or more function(s) of an mRNA, such as initiation of translation, translation efficiency, and/or transcriptional termination. Such histone stem-loop sequences may be histone stem-loop sequences disclosed in WO 2012/019780, the disclosure of which is incorporated herein by reference in its entirety. Other non-limiting examples of histone stem loop structures and nucleic acid sequences encoding such structures can be found in, e.g., WO 2016/091391, the disclosure of which is incorporated by reference herein in its entirety. In some aspects, the combination of a poly-A sequence or polyadenylation signal and at least one histone stem-loop, even though both represent alternative mechanisms in nature, acts synergistically to increase the protein expression beyond the level observed with either of the individual elements. In some aspects, the synergistic effect of the combination of poly-A and at least one histone stem-loop does not depend on the order of the elements and/or the length of the poly-A sequence. In some aspects, the RNA does not comprise a histone downstream element (HDE). An HDE includes a purine-rich polynucleotide stretch of approximately 15 to 20 nucleotides 3′ of naturally occurring stem-loops, representing the binding site for the U7 snRNA, which is involved in processing of histone pre-mRNA into mature histone mRNA. In some aspects, the histone stem-loop is generally derived from histone genes, and includes an intramolecular base pairing of two neighbored partially or entirely reverse complementary sequences separated by a spacer, consisting of a short sequence, which forms the loop of the structure. The unpaired loop region is typically unable to base pair with either of the stem loop elements. Stability of the stem-loop structure generally depends on the length, number of mismatches or bulges, and/or base composition of the paired region. In some aspects, wobble base pairing (non-Watson-Crick base pairing) may result. In some aspects, the at least one histone stem-loop sequence comprises a length of 15 to 45 nucleotides. In some aspects, the RNA molecules include (e.g., within the 3′ UTR) a poly(C) sequence. In some aspects, the poly-C sequences has at least, at most, exactly, or between (inclusive or exclusive) any two of 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 cytidines. In some aspects, the poly-C sequences has or has about 30 cytidines. In some aspects, the RNA molecules include an internal ribosome entry site (IRES) sequence or IRES-motif. In some aspects, an IRES sequence separates ORFs, e.g., if the RNA encodes two or more peptides or proteins. An IRES-sequence may therefore be useful if the RNA molecule is a bi- or multicistronic nucleic acid molecule. In some aspects, the RNA does not comprise an intron. In some aspects, the RNA may instead or additionally include a microRNA binding site. Representative RNA molecules including a combination of the elements disclosed herein can include, without limitation, in 5′-to-3′-direction, the following: ORF - poly-A sequence; ORF - IRES - ORF - poly-A sequence; ORF - 3′ UTR - poly-A sequence; ORF - poly-A sequence - 3′ UTR; ORF - 3′ UTR - poly-A sequence - poly(C) sequence - histone stem-loop; ORF - 3′ UTR - poly-A sequence - poly(C) sequence - poly-A sequence; ORF - 3′ UTR - poly-A sequence - histone stem-loop - poly-A sequence; 5′ UTR - ORF - 3′ UTR; 5′ UTR - ORF - poly-A sequence; 5′ UTR - ORF - poly-A sequence - poly(C) sequence - histone stem-loop; 5′ UTR - ORF - poly-A sequence - poly(C) sequence - poly-A sequence; 5′ UTR - ORF - poly-A sequence - histone stem-loop - poly-A sequence; 5′ UTR - ORF - 3′ UTR - poly-A sequence; 5′ UTR - ORF - 3′ UTR - poly-A sequence - poly(C) sequence 5′ UTR - ORF - 3′ UTR - poly-A sequence - poly(C) sequence - histone stem- loop; 5′-cap - 5′ UTR - ORF - 3′ UTR; 5′-cap - 5′ UTR - ORF - poly-A sequence; 5′-cap - 5′ UTR - ORF - 3′ UTR - poly-A sequence; 5′-cap - 5′ UTR - ORF - 3′ UTR - poly-A sequence - poly(C) sequence; or 5′-cap - 5′ UTR - ORF - 3′ UTR - poly-A sequence - poly(C) sequence - histone stem- loop. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing elements may be excluded from the RNA molecules disclosed herein. IV. RNA TRANSCRIPTION In some aspects, the RNA disclosed herein is produced by in vitro transcription or chemical synthesis. In the context of the present disclosure, the term “transcription” relates to a process, wherein the genetic code in a DNA sequence is transcribed into RNA. Subsequently, the RNA may be translated into peptide or protein. According to the present disclosure, “transcription” comprises “in vitro transcription” or “IVT,” which refers to the process whereby transcription occurs in vitro in a non-cellular system to produce a synthetic RNA product for use in various applications, including, e.g., production of protein or polypeptides. The methodology for in vitro transcription of mRNA is well known in the art. (see, e.g., Losick, R.1972. In vitro transcription, Ann Rev Biochem, 41409-46; Kamakaka, R. T. and Kraus, W. L. 2001. In vitro Transcription, Current Protocols in Cell Biology, 2:11.6:11.6.1-11.6.17; Beckert, B. And Masquida, B. 2010. Synthesis of RNA by In vitro Transcription in RNA, Methods in Molecular Biology, 703 (Neilson, H. Ed), New York, N.Y. Humana Press, 2010; Brunelle, J.L. and Green, R., 2013, Chapter Five – In vitro transcription from plasmid or PCR-amplified DNA, Methods in Enzymology 530:101-114; all of which are incorporated herein by reference). Cloning vectors may be applied for the generation of transcripts. These cloning vectors are generally designated as transcription vectors and are according to the present invention encompassed by the term “vector.” According to specific aspects, the RNA used is in vitro transcribed RNA (IVT-RNA) and may be obtained by in vitro transcription of an appropriate DNA template. Template DNA can be prepared for in vitro transcription from a number of sources with appropriate techniques which are well known in the art including, but not limited to, plasmid DNA and polymerase chain reaction amplification (see Linpinsel, J.L and Conn, G.L., General protocols for preparation of plasmid DNA template, and Bowman, J.C., Azizi, B., Lenz, T.K., Ray, P., and Williams, L.D. in RNA in vitro transcription and RNA purification by denaturing PAGE in Recombinant and in vitro RNA syntheses, Methods 941 Conn G.L. (ed), New York, N.Y. Humana Press, 2012, each incorporated herein by reference). The promoter for controlling transcription may be any promoter for any RNA polymerase. Particular examples of RNA polymerases are the T7, T3, and SP6 RNA polymerases. Preferably, the in vitro transcription according to the invention is controlled by a T7 or SP6 promoter. A DNA template for in vitro transcription may be obtained by cloning of a nucleic acid, in particular cDNA, and introducing it into an appropriate vector for in vitro transcription. The cDNA may be obtained by reverse transcription of RNA. Synthetic IVT RNA products may be translated in vitro or introduced directly into cells, where they may be translated. With respect to RNA, the term “expression” or “translation” relates to the process in the ribosomes of a cell by which a strand of mRNA directs the assembly of a sequence of amino acids to make a peptide or protein. Such synthetic RNA products include but are not limited to, e.g., mRNA molecules, saRNA molecules, antisense RNA molecules, shRNA molecules, long non-coding RNA molecules, ribozymes, aptamers, guide RNA molecules (e.g., for CRISPR), ribosomal RNA molecules, small nuclear RNA molecules, small nucleolar RNA molecules, and the like. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing synthetic RNA products may be excluded. An IVT reaction typically utilizes a DNA template (e.g., a linear DNA template) as described and/or utilized herein, ribonucleotides (e.g., non-modified ribonucleotide triphosphates or modified ribonucleotide triphosphates), and an appropriate RNA polymerase. In some aspects, an mRNA is produced by in vitro transcription using a DNA template where DNA refers to a nucleic acid that contains deoxyribonucleotides. In some aspects, an RNA disclosed herein is in vitro transcribed RNA (IVT-RNA) and may be obtained by in vitro transcription of an appropriate DNA template. The promoter for controlling transcription may be any promoter for any RNA polymerase. A DNA template for in vitro transcription may be obtained by cloning of a nucleic acid, in particular cDNA, and introducing it into an appropriate vector for in vitro transcription. The cDNA may be obtained by reverse transcription of RNA. In some aspects, starting material for IVT may include linearized DNA template, nucleotides, Rnase inhibitor, pyrophosphatase, and/or a polymerase (e.g., a T7 RNA polymerase). The nucleotides may be manufactured in house, may be obtained from a supplier, or may be synthesized. The nucleotides may be, but are not limited to, those described herein including natural and unnatural (modified) nucleotides. Any number of RNA polymerases or variants may be used, including, but not limited to, a phage RNA polymerase, e.g., a T7 RNA polymerase, a T3 RNA polymerase, a SP6 RNA polymerase, and/or mutant polymerases such as, but not limited to, polymerases able to incorporate modified nucleic acids and/or modified nucleotides, including chemically modified nucleic acids and/or nucleotides. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing RNA polymerases may be excluded from. Some embodiments exclude the use of Dnase. In some aspects, the IVT process is conducted in a bioreactor. The bioreactor may comprise a mixer. In some aspects, nucleotides may be added into the bioreactor throughout the IVT process. In some aspects, one or more post-IVT agents are added into the IVT mixture comprising RNA in the bioreactor after the IVT process. Exemplary post-IVT agents may include DNAse I configured to digest the linearized DNA template and/or proteinase K configured to digest DNAse I and T7 RNA polymerase. In some aspects, the post-IVT agents are incubated with the mixture in the bioreactor after IVT. In some aspects, the bioreactor may contain at least, at most, exactly, or between (inclusive or exclusive) any two of 60, 70, 80, 90, 100, 110, 120, 130, 140, 150 ,160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, and 500 or more liters IVT mixture. The IVT mixture may have an RNA concentration that is or is not at least, at most, exactly, or between (inclusive or exclusive) any two of 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 7.0, 8.0, 9.0, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50, 60, 70, 80, 90, and 100 mg/mL or more RNA. In some aspects, the IVT mixture may include residual spermidine, residual DNA, residual proteins, peptides, HEPES, EDTA, ammonium sulfate, cations (e.g., Mg2+, Na+, Ca2+), RNA fragments, residual nucleotides, free phosphates, or any combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing can be excluded from the IVT mixture. Isolation and/or purification of the nucleic acids described herein may include, but is not limited to, phenol/chloroform extraction and/or precipitation with either alcohol (ethanol, isopropanol) in the presence of monovalent cations or lithium chloride for nucleic acid clean-up, quality assurance and quality control. Additional, non-limiting examples of purification procedures include AGENCOURT® beads (Beckman Coulter Genomics, Danvers, MA), poly-T beads, LNATM oligo-T capture probes (EXIQON® Inc, Vedbaek, Denmark), HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC), size exclusion chromatography, and silica-based affinity chromatography and polyacrylamide gel electrophoresis. Purification can be performed using a variety of commercially available kits including, but not limited to SV Total Isolation System (Promega) and In vitro Transcription Cleanup and Concentration Kit (Norgen Biotek). In some aspects, 1, 2, 3, 4, 5, or more of the foregoing purification may be excluded. The term “purified” when used in relation to a nucleic acid such as a “purified nucleic acid” refers to one that is separated from at least one contaminant. A “contaminant” is any substance that makes another unfit, impure or inferior. Thus, a purified nucleic acid (e.g., DNA and RNA) is present in a form or setting different from that in which it is found in nature, or a form or setting different from that which existed prior to subjecting it to a treatment and/or purification method. In some aspects, at least a portion of the IVT mixture is filtered. The IVT mixture may be filtered via ultrafiltration and/or diafiltration to remove at least some impurities from the IVT mixture and/or to change buffer solution for the at least a portion of IVT mixture to produce a concentrated RNA solution as a retentate. In some aspects, both “ultrafiltration” and “diafiltration” refer to a membrane filtration process. Ultrafiltration typically uses membranes having pore sizes of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.001, 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, and 0.1 µm. In some aspects, ultrafiltration membranes are typically classified by molecular weight cutoff (MWCO) rather than pore size. For example, the MWCO may be at least, at most, exactly, or between (inclusive or exclusive) any two of 30 kDa, 40 kDa, 50 kDa, 60 kDa, 70 kDa, 80 kDa, 90 kDa, 100 kDa, 110 kDa, 120 kDa, 130 kDa, 140 kDa, 150 kDa, 160 kDa, 170 kDa, 180 kDa, 190 kDa, 200 kDa, 210 kDa, 220 kDa, 230 kDa, 240 kDa, 250 kDa, 260 kDa, 270 kDa, 280 kDa, 290 kDa, 300 kDa, 310 kDa, 320 kDa, 330 kDa, 340 kDa, 350 kDa, 360 kDa, 370 kDa, 380 kDa, 390 kDa, 400 kDa, 500 kDa, 600 kDa, 700 kDa, 800 kDa, 900 kDa, 1000 kDa, 2000 kDa, 3000 kDa, 4000 kDa, 5000 kDa, 6000 kDa, 7000 kDa, 8000 kDa, 9000 kDa, and 10000 kDa. A skilled artisan will understand that filtration membranes may comprise different suitable materials, including, e.g., polymers, cellulose, ceramic, etc., depending upon the application. In some aspects, membrane filtration may be more desirable for large volume purification process. In some aspects, ultrafiltration and diafiltration of the IVT mixture for purifying RNA may include (1) Direct Flow Filtration (DFF), also known as “dead-end” filtration, that applies a feed stream perpendicular to the membrane face and attempts to pass 100% of the fluid through the membrane, and/or (2) Tangential Flow Filtration (TFF), also known as crossflow filtration, where a feed stream passes parallel to the membrane face as one portion passes through the membrane (permeate) while the remainder (retentate) is retained and/or recirculated back to the feed tank. In some aspects, the filtering of the IVT mixture is conducted via TFF comprising an ultrafiltration step, a first diafiltration step, and a second diafiltration step. In some aspects, the first diafiltration step is conducted in the presence of ammonium sulfate. The first diafiltration step may be configured to remove a majority of impurities from the IVT mixture. In some aspects, the second diafiltration step is conducted without ammonium sulfate. The second diafiltration step may be configured to transfer the RNA into a DS buffer formulation. A filtration membrane with an appropriate MWCO may be selected for ultrafiltration in the TFF process. The MWCO of a TFF membrane determines which solutes may pass through the membrane into the filtrate and which are retained in the retentate. The MWCO of a TFF membrane may be selected such that substantially all of the solutes of interest (e.g., desired synthesized RNA species) remain in the retentate, whereas undesired components (e.g., excess ribonucleotides, small nucleic acid fragments such as digested or hydrolyzed DNA template, peptide fragments such as digested proteins and/or other impurities) pass into the filtrate. In some aspects, the retentate comprising desired synthesized RNA species may be re-circulated to a feed reservoir to be re-filtered in additional cycles. In some aspects, a TFF membrane may have a MWCO of at least, at most, exactly, or between (inclusive or exclusive) any two of 30 kDa, 40 kDa, 50 kDa, 60 kDa, 70 kDa, 80 kDa, 90 kDa, or more. In some aspects, a TFF membrane may have a MWCO of at least, at most, exactly, or between (inclusive or exclusive) any two of 100 kDa, 150 kDa, 200 kDa, 250 kDa, 300 kDa, 350 kDa, 400 kDa, or more. In some aspects, a TFF membrane may have a MWCO of or of about 250-350 kDa. In some aspects, a TFF membrane (e.g., a cellulose-based membrane) may have a MWCO of or of about 30-300 kDa; 50-300 kDa, 100-300 kDa, or 200-300 kDa. Diafiltration may be performed either discontinuously, or alternatively, continuously. For example, in continuous diafiltration, a diafiltration solution may be added to a sample feed reservoir at the same rate as filtrate is generated. In this way, the volume in the sample reservoir remains constant but small molecules (e.g., salts, solvents, etc.) that may freely permeate through a membrane are removed. Using solvent removal as an example, each additional diafiltration volume (DV) reduces the solvent concentration further. In discontinuous diafiltration, a solution is first diluted and then concentrated back to the starting volume. This process is then repeated until the desired concentration of small molecules (e.g., salts, solvents, etc.) remaining in the reservoir is reached. Each additional diafiltration volume (DV) reduces the small molecule (e.g., solvent) concentration further. Continuous diafiltration typically requires a minimum volume for a given reduction of molecules to be filtered. Discontinuous diafiltration, on the other hand, permits fast changes of the retentate condition, such as pH, salt content, and the like. In some aspects, the first diafiltration step is conducted with at least, at most, exactly, or between (inclusive or exclusive) any two of 2, 3, 4, 5, 6, 7, 8, 9, 10, or more diavolumes. In some aspects, the second diafiltration step is conducted with at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more diavolumes. In some aspects, the first diafiltration step is conducted with 5 diavolumes, and second diafiltration step is conducted with 10 diavolumes. In some aspects, for ultrafiltration and/or diafiltration, the IVT mixture is filtered at a rate of at least, at most, exactly, or between (inclusive or exclusive) any two of 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 500, 600, 700, 800, 900, or 1000 L/m2 of filter area per hour, or more. The concentrated RNA solution may comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5 mg/mL single stranded RNA. The bioburden of the concentrated RNA solution via filtration to obtain an RNA product solution may also be reduced, in some aspects. The filtration for reducing bioburden may be conducted using one or more filters. The one or more filters may include a filter with a pore size that is or is not at least, at most, exactly, or between (inclusive or exclusive) any two of 0.2 µm, 0.45 µm, 0.65 µm, 0.8 µm, or any other pore size configured to remove bioburdens. As one example, reducing the bioburden may include draining a retentate tank containing retentate obtained from the ultrafiltration and/or diafiltration to obtain the retentate. Reducing the bioburden may include flushing a filtration system for ultrafiltration and/or diafiltration using a wash buffer solution to obtain a wash pool solution comprising residue RNA remaining in the filtration system. The retentate may be filtered to obtain a filtered retentate. The wash pool solution may be filtered using a first 0.2 µm filter to obtain a filtered wash pool solution. The retentate may be filtered using the first 0.2 µm filter or another 0.2 µm filter. The filtered wash pool solution and the filtered retentate may be combined to form a combined pool solution. The combined pool solution may be filtered using a second 0.2 µm filter to obtain a filtered combined pool solution, which is further filtered using a third 0.2 µm filter to produce an RNA product solution. A quality assurance and/or quality control check may be conducted using methods such as, but not limited to, gel electrophoresis, UV absorbance, and/or analytical HPLC. In some aspects, the nucleic acids may be sequenced by methods including, but not limited to reverse-transcriptase-PCR. In some aspects, the nucleic acid may be quantified using methods such as, but not limited to, ultraviolet visible spectroscopy (UV/Vis). A non-limiting example of a UV/Vis spectrometer is a NANODROP® spectrometer (ThermoFisher, Waltham, MA). The quantified nucleic acid may be analyzed in order to determine if the nucleic acid may be of proper size and/or to assess degradation. Degradation of the nucleic acid may be assessed by methods such as, but not limited to, agarose gel electrophoresis, HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC), liquid chromatography-mass spectrometry (LCMS), capillary electrophoresis (CE) and capillary gel electrophoresis (CGE). In some aspects, 1, 2, 3, 4, 5, or more of the foregoing assessment methods may be excluded. V. RNA ENCAPSULATION The RNA in an RNA product solution may be encapsulated, and the RNA solution may further comprise at least one encapsulating agent. In one aspect, the encapsulating agent comprises a lipid, a lipid nanoparticle (LNP), lipoplexes, polymeric particles, polyplexes, monolithic delivery systems, or a combination thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing elements may be excluded as an encapsulating agent. In one aspect, the encapsulating agent is a lipid, and produced is lipid nanoparticle (LNP)- encapsulated RNA. Without intending to be bound by any theory, it is believed that the cationic or cationically ionizable lipid or lipid-like material and/or the cationic polymer combine together with the nucleic acid to form aggregates, and this aggregation results in colloidally stable particles. A lipid may be a naturally occurring lipid or a synthetic lipid. However, a lipid is usually a biological substance. Biological lipids are well known in the art, and include for example, neutral fats, phospholipids, phosphoglycerides, steroids, terpenes, lysolipids, glycosphingolipids, glucolipids, sulphatides, lipids with ether and ester-linked fatty acids and polymerizable lipids, and combinations thereof. A lipid is a substance that is insoluble in water and extractable with an organic solvent. Compounds other than those specifically described herein are understood by one of skill in the art as lipids and are encompassed by the compositions and methods of the present disclosure. A lipid component and a non-lipid may be attached to one another, either covalently or non-covalently. In some aspects, LNPs may be designed to protect RNA molecules (e.g., saRNA, mRNA) from extracellular Rnases and/or may be engineered for systemic delivery of the RNA to target cells. In some aspects, such LNPs may be particularly useful to deliver RNA molecules (e.g., saRNA, mRNA) when RNA molecules are intravenously administered to a subject in need thereof. In some aspects, such LNPs may be particularly useful to deliver RNA molecules (e.g., saRNA, mRNA) when RNA molecules are intramuscularly administered to a subject in need thereof. In some aspects, such LNPs may be particularly useful to deliver RNA molecules (e.g., saRNA, mRNA) when RNA molecules are intradermally administered to a subject in need thereof. In some aspects, such LNPs may be particularly useful to deliver RNA molecules (e.g., saRNA, mRNA) when RNA molecules are intranasally administered to a subject in need thereof. In one aspect, the RNA in the RNA product solution is at a concentration of < 1 mg/mL. In another aspect, the RNA is at a concentration of at least or at least about 0.05 mg/mL. In another aspect, the RNA is at a concentration of at least or at least about 0.5 mg/mL. In another aspect, the RNA is at a concentration of at least or at least about 1 mg/mL. In another aspect, the RNA concentration is from or from about 0.05 mg/mL to about 0.5 mg/mL. In another aspect, the RNA is at a concentration of at least 10 mg/mL. In another aspect, the RNA is at a concentration of at least 50 mg/mL. In some aspects, the RNA is or is not at a concentration of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 0.05 mg/mL, 0.06 mg/mL, 0.07 mg/mL, 0.08 mg/mL, 0.09 mg/mL, 0.10 mg/mL, 0.20 mg/mL, 0.30 mg/mL, 0.40 mg/mL, 0.5 mg/mL, 1 mg/mL, 10 mg/mL, 50 mg/mL, 75 mg/mL, 100 mg/mL, 150 mg/mL, 200 mg/mL, 250 mg/mL, 300 mg/mL, 400 mg/mL, or more. The present disclosure provides for an RNA product solution and a lipid preparation mixture or compositions thereof comprising at least one RNA encoding, e.g., an antigen (e.g., an RSV prefusion F protein) complexed with, encapsulated in, and/or formulated with one or more lipids, and forming lipid nanoparticles (LNPs), liposomes, lipoplexes and/or nanoliposomes. In some aspects, the composition comprises a lipid nanoparticle. A lipid nanoparticle or LNP refers to particles of any morphology generated when a cationic lipid and optionally one or more further lipids are combined, e.g., in an aqueous environment and/or in the presence of RNA. In some aspects, lipid nanoparticles are included in a formulation that may be used to deliver an active agent or therapeutic agent, such as a nucleic acid (e.g., mRNA) to a target site of interest (e.g., cell, tissue, organ, tumor, and the like). In some aspects, the lipid nanoparticles of the present disclosure comprise a nucleic acid (e.g., mRNA). Such lipid nanoparticles typically comprise a cationic lipid and one or more excipients, e.g., one or more neutral lipids, charged lipids, steroids, polymer conjugated lipids, or combinations thereof. In some aspects, the LNPs comprise at least one cationic (e.g., ionizable) lipid, at least one neutral (e.g., non-cationic) lipid, at least one structural lipid (e.g., a steroid), and/or at least one polymer conjugated lipid (e.g., a polyethylene glycol (PEG)-modified lipid). In some aspects, 1, 2, 3, or more of the foregoing excipients may be excluded from the LNPs. In some aspects, the LNPs comprise 20-60 mol% cationic (e.g., ionizable) lipid(s). For example, the LNPs may comprise 20-50 mol%, 20-40 mol%, 20-30 mol%, 30-60 mol%, 30-50 mol%, 30-40 mol%, 40-60 mol%, 40-50 mol%, or 50-60 mol% cationic (e.g., ionizable) lipid(s). In some aspects, the LNPs comprise or do not comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 20 mol%, 30 mol%, 40 mol%, 50, or 60 mol% cationic (e.g., ionizable) lipid(s). In some aspects, the LNPs comprise 45 to 55 mole percent (mol%) cationic (e.g., ionizable) lipid(s). For example, LNPs may comprise or not comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, or 55 mol% cationic (e.g., ionizable) lipid(s). In some aspects, the LNPs comprise 5-25 mol% neutral (e.g., non-cationic) lipid(s). For example, the LNPs may comprise 5-20 mol%, 5-15 mol%, 5-10 mol%, 10-25 mol%, 10-20 mol%, 10-25 mol%, 15-25 mol%, 15-20 mol%, or 20-25 mol% neutral (e.g., non-cationic) lipid(s). In some aspects, the LNPs are or are not at least, at most, exactly, or between (inclusive or exclusive) any two of 5 mol%, 10 mol%, 15 mol%, 20 mol%, or 25 mol% neutral (e.g., non- cationic) lipid(s). In some aspects, the LNPs comprise 5 to 15 mol% neutral (e.g., non-cationic) lipid(s). For example, LNPs may comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 mol% neutral (e.g., non-cationic) lipid(s). In some aspects, the LNPs comprise 25-55 mol% structural lipid(s) (e.g., a steroid). For example, the LNPs may comprise 25-50 mol%, 25-45 mol%, 25-40 mol%, 25-35 mol%, 25-30 mol%, 30-55 mol%, 30-50 mol%, 30-45 mol%, 30-40 mol%, 30-35 mol%, 35-55 mol%, 35-50 mol%, 35-45 mol%, 35-40 mol%, 40-55 mol%, 40-50 mol%, 40-45 mol%, 45-55 mol%, 45-50 mol%, or 50-55 mol% structural lipid(s) (e.g., a steroid). In some aspects, the LNPs are or are not at least, at most, exactly, or between (inclusive or exclusive) any two of 25 mol%, 30 mol%, 35 mol%, 40 mol%, 45 mol%, 50 mol%, or 55 mol% structural lipid(s) (e.g., a steroid). In some aspects, the LNPs comprise 35 to 40 mol% structural lipid(s) (e.g., a steroid). For example, LNPs may comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 35, 36, 37, 38, 39, or 40 mol% structural lipid(s) (e.g., a steroid). In some aspects, the LNPs comprise 0.5-15 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)-modified lipid). For example, the lipid nanoparticle may comprise 0.5- 10 mol%, 0.5-5 mol%, 1-15 mol%, 1-10 mol%, 1-5 mol%, 2-15 mol%, 2-10 mol%, 2-5 mol%, 5- 15 mol%, 5-10 mol%, or 10-15 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)-modified lipid). In some aspects, the lipid LNPs are or are not at least, at most, exactly, or between (inclusive or exclusive) any two of 0.5 mol%, 1 mol%, 2 mol%, 3 mol%, 4 mol%, 5 mol%, 6 mol%, 7 mol%, 8 mol%, 9 mol%, 10 mol%, 11 mol%, 12 mol%, 13 mol%, 14 mol%, or 15 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)-modified lipid). In some aspects, the LNPs comprise 1 to 2 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)- modified lipid). For example, LNPs may comprise at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 1.5, or 2 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)-modified lipid). In some aspects, the LNPs comprise 20-75 mol% cationic (e.g., ionizable) lipid(s) (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, and 75%), 0.5-25 mol% neutral (e.g., non-cationic) lipid(s) (e.g., at least, at most, exactly, or between (inclusive or exclusive) of 0.5%, 2.25%, 4%, 5.75%, 7.5%, 9.25%, 11%, 12.75%, 14.5%, 16.25%, 18%, 19.75%, 21.5%, 23.25%, and 25%), 5-55 mol% structural lipid(s) (e.g., a sterol) e.g., non-cationic) lipid(s) (e.g., at least, at most, exactly, or between (inclusive or exclusive) of 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, and 55%), and 0.5-20 mol% polymer conjugated lipid(s) (e.g., a polyethylene glycol (PEG)- modified lipid) (e.g., at least, at most, exactly, or between (inclusive or exclusive) of 0.5%, 2%, 3.5%, 5%, 6.5%, 8%, 9.5%, 11%, 12.5%, 14%, 15.5%, 17%, 18.5%, and 20%). In some aspects, 1, 2, 3, or more of the lipids may be excluded from the LNPs. In some non-limiting aspects, the molar lipid ratio is 50/10/38.5/1.5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 60/7.5/31/1.5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 57.5/7.5/31.5/3.5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 57.2/7.1/34.3/1.4 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 40/15/40/5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 50/10/35/4.5/0.5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 50/10/35/5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 47.5/10/40.7/1.8 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 40/10/40/10 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), 35/15/40/10 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid), or 52/13/30/5 (mol% cationic lipid/neutral lipid/structural lipid/polymer conjugated lipid). In some aspects, the active agent or therapeutic agent, such as a nucleic acid (e.g., mRNA), may be encapsulated in the lipid portion of the lipid nanoparticle and/or an aqueous space enveloped by some or all of the lipid portion of the lipid nanoparticle, thereby protecting it from enzymatic degradation or other undesirable effects induced by the mechanisms of the host organism or cells, e.g., an adverse immune response. The nucleic acid (e.g., mRNA) or a portion thereof may also be associated and complexed with the lipid nanoparticle. A lipid nanoparticle may comprise any lipid capable of forming a particle to which the nucleic acids are attached, and/or in which the one or more nucleic acids are encapsulated. In some aspects, provided RNA molecules (e.g., mRNA) may be formulated with LNPs. In some aspects, the lipid nanoparticles may or may not have a mean diameter of or of about 1 to 500 nm (e.g., at least, at most, exactly, or between (inclusive or exclusive) of 1, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, or 500 nm). In some aspects, the lipid nanoparticles have a mean diameter of or of from about 30 nm to about 150 nm, about 40 nm to about 150 nm, about 50 nm to about 150 nm, about 60 nm to about 130 nm, about 70 nm to about 110 nm, about 70 nm to about 100 nm, about 80 nm to about 100 nm, about 90 nm to about 100 nm, about 70 to about 90 nm, about 80 nm to about 90 nm, about 70 nm to about 80 nm, or at least, at most, exactly, or between (inclusive or exclusive) of 30 nm, 35 nm, 40 nm, 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, 80 nm, 85 nm, 90 nm, 95 nm, 100 nm, 105 nm, 110 nm, 115 nm, 120 nm, 125 nm, 130 nm, 135 nm, 140 nm, 145 nm, or 150 nm, and are substantially non-toxic. The term “mean diameter” refers to the mean hydrodynamic diameter of particles as measured by dynamic laser light scattering (DLS) with data analysis using the so-called cumulant algorithm, which provides as results the so-called Z-average with the dimension of a length, and the polydispersity index (PI), which is dimensionless (Koppel, D., J. Chem. Phys. 57, 1972, pp 4814-4820, ISO 13321). Here, “mean diameter,” “diameter,” or “size” for particles is used synonymously with the value of the Z-average. LNPs described herein may exhibit a polydispersity index less than or less than about 0.5, 0.4, 0.3, or 0.2 or less. By way of example, the LNPs may or may not exhibit a polydispersity index of at least, at most, exactly, or between (inclusive or exclusive) of 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, or 0.5. The polydispersity index is, in some aspects, calculated based on dynamic light scattering measurements by the so-called cumulant analysis referred to in the definition of “average diameter.” Under certain prerequisites, it may be taken as a measure of the size distribution of an ensemble of nanoparticles. In some aspects, an LNP of the disclosure comprises or does not comprise an N:P ratio of or of from about 2:1 to about 30:1, e.g., at least, at most, exactly, or between (inclusive or exclusive) of 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, 21:1, 22:1, 23:1, 24:1, 25:1, 26:1, 27:1, 28:1, 29:1, or 30:1. In some aspects, an LNP of the disclosure comprises an N:P ratio of or of about 6:1. In some aspects, an LNP of the disclosure comprises an N:P ratio of or of about 3:1. As used herein “N:P” is the ratio of cationic lipid to RNA. In some aspects, an LNP of the disclosure comprises or does not comprise a wt/wt ratio of the cationic lipid component to the RNA of or of from about 5:1 to about 100:1, e.g., at least, at most, exactly, or between (inclusive or exclusive) of 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, 21:1, 22:1, 23:1, 24:1, 25:1, 26:1, 27:1, 28:1, 29:1, 30:1, 31:1, 32:1, 33:1, 34:1, 35:1, 36:1, 37:1, 38:1, 39:1, 40:1, 41:1, 42:1, 43:1, 44:1, 45:1, 46:1, 47:1, 48:1, 49:1, 50:1, 51:1, 52:1, 53:1, 54:1, 55:1, 56:1, 57:1, 58:1, 59:1, 60:1, 61:1, 62:1, 63:1, 64:1, 65:1, 66:1, 67:1, 68:1, 69:1, 70:1, 71:1, 72:1, 73:1, 74:1, 75:1, 76:1, 77:1, 78:1, 79:1, 80:1, 81:1, 82:1, 83:1, 84:1, 85:1, 86:1, 87:1, 88:1, 89:1, 90:1, 91:1, 92:1, 93:1, 94:1, 95:1, 96:1, 97:1, 98:1, 99:1, or 100:1. In some aspects, an LNP of the disclosure comprises a wt/wt ratio of the ionizable cationic lipid component to the RNA of or of about 20:1. In some aspects, an LNP of the disclosure comprises a wt/wt ratio of the ionizable cationic lipid component to the RNA of or of about 10:1. In certain aspects, nucleic acids (e.g., RNA molecules), when present in provided LNPs, are resistant in aqueous solution to degradation with a nuclease. In some aspects, LNPs are liver- targeting lipid nanoparticles. In some aspects, LNPs are cationic lipid nanoparticles comprising one or more cationic lipids (e.g., those described herein). In some aspects, cationic LNPs may comprise at least one cationic lipid, at least one polymer conjugated lipid, and at least one helper lipid (e.g., at least one neutral lipid). In certain aspects, the RNA solution and lipid preparation mixture or compositions thereof may have at least, at most, exactly, between (inclusive or exclusive) of, or about 1%, 2%, 3%, 4% 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% of a particular lipid, lipid type, or non- lipid component such as lipid-like materials and/or cationic polymers and/or an adjuvant, antigen, peptide, polypeptide, sugar, nucleic acid or other material disclosed herein or as would be known to one of skill in the art. LNPs described herein can be generated using components, compositions, and methods as are generally known in the art, see , e.g., PCT/US2016/052352; PCT/US2016/068300; PCT/US2017/037551; PCT/US2015/027400; PCT/US2016/047406; PCT/US2016000129; PCT/US2016/014280; PCT/US2016/014280; PCT/US2017/038426; PCT/US2014/027077; PCT/US2014/055394; PCT/US2016/052117; PCT/US2012/069610; PCT/US2017/027492; PCT/US2016/059575 and PCT/US2016/069491, all of which are incorporated by reference herein in their entirety. Other non-limiting examples of methods for preparing LNPs can be found in, e.g., WO 2022/032154, the disclosure of which is incorporated by reference herein in its entirety. For example, methods of preparing LNPs may involve obtaining a colloid from at least one cationic or cationically ionizable lipid or lipid-like material and/or at least one cationic polymer and mixing the colloid with nucleic acid to obtain nucleic acid particles. The term “colloid” as used herein relates to a type of homogeneous mixture in which dispersed particles do not settle out. The insoluble particles in the mixture are microscopic, with particle sizes between 1 and 1000 nanometers. The mixture may be termed a colloid or a colloidal suspension. Sometimes the term “colloid” refers only to the particles in the mixture and not the entire suspension. For the preparation of colloids comprising at least one cationic or cationically ionizable lipid or lipid-like material and/or at least one cationic polymer, methods are applicable herein that are conventionally used for preparing liposomal vesicles and are appropriately adapted. The most commonly used methods for preparing liposomal vesicles share the following fundamental stages: (i) lipids dissolution in organic solvents, (ii) drying of the resultant solution, and (iii) hydration of dried lipid (using various aqueous media). In the film hydration method, lipids are first dissolved in a suitable organic solvent and dried down to yield a thin film at the bottom of the flask. The obtained lipid film is hydrated using an appropriate aqueous medium to produce a liposomal dispersion. Furthermore, an additional downsizing step may be included. Reverse phase evaporation is an alternative method to film hydration for preparing liposomal vesicles that involves formation of a water-in-oil emulsion between an aqueous phase and an organic phase containing lipids. A brief sonication of this mixture is required for system homogenization. The removal of the organic phase under reduced pressure yields a milky gel that subsequently turns into a liposomal suspension. The term “ethanol injection technique” refers to a process in which an ethanol solution comprising lipids is rapidly injected into an aqueous solution through a needle. This action disperses the lipids throughout the solution and promotes lipid structure formation, for example, lipid vesicle formation such as liposome formation. Generally, the RNA lipoplex particles described herein are obtainable by adding RNA to a colloidal liposome dispersion. Using the ethanol injection technique, such colloidal liposome dispersion is, in some aspects, formed as follows: an ethanol solution comprising lipids, such as cationic lipids and additional lipids, is injected into an aqueous solution under stirring. In some aspects, the RNA lipoplex particles described herein are obtainable without a step of extrusion. The term “extruding” or “extrusion” refers to the creation of particles having a fixed, cross-sectional profile. In particular, it refers to the downsizing of a particle, whereby the particle is forced through filters with defined pores. Other methods for preparing a colloid having organic solvent free characteristics may also be used according to the present disclosure. In some aspects, LNP-encapsulated RNA may be produced by rapid mixing of an RNA solution described herein (e.g., the RNA product solution) and a lipid preparation described herein (comprising, e.g., at least one cationic lipid and optionally one or more other lipid components, in an organic solvent) under conditions such that a sudden change in solubility of lipid component(s) is triggered, which drives the lipids towards self-assembly in the form of LNPs. In some aspects, suitable buffering agents comprise tris, histidine, citrate, acetate, phosphate, and/or succinate. In some aspects, 1, 2, 3, or more of the foregoing buffering agents are excluded. The pH of a liquid formulation relates to the pKa of the encapsulating agent (e.g., cationic lipid). The pH of the acidifying buffer may be at least half a pH scale less than the pKa of the encapsulating agent (e.g., cationic lipid), and the pH of the final buffer may be at least half a pH scale greater than the pKa of the encapsulating agent (e.g., cationic lipid). In some aspects, properties of a cationic lipid are chosen such that nascent formation of particles occurs by association with an oppositely charged backbone of a nucleic acid (e.g., RNA). In this way, particles are formed around the nucleic acid, which, for example, in some aspects, may result in much higher encapsulation efficiency than is achieved in the absence of interactions between nucleic acids and at least one of the lipid components. In certain aspects, nucleic acids, when present in the lipid nanoparticles, are resistant in aqueous solution to degradation with a nuclease. Lipid nanoparticles comprising nucleic acids and their method of preparation are disclosed in, e.g., U.S. Patent Publication Nos. 2004/0142025, 2007/0042031 and PCT Pub. Nos. WO 2013/016058 and WO 2013/086373, the full disclosures of which are herein incorporated by reference in their entirety for all purposes. Some aspects described herein relate to compositions, methods and uses involving more than one, e.g., 2, 3, 4, 5, 6 or even more nucleic acid species, such as RNA species. In an LNP formulation, it is possible that each nucleic acid species is separately formulated as an individual LNP formulation. In that case, each individual LNP formulation will comprise one nucleic acid species. The individual LNP formulations may be present as separate entities, e.g., in separate containers. Such formulations are obtainable by providing each nucleic acid species separately (typically each in the form of a nucleic acid-containing solution) together with suitable cationic or cationically ionizable lipids or lipid-like materials and cationic polymers that allow the formation of LNPs. Respective particles will contain exclusively the specific nucleic acid species that is being provided when the particles are formed (individual particulate formulations). In some aspects, a composition such as a pharmaceutical composition comprises more than one individual LNP formulation. Respective pharmaceutical compositions are referred to as mixed LNP formulations. Mixed LNP formulations according to the invention are obtainable by forming, separately, individual LNP formulations, as described above, followed by a step of mixing of the individual LNP formulations. By the step of mixing, a formulation comprising a mixed population of nucleic acid-containing LNPs is obtainable. Individual LNP populations may be together in one container, comprising a mixed population of individual LNP formulations. Alternatively, it is possible that different nucleic acid species are formulated together as a combined LNP formulation. Such formulations are obtainable by providing a combined formulation (typically combined solution) of different RNA species together with suitable cationic or cationically ionizable lipids or lipid-like materials and cationic polymers that allow the formation of LNPs. As opposed to a mixed LNP formulation, a combined LNP formulation will typically comprise LNPs that comprise more than one RNA species. In a combined LNP composition, different RNA species are typically present together in a single particle. A. CATIONIC POLYMERIC MATERIALS Given their high degree of chemical flexibility, polymeric materials are commonly used for nanoparticle-based delivery. Typically, cationic materials are used to electrostatically condense the negatively charged nucleic acid into nanoparticles. These positively charged groups often consist of amines that change their state of protonation in the pH range between 5.5 and 7.5, thought to lead to an ion imbalance that results in endosomal rupture. Polymers such as poly-L- lysine, polyamidoamine, protamine and polyethyleneimine, as well as naturally occurring polymers such as chitosan have all been applied to nucleic acid delivery and are suitable as cationic materials useful in some aspects herein. In addition, some investigators have synthesized polymeric materials specifically for nucleic acid delivery. Poly(P-amino esters), in particular, have gained widespread use in nucleic acid delivery owing to their ease of synthesis and biodegradability. In some aspects, such synthetic materials may be suitable for use as cationic materials herein. A “polymeric material,” as used herein, is given its ordinary meaning, e.g., a molecular structure comprising one or more repeat units (monomers), connected by covalent bonds. In some aspects, such repeat units may all be identical; alternatively, in some cases, there may be more than one type of repeat unit present within the polymeric material. In some cases, a polymeric material is biologically derived, e.g., a biopolymer such as a protein. In some cases, additional moieties may also be present in the polymeric material, for example targeting moieties such as those described herein. Those skilled in the art are aware that, when more than one type of repeat unit is present within a polymer (or polymeric moiety), then the polymer (or polymeric moiety) is said to be a “copolymer.” In some aspects, a polymer (or polymeric moiety) utilized in accordance with the present disclosure may be a copolymer. Repeat units forming the copolymer may be arranged in any fashion. For example, in some aspects, repeat units may be arranged in a random order; alternatively or additionally, in some aspects, repeat units may be arranged in an alternating order, or as a “block” copolymer, e.g., comprising one or more regions each comprising a first repeat unit (e.g., a first block), and one or more regions each comprising a second repeat unit (e.g., a second block), etc. Block copolymers may have two (a diblock copolymer), three (a triblock copolymer), or more numbers of distinct blocks. In certain aspects, a polymeric material for use in accordance with the present disclosure is biocompatible. Biocompatible materials are those that typically do not result in significant cell death at moderate concentrations. In certain aspects, a biocompatible material is biodegradable, e.g., is able to degrade, chemically and/or biologically, within a physiological environment, such as within the body. In certain aspects, a polymeric material may be or comprise protamine or polyalkyleneimine, in particular protamine. As those skilled in the art are aware term “protamine” is often used to refer to any of various strongly basic proteins of relatively low molecular weight that are rich in arginine and are found associated especially with DNA in place of somatic histones in the sperm cells of various animals (as fish). In particular, the term “protamine” is often used to refer to proteins found in fish sperm that are strongly basic, are soluble in water, are not coagulated by heat, and yield chiefly arginine upon hydrolysis. In purified form, they are used in a long-acting formulation of insulin and to neutralize the anticoagulant effects of heparin. In some aspects, the term “protamine” as used herein is refers to a protamine amino acid sequence obtained or derived from natural or biological sources, including fragments thereof and/or multimeric forms of said amino acid sequence or fragment thereof, as well as (synthesized) polypeptides which are artificial and specifically designed for specific purposes and cannot be isolated from native or biological sources. In some aspects, a polyalkyleneimine comprises polyethylenimine and/or polypropylenimine. In some aspects, the polyalkyleneimine is polyethyleneimine (PEI). In some aspects, the polyalkyleneimine is a linear polyalkyleneimine, e.g., linear polyethyleneimine (PEI). Cationic materials (e.g., polymeric materials, including polycationic polymers) contemplated for use herein include those which are able to electrostatically bind nucleic acid. In some aspects, cationic polymeric materials contemplated for use herein include any cationic polymeric materials with which nucleic acid may be associated, e.g. by forming complexes with the nucleic acid or forming vesicles in which the nucleic acid is enclosed or encapsulated. In some aspects, particles described herein may comprise polymers other than cationic polymers, e.g., non-cationic polymeric materials and/or anionic polymeric materials. Collectively, anionic and neutral polymeric materials are referred to herein as non-cationic polymeric materials. B. LIPIDS & LIPID-LIKE MATERIALS The terms “lipid” and “lipid-like material” are used herein to refer to molecules which comprise one or more hydrophobic moieties or groups and optionally also one or more hydrophilic moieties or groups. According to the disclosure, lipids and lipid-like materials may be cationic, anionic or neutral. Neutral lipids or lipid-like materials exist in an uncharged or neutral zwitterionic form at a selected pH. The term “lipid” refers to a group of organic compounds that are characterized by being insoluble in water but soluble in many organic solvents. Generally, lipids may be divided into eight categories: fatty acids and their derivatives (including tri-, di-, monoglycerides, and phospholipids), glycerolipids, glycerophospholipids, sphingolipids, saccharolipids, polyketides, sterol lipids as well as sterol-containing metabolites such as cholesterol, and prenol lipids. Examples of fatty acids include, but are not limited to, fatty esters and fatty amides. Examples of glycerolipids include, but are not limited to, glycosylglycerols and glycerophospholipids (e.g., phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine). Examples of sphingolipids include, but are not limited to, ceramides phosphosphingolipids (e.g., sphingomyelins, phosphocholine), and glycosphingolipids (e.g., cerebrosides, gangliosides). Examples of sterol lipids include, but are not limited to, cholesterol and its derivatives and tocopherol and its derivatives. In some aspects, 1, 2, 3, 4, 5, or more of the lipids may be excluded from the LNPs of the present disclosure. The term “lipid-like material,” “lipid-like compound,” or “lipid-like molecule” relates to substances that structurally and/or functionally relate to lipids but may not be considered as lipids in a strict sense. For example, the term includes compounds that are able to form amphiphilic layers as they are present in vesicles, multilamellar/unilamellar liposomes, or membranes in an aqueous environment and includes surfactants, or synthesized compounds with both hydrophilic and hydrophobic moieties. Generally speaking, the term refers to molecules, which comprise hydrophilic and hydrophobic moieties with different structural organization, which may or may not be similar to that of lipids. In some aspects, the RNA solution and lipid preparation mixture or compositions thereof may comprise cationic lipids, neutral lipids, cholesterol, and/or polymer (e.g., polyethylene glycol) conjugated lipids which form lipid nanoparticles that encompass the RNA molecules. Therefore, in some aspects, the LNP may comprise a cationic lipid and one or more excipients, e.g., one or more neutral lipids, charged lipids, steroids or steroid analogs (e.g., cholesterol), polymer conjugated lipids (e.g. PEG-lipid), or combinations thereof. In some aspects, 1, 2, 3, or more of the foregoing excipients may be excluded from the LNPs of the present disclosure. In some aspects, the lipids are present in a composition in an amount that is effective to form a lipid nanoparticle and deliver a therapeutic agent, e.g., an RNA molecule, for treating a particular disease or condition of interest, e.g., those related to RSV. In some aspects, the LNPs encompass, or encapsulate, the nucleic acid molecules. i. CATIONIC LIPIDS Cationic or cationically ionizable lipids or lipid-like materials refer to a lipid or lipid-like material capable of being positively charged and able to electrostatically bind nucleic acid. As used herein, a “cationic lipid” or “cationic lipid-like material” refers to a lipid or lipid-like material having a net positive charge. Cationic lipids or lipid-like materials bind negatively charged nucleic acid by electrostatic interaction. Generally, cationic lipids possess a lipophilic moiety, such as a sterol, an acyl chain, a diacyl, or more acyl chains, and the head group of the lipid typically carries the positive charge. Exemplary cationic lipids include one or more amine group(s) which bear the positive charge. Cationic lipids may encapsulate negatively charged RNA. In some aspects, cationic lipids are ionizable such that they may exist in a positively charged or neutral form depending on pH. The ionization of the cationic lipid affects the surface charge of the lipid nanoparticle under different pH conditions. Without wishing to be bound by theory, this ionizable behavior is thought to enhance efficacy through helping with endosomal escape and reducing toxicity as compared with particles that remain cationic at physiological pH. For purposes of the present disclosure, such “cationically ionizable” lipids or lipid-like materials are comprised by the term “cationic lipid” or “cationic lipid-like material” unless contradicted by the circumstances. In some aspects, a cationic lipid may comprise from or from about 10 mol % to about 100 mol %, about 20 mol % to about 100 mol %, about 30 mol % to about 100 mol %, about 40 mol % to about 100 mol %, or about 50 mol % to about 100 mol % of the total lipid present in the particle. In some aspects, a cationic lipid may or may not be at least, at most, exactly, or between (inclusive or exclusive) of 10 mol %, 20 mol %, 30 mol %, 40 mol %, 50 mol %, 60 mol %, 70 mol %, 80 mol %, 90 mol %, or 100 mol %, or any range or value derivable therein, of the total lipid present in the particle. In a preferred aspect, the cationic lipid comprises 47.5 mol% of the total lipid present in the LNP. Examples of cationic lipids include, but are not limited to: ((4- hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate), 1,2-dioleoyl-3- trimethylammonium propane (DOTAP), N,N-dimethyl-2,3-dioleyloxypropylamine (DODMA), 1,2- di-O-octadecenyl-3-trimethylammonium propane (DOTMA), 3-(N-( N′,N′-dimethylaminoethane)- carbamoyl)cholesterol (DC-Chol), dimethyldioctadecylammonium (DDAB), 1,2-dioleoyl-3- dimethylammonium-propane (DODAP), 1,2-diacyloxy-3-dimethylammonium propanes, 1,2- dialkyloxy-3-dimethylammonium propanes; dioctadecyldimethyl ammonium chloride (DODAC), 1,2-distearyloxy-N,N-dimethyl-3-aminopropane (DSDMA), 2,3-di(tetradecoxy)propyl-(2- hydroxyethyl)-dimethylazanium (DMRIE), 1,2-dimyristoyl-sn-glycero-3-ethylphosphocholine (DMEPC), 1,2-dimyristoyl-3-trimethylammonium propane (DMTAP), 1,2-dioleoyloxypropyl-3- dimethyl-hydroxyethyl ammonium bromide (DORIE), 2,3-dioleoyloxy-N-[2(spermine carboxamide)ethyl]-N,N-dimethyl-1-propanamium trifluoroacetate (DOSPA), 1,2-dilinoleyloxy- N,N-dimethylaminopropane (DLinDMA), 1,2-dilinolenyloxy-N,N-dimethylaminopropane (DLenDMA), dioctadecylamidoglycyl spermine (DOGS), 3-dimethylamino-2-(cholest-5-en-3- beta-oxybutan-4-oxy)-1-(cis,cis-9,12-oc-tadecadienoxy)propane (CLinDMA), 2-[5′-(cholest-5-en- 3-beta-oxy)-3′-oxapentoxy)-3-dimethyl-l-(cis,cis-9′,12′-octadecadienoxy)propane (CpLinDMA), N,N-dimethyl-3,4-dioleyloxybenzylamine (DMOBA), 1,2-N,N′-dioleylcarbamyl-3- dimethylaminopropane (DOcarbDAP), 2,3-dilinoleoyloxy-N,N-dimethylpropylamine (DLinDAP), 1,2-N,N′-dilinoleylcarbamyl-3-dimethylaminopropane (DLincarbDAP), 1,2-Dilinoleoylcarbamyl-3- dimethylaminopropane (DLinCDAP), 2,2-dilinoleyl-4-dimethylaminomethyl-[1,3]-dioxolane (DLin-K-DMA), 2,2-dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane (DLin-K-XTC2-DMA), 2,2- dilinoleyl-4-(2-dimethylaminoethyl)-[1,3]-dioxolane (DLin-KC2-DMA), heptatriaconta-6,9,28,31- tetraen-19-yl-4-(dimethylamino)butanoate (DLin-MC3-DMA), N-(2-Hydroxyethyl)-N,N-dimethyl- 2,3-bis(tetradecyloxy)-1-propanaminium bromide (DMRIE), (±)-N-(3-aminopropyl)-N,N-dimethyl- 2,3-bis(cis-9-tetradecenyloxy)-1-propanaminium bromide (GAP-DMORIE), (±)-N-(3- aminopropyl)-N,N-dimethyl-2,3-bis(dodecyloxy)-1-propanaminium bromide (GAP-DLRIE), (±)-N- (3-aminopropyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide (GAP-DMRIE), N-(2-Aminoethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide (bAE-DMRIE), N-(4-carboxybenzyl)-N,N-dimethyl-2,3-bis(oleoyloxy)propan-1-aminium (DOBAQ), 2-({8-[(3b)- cholest-5-en-3-yloxy]octyl}oxy)-N,N-dimethyl-3-[(9Z,12Z)-octadeca-9,12-dien-1-yloxy]propan-1- amine (Octyl-CLinDMA), 1,2-dimyristoyl-3-dimethylammonium-propane (DMDAP), 1,2- dipalmitoyl-3-dimethylammonium-propane (DPDAP), N1-[2-((1S)-1-[(3-aminopropyl)amino]-4- [di(3-amino-propyl)amino]butylcarboxamido)ethyl]-3,4-di[oleyloxy]-benzamide (MVL5), 1,2- dioleoyl-sn-glycero-3-ethylphosphocholine (DOEPC), 2,3-bis(dodecyloxy)-N-(2-hydroxyethyl)- N,N-dimethylpropan-1-amonium bromide (DLRIE), N-(2-aminoethyl)-N,N-dimethyl-2,3- bis(tetradecyloxy)propan-1-aminium bromide (DMORIE), di((Z)-non-2-en-l-yl) 8,8′- ((((2(dimethylamino)ethyl)thio)carbonyl)azanediyl)dioctanoate (ATX), N,N-dimethyl-2,3- bis(dodecyloxy)propan-1-amine (DLDMA), N,N-dimethyl-2,3-bis(tetradecyloxy)propan-1-amine (DMDMA), Di((Z)-non-2-en-l-yl)-9-((4-(dimethylaminobutanoyl)oxy)heptadecanedioate (L319), N-dodecyl-3-((2-dodecylcarbamoyl-ethyl)-{2-[(2-dodecylcarbamoyl-ethyl)-2-{(2- dodecylcarbamoyl-ethyl)-[2-(2-dodecylcarbamoyl-ethylamino)-ethyl]-amino}- ethylamino)propionamide (lipidoid 98N12-5), 1-[2-[bis(2-hydroxydodecyl)amino]ethyl-[2-[4-[2- [bis(2 hydroxydodecyl)amino]ethyl]piperazin-1-yl]ethyl]amino]dodecan-2-ol (lipidoid 02-200); C 12-200; or heptadecan-9-yl 8-((2-hydroxyethyl) (6-oxo-6-(undecyloxy)hexyl) amino) octanoate (SM-102). In some aspects, 1, 2, 3, 4, 5, or more of the foregoing cationic lipids may be excluded from the LNPs of the present disclosure. In some aspects, an ionizable cationic lipid of the disclosure comprises a compound of Formula (I): or a pharmaceutically acceptable salt, tautomer, prodrug, or stereoisomer thereof, wherein: R1 is a C5-30 alkyl, C5-20 alkenyl, -R*YR”, -YR”, or -R”M’R’; R2 and R3 are independently a H, C1-14 alkyl, C2-14 alkenyl, -R*YR”, -YR”, or -R*OR”, and/or R2 and R3, together with the atom to which they are attached, form a heterocycle or carbocycle; R4 is a C3-6 carbocycle, -(CH2)nQ, -(CH2)nCHQR, -CHQR, -CQ(R)2, or unsubstituted C1-6 alkyl, where Q is a carbocycle, heterocycle, -OR, -O(CH2)nN(R)2, -C(O)OR, -OC(O)R, -CX3, - CX2H, -CXH2, -CN, -N(R)2, -C(O)N(R)2, -N(R)C(O)R, -N(R)S(O)2R, -N(R)C(O)N(R)2, - N(R)C(S)N(R)2, -N(R)R8, -O(CH2)nOR, -N(R)C(=NR9)N(R)2, -N(R)C(=CHR9)N(R)2, -OC(O)N(R)2, -N(R)C(O)OR, -N(OR)C(O)R, -N(OR)S(O)2R, -N(OR)C(O)OR, -N(OR)C(O)N(R)2, - N(OR)C(S)N(R)2, -N(OR)C(=NR9)N(R)2, -N(OR)C(=CHR9)N(R)2, -C(=NR9)N(R)2, -C(=NR9)R, - C(O)N(R)OR, or -C(R)N(R)2C(O)OR, and/or each n is independently a 1, 2, 3, 4, or 5; each R5 is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R6 is independently a C1-3 alkyl, C2-3 alkenyl, or H; M and M’ are independently a -C(O)O-, -OC(O)-, -C(O)N(R’)-, -N(R’)C(O)-, -C(O)-, -C(S)- , -C(S)S-, -SC(S)-, -CH(OH)-, -P(O)(OR’)O-, -S(O)2-, -S-S-, an aryl group, or a heteroaryl group; R7 is a C1-3 alkyl, C2-3 alkenyl, or H; R8 is a C3-6 carbocycle or heterocycle; R9 is a H, CN, NO2, C1-6 alkyl, -OR, -S(O)2R, -S(O)2N(R)2, C2-6 alkenyl, C3-6 carbocycle, or heterocycle; each R is a C1-3 alkyl, C2-3 alkenyl, or H; each R' is a C1-18 alkyl, C2-18 alkenyl, -R*YR”, -YR”, or H; each R” is a C3-14 alkyl or C3-14 alkenyl; each R* is independently a C1-12 alkyl or C2-12 alkenyl; each Y is independently a C3-6 carbocycle; each X is independently a F, Cl, Br, or I; and m is a 5, 6, 7, 8, 9, 10, 11, 12, or 13. In some aspects, a subset of compounds of Formula (I) includes those in which when R4 is -(CH2)nQ, -(CH2)nCHQR, -CHQR, or -CQ(R)2, then (i) Q is not -N(R)2 when n is 1, 2, 3, 4, or 5, or (ii) Q is not 5, 6, or 7-membered heterocycloalkyl when n is 1 or 2. In some aspects, another subset of compounds of Formula (I) includes those in which R1 is a C5-30 alkyl, C5-20 alkenyl, -R*YR”, -YR”, or -R”M’R’; R2 and R3 are independently an H, C1-14 alkyl, C2-14 alkenyl, -R*YR”, -YR”, or -R*OR”, and/or R2 and R3, together with the atom to which they are attached, form a heterocycle or carbocycle; R4 is a C3-6 carbocycle, -(CH2)nQ, -(CH2)nCHQR, -CHQR, -CQ(R)2, or unsubstituted C1-6 alkyl, where Q is a C3-6 carbocycle, a 5- to 14-membered heteroaryl having one or more heteroatoms comprising N, O, or S, -OR, -O(CH2)nN(R)2, -C(O)OR, -OC(O)R, -CX3, -CX2H, - CXH2, -CN, -C(O)N(R)2, -N(R)C(O)R, -N(R)S(O)2R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, - CRN(R)2C(O)OR, -N(R)R8, -O(CH2)nOR, -N(R)C(=NR9)N(R)2, -N(R)C(=CHR9)N(R)2, - OC(O)N(R)2, -N(R)C(O)OR, -N(OR)C(O)R, -N(OR)S(O)2R, -N(OR)C(O)OR, -N(OR)C(O)N(R)2, - N(OR)C(S)N(R)2, -N(OR)C(=NR9)N(R)2, -N(OR)C(=CHR9)N(R)2, -C(=NR9)N(R)2, -C(=NR9)R, - C(O)N(R)OR, or a 5- to 14-membered heterocycloalkyl having one or more heteroatoms comprising N, O, and S which is substituted with one or more substituents comprising oxo (=O), OH, amino, mono- or di- alkylamino, or C1-3 alkyl, and/or each n is independently 1, 2, 3, 4, or 5; each R5 is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R6 is independently a C1-3 alkyl, C2-3 alkenyl, or H; M and M' are independently a -C(O)O-, -OC(O)-, -C(O)N(R’)-, -N(R’)C(O)-, -C(O)-, -C(S)- , -C(S)S-, -SC(S)-, -CH(OH)-, -P(O)(OR’)O-, -S(O)2-, -S-S-, an aryl group, or a heteroaryl group; R7 is a C1-3 alkyl, C2-3 alkenyl, or H; R8 is a C3-6 carbocycle or heterocycle; R9 is a H, CN, NO2, C1-6 alkyl, -OR, -S(O)2R, -S(O)2N(R)2, C2-6 alkenyl, C3-6 carbocycle or heterocycle; each R is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R’ is independently a C1-18 alkyl, C2-18 alkenyl, -R* YR”, -YR”, or H; each R” is independently a C3-14 alkyl or C3-14 alkenyl; each R* is independently a C1-12 alkyl or C2-12 alkenyl; each Y is independently a C3-6 carbocycle; each X is independently a F, Cl, Br, or I; and m is 5, 6, 7, 8, 9, 10, 11, 12, or 13, and/or pharmaceutically acceptable salts, tautomers, prodrugs, or stereoisomers thereof. In some aspects, another subset of compounds of Formula (I) includes those in which: R1 is a C5-30 alkyl, C5-20 alkenyl, -R*YR”, -YR”, or -R”M’R’; R2 and R3 are independently an H, C1-14 alkyl, C2-14 alkenyl, -R*YR”, -YR”, or -R*OR”, and/or R2 and R3, together with the atom to which they are attached, form a heterocycle or carbocycle; R4 is a C3-6 carbocycle, -(CH2)nQ, -(CH2)nCHQR, -CHQR, -CQ(R)2, or unsubstituted C1-6 alkyl, where Q is a C3-6 carbocycle, a 5- to 14-membered heterocycle having one or more heteroatoms comprising N, O, or S, -OR, -O(CH2)nN(R)2, -C(O)OR, -OC(O)R, -CX3, -CX2H, - CXH2, -CN, -C(O)N(R)2, -N(R)C(O)R, -N(R)S(O)2R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, - CRN(R)2C(O)OR, -N(R)R8, -O(CH2)nOR, -N(R)C(=NR9)N(R)2, -N(R)C(=CHR9)N(R)2, - OC(O)N(R)2, -N(R)C(O)OR, -N(OR)C(O)R, -N(OR)S(O)2R, -N(OR)C(O)OR, -N(OR)C(O)N(R)2, - N(OR)C(S)N(R)2, -N(OR)C(=NR9)N(R)2, -N(OR)C(=CHR9)N(R)2, -C(=NR9)R, -C(O)N(R)OR, or - C(=NR9)N(R)2, and/or each n is independently 1, 2, 3, 4, or 5; and/or when Q is a 5- to 14- membered heterocycle and (i) R4 is -(CH2)nQ in which n is 1 or 2, or (ii) R4 is -(CH2)nCHQR in which n is 1, or (iii) R4 is -CHQR, and -CQ(R)2, then Q is either a 5- to 14-membered heteroaryl or 8- to 14-membered heterocycloalkyl; each R5 is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R6 is independently a C1-3 alkyl, C2-3 alkenyl, or H; M and M' are independently a -C(O)O-, -OC(O)-, -C(O)N(R')-, -N(R')C(O)-, -C(O)-, -C(S)- , -C(S)S-, -SC(S)-, -CH(OH)-, -P(O)(OR’)O-, -S(O)2-, -S-S-, an aryl group, or a heteroaryl group; R7 is a C1-3 alkyl, C2-3 alkenyl, or H; R8 is C3-6 carbocycle or heterocycle; R9 is H, CN, NO2, C1-6 alkyl, -OR, -S(O)2R, -S(O)2N(R)2, C2-6 alkenyl, C3-6 carbocycle, or heterocycle; each R is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R’ is independently a C1-18 alkyl, C2-18 alkenyl, -R*YR”, -YR”, or H; each R” is independently a C3-14 alkyl or C3-14 alkenyl; each R* is independently a C1-12 alkyl or C2-12 alkenyl; each Y is independently a C3-6 carbocycle; each X is independently a F, Cl, Br, or I; and m is 5, 6, 7, 8, 9, 10, 11, 12, or 13, and/or pharmaceutically acceptable salts, tautomers, prodrugs, or stereoisomers thereof. In some aspects, another subset of compounds of Formula (I) includes those in which: R1 is a C5-30 alkyl, C5-20 alkenyl, -R*YR”, -YR”, or -R”M’R’; R2 and R3 are independently an H, C1-14 alkyl, C2-14 alkenyl, -R*YR”, -YR”, or -R*OR”, and/or R2 and R3, together with the atom to which they are attached, form a heterocycle or carbocycle; R4 is a C3-6 carbocycle, -(CH2)nQ, -(CH2)nCHQR, -CHQR, -CQ(R)2, or unsubstituted C1-6 alkyl, where Q is a C3-6 carbocycle, a 5- to 14-membered heteroaryl having one or more heteroatoms comprising N, O, or S, -OR, -O(CH2)nN(R)2, -C(O)OR, -OC(O)R, -CX3, -CX2H, - CXH2, -CN, -C(O)N(R)2, -N(R)C(O)R, -N(R)S(O)2R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, - CRN(R)2C(O)OR, -N(R)R8, -O(CH2)nOR, -N(R)C(=NR9)N(R)2, -N(R)C(=CHR9)N(R)2, - OC(O)N(R)2, -N(R)C(O)OR, -N(OR)C(O)R, -N(OR)S(O)2R, -N(OR)C(O)OR, -N(OR)C(O)N(R)2, - N(OR)C(S)N(R)2, -N(OR)C(=NR9)N(R)2, -N(OR)C(=CHR9)N(R)2, -C(=NR9)R, -C(O)N(R)OR, or - C(=NR9)N(R)2, and/or each n is independently 1, 2, 3, 4, or 5; each R5 is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R6 is independently a C1-3 alkyl, C2-3 alkenyl, or H; M and M' are independently a -C(O)O-, -OC(O)-, -C(O)N(R')-, -N(R’)C(O)-, -C(O)-, -C(S)- , -C(S)S-, -SC(S)-, -CH(OH)-, -P(O)(OR')O-, -S(O)2-, -S-S-, an aryl group, or a heteroaryl group; R7 is a C1-3 alkyl, C2-3 alkenyl, or H; R8 is a C3-6 carbocycle or heterocycle; R9 is an H, CN, NO2, C1-6 alkyl, -OR, -S(O)2R, -S(O)2N(R)2, C2-6 alkenyl, C3-6 carbocycle, or heterocycle; each R is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R' is independently a C1-18 alkyl, C2-18 alkenyl, -R*YR”, -YR”, or H; each R” is independently a C3-14 alkyl or C3-14 alkenyl; each R* is independently a C1-12 alkyl or C2-12 alkenyl; each Y is independently a C3-6 carbocycle; each X is independently a F, Cl, Br, or I; and m is 5, 6, 7, 8, 9, 10, 11, 12, or 13, and/or pharmaceutically acceptable salts, tautomers, prodrugs, or stereoisomers thereof. In some aspects, another subset of compounds of Formula (I) includes those in which: R1 is a C5-30 alkyl, C5-20 alkenyl, -R*YR”, -YR”, or -R”M’R’; R2 and R3 are independently an H, C2-14 alkyl, C2-14 alkenyl, -R*YR”, -YR”, or -R*OR”, and/or R2 and R3, together with the atom to which they are attached, form a heterocycle or carbocycle; R4 is -(CH2)nQ or -(CH2)nCHQR, where Q is -N(R)2, and/or n is 3, 4, or 5; each R5 is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R6 is independently a C1-3 alkyl, C2-3 alkenyl, or H; M and M’ are independently a -C(O)O-, -OC(O)-, -C(O)N(R')-, -N(R’)C(O)-, -C(O)-, -C(S)- , -C(S)S-, -SC(S)-, -CH(OH)-, -P(O)(OR')O-, -S(O)2-, -S-S-, an aryl group, or a heteroaryl group; R7 is a C1-3 alkyl, C2-3 alkenyl, or H; each R is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R’ is independently a C1-18 alkyl, C2-18 alkenyl, -R*YR”, -YR”, or H; each R” is independently a C3-14 alkyl or C3-14 alkenyl; each R* is independently a C1-12 alkyl or C1-12 alkenyl; each Y is independently a C3-6 carbocycle; each X is independently a F, Cl, Br, or I; and m is 5, 6, 7, 8, 9, 10, 11, 12, or 13, and/or pharmaceutically acceptable salts, tautomers, prodrugs, or stereoisomers thereof. In some aspects, another subset of compounds of Formula (I) includes those in which: R1 is a C5-30 alkyl, C5-20 alkenyl, -R*YR”, -YR”, or -R”M’R’; R2 and R3 are independently a C1-14 alkyl, C2-14 alkenyl, -R*YR”, -YR”, or -R*OR”, and/or R2 and R3, together with the atom to which they are attached, form a heterocycle or carbocycle; R4 is a -(CH2)nQ, -(CH2)nCHQR, -CHQR, or -CQ(R)2, where Q is -N(R)2, and/or n is 1, 2, 3, 4, or 5; each R5 is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R6 is independently a C1-3 alkyl, C2-3 alkenyl, or H; M and M' are independently a -C(O)O-, -OC(O)-, -C(O)N(R')-, -N(R’)C(O)-, -C(O)-, -C(S)- , -C(S)S-, -SC(S)-, -CH(OH)-, -P(O)(OR’)O-, -S(O)2-, -S-S-, an aryl group, or a heteroaryl group; R7 is a C1-3 alkyl, C2-3 alkenyl, or H; each R is independently a C1-3 alkyl, C2-3 alkenyl, or H; each R' is independently a C1-18 alkyl, C2-18 alkenyl, -R*YR”, -YR”, or H; each R” is independently a C3-14 alkyl or C3-14 alkenyl; each R* is independently a C1-12 alkyl or C1-12 alkenyl; each Y is independently a C3-6 carbocycle; each X is independently a F, Cl, Br, or I; and m is 5, 6, 7, 8, 9, 10, 11, 12, or 13, and/or pharmaceutically acceptable salts, tautomers, prodrugs, or stereoisomers thereof. In some aspects, a subset of compounds of Formula (I) includes those of Formula (IA): or a pharmaceutically acceptable salt, tautomer, prodrug, or stereoisomer thereof, wherein I is 1, 2, 3, 4, or 5; m is 5, 6, 7, 8, or 9; M1 is a bond or M’; R4 is unsubstituted C1-3 alkyl, or -(CH2)nQ, in which Q is OH, -NHC(S)N(R)2, -NHC(O)N(R)2, -N(R)C(O)R, -N(R)S(O)2R, - N(R)R8, -NHC(=NR9)N(R)2, -NHC(=CHR9)N(R)2, -OC(O)N(R)2, -N(R)C(O)OR, heteroaryl or heterocycloalkyl; M and M’ are independently a -C(O)O-, -OC(O)-, -C(O)N(R’)-, -P(O)(OR’)O-, - S-S-, an aryl group, or a heteroaryl group; and R2 and R3 are independently a H, C1-14 alkyl, or C2-14 alkenyl. In some aspects, a subset of compounds of Formula (I) includes those of Formula (II): or a pharmaceutically acceptable salt, tautomer, prodrug, or stereoisomer thereof, wherein I is 1, 2, 3, 4, or 5; M1 is a bond or M’; R4 is unsubstituted C1-3 alkyl, or -(CH2)nQ, in which n is 2, 3, or 4, and Q is OH, -NHC(S)N(R)2, -NHC(O)N(R)2, -N(R)C(O)R, -N(R)S(O)2R, -N(R)R8, -NHC(=NR9)N(R)2, -NHC(=CHR9)N(R)2, -OC(O)N(R)2, -N(R)C(O)OR, heteroaryl or heterocycloalkyl; M and M’ are independently a -C(O)O-, -OC(O)-, -C(O)N(R')-, -P(O)(OR’)O-, - S-S-, an aryl group, or a heteroaryl group; and R2 and R3 are independently a H, C1-14 alkyl, or C2-14 alkenyl. In some aspects, a subset of compounds of Formula (I) includes those of Formula (Ila), (lIb), (lIc), or (lIe):
Figure imgf000138_0001
or a pharmaceutically acceptable salt, tautomer, prodrug, or stereoisomer thereof, wherein R4 is as described herein. In some aspects, a subset of compounds of Formula (I) includes those of Formula (IId):
Figure imgf000138_0002
or a pharmaceutically acceptable salt, tautomer, prodrug, or stereoisomer thereof, wherein n is 2, 3, or 4; and m, R’, R”, and R2 through R6 are as described herein. For example, each of R2 and R3 may be independently a C5-14 alkyl or C5-14 alkenyl. In some aspects, an ionizable cationic lipid of the disclosure comprises a compound having structure:
Figure imgf000138_0003
In some aspects, an ionizable cationic lipid of the disclosure comprises a compound having structure:
Figure imgf000139_0001
LNPs described herein can also be generated using the cationic lipids and compositions comprising them as are known in the art, see , e.g., PCT Publication Nos. WO2015/199952, WO2017/004143, WO2017/075531, WO2017/117528, WO2016/176330, WO2018/191657, WO2018/081480, WO2018/107026, WO2018/200943, WO2018/078053, WO2019/036000, WO2019/036028, WO2019/036030, WO2019/036008, WO2020/061426, WO2020/081938, WO2020/146805, WO2021/030701, WO2022/016070, WO2023/114944, WO2023/114937, WO2023/114943, WO2024/054843, and WO2023/250427, all of which are incorporated by reference herein in their entirety for all purposes. In some aspects, an ionizable cationic lipid of the disclosure comprises a compound having structure:
Figure imgf000139_0002
or a pharmaceutically acceptable salt, tautomer, prodrug, or stereoisomer thereof, wherein: one of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, - NRaC(=O)-, -C(=O) =NRa-, NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O-, and the other of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, -NRaC(=O)-, - C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O- or a direct bond; G1 and G2 are each independently unsubstituted C1-C12 alkylene or C1-C12 alkenylene; G is C1-C24 alkylene, C1-C24 alkenylene, C3-C8 cycloalkylene, C3-C8 cycloalkenylene; Ra is H or C1-C12 alkyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is H, OR5, CN, -C(=O)OR4, -OC(=O)R4 or -NR5C(=O)R4; R4 is C1-C12 alkyl; R5 is H or C1- C6 alkyl; and x is 0, 1, or 2. In some of the foregoing aspects, the ionizable cationic lipid comprises a compound having one of the following structures:
Figure imgf000140_0001
or a pharmaceutically acceptable salt, tautomer, prodrug, or stereoisomer thereof, wherein: A is a 3 to 8-membered cycloalkyl or cycloalkylene ring; R6 is, at each occurrence, independently H, OH or C1-C24 alkyl; and n is an integer ranging from 1 to 15. In some of the foregoing aspects, the ionizable cationic lipid comprises a compound having one of the following structures:
Figure imgf000140_0002
or a pharmaceutically acceptable salt, tautomer, prodrug, or stereoisomer thereof, wherein y and z are each independently integers ranging from 1 to 12. In any of the foregoing aspects, one of L1 or L2 is -OCCO)-. For example, in some aspects, each of L1 and L2 are -O(C=O)-. In some aspects of any of the foregoing, L1 and L2 are each independently -(C=O)O- or -O(C=O)-. For example, in some aspects, each of L1 and L2 is - (C=O)O-. In some of the foregoing aspects, the ionizable cationic lipid comprises a compound having one of the following structures:
Figure imgf000140_0003
or a pharmaceutically acceptable salt, tautomer, prodrug, or stereoisomer thereof. In some of the foregoing aspects, the ionizable cationic lipid comprises a compound having one of the following structures:
Figure imgf000141_0001
Or a pharmaceutically acceptable salt, tautomer, prodrug, or stereoisomer thereof. In some of the foregoing aspects, n is an integer ranging from 2 to 12, for example from 2 to 8 or from 2 to 4. For example, in some aspects, n is 3, 4, 5, or 6. In some aspects, n is 3. In some aspects, n is 4. In some aspects, n is 5. In some aspects, n is 6. In some of the foregoing aspects, y and z are each independently an integer ranging from 2 to 10. For example, in some aspects, y and z are each independently an integer ranging from 4 to 9 or from 4 to 6. In some of the foregoing aspects, R6 is H. In other of the foregoing embodiments, R6 is C1-C24 alkyl. In other aspects, R6 is OH. In some embodiments, G is unsubstituted. In other aspects, G3 is substituted. In various different aspects, G3 is linear C1-C24 alkylene or linear C1- C24 alkenylene. In some other foregoing embodiments, R1 or R2, or both, is C6-C24 alkenyl. For example, in some embodiments, R1 and R2 each, independently have the following structure:
Figure imgf000141_0002
wherein: R7a and R7b are, at each occurrence, independently H or C1-C12 alkyl; and a is an integer from 2 to 12, wherein R7a, R7b, and a are each selected such that R1 and R2 each independently comprise from 6 to 20 carbon atoms. For example, in some embodiments a is an integer ranging from 5 to 9 or from 8 to 12. In some of the foregoing aspects, at least one occurrence of R7a is H. For example, in some aspects, R7a is H at each occurrence. In other different aspects of the foregoing, at least one occurrence of R7b is C1-C8 alkyl. For example, in some embodiments, C1-C8 alkyl is methyl, ethyl, n-propyl, iso-propyl, n- butyl, iso-butyl, tert-butyl, n-hexyl, or n-octyl. In different aspects, R1 or R2, or both, has one of the following:
Figure imgf000142_0001
In some of the foregoing aspects, R is OH, CN, -C(=O)OR4 -OC(=O)R4 or -NHC(=O)R4. In some aspects, R4 is methyl or ethyl. It is understood that any aspect of the compounds set forth above, and any specific substituent and/or variable in the compounds set forth above, may be independently combined with other aspects and/or substituents and/or variables of compounds to form aspects of the inventions not specifically set forth above. In addition, in the event that a list of substituents and/or variables is listed for any particular substituent and/or variable in a particular embodiment and/or claim, it is understood that each individual substituent and/or variable may be deleted from the particular aspect and/or claim and that the remaining list of substituents and/or variables will be considered to be within the scope of the disclosure. It is understood that in the present description, combinations of substituents and/or variables of the depicted formulae are permissible only if such contributions result in stable compounds. In some embodiments, the cationic lipid is
Figure imgf000142_0002
In some embodiments, the cationic lipid is
Figure imgf000142_0003
In some aspects, the lipid nanoparticles comprise one or more cationic lipids. In one aspect, the lipid nanoparticles comprise (4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2- hexyldecanoate) (ALC-0315), having the formula:
Figure imgf000143_0001
Exemplary cationic lipids are disclosed in, e.g., U.S. 10,166,298, the full disclosure of which is herein incorporated by reference in its entirety for all purposes. Representative cationic lipids include:
Figure imgf000143_0002
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0002
In yet another aspect, the ionizable cationic lipid is described in PCT Publication No. WO2015/199952, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of Formula (I):
Figure imgf000148_0001
or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: L1 and L2 are each independently -O(C=O)-, -(C=O)O- or a carbon- carbon double bond; R1a and R1b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently methyl or cycloalkyl; R7 is, at each occurrence, independently H or C1-C12 alkyl; R8 and R9 are each independently unsubstituted C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7- membered heterocyclic ring comprising one nitrogen atom; a and d are each independently an integer from 0 to 24; b and c are each independently an integer from 1 to 24; and e is 1 or 2. In other embodiments, the lipid compounds have the following Structure (
Figure imgf000149_0001
(Ia). In another embodiment, the lipid compounds have the following Structure (Ib):
Figure imgf000149_0002
(Ib). In yet other embodiments, the lipid compounds have the following Structure (Ic):
Figure imgf000149_0003
(Ic). Compound Nos. 1-41 as specifically exemplified in Table 1 of PCT Publication No. WO2015/199952, and falling within the generic scope of Structures (I), (Ia), (Ib) or (Ic) described above, are hereby incorporated herein by reference for all purposes. In yet another aspect, the ionizable cationic lipid is described in PCT Publication No. WO2016/176330, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of Formula (I):
Figure imgf000150_0001
or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: L1 and L2 are each independently -O(C=O)-, -(C=O)O- or a carbon carbon double bond; R1a and R1b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently methyl or cycloalkyl; R7 is, at each occurrence, independently H or C1-C12 alkyl; R8 and R9 are each independently unsubstituted C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring comprising one nitrogen atom; a and d are each independently an integer from 0 to 24; b and c are each independently an integer from 1 to 24; and e is 1 or 2. In one embodiment, the ionizable cationic lipid comprises a compound having a structure of Formula (II):
Figure imgf000151_0001
or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: L1 and L2 are each independently -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, -SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, -NRaC(=O)NRa, -OC(=O)NRa-, - NRaC(=O)O-, or a direct bond; G1 is C1-C2 alkylene, -(C=O)- , -O(C=O)-, -SC(=O)-, -NRaC(=O)- or a direct bond; G2 is -C(=O)- , -(C=O)O-, -C(=O)S-, -C(=O)NRa or a direct bond; G3 is C1-C6 alkylene; Ra is H or C1-C12 alkyl; R1a and R1b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4B are, at each occurrence, independently either: (a) H or C]-C12 alkyl; or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently H or methyl; R7 is C4-C20 alkyl; R8 and R9 are each independently C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring; a, b, c and d are each independently an integer from 1 to 24; and x is 0, 1 or 2. In one embodiment, the ionizable cationic lipid comprises a compound having a structure of Formula (III):
Figure imgf000152_0001
or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: one of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, - NRaC(=O)-, -C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O-, and the other of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, -NRaC(=O)-, - C(=O)NRa-, ,NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O- or a direct bond; G1 and G2 are each independently unsubstituted C1-C12 alkylene or C1-C12 alkenylene; G3 is C1-C24 alkylene, C1-C24 alkenylene, C3-C8 cycloalkylene, C3-C8 cycloalkenylene; Ra is H or C1-C12 alkyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is H, OR5, CN, -C(=O)OR4, - OC(=O)R4 or -NR5C(=O)R4; R4 is C1-C12 alkyl; R5 is H or C1-C6 alkyl; and x is 0, 1 or 2. Compound Nos. I-1 to I-41 as specifically exemplified in Table 1, Compound Nos. II-1 to II-34 as specifically exemplified in Table 2, and Compound Nos. III-1 to III-36 as specifically exemplified in Table 3, of PCT Publication No. WO2016/176330, and falling within the generic scope of Structures (I), (II) or (III) described above, are hereby incorporated herein by reference for all purposes. In yet another aspect, the ionizable cationic lipid is described in PCT Publication No. WO2017/004143, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of Formula (I):
Figure imgf000152_0002
or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: L1 and L2 are each independently -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, - C(=O)S-, -SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, -NRaC(=O)NRa-, -OC(=O) Ra-, -NRaC(=O)O- or a direct bond; G1 is C1-C2 alkylene, - (C=O)-, -O(C=O)-, -SC(=O)-, -NRaC(=O)- or a direct bond; G2 is -C(=O)-, -(C=O)O-, -C(=O)S-, -C(=O)NRa- or a direct bond; G3 is C1-C6 alkylene; Ra is H or C1-C12 alkyl; R1a and R1b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either (a): H or C1-C12 alkyl; or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently H or methyl; R7 is C4-C20 alkyl; R8 and R9 are each independently C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring; a, b, c and d are each independently an integer from 1 to 24; and x is 0, 1 or 2. In one embodiment, the cationic lipid comprises a compound having a structure of Formula (IA), (IB), (IC) or (ID):
Figure imgf000153_0001
(IA) (IB)
Figure imgf000154_0001
wherein e, f, g and h are each independently an integer from 1 to 12. In different aspects, Rb is branched C1-C15 alkyl. For
Figure imgf000154_0002
Compound Nos. 1-46 as specifically exemplified in Table 1 of PCT Publication No. WO2017/004143 described above, are hereby incorporated herein by reference for all purposes. In another aspect, the cationic lipid is described in PCT Publication No. WO2017/117528, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, a cationic lipid of the disclosure comprises a compound of Formula (I):
Figure imgf000154_0003
(I) or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof. Compound Nos. 1-3 as specifically exemplified in Table 1 of PCT Publication No. WO2017/117528 described above, are hereby incorporated herein by reference for all purposes. In another aspect, the cationic lipid is described in PCT Publication No. WO2018/191657, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, a cationic lipid of the disclosure comprises a compound of Formula (I):
Figure imgf000155_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: G1 is -OH, - R3R4, -(C=O)NR5 or -NR3(C=O)R5; G2 is -CH2- or -(C=O)-; R is, at each occurrence, independently H or OH; R1 and R2 are each independently optionally substituted branched, saturated or unsaturated C12- C36 alkyl; R3 and R4 are each independently H or optionally substituted straight or branched, saturated or unsaturated C1-C6 alkyl; R5 is optionally substituted straight or branched, saturated or unsaturated C1-C6 alkyl; and n is an integer from 2 to 6. In one embodiment, the cationic lipid comprises a compound having a structure of Formula (IA):
Figure imgf000155_0002
R6 and R7 are, at each occurrence, independently H or straight or branched, saturated or unsaturated C1-C14 alkyl; a and b are each independently an integer ranging from 1 to 15, provided that R6 and a, and R7 and b, are each independently selected such that R1 and R2, respectively, are each independently branched, saturated or unsaturated C12-C36 alkyl. In one embodiment, the cationic lipid comprises a compound having a structure of Formula (IB):
Figure imgf000156_0001
wherein R8, R9, R10 and R11 are each independently straight or branched, saturated or unsaturated C4-C12 alkyl, provided that R8 and R9, and R10 and R11, are each independently selected such that R1 and R2, respectively, are each independently branched, saturated or unsaturated C12-C36 alkyl. In different aspects, R1 and R2 have the following structure:
Figure imgf000156_0002
Compound Nos. 1-17 as specifically exemplified in Table 1 of PCT Publication No. WO2018/191657 described above, are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2018/081480, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, a cationic lipid of the disclosure comprises a compound of Structure (I), (II), (III), (IV) or (IV), or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof. Compound Nos. I-1 to I-41 as specifically exemplified in Table 1, Compound Nos. II-1 to II-46 as specifically exemplified in Table 2, Compound Nos. III-1 to III-49 as specifically exemplified in Table 3, and Compound Nos. IV-1 to IV-3 as specifically exemplified in Table 4 of PCT Publication No. WO2018/081480 described above, are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2018/107026, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, a cationic lipid of the disclosure is selected from compounds having the following Formulas (I, II, III and IV):
Figure imgf000157_0001
or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein, for Formula (I): L1 and L2 are each independently -O(C=O)-, (C=O)O- or a carbon-carbon double bond; R1a and R1b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently methyl or cycloalkyl; R7 is, at each occurrence, independently H or C1-C12 alkyl; R8 and R9 are each independently unsubstituted C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring comprising one nitrogen atom; a and d are each independently an integer from 0 to 24; b and c are each independently an integer from 1 to 24; and e is 1 or 2, for Formula (II): L1 and L2 are each independently -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, - C(=O)S-, -SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, -NRaC(=O)NRa-, -OC(=O)NRa-, -NRaC(=O)O- or a direct bond; G1 is C1-C2 alkylene, -(C=O)-, -O(C=O)-, -SC(=O)-, -NRaC(=O)- or a direct bond; G2 is -C(=O)-, -(C=O)O-, -C(=O)S-, -C(=O)NRa - or a direct bond; G3 is C1-C6 alkylene; Ra is H or C1-C12 alkyl; R1a and R1b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently H or methyl; R7 is C4-C20 alkyl; R8 and R9 are each independently C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring; a, b, c and d are each independently an integer from 1 to 24; and x is 0, 1 or 2; for Formula (III): one of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S- , -C(=O)S-, SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa- - RaC(=O)O-, and the other of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, ,NRaC(=O) Ra-, - OC(=O) Ra- or -NRaC(=O)O- or a direct bond; G1 and G2 are each independently unsubstituted C1-C 12 alkylene or C1-C12 alkenylene; G3 is C1-C24 alkylene, C1-C24 alkenylene, C3-C8 cycloalkylene, C3-C8 cycloalkenylene; Ra is H or C1-C12 alkyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is H, OR5, CN, -C(=O)OR4, -OC(=O)R4 or -NR5C(=O)R4; R4 is C1-C12 alkyl; R5 is H or C1-C6 alkyl; and x is 0, 1 or 2; and for Formula (IV): one of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S- , -C(=O)S-, SC(=O)-, - NRaC(=O)-, -C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa-, -NRaC(=O)O-, and the other of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, -NRaC(=O)-, -C(=O)NRa- ,_-NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O- or a direct bond; G1 and G2 are each independently unsubstituted C1-C12 alkylene or C1-C12 alkenylene; G3 is C1-C24 alkylene, C1-C24 alkenylene, C3-C8 cycloalkylene, C3-C8 cycloalkenylene; Ra is H or C1-C12 alkyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is H, OR5, CN, -C(=O)OR4, -OC(=O)R4 or -NR5C(=O)R4; R4 is C1-C12 alkyl; R5 is H or C1-C6 alkyl; and x is 0, 1 or 2. In one embodiment, the cationic lipid is selected from any one of Compound Nos. I-1 to I- 41 as specifically exemplified in Table 1, Compound Nos. II-1 to II-36 as specifically exemplified in Table 2, Compound Nos. III-1 to III-49 as specifically exemplified in Table 3, and Compound Nos. 1 to 17 as specifically exemplified in Table 4, of PCT Publication No. WO2018/107026, which Tables and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2018/200943, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, a cationic lipid of the disclosure is selected from compounds having the following Structures (I) or (II): (I) (II) or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: L1 is -O(C=O)R1, -(C=O)OR1, -C(=O)R1, -OR1, -S(O)XR1, -S-SR1, -C(=O)SR1, -SC(=O)R1, -NRaC(=O)R1, -C(=O)NRbRc, -NRaC(=O) RbRc, -OC(=O)NRbRc or -NRaC(=O)OR1; L2 is -O(C=O)R2, -(C=O)OR2, -C(=O)R2, -OR2, -S(O)XR2, -S-SR2, -C(=O)SR2, -SC(=O)R2, -NRdC(=O)R2, -C(=O)NReRf, -NRdC(=O)NReRf, -OC(=O)NReRf; -NRdC(=O)OR2 or a direct bond to R2; G1a and G2a are each independently C2-C12 alkylene or C2-C12 alkenylene; G1b and G2b are each independently C1-C12 alkylene or C2-C12 alkenylene; G3 is C1-C24 alkylene, C2-C24 alkenylene, C3-C8 cycloalkylene or C3-C8 cycloalkenylene; Ra, Rb, Rd and Re are each independently H or C1-C12 alkyl or C2-C12 alkenyl; Rc and Rf are each independently C1-C12 alkyl or C2-C12 alkenyl; R1 and R2 are each independently branched C6-C24 alkyl or branched C6-C24 alkenyl; R3a is -C(=O)N(R4a)R5a or -C(=O)OR6; R3b is -NR4bC(=O)R5b; R4a is C1-C12 alkyl; R4b is H, C1-C12 alkyl or C2-C12 alkenyl; R5a is H, C1-C8 alkyl or C2-C8 alkenyl; R5b is C2-C12 alkyl or C2-C12 alkenyl when R4b is H; or R5b is C1-C12 alkyl or C2- C12 alkenyl when R4b is C1-C12 alkyl or C2-C12 alkenyl; R6 is H, aryl or aralkyl; and x is 0, 1 or 2, and wherein each alkyl, alkenyl, alkylene, alkenylene, cycloalkylene, cycloalkenylene, aryl and aralkyl is independently substituted or unsubstituted. In one embodiment, a cationic lipid of the disclosure is selected from compounds having the following Structures (IA) or (IIA):
Figure imgf000161_0001
wherein y1 and z1 are each independently integers ranging from 2 to 12, and y2 and z2 are each independently integers ranging from 1 to 12. In another embodiment, a cationic lipid of the disclosure is selected from compounds having the following Structures (IB), (IC), (ID), (IE), (IIB) or (IIC):
Figure imgf000161_0002
In another embodiment, a cationic lipid of the disclosure is selected from compounds having the following Structures (IF), (IG), (IH), (IJ), (IID) or (IIE):
Figure imgf000161_0003
Figure imgf000162_0001
In different aspects, R1 or R2, or both, independently has one of the following structures:
Figure imgf000162_0002
In a further embodiment, the cationic lipid is selected from any one of Compound Nos. I- 1 to I-19 as specifically exemplified in Table 1, or Compound Nos. II-1 to II-20 as specifically exemplified in Table 2, of PCT Publication No. WO2018/200943, which Tables and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2018/078053, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, a cationic lipid of the disclosure is a compound having Formula (I):
Figure imgf000162_0003
(I) or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein: L1 and L2 are each independently —O(C═O)—, —(C═O)O— or a carbon-carbon double bond; R1a and R1b are, at each occurrence, independently either (a) H or C1-C12, alkyl, or (b) R1a is H or C1-C12, alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either (a) H or C1-C12, alkyl, or (b) R2a is H or C1-C12, alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R1a and R3b are, at each occurrence, independently either (a) H or C1-C12, alkyl, or (b) R1a is H or C1-C12, alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either (a) H or C1-C12, alkyl, or (b) R4a is H or C1-C12, alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently methyl or cycloalkyl; R7 is, at each occurrence, independently H or C1-C12, alkyl; R8 and R9 are each independently C1-C12, alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring comprising one nitrogen atom; a and d are each independently an integer from 0 to 24; b and c are each independently an integer from 1 to 24; and e is 1 or 2. In another embodiment, a cationic lipid of the disclosure is a compound having Formula (II):
Figure imgf000163_0001
or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein: L1 and L2 are each independently —O(C═O)—, —(C═O)O—, —C(═O)—, —O—, —S— S—, —C(═O)S—, —SC(═O)—, —NRaC(═O)—, —C(═O)NRa—, —NRaC(═O)NRa—, — OC(═O)NRa—, —NRaC(═O)O—, or a direct bond; G1 is C1-C2 alkylene, —(C═O)—, —O(C═O)—, —SC(═O)—, —NRaC(═O)— or a direct bond G2 is —C(═O)—, —(C═O)O—, —C(═O)S—, —C(═O)NRa— or a direct bond; G3 is C1-C6 alkylene; Ra is, at each occurrence, independently H or C1-C12 alkyl; R1a and R16 are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R1a is H or C1-C12, alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either: (a) H or C1-C12, alkyl; or (b) R2a is H or C1-C12, alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R1a and R3b are, at each occurrence, independently either: (a) H or C1-C12, alkyl; or (b) R1a is H or C1-C12, alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either: (a) H or C1-C12, alkyl; or (b) R4a is H or C1-C12, alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently H or methyl; R7 is C4-C20 alkyl; R8 and R9 are each independently C1-C12, alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring; a, b, c and d are each independently an integer from 1 to 24; and x is 0, 1 or 2. In another embodiment, a cationic lipid of the disclosure is a compound having Formula (III):
Figure imgf000164_0001
or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein: L1 and L2 are each independently —O(C═O)—, —(C═O)O—, —C(═O)—, —O—, — S(O)x—, —S—S—, —C(═O)S—, —SC(═O)—, —NRaC(═O)—, —C(═O)NRa—, — NRaC(═O)NRa—, —OC(═O)NRa— or —NRaC(═O)O—; G1 and G2 are each independently unsubstituted C1-C12 alkylene or C1-C12 alkenylene; G3 is C1-C24 alkylene, C1-C24 alkenylene, C3-C8 cycloalkylene, or C3-C8 cycloalkenylene; Ra is, at each occurrence, independently H or C1-C12, alkyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is OR5, CN, —C(═O)OR4, —OC(═O)R4 or —NR5C(═O)R4; R4 is C1-C12 alkyl; R5 is H or C1-C6 alkyl; and x is 0, 1 or 2. In a further embodiment, the cationic lipid is selected from any one of Compound Nos. I- 1 to I-41 as specifically exemplified in Table 7, or Compound Nos. II-1 to II-36 as specifically exemplified in Table 8, or Compound Nos. III-1 to III-36 as specifically exemplified in Table 9, of PCT Publication No. WO2018/078053, which Tables and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2019/036000, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of Formula (I):
Figure imgf000165_0001
or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: L1 and L2 are each independently —O(C═O)—, —(C═O)O—, —C(═O)—, —O—, — S(O)x—, —S—S—, —C(═O)S—, —SC(═O)—, —NRaC(═O)—, —C(═O)NRa—, — NRaC(═O)NRa—, —OC(═O)NRa—, —NRaC(═O)O— or a direct bond; G1 is C1-C2 alkylene, —(C═O)—, —O(C═O)—, —SC(═O)—, —NRaC(═O)— or a direct bond; G2 is —C(═O)—, —(C═O)O—, —C(═O)S—, —C(═O)NRa— or a direct bond; G3 is C1-C6 alkylene; Ra is H or C1-C12 alkyl; R1a and R1b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R′b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either (a): H or C1-C12 alkyl; or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently H or methyl; R7 is H or C1-C20 alkyl; R8 is OH, —N(R9)(C═O)R10, —(C═O)NR9R10, —NR9R10, —(C═O)OR11 or —O(C═O)R11, provided that G3 is C4-C6 alkylene when R8 is —NR9R10, R9 and R10 are each independently H or C1-C12 alkyl; R11 is aralkyl; a, b, c and d are each independently an integer from 1 to 24; and x is 0, 1 or 2, wherein each alkyl, alkylene and aralkyl is optionally substituted. In another embodiment, a cationic lipid of the disclosure is a compound having the following structures (IA) or (IB):
