Coronavirus vaccine

文档序号:121273 发布日期:2021-10-22 浏览:17次 中文

阅读说明:本技术 冠状病毒疫苗 (Coronavirus vaccine ) 是由 U·沙欣 A·居勒 A·库恩 A·穆伊克 A·福格尔 K·瓦尔策 S·威策尔 S·海因 于 2021-04-16 设计创作,主要内容包括:本公开涉及用于预防或治疗冠状病毒感染的RNA的领域。特别地,本公开涉及用于针对冠状病毒感染进行疫苗接种并诱导有效的冠状病毒抗原特异性免疫应答如抗体和/或T细胞应答的方法和药剂。具体地,在一实施方案中,本公开涉及这样的方法,其包括向受试者给药RNA,所述RNA编码包含SARS-CoV-2刺突蛋白(S蛋白)表位的肽或蛋白(即编码疫苗抗原的疫苗RNA),用于在受试者中诱导针对冠状病毒S蛋白(特别是SARS-CoV-2的S蛋白)的免疫应答。(The present disclosure relates to the field of RNA for preventing or treating coronavirus infection. In particular, the present disclosure relates to methods and agents for vaccinating against coronavirus infection and inducing an effective coronavirus antigen-specific immune response, such as an antibody and/or T cell response. In particular, in one embodiment, the disclosure relates to methods comprising administering to a subject an RNA encoding a peptide or protein comprising an epitope of the spike protein (S protein) of SARS-CoV-2 (i.e., a vaccine RNA encoding a vaccine antigen) for inducing an immune response against the S protein of coronavirus, particularly the S protein of SARS-CoV-2, in the subject.)

1. A pharmaceutical composition comprising:

an RNA comprising an open reading frame encoding a polypeptide comprising a SARS-CoV-2S protein or an immunogenic fragment or variant thereof, wherein the RNA is formulated in a lipid nanoparticle,

the lipid nanoparticle comprises a cationic ionizable lipid, a neutral lipid, a steroid, and a polyethylene glycol (PEG) -lipid.

2. The composition of claim 1, wherein the immunogenic fragment is or comprises an RBD.

3. The composition of claim 1, wherein said RNA is a modified RNA that is modified by substituting some or all of the uridine residues with modified uridine residues.

4. The composition of claim 1, wherein the RNA is formulated in a lipid nanoparticle comprising a cationic ionizable lipid, a phospholipid, cholesterol, and a polyethylene glycol (PEG) -lipid.

5. The composition of claim 4, wherein the phospholipid is or comprises Distearoylphosphatidylcholine (DSPC).

6. The composition of claim 1, wherein the steroid is cholesterol.

7. The composition of claim 1, wherein the (PEG) -lipid is or comprises the structure:

or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein:

R12and R13Each independently is 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 linkages; and w has an average value of 30 to 60.

8. The composition of claim 1, wherein the cationic ionizable lipid has the following structure:

or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein:

G1and G2Each independently is unsubstituted C1-C12Alkylene or C1-C12An alkenylene group;

G3is C1-C24Alkylene radical, C1-C24Alkenylene radical, C3-C8Cycloalkylene radical, C3-C8Cycloalkenylene;

R1and R2Each independently is C 6-C24Alkyl or C6-C24An alkenyl group;

R3is H, OR5、CN、-C(=O)OR4、-OC(=O)R4or-NR5C(=O)R4

R4Is C1-C12An alkyl group;

R5is H or C1-C6An alkyl group.

9. The composition of claim 8, wherein R1Or R2Or both having one of the following structures:

10. the composition of claim 1, wherein the neutral lipids are present at a concentration of 5-15 mole percent of total lipid.

11. The composition of claim 1, wherein the cationic ionizable lipid is present at a concentration of 40-55 mole percent of total lipid.

12. The composition of claim 1, wherein the steroid is present at a concentration of 30-50 mole percent of total lipid.

13. The composition of claim 1, wherein the pegylated lipid is present at a concentration of 1-10 mole percent of total lipid.

14. The composition of claim 1, wherein the lipid nanoparticle comprises 40-55 mole percent cationic ionizable lipid, 5-15 mole percent neutral lipid, 30-50 mole percent steroid, and 1-10 mole percent pegylated lipid.

15. The composition of claim 1, wherein the RNA further comprises: 5 ' cap, 5 ' UTR, 3 ' UTR and polyA sequences.

16. The composition of claim 15, wherein the polyA sequence comprises at least 100 a nucleotides.

17. The composition of claim 16, wherein the polyA sequence is a break sequence of a nucleotides.

18. The composition of claim 1, wherein the RNA comprises a 5' cap.

19. The composition of claim 18, wherein the 5' cap is or comprises a cap1 structure.

20. The composition of claim 1, further comprising at least one salt and/or cryoprotectant.

21. The composition of claim 20, wherein the cryoprotectant is or comprises sucrose.

22. The composition of claim 1, wherein the composition is characterized by,

serum from mouse subjects administered doses of 0.2 μ g-5 μ g RNA showed production of antibodies to the polypeptides encoded by the open reading frames 7 days after administration.

23. The composition of claim 1, wherein the composition is characterized by,

sera from mouse subjects dosed with 1 μ g to 5 μ g of modified RNA showed pseudovirus neutralizing activity 14 days after dosing.

24. The composition of claim 1, wherein the RNA is present in the composition in an amount ranging from 1 μ g to 100 μ g per dose.

25. Use of a pharmaceutical composition in the manufacture of a vaccine for vaccinating a subject, the pharmaceutical composition comprising:

An RNA comprising an open reading frame encoding a polypeptide comprising a SARS-CoV-2S protein or an immunogenic fragment or variant thereof, wherein the RNA is formulated in a lipid nanoparticle comprising a cationic ionizable lipid, a neutral lipid, a steroid, and a pegylated lipid.

26. The use of claim 25, wherein a dose of the pharmaceutical composition is administered to the subject.

27. The use of claim 26, wherein said dose comprises an amount of said RNA in the range of 1 μ g to 100 μ g.

28. The use of claim 26, wherein said administration is by intramuscular administration.

29. The use of claim 26, further comprising administering to the subject at least a second dose a period of time after the first dose.

30. The use of claim 29, wherein the second dose is administered 7-28 days after the administration of the first dose.

Technical Field

The present disclosure relates to the field of RNA for preventing or treating coronavirus infection. In particular, the present disclosure relates to methods and agents for vaccinating against coronavirus infection and inducing an effective coronavirus antigen-specific immune response, such as an antibody and/or T cell response. The methods and agents are particularly useful for preventing or treating coronavirus infection. Administration of the RNA disclosed herein to a subject can protect the subject from coronavirus infection. In particular, in one embodiment, the disclosure relates to methods comprising administering to a subject an RNA encoding a peptide or protein comprising a SARS-CoV-2 spike protein (S protein) epitope (i.e., a vaccine RNA encoding a vaccine antigen) for inducing an immune response against a coronavirus S protein, particularly the S protein of SARS-CoV-2, in the subject. Administration of RNA encoding a vaccine antigen to a subject can provide the vaccine antigen (after expression of the RNA by appropriate target cells) for inducing an immune response against the vaccine antigen (and disease-associated antigen) in the subject.

Background

The gene sequence of SARS-CoV-2 (Severe acute respiratory syndrome coronavirus 2) has been used by WHO and the public (MN908947.3), and the virus is classified in the sub-family β -coronavirus. By sequence analysis, the phylogenetic tree shows a closer relationship to a Severe Acute Respiratory Syndrome (SARS) virus isolate than to another human-infecting coronavirus, the Middle East Respiratory Syndrome (MERS) virus. 2 months and 2 days, a total of 14,557 cases were globally confirmed in 24 countries, including Germany, and SARS-CoV-2 was then caused to become a global epidemic by self-sustained interpersonal viral spread.

Coronaviruses are positive-sense single-stranded RNA ((+) ssRNA) enveloped viruses that encode a total of 4 structural proteins, spike protein (S), envelope protein (E), membrane protein (M), and nucleocapsid protein (N). The spike protein (S protein) is responsible for receptor recognition, attachment to cells, infection by the endosomal pathway and genomic release driven by fusion of the virus and the endosomal membrane. Although the sequence differs between different family members, there are conserved regions and motifs within the S protein that allow the S protein to be divided into two subdomains: s1 and S2. S2, with its transmembrane domain, is responsible for membrane fusion, while the S1 domain recognizes the virus-specific receptor and binds to the target host cell. Within several coronavirus isolates, a Receptor Binding Domain (RBD) was identified and the general structure of the S protein was defined (figure 1).

At present, there is no vaccine method and therapy against SARS-CoV-2, but there is an urgent need.

Due to the importance of the S protein in host cell recognition and entry and in the induction of virus neutralizing antibodies by the host immune system, we decided to target the viral S protein of SARS-CoV-2 and the subdomain of the S protein such as S1 or RBD for vaccine development. Mutations in regions important for conformation may be beneficial in inducing a stronger protective immune response. Therefore, we envisage testing several constructs (fig. 2). Since the naive S protein is a trimer and this trimeric structure is likely to affect the stability and antigenicity of the protein, we introduced a strategy based on the stable construct, introducing the T4 bacteriophage secondary fibrin (fibritin) domain, which is also used in HIV to generate a stable gp140 trimer and act on the SARS RBD-construct.

Disclosure of Invention

The present invention generally includes immunotherapy of a subject comprising administering RNA, i.e., vaccine RNA, that encodes an amino acid sequence, i.e., a vaccine antigen, comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof, i.e., an antigenic peptide or protein. Thus, the vaccine antigen comprises an epitope of the SARS-CoV-2S protein for use in inducing an immune response in a subject against the coronavirus S protein, in particular the SARS-CoV-2S protein. The RNA encoding the vaccine antigen is administered to provide the antigen (after expression of the polynucleotide by an appropriate target cell) for inducing, i.e. stimulating, eliciting and/or amplifying an immune response, e.g. antibodies and/or immune effector cells, which target the target antigen (coronavirus S protein, in particular SARS-CoV-2S protein) or a processed product thereof. In one embodiment, the immune response induced according to the present disclosure is a B cell-mediated immune response, i.e., an antibody-mediated immune response. Additionally or alternatively, in one embodiment, the immune response induced according to the present disclosure is a T cell mediated immune response. In one embodiment, the immune response is an anti-coronavirus, in particular an anti-SARS-CoV-2 immune response.

The vaccines described herein comprise as an active ingredient a single-stranded RNA that can be translated into the corresponding protein upon entry into the recipient cell. In addition to the wild-type or codon-optimized sequence encoding the antigen sequence, the RNA may also comprise one or more structural elements optimized for the maximum efficacy of the RNA in terms of stability and translational efficiency (5' cap, 5' UTR, 3' UTR, poly (a) -tail). In one embodiment, the RNA comprises all of these elements. In one embodiment, β -S-ARCA (D1) (m)2 7,2'-OGppSpG) or m2 7,3’-OGppp(m1 2’-O) ApG can be used as a specific capping structure for the 5' -end of RNA drug substance. As 5'-UTR sequence, the 5' -UTR sequence of human α -globin mRNA can be used, optionally with an optimized "Kozak sequence" to improve translation efficiency. As a 3' -UTR sequence, a combination of two sequence elements (FI element) derived from a "split amino terminal enhancer of split" (AES) mRNA (referred to as F) and a mitochondrially encoded 12S ribosomal RNA (referred to as I) placed between the coding sequence and the poly (a) -tail can be used to ensure higher maximum protein levels and prolonged mRNA persistence. These have been selected ex vivo by sequences that confer RNA stability and increased total protein expression The procedure was identified (see WO 2017/060314, incorporated herein by reference). Alternatively, the 3'-UTR may be two repeated 3' -UTRs of human β -globin mRNA. In addition, a poly (a) -tail of 110 nucleotides in length can be used, consisting of a stretch of 30 adenosine residues, followed by a 10 nucleotide linker sequence (random nucleotides) and another 70 adenosine residues. This poly (A) -tail sequence was designed to enhance RNA stability and translation efficiency.

In addition, a secretory signal peptide (sec) may be fused to the antigen coding region, preferably in such a manner that sec is translated into an N-terminal tag. In one embodiment, sec corresponds to the secretory signal peptide of the S protein. Sequences encoding short linker peptides consisting mainly of the amino acids glycine (G) and serine (S) commonly used for fusion proteins can be used as GS/linker.

The vaccine RNAs described herein may be complexed with proteins and/or lipids (preferably lipids) to produce RNA-particles for administration. If a combination of different RNAs is used, the RNAs may be complexed together or separately with proteins and/or lipids to produce RNA-particles for administration.

In one aspect, the invention relates to a composition or pharmaceutical product (medical preparation) comprising an RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof.

In one embodiment, the immunogenic fragment of SARS-CoV-2S protein comprises the S1 subunit of SARS-CoV-2S protein or the Receptor Binding Domain (RBD) of the S1 subunit of SARS-CoV-2S protein.

In one embodiment, the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof is capable of forming a multimeric complex, in particular a trimeric complex. To this end, the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof may comprise a domain which allows the formation of a multimeric complex, in particular a trimeric complex comprising the amino acid sequence of the SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof. In one embodiment, the domain that allows for the formation of a multimeric complex comprises a trimerization domain, e.g., a trimerization domain described herein.

In one embodiment, the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof is encoded by a coding sequence that is codon optimized and/or has increased G/C content compared to the wild-type coding sequence, wherein said codon optimization and/or increased G/C content preferably does not alter the sequence of the encoded amino acid sequence.

In one embodiment of the process of the present invention,

(i) the RNA encoding the SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises the nucleotide sequence of nucleotide 979-1584 of SEQ ID NO:2, 8 or 9, and the sequence of SEQ ID NO: 2. 8 or 9 nucleotide 979-minus 1584 has a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical or a fragment of the nucleotide sequence of SEQ ID NO. 2, 8 or 9 nucleotide 979-minus 1584 or a fragment of a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of SEQ ID NO. 2, 8 or 9 nucleotide 979-minus 1584; and/or

(ii) a SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises the amino acid sequence of amino acid 327-528 of SEQ ID NO:1 in comparison with the sequence of SEQ ID NO:1 amino acid 327-528 has an amino acid sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical, or an immunogenic fragment of the amino acid sequence of SEQ ID No. 1 amino acid 327-528 or an immunogenic fragment of an amino acid sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of SEQ ID No. 1 amino acid 327-528.

In one embodiment of the process of the present invention,

an RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises the amino acid sequence of SEQ ID NO: 2. nucleotide sequence of nucleotides 49 to 2055 of 8 or 9, and SEQ ID NO: 2. 8 or 9 from nucleotide 49 to 2055 having a nucleotide sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity, or SEQ ID NO: 2. 8 or 9 or a fragment of a nucleotide sequence from nucleotides 49 to 2055 of SEQ ID NO: 2. 8 or 9, nucleotides 49-2055 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity; and/or

(ii) a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises the amino acid sequence of SEQ ID NO:1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-685 of SEQ ID No. 1, or an amino acid sequence of SEQ ID NO:1 or an immunogenic fragment of an amino acid sequence of amino acids 17 to 685 of SEQ ID NO:1, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the amino acid sequence of amino acids 17-685.

In one embodiment of the process of the present invention,

an RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises a nucleotide sequence of nucleotides 49 to 3819 of SEQ ID NO:2, 8 or 9, a sequence that hybridizes to SEQ ID NO: 2. 8 or 9, nucleotides 49 to 3819, or a fragment of a nucleotide sequence of SEQ ID No. 2, 8 or 9, or a nucleotide sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 49 to 3819 of SEQ ID No. 2, 8 or 9, or a fragment of a nucleotide sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 49 to 3819 of SEQ ID No. 2, 8 or 9; and/or

(ii) a SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises the amino acid sequence of amino acids 17-1273 of SEQ ID NO:1 or 7, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273 of SEQ ID NO:1 or 7, or the amino acid sequence of SEQ ID NO:1 or 7 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273 of SEQ ID No. 1 or 7.

In one embodiment, the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a secretory signal peptide.

In one embodiment, the secretory signal peptide is fused, preferably by N-terminal fusion, to the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of the SARS-CoV-2S protein or an immunogenic variant thereof.

In one embodiment of the process of the present invention,

(i) the RNA encoding the secretion signal peptide comprises the nucleotide sequence of nucleotides 1 to 48 of SEQ ID NO. 2, 8 or 9, a nucleotide sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotides 1 to 48 of SEQ ID NO. 2, 8 or 9, or the sequence of SEQ ID NO: 2. 8 or 9 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 1 to 48 of SEQ ID No. 2, 8 or 9; and/or

(ii) the secretion signal peptide comprises SEQ ID NO:1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 16 of SEQ ID No. 1, or a functional fragment of the amino acid sequence of amino acids 1 to 16 of SEQ ID No. 1: 1, having an amino acid sequence at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical.

In one embodiment of the process of the present invention,

an RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises the nucleotide sequence of SEQ ID NO:6, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO:6, or a fragment of the nucleotide sequence of SEQ ID NO:6 or a fragment of the nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO: 6; and/or

(ii) a SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises the amino acid sequence of SEQ ID NO:5, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO:5 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO: 5.

In one embodiment, the RNA is a modified RNA, in particular a stable mRNA. In one embodiment, the RNA comprises a modified nucleoside in place of at least one uridine. In one embodiment, the RNA comprises modified nucleosides in place of each uridine. In one embodiment, the modified nucleoside is independently selected from the group consisting of pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ), and 5-methyl-uridine (m 5U).

In one embodiment, the RNA comprises modified nucleosides in place of uridine.

In one embodiment, the modified nucleoside is selected from the group consisting of pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ) and 5-methyl-uridine (m 5U).

In one embodiment, the RNA comprises a 5' cap.

In one embodiment, an RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a 5' UTR comprising the nucleotide sequence of SEQ ID No. 12, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the nucleotide sequence of SEQ ID No. 12.

In one embodiment, the RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a 3' UTR comprising the amino acid sequence of SEQ ID NO: 13, or a nucleotide sequence identical to SEQ ID NO: 13 has a nucleotide sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical.

In one embodiment, the RNA encoding an amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a poly-A sequence.

In one embodiment, the poly-A sequence comprises at least 100 nucleotides.

In one embodiment, the poly-a sequence comprises SEQ ID NO:14, or consists of the nucleotide sequence of SEQ ID No. 14.

In one embodiment, the RNA is formulated as or to be formulated as a liquid, a solid, or a combination thereof.

In one embodiment, the RNA is or is to be formulated for injection.

In one embodiment, the RNA is or is to be formulated for intramuscular administration.

In one embodiment, the RNA is or is to be formulated as a particle.

In one embodiment, the particle is a Lipid Nanoparticle (LNP) or lipoplex (lpx) particle.

In one embodiment, the LNP particles comprise ((4-hydroxybutyl) azendiyl) bis (hexane-6,1-diyl) bis (2-hexyl decanoate) ((4-hydroxybutyl) azanediyl) bis (2-hexyldecanoate)), 2- [ (polyethylene glycol) -2000] -N, N-ditetradecylacetamide (2- [ (polyethylene glycol) -2000] -N, N-didecyllacetamide), 1, 2-Distearoyl-sn-glycerol-3-phosphocholine (1, 2-Distearoyl-sn-glycerol-3-phosphate) and cholesterol (cholestrol).

In one embodiment, the RNA lipoplex particles can be obtained by mixing RNA with liposomes. In one embodiment, the RNA lipoplex particles can be obtained by mixing RNA with a lipid.

In one embodiment, the RNA is or is to be formulated as a colloid. In one embodiment, the RNA is or is to be formulated as particles, forming a colloidal dispersed phase. In one embodiment, 50% or more, 75% or more, or 85% or more of RNA is present in the dispersed phase. In one embodiment, the RNA is formulated or to be formulated as a particle comprising RNA and a lipid. In one embodiment, the particles are formed by exposing RNA dissolved in an aqueous phase to lipids dissolved in an organic phase. In one embodiment, the organic phase comprises ethanol. In one embodiment, the particles are formed by exposing RNA dissolved in an aqueous phase to lipids dispersed in the aqueous phase. In one embodiment, the lipids dispersed in the aqueous phase form liposomes.

In one embodiment, the RNA is mRNA or saRNA.

In one embodiment, the composition or pharmaceutical product is a pharmaceutical composition.

In one embodiment, the composition or pharmaceutical product is a vaccine.

In one embodiment, the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers, diluents and/or excipients.

In one embodiment, the composition or pharmaceutical product is a kit.

In one embodiment, the RNA and optionally the particle-forming components are in separate vials.

In one embodiment, the kit further comprises instructions for using the composition or pharmaceutical preparation to induce an immune response against a coronavirus in a subject.

In one aspect, the invention relates to a composition or pharmaceutical product as described herein for pharmaceutical use.

In one embodiment, the pharmaceutical use comprises inducing an immune response against a coronavirus in a subject.

In one embodiment, the pharmaceutical use comprises the treatment or prophylactic treatment of a coronavirus infection.

In one embodiment, the compositions or pharmaceutical preparations described herein are for administration to a human.

In one embodiment, the coronavirus is a beta coronavirus.

In one embodiment, the coronavirus is sabevivirus (sarbecovirus).

In one embodiment, the coronavirus is SARS-CoV-2.

In one aspect, the invention relates to a method of inducing an immune response against a coronavirus in a subject, the method comprising administering to the subject a composition comprising an RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof.

In one embodiment, the immunogenic fragment of SARS-CoV-2S protein comprises the S1 subunit of SARS-CoV-2S protein or the Receptor Binding Domain (RBD) of the S1 subunit of SARS-CoV-2S protein.

In one embodiment, the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof is capable of forming a multimeric complex, in particular a trimeric complex. To this end, the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof may comprise a domain allowing the formation of multimeric complexes, in particular trimeric complexes of amino acid sequences comprising the SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof. In one embodiment, the domain that allows for the formation of a multimeric complex comprises a trimerization domain, e.g., as described herein.

In one embodiment, the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof is encoded by a coding sequence that is codon optimized and/or has increased G/C content compared to the wild-type coding sequence, wherein said codon optimization and/or increased G/C content preferably does not alter the sequence of the encoded amino acid sequence.

In one embodiment of the process of the present invention,

(i) the RNA encoding the SARS-CoV-2S protein, the immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or the immunogenic variant thereof comprises the nucleotide sequence of nucleotide 979-1584 of SEQ ID NO:2, 8 or 9, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotide 979-1584 of SEQ ID NO:2, 8 or 9, or a fragment of the nucleotide sequence of nucleotide 979-1584 of SEQ ID NO:2, 8 or 9 or a fragment of the nucleotide sequence of SEQ ID NO:2, 8 or 9: 2. a fragment of nucleotide 979-1584 of nucleotide 8 or 9 having a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical; and/or

(ii) a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises the amino acid sequence of SEQ ID NO:1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acid 327-528 of SEQ ID No. 1, or an immunogenic fragment of the amino acid sequence of amino acid 327-528 of SEQ ID No. 1 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acid 327-528 of SEQ ID No. 1.

In one embodiment of the process of the present invention,

an RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises a nucleotide sequence of nucleotides 49 to 2055 of SEQ ID NO:2, 8 or 9, a sequence that differs from the sequence of nucleotides 49 to 2055 of SEQ ID NO: 2. 8 or 9, nucleotide sequence from nucleotides 49 to 2055 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity, or a nucleotide sequence of SEQ ID NO: 2. 8 or 9 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 49-2055 of SEQ ID No. 2, 8 or 9; and/or

(ii) a SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises an amino acid sequence of amino acids 17 to 685 of SEQ ID NO:1, a variant of SEQ ID NO:1, or an amino acid sequence of amino acids 17 to 685 of SEQ ID NO:1 or an immunogenic fragment of an amino acid sequence of amino acids 17 to 685 of SEQ ID NO:1, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the amino acid sequence of amino acids 17-685.

In one embodiment of the process of the present invention,

an RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises the amino acid sequence of SEQ ID NO: 2. 8 or 9, and the nucleotide sequence of nucleotides 49-3819 of SEQ ID NO: 2. 8 or 9, nucleotide sequence 49-3819 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity, or a nucleotide sequence of SEQ ID NO: 2. 8 or 9 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 49-3819 of SEQ ID No. 2, 8 or 9; and/or

(ii) a SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises the amino acid sequence of amino acids 17-1273 of SEQ ID NO:1 or 7, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273 of SEQ ID NO:1 or 7, or the amino acid sequence of SEQ ID NO:1 or 7 or an immunogenic fragment of the amino acid sequence of amino acids 17-1273 that differs from SEQ ID NO:1 or 7, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273.

In one embodiment, the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a secretory signal peptide.

In one embodiment, the secretory signal peptide is fused, preferably by N-terminal fusion, to the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of the SARS-CoV-2S protein or an immunogenic variant thereof.

In one embodiment of the process of the present invention,

(i) the RNA encoding the secretion signal peptide comprises the nucleotide sequence of nucleotides 1 to 48 of SEQ ID NO. 2, 8 or 9, a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotides 1 to 48 of SEQ ID NO. 2, 8 or 9, or a fragment of the nucleotide sequence of nucleotides 1 to 48 of SEQ ID NO. 2, 8 or 9 or a fragment of the nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotides 1 to 48 of SEQ ID NO. 2, 8 or 9; and/or

(ii) the signal peptide comprises SEQ ID NO:1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-16 of SEQ ID No. 1, or SEQ ID NO:1 or a functional fragment of an amino acid sequence corresponding to amino acids 1 to 16 of SEQ ID NO:1, having an amino acid sequence at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical.

In one embodiment of the process of the present invention,

an RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises the nucleotide sequence of SEQ ID NO:6, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO:6, or the sequence of SEQ ID NO:6 or a fragment of the nucleotide sequence of SEQ ID NO:6, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity; and/or

(ii) a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises the amino acid sequence of SEQ ID NO:5, and SEQ ID NO:5, or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO:5 or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID No. 5.

In one embodiment, the RNA is a modified RNA, in particular a stable mRNA. In one embodiment, the RNA comprises a modified nucleoside in place of at least one uridine. In one embodiment, the RNA comprises modified nucleosides in place of each uridine. In one embodiment, the modified nucleoside is independently selected from the group consisting of pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ), and 5-methyl-uridine (m 5U).

In one embodiment, the RNA comprises modified nucleosides in place of uridine.

In one embodiment, the modified nucleoside is selected from the group consisting of pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ) and 5-methyl-uridine (m 5U).

In one embodiment, the RNA comprises a cap.

In one embodiment, an RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a 5' UTR comprising the nucleotide sequence of SEQ ID No. 12, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the nucleotide sequence of SEQ ID No. 12.

In one embodiment, an RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a 3' UTR comprising the nucleotide sequence of SEQ ID No. 13, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the nucleotide sequence of SEQ ID No. 13.

In one embodiment, the RNA encoding an amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a poly-A sequence.

In one embodiment, the poly-A sequence comprises at least 100 nucleotides.

In one embodiment, the poly-A sequence comprises the nucleotide sequence of SEQ ID NO 14, or consists of SEQ ID NO:14, or a nucleotide sequence of seq id no.

In one embodiment, the RNA is formulated as a liquid, a solid, or a combination thereof.

In one embodiment, the RNA is administered by injection.

In one embodiment, the RNA is administered by intramuscular administration.

In one embodiment, the RNA is formulated as a particle.

In one embodiment, the particle is a Lipid Nanoparticle (LNP) or lipoplex (lpx) particle.

In one embodiment, the LNP particles comprise ((4-hydroxybutyl) azelidinyl) bis (hexane-6, 1-diyl) bis (hexyl 2-decanoate), 2- [ (polyethylene glycol) -2000] -N, N-ditetradecylacetamide, 1, 2-distearoyl-sn-glycerol-3-phosphocholine, and cholesterol.

In one embodiment, the RNA lipoplex particles can be obtained by mixing RNA with liposomes. In one embodiment, the RNA lipoplex particles can be obtained by mixing RNA with a lipid.

In one embodiment, the RNA is formulated as a colloid. In one embodiment, the RNA is formulated as particles, forming a dispersed phase of colloid. In one embodiment, 50% or more, 75% or more, or 85% or more of RNA is present in the dispersed phase. In one embodiment, the RNA is formulated as a particle comprising RNA and a lipid. In one embodiment, the particles are formed by exposing RNA dissolved in an aqueous phase to lipids dissolved in an organic phase. In one embodiment, the organic phase comprises ethanol. In one embodiment, the particles are formed by exposing RNA dissolved in an aqueous phase to lipids dispersed in the aqueous phase. In one embodiment, the lipids dispersed in the aqueous phase form liposomes.

In one embodiment, the RNA is mRNA or saRNA.

In one embodiment, the method is a method of vaccinating against coronavirus.

In one embodiment, the method is a method for the therapeutic or prophylactic treatment of a coronavirus infection.

In one embodiment, the subject is a human.

In one embodiment, the coronavirus is a beta coronavirus.

In one embodiment, the coronavirus is sabevivirus (sarbecovirus).

In one embodiment, the coronavirus is SARS-CoV-2.

In one embodiment of the methods described herein, the composition is a composition described herein.

In one aspect, the invention relates to a composition or pharmaceutical product as described herein for use in the methods described herein.

Among other things, the present disclosure demonstrates that a composition comprising lipid nanoparticle-encapsulated mRNA encoding at least a portion of (e.g., is or comprises) a polypeptide encoded by SARS-CoV-2 (e.g., an S protein encoded by SARS-CoV-2) can achieve a detectable antibody titer against an epitope in serum within 7 days after administration to a population of adult human subjects according to a protocol comprising administration of at least one dose of a vaccine composition. In addition, the present disclosure demonstrates the persistence of such antibody titers. In some embodiments, the present disclosure demonstrates this increase in antibody titer when using modified mRNA compared to that achieved with the corresponding unmodified mRNA.

In some embodiments, the provided regimen comprises at least one dose. In some embodiments, a provided regimen comprises a first dose and at least one subsequent dose. In some embodiments, the first dose is the same amount as at least one subsequent dose. In some embodiments, the first dose is the same amount as all subsequent doses. In some embodiments, the first dose is a different amount than at least one subsequent dose. In some embodiments, the first dose is an amount that is different from all subsequent doses. In some embodiments, the provided regimen comprises two doses. In some embodiments, the provided regimen consists of two doses.

In particular embodiments, the immunogenic composition is formulated as a single dose in a container (e.g., a vial). In some embodiments, the immunogenic composition is formulated as a multi-dose formulation in a vial. In some embodiments, the multi-dose formulation comprises at least 2 doses per vial. In some embodiments, the multi-dose formulation comprises a total of 2-20 doses/vial, e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 doses/vial. In some embodiments, each dose in the vial is equal in volume. In some embodiments, the first dose is a different volume than the subsequent dose.

"stable" multi-dose formulations do not exhibit unacceptable levels of microbial growth and are substantially free or free of decomposition or degradation of active biomolecule components. As used herein, a "stable" immunogenic composition includes a formulation that is capable of eliciting a desired immune response when administered to a subject.

In some embodiments, the multi-dose formulation remains stable for a specified period of time after multiple or repeated accesses (injections)/insertion of the multi-dose container. For example, in some embodiments, the multi-dose formulation, when contained within a multi-dose container, may be stable for at least 3 days, up to 10 uses. In some embodiments, the multi-dose formulation remains stable after 2-20 inoculations/insertions.

In some embodiments, administration of a composition comprising lipid nanoparticle-encapsulated mRNA encoding at least a portion (e.g., is or comprises an epitope) of a SARS-CoV-2-encoding polypeptide (e.g., a SARS-CoV-2-encoding S protein) can result in lymphocyte depletion in some subjects (e.g., in all subjects, in a majority of subjects, in about 50% or less, in about 40% or less, in about 25% or less, in about 20% or less, in about 15% or less, in about 10% or less, in about 5% or less, etc.) according to a protocol as described herein. Among other things, the present disclosure demonstrates that this lymphopenia may resolve over time (resolve). For example, in some embodiments, the lymphopenia is resolved in about 14, about 10, about 9, about 8, about 7 days, or less. In some embodiments, the lymphopenia is grade 3, grade 2, or less.

Accordingly, the present disclosure provides compositions comprising lipid nanoparticle-encapsulated mrnas encoding at least a portion (e.g., is or comprises an epitope) of a SARS-CoV-2-encoded polypeptide (e.g., an S protein encoded by SARS-CoV-2), the compositions characterized by exhibiting certain characteristics (e.g., achieving certain effects) as described herein, when administered to a population of related adults. In some embodiments, provided compositions can be prepared, stored, transported, characterized, and/or used under conditions in which the temperature does not exceed a particular threshold. Alternatively or additionally, in some embodiments, provided compositions may be protected from light (e.g., from certain wavelengths) in some or all of their preparation, storage, transport, characterization, and/or use. In some embodiments, one or more characteristics of a provided composition (e.g., mRNA stability, e.g., as may be assessed by one or more of size, presence or modification of a particular moiety, etc.; lipid nanoparticle stability or aggregation, pH, etc.) may or may have been assessed at one or more points in preparation, storage, transport, and/or use prior to administration.

Among other things, the present disclosure notes that certain provided compositions in which nucleotides within the mRNA are unmodified (e.g., are naturally occurring A, U, C, G) and/or methods involving the provision of such compositions (e.g., which may be or comprise, in some embodiments, an adult population when administered to a relevant population) are characterized by intrinsic adjuvant effects. In some embodiments, such compositions and/or methods may induce an antibody and/or T cell response. In some embodiments, such compositions and/or methods may induce a higher T cell response compared to conventional vaccines (e.g., non-mRNA vaccines such as protein vaccines).

Alternatively or additionally, the present disclosure records provided compositions in which nucleotides within the mRNA are modified (e.g., compositions comprising lipid nanoparticle-encapsulated mrnas encoding at least a portion (e.g., is or comprises an epitope) of a SARS-CoV-2 encoded polypeptide (e.g., S protein encoded by SARS-CoV-2)) and/or methods involving the provision of such compositions (e.g., which may be or comprise an adult human population in some embodiments when administered to a relevant population) are characterized by the absence of an intrinsic adjuvant effect, or a reduced intrinsic adjuvant effect as compared to other comparable compositions (or methods) having an unmodified outcome. Alternatively or additionally, in some embodiments, such compositions (or methods) are characterized in that they (e.g., when administered to a relevant population, which in some embodiments may be or comprise an adult population) induce an antibody response and/or a CD4+ T cell response. Further alternatively or additionally, in some embodiments such compositions (or methods) are characterized in that they (e.g., when administered to a relevant population, which in some embodiments may be or comprise an adult population) induce a higher CD4+ T cell response than observed with alternative vaccine formats (e.g., peptide vaccines). In some embodiments involving modified nucleotides, such modified nucleotides may be present in, for example, a 3 'UTR sequence, an antigen coding sequence, and/or a 5' UTR sequence. In some embodiments, the modified nucleotide is or includes one or more modified uracil residues and/or one or more modified cytosine residues.

Among other things, the present disclosure records sustained expression of provided compositions (e.g., compositions comprising lipid nanoparticle-encapsulated mrnas encoding at least a portion (e.g., is or comprises an epitope) of a SARS-CoV-2-encoded polypeptide (e.g., an S protein encoded by SARS-CoV-2)) and/or methods (e.g., which can be or comprise an adult human population in some embodiments when administered to a relevant population) characterized by the encoded polypeptide (e.g., a SARS-CoV-2-encoded protein [ e.g., S protein ] or portion thereof, which can be or comprise an epitope thereof in some embodiments). For example, in some embodiments, such compositions and/or methods are characterized in that, when administered to a human, they effect detectable polypeptide expression in a biological sample (e.g., serum) from such human, and in some embodiments, such expression is for a period of at least 36 hours or longer, including, e.g., at least 48 hours, at least 60 hours, at least 72 hours, at least 96 hours, at least 120 hours, at least 148 hours, or longer.

One of skill in the art reading this disclosure will understand that it describes various mRNA constructs that encode at least a portion (e.g., are or comprise an epitope) of a polypeptide encoded by SARS-CoV-2 (e.g., an S protein encoded by SARS-CoV-2). It will be particularly understood by those of ordinary skill in the art reading this disclosure that it describes various mRNA constructs that encode at least a portion of the SARS-CoV-2S protein, e.g., at least the RBD portion of the SARS-CoV-2S protein. Still further, one of ordinary skill in the art reading this disclosure will appreciate that it describes particular features and/or advantages of an mRNA construct that encodes at least a portion (e.g., is or comprises an epitope) of a polypeptide encoded by SARS-CoV-2 (e.g., an S protein encoded by SARS-CoV-2). Among other things, the present disclosure specifically notes the surprising and useful features and/or advantages of certain mRNA constructs that encode a SARS-CoV-2 RBD portion, and in some embodiments, do not encode a full-length SARS-CoV-2S protein. Without wishing to be bound by any particular theory, the present disclosure demonstrates that mRNA constructs provided that encode less than full-length SARS-CoV-2S proteins, and in particular mRNA constructs that encode at least the RBD portion of such SARS-CoV-2S proteins, are particularly useful and/or effective for use as or in immunogenic compositions (e.g., vaccines) and/or to achieve an immune effect as described herein (e.g., production of SARS-CoV-2 neutralizing antibodies, and/or a T cell response (e.g., CD4+ and/or CD8+ T cell response)).

In some embodiments, the present disclosure provides an RNA (e.g., mRNA) comprising an open reading frame encoding a polypeptide comprising a receptor binding portion of a SARS-CoV-2S protein, the RNA suitable for intracellular expression of the polypeptide. In some embodiments, such encoded polypeptides do not comprise a complete S protein. In some embodiments, the encoded polypeptide comprises a Receptor Binding Domain (RBD), e.g., as set forth in SEQ ID NO: 5, respectively. In some embodiments, the encoded polypeptide comprises a peptide according to SEQ ID NO:29 or 31. In some embodiments, such RNA (e.g., mRNA) can be complexed by (poly) cationic polymers, polyplex, proteins, or peptides. In some embodiments, such RNA can be formulated in a lipid nanoparticle (e.g., a lipid nanoparticle described herein). In some embodiments, such RNAs (e.g., mrnas) may be particularly useful and/or effective for use as or in immunogenic compositions (e.g., vaccines), and/or for achieving an immune effect as described herein (e.g., production of SARS-CoV-2 neutralizing antibodies, and/or T cell responses (e.g., CD4+ and/or CD8+ T cell responses)). In some embodiments, such RNA (e.g., mRNA) can be used to vaccinate a human (including, for example, a human known to be exposed and/or infected by SARS-CoV-2, and/or a human not known to be exposed to SARS-CoV-2).

One of skill in the art reading this disclosure will further understand that it describes various mRNA constructs comprising a nucleic acid sequence encoding a full-length SARS-CoV-2 spike protein (e.g., including embodiments in which such encoded SARS-CoV-2 spike protein can comprise at least one or more amino acid substitutions, e.g., a proline substitution as described herein, and/or embodiments in which the mRNA sequence is codon optimized for a subject (e.g., a mammal, e.g., a human)). In some embodiments, such a full-length SARS-CoV-2 spike protein can have an amino acid sequence that is or comprises the amino acid sequence set forth in SEQ ID NO. 7. Still further, one of ordinary skill in the art reading this disclosure would understand that it describes certain features and/or advantages of certain mRNA constructs comprising a nucleic acid sequence encoding the full-length SARS-CoV-2 spike protein. Without wishing to be bound by any particular theory, the present disclosure demonstrates that the mRNA constructs provided that encode the full-length SARS-CoV-2S protein can be particularly useful and/or effective for use as or in immunogenic compositions (e.g., vaccines) in a particular population of subjects (e.g., a population of a particular age). For example, in some embodiments, such mRNA compositions may be particularly useful in young (e.g., under 25 years old, under 20 years old, under 18 years old, under 15 years old, 10 years old, or lower) subjects; alternatively or additionally, in some embodiments, such mRNA compositions may be particularly useful in elderly subjects (e.g., over 55 years old, over 60 years old, over 65 years old, over 70 years old, over 75 years old, over 80 years old, over 85 years old, or higher). In particular embodiments, immunogenic compositions comprising such mRNA constructs provided herein exhibit minimal to moderate increases (e.g., no more than 30% increase, no more than 20% increase, or no more than 10% increase or less) in dose level and/or dose number dependent systemic reactogenicity (e.g., fever, fatigue, headache, chills, diarrhea, muscle pain, and/or joint pain, etc.) and/or local tolerance (e.g., pain, redness and/or swelling, etc.), at least in some subjects (e.g., in some subject age groups); in some embodiments, such reactogenicity and/or local tolerance is observed particularly in subjects of a younger age group (e.g., under 25 years, under 20 years, 18 years or less) and/or in an older (e.g., elderly) age group (e.g., 65-85 years). In some embodiments, the mRNA constructs provided that encode full-length SARS-CoV-2S proteins can be particularly useful and/or effective for use as or in an immunogenic composition (e.g., a vaccine) to induce levels of SARS-CoV-2 neutralizing antibody responses in a population of subjects (e.g., an elderly population, e.g., a 65-85 year old group) that has a high risk of severe disease associated with SARS-CoV-2 infection. In some embodiments, one of ordinary skill in the art reading this disclosure will appreciate that mRNA constructs encoding full-length SARS-CoV-2S proteins provided that exhibit favorable responsiveness profiles in young and old populations (e.g., as described herein) may be particularly useful and/or effective for use as or in immunogenic compositions (e.g., vaccines) to achieve the immune effects as described herein (e.g., production of SARS-CoV-2 neutralizing antibodies, and/or T cell responses (e.g., CD4+ and/or CD8+ T cell responses)). In some embodiments, the disclosure also indicates that the mRNA constructs provided encoding the full-length SARS-CoV-2S protein can be particularly effective for providing protection against SARS-CoV-2 infection, characterized by early clearance of SARS-CoV-2 viral RNA in a non-human mammalian subject (e.g., rhesus monkey) who is immunized with an immunogenic composition comprising such mRNA constructs and then challenged with a SARS-CoV-2 strain. In some embodiments, early clearance of such SARS-CoV-2 viral RNA can be observed in the nose of a non-human mammalian subject (e.g., rhesus monkey) immunized with an immunogenic composition comprising such mRNA construct and then challenged with a SARS-CoV-2 strain.

In some embodiments, the disclosure provides an RNA (e.g., mRNA) comprising an open reading frame encoding a full-length SARS-CoV-2S protein (e.g., a full-length SARS-CoV-2S protein having one or more amino acid substitutions), the RNA suitable for intracellular expression of the polypeptide. In some embodiments, the encoded polypeptide comprises SEQ ID NO: 7. In some embodiments, such RNA (e.g., mRNA) can be complexed by (poly) cationic polymers, polyplex, proteins, or peptides. In some embodiments, such RNA can be formulated in a lipid nanoparticle (e.g., a lipid nanoparticle described herein).

In some embodiments, the immunogenic compositions provided herein can comprise a plurality (e.g., at least 2 or more, including, e.g., at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, etc.) immunogenic epitopes of a SARS-CoV-2 polypeptide or variant thereof. In some such embodiments, such multiple immunogenic epitopes can be encoded by multiple RNAs (e.g., mrnas). In some such embodiments, such multiple immunogenic epitopes can be encoded by a single RNA (e.g., mRNA). In some embodiments, nucleic acid sequences encoding multiple immunogenic epitopes can be separated from each other in a single RNA (e.g., mRNA) by a linker (e.g., a peptide linker in some embodiments). Without wishing to be bound by any particular theory, in some embodiments, when considering the genetic diversity of SARS-CoV-2 variants, the provided polyepitope immunogenic compositions (including, e.g., those encoding full-length SARS-CoV-2 spike protein) may be particularly useful for providing protection against multiple viral variants and/or may provide greater opportunity to develop diverse and/or robust (e.g., durable, e.g., detectable about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60 or more days after administration of one or more doses) neutralizing antibodies and/or T cell responses, particularly robust T cell responses HType 1T cells (e.g., CD4+ and/or CD8+ T cells) respond.

In some embodiments, the disclosure records that compositions and/or methods provided are characterized (e.g., when administered to a relevant population, which in some embodiments may be or comprise an adult population) in that they achieve one or more specific therapeutic outcomes with a single administration (e.g., an effective immune response and/or detectable expression of an encoded SARS-CoV-2S protein or immunogenic fragment thereof as described herein); in some such embodiments, for example, results can be evaluated in comparison to results observed in the absence of the mRNA vaccine described herein. In some embodiments, a particular result may be achieved at a lower dose than is required by one or more alternative strategies.

In some embodiments, the present disclosure provides an immunogenic composition comprising an isolated messenger ribonucleic acid (mRNA) polynucleotide, wherein the isolated mRNA polynucleotide comprises an open reading frame encoding a polypeptide comprising a receptor binding portion of SARs-CoV-2S protein, and wherein the isolated mRNA polynucleotide is formulated in at least one lipid nanoparticle. For example, in some embodiments, such lipid nanoparticles may comprise a molar ratio of 20-60% ionizable cationic lipid, 5-25% noncationic lipid (e.g., neutral lipid), 25-55% sterol or steroid, and 0.5-15% polymer-conjugated lipid (e.g., PEG-modified lipid). In some embodiments, the sterol or steroid comprised in the lipid nanoparticle may be or comprise cholesterol. In some embodiments, the neutral lipid may be or comprise 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC). In some embodiments, the polymer-conjugated lipid may be or comprise PEG2000 DMG. In some embodiments, such immunogenic compositions may comprise a total lipid content of about 1mg to 10mg, or 3mg to 8mg, or 4mg to 6 mg. In some embodiments, such immunogenic compositions may comprise a total lipid content of about 5mg/mL to 15mg/mL or 7.5mg/mL to 12.5 mg/mL or 9 to 11 mg/mL. In some embodiments, such isolated mRNA polynucleotides are provided in an effective amount to induce an immune response in a subject administered at least one dose of an immunogenic composition. In some embodiments, provided isolated mRNA polynucleotides encode polypeptides that do not comprise an intact S protein. In some embodiments, such isolated mRNA polynucleotides provided in the immunogenic composition are not self-replicating RNA.

In some embodiments, the immune response may include the generation of a binding antibody titer against the SARS-CoV-2 protein (including, for example, a stable prefusion spike trimer in some embodiments) or a fragment thereof. In some embodiments, the immune response may comprise generating a binding antibody titer against the Receptor Binding Domain (RBD) of the SARS-CoV-2 spike protein. In some embodiments, the provided immunogenic compositions have been established to achieve a detectable binding antibody titer following administration of the first dose with seroconversion, e.g., to about 2 weeks, in at least 70% (including, e.g., at least 80%, at least 90%, at least 95%, and up to 100%) of a population of subjects receiving such provided immunogenic compositions.

In some embodiments, the immune response may include the generation of neutralizing antibody titers against SARS-CoV-2 protein (including, for example, stable pre-fusion spike trimers in some embodiments) or fragments thereof. In some embodiments, the immune response may include the generation of neutralizing antibody titers against the Receptor Binding Domain (RBD) of the SARS-CoV-2 spike protein. In some embodiments, the immunogenic compositions provided have been established to achieve neutralizing antibody titers in appropriate systems (e.g., in a human infected with SARS-CoV-2 and/or a population thereof, and/or in a model system thereof). For example, in some embodiments, such neutralizing antibody titers may have been demonstrated in one or more human populations, non-human primate models (e.g., rhesus monkeys), and/or mouse models.

In some embodiments, the neutralizing antibody titer is (e.g., has been established as) a titer sufficient to reduce viral infection of B cells observed relative to an appropriate control (e.g., an unvaccinated control subject, or a subject vaccinated with a live attenuated virus vaccine, an inactivated virus vaccine, or a protein subunit (subbunit) virus vaccine, or a combination thereof). In some such embodiments, such a reduction is at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more.

In some embodiments, the neutralizing antibody titer is (e.g., has been established as) a titer sufficient to reduce the rate of asymptomatic viral infection observed relative to an appropriate control (e.g., an unvaccinated control subject, or a subject vaccinated with a live attenuated virus vaccine, an inactivated virus vaccine, or a protein subunit virus vaccine, or a combination thereof). In some such embodiments, such a reduction is at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more. In some embodiments, such a reduction can be characterized by assessing SARS-CoV-2N protein serology. Significant protection against asymptomatic infection was also demonstrated by real life observations (see also Dagan N. et al, N Engl J med.2021, doi: 10.1056/nejmoa2101765.epub ahead of print. pmid: 33626250).

In some embodiments, the neutralizing antibody titer is (e.g., has been established as) a titer sufficient to reduce or block fusion of the virus with epithelial cells and/or B cells of the vaccinated subject relative to that observed for an appropriate control (e.g., an unvaccinated control subject, or a subject vaccinated with a live attenuated virus vaccine, an inactivated virus vaccine, or a protein subunit virus vaccine, or a combination thereof). In some such embodiments, such a reduction is at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more.

In some embodiments, induction of neutralizing antibody titers may be characterized by an increase in B cell numbers, which in some embodiments may include plasma cells, class-switched IgG 1-and IgG 2-positive B cells, and/or germinal center B cells. In some embodiments, the immunogenic compositions provided have been established to achieve such an increase in the number of B cells in an appropriate system (e.g., in a human infected with SARS-CoV-2 and/or a population thereof, and/or in a model system thereof). For example, in some embodiments, such an increase in B cell number may have been demonstrated in one or more human populations, non-human primate models (e.g., rhesus monkeys), and/or mouse models. In some embodiments, such an increase in the number of B cells may have been demonstrated in draining lymph nodes and/or spleen of a mouse model after immunization of such a mouse model with a provided immunogenic composition (e.g., after at least 7 days, at least 8 days, at least 9 days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least 14 days). In some embodiments, inducing neutralizing antibody titers may be characterized by a reduction in the number of circulating B cells in the blood. In some embodiments, the immunogenic compositions provided have been established to achieve such a reduction in the number of circulating B cells in the blood of an appropriate system (e.g., in a human infected with SARS-CoV-2 and/or a population thereof, and/or in a model system thereof). For example, in some embodiments, such a reduction in the number of circulating B cells in the blood may have been demonstrated in one or more human populations, non-human primate models (e.g., rhesus monkeys), and/or mouse models. In some embodiments, such a reduction in the number of circulating B cells in the blood may have been demonstrated in a mouse model after immunizing such a mouse model with a provided immunogenic composition (e.g., after at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 8 days, at least 9 days, at least 10 days). Without wishing to be bound by theory, the reduction in circulating B cells in the blood may be due to B cells homing to the lymphatic compartment.

In some embodiments, the immune response induced by the provided immunogenic compositions can include an increase in T cell numbers. In some embodiments, such an increase in T cell number may include follicular helper T (T)FH) The number of cells is increased, which in some embodiments may include one or more subsets with ICOS upregulation. Those skilled in the art will understand TFHProliferation in germinal centers is essential for the generation of adaptive B cell responses, while and in humans, T appears in the circulation after vaccinationFHAre often associated with a high frequency of antigen-specific antibodies. In some embodiments, the immunogenic compositions provided have been established to achieve such T cells (e.g., T cells) in an appropriate system (e.g., in a human infected with SARS-CoV-2 and/or a population thereof, and/or in a model system thereof)FHCells) increase in number. For example, in some embodiments, such a T cell (e.g., T)FHCells) may already be in oneOr in multiple human populations, non-human primate models (e.g., rhesus monkey), and/or mouse models. In some embodiments, such T cells (e.g., T cells) may have been confirmed in draining lymph nodes, spleen, and/or blood of a mouse model (e.g., after at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 8 days, at least 9 days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least 14 days) after immunization of such a mouse model with a provided immunogenic composition FHCells) increase in number.

In some embodiments, the protective response against SARS-CoV-2 induced by the provided immunogenic composition has been established in an appropriate model system for SARS-CoV-2. For example, in some embodiments, such a protective response may have been demonstrated in an animal model, such as a non-human primate model (e.g., rhesus monkey) and/or a mouse model. In some embodiments, a non-human primate (e.g., rhesus monkey) or population thereof that has received at least one immunization with the provided immunogenic composition is challenged with SARS-CoV-2, e.g., by intranasal and/or intratracheal routes. In some embodiments, such challenge may be performed weeks (e.g., 5-10 weeks) after at least one immunization with the provided immunogenic composition (including, e.g., at least two immunizations). In some embodiments, such a challenge can be performed when a detectable level of SARS-CoV-2 neutralizing titer (e.g., an antibody response to the SARS-CoV-2 spike protein and/or fragment thereof, including, for example, but not limited to, stable pre-fusion spike trimer, S-2P, and/or an antibody response to the receptor-binding portion of SARS-CoV-2) is achieved in a non-human primate (e.g., rhesus monkey) that has been immunized (including, for example, at least twice) with the provided immunogenic composition. In some embodiments, the protective response is characterized by the absence or detectable reduction of viral RNA in bronchoalveolar lavage (BAL) and/or nasal swabs of a challenged non-human primate (e.g., rhesus monkey). In some embodiments, the immunogenic compositions described herein can be characterized in that a greater percentage of challenged animals (e.g., non-human primates (e.g., rhesus monkeys) in a population) that have received at least one immunization (including, e.g., at least two immunizations) with the provided immunogenic composition, as compared to a non-immunized animal population (e.g., non-human primates (e.g., rhesus monkeys)) show an absence of detectable RNA in their BAL and/or nasal swab. In some embodiments, the immunogenic compositions described herein can be characterized by an animal (e.g., a non-human (e.g., rhesus monkey) in a population) that has received a challenge immunized at least once (including, e.g., at least twice) with a provided immunogenic composition, as compared to a non-immunized animal population (e.g., a non-human primate (e.g., rhesus monkey)), can exhibit clearance of viral RNA in a nasal swab no later than 10 days, including, e.g., no later than 8 days, no later than 6 days, no later than 4 days, etc.

In some embodiments, the immunogenic compositions described herein do not substantially increase the risk of vaccine-related enhanced respiratory disease when administered to a subject in need thereof. In some embodiments, such vaccine-associated enhanced respiratory diseases may be associated with antibody-dependent enhanced replication and/or with vaccine antigens that induce poorly neutralizing antibodies and Th2 biased responses. In some embodiments, the immunogenic compositions described herein do not substantially increase the risk of antibody-dependent replication enhancement when administered to a subject in need thereof.

In some embodiments, a single dose of an mRNA composition (e.g., formulated in a lipid nanoparticle) can induce a therapeutic antibody response in a vaccination of less than 10 days. In some embodiments, such a therapeutic antibody response may be characterized in that such an mRNA vaccine may induce the production of about 10-100 μ g/mL IgG in an animal model when measured 10 days after vaccination at a dose of 0.1-10 μ g or 0.2-5 μ g. In some embodiments, such therapeutic antibody responses may be characterized by the induction of about 100-1000. mu.g/mL IgG of such mRNA vaccines as measured in an animal model at 20 days of vaccination at a dose of 0.1-10. mu.g or 0.2-5. mu.g. In some embodiments, a single dose can induce pseudovirus neutralization titers of 10-200pVN50 drops 15 days after vaccination, as measured in animal models. In some embodiments, a single dose can induce a pseudovirus neutralization titer of 50-500pVN50 titers 15 days after vaccination, as measured in an animal model.

In some embodiments, a single dose of an mRNA composition can amplify an antigen-specific CD8 and/or CD 4T cell response by at least 50% or more (including, e.g., at least 60%, at least 70%, at least 80%, at least 90%, at least 95% or more) compared to that observed in the absence of such an mRNA construct encoding a SARS-COV-2 immunogenic protein or fragment thereof (e.g., spike protein and/or receptor binding domain). In some embodiments, a single dose of the mRNA composition can amplify the antigen-specific CD8 and/or CD 4T cell response by at least 1.5-fold or more (including, e.g., at least 2-fold, at least 3-fold, at least 5-fold, at least 10-fold, at least 50-fold, at least 100-fold, at least 500-fold, at least 1000-fold or more) as compared to that observed in the absence of such an mRNA construct encoding a SARS-COV-2 immunogenic protein or fragment thereof (e.g., spike protein and/or receptor binding domain).

In some embodiments, a regimen (e.g., a single dose of an mRNA composition) can expand T cells exhibiting a Th1 phenotype (e.g., characterized by expression of IFN- γ, IL-2, IL-4, and/or IL-5) by at least 50% or more (including, e.g., at least 60%, at least 70%, at least 80%, at least 90%, at least 95% or more) as compared to that observed in the absence of such an mRNA construct encoding a SARS-COV-2 immunogenic protein or fragment thereof (e.g., spike protein and/or receptor binding domain). In some embodiments, a regimen (e.g., a single dose of an mRNA composition) can expand T cells exhibiting a Th1 phenotype (e.g., characterized by expression of IFN- γ, IL-2, IL-4, and/or IL-5) by, for example, at least 1.5-fold or more (including, e.g., at least 2-fold, at least 3-fold, at least 5-fold, at least 10-fold, at least 50-fold, at least 100-fold, at least 500-fold, at least 1000-fold or more) as compared to that observed in the absence of such an mRNA construct encoding a SARS-COV-2 immunogenic protein or fragment thereof (e.g., spike protein and/or receptor binding domain). In some embodiments, the T cell phenotype may be or include a cytokine profile dominated by Th1 (e.g., characterized by IFN- γ positive and/or IL-2 positive), and/or no or biologically insignificant secretion of IL-4.

In some embodiments, a regimen as described herein (e.g., one or more doses of an mRNA composition) induces and/or effects the production of RBD-specific CD4+ T cells. Among other things, the present disclosure notes that mRNA compositions encoding a portion of a SARS-CoV-2 spike protein that comprises an RBD (e.g., does not encode a full-length SARS-CoV-2 spike protein) can be particularly useful and/or effective in such inducing and/or generating RBD-specific CD4+ T cells. In some embodiments, RBD-specific CD4+ T cells induced by an mRNA composition described herein (e.g., by an mRNA composition encoding a portion of SARS-CoV-2 spike protein that comprises an RBD, and in some embodiments does not encode a full-length SARS-CoV-2 spike protein) demonstrate a Th1 predominant cytokine profile (e.g., characterized by IFN- γ positive and/or IL-2 positive), and/or no or biologically insignificant IL-4 secretion.

In some embodiments, characterization of CD4+ and/or CD8+ T cell responses (e.g., as described herein) in a subject receiving an mRNA composition (e.g., as described herein) can be performed using ex vivo assays using PBMCs collected from the subject, e.g., the assays described in the examples.

In some embodiments, the immunogenicity of an mRNA composition described herein can be assessed by one or more of the following serological immunogenicity assays: detecting IgG, IgM and/or IgA present in a blood sample of a subject receiving the provided mRNA composition against SARS-CoV-2S protein, and/or a neutralization assay using SARS-CoV-2 pseudovirus and/or wild-type SARS-CoV-2 virus.

In some embodiments, the mRNA composition (e.g., as described herein) provides relatively low adverse effects (e.g., grade 1-grade 2 pain, redness and/or swelling) within 7 days after vaccination at a dose of 10 μ g-100 μ g or 1 μ g-50 μ g. In some embodiments, the mRNA composition (e.g., as described herein) provides relatively low observed values for systemic events (e.g., grade 1-grade 2 fever, fatigue, headache, chills, vomiting, diarrhea, muscle pain, joint pain, medication (therapy), and combinations thereof) within 7 days after vaccination at a dose of 10 μ g-100 μ g.

In some embodiments, the mRNA composition is characterized by producing IgG against the SARS-CoV-2 immunogenic protein or fragment thereof (e.g., spike protein and/or receptor binding domain) at a level of 100-100,000U/mL or 500-50,000U/mL 21 days after vaccination when administered to a subject at a dose of 10-100 μ g or 1 μ g-50 μ g.

In some embodiments, the mRNA encodes a naturally folded trimeric receptor binding protein of SARS-CoV-2. In some embodiments, the mRNA encodes a variant of such a receptor binding protein, such that the encoded variant binds to ACE2 with a Kd of 10 pM or less, including, for example, with a Kd of 9pM, 8pM, 7pM, 6pM, 5pM, 4 pM or less. In some embodiments, the mRNA encodes a variant of such a receptor binding protein, whereby the encoded variant binds to ACE2 with a Kd of 5 pM. In some embodiments, the mRNA encodes a trimeric receptor binding portion of SARS-CoV-2 comprising an ACE2 receptor binding site. In some embodiments, the mRNA comprises a coding sequence for a receptor binding portion and a trimerization domain of SARS-CoV-2 (e.g., the native trimerization domain of T4 minor fibrin (foldon)), such that the coding sequence directs the expression of a trimeric protein having an ACE2 receptor binding site and binding to ACE 2. in some embodiments, the mRNA encodes a trimeric receptor binding portion of SARS-CoV-2 or a variant thereof such that its Kd is less than Kd. of the monomeric Receptor Binding Domain (RBD) of SARS-CoV-2. for example, in some embodiments, the mRNA encodes a trimeric receptor binding portion of SARS-CoV-2 or a variant thereof such that its Kd is at least 10 times (including, e.g., at least 50 times, at least 100 times, at least 500 times) less than the Kd of the RBD of SARS-CoV-2, At least 1000-fold, etc.).

In some embodiments, when combined with ACE2 and B, as characterized by cryo-electron microscopy (cryoEM)0When the AT1 neutral amino acid transporter is complexed in a closed conformation, the trimeric receptor binding portion of SARS-CoV-2 encoded by the mRNA (e.g., as described herein) can be determined to be about 3-4 angstroms in size. In some embodiments, the geometric mean SARS-CoV-2 neutralization titer characterized and/or achieved by an mRNA composition or method as described herein can be at least 1.5-fold, including at least 2-fold, at least 2.5-fold, at least 3-fold or more, of a COVID-19 convalescent human cohort (e.g., a serogroup from a COVID-19 convalescent human obtained 20-40 days after symptom onset and at least 14 days after onset of asymptomatic rehabilitation).

In some embodiments, mRNA compositions as provided herein can be characterized by subjects that have been treated with such compositions (e.g., with at least one dose, at least two doses, etc.) can exhibit a reduction and/or short temporal persistence of viral RNA at the relevant site (e.g., the nose and/or lung, etc., and/or any other susceptible tissue) as compared to an appropriate control (e.g., a given expected level of a comparable subject or population that has not been so treated and has been exposed to the virus under reasonably comparable exposure conditions).

In some embodiments, the RBD antigen expressed by an mRNA construct (e.g., as described herein) may be modified, e.g., to increase its immunogenicity, by the addition of a T4 minor fibrin-derived "folder" trimerization domain.

In some embodiments, mRNA compositions and/or methods described herein are characterized by certain local reactions (e.g., pain, redness and/or swelling, etc.) and/or systemic events (e.g., fever, fatigue, headache, etc.) that may appear and/or peak at day 2 after vaccination. In some embodiments, the mRNA compositions described herein are characterized by that certain local reactions (e.g., pain, redness and/or swelling, etc.) and/or systemic events (e.g., fever, fatigue, headache, etc.) can resolve by day 7 after vaccination.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by no observed grade 1 or greater change in conventional clinical laboratory values or laboratory abnormalities in a subject receiving the mRNA composition (as described herein). Examples of such clinical laboratory assays may include lymphocyte counts, hematological changes, and the like.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by a Geometric Mean Concentration (GMC) of IgG to a SARS-CoV-2S polypeptide or immunogenic fragment thereof (e.g., RBD) of 200 units/mL or 500 units/mL 2000 units/mL at 21 days after a first dose (e.g., 10-100 μ g, including 10 and 100 μ g or 1 μ g-50 μ g, including 1 μ g and 50 μ g) as compared to 602 units/mL for a panel of COVID-19 convalescent human serum. In some embodiments, the mRNA compositions described herein are characterized in that the Geometric Mean Concentration (GMC) of IgG against the SARS-CoV-2 spike polypeptide or immunogenic fragment thereof (e.g., RBD) can be increased by at least 8-fold or more, including, e.g., at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold or more, 7 days after the second dose (e.g., 10-30 μ g, including 10 and 30 μ g; or 1 μ g-50 μ g, including 1 μ g and 50 μ g). In some embodiments, the mRNA compositions described herein are characterized by a Geometric Mean Concentration (GMC) of IgG to a SARS-CoV-2S polypeptide or immunogenic fragment thereof (e.g., RBD) that can increase to 1500 units/mL to 40,000 units/mL or 4000 units/mL to 40,000 units/mL 7 days after the second dose (e.g., 10-30 μ g, including 10 and 30 μ g; or 1 μ g to 50 μ g, including 1 μ g and 50 μ g). In some embodiments, the antibody concentrations described herein can last at least 20 days or more, including, e.g., at least 25 days, at least 30 days, at least 35 days, at least 40 days, at least 45 days, at least 50 days, after the first dose, or at least 10 days or more, including, e.g., at least 15 days, at least 20 days, at least 25 days, or more, after the second dose. In some embodiments, the antibody concentration may last 35 days after the first dose, or at least 14 days after the second dose.

In some embodiments, the mRNA compositions described herein are characterized by a GMC that is at least 30% higher (including, e.g., at least 40% higher, at least 50% higher, at least 60% higher, at least 70% higher, at least 80% higher, at least 90% higher, at least 95% higher) for IgG that is directed against a SARS-CoV-2S polypeptide or immunogenic fragment thereof (e.g., RBD) when measured 7 days after a second dose (e.g., 1-50 μ g, including 1 and 50 μ g) as compared to the concentration of antibodies observed in serum of a panel of covi-19 recovery stage humans. In many embodiments, the Geometric Mean Concentration (GMC) of IgG described herein is the GMC of IgG that binds RBD.

In some embodiments, the mRNA compositions described herein are characterized by a GMC that is at least 1.1 fold (including, e.g., at least 1.5 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 15 fold, at least 20 fold, at least 25 fold, at least 30 fold) IgG to a SARS-CoV-2S polypeptide or immunogenic fragment thereof (e.g., RBD) when measured 7 days after a second dose (e.g., 10-50 μ g, including 1 and 50 μ g) as compared to the concentration of antibodies observed in serum of a panel of cove-19 convalescence humans. In many embodiments, the Geometric Mean Concentration (GMC) of IgG described herein is the GMC of IgG that binds RBD.

In some embodiments, the mRNA compositions described herein are characterized by a GMC of IgG directed to a SARS-CoV-2S polypeptide or immunogenic fragment thereof (e.g., RBD) that is at least 5 fold (including, e.g., at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 15 fold, at least 20 fold, at least 25 fold, at least 30 fold) when measured 21 days after the second dose as compared to the concentration of antibodies observed in serum of a panel of covi-19 convalescent human subjects. In many embodiments, the Geometric Mean Concentration (GMC) of IgG described herein is the GMC of IgG that binds RBD.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by an increase (e.g., at least 30%, at least 40%, at least 50% or more) in SARS-CoV-2 neutralization Geometric Mean Titer (GMT) observed 21 days after the first dose. In some embodiments, the mRNA compositions described herein are characterized by achieving significantly higher serum neutralization GMT up to 150-.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by a protective efficacy (efficacy) of at least 60%, e.g., at least 70%, at least 80%, at least 90%, or at least 95%, 7 days after administration of the second dose. In one embodiment, the mRNA compositions and/or methods described herein are characterized by a protective efficacy of at least 70% 7 days after administration of the second dose. In one embodiment, the mRNA compositions and/or methods described herein are characterized by a protective efficiency of at least 80% 7 days after administration of the second dose. In one embodiment, the mRNA compositions and/or methods described herein are characterized by a protective efficiency of at least 90% 7 days after administration of the second dose. In one embodiment, the mRNA compositions and/or methods described herein are characterized by a protective efficacy of at least 95% 7 days after administration of the second dose.

In some embodiments, the RNA composition provided herein is characterized in that it induces an immune response against SARS-CoV-2 after at least 7 days after dosing (e.g., after the second dose). In some embodiments, the RNA composition provided herein is characterized in that it induces an immune response against SARS-CoV-2 less than 14 days after dosing (e.g., after the second dose). In some embodiments, the RNA composition provided herein is characterized in that it induces an immune response against SARS-CoV-2 after at least 7 days following the vaccination regimen. In some embodiments, the vaccination regimen comprises a first dose and a second dose. In some embodiments, the first dose and the second dose are administered at least 21 days apart. In some such embodiments, an immune response against SARS-CoV-2 is induced at least after 28 days after the first dose.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by a Geometric Mean Concentration (GMC) of antibodies directed against SARS-CoV-2 spike polypeptide or immunogenic fragment thereof (e.g., RBD) that is significantly higher than in the convalescent serogroup (e.g., as described herein), as measured in serum from a subject receiving the mRNA compositions of the present disclosure (e.g., at a dose of 10-30 μ g, including 10 and 30 μ g). In some embodiments where the subject may receive a second dose (e.g., 21 days after 1 first dose), the Geometric Mean Concentration (GMC) of antibodies directed against the SARS-CoV-2 spike polypeptide or immunogenic fragment thereof (e.g., RBD), as measured in serum from the subject, may be 8.0-fold to 50-fold that of the convalescent serogroup GMC. In some embodiments where the subject can receive a second dose (e.g., 21 days after 1 first dose), the Geometric Mean Concentration (GMC) of antibodies directed against SARS-CoV-2 spike polypeptide or immunogenic fragment thereof (e.g., RBD), as measured in serum from the subject, can be at least 8.0-fold or more, including, e.g., at least 10-fold, at least 20-fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold or more, as compared to convalescent serogroup GMC.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by a SARS-CoV-2 neutralization geometric mean titer that can be at least 1.5-fold or greater (including, e.g., at least 2-fold, at least 2.5-fold, at least 3-fold, at least 3.5-fold or greater) compared to the neutralizing GMT of the convalescent serogroup, as measured 28 days after the first dose or 7 days after the second dose.