Figure imgf000166_0001
In another embodiment, a cationic lipid of the disclosure is a compound having the following structures (IC) or (ID):
Figure imgf000167_0001
wherein e, f, g and h are each independently an integer from 1 to 12. In a further embodiment, the cationic lipid is selected from any one of Compound Nos.1- 37 as specifically exemplified in Table 1 of PCT Publication No. WO2019/036000, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2019/036028, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of structure (I):
Figure imgf000167_0002
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: X and X′ are each independently N or CR; Y and Y′ are each independently absent, —O(C═O)—, —(C═O)O— or NR, provided that: a) Y is absent when X is N; b) Y′ is absent when X′ is N; c) Y is —O(C═O)—, —(C═O)O— or NR when X is CR; and d) Y′ is —O(C═O)—, —(C═O)O— or NR when X′ is CR, L1 and L1′ are each independently —O(C═O)R1, —(C═O)OR1, —C(═O)R1, —OR1, — S(O)zR1, —S—SR1, —C(═O)SR1, —SC(═O)R1, —NRaC(═O)R1, —C(═O)NRbRc, — NRaC(═O)NRbRc, —OC(═O)NRbRc or —NRaC(═O)OR1; L2 and L2′ are each independently —O(C═O)R2, —(C═O)OR2, —C(═O)R2, —OR2, — S(O)zR2, —S—SR2, —C(═O)SR2, —SC(═O)R2, —NRdC(═O)R2, —C(═O)NReRf, — NRdC(═O)NReRf, —OC(═O)NReRf; —NRdC(═O)OR2 or a direct bond to R2; G1, G1′, G2 and G2′ are each independently C4-C8 alkylene or C2-C8 alkenylene; G3 is C2-C24 heteroalkylene or C2-C24 heteroalkenylene; Ra, Rb, Rd and Re are, at each occurrence, independently H, C1-C12 alkyl or C2-C12 alkenyl; Re and Rf are, at each occurrence, independently C1-C12 alkyl or C2-C12 alkenyl; R is, at each occurrence, independently H or C1-C12 alkyl; R1 and R2 are, at each occurrence, independently branched C6-C24 alkyl or branched C6- C24 alkenyl; z is 0, 1 or 2, and wherein each alkyl, alkenyl, alkylene, alkenylene, heteroalkylene and heteroalkenylene is independently substituted or unsubstituted unless otherwise specified. In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IA), (IB), (IC), (ID), (IE), (IF), (IG) or (IH):
Figure imgf000169_0001
wherein Rd is, at each occurrence, independently H or optionally substituted C1-C6 alkyl. In a further embodiment, the cationic lipid is selected from any one of Compound Nos.1- 11 as specifically exemplified in Table 1 of PCT Publication No. WO2019/036028, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2019/036030, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of structure (I): or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: one of L1 or L2 is —O (C═O)—, —(C═O)O—, —C(═O)—, —O—, —S—S—, —C(═O)S— , —SC(═O)—, —NRaC(═O)—, —C(═O)NRa—, NRaC(═O)NRa—, —OC(═O)NRa— or — NRaC(═O)O—, and the other of L1 or L2 is —O(C═O)—, —(C═O)O—, —C(═O)—, —O—, — S(O)x—, —S—S—, —C(═O)S—, SC(═O)—, —NRaC(═O)—, —C(═O)NRa—NRaC(═O)NRa—, —OC(═O)NRa— or —NRaC(═O)O— or a direct bond; G1 and G2 are each independently unsubstituted C1-C12 alkylene or C1-C12 alkenylene; G3 is C1-C24 alkylene, C1-C24 alkenylene, C3-C8 cycloalkylene, C3-C8 cycloalkenylene; Ra is H or C1-C12 alkyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is H, OR S , CN, —C(═O)OR4, —OC(═O)R4 or —NR5 C(═O)R4; R4 is C1-C12 alkyl; R5 is H or C1-C6 alkyl; and x is 0, 1 or 2. In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IA), (IB), (IC) or (ID):
Figure imgf000171_0001
In a further embodiment, the cationic lipid is selected from any one of Compound Nos.1- 12 as specifically exemplified in Table 1 of PCT Publication No. WO2019/036030, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2019/036008, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of structure (I):
Figure imgf000171_0002
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: L1 is —O(C═O)R1, —(C═O)OR1, —C(═O)R1, —OR1, —S(O)xR1, —S—SR1, —C(═O)SR1, —SC(═O)R1, —NRaC(═O)R1, —C(═O)NRbRc, —NRaC(═O)NRbRc, —OC(═O)NRbRc or — NRaC(═O)OR1; L2 is —O(C═O)R2, —(C═O)OR2, —C(═O)R2, —OR2, —S(O)xR2, —S—SR2, —C(═O)SR2, —SC(═O)R2, —NRdC(═O)R2, —C(═O)NReRf, —NRdC(═O)NReRf, —OC(═O)NReRf; — NRdC(═O)OR2 or a direct bond to R2; G1 and G2 are each independently C2-C12 alkylene or C2-C12 alkenylene; G3 is C1-C24 alkylene, C2-C24 alkenylene, C3-C8 cycloalkylene or C3-C8 cycloalkenylene; Ra, Rb, Rd and Re are each independently H, C1-C12 alkyl or C1-C12 alkenyl; Rc and Rf are each independently C1-C12 alkyl or C2-C12 alkenyl; R1 and R2 are each independently branched C6-C24 alkyl or branched C6-C24 alkenyl; R3 is —N(R4)R5; R4 is C1-C12 alkyl; R5 is substituted C1-C12 alkyl, wherein R5 is substituted with one or more substituents selected from the group consisting of —ORg, —NRgC(═O)Rh, —C(═O)NRgRh, —C(═O)Rh, — OC(═O)Rh, —C(═O)ORh and —ORiOH, wherein: Rg is, at each occurrence independently H or C1-C6 alkyl; Rh is at each occurrence independently C1-C6 alkyl; and Ri is, at each occurrence independently C1-C6 alkylene; and x is 0, 1 or 2, and wherein each alkyl, alkenyl, alkylene, alkenylene, cycloalkylene and cycloalkenylene is independently substituted or unsubstituted unless otherwise specified. In another embodiment, a cationic lipid of the disclosure is a compound having the following structures (IA),
Figure imgf000172_0001
wherein y and z are each independently integers ranging from 2 to 12. In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IB), (IC), (ID) or (IE):
Figure imgf000173_0001
In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IF), (IG), (IH) or (IJ):
Figure imgf000174_0001
wherein y and z are each independently integers ranging from 2-12. In a further embodiment, the cationic lipid is selected from any one of Compound Nos.1- 18 as specifically exemplified in Table 1 of PCT Publication No. WO2019/036008, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2020/081938, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of structure (I):
Figure imgf000174_0002
or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: G1 is —N(R3)R4 or —OR′; R1 is optionally substituted branched, saturated or unsaturated C12-C36 alkyl; R2 is optionally substituted branched or unbranched, saturated or unsaturated C12- C36 alkyl when L is —C(═O)—; or R2 is optionally substituted branched or unbranched, saturated or unsaturated C4-C36 alkyl when L is C6-C12 alkylene, C6-C12 alkenylene, or C2-C6 alkynylene; R3 and R4 are each independently H, optionally substituted branched or unbranched, saturated or unsaturated C1-C6 alkyl; or R3 and R4 are each independently optionally substituted branched or unbranched, saturated or unsaturated C1-C6 alkyl when L is C6-C12 alkylene, C6- C12 alkenylene, or C2-C6 alkynylene; or R3 and R4, together with the nitrogen to which they are attached, join to form a heterocyclyl; R5 is H or optionally substituted C1-C6 alkyl; L is —C(═O)—, C6-C12 alkylene, C6-C12 alkenylene, or C2-C6 alkynylene; and n is an integer from 1 to 12. In another embodiment, a cationic lipid of the disclosure is a compound having the following structure (IA):
Figure imgf000175_0001
wherein R8 and R9 are each independently H or optionally substituted branched or unbranched, saturated or unsaturated C2-C12 alkyl, provided that R8 and R9 are each independently selected such that R1 is optionally substituted branched, saturated or unsaturated C12-C36 alkyl; and R10 and R11 are each independently H or optionally substituted branched or unbranched, saturated or unsaturated C2-C12 alkyl, provided that R10 and R11 are each independently selected such that: R2 is optionally substituted branched or unbranched, saturated or unsaturated C12- C36 alkyl when L is —C(═O)—; and R2 is optionally substituted branched or unbranched, saturated or unsaturated C4-C36 alkyl when L is C6-C12 alkylene, C6-C12 alkenylene, or C2- C6 alkynylene. In a further embodiment, the cationic lipid is selected from any one of Compound Nos. I- 1 to I-22 as specifically exemplified in Table 1 of PCT Publication No. WO2020/081938, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2020/146805, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of structure (I):
Figure imgf000175_0002
or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: R1 is optionally substituted C1-C24 alkyl or optionally substituted C2-C24 alkenyl; R2 and R3 are each independently optionally substituted C1-C36 alkyl; R4 and R5 are each independently optionally substituted C1-C6 alkyl, or R4 and R5 join, along with the N to which they are attached, to form a heterocyclyl or heteroaryl; L1, L2, and L3 are each independently optionally substituted C I-C I X alkylene; G1 is a direct bond, -(CH2)n0(C=0)-, -(CH2)n(C=0)0-, or -(C=0)-; G2 and G3 are each independently -(C=0)0- or -0(C=0)-; and n is an integer greater than 0. In another embodiment, a cationic lipid of the disclosure is a compound having the following structure (IA):
Figure imgf000176_0001
. In another embodiment, a cationic lipid of the disclosure is a compound having the following structure (IB):
Figure imgf000176_0002
In a further embodiment, the cationic lipid is selected from any one of Compound Nos. I- 1 to I-40 as specifically exemplified in Table 1 of PCT Publication No. WO2020/146805, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2022/016070, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of structure (I):
Figure imgf000177_0001
or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein: G1 and G2 are each independently C1-C6 alkylene; L1 and L2 are each independently —O(C═O)— or —(C═O)O—; R1a and R1b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either (a): H or C1-C12 alkyl; or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently H or methyl; R7 is —O(C═O)R10, —(C═O)OR10, —NR9(C═O)R10 or —(C═O)NR9R10; R8 is OH, —N(R11)(C═O)R12, —(C═O)NR11R12, —NR11R12, —(C═O)OR′2 or — O(C═O)R′2; R9 is H or C1-C15 alkyl; R10 is C1-C15 alkyl; R11 is H or C1-C6 alkyl; R12 is C1-C6 alkyl; X is —(C═O)— or a direct bond; and a, b, c and d are each independently an integer from 1 to 24; wherein each methyl, alkyl and alkylene is independently optionally substituted. In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IA) or (IB):
Figure imgf000178_0001
or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof. In a further embodiment, the cationic lipid is selected from any one of Compound Nos. I- 1 to I-23 as specifically exemplified in Table 1 of PCT Publication No. WO2022/016070, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2023/114944, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of structure (I):
Figure imgf000178_0002
or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: L1 is - O(C=O)R1a, -(C=O)OR1a , -C(=O)R1a , -OR1a , -S(O)xR1a , -S-SR1a , - C(=O)SR1a , -SC(=O)R1a , - NRaC(=O)R1a , -C(=O)NRaR1a , -NRaC(=O)NRaR1a , - OC(=O)NRaR1a, -NRaC(=O)OR1a or R1b; L2 is -O(C=O)R4a , -(C=O)OR4a , -C(=O)R4a , -OR4a , -S(O)xR4a , -S-SR4a , - C(=O)SR4a , - SC(=O)R4a , -NRaC(=O)R4a , -C(=O)NRaR4a , -NRaC(=O)NRaR4a , - OC(=O)NRaR4a , - NRaC(=O)OR4a, or R4b; G1 is C1-C2 alkylene, - (C=O)-, -O(C=O)-, -SC(=O)-, -NRaC(=O)- or a direct bond; G2 is –C(=O)-, -(C=O)O-, -C(=O)S-, -C(=O)NRa- or a direct bond; G3 is C1- C6 alkylene; Ra is H or C1-C12 alkyl; R1a and R4a are each independently branched C6-C24 alkyl, branched C6-C24 alkenyl, branched C6-C24 fluoroalkyl, branched C6-C24 fluoroalkenyl, C6-C24 alkylacetal or C6-C24 fluoroalkylacetal; R1b and R4b are each independently –CH(OR)(OR), wherein each R is independently linear or branched C6-C18 alkyl, linear or branched C6- C18 alkenyl, linear or branched C6-C18 fluoroalkyl, or linear or branched C6-C18 fluoroalkenyl R2a and R2b are, at each occurrence, independently H, F, C1-C12 alkyl, or C1-C12 fluoroalkyl; R3a and R3b are, at each occurrence, independently H, F, C1-C12 alkyl, or C1-C12 fluoroalkyl; R7 is H, C4-C20 alkyl, or C2-C10 fluoroalkyl; R8 and R9 are each independently C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring; b and c are each independently an integer from 1 to 24; and wherein at least one of R2a, R2b, R3a, and R3b is F or C1-C12 fluoroalkyl; at least one of R1a and R4a is present and selected from branched C6-C24 fluoroalkyl, branched C6-C24 fluoroalkenyl and C6- C24 fluoroalkylacetal; at least one of R1b and R4b is present and selected from linear or branched C6-C18 fluoroalkyl and linear or branched C6-C18 fluoroalkenyl; G3 is C1-C6 fluoroalkylene; and/or R7 is C2-C10 fluoroalkyl. In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IA) or (IB):
Figure imgf000179_0001
. In a further embodiment, the cationic lipid is selected from any one of Compound Nos. I- 1 to I-25 as specifically exemplified in Table 1 of PCT Publication No. WO2023/114944, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2023/114937, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of structure (I):
Figure imgf000179_0002
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: L1 is -O(C=O)Rla, -(C=O)ORla , -C(=O)Rla , -ORla , -S(O)xRla , -S-SRla , - C(=O)SRla , - SC(=O)Rla , -NRaC(=O)Rla , -C(=O)NRaRla , -NRaC(=O)NRaRla , - OC(=O)NRaRla, - NRaC(=O)ORla or Rlb; L2 is -O(C=O)R2a , -(C=O)OR2a , -C(=O)R2a , -OR2a , -S(O)xR2a , -S-SR2a , - C(=O)SR2a , - SC(=O)R2a , -NRaC(=O)R2a , -C(=O)NRaR2a , -NRaC(=O)NRaR2a , - OC(=O)NRaR2a , - NRaC(=O)OR2a, or R2b; G1 and G2 are each independently linear or branched C1-C12 alkylene or linear or branched C1-C12 fluoroalkylene; G3 is linear or branched C1-C12 alkylene or linear or branched C1-C12 fluoroalkylene; each Ra is independently H or C1-C12 alkyl; Rla and R2a are each independently branched C6-C24 alkyl, branched C6-C24 alkenyl, branched C6-C24 fluoroalkyl, branched C6-C24 fluoroalkenyl, C6-C24 alkylacetal or C6-C24 fluoroalkylacetal; Rlb and R2b are each independently -CH(OR)(OR), wherein each R is independently linear or branched Ce-Cis alkyl, linear or branched Ce-Cis alkenyl, linear or branched Ce-Cis fluoroalkyl, or linear or branched Ce-Cis fluoroalkenyl; R3 is H, -OR5, -CN, -C(=O)OR4, -OC(=O)R4, -N(R5)N4, -C(=O)N(R4)R5, or - NR5C(=O)R4; and R4 is H, C1-C12 alkyl, or aryl, and R5 is H or Ci-Ce alkyl; or R4 and R5, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring, and 66 wherein at least one of G1 and G2 is linear or branched C1-C12 fluoroalkylene; G3 is linear or branched C1-C12 fluoroalkylene; at least one of Rla and R2a is present and selected from branched C6-C24 fluoroalkyl, branched C6-C24 fluoroalkenyl and C6-C24 fluoroalkylacetal; and/or at least one of Rlb and R2b is present and selected from linear or branched Ce-Cis fluoroalkyl and linear or branched Ce-Cis fluoroalkenyl. In another embodiment, a cationic lipid of the disclosure is a compound having the following structure (IA):
Figure imgf000180_0001
wherein: R6 is, at each occurrence, independently H, F, OH or C1-C24 alkyl; and n is an integer ranging from 1 to 15. In another embodiment, a cationic lipid of the disclosure is a compound having the following structure (IB):
Figure imgf000181_0001
wherein: y and z are each independently integers ranging from 1 to 12; and R7 is, at each occurrence, independently H or F. In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IC) or (ID):
Figure imgf000181_0002
In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IE) or (IF):
Figure imgf000181_0003
(IE) (IF) In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IG) or (IH):
Figure imgf000181_0004
(IG) (IH) wherein R11 and R12 are each independently C1-C12 alkyl; or R11 and R12, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring comprising one nitrogen atom. In a further embodiment, the cationic lipid is selected from any one of Compound Nos. I- 1 to I-10 as specifically exemplified in Table 1 of PCT Publication No. WO2023/114937, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the cationic lipid is described in PCT Publication No. WO2023/114943, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of structure (I):
Figure imgf000182_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: L1 is - NRaC(=O)R1 or -C(=O)NRbRc; L2 is -NRdC(=O)R2 or -C(=O)NReRf; G1 and G2 are each independently C2-C12 alkylene, or C2-C12 alkenylene; G3 is C1-C24 alkylene, or C2-C24 alkenylene; Ra, Rb, Rd and Re are each independently H, C1-C16 alkyl or C2-C16 alkenyl; Rc and Rf are each independently C1-C16 alkyl or C2-C16 alkenyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is H, -OH, CN, -N(R4)R5; -C(=O)N(R4)R5; - N(R4)C(=O)R5; -N(R4)C(=O)OR5;- C(=O)OR6, -OC(=O)R6, -OR7, heteroaryl or aryl; R4 and R5 are each independently is H, C1-C12 alkyl, C3-C6 cycloalkyl or C3-C6 cycloalkenyl, or R4 and R5, together with the nitrogen or carbon atom to which they are bound, form a 5 to 7-membered heterocyclic ring; R6 is H, C1-C12 alkyl, C2-C12 alkenyl or aralkyl; R7 is C1-C12 alkyl optionally substituted with hydroxyl or alkoxy; and wherein each alkyl, alkenyl, alkylene, alkenylene, aryl and aralkyl is independently substituted or unsubstituted unless otherwise specified. In another embodiment, a cationic lipid of the disclosure is a compound having the following structure (IA):
Figure imgf000182_0002
wherein y and z are each independently integers ranging from 2 to 12. In another embodiment, a cationic lipid of the disclosure is a compound having any one of the following structures (IB) or (IC):
Figure imgf000182_0003
(IB) (IC) In a further embodiment, the cationic lipid is selected from any one of Compound Nos. I- 1 to I-85 as specifically exemplified in Table 1 of PCT Publication No. WO2023/114943, which Table and compounds listed therein are hereby incorporated herein by reference for all purposes. In yet another aspect, the ionizable cationic lipid is described in PCT Publication No. WO2024/054843, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound of Structure (I):
Figure imgf000183_0001
or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: L1a and L1b are each independently optionally substituted C3-C12 alkyl; R1a is -C(=O)OR4a or -O(C=O)R4a; R1b is -C(=O)OR4b or -O(C=O)R4b; R2 is -NR6(C=O)R5, -(C=O)N(R6)R5 or -(C=O)OR7; R3 and R6 are each independently hydrogen or optionally substituted C1-C6 alkyl; R4a, R4b, and R5 are each independently optionally substituted alkyl; R7 is optionally substituted arylalkyl; n1 is 2, 3, 4, 5, or 6; and X is C2-C6 alkylene or C4-C20 alkyleneoxide. One embodiment provides a compound having Structure (I):
Figure imgf000183_0002
or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: L1a and L1b are each independently optionally substituted C3-C12 alkyl; R1a is -C(=O)OR4a or -O(C=O)R4a; R1b is -C(=O)OR4b or -O(C=O)R4b; R2 is -NR6(C=O)R5, -(C=O)N(R6)R5 or -(C=O)OR7; R3 and R6 are each independently hydrogen or optionally substituted C1-C12 alkyl; R4a, R4b, and R5 are each independently optionally substituted alkyl; R7 is optionally substituted C1-C6 alkyl or optionally substituted arylalkyl; n1 is 2, 3, 4, 5, or 6; and X is C2-C6 alkylene or C4-C20 alkyleneoxide. In some embodiments, wherein X is:
Figure imgf000184_0001
wherein: n2 is 2, 3, 4, 5, or 6; n3 is 0, 1, 2, 3, or 4; n4 is 2, 3, or 4; and n5 is 2, 3, 4, or 5. In some embodiments, L1a is C5-C9 alkyl. In certain embodiments, L1b is C5-C9 alkyl. In some embodiments, L1a is C5-, C6-, C7-, or C9-alkyl. In certain embodiments, L1b is C5-, C6-, C7-, or C9-alkyl. In some embodiments, L1a is C5-alkyl. In certain embodiments, L1a is C6-alkyl. In some embodiments, L1a is C7-alkyl. In certain embodiments, L1a is C9-alkyl. In some embodiments, L1b is C5-alkyl. In certain embodiments, L1b is C6-alkyl. In some embodiments, L1b is C7-alkyl. In certain embodiments, L1b is C9-alkyl. In some embodiments, L1a is unsubstituted. In certain embodiments, L1b is unsubstituted. In some embodiments, L1a is unbranched. In certain embodiments, L1b is unbranched. In some embodiments, one of R1a is -O(C=O)R4a. In certain embodiments, one of R1a is - (C=O)OR4a. In some embodiments, R1b is -O(C=O)R4b. In certain embodiments, one of R1b is - (C=O)OR4b. In some embodiments, R4a is C8-C24-alkyl. In certain embodiments, R4a is C10-C18-alkyl. In certain embodiments, R4a is C11-C16-alkyl. In some embodiments, R4a is C11-alkyl. In certain embodiments, R4a is C15-alkyl. In some embodiments, R4a isC16-alkyl. In certain embodiments, R4b is C8-C24-alkyl. In some embodiments, R4b is C10-C18-alkyl. In certain embodiments, R4b is C11-C16-alkyl. In some embodiments, R4b is C11-alkyl. In certain embodiments, R4b is C15-alkyl. In some embodiments, R4b is C16-alkyl. In certain embodiments, R4a is branched. In some embodiments, R4b is branched. In certain embodiments, R4a is unsubstituted. In some embodiments, R4b is unsubstituted. In certain embodiments, R4a has one of the following structures:
Figure imgf000185_0001
In some embodiments, R4b has one of the following structures:
Figure imgf000185_0002
In some embodiments, R2 is -NR6(C=O)R5. In certain embodiments, R2 is -(C=O)N(R6)R5. In some embodiments, R5 is C2-C16-alkyl. In certain embodiments, R5 is C4-C13-alkyl. In some embodiments, R5 is C4-, C7-, C8-, C10-, or C13-alkyl. In certain embodiments, R5 is unsubstituted. In some embodiments, R5 is substituted with hydroxyl. In some embodiments, R5 is branched. In certain embodiments, R5 is unbranched. In some embodiments, R5 has one of the following structures:
Figure imgf000185_0003
In certain embodiments, R5 is unbranched. In some embodiments, R5 has one of the following structures:
Figure imgf000186_0001
In some embodiments, R6 is C1-C6 alkyl. In some embodiments, R6 is C1-C10 alkyl. In certain embodiments, R6 is C1-C4-alkyl. In some embodiments, R6 is C1-, C2-, C3-, C6-, C8-, or C10-alkyl. In certain embodiments, methyl, ethyl, n-butyl, n-hexyl, n-octyl, or n-decyl. In some embodiments, R6 is unbranched. In certain embodiments, R6 is methyl or n-butyl. In some embodiments, R6 is unsubstituted. In some embodiments, R6 is substituted. In some embodiments, R6 is C1-C6 alkyl substituted with one or more hydroxyl. In some embodiments, R6 is C2-, C3-, C4-, or C6- alkyl substituted with one or more hydroxyl. In certain embodiments, R6 is hydrogen. In some embodiments, R2 is -(C=O)OR7. In some embodiments, R7 is C1-C3 alkyl or C7-C16 arylalkyl. In certain embodiments, R7 is C7-C16 arylalkyl. In some embodiments, R7 is C1-C3 alkyl. In some embodiments, R7 is unsubstituted. In certain embodiments, R7 is -CH3 or has the following structure:
Figure imgf000186_0002
In certain embodiments, R7 has the following structure:
Figure imgf000186_0003
In some embodiments, R3 is optionally substituted C1-C6 alkyl. In certain embodiments, R3 is optionally substituted methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, n-hexyl. In some embodiments, R3 is optionally substituted methyl. In some embodiments, R3 is C1-C6 alkyl substituted with one or more hydroxyl. In some embodiments, R3 is C2- or C4- alkyl substituted with one or more hydroxyl. In certain embodiments, R3 is unsubstituted. In some embodiments, R3 is hydrogen. In some embodiments, X is
Figure imgf000186_0004
For example, in certain aspects the compound has the following Structure (II):
Figure imgf000187_0001
or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. For example, in some of these embodiments n2 is 3, 4, or 5. In other embodiments X is
Figure imgf000187_0002
In some such embodiments, the compound has the following Structure (III):
Figure imgf000187_0003
or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. In different of these embodiments, n3 is 0 or 1. In other embodiments, n4 is 2 or 3. In some other different embodiments, n5 is 3. In some embodiments, n1 is 3, 4, or 5. In certain embodiments, n1 is 2. In some embodiments, the compound has one of the structures set forth in the Table below or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof:
Figure imgf000187_0004
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
 The names of exemplary Compounds I-1 to I-53 set forth in the Table above are listed below:
Figure imgf000195_0001
Figure imgf000196_0002
In a further aspect, the ionizable cationic lipid is described in PCT Publication No. WO2024/233387, the contents of which is hereby incorporated herein by reference in its entirety for all purposes. In one embodiment therein, an ionizable cationic lipid of the disclosure comprises a compound having the following structure (I):
Figure imgf000196_0001
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: A is a 3-10 membered carbocyclic or oxygen-containing heterocyclic ring optionally substituted with one or more fluoro, hydroxyl, C1-C6 alkyl, or C1-C6 alkylhydroxyl substituents; R1 is −NRaC(=O)R3 or −C(=O)NRbRc; R2 is −NRdC(=O)R4 or −C(=O)NReRf; R3 and R4 are each independently C6-C24 alkyl or C6-C24 alkenyl; Ra, Rb, Rd and Re are each independently H, C1-C20 alkyl or C2-C20 alkenyl; Rc and Rf are each independently C1-C20 alkyl or C2-C20 alkenyl; L1 and L2 are each independently a direct bond or C1-C6 alkylene; and L2a and L2b are each independently C4-C12 alkylene; wherein each alkyl, alkylene, and alkenyl is optionally substituted with one or more fluoro. In a further embodiment, the cationic lipid is selected from any one of Compound Nos. I- 1 to I-63 as specifically exemplified in Table 1 of PCT Publication No. WO2024/233387, including any racemic mixtures of enantiomer pairs of such compounds as described therein, which Table, compounds and racemic mixtures thereof listed therein are hereby incorporated herein by reference for all purposes. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing cationic lipids may be excluded from the LNPs of the present disclosure. In some aspects, the RNA-LNPs comprise a cationic lipid, a RNA molecule as described herein and one or more of neutral lipids, steroids, pegylated lipids, or combinations thereof. If more than one cationic lipid is incorporated within the LNP, such percentages apply to the combined cationic lipids. In one aspect, the cationic lipid is present in the LNP in an amount such as at least, at most, exactly, or between (inclusive or exclusive) of, or about 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59 or 60 mole percent (mol%). In some aspects, two or more cationic lipids are incorporated within the LNP. If more than one cationic lipid is incorporated within the LNP, the foregoing percentages apply to the combined cationic lipids. In some aspects of the disclosure the LNP comprises a combination or mixture of any the lipids described above. ii. POLYMER CONJUGATED LIPID In some aspects, the LNPs comprise a polymer conjugated lipid. The term “polymer conjugated lipid” refers to a molecule comprising both a lipid portion and a polymer portion. An example of a polymer conjugated lipid is a pegylated lipid (e.g., polyethylene glycol-lipid, PEG- lipid). In certain aspects, the LNP comprises an additional, stabilizing lipid that is a pegylated lipid. The term “pegylated lipid” refers to a molecule comprising both a lipid portion and a polyethylene glycol portion. Pegylated lipids are known in the art and include, but are not limited to, PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramides (e.g., PEG-CerC14 or PEG-CerC20), PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, 2-[(polyethylene glycol)-2000]-N,N-ditetradecylacetamide, and mixtures thereof. Representative polyethylene glycol-lipids include PEG-c-DOMG, PEG-c-DMA, PEG-DSG, PEG-DPG, and PEG-s-DMG (1-(monomethoxy-polyethyleneglycol)-2,3- dimyristoylglycerol). In one aspect, the polyethylene glycol-lipid is N-[(methoxy polyethylene glycol)2000)carbamoyl]-1,2-dimyristyloxlpropyl-3-amine (PEG-c-DMA). In one aspect, the polyethylene glycol-lipid is PEG-2000-DMG. In one aspect, the polyethylene glycol-lipid is PEG- c-DOMG. In other aspects, the LNPs comprise a PEGylated diacylglycerol (PEG-DAG) such as 1-(monomethoxy-polyethyleneglycol)-2,3-dimyristoylglycerol (PEG-DMG), a PEGylated phosphatidylethanolamine (PEG-PE), a PEG succinate diacylglycerol (PEG-S-DAG) such as 4- O-(2′,3′-di(tetradecanoyloxy)propyl-1-O-((O-methoxy(polyethoxy)ethyl)butanedioate (PEG-S- DMG), a PEGylated ceramide (PEG-cer), or a PEG dialkoxypropylcarbamate such as co- methoxy(polyethoxy)ethyl-N-(2,3di(tetradecanoxy)propyl)carbamate or 2,3- di(tetradecanoxy)propyl-N-(ω-methoxy(polyethoxy)ethyl)carbamate. PEG-lipids are disclosed in, e.g., U.S.9,737,619, the full disclosures of which is herein incorporated by reference in its entirety for all purposes. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing pegylated lipids may be excluded from the LNPs of the present disclosure. In some aspects, the composition comprises a pegylated lipid having the following structure:
Figure imgf000198_0001
or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: R8 and R9 are each independently a straight or branched, saturated or unsaturated alkyl chain containing from 10 to 30 carbon atoms, wherein the alkyl chain is optionally interrupted by one or more ester bonds; and w has a mean value ranging from 30 to 60. In some aspects, R8 and R9 are each independently straight, saturated alkyl chains containing from 12 to 16 carbon atoms. In some aspects, w has a mean value ranging from 43 to 53. In other aspects, the average w is or is about 45. In other different embodiments, the average w is or is about 49. In some aspects, the lipid nanoparticles comprise a polymer conjugated lipid. In one aspect, the lipid nanoparticle comprises 2-[(polyethylene glycol)-2000]-N,N- ditetradecylacetamide (ALC-0159), having the formula:
Figure imgf000198_0002
In various aspects, the molar ratio of the cationic lipid to the pegylated lipid ranges from or from about 100:1 to about 20:1, e.g., 20:1, 25:1, 30:1, 35:1, 40:1, 45:1, 50:1, 55:1, 60:1, 65:1, 70:1, 75:1, 80:1, 85:1, 90:1, 95:1, or 100:1, or any range or value derivable therein. In certain aspects, the PEG-lipid is or is not present in the LNP in an amount from or from about 1 to about 10 mole percent (mol %) (e.g., at least, at most, exactly, or between (inclusive or exclusive) of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 mol %), relative to the total lipid content of the nanoparticle. In some aspects, the ratio of PEG in the lipid nanoparticle formulations may be increased or decreased and/or the carbon chain length of the PEG lipid may be modified to alter the pharmacokinetics and/or biodistribution of the lipid nanoparticle formulations. iii. ADDITIONAL LIPIDS In certain aspects, the LNP comprises one or more additional lipids or lipid-like materials that stabilize particles during their formation. Suitable stabilizing or structural lipids include non- cationic lipids, e.g., neutral lipids and anionic lipids. Without being bound by any theory, optimizing the formulation of LNPs by addition of other hydrophobic moieties, such as cholesterol and lipids, in addition to an ionizable/cationic lipid or lipid-like material may enhance particle stability and efficacy of nucleic acid delivery. As used herein, an “anionic lipid” refers to any lipid that is negatively charged at a selected pH. The term “neutral lipid” refers to any one of a number of lipid species that exist in either an uncharged or neutral zwitterionic form at physiological pH. In some aspects, additional lipids comprise one of the following neutral lipid components: (1) a phospholipid, (2) cholesterol or a derivative thereof; or (3) a mixture of a phospholipid and cholesterol or a derivative thereof. Representative neutral lipids include phosphatidylcholines, phosphatidylethanolamines, phosphatidylglycerols, phosphatidic acids, phosphatidylserines, ceramides, sphingomyelins, dihydro-sphingomyelins, cephalins, and cerebrosides. Exemplary phospholipids include, for example, phosphatidylcholines, e.g., diacylphosphatidylcholines, such as distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dimyristoylphosphatidylcholine (DMPC), dipentadecanoylphosphatidylcholine, dilauroylphosphatidylcholine, dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), diarachidoylphosphatidylcholine (DAPC), dibehenoylphosphatidylcholine (DBPC), ditricosanoylphosphatidylcholine (DTPC), dilignoceroylphatidylcholine (DLPC), palmitoyloleoyl- phosphatidylcholine (POPC), 1,2-di-O-octadecenyl-sn-glycero-3-phosphocholine (18:0 Diether PC), 1-oleoyl-2-cholesterylhemisuccinoyl-sn-glycero-3-phosphocholine (OChemsPC), and 1- hexadecyl-sn-glycero-3-phosphocholine (C16 Lyso PC); and phosphatidylethanolamines, e.g., diacylphosphatidylethanolamines, such as dioleoyl-phosphatidylethanolamine (DOPE), 1,2- diundecanoyl-sn-glycero-phosphocholine (DUPC), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE) and dioleoyl-phosphatidylethanolamine 4-(N- maleimidomethyl)-cyclohexane-1-carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), dilauroyl- phosphatidylethanolamine (DLPE), distearoyl-phosphatidylethanolamine (DSPE), 1-phytanoyl- phosphatidylethanolamine (DpyPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, 1- stearoyl-2-oleoylphosphatidyethanolamine (SOPE), 1,2-dielaidoyl-sn-glycero-3- phosphoethanolamine (transDOPE), 1,2- dilinolenoyl-sn-glycero-3-phosphocholine,1,2- diarachidonoyl-sn-glycero-3-phosphocholine, 1,2- didocosahexaenoyl-sn-glycero-3- phosphocholine, 1,2-diphytanoyl-sn-glycero-3- phosphoethanolamine (ME 16.0 PE), 1,2- distearoyl-sn-glycero-3-phosphoethanolamine, 1,2- dilinoleoyl-sn-glycero-3- phosphoethanolamine, 1,2-dilinolenoyl-sn-glycero-3- phosphoethanolamine, 1,2-diarachidonoyl- sn-glycero-3-phosphoethanolamine, 1,2- didocosahexaenoyl-sn-glycero-3- phosphoethanolamine, 1,2-dioleoyl-sn-glycero-3-phospho-rac- (1-glycerol) sodium salt (DOPG), sphingomyelin, and mixtures thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing neutral lipids may be excluded from the LNPs of the present disclosure. In one aspect, the neutral lipid is 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), having the formula:
Figure imgf000200_0001
In some aspects, the LNPs comprise a neutral lipid, and the neutral lipid comprises one or more of DSPC, DPPC, DMPC, DOPC, POPC, DOPE, and/or SM. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing neutral lipids may be excluded from the LNPs of the present disclosure. In various aspects, the LNPs further comprise a steroid or steroid analogue. A “steroid” is a compound comprising the following carbon skeleton:
Figure imgf000200_0002
In certain aspects, the steroid or steroid analogue is cholesterol, fecosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, ursolic acid, alpha- tocopherol, and mixtures thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing steroid or steroid analogues may be excluded from the LNPs of the present disclosure. In certain aspects, the steroid or steroid analogue is cholesterol. Examples of cholesterol derivatives include, but are not limited to, cholestanol, cholestanone, cholestenone, coprostanol, cholesteryl-2′-hydroxyethyl ether, cholesteryl-4′-hydroxybutyl ether, tocopherol and derivatives thereof, and mixtures thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing cholesterol derivatives may be excluded from the LNPs of the present disclosure. In one aspect, the cholesterol has the formula:
Figure imgf000200_0003
Without being bound by any theory, the amount of the at least one cationic lipid compared to the amount of the at least one additional lipid may affect important nucleic acid particle characteristics, such as charge, particle size, stability, tissue selectivity, and bioactivity of the nucleic acid. Accordingly, in some aspects, the molar ratio of the cationic lipid to the neutral lipid ranges from or from about 2:1 to about 8:1, or from or from about 10:0 to about 1:9, about 4:1 to about 1:2, or about 3:1 to about 1:1. In another aspect, the molar ratio of the cationic lipid to cholesterol ranges from or from about 2:1 to 1:1. In a further aspect, the molar ratio of the cationic lipid to the pegylated lipid ranges from or from about 100:1 to about 10:1 or from about 100:1 to about 25.1. In some aspects, the non-cationic lipid, e.g., neutral lipid (e.g., one or more phospholipids and/or cholesterol), may comprise from or from about 0 mol % to about 90 mol %, from or from about 0 mol % to about 80 mol %, from or from about 0 mol % to about 70 mol %, from or from about 0 mol % to about 60 mol %, or from or from about 0 mol % to about 50 mol %, of the total lipid present in the particle. In some aspects, the non-cationic lipid, e.g., neutral lipid (e.g., one or more phospholipids and/or cholesterol), may or may not be at least, at most, exactly, or between (inclusive or exclusive) of 0 mol %, 10 mol %, 20 mol %, 30 mol %, 40 mol %, 50 mol %, 60 mol %, 70 mol %, 80 mol %, or 90 mol % of the total lipid present in the particle. VI. CHARACTERIZATION AND ANALYSIS OF RNA MOLECULE The RNA molecule described herein may be analyzed and characterized using various methods. Analysis may be performed before and/or after capping. Alternatively, analysis may be performed before and/or after poly-A capture-based affinity purification. In another aspect, analysis may be performed before and/or after additional purification steps, e.g., anion exchange chromatography and the like. For example, RNA template quality may be determined using a Bioanalyzer chip-based electrophoresis system. In other aspects, RNA template purity is analyzed using analytical reverse phase HPLC. Capping efficiency may be analyzed using, e.g., total nuclease digestion followed by MS/MS quantitation of the dinucleotide cap species vs. uncapped GTP species. In vitro efficacy may be analyzed by, e.g., transfecting an RNA molecule into a human cell line. Protein expression of the polypeptide of interest may be quantified using methods such as ELISA and/or flow cytometry. Immunogenicity may be analyzed by, e.g., transfecting RNA molecules into cell lines that indicate innate immune stimulation, e.g., PBMCs. Cytokine induction may be analyzed using, e.g., methods such as ELISA to quantify a cytokine, e.g., Interferon-α. Biodistribution may be analyzed by, e.g., bioluminescence measurements. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing analytic methods may be excluded. In some aspects, an RNA polynucleotide disclosed herein is characterized in that, when assessed in an organism administered a composition or medical preparation comprising an RNA polynucleotide, elevated expression of a gene of interest (e.g., an antigen); increased duration of expression (e.g., prolonged expression) of a gene of interest (e.g., an antigen); elevated expression and increased duration of expression (e.g., prolonged expression) of a gene of interest (e.g., an antigen); decreased interaction with IFIT1 of an RNA polynucleotide; and/or increased translation of an RNA polynucleotide; is observed relative to an appropriate reference. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing characteristics may not be observed after administration of a composition or medical preparation comprising an RNA molecule of the present disclosure. In some aspects, a reference comprises an organism administered an otherwise similar RNA polynucleotide without a m7(3′OMeG)(5′)ppp(5′)(2′OMeAi)pG2 cap. In some aspects, a reference comprises an organism administered an otherwise similar RNA polynucleotide without a cap proximal sequence disclosed herein. In some aspects, a reference comprises an organism administered an otherwise similar RNA polynucleotide with a self-hybridizing sequence. In some aspects, elevated expression is determined at least 24 hours, at least 48 hours, at least 72 hours, at least 96 hours, or at least 120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at least 24 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at least 48 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at least 72 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at least 96 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at least 120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at or at about 24-120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression is determined at or at about 24-110 hours, 24-100 hours, 24- 90 hours, 24-80 hours, 24-70 hours, 24-60 hours, 24-50 hours, 24-40 hours, 24-30 hours, 30-120 hours, 40-120 hours, 50-120 hours, 60-120 hours, 70-120 hours, 80-120 hours, 90-120 hours, 100-120 hours, or 110-120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, expression of a gene of interest (e.g., an antigen) is or is not elevated at least 2-fold to at least 10-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated at least 2-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated at least 3-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated at least 4-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated at least 6-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated at least 8-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated at least 10-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated or elevated about 2-fold to about 50-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is elevated or elevated about 2-fold to about 45-fold, about 2-fold to about 40-fold, about 2-fold to about 30-fold, about 2-fold to about 25-fold, about 2-fold to about 20-fold, about 2-fold to about 15-fold, about 2-fold to about 10-fold, about 2-fold to about 8-fold, about 2-fold to about 5-fold, about 5-fold to about 50-fold, about 10-fold to about 50-fold, about 15-fold to about 50-fold, about 20-fold to about 50-fold, about 25-fold to about 50-fold, about 30-fold to about 50-fold, about 40- fold to about 50-fold, or about 45-fold to about 50-fold. In some aspects, expression of a gene of interest (e.g., an antigen) is or is not elevated at least, at most, exactly, or between (inclusive or exclusive) of 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 11-fold, 12-fold, 13- fold, 14-fold, 15-fold, 16-fold, 17-fold, 18-fold, 19-fold, 20-fold, 21-fold, 22-fold, 23-fold, 24-fold, 25-fold, 26-fold, 27-fold, 28-fold, 29-fold, 30-fold, 31-fold, 32-fold, 33-fold, 34-fold, 35-fold, 36- fold, 37-fold, 38-fold, 39-fold, 40-fold, 41-fold, 42-fold, 43-fold, 44-fold, 45-fold, 46-fold, 47-fold, 48-fold, 49-fold, or 50-fold, or any range or value derivable therein. In some aspects, elevated expression (e.g., increased duration of expression) of a gene of interest (e.g., an antigen) persists for at least, at most, exactly, or between (inclusive or exclusive) of 24 hours, 48 hours, 72 hours, 96 hours, or 120 hours after administration of a composition or a medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression of a gene of interest (e.g., an antigen) persists for at least 24 hours after administration. In some aspects, elevated expression of a gene of interest (e.g., an antigen) persists for at least 48 hours after administration. In some aspects, elevated expression of a gene of interest (e.g., an antigen) persists for at least 72 hours after administration. In some aspects, elevated expression of a gene of interest (e.g., an antigen) persists for at least 96 hours after administration. In some aspects, elevated expression of a gene of interest (e.g., an antigen) persists for at least 120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression of a gene of interest (e.g., an antigen) persists for or for about 24-120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression persists for or for about 24-110 hours, 24-100 hours, 24-90 hours, 24-80 hours, 24-70 hours, 24-60 hours, 24-50 hours, 24-40 hours, 24-30 hours, 30-120 hours, 40-120 hours, 50-120 hours, 60-120 hours, 70-120 hours, 80- 120 hours, 90-120 hours, 100-120 hours, or 110-120 hours after administration of a composition or medical preparation comprising an RNA polynucleotide. In some aspects, elevated expression of a gene of interest (e.g., an antigen) persists or does not persist for at least, at most, exactly, or between (inclusive or exclusive) of 24 hours, 36 hours, 48 hours, 60 hours, 72 hours, 84 hours, 96 hours, 108 hours, or 120 hours, or any range or value derivable therein. VII. IMMUNE RESPONSE AND ASSAYS As discussed herein, the disclosure concerns evoking or inducing an immune response in a subject against an RSV protein, e.g., a wild type or variant RSV F protein. In one aspect, the immune response may protect against or treat a subject having, suspected of having, or at risk of developing an infection or related disease, particularly those related to RSV. One use of the immunogenic compositions of the disclosure is to prevent RSV infections by inoculating or vaccination of a subject. In some aspects, the immunogenic compositions immunize the subject against RSV up to 1 year (e.g., for a single RSV season). In some aspects, the immunogenic compositions immunize the subject against RSV for up to 2 years. In some aspects, the immunogenic compositions immunize the subject against RSV for more than 2 years. In some aspects, the immunogenic compositions immunize the subject against RSV for more than 3 years. In some aspects, the immunogenic compositions immunize the subject against RSV for more than 4 years. In some aspects, the immunogenic compositions immunize the subject against RSV for 5-10 years. A. IMMUNOASSAYS The present disclosure includes the implementation of serological assays to evaluate whether and to what extent an immune response is induced or evoked by compositions of the disclosure. There are many types of immunoassays that may be implemented. Immunoassays encompassed by the present disclosure include, but are not limited to, those described in U.S. Patent 4,367,110 (double monoclonal antibody sandwich assay) and U.S. Patent 4,452,901 (western blot). Other assays include immunoprecipitation of labeled ligands and immunocytochemistry, both in vitro and in vivo. Immunoassays generally are binding assays. In some aspects, the immunoassays are the various types of enzyme linked immunosorbent assays (ELISAs) and radioimmunoassays (RIA) known in the art. Immunohistochemical detection using tissue sections is also particularly useful. In one example, antibodies or antigens are immobilized on a selected surface, such as a well in a polystyrene microtiter plate, dipstick, or column support. Then, a test composition suspected of containing the desired antigen or antibody, such as a clinical sample, is added to the wells. After binding and washing to remove non-specifically bound immune complexes, the bound antigen or antibody may be detected. Detection is generally achieved by the addition of another antibody, specific for the desired antigen or antibody, that is linked to a detectable label. This type of ELISA is known as a “sandwich ELISA.” Detection also may be achieved by the addition of a second antibody specific for the desired antigen, followed by the addition of a third antibody that has binding affinity for the second antibody, with the third antibody being linked to a detectable label. Competition ELISAs are also possible implementations in which test samples compete for binding with known amounts of labeled antigens or antibodies. The amount of reactive species in the unknown sample is determined by mixing the sample with the known labeled species before or during incubation with coated wells. The presence of reactive species in the sample acts to reduce the amount of labeled species available for binding to the well and thus reduces the ultimate signal. Irrespective of the format employed, ELISAs have certain features in common, such as coating, incubating or binding, washing to remove non-specifically bound species, and detecting the bound immune complexes. Antigen or antibodies may also be linked to a solid support, such as in the form of plate, beads, dipstick, membrane, or column matrix, and the sample to be analyzed is applied to the immobilized antigen or antibody. In coating a plate with either antigen or antibody, one will generally incubate the wells of the plate with a solution of the antigen or antibody, either overnight or for a specified period. The wells of the plate will then be washed to remove incompletely- adsorbed material. Any remaining available surfaces of the wells are then “coated” with a nonspecific protein that is antigenically neutral with regard to the test antisera. These include bovine serum albumin (BSA), casein, and solutions of milk powder. The coating allows for blocking of nonspecific adsorption sites on the immobilizing surface and thus reduces the background caused by nonspecific binding of antisera onto the surface. B. DIAGNOSIS OF RSV INFECTION The present disclosure contemplates the use of RSV polypeptides, proteins, and/or peptides in a variety of ways, including the detection of the presence of RSV to diagnose an infection. In accordance with the disclosure, a method of detecting the presence of infections involves the steps of obtaining a sample suspected of being infected by one or more RSV strains, such as a sample taken from an individual, for example, from one’s blood, saliva, tissues, bone, muscle, cartilage, or skin. Following isolation of the sample, diagnostic assays utilizing the polypeptides, proteins, and/or peptides of the present disclosure may be carried out to detect the presence of RSV, and such assay techniques for determining such presence in a sample are well known to those skilled in the art and include methods such as radioimmunoassay, western blot analysis and ELISA assays. In general, in accordance with the disclosure, a method of diagnosing an infection is contemplated wherein a sample suspected of being infected with RSV has added to it the polypeptide, protein, or peptide, in accordance with the present disclosure, and RSV is indicated by antibody binding to the polypeptides, proteins, and/or peptides, or polypeptides, proteins, and/or peptides binding to the antibodies in the sample. Also contemplated is a method of testing a sample suspected of being infected with, previously infected with, or infected with, RSV has added to it the polypeptide, protein, or peptide, in accordance with the present disclosure, and RSV is indicated by antibody binding to the polypeptides, proteins, and/or peptides, or polypeptides, proteins, and/or peptides binding to the antibodies in the sample. Accordingly, RNA molecules encoding RSV polypeptides, proteins, and/or peptides in accordance with the disclosure may be used for to treat, prevent, or reduce the severity of illness due to RSV infection (e.g., active or passive immunization) or for use as research tools. Any of the above described polypeptides, proteins, and/or peptides may be labeled directly with a detectable label for identification and quantification of RSV. Labels for use in immunoassays are generally known to those skilled in the art and include enzymes, radioisotopes, and fluorescent, luminescent and chromogenic substances, including colored particles such as colloidal gold or latex beads. Suitable immunoassays include enzyme-linked immunosorbent assays (ELISA). C. PROTECTIVE IMMUNITY In some aspects of the disclosure, RNA molecules encoding RSV preF polypeptides, RNA-LNPs and compositions thereof, confer protective immunity to a subject. Protective immunity refers to a body’s ability to mount a specific immune response that protects the subject from developing a particular disease or condition that involves the agent against which there is an immune response. An immunogenically effective amount is capable of conferring protective immunity to the subject. In some aspects, the RNA molecules encoding RSV polypeptides, RNA- LNPs and compositions thereof of the present disclosure may be used to induce a balanced immune response against RSV comprising both cellular and humoral immunity, without many of the risks associated with attenuated virus vaccination. A “humoral” immune response refers to an immune response mediated by antibody molecules, including, e.g., secretory (IgA) or IgG molecules, while a “cellular” immune response is one mediated by T- lymphocytes (e.g., CD4+ helper and/or CD8+ T cells (e.g., CTLs) and/or other white blood cells. As used herein the phrase “immune response” or its equivalent “immunological response” refers to the development of a humoral (antibody mediated), cellular (mediated by antigen- specific T cells or their secretion products) or both humoral and cellular response directed against an antigen. Such a response may be an active response or a passive response. A cellular immune response is elicited by the presentation of polypeptide epitopes in association with Class I or Class II MHC molecules, to activate antigen-specific CD4 (+) T helper cells and/or CD8 (+) cytotoxic T cells. The response may also involve activation of monocytes, macrophages, NK cells, basophils, dendritic cells, astrocytes, microglia cells, eosinophils or other components of innate immunity. As used herein “active immunity” refers to any immunity conferred upon a subject from the production of antibodies in response to the presence of an of an antigen, e.g. an RSV F protein encoded by an RNA molecule of the present disclosure. As used herein “passive immunity” includes, but is not limited to, administration of activated immune effectors including cellular mediators or protein mediators (e.g., monoclonal and/or polyclonal antibodies) of an immune response. A monoclonal or polyclonal antibody composition may be used in passive immunization to treat, prevent, or reduce the severity of illness caused by infection by organisms that carry the antigen recognized by the antibody. An antibody composition may include antibodies that bind to a variety of antigens that may in turn be associated with various organisms. The antibody component may be a polyclonal antiserum. In certain aspects the antibody or antibodies are affinity purified from an animal or second subject that has been challenged with an antigen(s). Alternatively, an antibody mixture may be used, which is a mixture of monoclonal and/or polyclonal antibodies to antigens present in the same, related, or different microbes or organisms, such as viruses, including but not limited to RSV. Passive immunity may be imparted to a patient or subject by administering to the patient immunoglobulins (Ig) and/or other immune factors obtained from a donor or other non-patient source having a known immunoreactivity. In other aspects, an immunogenic composition of the present disclosure may be administered to a subject who then acts as a source or donor for globulin, produced in response to challenge with the immunogenic composition (“hyperimmune globulin”), that contains antibodies directed against a RSV or other organism. A subject thus treated would donate plasma from which hyperimmune globulin would then be obtained, via conventional plasma-fractionation methodology, and administered to another subject in order to impart resistance against or to treat RSV infection. For purposes of this specification and the accompanying claims the terms “epitope” and “antigenic determinant” are used interchangeably to refer to a site on an antigen to which B and/or T cells respond or recognize. B-cell epitopes may be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents. An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation. Methods of determining spatial conformation of epitopes include, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols (1996). Antibodies that recognize the same epitope may be identified in a simple immunoassay showing the ability of one antibody to block the binding of another antibody to a target antigen. T-cells recognize continuous epitopes of about nine amino acids for CD8 cells or about 13-15 amino acids for CD4 cells. T cells that recognize the epitope may be identified by in vitro assays that measure antigen-dependent proliferation, as determined by 3H- thymidine incorporation by primed T cells in response to an epitope (Burke et al., 1994), by antigen-dependent killing (cytotoxic T lymphocyte assay, Tigges et al., 1996) or by cytokine secretion. The presence of a cell-mediated immunological response may be determined by proliferation assays (CD4 (+) T cells) or CTL (cytotoxic T lymphocyte) assays. The relative contributions of humoral and cellular responses to the protective or therapeutic effect of an immunogenic composition may be distinguished by separately isolating IgG and T-cells from an immunized syngeneic animal and measuring protective or therapeutic effect in a second subject. As used herein, the terms “antibody” or “immunoglobulin” are used interchangeably and refer to any of several classes of structurally related proteins that function as part of the immune response of an animal or recipient, which proteins include IgG, IgD, IgE, IgA, IgM and related proteins. Under normal physiological conditions antibodies are found in plasma and other body fluids and in the membrane of certain cells and are produced by lymphocytes of the type denoted B cells or their functional equivalent. As used herein the terms “immunogenic agent” or “immunogen” or “antigen” are used interchangeably to describe a molecule capable of inducing an immunological response against itself on administration to a recipient, either alone, in conjunction with an adjuvant, or presented on a display vehicle. VIII. COMPOSITIONS In some aspects, RNA molecules and/or RNA-LNPs disclosed herein may be administered in a pharmaceutical composition or a medicament and may be administered in the form of any suitable pharmaceutical composition. In some aspects, a pharmaceutical composition is for therapeutic and/or prophylactic treatment. In one aspect, the disclosure relates to a composition for administration to a host. In some aspects, the host is a human. In other aspects, the host is a non-human. Formulations of the compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing an active ingredient (e.g., RNA molecules and/or RNA-LNPs) into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi-dose unit. A pharmaceutical composition or formulation in accordance with the present disclosure can be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a composition in accordance with the disclosure will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1% and 100%, e.g., between 0.5 and 50%, between 1-30%, between 5-80%, at least 80% (w/w), or at least, at most, exactly, or between (inclusive or exclusive) any two of 0.1%, 0.15%, 0.2%, 0.25%, 0.3%, 0.35%, 0.4%, 0.45%, 0.5%, 0.55%, 0.6%, 0.65%, 0.7%, 0.75%, 0.8%, 0.85%, 0.9%, 0.95%, 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% (w/w) active ingredient. General considerations in the formulation and/or manufacture of pharmaceutical agents, such as the compositions described herein, may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference in its entirety). In some aspects, RNA molecules and/or RNA-LNPs disclosed herein may be administered in a pharmaceutical composition which may be formulated into preparations in solid, semi-solid, liquid, lyophilized, frozen, and/or gaseous forms. In some aspects, an RNA molecule and/or RNA-LNPs disclosed herein may be administered in a pharmaceutical composition which may comprise a pharmaceutically acceptable carrier and may optionally comprise one or more adjuvants, stabilizers, salts, buffers, preservatives, and optionally other therapeutic agents. In some aspects, a pharmaceutical composition disclosed herein comprises one or more pharmaceutically acceptable carriers, diluents and/or excipients. In some aspects, pharmaceutical compositions do not include an adjuvant (e.g., they are adjuvant free). The term “excipient” as used herein refers to a substance which may be present in a pharmaceutical composition of the present disclosure but is not an active ingredient. Examples of excipients, include without limitation, carriers, diluents (e.g., solvents, dispersion media, and/or other liquid vehicles, dispersion or suspension aids), granulating and/or dispersing agents, surface active agents, isotonic agents, thickening and/or emulsifying agents, preservatives, binders, lubricants and/or oil, coloring, sweetening and/or flavoring agents, stabilizers, antioxidants, antimicrobial and/or antifungal agents, osmolality adjusting agents, pH adjusting agents, buffers, chelants, cryoprotectants, and/or bulking agents. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing excipients may be excluded from the pharmaceutical compositions disclosed herein. The term “carrier” refers to a component which may be natural, synthetic, organic, or inorganic, in which the active component is combined in order to facilitate, enhance and/or enable administration of the pharmaceutical composition. A carrier as used herein may be one or more compatible solid or liquid fillers, diluents or encapsulating substances, which are suitable for administration to subject. Suitable carriers include, without limitation, sterile water, Ringer’s solution, Ringer’s lactate solution, sterile sodium chloride solution, isotonic saline, polyalkylene glycols, hydrogenated naphthalenes and, in particular, biocompatible lactide polymers, lactide/glycolide copolymers or polyoxyethylene/polyoxy-propylene copolymers. In some aspects, the pharmaceutical composition of the present disclosure includes sodium chloride. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing carriers may be excluded from the pharmaceutical compositions disclosed herein. The term “diluent” relates a diluting and/or thinning agent. Moreover, the term “diluent” includes any one or more of fluid, liquid or solid suspension and/or mixing media. Examples of suitable diluents for use in a pharmaceutical compositions of the present disclosure include, without limitation, ethanol, glycerol, saline, water, calcium or sodium carbonate, calcium phosphate, calcium hydrogen phosphate, sodium phosphate, lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, etc., and/or combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing diluents may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable granulating and/or dispersing agents include, without limitation, starches, pregelatinized starches, or microcrystalline starch, alginic acid, guar gum, agar, poly(vinyl-pyrrolidone), (providone), cross-linked poly(vinyl-pyrrolidone) (crospovidone), cellulose, methylcellulose, carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), magnesium aluminum silicate (VEEGUM®), sodium lauryl sulfate, etc., and/or combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing granulating and/or dispersing agents may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable surface active agents for use in a pharmaceutical compositions of the present disclosure include, without limitation, natural emulsifiers (e.g., acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), sorbitan fatty acid esters (e.g., polyoxyethylene sorbitan monooleate [TWEEN®80], sorbitan monopalmitate [SPAN®40], glyceryl monooleate, polyoxyethylene esters, polyethylene glycol fatty acid esters (e.g., CREMOPHOR®), polyoxyethylene ethers (e.g., polyoxyethylene lauryl ether [BRIJ®30]), PLUORINC®F 68, POLOXAMER®188, etc. and/or combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing surface active agents may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable preservatives for use in a pharmaceutical compositions of the present disclosure include, without limitation, benzalkonium chloride, chlorobutanol, paraben, thimerosal, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, ascorbic acid, butylated hydroxyanisole, ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), etc., and combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing preservatives may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable antimicrobial and/or antifungal agents for use in a pharmaceutical compositions of the present disclosure include, without limitation, benzalkonium chloride, benzethonium chloride, methyl paraben, ethyl paraben, propyl paraben, butyl paraben, benzoic acid, hydroxybenzoic acid, potassium or sodium benzoate, potassium or sodium sorbate, sodium propionate, sorbic acid, etc., and combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing antimicrobial and/or antifungal agents may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable binders for use in a pharmaceutical compositions of the present disclosure include, without limitation, starch, gelatin, sugars (e.g., sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol), amino acids (e.g., glycine), natural and synthetic gums (e.g., acacia, sodium alginate), ethylcellulose, hydroxyethylcellulose, hydroxypropyl methylcellulose, etc., and combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing binders may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable lubricants and/or oil for use in a pharmaceutical compositions of the present disclosure include, without limitation, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium or magnesium lauryl sulfate, etc., and combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing lubricants and/or oils may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable antioxidants for use in a pharmaceutical compositions of the present disclosure include, without limitation, alpha tocopherol, ascorbic acid, ascorbyl palmitate, benzyl alcohol, butylated hydroxyanisole, m-cresol, methionine, butylated hydroxytoluene, monothioglycerol, sodium or potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, etc., and combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing antioxidants may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable osmolality adjusting agents, pH adjusting agents, and buffers for use in a pharmaceutical compositions of the present disclosure include, without limitation, sodium phosphate, sodium citrate, sodium succinate, histidine (or histidine-HCl), sodium malate, sodium carbonate, etc., and/or combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing osmolality adjusting agents may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable chelating agents for use in a pharmaceutical compositions of the present disclosure include, without limitation, ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, trisodium edetate, etc., and combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing chelating agents may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable cryoprotectants for use in a pharmaceutical compositions of the present disclosure include, without limitation, mannitol, sucrose, trehalose, lactose, glycerol, dextrose, etc., and combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing cryoprotectants may be excluded from the pharmaceutical compositions disclosed herein. Examples of suitable bulking agents include, without limitation, sucrose, trehalose, mannitol, glycine, lactose, raffinose, and combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing bulking agents may be excluded from the pharmaceutical compositions disclosed herein. Compositions can be formulated using one or more excipients (e.g., one or more carriers and/or diluents) to, e.g.: (1) increase stability; (2) increase cell transfection; (3) permit the sustained and/or delayed release (e.g., from a depot formulation); (4) alter the biodistribution (e.g., target to specific tissues and/or cell types); (5) increase the translation of encoded protein in vivo; and/or (6) alter the release profile of encoded protein (antigen) in vivo. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing excipient purposes may be excluded. Pharmaceutically acceptable excipients (e.g., carriers and/or diluents) for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington’s Pharmaceutical Sciences, Mack Publishing Co. (A. R Gennaro edit.1985). Pharmaceutical excipients (e.g., carriers and/or diluents) may be selected with regard to the intended route of administration and standard pharmaceutical practice. In some aspects, the composition comprises an RNA molecule comprising an open reading frame encoding an immunogenic polypeptide. In some aspects, the immunogenic polypeptide comprises a RSV antigen. In some aspects, the RSV antigen is a RSV F protein or a fragment or variant thereof. In some aspects, the composition comprises an RNA molecule comprising an open reading frame encoding a full-length RSV F protein. In some aspects, the encoded immunogenic polypeptide is a truncated RSV F protein. In some aspects, the encoded immunogenic polypeptide is a variant of a RSV F protein. In some aspects, the encoded immunogenic polypeptide is a fragment of a RSV F protein. A. IMMUNOGENIC COMPOSITIONS INCLUDING LNPS In some aspects, a pharmaceutical composition comprises an RNA molecule (e.g., polynucleotide) disclosed herein formulated with a lipid-based delivery system. Thus, some aspects, the composition includes a lipid-based delivery system (e.g., LNPs) (e.g., a lipid-based vaccine), which delivers a nucleic acid molecule to the interior of a cell, where it may then replicate, inhibit protein expression of interest, and/or express the encoded polypeptide of interest. The delivery system may have adjuvant effects which enhance the immunogenicity of an encoded antigen. In some aspects, the composition comprises at least one RNA molecule encoding a RSV polypeptide complexed with, encapsulated in, and/or formulated with one or more lipids, and forming lipid nanoparticles (LNPs), liposomes, lipoplexes and/or nanoliposomes. In some aspects, the composition comprises a lipid nanoparticle. Thus, in certain aspects, the present disclosure concerns compositions comprising one or more lipids associated with a nucleic acid or a polypeptide/peptide (e.g., RSV RNA-LNPs). In one aspect, the disclosure relates to an immunogenic composition may be multivalent, wherein the immunogenic composition comprises (i) a first ribonucleic acid (RNA) polynucleotide having an open reading frame encoding a first antigen, said antigen including at least one RSV F protein polypeptide or an immunogenic fragment thereof, and (ii) a second RNA polynucleotide having an open reading frame encoding a second antigen, said second antigen including at least one RSV F protein polypeptide or an immunogenic fragment thereof, wherein the first and second RNA polynucleotides are formulated in a lipid nanoparticle (LNP). In some embodiments, the first and second antigens include RSV F protein (F) as described herein, or an immunogenic fragment or variant thereof. In some embodiments, the first antigen includes an RSV F protein from a different subtype of RSV to the RSV antigenic polypeptide or an immunogenic fragment thereof of the second antigen. In some embodiments, the first and second RNA polynucleotides are formulated in a lipid nanoparticle. In one aspect, the immunogenic composition is a bivalent immunogenic composition comprising a first RSV molecule encoding an F protein as described herein which is RSV subtype A and a second RSV molecule encoding an F protein as described herein which is RSV subtype B. In one aspect, the ratio of the first RSV molecule to the second RSV molecule in the immunogenic composition is 1:1. In another aspect, the multivalent immunogenic composition may be a (i) "pre-mix" formulation wherein two or more RNA polynucleotides are combined in equal ratio followed by co-formulation into LNPs, or (ii) a "post- mix" formulation wherein each RNA polynucleotide is individually formulated into LNPs and the two or more LNPs are combined either in equal ratio or not in equal ratio. The immunogenic composition including a lipid-based delivery system may further include one or more salts and/or one or more pharmaceutically acceptable surfactants, preservatives, carriers, diluents, and/or excipients, in some cases. In some aspects, the immunogenic composition including a lipid-based delivery system further includes a pharmaceutically acceptable vehicle. In some aspects, each of a buffer, stabilizing agent, and optionally a salt, may be included in the immunogenic composition including a lipid-based delivery system. In other aspects, any one or more of a buffer, stabilizing agent, salt, surfactant, preservative, and excipient may be excluded from the immunogenic composition including a lipid-based delivery system. In a further aspect, the immunogenic composition including a lipid-based delivery system further comprises a stabilizing agent. In some aspects, the stabilizing agent comprises sucrose, mannose, sorbitol, raffinose, trehalose, mannitol, inositol, sodium chloride, arginine, lactose, hydroxyethyl starch, dextran, polyvinylpyrolidone, glycine, or a combination thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing stabilizing agents may be excluded from the immunogenic compositions disclosed herein. In some aspects, the stabilizing agent is a disaccharide, or sugar. In one aspect, the stabilizing agent is sucrose. In another aspect, the stabilizing agent is trehalose. In a further aspect, the stabilizing agent is a combination of sucrose and trehalose. In some aspects, the total concentration of the stabilizing agent(s) in the composition is or is about 5% to about 10% w/v. For example, the total concentration of the stabilizing agent may or may not be equal to at least, at most, exactly, or between (inclusive or exclusive) of 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, or 20% w/v or any range or value derivable therein. In some aspects, the stabilizing agent concentration includes, but is not limited to, a concentration of or of about 10 mg/mL to about 400 mg/mL, about 100 mg/mL to about 200 mg/mL, about 100 mg/mL to about 150 mg/mL, about 100 mg/mL to about 140 mg/mL, about 100 mg/mL to about 130 mg/mL, about 100 mg/mL to about 120 mg/mL, about 100 mg/mL to about 110 mg/mL, or about 100 mg/mL to about 105 mg/mL. In some aspects, the concentration of the stabilizing agent is or is not equal to at least, at most, exactly, or between (inclusive or exclusive) of 10 mg/mL, 20 mg/mL, 50 mg/mL, 100 mg/mL, 101 mg/mL, 102 mg/mL, 103 mg/mL, 104 mg/mL, 105 mg/mL, 106 mg/mL, 107 mg/mL, 108 mg/mL, 109 mg/mL, 110 mg/mL, 150 mg/mL, 200 mg/mL, 300 mg/mL, 400 mg/mL, or more. In a further aspect, the mass amount of the stabilizing agent and the mass amount of the RNA are in a specific ratio. In one aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 5000. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 2000. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 1000. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 500. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 100. In another aspect, the ratio of the mass amount of the stabilizing agent and the pharmaceutical substance is no greater than 50. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 10. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 1. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 0.5. In another aspect, the ratio of the mass amount of the stabilizing agent and the RNA is no greater than 0.1. In another aspect, the stabilizing agent and RNA comprise a mass ratio of or of about 200 – 2000 of the stabilizing agent : 1 of the RNA. In some aspects, the immunogenic composition including a lipid-based delivery system further comprises a buffer. Examples of buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, d-gluconic acid, calcium glycerophosphate, calcium lactate, calcium lactobionate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, Tris hydrochloride (HCl), amino-sulfonate buffers (e.g., HEPES), magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, isotonic saline, Ringer’s solution, ethyl alcohol, and/or combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing buffering agents may be excluded from the immunogenic compositions disclosed herein. In some aspects, the buffer is a HEPES buffer, a Tris buffer, and/or a PBS buffer. In one aspect, the buffer is Tris buffer. In another aspect, the buffer is a HEPES buffer. In a further aspect, the buffer is a PBS buffer. For example, the buffer concentration may or may not be equal to at least, at most, exactly, or between (inclusive or exclusive) of 1 mM, 2 mM, 3 mM, 4 mM, 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, 10 mM, 11 mM, 12 mM, 13 mM, 14 mM, 15 mM, 16 mM, 17 mM, 18 mM, 19 mM, or 20 mM, or any range or value derivable therein. The buffer may be at a neutral pH, pH 6.5 to 8.5, pH 7.0 to pH 8.0, or pH 7.2 to pH 7.6. For example, the buffer may or may not be at least, at most, exactly, or between (inclusive or exclusive) of pH 6.5, 6.6, 6.7, 6.8, 6.9, 7, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8, 8.1, 8.2, 8.3, 8.4, or 8.5, or any range or value derivable therein. In specific aspects, the buffer is at pH 7.4. In some aspects, the immunogenic composition including a lipid-based delivery system may further comprise a salt. Examples of salts include but not limited to sodium salts and/or potassium salts. In one aspect, the salt is a sodium salt. In a specific aspect, the sodium salt is sodium chloride. In one aspect, the salt is a potassium salt. In some aspects, the potassium salt comprises potassium chloride. In some aspects, any one or more of the foregoing salts may be excluded from the immunogenic compositions disclosed herein. The concentration of the salts in the composition may be or be about 70 mM to about 140 mM. For example, the salt concentration may or may not be equal to at least, at most, exactly, or between (inclusive or exclusive) of 50 mM, 60 mM, 70 mM, 80 mM, 90 mM, 100 mM, 120 mM, 130 mM, 140 mM, 150 mM, 160 mM, 170 mM, 180 mM, 190 mM, or 200 mM. In some aspects, the salt concentration includes, but is not limited to, a concentration of or of about 1 mg/mL to about 100 mg/mL, about 1 mg/mL to about 50 mg/mL, about 1 mg/mL to about 40 mg/mL, about 1 mg/mL to about 30 mg/mL, about 1 mg/mL to about 20 mg/mL, about 1 mg/mL to about 10 mg/mL, or about 1 mg/mL to about 15 mg/mL. In some aspects, the concentration of the salt is or is not equal to at least, at most, exactly, or between (inclusive or exclusive) of 1 mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, 5 mg/mL, 6 mg/mL, 7 mg/mL, 8 mg/mL, 9 mg/mL, 10 mg/mL, 11 mg/mL, 12 mg/mL, 13 mg/mL, 14 mg/mL, 15 mg/mL, 16 mg/mL, 17 mg/mL, 18 mg/mL, 19 mg/mL, 20 mg/mL, or more. The salt may be at a neutral pH, pH 6.5 to 8.5, pH 7.0 to pH 8.0, or pH 7.2 to pH 7.6. For example, the salt may or may not be at a pH equal to at least, at most, exactly, or between (inclusive or exclusive) of 6.5, 6.6, 6.7, 6.8, 6.9, 7, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8, 8.1, 8.2, 8.3, 8.4, or 8.5. In some aspects, the immunogenic composition including a lipid-based delivery system further comprises a surfactant, a preservative, any other excipient, or a combination thereof. As used herein, “any other excipient” includes, but is not limited to, antioxidants, glutathione, EDTA, methionine, desferal, antioxidants, metal scavengers, and/or free radical scavengers. In one aspect, the surfactant, preservative, excipient or combination thereof is sterile water for injection (sWFI), bacteriostatic water for injection (BWFI), saline, dextrose solution, polysorbates, poloxamers, Triton, divalent cations, Ringer’s lactate, amino acids, sugars, polyols, polymers, and/or cyclodextrins. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing excipients may be excluded from the immunogenic compositions disclosed herein. Further examples of excipients, which refer to ingredients in the immunogenic compositions that are not active ingredients, include but are not limited to carriers, binders, diluents, lubricants, thickeners, surface active agents, preservatives, stabilizers, emulsifiers, buffers, flavoring agents, disintegrants, coatings, plasticizers, compression agents, wet granulation agents, and/or colorants. As used herein, “pharmaceutically acceptable carrier” includes any and all aqueous solvents (e.g., water, alcoholic/aqueous solutions, saline solutions, parenteral vehicles, such as sodium chloride, Ringer’s dextrose, etc.), non-aqueous solvents (e.g., propylene glycol, polyethylene glycol, vegetable oil, and injectable organic esters, such as ethyl oleate), dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial and/or antifungal agents, anti-oxidants, chelating agents, and inert gases), isotonic agents, absorption delaying agents, salts, drugs, drug stabilizers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, fluid and nutrient replenishers, such like materials and combinations thereof, as would be known to one of ordinary skill in the art. Diluents, or diluting or thinning agents, include but are not limited to ethanol, glycerol, water, sugars such as lactose, sucrose, mannitol, and sorbitol, and starches derived from wheat, corn rice, and potato; and celluloses such as microcrystalline cellulose. The amount of diluent in the composition may range from or from about 10% to about 90% by weight of the total composition, e.g., from or from about 25% to about 75%, about 30% to about 60% by weight, or about 12% to about 60%. Preservatives for use in the compositions disclosed herein include but are not limited to benzalkonium chloride, chlorobutanol, paraben and thimerosal. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing excipients may be excluded from the immunogenic compositions disclosed herein. The pH and exact concentration of the various components in the immunogenic composition including a lipid-based delivery system are adjusted according to well-known parameters. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredients, its use in immunogenic, prophylactic and/or therapeutic compositions is contemplated. In one aspect, a pharmaceutical composition comprises an RSV RNA molecule encoding a RSV polypeptide as disclosed herein that is complexed with, encapsulated in, and/or formulated with one or more lipids to form RSV RNA-LNPs. In some aspects, the RSV RNA-LNP composition is a liquid. In some aspects, the RSV RNA-LNP composition is frozen. In some aspects, the RSV RNA-LNP composition is lyophilized. In some aspects, a RSV RNA-LNP composition comprises a RSV RNA polynucleotide molecule encoding a RSV polypeptide as disclosed herein, encapsulated in LNPs with a lipid composition of a cationic lipid, a PEGylated lipid (e.g. PEG- lipid), and one or more structural lipids (e.g., a neutral lipid). In some aspects, any one or more of the foregoing lipids may be excluded from the LNPs of the pharmaceutical compositions disclosed herein. In some aspects, a RSV RNA-LNP composition comprises a cationic lipid. The cationic lipid may comprise any one or more cationic lipids disclosed herein. The cationic lipid may comprise any one or more cationic lipids disclosed herein. In specific aspects, the cationic lipid comprises ((4-hydroxybutyl)azanediyl)bis(hexane-6,1- diyl)bis(2-hexyldecanoate) (ALC-0315). In some aspects, the cationic lipid (e.g., ALC-0315) is or is not included in the composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, 0.5, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.6, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.7, 0.71, 0.72, 0.73, 0.74, 0.75, 0.76, 0.77, 0.78, 0.79, 0.8, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.9, 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, 0.99, 1, 1.01, 1.02, 1.03, 1.04, 1.05, 1.06, 1.07, 1.08, 1.09, 1.1, 1.11, 1.12, 1.13, 1.14, 1.15, 1.16, 1.17, 1.18, 1.19, 1.2, 1.21, 1.22, 1.23, 1.24, 1.25, 1.26, 1.27, 1.28, 1.29, 1.3, 1.31, 1.32, 1.33, 1.34, 1.35, 1.36, 1.37, 1.38, 1.39, 1.4, 1.41, 1.42, 1.43, 1.44, 1.45, 1.46, 1.47, 1.48, 1.49, 1.5, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.6, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.7, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.8, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, 1.9, 1.91, 1.92, 1.93, 1.94, 1.95, 1.96, 1.97, 1.98, 1.99, 2, 2.01, 2.02, 2.03, 2.04, 2.05, 2.06, 2.07, 2.08, 2.09, 2.1, 2.11, 2.12, 2.13, 2.14, 2.15, 2.16, 2.17, 2.18, 2.19, 2.2, 2.21, 2.22, 2.23, 2.24, 2.25, 2.26, 2.27, 2.28, 2.29, 2.3, 2.31, 2.32, 2.33, 2.34, 2.35, 2.36, 2.37, 2.38, 2.39, 2.4, 2.41, 2.42, 2.43, 2.44, 2.45, 2.46, 2.47, 2.48, 2.49, 2.5, 2.51, 2.52, 2.53, 2.54, 2.55, 2.56, 2.57, 2.58, 2.59, 2.6, 2.61, 2.62, 2.63, 2.64, 2.65, 2.66, 2.67, 2.68, 2.69, 2.7, 2.71, 2.72, 2.73, 2.74, 2.75, 2.76, 2.77, 2.78, 2.79, 2.8, 2.81, 2.82, 2.83, 2.84, 2.85, 2.86, 2.87, 2.88, 2.89, 2.9, 2.91, 2.92, 2.93, 2.94, 2.95, 2.96, 2.97, 2.98, 2.99 ng/µg/mg per mL. In some aspects, the cationic lipid (e.g., ALC-0315) is or is not included in the composition at a concentration of at least, at most, between (inclusive or exclusive) of, or 2, 2.01, 2.02, 2.03, 2.04, 2.05, 2.06, 2.07, 2.08, 2.09, 2.1, 2.11, 2.12, 2.13, 2.14, 2.15, 2.16, 2.17, 2.18, 2.19, 2.2, 2.21, 2.22, 2.23, 2.24, 2.25, 2.26, 2.27, 2.28, 2.29, 2.3, 2.31, 2.32, 2.33, 2.34, 2.35, 2.36, 2.37, 2.38, 2.39, 2.4, 2.41, 2.42, 2.43, 2.44, 2.45, 2.46, 2.47, 2.48, 2.49, 2.5, 2.51, 2.52, 2.53, 2.54, 2.55, 2.56, 2.57, 2.58, 2.59, 2.6, 2.61, 2.62, 2.63, 2.64, 2.65, 2.66, 2.67, 2.68, 2.69, 2.7, 2.71, 2.72, 2.73, 2.74, 2.75, 2.76, 2.77, 2.78, 2.79, 2.8, 2.81, 2.82, 2.83, 2.84, 2.85, 2.86, 2.87, 2.88, 2.89, 2.9, 2.91, 2.92, 2.93, 2.94, 2.95, 2.96, 2.97, 2.98, 2.99 mg/mL. In some aspects, the cationic lipid (e.g., ALC-0315) is included in the composition at a concentration of at least 2, at least 2.1, at least 2.2, at least 2.3, at least 2.4, at least 2.5, at least 2.6, at least 2.7, at least 2.8, at least 2.9 mg/mL. In some aspects, the cationic lipid (e.g., ALC-0315) is included in the composition at a concentration of between 2.1 and 2.2, between 2.2 and 2.3, between 2.3 and 2.4, between 2.4 and 2.5, between 2.5 and 2.6, between 2.6 and 2.7, between 2.7 and 2.8, between 2.8 and 2.9, or between 2.9 and 3 mg/mL. In some aspects, the cationic lipid (e.g., ALC-0315) is included in the composition at a concentration of between 2 and 2.05, between 2.05 and 2.10, between 2.10 and 2.15, between 2.15 and 2.20, between 2.20 and 2.25, between 2.25 and 2.30, between 2.30 and 2.35, between 2.35 and 2.40, between 2.40 and 2.45, between 2.45 and 2.50, between 2.50 and 2.55, between 2.55 and 2.60, between 2.60 and 2.65, between 2.65 and 2.70, between 2.70 and 2.75, between 2.75 and 2.80, between 2.80 and 2.85, between 2.85 and 2.90, between 2.90 and 2.95 or between 2.95 and 3 mg/mL. In specific aspects, the cationic lipid (e.g., ALC-0315) is included in the composition at a concentration of or of about 2.58 mg/mL. In some aspects, the cationic lipid (e.g., ALC-0315) is or is not included in the composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, 0.5, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.6, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.7, 0.71, 0.72, 0.73, 0.74, 0.75, 0.76, 0.77, 0.78, 0.79, 0.8, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.9, 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, 0.99, or 1 mg/mL. In some aspects, the cationic lipid (e.g., ALC- 0315) is included in the composition at a concentration of at least 0.4, at least 0.45, at least 0.5, at least 0.55, at least 0.6, at least 0.65, at least 0.7, at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95, or at least 1 mg/mL. In some aspects, the cationic lipid (e.g., ALC-0315) is included in the composition at a concentration of between 0.4 and 0.5, between 0.5 and 0.6, between 0.6 and 0.7, between 0.7 and 0.8, between 0.8 and 0.9, or between 0.9 and 1. In some aspects, the cationic lipid (e.g., ALC-0315) is included in the composition at a concentration of between 0.4 and 0.45, between 0.45 and 0.5, between 0.5 and 0.55, between 0.55 and 0.6, between 0.6 and 0.65, between 0.65 and 0.7, between 0.7 and 0.75, between 0.75 and 0.8, between 0.8 and 0.85, between 0.85 and 0.9, between 0.9 and 0.95, or between 0.95 and 1 mg/mL. In specific aspects, the cationic lipid (e.g., ALC-0315) is included in the composition at a concentration of 0.8 to 0.95 mg/mL. In specific aspects, the cationic lipid (e.g., ALC-0315) is included in the composition at a concentration of or of about 0.8 to 0.9 mg/mL. In specific aspects, the cationic lipid (e.g., ALC-0315) is included in the composition at a concentration of or of about 0.85 to 0.9 mg/mL. In specific aspects, the cationic lipid (e.g., ALC-0315) is or is not included in the composition at a concentration of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 0.8, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.9, 0.91, 0.92, 0.93, 0.94, or 0.95 mg/mL. In specific aspects, the cationic lipid (e.g., ALC-0315) is included in the composition at a concentration of or of about 0.86 mg/mL. Concentrations for lyophilized compositions are determined post-reconstitution. In some aspects, a RSV RNA-LNP composition further comprises a PEGylated lipid (e.g., PEG-lipid). The PEGylated lipid may comprise any one or more PEGylated lipids disclosed herein. In specific aspects, the PEGylated lipid comprises 2-[(polyethylene glycol)-2000]-N,N- ditetradecylacetamide (ALC-0159). In some aspects, the PEGylated lipid (e.g., ALC-0159) is or is not included in the composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, 0.5, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.6, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.7, 0.71, 0.72, 0.73, 0.74, 0.75, 0.76, 0.77, 0.78, 0.79, 0.8, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.9, 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, 0.99, 1, 1.01, 1.02, 1.03, 1.04, 1.05, 1.06, 1.07, 1.08, 1.09, 1.1, 1.11, 1.12, 1.13, 1.14, 1.15, 1.16, 1.17, 1.18, 1.19, 1.2, 1.21, 1.22, 1.23, 1.24, 1.25, 1.26, 1.27, 1.28, 1.29, 1.3, 1.31, 1.32, 1.33, 1.34, 1.35, 1.36, 1.37, 1.38, 1.39, 1.4, 1.41, 1.42, 1.43, 1.44, 1.45, 1.46, 1.47, 1.48, 1.49, 1.5, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.6, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.7, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.8, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, 1.9, 1.91, 1.92, 1.93, 1.94, 1.95, 1.96, 1.97, 1.98, 1.99, or 2 ng/µg/mg per mL. In some aspects, the PEGylated lipid (e.g., ALC-0159) is or is not included in the composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, or 0.5 mg/mL. In some aspects, the PEGylated lipid (e.g., ALC-0159) is included in the composition at a concentration of at least 0.01, at least 0.05, at least 0.1, at least 0.15, at least 0.2, at least 0.25 mg/mL, at least 0.3 mg/mL, at least 0.35 mg/mL, at least 0.4 mg/mL, at least 0.45 mg/mL, or at least 0.5 mg/mL. In some aspects, the PEGylated lipid (e.g., ALC-0159) is included in the composition at a concentration of between 0.01 and 0.05, between 0.05 and 0.1, between 0.1 and 0.15, between 0.15 and 0.2, between 0.2 and 0.25, between 0.25 and 0.3, between 0.3 and 0.35, or between 0.35 and 0.4 mg/mL. In specific aspects, the PEGylated lipid (e.g., ALC-0159) is included in the composition at a concentration of or of about 0.32 mg/mL. In specific aspects, the PEGylated lipid (e.g., ALC-0159) is included in the composition at a concentration of or of about 0.05 to 0.15 mg/mL. In specific aspects, the PEGylated lipid (e.g., ALC-0159) is included in the composition at a concentration of or of about 0.10 to 0.15 mg/mL. In specific aspects, the PEGylated lipid (e.g., ALC-0159) is or is not included in the composition at a concentration of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.11, 0.12, 0.13, 0.14, or 0.15 mg/mL. In specific aspects, the PEGylated lipid (e.g., ALC-0159) is included in the composition at a concentration of or of about 0.11 mg/mL. Concentrations for lyophilized compositions are determined post- reconstitution. In some aspects, a RSV RNA-LNP composition further comprises one or more structural lipids. The one or more structural lipids may comprise any one or more structural lipids disclosed herein. In specific aspects, the one or more structural lipids comprise a neutral lipid and a steroid or steroid analog. In specific aspects, the one or more structural lipids comprise 1,2-distearoyl- sn-glycero-3-phosphocholine (DSPC) and cholesterol. In some aspects, the one or more structural lipids (e.g., DSPC or cholesterol) are or are not included in the composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, 0.5, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.6, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.7, 0.71, 0.72, 0.73, 0.74, 0.75, 0.76, 0.77, 0.78, 0.79, 0.8, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.9, 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, 0.99, 1, 1.01, 1.02, 1.03, 1.04, 1.05, 1.06, 1.07, 1.08, 1.09, 1.1, 1.11, 1.12, 1.13, 1.14, 1.15, 1.16, 1.17, 1.18, 1.19, 1.2, 1.21, 1.22, 1.23, 1.24, 1.25, 1.26, 1.27, 1.28, 1.29, 1.3, 1.31, 1.32, 1.33, 1.34, 1.35, 1.36, 1.37, 1.38, 1.39, 1.4, 1.41, 1.42, 1.43, 1.44, 1.45, 1.46, 1.47, 1.48, 1.49, 1.5, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.6, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.7, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.8, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, 1.9, 1.91, 1.92, 1.93, 1.94, 1.95, 1.96, 1.97, 1.98, 1.99, or 2 ng/µg/mg per mL. In some aspects, the one or more structural lipids (e.g., DSPC or cholesterol) are or are not included in the composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, 0.5, 0.51. 0.52. 