In some embodiments, the regimen administered to the subject may be or comprise a single dose. In some embodiments, the regimen administered to the subject can comprise a plurality of doses (e.g., at least two doses, at least three doses, or more). In some embodiments, the regimen administered to the subject may comprise a first dose and a second dose, which are administered at least 2 weeks apart, at least 3 weeks apart, at least 4 weeks apart, or longer. In some embodiments, such dosages may be separated by at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, at least 12 months, or longer. In some embodiments, the dose may be administered several days apart, such as 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 days or more apart. In some embodiments, the doses may be administered at about 1 to about 3 weeks apart, or at about 1 to about 4 weeks apart, or at about 1 to about 5 weeks apart, or at about 1 to about 6 weeks apart, or at about 1 to more than 6 weeks apart. In some embodiments, the doses may be separated by a period of about 7 to about 60 days, such as about 14 to about 48 days, and the like. In some embodiments, the minimum number of days between doses can be about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more. In some embodiments, the maximum number of days between doses can be about 60, 59, 58, 57, 56, 55, 54, 53, 52, 51, 50, 49, 48, 47, 46, 45, 44, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21 or less. In some embodiments, the doses may be separated by about 21 to about 28 days. In some embodiments, the doses may be separated by about 19 to about 42 days. In some embodiments, the doses may be separated by about 7 to about 28 days. In some embodiments, the dosage interval may be from about 14 to about 24 days. In some embodiments, the dosage interval may be from about 21 to about 42 days.

In some embodiments, particularly for compositions established to achieve elevated antibody and/or T cell titers over a period of time longer than about 3 weeks-e.g., in some embodiments, the compositions provided are established to achieve elevated antibody and/or T cell titers over a period of time longer than about 3 weeks (e.g., specific for the relevant portion of SARS-CoV-2 spike protein), in some such embodiments, the dosing regimen may involve only a single dose, or may involve two or more doses, which may be separated from each other over a period of time longer than about 21 days or 3 weeks. For example, in some such embodiments, such a period of time may be about 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks or longer, or about 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months or longer, or in some embodiments about one year or longer.

In some embodiments, the first dose and the second dose (and/or other subsequent doses) may be administered by intramuscular injection. In some embodiments, the first dose and the second dose can be administered in the deltoid muscle. In some embodiments, the first dose and the second dose can be administered in the same arm. In some embodiments, the mRNA compositions described herein are administered (e.g., by intramuscular injection) as a series of two doses (e.g., 0.3mL each) 21 days apart. In some embodiments, each dose is about 30 μ g. In some embodiments, each dose can be greater than 30 μ g, e.g., about 40 μ g, about 50 μ g, about 60 μ g. In some embodiments, each dose can be less than 30 μ g, e.g., about 20 μ g, about 10 μ g, about 5 μ g, etc. In some embodiments, each dose is about 3 μ g or less, e.g., about 1 μ g. In some such embodiments, the mRNA compositions described herein are administered to a subject that is 16 years or older (including, e.g., 16-85 years old). In some such embodiments, the mRNA compositions described herein are administered to a subject between 18 and 55 years of age. In some such embodiments, the mRNA compositions described herein are administered to a subject between 56 and 85 years of age. In some embodiments, the mRNA compositions described herein are administered as a single dose (e.g., by intramuscular injection).

In some embodiments, the mRNA compositions and/or methods described herein are characterized by having an mRNA composition and/or method that induces RBD-specific IgG (e.g., polyclonal response) that exhibits a higher binding affinity for a SARS-CoV-2 RBD as compared to a reference human monoclonal antibody (e.g., CR3022 as described in j.ter Meulen et al, PLOS med.3, e237 (2006)).

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity in the entire (across) group of SARs-CoV-2 spike variants (e.g., at least 10, at least 15, or more). In some embodiments, such SARs-CoV-2 spike variants include mutations in RBDs (e.g., without limitation, Q321L, V341I, a348T, N354D, S359N, V367F, K378R, R408I, Q409E, a435S, N439K, K458R, I472V, G476S, S477N, V483A, Y508H, H519P, etc., as compared to SEQ ID NO: 1) and/or mutations in spike proteins (e.g., without limitation, D614G, etc., as compared to SEQ ID NO: 1). Those skilled in the art will know the various spike variants and/or record their resources (e.g., a table of mutation sites in spikes maintained by the covi-19 viral genome analysis channel and found at https:// COV. lanl. gov/compositions/sequence/COV/int _ sites _ tbls. comp) (last visit 2020, 8/24 days), and will understand from this disclosure that the mRNA compositions and/or methods described herein may be characterized by their ability to induce sera in vaccinated subjects that exhibit neutralizing activity with respect to any or all such variants and/or combinations thereof.

In particular embodiments, the mRNA composition of the RBD encoding SARS-CoV-2 spike protein is characterized in that sera of vaccinated subjects exhibit neutralizing activity in the entire group of (e.g., at least 10, at least 15, or more) SARS-CoV-2 spike variants, including RBD variants (e.g., without limitation, Q321L, V341I, a348T, N354D, S359N, V367F, K378R, R408I, Q409E, a435S, N39439 38, K458R, I472V, G476S, S477N, V483A, Y508H, H519P, etc., as compared to SEQ ID NO: 1) and spike protein variants (e.g., without limitation, D G, as compared to SEQ ID NO: 1).

In particular embodiments, mRNA compositions encoding SARS-CoV-2 spike protein variants comprising two consecutive proline substitutions at amino acid positions 986 and 987 at the top of the central helix of the S2 subunit are characterized in that sera of vaccinated subjects exhibit neutralizing activity in the entire group of SARS-CoV-2 spike mutants (e.g., at least 10, at least 15, or more) including RBD variants (e.g., without limitation, Q321L, V341I, a348T, N354D, S359N, V367F, K378R, R408 39435, Q E, a S, N K, K458R, I V, G476S, S477 737 8, V483 6866, Y35508, 519H P, etc., as compared to SEQ ID NO: 1) and spike protein variants (e.g., without limitation, as compared to SEQ ID No. G: 1). For example, in some embodiments, mRNA compositions encoding SEQ ID NO:7(S P2) elicit immune responses against any one of the SARs-CoV-2 spike variants, including RBD variants (e.g., without limitation, Q321L, V341I, a348T, N354D, S359N, V367F, K378R, R408I, Q409E, a435S, N439K, K458R, I458 472V, G476S, S477N, V483A, Y508H, H519P, etc., as compared to SEQ ID NO: 1) and spike protein variants (e.g., without limitation, D614G as compared to SEQ ID NO: 1).

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants, as compared to the mRNA of SEQ ID NO:1 in contrast, the SARs-CoV-2 spike variant includes a mutation at position 501 in the spike protein. In some embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against one or more SARs-CoV-2 spike variants that are identical to the mRNA of SEQ ID NO:1 in contrast, the SARs-CoV-2 spike variant included an N501Y mutation in the spike protein.

And SEQ ID NO:1, compared to SEQ ID NO: one or more SARs-CoV-2 spike variants comprising a mutation at position 501 in the spike protein compared to SEQ ID NO:1 or one or more SARs-CoV-2 spike variants comprising the N501Y mutation in the spike protein compared to SEQ ID NO:1 may comprise one or more further mutations (e.g., without limitation, H69/V70 deletion, Y144 deletion, a570D, D614G, P681H, T716I, S982A, D1118H, D68180 80A, D215G, E484K, a701V, L18F, R246I, K417N, L242/a243/L244 deletion, etc., compared to SEQ ID NO: 1).

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against the SARs-CoV-2 spike variant "variant of interest 202012/01" (VOC-202012/01; also referred to as lineage B.1.1.7). This variant has previously been named the first variant under investigation (VUI-202012/01) by the United kingdom Public Health England (Public Health England) at 12 months 2020, but was reclassified as the variant of interest (VOC-202012/01). VOC-202012/01 is a variant of SARS-CoV-2 which is first detected in samples collected from the previous month during the United kingdom COVID-19 pandemic at 10 months of 2020 and which begins spreading rapidly in mid 12 months. It is associated with a significant increase in uk covi-19 infection rate; this increase is believed to be due, at least in part, to a change in N501Y within the receptor binding domain of the spike glycoprotein, which is required for binding to ACE2 in human cells. The VOC-202012/01 variant was defined by 23 mutations: 13 non-synonymous mutations, 4 deletions and 6 synonymous mutations (i.e., there were 17 mutations that changed the protein and 6 mutations that did not change the protein). Spike protein changes in VOC 202012/01 include deletions 69-70, deletion 144, N501Y, a570D, D614G, P681H, T716I, S982A, and D1118H. One of the most important changes in VOC-202012/01 appears to be N501Y, the change from asparagine (N) to tyrosine (Y) at amino acid position 501. This mutation alone or in combination with a deletion at position 69/70 in the N-terminal domain (NTD) can enhance the transmission capability of the virus.

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that sera of vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variants that include the following mutations compared to SEQ ID NO: 1: deletions 69-70, deletion 144, N501Y, a570D, D614G, P681H, T716I, S982A, and D1118H.

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against SARs-CoV-2 spike variant "501. V2". This variant was first observed in the sample starting at 10 months 2020, and since then more than 300 cases have been identified in south africa by Whole Genome Sequencing (WGS) with the 501.V2 variant, which was the predominant form of the virus at 12 months 2020. Preliminary results indicate that such variants may have increased transmission capacity. The V2 variant is defined by a plurality of spike protein changes, comprising: D80A, D215G, E484K, N501Y and a701V, and the most recently collected viruses had additional changes: L18F, R246I, K417N and deletions 242-244.

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against SARs-CoV-2 spike variants, which comprise the following mutations: and SEQ ID NO:1 compared to D80A, D215G, E484K, N501Y and a701V, and optionally: compared with SEQ ID NO:1, L18F, R246I, K417N and deletions 242-244. The SARs-CoV-2 spike variant may also include the D614G mutation as compared to SEQ ID NO: 1.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants, as compared to the mRNA of SEQ ID NO:1 by contrast, the SARs-CoV-2 spike variant included a deletion of H69/V70 in the spike protein.

In some embodiments, the polypeptide of SEQ ID NO:1, compared to SEQ ID NO: one or more SARs-CoV-2 spike variants including H69/V70 deletions in the spike protein may include one or more additional mutations (e.g., without limitation, Y144 deletion, N501Y, a570D, D614G, P681H, T716I, S982A, D1118H, D80A, D215G, E484K, a701V, L18F, R246I, K417N, L242/a243/L244 deletion, Y453F, I692V, S1147L, M1229I, etc., as compared to SEQ ID NO: 1).

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against the SARs-CoV-2 spike variant "variant of interest 202012/01" (VOC-202012/01; also referred to as lineage B.1.1.7).

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that sera of vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variants that include the following mutations compared to SEQ ID NO: 1: deletions 69-70, deletion 144, N501Y, a570D, D614G, P681H, T716I, S982A, and D1118H.

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against SARs-CoV-2 spike variant "Cluster (Cluster) 5", also known as Δ FVI-spike by State Serum Institute (SSI). It is found in North judland peninsula (North Jutland) denland, denmark, and is believed to have been transmitted from minks to humans through mink farms. In cluster 5, several different mutations in the spike protein of the virus have been identified. Specific mutations include 69-70 deltaHV (deletion of histidine and valine residues at positions 69 and 70 in the protein), Y453F (change from tyrosine to phenylalanine at position 453), I692V (change from isoleucine to valine at position 692), M1229I (change from methionine to isoleucine at position 1229), and optionally S1147L (change from serine to leucine at position 1147).

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that sera of vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variants that include the following mutations compared to SEQ ID NO: 1: deletion 69-70, Y453F, I692V, M1229I and optionally S1147L.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants, as compared to the mRNA of SEQ ID NO:1 in contrast, the SARs-CoV-2 spike variant includes a mutation at position 614 in the spike protein. In some embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against one or more SARs-CoV-2 spike variants that include a D614G mutation in the spike protein, as compared to SEQ ID NO: 1.

In some embodiments, the polypeptide of SEQ ID NO:1, compared to SEQ ID NO:1 one or more SARs-CoV-2 spike variants comprising a mutation at position 614 in the spike protein or said mutation compared to SEQ ID NO: one or more SARs-CoV-2 spike variants that include a D614G mutation in the spike protein compared to SEQ ID NO:1 may include one or more additional mutations (e.g., without limitation, H69/V70 deletion, Y144 deletion, N501Y, a570D, P681H, T716I, S982A, D1118H, D80A, D215G, E484K, a701V, L18F, R246I, K417N, L242/a243/L244 deletion, Y453F, I692V, S1147L, M1229I, etc.).

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against the SARs-CoV-2 spike variant "variant of interest 202012/01" (VOC-202012/01; also referred to as lineage B.1.1.7).

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that sera of vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variants that include the following mutations compared to SEQ ID NO: 1: deletions 69-70, deletion 144, N501Y, a570D, D614G, P681H, T716I, S982A, and D1118H.

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against SARs-CoV-2 spike variants, which comprise the following mutations: and SEQ ID NO:1 compared to D80A, D215G, E484K, N501Y, a701V and D614G, and optionally: and SEQ ID NO:1 compared to L18F, R246I, K417N and deletions 242-244.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants comprising mutations at positions 501 and 614 in the spike protein compared to SEQ ID NO: 1. In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants that include a N501Y mutation and a D614G mutation in the spike protein, as compared to SEQ ID NO: 1.

In some embodiments, one or more SARs-CoV-2 spike variants comprising mutations at positions 501 and 614 in the spike protein compared to SEQ ID NO:1 or said mutation in position 501 and 614 in comparison to SEQ ID NO:1 or said mutation in position 501 and said mutation in position 614 in comparison to SEQ ID NO:1 compared to one or more SARs-CoV-2 spike mutants that include N501Y and D614G mutations in the spike protein may include one or more additional mutations (e.g., without limitation, H69/V70 deletions, Y144 deletions, a570D, P681H, T716I, S982A, D1118H, D80A, D215G, E484K, a701V, L18F, R246I, K417N, L242/a243/L244 deletions, Y453F, I692V, S1147L, M1229I, etc., compared to SEQ ID NO: 1).

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against the SARs-CoV-2 spike variant "variant of interest 202012/01" (VOC-202012/01; also referred to as lineage B.1.1.7).

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that sera of vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variants that include the following mutations compared to SEQ ID NO: 1: deletions 69-70, deletion 144, N501Y, a570D, D614G, P681H, T716I, S982A, and D1118H.

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against SARs-CoV-2 spike variants, which comprise the following mutations: compared to SEQ ID No. 1, D80A, D215G, E484K, N501Y, a701V and D614G, and optionally: and SEQ ID NO:1 compared to L18F, R246I, K417N and deletions 242-244.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants, as compared to the mRNA of SEQ ID NO:1 in contrast, the SARs-CoV-2 spike variant included a mutation at position 484 in the spike protein. In some embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against one or more SARs-CoV-2 spike variants that are identical to the mRNA of SEQ ID NO:1 in contrast, the SARs-CoV-2 spike variant included an E484K mutation in the spike protein.

In some embodiments, the polypeptide of SEQ ID NO:1, compared to SEQ ID NO: one or more SARs-CoV-2 spike variants comprising a mutation at position 484 in the spike protein compared to SEQ ID NO:1 or said one or more SARs-CoV-2 spike variants comprising a mutation of E484K in the spike protein compared to SEQ ID NO:1 may comprise one or more further mutations (e.g., but not limited to H69/V70 deletion, Y144 deletion, N501Y, a570D, D614G, P681H, T716I, S982A, D1118H, D80A, D215G, a701V, L18F, R246I, K417N, L242/a243/L244 deletion, Y453N, I692N, S1147N, M1229N, T20N, P26N, D138N, R190N, K417K 10272, T10236655, T1023672, V1176: N, etc.).

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against SARs-CoV-2 spike variant "501. V2".

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against SARs-CoV-2 spike variants, which comprise the following mutations: compared to SEQ ID No. 1, D80A, D215G, E484K, N501Y and a701V, and optionally: and SEQ ID NO:1 compared to L18F, R246I, K417N and deletions 242-244. The SARs-CoV-2 spike variant may also include the D614G mutation as compared to SEQ ID NO: 1.

Lineage b.1.1.248, termed brazilian variant, is one of the variants of SARS-CoV-2, which has been designated as p.1 lineage, with 17 unique amino acid changes, 10 of which are in its spike protein, including N501Y and E484K. B.1.1.248 is derived from b.1.1.28. E484K is present in b.1.1.28 and b.1.1.248. B.1.1.248 has a number of S-protein polymorphisms [ L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, H655Y, T1027I, V1176F ], and are similar to the variants described in south africa at certain key RBD positions (K417, E484, N501).

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the sera of the vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variant "b.1.1.28".

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the sera of the vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variant "b.1.1.248".

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that sera of vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variants, as compared to the mRNA of SEQ ID NO:1 in contrast, the SARs-CoV-2 spike variant includes the following mutations: L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, H655Y, T1027I and V1176F.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants, as compared to the mRNA of SEQ ID NO:1, the SARs-CoV-2 spike variant includes mutations at positions 501 and 484 in the spike protein. In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants that include a N501Y mutation and an E484K mutation in the spike protein, as compared to SEQ ID NO: 1.

In some embodiments, one or more SARs-CoV-2 spike variants comprising mutations at positions 501 and 484 in the spike protein compared to SEQ ID NO:1 or said one or more SARs-CoV-2 spike mutants comprising N501Y and E484K mutations in the spike protein compared to SEQ ID NO:1 may comprise one or more further mutations (e.g., but not limited to H69/V70 deletion, Y144 deletion, a570D, D614G, P681H, T716I, S982A, D1118H, D80A, D215G, a701V, L18F, R I, K417N, L242/a243/L244 deletion, Y453N, I N, S1147N, M122369N, T20, P72, P417, R N, T1023672, K N, H N, T N, H N, T1023672, etc.) compared to SEQ ID NO: 1).

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against SARs-CoV-2 spike variant "501. V2".

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against SARs-CoV-2 spike variants, which comprise the following mutations: compared to SEQ ID No. 1, D80A, D215G, E484K, N501Y and a701V, and optionally: and SEQ ID NO:1 compared to L18F, R246I, K417N and deletions 242-244. The SARs-CoV-2 spike variant may also include the D614G mutation as compared to SEQ ID NO: 1.

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the sera of the vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variant "b.1.1.248".

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that sera of vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variants that include the following mutations compared to SEQ ID NO: 1: L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, H655Y, T1027I and V1176F.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants, as compared to the mRNA of SEQ ID NO:1, the SARs-CoV-2 spike variant includes mutations at positions 501, 484, and 614 in the spike protein. In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants that include a N501Y mutation, an E484K mutation, and a D614G mutation in the spike protein, as compared to SEQ ID NO: 1.

In some embodiments, the polypeptide of SEQ ID NO:1, compared to SEQ ID NO: one or more SARs-CoV-2 spike variants comprising mutations at positions 501, 484 and 614 in the spike protein compared to SEQ ID NO:1 or one or more SARs-CoV-2 spike variants comprising a N501Y mutation, an E484K mutation and a D614G mutation in the spike protein compared to SEQ ID NO:1 may comprise one or more further mutations (e.g., without limitation, H69/V70 deletion, Y144 deletion, a570D, P681H, T716I, S982A, D1118H, D80A, D215G, a701V, L18F, R246I, K417N, L242/a243/L244 deletion, Y453N, I N, S1147N, M1229N, T20N, P26N, D138N, R190, K N, K417, T102655, T1023672, T1176, V1176, etc.).

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against SARs-CoV-2 spike variants, which comprise the following mutations: compared to SEQ ID No. 1, D80A, D215G, E484K, N501Y, a701V and D614G, and optionally: compared with SEQ ID NO:1, L18F, R246I, K417N and deletions 242-244.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants, as compared to the mRNA of SEQ ID NO:1 in comparison, the SARs-CoV-2 spike variant includes a deletion of L242/A243/L244 in the spike protein.

In some embodiments, the polypeptide of SEQ ID NO:1, compared to SEQ ID NO:1 compared to one or more SARs-CoV-2 spike variants comprising a L242/a243/L244 deletion in the spike protein may comprise one or more further mutations (e.g., without limitation, H69/V70 deletion, Y144 deletion, N501Y, a570D, D614G, P681H, T716I, S982A, D1118H, D80A, D215G, E484K, a701V, L18F, R246I, K417N, Y453F, I692V, S1147L, M1229I, T20N, P26S, D138Y, R190S, K417T, H655Y, T1027I, V1176F, etc., compared to SEQ ID NO: 1).

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against SARs-CoV-2 spike variant "501. V2".

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against SARs-CoV-2 spike variants, which comprise the following mutations: and SEQ ID NO:1 compared to D80A, D215G, E484K, N501Y, a701V and deletions 242 and 244, and optionally: L18F, R246I and K417N compared to SEQ ID NO: 1. The SARs-CoV-2 spike variant may also include the D614G mutation as compared to SEQ ID NO: 1.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants, as compared to the mRNA of SEQ ID NO:1 in contrast, the SARs-CoV-2 spike variant includes a mutation at position 417 in the spike protein. In some embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against one or more SARs-CoV-2 spike variants that are identical to the mRNA of SEQ ID NO:1 in contrast, the SARs-CoV-2 spike variant included a K417N or K417T mutation in the spike protein.

In some embodiments, the polypeptide of SEQ ID NO:1, compared to SEQ ID NO:1 one or more SARs-CoV-2 spike variants comprising a mutation at position 417 in the spike protein or said mutation compared to SEQ ID NO: one or more SARs-CoV-2 spike variants that include K417N or K417T mutations in the spike protein compared to SEQ ID NO:1 may include one or more additional mutations (e.g., without limitation, H69/V70 deletion, Y144 deletion, N501Y, a570D, D614G, P681H, T716I, S982A, D1118H, D80A, D215G, E484K, a701V, L18F, R246F, L242/a243/L244 deletion, Y36453, I692F, S1147F, M1229F, T20F, P26F, D138F, R190F, H655F, T1027F, V1176F, etc. compared to SEQ ID NO: 1).

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against SARs-CoV-2 spike variant "501. V2".

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against SARs-CoV-2 spike variants, which comprise the following mutations: compared to SEQ ID No. 1, D80A, D215G, E484K, N501Y, a701V and K417N, and optionally: L18F, R246I and deletions 242 and 244 compared to SEQ ID NO: 1. And SEQ ID NO:1 in comparison, the SARs-CoV-2 spike variant may also include the D614G mutation.

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the sera of the vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variant "b.1.1.248".

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that sera of vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variants, as compared to the mRNA of SEQ ID NO:1 in contrast, the SARs-CoV-2 spike variant includes the following mutations: L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, H655Y, T1027I and V1176F.

In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants comprising mutations at positions 417 and 484 and/or 501 in the spike protein as compared to SEQ ID NO: 1. In some embodiments, the mRNA compositions and/or methods described herein are characterized by sera of vaccinated subjects exhibiting neutralizing activity against one or more SARs-CoV-2 spike variants that include a K417N or K417T mutation and an E484K and/or N501Y mutation in the spike protein, as compared to SEQ ID NO: 1.

In some embodiments, the polypeptide of SEQ ID NO:1, compared to SEQ ID NO: one or more SARs-CoV-2 spike variants comprising mutations at positions 417 and 484 and/or 501 in the spike protein compared to 1 or one or more SARs-CoV-2 spike variants comprising K417N or K417T mutations and E484K and/or N501Y mutations in the spike protein compared to SEQ ID NO:1 may comprise one or more further mutations (e.g., without limitation, H69/V70 deletion, Y144 deletion, a570D, D614G, P681 695 2, T716I, S982A, D1118H, D80A, D215G, a701V, L18F, R246F, L242/a243/L244 deletion, Y453F, I F, S1147, M122369, T20F, P F, D F, R190, R655, T11772, T F, T1176, F, etc.).

In particular embodiments, the mRNA compositions and/or methods described herein are characterized by sera from vaccinated subjects exhibiting neutralizing activity against SARs-CoV-2 spike variant "501. V2".

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the serum of the vaccinated subject exhibits neutralizing activity against SARs-CoV-2 spike variants, which comprise the following mutations: compared to SEQ ID No. 1, D80A, D215G, E484K, N501Y, a701V and K417N, and optionally: L18F, R246I and deletions 242 and 244 compared to SEQ ID NO: 1. And SEQ ID NO:1 in comparison, the SARs-CoV-2 spike variant may also include the D614G mutation.

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that the sera of the vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variant "b.1.1.248".

In certain embodiments, the mRNA compositions and/or methods described herein are characterized in that sera of vaccinated subjects exhibit neutralizing activity against SARs-CoV-2 spike variants that include the following mutations compared to SEQ ID NO: 1: L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, H655Y, T1027I and V1176F.

The SARs-CoV-2 spike variants described herein may or may not include the D614G mutation, as compared to SEQ ID NO: 1.

In some embodiments, the mRNA compositions and/or methods described herein can provide protection against SARS-CoV-2 and/or reduce the severity of SARS-CoV-2 infection in at least 50% of subjects receiving such mRNA compositions and/or methods.

In some embodiments, the population treated with an mRNA composition described herein comprises subjects 18-55 years of age. In some embodiments, the population treated with an mRNA composition described herein comprises subjects between 56-85 years of age. In some embodiments, populations treated with mRNA compositions described herein include older subjects (e.g., subjects over 60 years of age, over 65 years of age, over 70 years of age, over 75 years of age, over 80 years of age, over 85 years of age, etc., e.g., 65-85 years of age). In some embodiments, the population treated with an mRNA composition described herein comprises subjects 18-85 years old. In some embodiments, the population treated with an mRNA composition described herein includes subjects 18 years of age or younger. In some embodiments, the population treated with an mRNA composition described herein includes subjects 12 years old or younger. In some embodiments, the population treated with an mRNA composition described herein comprises subjects 10 years old or younger. In some embodiments, a population treated with an mRNA composition described herein can include a pubertal population (e.g., an individual from about 12 to about 17 years of age). In some embodiments, the population treated with an mRNA composition described herein includes infants (e.g., under 1 year of age). In some embodiments, a population treated with an mRNA composition described herein does not include infants whose mothers had received such an mRNA composition described herein during pregnancy (e.g., under 1 year of age). Without wishing to be bound by any particular theory, rat studies such as those shown in example 31 indicate that SARS-CoV-2 neutralizing antibody responses induced in female rats given such mRNA compositions during pregnancy can be transmitted to the fetus. In some embodiments, populations treated with mRNA compositions described herein include infants whose mothers did not receive such mRNA compositions described herein during pregnancy (e.g., under 1 year of age). In some embodiments, a population treated with an mRNA composition described herein can include a pregnant woman; in some embodiments, an infant whose mother has been vaccinated during pregnancy (e.g., receives at least one dose, or only two doses) is not vaccinated for the first weeks, months, or even years (e.g., 1, 2, 3, 4, 5, 6, 7, 8 weeks or more, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 months or more, or 1, 2, 3, 4, 5 years or more) of birth. Alternatively or additionally, in some embodiments, an infant whose mother has been vaccinated during pregnancy (e.g., receives at least one dose, or receives only two doses) may receive reduced vaccination (e.g., lower doses and/or smaller number of administrations-e.g., boost-and/or lower total exposure for a given period of time) after birth, e.g., during the first few weeks, months, or even years (e.g., 1, 2, 3, 4, 5, 6, 7, 8 weeks or more, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 months or more, or 1, 2, 3, 4, 5 years or more) after birth, lower doses and/or smaller number of administrations-e.g., booster-over a given time period). In some embodiments, the compositions provided herein are administered to a population that does not include pregnant women.

In some particular embodiments, the compositions provided herein are administered to a pregnant woman according to a regimen that includes a first dose administered after about 24 weeks of gestation (e.g., after about 22, 23, 24, 25, 26, 27, 28 or more weeks of gestation); in some embodiments, the compositions provided herein are administered to a pregnant woman according to a regimen that includes a first dose administered about 34 weeks prior to gestation (e.g., about 30, 31, 32, 33, 34, 35, 36, 37, 38 weeks prior to gestation). In some embodiments, the compositions provided herein are administered to a pregnant woman according to a regimen that includes a first dose administered after about 24 weeks of gestation (e.g., after about 27 weeks of gestation, e.g., between about 24 weeks and 34 weeks, or between about 27 weeks and 34 weeks) and a second dose administered after about 21 days; in some embodiments, both doses are administered prior to parturition. Without wishing to be bound by any particular theory, it is suggested that such a regimen (e.g., including administration of a first dose after about 24 or 27 weeks of gestation and optionally before about 34 weeks of gestation), and optionally a second dose within about 21 days (ideally before delivery), may have certain advantages with respect to safety (e.g., reducing the risk of preterm birth or fetal morbidity or mortality) and/or efficacy (e.g., administration of a vaccine to an infant) relative to an alternative dosing regimen (e.g., administration at any time during gestation, avoiding dosing during gestation and/or e.g., dosing in the late term so that only one dose is administered during gestation). In some embodiments, as described herein (see also example 34), an infant born to a mother vaccinated during pregnancy (e.g., according to the particular protocols described herein) may not require further vaccination during a period of time after birth (e.g., as described herein), or may require reduced vaccination (e.g., lower dose and/or lower number of administrations-e.g., boost-, and/or lower total exposure over a given period of time).

In some embodiments, the compositions provided herein are administered to a population in which women are advised not to become pregnant for a period of time after vaccination (e.g., after vaccination with a first dose of vaccine, after vaccination with a final dose of vaccine, etc.); in some such embodiments, the period of time may be at least 1 week, at least 2 weeks, at least 3 weeks, at least 4 weeks, at least 5 weeks, at least 6 weeks, at least 7 weeks, at least 8 weeks, at least 9 weeks, at least 10 weeks, or longer, or may be at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, or longer.

In some embodiments, a population treated with an mRNA composition described herein can include a population having one or more particularly high risk conditions or histories, e.g., as described herein. For example, in some embodiments, a population treated with an mRNA composition described herein can include subjects (including, for example, but not limited to, large scale transportation, prisoners, grocery store workers, residents in a long term care facility, butcher or other meat processing workers, paramedics, and/or emergency personnel, e.g., emergency response personnel) whose occupational and/or environmental exposure can greatly increase their risk of being infected with SARS-CoV-2. In particular embodiments, a population treated with an mRNA composition described herein can include health care workers and/or emergency workers, e.g., emergency response workers. In some embodiments, populations treated with mRNA compositions described herein can include those with smoking or e-cigarette history (e.g., over 6 months, 12 months, or longer, including chronic smoking or e-cigarette history). In some embodiments, populations treated with the mRNA compositions described herein can include certain ethnicities that have been determined to be more susceptible to SARS-CoV-2 infection.

In some embodiments, the populations treated with the mRNA compositions described herein can include populations of certain blood types, which may have been determined to be more susceptible to SARS-CoV-2 infection. In some embodiments, populations treated with the mRNA compositions described herein can include immunocompromised subjects (e.g., those with HIV/AIDS; cancer and transplant patients who are taking certain immunosuppressive drugs; autoimmune diseases or other physiological conditions where immunosuppressive therapy is expected to be needed (e.g., within 3 months, within 6 months or more), and those with genetic diseases that affect the immune system (e.g., congenital agammaglobulinemia, congenital IgA deficiency). In some embodiments, populations treated with mRNA compositions described herein can include those having infectious diseases. For example, in some embodiments, populations treated with mRNA compositions described herein can include those infected with Human Immunodeficiency Virus (HIV) and/or hepatitis virus (e.g., HBV, HCV). In some embodiments, populations treated with mRNA compositions described herein can include those with potential medical conditions. Examples of such potential medical conditions may include, but are not limited to, hypertension, cardiovascular disease, diabetes, chronic Sexual respiratory diseases, e.g., chronic lung disease, asthma, etc., cancer and other chronic diseases, e.g., lupus, rheumatoid arthritis, chronic liver disease (e.g., stage 3 or more, e.g., in some embodiments characterized by a Glomerular Filtration Rate (GFR) of 60mL/min/1.73m2Below). In some embodiments, populations treated with mRNA compositions described herein can include overweight or obese subjects, e.g., including in particular a Body Mass Index (BMI) of about 30kg/m2Those above. In some embodiments, populations treated with the mRNA compositions described herein can include subjects who have been previously diagnosed with COVID-19 or evidence of current or previous SARS-CoV-2 infection, e.g., based on serology or nasal swabs. In some embodiments, the population to be treated comprises white and/or non-hispanic/non-hispanic.

In some embodiments, certain mRNA compositions described herein (e.g., BNT162b1) can be selected for administration to an asian population (e.g., a chinese population), or in particular embodiments, to an older asian population (e.g., 60 years or older, such as 60-85 or 65-85 years old).

In some embodiments, mRNA compositions provided herein are administered to and/or evaluated in subjects who have been determined to not show prior infection and/or evidence of current infection prior to administration; in some embodiments, evidence of a prior infection and/or a current infection may be or include evidence of an intact virus or any viral nucleic acid, protein, lipid, etc. (e.g., in a biological sample thereof, such as blood, cells, mucosa, and/or tissue) present in the subject, and/or an immune response by the subject thereto. In some embodiments, mRNA compositions provided herein are administered to and/or evaluated in subjects who have been determined to show evidence of a prior infection and/or current infection prior to administration; in some embodiments, evidence of a prior infection and/or a current infection may be or include evidence of an intact virus or any viral nucleic acid, protein, lipid, etc. (e.g., in a biological sample thereof, such as blood, cells, mucosa, and/or tissue) present in the subject, and/or an immune response by the subject thereto. In some embodiments, the subject is considered to have a prior infection based on the day of dose 1 having a positive N-binding antibody test result or a positive Nucleic Acid Amplification Test (NAAT) result.

In some embodiments, an RNA (e.g., mRNA) composition provided herein is administered to a subject who has been told a risk of side effects, which may include one or more of the following, for example: chills, fever, headache, pain at injection site, muscle pain, fatigue; in some embodiments, an RNA (e.g., mRNA) composition is administered to a subject, who has been invited to notify a healthcare provider when: if one or more such side effects occur, more than mild or moderate side effects are experienced for more than one or several days, or if any serious or unexpected event is experienced by the subject that is reasonably considered likely to be associated with receiving the composition. In some embodiments, the RNA (e.g., mRNA) compositions provided herein are administered to a subject who has been invited to inform a medical institution of a particular medical condition, which may include, for example, one or more of allergy, bleeding disorder or taking blood thinning medication, breast feeding, fever, immune compromised state or taking medications that affect the immune system, pregnancy or planned pregnancy, or the like. In some embodiments, an RNA (e.g., mRNA) composition provided herein is administered to a subject who has been invited to advise a medical facility that another COVID-19 vaccine has been received. In some embodiments, an RNA (e.g., mRNA) composition provided herein is administered to a subject who is free of one of the following medical conditions: undergoing febrile illness, receiving immunosuppressant therapy, receiving anticoagulant therapy, having a hemorrhagic condition (e.g., a condition that prohibits intramuscular injection), or pregnancy and/or breastfeeding/nursing. In some embodiments, an RNA (e.g., mRNA) composition provided herein is administered to a subject who does not receive another COVID-19 vaccine. In some embodiments, an RNA (e.g., mRNA) composition provided herein is administered to a subject that is not allergic to any component of the RNA (e.g., mRNA) composition. Examples of such allergic reactions may include, but are not limited to, dyspnea, swollen airways (fact) and/or throat, rapid heartbeat, rash, dizziness, and/or weakness. In some embodiments, an RNA (e.g., mRNA) composition provided herein is administered to a subject that received a first dose and has no allergic reaction (e.g., as described herein) to the first dose. In some embodiments where an allergic reaction occurs in a subject after receiving a dose of an RNA (e.g., mRNA) composition provided herein, such a subject may be administered one or more interventions such as treatments to address and/or reduce the symptoms of such allergic reactions, e.g., antipyretics and/or anti-inflammatory substances.

In some embodiments, a subject that has received at least one dose of an RNA (e.g., mRNA) composition provided herein is told to avoid exposure to coronavirus (e.g., SARS-CoV-2) unless and until several days (e.g., at least 7 days, at least 8 days, 9 days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least 14 days, etc.) have elapsed since administration of the second dose. For example, a subject who has received at least one dose of an RNA (e.g., mRNA) composition provided herein is informed to take preventative measures against SARS-CoV-2 infection (e.g., maintain social distance, wear a mask, wash hands often, etc.) unless and until several days have passed since the administration of the second dose (e.g., at least 7 days, at least 8 days, 9 days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least 14 days, etc.). Thus, in some embodiments, a method of administering an RNA (e.g., mRNA) composition provided herein comprises administering a second dose of such an RNA (e.g., mRNA) composition provided herein to a subject that receives the first dose and takes precautions to avoid exposure to coronaviruses (e.g., SARS-CoV-2).

In some embodiments, the mRNA compositions described herein can be delivered to a draining lymph node of a subject in need thereof, e.g., for vaccine priming. In some embodiments, such delivery may be by intramuscular administration of the provided mRNA composition.

In some embodiments, different specific mRNA compositions can be administered to different populations of subjects;alternatively or additionally, in some embodiments, different dosing regimens may be administered to different subject populations. For example, in some embodiments, an mRNA composition administered to a particular population of subjects can be characterized by one or more particular effects (e.g., incidence and/or degree of effect) in those population of subjects. In some embodiments, such effects can be or include, for example, neutralizing antibodies and/or T cells (e.g., THT cells of type 1, e.g. CD4+ and/or CD8+T cells), protection against challenge (e.g., by injection and/or nasal exposure, etc.), incidence, severity, and/or persistence of side effects (e.g., reactogenicity), and/or the like.

In some embodiments, one or more of the mRNA compositions described herein may be administered according to established protocols to reduce the incidence of COVID-19 per 1000 human-years, e.g., based on laboratory tests such as Nucleic Acid Amplification Tests (NAATs). In some embodiments, one or more of the mRNA compositions described herein can be administered according to established protocols in order to reduce the incidence of codv-19 every 1000 human-years based on laboratory tests such as Nucleic Acid Amplification Tests (NAATs) in subjects who receive at least one dose of the provided mRNA composition without serological or virological evidence of past SARS-CoV-2 infection (e.g., up to 7 days after receiving the last dose). In some embodiments, one or more of the mRNA compositions described herein may be administered according to established protocols to reduce the incidence of confirmed severe COVID-19 every 1000 human-years. In some embodiments, one or more of the mRNA compositions described herein may be administered according to established protocols in order to reduce the incidence of confirmed severe COVID-19 every 1000 human-years in a subject who received at least one dose of the mRNA composition provided without serological or virological evidence of the prior SARS-CoV-2 infection.

In some embodiments, one or more of the mRNA compositions described herein can be administered according to established protocols such that neutralizing antibodies against the SARS-CoV-2 spike polypeptide and/or immunogenic fragment thereof (e.g., RBD) are produced over a period of time, as measured in serum from the subject, that reaches or exceeds a reference level (e.g., a reference level determined based on human SARS-CoV-2 infection/COVID-19 convalescent serum), and/or a cell-mediated immune response (e.g., a T cell response against SARS-CoV-2) is induced over a period of time, including, for example, in some embodiments, inducing at least one or more MHC restrictions within the SARS-CoV-2 spike polypeptide and/or immunogenic fragment thereof (e.g., RBD), MHC class I restricted) epitopes. In some such embodiments, the period of time may be at least 2 months, 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 8 months, at least 9 months, at least 10 months, at least 11 months, at least 12 months, or longer. In some embodiments, one or more epitopes recognized by vaccine-induced T cells (e.g., CD8+ T cells) can be presented on MHC class I alleles present in at least 50% of the subjects in a population, including, e.g., at least 60%, at least 70%, at least 80%, at least 90% or more; in some such embodiments, the MHC class I allele may be HLA-B0702, HLA-a 2402, HLA-B3501, HLA-B4401, or HLA-a 0201. In some embodiments, the epitope may comprise HLA-a x 0201 YLQPRTFLL; HLA-a 0201 RLQSLQTYV; HLA-a 2402 QYIKWPWYI; HLA-a 2402 NYNYLYRLF; HLA-a 2402 KWPWYIWLGF; HLA-B3501 QPTESIVRF; HLA-B3501 IPFAMQMAY; or HLA-B3501 LPFNDGVYF.

In some embodiments, efficacy is assessed as the incidence of COVID-19 per 1000 human-years in individuals who have no serological or virological evidence of past SARS-CoV-2 infection prior to and during the vaccination regimen; alternatively or additionally, in some embodiments, efficacy is assessed as the incidence of COVID-19 per 1000 human-years in subjects with and without evidence of previous SARS-CoV-2 infection prior to and during the vaccination regimen. In some such embodiments, this incidence is the number of cases of covd-19 confirmed within a specified time period after the final vaccination dose (e.g., the first dose in a single dose regimen; the second dose in two dose regimens, etc.); in some embodiments, such a period of time may be within (i.e., up to and including 7 days) a particular number of days (e.g., 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 or more days). In some embodiments, such a period of time may be within 7 days or within 14 days or within 21 days or within 28 days. In some embodiments, such a period of time may be within 7 days. In some embodiments, such a period of time may be within 14 days.

In some embodiments (e.g., in some embodiments where efficacy is assessed), a subject is determined to have experienced a COVID-19 infection if one or more of the following is established: detecting a SARS-CoV-2 nucleic acid in a sample from the subject, detecting an antibody that specifically recognizes SARS-CoV-2 (e.g., SARS-Co-V-2 spike protein), one or more symptoms of a COVID-19 infection, and combinations thereof. In some such embodiments, detection of SARS-CoV-2 nucleic acid can involve, for example, NAAT testing of a turbinate swab sample. In some such embodiments, the detection of the relevant antibodies may involve a serological test of the blood sample or portion thereof. In some such embodiments, the symptoms of a COVID-19 infection may be or include: fever, new or increased cough, new or increased breathlessness, chills, new or increased muscle pain, new loss of taste or smell, sore throat, diarrhea, vomiting, and combinations thereof. In some such embodiments, the symptoms of a COVID-19 infection may be or include: fever, new or increased cough, new or increased breathlessness, chills, new or increased muscle pain, new loss of taste or smell, sore throat, diarrhea, vomiting, fatigue, headache, nasal congestion or discharge, nausea, and combinations thereof. In some such embodiments, a subject is determined to have experienced a codv-19 infection if such subject has experienced one such symptom and has also received a positive test for SARS-CoV-2 nucleic acid or antibody or both. In some such embodiments, if a subject has experienced one such symptom, and has also received a positive test for SARS-CoV-2 nucleic acid, then such subject is determined to have experienced a COVID-19 infection. In some such embodiments, if a subject has experienced one such symptom, and has also received a positive test for SARS-CoV-2 antibody, then such subject is determined to have experienced a COVID-19 infection.

In some embodiments (e.g., in some embodiments where efficacy is assessed), a subject is determined to have experienced a severe COVID-19 infection if such subject has experienced one or more of: clinical manifestations of a systemic disease indicative or severe at rest (e.g., respiratory rate greater than or equal to 30 breaths per minute, heart rate of 125 or more beats per minute, SpO in the air of the room at sea level2Less than or equal to 93% or less than 300m Hg of PaO2/FiO2One or more of), respiratory failure (e.g., requiring one or more of high flow oxygen, non-invasive ventilation, mechanical ventilation, ECMO), evidence of shock (systolic pressure below 90mm Hg, diastolic pressure below 60mm Hg, vasopressors required), severe acute renal, hepatic, or neurological dysfunction, entry into an intensive care unit, death, and combinations thereof.

In some embodiments, one or more mRNA compositions described herein can be administered according to established protocols to reduce the percentage of subjects reporting at least one of: one or more local responses (e.g., as described herein) for up to 7 days after each dose; (ii) one or more systemic events up to 7 days after each dose; (iii) adverse events from the first dose to 1 month after the last dose (e.g., as described herein); and/or (iv) a serious adverse event (e.g., as described herein) 6 months after the first dose to the last dose.

In some embodiments, one or more subjects that have received an RNA (e.g., mRNA) composition described herein can be monitored (e.g., for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 days or longer, including, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 weeks or longer, including, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 months or longer, including, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 years or longer) to assess, e.g., the presence of evidence of an immune response to a SARS-CoV-2 or another coronavirus exposure and/or immune response, evidence of any adverse events, etc. In some embodiments, the monitoring may be accessible over the telephone. Alternatively or additionally, in some embodiments, the monitoring may be face-to-face.

In some embodiments, the therapeutic effect conferred by one or more mRNA compositions described herein may be characterized by: (i) SARS-CoV-2 anti-S1 binding antibody level above a predetermined threshold; (ii) a SARS-CoV-2 anti-RBD binding antibody level above a predetermined threshold; and/or (iii) SARS-CoV-2 serum neutralizing titer above a threshold level, e.g., 1 month, 3 months, 6 months, 9 months, 12 months, 18 months, and/or 24 months after completion of vaccination at baseline. In some embodiments, anti-S1 binding antibody levels and/or anti-RBD binding antibody levels and/or serum neutralization titers can be characterized by Geometric Mean Concentration (GMC), Geometric Mean Titer (GMT), or geometric mean fold increase (GMFR).

In some embodiments, the therapeutic effect conferred by one or more mRNA compositions described herein may be characterized by a percentage of treated subjects exhibiting SARS-CoV-2 serum neutralization titers above a predetermined threshold (e.g., at baseline, 1 month, 3 months, 6 months, 9 months, 12 months, 18 months, and/or 24 months after completion of vaccination) that is higher than a percentage of untreated subjects exhibiting SARS-CoV-2 serum neutralization titers above such a predetermined threshold (e.g., as described herein). In some embodiments, the serum neutralization titer can be characterized by Geometric Mean Concentration (GMC), Geometric Mean Titer (GMT), or geometric mean fold increase (GMFR).

In some embodiments, the therapeutic effect conferred by one or more mRNA compositions described herein can be characterized by detecting SARS-CoV-2 NVA-specific binding antibodies.

In some embodiments, the therapeutic effect conferred by one or more mRNA compositions described herein can be characterized by the SARS-CoV-2 detection of a nucleic acid amplification assay.

In some embodiments, the therapeutic effect conferred by one or more mRNA compositions described herein may be characterized by induction of a cell-mediated immune response (e.g., a T cell response against SARS-CoV-2), including, for example, in some embodiments, induction of T cells that recognize at least one or more MHC-restricted (e.g., MHC class I-restricted) epitopes within a SARS-CoV-2 spike polypeptide and/or an immunogenic fragment thereof (e.g., RBD). In some embodiments, one or more epitopes recognized by vaccine-induced T cells (e.g., CD8+ T cells) can be presented on MHC class I alleles present in at least 50% of subjects in a population, including, e.g., at least 60%, at least 70%, at least 80%, at least 90% or more; in some such embodiments, the MHC class I allele may be HLA-B0702, HLA-a 2402, HLA-B3501, HLA-B4401, or HLA-a 0201. In some embodiments, the epitope may comprise HLA-a x 0201 YLQPRTFLL; HLA-a 0201 RLQSLQTYV; HLA-a 2402 QYIKWPWYI; HLA-a 2402 NYNYLYRLF; HLA-a 2402 KWPWYIWLGF; HLA-B3501 QPTESIVRF; HLA-B3501 IPFAMQMAY; or HLA-B3501 LPFNDGVYF.

In some embodiments, a primary vaccine potency (VE) for one or more mRNA compositions described herein can be established when there is sufficient evidence (a posteriori probability) that primary VE1 or both primary VE1 and primary VE2 are > 30% or greater (including, e.g., 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, 90% or more, 95% or more, 96% or more, 97% or more, 98% or more), where primary VE is defined as primary VE 100 x (1-IRR); and IRR was calculated as the ratio of COVID-19 prevalence in the vaccine group to the corresponding prevalence in the placebo group. Primary VE1 represents the VE for confirmed COVID-19 for the prophylactic mRNA compositions described herein in participants with no evidence of infection prior to vaccination, while primary VE2 represents the VE for confirmed COVID-19 for the prophylactic mRNA compositions described herein in all participants after vaccination. In some embodiments, primary VE1 and VE2 may be evaluated sequentially to control overall type I errors at 2.5% (layered testing). In some embodiments in which it is confirmed that one or more RNA (e.g., mRNA) compositions described herein reach a primary VE endpoint as discussed above, the secondary VE endpoint (e.g., confirmation of severe COVID-19 in participants who have no evidence of infection prior to vaccination and confirmation of severe COVID-19 in all participants) may be evaluated sequentially, e.g., by the same methods as discussed above for primary VE endpoint evaluation (stratification test). In some embodiments, the evaluation of the primary and/or secondary VE endpoints may be based on at least 20,000 or more subjects (e.g., at least 25,000 or more subjects) randomly assigned to the vaccine or placebo group in a 1:1 ratio, e.g., based on the following assumptions: (i) an annual prevalence of 1.0% in the placebo group, and (ii) 20% of participants were unable to evaluate or had serological evidence of prior infection with SARS-CoV-2, potentially immunizing them against further infection.

In some embodiments, one or more mRNA compositions described herein can be administered according to established protocols to achieve maintenance and/or continued enhancement of an immune response. For example, in some embodiments, a dosing regimen may comprise a first dose, optionally followed by one or more subsequent doses; in some embodiments, the need, timing, and/or scale (magnitude) of any such subsequent dose may be selected to maintain, enhance and/or modify one or more immune responses or characteristics thereof. In some embodiments, the number, timing, and/or amount of doses effective in administering to the relevant population has been established. In some embodiments, the number, timing, and/or amount of doses can be adjusted for individual subjects; for example, in some embodiments, one or more characteristics of an immune response in an individual subject can be evaluated at least once (and optionally more than once, e.g., multiple times, typically separated, typically at pre-selected intervals) after receiving a first dose. For example, antibodies, B cells, and/or T cells (e.g., CD4+ and/or CD 8) can be evaluated+T cells) and/or the cells secreted thereby The presence of the seed and/or the nature (identity) and/or extent of the response to a particular antigen and/or epitope. In some embodiments, the need, timing, and/or amount for subsequent doses can be determined based on such evaluations.

As described above, in some embodiments, one or more subjects (e.g., for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 days or more, including, for example, 1, 2, 3, 4, 5, 10, 11, 12 weeks or more, including, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 months or more, including, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 years or more) that have received an RNA (e.g., mRNA) composition described herein can be monitored from the time of any particular dose to assess, for example, the presence of an immune response to the components of the administered composition, evidence of SARS-CoV-2 exposure and/or another coronavirus, evidence of any adverse events, etc., including the evaluation of antibodies, B cells and/or T cells (e.g., CD 4)+And/or CD8+T cells) and/or cytokines secreted thereby, and/or may assess the nature and/or extent of a response to a particular antigen and/or epitope. The compositions described herein may be administered according to a regimen that includes one or more such monitoring steps.

For example, in some embodiments, the need, timing, and/or amount of a second dose relative to a first dose (and/or subsequent doses relative to a previous dose) is assessed, determined, and/or selected such that administration of such a second (or subsequent) dose effects a magnification or modification of the immune response (e.g., as described herein) observed after the first (or other previous) dose. In some embodiments, such amplification of an immune response (e.g., an immune response described herein) can be at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or higher compared to the level of immune response observed after the first dose. In some embodiments, the immune response at such magnification may be at least 1.5-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 20-fold, at least 30-fold, or higher compared to the level of immune response observed after the first dose.

In some embodiments, the need, timing, and/or amount of a second (or subsequent) dose relative to a first (or other previous) dose is evaluated, determined, and/or selected such that administration of a later dose extends the persistence of an immune response (e.g., as described herein) observed after an earlier dose; in some such embodiments, persistence may be extended for at least 1 week, at least 2 weeks, at least 3 weeks, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 8 months, at least 9 months, or longer. In some embodiments, the immune response observed after the first dose may be characterized by the production of neutralizing antibodies against SARS-CoV-2 spike polypeptide and/or immunogenic fragment thereof (e.g., RBD) and/or the induction of a cell-mediated immune response (e.g., a T cell response against SARS-CoV-2) as measured in serum from the subject, including, for example, in some embodiments, the induction of T cells that recognize one or more MHC-restricted (e.g., MHC class I-restricted) epitopes within SARS-CoV-2 spike polypeptide and/or immunogenic fragment thereof (e.g., RBD). In some embodiments, one or more epitopes recognized by vaccine-induced T cells (e.g., CD8+ T cells) can be presented on MHC class I alleles present in at least 50% of the subjects in a population, including, e.g., at least 60%, at least 70%, at least 80%, at least 90% or more; in some such embodiments, the MHC class I allele may be HLA-B0702, HLA-a 2402, HLA-B3501, HLA-B4401, or HLA-a 0201. In some embodiments, the epitope may comprise HLA-a x 0201 YLQPRTFLL; HLA-a 0201 RLQSLQTYV; HLA-a 2402 QYIKWPWYI; HLA-a 2402 NYNYLYRLF; HLA-a 2402 KWPWYIWLGF; HLA-B3501 QPTESIVRF; HLA-B3501 IPFAMQMAY; or HLA-B3501 LPFNDGVYF.

In some embodiments, the need, timing, and/or amount of a second dose relative to a first dose (or other subsequent dose relative to a previous dose) is assessed, determined, and/or selected such that administration of such second (or subsequent) dose maintains or exceeds a reference level of immune response; in some such embodiments, the reference level is determined based on human SARS-CoV-2 infection/COVID-19 convalescent serum and/or PBMC samples drawn from the subject (e.g., at least a period of time after PCR confirmation, such as at least 14 days or more, including, e.g., 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 25 days, 30 days, 35 days, 40 days, 45 days, 50 days, 55 days, 60 days or more, when the subject is asymptomatic). In some embodiments, the immune response can be characterized by the production of neutralizing antibodies against SARS-CoV-2 spike polypeptide and/or immunogenic fragment thereof (e.g., RBD) and/or the induction of a cell-mediated immune response (e.g., a T cell response against SARS-CoV-2) as measured in serum from a subject, including, for example, in some embodiments, the induction of T cells that recognize one or more MHC-restricted (e.g., MHC class I-restricted) epitopes within SARS-CoV-2 spike polypeptide and/or immunogenic fragment thereof (e.g., RBD). In some embodiments, one or more epitopes recognized by vaccine-induced T cells (e.g., CD8+ T cells) can be presented on MHC class I alleles present in at least 50% of the subjects in a population, including, e.g., at least 60%, at least 70%, at least 80%, at least 90% or more; in some such embodiments, the MHC class I allele may be HLA-B0702, HLA-a 2402, HLA-B3501, HLA-B4401, or HLA-a 0201. In some embodiments, the epitope may comprise HLA-a x 0201 YLQPRTFLL; HLA-a 0201 RLQSLQTYV; HLA-a 2402 QYIKWPWYI; HLA-a 2402 NYNYLYRLF; HLA-a 2402 KWPWYIWLGF; HLA-B3501 QPTESIVRF; HLA-B3501 IPFAMQMAY; or HLA-B3501 LPFNDGVYF.

In some embodiments, determining the need, timing, and/or amount of a second (or subsequent) dose may comprise one or more of the following steps: after the first (or other prior) dose (e.g., after 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 days or more), assessing the presence and/or expression level of neutralizing antibodies against SARS-CoV-2 spike polypeptide and/or immunogenic fragment thereof (e.g., RBD), as measured in serum from the subject, and/or assessing induction of a cell-mediated immune response (e.g., a T cell response against SARS-CoV-2), including, for example, in some embodiments, the induction of T cells that recognize one or more MHC-restricted (e.g., MHC class I-restricted) epitopes within SARS-CoV-2 spike polypeptide and/or immunogenic fragment thereof (e.g., RBD). In some embodiments, one or more epitopes recognized by vaccine-induced T cells (e.g., CD8+ T cells) can be presented on MHC class I alleles present in at least 50% of the subjects in a population, including, e.g., at least 60%, at least 70%, at least 80%, at least 90% or more; in some such embodiments, the MHC class I allele may be HLA-B0702, HLA-a 2402, HLA-B3501, HLA-B4401, or HLA-a 0201. In some embodiments, the epitope may comprise HLA-a x 0201 YLQPRTFLL; HLA-a 0201 RLQSLQTYV; HLA-a 2402 QYIKWPWYI; HLA-a 2402 NYNYLYRLF; HLA-a 2402 KWPWYIWLGF; HLA-B3501 QPTESIVRF; HLA-B3501 IPFAMQMAY; or HLA-B3501 LPFNDGVYF.

In some embodiments, the kits provided herein may comprise a real-time monitoring recording device, e.g., in some embodiments, capable of providing a transport temperature, a transport time, and/or a location.

In some embodiments, the RNA (e.g., mRNA) compositions described herein can be transported, stored, and/or utilized in a container (e.g., a vial or syringe), for example, a glass container (e.g., a glass vial or syringe), which in some embodiments can be a single dose container or a multi-dose container (e.g., which can be arranged or configured to hold, and/or which in some embodiments can hold, a single dose or multiple doses of a product for administration). In some embodiments, a multi-dose container (e.g., a multi-dose vial or syringe) may be arranged or configured to hold, and/or may hold 2, 3, 4, 5, 6, 7, 8, 9, 10 or more doses; in some particular embodiments, it may be designed to hold and/or may hold 5 doses. In some embodiments, a single-dose or multi-dose container (e.g., a single-dose or multi-dose vial or syringe) may be arranged or configured to hold and/or may hold a volume or amount that is greater than the number of doses shown, e.g., to allow for some loss in transfer and/or administration. In some embodiments, the RNA (e.g., mRNA) compositions described herein can be transported, stored, and/or used in preservative-free glass containers (e.g., preservative-free glass vials or syringes, e.g., single-dose or multi-dose preservative-free glass vials or syringes). In some embodiments, the RNA (e.g., mRNA) compositions described herein can be shipped, stored, and/or used in preservative-free glass containers (e.g., preservative-free glass vials or syringes, e.g., single-dose or multi-dose preservative-free glass vials or syringes) containing 0.45ml of a frozen liquid (e.g., including 5 doses). In some embodiments, the RNA (e.g., mRNA) compositions described herein and/or the containers (e.g., vials or syringes) in which the compositions are placed, transported, stored, and/or used may be maintained at a temperature below room temperature, at 4 ℃ or below 4 ℃, at 0 ℃ or below 0 ℃, at-20 ℃ or below-20 ℃, at-60 ℃ or below-60 ℃, at-70 ℃ or below-70 ℃, at-80 ℃ or below-80 ℃, at-90 ℃ or below-90 ℃, and the like. In some embodiments, the RNA (e.g., mRNA) compositions described herein and/or the container (e.g., vial or syringe) in which the RNA (e.g., mRNA) compositions described herein are placed, transported, stored, and/or utilized may be maintained at a temperature between-80 ℃ and-60 ℃, and in some embodiments protected from light. In some embodiments, the RNA (e.g., mRNA) compositions described herein and/or the container (e.g., vial or syringe) in which the compositions are placed, transported, stored, and/or used may be maintained at a temperature below about 25 ℃, and in some embodiments protected from light. In some embodiments, the RNA (e.g., mRNA) compositions described herein and/or the container (e.g., vial or syringe) in which the composition is placed, transported, stored, and/or used may be maintained at a temperature below about 5 ℃ (e.g., below about 4 ℃) and, in some embodiments, protected from light. In some embodiments, the RNA (e.g., mRNA) compositions described herein and/or the container (e.g., vial or syringe) in which the composition is placed, transported, stored, and/or used may be maintained at a temperature below about-20 ℃, and in some embodiments protected from light. In some embodiments, the RNA (e.g., mRNA) compositions described herein and/or the container (e.g., vial or syringe) in which the compositions are placed, transported, stored, and/or used may be maintained at a temperature above about-60 ℃ (e.g., in some embodiments at or above about-20 ℃, in some embodiments at or above about 4-5 ℃, in either case, optionally below about 25 ℃), and in some embodiments protected from light, or otherwise not employed with an affirmative step (e.g., cooling means) to substantially bring the storage temperature below about-20 ℃.

In some embodiments, the RNA (e.g., mRNA) compositions described herein and/or the container (e.g., vial or syringe) in which the composition is placed can be transported, stored, and/or utilized with and/or in the environment of a thermal protection material or container and/or a temperature regulating material. For example, in some embodiments, an RNA (e.g., mRNA) composition described herein and/or a container (e.g., vial or syringe) in which the composition is placed can be shipped, stored, and/or utilized with ice and/or dry ice and/or with a thermally insulating material. In some particular embodiments, the container (e.g., vial or syringe) in which the RNA (e.g., mRNA) composition is placed in a tray or other holding device and is further contacted with (or otherwise present in) a temperature regulating (e.g., ice and/or dry ice) material and/or an insulating material. In some embodiments, a plurality of containers (e.g., a plurality of vials or syringes, such as single-use or multi-use vials or syringes as described herein) in which provided RNA (e.g., mRNA) compositions are placed are co-located (e.g., in a common tray, rack, box, etc.) and packaged (or otherwise present) with temperature regulating (e.g., ice and/or dry ice) material and/or insulating material. As one example, in some embodiments, a plurality of containers (e.g., a plurality of vials or syringes, such as single-use or multi-use vials or syringes as described herein) in which RNA (e.g., mRNA) compositions are placed in a common tray or rack, and a plurality of such trays or racks are stacked in a carton surrounded by temperature regulating material (e.g., dry ice) in an insulated (e.g., insulated) transport. In some embodiments, the temperature regulating material is replenished periodically (e.g., within 24 hours of arrival at the site, and/or every 2 hours, 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, etc.). Preferably, the insulated vehicle should be re-entered infrequently, and desirably should not occur more than twice a day. In some embodiments, the insulated transport is reclosed 5, 4, 3, 2, or 1 minute or less after opening. In some embodiments, provided RNA (e.g., mRNA) compositions that have been stored in an insulated transport for a period of time, optionally still are usable within a particular temperature range. For example, in some embodiments, an RNA (e.g., mRNA) composition described herein can be used for up to 10 days if an insulated vehicle comprising the provided RNA (e.g., mRNA) composition is at or maintained (e.g., stored) at a temperature in a range of about 15 ℃ to about 25 ℃; that is, in some embodiments, the provided RNA (e.g., mRNA) composition is administered to the subject for no more than 10 days while remaining in an insulated vehicle at a temperature range of about 15 ℃ to about 25 ℃. Alternatively or additionally, in some embodiments, an RNA (e.g., mRNA) composition provided can be used for up to 10 days if it is in or maintained (e.g., stored) within an incubation vehicle that has been maintained (e.g., stored) at a temperature in a range of about 15 ℃ to about 25 ℃; that is, in some embodiments, the provided RNA (e.g., mRNA) composition is administered to the subject for no more than 10 days while remaining in an insulated vehicle that has been maintained at a temperature in the range of about 15 ℃ to about 25 ℃.

In some embodiments, provided RNA (e.g., mRNA) compositions are shipped and/or stored in a frozen state. In some embodiments, the provided RNA (e.g., mRNA) compositions are shipped and/or stored as frozen suspensions, which, in some embodiments, are preservative-free. In some embodiments, the frozen RNA (e.g., mRNA) composition is thawed. In some embodiments, a thawed RNA (e.g., mRNA) composition (e.g., suspension) can comprise opaque amorphous particles that are white to off-white in color. In some embodiments, a thawed RNA (e.g., mRNA) composition can be used for a few days (e.g., 1, 2, 3, 4, 5, or 6 days) after thawing if maintained (e.g., stored) at or below room temperature (e.g., about 30 ℃ or less, about 25 ℃ or less, about 20 ℃ or less, about 15 ℃ or less, about 10 ℃ or less, about 8 ℃ or less, about 4 ℃ or less, etc.). In some embodiments, the thawed RNA (e.g., mRNA) composition can be used after storage at a temperature between about 2 ℃ and about 8 ℃ (e.g., such a small number of days); alternatively or additionally, the thawed RNA (e.g., mRNA) composition can be used within a few (e.g., 1, 2, 3, 4, 5, or 6) hours after thawing at room temperature. Thus, in some embodiments, a provided RNA (e.g., mRNA) composition that has been thawed and maintained at or below room temperature (in some embodiments between about 2 ℃ and about 8 ℃) for no more than 6, 5, 4, 3, 2, or 1 day is administered to a subject. Alternatively or additionally, in some embodiments, a provided RNA (e.g., mRNA) composition that has been thawed and maintained at room temperature for no more than 6, 5, 4, 3, 2, or 1 hour is administered to a subject. In some embodiments, the provided RNA (e.g., mRNA) compositions are transported and/or stored in a concentrated state. In some embodiments, such concentrated compositions are diluted prior to administration. In some embodiments, the diluted composition is administered within about 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 hour after dilution; in some embodiments, such administration is within 6 hours after dilution. Thus, in some embodiments, a provided diluted preparation of an RNA (e.g., mRNA) composition is administered to a subject within 6 hours after dilution (e.g., after having been maintained at an appropriate temperature, e.g., at a temperature below room temperature, at 4 ℃ or below 4 ℃, at 0 ℃ or below 0 ℃, at-20 ℃ or below-20 ℃, at-60 ℃ or below-60 ℃, at-70 ℃ or below-70 ℃, at-80 ℃ or below-80 ℃, etc., and typically at 2 ℃ or above about 2 ℃, e.g., between about 2 ℃ and about 8 ℃ or between about 2 ℃ and about 25 ℃, as described herein). In some embodiments, unused composition is discarded within hours (e.g., about 10, about 9, about 8, about 7, about 6, about 5 hours or less) after dilution; in some embodiments, unused composition is discarded within 6 hours of dilution.

In some embodiments, an RNA (e.g., mRNA) composition (e.g., frozen composition, liquid concentrated composition, diluted liquid composition, etc.) that is stored, transported, or used may have been maintained at a temperature substantially above-60 ℃ for a period of at least 1, 2, 3, 4, 5, 6, 7 days or longer, or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 weeks or more, or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 months or more; in some such embodiments, such compositions may have been maintained at a temperature of about-20 ℃ or above about-20 ℃ for such a period of time, and/or at a temperature of up to or about 4-5 ℃ for such a period of time, and/or may have been maintained at a temperature of above about 4-5 ℃ and optionally about 25 ℃ for a period of time of less than 2 months and/or optionally to about 1 month. In some embodiments, such compositions may not be substantially at a temperature above about 4-5 ℃, and in particular are not stored, transported, or used (or exposed) at a temperature of about 25 ℃ or close to about 25 ℃ for a period of about 2 weeks, or in some embodiments, for a period of 1 week. In some embodiments, such compositions may not be stored, transported, or used (or exposed) at temperatures substantially above about-20 ℃, and in particular not at about 4-5 ℃ or at temperatures approaching about 4-5 ℃ for a period of about 12 months, 11 months, 10 months, 9 months, 8 months, 7 months, 6 months, 5 months, 4 months, 3 months, 2 months, or, in some embodiments, for a period of about 8 weeks or 6 weeks or substantially above about 2 months, or, in some embodiments, 3 months, or, in some embodiments, 4 months.

In some embodiments, RNA (e.g., mRNA) compositions (e.g., frozen compositions, liquid concentrated compositions, diluted liquid compositions, etc.) that are stored, transported, or used can be protected from light. In some embodiments, one or more steps may be taken to reduce or minimize exposure of such compositions to light (e.g., they may be placed in a container such as a vial or syringe). In some embodiments, exposure to direct sunlight and/or ultraviolet light is avoided. In some embodiments, the diluted solution may be handled and/or utilized under normal room light conditions (e.g., no specific steps are taken to minimize or reduce exposure to room light). It will be appreciated that strict adherence to aseptic techniques is desirable during handling (e.g., dilution and/or administration) of the RNA (e.g., mRNA) compositions described herein. In some embodiments, the RNA (e.g., mRNA) compositions described herein are not administered intravenously (e.g., are not injected). In some embodiments, the RNA (e.g., mRNA) compositions described herein are not administered intradermally (e.g., are not injected). In some embodiments, the RNA (e.g., mRNA) compositions described herein are not administered subcutaneously (e.g., are not injected). In some embodiments, the RNA (e.g., mRNA) compositions described herein are not administered (e.g., are not injected) intravenously, intradermally, or subcutaneously. In some embodiments, an RNA (e.g., mRNA) composition described herein is not administered to a subject having a known hypersensitivity reaction to any of its components. In some embodiments, a subject to whom an RNA (e.g., mRNA) composition has been administered is monitored for one or more signs of anaphylaxis. In some embodiments, a subject administered an RNA (e.g., mRNA) composition has previously received at least one dose of a different SARS-CoV-2 vaccine; in some embodiments, a subject administered an RNA (e.g., mRNA) composition has not previously received a different SARS-CoV-2 vaccine. In some embodiments, the body temperature of a subject is measured immediately prior to administration of an RNA (e.g., mRNA) composition (e.g., before or shortly after thawing, dilution, and/or administration of such a composition); in some embodiments, the administration is delayed or cancelled if the subject is determined to have a fever. In some embodiments, an RNA (e.g., mRNA) composition described herein is not administered to a subject receiving anticoagulant therapy or having or susceptible to a bleeding disorder or disease condition that prohibits intramuscular injection. In some embodiments, an RNA (e.g., mRNA) composition described herein is administered by a healthcare professional who has communicated to the subject receiving the composition and information regarding side effects and risk. In some embodiments, an RNA (e.g., mRNA) composition described herein is administered by a healthcare professional who has agreed to submit an adverse event report for any serious adverse event, which may include, for example, one or more of the following: the development of death, disability or congenital abnormality/birth defects (e.g., in the child of the subject), hospitalization (including prolonged existing hospitalization), life-threatening events, medical or surgical intervention to prevent death, the ability to perform normal life functions being sustained or severely or substantially disrupted; or another significant medical event that may endanger the individual and may require medical or surgical intervention (treatment) to prevent one of the other consequences.

In some embodiments, the provided RNA compositions are administered to a population under 18 years of age, or under 17 years of age, or under 16 years of age, or under 15 years of age, or under 14 years of age, or under 13 years of age, e.g., according to established protocols, such that the incidence of one or more local response events, shown below, does not exceed the incidence shown below:

injection site pain (75% after the first dose and/or the second dose, and/or a lower incidence after the second dose, e.g., 65% after the second dose);

injection site redness (less than 5% after the first dose and/or the second dose); and/or

Swelling of the injection site (less than 5% after the first dose and/or the second dose).