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59 or 0.6 mg/mL. In some aspects, the one or more structural lipids (e.g., DSPC or cholesterol) are included in the composition at a concentration of at least .05, at least 0.1, at least 0.15, at least 0.2, at least 0.25, at least 0.3, at least 0.35, at least 0.4, at least 0.45, at least 0.5, at least 0.55, at least 0.6, at least 0.65, at least 0.7, at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or at least 1 mg/mL. In some aspects, the one or more structural lipids (e.g., DSPC or cholesterol) are included in the composition at a concentration of between 0.05 and 0.1, between 0.1 and 0.15, between 0.15 and 0.2, between 0.2 and 0.25, between 0.25 and 0.3, between 0.3 and 0.35, between 0.35 and 0.4, between 0.4 and 0.45, between 0.45 and 0.5, between 0.5 and 0.55, between 0.55 and 0.6, between 0.6 and 0.65, between 0.65 and 0.7, between 0.7 and 0.75, between 0.75 and 0.8, between 0.8 and 0.85, between 0.85 and 0.9, between 0.9 and 0.95, or between 0.95 and 1 mg/mL. In specific aspects, the one or more structural lipids include DSPC, and the DSPC is included in the composition at a concentration of or of about 0.1 to 0.25 mg/mL. In specific aspects, the one or more structural lipids include DSPC, and the DSPC is included in the composition at a concentration of or of about 0.15 to 0.25 mg/mL. In specific aspects, the one or more structural lipids include DSPC, and the DSPC is or is not included in the composition at a concentration of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, or 0.25 mg/mL. In specific aspects, the DSPC is included in the composition at a concentration of or of about 0.19 mg/mL. In specific aspects, the DSPC is included in the composition at a concentration of or of about 0.56 mg/mL. In specific aspects, the one or more structural lipids include cholesterol, and the cholesterol is included in the composition at a concentration of or of about 0.3 to 0.45 mg/mL. In specific aspects, the one or more structural lipids include cholesterol, and the cholesterol is included in the composition at a concentration of or of about 0.3 to 0.4. In specific aspects, the one or more structural lipids include cholesterol, and the cholesterol is included in the composition at a concentration of or of about 0.35 to 0.45. In specific aspects, the one or more structural lipids include cholesterol, and the cholesterol is or is not included in the composition at a concentration of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 0.30, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.40, 0.41, 0.42, 0.43, 0.44, or 0.45 mg/mL. In specific aspects, the cholesterol is included in the composition at a concentration of and/or of about 0.37 mg/mL. Concentrations for lyophilized compositions are determined post- reconstitution. In specific aspects, the one or more structural lipids include cholesterol, and the cholesterol is included in the composition at a concentration of or of about 1 to 1.25 mg/mL. In specific aspects, the one or more structural lipids include cholesterol, and the cholesterol is included in the composition at a concentration of or of about 1.10 to 1.15. In specific aspects, the one or more structural lipids include cholesterol, and the cholesterol is included in the composition at a concentration of or of about 1.15 to 1.20. In specific aspects, the one or more structural lipids include cholesterol, and the cholesterol is or is not included in the composition at a concentration of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 1, 1.01, 1.02, 1.03, 1.04, 1.05, 1.06, 1.07, 1.08, 1.09, 1.10, 1.11, 1.12, 1.13, 1.14, 1.15, 1.16, 1.17, 1.18, 1.19, 1.20, 1.21, 1.22, 1.23, 1.24 or 1.25 mg/mL. In specific aspects, the cholesterol is included in the composition at a concentration of and/or of about 1.12 mg/mL. Concentrations for lyophilized compositions are determined post-reconstitution. In some aspects, the RSV RNA-LNP composition further comprises one or more buffers and stabilizing agents, and optionally, salt diluents. Thus, in some aspects, the RSV RNA-LNP composition comprises a cationic lipid, a PEGylated lipid, one or more structural lipids, one or more buffers, a stabilizing agent, and optionally, a salt diluent. In some aspects, 1, 2, 3, or more of the foregoing elements are excluded from the RSV RNA-LNP composition. In some aspects, a RSV RNA-LNP composition comprises one or more buffers. The one or more buffers may comprise any one or more buffering agents disclosed herein. In specific aspects, the composition comprises a Tris buffer comprising at least a first buffer and a second buffer. In some aspects, the first buffer is tromethamine. In some aspects, the second buffer is Tris hydrochloride (HCl). In some aspects, the first buffer and second buffer of the Tris buffer (e.g., tromethamine and Tris HCl) are or are not included in the composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, 0.5, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.6, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.7, 0.71, 0.72, 0.73, 0.74, 0.75, 0.76, 0.77, 0.78, 0.79, 0.8, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.9, 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, 0.99, 1, 1.01, 1.02, 1.03, 1.04, 1.05, 1.06, 1.07, 1.08, 1.09, 1.1, 1.11, 1.12, 1.13, 1.14, 1.15, 1.16, 1.17, 1.18, 1.19, 1.2, 1.21, 1.22, 1.23, 1.24, 1.25, 1.26, 1.27, 1.28, 1.29, 1.3, 1.31, 1.32, 1.33, 1.34, 1.35, 1.36, 1.37, 1.38, 1.39, 1.4, 1.41, 1.42, 1.43, 1.44, 1.45, 1.46, 1.47, 1.48, 1.49, 1.5, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.6, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.7, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.8, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, 1.9, 1.91, 1.92, 1.93, 1.94, 1.95, 1.96, 1.97, 1.98, 1.99, or 2 ng/µg/mg per mL. Concentrations for lyophilized compositions are determined post-reconstitution. In some aspects, the RSV RNA-LNP composition is a liquid composition comprising a Tris buffer. In some aspects, the Tris buffer comprises a first buffer. In some aspects, the first buffer is tromethamine. In some aspects, the first buffer (e.g., tromethamine) is or is not included in the liquid composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, or 0.5 mg/mL. In some aspects, the first buffer (e.g., tromethamine) is included in the liquid composition at a concentration of at least 0.01, at least 0.05, at least 0.1, at least 0.15, at least 0.2, at least 0.25, at least 0.3, at least 0.35, at least 0.4, at least 0.45, at least 0.5, at least 0.55, at least 0.6, at least 0.65, at least 0.7, at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95, or at least 1 mg/mL. In some aspects, the first buffer (e.g., tromethamine) is included in the liquid composition at a concentration of between 0.05 and 0.15, between 0.15 and 0.25, between 0.25 and 0.35, between 0.35 and 0.45, between 0.45 and 0.55, between 0.55 and 0.65, between 0.65 and 0.75, between 0.75 and 0.85, or between 0.85 and 0.95. In some aspects, the first buffer (e.g., tromethamine) is included in the liquid composition at a concentration of between 0.05 and 0.1, between 0.1 and 0.15, between 0.15 and 0.2, between 0.2 and 0.25, between 0.25 and 0.3, between 0.3 and 0.35, between 0.35 and 0.4, between 0.4 and 0.45, between 0.45 and 0.5, between 0.5 and 0.55, between 0.55 and 0.6, between 0.6 and 0.65, between 0.65 and 0.7, between 0.7 and 0.75, between 0.75 and 0.8, between 0.8 and 0.85, between 0.85 and 0.9, between 0.9 and 0.95, or between 0.95 and 1 mg/mL. In specific aspects, the first buffer (e.g., tromethamine) is included in the liquid composition at a concentration of or of about 0.1 to 0.3 mg/mL. In specific aspects, the first buffer (e.g., tromethamine) is included in the liquid composition at a concentration of or of about 0.15 to 0.25 mg/mL. In specific aspects, the first buffer (e.g., tromethamine) is or is not included in the liquid composition at a concentration of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, or 0.3 mg/mL. In specific aspects, the first buffer (e.g., tromethamine) is included in the liquid composition at a concentration of or of about 0.20 mg/mL. In some aspects, the RSV RNA-LNP composition is a liquid composition comprising a Tris buffer comprising a second buffer. In some aspects, the second buffer comprises Tris HCl. In some aspects, the second buffer (e.g., Tris HCl) is or is not included in the liquid composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 0.5, 0.55, 1, 1.01, 1.02, 1.03, 1.04, 1.05, 1.06, 1.07, 1.08, 1.09, 1.1, 1.11, 1.12, 1.13, 1.14, 1.15, 1.16, 1.17, 1.18, 1.19, 1.2, 1.21, 1.22, 1.23, 1.24, 1.25, 1.26, 1.27, 1.28, 1.29, 1.3, 1.31, 1.32, 1.33, 1.34, 1.35, 1.36, 1.37, 1.38, 1.39, 1.4, 1.41, 1.42, 1.43, 1.44, 1.45, 1.46, 1.47, 1.48, 1.49, or 1.5 mg/mL. In some aspects, the second buffer (e.g., Tris HCl) is included in the liquid composition at a concentration of at least 0.5, at least 0.55, at least 0.6, at least 0.65, at least 0.7, at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95, at least 1, at least 1.05, at least 1.10, at least 1.15, at least 1.20, at least 1.25, at least 1.30, at least 1.35, at least 1.40, at least 1.45, or at least 1.50 mg/mL. In some aspects, the second buffer (e.g., Tris HCl) is included in the liquid composition at a concentration of between 0.5 and 0.6, between 0.6 and 0.7, between 0.7 and 0.8, between 0.8 and 0.9, between 0.9 and 1, between 1 and 1.10, between 1.10 and 1.20, between 1.20 and 1.30, between 1.30 and 1.40, or between 1.40 and 1.50 mg/mL. In specific aspects, the second buffer (e.g., Tris HCl) is included in the liquid composition at a concentration of or of about 1.25 to 1.40 mg/mL. In specific aspects, the second buffer (e.g., Tris HCl) is included in the liquid composition at a concentration of or of about 1.30 to 1.40 mg/mL. In specific aspects, the second buffer (e.g., Tris HCl) is or is not included in the liquid composition at a concentration of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 1.25, 1.26, 1.27, 1.28, 1.29, 1.30, 1.31, 1.32, 1.33, 1.34, or 1.35, 1.36, 1.37, 1.38, 1.39, or 1.40 mg/mL. In one specific aspect, the second buffer (e.g., Tris HCl) is included in the liquid composition at a concentration of or of about 1.32 mg/mL. In some aspects, the RSV RNA-LNP composition is a lyophilized composition comprising a Tris buffer. In some aspects, the Tris buffer comprises a first buffer. In some aspects, the first buffer is tromethamine. In some aspects, the first buffer (e.g., tromethamine) is or is not included in the lyophilized composition at a concentration, after reconstitution, of at least, at most, between (inclusive or exclusive) of, or exactly 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, or 0.5 mg/mL. In some aspects, the first buffer (e.g., tromethamine) is included in the lyophilized composition at a concentration, after reconstitution, of at least 0.01, of at least 0.05, of at least 0.1, of at least 0.15, of at least 0.2, of at least 0.25, of at least 0.3, of at least 0.35, of at least 0.4, of at least 0.45, or of at least 0.5 mg/mL. In some aspects, the first buffer (e.g., tromethamine (Tris base)) is included in the lyophilized composition at a concentration, after reconstitution, of between 0.01 and 0.05, between 0.05 and 0.1, between 0.1 and 0.15, between 0.15 and 0.2, between 0.2 and 0.25 mg/mL, between 0.25 and 0.3 mg/mL, between 0.3 and 0.35 mg/mL, between 0.35 and 0.4 mg/mL, between 0.4 and 0.45 mg/mL, or between 0.45 and 0.5 mg/mL. In specific aspects, the first buffer (e.g., tromethamine) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 0.01 and 0.20 mg/mL. In specific aspects, the first buffer (e.g., tromethamine) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 0.01 and 0.10 mg/mL. In specific aspects, the first buffer (e.g., tromethamine) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 0.05 and 0.15 mg/mL. In specific aspects, the first buffer (e.g., tromethamine) is or is not included in the lyophilized composition at a concentration, after reconstitution, of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19 or 0.20 mg/mL. In one specific aspect, the first buffer (e.g., tromethamine) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 0.09 mg/mL. In one specific aspect, the first buffer (e.g., tromethamine) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 0.09 mg/mL. In some aspects, the RSV RNA-LNP composition is a lyophilized composition comprising a Tris buffer comprising a second buffer. In some aspects, the second buffer comprises Tris HCl. In some aspects, the second buffer (e.g., Tris HCl) is or is not included in the lyophilized composition at a concentration, after reconstitution, of at least, at most, between (inclusive or exclusive) of, or exactly 0.1, 0.15, 0.2, 0.25, 0.3, 0.35, 0.4, 0.45, 0.5, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.6, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.7, 0.71, 0.72, 0.73, 0.74, 0.75, 0.76, 0.77, 0.78, 0.79, 0.8, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.9, 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, 0.99, or 1 mg/mL. In some aspects, the second buffer (e.g., Tris HCl) is included in the lyophilized composition at a concentration, after reconstitution, of at least 0.1, at least 0.2, at least 0.3, at least 0.4, at least 0.5, at least 0.6, at least 0.7, at least 0.8, at least 0.9, or at least 1 mg/mL. In some aspects, the second buffer (e.g., Tris HCl) is included in the lyophilized composition at a concentration, after reconstitution, of between 0.1 and 0.2, between 0.2 and 0.3, between 0.3 and 0.4, between 0.4 and 0.5, between 0.5 and 0.6, between 0.6 and 0.7, between 0.7 and 0.8, between 0.8 and 0.9, or between 0.9 and 1 mg/mL. In specific aspects, the second buffer (e.g., Tris HCl) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 0.5 and 0.65 mg/mL. In specific aspects, the second buffer (e.g., Tris HCl) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 0.5 and 0.6 mg/mL. In specific aspects, the second buffer (e.g., Tris HCl) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 0.55 and 0.65 mg/mL. In specific aspects, the second buffer (e.g., Tris HCl) is or is not included in the lyophilized composition at a concentration, after reconstitution, of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 0.5, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.6, 0.61, 0.62, 0.63, 0.64, or 0.65 mg/mL. In specific aspects, the second buffer (e.g., Tris HCl) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 0.57 mg/mL. In some aspects, a RSV RNA-LNP composition comprises a stabilizing agent. The stabilizing agent may comprise any one or more stabilizing agents disclosed herein. In some aspects, the stabilizing agent also functions as a cryoprotectant. In specific aspects, the stabilizing agent comprises sucrose. In some aspects, the stabilizing agent (e.g., sucrose) is or is not included in the composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, or 200 ng/µg/mg per mL. In some aspects, the RSV RNA-LNP composition is a liquid composition, and the stabilizing agent (e.g., sucrose) is or is not included in the liquid composition at a concentration of at least, at most, between (inclusive or exclusive) of, or exactly 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, or 130 mg/mL. In some aspects, the stabilizing agent (e.g., sucrose) is included in the liquid composition at a concentration of at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 105, at least 110, at least 115, at least 120, at least 125, or at least 130 mg/mL. In some aspects, the stabilizing agent (e.g., sucrose) is included in the liquid composition at a concentration of between 70 and 80, between 80 and 90, between 90 and 100, between 100 and 110, between 110 and 120, or between 120 and 130 mg/mL. In specific aspects, the stabilizing agent (e.g., sucrose) is included in the liquid composition at a concentration of or of about 95 to 110 mg/mL. In specific aspects, the stabilizing agent (e.g., sucrose) is included in the liquid composition at a concentration of or of about 95 to 105 mg/mL. In specific aspects, the stabilizing agent (e.g., sucrose) is included in the liquid composition at a concentration of or of about 100 to 110 mg/mL. In specific aspects, the stabilizing agent (e.g., sucrose) is or is not included in the liquid composition at a concentration of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, or 110 mg/mL. In specific aspects, the stabilizing agent (e.g., sucrose) is included in the liquid composition at a concentration of or of about 103 mg/mL. In some aspects, the RSV RNA-LNP composition is a lyophilized composition, and the stabilizing agent (e.g., sucrose) is or is not included in the lyophilized composition at a concentration, after reconstitution, of at least, at most, between (inclusive or exclusive) of, or exactly 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80 mg/mL. In some aspects, the stabilizing agent (e.g., sucrose) is included in the lyophilized composition at a concentration, after reconstitution, of at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 mg/mL. In some aspects, the stabilizing agent (e.g., sucrose) is included in the lyophilized composition at a concentration, after reconstitution, of between 20 to 30, between 30 to 40, between 40 to 50, between 50 to 60, between 60 to 70, or between 70 to 80 mg/mL. In specific aspects, the stabilizing agent (e.g., sucrose) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 35 to 50 mg/mL. In specific aspects, the stabilizing agent (e.g., sucrose) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 35 to 45 mg/mL. In specific aspects, the stabilizing agent (e.g., sucrose) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 40 to 50 mg/mL. In specific aspects, the stabilizing agent (e.g., sucrose) is or is not included in the lyophilized composition at a concentration, after reconstitution, of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 mg/mL. In specific aspects, the stabilizing agent (e.g., sucrose) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 44 /mL. In some aspects, lyophilized compositions are reconstituted in a suitable carrier and/or diluent. The carrier and/or diluent may comprise any one or more carriers and/or diluents disclosed herein. In specific aspects, the carrier and/or diluent comprises a salt diluent, such as sodium chloride (NaCl) (e.g., saline, e.g., physiological or normal saline). The sodium chloride may comprise 0.9% sodium chloride for injection. In some aspects, the lyophilized compositions are or are not reconstituted in at least, at most, between (inclusive or exclusive) of, or exactly 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.30, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.40, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, 0.50, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.60, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.70, 0.71, 0.72, 0.73, 0.74, 0.75, 0.76, 0.77, 0.78, 0.79, 0.80, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.90, 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, 0.99, or 1 mL of saline. In some aspects, the lyophilized compositions are reconstituted in at least 0.1, at least 0.2, at least 0.3, at least 0.4, at least 0.5, at least 0.6, at least 0.7, at least 0.8, at least 0.9, or at least 1 mL of sodium chloride. In specific aspects, the lyophilized compositions are reconstituted in or in about 0.6 to 0.75 mL of sodium chloride/saline. In specific aspects, the lyophilized compositions are reconstituted in or in about 0.65 to 0.75 mL of sodium chloride/saline. In specific aspects, the lyophilized compositions are or are not reconstituted in or in at least, at most, exactly, between (inclusive or exclusive) any two of, or about 0.6, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.7, 0.71, 0.72, 0.73, 0,74, or 0.75 mL of sodium chloride/saline. In some aspects, the salt diluent (e.g., NaCl) is or is not included in the lyophilized composition at a concentration, after reconstitution, of at least, at most, between (inclusive or exclusive) of, or exactly 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, 14, 14.5, 15, 15.5, 16, 16.5, 17, 17.5, 18, 18.5, 19, 19.5, 20, 20.5, 21, 21.5, 22, 22.5, 23, 23.5, 24, 24.5, 25, 25.5, 26, 26.5, 27, 27.5, 28, 28.5, 29, 29.5, 30, 30.5, 31, 31.5, 32, 32.5, 33, 33.5, 34, 34.5, 35, 35.5, 36, 36.5, 37, 37.5, 38, 38.5, 39, 39.5, 40, 40.5, 41, 41.5, 42, 42.5, 43, 43.5, 44, 44.5, 45, 45.5, 46, 46.5, 47, 47.5, 48, 48.5, 49, 49.5, or 50 ng/µg/mg per mL. In some aspects, the salt diluent (e.g., NaCl) is or is not included in the lyophilized composition at a concentration, after reconstitution, of in at least, at most, between (inclusive or exclusive) of, or exactly 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, 14, 14.5, 15, 15.5, 16, 16.5, 17, 17.5, 18, 18.5, 19, 19.5, or 20 mg/mL. In some aspects, the salt diluent (e.g., NaCl) is included in the lyophilized composition at a concentration, after reconstitution, of at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, or at least 20 mg/mL. In specific aspects, the salt diluent (e.g., NaCl) is included in the lyophilized composition at a concentration, after reconstitution, of between or between about 5 and 15 mg/mL. In some aspects, the salt diluent (e.g., NaCl) is included in the lyophilized composition at a concentration, after reconstitution, of between or between about 5 and 10 mg/mL. In specific aspects, the salt diluent (e.g., NaCl) is or is not included in the lyophilized composition at a concentration, after reconstitution, of or of at least, at most, exactly, between (inclusive or exclusive) any two of, or about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 mg/mL. In specific aspects, the salt diluent (e.g., NaCl) is included in the lyophilized composition at a concentration, after reconstitution, of or of about 9 mg/mL. The pH of the RSV RNA-LNP composition may or may not be at least, at most, exactly, or between (inclusive or exclusive) of pH 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, 8.1, 8.2, 8.3, 8.4, or 8.5, or any range or value derivable therein. In some aspects, the RSV RNA-LNP composition is at a pH of at least 6.5, at least 7.0, at least 7.5, at least 8.0, or at least 8.5. In specific aspects, the RSV RNA-LNP composition is at a pH between 6.0 and 7.5, between 6.5 and 7.5, between 7.0 and 8.0, between and 7.5 and 8.5. In specific aspects, the RSV RNA-LNP composition is between 7.0 and 8.0. In specific aspects, the RSV RNA-LNP composition is or is not at least, at most, exactly, between (inclusive or exclusive) any two of, or about pH 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0. In specific aspects, the RSV RNA-LNP composition is at or at about pH 7.4. In some aspects, sodium hydroxide buffer may be used for a buffer pH adjustment. In specific aspects, a RSV RNA-LNP composition comprises a RSV RNA polynucleotide encoding a RSV polypeptide as disclosed herein, encapsulated in LNPs with a lipid composition of a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL, a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL, a first structural lipid (e.g. DSPC) at a concentration of or of about 0.1 to 0.25 mg/mL, and a second structural lipid (e.g. cholesterol) at a concentration of or of about 0.3 to 0.45 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. In specific aspects, a RSV RNA-LNP composition comprises a RSV RNA polynucleotide encoding a RSV polypeptide as disclosed herein, encapsulated in LNPs with a lipid composition of ALC-0315 at a concentration of or of about 0.8 to 0.95 mg/mL, ALC-0159 at a concentration of or of about 0.05 to 0.15 mg/mL, DSPC at a concentration of or of about 0.1 to 0.25 mg/mL, and cholesterol at a concentration of about 0.3 to 0.45 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. In specific aspects, the RSV RNA-LNP composition is a liquid RSV RNA-LNP composition, and the liquid RSV RNA-LNP composition further comprises a buffer composition comprising a first buffer at a concentration of or of about 0.15 to 0.3 mg/mL, a second buffer at a concentration of or of about 1.25 to 1.4 mg/mL, and a stabilizing agent at a concentration of or of about 95 to 110 mg/mL. In specific aspects, the RSV RNA-LNP composition is a liquid RSV RNA- LNP composition, and the liquid RSV RNA-LNP composition further comprises a Tris buffer composition comprising tromethamine at a concentration of or of about 0.1 to 0.3 mg/mL, Tris HCl at a concentration of or of about 1.25 to 1.4 mg/mL, and sucrose at a concentration of or of about 95 to 110 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. Thus, in specific aspects, a liquid RSV RNA-LNP composition comprises an cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL, a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL, a first structural lipid at a concentration of or of about 0.1 to 0.25 mg/mL, a second structural lipid at a concentration of or of about 0.3 to 0.45 mg/mL, and further comprises a first buffer at a concentration of or of about 0.1 to 0.3 mg/mL, a second buffer at a concentration of or of about 1.25 to 1.4 mg/mL, and a stabilizing agent at a concentration of or of about 95 to 110 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. Thus, in specific aspects, a liquid RSV RNA-LNP composition comprises ALC-0315 at a concentration of or of about 0.8 to 0.95 mg/mL, ALC-0159 at a concentration of or of about 0.05 to 0.15 mg/mL, DSPC at a concentration of or of about 0.1 to 0.25 mg/mL, cholesterol at a concentration of or of about 0.3 to 0.45 mg/mL, and further comprises a Tris buffer composition comprising tromethamine at a concentration of or of about 0.1 to 0.3 mg/mL, Tris HCl at a concentration of or of about 1.25 to 1.4 mg/mL, and sucrose at a concentration of or of about 95 to 110 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. In specific aspects, the RSV RNA-LNP composition is a lyophilized RSV RNA-LNP composition, and the lyophilized RSV RNA-LNP composition further comprises (after reconstitution) a first buffer at a concentration of or of about 0.01 and 0.15 mg/mL, a second buffer at a concentration of or of about 0.5 and 0.65 mg/mL, a stabilizing agent at a concentration of or of about 35 to 50 mg/mL, and a salt diluent at a concentration of between or between about 5 and 15 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. In specific aspects, the RSV RNA-LNP composition is a lyophilized RSV RNA-LNP composition, and the lyophilized RSV RNA-LNP composition further comprises (after reconstitution) a Tris buffer composition comprising tromethamine at a concentration of or of about 0.01 and 0.15 mg/mL, Tris HCl at a concentration of or of about 0.5 and 0.65 mg/mL, sucrose at a concentration of or of about 35 to 50 mg/mL, and sodium chloride (NaCl) at a concentration of or of about 5 to 15 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. Thus, in specific aspects, a lyophilized RSV RNA-LNP composition comprises (after reconstitution) a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL, a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL, a first structural lipid at a concentration of or of about 0.1 to 0.25 mg/mL, a second structural lipid at a concentration of or of about 0.3 to 0.45 mg/mL, and further comprises a first buffer at a concentration of or of about 0.01 and 0.15 mg/mL, a second buffer at a concentration of or of about 0.5 and 0.65 mg/mL, a stabilizing agent at a concentration of or of about 35 to 50 mg/mL, and a salt diluent at a concentration of or of about 5 to 15 mg/mL. In specific aspects, the lyophilized compositions are reconstituted in 0.6 to 0.75 mL of the salt diluent. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. Thus, in some aspects, a lyophilized RSV RNA-LNP composition comprises (after reconstitution) ALC-0315 at a concentration of or of about 0.8 to 0.95 mg/mL, ALC-0159 at a concentration of or of about 0.05 to 0.15 mg/mL, DSPC at a concentration of or of about 0.1 to 0.25 mg/mL, cholesterol at a concentration of or of about 0.3 to 0.45 mg/mL, and further comprises tromethamine at a concentration of or of about 0.01 and 0.15 mg/mL, Tris HCl at a concentration of or of about 0.5 and 0.65 mg/mL, sucrose at a concentration of or of about 35 to 50 mg/mL, and NaCl at a concentration of or of about 5 to 15 mg/mL. In specific aspects, the lyophilized compositions are reconstituted in 0.6 to 0.75 mL of NaCl (saline). In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. Concentrations in the lyophilized RSV RNA-LNP composition above are determined post- reconstitution. In some aspects, a RSV RNA-LNP composition (pre-lyophilization) comprises a cationic lipid at a concentration of or of about 1.0 to 3.0 mg/mL, a PEGylated lipid at a concentration of or of about 0.10 to 0.35 mg/mL, a first structural lipid at a concentration of or of about 0.4 to 0.55 mg/mL, a second structural lipid at a concentration of or of about 0.85 to 1.0 mg/mL, and further comprises a first buffer at a concentration of or of about 0.1 and 0.3 mg/mL, a second buffer at a concentration of or of about 1.25 and 1.40 mg/mL, a stabilizing agent at a concentration of or of about 95 to 110 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. Thus, in some aspects, a RSV RNA-LNP composition (pre-lyophilization) comprises ALC- 0315 at a concentration of or of about 1.0 to 3.0 mg/mL, ALC-0159 at a concentration of or of about 0.10 to 0.35 mg/mL, DSPC at a concentration of or of about 0.4 to 0.55 mg/mL, cholesterol at a concentration of or of about 0.85 to 1.0 mg/mL, and further comprises tromethamine at a concentration of or of about 0.1 and 0.3 mg/mL, Tris HCl at a concentration of or of about 1.25 and 1.40 mg/mL, sucrose at a concentration of or of about 95 to 110 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. The RSV RNA-LNP compositions further comprise RSV RNA described herein encapsulated in LNPs. In specific aspects, a RSV RNA-LNP composition is a liquid RSV RNA-LNP composition comprising a RSV RNA polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition of an cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL, a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL, a first structural lipid at a concentration of or of about 0.1 to 0.25 mg/mL, and a second structural lipid at a concentration of or of about 0.3 to 0.45 mg/mL, and further comprising a buffer composition comprising a first buffer at a concentration of or of about 0.15 to 0.3 mg/mL, a second buffer at a concentration of or of about 1.25 to 1.4 mg/mL, and a stabilizing agent at a concentration of or of about 95 to 110 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. In specific aspects, a liquid RSV RNA-LNP composition comprises a RSV RNA polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably of or of about 0.06 mg/mL, encapsulated in LNPs with a lipid composition of ALC-0315 at a concentration of or of about 0.8 to 0.95 mg/mL, ALC-0159 at a concentration of or of about 0.05 to 0.15 mg/mL, DSPC at a concentration of or of about 0.1 to 0.25 mg/mL, and cholesterol at a concentration of or of about 0.3 to 0.45 mg/mL, and further comprising a Tris buffer composition comprising tromethamine at a concentration of or of about 0.1 to 0.3 mg/mL, Tris HCl at a concentration of or of about 1.25 to 1.4 mg/mL, and sucrose at a concentration of or of about 95 to 110 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. In specific aspects, the RSV RNA-LNP composition is a lyophilized RSV RNA-LNP composition comprising a RSV RNA polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition of a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL, a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL, a first structural lipid at a concentration of or of about 0.1 to 0.25 mg/mL, and a second structural lipid at a concentration of or of about 0.3 to 0.45 mg/mL, and further comprising a first buffer at a concentration of or of about 0.01 and 0.15 mg/mL, a second buffer at a concentration of or of about 0.5 and 0.65 mg/mL, a stabilizing agent at a concentration of or of about 35 to 50 mg/mL, and a salt diluent at a concentration of 5 to 15 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. In specific aspects, the lyophilized compositions are reconstituted in 0.6 to 0.75 mL of the salt diluent. Concentrations in the lyophilized RSV RNA-LNP composition are determined post-reconstitution. In specific aspects, a lyophilized RSV RNA-LNP composition comprises a RSV RNA polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably of or of about 0.06 mg/mL, encapsulated in LNPs with a lipid composition of ALC-0315 at a concentration of or of about 0.8 to 0.95 mg/mL, ALC-0159 at a concentration of or of about 0.05 to 0.15 mg/mL, DSPC at a concentration of or of about 0.1 to 0.25 mg/mL, and cholesterol at a concentration of or of about 0.3 to 0.45 mg/mL, and further comprising tromethamine at a concentration of or of about 0.01 and 0.15 mg/mL, Tris HCl at a concentration of or of about 0.5 and 0.65 mg/mL, sucrose at a concentration of or of about 35 to 50 mg/mL, and NaCl at a concentration of or of about 5 to 15 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. In specific aspects, the lyophilized compositions are reconstituted in 0.6 to 0.75 mL of the NaCl diluent (saline). Concentrations in the lyophilized RSV RNA-LNP composition are determined post-reconstitution. In some aspects, a RSV RNA-LNP composition (pre-lyophilization) comprises a RSV RNA polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in LNPs with a lipid composition of a cationic lipid at a concentration of or of about 1.0 to 3.0 mg/mL, a PEGylated lipid at a concentration of or of about 0.10 to 0.35 mg/mL, a first structural lipid at a concentration of or of about 0.4 to 0.55 mg/mL, a second structural lipid at a concentration of or of about 0.85 to 1.0 mg/mL, and further comprises a first buffer at a concentration of or of about 0.1 and 0.3 mg/mL, a second buffer at a concentration of or of about 1.25 and 1.40 mg/mL, a stabilizing agent at a concentration of or of about 95 to 110 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. Thus, in some aspects, a RSV RNA-LNP composition (pre-lyophilization) comprises a RSV RNA polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably 0.15 mg/mL, encapsulated in LNPs with a lipid composition of comprises ALC-0315 at a concentration of or of about 1.0 to 3.0 mg/mL, ALC-0159 at a concentration of or of about 0.10 to 0.35 mg/mL, DSPC at a concentration of or of about 0.4 to 0.55 mg/mL, cholesterol at a concentration of or of about 0.85 to 1.0 mg/mL, and further comprises tromethamine at a concentration of or of about 0.1 and 0.3 mg/mL, Tris HCl at a concentration of or of about 1.25 and 1.40 mg/mL, sucrose at a concentration of or of about 95 to 110 mg/mL. In some aspects, one or more of the foregoing elements may be excluded from the RSV RNA-LNP composition. In some aspects, the liquid RNA-LNP immunogenic composition comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in a LNP, and further comprising or comprising about 5 to 15 mM Tris buffer and about 200 to 400 mM sucrose at a pH of or of about 7.0 to 8.0. In some aspects, one or more of the foregoing elements may be excluded from the liquid RNA-LNP immunogenic composition. In some aspects, the liquid RNA-LNP immunogenic composition comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably of or of about 0.06 mg/mL, encapsulated in a LNP, and further comprising or comprising about 10 mM Tris buffer and 300 mM sucrose at a pH of or of about 7.4. In some aspects, one or more of the foregoing elements may be excluded from the liquid RNA-LNP immunogenic composition. In some aspects, the RNA-LNP immunogenic composition (pre-lyophilized) comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, encapsulated in a LNP, and further comprising or comprising about 5 to 15 mM Tris buffer and 200 to 400 mM sucrose at a pH of or of about 7.0 to 8.0, and reconstituted with 0.9% sodium chloride diluent. In some aspects, one or more of the foregoing elements may be excluded from the RNA-LNP immunogenic composition. In some aspects, the RNA-LNP immunogenic composition (pre-lyophilized) comprises an RNA molecule/polynucleotide encoding a RSV polypeptide as disclosed herein at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably 0.15 mg/mL, encapsulated in a LNP, and further comprising or comprising about 10 mM Tris buffer and 300 mM sucrose at a pH of or of about 7.4, and reconstituted with 0.9% sodium chloride diluent. In some aspects, one or more of the foregoing elements may be excluded from the RNA- LNP immunogenic composition. In some aspects, In some aspects, the RNA-LNP immunogenic composition (pre- lyophilized) comprises a first RNA molecule/polynucleotide encoding a RSV subtype A polypeptide as disclosed herein and a second RNA molecule/polynucleotide encoding a RSV subtype B as disclosed herein, each RNA molecule/polynucleotide at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably 0.09 mg/mL, encapsulated in LNPs with a lipid composition of a cationic lipid at a concentration of or of about 1.0 to 3.0 mg/mL, a PEGylated lipid at a concentration of or of about 0.10 to 0.35 mg/mL, a first structural lipid at a concentration of or of about 0.4 to 0.6 mg/mL, a second structural lipid at a concentration of or of about 0.85 to 1.20 mg/mL, and further comprising or comprising about 10 mM Tris buffer and 300 mM sucrose at a pH of or of about 7.4, and reconstituted with 0.9% sodium chloride diluent. In some aspects, one or more of the foregoing elements may be excluded from the RNA-LNP immunogenic composition. In some aspects, the RNA-LNP immunogenic composition (pre-lyophilized) comprises a first RNA molecule/polynucleotide encoding a first RSV subtype A polypeptide as disclosed herein and a second RNA molecule/polynucleotide encoding a RSV subtype B as disclosed herein, each RNA molecule/polynucleotide at a concentration of at least, at most, exactly, or between (inclusive or exclusive) of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL, preferably of or of about 0.01 to 0.09 mg/mL, and more preferably 0.09 mg/mL, encapsulated in LNPs with a lipid composition of ALC-0315 at a concentration of or of about 1.0 to 3.0 mg/mL, preferably about 2.56 mg/mL, ALC-0519 at a concentration of or of about 0.10 to 0.35 mg/mL, preferably about 0.32 mg/mL, DSPC at a concentration of or of about 0.4 to 0.6 mg/mL, preferably about 0.56 mg/mL, cholesterol at a concentration of or of about 0.85 to 1.20 mg/mL, preferably about 1.12 mg/mL, and further comprising or comprising about 10 mM Tris buffer and 300 mM sucrose at a pH of or of about 7.4, and reconstituted with 0.9% sodium chloride diluent. In some aspects, one or more of the foregoing elements may be excluded from the RNA-LNP immunogenic composition. B. VACCINES In some aspects, a pharmaceutical composition described herein is an immunogenic composition for inducing an immune response. For example, in some aspects, an immunogenic composition is a vaccine. In some aspects, the compositions described herein include at least one isolated nucleic acid or polypeptide molecule as described herein. In specific aspects, the immunogenic compositions comprise nucleic acids, and the immunogenic compositions are nucleic acid vaccines. In some aspects, the immunogenic compositions comprise RNA (e.g., mRNA, saRNA), and vaccines are RNA vaccines. In other aspects, the immunogenic compositions comprise DNA, and vaccines are DNA vaccines. In yet other aspects, the immunogenic compositions comprise a polypeptide, and vaccines are polypeptide vaccines. Conditions and/or diseases that may be treated with the nucleic acid and/or peptide or polypeptide compositions include, but are not limited to, those caused and/or impacted by infection, cancer, rare diseases, and other diseases or conditions caused by overproduction, underproduction, and/or improper production of protein or nucleic acids. In some aspects, the composition is substantially free of one or more impurities or contaminants and, for instance, includes nucleic acid or polypeptide molecules that are equal to at least, at most, exactly, or between (inclusive or exclusive) of 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% pure; at least 98% pure, or at least 99% pure. The present disclosure includes methods for preventing, treating and/or ameliorating an infection, disease or condition in a subject, including administering to a subject an effective amount of an RNA molecule that includes at least one open reading frame encoding a polypeptide or composition described herein. As such, the disclosure contemplates vaccines for use in both active and passive immunization aspects. Immunogenic compositions, proposed to be suitable for use as a vaccine, may be prepared from RNA molecules encoding polypeptide(s), such as RSV preF polypeptides. In certain aspects, immunogenic compositions are lyophilized for more ready formulation into a desired vehicle. The preparation of vaccines that contain nucleic acid and/or peptide or polypeptide as active ingredients is generally well understood in the art, as exemplified by U.S. Patents 4,608,251; 4,601,903; 4,599,231; 4,599,230; 4,596,792; and 4,578,770, all of which are incorporated herein by reference. Typically, such vaccines are prepared as injectables either as liquid solutions or suspensions; solid forms suitable for solution in or suspension in liquid prior to injection may also be prepared. The preparation may also be emulsified. The active immunogenic ingredient is often mixed with excipients that are pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like and combinations thereof. In addition, if desired, the vaccine may contain amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, or adjuvants that enhance the effectiveness of the vaccines. In specific aspects, vaccines are formulated with a combination of substances, as described in U.S. Patents 6,793,923 and 6,733,754, which are incorporated herein by reference. In some aspects, one or more of the foregoing elements may be excluded from a vaccine. Vaccines may be conventionally administered parenterally, by injection, for example, either subcutaneously or intramuscularly. Additional formulations which are suitable for other modes of administration include suppositories and, in some cases, oral formulations. For suppositories, traditional binders and carriers may include, for example, polyalkylene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in the range of or of about 0.5% to about 10%. In some aspects, suppositories may be formed from mixtures containing the active ingredient in the range of or of about 1% to about 2%. Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing excipients may be excluded from an oral formulation. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and contain or contain about 10% to about 95% of active ingredient. The polypeptide-encoding nucleic acid constructs and polypeptides may be formulated into a vaccine as neutral or salt forms. “Pharmaceutically acceptable salt” includes both acid and base addition salts. “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2- oxo-glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-1,5-disulfonic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2- naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing inorganic acids may be excluded. “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing inorganic bases may be excluded. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing organic bases may be excluded. The polypeptide-encoding nucleic acid constructs and polypeptides, or their pharmaceutically acceptable salts, may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. The present disclosure is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and (5)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included. A “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three- dimensional structures, which are not interchangeable. The present invention contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are non-superimposable mirror images of one another. A “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present invention disclosure tautomers of any said compounds. Compounds described herein that exist in free base or acid form can be converted to their pharmaceutically acceptable salts by treatment with the appropriate inorganic or organic base or acid by methods known to one skilled in the art. Salts of the compounds can be converted to their free base or acid form by standard techniques. It will be appreciated by those skilled in the art that in the process described herein the functional groups of intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxy, amino, mercapto and carboxylic acid. Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (for example, t- butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butoxycarbonyl, benzyloxycarbonyl, and the like. Suitable protecting groups for mercapto include -C(O)-R" (where R" is alkyl, aryl or arylalkyl), p-methoxybenzyl, trityl and the like. Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters. As one of skill in the art would appreciate, the protecting group may also be a polymer resin such as a Wang resin, Rink resin or a 2- chlorotrityl-chloride resin. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing protecting groups may be excluded. Protecting groups may be added or removed in accordance with standard techniques, which are known to one skilled in the art (see, e.g., Green, T.W. and P.G.M. Wutz, Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley) and as described herein. It will also be appreciated by those skilled in the art, although such protected derivatives of compounds of this invention may not possess pharmacological activity as such, they may be administered to a mammal and thereafter metabolized in the body to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as "prodrugs". All prodrugs of compounds of this invention are included within the scope of the invention. Typically, vaccines are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective and immunogenic. The quantity to be administered depends on the subject to be treated, including the capacity of the individual’s immune system to synthesize antibodies and the degree of protection desired. Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner. However, suitable dosage ranges are of the order of several hundred micrograms of active ingredient per vaccination. Suitable regimes for initial administration and booster shots are also variable, but are typified by an initial administration followed by subsequent inoculations and/or other administrations. The manner of application may be varied widely. Any of the conventional methods for administration of a vaccine are applicable. These are believed to include oral application within a solid physiologically acceptable base or in a physiologically acceptable dispersion, parenterally, by injection and the like. The dosage of the vaccine will depend on the route of administration and will vary according to the size and health of the subject. In certain aspects, it will be desirable to have one administration of the vaccine. In some aspects, it will be desirable to have multiple administrations of the vaccine, e.g., 2, 3, 4, 5, 6, or more administrations. The vaccinations may be at 1, 2, 3, 4, 5, 6, 7, 8, to 5, 6, 7, 8, 9 ,10, 11, or 12 twelve week intervals, including all ranges there between. In some aspects, vaccinations may be at 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 month intervals, including all ranges there between. Periodic boosters at intervals of 1-5 years may be desirable to maintain protective levels of the antibodies. i. CARRIERS A pharmaceutically acceptable carrier may include the liquid or non-liquid basis of a composition. If a composition is provided in liquid form, the carrier may be water, such as pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g. phosphate, citrate buffered solutions. Water or a buffer, such as an aqueous buffer, may be used, containing a sodium salt, a calcium salt, and and/or a potassium salt. The sodium, calcium and/or potassium salts may occur in the form of their halogenides, e.g. chlorides, iodides, or bromides, in the form of their hydroxides, carbonates, hydrogen carbonates, or sulfates, etc. Examples of sodium salts include, but are not limited to, NaCI, Nal, NaBr, Na2CO3, NaHCO3, Na2SO4, Na2HPO4, Na2HPO4·2H2O, examples of potassium salts include, but are not limited to, KCI, Kl, KBr, K2CO3, KHCO3, K2SO4, KH2PO4, and examples of calcium salts include, but are not limited to, CaCl2, Cal2, CaBr2, CaCO3, CaSO4, Ca(OH)2. Examples of further carriers may include sugars, such as, for example, lactose, glucose, trehalose and sucrose; starches, such as, for example, com starch or potato starch; dextrose; cellulose and its derivatives, such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate; powdered tragacanth; malt; gelatin; tallow; solid glidants, such as, for example, stearic acid, magnesium stearate; calcium sulfate; vegetable oils, such as, for example, groundnut oil, cottonseed oil, sesame oil, olive oil, corn oil and oil from theobroma; polyols, such as, for example, polypropylene glycol, glycerol, sorbitol, mannitol and polyethylene glycol; alginic acid. Examples of further carriers may include colloidal silicon oxide, magnesium stearate, cellulose, and sodium lauryl sulfate. Additional suitable pharmaceutical carriers and diluents, as well as pharmaceutical necessities for their use, are described in Remington’s Pharmaceutical Sciences. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing carriers may be excluded. ii. ADJUVANTS Suitable adjuvants include all acceptable immunostimulatory compounds, such as cytokines, toxins, or synthetic compositions. A number of adjuvants may be used to enhance an antibody response. Adjuvants include, but are not limited to, oil-in-water emulsions, water-in-oil emulsions, mineral salts, polynucleotides, and natural substances. Specific adjuvants that may be used include oil such as MONTANIDE® ISA51, IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL12, alpha-interferon, PTNGg, GM-CSF, GMCSP, BCG, LT-a, aluminum salts, such as aluminum hydroxide or other aluminum compound, MDP compounds, such as thur-MDP and nor- MDP, CGP (MTP-PE), lipid A, monophosphoryl lipid A (MPL), lipopeptides (e.g., Pam3Cys). RIBI, which contains three components extracted from bacteria, MPL, trehalose dimycolate (TDM), and cell wall skeleton (CWS) in a 2% squalene/Tween 80 emulsion. MHC antigens may even be used. Various methods of achieving adjuvant affect for the vaccine includes use of agents such as aluminum hydroxide or phosphate (alum), commonly used as about 0.05 to about 0.1% solution in phosphate buffered saline, admixture with synthetic polymers of sugars (CARBOPOL®) used as an about 0.25% solution, aggregation of the protein in the vaccine by heat treatment with temperatures ranging between about 70° to about 101°C for a 30-second to 2-minute period, respectively. Aggregation by reactivating with pepsin-treated (Fab) antibodies to albumin; mixture with bacterial cells (e.g., C. parvum), endotoxins or lipopolysaccharide components of Gram-negative bacteria; emulsion in physiologically acceptable oil vehicles (e.g., mannide mono-oleate (Aracel A)); or emulsion with a 20% solution of a perfluorocarbon (FLUOSOL-DA®) used as a block substitute may also be employed to produce an adjuvant effect. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing adjuvants may be excluded. In addition to adjuvants, it may be desirable to co-administer biologic response modifiers (BRM) to enhance immune responses. BRMs have been shown to upregulate T cell immunity or downregulate suppresser cell activity. Such BRMs include, but are not limited to, Cimetidine (CIM; 1200 mg/d) (Smith/Kline, PA); or low-dose Cyclophosphamide (CYP; 300 mg/m2) (Johnson/ Mead, NJ) and cytokines such as γ-interferon, IL-2, or IL-12 or genes encoding proteins involved in immune helper functions, such as B-7. C. COMBINATION THERAPY The compositions and related methods of the present disclosure, particularly administration of an RNA molecule encoding a RSV preF polypeptide, may also be used in combination with the administration of one or more other therapeutic agents. These include, but are not limited to, the administration of traditional therapies, e.g., antiviral therapies such as acyclovir, valacyclovir, and famciclovir, or various combinations of antivirals. Also included are the administration of one or more therapies to treat one or more symptoms of RSV infection, including, but not limited to, steroids including corticosteroids, anti- inflammatories including acetaminophen or ibuprofen, pain-relief agents, creams or lotions to relieve itching, cool compresses, or various combinations thereof. In some aspects, 1, 2, 3, 4, 5, or more of the foregoing therapeutic agents may be excluded. Such combination therapy includes administration of a single pharmaceutical dosage formulation of a composition of the invention and one or more additional active agents, as well as administration of the composition of the invention and each active agent in its own separate pharmaceutical dosage formulation. For example, a composition of the invention and the other active agent can be administered to the patient together in a single dosage composition such as an injection or tablet or capsule, or each agent administered in separate oral dosage formulations. Where separate dosage formulations are used, the compounds of the invention and one or more additional active agents can be administered at essentially the same time, e.g., concurrently, or at separately staggered times, e.g., sequentially; combination therapy is understood to include all these regimens. In one aspect, it is contemplated that a vaccine and/or therapy is used in conjunction with antiviral treatment. Alternatively, the vaccine and/or therapy may precede or follow treatment with another agent by intervals ranging from minutes to weeks. In aspects where the other agents and/or vaccines are administered separately, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and immunogenic composition would still be able to exert an advantageously combined effect on the subject. In such aspects, it is contemplated that one may administer both modalities within or within about 12-24 h of each other or within or within about 6-12 h of each other (e.g., within at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours). In some situations, it may be desirable to extend the time period for administration significantly, where several days (2, 3, 4, 5, 6, 7, or more) to several weeks (1, 2, 3, 4, 5, 6, 7, 8, or more) lapse between the respective administrations. Various combinations may be employed, for example antiviral therapy “A” and immunogenic polypeptide given as part of an immune therapy regime “B”: A/B/A B/A/B B/B/A A/A/B A/B/B B/A/A A/B/B/B B/A/B/B B/B/B/A B/B/A/B A/A/B/B A/B/A/B A/B/B/A B/B/A/A B/A/B/A B/A/A/B A/A/A/B B/A/A/A A/B/A/A A/A/B/A Administration of the immunogenic compositions of the present disclosure to a patient/subject will follow general protocols for the administration of such compounds, taking into account the toxicity, if any, of the RSV RNA vaccine composition, or other compositions described herein. It is expected that the treatment cycles would be repeated as necessary. It also is contemplated that various standard therapies, such as hydration, may be applied in combination with the described therapy. D. ADMINISTRATION Administration of the compositions described herein may be carried out via any of the accepted modes of administration of agents for serving similar utilities. In some aspects, a pharmaceutical composition described herein may be administered intranasally, subcutaneously, intradermally or intramuscularly. In specific aspects, the RSV RNA molecules and/or RNA-LNP compositions are administered intramuscularly. In certain aspects, the pharmaceutical composition is formulated for local administration or systemic administration. Systemic administration may include enteral administration, which involves absorption through the gastrointestinal tract, or parenteral administration. As used herein, “parenteral administration” refers to the administration in any manner other than through the gastrointestinal tract, such as by intravenous injection. In one aspect, the pharmaceutical composition is formulated for intramuscular administration. In another aspect, the pharmaceutical composition is formulated for systemic administration, e.g., for intravenous administration. In some aspects, 1, 2, 3, or more of the foregoing administration routes may be excluded. Pharmaceutical compositions may be formulated into preparations in solid, semi-solid, liquid, lyophilized, frozen, and/or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suspensions, suppositories, injections, inhalants, gels, microspheres, and aerosols. In some aspects, 1, 2, 3, or more of the foregoing preparations may be excluded. Typical routes of administering such pharmaceutical compositions include, without limitation, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal, and intranasal. The term parenteral as used herein includes subcutaneous injections, intramuscular, intradermal, intranasal, intrasternal injection, or infusion techniques. Pharmaceutical compositions described herein are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient. Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound in aerosol form may hold a plurality of dosage units. The composition to be administered will, in any event, contain a therapeutically and/or prophylactically effective amount of a compound within the scope of this disclosure, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings described herein. A pharmaceutical composition within the scope of this disclosure may be in the form of a solid or liquid and may be frozen or lyophilized. In one aspect, the carrier(s) are particulate, so that the compositions are, for example, in tablet or powder form. The carrier(s) may be liquid, with the compositions being, for example, an oral syrup, injectable liquid, or an aerosol, which is useful in, for example, inhalatory administration. In some aspects, when intended for oral administration, the pharmaceutical composition is in either solid or liquid form, where semi-solid, semi-liquid, suspension, and gel forms are included within the forms considered herein as either solid or liquid. As a solid composition for oral administration, the pharmaceutical composition may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form. Such a solid composition will typically contain one or more inert diluents or edible carriers. In addition, one or more of the following may be present or exclude: binders such as carboxymethylcellulose, ethyl cellulose, microcrystalline cellulose, gum tragacanth, or gelatin; excipients such as starch, lactose, or dextrins; disintegrating agents such as alginic acid, sodium alginate, PRIMOJEL®, corn starch and the like; lubricants such as magnesium stearate or STEROTEX®; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate, or orange flavoring; and a coloring agent. When the pharmaceutical composition is in the form of a capsule, for example, a gelatin capsule, it may contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol or oil. In some aspects, 1, 2, 3, or more of the foregoing elements may be excluded from a solid composition. The pharmaceutical composition may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension. The liquid may be for oral administration or for delivery by injection, as two examples. In some aspects, when intended for oral administration, compositions contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant, and flavor enhancer. In a composition intended to be administered by injection, one or more of a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer, and isotonic agent may be included or excluded. A liquid pharmaceutical composition, whether it be a solution, suspension, or other like form, may include or exclude one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, e.g., physiological saline, Ringer’s 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; agents to act as cryoprotectants such as sucrose or trehalose. The parenteral preparation may be enclosed in ampoules, disposable syringes, or multiple dose vials made of glass or plastic. In one aspect, physiological saline is the adjuvant. In one aspect, an injectable pharmaceutical composition is sterile. A liquid pharmaceutical composition intended for either parenteral or oral administration should contain an amount of a compound such that a suitable dosage will be obtained. The pharmaceutical composition may include various materials, which modify the physical form of a solid or liquid dosage unit. For example, the composition may include materials that form a coating shell around the active ingredients. The materials that form the coating shell are typically inert, and may be, for example, sugar, shellac, or other enteric coating agents. The pharmaceutical composition may include dosage units that can be administered as an aerosol. The term aerosol denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of compounds may be delivered in single phase, bi-phasic, or tri-phasic systems in order to deliver the active ingredient(s). Delivery of the aerosol includes the necessary container, activators, valves, subcontainers, and the like, which together may form a kit. One skilled in the art, without undue experimentation may determine preferred aerosols. The pharmaceutical compositions may be prepared by methodology well known in the pharmaceutical art. For example, a pharmaceutical composition intended to be administered by injection may be prepared by combining the nucleic acid or polypeptide with sterile, distilled water or other carrier so as to form a solution. A surfactant may be added to facilitate the formation of a homogeneous solution or suspension. Surfactants are compounds that non-covalently interact with a compound consistent with the teachings herein so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system. The pharmaceutical compositions according to the present disclosure, or their pharmaceutically acceptable salts, are generally applied in a “therapeutically effective amount” or a “prophylactically effective amount” and in “a pharmaceutically acceptable preparation.” The term “pharmaceutically acceptable” refers to the non-toxicity of a material which does not interact with the action of the active component of the pharmaceutical composition. The terms “therapeutically effective amount” and “prophylactically effective amount” refer to the amount which achieves a desired reaction or a desired effect alone or together with further doses. In the case of the treatment of a particular disease, in one aspect, the desired reaction relates to inhibition of the course of the disease. This comprises slowing down the progress of the disease and, in particular, interrupting and/or reversing the progress of the disease. The desired reaction in a treatment of a disease may also be delay of the onset and/or a prevention of the onset of said disease or said condition. The compositions within the scope of the disclosure are administered in a therapeutically and/or prophylactically effective amount, which will vary depending upon a variety of factors including the activity of the specific therapeutic and/or prophylactic agent employed; the metabolic stability and length of action of the therapeutic and/or prophylactic agent; the individual parameters of the patient, including the age, body weight, general health, gender, and diet of the patient; the mode, time, and/or duration of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy. In some aspects, 1, 2, 3, 4, 5, or more of the factors may be excluded from determining a therapeutically and/or prophylactically effective amount. Accordingly, the doses administered of the compositions described herein may depend on various of such parameters. In the case that a reaction in a patient is insufficient with an initial dose, higher doses (or effectively higher doses achieved by a different, more localized route of administration) may be used. In some aspects, compositions (e.g., RSV RNA-LNP compositions) may be administered at dosage levels sufficient to deliver 0.0001 ng/µg/mg per kg to 100 ng/µg/mg per kg, 0.001 ng/µg/mg per kg to 0.05 ng/µg/mg per kg, 0.005 ng/µg/mg per kg to 0.05 ng/µg/mg per kg, 0.001 ng/µg/mg per kg to 0.005 ng/µg/mg per kg, 0.05 ng/µg/mg per kg to 0.5 ng/µg/mg per kg, 0.01 ng/µg/mg per kg to 50 ng/µg/mg per kg, 0.1 ng/µg/mg per kg to 40 ng/µg/mg per kg, 0.5 ng/µg/mg per kg to 30 ng/µg/mg per kg, 0.01 ng/µg/mg per kg to 10 ng/µg/mg per kg, 0.1 ng/µg/mg per kg to 10 ng/µg/mg per kg, or 1 ng/µg/mg per kg to 25 ng/µg/mg per kg, of subject body weight per day, one or more times a day, per week, per month, etc. to obtain the desired therapeutic, diagnostic, prophylactic, and/or imaging effect (see, e.g., the range of unit doses described in International Publication No. WO2013/078199, herein incorporated by reference in its entirety). In some aspects, compositions (e.g., RSV RNA-LNP compositions) may or may not be administered at dosage levels sufficient to deliver at least, at most, exactly, or between (inclusive or exclusive) any two of 0.0001, 0.0002, 0.0003, 0.0004, 0.0005, 0.0006, 0.0007, 0.0008, 0.0009, 0.001,0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100 ng/µg/mg per kg, of subject body weight per day, one or more times a day, per week, per month, etc. to obtain the desired therapeutic, diagnostic, prophylactic, and/or imaging effect. In some aspects, compositions (e.g., RSV RNA-LNP compositions) may or may not be administered at a total dose of or at dosage levels sufficient to deliver a total dose of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.0001, 0.0002, 0.0003, 0.0004, 0.0005, 0.0006, 0.0007, 0.0008, 0.0009, 0.001, 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100 ng/µg/mg per day, one or more times a day, per week, per month, etc. to obtain the desired therapeutic, diagnostic, prophylactic, and/or imaging effect. In specific aspects, compositions (e.g., RSV RNA-LNP compositions) may or may not be administered at a total dose of or at dosage levels sufficient to deliver a total dose of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.0001, 0.0002, 0.0003, 0.0004, 0.0005, 0.0006, 0.0007, 0.0008, 0.0009, 0.001, 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100 mg/mL RSV RNA encapsulated in LNP. In exemplary aspects, compositions (e.g., RSV RNA-LNP compositions) may or may not be administered at dose levels of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg/mL RSV RNA encapsulated in LNP. In exemplary aspects, compositions (e.g., RSV RNA-LNP compositions) may or may not be administered at dose levels of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.01, 0.15, 0.30, 0.45, 0.60, 0.75, or 0.90 mg RSV RNA encapsulated in LNP. In specific aspects, compositions (e.g., RSV RNA-LNP compositions) may or may not be administered at a total dose of or at dosage levels sufficient to deliver a total dose of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.0001, 0.0002, 0.0003, 0.0004, 0.0005, 0.0006, 0.0007, 0.0008, 0.0009, 0.001, 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100 µg/mL RSV RNA encapsulated in LNP. In exemplary aspects, compositions (e.g., RSV RNA-LNP compositions) may or may not be administered at dose levels of at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 15, 30, 45, 60, 75, 90, 100 or higher µg/mL RSV RNA encapsulated in LNP. In exemplary aspects, compositions (e.g., RSV RNA-LNP compositions) may or may not be administered at dose levels of at least, at most, exactly, or between (inclusive or exclusive) any two of 1, 15, 30, 45, 60, 75, 90, 100 or higher µg RSV RNA encapsulated in LNP. The desired dosage may be delivered multiple times a day (e.g., 1, 2, 3, 4, 5, or more times a day), every other day, every third day, every week, every two weeks, every three weeks, every four weeks, every 2 months, every three months, every 6 months, every year, etc. In certain aspects, the desired dosage may be delivered using a single-dose administration. In certain aspects, the desired dosage may be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations). When multiple administrations are employed, split dosing regimens may be used. The time of administration between the initial administration of the composition and a subsequent administration of the composition may be, but is not limited to, 1 minute, 2 minutes, 3 minutes, 4 minutes, 5 minutes, 6 minutes, 7 minutes, 8 minutes, 9 minutes, 10 minutes, 15 minutes, 20 minutes 35 minutes, 40 minutes, 45 minutes, 50 minutes, 55 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, 1 day, 36 hours, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 10 days, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 18 months, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, 10 years, 11 years, 12 years, 13 years, 14 years, 15 years, 16 years, 17 years, 18 years, 19 years, 20 years, 25 years, 30 years, 35 years, 40 years, 45 years, 50 years, 55 years, 60 years, 65 years, 70 years, 75 years, 80 years, 85 years, 90 years, 95 years or more than 99 years. In some aspects, compositions (e.g., RSV RNA-LNP compositions) may be administered in a single dose. In some aspects, compositions (e.g., RSV RNA-LNP compositions) may be administered twice (e.g., Day 0 and on or about Day 7, Day 0 and on or about Day 14, Day 0 and on or about Day 21, Day 0 and on or about Day 28, Day 0 and on or about Day 60, Day 0 and on or about Day 90, Day 0 and on or about Day 120, Day 0 and on or about Day 150, Day 0 and on or about Day 180, Day 0 and on or about 1 month later, Day 0 and on or about 2 months later, Day 0 and on or about 3 months later, Day 0 and on or about 6 months later, Day 0 and on or about 9 months later, Day 0 and on or about 12 months later, Day 0 and on or about 18 months later, Day 0 and on or about 2 years later, Day 0 and on or about 5 years later, or Day 0 and on or about 10 years later), with each administration at a total dose of or at dosage levels sufficient to deliver a total dose of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.0001, 0.0002, 0.0003, 0.0004, 0.0005, 0.0006, 0.0007, 0.0008, 0.0009, 0.001, 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100 ng/µg/mg RSV RNA encapsulated in LNP. Higher and lower dosages and frequency of administration are encompassed by the present disclosure. For example, compositions (e.g., RSV RNA-LNP compositions) may be administered three or four times. Periodic boosters at intervals of 1-5 years may be desirable to maintain protective levels of the antibodies. As used herein, the term “booster” refers to an extra administration of a composition (e.g., a RSV RNA-LNP composition). A booster may be given after an earlier administration of the composition. In some aspects, the compositions (e.g., RSV RNA-LNP compositions) are or are not administered to a subject as a single dose of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.0001, 0.0002, 0.0003, 0.0004, 0.0005, 0.0006, 0.0007, 0.0008, 0.0009, 0.001, 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100 ng/µg/mg of RSV RNA encapsulated in LNP. In some aspects, the compositions (e.g., RSV RNA-LNP compositions) are or are not administered the subject as a single dose of at least, at most, exactly, or between (inclusive or exclusive) any two of 1 µg, 15 µg, 30 µg, 45 µg, 60 µg, 75 µg, 90 µg, 100 µg or higher of RSV RNA encapsulated in LNP. In some aspects, the compositions (e.g., RSV RNA-LNP compositions) are or are not administered to a subject as two doses of at least, at most, exactly, or between (inclusive or exclusive) any two of 0.0001, 0.0002, 0.0003, 0.0004, 0.0005, 0.0006, 0.0007, 0.0008, 0.0009, 0.001, 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100 ng/µg/mg of RSV RNA encapsulated in LNP. In some aspects, the compositions (e.g., RSV RNA-LNP compositions) are or are not administered the subject as two doses of at least, at most, exactly, or between (inclusive or exclusive) any two of 1 µg, 15 µg, 30 µg, 45 µg, 60 µg, 75 µg, 90 µg, 100 µg or higher of RSV RNA encapsulated in LNP. In specific aspects, compositions (e.g., RSV RNA-LNP compositions) may or may not be administered twice (e.g., Day 0 and Day 28, Day 0 and Day 60, Day 0 and Day 180, Day 0 and 2 months later, Day 0 and 6 months later, Day 0 and one year later, etc.), with each administration at a total dose of or at dosage levels sufficient to deliver a total dose of at least, at most, exactly, or between (inclusive or exclusive) any two of 1 µg, 15 µg, 30 µg, 45 µg, 60 µg, 75 µg, 90 µg, 100 µg or higher RSV RNA encapsulated in LNP. IX. METHODS OF USE Provided herein are compositions (e.g., pharmaceutical compositions comprising RSV RNA molecules and/or RSV RNA-LNPs), methods, kits and reagents for prevention and/or treatment of RSV in humans and other mammals. RSV RNA compositions (e.g., RSV RNA-LNP compositions) may be used as therapeutic and/or prophylactic agents. They may be used in medicine to prevent and/or treat infectious disease. In exemplary aspects, the RSV RNA compositions are used to provide prophylactic protection from acute lower respiratory infection (ALRI) of any genotype, strain, or isolate. It is envisioned that there may be situations where persons are at risk for infection with more than one strain of RSV. RSV RNA compositions (e.g., RSV RNA-LNP compositions) are particularly amenable to combination vaccination approaches due to a number of factors including, but not limited to, speed of manufacture, ability to rapidly tailor vaccines to accommodate perceived geographical threat, and the like. Moreover, because the RSV RNA compositions (e.g., RSV RNA-LNP compositions) utilize the human body to produce the antigenic protein, the RSV RNA compositions (e.g., RSV RNA-LNP compositions) are amenable to the production of larger, more complex antigenic proteins, allowing for proper folding, surface expression, antigen presentation, etc. in the human subject. To protect against more than one strain of RSV, a combination RSV RNA composition can be administered that includes RNA encoding at least one antigenic polypeptide protein (or antigenic portion thereof) of a first RSV and further includes RNA encoding at least one antigenic polypeptide protein (or antigenic portion thereof) of a second RSV. In one embodiment of this invention, the RSV RNA-LNP composition is bivalent, wherein the composition comprises a first RSV RNA of subtype A and a second RSV RNA of subtype B. The RSV vaccines of the present disclosure may be used to prevent RSV (infection-associated illness, including pneumonia and bronchitis) and may be particularly useful for prevention and/or treatment of immunocompromised and elderly patients to prevent or to reduce the severity and/or duration of RSV infection. In some aspects, the RSV RNA compositions (e.g., RSV RNA-LNP compositions) of the disclosure are administered to a subject (e.g., a mammalian subject, such as a human subject), and the RNA polynucleotides are translated in vivo to produce an antigenic polypeptide. The RSV RNA compositions (e.g., RSV RNA-LNP compositions) may be induced for translation of a polypeptide (e.g., antigen or immunogen) in a cell, tissue or organism. In exemplary embodiments, such translation occurs in vivo, although there can be envisioned embodiments where such translation occurs ex vivo, in culture or in vitro. In exemplary embodiments, the cell, tissue or organism is contacted with an effective amount of a RSV RNA composition (e.g., a RSV RNA-LNP composition) including an RNA molecule having at least one translatable region encoding an antigenic polypeptide (e.g., a RSV antigen). In some aspects, the RSV RNA compositions of the disclosure may be used to prime immune effector cells, for example, to activate peripheral blood mononuclear cells (PBMCs) ex vivo, which are then infused (re-infused) into a subject. In some aspects, after administration of a RSV RNA molecule described herein, e.g., formulated as RNA-LNPs, at least a portion of the RNA is delivered to a target cell. In some aspects, at least a portion of the RNA is delivered to the cytosol of the target cell. In some aspects, the RNA is translated by the target cell to produce the polypeptide or protein it encodes. In some aspects, the target cell is a spleen cell. In some aspects, the target cell is an antigen presenting cell such as a professional antigen presenting cell in the spleen. In some aspects, the target cell is a dendritic cell and/or macrophage. RNA molecules such as RNA-LNPs described herein may be used for delivering RNA to such target cell. Accordingly, the present disclosure also relates to a method for delivering RNA to a target cell in a subject comprising the administration of the RNA-particles described herein to the subject. In some aspects, the RNA is delivered to the cytosol of the target cell. In some aspects, the RNA is translated by the target cell to produce the polypeptide or protein encoded by the RNA. “Encoding” refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (e.g., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom. Thus, a gene encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system. Both the coding strand, the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, may be referred to as encoding the protein or other product of that gene or cDNA. In some aspects, nucleic acid compositions described herein, e.g., compositions comprising a RSV RNA-LNP, are characterized by (e.g., when administered to a subject) an induced and/or boosted immune response as a function of antigen production in the cell. Increased antigen production may be demonstrated by, e.g., increased cell transfection (the percentage of cells transfected with the RNA vaccine), increased protein translation from the polynucleotide, decreased nucleic acid degradation (as demonstrated, for example, by increased duration of protein translation from a modified polynucleotide), and/or altered antigen specific immune response of the host cell. In some aspects, the disclosure relates to a method of inducing an immune response against RSV in a subject. The method includes administering to the subject an effective amount of an RNA molecule, RNA-LNP and/or composition as described herein to produce an immune response against RSV. In another aspect, the disclosure relates to a method of vaccinating a subject. The method includes administering to the subject in need thereof an effective amount of an RNA molecule, RNA-LNP and/or composition described herein. In another aspect, the disclosure relates to a method of treating and/or preventing an infectious disease. The method includes administering to the subject an effective amount of an RNA molecule RNA-LNP and/or composition as described herein. In another aspect, the disclosure relates to a method of treating and/or preventing and/or reducing the severity of a RSV infection and/or illness caused by RSV . The method includes administering to the subject an effective amount of an RNA molecule, RNA-LNP and/or composition as described herein. In another aspect, the disclosure relates to a method of treating and/or preventing and/or reducing the severity of an infectious disease in a subject by, for example, inducing an immune response to an infectious disease in the subject. In some aspects, the method includes administering a priming composition that includes an effective amount of an RNA molecule, RNA- LNP and/or composition described herein, and administering a booster composition including an effective amount of an RNA molecule, RNA-LNP and/or composition. In some aspects, the composition elicits an immune response including an antibody response. In some aspects, the composition elicits an immune response including a T cell response and/or a B cell response. In some aspects, an immune response comprises a T cell response and a B cell response. In some aspects, the composition elicits a neutralizing immune response. A neutralizing immune response is an immune response that is a neutralizing antibody response and/or an effective neutralizing T cell response. In some embodiments a neutralizing antibody response produces a level of antibodies that meet or exceed a seroprotection threshold. In some aspects, the composition elicits an effective T cell response. An effective T cell response is a response which produces a baseline level of infectious disease-activated and/or infectious disease-specific T cells including CD8+ and CD4+ T helper type 1 cells. In some aspects, the effective T cells comprises a high proportion of CD8+ T cells and/or CD4+ T cells, relative to a baseline level (in a naive subject). In some embodiments these T cells are differentiated towards an early- differentiated memory phenotype with co-expression of CD27 and CD28. In another aspect, the disclosure relates to a method of treating and/or preventing and/or reducing the severity of a RSV infection and/or illness caused by RSV in a subject by, for example, inducing an immune response to RSV in the subject. In some aspects, the method includes administering a priming composition that includes an effective amount of an RNA molecule, RNA- LNP and/or composition described herein, and administering a booster composition including an effective amount of an RNA molecule RNA-LNP and/or composition as described herein. In some aspects, the composition elicits an immune response including an antibody response. In some aspects, the composition elicits an immune response including a T cell response and/or a B cell response. In some aspects, an immune response comprises a T cell response and a B cell response. In some aspects, the composition elicits a neutralizing immune response. A neutralizing immune response is an immune response that is a neutralizing antibody response and/or an effective neutralizing T cell response. In some embodiments a neutralizing antibody response produces a level of antibodies that meet or exceed a seroprotection threshold. In some aspects, the composition elicits an effective T cell response. An effective T cell response is a response which produces a baseline level of infectious disease-activated and/or infectious disease-specific T cells including CD8+ and CD4+ T helper type 1 cells. In some aspects, the effective T cells comprises a high proportion of CD8+ T cells and/or CD4+ T cells, relative to a baseline level (in a naive subject). In some embodiments these T cells are differentiated towards an early-differentiated memory phenotype with co-expression of CD27 and CD28. The methods disclosed herein may involve administering to the subject a RSV RNA-LNP composition comprising at least one RSV RNA molecule having an open reading frame encoding at least one RSV antigenic polypeptide, thereby inducing in the subject an immune response specific to RSV antigenic polypeptide, wherein anti-antigenic polypeptide antibody titer in the subject is increased following vaccination relative to anti-antigenic polypeptide antibody titer in a subject vaccinated with a prophylactically effective dose (e.g., a therapeutically effective dose that prevents infection with the virus at a clinically acceptable level) of a traditional vaccine against the RSV. An “anti-antigenic polypeptide antibody” is a serum antibody the binds specifically to the antigenic polypeptide. In some aspects, the anti-antigenic polypeptide antibody titer in the subject is or is not increased at least, at most, between (inclusive or exclusive) any two of, or exactly 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 log following administration of the RSV RNA-LNP composition relative to anti-antigenic polypeptide antibody titer in a subject administered a prophylactically effective dose of a traditional composition against RSV (e.g., the standard of care dose of a recombinant or purified RSV protein vaccine, a live attenuated or inactivated RSV vaccine, or a RSV VLP vaccine). In some aspects, the anti-antigenic polypeptide antibody titer in the subject is or is not increased at least, at most, between (inclusive or exclusive) any two of, or exactly 1-, 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, 10-, 100-, or 1000-fold following administration of the RSV RNA-LNP composition relative to anti-antigenic polypeptide antibody titer in a subject administered a prophylactically effective dose of a traditional composition against RSV (e.g., the standard of care dose of a recombinant or purified RSV protein vaccine, a live attenuated or inactivated RSV vaccine, or a RSV VLP vaccine). In some aspects, an effective amount of a RSV RNA-LNP composition comprising at least one RSV RNA molecule having an open reading frame encoding at least one RSV antigenic polypeptide results in a 2-fold to 200-fold (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of a 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, 10-, 20-, 30-, 40-, 50-, 60-, 70-, 80-, 90-, 100-, 110-, 120-, 130-, 140-, 150-, 160-, 170-, 180-, 190-, or 200-fold) increase in serum neutralizing antibodies against RSV , relative to a traditional composition against RSV (e.g., the standard of care dose of a recombinant or purified RSV protein vaccine, a live attenuated or inactivated RSV vaccine, or a RSV VLP vaccine). In some aspects, an effective amount of a RSV RNA-LNP composition comprising at least one RSV RNA molecule having an open reading frame encoding at least one RSV antigenic polypeptide is a dose equivalent to an at least 2-fold reduction in a standard of care dose of a traditional composition against RSV . For example, an effective amount of a RSV RNA-LNP composition may or may not be a dose equivalent to a 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, 10-, 20-, 50-, 100-, 250-, 500-, or 1000-fold reduction in a standard of care dose of a traditional composition against RSV . In some embodiments, the anti-RSV antigenic polypeptide antibody titer produced in a subject administered an effective amount of a RSV RNA-LNP composition is equivalent to an anti-RSV antigenic polypeptide antibody titer produced in a control subject administered the standard of care dose of a traditional composition against RSV . In some embodiments, an effective amount of a RSV RNA-LNP composition is or is not a dose equivalent to a 2-fold to 1000-fold reduction (e.g., at least, at most, exactly, or between (inclusive or exclusive) any two of a 2-, 3 -,4 -,5 -,6-, 7-, 8-, 9-, 10-, 20-, 30-, 40-, 50-, 60-, 70-, 80-, 90-, 100-, 110-, 120-, 130-, 140-, 150-, 160-, 170-, 1280-, 190-, 200-, 210-, 220-, 230-, 240-, 250-, 260-, 270-, 280-, 290-, 300-, 310-, 320-, 330-, 340-, 350-, 360-, 370-, 380-, 390-, 400-, 410-, 420-, 430-, 440-, 450-, 4360-, 470-, 480-, 490-, 500-, 510-, 520-, 530-, 540-, 550-, 560-, 5760-, 580-, 590-, 600-, 610-, 620-, 630-, 640-, 650-, 660-, 670-, 680-, 690-, 700-, 710-, 720-, 730-, 740-, 750-, 760-, 770-, 780- , 790-, 800-, 810-, 820-, 830-, 840-, 850-, 860-, 870-, 880-, 890-, 900-, 910-, 920-, 930-, 940-, 950-, 960-, 970-, 980-, 990-, or 1000-fold reduction) in the standard of care dose of a traditional composition against RSV , wherein the anti-RSV antigenic polypeptide antibody titer produced in the subject is equivalent to an anti-RSV antigenic polypeptide antibody titer produced in a control subject administered the standard of care dose of a traditional composition against RSV . In some aspects, an anti-RSV antigenic polypeptide antibody titer produced in the subject is equivalent to an anti-RSV antigenic polypeptide antibody titer produced in a control subject administered the standard of care dose of a traditional composition against RSV . A traditional composition against RSV , as used herein, refers to a composition other than the RNA molecules, RNA-LNPs and/or compositions described herein. For instance, a traditional composition includes, but is not limited, to live microorganism vaccines, killed microorganism vaccines, attenuated vaccines, subunit vaccines, protein antigen vaccines containing recombinant protein produced in a heterologous expression system or purified from large amounts of the pathogenic organism, DNA vaccines, virus-like particle (VLP) vaccines containing viral capsid proteins (e.g., pre- and/or post-fusion F proteins) but lacking viral genome, etc. In exemplary embodiments, a traditional vaccine is a vaccine that has achieved regulatory approval and/or is registered by a national drug regulatory body, for example the Food and Drug Administration (FDA) in the United States or the European Medicines Agency (EMA). A “standard of care,” as provided herein, refers to a medical or psychological treatment guideline and can be general or specific. “Standard of care” specifies appropriate treatment based on scientific evidence and collaboration between medical professionals involved in the treatment of a given condition. It is the diagnostic and treatment process that a physician/ clinician should follow for a certain type of patient, illness or clinical circumstance. A “standard of care dose,” as provided herein, refers to the dose of a traditional composition against RSV that a physician/clinician or other medical professional would administer to a subject to treat and/or prevent RSV, or a RSV - related condition, while following the standard of care guideline for treating and/or preventing RSV, or a RSV-related condition. In some aspects, an RNA molecule, RNA-LNP and/or composition described herein (e.g., a RSV RNA-LNP composition comprising at least one RSV RNA molecule having an open reading frame encoding at least one RSV antigenic polypeptide) produces prophylactically- and/or therapeutically- efficacious levels, concentrations and/or titers of antigen-specific antibodies in the blood or serum of a subject. As defined herein, the term antibody titer refers to the amount of antigen-specific antibody produces in a subject, e.g., a human subject. In exemplary embodiments, antibody titer is expressed as the inverse of the greatest dilution (in a serial dilution) that still gives a positive result. In exemplary aspects, antibody titer is determined or measured by enzyme-linked immunosorbent assay (ELISA). In exemplary embodiments, antibody titer is determined or measured by neutralization assay, e.g., by microneutralization assay. In certain aspects, antibody titer measurement is expressed as a ratio, such as 1:40, 1:100, etc. In exemplary aspects, an efficacious an RNA molecule, RNA-LNP and/or composition described herein (e.g., a RSV RNA-LNP composition comprising at least one RSV RNA molecule having an open reading frame encoding at least one RSV antigenic polypeptide) produces an antibody titer of greater than 1:10, greater that 1:100, greater than 1:400, greater than 1:1000, greater than 1:2000, greater than 1:3000, greater than 1:4000, greater than 1:5000, greater than 1:6000, greater than 1:7500, or greater than 1:10000. In exemplary aspects, the antibody titer is produced or reached by 10 days following vaccination, by 20 days following vaccination, by 30 days following vaccination, by 40 days following vaccination, or by 50 or more days following vaccination. In exemplary aspects, the titer is produced or reached following a single dose of vaccine administered to the subject. In other aspects, the titer is produced or reached following multiple doses, e.g., following a first and a second dose (e.g., a booster dose). The methods disclosed herein may involve administering to the subject a RSV RNA-LNP composition comprising at least one RSV RNA molecule having an open reading frame encoding at least one RSV antigenic polypeptide, thereby inducing in the subject an immune response specific to RSV antigenic polypeptide, wherein the immune response in the subject is equivalent to an immune response in a subject administered with a traditional composition against the RSV that is or is not at least, at most, in between (inclusive or exclusive) any two of, or exactly 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, or 100 times the dosage level relative to the RNA composition. In some aspects, the RNA molecule, RNA-LNP and/or composition is used as a vaccine. In some aspects, the RNA molecule, RNA-LNP and/or composition may be used in various therapeutic or prophylactic methods for preventing, treating or ameliorating of acute lower respiratory infection (ALRI), or a disorder related to respiratory illness, including pneumonia and bronchitis. In some aspects, the RNA molecule, RNA-LNP and/or composition may be used in various therapeutic or prophylactic methods for preventing, treating or ameliorating of acute lower respiratory infection (ALRI), including pneumonia and bronchitis. RSV RNA compositions may be administered prophylactically to healthy subjects. In some aspects, the subject is immunocompetent. In some aspects, the subject is immunocompromised. In some aspects, the RNA molecule, RNA-LNP and/or composition is administered in a single dose. In some aspects, a second, third or fourth dose may be given. In some aspects, the RNA molecule, RNA-LNP and/or composition is administered in multiple doses. In some aspects, the RNA molecule, RNA-LNP and/or composition is administered intramuscularly (IM) or intradermally (ID). The present disclosure further provides a kit comprising the RNA molecule, RNA-LNP, and/or composition. In some aspects, the RNA molecule, RNA-LNP and/or composition described herein is administered to a subject that is less than about 1 years old, or about 1 years old to about 10 years old, or about 10 years old to about 20 years old, or about 20 years old to about 50 years old, or about 60 years old to about 70 years old, or older. In some aspects the subject is at least, at most, exactly, or between any two of less than 1 year of age, greater than 1 year of age, greater than 5 years of age, greater than 10 years of age, greater than 20 years of age, greater than 30 years of age, greater than 40 years of age, greater than 50 years of age, greater than 60 years of age, greater than 70 years of age, or older. In some aspects, the subject is greater than 50 years of age. In some aspects the subject is at least, at most, exactly, or between (inclusive or exclusive) any two of about 4 months of age or older, about 1 year of age or older, about 5 years of age or older, about 10 years of age or older, about 20 years of age or older, about 30 years of age or older, about 40 years of age or older, about 50 years of age or older, about 60 years of age or older, about 70 years of age or older, or older. In some aspects, the subject may be about 50 years of age or older. In some embodiments, the composition of the present invention further comprises a human metapneumovirus subtype A (hMPV A) F protein mutant or a nucleic acid encoding a hMPV A F protein mutant. In some embodiments, the composition of the present invention further comprises a human metapneumovirus subtype B (hMPV B) F protein mutant or a nucleic acid encoding a hMPV B F protein mutant. In further embodiments, the composition of the present invention further comprises both hMPV A and hMPV B F protein mutants or a nucleic acids encoding a hMPV A F protein mutant and a hMPV B F protein mutant. hMPV F protein mutants are described in any of PCT Publication Nos. WO16103238, WO20234300, WO21222639, WO22076669, WO22214678, WO23102373, WO23110618, WO23217988, and WO23102388, the full disclosures of which are herein incorporated by reference in their entirety for all purposes. In some embodiments, the composition of the present invention further comprises a parainfluenza virus type 1 (PIV1) F protein mutant or a nucleic acid encoding a PIV1 F protein mutant. In some embodiments, the composition of the present invention further comprises a parainfluenza virus type 3 (PIV3) F protein mutant or a nucleic acid encoding a PIV3 F protein mutant. In further embodiments, the composition of the present invention further comprises both PIV1 and PIV3 F protein mutants or a nucleic acids encoding a PIV1 F protein mutant and a PIV3 F protein mutant. PIV1 and PIV3 F protein mutants are described in any of PCT Publication Nos. WO2018081289, and WO2022207839, the full disclosures of which are herein incorporated by reference in their entirety for all purposes. The vaccines provided by the present disclosure may be used together with one or more other vaccines. For example, in adults they may be used together with an influenza vaccine, Prevnar, tetanus vaccine, diphtheria vaccine, COVID19 vaccine and pertussis vaccine. For pediatric use, vaccines provided by the present disclosure may be used with any other vaccine indicated for pediatric patients. X. CLINICAL STUDIES The RSV RNA-LNP vaccines of the present disclosure comprise nucleoside-modified mRNA encoding prefusion F (preF) polypeptide from RSV (modified RNA; modRNA). The RSV RNA-LNP vaccines may comprise RNA comprising a single-stranded, 5′-capped and polyadenylated modified RNA that is translated after entering the cell. The RNA comprises an open reading frame (ORF) that encodes variations of the RSV preF polypeptide. Further, as described herein, the RNA may comprise structural elements, such as untranslated regions (UTRs), optimized for high efficacy of the RNA. The RSV RNA-LNPs may comprise RNA as provided in Table 7 of Example 1 disclosed herein. The RSV RNA-LNPs may comprise RNA as provided in Tables 1 to 3 of Example 6 disclosed herein. The RNA may also comprise a substitution of 1-methyl-pseudouridine for uridine to decrease recognition of the vaccine RNA by innate immune sensors, such as toll-like receptors (TLRs) 7 and 8, resulting in decreased innate immune activation and increased protein translation. The RNA molecules described herein are formulated/encapsulated into lipid nanoparticles (LNPs) to enable delivery of the RNA into host cells after, e.g., intramuscular (IM), intradermal (ID), or intranasal (IN) injection. The LNP formulation may comprise two functional lipids, ALC- 0315 and ALC-0159, and two structural lipids, DSPC (1,2-distearoyl-sn-glycero-3- phosphocholine) and cholesterol. In some aspects, 1, 2, 3, or more of the forgoing lipids may be excluded from the LNP formulation. The potency of RNA vaccines is optimized by LNP encapsulation, which protects the RNA from degradation by extracellular RNases and facilitates delivery in the cell. After IM injection of RSV RNA-LNP vaccines, the LNPs are taken up by the cells, and the RNA is released into the cytosol. In the cytosol, the RNA is translated, and the encoded viral antigen is produced. The Examples herein demonstrate the RSV RNA-LNP vaccines of the present disclosure are immunogenic in mice and induce both humoral and cell mediated immune responses in mice. Clinical studies of the present disclosure evaluate the safety, tolerability, and immunogenicity of RSV RNA-LNP vaccines against RSV. For example, the RSV RNA-LNPs vaccines may be indicated for active immunization for the prevention of acute lower respiratory infection (ALRI), including pneumonia and bronchitis caused by RSV for adults (e.g., ≥45, ≥50, ≥55, ≥60, ≥70…etc. years of age or 50 through 69 years of age), or prevention of lower respiratory tract illnesses (LRTI) and severe LRTI, including pneumonia and bronchiolitis caused by RSV in children 4 months to < 5 years of age. RSV RNA-LNP vaccines may be administered in different dose level(s), dose formulation, number of doses and dosing schedules, as described herein, including but not limited to: - As a single-dose schedule or a two-dose schedule (e.g., Day 0 and on or about 2 months after or Day 0 and on or about 6 months after) - At different dose levels (e.g., of or of about 15 µg, 30 µg, 45 µg, 60 µg, 75 µg, 90 µg, 100 µg or higher per administration) - At different formulations (non-lyophilized and/or lyophilized) The RSV RNA-LNPs may be presented as a liquid or lyophilized formulation. Administration of the RSV RNA-LNP vaccines may or may not be dosed in the range of or of about 15 µg, 30 µg, 45 µg, 60 µg, 75 µg, 90 µg, 100 µg or higher per dose with an injection volume of or of about 0.25 to 1 mL (e.g., of or of about 0.25, 0.5, 1 mL). Dilution with sterile 0.9% sodium chloride (normal saline) may be required. The objectives of RSV RNA-LNP clinical studies may include, but are not limited to: -To describe the safety and tolerability profile of RSV RNA-LNP vaccines administered at selected dose levels and schedules in participants. -To describe the immune responses elicited by commercially-available vaccines against RSV and RSV RNA-LNP vaccines administered at selected dose levels and schedules in participants. In some aspects, the efficacy (or effectiveness) against RSV of the RNA molecules encoding RSV polypeptides, RNA-LNPs and compositions thereof disclosed herein is or is not greater than 50% (e.g., at least, at most, exactly, or between any two of 50%, 60%, 70%, 80%, 90%, or more). Vaccine efficacy may be assessed using standard analyses (see, e.g., Weinberg et al, J Infect Dis.2010 Jun 1;201(11): 1607-10). For example, vaccine efficacy may be measured by double-blind, randomized, clinical controlled trials such as those described herein. Vaccine efficacy may be expressed as a proportionate reduction in disease attack rate (AR) between the unvaccinated (ARU) and vaccinated (ARV) study cohorts and can be calculated from the relative risk (RR) of disease among the vaccinated group with use of the following formulas: Efficacy = (ARU ‒ ARV)/ARU x 100; and Efficacy = (1-RR) x 100. Likewise, vaccine effectiveness may be assessed using standard analyses (see, e.g., Weinberg et al., J Infect Dis. 2010 Jun 1;201(11): 1607-10). Vaccine effectiveness is an assessment of how a vaccine (which may have already proven to have high vaccine efficacy) reduces disease in a population. This measure can assess the net balance of benefits and adverse effects of a vaccination program, not just the vaccine itself, under natural field conditions rather than in a controlled clinical trial. Vaccine effectiveness is proportional to vaccine efficacy (potency) but is also affected by how well target groups in the population are immunized, as well as by other non-vaccine-related factors that influence the real-world outcomes of hospitalizations, ambulatory visits, and/or costs. For example, a retrospective case control analysis may be used, in which the rates of vaccination among a set of infected cases and appropriate controls are compared. Vaccine effectiveness may be expressed as a rate difference, with use of the odds ratio (OR) for developing infection despite vaccination: Effectiveness = (1 ‒ OR) x 100. In some aspects, efficacy of the RSV polypeptides, RNA-LNPs and compositions thereof is at least 60% relative to unvaccinated control subjects. For example, efficacy may be at least, at most, exactly, or between (inclusive or exclusive) any two of 65%, 70%, 75%, 80%, 85%, 95%, 98%, or 100% relative to unvaccinated control subjects. EXAMPLES Below are examples of specific aspects for carrying out the present disclosure. The following examples are included to demonstrate aspects of the disclosure. The examples are offered for illustrative purposes only and are not intended to limit the scope of the present disclosure in any way. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the disclosure. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes may be made in the specific aspects which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the disclosure. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should, of course, be allowed for. Example 1. Generation of RSV preF modRNA constructs RNA constructs generated herein encode RSV F protein wild-type (WT) and RSV F protein variants/mutants (e.g. RSV pre-fusion F protein). Table 6 describes WT F (WT F) and variant RSV preF designs. Table 6. RSV F proteins and description
Figure imgf000257_0001
Figure imgf000258_0001
DNA sequences encoding RSV F proteins were prepared and utilized for in vitro transcription reactions to generate RNA. In vitro transcription of RNA is known in the art and is described herein. DNA templates were cloned into a plasmid vector with backbone sequence elements (T7 promoter, 5′ and 3′ UTR, poly-A tail) for improved RNA stability and translational efficiency. The DNA was purified, spectrophotometrically quantified and in vitro-transcribed by T7 RNA polymerase in the presence of a trinucleotide cap1 analogue ((m2 7,3′-O)Gppp(m2’-O)ApG) (TriLink) and with N1-methylpseudouridine (m1Ψ) replacing uridine (modified RNA (modRNA)). The RSV RNA was generated from codon-optimized (CO) DNA for stabilization and superior protein expression. Table 7 shows RNA constructs of the present disclosure, and corresponding sequences, comprising a 5’ UTR, an open reading frame encoding a respiratory syncytial virus (RSV) polypeptide, a 3’ UTR and a poly-A tail. Table 7. RSV F modRNA constructs ’ ’
Figure imgf000258_0002
Figure imgf000259_0001
Figure imgf000260_0001
5 UTR sequence includes 5 cap sequence ** Poly-A tail length may contain +/-1 A or +/-2 A Example 2. Preparation of RSV preF RNA formulated in LNP The LNP formulation used in the following Examples contains 2 functional lipids, ALC- 0315 and ALC-0159, and 2 structural lipids DSPC (1,2distearoyl-sn-glycero-3-phosphocholine) and cholesterol. The physicochemical properties and the structures of the 4 lipids are shown in Table 8 below. Lipid nanoparticles were prepared and tested according to the general procedures described in US Patent 9737619 (PCT Pub. No. WO2015/199952) and US Patent 10166298 (WO 2017/075531) and WO2020/146805, each of which is hereby incorporated by reference in its entirety. Briefly, cationic lipid, DSPC, cholesterol and PEG-lipid were solubilized in ethanol at a molar ratio of about 47.5: 10: 40.7: 1.8. Table 8. Lipids in the LNP Formulation
Figure imgf000261_0001
CAS=Chemical Abstract Service; DSPC=1,2-disteroyl-sn-glycero-3-phosphocholine Example 3. RSV preF expression 135 unique UTR constructs were tested for RSV-F expression in both HEK (human embryonic kidney) cells and hDCs (human dendritic cells) in multiple subsets. A representative dataset of 64 constructs were tested for RSV-F expression (FIG.2) in both HEK and hDCs. A majority of the UTR constructs yielded higher expression compared to the WHO UTRs (5UTR_1_RSV_3UTR_1) (5’UTR sequence SEQ ID NO: 111 and 3’UTR sequence SEQ ID NO: 280) and the human hemoglobin (5UTR_2_RSV_3UTR_2) (5’UTR sequence SEQ ID NO: 73 and 3’UTR sequence SEQ ID NO: 281) benchmarks. Based on expression in HEK and hDC cells, 11 constructs were chosen for testing by assessing immunogenicity (neutralizing antibody response) of RSV-F encoding modRNA-LNPs in multiple rounds of screening. Not all sequences from the in silico design methods worked equally well. A low-performing UTR, 5UTR_261_RSV_3UTR_2 (5' UTR sequence SEQ ID NO: 59), was included as a low-expression control, and to evaluate translation of results from in vitro protein expression to in vivo immunogenicity (FIG.2, labeled constructs). Drug substance and subsequently drug product was generated for a total of 15 constructs which included the four benchmarks: WHO (5UTR_1/3UTR_1, or SEQ ID NO:111 and SEQ ID NO:280) with both the contiguous (80A) and the interrupted (30L70, consisting of 30A, then the linker of SEQ ID NO: 23, then an additional 70A) polyA, another marketed modRNA UTRs as benchmarks (5UTR_444, SEQ ID NO:90 and 3UTR_17, SEQ ID NO:287), and human hemoglobin UTRs (5UTR_2 and 3UTR_2, or SEQ ID NO:73 and SEQ ID NO:281) (Table 9). Table 9: Quality attributes of RSV modRNA LNPs generated for in vivo study
Figure imgf000262_0001
All modRNA LNPs were of high quality with >80% full length capped RNAs, with high encapsulation efficiency within lipid nanoparticles of low polydispersity index (Table 9). Furthermore, in vitro expression of these modRNA LNPs was tested in HEK293T cells and hDCs (Table 10). Table 10: In vitro expression profile of UTR candidates for RSV-F mouse study
Figure imgf000262_0002
Figure imgf000263_0001
There was a reasonable agreement in performance of the 5' UTRs in HEK cells in both of the experiments (FIG.2 and Table 10), e.g., the highest expression in both experiments was observed for mRNA-RSV-0037 (5UTR_73/3UTR_7). Additionally, the low expressing control UTR-OPT-1030 (5UTR_261/3UTR_2) also performed similarly in both experiments. In human dendritic cells, the novel UTR constructs performed better than the WHO benchmark with 80 polyA, except for the low expressing control, which was only about 10% as potent. Drug products were then transferred for immunogenicity testing in mice. Groups of female BALB/C mice received RSV-F modRNA-LNPs representing different UTR candidates intramuscularly on day 0 and day 21. Sera collected on day 21 (post dose 1, PD1) and day 35 (14 days PD2) were assessed for RSV neutralizing response. After the first dose, it was found that several top performing UTRs from the in vitro screening induced higher or similar levels of 50% neutralizing titers compared to several benchmark UTRs including the human hemoglobin (hHBB 5UTR_2/3UTR_2), WHO (5UTR_1/3UTR_1), and marketed Covid-19 vaccine modRNA benchmark UTRs with both contiguous and interrupted polyA (FIG.3A). Particularly, 5UTR_105 (SEQ ID NO:55) and 5UTR_15 (SEQ ID NO:50) had higher neutralizing titer levels than at least one of the WHO benchmarks, albeit not statistically significant. Consistent with the in vitro expression results, the low expressing control 5UTR_261(SEQ ID NO:59)/hHBB (SEQ ID NO:281) showed lower levels of neutralizing antibody titers. The UTR benchmark (5UTR_444/3UTR_17) based on a marketed vaccine modRNA also showed poor response in mice which was even lower than the 5UTR_261 low-performing control. Interestingly, after the second dose (day 35), other novel UTRs induced a neutralizing response comparable to top-performing UTRs in post-dose 1 (FIG. 3B). For example, the 5UTR_260 (SEQ ID NO:58) and the hHBB benchmark (5UTR_2, SEQ ID NO:73) showed similar neutralizing antibody responses as novel-designed UTRs such as 5UTR_15 (SEQ ID NO:50) and 5UTR_105 (SEQ ID NO:55). Consistent with previous observations in vitro and PD-1 data, 5UTR_261 induced a lower immune response. Furthermore, the data indicate that benchmark UTRs designed by others may not be useful in a gene-agnostic manner. Based on immunogenicity data described in FIG. 3, several additional novel UTRs were designed and tested for immunogenicity in mice. For example, the seed sequence for 5UTR_562 (SEQ ID NO:86) and 5UTR_563 (SEQ ID NO:87) is 5UTR_15 (SEQ ID NO:50); this parent UTR showed robust immunogenicity in mice using RSV-F as the antigen. Remarkably, 5UTR_563 (SEQ ID NO:87) showed comparable immunogenicity to its parent 5UTR_15 (SEQ ID NO:50) (FIG. 5A) and 5UTR_562 was the second-best performer in an additional follow-up immunogenicity study in mice (FIG.5B, below). The RSV 50% geometric mean neutralizing titers for 5UTR_563 (FIG.5A) were over 3-fold above 5UTR_1/3UTR_1 benchmark (WHO/WHO-80A) and close to 4-fold higher than the 5UTR_444/3UTR_17 benchmark. UTR construct 5UTR_273 (SEQ ID NO:78) was among the best performers with over 4-fold higher neutralizing titers compared to the WHO/WHO-80A benchmark. The 5UTR_273 UTR has a unique feature in that it contains three upstream start codons in the untranslated region itself, which are all in-frame of the start site downstream of the Kozak sequence. The 5UTR_105 sequence has one such upstream in-frame start codon. However, initiation of translation at an alternate upstream start site may add unwanted amino acids to the protein terminus. Therefore, UTR variants based on 5UTR_105 and 5UTR_273 were designed, in which these upstream start codons were removed. A subsequent mouse study (FIG.5B) examined additional UTR variants (including 5UTR_576 based on 5UTR_105, and 5UTR_581/582/583/599 based on 5UTR_273). In this study, notably, RSV-F modRNA-LNP based on 5UTR_582 induced robust neutralizing responses higher than the 5UTR_1/3UTR_1 benchmark as well as other UTRs. These examples also demonstrate that minimizing secondary structures in the 5' UTR, can drive robust expression of the encoded gene both in vitro and in vivo. For example, the activity (data not shown) of the 5UTR_15 (SEQ ID NO:50) was comparable to benchmark sequences and also to a previously published UTR sequence (NCA7d/TPRKB-80A), which was engineered for high expression ((Zeng et al., Adv Mater 32(40):e2004452 [2020]). (The NCA7d sequence used herein, 5UTR_5, is modified from Table S2 of Zeng to use a GCCACC Kozak motif, and to contain a 5' end suitable for cleancap reagent). This UTR also robustly expressed RSV-F protein in vitro and elicited strong RSV immune responses in mice. Variants of 5UTR_15 (including 5UTR_562 (SEQ ID NO: 86) and 5UTR_563 (SEQ ID NO:87)) that were also designed to minimize secondary structure also gave robust RSV neutralizing responses in mice (FIG.5B, see 5UTR_562). Together, these results suggest that rational designs may generate UTRs that can drive expression of multiple genes of interest that yield potent immune responses to the target protein of interest encoded from the mRNA. In Vitro protocols In vitro expression (IVE) for the modRNA LNP drug products was assessed by transfecting HEK293F cells with a dose response curves and staining for antibodies, RSV mAb1 specific for the trimeric RSV prefusion F-protein and L4-6 specific for total RSV F-protein. These antibodies have been shown to recognize both the A-strain and B-strain RSV F-protein and were used in the assay with either permeabilizing or non-permeabilizing conditions to assess the total cell vs cell surface content of the RSV F-protein. The description of the protocol below is for total staining (surface + intracellular proteins), for surface only staining protocol, the fixation and wash buffers are changed to remove the permeabilization agent and all other steps and reagents are the same. In brief, 96-well culture plates were seeded with HEK293F cells at a density of 2.5x105 cells/well and were placed in a shaking incubator (350 RPM, 37°C, humidified, 8% CO2) while the samples titrations were prepared. The LNP drug product was diluted in DPBS to a concentration of 160 ng/uL and serially diluted 11-points with a 2.5 dilution factor. After the sample preparation was completed, the 96-well culture plate was removed from the incubator and 50 uL of each point of the diluted LNP were added to duplicate wells of the 96-well culture plate to generate a titration curve ranging from 4,000 ng/well – 0.42 ng/well. The 96-well culture plate was placed back into the shaking incubator overnight. After incubation, 250 uL of cells are transferred to a 96-well u-bottom polystyrene plate and pelleted using a swinging bucket centrifuge at 500 radial centrifugal force (rcf) for 5 minutes (min) at room temperature (RT). The supernatant is aspirated, and cells resuspended in 100 uL solution of Aqua405 live/dead stain [Invitrogen, cat# L34966]. The plate is incubated 15 to 30 min at room temperature, protected from light. After incubation, cells are washed with wash buffer [BD, cat#51-2091KZ] and pelleted using swinging bucket centrifuge at 500 rcf for 5 min at RT. The supernatant is aspirated, cells are resuspended in 100 uL fixation/permeabilization buffer [BD, cat# 51-2091KZ] and the plate is incubated 30 minutes at 2 -8° C, protected from light. Once incubation is complete, the cells are pelleted using a swinging bucket centrifuge at 500 rcf for 5 min at RT. The supernatant is aspirated, and cells resuspended with 250 uL wash buffer, the washing of the cell pellet is repeated twice. After the final wash step, the cells are pelleted, supernatant aspirated, and resuspended in 50 uL of primary antibody solution. The plates are sealed and incubated for 45 minutes at 2-8° C, protected from light. Once incubation is completed, the cells are pelleted using a swinging bucket centrifuge at 500 rcf for 5 min at RT. The supernatant is aspirated, and cells resuspended with 250 uL wash buffer, the washing of the cell pellet is repeated twice. After the final wash step, the cells are pelleted, supernatant aspirated, and resuspended in 50 uL of secondary antibody solution. The plates are sealed and incubated for 45 minutes at 2-8° C, protected from light. Once the incubation is completed the cells are pelleted using a swinging bucket centrifuge at 500 rcf for 5 min at RT. The supernatant is removed and cells resuspended with 250 uL wash buffer, this is repeated twice. After the final wash step cells are pelleted, supernatant removed, and resuspended in 200 uL of wash buffer and data acquired by flow cytometry. In Vivo Protocol (mouse study) Female BALB/c mice (10/group) were immunized intramuscularly at day 0 and 21 with RSV-F subgroup A modRNA-LNPs with novel UTR candidates at 0.2 µg dose as shown in FIG 5A & 5B. Immunogenicity was evaluated by measuring RSV neutralizing antibody response. Serum was collected on day 21 and day 35 (2 weeks post dose 2, PD2) for RSV neutralization assays. Neutralization Assay The RSV microneutralization assay is a 3-day assay using A549 cells (human alveolar basal epithelial cells) to measure functional antibodies in serum that neutralize RSV activity, preventing infection of a host cell monolayer. On Day 0, A549 cells (human alveolar basal epithelial cells; ATCC, cat # CCL-185) are seeded in 96-well tissue-culture treated plates at 2.5 x 104 cells per well and incubated for at least 20 hours to form a confluent monolayer. On Day 1, diluted virus (RSV A, M37; RSV B, B18537; 500 FFU/well) is added to 3-fold serial dilutions of heat inactivated test serum prepared in duplicate and incubated for 1 hour to allow antibodies to bind to the virus. The neutralization reaction is then transferred onto the prepared A549 cell monolayers and incubated for 2 hours. Additional media is supplemented onto the plates prior to an overnight incubation (at least 16 hours). On Day 2, the plates are fixed with methanol and stained with a human anti-RSV F (Sobi, Synagis) primary antibody followed by an Alexa 488 fluorescently labeled secondary antibody to detect viral foci. A 50% neutralization titer is calculated as the last reciprocal serum dilution at which 50% of the virus is neutralized compared to wells containing virus only. A titer is reported as the geometric mean titer (GMT) of the two replicate titers of each sample. The assay titer range is 20 to 43,740. Any samples with a titer >43,740 are prediluted and repeated to extend the upper titer limit. Any samples below the lower limit of detection (LLOD) are reported at LLOD of 20. Example 4. Generation and Selection of UTRs for modRNA RSV Based on the in vitro and in vivo screening described in Example 3, the immunogenicity of the three lead UTR candidates, 5UTR_563, 5UTR_582 and 5UTR_599, was assessed using RSV A preF carrying 847 stabilizing mutations as the GOI. A 5'/3'-UTR pair (5UTR_1 (SEQ ID NO: 397)/ 3UTR_1 (SEQ ID NO: 566), named “WHO”) was selected as a benchmark comparison (Demongeot & Fougere, 2023). The in vivo study was performed similarly to what was described in Example 3. Briefly, BALB/c mice were immunized on day 0 and 21 with 0.05 μg or 0.2 μg of RSV modRNAs. Serum was collected on day 35 (2 weeks post dose 2, PD2) for RSV neutralizing response and T cell responses. Neutralization assays were performed as described in Example 3. The detailed T cell response evaluation by intracellular cytokine staining is described in this example below. RSV modRNAs containing 5UTR_563 (also known as “BMD563”; SEQ ID NO: 373), 5UTR_582 (also known as “BMD582”; SEQ ID NO: 380), and 5UTR_599 (also known as “BMD599”; SEQ ID NO: 385) novel 5’UTRs induced robust and comparable RSV neutralizing responses at both the 0.5 µg and 0.2 µg dose levels (Table 11 and FIG.6A). At the low dose of 0.05 µg, compared to the WHO benchmark, the BMD582 demonstrated a 7.3-fold higher neutralizing response. BMD599 neutralizing titers were similar to WHO. All modRNA groups also induced robust RSVpreF-specific CD4+ and CD8+ T-cell responses, with responses trending slightly higher with BMD582 (Table 12 and FIG.6B – 6D). Table 11. RSV A Neutralization Response
Figure imgf000267_0001
Table 12. RSV F-Specific T-Cell Responses
Figure imgf000267_0002
T Cell Response Evaluation Procedure RSV F-specific T cell responses were measured from splenocytes of selected groups by a flow cytometry-based intracellular cytokine staining assay (ICS). Splenocytes were cultured under unstimulated or RSV F peptide pool stimulated conditions in the presence of anti-CD107a APC antibody and protein transport inhibitors. Following stimulation, cells were incubated with fluorescently-conjugated antibodies to the surface proteins CD19, CD3, CD4, and CD8 followed by fixation, permeabilization and staining with IFN-γ, TNF-α, IL-4 and CD40L/CD154 antibodies. After staining, the cells were washed and resuspended in buffer. Cells were acquired on an LSR Fortessa and data analyzed by FlowJo (10.7.1). Results were background subtracted using the DMSO condition and shown as percentage of cytokine-expressing CD4+ T cells and CD8+ T cells, respectively. Example 5. In Vivo Evaluation of modRNA RSV Formulations Bivalent modRNA RSV composed of stabilized-preF antigens representing the two major RSV subgroups, A and B, is expected to offer the potential for the greatest breadth of protection against RSV disease. The balanced RSV A and RSV B neutralizing responses and equivalent high efficacy against RSV A and RSV B disease in humans has recently been demonstrated with Pfizer’s RSVpreF (ABRYSVO®) subunit vaccine (Walsh et al, 2022; Walsh et al, 2023; Wilson et al, 2023). For the bivalent modRNA, we evaluated immunogenicity of two different LNP- formulation approaches: 1) a "pre-mix" approach where both mRNAs are combined in equal ratio followed by co-formulation into LNPs, or 2) a "post-mix" approach where each mRNA is individually formulated into LNPs and the two LNP preparations combined in equal ratio. modRNA RSV formulation options were evaluated using both 5’ UTR BMD563 (Study 1 shown in Table 13, FIG.7A and 7B) and 5’ UTR BMD582 (Study 2 shown in Table 14, FIG.7C and 7D) in modRNA encoding RSV 847 A or B preF design or bivalent modRNA encoding RSV 847 A and B preF designs, comprising 3’UTR_2 (hHBB) and 80A polyA tail as described in Example 1 and formulated in LNPs described in Example 2. Pre-mix and post-mix bivalent formulations were compared to monovalent RSV A and monovalent RSV B formulations. Female BALB/c mice (N=10 per group) were immunized IM with 0.25 or 0.50 μg of monovalent LNP-formulated modRNA encoding RSV A or B 847 preF design, or bivalent modRNA (0.25 μg RSV A + 0.25 μg RSV B) prepared using post-mix or pre-mixed LNP- formulation approaches. Virus neutralization responses against a RSV A, M37 strain (Memphis 37 strain) (FIG.7A and FIG.7C), or RSV B, B18537 strain (FIG.7B and FIG.7D), were measured by neutralization assays on Day 35 (2 weeks post-dose 2) with procedure as described in Example 3. As RSV F has 90% amino acid sequence identity between RSV A and B subgroups and sequence variability in surface exposed residues clusters in prefusion specific antigenic sites, such as site Ø, the control monovalent RSV A and RSV B modRNA groups elicited cross-reactive neutralizing responses, with the highest neutralizing response to the subgroup matched to the vaccine. In the bivalent pre-mix and post-mix groups (0.25 μg RSV A + 0.25 μg RSV B), RSV A neutralizing responses were similar in both studies; RSV B neutralizing responses were similar or slightly lower in the pre-mix group compared to the post-mix group in the first and second study, respectively. Across both studies, RSV A and RSV B neutralizing responses for pre-mix and post-mix groups were similar or higher compared to the monovalent control groups. Overall, the data support the benefits of a bivalent formulation in eliciting high neutralizing responses against both RSV A and RSV B and suggest both pre-mix and post-mix formulation approaches elicit robust immune responses. Table 13. RSV Neutralization Responses with 563-UTR
Figure imgf000269_0001
Table 14. RSV Neutralization Responses with 582-UTR
Figure imgf000269_0002
Figure imgf000270_0001
Example 6. RSV F Antigens Sequences of the RSV antigens/polypeptides, RSV DNA and RSV RNA of the present invention are provided in Tables 1-3. The sequences may comprise any stop codon, including but not limited to the stop codons provided in the Tables below. Table 1. RSV Polypeptides
Figure imgf000270_0002
Figure imgf000271_0001
Figure imgf000272_0001
Figure imgf000273_0002
Table 2. RSV DNA
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000285_0001
Figure imgf000286_0001
Table 3. RSV RNA
Figure imgf000286_0002
Figure imgf000287_0001
Figure imgf000288_0001
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
The following paragraphs describe additional aspects of the disclosure: 1. An RNA molecule comprising at least one open reading frame encoding a respiratory syncytial virus (RSV) polypeptide and at least one untranslated region (UTR) selected from Table 5, provided that the UTR does not consist of only SEQ ID NOs: 397 and 566. 2. An RNA molecule comprising at least one open reading frame encoding a respiratory syncytial virus (RSV) fusion protein F (F) polypeptide and a 5’ untranslated region (5’ UTR), wherein the 5’ UTR comprises a nucleic acid sequence at least 90% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336-344, 356- 358, 360, 362-367, 370-375, and 378-396. 3. The RNA molecule of any one of paragraphs 1-2, wherein the 5’ UTR comprises a nucleic acid sequence at least 92% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336, 341, 356-358, 372-375, and 378-396. 4. The RNA molecule of any one of paragraphs 1-3, wherein the 5’ UTR comprises a nucleic acid sequence at least 95% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336, 357, 372-374, 378, 380, 384 and 385. 5. The RNA molecule of any one of paragraphs 1-4, wherein the 5’ UTR comprises a nucleic acid sequence at least 98% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 357, 372-374, 378, 380, 384 and 385. 6. The RNA molecule of any one of paragraphs 1-5, wherein the 5’ UTR comprises a nucleic acid sequence at least 98% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 357, 373, 378, 380, 384 and 385. 7. The RNA molecule of any one of paragraphs 1-6, wherein the 5’ UTR comprises a nucleic acid sequence selected from the group consisting of: SEQ ID NO: 373 (5UTR_563); SEQ ID NO: 380 (5UTR_582); and SEQ ID NO: 385 (5UTR_599). 8. The RNA molecule of any one of paragraphs 1-7, wherein the UTR is a 3’ untranslated region (3’ UTR). 9. The RNA molecule of paragraph 8, wherein the 3’ UTR comprises nucleotides having sequence SEQ ID NO: 567 (3UTR_2 (hHBB)). 10. The RNA molecule of paragraphs 1-9, wherein the RSV polypeptide is an RSV glycoprotein. 11. The RNA molecule of paragraphs 1-10, wherein the RSV glycoprotein is RSV F protein. 12. The RNA molecule of any one of paragraphs 1 to 11, wherein the RSV polypeptide is a full-length, truncated, fragment or variant thereof. 13. The RNA molecule of any one of paragraphs 1 to 12, wherein the RSV polypeptide comprises at least one mutation. 14. The RNA molecule of any one of paragraphs 1 -13, wherein the RSV F polypeptide is a prefusion F polypeptide. 15. The RNA molecule of any one of paragraphs 1 to 14, wherein the RSV F polypeptide comprises an amino acid of Table 1, including but not limited to any of SEQ ID NO: 1 to 6 or 621-626. 16. The RNA molecule of any one of paragraphs 1 to 15, wherein the RSV F polypeptide has at least 90%, 95%, 96%, 97%, 98% or 99% identity to the amino acid sequence selected from SEQ ID NO: 1 to 6 or 621-626. 17. The RNA molecule of paragraph 16, wherein the RSV F polypeptide has an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 6 and 621-626. 18. The RNA molecule of paragraph 17, wherein the RSV F polypeptide has an amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 6. 19. The RNA molecule of any one of paragraphs 1 to 18, wherein the open reading frame is transcribed from a nucleic acid sequence of Table 2, including by not limited to any of SEQ ID NO: 7 to 10 or 627-632. 20. The RNA molecule of any one of paragraphs 1 to 19, wherein the open reading frame is transcribed from a nucleic acid comprising a nucleotide sequence having at least 90%, 95%, 96%, 97%, 98% or 99% identity to any of the sequences of SEQ ID NOs: 7 to 10 or 627-632. 21. The RNA molecule of paragraph 20, wherein the open reading frame is transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 8, 10, and 627-632. 22. The RNA molecule of paragraph 21, wherein the open reading frame is transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 8 and 10. 23. The RNA molecule of any one of paragraphs 1 to 22, wherein the open reading frame comprises a nucleic acid sequence of Table 3, including but not limited to any of SEQ ID NO: 15-16 or 633-638. 24. The RNA molecule of any one of paragraphs 1-23, wherein the open reading frame comprises a nucleic acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to any one of the sequences of SEQ ID NOs: 15-16 or 633-638. 25. The RNA molecule of paragraph 24, wherein the open reading frame comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 15-16 and 633-638. 26. The RNA molecule of paragraph 25, wherein the open reading frame comprises a nucleic acid sequence selected from the group consisting of SEQ ID Nos: 15 and 16. 27. The RNA molecule of any one of paragraphs 1-26, wherein the RNA molecule further comprises a 5’ cap moiety or a 3’ poly-A tail. 28. The RNA molecule of paragraph 27, wherein the poly-A tail comprises a sequence having SEQ ID NO: 22. 29. The RNA molecule of paragraph 27, wherein the poly-A tail comprises a sequence having SEQ ID NO: 26, optionally comprising +/-1 or +/-2 adenosine (A). 30. The RNA molecule of paragraph 1, comprising a 5′ UTR comprising the sequence of SEQ ID NOs: 373, 380 or 385, an open reading frame comprising the sequence of any of SEQ ID NOs: 15 or 16 and a 3′ UTR comprising the sequence of SEQ ID NO: 567. 31. The RNA molecule of paragraph 1, comprising a 5′ UTR comprising the sequence of any of SEQ ID NOs: 373, 380 or 385, an open reading frame comprising the sequence of any of SEQ ID NOs: 15 or 16, a 3′ UTR comprising the sequence of SEQ ID NO: 567, and a poly-A tail comprising a sequences of SEQ ID NO: 22. 32. The RNA molecule of paragraph 1, wherein the RNA molecule is transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 11-14. 33. The RNA molecule of paragraph 1, wherein the RNA molecule comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 17-20. 34. The RNA molecule of any one of paragraphs 1 to 33, wherein the open reading frame was generated from codon-optimized DNA. 35. The RNA molecule of any one of paragraphs 1 to 34, wherein the open reading frame comprises a G/C content of at least 55%, at least 60%, at least 65%, at least 70%, or at least 75%, of or of about 50% to 75% or 55% to 70%, or of or of about 58%, 66%, or 62%. 36. The RNA molecule of any one of paragraphs 1 to 35, wherein the encoded RSV polypeptide localizes in the cellular membrane, localizes in the Golgi and/or is anchored in the membrane and is secreted. 37. The RNA molecule of any of paragraphs 1 to 36, wherein the RNA molecule comprises stabilized RNA. 38. The RNA molecule of any one of paragraphs 1 to 38, wherein the RNA comprises at least one modified nucleotide. 39. The RNA molecule of paragraph 38, wherein the modified nucleotide is pseudouridine, N1-methylpseudouridine, N1-ethylpseudouridine, 2-thiouridine, 4′-thiouridine, 5- methylcytosine, 5-methyluridine, 2-thio-1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl- pseudouridine, 2-thio-5-aza-uridine, 2-thio-dihydropseudouridine, 2-thio-dihydrouridine, 2- thio-pseudouridine, 4-methoxy-2-thio-pseudouridine, 4-methoxy-pseudouridine, 4-thio-1- methyl-pseudouridine, 4-thio-pseudouridine, 5-aza-uridine, dihydropseudouridine, 5- methoxyuridine OR 2′-O-methyl uridine. 40. The RNA molecule of paragraph 39, wherein the modified nucleotide is N1- methylpseudouridine (m1Ψ). 41. The RNA molecule of any one of paragraphs 1 to 40, wherein the RNA is mRNA. 42. The RNA molecule of paragraph 41, wherein the RNA is modRNA. 43. A composition comprising the RNA molecule of any one of paragraphs 1 to 42, wherein the RNA molecule is formulated in a lipid nanoparticle (RNA-LNP). 44. The composition of paragraph 43, wherein lipid nanoparticle comprises at least one of a cationic ionizable lipid, a PEG-lipid, a neutral lipid, and a steroid or steroid analog. 45. The composition of paragraph 44, wherein the cationic lipid is ((4- hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate), 1,2-dioleoyl-3- trimethylammonium propane (DOTAP), N,N-dimethyl-2,3-dioleyloxypropylamine (DODMA), 1,2-di-O-octadecenyl-3-trimethylammonium propane (DOTMA), 3-(N-( N′,N′- dimethylaminoethane)-carbamoyl)cholesterol (DC-Chol), dimethyldioctadecylammonium (DDAB), 1,2-dioleoyl-3-dimethylammonium-propane (DODAP), 1,2-diacyloxy-3- dimethylammonium propanes, 1,2-dialkyloxy-3-dimethylammonium propanes; dioctadecyldimethyl ammonium chloride (DODAC), 1,2-distearyloxy-N,N-dimethyl-3- aminopropane (DSDMA), 2,3-di(tetradecoxy)propyl-(2-hydroxyethyl)-dimethylazanium (DMRIE), 1,2-dimyristoyl-sn-glycero-3-ethylphosphocholine (DMEPC), 1,2-dimyristoyl-3- trimethylammonium propane (DMTAP), 1,2-dioleoyloxypropyl-3-dimethyl-hydroxyethyl ammonium bromide (DORIE), 2,3-dioleoyloxy-N-[2(spermine carboxamide)ethyl]-N,N- dimethyl-1-propanamium trifluoroacetate (DOSPA), 1,2-dilinoleyloxy-N,N- dimethylaminopropane (DLinDMA), 1,2-dilinolenyloxy-N,N-dimethylaminopropane (DLenDMA), dioctadecylamidoglycyl spermine (DOGS), 3-dimethylamino-2-(cholest-5-en- 3-beta-oxybutan-4-oxy)-1-(cis,cis-9,12-oc-tadecadienoxy)propane (CLinDMA), 2-[5′- (cholest-5-en-3-beta-oxy)-3′-oxapentoxy)-3-dimethyl-l-(cis,cis-9′,12′- octadecadienoxy)propane (CpLinDMA), N,N-dimethyl-3,4-dioleyloxybenzylamine (DMOBA), 1,2-N,N′-dioleylcarbamyl-3-dimethylaminopropane (DOcarbDAP), 2,3- dilinoleoyloxy-N,N-dimethylpropylamine (DLinDAP), 1,2-N,N′-dilinoleylcarbamyl-3- dimethylaminopropane (DLincarbDAP), 1,2-Dilinoleoylcarbamyl-3-dimethylaminopropane (DLinCDAP), 2,2-dilinoleyl-4-dimethylaminomethyl-[1,3]-dioxolane (DLin-K-DMA), 2,2- dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane (DLin-K-XTC2-DMA), 2,2-dilinoleyl-4-(2- dimethylaminoethyl)-[1,3]-dioxolane (DLin-KC2-DMA), heptatriaconta-6,9,28,31-tetraen- 19-yl-4-(dimethylamino)butanoate (DLin-MC3-DMA), N-(2-Hydroxyethyl)-N,N-dimethyl- 2,3-bis(tetradecyloxy)-1-propanaminium bromide (DMRIE), (±)-N-(3-aminopropyl)-N,N- dimethyl-2,3-bis(cis-9-tetradecenyloxy)-1-propanaminium bromide (GAP-DMORIE), (±)-N- (3-aminopropyl)-N,N-dimethyl-2,3-bis(dodecyloxy)-1-propanaminium bromide (GAP- DLRIE), (±)-N-(3-aminopropyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide (GAP-DMRIE), N-(2-Aminoethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1- propanaminium bromide (bAE-DMRIE), N-(4-carboxybenzyl)-N,N-dimethyl-2,3- bis(oleoyloxy)propan-1-aminium (DOBAQ), 2-({8-[(3b)-cholest-5-en-3-yloxy]octyl}oxy)- N,N-dimethyl-3-[(9Z,12Z)-octadeca-9,12-dien-1-yloxy]propan-1-amine (Octyl-CLinDMA), 1,2-dimyristoyl-3-dimethylammonium-propane (DMDAP), 1,2-dipalmitoyl-3- dimethylammonium-propane (DPDAP), N1-[2-((1S)-1-[(3-aminopropyl)amino]-4-[di(3- amino-propyl)amino]butylcarboxamido)ethyl]-3,4-di[oleyloxy]-benzamide (MVL5), 1,2- dioleoyl-sn-glycero-3-ethylphosphocholine (DOEPC), 2,3-bis(dodecyloxy)-N-(2- hydroxyethyl)-N,N-dimethylpropan-1-amonium bromide (DLRIE), N-(2-aminoethyl)-N,N- dimethyl-2,3-bis(tetradecyloxy)propan-1-aminium bromide (DMORIE), di((Z)-non-2-en-l-yl) 8,8′-((((2(dimethylamino)ethyl)thio)carbonyl)azanediyl)dioctanoate (ATX), N,N-dimethyl- 2,3-bis(dodecyloxy)propan-1-amine (DLDMA), N,N-dimethyl-2,3-bis(tetradecyloxy)propan- 1-amine (DMDMA), Di((Z)-non-2-en-l-yl)-9-((4- (dimethylaminobutanoyl)oxy)heptadecanedioate (L319), N-dodecyl-3-((2- dodecylcarbamoyl-ethyl)-{2-[(2-dodecylcarbamoyl-ethyl)-2-{(2-dodecylcarbamoyl-ethyl)-[2- (2-dodecylcarbamoyl-ethylamino)-ethyl]-amino}-ethylamino)propionamide (lipidoid 98N12- 5), 1-[2-[bis(2-hydroxydodecyl)amino]ethyl-[2-[4-[2-[bis(2 hydroxydodecyl)amino]ethyl]piperazin-1-yl]ethyl]amino]dodecan-2-ol (lipidoid 02-200); C 12-200; heptadecan-9-yl 8-((2-hydroxyethyl) (6-oxo-6-(undecyloxy)hexyl) amino) octanoate (SM-102), bis(2-hexyldecyl) 6,6'-((2-(methyl(4- octanamidobutyl)amino)ethyl)azanediyl)dihexanoate, bis(2-hexyldecyl) 6,6'-((2-((4- octanamidobutyl)amino)ethyl)azanediyl)dihexanoate, bis(2-hexyldecyl) 6,6'-((2-(methyl(4- tetradecanamidobutyl)amino)ethyl)azanediyl)dihexanoate, bis(2-hexyldecyl) 8,8'-((2- (methyl(3- nonanamidopropyl)amino)ethyl)azanediyl)dioctanoate, bis(2-hexyldecyl) 8,8'- ((2-(methyl(4- octanamidobutyl)amino)ethyl)azanediyl)dioctanoate, ((2-(methyl(4- octanamidobutyl)amino)ethyl)azanediyl)bis(nonane- 9,1-diyl) bis(2-butyloctanoate), bis(2- hexyldecyl) 6,6'-((2-((6-(decylamino)-6- oxohexyl)(methyl)amino)ethyl)azanediyl)dihexanoate, ((2-((4-(dibutylamino)-4- oxobutyl)(methyl)amino)ethyl)azanediyl)bis(hexane-6,1-diyl) bis(2- hexyldec anoate), bis(2-hexyldecyl) 6,6'-((2-((6-(benzyloxy)-6- oxohexyl)(methyl)amino)ethyl)azanediyl)dihexanoate, bis(2-hexyldecyl) 6,6'-((2-(methyl(6- (methyl(octyl)amino)-6- oxohexyl)amino)ethyl)azanediyl)dihexanoate, bis(2-hexyldecyl) 6,6'-((2-((6-((2-ethylhexyl)amino)-6- oxohexyl)(methyl)amino)ethyl)azanediyl)dihexanoate, ((4-((5-(dihexylamino)-5-oxopentyl)(methyl)amino)butyl)azanediyl)bis(hexane- 6,1-diyl) bis(2-hexyldecanoate), ((4-((5-(didecylamino)-5- oxopentyl)(methyl)amino)butyl)azanediyl)bis(hexane- 6,1-diyl) bis(2-hexyldecanoate), bis(2-hexyldecyl) 6,6'-((3-((5-(dihexylamino)-5- oxopentyl)(methyl)amino)propyl)azanediyl)dihexanoate, bis(2-hexyldecyl) 6,6'-((3-((5- methoxy-5- oxopentyl)(methyl)amino)propyl)azanediyl)dihexanoate, ((4-((5-(dioctylamino)- 5-oxopentyl)(methyl)amino)butyl)azanediyl)bis(hexane- 6,1-diyl) bis(2-hexyldecanoate), 6- ((2-((3-(a,a-diethylcarbamoyl)propyl)-n-methylamino)ethyl)(6-(1- hexylnonylcarbonyloxy)hexyl)amino)hexyl 2-hexyldecanoate, or 6-((2-((3-(7v,7v- dihexylcarbamoyl)propyl)-n-methylamino)ethyl)(6-(1- hexylnonylcarbonyloxy)hexyl)amino)hexyl 2-hexyldecanoate. 46. The composition of paragraph 45 wherein the cationic lipid is (4- hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate) (ALC-0315). 47. The composition of paragraph 45 or 46, wherein the PEGylatedlipid is PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramides (e.g. PEG-CerC14 or PEG-CerC20), PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, 2-[(polyethylene glycol)-2000]-N,N- ditetradecylacetamide, glycol-lipids including PEG-c-DOMG, PEG-c-DMA, PEG-s-DMG,N- [(methoxy polyethylene glycol)2000)carbamyl]-1,2-dimyristyloxlpropyl-3-amine (PEG-c- DMA), and PEG-2000-DMG, PEGylated diacylglycerol (PEG-DAG) such as 1 - (monomethoxy-polyethyleneglycol)-2,3-dimyristoylglycerol (PEG-DMG), a PEGylated phosphatidylethanoloamine (PEG-PE), a PEG succinate diacylglycerol (PEG-S-DAG) such as 4-O-(2’,3’- di(tetradecanoyloxy)propyl-1-O-((o- methoxy(polyethoxy)ethyl)butanedioate (PEG-S-DMG), a PEGylated ceramide (PEG-cer), or a PEG dialkoxypropylcarbamate such as co-methoxy(polyethoxy)ethyl-N- (2,3di(tetradecanoxy)propyl)carbamate or 2,3-di(tetradecanoxy)propyl-N-(u>- methoxy(polyethoxy)ethyl)carbamate. 48. The composition of paragraph 47, wherein the PEG-lipid is 2-[(polyethylene glycol)-2000]- N,N-ditetradecylacetamide (ALC-0159). 49. The composition of any one of paragraphs 44 to 47, wherein the neutral lipid is distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl-phosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE) and dioleoyl-phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane- 1carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl- phosphatidylethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, 1-stearioyl-2- oleoylphosphatidyethanol amine (SOPE), or 1,2-dielaidoyl-sn-glycero-3- phophoethanolamine (transDOPE). 50. The composition of paragraph 49, wherein the neutral lipid is 1,2-distearoyl-sn-glycero-3- phosphocholine (DSPC). 51. The composition of any one of paragraphs 45 to 50, wherein the lipid nanoparticle comprises a steroid or steroid analog. 52. The composition of paragraph 51, wherein the steroid or steroid analog is cholesterol. 53. The composition of any one of paragraphs 45 to 52, wherein lipid nanoparticle wherein has a mean diameter of about 1 to about 500 nm. 54. The composition of any one of paragraphs 45 to 53, wherein the RNA is from RSV subtype A. 55. The composition of any one of paragraphs 45 to 53, wherein the RNA is from RSV subtype B. 56. The composition of any one of paragraphs 45 to 53, wherein the RNA is from: (i) RSV subtype A and (ii) RSV subtype B. 57. The composition of any one of paragraphs 45 to 56, comprising an RNA molecule at a concentration of or of about 0.01 to 0.09 mg/mL formulated in a lipid nanoparticle (LNP) comprising a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL, a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL, a neutral lipid at a concentration of or of about 0.1 to 0.25 mg/mL and a steroid or steroid analog at a concentration of or of about 0.3 to 0.45 mg/mL. 58. The composition of any one of paragraphs 45 to 57, comprising an RNA molecule at a concentration of or of about 0.01 to 0.09 mg/mL formulated in a lipid nanoparticle (LNP) comprising (4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate) (ALC- 0315) at a concentration of or of about 0.8 to 0.95 mg/mL, 2-[(polyethylene glycol)-2000]- N,N-ditetradecylacetamide (ALC-0159) at a concentration of or of about 0.05 to 0.15 mg/mL, 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) at a concentration of or of about 0.1 to 0.25 mg/mL and cholesterol at a concentration of or of about 0.3 to 0.45 mg/mL. 59. The composition of any one of paragraphs 45 to 58, comprising an RNA molecule at a concentration of or of about 0.06 mg/mL formulated in a lipid nanoparticle (LNP). 60. The composition of any one of paragraphs 45 to 59, further comprising at least one of a buffer, a stabilizing agent, salt, surfactant, preservative, excipient, and/or adjuvant. 61. The composition of any one of paragraphs 45 to 60, further comprising at least a buffer and a stabilizing agent, and optionally, a salt diluent. 62. The composition of paragraph 45 or 61, wherein the buffer is a Tris buffer. 63. The composition of paragraph 62, wherein the Tris buffer comprises tromethamine and Tris hydrochloride (HCl). 64. The composition of paragraph 63, wherein the tromethamine is at a concentration of or of about 0.1 to 0.3 mg/mL or of or of about 0.01 to 0.15 mg/mL. 65. The composition of paragraph 62 or 63, wherein and the Tris HCl is at a concentration of or of about 1.25 to 1.40 mg/mL or of or of about 0.5 to 0.65 mg/mL. 66. The composition of any one of paragraphs 63 to 65, wherein the stabilizing agent is sucrose. 67. The composition of paragraph 66, wherein the sucrose is at a concentration of or of about 95 to 110 mg/mL or of or of about 35 to 50 mg/mL. 68. The composition of any one of paragraphs 60 to 67, wherein the salt diluent for reconstitution is sodium chloride. 69. The composition of paragraph 68, wherein the sodium chloride is at a concentration of or of about 5 to 15 mg/mL. 70. The composition of any one of paragraphs 45 to 69, wherein the composition is a liquid or lyophilized. 71. The composition of paragraph 70, comprising an RNA molecule at a concentration of or of about 0.01 to 0.09 mg/mL formulated in a lipid nanoparticle (LNP) comprising a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL, a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL, a neutral lipid at a concentration of or of about 0.1 to 0.25 mg/mL and a steroid or steroid analog at a concentration of or of about 0.3 to 0.45 mg/mL, and further comprising a Tris buffer comprising tromethamine at a concentration of or of about 0.1 to 0.3 mg/mL and Tris hydrochloride (HCl) at a concentration of or of about 1.25 to 1.40 mg/mL, and sucrose at a concentration of or of about 95 to 110 mg/mL, wherein the composition is a liquid composition. 72. The composition of paragraph 70, comprising an RNA molecule at a concentration of or of about 0.01 to 0.09 mg/mL formulated in a lipid nanoparticle (LNP) comprising a cationic lipid at a concentration of or of about 0.8 to 0.95 mg/mL, a PEGylated lipid at a concentration of or of about 0.05 to 0.15 mg/mL, a neutral lipid at a concentration of or ofabout 0.1 to 0.25 mg/mL and a steroid or steroid analog at a concentration of or of about 0.3 to 0.45 mg/mL, and further comprising a Tris buffer comprising tromethamine at a concentration of or of about 0.01 to 0.15 mg/mL and Tris hydrochloride (HCl) at a concentration of or of about 0.5 to 0.65 mg/mL, sucrose at a concentration of or of about 35 to 50 mg/mL. 73. The composition of paragraph 70, wherein the composition is reconstituted with sodium chloride at a concentration of or of about 5 to 15 mg/mL. 74. The composition of paragraph 70, wherein the composition is reconstituted with or with about 0.6 to 0.75 mL sodium chloride. 75. The composition of paragraph 70, further comprising or comprising about 5 to 15 mM Tris buffer, 200 to 400 mM sucrose at a pH of or of about 7.0 to 8.0, and optionally, 0.9% sodium chloride diluent to reconstitute. 76. A method of inducing an immune response against RSV in a subject, comprising administering to the subject an effective amount of one or more of the RNA molecules, RNA-LNPs and/or compositions of any one of paragraphs 1 to 75. 77. A method of preventing, treating or ameliorating an infection, disease or condition associated with RSV in a subject, comprising administering to a subject an effective amount of one or more of the RNA molecules, RNA-LNPs and/or compositions of any one of paragraphs 1 to 75. 78. The method of paragraph 76 or 77, wherein the infection, disease or condition is RSV infection-induced acute respiratory tract illness, including pneumonia and bronchitis. 79. Use of the RNA molecule, RNA-LNP and/or composition of any one of paragraphs 1 to 75 in the manufacture of a medicament for use in inducing an immune response against RSV in a subject. 80. Use of the RNA molecule, RNA-LNP and/or composition of any one of paragraphs 1 to 75 in the manufacture of a medicament for use in preventing, treating or ameliorating an infection, disease or condition in a subject. 81. The use of paragraph 80, wherein the infection, disease or condition is associated with RSV. 82. The use of paragraph 79 or 80, wherein the infection, disease or condition is RSV infection-related respiratory illness, including pneumonia and bronchitis. 83. The method or use of any one of paragraphs 76 to 82, wherein the subject is less than about 1 year of age, about 1 year of age or older, about 5 years of age or older, about 10 years of age or older, about 20 years of age or older, about 30 years of age or older, about 40 years of age or older, about 50 years of age or older, about 60 years of age or older, about 70 years of age or older, or older. 84. The method or use of any one of paragraphs 76 to 83, wherein the subject the subject is about 50 years of age or older. 85. The method or use of any one of paragraphs 76 to 83, wherein the subject the subject is a pregnant woman. 86. The method or use of any one of paragraphs 76 to 85, wherein the RNA molecule or composition is administered as a vaccine. 87. The method or use of any one of paragraphs 76 to 86, wherein the RNA molecule or composition is administered by intradermal or intramuscular injection. 88. The RNA molecule of paragraph 87, wherein the modRNA comprises nucleotides having SEQ ID NO: 17. 89. The RNA molecule of paragraph 87, wherein the modRNA comprises nucleotides having SEQ ID NO: 18. 90. The RNA molecule of paragraph 87, wherein the modRNA comprises nucleotides having SEQ ID NO: 19. 91. The RNA molecule of paragraph 87, wherein the modRNA comprises nucleotides having SEQ ID NO: 20. 92. The method or use of any one of paragraphs 76 to 91, wherein the subject is administered a single dose, two doses, three doses, or more doses of the RNA molecule and/or composition. 93. The method or use of any one of paragraphs 76 to 92, wherein the subject is administered a single dose of the RNA molecule and/or composition. 94. The method or use of any one of paragraphs 76 to 93, wherein the subject is administered two doses of the RNA molecule and/or composition. 95. The method or use of any one of paragraphs 76 to 94, wherein the subject is administered two doses of the RNA molecule and/or composition on Day 0 and on or about 2 months later. 96. The method or use of any one of paragraphs 76 to 95, wherein the subject is administered two doses of the RNA molecule and/or composition on Day 0 and on or about 6 months later. 97. The method or use of any one of paragraphs 76 to 96, wherein the subject is administered at least one booster dose of the RNA molecule and/or composition. 98. The method or use of any one of paragraphs 76 to 97, wherein the subject is administered a dose of at least or at least about 15 μg, 30 μg, 60 μg, 90 μg, 100 μg or higher RNA molecule and/or composition per administration. 99. The method or use of any one of paragraphs 76 to 98, wherein the subject is administered an injection with a volume of or of about 0.25 to 1 mL, including but not limited to, of or of about 0.25, 0.5, 1 mL. 100. The method or use of any one of paragraphs 76 to 99, wherein the RNA molecule, LNP or composition is administered as a vaccine. All of the methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this disclosure have been described in terms of certain aspects, it will be apparent to those of skill in the art that variations may be applied to the methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the disclosure. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the disclosure as defined by the appended claims. The contents of all cited references (including literature references, issued patents, published patent applications, and GENBANK® Accession numbers as cited throughout this application) recited in the application, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are hereby specifically and expressly incorporated by reference. When definitions of terms in documents that are incorporated by reference herein conflict with those used herein, the definitions used herein govern.