In some embodiments, the provided RNA compositions are administered to a population under 18 years of age, or under 17 years of age, or under 16 years of age, or under 15 years of age, or under 14 years of age, or under 13 years of age, e.g., according to established protocols, such that the incidence of one or more systemic response events, shown below, does not exceed the incidence shown below:

fatigue (55% after the first and/or second dose);

headache (50% after the first and/or second dose);

muscle pain (40% after the first and/or second dose);

Chills (40% after the first and/or second dose);

joint pain (20% after the first and/or second dose);

fever (25% after the first and/or second dose);

emesis (10% after the first and/or second dose); and/or

Diarrhea (10% after the first and/or second dose).

In some embodiments, an individual under 18 years of age, or under 17 years of age, or under 16 years of age, or under 15 years of age, or under 14 years of age, or under 13 years of age who has been administered with a provided RNA composition and experienced one or more local and/or systemic response events (e.g., described herein) is administered a drug that ameliorates one or more symptoms of one or more local and/or systemic response events (e.g., described herein). In some embodiments, an antipyretic and/or analgesic may be administered to such individuals.

Drawings

FIG. 1: schematic representation of S protein organization of SARS-CoV-2S protein

The sequence within the S1 subunit consists of a Signal Sequence (SS) and a Receptor Binding Domain (RBD), a key subunit within the S protein that is involved in binding to the human cell receptor ACE 2. The S2 subunit contains an S2 protease cleavage site (S2'), followed by a Fusion Peptide (FP) for membrane fusion, a heptapeptide repeat with a Central Helical (CH) domain (HR1 and HR2), a transmembrane domain (TM) and a Cytoplasmic Tail (CT).

FIG. 2: contemplated constructs for the development of SARS-CoV-2 vaccines

Based on intact and wild-type S proteins, we have designed different constructs that encode (1) an intact protein with mutations within a short distance of the first heptad repeat (HRP1) that include stabilizing mutations that preserve the neutralization sensitive site, (2) an S1 domain or (3) an RB-only domain (RBD). In addition, to stabilize the protein fragment, the minor fibrin domain (F) is fused to the C-terminus. All constructs start with a Signal Peptide (SP) to ensure golgi transport to the cell membrane.

FIG. 3: antibody immune response to influenza HA using LNP formulated modRNA

BALB/c mice were immunized 2 times with 1. mu.g of vaccine candidate. The total amount of viral antigen specific immunoglobulin g (igg) was measured by ELISA. Antibody functionality was assessed by VNT.

FIG. 4: t cell response to influenza HA using an LNP formulated modRNA platform

BALB/c mice were IM immunized 2 times with 1. mu.g of the candidate vaccine. T cell response analysis was performed on T cell stimulation recovered from the spleen using antigen-specific peptides. IFN γ release was measured using ELISpot assay after peptide stimulation.

FIG. 5: anti-S protein IgG response 7, 14, 21 and 28 days after immunization with BNT162a1

BALB/c mice were immunized once with 1, 5 or 10 μ g of RBL063.3 IM in LNP. On days 7, 14, 21 and 28 after immunization, the animals were bled and serum samples were analyzed for the total amount of anti-S1 (left) and anti-RBD (right) antigen-specific immunoglobulin g (igg) measured by ELISA. Values for serum dilution of 1:100 are included in the figure for day 7, 14, 21 and 28. One dot in the figure represents one mouse, and each mouse sample was measured in duplicate (group size n-8; each group included mean + SEM).

FIG. 6: anti-S protein IgG response 7, 14, 21 and 28 days after immunization with BNT162b1

BALB/c mice were immunized once with 0.2, 1 or 5 μ g of RBP020.3 IM in LNP. On days 7, 14, 21 and 28 after immunization, the animals were bled and serum samples were analyzed for the total amount of anti-S1 (left) and anti-RBD (right) antigen-specific immunoglobulin g (igg) measured by ELISA. Different serum dilutions were included in the figure for day 7 (1:100), day 14 (1: 300), day 21 (1: 900) and day 28 (1: 2700). One dot in the figure represents one mouse, and each mouse sample was measured in duplicate (group size n-8; each group includes mean + SEM).

FIG. 7: neutralization of SARS-CoV-2 pseudovirus 14, 21, and 28 days after immunization with BNT162b1

BALB/c mice were immunized once with 0.2, 1 or 5 μ g of RBP020.3 IM in LNP. At 14, 21 and 28 days after immunization, animals were bled and sera tested for SARS CoV-2 pseudovirus neutralization. The figure shows pVN50 serum dilutions (50% reduction in infection events compared to positive control without serum). One dot in the figure represents one mouse. Samples from each mouse were measured in duplicate. Group size n is 8. The mean + SEM is shown for each group using horizontal bars with whiskers (whisker). LLOQ, lower limit of quantitation. ULOQ, upper limit of quantitation.

FIG. 8: anti-S protein IgG response 7, 14 and 21 days after immunization with BNT162c1

BALB/c mice were immunized once with 0.2, 1 or 5 μ g of RBS004.3 IM formulated in LNP. On days 7, 14 and 21 after immunization, the animals were bled and serum samples were analyzed for the total amount of anti-S1 (left) and anti-RBD (right) antigen-specific immunoglobulin g (igg) measured by ELISA. Different serum dilutions were included in the figure for day 7 (1:100), day 14 (1:300) and day 21 (1: 900). One dot in the figure represents one mouse, and each mouse sample was measured in duplicate (group size n-8; each group included mean + SEM).

FIG. 9: neutralization of SARS-CoV-2 pseudovirus 14 and 21 days after immunization with BNT162c1

BALB/c mice were immunized once with 0.2, 1 or 5 μ g of RBS004.3 IM formulated in LNP. At 14 and 21 days after immunization, animals were bled and sera tested for SARS CoV-2 pseudovirus neutralization. The figure shows pVN50 serum dilutions (50% reduction in infection events compared to positive control without serum). One dot in the figure represents one mouse. Samples from each mouse were measured in duplicate. Group size n is 8. Each group shows mean + SEM with horizontal bars with whiskers. LLOQ, lower limit of quantitation. ULOQ, upper limit of quantitation.

FIG. 10: anti-S protein IgG responses 7, 14, 21, and 28 days after immunization with LNP formulated RBL063.1

BALB/c mice were immunized once with 1, 5 or 10 μ g of RBL063.1 IM in LNP. On days 7, 14, 21 and 28 after immunization, the animals were bled and serum samples were analyzed for the total amount of anti-S1 (left) and anti-RBD (right) antigen-specific immunoglobulin g (igg) measured by ELISA. Different serum dilutions were included in the figure for day 7 (1:100), day 14 (1:100), day 21 (1:300) and day 28 (1: 900). One dot in the figure represents one mouse, and each mouse sample was measured in duplicate (group size n-8; each group includes mean + SEM).

FIG. 11: neutralization of SARS-CoV-2 pseudovirus 14, 21 and 28 days after immunization with RBL063.1 formulated in LNP

BALB/c mice were immunized once with 1, 5 or 10 μ g of RBL063.1 IM in LNP. At 14, 21 and 28 days after immunization, animals were bled and sera tested for SARS CoV-2 pseudovirus neutralization. The figure shows pVN50 serum dilutions (50% reduction in infection events compared to positive control without serum). One dot in the figure represents one mouse. Samples from each mouse were measured in duplicate. Group size n is 8. Each group shows mean + SEM with horizontal bars with whiskers. LLOQ, lower limit of quantitation. ULOQ, upper limit of quantitation.

FIG. 12: anti-S protein IgG response 7, 14 and 21 days after immunization with BNT162b2 (RBP 020.1 formulated in LNP)

BALB/c mice were immunized once with 0.2, 1 or 5 μ g of RBP020.1 IM in LNP. On days 7, 14 and 21 after immunization, the animals were bled and serum samples were analyzed for the total amount of anti-S1 (left) and anti-RBD (right) antigen-specific immunoglobulin g (igg) measured by ELISA. Different serum dilutions were included in the figure for day 7 (1:100), day 14 (1:300) and day 21 (1: 1100). One dot in the figure represents one mouse, and each mouse sample was measured in duplicate (group size n-8; each group included mean + SEM).

FIG. 13: neutralization of SARS-CoV-2 pseudovirus 14 and 21 days after immunization with BNT162b2 (RBP 020.1 formulated in LNP)

BALB/c mice were immunized once with 0.2, 1 or 5 μ g of RBP020.1 IM in LNP. On days 14 and 21 after immunization, animals were bled and sera tested for SARS CoV-2 pseudovirus neutralization. The figure shows pVN50 serum dilutions (50% reduction in infection events compared to positive control without serum). One dot in the figure represents one mouse. Samples from each mouse were measured in duplicate. Group size n is 8. Each group shows mean + SEM with horizontal bars with whiskers. LLOQ, lower limit of quantitation. ULOQ, upper limit of quantitation.

FIG. 14: anti-S protein IgG responses 7, 14 and 21 days after immunization with LNP formulated RBS004.2

BALB/c mice were immunized once with 0.2, 1 or 5 μ g of RBS004.2 IM formulated in LNP. On days 7, 14 and 21 after immunization, the animals were bled and serum samples were analyzed for the total amount of anti-S1 (left) and anti-RBD (right) antigen-specific immunoglobulin g (igg) measured by ELISA. Different serum dilutions were included in the figure for day 7 (1:100), day 14 (1:300) and day 21 (1: 900). One dot in the figure represents one mouse, and each mouse sample was measured in duplicate (group size n-8; each group included mean + SEM).

FIG. 15: neutralization of SARS-CoV-2 pseudovirus 14 and 21 days after immunization with LNP formulated RBS004.2

BALB/c mice were immunized once with 0.2, 1 or 5 μ g of RBS004.2 IM formulated in LNP. At 14 and 21 days after immunization, animals were bled and sera tested for SARS CoV-2 pseudovirus neutralization. The figure shows pVN50 serum dilutions (50% reduction in infection events compared to positive control without serum). One dot in the figure represents one mouse. Samples from each mouse were measured in duplicate. Group size n is 8. Each group shows mean + SEM with horizontal bars with whiskers. LLOQ, lower limit of quantitation. ULOQ, upper limit of quantitation.

FIG. 16: ALC-0315 Activity in the screening Process

FIG. 17: luciferase expression was monitored on the right (injection site), dorsal (injection site) and ventral (drainage to liver) sides of the animals following intramuscular administration in Wild Type (WT) or ApoE knockout C57Bl/6 mice in the presence or absence of ApoE 3. Luciferase expression was detected at 4, 24, 72 and 96 hours post-dose using Xenolight D-fluorescein reject.

FIG. 18: luciferase activity following Intravenous (IV) and Intramuscular (IM) administration in Wild Type (WT) or ApoE knockout C57Bl/6 mice in the presence (KO +) or absence (KO) ApoE 3. Luciferase expression was detected 4 hours after dosing using Xenolight D-luciferin reject.

FIG. 19: general Structure of RNA

Schematic representation of the general structure of an RNA vaccine with a 5' -cap, 5' -and 3' -untranslated regions, coding sequence with an intrinsic secretion signal peptide and GS-linker, and a poly (a) -tail. Note that individual elements are not drawn to scale exactly as compared to the sequence length of the individual elements.

UTR ═ untranslated region; sec is a secretory signal peptide; RBD ═ receptor binding domain; GS-serine linker.

FIG. 20: general Structure of RNA

Schematic representation of the general structure of an RNA drug substance with a 5' -cap, 5' -and 3' -untranslated regions, coding sequence with an intrinsic secretion signal peptide and GS-linker, and a poly (a) -tail. Note that individual elements are not drawn to scale exactly as compared to the sequence length of the individual elements.

GS-glycine-serine linker; UTR ═ untranslated region; sec is a secretory signal peptide; RBD is a receptor binding domain.

FIG. 21: general Structure of RNA

Schematic representation of the general structure of an RNA vaccine with a 5' -cap, 5' -and 3' -untranslated regions, Venezuelan Equine Encephalitis Virus (VEEV) RNA-dependent RNA polymerase replicase with intrinsic secretory signal peptide and GS-linker and the coding sequence of SARS-CoV-2 antigen, and poly (A) -tail. Note that individual elements are not drawn to scale exactly as compared to the sequence length of the individual elements.

UTR ═ untranslated region; sec is a secretory signal peptide; RBD ═ receptor binding domain; GS-serine-glycine linker.

FIG. 22: ELISpot assay 28 days after immunization with BNT162b1

BALB/c mice were immunized once with 1. mu.g of RBP020.3 IM in LNP. On day 28 after immunization, mice were euthanized and splenocytes were prepared. ELISpot assays were performed using MACS-sorted CD4+ and CD8+ T cells. T cells were stimulated with S protein or RBD specific overlapping peptide pools and IFN- γ secretion was measured to assess T cell responses. One dot in the graph represents the individual spot count of one mouse, and each mouse sample was measured in duplicate (group size n-8; each group includes the mean).

FIG. 23: cytokine concentrations in the restimulated splenocyte supernatants 12 days after immunization with BNT162b1

BALB/c mice were immunized once with 5. mu.g of RBP020.3 IM in LNP. On day 12 post immunization, mice were euthanized. Splenocytes were prepared and stimulated with a pool of S protein-specific overlapping peptides. After 48h of stimulation, supernatants were collected and cytokine concentrations were determined. One dot in the graph represents the individual cytokine concentration of one mouse, each mouse sample being measured in duplicate (group size n-8; each group includes mean).

FIG. 24: t cell immunophenotyping in PBMC 7 days after immunization with BNT162b1

BALB/c mice were immunized once with 5. mu.g of RBP020.3 IM in LNP. On day 7 post immunization, mice were bled. Flow cytometry analysis of PBMCs was performed on T cells. Definition of T cells as viable CD3+CD4+And CD3+CD8+T cells. Additional phenotyping markers are included in the figure. Tfh cell selection from CD4+T cell gated and defined as CD4+T-bet-GATA3-CD44+CD62L-PD-1+CXCR5+A cell. One dot in the figure represents the individual cell fraction of one mouse (group size n-8; each group includes the mean).

FIG. 25: immunophenotyping of B cells in draining lymph nodes 12 days after immunization with BNT162B1

BALB/c mice were immunized once with 5. mu.g of RBP020.3 IM in LNP. On day 12 post immunization, mice were euthanized. Flow cytometric analysis of lymphocytes was performed on B cells. Activated B cells are gated within single, living lymphocytes and are defined as IgD-Dump (CD4, CD8, F4/80, GR-1)-A cell. Plasma cells defined as CD138+B220low/-A cell. Transformed B cells were gated from non-plasma cells and were defined as CD19+CD138- IgM-. Germinal Center (GC) B cells are gated from transformed B cells and are defined as CD19+IgM-CD38-CD95+Cells and gated for IgG1 and IgG2 a. One dot in the figure represents the individual cell fraction of one mouse (group size n-8; each group includes the mean).

FIG. 26: ELISpot assay 28 days after immunization with LNP formulated modRNA RBP020.1

BALB/c mice were immunized once with 5. mu.g of RBP020.1 IM in LNP. On day 28 after immunization, mice were euthanized and splenocytes were prepared. ELISpot assays were performed using MACS-sorted CD4+ and CD8+ T cells. T cells were stimulated with a pool of S protein-specific overlapping peptides, and IFN- γ secretion was measured to assess T cell responses. One dot in the graph represents the individual spot count of one mouse, and each mouse sample was measured in duplicate (group size n-8; each group includes the mean).

FIG. 27 is a schematic view showing: cytokine concentrations in the supernatants of restimulated splenocytes 28 days after immunization with modRNA RBP020.1 formulated in LNP

BALB/c mice were immunized once with 5. mu.g of RBP020.1 IM in LNP. On day 28 post immunization, mice were euthanized. Splenocytes were prepared and stimulated with a pool of S protein-specific overlapping peptides. After 48h of stimulation, supernatants were collected and cytokine concentrations were determined. One dot in the graph represents the individual cytokine concentration of one mouse, each mouse sample being measured in duplicate (group size n-8; each group includes mean).

FIG. 28: ELISpot assay 28 days after immunization with LNP-formulated sarRNA RBS004.2

BALB/c mice were immunized once with 5. mu.g of RBS004.2 IM in LNP. On day 28 after immunization, mice were euthanized and splenocytes were prepared. ELISpot assays were performed using MACS-sorted CD4+ and CD8+ T cells. T cells were stimulated with a pool of S protein-specific overlapping peptides, and IFN- γ secretion was measured to assess T cell responses. One dot in the graph represents the individual spot count of one mouse, and each mouse sample was measured in duplicate (group size n-8; each group includes the mean).

FIG. 29: cytokine concentrations in the supernatants of restimulated splenocytes 28 days after immunization with LNP formulated sarRNA RBS004.2

BALB/c mice were immunized once with 1. mu.g of RBS004.2 IM in LNP. On day 28 post immunization, mice were euthanized. Splenocytes were prepared and stimulated with a pool of S protein-specific overlapping peptides. After 48h of stimulation, supernatants were collected and cytokine concentrations were determined. One dot in the graph represents the individual cytokine concentration of one mouse, each mouse sample being measured in duplicate (group size n-8; each group includes mean).

FIG. 30: schematic representation of S protein tissue of SARS-CoV-2S protein and novel constructs for the development of SARS-CoV-2 vaccines

Based on the wild-type S protein, we designed two different transmembrane anchored RBD-based vaccine constructs encoding RBD fragments fused to the T4 secondary fibrin trimerization domain (F) and the native (autochthonous) transmembrane domain (TM). Construct (1) starts with the SARS-CoV-2-S signal peptide (SP; AA 1-19 of the S protein) and construct (2) starts with the human Ig heavy chain signal peptide (huSec) to ensure Golgi transport to the cell membrane.

FIG. 31: anti-S protein IgG responses 6, 14, and 21 days after immunization with modRNA formulated in LNP-C12 encoding transmembrane anchored RBD-based vaccine constructs

BALB/C mice were IM immunized once with 4 μ g of the transmembrane anchored RBD based vaccine construct (replacing BNT162b3C/BNT162b3d) formulated in LNP-C12. On days 6, 14 and 21 after immunization, the animals were bled and serum samples were analyzed for the total amount of anti-S1 (left) and anti-RBD (right) antigen-specific immunoglobulin g (igg) measured by ELISA. Different serum dilutions were included in the figure for day 6 (1:50), day 14 (1:300) and day 21 (1: 900). One dot in the figure represents one mouse, and each mouse sample was measured in duplicate (group size n-8; each group includes mean + SEM).

FIG. 32: neutralization of SARS-CoV-2 pseudovirus 6, 14 and 21 days after immunization with modRNA formulated in LNP-C12 encoding a transmembrane anchored RBD based vaccine construct

BALB/C mice were IM immunized once with 4 μ g of the transmembrane anchored RBD based vaccine construct (replacing BNT162b3C/BNT162b3d) formulated in LNP-C12. On days 6, 14 and 21 after immunization, animals were bled and sera tested for SARS CoV-2 pseudovirus neutralization. The figure shows pVN50 serum dilutions (50% reduction in infection events compared to positive control without serum). One dot in the figure represents one mouse. Each mouse sample was measured in duplicate. Group size n is 8. Each group shows mean + SEM with horizontal bars with whiskers. LLOQ, lower limit of quantitation. ULOQ, upper limit of quantitation.

FIG. 33: immunogenicity of BNT162b1 in rhesus monkeys and comparison with human convalescent serum

Rhesus monkeys were immunized with 30 μ g or 100 μ g of BNT162b1 or placebo (0.9% NaCl) on days 0 and 21. Sera were obtained before and 14, 21, 28 and 35 days after immunization; PBMCs were obtained before immunization and 14 and 42 days after immunization. Serum from patients with COVID-19 was obtained 20-40 days after the onset of symptoms and at least 14 days after the asymptomatic convalescence phase. a, geometric mean concentration of IgG bound to recombinant S1 protease fragment of SARS-CoV-2S in rhesus monkey sera extracted at the indicated times after immunization (n 6/group, all measurement time points for placebo group are indicated under "control") and in human convalescent sera (n 62). b, SARS-CoV-2 geometric mean 50% neutralization titers of rhesus monkey serum (n 6/group, all measurement time points for the placebo group are shown under "control") and human convalescent serum (n 38). Multiple comparison test by two-tailed one-way ANOVA and Dunnett' s The P value is determined a priori. c, IFN γ, IL-2, TNF (T) production in rhesus PBMC on day 42H1) IL-21 or IL-4 (T)H2) Cytokine CD4+Flow cytometric analysis of T cells. P values were determined by the two-tailed Kruskal-Wallis test followed by the Dunn's multiple comparison test. Each data point corresponds to an individual animal.

FIG. 34: overview of the study population

FIG. 35: local response reported within 7 days of all dose levels of vaccination

The injection site (local) response solicited was: pain at the injection site (mild-no-activity-disturbed; moderate-no-activity-disturbed; severe-no-daily-activity; grade 4-emergency or hospitalization) and redness and swelling (mild-2.5-5.0 cm in diameter; medium-5.5-10.0 cm in diameter; severe-10.0 cm in diameter; grade 4-redness as necrosis or exfoliative dermatitis, swelling as necrosis). Data were collected using an electronic diary 14 days after each vaccination.

FIG. 36: a: systemic events reported within 7 days after vaccination 1: all dosage levels; b: systemic events reported within 7 days after vaccination 2: 10 μ g &30 μ g dose levels

The systemic events solicited were: nausea/vomiting (mild-no interference to activity or 1-2 times within 24 hours; moderate-no interference to activity or >2 times within 24 hours; severe-prevention of daily activity or requirement of intravenous water; grade 4-emergency or hospitalization due to hypotensive shock), diarrhea (mild-no interference to activity; moderate-no interference to activity; repeated use of non-narcotic analgesics >24 hours or some interference to activity; severe-no interference to activity, any use of narcotic analgesics or prevention of daily activity; grade 4-emergency or hospitalization), headache (mild-no interference to activity; moderate-no interference to activity; severe-prevention of activity; grade 4-emergency or hospitalization), fatigue/fatigue (mild-no interference to activity; moderate-no interference to activity; severe-no interference to activity; grade 4; severe-prevention of daily activity; grade 4 or hospitalization) Hospital), muscle pain (pain occurring in areas other than the injection site; mild-no interference to activity; moderate-some interference with activity; severe is significant; blocking daily activities; grade 4, emergency or hospitalization), joint pain (mild, no interference to activity; moderate-some interference with activity; severe is significant; blocking daily activities; grade 4 ═ emergency or hospitalization), and fever (mild ═ 100.4 ° F-101.1 ° F [38.0 ℃ to 38.4 ℃); medium 101.2 ° F-102.0 ° F [38.5 ℃ -38.9 ℃; severe 102.1 ° F-104.0 ° F [39.0 ℃ -40.0 ℃) ]; grade 4 >104.0 ° F [ >40.0 ℃ ]).

FIG. 37: immunogenicity of BNT162b1 IgG GMC and SARS CoV binding to RBD 250% neutralizing titers after 1 or 2 doses

On day 1 (all dose levels and placebo) and day 21 (10 μ g and 30 μ g dose levels and placebo), subjects from 15 groups were immunized with BNT162b1 (n-12) or placebo (P, n-3) at the indicated dose levels. Sera were obtained before immunization (day 1) and 7, 21 and 28 days after the first immunization. Human COVID-19 convalescent serum (HCS) (n-38) was obtained 20-40 days after the onset of symptoms and at least 14 days after the asymptomatic convalescent period. a, GMC binding IgG to recombinant RBD. Lower limit of quantitation (LLOQ)1.15 (dotted line). b, 50% SARS-CoV-2 neutralized GMT. Each data point represents a serum sample and each vertical bar represents the geometric mean with a 95% confidence interval.

FIG. 38: BNT162b1 induced strong CD4 and CD 8T cell responses in humans

BNT162 induced T cells: ex vivo INF γ ELISpot; t cell responses of 8 tested subjects. Here: subjects vaccinated with 10 μ g BNT162b1 prime/boost; CEF: CMV, EBV, influenza CD 8T cell epitope cocktail, CEFT: CMV, EBV, influenza, tetanus CD 4T cell epitope mixture.

FIG. 39: BNT162b1 induced IgG concentration

On day 1 (all dose levels) and day 22 (all dose levels except 60 μ g), subjects were immunized with BNT162b1 (n 12/group, starting from day 22, n 11 for the 10 μ g and 50 μ g cohorts (cohort). Sera were obtained on day 1 (pre-prime) and on days 8, 22 (pre-boost), 29 and 43. The pre-dose responses at all dose levels were pooled. Human COVID-19 convalescent sera (HCS, n-38) were obtained at least 14 days after PCR confirmation and when the donor was no longer symptomatic. LLOQ/2 values were plotted for RBD-binding IgG concentrations below the lower limit of quantitation (LLOQ ═ 1.15). Arrows indicate vaccination. Bars indicate no booster immunizations. The values above the bars are geometric means with 95% confidence intervals. At the time of submission, day 43 data waited for 5 subjects in a 50 μ g cohort and all subjects in a 60 μ g cohort.

FIG. 40: viral neutralization titer induced by BNT162b1

The vaccination schedule and serum sampling were the same as in figure 39. a, SARS-CoV-250% neutralization titre (VNT) in immunized subjects and COVID-19 convalescent patients (HCS)50). For values below the lower limit of quantitation (LLOQ) ═ 20, LLOQ/2 values were plotted. Arrows indicate day of immunization. Bars indicate no booster immunizations. Geometric mean with 95% confidence interval (values above the bars). Day 43 data for 5 subjects who had no 50 μ g cohort and all subjects in the 60 μ g cohort at the time of filing, b mean IgG binding concentration to RBD (GMC) (see fig. 39) and VNT at day 29 50Correlation of (all evaluable subject sera). Non-parametric Spearman correlation. c, shows a 50% neutralization titer of pseudoviruses in the pseudoviral group of 17 SARS-CoV-2 spike protein variants (including 16 RBD mutants and the predominant spike protein variant D614G) (pVNT)50) (dose levels 10, 30 and 50 μ g, n-1-2 per group; day 29). Lower limit of quantitation (LLOQ) is 40. Geometric mean value.

FIG. 41: BNT162b 1-induced CD4+And CD8+Frequency and size of T cell response

The vaccination schedule is shown in figure 39. PBMCs obtained on day 1 (before) and day 29 (after, 7 days after boosting) (1 and 50. mu.g, 8 for each n; 10 and 30. mu.g, 10 for each n) were CD 4-enriched in PBMCs+Or CD8+T cell effectors and stimulation with overlapping peptide pools representing vaccine-encoded RBDs alone overnight were used for evaluation in direct ex vivo IFN γ ELISpot. Common pathogen T cell epitope pools CEF (CMV, EBV, influenza virus HLA class I epitopes) and CEFT (CMV, E)BV, influenza virus, tetanus toxoid HLA class II epitopes) were used to evaluate general T cell reactivity, and the medium was used as a negative control. Each dot represents the normalized mean spot count of duplicate wells from one study subject after subtraction of the medium only control. a, the proportion above the data points after vaccination is detectable CD4 in the total number of test subjects per dose cohort +Or CD8+Number of subjects responding with T cells. b, exemplary CD4 of 10 μ g cohort subjects+And CD8+ELISpot. c RBD-specific CD4 in all prime/boost vaccinated subjects+And CD8+T cell responses and their baseline CEFT and CEF specific T cell responses. dose cohorts 10-50 μ g (1 and 50 μ g, 8 for each n; 10 and 30 μ g, 10 for each n) of VNT50(see FIG. 40a) and CD4+Correlation of T cell responses (as in figure 41). Non-parametric Spearman correlation.

FIG. 42: BNT162b 1-induced cytokine polarization of T cells

Vaccination schedules and PBMC samples are as in figure 41. PBMCs of vaccinees and COVID-19 convalescent donors were stimulated with overlapping peptide pools representing the vaccine-encoded RBD (HCS n ═ 6; in (c)) and analyzed by flow cytometry (a-c) and bead-based immunoassay (d). a, cytokine-producing CD4 from 10 μ g cohort subjects+And CD8+Exemplary pseudo-color flow cytometry plots of T cells. b, RBD-specific CD4 producing the indicated cytokines+RBD-specific CD4 with total cytokine production by T cells+Fraction of T cells (fraction), and c, RBD-specific CD8 producing the indicated cytokines+(left) or CD4+(right) fraction of total circulating T cells (fraction) of T cells in the same subset. (b) One CD4 non-responder from the 30 μ g queue was excluded ( <0.02% total cytokine-producing T cells) and one CD8 non-responder(s) ((ii)<0.01% total cytokine-producing T cells). The value above the data point is the average fraction of all dose cohorts. d, PBMC from a 50- μ g cohort. Each dot represents the average of one study subject subtracted from duplicate wells for DMSO control. The lower limit of quantitation (LLOQ) is 6.3pg/m for TNFL, 2.5pg/mL for IL-1. beta. and 7.6pg/mL for IL-12p 70. Average value (b).

FIG. 43: schedules of vaccination and evaluation

FIG. 44: solicited adverse events

On day 1 (all dose levels) and day 22 (all dose levels except 60 μ g), subjects were immunized with BNT162b1 at the dose levels indicated (n-12/group, starting from day 22, n-11 for the 10 μ g and 50 μ g cohorts). a, b, by day (days 1-9, days 22-30) and the number of subjects in the cohort who had a local (a) or systemic response (b). Classification of adverse events is done according to FDA recommendations (U.S. department of Health and Human Services, Administration, F.and D. & Research, C.for B.E.and. perception mapping scale for help Administration and Administration volentifier Administration in predictive significance standards (2007.) see https: t/www.fda.gov/correlation-information/search-FDA-identification-statistics/perception-mapping-hierarchy-Administration-and-Administration-volentifier-predictive-prediction-significance).

FIG. 45: pharmacodynamic marker

Subjects were immunized with BNT162b1 at the indicated dose levels on day 1 (all dose levels) and day 22 (all dose levels except 60 μ g). a, kinetics of C-reactive protein (CRP) levels and b, kinetics of lymphocyte counts. The dotted lines represent the upper and lower limits of the reference range. For values below the lower limit of quantitation (LLOQ ═ 0.3), LLOQ/2 values were plotted (a).

FIG. 46: correlation of antibodies and T-response

Subjects were immunized with BNT162b1 at the indicated dose levels on day 1 (all dose levels) and day 22 (all dose levels except 60 μ g). a, RBD-specific IgG response at day 29 (from FIG. 39) and CD4+Correlation of T cell responses (1 and 50 μ g, 8 for each n; 10 and 30 μ g, 10 for each n). Non-parametric Spearman correlation. b dose cohorts 10-50 μ g (1 and 50 μ g, 8 for each n; 10 and 30 μ g, 10 for each n) of CD4+And CD8+Correlation of T cell responses (as in figure 41). The parameter Pearson correlation. C, RBD specificity I on day 29gG response (from FIG. 39) and CD8+Correlation of T cell responses (1 and 50 μ g, 8 for each n; 10 and 30 μ g, 10 for each n). Non-parametric Spearman correlation.

FIG. 47: gating strategy for flow cytometry analysis of the data shown in fig. 42

Flow cytometry gating strategies for identifying IFN γ, IL-2 and IL-4 secreting T cells in a subject PBMC sample. a, mixing CD4+And CD8+T cells are gated within single, living lymphocytes. b, c, CD4+Gating of IFN γ, IL-2 and IL-4 in T cells (b) and CD8+Gating of IFN γ and IL-2 in T cells (c).

FIG. 48: BNT162b 118-55 years old: local response after each dose

FIG. 49: BNT162b 118-55 years old: systemic events after each dose

FIG. 50: BNT162b 165-85 years old: RBD-binding IgG GMC

FIG. 51: BNT162b 165-85 years old: 50% SARS-CoV-2 neutralization GMT

FIG. 52: BNT162b 218-55 years old: local response after each dose

FIG. 53: BNT162b 218-55 years old: systemic events after each dose

FIG. 54: BNT162b 265-85 years old: local response after each dose

FIG. 55: BNT162b 265-85 years old: systemic events after each dose

FIG. 56: BNT162b 218-55 years old: IgG GMC binding to S1

FIG. 57: BNT162b 218-55 years old: 50% SARS-CoV-2 neutralization GMT

FIG. 58: BNT162b 265-85 years old: IgG GMC binding to S1

FIG. 59: BNT162b 265-85 years old: 50% SARS-CoV-2 neutralization GMT

FIG. 60: BNT162b 2-elicited T cell responses in mice

Splenocytes from BALB/c mice immunized with BNT162b2 or buffer IM were re-stimulated ex vivo with either the full length S peptide cocktail or a negative control (irrelevant peptide in a, right); there are no peptides in (a, left) and (c). P-values were determined by two-tailed paired t-test. (a) IFN γ ELISpot (left) from splenocytes collected 12 days after immunization of mice with 5 μ g of BNT162b2 (n-8/group). IFN γ ELISpot from isolated splenic CD4+ T cells or CD8+ T cells 28 days after immunization of mice with 1 μ g BNT162b2 (n-8 mice/group) (neutralization right). (b) Splenocytes from mice immunized with 5 μ g BNT162b2 or buffer (control) (n-8/group) released CD8+ T cell specific cytokines as determined by flow cytometry. The S peptide specific response was corrected for background (no peptide). (c) Mice were immunized with 1 μ g BNT162b2 (n 8/group, n 7 for IL-4, IL-5 and IL-13, since for S-peptide stimulated samples one anomaly [ Q1% ]) was removed by the routs test followed by cytokine production by splenocytes obtained 28 days, as determined by bead-based multiplex analysis.

FIG. 61: IFN γ ELISpot data for 5 subjects vaccinated with 10 μ g BNT162b2 vaccine

Every 10 th6Individual cells were counted in background-subtracted spots from replicate samples before (before) and on day 29 (after-7 days post-boost). T cell response analysis was performed in a GCLP-compliant manner using a validated ex vivo IFN γ ELISpot assay. All tests were performed in duplicate and included negative and positive controls (medium only and anti-CD 3). Furthermore, peptide epitopes derived from Cytomegalovirus (CMV), Epstein Barr Virus (EBV) and influenza virus were used as positive controls. PBMCs that remove CD4 or CD8 were stimulated with overlapping peptides covering the N-and C-terminal portions of the spike glycoprotein in pre-coated ELISpot plates (Mabtech) for 16-20 h. To analyze ex vivo T cell responses, bound IFN γ was visualized by alkaline phosphatase conjugated secondary antibodies. The plates were scanned using a Robot ELISPOT reader and analyzed by ImmunoCapture V6.3 or AID ELISPOT 7.0 software. The spot counts are summarized as the average of each replicate sample. T cell counts were calculated as the sum of spot counts detected after stimulation with S-pool 1 and S-pool 2. Peptide-stimulated T cell responses were compared with medium-only incubated effects as negative controls using an ELISpot data analysis tool (EDA) based on two statistical tests (no partition resampling) according to Moodie et al (Moodie Z.et al., J Immunol Methods 315,2006,121-32; Moodie Z.et al., Cancer Immunol other 59,2010,1489-501) The comparison is performed, thus providing sensitivity while maintaining control over the false positive rate. No significant change was observed between the previous and 29 day T cell responses to positive control peptides from CMV, EBV and influenza viruses (not shown).

FIG. 62: examples of CD4+ and CD8+ IFN γ ELISpot data

As figure 61, IFN γ ELISpot was performed using PBMCs obtained from subjects before immunization and after dose 1 of 10 μ g BNT162b2 on day 29 (7 days after dose 2). HLA class I and class II peptide pools CEF (cytomegalovirus [ CMV ], Epstein Barr virus [ EBV ] (7 days post dose 2) and influenza virus, HLA class I epitope cocktail) and CEFT (CMV, EBV, influenza virus and tetanus toxoid HLA class II epitope cocktail) were used as benchmark controls to evaluate CD8+ and CD4+ T cell reactivity.

FIG. 63: comparison of BNT162b2 elicited response to baseline INF γ ELISpot response

IFN γ spot counts from PBMC samples from day 29 (7 days after dose 2) obtained from 5 subjects immunized with 10 μ g of BNT162b2 on days 1 and 22. CEF (mixture of CMV, EBV and influenza virus HLA class I epitopes) and CEFT (mixture of CMV, EBV, influenza virus and tetanus toxoid HLA class II cell epitopes) were used as benchmark controls to evaluate CD8+ and CD4+ T cell reactivity. The horizontal line represents the median value.

FIG. 64: design and characterization of immunogens

a, BNT162b 1. A line graph of RNA (left) and a cartoon graph of LNP (right). UTR, untranslated region; SP, signal peptide. b, representative 2D-like mean (2D) from electron microscopy of negatively stained RBD-foldon trimers. Frame edge: 37 nm. c, concentration refinement of the extracellular domain of ACE2 bound to RBD monomer, ACE2/B0AT 1/RBD-foldon trimer complex inDensity map of (a). Surface color-coded in subunits. The band model, refined to density, shows the RBD-ACE2 binding interface, labeling residues likely to mediate polar interactions.

FIG. 65: mouse immunogenicity

a-c, BALB/c mice (n ═ 8/group) were immunized Intramuscularly (IM) with 0.2, 1 or 5 μ g of BNT162b1 or buffer. Geometric mean ± 95% CI, P-value of each group compared day 28 with baseline sera (multiple comparisons analyzed for mixed effects using Dunnett's multiple comparison test) that were not immunized (0 μ g; n ═ 8) (a, c). a, IgG responses to RBD binding in sera obtained 7, 14, 21 and 28 days after immunization, determined by ELISA. For day 0, pre-screening of randomized animals was performed (n-4). b, representative surface plasmon resonance sensorgram of binding kinetics of His-tagged RBDs to immobilized mouse IgG from serum 28 days after immunization with 5 μ g BNT162b1 (n-8). Actual binding (green) and best fit of data to the 1:1 binding model (black). c, VSV-SARS-CoV-2 pseudovirus 50% serum neutralization Titer (pVNT) 50). d-f, splenocytes from BALB/c mice immunized with IM using BNT162b1 or buffer (control) were re-stimulated ex vivo with either the full-length S peptide mixture or the negative control (no peptide in d, left and (e, f); irrelevant peptide in d, right). P-values were determined by two-tailed paired t-test. d, IFN γ ELISpot (left) of splenocytes collected 12 days after immunization of mice with 5 μ g of BNT162b1 (n-8/group). Spleen CD4 isolated 28 days after immunization with 1. mu.g BNT162b1+T cells (n-7, one outlier removed by Grubbs test, α -0.05) or CD8+IFN γ ELISpot (on the right of neutralization) of T cells (n ═ 8). e, T cell specific cytokine release from splenocytes of mice immunized with 5 μ g BNT162b1 (n-8/group) was determined by flow cytometry. The S peptide specific response was corrected for background (no peptide). Cytokine production by splenocytes obtained 28 days after immunization of mice with 0.2 μ g BNT162b1 (n-8/group) was determined by bead-based multiplex analysis.

FIG. 66: immunogenicity of BNT162b1 in rhesus monkeys and comparison with human convalescent serum

a, b, male rhesus monkeys 2-4 years old (n ═ 6/group) were immunized with 30 μ g or 100 μ g of BNT162b1 or with buffer IM on days 0 and 21, and sera were obtained before and 14, 21, 28, 35, and 42 days after immunization. At least 14 days after PCR confirmation and already at acute COVID-19 symptoms Human Convalescent Serum (HCS) (n 38) was obtained from SARS-CoV-2 infected patients at remission. The values above the bars give the geometric mean. a, geometric mean concentration of IgG binding to recombinant SARS-CoV-2 RBD (GMC). The dashed line represents the geometric mean (1.72U/mL) of sera from all time points in the placebo group. Statistical significance of group IgG titers at each time point to HCS samples was analyzed using one-way ANOVA with Dunnett's multiple comparison correction and at the 30 μ g dose level group (day 28, p)<0.0001; day 35, p ═ 0.0016) and 100 μ g dose level groups (days 28, 35, and 42, all p<0.0001) confirmed statistical significance. b, SARS-CoV-250% neutralization Titer (VNT)50). The dashed line represents the geometric mean (10.31U/mL) of sera from all time points in the placebo group. Analysis of group VNT at each time point with Dunnett's multiple comparison correction Using one-way ANOVA50Statistical significance for HCS samples, and at a 30 μ g dose level group (day 28, p)<0.0001) and 100 μ g dose level groups (days 28 and 35, p)<0.0001; p-0.007) on day 42).

FIG. 67: viral RNA in non-immunized and immunized rhesus monkeys after SARS-CoV-2 challenge

Rhesus monkeys (n-6/group) were immunized with 100 μ g BNT162b1 or buffer (control) on days 0 and 21 as described in figure 66. 41-48 days after the second immunization, animals were split 1X 10 equally between IN and IT routes6SARS-CoV-2 challenge of total pfu. 3 non-immunized age-matched male rhesus monkeys were challenged with cell culture medium (Sentinel). Viral RNA levels were detected by RT-qPCR. The ratio above the data points is the number of viral RNA positive animals in all animals per group. a, viral RNA in bronchoalveolar lavage fluid (BAL) obtained before challenge, and on days 3 and 6 after challenge. At day 6, the viral load between control and BNT162b1 immunized animals was statistically significant (p ═ 0.0131). b, viral RNA in nasal swabs obtained before challenge and on days 1, 3 and 6 after challenge. At day 3, the viral load between control and BNT162b1 immunized animals was statistically significant (p ═ 0.0229). The dotted line indicates the lower limit of detection (LLOD). Setting the negative sample1/2 for LLOD. The P-value of the binomial response was determined by classification analysis (undetectable viral load after challenge was successful and detectable viral load after challenge was failed).

FIG. 68: BNT162b1 and b 2V 8 reduce viral RNA in rhesus monkeys after challenge with SARS-CoV-2; b2 shows early clearance in the nose

FIG. 69: overview of exemplary pandemic supplies product packaging

FIG. 70: exemplary vaccine storage & handling at vaccination site

FIG. 71: exemplary Multi-dose articles

Figure 72 geometric mean titer and 95% CI: SARS-CoV-2 neutralization assay-NT 50-phase 1, 2 doses, 21 days-18-55 years-BNT 162b 1-evaluable immunogenic population

Figure 73 geometric mean titer and 95% CI: SARS-CoV-2 neutralization assay-NT 50-phase 1, 2 doses, 21 days-65-85 years-BNT 162b 1-evaluable immunogenic population

Figure 74 geometric mean titer and 95% CI: SARS-CoV-2 neutralization assay-NT 50-phase 1, 2 doses, 21 days-18-55 years-BNT 162b 2-evaluable immunogenic population

Figure 75 geometric mean titer and 95% CI: SARS-CoV-2 neutralization assay-NT 50-phase 1, 2 doses, 21 days-65-85 years-BNT 162b 2-evaluable immunogenic population

Figure 76 geometric mean concentration and 95% CI: determination of IgG levels binding to SARS-CoV-2 RBD phase 1, 2 doses, 21 days-18-55 years-BNT 162b 1-evaluable immunogenic population

Figure 77 geometric mean concentration and 95% CI: determination of IgG levels binding to SARS-CoV-2 RBD phase 1, 2 doses, 21 days apart-65-85 years old, BNT162b 1-evaluable immunogenic population

Figure 78 geometric mean concentration and 95% CI: IgG level assay binding to SARS-CoV-2S 1 phase 1, 2 doses, 21 days-18-55 years-BNT 162b 1-evaluable immunogenic population

FIG. 79 geometric mean concentration and 95% CI: IgG level assay binding to SARS-CoV-2S 1 phase 1, 2 doses, 21 days-65-85 years-BNT 162b 1-evaluable immunogenic population

Figure 80 geometric mean concentration and 95% CI: IgG level assay binding to SARS-CoV-2S 1 phase 1, 2 doses, 21 days-18-55 years-BNT 162b 2-evaluable immunogenic population

Figure 81 geometric mean concentration and 95% CI: IgG level assay binding to SARS-CoV-2S 1 phase 1, 2 doses, 21 days-65-85 years-BNT 162b 2-evaluable immunogenic population

Figure 82 geometric mean concentration and 95% CI: determination of IgG levels binding to SARS-CoV-2 RBD phase 1, 2 doses, 21 days-18-55 years-BNT 162b 2-evaluable immunogenic population

Figure 83 geometric mean concentration and 95% CI: determination of IgG levels binding to SARS-CoV-2 RBD phase 1, 2 doses, 21 days-65-85 years-BNT 162b 2-evaluable immunogenic population

FIG. 84. subjects who reported a local response with maximal severity within 7 days after each dose-phase 1, 2 doses, 21 days-18-55 years old-BNT 162b 1-safety population

FIG. 85 subjects who reported a local response with maximal severity in 7 days after each dose-phase 1, 2 doses, 21 days-65-85 years-BNT 162b 1-safety population

FIG. 86 subjects who reported a local response with maximal severity within 7 days after each dose-phase 1, 2 doses, 21 days-18-55 years old-BNT 162b 2-safety population

FIG. 87 subjects who reported a local response with maximal severity within 7 days after each dose-phase 1, 2 doses, 21 days-65-85 years-BNT 162b 2-safety population

Figure 88 subjects who reported systemic events with maximal severity within 7 days after each dose-phase 1, 2 doses, 21 days-18-55 years-BNT 162b 1-safety population

Figure 89 subjects who reported systemic events with maximal severity within 7 days after each dose-phase 1, 2 doses, 21 days-65-85 years-BNT 162b 1-safety population

Figure 90. subjects who reported systemic events with maximal severity within 7 days after each dose-phase 1, 2 doses, 21 days-18-55 years-BNT 162b 2-safety population

Figure 91 subjects who reported systemic events with maximal severity within 7 days after each dose-phase 1, 2 doses, 21 days-65-85 years-BNT 162b 2-safety population

FIG. 92. subjects reporting a regional response with maximal severity within 7 days after each dose, age group 18-55 years-2-safe population

FIG. 93. subjects reporting a regional response with maximal severity within 7 days after each dose, age group 56-85 years-2-safe population

FIG. 94 subjects reporting systemic events with maximal severity within 7 days after each dose, age group 18-55 years-2-safe population

Figure 95. subjects reporting systemic events with maximal severity within 7 days after each dose, age group 56-85 years-2-safe population

Figure 96 subjects reporting local reactions with maximal severity within 7 days after each dose, age group 1855 years-6000 subjects for 2/3 days-safety population

Figure 97 subjects reporting a local response with maximal severity within 7 days after each dose, age group 5685 years-for 2/3 days, -6000 subjects-safety population

Figure 98. subjects reporting systemic events with maximal severity within 7 days after each dose, age group 18-55 years-for period 2/3, -6000 subjects-safety population

Figure 99. subjects reporting systemic events with maximal severity within 7 days after each dose, age group 56-85 years-for period 2/3, -6000 subjects-safety population

Figure 100. cumulative incidence curve of first COVID-19 after dose 1-dose 1 all available efficacy population figure 101.BNT162b 2-exemplary functional 50% SARS-CoV-2 neutralizing antibody titer (VN)50)

Young adults (18-55 years) and older adults (56-85 years) were immunized with BNT162b2 on days 1 and 22 (n-12/group). Sera were obtained from young adults on day 1 (baseline) and on days 8, 22 (pre-boost), 29, 43, 50 and 85. Sera were obtained from older adults on day 1 (baseline) and on days 8, 22 and 29. Human COVID-19 convalescent serum (HSC, n 38) was obtained at least 14 days after confirmation and when the donor had no more symptoms. SARS-CoV-250% neutralization titers (VN) with 95% confidence intervals are shown for young adults immunized with 1, 3, 10, 20 or 30 μ g BNT162b2 and older adults immunized with 20 μ g BNT162b250Titer). Values below the limit of detection (LOD) were plotted as 0.5 LOD. Arrows indicate baseline (pre-dose 1, day 1) and dose 2 (day 22). The horizontal dashed line represents the LOD. VN 50SARS-CoV-2 neutralizing antibody titer 50%; HCS-human COVID-19 convalescent serum.

FIG. 102.BNT162b 1-neutralizing antibody titers at functional 50% SARS-CoV-2 (VN)50) Exemplary fold increase from baseline

Vaccination schedule and serum sampling were the same as in figure 39 (n-12/group). VN with 95% confidence intervals was shown for young participants (18-55 years) immunized with 1, 10, 30, 50 or 60 μ g BNT162b150Geometric mean fold increase from baseline in titer (GMFI). Arrows indicate baseline (before dose 1, day 1) and dose 2 (day 22). Dose 2 was not performed in the 60 μ g dose group. The dashed horizontal line indicates the threshold for seroconversion (fold increase ≧ 4). VN50SARS-CoV-2 neutralizing antibody titers were 50%.

FIG. 103.BNT162b 2-neutralizing antibody titer (VN) at functional 50% SARS-CoV-250) Exemplary fold increase from baseline

The vaccination schedule and serum sampling were the same as in figure 101. VNs with 95% confidence intervals were shown for (A) young participants (18-55 years) immunized with 1, 3, 10, 20, or 30 μ g BNT162B2, and (B) older participants (56-85 years) immunized with 20 μ g BNT162B250From baseline in titerWhich mean fold increase (GMFI). Arrows indicate baseline (pre-dose 1, day 1) and dose 2 (day 22). The dashed horizontal line indicates the threshold for serum turnover (fold increase ≧ 4). VN 50SARS-CoV-2 neutralizing antibody titers were 50%.

FIG. 104 exemplary frequencies of participants with SARS-CoV-2GMT seroconversion following immunization with BNT162b1

Vaccination schedule and serum sampling were the same as in figure 39 (n-12/group). For young participants (18-55 years) immunized with 1, 10, 30, 50 or 60 μ g BNT162b1, titration (VN) for 50% SARS-CoV-2 neutralizing antibodies was shown50) Seroconversion of (4). Seroconversion was defined as the minimum 4-fold increase in functional antibody response compared to baseline. Arrows indicate baseline (before dose 1, day 1) and dose 2 (day 22). Dose 2 was not performed in the 60 μ g dose group. GMT-geometric mean titer.

FIG. 105 exemplary frequencies of participants with SARS-CoV-2GMT seroconversion following immunization with BNT162b2

The vaccination schedule and serum sampling were the same as in figure 101. Neutralization antibody titers (VN) for 50% SARS-CoV-2 are shown for (A) young participants (18-55 years) dosed with 1, 3, 10, 20, or 30 μ g BNT162B2, and (B) older participants (56-85 years) dosed with 20 μ g BNT162B250) Seroconversion of (c). Seroconversion was defined as the minimum 4-fold increase in functional antibody response compared to baseline. Arrows indicate baseline (pre-dose 1, day 1) and dose 2 (day 22). GMT-geometric mean titer.

FIG. 106 exemplary fold increase from baseline in antibody concentration binding to S1 following immunization with BNT162b1

Vaccination schedule and serum sampling were the same as in figure 39 (n-12/group). For young participants (18-55 years) immunized with 1, 10, 30, 50, or 60 μ g BNT162b1, a geometric mean fold increase from baseline (GMFI) in antibody concentration bound to S1 with 95% confidence intervals was shown. Arrows indicate baseline (before dose 1, day 1) and dose 2 (day 22). Dose 2 was not performed in the 60 μ g dose group. The dashed horizontal line indicates the threshold for seroconversion (fold increase ≧ 4).

FIG. 107 exemplary fold increase from baseline in antibody concentration binding to S1 following immunization with BNT162b2

The vaccination schedule and serum sampling were the same as in figure 101. Geometric mean fold increase from baseline (GMFI) in antibody concentration bound to S1 with 95% confidence interval was shown for (a) young participants (18-55 years) immunized with 1, 3, 10, 20, or 30 μ g BNT162B2, and (B) older participants (56-85 years) immunized with 20 μ g BNT162B 2. Arrows indicate baseline (pre-dose 1, day 1) and dose 2 (day 22). Horizontal dotted line indicates the threshold for seroconversion (fold increase ≧ 4)

Figure 108 exemplary frequency of participants with IgG GMC seroconversion binding to S1 after immunization with BNT162b 1.

Vaccination schedule and serum sampling were the same as in figure 39 (n-12/group). Serum turnover with respect to antibody GMC binding to S1 was shown for young participants (18-55 years) immunized with 1, 10, 30, 50 or 60 μ g BNT162b 1. Seroconversion was defined as at least a 4-fold increase in IgG GMC response to S1 compared to baseline. Arrows indicate baseline (pre-dose 1, day 1) and dose 2 (day 22). Dose 2 was not performed in the 60 μ g dose group. GMC-geometric mean concentration.

FIG. 109 exemplary frequencies of participants with IgG GMC seroconversion binding to S1 following immunization with BNT162b2

The vaccination schedule and serum sampling were the same as in figure 101. Seroconversion for the antibody GMC binding to S1 was shown for (a) young participants (18-55 years) immunized with 1, 3, 10, 20 or 30 μ g BNT162B2, and (B) older participants (56-85 years) dosed with 20 μ g BNT162B 2. Seroconversion was defined as at least a 4-fold increase in IgG GMC response to S1 compared to baseline. Arrows indicate baseline (before dose 1, day 1) and dose 2 (day 22). GMC-geometric mean concentration.

FIG. 110S-specific CD4 from young participants immunized with BNT162b2+Exemplary results of cytokine production by T cells

Treatment with different doses of BNT162b2 was collected at baseline (before dose 1) and 29 days (+ -3 days) after dose 1Peripheral Blood Mononuclear Cell (PBMC) cell fractions isolated from blood of participants in the treatment were analyzed. Participants included young participants (age 18-55 years) dosed at 1 μ g (n-8), 3 μ g (n-9), 10 μ g (n-10), 20 μ g (n-9) or 30 μ g (n-10). The bar graph shows the arithmetic mean with 95% confidence intervals. By mixing all CD4 positive for IFN gamma, IL-2 or IL-4+Cytokine production was calculated by adding fractions of T cells, setting this sum to 100% and calculating the fraction of each subset that produced a particular cytokine. This analysis excluded 2 participants from the 1 μ g cohort, 1 participant from the 3 μ g cohort and 1 participant from the 10 μ g cohort (total cytokine producing CD 4)+T cell frequency<0.03%). IFN-interferon; IL ═ interleukin; young participants were 18-55 years old; SARS-CoV-2 spike protein.

FIG. 111S-specific CD4 from older participants immunized with BNT162b2 +Exemplary results of cytokine production by T cells

Peripheral Blood Mononuclear Cell (PBMC) cell fractions isolated from participant blood treated with different doses of BNT162b2 were collected at baseline (before dose 1) and 29 days after dose 1 (± 3 days) and analyzed. Participants included older participants (age 56-85 years) dosed at 10 μ g (n-11), 20 μ g (n-8) or 30 μ g (n-9). The bar graph shows the arithmetic mean with 95% CI. By mixing all CD4 positive for IFN gamma, IL-2 or IL-4+Cytokine production was calculated by adding fractions of T cells, setting this sum to 100% and calculating the fraction of each subset that produced a particular cytokine. This analysis excluded 6 participants from the 10 μ g cohort and 1 participant from the 20 μ g cohort (total cytokine producing CD 4)+T cell frequency<0.03%). IFN-interferon; IL ═ interleukin; elderly participants were 56-85 years old participants; SARS-CoV-2 spike protein.

FIG. 112 incidence and Scale of BNT162b 2-induced T cell response

At day 1 (pre-prime) and day 29 (7 days post-boost) (dose cohorts 1, 10 and 20 μg, each n is 9; 30 μ g, n ═ 10) of PBMC obtained enriched in CD4 +Or CD8+T cell effectors and stimulated overnight with 3 overlapping peptide pools (N-terminal pool S pool 1 and RBD, and C-terminal pool S pool 2) representing different portions of the wild-type sequence of SARS-CoV-2S, respectively, for evaluation in direct ex vivo IFN γ ELISpot. The common pathogen T cell epitope pools CEF (immunodominant HLA class I epitopes for CMV, EBV, influenza virus) and CEFT (immunodominant HLA class II epitopes for CMV, EBV, influenza virus, tetanus toxoid) were used as controls. Cell culture medium was used as a negative control. Each dot represents the normalized mean spot count of duplicate wells from one study participant after subtraction of the medium only control (a, c). a, antigen-specific CD4 for each dose cohort+And CD8+T cell response. The number of participants with detectable T cell responses at day 29 out of the total number of participants tested per dose cohort is provided. Spot count data from 2 participants in the 20 μ g dose cohort could not be normalized and were not plotted. b, CD4 of 30 μ g dose cohort participants+And CD8+An example of ELISpot. c, recognition of S-specific T cell responses in all participants of any S-peptide pool and their baseline CEFT and CEF-specific T cell responses. The horizontal bars represent median values.

FIG. 113 BNT162b 2-induced S-specific CD8+And CD4+T cells

CD4 of participants to be immunized+Or CD8+T cell effector-enriched fractions were stimulated overnight with two overlapping peptide pools (S pool 1 and S pool 2) covering wild-type SARS-CoV-2S for evaluation in direct ex vivo IFN γ ELISpot (a-c), the CD4+Or CD8+The T cell effector-enriched fractions were derived from PBMCs obtained from day 1 (pre-prime) and day 29 (7 days post-boost) (1, 10 and 20 μ g dose cohorts, each n ═ 9; 30 μ g dose cohort, n ═ 10). Each dot represents the normalized mean spot count of duplicate wells from one study participant after subtraction of the medium only control. The T cell responses of each participant to S pool 1 and S pool 2 were pooled. Spot count data from 2 participants in a 20 μ g dose cohort could not be normalizedAnd not plotted. PBMCs from vaccinated participants on day 29 of stimulation (7 days post boost) as described above (dose cohort 1 μ g, n-7; 10 and 30 μ g, n-10; 20 μ g, n-9) were analyzed by flow cytometry. a, S-specific CD4 for each dose cohort+And CD8+T cell response. The number of participants with detectable T cell responses at day 29 out of the total number of participants tested per dose cohort is provided. Participant with evaluable baseline data (for CD 4) +T cell response, n-34, for CD8+T cell response, n-37) was plotted against vaccine-induced responses of different parts of S. De novo induced or amplified responses were classified as those induced by BNT162b 2; a non-response or a pre-existing response that is not amplified by vaccination is classified as a non-vaccine response (none). c, intensity of response to S pool 1 in individuals with or without pre-existing response to S pool 2. Data from the 1 μ g dose cohort was excluded because there was no baseline response to S pool 2 in this dose cohort. Horizontal bars represent median of each group. d, cytokine-producing CD4 from participants vaccinated with 30 μ g BNT162b prime/boost+And CD8+Examples of pseudo-color flow cytometry plots of T cells. e, vaccine induced S-specific IFN γ+CD4+T cell pair IL4+CD4+Frequency of T cells. ICS stimulation was performed using peptide mixtures of S-pool 1 and S-pool 2. Each data point represents one study participant (1 μ g dose cohort, n-8; 20 μ g dose cohort, n-8; 10 and 30 μ g, each n-10). Exclude pre-existing CD4 from the 20 μ g dose queue that is strong for S pool 2+One participant in T cell responses. f, for 3 participants analyzed in FIG. 116, staining by pMHC class I multimers (CD 8) +Of+) ICS and ELISpot (CD 8)+Of (2) IFN gamma+) Defined antigen specificity CD8+T cell frequency. The signals of S pool 1 and S pool 2 are combined.

FIG. 114 correlation of antibodies and T-response

All participants of the prime/boost vaccination since day 29 (dose cohorts 1, 10, 2)0 and 30 μ g), data points of participants with no detectable T cell response were excluded from the correlation analysis (open circles; b, c). a, S1-specific IgG response vs S-specific CD4+Correlation of T cell responses. b, S specific CD4+And CD8+Correlation of T cell responses. c, S1-specific IgG response vs S-specific CD8+Correlation of T cell responses.

FIG. 115 cytokine polarization of T cells induced by BNT162b2

PBMCs obtained on day 1 (pre-prime) and day 29 (7 days post-boost) (dose cohorts 1 μ g, N-8; 10 and 30 μ g, each N-10; 20 μ g, N-9) and PBMCs from COVID-19 convalescent donors (HCS, N-18; c, d) were pooled with 3 overlapping peptides representing different parts of the SARS-CoV-2S wild-type sequence (N-terminal pool S pool 1[ aa 1-643)]And RBD [ aa1-16 fused to aa 327-528 of S]And C-terminal S cell 2[ aa 633-]) The stimulation was overnight and analyzed by flow cytometry. a cytokine-producing CD4 from 30 μ g dose cohort participants in response to S pool 1 +And CD8+Examples of pseudo-color flow cytometry plots of T cells. b, S-specific CD4 producing the indicated cytokines+T cells, S-specific CD4 for total cytokine production in response to S pool 1 and S pool 2+Fraction of T cells. Excluding CD4 non-responders (<0.03% total cytokine-producing T cells: 1 ug, n ═ 2 [ S pool 1]And n is 1[ S pool 2 ]](ii) a 10 μ g, n ═ 1). Arithmetic mean with 95% confidence interval. C, S-specific CD4 producing the indicated cytokines+(S pool 1, S pool 2 and RBD) and d, CD8+T cells (S pool 1, S pool 2 and RBD), account for the fraction of total circulating T cells of the same subset. The values above the data points represent the average fraction of each dose cohort. Participant PBMCs were tested as a single example (b-d).

FIG. 116 characterization of BNT162b 2-induced T cells at the single epitope level

PBMCs from 3 vaccination participants (dose cohort 10 μ g, n-1; 30 μ g, n-2) obtained on day 1 (pre-prime) and day 29 (7 days post-boost) were stained with pMHC class I multimer cocktail of individual (individual) and analyzed by flow cytometry for T-cell epitope specificity (a) and phenotype (b; example from participant 3; YLQPRTFLL). The peptide sequences above the dot plot indicate pMHC class I multimer epitope specificity, and the numbers above the dot plot indicate amino acids corresponding to the epitopes within S. c, localization of the identified MHC class I restricted epitope within S.

FIG. 117 ELISA screening assay of exemplary cohort sera to detect antibody response against the S1 domain of recombinant SARS-CoV-2 spike protein

Serum samples collected on day 10 after two immunizations (prime/boost on day 1 and 8) with BNT162c1, or day 17 after three doses of BNT162a1, BNT162b1, or BNT162b2 (prime/boost on day 1/8/15) were subjected to ELISA to analyze elicited antibody responses. Serum samples were tested against the S1 protein. By spanning 1: the points at serum dilution from 100 to 1:24,300 show the group mean Δ OD values for n-20 mice/group.

FIG. 118 ELISA screening assay of exemplary cohort sera to detect antibody response against the RBD domain of recombinant SARS-CoV-2 spike protein

ELISA was performed using serum samples collected at day 10 after two immunizations with BNT162c1 (prime/boost on day 1 and day 8), or day 17 after three doses of BNT162a1, BNT162b1, or BNT162b2 to analyze elicited antibody responses. Serum samples were tested against the RBD domain. Group mean Δ OD values for n-20 mice/group are shown by the points spanning serum dilutions from 1:100 to 1:24,300.

FIG. 119 is a drawing pVN50Exemplary cohort sera for titer pseudovirus neutralization Activity

Serum samples were collected either at day 10 (BNT162c1, saRNA) or day 17 (all other cohorts) after the first immunization of the animals and the titer of virus neutralizing antibodies was determined by a pseudovirus-based neutralization test (pVNT). The dots indicate that 50% pseudovirus neutralization was caused (pVN)50) Individual VNT titer of (a); group means are represented by horizontal bars (± SEM, standard error of mean).

FIG. 120 virus neutralizing antibodies and specific binding antibody responses to RBD and S1 in participants

RBD is a receptor binding domain. GMT-geometric mean titer. In the young adult group, serum samples were obtained before vaccination (day 1) and on days 8, 22, 29 and 43 after primary immunization, and in the older adult group, serum samples were obtained before vaccination (day 1) and on days 22, 29 and 43 after primary immunization. In covd-19 patients, a panel of human covd-19 recovery-stage sera was obtained at least 14 days after PCR confirmation (n-24). (A) SARS-CoV-2 neutralizes GMT of the antibody. (B) GMT of RBD binding antibody measured by ELISA. (C) GMT of ELISA antibody of S1. Each dot represents a serum sample and each vertical bar represents a geometric mean with 95% CI.

FIG. 121T cell responses in participants before and after vaccination as measured by IFN- γ ELISpot

IFN is interferon. PBMC-peripheral blood mononuclear cells. The S1 peptide pool covered the N-terminal half of the SARS-CoV-2 spike, including the RBD. The S2 peptide pool covered the C-terminal half of the SARS-CoV-2 spike, excluding the RBD. The CEF peptide pool consists of 32 MHC class I-restricted viral peptides from human cytomegalovirus, Epstein Barr virus and influenza virus. Panel A shows the number of IFN- γ secreting specific T cells on days 1, 29 and 43 in young participants at 18-55 years of age. Panel B shows the number of IFN- γ secreting specific T cells on days 1, 29 and 43 in 65-85 year old participants.

FIG. 122. 50% pseudovirus neutralization titer of 16 sera from BNT162b2 vaccine recipients against VSV-SARS-CoV-2-S pseudovirus comprising the spike protein of either Wuhan or lineage B.1.1.7

Each N-8 representative sera from young adults (18-55 years old; indicated by triangles) and older adults (56-85 years old; indicated by circles) extracted on day 43 (21 days after dose 2) was tested.

FIG. 123 is a schematic representation of the production of VSV pseudoviruses comprising SARS-CoV-2S protein. (1) The SARS-CoV-2-S expression plasmid was transfected into HEK293/T17 cells. (2) Cells expressing SARS-CoV-2S are infected with a VSV-G supplemented import virus (VSV Δ G) lacking VSV-G in its genome and encoding a reporter gene. (3) Neutralizing residual VSV-G supplemented input virus by adding anti-VSV-G antibody, producing SARS-CoV-2S pseudotyped VSV Δ G as a replacement for live SARS-CoV-2.

FIG. 124. titration of SARS-CoV-2 Wuhan reference strain and lineage B.1.1.7 spike-pseudotyped VSV on Vero 76 cells using GFP infected cells as a readout.

Figure 125 protocol for BNT162b2 vaccination and serum sampling.

FIG. 126 pVNT between SARS-CoV-2 lineage B.1.1.7 and Wuhan reference strain spike-pseudotyped VSV50A graph of the ratio. Triangles represent serum from young adults (18-55 years) and circles represent serum from older adults (56-85 years). Serum was drawn on day 43 (21 days after dose 2).

FIG. 127 50% pseudovirus neutralization titer of 12 sera from BNT162b2 vaccine recipients against VSV-SARS-CoV-2-S pseudovirus comprising the Wuhan Hu-1 reference, lineage B.1.1.298, or lineage B.1.351 spike protein (pVNT50)

N-12 sera from young adults immunized with 30 μ g BNT162b2, drawn on day 29 or day 43 (7 or 21 days after dose 2) were tested. Geometric mean titers are shown. Statistical significance of differences between wuhan Hu-1 reference pseudoviruses and either lineage b.1.1.298 or lineage b.1.351 pseudoviruses was calculated by Wilcoxon paired signed rank test. Two-tailed p-values are reported. ns, not significant; p < 0.001; LLOQ, lower limit of quantitation.

FIG. 128 50% plaque reduction neutralization titers against N501 and Y501 SARS-CoV-2 from 20 sera of BNT162b2 vaccine recipients

7 sera (represented by triangles) were drawn 2 weeks after the second dose of vaccine; 13 sera (indicated by circles) were drawn 4 weeks after the second dose.

FIG. 129. FIG. N501Y substituted graph. An L-leader sequence; ORF-open reading frame; an RBD-receptor binding domain; (ii) S-spike glycoprotein; an N-terminal furin cleavage fragment of S1-S; a C-terminal furin enzyme cleaved fragment of S2-S; e-envelope protein; an M-membrane protein; an N-nucleoprotein; UTR-untranslated region.

FIG. 130 plaque morphology on Vero E6 cells of N501 and Y501 SARS-CoV-2.

Figure 131.BNT162 vaccination and serum sampling protocol.

FIG. 132 PRNT between Y501 and N501 viruses50A graph of the ratio. Triangles represent sera drawn 2 weeks after the second dose; circles represent serum drawn 4 weeks after the second dose.

FIG. 133 engineered mutations

Nucleotide and amino acid positions are shown. The absence is shown by the dashed line. The mutant nucleotide was red. L, leader sequence; ORF, open reading frame; RBD, receptor binding domain; s, spike glycoprotein; s1, N-terminal furin cleavage fragment of S; s2, a C-terminal furin cleavage fragment of S; e, envelope protein; m, membrane protein; n, nucleoprotein; UTR, untranslated region.

FIG. 134, plaque morphology of WT (USA-WA1/2020), mutant N501Y, Δ 69/70+ N501Y + D614G and E484K + N501Y + D614G SARS-CoV-2s on Vero E6 cells

FIG. 135.BNT162 Vaccination and serum sampling protocol

FIG. 136.20 PRNT of BNT162b2 vaccinated human sera against Wild Type (WT) and mutant SARS-CoV-250

(a) WT (USA-WA1/2020) and mutant N501Y. (b) WT and Δ 69/70+ N501Y + D614G. (c) WT and E484K + N501Y + D614G. 7 (triangle) and 13 (circle) sera were withdrawn 2 and 4 weeks after the second dose of vaccination, respectively. Will have different PRNT to WT and mutant virus50The sera of (a) were linked by a line. (a) The results in (a) were from one experiment; (b) the results in (a) and (c) were from another set of experiments. Each data point is the average of duplicate determinations.

FIG. 137 ratio of neutralizing GMT against mutant virus to GMT against WT virus

Triangles represent sera drawn 2 weeks after the second dose of vaccination; circles represent sera drawn 4 weeks after the second dose of vaccination.

FIG. 138 schematic representation of engineered spike substitutions and deletions

In this study, the genome and sequence of clinical isolate USA-WA1/2020 WAs used as the wild-type virus. There are mutations from the british b.1.1.7, brazil p.1 and south africa b.1.351 lineages. The absence is shown by the dashed line. The mutated nucleotide is red. Nucleotide and amino acid positions are shown. An L-leader sequence; ORF-open reading frame; an RBD-receptor binding domain; (ii) S-spike glycoprotein; an N-terminal furin (furin) cleavage fragment of S1-S; a C-terminal furin cleavage fragment of S2-S; e-envelope protein; an M-membrane protein; an N-nucleoprotein; UTR-untranslated region.

FIG. 139, USA-WA1/2020 and mutant SARS-CoV-2 plaque morphology

Plaque assays were performed on Vero E6 cells in 6-well plates.

FIG. 140.BNT162 immunization and serum sampling protocol

FIG. 141 serum neutralization of variant strains of SARS-CoV-2 after a second dose of BNT162b2 vaccine

Results of a 50% plaque reduction neutralization test (PRNT50) using 20 samples obtained from 15 test participants are shown 2 weeks (circles) or 4 weeks (triangles) after administration of the second dose of BNT162b2 vaccine. Mutant viruses were obtained by designing a full set of mutations in the B.1.1.7, P.1. or B.1.351 lineage or a subset of the mutations in the S gene in the B.1.351 lineage (B.1.351-. DELTA.242-244 + D614G and B.1.351-RBD-614G) into USA-WA 1/2020. Each data point represents the geometric mean PRNT50 obtained with serum samples for the virus shown, including data from replicate experiments, as detailed in table 31. Data in USA-WA1/2020 were from three experiments; data from one experiment for each of the B.1.1.7 spike, B.1.351-. DELTA.242-244 + D614G, and B.1.351-RBD-D614G viruses; data were from two experiments for each of the p.1 spike and b.1.351 spike viruses. In each experiment, neutralization titers were determined in duplicate assays, and geometric means were taken. LOD: the limit of detection.

FIG. 142 persistence of T cell response induced by BNT162b2

PBMCs obtained on day 1 (pre-prime), day 29, day 85 and day 184 (7, 9 and 23 weeks post boost, respectively) were analyzed in ex vivo IFN γ ELISpot (see GA-RB-022-01A for details). Common pathogen T cell epitope pools CEF (CMV, EBV and influenza virus HLA class I epitopes) and CEFT (CMV, EBV, influenza virus and tetanus toxoid HLA class II epitopes) were used to evaluate general T cell reactivity, and the medium was used as a negative control. Each dot represents the sum of the normalized mean spot counts of duplicate wells from one study subject stimulated with two peptide pools corresponding to full-length wt S protein after subtraction of the medium-only control. The proportion above the data points after vaccination is the number of subjects with detectable CD4+ or CD8+ T cell responses within the total number of test subjects per dose cohort and time point.

FIG. 143. in a dual IHC-ISH assay, specific vaccine mRNA signals (red) were detected in LN 6 hours post injection using the modV9 probe. The vaccine was mainly localized to subcapsular sinus (LN in positions 9 and 5) and B cell follicles (LN in positions 12 and 1). Dendritic cells were visualized by CD11c staining (blue-green, upper panel) and only some of them absorbed the vaccine. Most CD169+ macrophages (subcapsular sinus lymphocytes macrophages, blue-green, middle panel) were positive for the vaccine. B cells (CD19+, blue-green, lower panel) were the second major population exhibiting vaccine signaling.

FIG. 144. in a dual IHC-ISH assay, a specific vaccine mRNA signal (red) was detected in the spleen 6 hours post injection using the modV9 probe. The major vaccine signal was detected in the white marrow. Dendritic cells were visualized by CD11c staining (blue-green, upper panel) and only some of them absorbed vaccine. A small fraction of F4/80+ macrophages (blue-green, middle panel) absorbed the vaccine. B cells (CD19+, blue-green, lower panel) are the predominant population exhibiting vaccine signaling.

FIG. 145. exemplary stability data

Exemplary data from certain stability studies (see, e.g., example 42) show that BNT162b2 LNP preparations at the indicated concentration and temperature conditions were evaluated by ELISA to characterize the reactivity of the antibodies to S1 spike protein.

DESCRIPTION OF THE SEQUENCES

The following table provides a list of certain sequences referenced herein.

Detailed Description

Although the present disclosure is described in detail below, it is to be understood that this disclosure is not limited to the particular methodology, protocols, and reagents described herein as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present disclosure, which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art.

Preferably, terms used herein such as "A multilingual collaboration of biological entities" (IUPAC Recommendations) ", H.G.W.Leuenberger, B.Nagel, and H.Eds., Helvetica Chimica Acta, CH-4010Basel, Switzerland, (1995).

Unless otherwise indicated, practice of the present disclosure will employ conventional methods of chemistry, biochemistry, cell biology, immunology and recombinant DNA techniques, which are explained in the literature of the art (see, e.g., Molecular Cloning: A Laboratory Manual, 2)nd Edition,J.Sambrook et al.eds.,Cold Spring Harbor Laboratory Press,Cold Spring Harbor 1989)。

Elements of the present disclosure are described below. These elements are listed with particular embodiments, however, it should be understood that they may be combined in any manner and in any number to produce additional embodiments. The foregoing examples and embodiments should not be construed as limiting the disclosure to only the explicitly described embodiments. This specification should be understood to disclose and encompass embodiments combining the explicitly described embodiments with any number of the disclosed elements. Moreover, any arrangement or combination of all described elements is deemed to be disclosed by the present specification unless otherwise indicated by the context.

The term "about" means approximately or nearly, and in one embodiment means plus or minus 20%, + -10%, + -5% or + -3% of the recited or claimed value or range above or below the value or range shown herein.

The use of the terms "a" and "an" and "the" and similar referents in the context of describing the disclosure (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein are 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 were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the disclosure and does not pose a limitation on the scope of the claims. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the disclosure.

Unless expressly stated otherwise, the term "comprising" is used in the context of this document to indicate that, in addition to the list members introduced by "comprising," other members may optionally be present. However, as a specific embodiment of this disclosure, the term "comprising" is contemplated to encompass the possibility that no other member is present, i.e., for the purposes of this embodiment, "comprising" may be understood to have the meaning of "consisting of …" or "consisting essentially of …".

Several documents are cited throughout the text of this specification. Each document cited herein (including all patents, patent applications, scientific publications, manufacturer's specifications, instructions, etc.), whether supra or infra, is hereby incorporated by reference in its entirety. Nothing herein is to be construed as an admission that the present disclosure is not entitled to antedate such disclosure by virtue of prior disclosure.

Definition of

In the following, definitions will be provided that apply to all aspects of the present disclosure. Unless otherwise indicated, the following terms have the following meanings. Any undefined terms have their accepted meanings.

As used herein, terms such as "reduce", "inhibit" or "impair" relate to the ability to generally reduce or cause a general reduction, preferably by at least 5%, at least 10%, at least 20%, at least 50%, at least 75% or even more in level. These terms include complete or substantially complete inhibition, i.e., reduction to 0 or substantially to 0.

Terms such as "increase", "enhance" or "over" preferably relate to an increase or enhancement of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 80%, at least 100%, at least 200%, at least 500% or even more.

According to the present disclosure, the term "peptide" encompasses oligopeptides and polypeptides, and refers to a substance comprising about 2 or more, about 3 or more, about 4 or more, about 6 or more, about 8 or more, about 10 or more, about 13 or more, about 16 or more, about 20 or more, and up to about 50, about 100, or about 150 consecutive amino acids linked to each other by peptide bonds. The term "protein" or "polypeptide" refers to a large peptide, particularly a peptide having at least about 150 amino acids, although the terms "peptide," "protein," and "polypeptide" are generally used synonymously herein.

A "therapeutic protein" has a positive or beneficial effect on the condition or disease state of a subject when provided to the subject in a therapeutically effective amount. In one embodiment, the therapeutic protein has therapeutic or palliative properties and can be administered to improve, reduce, alleviate, reverse, delay the onset of, or reduce the severity of one or more symptoms of a disease or disorder. Therapeutic proteins may have prophylactic properties and may be used to delay the onset of a disease or to reduce the severity of such a disease or pathological condition. The term "therapeutic protein" includes intact proteins or peptides, and may also refer to therapeutically active fragments thereof. It may also include therapeutically active variants of the protein. Examples of therapeutically active proteins include, but are not limited to, antigens for vaccination and immunostimulants such as cytokines.

With respect to amino acid sequences (peptides or proteins), "fragments" refer to a portion of an amino acid sequence, i.e., a sequence representing an amino acid sequence that is shortened at the N-terminus and/or C-terminus. A fragment shortened at the C-terminus (N-terminal fragment) is obtainable, for example, by translation of a truncated open reading frame lacking the 3' end of the open reading frame. A shortened segment at the N-terminus (C-terminal segment) is obtainable, for example, by translation of a truncated open reading frame lacking the 5' end of the open reading frame, provided that the truncated open reading frame comprises a start codon for initiation of translation. Fragments of an amino acid sequence comprise, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% of the amino acid residues from the amino acid sequence. A fragment of an amino acid sequence preferably comprises at least 6, in particular at least 8, at least 12, at least 15, at least 20, at least 30, at least 50 or at least 100 consecutive amino acids from the amino acid sequence.

"variant" herein means an amino acid sequence that differs from a parent amino acid sequence due to at least one amino acid modification. The parent amino acid sequence may be a naturally occurring or Wild Type (WT) amino acid sequence, or may be a modified form of a wild type amino acid. Preferably, the variant amino acid sequence has at least one amino acid modification as compared to the parent amino acid sequence, e.g., from 1 to about 20 amino acid modifications as compared to the parent, and preferably from 1 to about 10 or from 1 to about 5 amino acid modifications.

"wild-type" or "WT" or "native" herein refers to an amino acid sequence found in nature, including allelic variations. The wild-type amino acid sequence, peptide or protein has a fragment of the amino acid sequence which has not been deliberately modified.

For the purposes of the present disclosure, "variants" of an amino acid sequence (peptide, protein, or polypeptide) include amino acid insertion variants, amino acid addition variants, amino acid deletion variants, and/or amino acid substitution variants. The term "variant" includes all mutants, splice variants, post-translationally modified variants, conformations, isoforms, allelic variants, species variants and species homologues, in particular those occurring in nature. In particular, the term "variant" includes amino acid sequences.

Amino acid insertion variants include the insertion of a single or two or more amino acids in a particular amino acid sequence. In the case of amino acid sequence variants having insertions, one or more amino acid residues are inserted at specific positions in the amino acid sequence, however random insertions are also possible with appropriate screening of the resulting product. Amino acid addition variants comprise amino-and/or carboxy-terminal fusions of one or more amino acids, such as 1, 2, 3, 5, 10, 20, 30, 50 or more amino acids. Amino acid deletion variants are characterized by the removal of one or more amino acids from the sequence, such as the removal of 1, 2, 3, 5, 10, 20, 30, 50, or more amino acids. The deletion may be at any position of the protein. Amino acid deletion variants that contain deletions at the N-terminus and/or C-terminus of the protein are also referred to as N-terminal and/or C-terminal truncated variants. Amino acid substitution variants are characterized by the removal of at least one residue in the sequence and the insertion of another residue at its position. Modifications in amino acid sequence positions that are not conserved between homologous proteins or peptides and/or the replacement of amino acids with other amino acids having similar properties are preferred. Preferably, the amino acid changes in peptide and protein variants are conservative amino acid changes, i.e., substitutions of similarly charged or uncharged amino acids. Conservative amino acid changes include substitutions in one of the related families of amino acids in their side chains. Naturally occurring amino acids are generally divided into 4 families: acidic (aspartic acid, glutamic acid), basic (lysine, arginine, histidine), nonpolar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), and uncharged polar (glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine) amino acids. Phenylalanine, tryptophan, and tyrosine are sometimes collectively classified as aromatic amino acids. In one embodiment, conservative amino acid substitutions involve substitutions within the following groups:

Glycine, alanine;

valine, isoleucine, leucine;

aspartic acid, glutamic acid;

asparagine, glutamine;

serine, threonine;

lysine, arginine; and

phenylalanine, tyrosine.

Preferably, the degree of similarity, preferably the degree of identity, between a given amino acid sequence and the amino acid sequence of a variant of said given amino acid sequence is at least about 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%. Preferably, the degree of similarity or identity is given to a region of amino acids that is at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or about 100% of the entire length of the reference amino acid sequence. For example, if a reference amino acid sequence consists of 200 amino acids, it is preferred to give a degree of similarity or identity, in some embodiments, consecutive amino acids, to at least about 20, at least about 40, at least about 60, at least about 80, at least about 100, at least about 120, at least about 140, at least about 160, at least about 180, or about 200 amino acids. In some embodiments, the entire length of a reference amino acid sequence is given a degree of similarity or identity. Alignments to determine sequence similarity, preferably sequence identity, preferably using optimal sequence alignments, e.g., using Align, using standard settings, preferably EMBOSS: needle, Matrix, Blosum62, Gap Open 10.0, Gap extended 0.5.

"sequence similarity" refers to the percentage of amino acids that are identical or represent conservative amino acid substitutions. "sequence identity" between two amino acid sequences means the percentage of amino acids that are identical between the sequences. "sequence identity" between two nucleic acid sequences means the percentage of nucleotides that are identical between the sequences.

In particular, the terms "% identity", "% identity" or similar terms refer to the percentage of nucleotides or amino acids that are identical in the best alignment between the sequences to be compared. The percentages are purely statistical and the differences between the two sequences may, but need not necessarily, be randomly distributed over the entire length of the sequences to be compared. Usually after optimal alignment of the segments or "comparison windows", a comparison of the two sequences is carried out by comparing the sequences in order to identify local regions of the corresponding sequences. The optimal alignment for comparison can be performed manually or by means of the local homology algorithm of Smith and Waterman,1981, Ads App.Math.2,482, by means of the local homology algorithm of Neddleman and Wunsch,1970, J.mol.biol.48,443, by means of the similarity search method of Pearson and Lipman,1988, Proc.Natl Acad.Sci.USA 88,2444, or by means of a Computer program using said algorithms (Wisconsin Genetics Software Package, Genetics Computer Group,575Science Drive, Madison, GAP in Wis, BESTFIT, FASTA, BLAST P, BLAST N and TFASTA). In some embodiments, percent identity of two sequences is determined using the BLASTN or BLASTP algorithm, which can be obtained at the National Center for Biotechnology Information (NCBI) website (e.g., at BLAST. NCBI. nlm. nih. gov/BLAST. cgippage _ TYPE ═ BLAST search & BLAST _ SPEC ═ BLAST2seq & LIN _ LOC ═ align2 seq). In some embodiments, the algorithm parameters for the BLASTN algorithm on the NCBI website include: (i) the desired threshold is set to 10; (ii) the word size is set to 28; (iii) the maximum match in the query range is set to 0; (iv) match/mismatch scores are set to 1, -2; (v) the notch cost is set to be linear; and (vi) a filter for low complexity regions. In some embodiments, the algorithm parameters for the BLASTP algorithm on the NCBI website include: (i) the desired threshold is set to 10; (ii) the word length is set to 3; (iii) the maximum match in the query range is set to 0; (iv) the matrix is set to BLOSUM 62; (v) the notch cost is set to exist: 11, extension: 1; and (vi) conditional component scoring matrix adjustment.

Percent identity is obtained by determining the number of identical positions corresponding to the sequences to be compared, 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 embodiments, a degree of similarity or identity is given to a region of at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or about 100% of the entire length of a reference sequence. For example, if a reference nucleic acid sequence consists of 200 nucleotides, the degree of identity is given for at least about 100, at least about 120, at least about 140, at least about 160, at least about 180, or about 200 nucleotides, and in some embodiments, consecutive nucleotides. In some embodiments, the entire length of the reference sequence is given a degree of similarity or identity.

According to the present disclosure, homologous amino acid sequences exhibit at least 40%, in particular at least 50%, at least 60%, at least 70%, at least 80%, at least 90% and preferably at least 95%, at least 98 or at least 99% identity of amino acid residues.

The skilled person can readily prepare amino acid sequence variants described herein, e.g. by recombinant DNA manipulation. For example, procedures for preparing a DNA sequence having a peptide or protein having a substitution, addition, insertion or deletion are described in detail in Sambrook et al (1989). In addition, the peptides and amino acid variants described herein can be readily prepared by known peptide synthesis techniques, e.g., by solid phase synthesis and similar methods.

In one embodiment, a fragment or variant of an amino acid sequence (peptide or protein) is preferably a "functional fragment" or a "functional variant". The term "functional fragment" or "functional variant" of an amino acid sequence refers to any fragment or variant that exhibits one or more functional properties that are the same as or similar to the amino acid sequence from which it is derived, i.e., it is functionally equivalent. With respect to an antigen or antigenic sequence, one particular function is one or more immunogenic activities exhibited by the amino acid sequence from which the fragment or variant is derived. As used herein, the term "functional fragment" or "functional variant" refers in particular to a variant molecule or sequence comprising 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 still be capable of performing one or more functions of the parent molecule or sequence, e.g., inducing an immune response. In one embodiment, the modification in the amino acid sequence of the parent molecule or sequence does not significantly affect or alter the characteristics of the molecule or sequence. In various embodiments, the function of a functional fragment or functional variant may be reduced but still significantly present, e.g., the immunogenicity of a functional variant may be at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% of the parent molecule or sequence. However, in other embodiments, the immunogenicity of the functional fragment or functional variant may be enhanced as compared to the parent molecule or sequence.

An amino acid sequence (peptide, protein, or polypeptide) "derived from" a given amino acid sequence (peptide, protein, or polypeptide) refers to the source of the first amino acid sequence. Preferably, the amino acid sequence derived from a particular amino acid sequence has an amino acid sequence that is identical, substantially identical, or homologous to the particular amino acid sequence or fragment thereof. An amino acid sequence derived from a particular amino acid sequence may be a variant of that particular sequence or a fragment thereof. For example, one of ordinary skill in the art will appreciate that antigens suitable for use herein can be altered such that their sequence differs from the naturally occurring sequence or native sequence from which they are derived, while retaining the desirable activity of the native sequence.

As used herein, "instructional material" or "instructions" includes a publication, a record, a diagram, or any other medium of expression that can be used to convey the usefulness of the compositions and methods of the present invention. The instructional material of the kit of the invention may, for example, be affixed to the container containing the composition of the invention or shipped together with the container containing the composition. Alternatively, the instructional material may be shipped separately from the container for the purpose of instructing the material and the compound to be used cooperatively by the recipient.

"isolated" means changed or removed from a natural state. For example, a nucleic acid or peptide naturally occurring in a living animal is not "isolated," but the same nucleic acid or peptide partially or completely separated from the coexisting materials of its natural state is "isolated. An isolated nucleic acid or protein may exist in a substantially pure form, or may exist in a non-natural environment, such as a host cell.

The term "recombinant" in the context of the present invention means prepared by "genetic engineering". Preferably, a "recombinant object" such as a recombinant nucleic acid is not naturally occurring in the context of the present invention.

The term "naturally occurring" as used herein refers to the fact that an object can be found in nature. For example, a peptide or nucleic acid that is present in an organism (including viruses) and that can be isolated from a natural source and that has not been intentionally modified by man in the laboratory is naturally occurring.

As used herein, "physiological pH" refers to a pH of about 7.5.

The term "genetically modified" or simply "modifying" includes transfecting a cell with a nucleic acid. The term "transfection" relates to the introduction of nucleic acids, in particular RNA, into cells. For the purposes of the present invention, the term "transfection" also includes the introduction of nucleic acids into cells or the uptake of nucleic acids by such cells, wherein the cells may be present in a subject, e.g., a patient. Thus, according to the present invention, the cells used to transfect the nucleic acids described herein may be present in vitro or in vivo, e.g., the cells may form part of an organ, a tissue, and/or an organism of a patient. According to the invention, transfection may be transient or stable. For some applications of transfection, it is sufficient if the transfected genetic material is only transiently expressed. The RNA can be transfected into cells to transiently express the protein it encodes. Because the nucleic acid introduced during transfection is not normally integrated into the nuclear genome, exogenous nucleic acid is diluted or degraded through mitosis. Cells that allow free expansion of nucleic acids greatly reduce the dilution rate. If it is desired that the transfected nucleic acid actually remains in the genome of the cell and its daughter cells, stable transfection must be performed. Such stable transfection can be achieved by transfection using a virus-based system or a transposon-based system. Typically, a nucleic acid encoding an antigen is transiently transfected into a cell. The RNA can be transfected into cells to transiently express the protein it encodes.

The term "seroconversion" includes a > 4-fold increase from before vaccination to 1 month after dose 2.

Coronavirus (coronavirus)

Coronaviruses are enveloped, positive-sense, single-stranded RNA ((+) ssRNA) viruses. They have the largest genome among known RNA viruses (26-32 kb) and are phylogenetically divided into 4 genera (α, β, γ and δ), while β coronaviruses are further subdivided into 4 lineages (A, B, C and D). Coronaviruses infect a wide range of avian and mammalian species, including humans. Some human coronaviruses generally cause mild respiratory disease, although the severity may be higher in infants, the elderly, and immunocompromised. The middle east respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus (SARS-CoV), belonging to beta coronavirus lineages C and B, respectively, are highly pathogenic. Both viruses have entered the human population from animal hosts within the last 15 years and have resulted in outbreaks of high mortality. Since 12 months of 2019, outbreaks of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) causing atypical pneumonia (coronavirus disease in 2019; COVID-19) have persisted in china and have developed into emergent public health events of international concern. SARS-CoV-2(MN908947.3) belongs to the beta coronavirus lineage B. It has at least 70% sequence similarity to SARS-CoV.

Generally, coronaviruses have 4 structural proteins, i.e., envelope (E), membrane (M), nucleocapsid (N), and spike (S). The E and M proteins have important functions in viral assembly, while the N protein is essential for viral RNA synthesis. The key glycoprotein S is responsible for viral binding and entry into target cells. The S protein is synthesized as a single-chain inactive precursor and cleaved by furin-like host proteases into two non-covalently associated subunits S1 and S2 in the producing cell. The S1 subunit contains a Receptor Binding Domain (RBD), which recognizes a host cell receptor. The S2 subunit comprises a fusion peptide, two heptad repeats, and a transmembrane domain, all of which are required to mediate fusion of viral and host cell membranes by undergoing large conformational rearrangements. Trimerization of the S1 and S2 subunits forms large prefusion spikes.

The S precursor protein of SARS-CoV-2 can be proteolytically cleaved into S1(685aa) and S2(588aa) subunits. The S1 subunit consists of a Receptor Binding Domain (RBD) that mediates entry of the virus into sensitive cells via the host angiotensin converting enzyme 2 (ACE2) receptor.

Antigens

The present invention includes the use of RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof. Thus, the RNA encodes a peptide or protein comprising at least the epitope SARS-CoV-2S protein or an immunogenic variant thereof for use in inducing an immune response in a subject against a coronavirus S protein, particularly a SARS-CoV-2S protein. The amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment (i.e., antigenic peptide or protein) of said SARS-CoV-2S protein or immunogenic variant thereof is also designated herein as a "vaccine antigen", "peptide and protein antigen", "antigenic molecule", or simply "antigen". The SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof is also designated herein as an "antigenic peptide or protein" or "antigenic sequence".

The SARS-CoV-2 coronavirus full-length spike (S) protein consists of 1273 amino acids (see SEQ ID NO: 1). In a specific embodiment, the full length spike (S) protein according to SEQ ID NO:1 is modified in such a way that the prototypic prefusion conformation is stabilized. Stabilization of the prefusion conformation can be achieved by introducing two consecutive substitutions of prolines at residues 986 and 987 of the AS in the full length spike protein. Specifically, a spike (S) protein-stabilized protein variant was obtained in such a manner that the amino acid residue at position 986 was exchanged for proline, and the amino acid residue at position 987 was also exchanged for proline. In one embodiment, the SARS-CoV-2S protein variant comprises the amino acid sequence of SEQ ID NO: 7.

In one embodiment, the vaccine antigens described herein comprise, consist essentially of, or consist of the spike protein (S) of SARS-CoV-2, a variant thereof, or a fragment thereof, of SARS-CoV-2.

In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 17-1273 of SEQ ID NO. 1 or 7, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273 of SEQ ID NO. 1 or 7, or an immunogenic fragment of the amino acid sequence of amino acids 17-1273 of SEQ ID NO. 1 or 7 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273 of SEQ ID NO. 1 or 7. In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 17-1273 of SEQ ID NO 1 or 7.

In one embodiment, the rna encoding the vaccine antigen (i) comprises a nucleotide sequence of nucleotides 49-3819 of SEQ ID No. 2, 8 or 9, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 49-3819 of SEQ ID No. 2, 8 or 9, or a fragment of the nucleotide sequence of nucleotides 49-3819 of SEQ ID No. 2, 8 or 9 or a fragment of the nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 49-3819 of SEQ ID No. 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 17-1273 of SEQ ID NO:1 or 7, which is identical to the amino acid sequence of SEQ ID NO:1 or 7, or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273 of SEQ ID NO:1 or 7 or an immunogenic fragment of an amino acid sequence of amino acids 17-1273 that is identical to SEQ ID NO:1 or 7, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273. In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO: 2. 8 or 9 from nucleotide 49 to 3819; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 17-1273 of SEQ ID NO:1 or 7.

In one embodiment, the vaccine antigen comprises, consists essentially of, or consists of a SARS-CoV-2 spike S1 fragment (S1), a variant thereof, or a fragment thereof (S1) (S1 subunit of the spike protein (S) of SARS-CoV-2), a variant thereof, or a fragment thereof, or a SARS-CoV-2 spike S1 fragment (S1), a variant thereof, or a fragment thereof.

In one embodiment, the vaccine antigen comprises SEQ ID NO:1, amino acid sequence corresponding to amino acids 17-683 of SEQ ID NO:1, or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-683 of SEQ ID NO:1 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-683 of SEQ ID No. 1. In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 17-683 of SEQ ID NO. 1.

In one embodiment, the rna encoding the vaccine antigen (i) comprises a nucleotide sequence of nucleotides 49 to 2049 of SEQ ID No. 2, 8 or 9, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 49 to 2049 of SEQ ID No. 2, 8 or 9, or a fragment of a nucleotide sequence of nucleotides 49 to 2049 of SEQ ID No. 2, 8 or 9 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 49 to 2049 of SEQ ID No. 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 17-683 of SEQ ID NO:1, identical to SEQ ID NO:1, or an immunogenic fragment of an amino acid sequence of amino acids 17-683 of SEQ ID No. 1, or an amino acid sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identical to an amino acid sequence of amino acids 17-683 of SEQ ID No. 1. In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of nucleotides 49-2049 of SEQ ID NO 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO:1, amino acid sequence of amino acids 17-683.

In one embodiment, the vaccine antigen comprises SEQ ID NO:1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-685 of SEQ ID No. 1, or an immunogenic fragment of the amino acid sequence of amino acids 17-685 of SEQ ID No. 1 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-685 of SEQ ID No. 1. In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 17-685 of SEQ ID NO. 1.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO: 2. 8 or 9, nucleotide sequence from nucleotides 49 to 2055 of SEQ ID NO: 2. 8 or 9, nucleotides 49 to 2055, or a fragment of a nucleotide sequence of nucleotides 49 to 2055 of SEQ ID NO. 2, 8 or 9 or a nucleotide sequence of nucleotides at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to a nucleotide sequence of nucleotides 49 to 2055 of SEQ ID NO. 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:1, amino acid sequence corresponding to amino acids 17-685 of SEQ ID NO:1, or an immunogenic fragment of an amino acid sequence of amino acids 17-685 of SEQ ID No. 1 or an amino acid sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to an amino acid sequence of amino acids 17-685 of SEQ ID No. 1. In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO: 2. a nucleotide sequence of nucleotides 49 to 2055 of 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO:1, amino acid sequence of amino acids 17-685.

In one embodiment, the vaccine antigen comprises, consists essentially of, or consists of the Receptor Binding Domain (RBD) of the S1 subunit of the spike protein (S) of SARS-CoV-2, a variant thereof, or a fragment thereof, or the Receptor Binding Domain (RBD) of the S1 subunit of the spike protein (S) of SARS-CoV-2, a variant thereof, or a fragment thereof. The amino acid sequence of amino acid 327-528 of SEQ ID NO:1, variants thereof or fragments thereof are also referred to herein as "RBD" or "RBD domain".

In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acid 327-528 of SEQ ID NO:1, which is identical to the amino acid sequence of SEQ ID NO:1, or the amino acid sequence of amino acid 327-528 of SEQ ID NO:1 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acid 327-528 of SEQ ID NO: 1. In one embodiment, the vaccine antigen comprises SEQ ID NO:1 amino acid 327-528.

In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of nucleotide 979-1584 of SEQ ID NO 2, 8 or 9, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotide 979-1584 of SEQ ID NO 2, 8 or 9, or a fragment of the nucleotide sequence of nucleotide 979-1584 of SEQ ID NO 2, 8 or 9: 2. a fragment of nucleotide sequence of nucleotide 979-1584 of 8 or 9 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acid 327-528 of SEQ ID NO:1, which is identical to the amino acid sequence of SEQ ID NO:1 amino acid 327-528 has an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical, or an immunogenic fragment of the amino acid sequence of SEQ ID No. 1 amino acid 327-528 or an immunogenic fragment of an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of SEQ ID No. 1 amino acid 327-528. In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of nucleotides 979-1584 of SEQ ID NO 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acid 327-528 of SEQ ID NO: 1.

According to certain embodiments, the signal peptide is fused to the SARS-CoV-2S protein, variant thereof, or fragment thereof, i.e., the antigenic peptide or protein, either directly or through a linker. Thus, in one embodiment, the signal peptide is fused to the above amino acid sequence derived from the SARS-CoV-2S protein or an immunogenic fragment thereof (antigenic peptide or protein) comprised by the above vaccine antigen.

Such signal peptides are sequences which generally exhibit a length of about 15 to 30 amino acids and are preferably located at the N-terminus of the antigen peptide or protein, but are not limited thereto. The signal peptide as defined herein preferably allows the transport of the RNA-encoded antigenic peptide or protein to a defined cellular compartment, preferably the cell surface, the Endoplasmic Reticulum (ER) or the endosome-lysosome compartment. In one embodiment, a signal peptide sequence as defined herein includes, but is not limited to, the signal peptide sequence of the SARS-CoV-2S protein, particularly a sequence comprising the amino acid sequence of amino acids 1-16 or 1-19 of SEQ ID NO:1 or a functional variant thereof.

In one embodiment, the signal sequence comprises SEQ ID NO:1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 16 of SEQ ID No. 1, or a functional fragment of the amino acid sequence of amino acids 1 to 16 of SEQ ID No. 1 or of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 16 of SEQ ID No. 1. In one embodiment, the signal sequence comprises SEQ ID NO:1, amino acid sequence of amino acids 1-16.

In one embodiment, the RNA encoding a signal sequence (i) comprises a nucleotide sequence of nucleotides 1-48 of SEQ ID NO 2, 8 or 9, a nucleotide sequence that is identical to the nucleotide sequence of SEQ ID NO: 2. 8 or 9, or a fragment of a nucleotide sequence of nucleotides 1 to 48 of SEQ ID No. 2, 8 or 9 or a nucleotide sequence of nucleotides 1 to 48 of SEQ ID No. 2, 8 or 9, or a fragment of a nucleotide sequence of nucleotides 1 to 48 of SEQ ID No. 99, 98, 97, 96, 95, 90, 85 or 80% identity; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1 to 16 of SEQ ID NO. 1, substantially identical to the amino acid sequence of SEQ ID NO:1 with an amino acid sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity, or a functional fragment of an amino acid sequence of amino acids 1 to 16 of SEQ ID No. 1 or an amino acid sequence with at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to an amino acid sequence of amino acids 1 to 16 of SEQ ID No. 1. In one embodiment, RNA encoding a signal sequence (i) comprises the nucleotide sequence of nucleotides 1-48 of SEQ ID NO. 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1-16 of SEQ ID NO: 1.

In one embodiment, the signal sequence comprises SEQ ID NO:1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 19 of SEQ ID No. 1, or a functional fragment of the amino acid sequence of amino acids 1 to 19 of SEQ ID No. 1 or of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 19 of SEQ ID No. 1. In one embodiment, the signal sequence comprises SEQ ID NO:1, amino acid sequence of amino acids 1-19.

In one embodiment, the RNA encoding a signal sequence (i) comprises a nucleotide sequence of nucleotides 1-57 of SEQ ID NO 2, 8 or 9, a nucleotide sequence that is identical to the nucleotide sequence of SEQ ID NO: 2. 8 or 9, or a fragment of a nucleotide sequence of nucleotides 1 to 57 of SEQ ID No. 2, 8 or 9 or a nucleotide sequence of nucleotides 1 to 57 of SEQ ID No. 2, 8 or 9, or a fragment of a nucleotide sequence of nucleotides 1 to 57 of SEQ ID No. 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-19 of SEQ ID No. 1, or the amino acid sequence of SEQ ID NO:1 or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-19 of SEQ ID No. 1. In one embodiment, RNA encoding a signal sequence (i) a nucleotide sequence comprising nucleotides 1-57 of SEQ ID NOs 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1-19 of SEQ ID NO: 1.

Signal peptides as defined herein further include, but are not limited to, signal peptide sequences of immunoglobulins, such as signal peptide sequences of immunoglobulin heavy chain variable regions, wherein the immunoglobulins may be human immunoglobulins. In particular, a signal peptide sequence as defined herein includes a sequence comprising the amino acid sequence of amino acids 1 to 22 of SEQ ID NO. 31 or a functional variant thereof.

In one embodiment, the signal sequence comprises the amino acid sequence of amino acids 1-22 of SEQ ID NO 31, which is identical to the amino acid sequence of SEQ ID NO:31 has an amino acid sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of amino acids 1 to 22 of SEQ ID No. 31, or a functional fragment of an amino acid sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of amino acids 1 to 22 of SEQ ID No. 31. In one embodiment, the signal sequence comprises SEQ ID NO:31, amino acid sequence of amino acids 1-22.

In one embodiment, the rna encoding the signal sequence (i) comprises SEQ ID NO:32, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 54-119 of SEQ ID No. 32, or a fragment of the nucleotide sequence of nucleotides 54-119 of SEQ ID No. 32 or a nucleotide sequence which is identical to SEQ ID NO:32, having a nucleotide sequence of nucleotides 54 to 119 of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:31, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 22 of SEQ ID No. 31, or a functional fragment of an amino acid sequence of amino acids 1 to 22 of SEQ ID No. 31 or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 22 of SEQ ID No. 31. In one embodiment, RNA encoding a signal sequence (i) a nucleotide sequence comprising nucleotides 54-119 of SEQ ID NO 32; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1-22 of SEQ ID NO 31.

Such signal peptides are preferably used to facilitate secretion of the encoded antigenic peptide or protein. More preferably, a signal peptide as defined herein is fused to an encoded antigenic peptide or protein as defined herein.

Thus, in a particularly preferred embodiment, the RNA described herein comprises at least one coding region encoding an antigenic peptide or protein and a signal peptide, preferably fused to the antigenic peptide or protein, more preferably fused to the N-terminus of the antigenic peptide or protein described herein.

In one embodiment, the vaccine antigen comprises SEQ ID NO:1 or 7, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID No. 1 or 7, or SEQ ID NO:1 or 7 or an immunogenic fragment thereof corresponding to the amino acid sequence of SEQ ID NO:1 or 7 has an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical. In one embodiment, the vaccine antigen comprises the amino acid sequence of SEQ ID NO 1 or 7.

In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of SEQ ID NO 2, 8 or 9, a nucleotide sequence that is complementary to the nucleotide sequence of SEQ ID NO: 2. 8 or 9, or a fragment of a nucleotide sequence of SEQ ID No. 2, 8 or 9 or a fragment of a nucleotide sequence of at least 99%, 98%, 97%, 96%, 95%, 85% or 80% identical to a nucleotide sequence of SEQ ID No. 2, 8 or 9 or a fragment of a nucleotide sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to a nucleotide sequence of SEQ ID No. 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:1 or 7, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID No. 1 or 7, or an immunogenic fragment of the amino acid sequence of SEQ ID No. 1 or 7 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID No. 1 or 7. In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of SEQ ID NO 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 1 or 7.

In one embodiment, the vaccine antigen comprises SEQ ID NO:7, and SEQ ID NO:7, or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO:7 or an immunogenic fragment of the amino acid sequence of SEQ ID NO:7, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity. In one embodiment, the vaccine antigen comprises the amino acid sequence of SEQ ID NO. 7.

In one embodiment, the rna encoding the vaccine antigen (i) comprises the nucleotide sequence of SEQ ID NO 15, 16, 19, 20, 24 or 25, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO 15, 16, 19, 20, 24 or 25, or a fragment of the nucleotide sequence of SEQ ID NO 15, 16, 19, 20, 24 or 25 or a fragment of the nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO 15, 16, 19, 20, 24 or 25; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID No. 7, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID No. 7, or an immunogenic fragment of the amino acid sequence of SEQ ID No. 7 or a polypeptide which is identical to the amino acid sequence of SEQ ID NO:7, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity. In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of SEQ ID NO 15, 16, 19, 20, 24 or 25; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO: 7.

In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 1-683 of SEQ ID No. 1, an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identical to the amino acid sequence of amino acids 1-683 of SEQ ID No. 1, or an immunogenic fragment of the amino acid sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identical to the amino acid sequence of amino acids 1-683 of SEQ ID No. 1. In one embodiment, the vaccine antigen comprises SEQ ID NO:1, amino acid sequence of amino acids 1-683.

In one embodiment, the rna encoding the vaccine antigen (i) comprises a nucleotide sequence of nucleotides 1 to 2049 of SEQ ID No. 2, 8 or 9, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 1 to 2049 of SEQ ID No. 2, 8 or 9, or a fragment of a nucleotide sequence of nucleotides 1 to 2049 of SEQ ID No. 2, 8 or 9 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 1 to 2049 of SEQ ID No. 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1-683 of SEQ ID No. 1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-683 of SEQ ID No. 1, or an immunogenic fragment of the amino acid sequence of amino acids 1-683 of SEQ ID No. 1 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-683 of SEQ ID No. 1. In one embodiment, the RNA encoding the vaccine antigen (i) comprises a nucleotide sequence of nucleotides 1-2049 of SEQ ID NO 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:1, amino acid sequence of amino acids 1-683.

In one embodiment, the vaccine antigen comprises an amino acid sequence of amino acids 1-685 of SEQ ID NO. 1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-685 of SEQ ID NO. 1, or an immunogenic fragment of an amino acid sequence of amino acids 1-685 of SEQ ID NO. 1 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-685 of SEQ ID NO. 1. In one embodiment, the vaccine antigen comprises SEQ ID NO:1, amino acid sequence of amino acids 1-685.

In one embodiment, the rna encoding the vaccine antigen (i) comprises the nucleotide sequence of nucleotides 1-2055 of SEQ ID No. 2, 8 or 9, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 1-2055 of SEQ ID No. 2, 8 or 9, or the nucleotide sequence of SEQ ID NO: 2. 8 or 9 or a fragment of a nucleotide sequence from nucleotides 1 to 2055 of SEQ ID No. 2, 8 or 9 or a fragment of a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence from nucleotides 1 to 2055 of SEQ ID NO; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1 to 685 of SEQ ID No. 1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 685 of SEQ ID No. 1, or an immunogenic fragment of the amino acid sequence of amino acids 1 to 685 of SEQ ID No. 1 or a polypeptide that is identical to the amino acid sequence of SEQ ID No. 1: 1 has an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical. In one embodiment, the RNA encoding the vaccine antigen (i) comprises a nucleotide sequence from nucleotides 1 to 2055 of SEQ ID NO. 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:1, amino acid sequence of amino acids 1-685.

In one embodiment, the vaccine antigen comprises SEQ ID NO:3, and SEQ ID NO:3, or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO:3 or an immunogenic fragment thereof corresponding to the amino acid sequence of SEQ ID NO:3, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the amino acid sequence of said polypeptide. In one embodiment, the vaccine antigen comprises the amino acid sequence of SEQ ID NO. 3.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:4, and SEQ ID NO:4, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO:4 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 4; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO. 3, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO. 3, or an immunogenic fragment of the amino acid sequence of SEQ ID NO. 3 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO. 3. In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO: 4; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 3.

In one embodiment, the vaccine antigen comprises SEQ ID NO:29, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-221 of SEQ ID No. 29, or an immunogenic fragment of the amino acid sequence of amino acids 1-221 of SEQ ID No. 29 or a polypeptide that is substantially identical to the amino acid sequence of amino acids 1-221 of SEQ ID NO:29, having an amino acid sequence at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical. In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 1-221 of SEQ ID NO. 29.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:30, and nucleotides 54 to 716 of SEQ ID NO:30, nucleotides 54-716, or a fragment of a nucleotide sequence of nucleotides 54-716 of SEQ ID No. 30 or a nucleotide sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to a nucleotide sequence of nucleotides 54-716 of SEQ ID No. 30; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:29, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-221 of SEQ ID No. 29, or the amino acid sequence of SEQ ID NO:29 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-221 of SEQ ID No. 29. In one embodiment, RNA encoding a vaccine antigen (i) comprises the nucleotide sequence of nucleotides 54-716 of SEQ ID NO. 30; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1-221 of SEQ ID NO: 29.

In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 1-224 of SEQ ID NO 31, identical to the amino acid sequence of SEQ ID NO:31, or an amino acid sequence of amino acids 1-224 of SEQ ID NO:31 or an immunogenic fragment of an amino acid sequence of amino acids 1-224 of SEQ ID NO:31, an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-224 of seq id no. In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 1-224 of SEQ ID NO. 31.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:32, nucleotide sequence corresponding to nucleotides 54 to 725 of SEQ ID NO:32 has a nucleotide sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 54-725 of SEQ ID NO:32 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 54-725 of SEQ ID NO: 32; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:31, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-224 of SEQ ID No. 31, or the amino acid sequence of SEQ ID NO:31 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-224 of SEQ ID No. 31. In one embodiment, RNA encoding a vaccine antigen (i) comprises the nucleotide sequence of nucleotides 54-725 of SEQ ID NO. 32; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:31, amino acid sequence of amino acids 1-224.

According to certain embodiments, the trimerizing domain is fused to the SARS-CoV-2S protein, variant thereof or fragment thereof, i.e., the antigenic peptide or protein, either directly or through a linker (e.g., a glycine/serine linker). Thus, in one embodiment, the trimerization domain is fused to the above amino acid sequence derived from the SARS-CoV-2S protein or an immunogenic fragment thereof (antigenic peptide or protein) comprised by the above vaccine antigen (which may optionally be fused to the above signal peptide).

Such trimerization domains are preferably located at the C-terminus of the antigenic peptide or protein, but are not limited thereto. The trimeric domain as defined herein preferably allows trimerization of the antigenic peptide or protein encoded by said RNA. Examples of trimerization domains as defined herein include, but are not limited to, the native trimerization domain of foldon, T4 secondary fibrin (fibritin). The C-terminal domain (foldon) of T4 minor fibrin is essential for the formation of the minor fibrin trimer structure and can be used as an artificial trimerization domain. In one embodiment, a trimerization domain as defined herein includes, but is not limited to, a sequence comprising SEQ ID NO: 10 or a functional variant thereof. In one embodiment, a trimerization domain as defined herein includes, but is not limited to, a sequence comprising SEQ ID NO: 10 or a functional variant thereof.

In one embodiment, the trimerization domain comprises SEQ ID NO:10, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 3 to 29 of SEQ ID No. 10, or a functional fragment of the amino acid sequence of amino acids 3 to 29 of SEQ ID No. 10 or of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 3 to 29 of SEQ ID No. 10. In one embodiment, the trimerization domain comprises the amino acid sequence of amino acids 3-29 of SEQ ID NO 10.

In one embodiment, the rna encoding the trimerization domain (i) comprises the nucleotide sequence of nucleotides 7 to 87 of SEQ ID No. 11, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 7 to 87 of SEQ ID No. 11, or the nucleotide sequence of SEQ ID NO:11 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 7 to 87 of SEQ ID No. 11; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 3 to 29 of SEQ ID NO. 10, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 3 to 29 of SEQ ID NO. 10, or a functional fragment of the amino acid sequence of amino acids 3 to 29 of SEQ ID NO. 10 or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 3 to 29 of SEQ ID NO. 10. In one embodiment, the RNA encoding the trimerization domain (i) comprises the nucleotide sequence of nucleotides 7-87 of SEQ ID NO. 11; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:10, amino acid sequence of amino acids 3-29.

In one embodiment, the trimerization domain comprises SEQ ID NO:10, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID No. 10, or a functional fragment of the amino acid sequence of SEQ ID No. 10 or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID No. 10. In one embodiment, the trimerization domain comprises the amino acid sequence of SEQ ID NO 10.

In one embodiment, the rna encoding the trimerization domain (i) comprises the nucleotide sequence of SEQ ID No. 11, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 11, or the nucleotide sequence of SEQ ID NO:11 or a fragment of the nucleotide sequence of SEQ ID NO:11, a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the nucleotide sequence of seq id no; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO:10, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO:10, or a functional fragment of the amino acid sequence of SEQ ID NO:10 or a functional fragment of the amino acid sequence of SEQ ID NO:10, or a functional fragment thereof having an amino acid sequence at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identical thereto. In one embodiment, the RNA encoding the trimerization domain (i) comprises the nucleotide sequence of SEQ ID NO 11; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 10.

Such trimerization domains are preferably used to facilitate trimerization of the encoded antigenic peptide or protein. More preferably, the trimerization domain as defined herein is fused to an antigenic peptide or protein as defined herein.

Thus, in a particularly preferred embodiment, the RNA described herein comprises at least one coding region encoding an antigenic peptide or protein and a trimerization domain as defined herein, preferably fused to the antigenic peptide or protein, more preferably fused to the C-terminus of the antigenic peptide or protein.

In one embodiment, the vaccine antigen comprises SEQ ID NO:5, and SEQ ID NO:5, or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO:5 or an immunogenic fragment of the amino acid sequence of SEQ ID NO:5 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity. In one embodiment, the vaccine antigen comprises the amino acid sequence of SEQ ID NO. 5.

In one embodiment, the rna encoding the vaccine antigen (i) comprises the nucleotide sequence of SEQ ID No. 6, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 6, or a fragment of the nucleotide sequence of SEQ ID No. 6 or a fragment of the nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 6; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:5, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID No. 5, or an immunogenic fragment of the amino acid sequence of SEQ ID No. 5 or a polypeptide which is identical to the amino acid sequence of SEQ ID NO:5 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity. In one embodiment, RNA encoding a vaccine antigen (i) comprises the nucleotide sequence of SEQ ID NO 6; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 5.

In one embodiment, the rna encoding the vaccine antigen (i) comprises the nucleotide sequence of SEQ ID NO 17, 21 or 26, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO 17, 21 or 26, or a fragment of the nucleotide sequence of SEQ ID NO 17, 21 or 26 or a nucleotide sequence that is identical to SEQ ID NO: 17. 21 or 26, a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:5, and SEQ ID NO:5, or an immunogenic fragment of the amino acid sequence of SEQ ID No. 5 or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID No. 5. In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO: 17. 21 or 26; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO: 5.

In one embodiment, the vaccine antigen comprises the amino acid sequence of SEQ ID NO. 18, an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of SEQ ID NO. 18, or the amino acid sequence of SEQ ID NO:18 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID No. 18. In one embodiment, the vaccine antigen comprises SEQ ID NO: 18.

In one embodiment, the vaccine antigen comprises SEQ ID NO:29, amino acid sequence corresponding to amino acids 1-257 of SEQ ID NO:29, or an immunogenic fragment of the amino acid sequence of amino acids 1-257 of SEQ ID No. 29 or an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of amino acids 1-257 of SEQ ID NO:29, amino acid sequence of amino acids 1-257 of at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity. In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 1-257 of SEQ ID NO. 29.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:30, and nucleotides 54 to 824 of SEQ ID NO:30, nucleotide 54-824, or a fragment of a nucleotide sequence of nucleotides 54-824 of SEQ ID No. 30 or a nucleotide sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to a nucleotide sequence of nucleotides 54-824 of SEQ ID No. 30; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1-257 of SEQ ID NO:29, which is identical to the amino acid sequence of SEQ ID NO:29, or an immunogenic fragment of the amino acid sequence of amino acids 1-257 of SEQ ID No. 29 or an amino acid sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of amino acids 1-257 of SEQ ID No. 29. In one embodiment, RNA encoding a vaccine antigen (i) comprises the nucleotide sequence of nucleotides 54-824 of SEQ ID NO. 30; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:29, amino acid sequence of amino acids 1 to 257.

In one embodiment, the vaccine antigen comprises SEQ ID NO:31, and amino acid sequence of amino acids 1-260 of SEQ ID NO:31, or an immunogenic fragment of an amino acid sequence of amino acids 1-260 of SEQ ID No. 31 or an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of amino acids 1-260 of SEQ ID NO:31, an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-260. In one embodiment, the vaccine antigen comprises SEQ ID NO:31, amino acid sequence of amino acids 1-260.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:32, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 54-833 of SEQ ID No. 32, or the nucleotide sequence of SEQ ID NO:32 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 54-833 of SEQ ID No. 32; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1-260 of SEQ ID NO:31, substantially identical to the amino acid sequence of SEQ ID NO:31, or an immunogenic fragment of the amino acid sequence of amino acids 1-260 of SEQ ID No. 31 or an amino acid sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of amino acids 1-260 of SEQ ID No. 31. In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:32, nucleotide sequence 54-833; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:31, amino acid sequence of amino acids 1-260.

In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 20-257 of SEQ ID No. 29, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 20-257 of SEQ ID No. 29, or the amino acid sequence of SEQ ID NO:29 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 20-257 of SEQ ID No. 29. In one embodiment, the vaccine antigen comprises SEQ ID NO:29 amino acids 20-257.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:30, a nucleotide sequence of nucleotide 111-824 of SEQ ID NO. 30 which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotide 111-824 of SEQ ID NO. 30, or a fragment of the nucleotide sequence of nucleotide 111-824 of SEQ ID NO. 30 which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotide 111-824 of SEQ ID NO. 30; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:29, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 20-257 of SEQ ID No. 29, or an immunogenic fragment of the amino acid sequence of amino acids 20-257 of SEQ ID No. 29 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 20-257 of SEQ ID No. 29. In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of nucleotides 111-824 of SEQ ID NO: 30; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 20-257 of SEQ ID NO: 29.

In one embodiment, the vaccine antigen comprises SEQ ID NO:31, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 23-260 of SEQ ID No. 31, or an immunogenic fragment of the amino acid sequence of amino acids 23-260 of SEQ ID No. 31 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 23-260 of SEQ ID No. 31. In one embodiment, the vaccine antigen comprises SEQ ID NO:31, amino acid sequence of amino acids 23-260.

In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of nucleotide 120-833 of SEQ ID NO. 32, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotide 120-833 of SEQ ID NO. 32, or a fragment of the nucleotide sequence of nucleotide 120-833 of SEQ ID NO. 32 or a fragment of the nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotide 120-833 of SEQ ID NO. 32; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 23 to 260 of SEQ ID NO. 31, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 23 to 260 of SEQ ID NO. 31, or an immunogenic fragment of the amino acid sequence of amino acids 23 to 260 of SEQ ID NO. 31 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 23 to 260 of SEQ ID NO. 31. In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of nucleotides 120-833 of SEQ ID NO. 32; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 23-260 of SEQ ID NO: 31.

According to certain embodiments, the transmembrane domain is fused to the SARS-CoV-2S protein, variant thereof, or fragment thereof, i.e., the antigenic peptide or protein, either directly or through a linker (e.g., a glycine/serine linker). Thus, in one embodiment, the transmembrane domain is fused to the above amino acid sequence derived from the SARS-CoV-2S protein or immunogenic fragment thereof (antigenic peptide or protein) comprised by the above vaccine antigen (which may optionally be fused to the above signal peptide and/or trimerization domain).

Such transmembrane domains are preferably located at the C-terminus of the antigenic peptide or protein, but are not limited thereto. Preferably, such a transmembrane domain is located at the C-terminus of the trimerization domain (if present), but is not limited thereto. In one embodiment, the trimerization domain is present between the SARS-CoV-2S protein, variant thereof or fragment thereof (i.e., antigenic peptide or protein), and the transmembrane domain.

The transmembrane domain as defined herein preferably allows the antigenic peptide or protein encoded by the RNA to be anchored in the cell membrane.

In one embodiment, the transmembrane domain sequence as defined herein includes, but is not limited to, the transmembrane domain sequence of the SARS-CoV-2S protein, in particular a transmembrane domain sequence comprising the amino acid sequence of SEQ ID NO: 1, amino acid 1207-1254 or a functional variant thereof.

In one embodiment, the transmembrane domain sequence comprises the amino acid sequence of amino acid 1207-1254 of SEQ ID NO. 1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acid 1207-1254 of SEQ ID NO. 1, or a functional fragment of the amino acid sequence of amino acid 1207-1254 of SEQ ID NO. 1 or a functional fragment of the amino acid sequence of SEQ ID NO. 1: 1 amino acid 1207-1254 has an amino acid sequence at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical. In one embodiment, the transmembrane domain sequence comprises SEQ ID NO:1 amino acid 1207-1254.

In one embodiment, the RNA encoding the transmembrane domain sequence (i) comprises the nucleotide sequence of nucleotides 3619-3762 of SEQ ID NO 2, 8 or 9, and the sequence of nucleotides corresponding to SEQ ID NO: 2. 8 or 9 nucleotide 3619-3762 has a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical, or a fragment of the nucleotide sequence 3619-3762 of SEQ ID NO. 2, 8 or 9 or a fragment of the nucleotide sequence at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence 3619-3762 of SEQ ID NO. 2, 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acid 1207-1254 of SEQ ID NO:1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acid 1207-1254 of SEQ ID NO:1, or an amino acid sequence of SEQ ID NO:1 or a functional fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acid 1207-1254 of SEQ ID NO: 1. In one embodiment, the rna encoding the transmembrane domain sequence (i) comprises SEQ ID NO: 2. nucleotide 3619-3762 of 8 or 9; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1207-1254 of SEQ ID NO: 1.

In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 1-311 of SEQ ID NO. 29, having a sequence identical to the sequence set forth in SEQ ID NO:29 has an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-311 of SEQ ID NO:29 or an immunogenic fragment of the amino acid sequence of amino acids 1-311 of SEQ ID NO:29, having an amino acid sequence at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical. In one embodiment, the vaccine antigen comprises SEQ ID NO:29, amino acid sequence of amino acids 1-311.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:30, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 54 to 986 of SEQ ID No. 30, or a fragment of the nucleotide sequence of nucleotides 54 to 986 of SEQ ID No. 30 or a fragment of the nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 54 to 986 of SEQ ID No. 30; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:29, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-311 of SEQ ID No. 29, or an immunogenic fragment of the amino acid sequence of amino acids 1-311 of SEQ ID No. 29 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-311 of SEQ ID No. 29. In one embodiment, RNA encoding a vaccine antigen (i) comprises the nucleotide sequence of nucleotides 54-986 of SEQ ID NO. 30; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:29, amino acid sequence of amino acids 1-311.

In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 1-314 of SEQ ID NO. 31, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-314 of SEQ ID NO. 31, or the amino acid sequence of SEQ ID NO:31 or an immunogenic fragment of an amino acid sequence of amino acids 1-314 of SEQ ID NO:31, an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-314. In one embodiment, the vaccine antigen comprises SEQ ID NO:31, amino acid sequence of amino acids 1-314.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:32, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 54-995 of SEQ ID NO:32, or a fragment of the nucleotide sequence of nucleotides 54-995 of SEQ ID NO:32 or a fragment of the nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 54-995 of SEQ ID NO: 32; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1 to 314 of SEQ ID NO. 31, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 314 of SEQ ID NO. 31, or an immunogenic fragment of the amino acid sequence of amino acids 1 to 314 of SEQ ID NO. 31 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 314 of SEQ ID NO. 31. In one embodiment, RNA encoding a vaccine antigen (i) comprises the nucleotide sequence of nucleotides 54-995 of SEQ ID NO: 32; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 1-314 of SEQ ID NO 31.

In one embodiment, the vaccine antigen comprises SEQ ID NO:29, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 20-311 of SEQ ID No. 29, or an immunogenic fragment of the amino acid sequence of amino acids 20-311 of SEQ ID No. 29 or a polypeptide which is identical to the amino acid sequence of amino acids 20-311 of SEQ ID NO:29, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity thereto. In one embodiment, the vaccine antigen comprises SEQ ID NO:29 amino acid sequence of amino acids 20-311.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:30, nucleotide 111-986, and the nucleotide sequence of SEQ ID NO:30, or a fragment of the nucleotide sequence of nucleotide 111-986 of SEQ ID No. 30 or a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotide 111-986 of SEQ ID NO:30, nucleotide 111-986 has at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical nucleotide sequence of the fragment; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:29, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 20-311 of SEQ ID No. 29, or the amino acid sequence of SEQ ID NO:29 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 20-311 of SEQ ID No. 29. In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of nucleotides 111-986 of SEQ ID NO. 30; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 20-311 of SEQ ID NO: 29.

In one embodiment, the vaccine antigen comprises the amino acid sequence of amino acids 23-314 of SEQ ID No. 31, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 23-314 of SEQ ID No. 31, or an immunogenic fragment of the amino acid sequence of amino acids 23-314 of SEQ ID No. 31 or a peptide of SEQ ID NO:31 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity thereto. In one embodiment, the vaccine antigen comprises SEQ ID NO:31, amino acids 23-314.

In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO:32, the nucleotide sequence of nucleotide 120-995 of SEQ ID NO:32, a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotide 120-995 of SEQ ID NO:32, or a fragment of the nucleotide sequence of nucleotide 120-995 of SEQ ID NO:32 or a fragment of the nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotide 120-995 of SEQ ID NO: 32; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO:31, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 23-314 of SEQ ID No. 31, or an immunogenic fragment of the amino acid sequence of amino acids 23-314 of SEQ ID No. 31 or a polypeptide that is substantially identical to the amino acid sequence of amino acids 23-314 of SEQ ID NO:31 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity thereto. In one embodiment, the RNA encoding the vaccine antigen (i) comprises the nucleotide sequence of nucleotides 120-995 of SEQ ID NO: 32; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of amino acids 23-314 of SEQ ID NO 31.

In one embodiment, the rna encoding the vaccine antigen (i) comprises the nucleotide sequence of SEQ ID No. 30, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 30, or a fragment of the nucleotide sequence of SEQ ID No. 30 or a fragment of the nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 30; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO. 29, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO. 29, or an immunogenic fragment of the amino acid sequence of SEQ ID NO. 29 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO. 29. In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO: 30; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO: 29.

In one embodiment, RNA encoding a vaccine antigen (i) comprises the nucleotide sequence of SEQ ID NO 32: 32, or a fragment of the nucleotide sequence of SEQ ID No. 32 or a nucleotide sequence at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of SEQ ID No. 32; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 31, a sequence identical to SEQ ID NO:31, or an immunogenic fragment of the amino acid sequence of SEQ ID No. 31 or an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of SEQ ID NO:31 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity thereto. In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO: 32; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO: 31.

In one embodiment, the vaccine antigen comprises the amino acid sequence of SEQ ID NO. 28, an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of SEQ ID NO. 28, or the amino acid sequence of SEQ ID NO:28 or an immunogenic fragment thereof that hybridizes to the amino acid sequence of SEQ ID NO:28, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity thereto. In one embodiment, the vaccine antigen comprises the amino acid sequence of SEQ ID NO 28.

In one embodiment, the rna encoding the vaccine antigen (i) comprises the nucleotide sequence of SEQ ID No. 27, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 27, or a fragment of the nucleotide sequence of SEQ ID No. 27 or a fragment of the nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 27; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO. 28, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO. 28, or an immunogenic fragment of the amino acid sequence of SEQ ID NO. 28 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO. 28. In one embodiment, the rna encoding the vaccine antigen (i) comprises SEQ ID NO: 27; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO: 28.

In one embodiment, the vaccine antigen comprises a contiguous sequence of SARS-CoV-2 coronavirus spike (S) protein consisting of, or consisting essentially of, the amino acid sequence from SARS-CoV-2S protein or an immunogenic fragment thereof (antigenic peptide or protein) comprised by the vaccine antigen. In one embodiment, the vaccine antigen comprises a contiguous sequence of SARS-CoV-2 coronavirus spike (S) protein of no more than 220 amino acids, 215 amino acids, 210 amino acids, or 205 amino acids.

In one embodiment, the RNA encoding the vaccine antigen is a nucleoside-modified messenger RNA (modrna) described herein as BNT162b1(RBP020.3), BNT162b2(RBP020.1 or RBP 020.2). In one embodiment, the RNA encoding the vaccine antigen is a nucleoside-modified messenger RNA (modrna) described herein as RBP 020.2.

In one embodiment, the RNA encoding the vaccine antigen is a nucleoside modified messenger RNA (modrna) and (i) comprises the nucleotide sequence of SEQ ID NO:21, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO:21, and/or (ii) encodes an amino acid sequence comprising the nucleotide sequence of SEQ ID NO: 5, or an amino acid sequence identical to SEQ ID NO: 5 has an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical. In one embodiment, the RNA encoding the vaccine antigen is a nucleoside-modified messenger RNA (modRNA), and (i) comprises the nucleotide sequence of SEQ ID NO: 21; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO: 5.

In one embodiment, the RNA encoding the vaccine antigen is a nucleoside-modified messenger RNA (modrna), and (i) comprises SEQ ID NO:19 or 20, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO 19 or 20, and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 7, or a nucleotide sequence which is identical to the nucleotide sequence of SEQ ID NO:7 has an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical. In one embodiment, the RNA encoding the vaccine antigen is a nucleoside-modified messenger RNA (modRNA), and (i) comprises the nucleotide sequence of SEQ ID NO 19 or 20; and/or (ii) encodes an amino acid sequence comprising SEQ ID NO: 7.

In one embodiment, the RNA encoding the vaccine antigen is a nucleoside-modified messenger RNA (modrna), and (i) comprises SEQ ID NO:20, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO:20, and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO:7, or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO: 7. In one embodiment, the RNA encoding the vaccine antigen is a nucleoside-modified messenger RNA (modRNA), and (i) comprises the nucleotide sequence of SEQ ID NO: 20; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 7.

As used herein, the term "vaccine" refers to a composition that induces an immune response upon vaccination into a subject. In some embodiments, the induced immune response provides protective immunity.

In one embodiment, RNA encoding an antigenic molecule is expressed in cells of the subject to provide the antigenic molecule.

In one embodiment, the expression of the antigenic molecule is on the surface of the cell or into the extracellular space. In one embodiment, the antigenic molecule is presented in the context of an MHC. In one embodiment, the RNA encoding the antigenic molecule is transiently expressed in the cells of the subject. In one embodiment, the RNA encoding the antigenic molecule is expressed in muscle after administration of the RNA encoding the antigenic molecule, in particular after intramuscular administration of the RNA encoding the antigenic molecule. In one embodiment, following administration of the RNA encoding the antigenic molecule, the RNA encoding the antigenic molecule is expressed in the spleen.

In one embodiment, the RNA encoding the antigenic molecule is expressed in antigen presenting cells, preferably professional antigen presenting cells, after administration of the RNA encoding the antigenic molecule. In one embodiment, the antigen presenting cell is selected from the group consisting of a dendritic cell, a macrophage, and a B cell. In one embodiment, no or substantially no expression of the RNA encoding the antigenic molecule occurs in the lung and/or liver following administration of the RNA encoding the antigenic molecule. In one embodiment, the RNA encoding the antigenic molecule is expressed in the spleen in an amount at least 5-fold greater than the amount expressed in the lung following administration of the RNA encoding the antigenic molecule.

In some embodiments, following administration to a subject, particularly intramuscular administration, the methods and agents described herein, e.g., mRNA compositions, result in delivery of RNA encoding a vaccine antigen to the lymph nodes and/or spleen. In some embodiments, RNA encoding the vaccine antigen is detectable in the lymph nodes and/or spleen 6 hours or more after administration, and preferably up to 6 days or more.

In some embodiments, following administration to a subject, particularly intramuscular administration, the methods and agents described herein, e.g., mRNA compositions, result in delivery of RNA encoding a vaccine antigen to the B cell follicular, the subcapsular sinus, and/or the T cell region, particularly the B cell follicular and/or the subcapsular sinus of a lymph node.

In some embodiments, following administration to a subject, particularly intramuscular administration, the methods and agents described herein, e.g., mRNA compositions, result in delivery of RNA encoding a vaccine antigen to B cells in the T cell region and the middle sinus of the lymph node (CD19+), subcapsular sinus macrophages (CD169+) and/or dendritic cells (CD11c +), particularly B cells of lymph nodes (CD19+) and/or subcapsular sinus macrophages (CD169 +).

In some embodiments, following administration to a subject, particularly intramuscular administration, the methods and agents described herein, e.g., mRNA compositions, result in delivery of RNA encoding a vaccine antigen to the white marrow of the spleen.

In some embodiments, following administration to a subject, particularly intramuscular administration, the methods and agents described herein, e.g., mRNA compositions, result in the delivery of RNA encoding a vaccine antigen to B cells, DCs (CD11c +), particularly those surrounding B cells, and/or macrophages of the spleen, particularly B cells and/or DCs (CD11c +).

In one embodiment, the vaccine antigen is expressed in the lymph nodes and/or spleen, particularly in cells of the lymph nodes and/or spleen as described above.

Peptide and protein antigens suitable for use in accordance with the present disclosure generally include peptides or proteins comprising an epitope of the SARS-CoV-2S protein or a functional fragment thereof for inducing an immune response. The peptide or protein or epitope may be derived from a target antigen, i.e. an antigen against which an immune response is elicited. For example, a peptide or protein antigen or an epitope contained within a peptide or protein antigen may be a target antigen or a fragment or variant of a target antigen. The target antigen may be a coronavirus S protein, particularly a SARS-CoV-2S protein.

The antigenic molecule or its processed product, e.g., a fragment thereof, can bind to an antigen receptor, such as a BCR or TCR, carried by an immune effector cell, or to an antibody.

Peptide or protein antigens, i.e., vaccine antigens, provided to a subject by administering RNA encoding the peptide or protein antigens according to the invention preferably result in the induction of an immune response, e.g., a humoral and/or cellular immune response, in the subject providing the peptide or protein antigens. The immune response is preferably directed against a target antigen, in particular a coronavirus S protein, in particular a SARS-CoV-2S protein. Thus, a vaccine antigen may comprise a target antigen, a variant thereof, or a fragment thereof. In one embodiment, such a fragment or variant is immunologically equivalent to the target antigen. In the context of the present disclosure, the term "fragment of an antigen" or "variant of an antigen" denotes a substance that results in the induction of an immune response that targets the antigen, i.e. the target antigen. Thus, a vaccine antigen may correspond to or may comprise a target antigen, may correspond to or may comprise a fragment of a target antigen, or may correspond to or may comprise an antigen that is homologous to a target antigen or fragment thereof. Thus, according to the present disclosure, a vaccine antigen may comprise an immunogenic fragment of a target antigen or an amino acid sequence homologous to an immunogenic fragment of a target antigen. An "immunogenic fragment of an antigen" according to the present disclosure preferably relates to a fragment of an antigen capable of inducing an immune response against a target antigen. The vaccine antigen may be a recombinant antigen.

The term "immunologically equivalent" means that the immunologically equivalent molecules, such as the immunologically equivalent amino acid sequences, exhibit the same or substantially the same immunological properties and/or exert the same or substantially the same immunological effect, e.g., in terms of the type of immunological effect. In the context of the present disclosure, the term "immunologically equivalent" is preferably used in relation to the immunological effect or property of the antigen or antigen variant used for immunization. For example, an amino acid sequence is immunologically equivalent to a reference amino acid sequence if, upon exposure to the immune system of the subject, the amino acid sequence induces an immune response having reaction specificity for the reference amino acid sequence.

As used herein, "activation" or "stimulation" refers to the state of an immune effector cell, such as a T cell, that has been sufficiently stimulated to induce detectable cell proliferation. Activation can also be associated with initiation of signaling pathways, induced cytokine production, and detectable effector functions. The term "activated immune effector cells" refers to immune effector cells that undergo cell division.

The term "priming" refers to a process in which an immune effector cell, such as a T cell, is first contacted with its specific antigen and results in differentiation into an effector cell, such as an effector T cell.

The term "clonal amplification" or "amplification" refers to a process in which specific entities are increased. In the context of the present disclosure, the term is preferably used in the context of an immunological response, wherein immune effector cells are stimulated by an antigen, proliferate, and the specific immune effector cells recognizing the antigen expand. Preferably, clonal expansion results in differentiation of immune effector cells.

The term "antigen" relates to a substance comprising an epitope against which an immune response can be generated. In particular, the term "antigen" includes proteins and peptides. In one embodiment, the antigen is presented by a cell of the immune system, e.g., an antigen presenting cell, such as a dendritic cell or macrophage. In one embodiment, the antigen or processed product thereof, such as a T-cell epitope, binds to a T-or B-cell receptor, or to an immunoglobulin molecule, such as an antibody. Thus, the antigen or its processed product can specifically react with an antibody or a T lymphocyte (T cell). In one embodiment, the antigen is a viral antigen, such as a coronavirus S protein, e.g., a SARS-CoV-2S protein, and the epitope is derived from such an antigen.

The term "viral antigen" refers to any viral component having antigenic properties, i.e. capable of eliciting an immune response in an individual. The viral antigen can be a coronavirus S protein, e.g., a SARS-CoV-2S protein.

The term "expressed on the surface of a cell" or "associated with the surface of a cell" means that a molecule, such as an antigen, is associated with and located on the plasma membrane of a cell, wherein at least a portion of the molecule faces the extracellular space of the cell and is accessible from the outside of the cell, e.g., by an antibody located outside the cell. In this context, a portion is preferably at least 4, preferably at least 8, preferably at least 12, more preferably at least 20 amino acids. The association may be direct or indirect. For example, the association may be through one or more transmembrane domains, one or more lipid anchors, or through interaction with any protein, lipid, carbohydrate or other structure that may be found on the outer leaflet of the plasma membrane of a cell. For example, a molecule associated with the cell surface may be a transmembrane protein with an extracellular portion, or may be a protein associated with the cell surface by interacting with another protein (which is a transmembrane protein).