Claims

CLAIMS 1. An RNA molecule comprising at least one open reading frame encoding a respiratory syncytial virus (RSV) fusion protein F (F) polypeptide and a 5’ untranslated region (5’ UTR), wherein the 5’ UTR comprises a nucleic acid sequence having at least 90% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336-344, 356-358, 360, 362-367, 370-375, and 378-396.
2. The RNA molecule of claim 1, wherein the 5’ UTR comprises a nucleic acid sequence having at least 92% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336, 341, 356-358, 372-375, and 378-396.
3. The RNA molecule of any one of claims 1-2, wherein the 5’ UTR comprises a nucleic acid sequence having at least 95% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 336, 357, 372-374, 378, 380, 384 and 385.
4. The RNA molecule of any one of claims 1-3, wherein the 5’ UTR comprises a nucleic acid sequence having at least 98% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 357, 372-374, 378, 380, 384 and 385.
5. The RNA molecule of any one of claims 1-4, wherein the 5’ UTR comprises a nucleic acid sequence having at least 98% identity to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 357, 373, 378, 380, 384 and 385.
6. The RNA molecule of any one of claims 1-5, wherein the 5’ UTR comprises a nucleic acid sequence selected from the group consisting of: SEQ ID NO: 373 (5UTR_563); SEQ ID NO: 380 (5UTR_582); and SEQ ID NO: 385 (5UTR_599).
7. The RNA molecule of any one of claims 1-6, wherein the UTR is a 3’ untranslated region (3’ UTR).
8. The RNA molecule of claim 7, wherein the 3’ UTR comprises nucleotides having sequence SEQ ID NO: 567 (3UTR_2 (hHBB)).
9. The RNA molecule of any one of claims 1-8, wherein the RNA molecule further comprises a 5’ cap moiety and/or a 3’ poly-A tail.
10. The RNA molecule of claim 9, wherein the poly-A tail comprises a sequence having SEQ ID NO: 22.
11. The RNA molecule of any one of claims 1-10, wherein the RSV F polypeptide is a full- length, truncated, fragment or variant thereof.
12. The RNA molecule of any one of claims 1-11, wherein the RSV F polypeptide comprises at least one mutation.
13. The RNA molecule of any one of claims 1 -12, wherein the RSV F polypeptide is a prefusion F polypeptide.
14. The RNA molecule of any one of claims 1-13, wherein the RSV F polypeptide comprises a combination of introduced amino acid mutations, wherein amino acid mutation positions are numbered according to SEQ ID NO: 1, and are selected from: (1) combination of 103C, 148C, 190I, and 486S; (2) combination of 54H, 103C, 148C, 190I, 296I, and 486S; or (3) combination of 54H, 55C, 188C, and 486S.
15. The RNA molecule of any one of claims 1-14, where in the RSV F polypeptide is linked to a trimerization domain.
16. The RNA molecule of claim 15, wherein the trimerization domain is a phage T4 fibritin foldon domain.
17. The RNA molecule of any one of claims 1-16, wherein the RSV F polypeptide has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to an amino acid sequence selected from SEQ ID NO: 1-6 or 621-626.
18. The RNA molecule of claim 17, wherein the RSV F polypeptide has an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 6 and 621-626.
19. The RNA molecule of claim 18, wherein the RSV F polypeptide has an amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 6.
20. The RNA molecule of any one of claims 1-19, wherein the open reading frame is transcribed from a nucleic acid comprising a nucleotide sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to any one of the sequences of SEQ ID NOs: 7-10 or 627-632.
21. The RNA molecule of claim 20, wherein the open reading frame is transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 8, 10, and 627-632.
22. The RNA molecule of claim 21, wherein the open reading frame is transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 8 and 10.
23. The RNA molecule of any one of claims 1-22, wherein the open reading frame comprises a nucleic acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to any one of the sequences of SEQ ID NOs: 15-16 or 633-638.
24. The RNA molecule of claim 23, wherein the open reading frame comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 15-16 and 633-638.
25. The RNA molecule of claim 24, wherein the open reading frame comprises a nucleic acid sequence selected from the group consisting of SEQ ID Nos: 15 and 16.
26. The RNA molecule of claim 1, wherein the RNA molecule is transcribed from a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 11-14 and 639-640.
27. The RNA molecule of claim 1, wherein the RNA molecule comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 17-20 and 641-642.
28. The RNA molecule of any one of claims 1-27, wherein the open reading frame comprises a G/C content of at least 55%, 60%, 65%, 70%, or 75%, or of or of about 50% to 75% or 55% to 70%.
29. The RNA molecule of any one of claims 1-28, wherein the encoded RSV polypeptide localizes in the cellular membrane, localizes in the Golgi and/or is secreted.
30. The RNA molecule of any one of claims 1-29, wherein the RNA comprises at least one modified nucleotide.
31. The RNA molecule of claim 30, wherein the modified nucleotide is pseudouridine (Ψ), N1-methylpseudouridine (m1 Ψ), N1-ethylpseudouridine, 2-thiouridine, 4′-thiouridine, 5-methylcytosine, 5-methyluridine, 2-thio-1-methyl-1-deaza-pseudouridine, 2-thio-1- methyl-pseudouridine, 2-thio-5-aza-uridine, 2-thio-dihydropseudouridine, 2-thio- dihydrouridine, 2-thio-pseudouridine, 4-methoxy-2-thio-pseudouridine, 4-methoxy- pseudouridine, 4-thio-1-methyl-pseudouridine, 4-thio-pseudouridine, 5-aza-uridine, dihydropseudouridine, 5-methoxyuridine or 2′-O-methyl uridine.
32. The RNA molecule of any one of claims 1-31, wherein the RNA is mRNA.
33. The RNA molecule of claim 32, wherein the RNA is modRNA.
34. The RNA molecule of claim 33, wherein the modified nucleotide is N1- methylpseudouridine (m1Ψ).
35. The RNA molecule of claim 34, wherein all uridine nucleotides in the RNA molecule have been replaced with N1-methylpseudouridine (m1Ψ).
36. A composition comprising the RNA molecule of any one of claims 1-35, wherein the RNA molecule is formulated in a lipid nanoparticle (RNA-LNP).
37. The composition according to claim 36, wherein the RSV molecule encodes an F protein of RSV subtype A.
38. The composition according to claim 36, wherein the RSV molecule encodes an F protein of RSV subtype B.
39. The composition according to claim 37, further comprising a second RNA molecule encoding an F protein of RSV subtype B.
40. The composition of any one of claims 36-39, wherein lipid nanoparticle comprises at least one of a cationic lipid, a PEGylated lipid, a neutral lipid, and a steroid or steroid analog.
41. The composition of claim 40, wherein the cationic lipid is (4- hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate) (ALC-0315).
42. The composition of claims 40 or 41, wherein the PEGylated lipid is PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramides (e.g. PEG-CerC14 or PEG-CerC20), PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, 2-[(polyethylene glycol)-2000]-N,N- ditetradecylacetamide, glycol-lipids including PEG-c-DOMG, PEG-c-DMA, PEG-s- DMG,N-[(methoxy polyethylene glycol)2000)carbamyl]-1,2-dimyristyloxlpropyl-3-amine (PEG-c-DMA), and PEG-2000-DMG, PEGylated diacylglycerol (PEG-DAG) such as 1 - (monomethoxy-polyethyleneglycol)-2,3-dimyristoylglycerol (PEG-DMG), a PEGylated phosphatidylethanoloamine (PEG-PE), a PEG succinate diacylglycerol (PEG-S-DAG) such as 4-O-(2’,3’- di(tetradecanoyloxy)propyl-1-O-((o- methoxy(polyethoxy)ethyl)butanedioate (PEG-S-DMG), a PEGylated ceramide (PEG- cer), or a PEG dialkoxypropylcarbamate such as co-methoxy(polyethoxy)ethyl-N- (2,3di(tetradecanoxy)propyl)carbamate or 2,3-di(tetradecanoxy)propyl-N-(u>- methoxy(polyethoxy)ethyl)carbamate.
43. The composition of claim 42, wherein the PEGylated lipid is 2-[(polyethylene glycol)- 2000]-N,N-ditetradecylacetamide (ALC-0159).
44. The composition of any one of claims 40-43, wherein the neutral lipid is distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl-phosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE) and dioleoyl-phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane- 1carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl- phosphatidylethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, 1-stearioyl-2- oleoylphosphatidyethanol amine (SOPE), or 1,2-dielaidoyl-sn-glycero-3- phophoethanolamine (transDOPE).
45. The composition of claim 44, wherein the neutral lipid is 1,2-distearoyl-sn-glycero-3- phosphocholine (DSPC).
46. The composition of any one of claims 40-45, wherein the steroid or steroid analog is cholesterol.
47. A method of inducing an immune response against RSV in a subject, comprising administering to the subject an effective amount of the RNA molecule of any one of claims 1-35.
48. A method of preventing, treating or ameliorating an infection, disease or condition associated with RSV in a subject, comprising administering to a subject an effective amount of the RNA molecule of any one of claims 1-35.
49. A method of inducing an immune response against RSV in a subject, comprising administering to the subject an effective amount of the composition of any one of claims 36-46.
50. A method of preventing, treating or ameliorating an infection, disease or condition associated with RSV in a subject, comprising administering to a subject an effective amount of the composition of any one of claims 36-46.
51. The method of claims 48 or 50, wherein the infection, disease or condition is RSV infection-induced acute respiratory tract illness, including pneumonia and bronchitis.
52. The method of any one of claims 47-51, wherein the subject is less than about 1 year of age, about 1 year of age or older, about 5 years of age or older, about 10 years of age or older, about 20 years of age or older, about 30 years of age or older, about 40 years of age or older, about 50 years of age or older, about 60 years of age or older, about 70 years of age or older, or older.
53. The method of any one of claims 47-52, wherein the RNA molecule or composition is administered as a vaccine.
54. The method of any one of claims 47-53, wherein the subject is administered a single dose, two doses, three doses, or more, and optionally, a booster dose of the RNA molecule, composition or vaccine.
PCT/IB2024/062510 2023-12-14 2024-12-11 Rna molecules Pending WO2025126071A1 (en)

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US202363610042P 2023-12-14 2023-12-14
US63/610,042 2023-12-14
US202463553480P 2024-02-14 2024-02-14
US63/553,480 2024-02-14
US202463565428P 2024-03-14 2024-03-14
US63/565,428 2024-03-14
US202463567866P 2024-03-20 2024-03-20
US63/567,866 2024-03-20
US202463639366P 2024-04-26 2024-04-26
US63/639,366 2024-04-26
US202463725096P 2024-11-26 2024-11-26
US63/725,096 2024-11-26

Publications (1)

Publication Number Publication Date
WO2025126071A1 true WO2025126071A1 (en) 2025-06-19

Family

ID=94216532

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2024/062510 Pending WO2025126071A1 (en) 2023-12-14 2024-12-11 Rna molecules

Country Status (1)

Country Link
WO (1) WO2025126071A1 (en)

Citations (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4367110A (en) 1979-07-02 1983-01-04 Toppan Printing Co. Decorative laminate and a manufacturing method therefor
US4452901A (en) 1980-03-20 1984-06-05 Ciba-Geigy Corporation Electrophoretically transferring electropherograms to nitrocellulose sheets for immuno-assays
US4578770A (en) 1982-08-30 1986-03-25 Musashi Engineering Kabushiki Kaisha Method of discriminating sheet
US4596792A (en) 1981-09-04 1986-06-24 The Regents Of The University Of California Safe vaccine for hepatitis containing polymerized serum albumin
US4599231A (en) 1984-03-09 1986-07-08 Scripps Clinic And Research Foundation Synthetic hepatitis B virus vaccine including both T cell and B cell determinants
US4599230A (en) 1984-03-09 1986-07-08 Scripps Clinic And Research Foundation Synthetic hepatitis B virus vaccine including both T cell and B cell determinants
US4601903A (en) 1985-05-01 1986-07-22 The United States Of America As Represented By The Department Of Health And Human Services Vaccine against Neisseria meningitidis Group B serotype 2 invasive disease
US4608251A (en) 1984-11-09 1986-08-26 Pitman-Moore, Inc. LHRH analogues useful in stimulating anti-LHRH antibodies and vaccines containing such analogues
US6733754B2 (en) 1999-01-29 2004-05-11 Pfizer, Inc. Adjuvants for use in vaccines
US20040142025A1 (en) 2002-06-28 2004-07-22 Protiva Biotherapeutics Ltd. Liposomal apparatus and manufacturing methods
US6793923B2 (en) 2000-11-07 2004-09-21 Immunovaccine Technologies, Inc. Vaccines with enhanced immune response and methods for their preparation
US20050147621A1 (en) * 2003-10-10 2005-07-07 Higgins Darren E. Use of bacterial 5' untranslated regions for nucleic acid expression
US20060148033A1 (en) * 2002-12-11 2006-07-06 Takara Bio Inc. Cold-induced expression vector
US20070042031A1 (en) 2005-07-27 2007-02-22 Protiva Biotherapeutics, Inc. Systems and methods for manufacturing liposomes
WO2008147196A2 (en) 2007-06-01 2008-12-04 Aimm Therapeutics B.V. Rsv-specific binding molecules and means for producing them
US20080312179A1 (en) * 2004-01-20 2008-12-18 Insight Biopharmaceuticals Ltd. High level expression of recombinant human erythropoietin having a modified 5'-UTR
WO2009079796A1 (en) 2007-12-24 2009-07-02 Id Biomedical Corporation Of Quebec Recombinant rsv antigens
US20090226470A1 (en) 2007-12-11 2009-09-10 Mauro Vincent P Compositions and methods related to mRNA translational enhancer elements
WO2010149745A1 (en) 2009-06-24 2010-12-29 Glaxosmithkline Biologicals S.A. Recombinant rsv antigens
WO2011008974A2 (en) 2009-07-15 2011-01-20 Novartis Ag Rsv f protein compositions and methods for making same
WO2011043643A1 (en) 2009-10-06 2011-04-14 Medimmune Ltd Rsv-specific binding molecule
WO2012019780A1 (en) 2010-08-13 2012-02-16 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
WO2013016058A1 (en) 2011-07-22 2013-01-31 Merck Sharp & Dohme Corp. Novel bis-nitrogen containing cationic lipids for oligonucleotide delivery
WO2013078199A2 (en) 2011-11-23 2013-05-30 Children's Medical Center Corporation Methods for enhanced in vivo delivery of synthetic, modified rnas
WO2013086373A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
WO2013103659A1 (en) 2012-01-04 2013-07-11 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Stabilizing rna by incorporating chain-terminating nucleosides at the 3'-terminus
WO2014132064A2 (en) * 2013-02-27 2014-09-04 University Of Newcastle Upon Tyne Cells and methods for fatty acid synthesis
WO2014160463A1 (en) 2013-03-13 2014-10-02 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Prefusion rsv f proteins and their use
WO2014174018A1 (en) 2013-04-25 2014-10-30 Crucell Holland B.V. Stabilized soluble prefusion rsv f polypeptides
WO2014202570A1 (en) 2013-06-17 2014-12-24 Crucell Holland B.V. Stabilized soluble pre-fusion rsv f polypeptides
WO2015013551A1 (en) 2013-07-25 2015-01-29 Marshall Christopher Patrick Conformationally stabilized rsv pre-fusion f proteins
WO2015177312A1 (en) 2014-05-22 2015-11-26 Glaxosmithkline Biologicals Sa Rsvf trimerization domains
WO2015199952A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2016005324A1 (en) 2014-07-11 2016-01-14 Biontech Rna Pharmaceuticals Gmbh Stabilization of poly(a) sequence encoding dna sequences
WO2016091391A1 (en) 2014-12-12 2016-06-16 Curevac Ag Artificial nucleic acid molecules for improved protein expression
WO2016103238A1 (en) 2014-12-24 2016-06-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Recombinant metapneumovirus f proteins and their use
WO2016176330A1 (en) 2015-04-27 2016-11-03 The Trustees Of The University Of Pennsylvania Nucleoside-modified rna for inducing an adaptive immune response
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017005848A1 (en) 2015-07-07 2017-01-12 Janssen Vaccines & Prevention B.V. Stabilized soluble pre-fusion rsv f polypeptides
WO2017005844A1 (en) 2015-07-07 2017-01-12 Janssen Vaccines & Prevention B.V. Vaccine against rsv
WO2017075531A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017109629A1 (en) 2015-12-23 2017-06-29 Pfizer Inc. Rsv f protein mutants
WO2017117528A1 (en) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017174564A1 (en) 2016-04-05 2017-10-12 Janssen Vaccines & Prevention B.V. Vaccine against rsv
WO2018081289A2 (en) 2016-10-25 2018-05-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Prefusion piv f immunogens and their use
WO2018078053A1 (en) 2016-10-26 2018-05-03 Curevac Ag Lipid nanoparticle mrna vaccines
WO2018081480A1 (en) 2016-10-26 2018-05-03 Acuitas Therapeutics, Inc. Lipid nanoparticle formulations
WO2018107026A1 (en) 2016-12-09 2018-06-14 Sangamo Therapeutics, Inc. Delivery of target specific nucleases
WO2018109220A2 (en) 2016-12-16 2018-06-21 Institute For Research In Biomedicine Novel recombinant prefusion rsv f proteins and uses thereof
WO2018191657A1 (en) 2017-04-13 2018-10-18 Acuitas Therapeutics, Inc. Lipids for delivery of active agents
WO2018200943A1 (en) 2017-04-28 2018-11-01 Acuitas Therapeutics, Inc. Novel carbonyl lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2019036028A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036030A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036000A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036008A1 (en) 2017-08-16 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019202035A1 (en) * 2018-04-17 2019-10-24 Curevac Ag Novel rsv rna molecules and compositions for vaccination
WO2020061426A2 (en) 2018-09-21 2020-03-26 Acuitas Therapeutics, Inc. Systems and methods for manufacturing lipid nanoparticles and liposomes
WO2020081938A1 (en) 2018-10-18 2020-04-23 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020146805A1 (en) 2019-01-11 2020-07-16 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020234300A1 (en) 2019-05-20 2020-11-26 Valneva Se A subunit vaccine for treatment or prevention of a respiratory tract infection
WO2021030701A1 (en) 2019-08-14 2021-02-18 Acuitas Therapeutics, Inc. Improved lipid nanoparticles for delivery of nucleic acids
WO2021222639A2 (en) 2020-04-29 2021-11-04 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Recombinant human metapneumovirus f proteins and their use
WO2022016070A1 (en) 2020-07-16 2022-01-20 Acuitas Therapeutics, Inc. Cationic lipids for use in lipid nanoparticles
WO2022032154A2 (en) 2020-08-06 2022-02-10 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
WO2022067010A1 (en) 2020-09-25 2022-03-31 Modernatx, Inc. Multi-proline-substituted coronavirus spike protein vaccines
CN114277039A (en) * 2021-10-25 2022-04-05 浙江君怡生物科技有限公司 Respiratory syncytial virus mRNA vaccine and preparation method and application thereof
WO2022076669A1 (en) 2020-10-09 2022-04-14 Board Of Regents, The University Of Texas System Prefusion-stabilized hmpv f proteins
WO2022175815A1 (en) * 2021-02-19 2022-08-25 Pfizer Inc. Methods of protecting rna
WO2022207839A2 (en) 2021-04-01 2022-10-06 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion piv3 f proteins
WO2022214678A2 (en) 2021-04-09 2022-10-13 Valneva Se Human metapneumo virus vaccine
WO2023283600A1 (en) * 2021-07-08 2023-01-12 Fred Hutchinson Cancer Center Method for analyzing an ability of target nucleic acid sequences to impact gene expression
WO2023102373A1 (en) 2021-11-30 2023-06-08 Sanofi Pasteur Inc. Human metapneumovirus vaccines
WO2023102388A1 (en) 2021-11-30 2023-06-08 Sanofi Pasteur Inc. Human metapneumovirus viral vector-based vaccines
WO2023114943A2 (en) 2021-12-16 2023-06-22 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2023114937A2 (en) 2021-12-16 2023-06-22 Acuitas Therapeutics, Inc. Fluorinated cationic lipids for use in lipid nanoparticles
WO2023110618A1 (en) 2021-12-16 2023-06-22 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion hmpv fusion proteins
WO2023114944A1 (en) 2021-12-16 2023-06-22 Acuitas Therapeutics, Inc. Fluorinated cationic lipids for use in lipid nanoparticles
WO2023217988A1 (en) 2022-05-12 2023-11-16 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion hmpv fusion proteins
WO2023250427A2 (en) 2022-06-22 2023-12-28 Flagship Pioneering Innovations V, Inc. Formulations for modulating myc expression
WO2024054843A2 (en) 2022-09-07 2024-03-14 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2024233387A1 (en) 2023-05-05 2024-11-14 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations

Patent Citations (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4367110A (en) 1979-07-02 1983-01-04 Toppan Printing Co. Decorative laminate and a manufacturing method therefor
US4452901A (en) 1980-03-20 1984-06-05 Ciba-Geigy Corporation Electrophoretically transferring electropherograms to nitrocellulose sheets for immuno-assays
US4596792A (en) 1981-09-04 1986-06-24 The Regents Of The University Of California Safe vaccine for hepatitis containing polymerized serum albumin
US4578770A (en) 1982-08-30 1986-03-25 Musashi Engineering Kabushiki Kaisha Method of discriminating sheet
US4599231A (en) 1984-03-09 1986-07-08 Scripps Clinic And Research Foundation Synthetic hepatitis B virus vaccine including both T cell and B cell determinants
US4599230A (en) 1984-03-09 1986-07-08 Scripps Clinic And Research Foundation Synthetic hepatitis B virus vaccine including both T cell and B cell determinants
US4608251A (en) 1984-11-09 1986-08-26 Pitman-Moore, Inc. LHRH analogues useful in stimulating anti-LHRH antibodies and vaccines containing such analogues
US4601903A (en) 1985-05-01 1986-07-22 The United States Of America As Represented By The Department Of Health And Human Services Vaccine against Neisseria meningitidis Group B serotype 2 invasive disease
US6733754B2 (en) 1999-01-29 2004-05-11 Pfizer, Inc. Adjuvants for use in vaccines
US6793923B2 (en) 2000-11-07 2004-09-21 Immunovaccine Technologies, Inc. Vaccines with enhanced immune response and methods for their preparation
US20040142025A1 (en) 2002-06-28 2004-07-22 Protiva Biotherapeutics Ltd. Liposomal apparatus and manufacturing methods
US20060148033A1 (en) * 2002-12-11 2006-07-06 Takara Bio Inc. Cold-induced expression vector
US20050147621A1 (en) * 2003-10-10 2005-07-07 Higgins Darren E. Use of bacterial 5' untranslated regions for nucleic acid expression
US20080312179A1 (en) * 2004-01-20 2008-12-18 Insight Biopharmaceuticals Ltd. High level expression of recombinant human erythropoietin having a modified 5'-UTR
US20070042031A1 (en) 2005-07-27 2007-02-22 Protiva Biotherapeutics, Inc. Systems and methods for manufacturing liposomes
WO2008147196A2 (en) 2007-06-01 2008-12-04 Aimm Therapeutics B.V. Rsv-specific binding molecules and means for producing them
US20090226470A1 (en) 2007-12-11 2009-09-10 Mauro Vincent P Compositions and methods related to mRNA translational enhancer elements
WO2009079796A1 (en) 2007-12-24 2009-07-02 Id Biomedical Corporation Of Quebec Recombinant rsv antigens
WO2010149745A1 (en) 2009-06-24 2010-12-29 Glaxosmithkline Biologicals S.A. Recombinant rsv antigens
WO2011008974A2 (en) 2009-07-15 2011-01-20 Novartis Ag Rsv f protein compositions and methods for making same
WO2011043643A1 (en) 2009-10-06 2011-04-14 Medimmune Ltd Rsv-specific binding molecule
WO2012019780A1 (en) 2010-08-13 2012-02-16 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
WO2013016058A1 (en) 2011-07-22 2013-01-31 Merck Sharp & Dohme Corp. Novel bis-nitrogen containing cationic lipids for oligonucleotide delivery
WO2013078199A2 (en) 2011-11-23 2013-05-30 Children's Medical Center Corporation Methods for enhanced in vivo delivery of synthetic, modified rnas
WO2013086373A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
WO2013103659A1 (en) 2012-01-04 2013-07-11 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Stabilizing rna by incorporating chain-terminating nucleosides at the 3'-terminus
WO2014132064A2 (en) * 2013-02-27 2014-09-04 University Of Newcastle Upon Tyne Cells and methods for fatty acid synthesis
WO2014160463A1 (en) 2013-03-13 2014-10-02 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Prefusion rsv f proteins and their use
WO2014174018A1 (en) 2013-04-25 2014-10-30 Crucell Holland B.V. Stabilized soluble prefusion rsv f polypeptides
WO2014202570A1 (en) 2013-06-17 2014-12-24 Crucell Holland B.V. Stabilized soluble pre-fusion rsv f polypeptides
WO2015013551A1 (en) 2013-07-25 2015-01-29 Marshall Christopher Patrick Conformationally stabilized rsv pre-fusion f proteins
WO2015177312A1 (en) 2014-05-22 2015-11-26 Glaxosmithkline Biologicals Sa Rsvf trimerization domains
WO2015199952A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
US9737619B2 (en) 2014-06-25 2017-08-22 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2016005324A1 (en) 2014-07-11 2016-01-14 Biontech Rna Pharmaceuticals Gmbh Stabilization of poly(a) sequence encoding dna sequences
WO2016091391A1 (en) 2014-12-12 2016-06-16 Curevac Ag Artificial nucleic acid molecules for improved protein expression
WO2016103238A1 (en) 2014-12-24 2016-06-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Recombinant metapneumovirus f proteins and their use
WO2016176330A1 (en) 2015-04-27 2016-11-03 The Trustees Of The University Of Pennsylvania Nucleoside-modified rna for inducing an adaptive immune response
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017005848A1 (en) 2015-07-07 2017-01-12 Janssen Vaccines & Prevention B.V. Stabilized soluble pre-fusion rsv f polypeptides
WO2017005844A1 (en) 2015-07-07 2017-01-12 Janssen Vaccines & Prevention B.V. Vaccine against rsv
WO2017075531A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
US10166298B2 (en) 2015-10-28 2019-01-01 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017109629A1 (en) 2015-12-23 2017-06-29 Pfizer Inc. Rsv f protein mutants
WO2017117528A1 (en) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017174564A1 (en) 2016-04-05 2017-10-12 Janssen Vaccines & Prevention B.V. Vaccine against rsv
WO2018081289A2 (en) 2016-10-25 2018-05-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Prefusion piv f immunogens and their use
WO2018081480A1 (en) 2016-10-26 2018-05-03 Acuitas Therapeutics, Inc. Lipid nanoparticle formulations
WO2018078053A1 (en) 2016-10-26 2018-05-03 Curevac Ag Lipid nanoparticle mrna vaccines
WO2018107026A1 (en) 2016-12-09 2018-06-14 Sangamo Therapeutics, Inc. Delivery of target specific nucleases
WO2018109220A2 (en) 2016-12-16 2018-06-21 Institute For Research In Biomedicine Novel recombinant prefusion rsv f proteins and uses thereof
WO2018191657A1 (en) 2017-04-13 2018-10-18 Acuitas Therapeutics, Inc. Lipids for delivery of active agents
WO2018200943A1 (en) 2017-04-28 2018-11-01 Acuitas Therapeutics, Inc. Novel carbonyl lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2019036008A1 (en) 2017-08-16 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036030A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036000A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036028A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019202035A1 (en) * 2018-04-17 2019-10-24 Curevac Ag Novel rsv rna molecules and compositions for vaccination
WO2020061426A2 (en) 2018-09-21 2020-03-26 Acuitas Therapeutics, Inc. Systems and methods for manufacturing lipid nanoparticles and liposomes
WO2020081938A1 (en) 2018-10-18 2020-04-23 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020146805A1 (en) 2019-01-11 2020-07-16 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020234300A1 (en) 2019-05-20 2020-11-26 Valneva Se A subunit vaccine for treatment or prevention of a respiratory tract infection
WO2021030701A1 (en) 2019-08-14 2021-02-18 Acuitas Therapeutics, Inc. Improved lipid nanoparticles for delivery of nucleic acids
WO2021222639A2 (en) 2020-04-29 2021-11-04 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Recombinant human metapneumovirus f proteins and their use
WO2022016070A1 (en) 2020-07-16 2022-01-20 Acuitas Therapeutics, Inc. Cationic lipids for use in lipid nanoparticles
WO2022032154A2 (en) 2020-08-06 2022-02-10 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
WO2022067010A1 (en) 2020-09-25 2022-03-31 Modernatx, Inc. Multi-proline-substituted coronavirus spike protein vaccines
WO2022076669A1 (en) 2020-10-09 2022-04-14 Board Of Regents, The University Of Texas System Prefusion-stabilized hmpv f proteins
WO2022175815A1 (en) * 2021-02-19 2022-08-25 Pfizer Inc. Methods of protecting rna
WO2022207839A2 (en) 2021-04-01 2022-10-06 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion piv3 f proteins
WO2022214678A2 (en) 2021-04-09 2022-10-13 Valneva Se Human metapneumo virus vaccine
WO2023283600A1 (en) * 2021-07-08 2023-01-12 Fred Hutchinson Cancer Center Method for analyzing an ability of target nucleic acid sequences to impact gene expression
CN114277039A (en) * 2021-10-25 2022-04-05 浙江君怡生物科技有限公司 Respiratory syncytial virus mRNA vaccine and preparation method and application thereof
WO2023102373A1 (en) 2021-11-30 2023-06-08 Sanofi Pasteur Inc. Human metapneumovirus vaccines
WO2023102388A1 (en) 2021-11-30 2023-06-08 Sanofi Pasteur Inc. Human metapneumovirus viral vector-based vaccines
WO2023114943A2 (en) 2021-12-16 2023-06-22 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2023114937A2 (en) 2021-12-16 2023-06-22 Acuitas Therapeutics, Inc. Fluorinated cationic lipids for use in lipid nanoparticles
WO2023110618A1 (en) 2021-12-16 2023-06-22 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion hmpv fusion proteins
WO2023114944A1 (en) 2021-12-16 2023-06-22 Acuitas Therapeutics, Inc. Fluorinated cationic lipids for use in lipid nanoparticles
WO2023217988A1 (en) 2022-05-12 2023-11-16 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion hmpv fusion proteins
WO2023250427A2 (en) 2022-06-22 2023-12-28 Flagship Pioneering Innovations V, Inc. Formulations for modulating myc expression
WO2024054843A2 (en) 2022-09-07 2024-03-14 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2024233387A1 (en) 2023-05-05 2024-11-14 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING CO
BECKERT, BMASQUIDA, B: "Methods in Molecular Biology", 2010, HUMANA PRESS, article "Synthesis of RNA by In vitro Transcription in RNA", pages: 703
BOWMAN, J.C.AZIZI, B.LENZ, T.K.RAY, P.WILLIAMS, L.D: "Methods", 2012, HUMANA PRESS, article "RNA in vitro transcription and RNA purification by denaturing PAGE in Recombinant and in vitro RNA syntheses", pages: 941
BRUNELLE, J.LGREEN, R.: "Chapter Five - In vitro transcription from plasmid or PCR-amplified DNA", METHODS IN ENZYMOLOGY, vol. 530, 2013, pages 101 - 114
CHAIWATPONGSAKORN, S.EPAND, R.F.COLLINS, P.L.EPAND R.M.PEEPLES, M.E., J VIROL., vol. 85, no. 8, 2011, pages 3968 - 77
DORMITZER, P.R.GRANDI, G.RAPPUOLI, R., NATURE REVIEWS MICROBIOL, vol. 10, 2012, pages 807
GILMAN MSMOIN SMMAS V ET AL., PLOS PATHOGENS, vol. 11, no. 7, 2015
GREEN, T.WP.G.M. WUTZ: "Protective Groups in Organic Synthesis", 1999, WILEY
GRUDZIEN-NOGALSKA, E.KOWALSKA, J.SU, W.KUHN, A.N.SLEPENKOV, S.V.DARYNKIEWICZ, E.SAHIN, U.JEMIELITY, J.RHOADS, R.E.: "Synthetic mRNAs with superior translation and stability properties in Synthetic Messenger RNA and Cell Metabolism Modulation in Methods", MOLECULAR BIOLOGY, 2013, pages 69
KAMAKAKA, R. TKRAUS, W. L: "In vitro Transcription", CURRENT PROTOCOLS IN CELL BIOLOGY, vol. 2, 2001
KOPPEL, D., J. CHEM. PHYS., vol. 57, 1972, pages 4814 - 4820
LEPPEK ET AL., NAT REV MOL CELL BIOL, 2018
LIM YITING ET AL: "Multiplexed functional genomic analysis of 5' untranslated region mutations across the spectrum of prostate cancer", NATURE COMMUNICATIONS, vol. 12, no. 1, 9 July 2021 (2021-07-09), XP093023532, Retrieved from the Internet <URL:https://www.nature.com/articles/s41467-021-24445-6> DOI: 10.1038/s41467-021-24445-6 *
LOSICK, R., ANN REV BIOCHEM, vol. 41, 1972, pages 409 - 46
MCLELLAN JSCHEN MLEUNG S ET AL.: "Structure of RSV fusion glycoprotein trimer bound to a pre-fusion-specific neutralizing antibody", SCIENCE, vol. 340, 2013, pages 1113 - 1117
NEDDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
NGWUTA, J.O.CHEN, M.MODJARRAD, K.JOYCE, M.G.KANEKIYO, M.KUMAR, A.YASSINE, H.M.MOIN, S.M.KILLIKELLY, A.M.CHUANG, G.Y., SCIENCE TRANSLAT. MED., vol. 14, no. 7, 2015, pages 309
PEARSONLIPMAN, PROC. NATL ACAD. SCI. USA, vol. 88, 1988, pages 2444
REMINGTON: "The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
SMITHWATERMAN, ADS APP. MATH, vol. 2, 1981, pages 482
WEINBERG ET AL., J INFECT DIS., vol. 201, no. 11, 1 June 2010 (2010-06-01), pages 1607 - 10
YUNUS, A.S.JACKSON T.P.CRISAFI, K.BURIMSKI, I.KILGORE, N.R.ZOUMPLIS, D.ALLAWAY, G.P.WILD, CTSALZWEDEL, K, VIROLOGY, vol. 396, no. 2, 20 January 2010 (2010-01-20), pages 226 - 37
ZENG ET AL., ADV MATER, vol. 32, no. 40, 2020, pages e2004452

Similar Documents

Publication Publication Date Title
US20230233671A1 (en) Rna molecules
EP4448002A1 (en) Polynucleotide compositions and uses thereof
TWI888970B (en) Rna molecules
US20240252612A1 (en) Immunogenic compositions and uses thereof
US20250000960A1 (en) Polynucleotide compositions and uses thereof
EP4608441A1 (en) Immunogenic compositions against influenza and rsv
JP2024151316A (en) RNA molecule
WO2025126071A1 (en) Rna molecules
AU2022343710A1 (en) Universal influenza vaccine and methods of use
EP4651890A1 (en) Immunogenic compositions and methods of inducing an immune response against varicella zoster virus
WO2025126019A1 (en) Rna molecules
KR20240152232A (en) Rna molecules
JP2025166811A (en) RNA molecules encoding RSV-F and vaccines containing the same
WO2025186719A1 (en) Immunogenic compositions and uses thereof
WO2025027492A1 (en) Coronavirus antigen variants
CN118574637A (en) RNA molecules
WO2025186725A2 (en) Improved lnp formulations and uses thereof
US20240376157A1 (en) Polynucleotides encoding norovirus vp1 antigens and uses thereof
HK40125987A (en) Immunogenic compositions and methods of inducing an immune response against varicella zoster virus
AU2024302344A1 (en) Method for stabilizing rna
WO2025202834A1 (en) Polynucleotide compositions and uses thereof
WO2025213131A1 (en) Rna compositions for delivery of orthopox antigens and related methods
CN118647397A (en) Polynucleotide compositions and uses thereof

Legal Events

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

Ref document number: 24834449

Country of ref document: EP

Kind code of ref document: A1