"cell surface" or "surface of a cell" is used in its normal meaning in the art and thus includes the exterior of a cell that is accessible for binding by proteins and other molecules. An antigen is expressed on the surface of a cell if it is located on the surface of the cell and can be bound, for example, by an antigen-specific antibody added to the cell.

The term "extracellular portion" or "extracellular domain (exodomain)" in the context of the present invention refers to a portion of a molecule, such as a protein, which faces the extracellular space of a cell and is preferably accessible from outside said cell, e.g. by means of a binding molecule, such as an antibody, located outside the cell. Preferably, the term refers to one or more extracellular loops or domains or fragments thereof.

The term "epitope" refers to a portion or fragment of a molecule, such as an antigen, that is recognized by the immune system. For example, the epitope may be recognized by a T cell, B cell, or antibody. An epitope of an antigen may comprise a continuous or discontinuous portion of the antigen and may be from about 5 to about 100, such as from about 5 to about 50, more preferably from about 8 to about 30, most preferably from about 8 to about 25 amino acids in length, for example, an epitope may preferably be 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids in length. In one embodiment, the epitope is about 10 to about 25 amino acids in length. The term "epitope" includes T cell epitopes.

The term "T cell epitope" refers to a portion or fragment of a protein that is recognized by T cells when presented in the context of MHC molecules. The term "major histocompatibility complex" and the abbreviation "MHC" include both MHC class I and MHC class II molecules and relate to the genetic complex present in all vertebrates. MHC proteins or molecules are important for signaling between lymphocytes and antigen presenting or diseased cells in immune responses, where they bind peptide epitopes and present them for T cell receptor recognition on T cells. MHC-encoded proteins are expressed on the cell surface and display self-antigens (peptide fragments from the cell itself) and non-self-antigens (e.g., fragments of invading microorganisms) to T cells. In the case of class I MHC/peptide complexes, the binding peptides are typically about 8 to about 10 amino acids in length, although longer or shorter peptides may be effective. In the case of MHC class II/peptide complexes, the binding peptides are generally about 10 to about 25 amino acids, particularly about 13 to about 18 amino acids in length, while longer or shorter peptides may be effective.

Peptide and protein antigens may be 2-100 amino acids in length, including, for example, 5 amino acids, 10 amino acids, 15 amino acids, 20 amino acids, 25 amino acids, 30 amino acids, 35 amino acids, 40 amino acids, 45 amino acids, or 50 amino acids. In some embodiments, the peptide may be more than 50 amino acids in length. In some embodiments, the peptide may be 100 amino acids or more in length.

The peptide or protein antigen may be any peptide or protein that can induce or increase the ability of the immune system to develop antibody and T cell responses to the peptide or protein.

In one embodiment, the vaccine antigen is recognized by immune effector cells. Preferably, if the vaccine antigen is recognized by an immune effector cell, it is capable of inducing stimulation, priming and/or expansion of the immune effector cell carrying the antigen receptor recognizing the vaccine antigen in the presence of an appropriate co-stimulatory signal. In the context of embodiments of the present invention, the vaccine antigen is preferably presented or present on the surface of a cell, preferably an antigen presenting cell. In one embodiment, the antigen is presented by a diseased cell (e.g., a virally infected cell). In one embodiment, the antigen receptor is a TCR, which binds to an epitope presented in the context of an MHC. In one embodiment, binding of the TCR to an antigen presented by a cell (e.g., an antigen presenting cell) results in stimulation, priming, and/or expansion of the T cell when expressed by and/or present on the T cell. In one embodiment, binding of the TCR to an antigen presented on a diseased cell, when expressed by and/or present on a T cell, which preferably releases cytotoxic factors such as perforin and granzyme, results in cytolysis and/or apoptosis of the diseased cell.

In one embodiment, the antigen receptor is an antibody or B cell receptor that binds to an epitope in the antigen. In one embodiment, the antibody or B cell receptor binds to a native epitope of the antigen.

Nucleic acids

As used herein, the term "polynucleotide" or "nucleic acid" is intended to include DNA and RNA such as genomic DNA, cDNA, mRNA, recombinantly produced and chemically synthesized molecules. The nucleic acid may be single-stranded or double-stranded. RNA includes in vitro transcribed RNA, (IVT RNA) or synthetic RNA. According to the invention, the polynucleotide is preferably isolated.

The nucleic acid may be contained in a vector. The term "vector" as used herein includes any vector known to the skilled person, including plasmid vectors, cosmid vectors, phage vectors (such as lambda phage), viral vectors (such as retrovirus, adenovirus or baculovirus vectors) or artificial chromosome vectors (such as Bacterial Artificial Chromosome (BAC), Yeast Artificial Chromosome (YAC) or P1 Artificial Chromosome (PAC)). The vector includes an expression vector and a cloning vector. Expression vectors include plasmids as well as viral vectors, and typically contain the desired coding sequence and appropriate DNA sequences necessary for expression of the operably linked coding sequence in a particular host organism (e.g., bacterial, yeast, plant, insect, or mammalian) or in vitro expression system. Cloning vectors are generally used to engineer and amplify a desired DNA fragment and may lack the functional sequences required to express the desired DNA fragment.

In one embodiment of all aspects of the invention, RNA encoding a vaccine antigen is expressed in a cell (e.g., an antigen presenting cell of a subject treated to provide the vaccine antigen).

The nucleic acids described herein may be recombinant and/or isolated molecules.

In the present disclosure, the term "RNA" relates to a nucleic acid molecule comprising ribonucleotide residues. In a preferred embodiment, the RNA comprises all or most ribonucleotide residues. As used herein, "ribonucleotide" refers to a nucleotide having a hydroxyl group at the 2' -position of the β -D-ribofuranosyl (β -D-ribofuranosyl) group. RNA encompasses, but is not limited to, double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, substantially pure RNA, synthetic RNA, recombinantly produced RNA, and modified RNA that differs from naturally occurring RNA by the addition, deletion, substitution, and/or alteration of one or more nucleotides. Such alterations may refer to the addition of non-nucleotide species to the end of an internal RNA molecule or RNA. It is also contemplated herein that the nucleotides in the RNA can be non-standard nucleotides, such as chemically synthesized nucleotides or deoxynucleotides. For purposes of this disclosure, these altered RNAs are considered analogs of naturally occurring RNAs.

In certain embodiments of the present disclosure, the RNA is messenger RNA (mrna) associated with an RNA transcript encoding a peptide or protein. As established in the art, mRNAs typically comprise a 5 'untranslated region (5' -UTR), a peptide coding region, and a 3 'untranslated region (3' -UTR). In some embodiments, the RNA is produced by in vitro transcription or chemical synthesis. In one embodiment, the mRNA is produced by in vitro transcription using a DNA template, wherein DNA refers to a nucleic acid comprising deoxyribonucleotides.

In one embodiment, the RNA is in vitro transcribed RNA (IVT-RNA) and can be obtained by in vitro transcription of an appropriate DNA template. The promoter used to control transcription may be any promoter of any RNA polymerase. DNA templates for in vitro transcription can be obtained by cloning nucleic acids, in particular cDNA, and introducing them into an appropriate vector for in vitro transcription. cDNA can be obtained by reverse transcription of RNA.

In certain embodiments of the present disclosure, the RNA is a "replicon RNA" or simply a "replicon", in particular a "self-replicating RNA" or "self-amplifying RNA". In a particularly preferred embodiment, the replicon or self-replicating RNA is derived from or comprises elements derived from an ssRNA virus, in particular a positive-stranded ssRNA virus such as an alphavirus. Alphaviruses are a typical representation of positive-stranded RNA viruses. Alphaviruses replicate in the cytoplasm of infected cells (for reviews of the alphavirus life cycle, see Jos e et al, Future microbiol, 2009, vol.4, pp.837-856). The total genomic length of many alphaviruses typically ranges between 11,000 and 12,000 nucleotides, and the genomic RNA typically has a 5 '-cap and a 3' poly (a) tail. The genome of alphaviruses encodes non-structural proteins (involved in transcription, modification and replication of viral RNA and protein modification) and structural proteins (forming viral particles). There are usually two Open Reading Frames (ORFs) in the genome. The 4 nonstructural proteins (nsP 1-nsP 4) are typically encoded together by a first ORF starting near the 5 'end of the genome, while the alphavirus structural proteins are encoded together by a second ORF located downstream of the first ORF and extending near the 3' end of the genome. Typically, the first ORF is larger than the second ORF, in a ratio of approximately 2: 1. In cells infected with an alphavirus, only nucleic acid sequences encoding non-structural proteins are translated from genomic RNA, whereas genetic information encoding structural proteins can be translated from subgenomic transcripts, which are RNA molecules similar to eukaryotic messenger RNA (mRNA; Gould et al.,2010, Antiviral Res., vol.87pp.111-124). Following infection, i.e., early in the viral life cycle, the (+) strand genomic RNA acts directly as messenger RNA for translation of the open reading frame encoding the nonstructural polyprotein (nsP 1234). Alphavirus-derived vectors have been proposed for the delivery of foreign genetic information into target cells or target organisms. In a simple method, the open reading frame encoding the alphavirus structural protein is replaced by an open reading frame encoding the protein of interest. The alphavirus-based trans-replication system relies on the alphavirus nucleotide sequence elements on two separate nucleic acid molecules: one nucleic acid molecule encodes a viral replicase, while the other nucleic acid molecule is capable of being replicated in trans by the replicase (hence the name trans-replication system). Trans-replication requires the presence of both nucleic acid molecules in a given host cell. Nucleic acid molecules capable of being replicated in trans by replicase enzymes must contain certain alphavirus sequence elements to allow recognition and RNA synthesis by the alphavirus replicase.

In one embodiment, the RNA described herein may have modified nucleosides. In some embodiments, the RNA comprises modified nucleosides in place of at least one (e.g., each) uridine.

As used herein, the term "uracil" describes one of the nucleobases that can occur in a nucleic acid of an RNA. The structure of uracil is:

as used herein, the term "uridine" describes one of the nucleosides that can be present in RNA. The structure of uridine is:

UTP (uridine 5' -triphosphate) has the following structure:

pseudo-UTP (pseudouridine 5' -triphosphate) has the following structure:

"pseudouridine" is an example of a modified nucleoside, which is an isomer of uridine, in which uracil is linked to the pentose ring by a carbon-carbon bond rather than a nitrogen-carbon glycosidic bond.

Another exemplary modified nucleoside is N1-methyl-pseudouridine (m1 Ψ), having the following structure:

N1-methyl-pseudo-UTP has the following structure:

another exemplary modified nucleoside is 5-methyl-uridine (m5U), which has the following structure:

in some embodiments, one or more uridines in the RNA described herein are replaced by modified uridines. In some embodiments, the modified nucleoside is a modified uridine.

In some embodiments, the RNA comprises a modified nucleoside in place of at least one uridine. In some embodiments, the RNA comprises modified nucleosides in place of each uridine.

In some embodiments, the modified nucleoside is independently selected from pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ), and 5-methyl-uridine (m 5U). In some embodiments, the modified nucleoside comprises pseudouridine (ψ). In some embodiments, the modified nucleoside comprises N1-methyl-pseudouridine (m1 ψ). In some embodiments, the modified nucleoside comprises 5-methyl-uridine (m 5U). In some embodiments, the RNA may comprise more than one type of modified nucleoside, and the modified nucleosides are independently selected from pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ), and 5-methyl-uridine (m 5U). In some embodiments, the modified nucleoside comprises pseudouridine (ψ) and N1-methyl-pseudouridine (m1 ψ). In some embodiments, the modified nucleoside comprises pseudouridine (ψ) and 5-methyl-uridine (m 5U). In some embodiments, the modified nucleoside comprises N1-methyl-pseudouridine (m1 ψ) and 5-methyl-uridine (m 5U). In some embodiments, the modified nucleoside comprises pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ), and 5-methyl-uridine (m 5U).

In some embodiments, the modified nucleoside that replaces one or more (e.g., all) uridines in the RNA can be any one or more of: 3-methyl-uridine (m)3U), 5-methoxy-uridine (mo)5U), 5-aza-uridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2-thio-uridine(s)2U), 4-thio-uridine(s)4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-uridine (ho)5U), 5-aminoallyl-uridine, 5-halo-uridine (e.g., 5-iodo-uridine or 5-bromo-uridine), uridine 5-oxoacetic acid (cmo)5U), uridine 5-oxoacetate methyl ester (mcmo)5U), 5-carboxymethyl-uridine (cm)5U), 1-carboxymethyl-pseudouridine, 5-carboxyhydroxymethyl-uridine (chm)5U), 5-carboxyhydroxymethyl-uridine methyl ester (mchm)5U), 5-methoxycarbonylmethyl-uridine (mcm)5U), 5-methoxycarbonylmethyl-2-thio-uridine (mcm)5s2U), 5-aminomethyl-2-thio-uridine (nm)5s2U), 5-methylaminomethyl-uridine (mnm)5U), 1-ethyl-pseudouridine, 5-methylaminomethyl-2-thio-uridine (mnm)5s2U), 5-methylaminomethyl-2-seleno-uridine (mnm)5se2U), 5-carbamoylmethyl-uridine (ncm)5U), 5-carboxymethylaminomethyl-uridine (cmnm)5U), 5-carboxymethyl aminomethyl-2-sulfur Substituted-uridine (cmnm)5s2U), 5-propynyl-uridine, 1-propynyl-pseudouridine, 5-taunomethyl-uridine (. tau.m)5U), 1-tauromethyl-pseudouridine, 5-tauromethyl-2-thio-uridine (. tau.m.sup.5s2U), 1-tauromethyl-4-thio-pseudouridine, 5-methyl-2-thio-uridine (m.sup.m)5s2U), 1-methyl-4-thio-pseudouridine (m)1s4Psi), 4-thio-1-methyl-pseudouridine, 3-methyl-pseudouridine (m)3Psi), 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 (m)5D) 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 (acp)3U), 1-methyl-3- (3-amino-3-carboxypropyl) pseudouridine (acp)3Psi), 5- (isopentenylaminomethyl) uridine (inm)5U), 5- (isopentenylaminomethyl) -2-thio-uridine (inm)5s2U), α -thio-uridine, 2 '-O-methyl-uridine (Um), 5, 2' -O-dimethyl-uridine (m)5Um), 2 '-O-methyl-pseudouridine (ψ m), 2-thio-2' -O-methyl-uridine(s) 2Um), 5-methoxycarbonylmethyl-2' -O-methyl-uridine (mcm)5Um), 5-carbamoylmethyl-2' -O-methyl-uridine (ncm)5Um), 5-carboxymethylaminomethyl-2' -O-methyl-uridine (cmnm)5Um), 3, 2' -O-dimethyl-uridine (m)3Um), 5- (isopentenylaminomethyl) -2' -O-methyl-uridine (inm)5Um), 1-thio-uridine, deoxythymidine, 2 ' -F-arabino-uridine (2 ' -F-ara-uridine), 2 ' -F-uridine, 2 ' -OH-arabino-uridine (2 ' -OH-ara-uridine), 5- (2-methoxycarbonylvinyl) uridine, 5- [3- (1-E-propenylamino) uridine, or any other modified uridine known in the art.

In one embodiment, the RNA comprises other modified nucleosides, or comprises further modified nucleosides, e.g., modified cytidine. For example, in one embodiment, 5-methylcytidine is partially or completely, preferably completely, substituted for cytidine in the RNA. In one embodiment, the RNA comprises 5-methylcytidine and one or more selected from pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ) and 5-methyl-uridine (m 5U). In one embodiment, the RNA comprises 5-methylcytidine and N1-methyl-pseudouridine (m1 ψ). In some embodiments, the RNA comprises 5-methylcytidine in place of each cytidine, and N1-methyl-pseudouridine (m1 ψ) in place of each uridine.

In some embodiments, an RNA according to the present disclosure comprises a 5' -cap. In one embodiment, the RNA of the present disclosure is devoid of uncapped 5' -triphosphates. In one embodiment, the RNA may be modified by a 5' -cap analog. The term "5 '-cap" refers to the structure found on the 5' end of an mRNA molecule and generally consists of guanosine nucleotides linked to the mRNA by 5 '-to 5' -triphosphate linkages. In one embodiment, this guanosine is methylated at the 7-position. Providing RNA with a 5' -cap or 5' -cap analogue can be achieved by in vitro transcription, wherein the 5' -cap is co-transcribed into the RNA strand, or can be post-transcribed to the RNA using a capping enzyme.

In some embodiments, the building block cap of the RNA is m2 7,3’-OGppp(m1 2’-O) ApG (sometimes also referred to as m)2 7,3`OG(5’)ppp(5’)m2’-OApG) having the following structure:

the following is an exemplary Cap1 RNA comprising RNA and m2 7,3`OG(5’)ppp(5’)m2’-OApG:

The following is another exemplary Cap1 RNA (without the Cap analog):

in some embodiments, the RNA is structurally modified with "Cap 0", and in some embodiments, a Cap analog anti-reverse Cap (anti-reverse Cap) (ARCA Cap (m)2 7,3`OG (5 ') ppp (5') G)), having the following structure:

the following are RNA-containing and m 2 7,3`OExemplary Cap0 RNA for G (5 ') ppp (5') G:

in some embodiments, a cap analog having the structure β -S-ARCA (m) is utilized2 7,2`OG (5 ') ppSp (5') G) gives rise to the "Cap 0" structure:

the following are compositions comprising beta-S-ARCA (m)2 7,2`OG (5 ') ppSp (5') G) and exemplary Cap0 RNA of RNA:

the "D1" diastereomer of β -S-ARCA or "β -S-ARCA (D1)" is the diastereomer of β -S-ARCA, which elutes first on the HPLC column and therefore exhibits a shorter retention time than the D2 diastereomer of β -S-ARCA (D2)) (see WO 2011/015347, incorporated herein by reference).

A particularly preferred cap is β -S-ARCA (D1) (m)2 7,2'-OGppSpG) or m2 7,3’-OGppp(m1 2’-O)ApG。

In some embodiments, an RNA according to the present disclosure comprises a 5'-UTR and/or a 3' -UTR. The term "untranslated region" or "UTR" refers to a region in a DNA molecule that is transcribed but not translated into an amino acid sequence, or to a corresponding region in an RNA molecule (e.g., an mRNA molecule). The untranslated region (UTR) may be present 5 '(upstream) (5' -UTR) and/or 3 '(downstream) (3' -UTR) of the open reading frame. If present, the 5'-UTR is located at the 5' end, upstream of the start codon of the protein coding region. The 5' -UTR is located downstream of the 5' -cap (if present), e.g., directly adjacent to the 5' -cap. If present, the 3' -UTR is located at the 3' end, downstream of the stop codon of the protein coding region, but the term "3 ' -UTR" preferably does not include the poly (A) sequence. Thus, the 3' -UTR is located upstream of the poly (A) sequence (if present), e.g., immediately adjacent to the poly (A) sequence.

In some embodiments, the RNA comprises a 5' -UTR comprising the nucleotide sequence of SEQ ID No. 12, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the nucleotide sequence of SEQ ID No. 12.

In some embodiments, the RNA comprises a 3' -UTR comprising the nucleotide sequence of SEQ ID No. 13, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the nucleotide sequence of SEQ ID No. 13.

A particularly preferred 5' -UTR comprises the nucleotide sequence of SEQ ID NO 12. A particularly preferred 3' -UTR comprises SEQ ID NO:13, or a pharmaceutically acceptable salt thereof.

In some embodiments, an RNA according to the present disclosure comprises a 3' -poly (a) sequence.

As used herein, the term "poly (a) sequence" or "poly-a tail" refers to an uninterrupted or interrupted sequence of adenylate residues, which are typically located at the 3' end of an RNA molecule. The poly (a) sequence is known to those skilled in the art and may follow the 3' -UTR in the RNA described herein. The uninterrupted poly (A) sequence is characterized by consecutive adenylate residues. In nature, uninterrupted poly (A) sequences are typical. The RNA disclosed herein may have a poly (a) sequence that is linked to the free 3' end of the RNA after transcription by a template-independent RNA polymerase, or a poly (a) sequence that is encoded by DNA and transcribed by a template-dependent RNA polymerase.

A poly (a) sequence of about 120 a nucleotides has been shown to have a beneficial effect on RNA levels in transfected eukaryotic cells as well as protein levels translated from an open reading frame present upstream (5') of the poly (a) sequence (Holtkamp et al, 2006, Blood, vol.108, pp.4009-4017).

The poly (A) sequence may be of any length. In some embodiments, the poly (a) sequence comprises, consists essentially of, or consists of: at least 20, at least 30, at least 40, at least 80 or at least 100 and up to 500, up to 400, up to 300, up to 200 or up to 150A nucleotides, in particular about 120A nucleotides. In this case, "consisting essentially of …" means that the majority of nucleotides in a 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% of the number of nucleotides in the poly (a) sequence, are a nucleotides, but that the remaining nucleotides are allowed to be nucleotides other than a nucleotides, such as U nucleotides (uridines), G nucleotides (guandines), or C nucleotides (cytidyls). In this case, "consisting of …" means that all nucleotides of the poly (A) sequence, i.e., 100% of the number of nucleotides in the poly (A) sequence, are A nucleotides. The term "A nucleotide" or "A" refers to an adenosine.

In some embodiments, the poly (a) sequence is ligated during RNA transcription, e.g., during preparation of in vitro transcribed RNA, based on a DNA template comprising repeated dT nucleotides (deoxythymidine) in the strand complementary to the coding strand. The DNA sequence (coding strand) encoding the poly (A) sequence is referred to as the poly (A) cassette.

In some embodiments, the poly (a) cassette present in the DNA coding strand consists essentially of dA nucleotides, but is interrupted by a random sequence of 4 nucleotides (dA, dC, dG, and dT). Such random sequences may be 5-50, 10-30, or 10-20 nucleotides in length. Such a cassette is disclosed in WO 2016/005324A 1, which is incorporated herein by reference. Any of the poly (A) cassettes disclosed in WO 2016/005324A 1 may be used in the present invention. A poly (a) cassette is contemplated which essentially consists of dA nucleotides, but is interrupted by a random sequence with 4 nucleotides (dA, dC, dG, dT) distributed equally and with a length of e.g. 5-50 nucleotides, shows a constant proliferation of plasmid DNA in e.coli (e. coli) at the DNA level and still is associated with beneficial properties in supporting RNA stability and translation efficiency at the RNA level. Thus, in some embodiments, the poly (a) sequence comprised in the RNA molecules described herein consists essentially of a nucleotides, but is interrupted by a random sequence of 4 nucleotides (A, C, G, U). Such random sequences may be 5-50, 10-30, or 10-20 nucleotides in length.

In some embodiments, the poly (a) sequence is flanked at its 3 'end by no nucleotides other than a nucleotides, i.e., the poly (a) sequence is not masked or followed at its 3' end by nucleotides other than a.

In some embodiments, the poly (a) sequence may comprise at least 20, at least 30, at least 40, at least 80, or at least 100 and up to 500, up to 400, up to 300, up to 200, or up to 150 nucleotides. In some embodiments, the poly (a) sequence may consist essentially of at least 20, at least 30, at least 40, at least 80, or at least 100 and up to 500, up to 400, up to 300, up to 200, or up to 150 nucleotides. In some embodiments, the poly (a) sequence may consist of at least 20, at least 30, at least 40, at least 80, or at least 100 and up to 500, up to 400, up to 300, up to 200, or up to 150 nucleotides. In some embodiments, the poly (a) sequence comprises at least 100 nucleotides. In some embodiments, the poly (a) sequence comprises about 150 nucleotides. In some embodiments, the poly (a) sequence comprises about 120 nucleotides.

In some embodiments, the RNA comprises a poly (a) sequence comprising the nucleotide sequence of SEQ ID No. 14, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the nucleotide sequence of SEQ ID No. 14.

Particularly preferred poly (A) sequences comprise the nucleotide sequence of SEQ ID NO 14.

According to the present disclosure, the vaccine antigen is preferably administered as a single-stranded, 5' -capped mRNA that is translated into the corresponding protein upon entry into the cells of the subject to which the RNA is administered. Preferably, the RNA comprises structural elements (5' -cap, 5' -UTR, 3' -UTR, poly (a) sequence) optimized for the maximum potency of the RNA in terms of stability and translational efficiency.

In one embodiment, β -S-ARCA (D1) is used as a specific capping structure at the 5' end of the RNA. In one embodiment, m2 7,3’-OGppp(m1 2’-O) ApG serve as a specific capping structure for the 5' end of the RNA. In one embodiment, the 5' -UTR sequence is derived from human α -globin mRNA, and optionally has an optimized "Kozak sequence" to improve translation efficiency. In one embodiment, a combination of two sequence elements derived from the "split amino terminal enhancer of split" (AES) mRNA (referred to as F) and the mitochondrially encoded 12S ribosomal RNA (referred to as I) (FI element) is placed between the coding sequence and the poly (a) sequence to ensure higher maximum protein levels and extended mRNA persistence. In one embodiment, two repeated 3' -UTRs derived from human β -globin mRNA are placed between the coding sequence and the poly (a) sequence to ensure higher maximum protein levels and extended mRNA persistence. In one embodiment, a poly (a) sequence of 110 nucleotides in length is used, consisting of a stretch of 30 adenosine residues, followed by a 10 nucleotide linker sequence and another 70 adenosine residues. This poly (A) sequence was designed to enhance RNA stability and translation efficiency.

In one embodiment of all aspects of the invention, RNA encoding a vaccine antigen is expressed in cells of the treated subject to provide the vaccine antigen. In one embodiment of all aspects of the invention, the RNA is transiently expressed in the cells of the subject. In one embodiment of all aspects of the invention, the RNA is an in vitro transcribed RNA. In one embodiment of all aspects of the invention, the expression of the vaccine antigen is at the cell surface. In one embodiment of all aspects of the invention, the vaccine antigen is expressed and presented in the context of MHC. In one embodiment of all aspects of the invention, the expression of the vaccine antigen into the extracellular space, i.e. the vaccine antigen is secreted.

In the context of the present disclosure, the term "transcription" relates to a process in which a genetic code in a DNA sequence is transcribed into RNA. Subsequently, the RNA can be translated into a peptide or protein.

According to the present invention, the term "transcription" comprises "in vitro transcription", wherein the term "in vitro transcription" relates to a process wherein RNA, in particular mRNA, is synthesized in vitro in a cell-free system, preferably using a suitable cell extract. Preferably, cloning vectors are used to produce transcripts. These cloning vectors are generally designated as transcription vectors and are encompassed within the term "vector" according to the present invention. According to the invention, the RNA used in the invention is preferably an in vitro transcribed RNA (IVT-RNA) and can be obtained by in vitro transcription of an appropriate DNA template. The promoter used to control transcription may be any promoter of any RNA polymerase. Particular examples of RNA polymerases are T7, T3 and SP6 RNA polymerase. Preferably, in vitro transcription according to the invention is controlled by the T7 or SP6 promoters. DNA templates for in vitro transcription can be obtained by cloning nucleic acids, in particular cDNA, and introducing them into an appropriate vector for in vitro transcription. cDNA can be obtained by reverse transcription of RNA.

With respect to RNA, the terms "expression" or "translation" relate to the process in the nuclear sugar body by which an mRNA strand directs the assembly of amino acid sequences to produce a peptide or protein.

In one embodiment, at least a portion of the RNA is delivered to the target cell following administration of the RNA described herein, e.g., formulated as an RNA lipid particle. In one embodiment, at least a portion of the RNA is delivered to the cytosol of the target cell. In one embodiment, the RNA is translated by the target cell to produce the peptide or protein encoded thereby. In one embodiment, the target cell is a spleen cell. In one embodiment, the target cell is an antigen presenting cell, such as a professional antigen presenting cell in the spleen. In one embodiment, the target cell is a dendritic cell or macrophage. The RNA particles (e.g., RNA lipid particles) described herein can be used to deliver RNA to such target cells. Accordingly, the present disclosure also relates to a method of delivering RNA to a target cell in a subject, the method comprising administering to the subject an RNA particle described herein. In one embodiment, the RNA is delivered to the cytosol of the target cell. In one embodiment, the RNA is translated by the target cell to produce a peptide or protein encoded by the RNA.

"encoding" refers to the inherent property of a particular nucleotide sequence in a polynucleotide (e.g., a gene, cDNA, or mRNA) to serve as a template for the synthesis of other polymers or macromolecules in biological processes having defined nucleotide sequences (i.e., rRNA, tRNA and mRNA) or defined amino acid sequences and the biological properties resulting therefrom. Thus, a gene encodes a protein if transcription and translation of the mRNA corresponding to the gene produces the protein in a cell or other biological system. Both the coding strand, whose nucleotide sequence is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand, which serves as a template for transcription of a gene or cDNA, can be referred to as the protein or other product that encodes the gene or cDNA.

In one embodiment, the RNA encoding the vaccine antigen administered according to the invention is non-immunogenic. RNA encoding an immunostimulant may be administered according to the invention to provide an adjuvant effect. The RNA encoding the immunostimulant may be a standard RNA or a non-immunogenic RNA.

The term "non-immunogenic RNA" as used herein refers to RNA that does not induce an immune system response when administered to, for example, a mammal, or that induces a response that differs from that induced by the same RNA that has not been modified and treated to render the non-immunogenic RNA non-immunogenic, i.e., weaker than that induced by standard RNA (stdrna). In a preferred embodiment, non-immunogenic RNA, also referred to herein as modified RNA (modrna), is rendered non-immunogenic by incorporating modified nucleosides into the RNA that inhibit RNA-mediated activation of innate immune receptors and removing double-stranded RNA (dsrna).

In order to render non-immunogenic R by incorporation of modified nucleosidesNA is rendered non-immunogenic and any modified nucleoside can be used so long as it reduces or inhibits the immunogenicity of the RNA. Particularly preferred are modified nucleosides that inhibit RNA-mediated activation of innate immune receptors. In one embodiment, the modified nucleoside comprises the replacement of one or more uridines with a nucleoside comprising a modified nucleobase. In one embodiment, the modified nucleobase is a modified uracil. In one embodiment, the nucleoside comprising a modified nucleobase is selected from 3-methyl-uridine (m)3U), 5-methoxy-uridine (mo)5U), 5-azauridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2-thio-uridine(s)2U), 4-thio-uridine(s)4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-uridine (ho)5U), 5-aminoallyl-uridine, 5-halo-uridine (e.g., 5-iodo-uridine or 5-bromo-uridine), uridine 5-oxoacetic acid (cmo)5U), uridine 5-oxoacetate methyl ester (mcmo)5U), 5-carboxymethyl-uridine (cm)5U), 1-carboxymethyl-pseudouridine, 5-carboxyhydroxymethyl-uridine (chm)5U), 5-carboxyhydroxymethyl-uridine methyl ester (mchm)5U), 5-methoxycarbonylmethyl-uridine (mcm) 5U), 5-methoxycarbonylmethyl-2-thio-uridine (mcm)5s2U), 5-aminomethyl-2-thio-uridine (nm)5s2U), 5-methylaminomethyl-uridine (mnm)5U), 1-ethyl-pseudouridine, 5-methylaminomethyl-2-thio-uridine (mnm)5s2U), 5-methylaminomethyl-2-seleno-uridine (mnm)5se2U), 5-carbamoylmethyl-uridine (ncm)5U), 5-carboxymethylaminomethyl-uridine (cmnm)5U), 5-carboxymethylaminomethyl-2-thio-uridine (cmnm)5s2U), 5-propynyl-uridine, 1-propynyl-pseudouridine, 5-taunomethyl-uridine (. tau.m)5U), 1-taunomethyl-pseudouridine, 5-taunomethyl-2-thio-uridine (. tau.m.sup.5s2U), 1-taunomethyl-4-thio-pseudouridine, 5-methyl-2-thio-uridine (m.sup.m)5s2U), 1-methyl-4-thio-pseudouridine (m)1s4Psi), 4-thio-1-methyl-pseudouridine, 3-methyl-pseudouridine (m)3Psi), 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 (m)5D) 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 (acp) 3U), 1-methyl-3- (3-amino-3-carboxypropyl) pseudouridine (acp)3Psi), 5- (isopentenylaminomethyl) uridine (inm)5U), 5- (isopentenylaminomethyl) -2-thio-uridine (inm)5s2U), α -thio-uridine, 2 '-O-methyl-uridine (Um), 5, 2' -O-dimethyl-uridine (m)5Um), 2 '-O-methyl-pseudouridine (ψ m), 2-thio-2' -O-methyl-uridine(s)2Um), 5-methoxycarbonylmethyl-2' -O-methyl-uridine (mcm)5Um), 5-carbamoylmethyl-2' -O-methyl-uridine (ncm)5Um), 5-carboxymethylaminomethyl-2' -O-methyl-uridine (cmnm)5Um), 3, 2' -O-dimethyl-uridine (m)3Um), 5- (isopentenylaminomethyl) -2' -O-methyl-uridine (inm)5Um), 1-thio-uridine, deoxythymidine, 2 ' -F-arabino-uridine, 2 ' -F-uridine, 2 ' -OH-arabino-uridine, 5- (2-methoxycarbonylethenyl) uridine and 5- [3- (1-E-propenylamino) uridine. In a particularly preferred embodiment, the nucleoside comprising a modified nucleobase is a pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ) or 5-methyl-uridine (m5U), in particular N1-methyl-pseudouridine.

In one embodiment, the replacement of one or more uridines with a nucleoside comprising a modified nucleobase comprises replacing at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 10%, at least 25%, at least 50%, at least 75%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of a uridine.

During the synthesis of mRNA by In Vitro Transcription (IVT) using T7 RNA polymerase, a number of abnormal products, including double-stranded RNA (dsrna), are produced due to the non-canonical activity of the enzyme. dsRNA induces inflammatory cytokines and activates effector enzymes, resulting in inhibition of protein synthesis. dsRNA can be removed from RNA (e.g., IVT RNA) by, for example, ion-pair reverse phase HPLC using a non-porous or porous C-18 polystyrene-divinylbenzene (PS-DVB) matrix. Alternatively, an enzyme-based approach can be used to eliminate dsRNA contaminants from IVT RNA preparations using e.coli RNaseIII that specifically hydrolyzes dsRNA but not ssRNA. In addition, dsRNA can be separated from ssRNA by using cellulosic material. In one embodiment, the RNA preparation is contacted with the cellulosic material and the ssRNA is isolated from the cellulosic material under conditions that allow the dsRNA to bind to the cellulosic material and do not allow the ssRNA to bind to the cellulosic material.

As the term is used herein, "remove" or "removal" refers to the feature of a first population of substances (e.g., non-immunogenic RNA) as being separated from the vicinity of a second population of substances (e.g., dsRNA), wherein the first population of substances is not necessarily completely free of the second substance, and the second population of substances is not necessarily completely free of the first substance. However, a first species population characterized by the removal of a second species population has a measurably lower content of the second species than an unseparated mixture of the first and second species.

In one embodiment, removing dsRNA from the non-immunogenic RNA comprises removing dsRNA such that 10% or less, 5% or less, 4% or less, 3% or less, 2% or less, 1% or less, 0.5% or less, 0.3% or less, or 0.1% or less of RNA in the non-immunogenic RNA composition is dsRNA. In one embodiment, the non-immunogenic RNA is free or substantially free of dsRNA. In some embodiments, the non-immunogenic RNA composition comprises a purified preparation of nucleoside-modified single-stranded RNA. For example, in some embodiments, a purified preparation of nucleoside-modified single-stranded RNA is substantially free of double-stranded RNA (dsrna). In some embodiments, the purified preparation is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, or at least 99.9% nucleoside-modified single-stranded RNA relative to all other nucleic acid molecules (DNA, dsRNA, etc.).

In one embodiment, the non-immunogenic RNA is translated more efficiently in the cell than a standard RNA having the same sequence. In one embodiment, the fold increase in translation is 2 fold relative to its unmodified counterpart. In one embodiment, the fold of translational enhancement is 3 fold. In one embodiment, the fold of translational enhancement is 4 fold. In one embodiment, the fold of translational enhancement is 5 fold. In one embodiment, the fold of translational enhancement is 6 fold. In one embodiment, the fold of translational enhancement is 7 fold. In one embodiment, the fold of translational enhancement is 8 fold. In one embodiment, the fold of translational enhancement is 9 fold. In one embodiment, the fold of translational enhancement is 10 fold. In one embodiment, the fold of translational enhancement is 15 fold. In one embodiment, the fold of translational enhancement is 20 fold. In one embodiment, the fold of translational enhancement is 50 fold. In one embodiment, the fold of translational enhancement is 100 fold. In one embodiment, the fold of translational enhancement is 200 fold. In one embodiment, the fold of translational enhancement is 500 fold. In one embodiment, the fold of translational enhancement is 1000 fold. In one embodiment, the fold increase in translation is 2000-fold. In one embodiment, the factor is 10 to 1000. In one embodiment, the factor is 10 to 100. In one embodiment, the factor is 10 to 200. In one embodiment, the factor is 10 to 300. In one embodiment, the factor is 10 to 500 times. In one embodiment, the factor is 20 to 1000. In one embodiment, the factor is 30 to 1000. In one embodiment, the factor is 50 to 1000. In one embodiment, the factor is 100-fold and 1000-fold. In one embodiment, the factor is 200-and 1000-fold. In one embodiment, translation is enhanced by any other significant amount or range of amounts.

In one embodiment, the non-immunogenic RNA exhibits significantly lower innate immunogenicity as compared to standard RNA having the same sequence. In one embodiment, the non-immunogenic RNA exhibits a 2-fold less innate immune response than its unmodified counterpart. In one embodiment, the reduction in innate immunogenicity is by a factor of 3. In one embodiment, the reduction in innate immunogenicity is by a factor of 4. In one embodiment, the reduction in innate immunogenicity is by a factor of 5. In one embodiment, the reduction in innate immunogenicity is by a factor of 6. In one embodiment, the reduction in innate immunogenicity is by a factor of 7. In one embodiment, the reduction in innate immunogenicity is by a factor of 8. In one embodiment, the reduction in innate immunogenicity is by a factor of 9. In one embodiment, the reduction in innate immunogenicity is by a factor of 10. In one embodiment, the reduction in innate immunogenicity is by a factor of 15. In one embodiment, the reduction in innate immunogenicity is by a factor of 20. In one embodiment, the reduction in innate immunogenicity is by a factor of 50. In one embodiment, the reduction in innate immunogenicity is by a factor of 100. In one embodiment, the reduction in innate immunogenicity is by a factor of 200. In one embodiment, the reduction in innate immunogenicity is by a factor of 500. In one embodiment, the reduction in innate immunogenicity is by a factor of 1000. In one embodiment, the reduction in innate immunogenicity is a factor of 2000.

The term "exhibits significantly less innate immunogenicity" refers to a detectable reduction in innate immunogenicity. In one embodiment, the term refers to a decrease such that an effective amount of non-immunogenic RNA can be administered without triggering a detectable innate immune response. In one embodiment, the term refers to a decrease such that the non-immunogenic RNA can be repeatedly administered without eliciting an innate immune response sufficient to detectably reduce production of the protein encoded by the non-immunogenic RNA. In one embodiment, the reduction allows for repeated administration of the non-immunogenic RNA without eliciting an innate immune response sufficient to abrogate detectable production of the protein encoded by the non-immunogenic RNA.

"immunogenicity" is the ability of a foreign substance (e.g., RNA) to elicit an immune response in a human or other animal. The innate immune system is an integral part of the immune system, which is relatively nonspecific and immediate. It is one of the two major components of the vertebrate immune system, along with the adaptive immune system.

As used herein, "endogenous" refers to any substance that is derived from or produced within an organism, cell, tissue, or system.

As used herein, the term "exogenous" refers to any substance introduced from or produced outside of an organism, cell, tissue, or system.

The term "expression" as used herein is defined as the transcription and/or translation of a particular nucleotide sequence.

As used herein, the terms "linked," "fused," or "fused" are used interchangeably. These terms refer to two or more elements or components or domains linked together.

Codon optimization/increase in G/C content

In some embodiments, the amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof described herein is encoded by a coding sequence that is codon optimized and/or has increased G/C content as compared to the wild-type coding sequence. This also includes embodiments wherein one or more sequence regions of the coding sequence are codon optimized and/or have an increased G/C content as compared to the corresponding sequence region of the wild-type coding sequence. In one embodiment, codon optimization and/or an increase in G/C content preferably does not alter the sequence of the encoded amino acid sequence.

The term "codon-optimized" refers to altering codons in the coding region of a nucleic acid molecule to reflect typical codon usage of a host organism, preferably without altering the amino acid sequence encoded by the nucleic acid molecule. In the context of the present invention, the coding region is preferably codon-optimized for optimal expression in the subject to be treated using the RNA molecules described herein. Codon optimization was based on the following findings: the efficiency of translation is also determined by the different frequencies of tRNA occurrence in the cell. Thus, the sequence of the RNA can be modified to insert codons that can acquire frequently occurring tRNA's in place of "rare codons".

In some embodiments of the invention, the guanosine/cytosine (G/C) content of the coding regions of the RNAs described herein is increased as compared to the G/C content of the corresponding coding sequence of the wild-type RNA, wherein the amino acid sequence encoded by the RNA is preferably unmodified as 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 the mRNA. Sequences with increased G (guanosine)/C (cytosine) content are more stable than sequences with increased a (adenosine)/U (uracil) content. With regard to the fact that several codons encode one and the same amino acid (so-called degeneracy of the genetic code), the codon that is most favorable for stability (so-called alternative codon usage) can be determined. Depending on the amino acid encoded by the RNA, there are various possibilities for modification of the RNA sequence compared to its wild-type sequence. In particular, codons comprising a and/or U nucleotides may be modified by substituting these codons with other codons encoding the same amino acid but lacking a and/or U or comprising lower amounts of a and/or U nucleotides.

In various embodiments, the G/C content of the coding region of an RNA described herein is increased by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 55%, or even more compared to the G/C content of the coding region of a wild-type RNA.

Embodiments of the RNA administered

In some embodiments, the compositions or pharmaceutical preparations described herein comprise an RNA that encodes an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof. Likewise, the methods described herein include the administration of such RNA.

The activity platform for use herein is based on an RNA vaccine encoding an antigen in order to induce robust neutralizing antibodies and concomitant T cell responses to achieve protective immunity with the preferred minimum vaccine dose. The RNA administered is preferably in vitro transcribed RNA.

Three different RNA platforms, namely unmodified uridine-containing mrna (unrna), nucleoside-modified mrna (modrna) and self-amplifying RNA (sarna), are particularly preferred. In a particularly preferred embodiment, the RNA is an in vitro transcribed RNA.

As described herein, embodiments of each of these platforms are evaluated herein (see, e.g., example 2), representing a novel and powerful method and system for rapid vaccine development. The methods and systems have met with significant and effective success, enabling the development of effective clinical candidates within several months of providing an antigen (e.g., SARS-CoV-2S 1 protein and/or its RBD) sequence (relevant sequence information (e.g., GenBank: MN908947.3) is available in 1 month 2020). The insights and advantages embodied in the methods and systems include, for example, the ability to directly compare one or more features of different policies to enable rapid, efficient, and effective development. Among other things, the present disclosure encompasses the insight that more typical strategies for vaccine development identify the source of the problem. Moreover, the discoveries included herein establish various advantages and benefits, particularly with particular benefits in rapid vaccine development, particularly in pandemics.

As described herein, in some embodiments, the vaccine candidate is evaluated for the titer of antibodies induced in the model organism (e.g., mouse; see, e.g., example 2) against the encoded antigen (e.g., S1 protein) or portion thereof (e.g., RBD). In some embodiments, vaccine candidates are evaluated for pseudovirus neutralization (see, e.g., example 2) activity of the induced antibodies. In some embodiments, vaccine candidates are characterized by the nature of the induced T cell response (e.g., T)H1 vs TH2, performing the following steps; see, e.g., example 4). In some embodiments, vaccine candidates are evaluated in more than one model organism (see, e.g., example 2, example 4, etc.).

In the following, embodiments of these three different RNA platforms are described, wherein certain terms used in describing the elements thereof have the following meanings:

S1S2 protein/S1S 2 RBD: sequences encoding the respective antigens of SARS-CoV-2.

nsP1, nsP2, nsP3 and nsP 4: wild-type sequences encoding Venezuelan Equine Encephalitis Virus (VEEV) RNA-dependent RNA polymerase replicase and subgenomic promoters, and conserved sequence elements that support replication and translation.

virUTR: a viral untranslated region encoding a portion of a subgenomic promoter and elements of replication and translation support sequences.

hAg-Kozak: 5' -UTR sequence of human alpha-globin mRNA with optimized "Kozak sequence" to improve translation efficiency.

And c: sec corresponds to the intrinsic S1S2 protein secretion signal peptide (Sec), which directs translocation of the nascent polypeptide chain to the endoplasmic reticulum.

Glycine-serine linker (GS): sequences encoding short linker peptides consisting essentially of the amino acids glycine (G) and serine (S) are commonly used for fusion proteins.

Secondary fibrin: t4 partial sequence of the minor fibrin (foldon), used as an artificial trimerization domain.

TM: the TM sequence corresponds to the transmembrane portion of the S1S2 protein.

An FI element: the 3' -UTR is a combination of two sequence elements derived from the "split amino terminal enhancer" (AES) mRNA (referred to as F) and the mitochondrially encoded 12S ribosomal RNA (referred to as I). These were identified by an ex vivo selection process for sequences that confer RNA stability and increased total protein expression.

A30L 70: a poly (a) -tail of 110 nucleotides in length, consisting of a stretch of 30 adenosine residues followed by a 10 nucleotide linker sequence and another stretch of 70 adenosine residues, designed to enhance RNA stability and translation efficiency in dendritic cells.

In general, the vaccine RNA described herein can comprise from 5 'to 3' one of the following structures:

cap-5 '-UTR-vaccine antigen-coding sequence-3' -UTR-Poly (A)

Or

beta-S-ARCA (D1) -hAg-Kozak-vaccine antigen-coding sequence-FI-A30L 70.

In general, a vaccine antigen described herein can comprise from N-terminus to C-terminus one of the following structures:

signal sequence-RBD-trimerization domain

Or

Signal sequence-RBD-trimerization domain-transmembrane domain.

The RBD and trimerization domains may be separated by a linker, in particular a GS linker such as a linker having the amino acid sequence GSPGSGSGS. The trimerization domain and the transmembrane domain may be separated by a linker, in particular a GS linker such as a linker having the amino acid sequence GSGSGS.

The signal sequence may be a signal sequence as described herein. The RBD may be an RBD domain as described herein. The trimerization domain may be a trimerization domain as described herein. The transmembrane domain may be a transmembrane domain as described herein.

In one embodiment of the process of the present invention,

the signal sequence comprises SEQ ID NO: 1 or 1-19 or SEQ ID NO: 31, or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to this amino acid sequence,

The RBD comprises SEQ ID NO:1, or an amino acid sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to this amino acid sequence,

the trimerizing domain comprises the amino acid sequence of amino acids 3-29 of SEQ ID NO. 10 or the amino acid sequence of SEQ ID NO. 10 or an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to this amino acid sequence; and is

The transmembrane domain comprises the amino acid sequence of amino acid 1207-1254 of SEQ ID NO. 1 or an amino acid sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to this amino acid sequence.

In one embodiment of the process of the present invention,

the signal sequence comprises SEQ ID NO:1 or 1-19 or SEQ ID NO: 31 of amino acids 1 to 22,

the RBD comprises SEQ ID NO:1 amino acid 327-528 of the sequence,

the trimerization domain comprises the amino acid sequence of amino acids 3-29 of SEQ ID NO. 10 or the amino acid sequence of SEQ ID NO. 10; and is

The transmembrane domain comprises the amino acid sequence of amino acids 1207-1254 of SEQ ID NO: 1.

The RNA or RNA encoding the vaccine antigen may be unmodified uridine-containing mrna (unrna), nucleoside-modified mrna (modrna), or self-amplifying RNA (sarna). In one embodiment, the RNA or RNA encoding the vaccine antigen is a nucleoside-modified mrna (modrna).

Unmodified uridine messenger RNA (uRNA)

The active ingredient of an unmodified messenger rna (unrna) drug is a single-stranded mRNA that is translated upon entry into a cell. In addition to the sequences encoding the coronavirus vaccine antigens (i.e. open reading frames), each uRNA preferably comprises common structural elements (5 ' -cap, 5 ' -UTR, 3 ' -UTR, poly (A) -tail) optimized for the maximal potency of the RNA in terms of stability and translation efficiency. A preferred 5' cap structure is β -S-ARCA (D1) (m)2 7,2'-OGppsg). Preferred 5 '-UTR and 3' -UTR comprise the nucleotide sequence of SEQ ID NO. 12 and the nucleotide sequence of SEQ ID NO. 13, respectively. A preferred poly (A) -tail comprises the sequence of SEQ ID NO 14.

Various embodiments of this platform are as follows:

RBL063.1(SEQ ID NO:15;SEQ ID NO:7)

the structure is beta-S-ARCA (D1) -hAg-Kozak-S1S2-PP-FI-A30L70

Viral spike protein (S1S2 protein) of the encoded antigen SARS-CoV-2 (S1S2 full-length protein, sequence variants)

RBL063.2(SEQ ID NO:16;SEQ ID NO:7)

The structure is beta-S-ARCA (D1) -hAg-Kozak-S1S2-PP-FI-A30L70

Viral spike protein (S1S2 protein) of the encoded antigen SARS-CoV-2 (S1S2 full-length protein, sequence variants)

BNT162a1;RBL063.3(SEQ ID NO:17;SEQ ID NO:5)

Structure beta-S-ARCA (D1) -hAg-Kozak-RBD-GS-Secondary fibrin-FI-A30L 70

Viral spike protein (S protein) of the encoded antigen SARS-CoV-2 (partial sequence, Receptor Binding Domain (RBD) of the S1S2 protein)

FIG. 19 illustrates the general structure of an RNA encoding an antigen.

Nucleotide sequence of RBL063.1

Nucleotide sequences are shown having individual sequence elements as indicated by bold letters. In addition, the sequence of the translated protein is shown in italics below the coding nucleotide sequence (═ stop codon).

Nucleotide sequence of RBL063.2

Nucleotide sequences are shown having individual sequence elements as indicated by bold letters. In addition, the sequence of the translated protein is shown in italics below the coding nucleotide sequence (═ stop codon).

Nucleotide sequence of RBL063.3

Nucleotide sequences are shown having individual sequence elements as indicated by bold letters. In addition, the sequence of the translated protein is shown in italics below the coding nucleotide sequence (═ stop codon).

Nucleoside modified messenger RNA (modRNA)

The active ingredient of a nucleoside-modified messenger rna (modrna) drug is also a single-stranded mRNA that is translated upon entry into a cell. In addition to sequences encoding coronavirus vaccine antigens (i.e. open reading frames),each modRNA contains common structural elements (5 ' -cap, 5 ' -UTR, 3 ' -UTR, poly (A) -tail) optimized for maximum potency of the RNA, as does uRNA. Compared to uRNA, modRNA contains 1-methyl-pseudouridine instead of uridine. The preferred 5' cap structure is m2 7,3’-OGppp(m1 2’-O) ApG are provided. Preferred 5 '-UTR and 3' -UTR comprise the nucleotide sequence of SEQ ID NO. 12 and the nucleotide sequence of SEQ ID NO. 13, respectively. A preferred poly (A) -tail comprises the sequence of SEQ ID NO 14. Additional purification steps were applied to modRNA to reduce dsRNA contaminants produced during the in vitro transcription reaction.

Various embodiments of this platform are as follows:

BNT162b2;RBP020.1(SEQ ID NO:19;SEQ ID NO:7)

structure m2 7,3’-OGppp(m1 2’-O)ApG)-hAg-Kozak-S1S2-PP-FI-A30L70

Viral spike protein (S1S2 protein) of the encoded antigen SARS-CoV-2 (S1S2 full-length protein, sequence variants)

BNT162b2;RBP020.2(SEQ ID NO:20;SEQ ID NO:7)

Structure m2 7,3’-OGppp(m1 2’-O)ApG)-hAg-Kozak-S1S2-PP-FI-A30L70

Viral spike protein (S1S2 protein) of the encoded antigen SARS-CoV-2 (S1S2 full-length protein, sequence variants)

BNT162b1;RBP020.3(SEQ ID NO:21;SEQ ID NO:5)

Structure m2 7,3’-OGppp(m1 2’-O) ApG) -hAg-Kozak-RBD-GS-Secondibrin-FI-A30L 70

Viral spike protein (S1S2 protein) of the encoded antigen SARS-CoV-2 (partial sequence, Receptor Binding Domain (RBD) of S1S2 protein fused to minor fibrin)

FIG. 20 illustrates the general structure of an RNA encoding an antigen.

Nucleotide sequence of RBP020.1

Nucleotide sequences are shown having individual sequence elements as indicated by bold letters. In addition, the sequence of the translated protein is shown in italics below the coding nucleotide sequence (═ stop codon).

Nucleotide sequence of RBP020.2

Nucleotide sequences are shown having individual sequence elements as indicated by bold letters. In addition, the sequence of the translated protein is shown in italics below the coding nucleotide sequence (═ stop codon).

RBP020.3 nucleotide sequence

Nucleotide sequences are shown having individual sequence elements as indicated by bold letters. In addition, the sequence of the translated protein is shown in italics below the coding nucleotide sequence (═ stop codon).

Other embodiments of the nucleoside-modified messenger rna (modrna) platform are as follows:

BNT162b3c(SEQ ID NO:29;SEQ ID NO:30)

structure m2 7,3’-OGppp(m1 2’-O) ApG-hAg-Kozak-RBD-GS-Secondary fibrin-GS-TM-FI-A30L 70

The viral spike protein (S1S2 protein) of the encoded antigen SARS-CoV-2 (partial sequence, Receptor Binding Domain (RBD) of S1S2 protein, fused to secondary fibrin, fused to transmembrane domain (TM) of S1S2 protein);

The intrinsic S1S2 protein secretion signal peptide at the N-terminus of the antigen sequence (aa 1-19)

BNT162b3d(SEQ ID NO:31;SEQ ID NO:32)

Structure m2 7,3’-OGppp(m1 2’-O) ApG-hAg-Kozak-RBD-GS-Secondary fibrin-GS-TM-FI-A30L 70

The viral spike protein (S1S2 protein) of the encoded antigen SARS-CoV-2 (partial sequence, Receptor Binding Domain (RBD) of S1S2 protein, fused to secondary fibrin, fused to transmembrane domain (TM) of S1S2 protein);

immunoglobulin secretion signal peptide at the N-terminus of the antigen sequence (aa 1-22)

Self-amplifying RNA (sarna)

The active ingredient of self-amplifying mrna (sarna) drugs is single-stranded RNA, which self-amplifies upon entry into cells, after which coronavirus vaccine antigens are translated. In contrast to the uRNA and modRNA, which preferably encode a single protein, the coding region of the sarRNA contains two Open Reading Frames (ORFs). The 5' -ORF encodes an RNA-dependent RNA polymerase such as Venezuelan Equine Encephalitis Virus (VEEV) RNA-dependent RNA polymerase (replicase). The replicase ORF is followed 3' by a subgenomic promoter and a second ORF encoding an antigen. In addition, the saRNA UTR contains 5 'and 3' Conserved Sequence Elements (CSEs) required for self-amplification. The saRNA comprisesCommon structural elements (5 ' -cap, 5 ' -UTR, 3 ' -UTR, poly (a) -tail) optimized for maximum potency of RNA, like u RNA. The saRNA preferably comprises uridine. A preferred 5' cap structure is β -S-ARCA (D1) (m) 2 7,2'-OGppSpG)。

Cytoplasmic delivery of saRNA initiates an alphavirus-like life cycle. However, saRNA does not encode the alphavirus structural proteins required for genomic packaging or cellular entry, and thus it is highly unlikely to be possible to produce replication-competent viral particles. Replication does not involve any intermediate steps in the production of DNA. Thus, the use/uptake of saRNA does not risk genomic integration or other permanent genetic modification in the target cell. In addition, saRNA itself prevents its sustained replication by recognizing dsRNA intermediates that effectively activate the innate immune response.

Various embodiments of this platform are as follows:

RBS004.1(SEQ ID NO:24;SEQ ID NO:7)

structure beta-S-ARCA (D1) -replicase-S1S 2-PP-FI-A30L70

Viral spike protein (S protein) of the encoded antigen SARS-CoV-2 (S1S2 full length protein, sequence variants)

RBS004.2(SEQ ID NO:25;SEQ ID NO:7)

Structure beta-S-ARCA (D1) -replicase-S1S 2-PP-FI-A30L70

Viral spike protein (S protein) of the encoded antigen SARS-CoV-2 (S1S2 full length protein, sequence variants)

BNT162c1;RBS004.3(SEQ ID NO:26;SEQ ID NO:5)

Structure beta-S-ARCA (D1) -replicase-RBD-GS-Secondary fibrin-FI-A30L 70

Viral spike protein (S protein) of the encoded antigen SARS-CoV-2 (partial sequence, Receptor Binding Domain (RBD) of the S1S2 protein)

RBS004.4(SEQ ID NO:27;SEQ ID NO:28)

Structure beta-S-ARCA (D1) -replicase-RBD-GS-Secondary fibrin-TM-FI-A30L 70

Viral spike protein (S protein) of the encoded antigen SARS-CoV-2 (partial sequence, Receptor Binding Domain (RBD) of the S1S2 protein)

FIG. 21 illustrates the general structure of an RNA encoding an antigen.

Nucleotide sequence of RBS004.1

Nucleotide sequences are shown having individual sequence elements as indicated by bold letters. In addition, the sequence of the translated protein is shown in italics below the coding nucleotide sequence (═ stop codon).

Nucleotide sequence of RBS004.2

Nucleotide sequences are shown having individual sequence elements as indicated by bold letters. In addition, the sequence of the translated protein is shown in italics below the coding nucleotide sequence (═ stop codon).

Nucleotide sequence of RBS004.3

Nucleotide sequences are shown having individual sequence elements as indicated by bold letters. In addition, the sequence of the translated protein is shown in italics below the coding nucleotide sequence (═ stop codon).

Nucleotide sequence of RBS004.4

Nucleotide sequences are shown having individual sequence elements as indicated by bold letters. In addition, the sequence of the translated protein is shown in italics below the coding nucleotide sequence (═ stop codon).

In some embodiments, the vaccine RNA described herein comprises a sequence selected from SEQ ID NOs: 15. 16, 17, 19, 20, 21, 24, 25, 26, 27, 30 and 32. Particularly preferred vaccine RNAs described herein comprise a sequence selected from SEQ ID NOs: 15. 17, 19, 21, 25, 26, 30 and 32, as selected from the group consisting of SEQ ID NOs: 17. 19, 21, 26, 30 and 32.

The RNA described herein is preferably formulated in Lipid Nanoparticles (LNPs). In one embodiment, the LNP comprises a cationic lipid, a neutral lipid, a steroid, a polymer conjugated lipid; and the RNA. In an embodiment, the cationic lipid is ALC-0315, the neutral lipid is DSPC, the steroid is cholesterol, and the polymer-conjugated lipid is ALC-0159. The preferred mode of administration is intramuscular, more preferably in aqueous cryoprotectant buffer for intramuscular administration. The pharmaceutical product is preferably a preservative-free sterile dispersion of RNA formulated in Lipid Nanoparticles (LNPs) in an aqueous cryoprotectant buffer for intramuscular administration.

In various embodiments, the pharmaceutical product comprises the components shown below, preferably in the ratios or concentrations shown below:

[1] ALC-0315 ═ bis (hexane-6,1-diyl) bis (hexyl 2-decanoate) ((4-hydroxybutyl) azanediyl) bis (hexyl-6- (2-hexyldecanoyloxy) hexyl-N- (4-hydroxybutyl) amino ] hexyl 2-decanoate (((4-hydroxybutanoyl) azanediyl) bis (2-hexanodecanoate)/6- [ N-6- (2-hexanoyldiyloxy) hexyl-N- (4-hydroxybutanoyl) amino ] hexanoyl 2-hexanodecanoate)

[2] ALC-0159 ═ 2- [ (polyethylene glycol) -2000] -N, N-ditetradecylacetamide/2- [2- (ω -methoxy (polyethylene glycol2000) ethoxy ] -N, N-ditetradecylacetamide (2- [ (polyethylene glycol) -2000] -N, N-ditetradecylacetamide/2- [2- (ω -methoxy (polyethylene glycol2000) ethoxy ] -N, N-ditetradecylacetamide)

[3] The amount of DSPC ═ 1,2-Distearoyl-sn-glycero-3-phosphocholine (1, 2-stearoyl-sn-glycero-3-phosphocholine) q.s. ═ right amount (as far as possible enough)

ALC-0315:

ALC-0159:

DSPC:

Cholesterol:

in one embodiment, the ratio of mRNA to total lipid (N/P) is between 6.0 and 6.5, such as about 6.0 or about 6.3.

Particles comprising nucleic acids

Nucleic acids described herein (e.g., RNA encoding a vaccine antigen) can be formulated for administration as particles.

In the context of the present disclosure, the term "particle" relates to a structured entity formed by molecules or molecular complexes. In one embodiment, the term "particle" relates to a micro-or nano-sized structure, such as a micro-or nano-sized dense structure dispersed in a medium. In one embodiment, the particle is a nucleic acid-containing particle, such as a particle comprising DNA, RNA, or a mixture thereof.

Electrostatic interactions between positively charged molecules (such as polymers and lipids) and negatively charged nucleic acids participate in particle formation. This results in the complexing and spontaneous formation of nucleic acid particles. In one embodiment, the nucleic acid particle is a nanoparticle.

As used in this disclosure, "nanoparticles" refers to particles having a mean diameter suitable for parenteral administration.

A "nucleic acid particle" can be used to deliver a nucleic acid to a target site of interest (e.g., a cell, tissue, organ, etc.). The nucleic acid particles may be formed from at least one cationic or cationic ionizable lipid or lipid-like material, at least one cationic polymer (such as protamine) or mixtures thereof, and nucleic acids. Nucleic acid particles include Lipid Nanoparticle (LNP) and lipoplex (lpx) based formulations.

Without being bound by any theory, it is believed that the cationic or cationic ionizable lipid or lipid-like material and/or the cationic polymer are bound together with the nucleic acid to form aggregates, and that such aggregation results in colloidally stable particles.

In one embodiment, the particles described herein further comprise at least one lipid or lipid-like material other than a cationic or cationic ionizable lipid or lipid-like material, at least one polymer other than a cationic polymer, or a mixture thereof.

In some embodiments, the nucleic acid particles comprise more than one type of nucleic acid molecule, wherein the molecular parameters of the nucleic acid molecules may be similar or different from each other, for example with respect to molar mass or basic structural elements such as molecular structure, capping, coding regions or other features.

The average diameter of the nucleic acid particles described herein may be, in one embodiment, from about 30nm to about 1000nm, from about 50nm to about 800nm, from about 70nm to about 600nm, from about 90nm to about 400nm, from about 100nm to about 300 nm.

The nucleic acid particles described herein can exhibit a polydispersity index of about 0.5 or less, about 0.4 or less, about 0.3 or less, or about 0.2 or less. For example, the nucleic acid particles can exhibit a polydispersity index in the range of about 0.1 to about 0.3 or about 0.2 to about 0.3.

With respect to RNA lipid particles, the N/P ratio gives the ratio of the number of nitrogen groups in the lipid to the number of phosphate groups in the RNA. It is related to the charge ratio, since the nitrogen atom (depending on the pH) is usually positively charged, while the phosphate group is negatively charged. When there is charge balance, the N/P ratio depends on the pH. Lipid formulations are typically formed with N/P ratios of 4 or more, up to 12, as positively charged nanoparticles are believed to facilitate transfection. In that case, the RNA is considered to be fully bound to the nanoparticle.

The nucleic acid particles described herein can be prepared using a wide range of methods, which can include obtaining a colloid from at least one cationic or cationic ionizable lipid or lipid-like material and/or at least one cationic polymer, and mixing the colloid with the nucleic acid to obtain the nucleic acid particles.

The term "colloid" as used herein relates to the type of homogeneous mixture in which the dispersed particles do not precipitate. The insoluble particles in the mixture are microscopic and have a particle size of between 1 and 1000 nanometers. This mixture may be referred to as a colloid or colloidal suspension. Sometimes the term "colloid" refers only to the particles in the mixture and not to the entire suspension.

For the preparation of colloids comprising at least one cationic or cationic ionizable lipid or lipid-like material and/or at least one cationic polymer, methods conventionally used for the preparation of liposomal vesicles and suitably adapted may be applicable herein. The most common methods for preparing liposome vesicles share the following basic steps: (i) dissolving lipids in an organic solvent, (ii) drying the resulting solution, and (iii) hydrating the dried lipids (using various aqueous media).

In the membrane hydration process, the lipids are first dissolved in a suitable organic solvent and then dried to produce a thin film at the bottom of the flask. The obtained lipid membrane is hydrated using a suitable aqueous medium to produce a liposome dispersion. In addition, additional reduction steps may be included.

Reverse phase evaporation is an alternative method for membrane hydration for the preparation of liposome vesicles, which involves the formation of a water-in-oil emulsion between an aqueous phase and an organic phase comprising lipids. For systematic homogenization, a brief sonication of this mixture is required. Removal of the organic phase under reduced pressure produced a milky gel which subsequently became a liposome suspension.

The term "ethanol injection technique" refers to a process in which an ethanol solution containing lipids is rapidly injected into an aqueous solution through a needle. This action disperses lipids throughout the solution and promotes lipid structure formation, e.g., lipid vesicle formation such as liposome formation. In general, the RNA lipoplex particles described herein can be obtained by adding RNA to a colloidal liposome dispersion. In one embodiment, such colloidal liposome dispersions are formed using ethanol injection techniques as follows: an ethanol solution comprising a lipid (e.g., a cationic lipid) and an additional lipid is injected into the aqueous solution with agitation. In one embodiment, the RNA lipoplex particles described herein are obtainable without an extrusion step.

The term "extrusion" refers to the production of particles having a fixed cross-sectional profile. In particular, it refers to the reduction of particles, thereby forcing the particles through a filter having defined pores.

Other methods characterized as being free of organic solvents may also be used to prepare colloids according to the present disclosure.

LNPs typically comprise 4 components: ionizable cationic lipids, neutral lipids such as phospholipids, steroids such as cholesterol, and polymer-conjugated lipids such as polyethylene glycol (PEG) -lipids. Each component is responsible for payload protection and enables efficient intracellular delivery. LNPs can be prepared by rapid mixing of ethanol-soluble lipids with nucleic acids in aqueous buffers.

The term "mean diameter" refers to the mean hydrodynamic diameter of a particle measured by dynamic laser light scattering (DLS) and data analysis using the so-called cumulant algorithm, which results in providing a so-called Z having a length dimensionaverageAnd dimensionless Polydispersity Index (PI) (Koppel, D., J.chem.Phys.57,1972, pp 4814-. Here the "average diameter", "diameter" or "size" of the particles and ZaverageThis value of (2) is used synonymously.

The "polydispersity index" is preferably calculated on the basis of dynamic light scattering measurements by so-called cumulant analysis, mentioned in the definition of "mean diameter". Under certain preconditions, it can be taken as a measure of the overall size distribution of the nanoparticles.

Different types of nucleic acid-containing particles have been previously described as suitable for delivering nucleic acids in particle form (e.g., Kaczmarek, j.c. et al, 2017, Genome Medicine 9, 60). For non-viral nucleic acid delivery vehicles, nanoparticle encapsulation of nucleic acids physically protects the nucleic acids from degradation and, depending on the particular chemistry, can aid in cellular uptake and endosomal escape.

The present disclosure describes particles comprising a nucleic acid, at least one cationic or cationic ionizable lipid or lipid-like material, and/or at least one cationic polymer associated with the nucleic acid to form nucleic acid particles, and compositions comprising such particles. The nucleic acid particles may comprise nucleic acids complexed with the particles in different forms by non-covalent interactions. The particles described herein are not viral particles, in particular infectious viral particles, i.e. they are not capable of virally infecting cells.

Suitable cationic or cationic ionizable lipid or lipid-like materials and cationic polymers are those that form nucleic acid particles, and are encompassed by the terms "particle-forming component" or "particle-forming agent". The term "particle-forming component" or "particle-forming agent" relates to any component that associates with a nucleic acid to form a nucleic acid particle. Such components include any component that may be part of a nucleic acid particle.

Cationic polymers

Polymers are common materials for nanoparticle-based delivery in view of their high degree of chemical flexibility. Typically, cationic polymers are used to electrostatically coagulate negatively charged nucleic acids into nanoparticles. These positively charged groups are usually composed of amines that change their protonation state in the pH range of 5.5-7.5, which is believed to cause ionic imbalance leading to endosomal disruption. Polymers such as poly-L-lysine, polyamidoamine (polyamidoamine), protamine and polyethyleneimine, as well as naturally occurring polymers such as chitosan, have all been used for nucleic acid delivery and are suitable as cationic polymers herein. In addition, some researchers have synthesized polymers specifically for nucleic acid delivery. In particular, poly (β -amino esters) have gained widespread use in nucleic acid delivery due to their ease of synthesis and biodegradability. Such synthetic polymers are also suitable as cationic polymers herein.

As used herein, "polymer" has its usual meaning, i.e., a molecular structure comprising one or more repeating units (monomers) linked by covalent bonds. The repeat units may all be the same, or in some cases more than one type of repeat unit may be present in the polymer. In some cases, the polymer is biologically derived, i.e., a biopolymer such as a protein. In certain instances, additional moieties, such as targeting moieties, e.g., as described herein, can also be present in the polymer.

If more than one type of repeating unit is present in the polymer, the polymer is referred to as a "copolymer". It is to be understood that the polymers employed herein may be copolymers. The repeat units forming the copolymer may be arranged in any manner. For example, the repeat units can be arranged in a random order, an alternating order, or as a "block" copolymer, i.e., comprising one or more regions, each region 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), and so forth. The block copolymer may have two (diblock copolymer), three (triblock copolymer) or more different blocks.

In certain embodiments, the polymer is biocompatible. Biocompatible polymers are polymers that do not normally cause significant cell death at moderate concentrations. In certain embodiments, the biocompatible polymer is biodegradable, i.e., the polymer is capable of chemical and/or biological degradation within a physiological environment, such as in vivo.

In certain embodiments, the polymer may be protamine or a polyalkyleneimine, particularly protamine.

The term "protamine" refers to any strongly basic protein of relatively low molecular weight that is rich in arginine and found in sperm cells of various animals (e.g., fish) in particular in association with DNA in place of somatic histones. In particular, the term "protamine" refers to a protein found in protamine that is strongly basic, soluble in water, does not coagulate upon heating, and produces primarily arginine upon hydrolysis. In purified form, they are useful in long acting formulations of insulin and for neutralizing the anticoagulant effect of heparin.

According to the present disclosure, the term "protamine" as used herein is meant to encompass any protamine amino acid sequence obtained or derived from a natural or biological source, including fragments thereof as well as multimeric forms of said amino acid sequence or fragments thereof as well as (synthetic) polypeptides which are artificial, specifically designed for a specific purpose, and which cannot be isolated from natural or biological sources.

In one embodiment, the polyalkyleneimine comprises a polyethyleneimine and/or a polypropyleneimine, preferably a polyethyleneimine. The preferred polyalkyleneimine is Polyethyleneimine (PEI). The average molecular weight of PEI is preferably 0.75. multidot.102-107Da, preferably 1000-10 5Da, more preferably 10000-40000Da, more preferably 15000-30000Da, and even more preferably 20000-25000 Da.

Linear polyalkyleneimines such as linear Polyethyleneimine (PEI) are preferred according to the present disclosure.

Cationic polymers (including polycationic polymers) contemplated for use herein include any cationic polymer capable of electrostatically binding nucleic acids. In one embodiment, cationic polymers contemplated for use herein include any cationic polymer to which a nucleic acid may be associated, for example by forming a complex with the nucleic acid or forming a vesicle in which the nucleic acid is enclosed or encapsulated.

The particles described herein may also comprise polymers other than cationic polymers, i.e., non-cationic polymers and/or anionic polymers. Anionic and neutral polymers are collectively referred to herein as non-cationic polymers.

Lipids and lipid-like materials

The terms "lipid" and "lipid-like material" are broadly defined herein as molecules comprising one or more hydrophobic moieties or groups and optionally also comprising one or more hydrophilic moieties or groups. Molecules comprising a hydrophobic portion and a hydrophilic portion are also commonly referred to as amphiphilic molecules. Lipids are generally poorly soluble in water. In an aqueous environment, the amphiphilic nature allows the molecules to self-assemble into organized structures and distinct phases. One of those phases consists of lipid bilayers, as they are present in vesicles, multilamellar/unilamellar liposomes or membranes in an aqueous environment. Hydrophobicity may be imparted by the inclusion of non-polar groups including, but not limited to, long chain saturated and unsaturated aliphatic hydrocarbon groups and such groups substituted with one or more aromatic, alicyclic or heterocyclic groups. Hydrophilic groups can comprise polar and/or charged groups and include carbohydrates, phosphates, carboxyl groups, sulfates, amino groups, sulfhydryls, nitro groups, hydroxyl groups, and other similar groups.

As used herein, the term "amphiphilic" refers to a molecule having a polar portion and a non-polar portion. Typically, amphiphilic compounds have a polar head attached to a long hydrophobic tail. In some embodiments, the polar portion is soluble in water, while the non-polar portion is insoluble in water. Further, the polar moiety may have a formal positive charge or a formal negative charge. Alternatively, the polar moiety may have formal positive and negative charges and may be a zwitterion or an inner salt. For the purposes of this disclosure, an amphiphilic compound can be, but is not limited to, one or more of natural or unnatural lipids and lipid-like compounds.

The term "lipid-like material", "lipid-like compound" or "lipid-like molecule" relates to a substance that is structurally and/or functionally related to lipids but may not be considered lipids in the strict sense. For example, the term includes compounds that are capable of forming an amphiphilic layer when present in a vesicle, multilamellar/unilamellar liposome or membrane in an aqueous environment, and includes surfactants or synthetic compounds having hydrophilic and hydrophobic portions. In general, the term refers to molecules comprising hydrophilic and hydrophobic portions having different structural organizations, which may or may not be similar to the lipid structural organizations. As used herein, the term "lipid" is to be construed to encompass lipids and lipid-like materials, unless otherwise indicated herein or clearly contradicted by context.

Specific examples of amphiphilic compounds that may be included in the amphiphilic layer include, but are not limited to, phospholipids, amino lipids, and sphingolipids.

In certain embodiments, the amphiphilic compound is a lipid. The term "lipid" refers to a group of organic compounds characterized by being insoluble in water, but soluble in many organic solvents. In general, lipids can be divided into 8 classes: fatty acids, glycerolipids, glycerophospholipids, sphingolipids, glycolipids, polyketides (derived from the condensation of ketoacyl subunits), sterol lipids, and terpene alcohol (prenol) lipids (derived from the condensation of isoprene subunits). Although the term "lipid" is sometimes used as a synonym for fat, fat is a subgroup of lipids called triglycerides. Lipids also encompass molecules such as fatty acids and their derivatives (including triglycerides, diglycerides, monoglycerides, and phospholipids), as well as metabolites comprising sterols such as cholesterol.

Fatty acids or fatty acid residues are a diverse group of molecules formed from hydrocarbon chains terminated with carboxylic acid groups; this arrangement imparts a polar, hydrophilic end to the molecule, as well as a non-polar, hydrophobic end that is insoluble in water. Carbon chains, typically 4-24 carbons in length, may be saturated or unsaturated, and may be linked to functional groups comprising oxygen, halogen, nitrogen and sulfur. If the fatty acid contains a double bond, it may have cis or trans geometric isomerism, which significantly affects the configuration of the molecule. The cis double bond bends the fatty acid chain, which action mixes with more double bonds in the chain. The other major lipid classes in the fatty acid class are fatty esters and fatty amides.

Glycerolipids include mono-, di-and tri-substituted glycerols, most notably fatty acid triesters of glycerol, known as triglycerides. The word "triacylglycerol" is sometimes used synonymously with "triglyceride". In these compounds, the three hydroxyl groups of glycerol are usually esterified with each of the different fatty acids. Other subclasses of glycerolipids are represented by the glycoglycerols, which are characterized by the presence of one or more sugar residues linked to the glycerol by a glycosidic bond.

Glycerophospholipids are amphiphilic molecules (comprising a hydrophobic region and a hydrophilic region) comprising a glycerol core linked by ester linkages to two fatty acid-derived "tails" and by phosphate linkages to one "head" group. Examples of glycerophospholipids commonly referred to as phospholipids (although sphingomyelin is also classified as a phospholipid) are phosphatidylcholine (also known as PC, GPCho or lecithin), phosphatidylethanolamine (PE or GPEtn) and phosphatidylserine (PS or GPSer).

Sphingolipids are a complex family of compounds that share a common structural feature, the sphingoid base (sphingoid base) backbone. The major sphingosine base in mammals is commonly referred to as sphingosine. Ceramides (N-acyl-sphingosine bases) are a major subset of sphingosine base derivatives, which have amide-linked fatty acids. Fatty acids are generally saturated or mono-unsaturated, having a chain length of 16 to 26 carbon atoms. The major phospholipid sphingoester of mammals is sphingomyelin (ceramide phosphorylcholine), while insects contain predominantly ceramide phosphorylethanolamine, and fungi have a plant ceramide phosphorylinositol and a head group containing mannose. Glycosphingolipids are a diverse family of molecules that include one or more sugar residues linked to a sphingoid base by a glycosidic linkage. Examples of these are simple and complex glycosphingolipids such as cerebrosides and gangliosides.

Sterol lipids, such as cholesterol and its derivatives, or tocopherol and its derivatives, are important components of membrane lipids, along with glycerophospholipids and sphingophospholipids.

Glycolipids describe compounds in which fatty acids are directly attached to a sugar backbone, forming a structure compatible with membrane bilayers. In glycolipids, monosaccharides replace the glycerol backbone present in glycerolipids and glycerophospholipids. The most familiar glycolipids are acylated glucosamine precursors of the lipid a component of lipopolysaccharides in gram-negative bacteria. A typical lipid a molecule is the disaccharide of glucosamine, which is derived from up to seven fatty-acyl chains. The smallest lipopolysaccharide required for growth in E.coli is Kdo 2-lipid A, a hexaacylated disaccharide of glucosamine glycosylated with two 3-deoxy-D-manno-octulosonic acid (3-deoxy-D-mano-octulosonic acid, Kdo) residues.

Polyketides are synthesized by the classical enzyme polymerization of acetyl and propionyl subunits, as well as the iterative and multimodular enzymes that share mechanistic features with fatty acid synthases. They contain a large number of secondary metabolites and natural products from animal, plant, bacterial, fungal and marine sources and have a great structural diversity. Many polyketides are cyclic molecules whose backbone is often further modified by glycosylation, methylation, hydroxylation, oxidation, or other processes.

In accordance with the present disclosure, the lipids and lipid-like materials may be cationic, anionic, or neutral. The neutral lipid or lipid-like material exists in an uncharged or neutral zwitterionic form at the selected pH.

Cationic or cationic ionizable lipid or lipid-like material

The nucleic acid particles described herein may comprise at least one cationic or cationic ionizable lipid or lipid-like material as a particle-forming agent. Cationic or cationic ionizable lipid or lipid-like material contemplated for use herein includes any cationic or cationic ionizable lipid or lipid-like material capable of electrostatically binding nucleic acids. In one embodiment, the cationic or cationic ionizable lipid or lipid-like material contemplated for use herein can be associated with a nucleic acid, for example, by forming a complex with the nucleic acid or forming a vesicle in which the nucleic acid is enclosed or encapsulated.

As used herein, "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 acids through electrostatic interactions. Generally, cationic lipids have 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 a positive charge.

In certain embodiments, the cationic lipid or lipid-like material has a net positive charge only at certain pH, in particular at acidic pH, whereas at a different, preferably higher pH, such as physiological pH, it preferably has no net positive charge, preferably no charge, i.e. it is neutral. This ionizable behavior is believed to enhance efficacy by helping endosomal escape and reducing toxicity, as compared to particles that remain cationic at physiological pH.

For the purposes of this disclosure, unless otherwise contradicted by context, the term "cationic lipid or lipid-like material" encompasses such "cationic ionizable" lipids or lipid-like materials.

In one embodiment, the cationic or cationic ionizable lipid or lipid-like material comprises a head group comprising at least one nitrogen atom (N) which is positively charged or capable of protonation.

Examples of cationic lipids include, but are not limited to, 1, 2-dioleoyl-3-trimethylammonium-propane (1, 2-dioleoyl-3-trimethyllammonium propane, DOTAP); n, N-dimethyl-2, 3-dioleyloxypropylamine (N, N-dimethyl-2, 3-dioleyloxypropylamine, DODMA), 1, 2-di-O-octadecenyl-3-trimethylammoniumpropane (1, 2-di-O-octadecenyl-3-trimethylammoniumpropane, DOTMA), 3- (N ', N' -dimethylaminoethane) -carbamoyl) cholesterol (3- (N ', N' -dimethylaminoethane) -carbamoyl) choleestenol, DC-Chol), dimethyldioctadecylammonium (DDAB); 1,2-dioleoyl-3-dimethylammonium-propane (1, 2-dioleoyl-3-dimethyllammonium-propane, DODAP); 1, 2-diacyloxy-3-dimethylammoniumpropane (1, 2-diacetoxy-3-dimethylammoniumpropane); 1, 2-dialkoxy-3-dimethylammoniumpropane (1, 2-dialkyloxy-3-dimethylammoniumpropane); dioctadecyl dimethyl ammonium chloride (dioc) tadecyl ammonium chloride, DODAC), 1,2-distearyloxy-N, N-dimethyl-3-aminopropane (1,2-distearyloxy-N, N-dimethyl-3-aminopropane, DSDMA), 2, 3-ditetradecyloxy propyl- (2-hydroxyethyl) -dimethylammonium (2,3-di (tetrahydroxy) propyl- (2-hydroxyethoxy) -dimethylammonium, DMRIE), 1, 2-dimyristoyl-sn-glycerol-3-ethylphosphonic acid choline (1, 2-dimyristoyl-sn-3-ethylphosphonic acid, DMEPC), l, 2-dimyristoyl-3-trimethylammonium propane (2-dimyristoyl-3-trimethylammoniumnioxide, DMEPC), 1, 2-dimyristoyl-3-dimethylpropylammonium bromide (1, 2-dimyristoyl-3-trimethylammoniumnioxide, dmec-dimethyloxypropyl, 1, 2-dimethyloxypropyl, 1-dimethylolpropyl-1-hydroxyethylammonium bromide, 2-dioleyloxypropylphenyl-3-dimethylnaphthylammonium bromide, DORIE), and 2,3-dioleoyloxy-N- [2(spermine carboxamide) ethyl]-N, N-dimethyl-l-propylaminium trifluoroacetate (2, 3-dioleoxy-N- [2 (speramine carboxamide) ethyl)]-N, N-dimethyl-l-propanamide trifluoracetate, DOSPA), 1, 2-dioleyloxy-N, N-dimethylaminopropane (1, 2-dilinoleyloxy-N, N-dilinominopropane, DLInDMA), 1, 2-dioleyloxy-N, N-dimethylaminopropane (1, 2-dilinoleyloxy-N, N-dimethyllaminopropane, DLenDMA), dioctadecylamidoglycyl spermine (DOGS), 3-dimethylamino-2- (cholest-5-en-3- β -oxybutane-4-oxy) -1- (cis, cis-9, 12-octadecenyloxy) propane (3-dime-2- (chost-5-en-3- β -oxybutane-4-oxy) -1- (cis, cis-9, 12-octadecenyloxy) -propane (1- (1, 4-oxo-2- (chost-5-diol-3-beta-oxo-4-diol) -1- (1, cis-9, 12-oc-tadecadienyloxy) propane, CLinDMA), 2- [5 ' - (cholest-5-en-3- β -oxy) -3 ' -oxapentoxy) -3-dimethyl-1- (cis, cis-9 ', 12 ' -octadecadienyloxy) propane (2- [5 ' - (cholest-5-en-3-beta-oxy) -3 ' -oxapentoxy) -3-dimethyl-1- (cis, cis-9 ', 12 ' -octadienyloxy) propane, CpDMA, N-dimethyl-3, 4-dioleyloxybenzylamine (N, N-dimethyl-3, 4-dioleyloxybenzamine, DMOBA), 1,2-N, N ' -dioleylcarbamoyl-3-dimethylaminopropane (1,2-N, N ' -Dilinolylcarbamoyl-3-dimethyllaminopropane, DOcarbDAP), 2,3-dioleoyloxy-N, N-dimethylpropylamine (2,3-Dilinoleoyloxy-N, N-dimethylpropylamine, DLINDAP), 1,2-N, N ' -dioleylcarbamoyl-3-dimethylaminopropane (1,2-N, N ' -Dilinolylcarbamoyl-3-dimethyllamino) propane, DLincarbDAP), 1, 2-dioleoylcarbamoyl-3-dimethylaminopropane (1, 2-dilinoleylcarbamoyl-3-dimethyllaminopropane, DLincdAP), 2-dioleyl-4-dimethylaminomethyl- [1, 3-dioleyl ] -4-dimethylaminomethyl- [1]2, 2-dilinoleyl-4-dimethylmenthyl- [1,3 ]]Dioxolane, DLin-K-DMA), 2-dioleyl-4-dimethylaminoethyl- [1,3]-dioxolane (2, 2-dilinoleyl-4-dimethylmenthyl- [1, 3)]Dioxolane, DLin-K-XTC2-DMA), 2-dioleyl-4- (2-dimethylaminoethyl) - [1, 3%]-dioxolane (2,2-dilinoleyl-4- (2-dimethylmenthyl) - [1,3]-dioxolane, DLin-KC 2-DMA), tridecyl-6, 9,28,31-tetraen-19-yl-4- (dimethylamino) butyrate (heptatriconta-6, 9,28, 31-tetra-en-19-yl-4- (dimethylamino) butanoate, DLin-MC3-DMA), N- (2-Hydroxyethyl) -N, N-dimethyl-2, 3-bis (tetradecyloxy) -1-propylaminium bromide (N- (2-hydroxyethy) -N, N-dimethyl-2, 3-bis (tetradecyloxy) -1-propylaminium bromide, DMRIE), (±) -N- (3-aminopropyl) -N, N-dimethyl-2, 3-bis (cis-9-tetradecyloxy) -1-propylaminium bromide ((±) (cis-9-tetradecyloxy) -1-propylaminium bromide) + -) -N- (3-aminopropoxy) -N, N-dimethyl-2, 3-bis (cis-9-tetradecyloxy) -1-propanaminium bromide, GAP-DMORIE), (+ -) -N- (3-aminopropyl) -N, N-dimethyl-2, 3-bis (dodecyloxy) -1-propanaminium bromide (+ -) -N- (3-aminopropoxy) -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 (+ -) -N- (3-aminopropoxy) -N, n-dimethyl-2, 3-bis (tetracyloxy) -1-propanaminium bromide, GAP-DMRIE, N- (2-Aminoethyl) -N, N-dimethyl-2, 3-bis (tetradecyloxy) -1-propanaminium bromide (N- (2-aminoethylexy) -N, N-dimethyl-2, 3-bis (tetradecyloxy) -1-propanaminium bromide, beta AE-DMRIE), N- (4-carboxybenzyl) -N, N-dimethyl-2, 3-bis (oleoyloxy) propan-1-aminium (N- (4-carboxybenzyl) -N, N-dimethyl-2, 3-bis (oleoyloxy) propan-1-aminium, DOQ), 2- ({8 BAb) -cholest-5-cholest-3-yloxy-yl ]Octyl } oxy) -N, N-dimethyl-3- [ (9Z,12Z) -octadeca-9, 12-dien-1-yloxy]Propane-1-amine (2- ({8- [ (3. beta.) -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-dintMyristoyl-3-dimethylammonium-propane (1, 2-dimyristoyl-3-dimthyllammonium-propane, DMDAP), 1, 2-dipalmitoyl-3-dimthylammonium-propane (1, 2-dipalmitoyl-3-dimthyllammonium-propane, DPTAP), N1- [2- ((1S) -1- [ (3-aminopropyl) amino group]-4- [ bis (3-amino-propyl) amino]Butyl carboxamide) ethyl]-3, 4-bis [ oleyloxy [ ] [ -O ]]Benzamide (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 (1, 2-dioleoyl-sn-glycero-3-ethylphosphocholine, DOEPC), 2,3-bis (dodecyloxy) -N- (2-hydroxyethyl) -N, N-dimethylpropane-1-ammonium bromide (2,3-bis (dodecyloxy) -N- (2-hydroxyethoxy) -N, N-dimethylpropan-1-amonium bromide, DLRIE), N- (2-aminoethyl) -N, N-dimethyl-2,3-bis (tetradecyloxy) propane-1-aminium bromide (N- (2-aminoethylyl) -N, N-dimethyll-2, 3-bis (tetracyloxy) propan-1-aminium bromide, DMORIE), bis ((Z) -non-2-en-1-yl) 8,8'- (((2(dimethylamino) ethyl) thio) carbonyl) azanediyl) dicaprylate (di ((Z) -non-2-en-1-yl) 8,8' - ((((2(dimethylamino) ethyl) thio) carbonyl) azanediyl) dicatanoate, ATX), N-dimethyl-2,3-bis (dodecyloxy) propan-1-amine (N, N-dimethyl-2,3-bis (dodecyloxy) propan-1-amine, DLDMA), N-dimethyl-2,3-bis (tetradecyloxy) propan-1-amine (N, N-dimethyl-2,3-bis (tetracyloxy) propan-1-amine, DMDMA), bis ((Z) -non-2-en-1-yl) -9- ((4- (dimethylaminobutyryl) oxy) heptadecanedioate (Di ((Z) -non-2-en-1-yl) -9- ((4- (dimethylaminobutyanoyl) oxy) heptadecenoate, l319), N-dodecyl-3- ((2-dodecylcarbamoyl-ethyl) - {2- [ (2-dodecylcarbamoyl-ethyl) -2- { (2-dodecylcarbamoyl-ethyl) - [2- (2-dodecylcarbamoyl-ethylamino) -ethyl. ]-amino } -ethylamino) propionamide (N-Dodecyclyl-3- ((2-docyclablecobamoyl-ethyl) - {2- [ (2-docyclablecobamoyl-ethyl) -2- { (2-docyclablecobamoyl-ethyl) - [2- (2-docyclablecobamoyl-ethyl) -ethyl } -ethyl]-amino}-ethylamino)propionamide,lipidoid 98N12-5), 1- [2- [ bis (2-hydroxydodecyl) amino]Ethyl- [2- [4- [2- [ bis (2 hydroxydodecyl) amino group]Ethyl radical]Piperazin-1-yl]Ethyl radical]Amino group]Dodecane-2-alcohol (1- [2- [ bis (2-hydroxydecacyl) amino)]ethyl-[2-[4-[2-[bis(2 hydroxydodecyl)amino]ethyl]piperazin-1-yl]ethyl]amino]dodecan-2-ol,lipidoid C12-200)。

In some embodiments, the cationic lipid may comprise from about 10 mole (mol)% -about 100 mol%, from about 20 mol% -about 100 mol%, from about 30 mol% -about 100 mol%, from about 40 mol% -about 100 mol%, or from about 50 mol% -about 100 mol% of the total lipid present in the particle.

Additional lipids or lipid-like materials

The particles described herein may also comprise a lipid or lipid-like material other than a cationic or cationic ionizable lipid or lipid-like material, i.e., a non-cationic lipid or lipid-like material (including a non-cationic ionizable lipid or lipid-like material). Anionic and neutral lipid or lipid-like materials are collectively referred to herein as non-cationic lipid or lipid-like materials. The particle stability and efficacy of nucleic acid delivery can be enhanced by optimizing the formulation of the nucleic acid particles by adding other hydrophobic moieties such as cholesterol and lipids in addition to ionizable/cationic lipids or lipid-like materials.

Additional lipids or lipid-like materials may be incorporated, which may or may not affect the overall charge of the nucleic acid particles. In certain embodiments, the additional lipid or lipid-like material is a non-cationic lipid or lipid-like material. The non-cationic lipid may comprise, for example, one or more anionic lipids and/or neutral lipids. As used herein, "anionic lipid" refers to any lipid that is negatively charged at a selected pH. As used herein, "neutral lipid" refers to any of a variety of lipid species that exist in an uncharged or neutral zwitterionic form at a selected pH. In a preferred embodiment, the additional lipids comprise one of the following neutral lipid components: (1) a phospholipid, (2) cholesterol or a derivative thereof; or (3) a mixture of phospholipids and cholesterol or derivatives thereof. Examples of cholesterol derivatives include, but are not limited to, cholestanol, cholestanone, cholestenone, coprosterol, cholesteryl-2 '-hydroxyethyl ether, cholesteryl-4' -hydroxybutyl ether, tocopherol and its derivatives, and mixtures thereof.

Specific phospholipids that may be used include, but are not limited to, phosphatidylcholine (phosphatidylethanolamine), phosphatidylethanolamine (phosphatidylethanolamine), phosphatidylglycerol (phosphatidylglycerol), phosphatidic acid (phosphatidylacid), phosphatidylserine (phosphatidylserine), or sphingomyelin (sphingomyelin). Such phospholipids include in particular diacyl-phosphatidyl choline (DSPC), Dioleoylphosphatidylcholine (DOPC), Dimyristoylphosphatidylcholine (DMPC), Dipentanoylphosphatidylcholine (DPPC), dilauroylphosphatidylcholine (dilauroylphosphatidylcholine), Dipalmitoylphosphatidylcholine (DPPC), diarachitoylphosphatidylcholine (DADYPC), docosanoylphosphatidylcholine (DBDYL-PC), Dioctadecylphosphatidylcholine (DPPC), dioctadecylphosphatidylcholine (DADYL-3-D-glycerophosphatidylcholine, didecylphosphatidylcholine (DBG-3-D-glycerophosphatidylcholine, DPC), 18:0Diether PC), 1-oleoyl-2-cholesteryl hemisuccinyl-sn-glycerol-3-phosphocholine (1-oleoyl-2-cholesteryl-sn-glycero-3-phosphocholine, OChemisPC), 1-hexadecyl-sn-glycerol-3-phosphocholine (1-hexadecyl-sn-glycero-3-phosphocholine, C16 Lyso PC) and phosphatidylethanolamine (phosphatidylethanolamine), in particular diacylphosphatidylethanolamine (diacylphosphatidylethanolamine), such as dioleoylphosphatidylethanolamine, DOPE), distearoylphosphatidylethanolamine (distearoylphosphatidylethanolamine, DSpalmitoylethanolamine), phosphatidylethanolamine (phosphatidylethanolamine, DPPE), DMPE), dilauroyl-phosphatidylethanolamine (DLPE), diphytanoyl-phosphatidylethanolamine (DPyPE), and other phosphatidylethanolamine lipids with different hydrophobic chains.

In certain preferred embodiments, the additional lipid is DSPC or DSPC and cholesterol.

In certain embodiments, the nucleic acid particle comprises a cationic lipid and an additional lipid.

In one embodiment, the particles described herein comprise a polymer-conjugated lipid, such as a pegylated lipid. The term "pegylated lipid" refers to a molecule comprising a lipid moiety and a polyethylene glycol moiety. Pegylated lipids are known in the art.

Without wishing to be bound by theory, the amount of the at least one cationic lipid may affect important nucleic acid particle characteristics, such as charge, particle size, stability, tissue selectivity, and biological activity of the nucleic acid, as compared to the amount of the at least one additional lipid. Thus, in some embodiments, the molar ratio of the at least one cationic lipid to the at least one additional lipid is from about 10:0 to about 1:9, about 4: 1 to about 1:2 or about 3:1 to about 1: 1.

In some embodiments, the non-cationic lipid, particularly the neutral lipid (e.g., one or more phospholipids and/or cholesterol), may comprise from about 0 mol% to about 90 mol%, from about 0 mol% to about 80 mol%, from about 0 mol% to about 70 mol%, from about 0 mol% to about 60 mol%, or from about 0 mol% to about 50 mol% of the total lipid present in the particle.

Lipopolex particles

In certain embodiments of the present disclosure, the RNA described herein may be present in an RNA lipoplex particle.

In the context of the present disclosure, the term "RNA lipoplex particle" relates to a particle comprising lipids, in particular cationic lipids and RNA. Electrostatic interactions between positively charged liposomes and negatively charged RNA lead to complexation (complexation) and spontaneous formation of RNA lipoplex particles. Positively charged liposomes can generally be synthesized using cationic lipids such as DOTMA and additional lipids such as DOPE. In one embodiment, the RNA lipoplex particle is a nanoparticle.

In certain embodiments, the RNA lipoplex particle comprises a cationic lipid and an additional lipid. In an exemplary embodiment, the cationic lipid is DOTMA and the additional lipid is DOPE.

In some embodiments, the molar ratio of the at least one cationic lipid to the at least one additional lipid is from about 10:0 to about 1: 9. about 4: 1 to about 1: 2 or about 3:1 to about 1: 1. In particular embodiments, the molar ratio may be about 3:1, about 2.75:1, about 2.5:1, about 2.25:1, about 2:1, about 1.75:1, about 1.5:1, about 1.25: 1 or about 1: 1. In an exemplary embodiment, the molar ratio of the at least one cationic lipid to the at least one additional lipid is about 2: 1.

The average diameter of the RNA lipoplex particles described herein may in one embodiment be from about 200nm to about 1000nm, from about 200nm to about 800nm, from about 250 to about 700nm, from about 400 to about 600nm, from about 300nm to about 500nm, or from about 350nm to about 400 nm. In particular embodiments, the average diameter of the RNA lipoplex particles is about 200nm, about 225nm, about 250nm, about 275nm, about 300nm, about 325nm, about 350nm, about 375nm, about 400nm, about 425nm, about 450nm, about 475nm, about 500nm, about 525nm, about 550nm, about 575nm, about 600nm, about 625nm, about 650nm, about 700nm, about 725nm, about 750nm, about 775nm, about 800nm, about 825nm, about 850nm, about 875nm, about 900nm, about 925nm, about 950nm, about 975nm, or about 1000 nm. In one embodiment, the average diameter of the RNA lipoplex particles is from about 250nm to about 700 nm. In another embodiment, the average diameter of the RNA lipoplex particles is from about 300nm to about 500 nm. In an exemplary embodiment, the average diameter of the RNA lipoplex particles is about 400 nm.

The RNA lipoplex particles and compositions comprising RNA lipoplex particles described herein are useful for delivering RNA to a target tissue following parenteral administration, in particular intravenous administration. RNA lipoplex particles can be prepared using liposomes, which can be obtained by injecting a solution of lipids in ethanol into water or a suitable aqueous phase. In one embodiment, the aqueous phase has an acidic pH. In one embodiment, the aqueous phase comprises acetic acid, e.g., in an amount of about 5 mM. Liposomes can be used to prepare RNA lipoplex particles by mixing the liposomes with RNA. In one embodiment, the liposome and RNA lipoplex particles comprise at least one cationic lipid and at least one additional lipid. In one embodiment, the at least one cationic lipid comprises 1, 2-di-O-octaalkenyl-3-trimethylammonium propane (1, 2-di-O-octadecenyl-3-trimethylammomum propane, DOTMA) and/or 1, 2-dioleoyl-3-trimethylammomum-propane (1, 2-dioleoyl-3-trimethylammomum-propane, DOTAP). In one embodiment, the at least one additional lipid comprises 1,2-di- (9Z-octadecenoyl) -sn-glycerol-3-phosphoethanolamine (1,2-di- (9Z-octadecenoyl) -sn-glycerol-3-phosphoethanolamine, DOPE), cholesterol (Chol) and/or 1, 2-dioleoyl-sn-glycerol-3-phosphocholine (1, 2-dioleoyl-sn-glycerol-3-phosphocholine, DOPC). In one embodiment, the at least one cationic lipid comprises 1,2-di-O-octadecenyl-3-trimethylammonium propane (1, 2-di-O-octadecenyl-3-trimethyllammonium propane, DOTMA) and the at least one additional lipid comprises 1,2-di- (9Z-octadecenoyl) -sn-glycerol-3-phosphoethanolamine (1,2-di- (9Z-octadecenyl) -sn-glycerol-3-phosphoethanolamine, DOPE). In one embodiment, the liposome and RNA lipoplex particles comprise 1,2-di-O-octadecenyl-3-trimethylammonium propane (1, 2-di-O-octadecenyl-3-trimethyllammonium propane, DOTMA) and 1,2-di- (9Z-octadecenoyl) -sn-glycerol-3-phosphoethanolamine (1,2-di- (9Z-octadecenyl) -sn-glycerol-3-phosphoethanolamine, DOPE).

Spleen-targeting RNA lipoplex particles are described in WO 2013/143683, which is incorporated herein by reference. It has been found that RNA lipoplex particles having a net negative charge can be used to preferentially target spleen tissue or spleen cells such as antigen presenting cells, in particular dendritic cells. Thus, following administration of RNA lipoplex particles, RNA accumulation and/or RNA expression occurs in the spleen. Thus, the RNA lipoplex particles of the present disclosure can be used to express RNA in the spleen. In one embodiment, no or substantially no RNA accumulation and/or RNA expression occurs in the lung and/or liver following administration of the RNA lipoplex particles. In one embodiment, RNA accumulation and/or RNA expression occurs in antigen presenting cells, such as professional antigen presenting cells in the spleen, following administration of the RNA lipoplex particles. Thus, the RNA lipoplex particles of the present disclosure may be used to express RNA in such antigen presenting cells. In one embodiment, the antigen presenting cell is a dendritic cell and/or a macrophage.

Lipid Nanoparticles (LNP)

In one embodiment, a nucleic acid, such as RNA, described herein is administered in the form of a Lipid Nanoparticle (LNP). The LNP may comprise any lipid capable of forming a particle, to which one or more nucleic acid molecules are attached, or in which one or more nucleic acid molecules are encapsulated.

In one embodiment, the LNP comprises one or more cationic lipids, and one or more stabilizing lipids. Stabilizing lipids include neutral lipids and pegylated lipids.

In one embodiment, the LNP comprises a cationic lipid, a neutral lipid, a steroid, a polymer conjugated lipid; and RNA encapsulated within or associated with the lipid nanoparticle.

In one embodiment, the LNP comprises 40-55 mole percent, 40-50 mole percent, 41-49 mole percent, 41-48 mole percent, 42-48 mole percent, 43-48 mole percent, 44-48 mole percent, 45-48 mole percent, 46-48 mole percent, 47-48 mole percent, or 47.2-47.8 mole percent of a cationic lipid. In one embodiment, the LNP comprises about 47.0, 47.1, 47.2, 47.3, 47.4, 47.5, 47.6, 47.7, 47.8, 47.9, or 48.0 mole percent of a cationic lipid.

In one embodiment, the neutral lipids are present at a concentration of 5-15 mole percent, 7-13 mole percent, or 9-11 mole percent. In one embodiment, the neutral lipids are present at a concentration of about 9.5, 10, or 10.5 mole percent.

In one embodiment, the steroid is present at a concentration of 30 to 50 mole percent, 35 to 45 mole percent, or 38 to 43 mole percent. In one embodiment, the steroid is present at a concentration of about 40, 41, 42, 43, 44, 45 or 46 mole percent.

In one embodiment, the LNP comprises 1-10 mole percent, 1-5 mole percent, or 1-2.5 mole percent of the polymer-conjugated lipid.

In one embodiment, the LNP comprises 40-50 mole percent of cationic lipid; 5-15 mole percent of neutral lipids; 35-45 mole percent of a steroid; 1-10 mole percent of a polymer-conjugated lipid; and RNA encapsulated within or associated with the lipid nanoparticle.

In one embodiment, the mole percentage is determined based on the total moles of lipid present in the lipid nanoparticle.

In one embodiment, the neutral lipid is selected from the group consisting of DSPC, DPPC, DMPC, DOPC, POPC, DOPE, DOPG, DPPG, POPE, DPPE, DMPE, DSPE and SM. In one embodiment, the neutral lipid is selected from the group consisting of DSPC, DPPC, DMPC, DOPC, POPC, DOPE and SM. In one embodiment, the neutral lipid is DSPC.

In one embodiment, the steroid is cholesterol.

In one embodiment, the polymer-conjugated lipid is a pegylated lipid. In one embodiment, the pegylated lipids have the following structure:

or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein:

R12And R13Each independently is 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 linkages; and w has an average value of 30 to 60. In one embodiment, R12And R13Each independently a straight, saturated alkyl chain containing from 12 to 16 carbon atoms. In one embodiment, w has an average value of 40 to 55. In one embodiment, the average w is about 45. At one endIn embodiments, R12And R13Each independently a straight, saturated alkyl chain containing about 14 carbon atoms, and w has an average value of about 45.

In one embodiment, the pegylated lipid is DMG-PEG 2000, for example, having the structure:

in some embodiments, the cationic lipid component of LNP has the structure of formula (III):

or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein:

L1or L2One of them 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 L1Or L2The other 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;

G1and G2Each independently is unsubstituted C 1-C12Alkylene or C1-C12An alkenylene group;

G3is C1-C24Alkylene radical, C1-C24Alkenylene radical, C3-C8Cycloalkylene radical, C3-C8Cycloalkenylene;

Rais H or C1-C12An alkyl group;

R1and R2Each independently is C6-C24Alkyl or C6-C24An alkenyl group;

R3is H, OR5、CN、-C(=O)OR4、-OC(=O)R4or-NR5C(=O)R4

R4Is C1-C12An alkyl group;

R5is H or C1-C6An alkyl group; and is

x is 0, 1 or 2.

In some of the foregoing embodiments of formula (III), the lipid has one of the following structures (IIIA) or (IIIB):

wherein:

a is a 3-8 membered cycloalkyl or cycloalkylene ring;

at each occurrence, R6Independently H, OH or C1-C24An alkyl group;

n is an integer of 1 to 15.

In some of the foregoing embodiments of formula (III), the lipid has structure (IIIA), and in other embodiments, the lipid has structure (IIIB).

In other embodiments of formula (III), the lipid has one of the following structures (IIIC) or (IIID):

wherein y and z are each independently an integer from 1 to 12.

In any of the preceding embodiments of formula (III), L1Or L2One is-O (C ═ O) -. For example, in some embodiments, L1And L2Each is-O (C ═ O) -. In some various embodiments of any of the foregoing, L1And L2Are each independently-(C ═ O) O — or-O (C ═ O) -. For example, in some embodiments, L1And L2Each is- (C ═ O) O-.

In some different embodiments of formula (III), the lipid has one of the following structures (IIIE) or (IIIF):

in some of the foregoing embodiments of formula (III), the lipid has one of the following structures (IIIG), (IIIH), (IIII), or (IIIJ):

in some of the foregoing embodiments of formula (III), n is an integer from 2 to 12, such as from 2 to 8 or from 2 to 4. For example, in some embodiments, n is 3, 4, 5, or 6. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments, n is 5. In some embodiments, n is 6.

In some other previous embodiments of formula (III), y and z are each independently an integer from 2 to 10. For example, in some embodiments, y and z are each independently an integer from 4 to 9 or from 4 to 6.

In some of the foregoing embodiments of formula (III), R6Is H. In other of the foregoing embodiments, R6Is C1-C24An alkyl group. In other embodiments, R6Is OH.

In some embodiments of formula (III), G3Is unsubstituted. In other embodiments, G3 is substituted. In various embodiments, G3Is linear C1-C24Alkylene or linear C1-C24An alkenylene group.

In-situ type(III) in some other of the preceding embodiments, R 1Or R2Or both are C6-C24An alkenyl group. For example, in some embodiments, R1And R2Each independently having the structure:

wherein:

at each occurrence, R7aAnd R7bIndependently is H or C1-C12An alkyl group; and is

a is an integer of 2 to 12,

wherein each is selected from R7a、R7bAnd a, whereby R1And R2Each independently containing from 6 to 20 carbon atoms. For example, in some embodiments, a is an integer from 5 to 9 or from 8 to 12.

In some of the foregoing embodiments of formula (III), R7Is H. For example, in some embodiments, R is present at each occurrence7aIs H. In various other embodiments of the foregoing, R7bIs C at least one occurrence of1-C8An alkyl group. For example, in some embodiments, C1-C8Alkyl is methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-hexyl or n-octyl.

In various embodiments of formula (III), R1Or R2Or both having one of the following structures:

in some of the foregoing embodiments of formula (III), R3Is OH, CN, -C (═ O) OR4、-OC(=O)R4or-NHC (═ O) R4. In some embodiments, R4Is methyl or ethyl.

In various embodiments, the cationic lipid of formula (III) has one of the structures shown in the table below.

Representative compounds of formula (III).

In some embodiments, the LNP comprises lipids of formula (III), RNA, neutral lipids, steroids, and pegylated lipids. In some embodiments, the lipid of formula (III) is compound III-3. In some embodiments, the neutral lipid is DSPC. In some embodiments, the steroid is cholesterol. In some embodiments, the pegylated lipid is ALC-0159.

In some embodiments, the cationic lipid is present in the LNP in an amount of about 40 to about 50 mole percent. In one embodiment, the neutral lipids are present in the LNP in an amount from about 5 to about 15 mole percent. In one embodiment, the steroid is present in the LNP in an amount of from about 35 to about 45 mole percent. In one embodiment, the pegylated lipid is present in the LNP in an amount of from about 1 to about 10 mole percent.

In some embodiments, the LNP comprises compound III-3 in an amount from about 40 to about 50 mole percent, DSPC in an amount from about 5 to about 15 mole percent, cholesterol in an amount from about 35 to about 45 mole percent, and ALC-0159 in an amount from about 1 to about 10 mole percent.

In some embodiments, the LNP comprises compound III-3 in an amount of about 47.5 mole percent, DSPC in an amount of about 10 mole percent, cholesterol in an amount of about 40.7 mole percent, and ALC-0159 in an amount of about 1.8 mole percent.

In various embodiments, the cationic lipid has one of the structures shown in the following table.

In some embodiments, the LNP comprises a cationic lipid (e.g., a cationic lipid of formula (B) or formula (D), particularly a cationic lipid of formula (D)), RNA, a neutral lipid, a steroid, and a pegylated lipid as shown in the above table. In some embodiments, the neutral lipid is DSPC. In some embodiments, the steroid is cholesterol. In some embodiments, the pegylated lipid is DMG-PEG 2000.

In one embodiment, the LNP comprises a cationic lipid which is an ionizable lipid-like material (lipidoid). In one embodiment, the cationic lipid has the following structure:

the N/P value is preferably at least about 4. In some embodiments, the N/P value ranges from 4 to 20, 4 to 12, 4 to 10, 4 to 8, or 5 to 7. In one embodiment, the N/P value is about 6.

The average diameter of the LNPs described herein can be from about 30nm to about 200nm or from about 60 nm to about 120nm in one embodiment.

RNA targeting

Some aspects of the disclosure include targeted delivery of the RNAs disclosed herein (e.g., RNAs encoding vaccine antigens and/or immunostimulants).

In one embodiment, the present disclosure includes targeting the lung. If the RNA administered is RNA encoding a vaccine antigen, it is particularly preferred to target the lung. For example, RNA can be delivered to the lung by inhalation, by administering RNA that can be formulated as particles as described herein, such as lipid particles.

In one embodiment, the present disclosure includes targeting the lymphatic system, particularly the secondary lymphatic organs, more particularly the spleen. If the RNA administered is that encoding a vaccine antigen, it is particularly preferred to target the lymphatic system, in particular the secondary lymphoid organs, more particularly the spleen.

In one embodiment, the target cell is a spleen cell. In one embodiment, the target cell is an antigen presenting cell, such as a professional antigen presenting cell in the spleen. In one embodiment, the target cell is a dendritic cell in the spleen.

The "lymphatic system" is a part of the circulatory system and is an important part of the immune system, comprising a network of lymphatic vessels carrying lymph. The lymphatic system is composed of lymphatic organs, the conducting network of lymphatic vessels, and circulating lymph. The major or central lymphoid organs produce lymphocytes from immature progenitor cells. The thymus and bone marrow constitute the major lymphoid organs. Secondary or peripheral lymphoid organs, including the lymph nodes and spleen, maintain mature naive lymphocytes and initiate an adaptive immune response.

RNA can be delivered to the spleen by so-called lipoplex formulations, where the RNA is bound to liposomes comprising cationic lipids and optionally additional or helper lipids to form injectable nanoparticle formulations. Liposomes can be obtained by injecting a solution of the lipid in ethanol into water or a suitable aqueous phase. RNA lipoplex particles can be prepared by mixing liposomes with RNA. Spleen-targeting RNA lipoplex particles are described in WO 2013/143683, which is incorporated herein by reference. It has been found that RNA lipoplex particles having a net negative charge can be used to preferentially target spleen tissue or spleen cells such as antigen presenting cells, in particular dendritic cells. Thus, following administration of RNA lipoplex particles, RNA accumulation and/or RNA expression occurs in the spleen. Thus, the RNA lipoplex particles of the present disclosure can be used to express RNA in the spleen. In one embodiment, no or substantially no RNA accumulation and/or RNA expression occurs in the lung and/or liver following administration of the RNA lipoplex particles. In one embodiment, RNA accumulation and/or RNA expression occurs in antigen presenting cells, such as professional antigen presenting cells in the spleen, following administration of the RNA lipoplex particles. Thus, the RNA lipoplex particles of the present disclosure may be used to express RNA in such antigen presenting cells. In one embodiment, the antigen presenting cell is a dendritic cell and/or a macrophage.

The charge of the RNA lipoplex particles of the present disclosure is the sum of the charge present in the at least one cationic lipid and the charge present in the RNA. The charge ratio is the ratio of the positive charge present in the at least one cationic lipid to the negative charge present in the RNA. The charge ratio of the positive charges present in the at least one cationic lipid to the negative charges present in the RNA is calculated by the following equation: charge ratio [ (cationic lipid concentration (moles)) × (total number of positive charges in cationic lipid) ]/[ (RNA concentration (moles)) × (total number of negative charges in RNA) ].

The spleen-targeting RNA lipoplex particles described herein preferably have a net negative charge at physiological pH, e.g., a charge ratio of positive to negative charge of about 1.9: 2 to about 1: 2, or about 1.6: 2 to about 1.1: 2. In particular embodiments, the charge ratio of positive to negative charges in the RNA lipoplex particle is about 1.9: 2.0, about 1.8: 2.0, about 1.7: 2.0, about 1.6: 2.0, about 1.5: 2.0, about 1.4: 2.0, about 1.3: 2.0, about 1.2: 2.0, about 1.1: 2.0, or about 1: 2.0 at physiological pH.

An immunostimulatory agent may be provided to a subject by administering an RNA encoding the immunostimulatory agent to the subject in a formulation that preferentially delivers the RNA to the liver or liver tissue. Preferably, the RNA is delivered to such a target organ or tissue, in particular in large amounts if expression of a large amount of the immunostimulant is desired and/or if systemic presence of the immunostimulant is desired or required.

RNA delivery systems have an inherent preference for the liver. This relates to lipid-based particles, cationic and neutral nanoparticles, in particular to lipophilic ligands in lipid nanoparticles such as liposomes, nanomicelles and bioconjugates. Liver accumulation is caused by the discrete nature of the hepatic vascular system or lipid metabolism (liposomes and lipid or cholesterol conjugates).

To deliver RNA to the liver in vivo, drug delivery systems may be used to transport RNA to the liver by preventing its degradation. For example, polyplex nanomicelles composed of a poly (ethylene glycol) (PEG) coated surface and a core comprising mRNA are useful systems because nanomicelles provide excellent RNA stability in vivo under physiological conditions. In addition, the stealth properties provided by the polyplex nanomicelle surface comprising a dense PEG cage effectively circumvent host immune defenses.

Examples of suitable immunostimulants for targeting the liver are cytokines involved in T cell proliferation and/or maintenance. Examples of suitable cytokines include IL2 or IL7, fragments and variants thereof, and fusion proteins of these cytokines, fragments and variants, such as extended PK cytokines.

In another embodiment, the RNA encoding the immunostimulatory agent may be administered in a formulation that preferentially delivers the RNA to the lymphatic system (particularly the secondary lymphoid organs, more particularly the spleen). It is preferred to deliver an immunostimulant to such a target tissue, particularly if it is desired to have an immunostimulant present in such an organ or tissue (e.g. in order to induce an immune response, particularly during T-cell priming or in order to activate resident immune cells in case an immunostimulant such as a cytokine is required), but it is not desired that the immunostimulant be present systemically, particularly in large amounts (e.g. because the immunostimulant is systemically toxic).

Examples of suitable immunostimulants are cytokines involved in T cell priming. Examples of suitable cytokines include IL12, IL15, IFN- α or IFN- β, fragments and variants thereof, and fusion proteins of these cytokines, fragments and variants, such as extended PK cytokines.

Immunostimulant

In one embodiment, the RNA encoding the vaccine antigen may be non-immunogenic. In this and other embodiments, the RNA encoding the vaccine antigen may be co-administered with the immunostimulant or the RNA encoding the immunostimulant. The methods and agents described herein are particularly effective if the immunostimulant is linked to a pharmacokinetic modifying group (hereinafter referred to as a "prolonged Pharmacokinetic (PK)" immunostimulant). The methods and agents described herein are particularly effective if the immunostimulant is administered in the form of an RNA encoding the immunostimulant. In one embodiment, the RNA is targeted to the liver for systemic availability. Hepatocytes can be transfected efficiently and are capable of producing large amounts of protein.

An "immunostimulant" is any substance that stimulates the immune system by inducing the activation or increasing the activity of any component of the immune system, in particular immune effector cells. The immunostimulant may be pro-inflammatory.

According to one aspect, the immunostimulant is a cytokine or a variant thereof. Examples of cytokines include interferons such as interferon-alpha (IFN-alpha) or interferon-gamma (IFN-gamma), interleukins such as IL2, IL7, IL12, IL15 and IL23, colony stimulating factors such as M-CSF and GM-CSF, and tumor necrosis factor. According to another aspect, the immunostimulant comprises an adjuvant-type immunostimulant such as an APC Toll-like receptor agonist or a costimulatory/cell adhesion membrane protein. Examples of Toll-like receptor agonists include costimulatory/adhesion proteins such as CD80, CD86, and ICAM-1.

Cytokines are a small class of proteins (5-20 kDa) important in cell signaling. Their release has an effect on the behavior of the cells around them. Cytokines are involved in autocrine signaling, paracrine signaling, and endocrine signaling as immunomodulators. Cytokines include chemokines, interferons, interleukins, lymphokines, and tumor necrosis factor, but generally do not include hormones or growth factors (although some overlap in terms). Cytokines are produced by a wide range of cells, including immune cells such as macrophages, B lymphocytes, T lymphocytes and mast cells, as well as endothelial cells, fibroblasts and various stromal cells. A given cytokine may be produced by more than one type of cell. Cytokines act through receptors and are particularly important in the immune system; cytokines regulate the balance between humoral and cell-based immune responses, and they regulate the maturation, growth, and reactivity of specific cell populations. Some cytokines enhance or inhibit the action of other cytokines in a complex manner.

According to the present disclosure, the cytokine may be a naturally occurring cytokine or a functional fragment or variant thereof. The cytokine may be a human cytokine and may be derived from any vertebrate, in particular any mammal. One particularly preferred cytokine is interferon- α.

Interferon

Interferons (IFNs) are a group of signaling proteins that are produced and released by host cells in response to the presence of several pathogens, such as viruses, bacteria, parasites, and also tumor cells. Typically, virus-infected cells release interferon, causing nearby cells to enhance their antiviral defenses.

Interferons are generally divided into three classes based on the type of receptor through which they conduct signals: type I interferons, type II interferons and type III interferons.

All type I interferons bind to a specific cell surface receptor complex called IFN- α/β receptor (IFNAR), which consists of IFNAR1 and IFNAR2 chains.

The type I interferons present in humans are IFN α, IFN β, IFN ∈, IFN κ, and IFN ω. Generally, type I interferons are produced when the body recognizes an invaded virus. They are produced by fibroblasts and monocytes. Once released, type I interferons bind to specific receptors on target cells, which results in the expression of proteins, thereby preventing the virus from producing and replicating its RNA and DNA.

IFN α protein is produced mainly by plasmacytoid dendritic cells (pdcs). They are primarily involved in innate immunity against viral infection. There are 13 subtypes of genes responsible for their synthesis, called IFNA1, IFNA2, IFNA4, IFNA5, IFNA6, IFNA7, IFNA8, IFNA10, IFNA13, IFNA14, IFNA16, IFNA17, IFNA21, respectively. These genes are found together in clusters on chromosome 9.

IFN beta protein by fibroblasts in large production. They have antiviral activity and are primarily involved in the innate immune response. Two types of IFN β, IFN β 1 and IFN β 3, have been described. Both natural and recombinant forms of IFN beta 1 have antiviral, antibacterial and anticancer properties.

Type II interferons (IFN γ in humans) are also known as immuno-interferons and are activated by IL 12. In addition, type II interferons are released by cytotoxic T cells and T helper cells.

Type III interferons signal through a receptor complex consisting of IL10R2 (also known as CRF2-4) and IFNLR1 (also known as CRF 2-12). Although more recently discovered than type I and type II IFNs, recent information confirms the importance of type III IFNs in certain types of viral or fungal infections.

In general, type I and type II interferons are responsible for modulating and activating immune responses.

According to the present disclosure, the type I interferon is preferably IFN α or IFN β, more preferably IFN α.

In accordance with the present disclosure, the interferon may be a naturally occurring interferon or a functional fragment or variant thereof. The interferon may be human interferon and may be derived from any vertebrate, in particular any mammal.

Interleukin

Interleukins (ILs) are a group of cytokines (secreted proteins or signaling molecules) that can be divided into four large groups based on distinguishing structural features. However, their amino acid sequence similarity is very weak (usually 15-25% identity). The human genome encodes more than 50 interleukins and related proteins.

In accordance with the present disclosure, the interleukin may be a naturally occurring interleukin or a functional fragment or variant thereof. The interleukin may be a human interleukin and may be derived from any vertebrate, in particular any mammal.

Extended PK groups

The immunostimulatory polypeptide described herein can be prepared as a fusion or chimeric polypeptide that includes an immunostimulatory portion and a heterologous polypeptide (i.e., a polypeptide that is not an immunostimulatory agent). An immunostimulant may be fused to an extended PK group, which increases the circulating half-life. Non-limiting examples of extended PK groups are described below. It is understood that other PK groups that increase the circulating half-life of an immune stimulant such as a cytokine or variant thereof are also suitable for use in the present disclosure. In certain embodiments, the extended PK group is a serum albumin domain (e.g., mouse serum albumin, human serum albumin).

As used herein, the term "PK" is an acronym for "pharmacokinetics" and encompasses the characteristics of a compound, including, for example, absorption, distribution, metabolism, and elimination in a subject. As used herein, an "extended PK group" refers to a protein, peptide or moiety that when fused or administered with a biologically active molecule increases the circulating half-life of the biologically active molecule. Examples of extended PK groups include serum albumin (e.g., HSA), immunoglobulin Fc or Fc fragments and variants thereof, transferrin and variants thereof, and Human Serum Albumin (HSA) binders (as disclosed in U.S. publication nos. 2005/0287153 and 2007/0003549). Kontermann, Expert Opin Biol Ther,2016 Jul; other exemplary extended PK groups are disclosed in 16(7) 903-15, which are incorporated herein by reference in their entirety. As used herein, an "extended PK" immunostimulant refers to the portion of the immunostimulant that is combined with an extended PK group. In one embodiment, the extended PK immunostimulant is a fusion protein, wherein the immunostimulant moiety is linked or fused to the extended PK group.

In certain embodiments, the serum half-life of the extended PK immunostimulant is increased relative to the immunostimulant alone (i.e., the immunostimulant not fused to the extended PK group). In certain embodiments, the serum half-life of the extended PK immune stimulator is at least 20, 40, 60, 80, 100, 120, 150, 180, 200, 400, 600, 800, or 1000% longer relative to the serum half-life of the immune stimulator alone. In certain embodiments, the serum half-life of the extended PK immunostimulant is at least 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4-fold, 4.5-fold, 5-fold, 6-fold, 7-fold, 8-fold, 10-fold, 12-fold, 13-fold, 15-fold, 17-fold, 20-fold, 22-fold, 25-fold, 27-fold, 30-fold, 35-fold, 40-fold, or 50-fold that of the immunostimulant alone. In certain embodiments, the extended serum half-life of the PK immunostimulant is at least 10 hours, 15 hours, 20 hours, 25 hours, 30 hours, 35 hours, 40 hours, 50 hours, 60 hours, 70 hours, 80 hours, 90 hours, 100 hours, 110 hours, 120 hours, 130 hours, 135 hours, 140 hours, 150 hours, 160 hours, or 200 hours.

As used herein, "half-life" refers to the time it takes for the serum or plasma concentration of a compound, such as a peptide or protein, to decrease by 50% in vivo, for example, due to degradation and/or clearance or sequestration by natural mechanisms. The extended PK immunostimulants suitable for use herein are stable in vivo and their half-life is increased by, for example, fusion to serum albumin (e.g., HSA or MSA) that is resistant to degradation and/or clearance or sequestration. The half-life can be determined by any means known per se, such as by pharmacokinetic analysis. Suitable techniques are well known to those skilled in the art and may for example generally comprise the steps of: suitably administering to the subject a suitable dose of the amino acid sequence or compound; periodically collecting a blood sample or other sample from the subject; determining the level or concentration of an amino acid sequence or compound in the blood sample; and calculating from (the graph of) the data thus obtained the time until the level or concentration of the amino acid sequence or compound is reduced by 50% compared to the initial level at the time of dose administration. Further details are provided, for example, in standard manuals, such as Kenneth, A.et al, Chemical Stability of Pharmaceuticals: A Handbook for Pharmaceuticals and in Peters et al, pharmaceutical Analysis: A Practical Approach (1996). Reference is also made to Gibaldi, m.et al, Pharmacokinetics,2nd rev.edition, Marcel Dekker (1982).

In certain embodiments, the extended PK group comprises serum albumin or a fragment thereof, or a variant of serum albumin or a fragment thereof (all of which are included in the term "albumin" for purposes of this disclosure). The polypeptides described herein may be fused to albumin (or fragments or variants thereof) to form an albumin fusion protein. Such albumin fusion proteins are described in U.S. publication No. 20070048282.

As used herein, "albumin fusion protein" refers to a protein formed by fusing at least one molecule of albumin (or a fragment or variant thereof) to at least one molecule of a protein (e.g., a therapeutic protein, particularly an immunostimulant). Albumin fusion proteins can be produced by translating a nucleic acid in which a polynucleotide encoding a therapeutic protein is linked in-frame to a polynucleotide encoding albumin. Once part of the albumin fusion protein, the therapeutic protein and albumin can each be referred to as a "portion", "region", or "moiety" (e.g., "therapeutic protein portion" or "albumin portion") of the albumin fusion protein. In a highly preferred embodiment, the albumin fusion protein comprises at least one molecule of a therapeutic protein (including but not limited to a mature form of the therapeutic protein) and at least one molecule of albumin (including but not limited to a mature form of albumin). In one embodiment, the albumin fusion protein is processed by a host cell, such as a cell of a target organ, for administration of RNA, e.g., a hepatocyte, and secreted into the circulation. Processing of the nascent albumin fusion protein occurring in the secretory pathway of a host cell for RNA expression may include, but is not limited to, signal peptide cleavage; formation of disulfide bonds; properly folding; carbohydrate addition and processing (e.g., N-and O-linked glycosylation); specific proteolytic cleavage; and/or assembly into multimeric proteins. The albumin fusion protein is preferably encoded by the RNA in unprocessed form, it has a signal peptide, in particular at the N-terminus, and is preferably present in processed form after secretion by the cell, in particular the signal peptide having been cleaved off. In a most preferred embodiment, "processed form of an albumin fusion protein" refers to an albumin fusion protein product that has undergone N-terminal signal peptide cleavage, also referred to herein as a "mature albumin fusion protein"

In preferred embodiments, an albumin fusion protein comprising a therapeutic protein has a higher plasma stability than the plasma stability of the same therapeutic protein when not fused to albumin. Plasma stability generally refers to the period of time from administration of a therapeutic protein into the blood stream in vivo to degradation of the therapeutic protein and clearance from the blood stream into the organ (e.g., kidney or liver) that ultimately clears the therapeutic protein from the body. Plasma stability was calculated from the half-life of the therapeutic protein in the bloodstream. The half-life of a therapeutic protein in the bloodstream can be readily determined by common assays known in the art.

As used herein, "albumin" refers collectively to albumin or an amino acid sequence, or a fragment or variant of albumin, that has one or more functional activities (e.g., biological activities) of albumin. In particular, "albumin" refers to human albumin or fragments or variants thereof, in particular the mature form of human albumin, or albumin or fragments thereof from other vertebrates, or variants of these molecules. The albumin may be derived from any vertebrate, in particular any mammal, such as a human, bovine, ovine or porcine. Non-mammalian albumins include, but are not limited to, chicken and salmon albumins. The albumin portion of the albumin fusion protein can be from a different animal than the therapeutic protein.

In certain embodiments, the albumin is Human Serum Albumin (HSA), or a fragment or variant thereof, such as those disclosed in US 5,876,969, WO 2011/124718, WO 2013/075066, and WO 2011/0514789.

The terms Human Serum Albumin (HSA) and Human Albumin (HA) are used interchangeably herein. The terms "albumin" and "serum albumin" are broader and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof).

As used herein, an albumin fragment sufficient to prolong the therapeutic activity or plasma stability of a therapeutic protein refers to an albumin fragment of sufficient length and structure to stabilize or prolong the therapeutic activity or plasma stability of the protein, such that the plasma stability of the therapeutic protein portion of the albumin fusion protein is prolonged or extended as compared to the plasma stability in a non-fused state.

The albumin portion of the albumin fusion protein may comprise the full length of the albumin sequence, or may include one or more fragments thereof capable of stabilizing or prolonging the therapeutic activity or plasma stability. Such fragments may be 10 or more amino acids in length, or may comprise about 15, 20, 25, 30, 50 or more contiguous amino acids from the albumin sequence, or may comprise a portion or all of a particular domain of albumin. For example, one or more fragments of HSA spanning the first two immunoglobulin-like domains may be used. In a preferred embodiment, the HSA fragment is the mature form of HSA.

Generally, the albumin fragment or variant is at least 100 amino acids in length, preferably at least 150 amino acids in length.

In accordance with the present disclosure, the albumin may be a naturally occurring albumin or a fragment or variant thereof. The albumin may be human albumin and may be derived from any vertebrate, in particular any mammal.

Preferably, the albumin fusion protein comprises albumin as the N-terminal portion and a therapeutic protein as the C-terminal portion. Alternatively, albumin fusion proteins comprising albumin as the C-terminal portion and a therapeutic protein as the N-terminal portion may also be used. In other embodiments, the albumin fusion protein has a therapeutic protein fused to the N-terminus and C-terminus of albumin. In a preferred embodiment, the therapeutic proteins fused at the N-and C-termini are the same therapeutic protein. In another preferred embodiment, the therapeutic proteins fused at the N-and C-termini are different therapeutic proteins. In one embodiment, the different therapeutic proteins are all cytokines.

In one embodiment, the therapeutic protein is linked to the albumin through a peptide linker. Linker peptides between fusion moieties may provide greater physical separation between moieties, thus maximizing accessibility of the therapeutic protein, e.g., for binding to its cognate receptor. The linker peptide may be composed of amino acids so that it is flexible or more rigid. The linker sequence may be protease or chemically cleavable.

The term "Fc region" as used herein refers to the portion of a native immunoglobulin formed by the Fc domains (or Fc portions) of each of its two heavy chains. As used herein, the term "Fc domain" refers to a portion or fragment of a single immunoglobulin (Ig) heavy chain, wherein the Fc domain does not contain an Fv domain. In certain embodiments, the Fc domain begins at the hinge region upstream of the papain cleavage site and terminates at the C-terminus of the antibody. Thus, a complete Fc domain comprises at least a hinge region, a CH2 domain, and a CH3 domain. In certain embodiments, the Fc domain comprises at least one of: a hinge (e.g., upper, middle, and/or lower hinge region) domain, a CH2 domain, a CH3 domain, a CH4 domain, or a variant, portion, or fragment thereof. In certain embodiments, the Fc domain comprises a complete Fc domain (i.e., the hinge domain, CH2 domain, and CH3 domain). In certain embodiments, the Fc domain comprises a hinge domain (or portion thereof) fused to a CH3 domain (or portion thereof). In certain embodiments, the Fc domain comprises a CH2 domain (or portion thereof) fused to a CH3 domain (or portion thereof). In certain embodiments, the Fc domain consists of a CH3 domain or portion thereof. In certain embodiments, the Fc domain consists of a hinge domain (or portion thereof) and a CH3 domain (or portion thereof). In certain embodiments, the Fc domain consists of a CH2 domain (or portion thereof) and a CH3 domain. In certain embodiments, the Fc domain consists of a hinge domain (or portion thereof) and a CH2 domain (or portion thereof). In certain embodiments, the Fc domain lacks at least a portion of a CH2 domain (e.g., all or part of a CH2 domain). An Fc domain herein generally refers to a polypeptide comprising all or part of the Fc domain of an immunoglobulin heavy chain. This includes, but is not limited to, polypeptides comprising the entire CH1, hinge, CH2, and/or CH3 domains, as well as fragments of such peptides comprising only the hinge, CH2, and CH3 domains, for example. The Fc domain may be derived from any species and/or any subtype of immunoglobulin, including but not limited to human IgG1, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM antibodies. Fc domains encompass native Fc and Fc variant molecules. As shown herein, one of ordinary skill in the art will appreciate that any Fc domain can be modified such that its amino acid sequence differs from the native Fc domain of a naturally occurring immunoglobulin molecule. In certain embodiments, the Fc domain has reduced effector function (e.g., fcyr binding).

The Fc domains of the polypeptides described herein may be derived from different immunoglobulin molecules. For example, the Fc domain of a polypeptide may comprise a CH2 and/or CH3 domain derived from an IgG1 molecule and a hinge region derived from an IgG3 molecule. In another example, the Fc domain may comprise a chimeric hinge region derived in part from an IgG1 molecule and in part from an IgG3 molecule. In another example, the Fc domain may comprise a chimeric hinge derived in part from an IgG1 molecule and in part from an IgG4 molecule.

In certain embodiments, the extended PK group comprises an Fc domain or fragment thereof, or a variant of an Fc domain or fragment thereof (all of which are included in the term "Fc domain" for purposes of this disclosure). The Fc domain does not contain a variable region that binds to an antigen. Suitable Fc domains for use in the present disclosure may be obtained from many different sources. In certain embodiments, the Fc domain is derived from a human immunoglobulin. In certain embodiments, the Fc domain is from a human IgG1 constant region. However, it is understood that the Fc domain may be derived from an immunoglobulin of another mammalian species, including, for example, a rodent (e.g., mouse, rat, rabbit, guinea pig) or non-human primate (e.g., chimpanzee, macaque) species.

Furthermore, the Fc domain (or fragment or variant thereof) may be derived from any immunoglobulin class, including IgM, IgG, IgD, IgA, and IgE, and any immunoglobulin isotype, including IgG1, IgG2, IgG3, and IgG 4.

Various Fc domain gene sequences (e.g., mouse and human constant region gene sequences) are available in the form of publicly available deposits. Constant region domains comprising Fc domain sequences may be selected that lack specific effector functions and/or have specific modifications to reduce immunogenicity. A number of antibodies and sequences of antibody-encoding genes have been disclosed, and suitable Fc domain sequences (e.g., hinge, CH2, and/or CH3 sequences, or fragments or variants thereof) can be derived from these sequences using techniques well known in the art.

In certain embodiments, the extended PK group is a serum albumin binding protein such as those described in US2005/0287153, US2007/0003549, US2007/0178082, US2007/0269422, US2010/0113339, WO2009/083804, and WO2009/133208, which are incorporated herein by reference in their entirety. In certain embodiments, the extended PK group is transferrin, as disclosed in US 7,176,278 and US 8,158,579, which are incorporated herein by reference in their entirety. In certain embodiments, the extended PK group is a serum immunoglobulin binding protein such as those disclosed in US2007/0178082, US2014/0220017 and US2017/0145062, which are incorporated herein by reference in their entirety. In certain embodiments, the extended PK group is a fibronectin (Fn) -based scaffold domain protein that binds to serum albumin, such as those disclosed in US2012/0094909, which is incorporated herein by reference in its entirety. Methods of making fibronectin based scaffold domain proteins are also disclosed in US 2012/0094909. A non-limiting example of an extended PK group based on Fn3 is Fn3(HSA), i.e., Fn3 protein that binds to human serum albumin.

In certain aspects, the extended PK immunostimulants suitable for use according to the present disclosure may employ one or more peptide linkers. As used herein, the term "peptide linker" refers to a peptide or polypeptide sequence that connects two or more domains (e.g., an extended PK moiety and an immunostimulant moiety) in a linear amino acid sequence of a polypeptide chain. For example, a peptide linker may be used to link the immunostimulant to the HSA domain.

Linkers suitable for fusing extended PK groups to, for example, an immunostimulant are well known in the art. Exemplary linkers include glycine-serine polypeptide linkers, glycine-proline polypeptide linkers, and proline-alanine polypeptide linkers. In certain embodiments, the linker is a glycine-serine polypeptide linker, i.e., a peptide consisting of glycine and serine residues.

The immunostimulatory polypeptide described herein may comprise a sequence encoding a "marker" or "reporter molecule" in addition to or instead of the heterologous polypeptide described above. Examples of markers or reporter genes include beta-lactamase, Chloramphenicol Acetyltransferase (CAT), Adenosine Deaminase (ADA), aminoglycoside phosphotransferase, dihydrofolate reductase (DHFR), hygromycin-B-phosphotransferase (HPH), Thymidine Kinase (TK), beta-galactosidase, and Xanthine Guanine Phosphoribosyltransferase (XGPRT).

Pharmaceutical composition

The agents (agents) described herein may be administered in a pharmaceutical composition or medicament, and may be administered in the form of any suitable pharmaceutical composition.

In one embodiment, the pharmaceutical composition described herein is an immunogenic composition for inducing an immune response against a coronavirus in a subject. For example, in one embodiment, the immunogenic composition is a vaccine.

In one embodiment of all aspects of the invention, the components described herein, such as RNA encoding a vaccine antigen, may be administered in a pharmaceutical composition, which may comprise a pharmaceutically acceptable carrier, and may optionally comprise one or more adjuvants, stabilizers, and the like. In one embodiment, the pharmaceutical composition is for use in a therapeutic or prophylactic treatment, e.g., for treating or preventing a coronavirus infection.

The term "pharmaceutical composition" relates to a formulation comprising a therapeutically effective substance, preferably together with a pharmaceutically acceptable carrier, diluent and/or excipient. The pharmaceutical composition can be used to treat, prevent, or reduce the severity of a disease or disorder by administering the pharmaceutical composition to a subject. Pharmaceutical compositions are also known in the art as pharmaceutical formulations.

The pharmaceutical compositions of the present disclosure may comprise, or may be administered with, one or more adjuvants. The term "adjuvant" relates to compounds that prolong, enhance or accelerate the immune response. Adjuvants include a group of xenogenic compounds such as oil emulsions (e.g., freund's adjuvant), minerals (e.g., alum), bacterial products (e.g., pertussis toxin), or immune stimulating complexes. Examples of adjuvants include, but are not limited to, LPS, GP96, CpG oligodeoxynucleotides, growth factors and cytokines such as monokines, lymphokines, interleukins, chemokines. The cytokine may be IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL12, IFN alpha, IFN gamma, GM-CSF, LT-a. Other known adjuvants are aluminum hydroxide, freund's adjuvant or oils (e.g.,ISA 51). Other suitable adjuvants for use in the present disclosure include lipopeptides, such as Pam3 Cys.

Pharmaceutical compositions according to the present disclosure are generally employed in "pharmaceutically effective amounts" and "pharmaceutically acceptable formulations".

The term "pharmaceutically acceptable" refers to the non-toxicity of a substance that does not interact with the active ingredients of a pharmaceutical composition.

The term "pharmaceutically effective amount" or "therapeutically effective amount" refers to an amount that alone or in combination with further dosages achieves a desired response or desired effect. In the case of treatment of a particular disease, the desired response preferably involves inhibition of the disease process. This includes slowing the progression of the disease and, in particular, interrupting or reversing the progression of the disease. The desired response in treating a disease may also be to delay the onset of the disease or the disease condition or to prevent the onset of the disease or the disease condition. The effective amount of the compositions described herein will depend upon the disease condition being treated, the severity of the disease, the individual parameters of the patient, including age, physiological condition, size and weight, the duration of treatment, the type of concomitant therapy (if any), the particular route of administration, and like factors. Thus, the dosage of administration of the compositions described herein may depend on many such parameters. In the event of an inadequate response in the patient with the initial dose, a higher dose (or a higher dose effectively achieved by a different, more localized route of administration) may be used.

The pharmaceutical compositions of the present disclosure may comprise salts, buffers, preservatives and optionally other therapeutic agents. In one embodiment, the pharmaceutical compositions of the present disclosure comprise one or more pharmaceutically acceptable carriers, diluents, and/or excipients.

Suitable preservatives for use in the pharmaceutical compositions of the present disclosure include, but are not limited to, benzalkonium chloride, chlorobutanol, parabens, and thimerosal.

The term "excipient" as used herein refers to a substance that may be present in a pharmaceutical composition of the present disclosure but is not an active ingredient. Examples of excipients include, but are not limited to, carriers, binders, diluents, lubricants, thickeners, surfactants, preservatives, stabilizers, emulsifiers, buffers, flavoring agents, or coloring agents.

The term "diluent" relates to a diluting (diluting) and/or diluting (thining) agent. Further, the term "diluent" includes any one or more of a fluid, liquid or solid suspension, and/or a mixing medium. Examples of suitable diluents include ethanol, glycerol and water.

The term "carrier" refers to a component that can be natural, synthetic, organic, inorganic, in which the active component is combined to facilitate, enhance, or effect 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 a subject. Suitable carriers include, but are not limited to, sterile water, ringer's solution, ringer's lactate solution, sterile sodium chloride solution, isotonic saline, polyalkylene glycols, hydrogenated naphthalenes, and particularly biocompatible lactide polymers, lactide/glycolide copolymers, or polyoxyethylene/polyoxypropylene copolymers. In one embodiment, the pharmaceutical composition of the present disclosure comprises isotonic saline.

Pharmaceutically acceptable carriers, excipients 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 edge.1985).

The pharmaceutical carrier, excipient or diluent may be selected according to the intended route of administration and standard pharmaceutical practice.

In one embodiment, the pharmaceutical compositions described herein may be administered intravenously, intraarterially, subcutaneously, intradermally, or intramuscularly. In certain embodiments, the pharmaceutical composition is formulated for local administration or systemic administration. Systemic administration may include enteral administration, which includes absorption through the gastrointestinal tract, or parenteral administration. As used herein, the term "parenteral administration" refers to administration by any means other than through the gastrointestinal tract, such as by intravenous injection. In a preferred embodiment, the pharmaceutical composition is formulated for intramuscular administration. In another embodiment, the pharmaceutical composition is formulated for systemic administration, e.g., for intravenous administration.

The term "co-administration" as used herein refers to the process of administering different compounds or compositions (e.g., an RNA encoding an antigen and an RNA encoding an immunostimulant) to the same patient. The different compounds or compositions may be administered simultaneously, substantially simultaneously or sequentially.

The pharmaceutical compositions and products described herein may be provided as a frozen concentrate of a solution for injection, e.g., at a concentration of 0.50 mg/mL. In one embodiment, to prepare a solution for injection, the drug is thawed and diluted with an isotonic sodium chloride solution (e.g., 0.9% NaCl, saline), e.g., by a one-step dilution method. In some embodiments, a bacteriostatic sodium chloride solution (e.g., 0.9% NaCl, saline) may not be used as a diluent. In some embodiments, the diluted pharmaceutical product is an off-white suspension. The concentration of the final solution for injection varies according to the respective dosage level to be administered.

In one embodiment, administration is performed within 6 hours after the start of preparation due to the risk of microbial contamination and considering the multi-dose approach of the preparation process. In one embodiment, during this 6 hour period, two conditions are allowed: room temperature for preparation, handling and transfer, and 2-8 ℃ for storage.

The compositions described herein can be transported and/or stored under controlled temperature conditions, for example, temperature conditions of about 4-5 ℃ or less, about-20 ℃ or less, -70 ℃ ± 10 ℃ (e.g., -80 ℃ to-60 ℃), for example, utilizing a cooling system (e.g., which can be or include dry ice) to maintain the desired temperature. In one embodiment, the compositions described herein may be transported in a controlled temperature insulated transport (thermal spreader). Such insulated vehicles may contain GPS-enabled thermal sensors to track the location and temperature of each transport. The composition may be stored by refilling with, for example, dry ice.

Treatment of

The present invention provides methods and agents for inducing an adaptive immune response against a coronavirus in a subject comprising administering an effective amount of a composition described herein comprising an RNA encoding a coronavirus vaccine antigen.

In one embodiment, the methods and agents described herein provide immunity to a coronavirus, a coronavirus infection, or a coronavirus-related disease or disorder in a subject. The present invention thus provides methods and agents for treating or preventing infections, diseases or disorders associated with coronaviruses.

In one embodiment, the methods and agents described herein are administered to a subject having an infection, disease, or condition associated with a coronavirus. In one embodiment, the methods and agents described herein are administered to a subject at risk of developing an infection, disease, or condition associated with a coronary virus. For example, the methods and agents described herein can be administered to a subject at risk of contact with a coronavirus. In one embodiment, the methods and agents described herein are administered to a subject who lives, travels, or is expected to travel to a geographic area where coronavirus epidemics occur. In one embodiment, the methods and agents described herein are administered to a subject in contact with or expected to be in contact with another person who lives, travels or is expected to travel to a geographic area where the coronavirus is prevalent. In one embodiment, the methods and agents described herein are administered to a subject known to be exposed to coronavirus by occupational or other exposure thereof. In one embodiment, the coronavirus is SARS-CoV-2. In some embodiments, the methods and agents described herein are administered to a subject who has evidence of prior exposure to and/or infection with SARS-CoV-2 and/or an antigen or epitope thereof or cross-reactivity therewith. For example, in some embodiments, the methods and agents described herein are administered to a subject in which antibodies reactive with one or more epitopes of the SARS-CoV-2 spike protein are detectable and/or have been detected.

For a composition to be used as a vaccine, the composition must induce an immune response against a coronavirus antigen in a cell, tissue, or subject (e.g., a human). In some embodiments, the composition induces an immune response against a coronavirus antigen in a cell, tissue, or subject (e.g., a human). In certain instances, the vaccine induces a protective immune response in a mammal. A therapeutic compound or composition of the invention can be administered prophylactically (i.e., to prevent a disease or condition) or therapeutically (i.e., to treat a disease or condition) to a subject suffering from or at risk of (or susceptible to) developing a disease or condition. Such subjects can be identified using standard clinical methods. In the context of the present invention, prophylactic administration occurs before overt clinical symptoms of the disease are manifested, thereby preventing the disease or disorder or delaying its development. In the context of the medical field, the term "prevention" encompasses any activity that reduces the mortality or morbidity burden caused by the disease. Prevention can occur at primary, secondary, and tertiary prevention levels. While primary prevention avoids the development of disease, secondary and tertiary levels of prevention encompass activities aimed at preventing disease development and the appearance of symptoms, as well as reducing the negative effects of established disease by restoring function and reducing disease-related complications.

The present disclosure reports various characterizations of the provided compositions (see, e.g., example 2; see also examples hereinafter), and further establishes parameters for vaccines that are effective in humans.

In some embodiments, administration of the immunogenic composition or vaccine of the invention may be by a single administration or boosted by multiple administrations.

In some embodiments, an amount of RNA described herein of 0.1 μ g to 300 μ g, 0.5 μ g to 200 μ g, or 1 μ g to 100 μ g, such as about 1 μ g, about 3 μ g, about 10 μ g, about 30 μ g, about 50 μ g, or about 100 μ g, can be administered per dose. In one embodiment, the present invention contemplates administration of a single dose. In one embodiment, the present invention envisions administration of a prime dose followed by one or more booster doses. The booster or first booster dose may be administered 7-28 days or 14-24 days after the administration of the prime dose.

In some embodiments, an amount of RNA described herein of 60 μ g or less, 50 μ g or less, 40 or less, 30 μ g or less, 20 μ g or less, 10 μ g or less, 5 μ g or less, 2.5 μ g or less, or 1 μ g or less may be administered per dose.

In some embodiments, an amount of at least 0.25 μ g, at least 0.5 μ g, at least 1 μ g, at least 2 μ g, at least 3 μ g, at least 4 μ g, at least 5 μ g, at least 10 μ g, at least 20 μ g, at least 30 μ g, or at least 40 μ g of an RNA described herein can be administered per dose.

In some embodiments, an amount of RNA described herein of 0.25 μ g to 60 μ g, 0.5 μ g to 55 μ g, 1 μ g to 50 μ g, 5 μ g to 40 μ g, or 10 μ g to 30 μ g may be administered per dose.

In one embodiment, an amount of about 30 μ g of RNA described herein is administered per dose. In one embodiment, at least two such doses are administered. For example, the second dose may be administered about 21 days after the first dose is administered.

In some embodiments, the potency of an RNA vaccine described herein (e.g., administered in two doses, wherein the second dose can be administered about 21 days after administration of the first dose, and e.g., administered in an amount of about 30 μ g per dose) is at least 70%, at least 80%, at least 90, or at least 95% at the beginning 7 days after administration of the second dose (e.g., at the beginning 28 days after administration of the first dose if the second dose is administered 21 days after administration of the first dose). In some embodiments, such efficacy is observed in a population at least 50 years of age, at least 55 years of age, at least 60 years of age, at least 65 years of age, at least 70 years of age, or older. In some embodiments, the potency of an RNA vaccine described herein (e.g., administered in two doses, wherein the second dose can be administered about 21 days after administration of the first dose, and, for example, administered in an amount of about 30 μ g per dose) is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, or at least 95% in a population that is at least 65 years old, such as 65-80, 65-75, or 65-70 years old, at least 7 days after administration of the second dose (e.g., 28 days after administration of the first dose if the second dose is administered 21 days after administration of the first dose). Such efficacy can be observed over a period of up to 1 month, 2 months, 3 months, 6 months, or more.

In one embodiment, vaccine efficacy is defined as the percentage of reduction in the number of subjects with evidence of infection (vaccinated subjects versus unvaccinated subjects).

In one embodiment, efficacy is assessed by monitoring potential cases of COVID-19. For purposes herein, a patient may be considered to have COVID-19 disease at any time if the patient develops an acute respiratory disease. The evaluation may include nasal (middle turbinate) swabs, which may be tested using reverse transcription-polymerase chain reaction (RT-PCR) to detect SARS-CoV-2. In addition, clinical information and results from local standard of care tests can be evaluated.

In some embodiments, efficacy assessment may utilize the definition of SARS-CoV-2 associated cases, wherein:

confirmed COVID-19: at least 1 or less symptom and positive SARS-CoV-2NAAT (nucleic acid amplification based test) was present during the period of symptoms or within 4 days before or after the period of symptoms: fever is caused; new or increased coughing; new or increased tachypnea; chills; new or increased muscle pain; loss of new taste or smell; sore throat; diarrhea; vomiting.

Alternatively or additionally, in some embodiments, efficacy assessment may utilize the definition of SARS-CoV-2 related cases, where one or more of the following additional symptoms defined by CDC may be considered: fatigue; headache, nasal obstruction or discharge; nausea.

In some embodiments, efficacy assessment can utilize the definition of SARS-CoV-2 associated severe cases

Confirmed severe COVID-19: diagnosed COVID-19 and at least 1 of the following: clinical manifestations of severe systemic disease are indicated at rest (e.g., RR ≧ 30 breaths/min, HR ≧ 125 beats/min, SpO in room air at sea level2Less than or equal to 93%, or PaO2/FiO2<300mm Hg); respiratory failure (which may be defined as a need for high flow of oxygen, non-invasive ventilation, mechanical ventilation or ECMO); evidence of shock (e.g., SBP)<90mm Hg 、DBP<60mm Hg or requiring vascular compressionAn agent); severe acute renal, hepatic or neurological dysfunction; entering an ICU; and death.

Alternatively or additionally, in some embodiments, the serological definition may be applied to patients without COVID-19 clinical manifestations: for example, serum has been demonstrated to switch to SARS-CoV-2 without the confirmed COVID-19: for example, a positive N-binding antibody results in a patient with a previous negative N-binding antibody result.

In some embodiments, any or all of the following assays may be performed on a serum sample: SARS-CoV-2 neutralization assay; s1 binding IgG level determination; RBD binding IgG level assay; n binding antibody assay.

In one embodiment, the methods and agents described herein can be administered to a pediatric population. In various embodiments, the pediatric population comprises or consists of subjects under 18 years of age, e.g., 5 to under 18 years of age, 12 to under 18 years of age, 16 to under 18 years of age, 12 to under 16 years of age, or 5 to under 12 years of age. In various embodiments, the pediatric population includes or consists of subjects under 5 years of age, e.g., under 2 to 5 years of age, under 12 to 24 months, under 7 to 12 months, or under 6 months.

In one embodiment, the pediatric population comprises or consists of subjects under 12 to 18 years of age, including subjects under 16 to 18 years of age and/or subjects under 12 to 16 years of age. In this embodiment, treatment may include 2 vaccinations separated by 21 days, wherein in one embodiment the vaccine is administered in an amount of 30 μ g RNA per dose, for example by intramuscular administration.

In one embodiment, the pediatric population comprises or consists of subjects under 5 to 18 years of age, including subjects under 12 to 18 years of age and/or subjects under 5 to 12 years of age. In this embodiment, treatment may include 2 vaccinations at 21 day intervals, wherein in various embodiments the vaccine is administered in an amount of 10 μ g, 20 μ g, or 30 μ g RNA per dose, e.g., by intramuscular administration.

In one embodiment, the pediatric population comprises or consists of subjects under 5 years of age, including subjects under 2 to 5 years of age, subjects under 12 to 24 months, subjects under 7 to 12 months, subjects under 6 to 12 months and/or subjects under 6 months. In this embodiment, treatment may comprise 2 vaccinations, e.g., 21-42 days apart, e.g., 21 days apart, wherein in various embodiments the vaccine is administered in an amount of 10 μ g, 20 μ g, or 30 μ g RNA per dose, e.g., by intramuscular administration.

In some embodiments, the mRNA compositions described herein can be evaluated for efficacy in a pediatric population (e.g., as described herein) by various indicators described herein (including, for example, but not limited to, the incidence of COVID-19 every 1000 human-years in subjects without serological or virological evidence of past SARS-CoV-2 infection; e.g., the geometric mean ratio of the neutralization titers of SARS CoV-2 measured 7 days after the second dose (GMR); etc.).

In some embodiments, following administration of an RNA composition (e.g., mRNA) described herein, the pediatric population described herein (e.g., 12 to under 16 years of age) can be monitored for the occurrence of Multisystem Inflammatory Syndrome (MIS) (e.g., inflammation in various body parts such as heart, lung, kidney, brain, skin, eye, and/or gastrointestinal organs). Exemplary symptoms of MIS in children may include, but are not limited to, fever, abdominal pain, vomiting, diarrhea, neck pain, rash, ocular congestion, extreme tiredness, and combinations thereof.

In one embodiment, the RNA administered as described above is a nucleoside-modified messenger RNA (modrna) described herein as BNT162b1(RBP020.3), BNT162b2(RBP020.1 or RBP 020.2). In one embodiment, the RNA administered as described above is a nucleoside-modified messenger RNA (modrna) described herein as RBP 020.2.

In one embodiment, the RNA administered as described above is a nucleoside-modified messenger RNA (modrna), and (i) comprises SEQ ID NO:21, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 21, and/or (ii) a nucleotide sequence encoding an amino acid sequence comprising the nucleotide sequence of SEQ ID NO:5, or an amino acid sequence identical to SEQ ID NO:5 has an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical. In one embodiment, the RNA administered as described above is a nucleoside-modified messenger RNA (modRNA), and (i) comprises the nucleotide sequence of SEQ ID NO: 21; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO: 5.

In one embodiment, the RNA administered as described above is a nucleotide modified messenger RNA (modRNA) and (i) comprises the nucleotide sequence of SEQ ID NO 19 or 20, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO 19 or 20, and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 7, or a nucleotide sequence that is identical to the nucleotide sequence of SEQ ID NO:7 has an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical. In one embodiment, the RNA administered as described above is a nucleoside-modified messenger RNA (modrna), and (i) comprises SEQ ID NO:19 or 20; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 7.

In one embodiment, the RNA administered as described above is a nucleotide modified messenger RNA (modrna) and (i) comprises the nucleotide sequence of SEQ ID NO:20, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO:20, and/or (ii) encodes an amino acid sequence comprising the nucleotide sequence of SEQ ID NO:7, or an amino acid sequence identical to SEQ ID NO:7 has an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical. In one embodiment, the RNA administered as described above is a nucleoside-modified messenger RNA (modRNA), and (i) comprises the nucleotide sequence of SEQ ID NO: 20; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 7.

In one embodiment, the RNA administered is a nucleoside-modified messenger RNA (modrna), and (i) comprises SEQ ID NO: 20; and/or (ii) encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NO 7 and is administered in an amount of about 30 μ g/dose. In one embodiment, at least two such doses are administered. For example, the second dose may be administered about 21 days after the first dose is administered.

In some embodiments, a population treated with an RNA described herein comprises, consists essentially of, or consists of: a subject at an age of at least 50 years, at least 55 years, at least 60 years, or at least 65 years. In some embodiments, a population treated with an RNA described herein comprises, consists essentially of, or consists of: subjects aged 55-90 years, 60-85 years, or 65-85 years.

In some embodiments, the time period between doses administered is at least 7 days, at least 14 days, or at least 21 days. In some embodiments, the time period between doses administered is 7 days to 28 days, such as 14 days to 23 days.

In some embodiments, no more than 5 doses, no more than 4 doses, or no more than 3 doses of RNA described herein can be administered to a subject.

In some embodiments, the methods and agents described herein are administered (e.g., in doses, dose frequencies, and/or dose amounts) such that Adverse Events (AEs), i.e., any undesirable medical condition in the patient, e.g., any adverse and undesirable signs, symptoms, or diseases associated with the use of the drug product, whether or not associated with the drug product, are mild or moderate in intensity. In some embodiments, the methods and agents described herein are administered such that Adverse Events (AEs) can be managed by intervention, such as treatment with paracetamol or other drugs that provide analgesic, antipyretic (reduced fever), and/or anti-inflammatory effects, e.g., non-steroidal anti-inflammatory drugs (NSAIDs), e.g., aspirin, ibuprofen, and naproxen. Paracetamol or "paracetamol", which is not classified as an NSAID, exerts a weak anti-inflammatory effect and may be administered as an analgesic according to the present invention.

In some embodiments, the methods and agents described herein provide a neutralizing effect on a coronavirus, a coronavirus infection, or a coronavirus-associated disease or disorder in a subject.

In some embodiments, the methods and agents described herein induce an immune response in a subject that blocks or neutralizes coronavirus after administration to the subject. In some embodiments, the methods and agents described herein induce the production of antibodies, such as IgG antibodies, that block or neutralize coronaviruses in a subject following administration to the subject. In some embodiments, the methods and agents described herein induce an immune response in a subject that blocks or neutralizes the binding of the coronary virus S protein to ACE2 following administration to the subject. In some embodiments, the methods and agents described herein induce the production of antibodies that block or neutralize binding of coronavirus S protein to ACE2 in a subject following administration to the subject.

In some embodiments, the methods and agents described herein induce a Geometric Mean Concentration (GMC) of RBD domain-binding antibodies, such as IgG antibodies, of at least 500U/ml, 1000U/ml, 2000U/ml, 3000U/ml, 4000U/ml, 5000U/ml, 10000U/ml, 15000U/ml, 20000U/ml, 25000U/ml, 30000U/ml, or even higher in a subject following administration to the subject. In some embodiments, the elevated GMC of the RBD domain-binding antibody is for at least 14 days, 21 days, 28 days, 1 month, 3 months, 6 months, 12 months, or even longer.

In some embodiments, the methods and agents described herein induce a Geometric Mean Titer (GMT) of neutralizing antibodies, such as IgG antibodies, in the subject of at least 100U/ml, 200U/ml, 300U/ml, 400U/ml, 500U/ml, 1000U/ml, 1500U/ml, or even higher after administration to the subject. In some embodiments, the elevated GMT of the neutralizing antibody persists for at least 14 days, 21 days, 28 days, 1 month, 3 months, 6 months, 12 months, or even longer.

As used herein, the term "neutralizing" refers to an event in which a binding agent, such as an antibody, binds to a biologically active site of a virus, such as a receptor binding protein, thereby inhibiting viral infection of a cell. As used herein, the term "neutralizing" with respect to a coronavirus, particularly a coronavirus S protein, refers to an event in which a binding agent, such as an antibody, binds to the RBD domain of the S protein, thereby inhibiting viral infection of a cell. In particular, the term "neutralizing" refers to an event in which the binding agent eliminates or significantly reduces the virulence (e.g., the ability to infect cells) of the virus of interest.

The type of immune response generated in response to an antigen challenge can generally be distinguished by a subset of T helper (Th) cells involved in the response. Immune responses can be broadly divided into two types: th1 and Th 2. Th1 immune activation is optimized for intracellular infections such as viruses, while Th2 immune responses are optimized for humoral (antibody) responses. Th1 cells produce interleukin 2(IL-2), tumor necrosis factor (TNF α), and interferon γ (IFN γ). Th2 cells produce IL-4, IL-5, IL-6, IL-9, IL-10 and IL-13. Th1 immune activation is most desirable in many clinical situations. Vaccine compositions that specifically elicit a Th2 or humoral immune response are generally not effective against most viral diseases.

In some embodiments, the methods and agents described herein induce or promote a Th 1-mediated immune response in a subject following administration to the subject. In some embodiments, the methods and agents described herein induce or promote a cytokine profile characteristic of a Th 1-mediated immune response in a subject following administration to the subject. In some embodiments, the methods and agents described herein induce or promote the production of interleukin 2(IL-2), tumor necrosis factor (TNF α), and/or interferon gamma (IFN γ) in a subject following administration to the subject. In some embodiments, the methods and agents described herein induce or promote the production of interleukin 2(IL-2) and interferon gamma (IFN γ) in a subject following administration to the subject. In some embodiments, the methods and agents described herein do not induce or promote a Th 2-mediated immune response in the subject, or induce or promote a significantly lower degree of a Th 2-mediated immune response in the subject than the induction or promotion of a Th 1-mediated immune response, after administration to the subject. In some embodiments, the methods and agents described herein do not induce or promote a cytokine profile typical of a Th 2-mediated immune response in a subject, or induce or promote a cytokine profile typical of a Th 2-mediated immune response to a significantly lower degree in a subject than the induction or promotion of a cytokine profile typical of a Th 1-mediated immune response after administration to a subject. In some embodiments, the methods and agents described herein do not induce or promote the production of IL-4, IL-5, IL-6, IL-9, IL-10, and/or IL-13, or induce or promote the production of IL-4, IL-5, IL-6, IL-9, IL-10, and/or IL-13 to a significantly lower degree in a subject than the induction or promotion of interleukin 2(IL-2), tumor necrosis factor (TNF α), and/or interferon γ (IFN γ) in the subject, following administration to the subject. In some embodiments, the methods and agents described herein do not induce or promote the production of IL-4 after administration to a subject, or induce or promote the production of IL-4 in a subject to a significantly lower degree than the induction or promotion of interleukin 2(IL-2) and interferon gamma (IFN γ) in a subject.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets a panel of different S protein variants, such as SARS-CoV-2S protein variants, particularly naturally occurring S protein variants, following administration to the subject. In some embodiments, the set of different S protein variants comprises at least 5, at least 10, at least 15, or even more S protein variants. In some embodiments, such S protein variants comprise variants having amino acid modifications in the RBD domain and/or variants having amino acid modifications outside the RBD domain. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof wherein the amino acid corresponding to position 321(Q) in SEQ ID NO:1 is S. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the amino acid corresponding to position 321(Q) in SEQ ID NO:1 is L. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the amino acid corresponding to position 341(V) in SEQ ID NO:1 is I. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the amino acid corresponding to position 348(A) in SEQ ID NO:1 is T. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the amino acid corresponding to position 354(N) in SEQ ID NO:1 is D. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof wherein the amino acid corresponding to position 359(S) in SEQ ID NO:1 is N. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the amino acid corresponding to position 367(V) in SEQ ID NO:1 is F. In one embodiment, such S protein variants comprise a SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the sequence corresponding to SEQ ID NO:1 the amino acid at position 378(K) is S. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof wherein the amino acid corresponding to position 378(K) in SEQ ID NO:1 is R. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the sequence corresponding to SEQ ID NO: the amino acid at position 408(R) in 1 is I. In one embodiment, such S protein variants comprise a SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the sequence corresponding to SEQ ID NO: the amino acid at position 409(Q) in 1 is E. In one embodiment, such S protein variants comprise a SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the sequence corresponding to SEQ ID NO: the amino acid at position 435(A) in 1 is S. In one embodiment, such S protein variants comprise a SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the sequence corresponding to SEQ ID NO: the amino acid at position 439(N) in 1 is K. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the amino acid corresponding to position 458(K) in SEQ ID NO:1 is R. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the amino acid corresponding to position 472(I) in SEQ ID NO:1 is V. In one embodiment, such S protein variants comprise a SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the sequence corresponding to SEQ ID NO: the amino acid at position 476(G) in 1 is S. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof wherein the amino acid corresponding to position 477(S) in SEQ ID NO:1 is N. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the amino acid corresponding to position 483(V) in SEQ ID NO:1 is A. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the amino acid corresponding to position 508(Y) in SEQ ID NO:1 is H. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof wherein the amino acid corresponding to position 519(H) in SEQ ID NO:1 is P. In one embodiment, such S protein variants comprise the SARS-CoV-2S protein or a naturally occurring variant thereof, wherein the amino acid corresponding to position 614(D) in SEQ ID NO:1 is G.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets an S protein variant, such as a SARS-CoV-2S protein variant, particularly a naturally occurring S protein variant comprising a mutation at a position corresponding to position 501(N) in SEQ ID NO: 1. In one embodiment, the amino acid corresponding to position 501(N) in SEQ ID NO:1 is Y.

The S protein variant comprising a mutation at a position corresponding to position 501(N) in SEQ ID NO. 1 may comprise one or more further mutations. Such one or more further mutations may be selected from mutations at positions corresponding to the following positions in SEQ ID NO: 1: 69(H), 70(V), 144(Y), 570(A), 614(D), 681(P), 716(T), 982(S), 1118(D), 80(D), 215(D), 484(E), 701(A), 18(L), 246(R), 417(K), 242(L), 243(A), and 244 (L). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid 69 (H). In one embodiment, the amino acid corresponding to position 70(V) in SEQ ID NO 1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid at position 144 (Y). In one embodiment, the amino acid corresponding to position 570(A) in SEQ ID NO:1 is D. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 614(D) in 1 is G. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 681(P) in 1 is H. In one embodiment, the amino acid corresponding to position 716(T) in SEQ ID NO:1 is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 982(S) is a. In one embodiment, the amino acid corresponding to position 1118(D) in SEQ ID NO:1 is H. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 80(D) is A. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 215(D) in 1 is G. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 484(E) in 1 is K. In one embodiment, the amino acid corresponding to position 701(A) in SEQ ID NO 1 is V. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 18(L) is F. In one embodiment, the amino acid corresponding to position 246(R) in SEQ ID NO:1 is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 417(K) in 1 is N. In one embodiment, the amino acid corresponding to position 242(L) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 243(A) in SEQ ID NO 1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 244 (L).

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets VOC-202012/01 after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: deletions 69-70, deletion 144, N501Y, a570D, D614G, P681H, T716I, S982A, and D1118H.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets 501.V2 after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: D80A, D215G, E484K, N501Y and a701V, and optionally: L18F, R246I, K417N and deletions 242-244. The S protein variant may also be modified in a sequence corresponding to SEQ ID NO:1 comprises a D- > G mutation at position 614.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets S protein variants, such as SARS-CoV-2S protein variants, particularly naturally occurring S protein variants comprising deletions at positions corresponding to positions 69(H) and 70(V) in SEQ ID NO: 1.

In some embodiments, a S protein variant comprising deletions at positions corresponding to positions 69(H) and 70(V) in SEQ ID No. 1 may comprise one or more further mutations. Such one or more further mutations may be selected from the group consisting of: 1 at the following positions: 144(Y), 501(N), 570(A), 614(D), 681(P), 716(T), 982(S), 1118(D), 80(D), 215(D), 484(E), 701(A), 18(L), 246(R), 417(K), 242(L), 243(A), 244(L), 453(Y), 692(I), 1147(S) and 1229 (M). In one embodiment, the amino acid corresponding to position 144(Y) in SEQ ID NO 1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 501(N) in 1 is Y. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 570(A) in 1 is D. In one embodiment, the amino acid corresponding to position 614(D) in SEQ ID NO 1 is G. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 681(P) in 1 is H. In one embodiment, the amino acid corresponding to position 716(T) in SEQ ID NO:1 is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 982(S) is a. In one embodiment, the amino acid corresponding to position 1118(D) in SEQ ID NO:1 is H. In one embodiment, the amino acid corresponding to position 80(D) in SEQ ID NO:1 is A. In one embodiment, the amino acid corresponding to position 215(D) in SEQ ID NO:1 is G. In one embodiment, the amino acid corresponding to position 484(E) in SEQ ID NO 1 is K. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 701(A) in 1 is V. In one embodiment, the amino acid corresponding to position 18(L) in SEQ ID NO:1 is F. In one embodiment, the amino acid corresponding to position 246(R) in SEQ ID NO:1 is I. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is N. In one embodiment, the amino acid corresponding to position 242(L) in SEQ ID NO 1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 243 (a). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 244 (L). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 453(Y) in 1 is F. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 692(I) in 1 is V. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 1147(S) is L. In one embodiment, the amino acid corresponding to position 1229(M) in SEQ ID NO:1 is I.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets VOC-202012/01 after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: deletions 69-70, deletion 144, N501Y, a570D, D614G, P681H, T716I, S982A, and D1118H.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets "Cluster 5" after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: deletion 69-70, Y453F, I692V, M1229I and optionally S1147L.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets an S protein variant, such as a SARS-CoV-2S protein variant, particularly a naturally occurring S protein variant comprising a mutation at a position corresponding to position 614(D) in SEQ ID NO: 1. In one embodiment, the amino acid corresponding to position 614(D) in SEQ ID NO 1 is G.

In some embodiments, the polypeptide is encoded in a sequence corresponding to SEQ ID NO: the S protein variant comprising a mutation at position 614(D) in 1 may comprise one or more further mutations. Such one or more further mutations may be selected from the group consisting of: 1 at the following positions: 69(H), 70(V), 144(Y), 501(N), 570(A), 681(P), 716(T), 982(S), 1118(D), 80(D), 215(D), 484(E), 701(A), 18(L), 246(R), 417(K), 242(L), 243(A), 244(L), 453(Y), 692(I), 1147(S), and 1229 (M). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid 69 (H). In one embodiment, the amino acid corresponding to position 70(V) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 144(Y) in SEQ ID NO 1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 501(N) in 1 is Y. In one embodiment, the amino acid corresponding to position 570(A) in SEQ ID NO:1 is D. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 681(P) in 1 is H. In one embodiment, the amino acid corresponding to position 716(T) in SEQ ID NO:1 is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 982(S) is a. In one embodiment, the amino acid corresponding to position 1118(D) in SEQ ID NO:1 is H. In one embodiment, the amino acid corresponding to position 80(D) in SEQ ID NO:1 is A. In one embodiment, the amino acid corresponding to position 215(D) in SEQ ID NO:1 is G. In one embodiment, the amino acid corresponding to position 484(E) in SEQ ID NO 1 is K. In one embodiment, the amino acid corresponding to position 701(A) in SEQ ID NO:1 is V. In one embodiment, the amino acid corresponding to position 18(L) in SEQ ID NO:1 is F. In one embodiment, the amino acid corresponding to position 246(R) in SEQ ID NO:1 is I. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is N. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 (L) amino acid deletion at position 242. In one embodiment, the amino acid corresponding to position 243(A) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 244(L) in SEQ ID NO:1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 453(Y) in 1 is F. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 692(I) in 1 is V. In one embodiment, the amino acid corresponding to position 1147(S) in SEQ ID NO 1 is L. In one embodiment, the amino acid corresponding to position 1229(M) in SEQ ID NO:1 is I.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets VOC-202012/01 after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: deletions 69-70, deletion 144, N501Y, a570D, D614G, P681H, T716I, S982A, and D1118H.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: D80A, D215G, E484K, N501Y, D614G and a701V, and optionally: L18F, R246I, K417N and deletions 242-244.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets S protein variants, such as SARS-CoV-2S protein variants, particularly naturally occurring S protein variants comprising mutations at positions corresponding to positions 501(N) and 614(D) in SEQ ID NO: 1. In one embodiment, the amino acid corresponding to position 501(N) in SEQ ID NO:1 is Y and the amino acid corresponding to position 614(D) in SEQ ID NO:1 is G.

In some embodiments, S protein variants comprising mutations at positions corresponding to positions 501(N) and 614(D) in SEQ ID NO. 1 may comprise one or more additional mutations. Such one or more further mutations may be selected from mutations at positions corresponding to the following positions in SEQ ID NO: 1: 69(H), 70(V), 144(Y), 570(A), 681(P), 716(T), 982(S), 1118(D), 80(D), 215(D), 484(E), 701(A), 18(L), 246(R), 417(K), 242(L), 243(A), 244(L), 453(Y), 692(I), 1147(S), and 1229 (M). In one embodiment, the amino acid corresponding to position 69(H) in SEQ ID NO:1 is deleted. In one embodiment, the amino acid corresponding to position 70(V) in SEQ ID NO 1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid at position 144 (Y). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 570(A) in 1 is D. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 681(P) in 1 is H. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 716(T) in 1 is I. In one embodiment, the amino acid corresponding to position 982(S) in SEQ ID NO 1 is A. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 1118(D) in 1 is H. In one embodiment, the amino acid corresponding to position 80(D) in SEQ ID NO:1 is A. In one embodiment, the amino acid corresponding to position 215(D) in SEQ ID NO:1 is G. In one embodiment, the amino acid corresponding to position 484(E) in SEQ ID NO 1 is K. In one embodiment, the amino acid corresponding to position 701(A) in SEQ ID NO 1 is V. In one embodiment, the amino acid corresponding to position 18(L) in SEQ ID NO:1 is F. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 246(R) is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 417(K) in 1 is N. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 (L) amino acid deletion at position 242. In one embodiment, the amino acid corresponding to position 243(A) in SEQ ID NO 1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 244 (L). In one embodiment, the amino acid corresponding to position 453(Y) in SEQ ID NO 1 is F. In one embodiment, the amino acid corresponding to position 692(I) in SEQ ID NO:1 is V. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 1147(S) is L. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 1229(M) in 1 is I.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets VOC-202012/01 after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: deletions 69-70, deletion 144, N501Y, a570D, D614G, P681H, T716I, S982A, and D1118H.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: D80A, D215G, E484K, N501Y, D614G and a701V, and optionally: L18F, R246I, K417N and deletions 242-244.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets an S protein variant, such as a SARS-CoV-2S protein variant, particularly a naturally occurring S protein variant comprising a mutation at a position corresponding to position 484(E) in SEQ ID NO: 1. In one embodiment, the amino acid corresponding to position 484(E) in SEQ ID NO 1 is K.

In some embodiments, a variant S protein comprising a mutation at a position corresponding to position 484(E) in SEQ ID No. 1 may comprise one or more further mutations. Such one or more further mutations may be selected from mutations at positions corresponding to the following positions in SEQ ID NO: 1: 69(H), 70(V), 144(Y), 501(N), 570(A), 614(D), 681(P), 716(T), 982(S), 1118(D), 80(D), 215(D), 701(A), 18(L), 246(R), 417(K), 242(L), 243(A), 244(L), 453(Y), 692(I), 1147(S), 1229(M), 20(T), 26(P), 138(D), 190(R), 417(K), 655(H), 1027(T) and 1176 (V). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid 69 (H). In one embodiment, the amino acid corresponding to position 70(V) in SEQ ID NO 1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid at position 144(Y) is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 501(N) in 1 is Y. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 570(A) in 1 is D. In one embodiment, the amino acid corresponding to position 614(D) in SEQ ID NO 1 is G. In one embodiment, the amino acid corresponding to position 681(P) in SEQ ID NO:1 is H. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 716(T) in 1 is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 982(S) is a. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid in position 1118(D) in 1 is H. In one embodiment, the amino acid corresponding to position 80(D) in SEQ ID NO:1 is A. In one embodiment, the amino acid corresponding to position 215(D) in SEQ ID NO:1 is G. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 701(A) in 1 is V. In one embodiment, the amino acid corresponding to position 18(L) in SEQ ID NO:1 is F. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 246(R) is I. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is N. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 (L) amino acid deletion at position 242. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 243 (a). In one embodiment, the amino acid corresponding to position 244(L) in SEQ ID NO:1 is deleted. In one embodiment, the amino acid corresponding to position 453(Y) in SEQ ID NO 1 is F. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 692(I) in 1 is V. In one embodiment, the amino acid corresponding to position 1147(S) in SEQ ID NO 1 is L. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 1229(M) is I. In one embodiment, the nucleic acid sequence corresponding to SEQ ID NO:1 the amino acid at position 20(T) is N. In one embodiment, the amino acid corresponding to position 26(P) in SEQ ID NO:1 is S. In one embodiment, the amino acid corresponding to position 138(D) in SEQ ID NO:1 is Y. In one embodiment, the amino acid corresponding to position 190(R) in SEQ ID NO:1 is S. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is T. In one embodiment, the amino acid corresponding to position 655(H) in SEQ ID NO:1 is Y. In one embodiment, the amino acid corresponding to position 1027(T) in SEQ ID NO 1 is I. In one embodiment, the amino acid corresponding to position 1176(V) of SEQ ID NO:1 is F.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets 501.V2 after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: D80A, D215G, E484K, N501Y and a701V, and optionally: L18F, R246I, K417N and deletions 242-244. The S protein variant may also comprise a D- > G mutation at a position corresponding to position 614 in SEQ ID NO 1.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets "b.1.1.28" after administration to the subject.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets "b.1.1.248" after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, H655Y, T1027I and V1176F.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets S protein variants, such as SARS-CoV-2S protein variants, particularly in a polypeptide corresponding to SEQ ID NO:1 at positions 501(N) and 484(E) comprising a mutation. In one embodiment, the amino acid corresponding to position 501(N) in SEQ ID NO:1 is Y and the amino acid corresponding to SEQ ID NO: the amino acid at position 484(E) in 1 is K.

In some embodiments, the polypeptide is encoded in a sequence corresponding to SEQ ID NO: the S protein variant comprising a mutation at positions 501(N) and 484(E) in position 1 may comprise one or more further mutations. Such one or more further mutations may be selected from the group consisting of: 1 at the following positions: 69(H), 70(V), 144(Y), 570(A), 614(D), 681(P), 716(T), 982(S), 1118(D), 80(D), 215(D), 701(A), 18(L), 246(R), 417(K), 242(L), 243(A), 244(L), 453(Y), 692(I), 1147(S), 1229(M), 20(T), 26(P), 138(D), 190(R), 417(K), 655(H), 1027(T), and 1176 (V). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid 69 (H). In one embodiment, the amino acid corresponding to position 70(V) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 144(Y) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 570(A) in SEQ ID NO:1 is D. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 614(D) in 1 is G. In one embodiment, the amino acid corresponding to position 681(P) in SEQ ID NO:1 is H. In one embodiment, the amino acid corresponding to position 716(T) in SEQ ID NO:1 is I. In one embodiment, the amino acid corresponding to position 982(S) in SEQ ID NO 1 is A. In one embodiment, the amino acid corresponding to position 1118(D) in SEQ ID NO:1 is H. In one embodiment, the amino acid corresponding to position 80(D) in SEQ ID NO:1 is A. In one embodiment, the amino acid corresponding to position 215(D) in SEQ ID NO:1 is G. In one embodiment, the amino acid corresponding to position 701(A) in SEQ ID NO 1 is V. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 18(L) is F. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 246(R) is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 417(K) in 1 is N. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 242 (L). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 243 (a). In one embodiment, the nucleic acid sequence corresponding to SEQ ID NO:1, amino acid deletion at position 244 (L). In one embodiment, the amino acid corresponding to position 453(Y) in SEQ ID NO 1 is F. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 692(I) in 1 is V. In one embodiment, the amino acid corresponding to position 1147(S) in SEQ ID NO 1 is L. In one embodiment, the amino acid corresponding to position 1229(M) in SEQ ID NO:1 is I. In one embodiment, the amino acid corresponding to position 20(T) in SEQ ID NO:1 is N. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 26(P) in 1 is S. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 138(D) in 1 is Y. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 190(R) is S. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is T. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 655(H) in 1 is Y. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 1027(T) is I. In one embodiment, the amino acid corresponding to position 1176(V) in SEQ ID NO 1 is F.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets 501.V2 after administration to the subject.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets a polypeptide that is in a position corresponding to SEQ ID NO: 1, a S protein variant comprising the following mutations at the following positions: D80A, D215G, E484K, N501Y and a701V, and optionally: L18F, R246I, K417N and deletions 242-244. The S protein variant may also be modified in a sequence corresponding to SEQ ID NO: 1 comprises a D- > G mutation at position 614.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets "b.1.1.248" after administration to the subject.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets a polypeptide that is in a position corresponding to SEQ ID NO: 1, a S protein variant comprising the following mutations at the following positions: L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, H655Y, T1027I and V1176F.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets S protein variants, such as SARS-CoV-2S protein variants, particularly in a polypeptide corresponding to SEQ ID NO:1 at positions 501(N), 484(E) and 614(D) comprising mutations. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 501(N) is Y, corresponding to SEQ ID NO: the amino acid at position 484(E) in SEQ ID NO. 1 is K and the amino acid corresponding to position 614(D) in SEQ ID NO. 1 is G.

In some embodiments, the polypeptide is encoded in a sequence corresponding to SEQ ID NO: the S protein variants comprising mutations at positions 501(N), 484(E) and 614(D) in position 1 may comprise one or more further mutations. Such one or more further mutations may be selected from mutations at positions corresponding to the following positions in SEQ ID NO: 1: 69(H), 70(V), 144(Y), 570(A), 681(P), 716(T), 982(S), 1118(D), 80(D), 215(D), 701(A), 18(L), 246(R), 417(K), 242(L), 243(A), 244(L), 453(Y), 692(I), 1147(S), 1229(M), 20(T), 26(P), 138(D), 190(R), 417(K), 655(H), 1027(T), and 1176 (V). In one embodiment, the amino acid corresponding to position 69(H) in SEQ ID NO:1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 70 (V). In one embodiment, the amino acid corresponding to position 144(Y) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 570(A) in SEQ ID NO:1 is D. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 681(P) in 1 is H. In one embodiment, the amino acid corresponding to position 716(T) in SEQ ID NO:1 is I. In one embodiment, the amino acid corresponding to position 982(S) in SEQ ID NO 1 is A. In one embodiment, the amino acid corresponding to position 1118(D) in SEQ ID NO:1 is H. In one embodiment, the amino acid corresponding to position 80(D) in SEQ ID NO:1 is A. In one embodiment, the amino acid corresponding to position 215(D) in SEQ ID NO:1 is G. In one embodiment, the amino acid corresponding to position 701(A) in SEQ ID NO 1 is V. In one embodiment, the amino acid corresponding to position 18(L) in SEQ ID NO:1 is F. In one embodiment, the amino acid corresponding to position 246(R) in SEQ ID NO:1 is I. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is N. In one embodiment, the amino acid corresponding to position 242(L) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 243(A) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 244(L) in SEQ ID NO:1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 453(Y) in 1 is F. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 692(I) in 1 is V. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 1147(S) is L. In one embodiment, the amino acid corresponding to position 1229(M) in SEQ ID NO:1 is I. In one embodiment, the amino acid corresponding to position 20(T) in SEQ ID NO:1 is N. In one embodiment, the amino acid corresponding to position 26(P) in SEQ ID NO:1 is S. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 138(D) in 1 is Y. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 190(R) is S. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is T. In one embodiment, the amino acid corresponding to position 655(H) in SEQ ID NO:1 is Y. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 1027(T) is I. In one embodiment, the amino acid corresponding to position 1176(V) in SEQ ID NO 1 is F.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: D80A, D215G, E484K, N501Y, a701V and D614G, and optionally: L18F, R246I, K417N and deletions 242-244.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets S protein variants, such as SARS-CoV-2S protein variants, particularly naturally occurring S protein variants comprising deletions at positions corresponding to positions 242(L), 243(a), and 244(L) in SEQ ID NO: 1.

In some embodiments, the polypeptide is encoded in a sequence corresponding to SEQ ID NO: the S protein variant comprising deletions at positions 242(L), 243(a) and 244(L) in 1 may comprise one or more further mutations. Such one or more further mutations may be selected from mutations at positions corresponding to the following positions in SEQ ID NO: 1: 69(H), 70(V), 144(Y), 501(N), 570(A), 614(D), 681(P), 716(T), 982(S), 1118(D), 80(D), 215(D), 484(E), 701(A), 18(L), 246(R), 417(K), 453(Y), 692(I), 1147(S), 1229(M), 20(T), 26(P), 138(D), 190(R), 417(K), 655(H), 1027(T) and 1176 (V). In one embodiment, the amino acid corresponding to position 69(H) in SEQ ID NO:1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 70 (V). In one embodiment, the amino acid corresponding to position 144(Y) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 501(N) in SEQ ID NO 1 is Y. In one embodiment, the amino acid corresponding to position 570(A) in SEQ ID NO:1 is D. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 614(D) in 1 is G. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 681(P) in 1 is H. In one embodiment, the amino acid corresponding to position 716(T) in SEQ ID NO:1 is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 982(S) in 1 is A. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 1118(D) in 1 is H. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 80(D) is A. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 215(D) in 1 is G. In one embodiment, the amino acid corresponding to position 484(E) in SEQ ID NO 1 is K. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 701(A) in 1 is V. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 18(L) is F. In one embodiment, the amino acid corresponding to position 246(R) in SEQ ID NO:1 is I. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is N. In one embodiment, the amino acid corresponding to position 453(Y) in SEQ ID NO 1 is F. In one embodiment, the amino acid corresponding to position 692(I) in SEQ ID NO:1 is V. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 1147(S) is L. In one embodiment, the amino acid corresponding to position 1229(M) in SEQ ID NO:1 is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 20(T) is N. In one embodiment, the amino acid corresponding to position 26(P) in SEQ ID NO:1 is S. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 138(D) in 1 is Y. In one embodiment, the amino acid corresponding to position 190(R) in SEQ ID NO:1 is S. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is T. In one embodiment, the amino acid corresponding to position 655(H) in SEQ ID NO:1 is Y. In one embodiment, the amino acid corresponding to position 1027(T) in SEQ ID NO 1 is I. In one embodiment, the amino acid corresponding to position 1176(V) in SEQ ID NO 1 is F.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets 501.V2 after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: D80A, D215G, E484K, N501Y, a701V and deletions 242 and 244, and optionally: L18F, R246I and K417N. The S protein variant may also comprise a D- > G mutation at a position corresponding to position 614 in SEQ ID NO 1.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets an S protein variant, such as a SARS-CoV-2S protein variant, particularly a naturally occurring S protein variant comprising a mutation at a position corresponding to position 417(K) in SEQ ID NO: 1. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 417(K) in 1 is N. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is T.

In some embodiments, a variant S protein comprising a mutation at a position corresponding to position 417(K) in SEQ ID No. 1 may comprise one or more additional mutations. Such one or more further mutations may be selected from mutations at positions corresponding to the following positions in SEQ ID NO: 1: 69(H), 70(V), 144(Y), 501(N), 570(A), 614(D), 681(P), 716(T), 982(S), 1118(D), 80(D), 215(D), 484(E), 701(A), 18(L), 246(R), 242(L), 243(A), 244(L), 453(Y), 692(I), 1147(S), 1229(M), 20(T), 26(P), 138(D), 190(R), 655(H), 1027(T) and 1176 (V). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid 69 (H). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 70 (V). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid at position 144 (Y). In one embodiment, the amino acid corresponding to position 501(N) in SEQ ID NO 1 is Y. In one embodiment, the amino acid corresponding to position 570(A) in SEQ ID NO:1 is D. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 614(D) in 1 is G. In one embodiment, the amino acid corresponding to position 681(P) in SEQ ID NO:1 is H. In one embodiment, the amino acid corresponding to position 716(T) in SEQ ID NO:1 is I. In one embodiment, the amino acid corresponding to position 982(S) in SEQ ID NO 1 is A. In one embodiment, the amino acid corresponding to position 1118(D) in SEQ ID NO:1 is H. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 80(D) is A. In one embodiment, the amino acid corresponding to position 215(D) in SEQ ID NO:1 is G. In one embodiment, the amino acid corresponding to position 484(E) in SEQ ID NO 1 is K. In one embodiment, the amino acid corresponding to position 701(A) in SEQ ID NO 1 is V. In one embodiment, the amino acid corresponding to position 18(L) in SEQ ID NO:1 is F. In one embodiment, the amino acid corresponding to position 246(R) in SEQ ID NO:1 is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 (L) amino acid deletion at position 242. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 243 (a). In one embodiment, the amino acid corresponding to position 244(L) in SEQ ID NO:1 is deleted. In one embodiment, the amino acid corresponding to position 453(Y) in SEQ ID NO 1 is F. In one embodiment, the amino acid corresponding to position 692(I) in SEQ ID NO:1 is V. In one embodiment, the amino acid corresponding to position 1147(S) in SEQ ID NO 1 is L. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 1229(M) is I. In one embodiment, the amino acid corresponding to position 20(T) in SEQ ID NO:1 is N. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 26(P) in 1 is S. In one embodiment, the amino acid corresponding to position 138(D) in SEQ ID NO:1 is Y. In one embodiment, the amino acid corresponding to position 190(R) in SEQ ID NO:1 is S. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 655(H) in 1 is Y. In one embodiment, the amino acid corresponding to position 1027(T) in SEQ ID NO 1 is I. In one embodiment, the amino acid corresponding to position 1176(V) in SEQ ID NO 1 is F.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets 501.V2 after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: D80A, D215G, E484K, N501Y, a701V and K417N, and optionally: L18F, R246I and deletions 242 and 244. The S protein variant may also be modified in a sequence corresponding to SEQ ID NO:1 comprises a D- > G mutation at position 614.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets "b.1.1.248" after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, H655Y, T1027I and V1176F.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets S protein variants, such as SARS-CoV-2S protein variants, particularly naturally occurring S protein variants comprising mutations at positions corresponding to positions 417(K) and 484(E) and/or 501(N) in SEQ ID NO: 1. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is N, the amino acid corresponding to position 484(E) in SEQ ID NO:1 is K and/or the amino acid corresponding to position 501(N) in SEQ ID NO:1 is Y. In one embodiment, the amino acid corresponding to position 417(K) in SEQ ID NO:1 is T, the amino acid corresponding to position 484(E) in SEQ ID NO:1 is K and/or the amino acid corresponding to position 501(N) in SEQ ID NO:1 is Y.

In some embodiments, S protein variants comprising mutations at positions corresponding to positions 417(K) and 484(E) and/or 501(N) in SEQ ID NO:1 may comprise one or more additional mutations. Such one or more further mutations may be selected from the group consisting of: 1 at the following positions: 69(H), 70(V), 144(Y), 570(A), 614(D), 681(P), 716(T), 982(S), 1118(D), 80(D), 215(D), 701(A), 18(L), 246(R), 242(L), 243(A), 244(L), 453(Y), 692(I), 1147(S), 1229(M), 20(T), 26(P), 138(D), 190(R), 655(H), 1027(T), and 1176 (V). In one embodiment, the amino acid corresponding to position 69(H) in SEQ ID NO:1 is deleted. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid deletion at position 70 (V). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1, amino acid at position 144 (Y). In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 570(A) in 1 is D. In one embodiment, the amino acid corresponding to position 614(D) in SEQ ID NO 1 is G. In one embodiment, the amino acid corresponding to position 681(P) in SEQ ID NO:1 is H. In one embodiment, the amino acid corresponding to position 716(T) in SEQ ID NO:1 is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 982(S) is a. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 1118(D) in 1 is H. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 80(D) in 1 is A. In one embodiment, the amino acid corresponding to position 215(D) in SEQ ID NO:1 is G. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 701(A) in 1 is V. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 18(L) is F. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 246(R) in 1 is I. In one embodiment, the amino acid corresponding to position 242(L) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 243(A) in SEQ ID NO 1 is deleted. In one embodiment, the amino acid corresponding to position 244(L) in SEQ ID NO:1 is deleted. In one embodiment, the amino acid corresponding to position 453(Y) in SEQ ID NO 1 is F. In one embodiment, the amino acid corresponding to position 692(I) in SEQ ID NO:1 is V. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 1147(S) is L. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 1229(M) in 1 is I. In one embodiment, the amino acid corresponding to position 20(T) in SEQ ID NO:1 is N. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 26(P) in 1 is S. In one embodiment, the amino acid corresponding to position 138(D) in SEQ ID NO:1 is Y. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 190(R) is S. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 655(H) in 1 is Y. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO:1 the amino acid at position 1027(T) is I. In one embodiment, the nucleotide sequence corresponding to SEQ ID NO: the amino acid at position 1176(V) in 1 is F.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets 501.V2 after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: D80A, D215G, E484K, N501Y, a701V and K417N, and optionally: L18F, R246I and deletions 242 and 244. The S protein variant may also comprise a D- > G mutation at a position corresponding to position 614 in SEQ ID NO 1.

In some embodiments, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in a subject that targets "b.1.1.248" after administration to the subject.

In some embodiments, upon administration to a subject, the methods and agents described herein induce an antibody response, particularly a neutralizing antibody response, in the subject that targets a S protein variant comprising the following mutations at positions corresponding to the following positions in SEQ ID NO: 1: L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, H655Y, T1027I and V1176F.

The term "amino acid corresponding to position …" as used herein refers to the amino acid position number corresponding to the amino acid position number in the SARS-CoV-2S protein, in particular the amino acid sequence shown in SEQ ID NO: 1. The corresponding amino acid positions in other coronavirus S protein variants, such as SARS-CoV-2S protein variants, can be found by alignment with the SARS-CoV-2S protein, in particular with the amino acid sequence shown in SEQ ID NO: 1. It is well known in the art how to align sequences or fragments within a sequence to determine the corresponding position of an amino acid sequence according to the invention within the sequence. Standard sequence alignment programs such as ALIGN, ClustalW or similar programs can be used, usually with default settings.

In some embodiments, the set of different S protein variants targeted by the antibody response comprises at least 5, at least 10, at least 15, or even more S protein variants selected from: Q321S, V341I, a348T, N354D, S359N, V367F, K378S, R408I, Q409E, a435S, K458R, I472V, G476S, V483A, Y508H, H519P, and D614G described above. In some embodiments, the set of different S protein variants targeted by the antibody response comprises all S protein variants of the group consisting of: Q321S, V341I, a348T, N354D, S359N, V367F, K378S, R408I, Q409E, a435S, K458R, I472V, G476S, V483A, Y508H, H519P, and D614G described above.

In some embodiments, the set of different S protein variants targeted by the antibody response comprises at least 5, at least 10, at least 15, or even more S protein variants selected from: Q321L, V341I, a348T, N354D, S359N, V367F, K378R, R408I, Q409E, a435S, N439K, K458R, I472V, G476S, S477N, V483A, Y508H, H519P and D614G described above. In some embodiments, the set of different S protein variants targeted by the antibody response comprises all S protein variants of the group consisting of: Q321L, V341I, a348T, N354D, S359N, V367F, K378R, R408I, Q409E, a435S, N439K, K458R, I472V, G476S, S477N, V483A, Y508H, H519P and D614G described above.

In some embodiments, a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof, e.g., as encoded by an RNA as described herein, comprises one or more mutations of an S protein variant as described herein, such as a SARS-CoV-2S protein variant, particularly a naturally occurring S protein variant. In one embodiment, a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof, e.g., as encoded by RNA described herein, comprises a mutation at a position corresponding to position 501(N) in SEQ ID NO: 1. In one embodiment, the nucleic acid sequence corresponding to SEQ ID NO: the amino acid at position 501(N) in 1 is Y. In some embodiments, the SARS-CoV-2S protein, immunogenic variant thereof, or immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof, e.g., as encoded by RNA as described herein, comprises one or more mutations, such as all mutations, of the SARS-CoV-2S protein of the SARS-CoV-2 variant selected from VOC-202012/01, 501.V2, cluster 5, and b.1.1.248. In some embodiments, a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof, e.g., encoded by an RNA described herein, comprises an amino acid sequence that is set forth in SEQ ID NO:1 with an alanine substitution at position 80, a glycine substitution at position 215, a lysine substitution at position 484, a tyrosine substitution at position 501, a valine substitution at position 701, a phenylalanine substitution at position 18, an isoleucine substitution at position 246, an asparagine substitution at position 417, a glycine substitution at position 614, a deletion at positions 242 and 244 and proline substitutions at positions 986 and 987.

In some embodiments, the methods and agents described herein, e.g., mRNA compositions, induce a cell-mediated immune response (e.g., CD4+ and/or CD8+ T cell response) following administration to a subject. In some embodiments, T cells are induced that recognize one or more epitopes (e.g., MHC class I restricted epitopes) selected from LPFNDGVYF, GVYFASTEK, YLQPRTFLL, QPTESIVRF, CVADYSVLY, KCYGVSPTK, NYNYLYRLF, FQPTNGVGY, IPFAMQMAY, RLQSLQTYV, GTHWFVTQR, VYDPLQPEL, QYIKWPWYI and KWPWYIWLGF. In one embodiment, T cells recognizing epitope YLQPRTFLL are induced. In one embodiment, T cells recognizing epitope NYNYLYRLF are induced. In one embodiment, T cells recognizing epitope QYIKWPWYI are induced. In one embodiment, T cells recognizing epitope KCYGVSPTK are induced. In one embodiment, T cells recognizing epitope RLQSLQTYV are induced. In some embodiments, the methods and agents described herein, e.g., mRNA compositions, are administered according to a protocol that achieves such induction of T cells.

In some embodiments, following administration to a subject, the methods and agents described herein, e.g., mRNA compositions, induce a cell-mediated immune response (e.g., CD4+ and/or CD8+ T cell response) that is detectable 15 weeks or later, 16 weeks or later, 17 weeks or later, 18 weeks or later, 19 weeks or later, 20 weeks or later, 21 weeks or later, 22 weeks or later, 23 weeks or later, 24 weeks or later, or 25 weeks or later after administration, e.g., using two doses of RNA described herein (wherein the second dose may be administered about 21 days after administration of the first dose). In some embodiments, the methods and agents described herein, e.g., mRNA compositions, are administered according to a regimen to achieve such induction of a cell-mediated immune response.

In one embodiment, the herein described vaccination against coronavirus, e.g. using RNA as described herein, which may be administered in the amounts and schedules described herein, e.g. in two doses of 30 μ g/dose, e.g. 21 days apart, may be repeated after a certain period of time, e.g. once a reduced protection against coronavirus infection is observed, using the same or a different vaccine as used for the first vaccination. This particular period of time may be at least 6 months, 1 year, 2 years, etc. In one embodiment, the same RNA as used for the first vaccination is used for the second or further vaccination, but is administered at a lower dose or less frequently. For example, a first vaccination may comprise vaccination with a dose of about 30 μ g/dose, wherein in one embodiment at least two such doses are administered, e.g., the second dose may be administered about 21 days after administration of the first dose), and a second or further vaccination may comprise vaccination with a dose of less than about 30 μ g/dose, wherein in one embodiment only one such dose is administered. In one embodiment, a different RNA is used for the second or further vaccination than for the first vaccination, e.g., BNT162B2 is used for the first vaccination and BNT162B1 or BNT162B3 is used for the second or further vaccination.

In one embodiment, a vaccination regimen comprises a first vaccination with at least two doses of an RNA described herein, e.g., two doses of an RNA described herein (wherein the second dose may be administered about 21 days after administration of the first dose), and a second vaccination with a single or multiple doses, e.g., two doses, of an RNA described herein. In various embodiments, the second vaccination is administered after administration of the first vaccination, e.g., 3-24 months, 6-18 months, 6-12 months, or 5-7 months after the initial two-dose regimen. The amount of RNA used in each dose of the second vaccination may be the same or different from the amount of RNA used in each dose of the first vaccination. In one embodiment, the amount of RNA used in each dose of the second vaccination is equal to the amount of RNA used in each dose of the first vaccination. In one embodiment, the amount of RNA used in each dose of the second vaccination and the amount of RNA used in each dose of the first vaccination is about 30 μ g/dose. In one embodiment, the same RNA as used for the first vaccination is used for the second vaccination. In one embodiment, the RNA used for the first and second vaccinations is BNT162b 2. In one embodiment, a different RNA is used for the second vaccination than for the first vaccination. In one embodiment, the RNA for the first vaccination is BNT162b2, and the RNA for the second vaccination is RNA encoding the SARS-CoV-2S protein of a SARS-CoV-2 variant strain (e.g., the strain discussed herein). In one embodiment, the RNA for the first vaccination is BNT162b2, and the RNA for the second vaccination is RNA encoding the SARS-CoV-2S protein of a SARS-CoV-2 variant strain that is circulating or rapidly spreading at the time of the second vaccination. In one embodiment, the RNA for the first vaccination is BNT162b2 and the RNA for the second vaccination is RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof, a SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprising one or more mutations of a S protein variant as described herein, such as a SARS-CoV-2S protein variant, in particular a naturally occurring S protein variant. In one embodiment, the RNA used for the first vaccination is BNT162b2, and the RNA used for the second vaccination is RNA encoding the SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof, the SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises one or more mutations, such as all mutations, of the SARS-CoV-2S protein of the SARS-CoV-2 variant selected from the group consisting of VOC-202012/01, 501.V2, cluster 5 and B.1.1.248. In one embodiment, the RNA used for the first vaccination encodes a polypeptide comprising an amino acid sequence with proline residue substitutions at positions 986 and 987 of SEQ ID NO:1, while the RNA used for the second vaccination is an RNA encoding a polypeptide comprising an amino acid sequence with alanine substitution at position 80, glycine substitution at position 215, lysine substitution at position 484, tyrosine substitution at position 501, valine substitution at position 701, phenylalanine substitution at position 18, isoleucine substitution at position 246, asparagine substitution at position 417, glycine substitution at position 614, deletions at positions 242 and 244 and proline substitutions at positions 986 and 987 of SEQ ID NO: 1.

In one embodiment, the vaccination regimen comprises a first vaccination with two doses of RNA encoding a polypeptide comprising the sequence set forth in SEQ ID NO:1, amino acid sequence having a substitution of a proline residue at positions 986 and 987, administered about 21 days apart; and a second vaccination using a single or multiple doses of RNA encoding a polypeptide comprising an amino acid sequence with proline residue substitutions at positions 986 and 987 of SEQ ID NO:1, administered about 6-12 months after administration of the first vaccination, i.e., after the initial two dose regimen. In one embodiment, each RNA dose comprises 30 μ g RNA.

In one embodiment, the vaccination regimen comprises a first vaccination with two doses of RNA encoding a polypeptide comprising the sequence set forth in SEQ ID NO:1, amino acid sequence having a substitution of a proline residue at positions 986 and 987, administered about 21 days apart; and a second vaccination using a single or multiple doses of RNA encoding a polypeptide comprising the amino acid sequence set forth in SEQ ID NO:1 with an alanine substitution at position 80, a glycine substitution at position 215, a lysine substitution at position 484, a tyrosine substitution at position 501, a valine substitution at position 701, a phenylalanine substitution at position 18, an isoleucine substitution at position 246, an asparagine substitution at position 417, a glycine substitution at position 614, a deletion at positions 242 and 244 and proline substitutions at positions 986 and 987, is administered about 6-12 months after administration of the first vaccination, i.e. after the initial two dose regimen. In one embodiment, each RNA dose comprises 30 μ g RNA.

In one embodiment, the second vaccination results in an enhancement of the immune response.

In one embodiment, the RNA described herein is co-administered with other vaccines. In some embodiments, the RNA compositions described herein are co-administered with an influenza vaccine. In some embodiments, the RNA compositions provided herein and other injectable vaccines are administered at different times. In some embodiments, the RNA compositions provided herein are administered simultaneously with other injectable vaccines. In some such embodiments, the RNA composition provided herein and at least one further injectable vaccine are administered at different injection sites. In some embodiments, the RNA compositions provided herein are not mixed with any other vaccine in the same syringe. In some embodiments, the RNA compositions provided herein are not combined with other coronavirus vaccines as part of a vaccination against a coronavirus (e.g., SARS-CoV-2).

The term "disease" refers to an abnormal condition affecting the body of an individual. A disease is generally understood as a medical condition associated with a particular symptom or sign. The disease may be caused by factors originating from an external source, such as an infectious disease, or it may be caused by an internal dysfunction, such as an autoimmune disease. In humans, "disease" is generally used more broadly to refer to any condition that causes pain, dysfunction, distress, social problems or death in a subject suffering from the disease or similar problems in a person in contact with the subject. In this broader sense it sometimes includes injuries, disabilities, disorders, syndromes, infections, isolated symptoms, deviant behaviors, and atypical changes in structure and function, but in other contexts and for other purposes these may be considered distinguishable categories. Diseases often affect individuals not only physically, but also emotionally, as infections and suffering from many diseases can change a person's appearance and a person's personality.

As used herein, the term "treatment" or "therapeutic intervention" relates to the management and care of a subject for the purpose of combating a disease condition, such as a disease or disorder. The term is intended to encompass the full range of treatment of a given disease condition suffered by a subject, such as the administration of therapeutically effective compounds to alleviate symptoms or complications, delay the development of the disease, disorder, or condition, alleviate or relieve symptoms and complications, and/or cure or eliminate the disease, disorder, or condition and to prevent the disease condition, wherein prevention is understood to be the management and care of the individual for the purpose of combating the disease, disorder, or condition, and includes the administration of active compounds to prevent the occurrence of symptoms or complications.

The term "therapeutic treatment" relates to any treatment that improves the health status and/or extends (increases) the longevity of an individual. The treatment can eliminate the disease in the subject, arrest or slow the progression of the disease in the subject, inhibit or slow the progression of the disease in the subject, reduce the frequency or severity of symptoms in the subject, and/or reduce relapse in a subject currently suffering from or previously suffering from the disease.

The term "prophylactic treatment" or "prophylactic treatment" relates to any treatment intended to prevent the occurrence of a disease in an individual. The terms "prophylactic treatment" or "prophylactic treatment" are used interchangeably herein.

The terms "individual" and "subject" are used interchangeably herein. They refer to a human or another mammal (e.g., mouse, rat, rabbit, dog, cat, cow, pig, sheep, horse, or primate) that may be suffering from or susceptible to a disease or disorder, but may or may not have the disease or disorder. In many embodiments, the subject is a human. Unless otherwise indicated, the terms "individual" and "subject" do not denote a particular age, and thus encompass adults, the elderly, children, and newborns. In some embodiments, the term "subject" includes a human that is at least 50 years old, at least 55 years old, at least 60 years old, at least 65 years old, at least 70 years old, or older. In some embodiments, the term "subject" includes a human having an age of at least 65 years, such as 65-80 years, 65-75 years, or 65-70 years. In embodiments of the present disclosure, an "individual" or "subject" is a "patient".

The term "patient" means an individual or subject to be treated, particularly an individual or subject suffering from a disease.

In one embodiment of the present disclosure, it is an object to provide an immune response against a coronavirus, as well as to prevent or treat a coronavirus infection.

A pharmaceutical composition comprising an RNA encoding a peptide or protein comprising an epitope may be administered to a subject to elicit an immune response in the subject against an antigen comprising the epitope, which may be therapeutic or partially or fully protective. One skilled in the art will appreciate that one of the principles of immunotherapy and vaccination is based on the following: an immunoprotective response to a disease is generated by immunizing a subject with an antigen or epitope that is immunologically relevant to the disease to be treated. Thus, the pharmaceutical compositions described herein may be used to induce or enhance an immune response. The pharmaceutical compositions described herein are therefore useful for the prophylactic and/or therapeutic treatment of diseases in which antigens or epitopes are involved.

As used herein, "immune response" refers to the overall bodily response to an antigen or cell expressing an antigen, and refers to a cellular immune response and/or a humoral immune response. The immune system is divided into the more primitive innate immune system, and the acquired or adaptive immune system of vertebrates, each comprising humoral and cellular components.

"cell-mediated immunity", "cellular immune response" or similar terms are meant to include cellular responses directed against cells characterized by expression of an antigen, particularly cells characterized by presentation of an antigen with class I or class II MHC. Cellular responses involve immune effector cells, particularly cells known as T cells or T lymphocytes, which act as "helpers" or "killers". Helper T cells (also known as CD 4)+T cells) play a central role by modulating immune responses, killing cells (also known as cytotoxic T cells, cytolytic T cells, CD 8)+T cells or CTLs) kill diseased cells such as virus-infected cells, preventing the production of more diseased cells.

Immune effector cells include any cell that is responsive to a vaccine antigen. Such responsiveness includes activation, differentiation, proliferation, survival and/or indication of one or more immune effector functions. In particular, the cells comprise cells with lytic potential, in particular lymphoid cells, and preferably T cells, in particular cytotoxic lymphocytes, preferably selected from the group consisting of cytotoxic T cells, Natural Killer (NK) cells and lymphokine-activated killer (LAK) cells. Each of these cytotoxic lymphocytes triggers the destruction of target cells when activated. For example, cytotoxic T cells trigger the destruction of target cells by one or both of the following means. First, when activated, T cells release cytotoxic agents such as perforin, granzyme and granulysin. Perforin and granulysin create pores in the target cell, while granzyme enters the cell and triggers a caspase cascade in the cytoplasm, inducing apoptosis (programmed cell death) of the cell. Second, apoptosis can be induced by Fas-Fas ligand interaction between T cells and target cells.

In the context of the present invention, the term "effector function" includes any function mediated by a component of the immune system, which, for example, leads to neutralization of pathogenic substances such as viruses and/or kills diseased cells such as virus-infected cells. In one embodiment, the inventionThe effector function in the context of (a) is a T cell mediated effector function. Such functions are in helper T cells (CD 4)+T cells) including cytokine release and/or activation of CD8+Lymphocytes (CTLs) and/or B cells, and in the case of CTLs includes depleted cells, i.e., cells characterized by expression of an antigen, e.g., by apoptosis or perforin-mediated cell lysis, production of cytokines such as IFN- γ and TNF- α, and specific cytolytic killing of target cells expressing the antigen.

The term "immune effector cell" or "immunoreactive cell" in the context of the present invention relates to a cell which exerts effector functions during an immune reaction. In one embodiment, an "immune effector cell" is capable of binding an antigen, such as an antigen that is presented on a cell in the context of MHC or expressed on the surface of a cell and mediates an immune response. For example, immune effector cells include T cells (cytotoxic T cells, helper T cells, tumor infiltrating T cells), B cells, natural killer cells, neutrophils, macrophages, and dendritic cells. Preferably, in the context of the present invention, an "immune effector cell" is a T cell, preferably CD4 +And/or CD8+T cells, most preferably CD8+T cells. According to the present invention, the term "immune effector cell" also includes cells that can mature into immune cells (e.g., T cells, particularly T helper cells, or cytolytic T cells) with a suitable stimulus. The immune effector cell comprises CD34+Hematopoietic stem cells, immature and mature T cells, and immature and mature B cells. Differentiation of T cell precursors into cytolytic T cells upon exposure to antigen is similar to clonal selection by the immune system.

"lymphoid cells" are cells capable of producing an immune response, such as a cellular immune response, or precursors of such cells, and include lymphocytes, preferably T lymphocytes, prolymphoblasts (lymphoblasts), and plasma cells. The lymphoblastoid cells may be immune effector cells as described herein. Preferred lymphoid cells are T cells.

The terms "T cell" and "T lymphocyte" are used interchangeably herein and include T helper cells (CD4+ T cells) and cytotoxic T cells (CTL, CD8+ T cells), which comprise cytolytic T cells. The term "antigen-specific T cell" or similar terms relate to a T cell that recognizes an antigen targeted by the T cell and preferably functions as an effector of the T cell.

T cells belong to a group of white blood cells called lymphocytes and play a central role in cell-mediated immunity. They can be distinguished from other lymphocyte types such as B cells and natural killer cells by the presence on their cell surface of a special receptor called T Cell Receptor (TCR). The thymus is the major organ responsible for T cell maturation. Several different subsets of T cells have been found, each with different functions.

T helper cells assist other leukocytes in the immune process, including B cells maturation into plasma cells and activation of cytotoxic T cells and macrophages, among others. These cells are also called CD4+ T cells because they express CD4 glycoprotein on the surface. Helper T cells are activated when presented with peptide antigens by MHC class II molecules expressed on the surface of Antigen Presenting Cells (APCs). Once activated, they rapidly divide and secrete small proteins called cytokines that regulate or assist in the active immune response.

Cytotoxic T cells destroy virus-infected cells and tumor cells, and are also involved in transplant rejection. These cells are also called CD8+ T cells because they express CD8 glycoprotein on the surface. These cells recognize their target by binding to MHC class I-associated antigens, which are present on the surface of almost every cell of the body.

Most T cells have a T Cell Receptor (TCR) that exists as a complex of several proteins. The TCR of a T cell is capable of interacting with an immunogenic peptide (epitope) that binds to a Major Histocompatibility Complex (MHC) molecule and is presented on the surface of a target cell. Specific binding of the TCR triggers a signaling cascade within the T cell, leading to proliferation and differentiation into mature effector T cells. The actual T cell receptor comprises two distinct peptide chains, which are generated from independent T cell receptor alpha and beta (TCR alpha and TCR beta) genes and are referred to as alpha-and beta-TCR chains. γ δ T cells (γ δ T cells) represent a small fraction of T cells with different T Cell Receptors (TCRs) on their surface. However, in γ δ T cells, the TCR consists of one γ -chain and one δ -chain. This group of T cells is less common (2% of total T cells) than α β T cells.

"humoral immunity" or "humoral immune response" is an aspect of immunity that is mediated by large molecules found in extracellular fluids, such as secreted antibodies, complement proteins, and certain antimicrobial peptides. This is in contrast to cell-mediated immunity. The aspect that it involves antibodies is often referred to as antibody-mediated immunity.

Humoral immunity refers to the production of antibodies and their attendant ancillary processes, including: th2 activation and cytokine production, germinal center formation and isotype switching, affinity maturation and memory cell production. It also refers to effector functions of the antibody, including pathogen neutralization, classical complement activation, and opsonization promotion of phagocytosis and pathogen elimination.

In a humoral immune response, B cells first mature in the bone marrow and acquire the B-cell receptor (BCR), which is abundantly displayed on the cell surface. These membrane-bound protein complexes have antibodies specific for antigen detection. Each B cell has a unique antibody that binds to the antigen. Mature B cells migrate from the bone marrow to lymph nodes or other lymphoid organs where they begin to encounter pathogens. When a B cell encounters an antigen, the antigen binds to the receptor and is taken up into the B cell by endocytosis. Antigens are processed by MHC-II proteins and re-presented on the surface of B cells. B cells wait for helper T cells (TH) to bind to the complex. This binding activates TH cells, which then release cytokines that induce B cells to divide rapidly, producing thousands of identical B cell clones. These daughter cells become plasma cells or memory cells. Memory B cells remain inactive here; later when these memory B cells encounter the same antigen due to reinfection, they divide and form plasma cells. In another aspect, the plasma cells produce large amounts of antibodies that are freely released into the circulatory system. These antibodies encounter antigens and bind to them. This can interfere with the chemical interaction between the host and foreign cells, or they can form bridges between their antigenic sites, preventing their normal function, or their presence can attract macrophages or killer cells to attack and phagocytose them.

The term "antibody" includes immunoglobulins comprising at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds. Each heavy chain comprises a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. Each light chain comprises a light chain variable region (abbreviated herein as VL) and a light chain constant region. The VH and VL regions may be further subdivided into regions of high denaturation, called Complementarity Determining Regions (CDRs), interspersed with regions that are more conserved, called Framework Regions (FRs). Each VH and VL comprises 3 CDRs and 4 FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR 4. The variable regions of the heavy and light chains comprise binding domains that interact with an antigen. The constant region of an antibody can mediate the binding of an immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component of the classical complement system (Clq). The antibody binds, preferably specifically binds, to the antigen.

Antibodies expressed by B cells are sometimes referred to as BCRs (B cell receptors) or antigen receptors. The 5 members included in this class of proteins are IgA, IgG, IgM, IgD and IgE. IgA is the predominant antibody present in body secretions such as saliva, tears, milk, gastrointestinal secretions and mucous secretions of the respiratory and genitourinary tracts. IgG is the most common circulating antibody. In most subjects, IgM is the primary immunoglobulin produced in the primary immune response. It is the most effective immunoglobulin in agglutination, complement fixation and other antibody responses, and is important in defense against bacteria and viruses. IgD is an immunoglobulin without known antibody function, but can act as an antigen receptor. IgE is an immunoglobulin that mediates immediate hypersensitivity by causing mast cells and basophils to release mediators upon exposure to a reactogen.

As used herein, "antibody heavy chain" refers to the larger of the two types of polypeptide chains present in an antibody molecule in their naturally occurring conformation.

As used herein, "antibody light chain" refers to the smaller of the two types of polypeptide chains present in an antibody molecule in their naturally occurring conformation, with kappa and lambda light chains referring to the two major antibody light chain isotypes.

The present disclosure contemplates immune responses that may be protective, prophylactic, and/or therapeutic. As used herein, "inducing an immune response" may mean that there is no immune response to a particular antigen prior to induction, or it may mean that there is a basal level of immune response to a particular antigen prior to induction, which is enhanced after induction. Thus, "inducing an immune response" includes "enhancing an immune response".

The term "immunotherapy" relates to the treatment of a disease or disease condition by inducing or enhancing an immune response. The term "immunotherapy" includes antigen immunization or antigen vaccination.

The term "immunization" or "vaccination" describes the process of administering an antigen to an individual, for the purpose of inducing an immune response, e.g. for therapeutic or prophylactic reasons.

The term "macrophage" refers to a subset of phagocytic cells produced by differentiation of monocytes. Macrophages activated by inflammatory, immune cytokines or microbial products non-specifically phagocytose and kill foreign pathogens within the macrophages through hydrolytic and oxidative attack, resulting in pathogen degradation. Peptides from the degraded proteins are displayed on the macrophage surface where they can be recognized by T cells and they can interact directly with antibodies on the B cell surface, resulting in T and B cell activation and further stimulation of the immune response. Macrophages belong to the class of antigen presenting cells. In one embodiment, the macrophage is a spleen macrophage.

The term "dendritic cell" (DC) refers to another subset of phagocytic cells that belongs to the class of antigen presenting cells. In one embodiment, the dendritic cells are derived from hematopoietic bone marrow progenitor cells. These progenitor cells are initially transformed into immature dendritic cells. These immature cells are characterized by high phagocytic activity and low T cell activation potential. Immature dendritic cells are constantly sampling the surrounding environment for pathogens such as viruses and bacteria. Once they are contacted with presentable antigens, they activate to mature dendritic cells and begin to migrate to the spleen or lymph nodes. Immature dendritic cells phagocytose pathogens and degrade their proteins into small pieces, and upon maturation present those fragments on their cell surface using MHC molecules. At the same time, they upregulate cell surface receptors such as CD80, CD86, and CD40 that act as co-receptors in T cell activation, greatly enhancing their ability to activate T cells. They also upregulate the chemotactic receptor CCR7, which induces dendritic cells to reach the spleen through the bloodstream or lymph nodes through the lymphatic system. Here they act as antigen presenting cells and activate helper and killer T cells as well as B cells by presenting antigen as well as non-antigen specific costimulatory signals. Thus, dendritic cells can actively induce a T cell or B cell-associated immune response. In one embodiment, the dendritic cells are splenic dendritic cells.

The term "antigen presenting cell" (APC) is a cell of a variety of cells that is capable of displaying, obtaining and/or presenting at least one antigen or antigen fragment on (or on) its cell surface. Antigen presenting cells can be distinguished between professional and non-professional antigen presenting cells.

The term "professional antigen presenting cell" relates to an antigen presenting cell that constitutively expresses the major histocompatibility complex class II (MHC class II) molecule required for interaction with naive T cells. If T cells interact with MHC class II molecule complexes on the membrane of antigen presenting cells, the antigen presenting cells produce co-stimulatory molecules that induce T cell activation. Professional antigen presenting cells include dendritic cells and macrophages.

The term "non-professional antigen presenting cell" relates to an antigen presenting cell that does not constitutively express MHC class II molecules, but expresses MHC class II molecules when stimulated by certain cytokines such as interferon-gamma. Exemplary non-professional antigen presenting cells include fibroblasts, thymic epithelial cells, thyroid epithelial cells, glial cells, pancreatic beta cells, or vascular endothelial cells.

"antigen processing" refers to the degradation of an antigen into a processed product that is a fragment of the antigen (e.g., the degradation of a protein into a peptide), and one or more of these fragments are associated with (e.g., by binding to) an MHC molecule for presentation by a cell, such as an antigen presenting cell of a particular T cell.

The term "disease involving an antigen" refers to any disease in which an antigen is involved, for example a disease characterized by the presence of an antigen. The disease in which the antigen is involved may be an infectious disease. As mentioned above, the antigen may be a disease-associated antigen, such as a viral antigen. In one embodiment, the disease in which an antigen is involved is a disease in which cells expressing the antigen are involved, preferably expressing the antigen on the cell surface.

The term "infectious disease" refers to any disease that can be transmitted from an individual to an individual or from an organism to an organism, and is caused by microbial matter (e.g., the common cold). Infectious diseases are known in the art and include, for example, viral diseases, bacterial diseases, or parasitic diseases, which are caused by viruses, bacteria, and parasites, respectively. In this regard, the infectious disease may be, for example, hepatitis, sexually transmitted diseases (e.g., chlamydia or gonorrhea), tuberculosis, HIV/acquired immunodeficiency syndrome (AIDS), diphtheria, hepatitis b, hepatitis c, cholera, Severe Acute Respiratory Syndrome (SARS), avian influenza, and influenza.

Certain exemplary embodiments:

1. a method of immunizing against SARS-CoV-2, the method comprising the steps of:

administering a composition comprising lipid nanoparticle encapsulated mRNA encoding at least one epitope of a SARS-CoV-2 encoded polypeptide according to a protocol established to achieve a detectable antibody titer in serum against the epitope within 7 days, the protocol comprising administering at least one dose of the composition.

2. The method of embodiment 1, wherein said regimen comprises administering at least two doses of said composition.

3. The method of embodiment 1, wherein said regimen consists of administering at least two doses of said composition.

4. The method of embodiment 2 or embodiment 3, wherein the first dose is an amount that is different from one or more subsequent doses.

5. The method of embodiment 1 or embodiment 4, wherein the first dose is administered a period of time prior to a subsequent dose, the period of time being at least 1 week, 1 month, 2 months, 3 months, 6 months, 1 year, 2 years, 3 years, or longer.

6. The method of any one of embodiments 1-6, wherein said regimen is established to have an incidence of adverse events of 60% or less when administered to a population of related adults.

7. The method of embodiment 6 wherein the regimen is established so as not to elicit a local injection site reaction of greater than moderate severity with an incidence of greater than about one-half (1in 75).

8. The method of any of embodiments 1-7, wherein each dose is no more than 60 μ g or less, including, for example, no more than 50 μ g, no more than 40 μ g, no more than 30 μ g, no more than 20 μ g, no more than 10 μ g, no more than 5 μ g, no more than 2.5 μ g, no more than 1 μ g.

9. The method of any one of embodiments 1-8, wherein each dose is at least 1 μ g or more, including, e.g., at least 2 μ g, at least 5 μ g, at least 10 μ g, at least 20 μ g, at least 30 μ g, at least 40 μ g, or more.

10. A method comprising administering to a subject a composition comprising a lipid nanoparticle-encapsulated mRNA, wherein the mRNA encodes an amino acid sequence comprising a SARS-COV-2S protein or fragment thereof, wherein the composition is administered in an effective amount to induce a SARS-COV-2S-protein specific immune response in the subject, wherein the effective amount is sufficient to provide an eliminating (sterizing) immunity in the subject at a dose that is at least 2-fold lower (including, e.g., at least 3-fold, at least 4-fold, at least 5-fold) relative to a reference composition (e.g., a reference RNA vaccine or composition).

11. A method comprising administering to a subject a composition comprising a lipid nanoparticle-encapsulated mRNA, wherein the mRNA encodes an amino acid sequence comprising a SARS-COV-2S protein or fragment thereof, wherein the composition is administered in an effective amount to reduce viral load in the subject by at least 80% relative to a control for 2 or more days (including, e.g., 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, or more) after exposure to SARS-COV-2, wherein the control is the viral load in a subject administered a reference composition (e.g., a reference RNA vaccine or composition).

12. A method comprising administering to a subject a composition comprising lipid nanoparticle encapsulated mRNA, wherein the mRNA encodes an amino acid sequence comprising a SARS-COV-2S protein or fragment thereof, wherein the composition is administered in an effective amount to induce a SARS-COV-2S-protein specific immune response in the subject, wherein the efficacy of the RNA vaccine is at least 80% relative to an unvaccinated control subject.

13. The method of any of embodiments 10-12, wherein the effective amount is sufficient to produce detectable levels of SARS-COV-2S protein or a fragment thereof, as measured in the serum of the subject 1-72 hours after administration.

14. The method of any of embodiments 10-12, wherein the effective amount is sufficient to produce a 1,000-10,000 neutralizing titer by neutralizing antibodies against the SARS-COV-2S protein, as measured in the serum of the subject 1-72 hours after administration.

15. The method of any of embodiments 10-14, wherein the titer of anti-SARS-COV-2S protein antibodies produced in the subject is increased by at least 1log relative to a control, wherein the control is the titer of anti-SARS-COV-2S protein antibodies produced in a subject that has not been administered a vaccine against SARS-COV-2.

16. The method of any of embodiments 10-15, wherein the titer of anti-SARS-COV-2S protein antibodies produced in the subject is increased at least 2-fold relative to a control, wherein the control is the titer of anti-SARS-COV-2S protein antibodies produced in a subject that has not been administered a vaccine against SARS-COV-2.

17. The method of any one of embodiments 1-16, wherein said administering is by intramuscular injection.

18. An immunogenic composition comprising a lipid nanoparticle encapsulated RNA (e.g., mRNA) encoding at least one epitope of a SARS-CoV-2 encoded polypeptide, the vaccine composition established to achieve a detectable antibody titer in serum against the epitope within 7 days after administration to a population of adult human subjects according to a regimen comprising administering at least one dose of the vaccine composition.

19. The immunogenic composition of embodiment 18, wherein at least 80% of the uridine in said RNA has a chemical modification.

20. The immunogenic composition of embodiment 18 or 19, wherein 100% of the uridine in said RNA has a chemical modification.

21. The immunogenic composition of any one of embodiments 18-20, wherein the 5' end cap is 7mG (5') ppp (5') NlmpNp.

22. The immunogenic composition of any one of embodiments 18-21, wherein the lipid nanoparticles in the composition comprise a cationic lipid, a PEG-modified lipid, a sterol, and a non-cationic lipid.

23. The immunogenic composition of any one of embodiments 18-22, wherein the lipid nanoparticles in the composition comprise a molar ratio of about 20-60% cationic lipid, 0.5-15% PEG-modified lipid, 25-55% sterol, and 5-25% non-cationic lipid.

24. The immunogenic composition of embodiment 22 or 23, wherein the cationic lipid is an ionizable cationic lipid, the non-cationic lipid is a neutral lipid, and the sterol is cholesterol.

25. The immunogenic composition of any one of embodiments 22 or 23 wherein the cationic lipid is selected from the group consisting of 2, 2-dioleyl-4-dimethylaminoethyl- [1,3] -dioxolane (2, 2-dilinoleyl-4-methyleneethyl- [1,3] -dioxolane, DLin-KC2-DMA), dioleyl-methyl-4-dimethylaminobutyrate ester (DLin-MC 3-DMA) and bis ((Z) -non-2-en-1-yl)9- ((4- (dimethylamino) butanoyl) oxy) heptadecanedioate (di ((Z) -non-2-en-1-yl)9- ((4- (dimethylamino) butanoyl) oxy) heptadecenoate (4- (dimethyllamino) butanoyl) oxy) heptadecenoate).

26. The immunogenic composition of any one of embodiments 18-25, wherein the RNA comprises a 5' end cap and a chemical modification, and the RNA is or is to be formulated as a lipid nanoparticle.

27. The immunogenic composition of any of embodiments 18-26, wherein the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof is linked to a signal peptide.

28. The immunogenic composition of embodiment 27, wherein the signal peptide is selected from the group consisting of HuIgGk signal peptide (METPAQLLFLLLLWLPDTTG); IgE heavy chain epsilon (epsilon) -1 signal peptide (MDWTWILFLVAAATRVHS); japanese encephalitis PRM signal sequence (MLGSNSGQRVVFTILLLLVAPAYS) and VSVg protein signal sequence (MKCLLYLAFLFIGVNCA).

29. A method of eliciting an immune response in a subject by activating T cells in vivo, the method comprising administering to the subject a composition comprising a lipid nanoparticle encapsulated modified nucleoside mRNA, wherein the mRNA encodes an amino acid sequence comprising a SARS-COV-2S protein or fragment thereof, thereby activating T cells in vivo against SARS-COV-2 infection in the subject.

30. A method of eliciting an immune response against SARS-COV-2 in a subject, the method comprising administering to the subject a composition comprising a lipid nanoparticle encapsulated modified nucleoside mRNA, wherein the mRNA encodes an amino acid sequence comprising a SARS-COV-2S protein or fragment thereof, wherein the composition causes increased production of SARS-COV-2 polypeptide or fragment thereof compared to a composition comprising a corresponding unmodified mRNA.

31. A method of eliciting an immune response against SARS-COV-2 in a subject, the method comprising administering to the subject a composition comprising a lipid nanoparticle encapsulated modified nucleoside mRNA, wherein the mRNA encodes an amino acid sequence comprising a SARS-COV-2S protein or fragment thereof, wherein the composition causes an increased antibody titer as compared to a composition comprising a corresponding unmodified mRNA.

32. A method of eliciting an immune response against SARS-COV-2 in a subject, the method comprising administering at least one dose of a composition comprising lipid nanoparticle encapsulated modified nucleoside mRNA, wherein the mRNA encodes an amino acid sequence comprising a SARS-COV-2S protein or fragment thereof, to the subject, wherein the composition causes an increased antibody titer in the subject at least 7 days after the first dose compared to the antibody titer against SARS-COV-2 prior to administration of the composition.

33. A method of eliciting an immune response against SARS-COV-2 in a subject, the method comprising administering at least one dose of a composition comprising lipid nanoparticle-encapsulated modified nucleoside mRNA, wherein the mRNA encodes an amino acid sequence comprising SARS-COV-2S protein or a fragment thereof, to the subject, wherein the composition causes an increased antibody titer in the subject at least 7 days after the first dose compared to a composition comprising the corresponding unmodified mRNA.

34. The method of any one of embodiments 29-33, wherein the mRNA is present in 90% or greater purity.

35. The method of any one of embodiments 29-34, wherein the composition does not further comprise unmodified RNA encoding a SARS-COV-2S protein or fragment thereof.

36. The method of any one of embodiments 29-35, wherein said composition is administered at least 2 times, the first and second administrations being separated by at least 7 days.

37. The method of any one of embodiments 29-36, wherein the subject is at risk of SARS-CoV-2 infection.

38. The method of any one of embodiments 29-37, wherein the subject is undergoing treatment for a cardiovascular disease.

39. The method of any one of embodiments 29-37, wherein the subject is undergoing treatment for diabetes.

40. The method of any one of embodiments 29-37, wherein the subject is undergoing treatment for chronic cardiopulmonary disease.

41. The method of any one of embodiments 29-37, wherein the subject is undergoing treatment for chronic kidney disease.

42. The method of any one of embodiments 29-41, wherein the immune response lasts at least about 30 days.

43. The method of any one of embodiments 29-42, wherein the immune response lasts at least about 60 days.

44. The method of any one of embodiments 29-43, wherein said immune response lasts at least about 180 days.

45. The method of any one of embodiments 29-44, wherein the immune response comprises a virus neutralization titer.

46. The method of any one of embodiments 29-45, wherein the subject is at least 18 years old.

47. The method of any one of embodiments 29-46, wherein the dose comprises 100 μ g or less of mRNA.

48. The method of any one of embodiments 29-47, wherein the dose comprises less than 100 μ g of mRNA and the composition elicits an immune response that is greater than the immune response elicited by a composition comprising at least 100 μ g of mRNA.

49. The method of any one of embodiments 29-48, wherein said dose comprises 30 μ g of mRNA.

50. The method of any of embodiments 29-49, wherein the immune response comprises an antibody directed against the receptor binding domain of the S protein of SARS-CoV-2.

51. The method of any one of embodiments 29-50, wherein said immune response comprises IgG that binds to RBD.

52. The method of any of embodiments 29-50, wherein the SARS-CoV-2S protein or fragment thereof comprises a receptor binding domain.

53. A kit comprising a) a composition comprising mRNA encapsulated by lipid nanoparticles; and b) a temperature monitoring system.

54. The kit of embodiment 53, wherein said temperature monitoring system comprises a temperature sensor and a display, wherein said temperature monitoring system displays or warns when the temperature of said composition reaches a temperature above about-80 ℃.

55. The kit of embodiment 53, wherein said temperature monitoring system comprises a temperature sensor and a display, wherein said temperature monitoring system displays or warns when the temperature of said composition reaches a temperature above about-60 ℃.

56. A kit comprising a) a composition comprising mRNA encapsulated by lipid nanoparticles; and b) a light sensor.

57. The kit of embodiment 56, wherein the light sensor comprises a photosensitive element configured to react to exposure to light resulting in a change in a material property of the photosensitive element.

58. The method of any one of embodiments 29-35, wherein said composition is administered at least 2 times, the first and second administrations being separated by at least 14 days.

59. The method of any one of embodiments 29-35, wherein the composition is administered at least 2 times, the first and second administrations being separated by at least 21 days.

60. The method of any one of embodiments 29-48, wherein said dose comprises 10 μ g of mRNA.

61. The method of any one of embodiments 29-35, wherein said composition is administered at least 2 times, the first and second administrations being separated by at least 28 days.

62. The method of any one of embodiments 29-35, wherein the mRNA encodes the amino acid sequence of SEQ ID No. 3, SEQ ID NO: 5 and SEQ ID NO. 7.

63. An immunogenic composition comprising an effective amount of a messenger ribonucleic acid (mRNA) polynucleotide comprising an open reading frame encoding a polypeptide comprising a receptor binding portion of SARs-CoV-2S protein to induce an immune response in a subject administered at least one dose of the immunogenic composition, the messenger ribonucleic acid (mRNA) polynucleotide formulated in at least one lipid nanoparticle comprising a cationic lipid, wherein the isolated mRNA polynucleotide is not a self-replicating RNA.

64. The immunogenic composition of embodiment 63, wherein the lipid nanoparticle further comprises any one of a non-cationic lipid, a sterol, and a PEG-modified lipid.

65. The immunogenic composition of embodiment 63 comprising an effective amount of an isolated messenger ribonucleic acid (mRNA) polynucleotide comprising an open reading frame encoding a polypeptide comprising a receptor binding portion of a SARs-CoV-2S protein to induce an immune response in a subject administered at least one dose of the immunogenic composition; the messenger ribonucleic acid (mRNA) polynucleotide is formulated in at least one lipid nanoparticle comprising a molar ratio of 20-60% ionizable cationic lipid, 5-25% non-cationic lipid, 25-55% sterol, and 0.5-15% PEG-modified lipid, wherein the isolated mRNA polynucleotide is not a self-replicating RNA.

66. The immunogenic composition of embodiment 63, wherein the polypeptide does not contain an intact S protein.

67. The immunogenic composition of embodiment 63, wherein the polypeptide comprises the Receptor Binding Domain (RBD) of the SARs-CoV-2S protein.

68. The immunogenic composition of embodiment 63, wherein the polypeptide comprises SEQ ID NO 5.

69. The immunogenic composition of embodiment 63, wherein the polypeptide comprises SEQ ID NO:29 or 31.

70. The immunogenic composition of embodiment 63, wherein the polypeptide comprises SEQ ID NO 3.

71. The immunogenic composition of embodiment 63, wherein the polypeptide comprises SEQ ID NO 7.

72. The immunogenic composition of any one of embodiments 63-71, wherein the isolated mRNA polynucleotide further comprises a 5' end cap, 7mG (5 ') ppp (5 ') NlmpNp.

73. The immunogenic composition of any one of embodiments 63-72, wherein at least 80% of the uracils in the open reading frame have a chemical modification selected from the group consisting of N1-methyl-pseudouridine or N1-ethyl-pseudouridine.

74. The immunogenic composition of any of embodiments 63-73, wherein the chemical modification is at the 5-position of uracil.

75. The immunogenic composition of any one of embodiments 63-74, wherein the potency of the immunogenic composition in a vaccinated subject is at least 60% after a single dose of the immunogenic composition relative to an unvaccinated subject.

76. The immunogenic composition of embodiment 75, wherein the potency of the immunogenic composition in a vaccinated subject is at least 70% after a single dose of the immunogenic composition relative to an unvaccinated subject.

77. The immunogenic composition of embodiment 75, wherein the potency of the immunogenic composition in a vaccinated subject is at least 80% after a single dose of the immunogenic composition relative to an unvaccinated subject.

78. The immunogenic composition of embodiment 75, wherein the potency of the immunogenic composition in a vaccinated subject is at least 90% after a single dose of the immunogenic composition relative to an unvaccinated subject.

79. The immunogenic composition of any one of embodiments 63-78, wherein the effective amount is sufficient to produce detectable levels of a polypeptide comprising a receptor binding portion of a SARS-CoV-2S protein as measured in the serum of a subject vaccinated with at least one dose of the immunogenic composition 1-72 hours after administration.

80. The immunogenic composition of any one of embodiments 63-79, wherein the effective amount is sufficient to produce a 1,000-10,000 neutralizing titer by neutralizing antibodies against an antigenic polypeptide comprising a receptor binding portion of a SARS-COV-2S protein, as measured in the serum of a subject vaccinated with at least one dose of the immunogenic composition 1-72 hours after administration.

81. The immunogenic composition of embodiment 80, wherein the 1,000-10,000 neutralization titer is generated in the absence of antibody-dependent enhancement (ADE) of SARS-CoV-2 associated disease.

82. The immunogenic composition of any one of embodiments 63-81, wherein the effective amount does not induce an Enhanced Respiratory Disease (ERD) associated with the immunogenic composition.

83. The immunogenic composition of any of embodiments 63-82, wherein the effective amount reduces the amount of SARS-CoV-2 viral RNA in the lung of the subject after infection with SARS-CoV-2 virus as compared to the amount of SARS-CoV-2 viral RNA in the lung of an unvaccinated subject after infection with SARS-CoV-2 virus.

84. The immunogenic composition of any of embodiments 63-82, wherein the effective amount reduces the amount of SARS-CoV-2 viral RNA in the lung of the subject at least 3 days after infection with SARS-CoV-2 virus as compared to the amount of SARS-CoV-2 viral RNA in the lung of the subject 3 days after infection with SARS-CoV-2 virus.

85. The immunogenic composition of any of embodiments 63-82, wherein the effective amount reduces the amount of SARS-CoV-2 viral RNA in a nasal swab sample of the subject after infection with SARS-CoV-2 virus as compared to the amount of SARS-CoV-2 viral RNA in a nasal swab sample of a subject not vaccinated after infection with SARS-CoV-2 virus.

86. The immunogenic composition of any of embodiments 63-82, wherein the effective amount does not increase the amount of SARS-CoV-2 viral RNA in a nasal swab sample of the subject 3 days after infection with SARS-CoV-2 virus as compared to the amount of SARS-CoV-2 viral RNA in a nasal swab sample of the subject 1 day after infection with SARS-CoV-2 virus.

87. The immunogenic composition of any of embodiments 63-87, wherein the anti-SARS-CoV-2 antibody titer produced in a subject vaccinated with at least one dose of the immunogenic composition increases by at least 1log relative to a control, wherein the control is the anti-SARS-CoV-2 antibody titer produced in a subject not administered the immunogenic composition against SARS-CoV-2.

88. The immunogenic composition of any of embodiments 63-87, wherein the anti-SARS-CoV-2 antibody titer produced in a subject vaccinated with at least one dose of the immunogenic composition increases by 2-fold relative to a control, wherein the control is the anti-SARS-CoV-2 antibody titer produced in a subject not administered the immunogenic composition against SARS-CoV-2.

89. The immunogenic composition of any one of embodiments 63-88, wherein the effective amount is a total dose of 2 μ g to 100 μ g.

90. The immunogenic composition of embodiment 89, wherein the effective amount is a total dose of 100 μ g.

91. The immunogenic composition of embodiment 89, wherein the effective amount is a total dose of 20 μ g to 50 μ g.

92. The immunogenic composition of embodiment 89, wherein the effective amount is a total dose of 10 μ g to 30 μ g.

93. The immunogenic composition of embodiment 89, wherein the effective amount is a total dose of 10 μ g.

94. The immunogenic composition of embodiment 89, wherein the effective amount is a total dose of 20 μ g.

95. The immunogenic composition of embodiment 89, wherein the effective amount is a total dose of 30 μ g.

96. The immunogenic composition of any one of embodiments 63-95, wherein the composition is formulated in a single dose vial.

97. The immunogenic composition of any one of embodiments 63-95, wherein the composition is formulated in a multi-dose vial.

98. The immunogenic composition of any one of embodiments 63-97, wherein intramuscular administration of an effective amount of the immunogenic composition to a subject induces a neutralizing antibody titer in the subject.

99. The immunogenic composition of embodiment 98, wherein the neutralizing antibody titer is sufficient to reduce viral infection of B cells by at least 50% relative to the neutralizing antibody titer of an unvaccinated control subject or relative to the neutralizing antibody titer of a subject vaccinated with a live attenuated virus vaccine, an inactivated virus vaccine, or a protein subunit virus vaccine.

100. The immunogenic composition of embodiment 98 or 99, wherein the neutralizing antibody titer is induced in a subject after 3 or fewer doses of the immunogenic composition.

101. The immunogenic composition of any one of embodiments 98-100, wherein the neutralizing antibody titer and/or T cell immune response is sufficient to reduce the asymptomatic viral infection rate relative to the neutralizing antibody titer of an unvaccinated control subject.

102. The immunogenic composition of any one of embodiments 98-101, wherein the neutralizing antibody titer and/or T cell immune response is sufficient to prevent viral latency in the subject.

103. The immunogenic composition of any one of embodiments 98-102, wherein the neutralizing antibody titer is sufficient to block viral fusion with epithelial cells and/or B cells of the subject.

104. The immunogenic composition of any one of embodiments 63-103, wherein intramuscular administration of an effective amount of the immunogenic composition to a subject induces a T cell immune response in the subject.

105. The immunogenic composition of embodiment 104, wherein the T cell immune response comprises CD4+T cell immune response and/or CD8+T cell immune response.

106. The immunogenic composition of any one of embodiments 63-105, wherein the encoded polypeptide is presented on the cell surface of a subject.

107. A method comprising administering to a subject the immunogenic composition of any one of embodiments 63-106, wherein the immunogenic composition is administered to the subject in an effective amount to induce an immune response in the subject.

108. The method of embodiment 107, wherein an immune response is induced against a SARs-CoV-2 virus having a mutation in RBD compared to SEQ ID NO: 5.

109. The method of embodiment 107, wherein an immune response is induced against a SARs-CoV-2 virus having a mutation in the spike protein compared to SEQ ID NO: 1.

110. The method of embodiment 108 or 109, wherein the immune response is induced against a SARs-CoV-2 virus having any one of the following mutations in the RBD compared to SEQ ID NO: 1: Q321L, V341I, a348T, N354D, S359N, V367F, K378R, R408I, Q409E, a435S, N439K, K458R, I472V, G476S, S477N, V483A, Y508H and H519P.

111. The method of embodiment 109, wherein the immune response is induced against a SARs-CoV-2 virus having a D614G mutation in the spike protein compared to SEQ ID NO: 1.

112. The method of embodiment 107, wherein the immunogenic composition is administered to the subject annually.

113. An RNA, optionally complexed by a (poly) cationic polymer, polyplex, protein or peptide, said RNA: (a) comprising an open reading frame encoding a polypeptide comprising a receptor binding portion of a SARS-CoV-2S protein; and (b) is suitable for intracellular expression of the polypeptide.

114. The RNA of embodiment 113, wherein the polypeptide does not comprise an intact S protein.

115. The RNA of embodiment 113 or 114, wherein the RNA further comprises a 5' end cap, 7mG (5 ') ppp (5 ') NlmpNp.

116. The RNA of any one of embodiments 113 and 115, wherein at least 80% of the uracils in the open reading frame have a chemical modification selected from the group consisting of N1-methyl-pseudouridine and N1-ethyl-pseudouridine.

117. The RNA of any one of embodiments 113 and 116, wherein the chemical modification is at the 5-position of uracil.

118. The RNA of any one of embodiments 113 and 117 for use in inducing an immune response in a human or for vaccination of a human.

119. The RNA for embodiment 118, wherein the human comprises a human known to be exposed to SARS-CoV-2.

120. The RNA for embodiment 118, wherein the human comprises a human known to be infected with SARS-CoV-2.

121. The RNA for embodiment 118, wherein the human comprises a human that is not known to be exposed to SARS-CoV-2.

122. Use of the RNA of any one of embodiments 113 and 117 for vaccination of humans.

123. The use of embodiment 122, wherein said human comprises a human known to be exposed to SARS-CoV-2.

124. The use of embodiment 122, wherein said human comprises a human known to be infected with SARS-CoV-2.

125. The use of embodiment 122, wherein said human comprises a human not known to be exposed to SARS-CoV-2.

126. A single dose formulation comprising the immunogenic composition of any one of embodiments 63-106.

127. A multi-dose formulation comprising the immunogenic composition of any one of embodiments 63-106 in one vial.

128. The formulation of embodiment 126, comprising at least 2 doses per vial.

129. The formulation of embodiment 126, comprising a total of 2-12 doses per vial.

130. The formulation of any one of embodiments 126-129, wherein the volume of each dose is equal.

131. The formulation of any one of embodiments 126-130, wherein each formulation comprises a total volume of 1-3mL in the vial.

132. The formulation of any one of embodiments 126-131, wherein the immunogenic composition is frozen.

133. A pre-filled (pre-filled) vaccine delivery device comprising the immunogenic composition of any one of embodiments 63-106.

Further certain exemplary embodiments:

1. a composition or pharmaceutical product comprising an RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of the SARS-CoV-2S protein or immunogenic variant thereof.

2. The composition or pharmaceutical product of embodiment 1, wherein the immunogenic fragment of SARS-CoV-2S protein comprises the S1 subunit of SARS-CoV-2S protein or the Receptor Binding Domain (RBD) of the S1 subunit of SARS-CoV-2S protein.

3. The composition or pharmaceutical product of embodiments 1 or 2, wherein the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof is encoded by a coding sequence that is codon optimized and/or has an increased G/C content compared to the wild type coding sequence, wherein said codon optimization and/or increased G/C content preferably does not alter the sequence of the encoded amino acid sequence.

4. A composition or a pharmaceutical product according to any one of embodiments 1 to 3, wherein

(i) An RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises SEQ ID NO: 2. nucleotide 979-1584 of nucleotide 8 or 9, and SEQ ID NO: 2. nucleotide sequence of nucleotide 979-1584 of 8 or 9 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity, or SEQ ID NO: 2. 8 or 9, or a fragment of the nucleotide sequence of nucleotide 979-1584 or a fragment corresponding to SEQ ID NO: 2. a fragment of nucleotide 979-1584 of nucleotide 8 or 9 having a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical; and/or

(ii) The SARS-CoV-2S protein, the immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or the immunogenic variant thereof comprises the amino acid sequence of amino acid 327-528 of SEQ ID NO:1 in combination with the sequence of SEQ ID NO:1 amino acid 327-528 has an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the amino acid sequence of SEQ ID NO 1, or an immunogenic fragment of the amino acid sequence of amino acid 327-528 of SEQ ID NO:1 amino acid 327-528 has an immunogenic fragment of an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical.

5. A composition or a pharmaceutical product according to any one of embodiments 1 to 4, wherein

(i) An RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises a nucleotide sequence of nucleotides 49-2055 of SEQ ID No. 2, 8 or 9, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 49-2055 of SEQ ID No. 2, 8 or 9, or a nucleotide sequence of SEQ ID NO: 2. 8 or 9 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 49-2055 of SEQ ID No. 2, 8 or 9; and/or

(ii) A SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprising the amino acid sequence of amino acids 17-685 of SEQ ID No. 1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-685 of SEQ ID No. 1, or an immunogenic fragment of SEQ ID NO:1 or an immunogenic fragment of an amino acid sequence of amino acids 17 to 685 of SEQ ID NO:1, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the amino acid sequence of amino acids 17-685.

6. A composition or a pharmaceutical product according to any one of embodiments 1 to 5, wherein

(i) An RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises SEQ ID NO: 2. 8 or 9, nucleotide 49-3819, and SEQ ID NO: 2. 8 or 9, nucleotides 49 to 3819, or a fragment of a nucleotide sequence of SEQ ID No. 2, 8 or 9, or a nucleotide sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 49 to 3819 of SEQ ID No. 2, 8 or 9, or a fragment of a nucleotide sequence of at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 49 to 3819 of SEQ ID No. 2, 8 or 9; and/or

(ii) A SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprising the amino acid sequence of amino acids 17-1273 of SEQ ID No. 1 or 7, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273 of SEQ ID No. 1 or 7, or an immunogenic fragment of the amino acid sequence of amino acids 17-1273 of SEQ ID No. 1 or 7 or an immunogenic fragment of the amino acid sequence of SEQ ID No. 1 or 7 or a variant thereof which is identical to SEQ ID NO:1 or 7, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273.

7. The composition or pharmaceutical product of any one of embodiments 1-6, wherein the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a secretory signal peptide.

8. The composition or pharmaceutical product of embodiment 7, wherein said secretory signal peptide is fused, preferably by N-terminal fusion, to a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof.

9. A composition or pharmaceutical product according to embodiment 7 or 8, wherein

(i) The RNA encoding the secretion signal peptide comprises SEQ ID NO: 2. 8 or 9, a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotides 1 to 48 of SEQ ID NO. 2, 8 or 9, or a fragment of the nucleotide sequence of nucleotides 1 to 48 of SEQ ID NO. 2, 8 or 9 or a fragment of the nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotides 1 to 48 of SEQ ID NO. 2, 8 or 9; and/or

(ii) The secretion signal peptide comprises an amino acid sequence of amino acids 1 to 16 of SEQ ID NO. 1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 16 of SEQ ID NO. 1, or a functional fragment of the amino acid sequence of amino acids 1 to 16 of SEQ ID NO. 1 or a functional fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1 to 16 of SEQ ID NO. 1.

10. A composition or a pharmaceutical product according to any one of embodiments 1 to 9, wherein

(i) An RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises SEQ ID NO:6, and SEQ ID NO:6, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the nucleotide sequence of SEQ ID NO:6 or a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 6; and/or

(ii) The SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises the amino acid sequence of SEQ ID NO. 5, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO. 5 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO. 5.

11. A composition or a pharmaceutical product according to any one of embodiments 1 to 10, wherein the RNA comprises a modified nucleoside instead of uridine.

12. The composition or pharmaceutical product of embodiment 11, wherein said modified nucleoside is selected from the group consisting of pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ) and 5-methyl-uridine (m 5U).

13. A composition or a pharmaceutical product according to any one of embodiments 1 to 12, wherein the RNA comprises a 5' cap.

14. A composition or a pharmaceutical product according to any one of embodiments 1 to 13, wherein

An RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a 5' UTR comprising the nucleotide sequence of SEQ ID No. 12, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the nucleotide sequence of SEQ ID No. 12.

15. The composition or pharmaceutical product of any one of embodiments 1 to 14, wherein the RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a 3' UTR comprising the nucleotide sequence of SEQ ID NO:13, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the nucleotide sequence of SEQ ID NO: 13.

16. The composition or pharmaceutical product of any one of embodiments 1-15, wherein the RNA encoding the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a poly-a sequence.

17. The composition or pharmaceutical product of embodiment 16, wherein said poly-a sequence comprises at least 100 nucleotides.

18. The composition or pharmaceutical product of embodiment 16 or 17, wherein said poly-a sequence comprises SEQ ID NO: 14, or a nucleotide sequence consisting of SEQ ID NO: 14, or a nucleotide sequence of seq id no.

19. The composition or pharmaceutical product of any one of embodiments 1 to 18, wherein said RNA is or is to be formulated as a liquid, a solid, or a combination thereof.

20. A composition or a pharmaceutical product according to any one of embodiments 1 to 19, wherein the RNA is or is to be formulated for injection.

21. A composition or a pharmaceutical product according to any one of embodiments 1 to 20, wherein the RNA is or is to be formulated for intramuscular administration.

22. A composition or a pharmaceutical product according to any one of embodiments 1 to 21, wherein the RNA is or is to be formulated as a particle.

23. A composition or a pharmaceutical product according to embodiment 22, wherein the particle is a Lipid Nanoparticle (LNP) or lipoplex (lpx) particle.

24. The composition or pharmaceutical product of embodiment 23 wherein said LNP particles comprise ((4-hydroxybutyl) azanediyl) bis (hexane-6, 1-diyl) bis (hexyl 2-decanoate), 2- [ (polyethylene glycol) -2000] -N, N-ditetradecylacetamide, 1, 2-distearoyl-sn-glycerol-3-phosphocholine, and cholesterol.

25. A composition or a pharmaceutical product according to embodiment 23, wherein the RNA lipoplex particles are obtainable by mixing RNA with liposomes.

26. A composition or a pharmaceutical product according to any one of embodiments 1 to 25, wherein the RNA is mRNA or saRNA.

27. A composition or a pharmaceutical product according to any one of embodiments 1 to 26, which is a pharmaceutical composition.

28. A composition or a pharmaceutical product according to any one of embodiments 1 to 27, which is a vaccine.

29. The composition or pharmaceutical product of embodiment 27 or 28, wherein said pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers, diluents and/or excipients.

30. A composition or a pharmaceutical product according to any one of embodiments 1 to 26, which is a kit.

31. The composition or pharmaceutical product of embodiment 30, wherein the RNA and optionally the particle-forming components are in separate vials.

32. The composition or pharmaceutical product of embodiment 30 or 31, further comprising instructions for using the composition or pharmaceutical product to induce an immune response against a coronavirus in a subject.

33. A composition or a pharmaceutical product according to any one of embodiments 1 to 32 for use in medicine.

34. The composition or pharmaceutical product of embodiment 33, wherein said pharmaceutical use comprises inducing an immune response against a coronavirus in a subject.

35. The composition or pharmaceutical product of embodiment 33 or 34, wherein said pharmaceutical use comprises the treatment or prophylactic treatment of a coronavirus infection.

36. A composition or a pharmaceutical product according to any one of embodiments 1 to 35 for administration to a human.

37. A composition or a pharmaceutical product according to any one of embodiments 32 to 36, wherein the coronavirus is a beta coronavirus.

38. A composition or a pharmaceutical product according to any one of embodiments 32 to 37 wherein the coronavirus is sabovirus (sarbecovirus).

39. A composition or a pharmaceutical product according to any one of embodiments 32 to 38, wherein the coronavirus is SARS-CoV-2.

40. A method of inducing an immune response against a coronavirus in a subject, the method comprising administering to the subject a composition comprising an RNA encoding an amino acid sequence comprising a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of the SARS-CoV-2S protein or immunogenic variant thereof.

41. The method of embodiment 40, wherein the immunogenic fragment of SARS-CoV-2S protein comprises the S1 subunit of SARS-CoV-2S protein or the Receptor Binding Domain (RBD) of the S1 subunit of SARS-CoV-2S protein.

42. The method of embodiment 40 or 41, wherein the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof is encoded by a coding sequence which is codon optimized and/or has an increased G/C content compared to the wild type coding sequence, wherein said codon optimization and/or increased G/C content preferably does not alter the sequence of the encoded amino acid sequence.

43. The method of any one of embodiments 40-42, wherein

(i) An RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises SEQ ID NO: 2. 8 or 9, nucleotide 979-1584, or a nucleotide sequence which is identical to SEQ ID NO: 2. 8 or 9 nucleotide 979-1584 has a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical to the nucleotide sequence of nucleotides 979-1584 of SEQ ID NO: 2, 8 or 9 or a fragment of the nucleotide sequence of nucleotides 979-1584 of SEQ ID NO: 2. a fragment of nucleotide 979-1584 of nucleotide 8 or 9 having a nucleotide sequence which is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical; and/or

(ii) The SARS-CoV-2S protein, the immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or the immunogenic variant thereof comprises the amino acid sequence of amino acid 327-528 of SEQ ID NO:1 in combination with the sequence of SEQ ID NO:1, or the amino acid sequence of amino acid 327-528 of SEQ ID NO:1 or an immunogenic fragment of the amino acid sequence of amino acid 327-528 of SEQ ID NO:1 amino acid 327-528 has an immunogenic fragment of an amino acid sequence that is at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identical.

44. The method of any one of embodiments 40-43, wherein

(i) An RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises a nucleotide sequence of nucleotides 49-2055 of SEQ ID No. 2, 8 or 9, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to a nucleotide sequence of nucleotides 49-2055 of SEQ ID No. 2, 8 or 9, or a nucleotide sequence of SEQ ID NO: 2. 8 or 9 or a fragment of a nucleotide sequence from nucleotides 49 to 2055 of SEQ ID NO: 2. 8 or 9, nucleotides 49-2055 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity; and/or

(ii) A SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof, comprising the amino acid sequence of SEQ ID NO: 1, amino acid sequence corresponding to amino acids 17-685 of SEQ ID NO: 1, or an amino acid sequence of amino acids 17 to 685 of SEQ ID NO: 1 or an immunogenic fragment of an amino acid sequence of amino acids 17 to 685 of SEQ ID NO: 1, having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the amino acid sequence of amino acids 17-685.

45. The method of any one of embodiments 40-44, wherein

(i) An RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises SEQ ID NO: 2. nucleotide 49 to 3819 of 8 or 9, and SEQ ID NO: 2. 8 or 9 from nucleotide 49 to 3819 having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity, or SEQ ID NO: 2. 8 or 9 or a fragment of a nucleotide sequence from nucleotides 49 to 3819 of SEQ ID NO: 2. 8 or 9, a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 49-3819; and/or

(ii) A SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprising the amino acid sequence of amino acids 17-1273 of SEQ ID NO:1 or 7, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273 of SEQ ID NO:1 or 7, or SEQ ID NO:1 or 7 or an immunogenic fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 17-1273 of SEQ ID No. 1 or 7.

46. The method of any of embodiments 40-45, wherein the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof comprises a secretory signal peptide.

47. The method of embodiment 46, wherein said secretory signal peptide is fused, preferably by N-terminal fusion, to a SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or immunogenic variant thereof.

48. The method of embodiment 46 or 47, wherein

(i) The RNA encoding the secretion signal peptide comprises SEQ ID NO: 2. 8 or 9, and a nucleotide sequence of nucleotides 1 to 48 of SEQ ID NO: 2. 8 or 9 has at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 1-48 of SEQ ID NO: 2. 8 or 9 or a fragment of a nucleotide sequence corresponding to nucleotides 1 to 48 of SEQ ID NO: 2. 8 or 9, a fragment of a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of nucleotides 1-48; and/or

(ii) The secretion signal peptide comprises an amino acid sequence of amino acids 1-16 of SEQ ID NO. 1, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-16 of SEQ ID NO. 1, or an amino acid sequence of SEQ ID NO:1 or a functional fragment of an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of amino acids 1-16 of SEQ ID No. 1.

49. The method of any one of embodiments 40-48, wherein

(i) An RNA encoding a SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises the nucleotide sequence of SEQ ID NO. 6, a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO. 6, or a fragment of the nucleotide sequence of SEQ ID NO. 6 or a fragment of the nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID NO. 6; and/or

(ii) The SARS-CoV-2S protein, an immunogenic variant thereof or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises the amino acid sequence of SEQ ID NO. 5, an amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO. 5 or an immunogenic fragment of the amino acid sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the amino acid sequence of SEQ ID NO. 5.

50. The method according to any one of embodiments 40-49, wherein said RNA comprises a modified nucleoside instead of uridine.

51. The method of embodiment 50, wherein said modified nucleoside is selected from the group consisting of pseudouridine (ψ), N1-methyl-pseudouridine (m1 ψ) and 5-methyl-uridine (m 5U).

52. The method of any one of embodiments 40-51, wherein said RNA comprises a cap.

53. The method of any of embodiments 40-52, wherein the RNA encoding an amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises a 5' UTR comprising the amino acid sequence of SEQ ID NO:12, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 12.

54. The method of any of embodiments 40-53, wherein the RNA encoding an amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises a 3' UTR comprising the amino acid sequence of SEQ ID NO:13, or a nucleotide sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85% or 80% identity to the nucleotide sequence of SEQ ID No. 13.

55. The method of any of embodiments 40-54, wherein the RNA encoding the amino acid sequence comprising the SARS-CoV-2S protein, an immunogenic variant thereof, or an immunogenic fragment of said SARS-CoV-2S protein or an immunogenic variant thereof comprises a poly-A sequence.

56. The method of embodiment 55, wherein said poly-A sequence comprises at least 100 nucleotides.

57. The method of embodiment 55 or 56, wherein said poly-A sequence comprises or consists of the nucleotide sequence of SEQ ID NO. 14.

58. The method of any one of embodiments 40-57, wherein said RNA is formulated as a liquid, a solid, or a combination thereof.

59. The method of any one of embodiments 40-58, wherein said RNA is administered by injection.

60. The method of any one of embodiments 40-59, wherein said RNA is administered by intramuscular administration.

61. The method of any one of embodiments 40-60, wherein said RNA is formulated as a particle.

62. The method of embodiment 61, wherein the particle is a Lipid Nanoparticle (LNP) or lipoplex (lpx) particle.

63. The method of embodiment 62, wherein said LNP particles comprise ((4-hydroxybutyl) azanediyl) bis (hexane-6, 1-diyl) bis (hexyl 2-decanoate), 2- [ (polyethylene glycol) -2000] -N, N-ditetradecylacetamide, 1, 2-distearoyl-sn-glycerol-3-phosphocholine, and cholesterol.

64. The method of embodiment 62, wherein said RNA lipoplex particles are obtainable by mixing RNA with liposomes.

65. A composition or a pharmaceutical product according to any one of embodiments 40 to 64, wherein the RNA is mRNA or saRNA.

66. The method of any one of embodiments 40-65, which is a method of vaccinating against a coronavirus.

67. The method of any one of embodiments 40-66, which is a method for the therapeutic or prophylactic treatment of a coronavirus infection.

68. The method according to any one of embodiments 40-67, wherein the subject is a human.

69. The method of any one of embodiments 40-68, wherein said coronavirus is a beta coronavirus.

70. The method of any one of embodiments 40-69, wherein said coronavirus is sabevivirus (sarbecovirus).

71. The method of any one of embodiments 40-70, wherein the coronavirus is SARS-CoV-2.

72. The method of any of embodiments 40-71, wherein the composition is the composition of any of embodiments 1-39.

73. A composition or a pharmaceutical product according to any one of embodiments 1 to 39 for use in a method according to any one of embodiments 40 to 72.

Certain exemplary embodiments of the invention

1. A pharmaceutical composition comprising:

An RNA comprising an open reading frame encoding a polypeptide comprising a SARS-CoV-2S protein or an immunogenic fragment or variant thereof, wherein the RNA is formulated in a lipid nanoparticle,

the lipid nanoparticle comprises a cationic ionizable lipid, a neutral lipid, a steroid, and a polyethylene glycol (PEG) -lipid.

2. The composition of embodiment 1, wherein said immunogenic fragment is or comprises an RBD.

3. The composition of embodiment 1, wherein said RNA is a modified RNA that is modified by substituting some or all of the uridine residues with modified uridine residues.

4. The composition of embodiment 1, wherein the RNA is formulated in a lipid nanoparticle comprising a cationic ionizable lipid, a phospholipid, cholesterol, and a polyethylene glycol (PEG) -lipid.

5. The composition of embodiment 4 wherein the phospholipid is or comprises Distearoylphosphatidylcholine (DSPC).

6. The composition of embodiment 1, wherein the steroid is cholesterol.

7. The composition of embodiment 1, wherein the (PEG) -lipid is or comprises the structure:

or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein:

R12And R13Each independently a linear 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 linkages; and w has an average value of 30 to 60.

8. The composition of embodiment 1, wherein the cationic ionizable lipid has the following structure:

or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein:

G1and G2Each independently is unsubstituted C1-C12Alkylene or C1-C12An alkenylene group;

G3is C1-C24Alkylene radical, C1-C24Alkenylene radical, C3-C8Cycloalkylene radical, C3-C8Cycloalkenylene;

R1and R2Each independently is C6-C24Alkyl or C6-C24An alkenyl group;

R3is H, OR5、CN、-C(=O)OR4、-OC(=O)R4or-NR5C(=O)R4

R4Is C1-C12An alkyl group;

R5is H or C1-C6An alkyl group.

9. The composition of embodiment 8 wherein R1Or R2Or both having one of the following structures:

10. the composition of embodiment 1, wherein the neutral lipids are present at a concentration of 5-15 mole percent of total lipid.

11. The composition of embodiment 1, wherein the cationic ionizable lipid is present at a concentration of 40-55 mole percent of total lipid.

12. The composition of embodiment 1, wherein the steroid is present at a concentration of 30-50 mole percent of total lipid.

13. The composition of embodiment 1, wherein the pegylated lipid is present at a concentration of 1-10 mole percent of total lipid.

14. The composition of embodiment 1, wherein the lipid nanoparticle comprises 40-55 mole percent cationic ionizable lipid, 5-15 mole percent neutral lipid, 30-50 mole percent steroid, and 1-10 mole percent pegylated lipid.

15. The composition of embodiment 1, wherein the RNA further comprises: 5 ' cap, 5 ' UTR, 3 ' UTR and polyA sequences.

16. The composition of embodiment 15, wherein the polyA sequence comprises at least 100 a nucleotides.

17. The composition of embodiment 16, wherein said polyA sequence is a break sequence of a nucleotides.

18. The composition of embodiment 1, wherein said RNA comprises a 5' cap.

19. The composition of embodiment 18, wherein said 5' cap is or comprises a cap1 structure.

20. The composition of embodiment 1, further comprising at least one salt and/or cryoprotectant.

21. The composition of embodiment 20, wherein the cryoprotectant is or comprises sucrose.

22. The composition of embodiment 1, wherein the composition is characterized in that,

serum from mouse subjects administered doses of 0.2 μ g-5 μ g RNA showed production of antibodies to the polypeptides encoded by the open reading frames 7 days after administration.

23. The composition of embodiment 1, wherein the composition is characterized in that,

serum from mouse subjects dosed with 1 μ g to 5 μ g of modified RNA showed pseudovirus neutralizing activity 14 days after dosing.

24. The composition of embodiment 1, wherein said RNA is present in said composition in an amount ranging from 1 μ g to 100 μ g per dose.

25. A method of vaccination by administering a pharmaceutical composition comprising:

an RNA comprising an open reading frame encoding a polypeptide comprising a SARS-CoV-2S protein or an immunogenic fragment or variant thereof, wherein the RNA is formulated in a lipid nanoparticle comprising a cationic ionizable lipid, a neutral lipid, a steroid, and a pegylated lipid.

26. A method comprising administering to a subject a dose of the composition of embodiment 25.

27. The method of embodiment 25, wherein said dose comprises an amount of said RNA in the range of 1 μ g to 100 μ g.

28. The method of embodiment 25, wherein said administering is by intramuscular administration.

29. The method of embodiment 25, further comprising administering to the subject at least a second dose at a time after the first dose.

30. The method of embodiment 29, wherein said second dose is administered 7 to 28 days after the administration of the first dose.

Citation of documents and studies referred to herein is not intended as an admission that any of the foregoing is pertinent prior art. All statements as to the contents of these documents are based on the information available to the applicants and do not constitute any admission as to the correctness of the contents of these documents.

The following description is presented to enable one of ordinary skill in the art to make and use various embodiments. Descriptions of specific devices, techniques, and applications are provided only as examples. Various modifications to the examples described herein will be readily apparent to those skilled in the art, and the generic principles defined herein may be applied to other examples and applications without departing from the spirit and scope of various embodiments. Thus, the various embodiments are not intended to limit the examples described and illustrated herein, but are consistent with the scope of the claims.

Examples

Example 1: in vivo immunogenicity using influenza Hemagglutinin (HA) as a model antigen

The efficacy of the RNA platform to be used in the coronavirus vaccines described herein was tested by extensive immunogenicity and challenge studies using influenza HA as a model antigen. These studies explored the induction of antibody responses determined by antigen-specific enzyme-linked immunosorbent assay (ELISA) tests and functional studies using Virus Neutralization (VNT) assays. One study evaluated the efficacy of LNP formulations using modRNA-LNP vaccines encoding influenza HA. Mice were IM injected with 1 μ g of LNP formulated influenza HA modRNA on days 0 and 28. On days 14, 28 and 49, blood samples were collected and tested for immunogenicity. The analysis showed a high antibody immune response, resulting in very high antigen-specific IgG titers in serum and high virus neutralizing activity (fig. 3). In addition, the modRNA vaccine induced strong Th1 CD4 and CD 8T cell responses (FIG. 4).

Example 2: immunogenicity studies of coronavirus vaccine candidates

Preliminary pharmacodynamic studies were performed in BALB/c mice to test the immunogenicity of the vaccine candidates shown in the table below.

Table 2: vaccine candidates

Vaccine mRNA type Vaccine-encoded antigens
BNT162a1 uRNA RBD (receptor binding Domain) of SARS-CoV-2 spike protein (S protein)
BNT162b1 modRNA SARRBD (receptor binding Domain) of S-CoV-2 spike protein (S protein)
BNT162b2 modRNA Modified forms of SARS-CoV-2 spike protein (S protein)
BNT162c1 saRNA RBD (receptor binding Domain) of SARS-CoV-2 spike protein (S protein)

Thus, it can be seen that various forms of embodiments were evaluated in parallel. This described method and system has enjoyed significant and effective success, enabling the development of effective clinical candidates within months of providing antigen (e.g., SARS-CoV-2S 1 protein and/or its RBD) sequences (relevant sequence information (e.g., GenBank: MN908947.3) is available in 1 month 2020).

Four groups of 8 female BALB/c mice were immunized once per study with three different doses of animal test material or buffer (control; see Table 3). Although the clinical test material will be diluted with saline, the animal test material is diluted in PBS containing 300mM sucrose. Since this is the storage buffer of the material itself, the test program is representative for a vaccine that will be used in the planned clinical trial. Immunization was performed using a dose volume of 20 μ L, IM.

Table 3: design of research

Blood from immunized animals was collected on days 7, 14, 21 and 28 and antibody immune responses were analyzed by ELISA and pseudovirus-based neutralization assay (pVNT).

SARS-CoV-2-S specific antibody responses to recombinant S1 subunit or RBD were detected by ELISA. Briefly, high protein-binding 96-well plates (MaxiSorp ELISA plates, VWR International GmbH, cat No.7341284) were coated overnight at 4 ℃ with 100ng of recombinant S1 subunit (nano Biological inc., cat No.40591-V08H) or RBD (nano Biological inc., cat No.40592-V02H) per well in 100 μ L of coating buffer (50mM sodium carbonate-sodium bicarbonate buffer, pH 9.6). Plates were washed three times with 300 μ L/well 1x phosphate buffered saline (PBS, VWR International GmbH, Cat No.0780-10L) supplemented with 0.01% Tween 20(Carl Roth GmbH & co. kg, Cat No.9127.1) and blocked with 250 μ L/well 1x casein blocking buffer (Sigma-Aldrich GmbH, Cat No. b6429-500ml) at 37 ℃ for 1 hour on a microplate shaker. The plate was again washed three times with 300 μ Ι/well of 1x PBS supplemented with 0.01% Tween 20 and incubated with mouse serum samples diluted in 1x casein blocking buffer for 1 hour at 37 ℃ on a microplate shaker. The plates were washed three times with 300. mu.L/well of 1 XPBS supplemented with 0.01% Tween 20, followed by incubation with peroxidase-conjugated goat anti-mouse secondary antibody (Jackson ImmunoResearch Ltd., Cat. No. 115-036-071; diluted 1:7500 in 1 XPS blocking buffer) at 37 ℃ for 45 minutes on a microplate shaker. Plates were washed three times with 300 μ L/well of 1x PBS supplemented with 0.01% Tween 20 and 100 μ L/well of TMB substrate (Biotrend Chemiekalien GmbH, cat. No.4380a) was added. The plates were incubated at room temperature for 8 minutes and the reaction was stopped by adding 100 μ L of 25% sulfuric acid (VWR International GmbH, cat. No. 1007161000). The plate was read on a microplate reader and the absorbance recorded at 450nm was corrected by subtracting the reference absorbance at 620 nm.

Functional antibody responses to vaccine candidates were detected by pVNT. pvntn uses a replication-defective Vesicular Stomatitis Virus (VSV) that lacks the genetic information of the VSV envelope glycoprotein G, but contains an open reading frame for Green Fluorescent Protein (GFP). VSV/SARS-CoV-2 pseudovirus was produced according to the published protocol (Hoffmann et al, Cell, 2020; PMID 32142651). The pseudotyped virus carries SARS-CoV-2S protein which mediates cell entry. Thus, pseudoviruses can be inactivated by neutralizing antibodies that bind SARS-CoV-2S. This inactivation can be analyzed by in vitro methods.

Briefly, 4X10 per well was added to 150. mu.L/well of DMEM (Thermo Fisher Scientific, Cat. No.61965059) supplemented with 10% fetal bovine serum (FBS, Sigma-Aldrich GmbH, Cat. No. F7524)4Vero76 cell (CRL-1587TM) Seeded in 96-well plates (Greiner Bio-One GmbH, Cat. No. 655160). Cells were incubated at 37 ℃ and 7.5% CO2Incubate for 4-6 hours. Meanwhile, mouse serum samples were diluted 1: 6 to 1: 768 in DMEM/10% FBS in a two-fold dilution procedure. The diluted serum samples were combined with equal volumes of titrated and prediluted VSV/SARS CoV-2 pseudovirus supernatant, which resulted in serum dilutions of 1: 12 to 1: 1536. The pseudovirus/serum dilution mixture was incubated at 750rpm on a microplate shaker at RT for 5 minutes and at room temperature for an additional 5 minutes without agitation. 50 μ L/well of a pseudovirus/serum dilution mix was added to the seeded Vero-76 cells, the pseudovirus volume applied per well corresponding to 200 Infectious Units (IU). Each dilution of serum samples was tested in duplicate wells. Cells were incubated at 37 ℃ and 7.5% CO 2Incubation was performed for 16-24 hours. Vero76 cells incubated with pseudovirus in the absence of mouse serum were used as positive controls. Vero76 cells not incubated with pseudovirus were used as negative control. After incubation, the cell culture plates were removed from the incubator, placed in the IncuCyte viable cell assay system (Essen Bioscience), and incubated for 30 minutes prior to analysis. Full-well scans of bright field and GFP fluorescence were performed using a 4 x objective. To calculate the neutralization titer, the number of infected GFP positive cells per well was compared to a pseudovirus positive control. Mean of pseudovirus controls multiplied by 0.5 indicated 50% pseudovirus neutralization (pVN)50). Serum samples with mean values below this cut-off value were each shown>50% virus neutralizing activity.

Immunogenicity Studies of BNT162a1(RBL063.3)

To investigate the efficacy of LNP formulated u rna vaccine encoding BNT162a1, BALB/c mice were IM immunized once as shown in table 3. The immunogenicity of RNA vaccines has been studied by focusing on antibody immune responses.

ELISA data at 7, 14, 21 and 28 days (d) after the first immunization showed early, dose-dependent immune activation against the S1 protein and the receptor binding domain (fig. 5).

Immunogenicity Studies of BNT162b1(RBP020.3)

To investigate the efficacy of the LNP formulated modRNA vaccine encoding BNT162b1, BALB/c mice were IM immunized once as shown in table 3. The immunogenicity of RNA vaccines has been studied by focusing on antibody immune responses.

ELISA data at 7, 14, 21 and 28 days after the first immunization showed early, dose-dependent immune activation against the S1 protein and the receptor binding domain (fig. 6). Sera obtained 14, 21 and 28 days after immunization showed high SARS-CoV-2 pseudovirus neutralization, especially of mice immunized with 1 or 5 μ g BNT162b1, and correlated with a strong increase in IgG antibody titer (fig. 7).

Immunogenicity Studies of BNT162c1(RBS004.3)

To investigate the efficacy of LNP formulated saRNA vaccine encoding BNT162c1, BALB/c mice were IM immunized once as shown in table 3. The immunogenicity of RNA vaccines has been studied by focusing on antibody immune responses.

ELISA data at 7, 14 and 21 days after the first immunization showed early, dose-dependent immune activation against the S1 protein and the receptor binding domain (fig. 8). Sera obtained 14 and 21 days after immunization showed dose-dependent SARS-CoV-2 pseudovirus neutralizing activity (figure 9).

Immunogenicity study of LNP-formulated uRNA (RBL063.1) encoding the viral S protein-V8 (SEQ ID NOS: 7, 8)

To investigate the efficacy of the LNP formulated uRNA vaccine (RBL063.1) encoding viral S protein-V8, BALB/c mice were IM immunized once as shown in Table 3. The immunogenicity of RNA vaccines has been studied by focusing on antibody immune responses.

ELISA data available 7, 14, 21 and 28 days after the first immunization showed early, dose-dependent immune activation against the S1 protein and the receptor binding domain (fig. 10). Sera obtained at 14, 21 and 28 days post-immunization showed dose-dependent SARS-CoV-2 pseudovirus neutralizing activity (figure 11).

Immunogenicity Studies of BNT162b2(RBP020.1)

To investigate the efficacy of the vaccine BNT162b2(RBP020.1), the immunogenicity of the construct was investigated. For this reason, dose titration studies were performed in BALB/c mice, where the analysis of immune responses was focused on antibody immune responses.

ELISA data available 7, 14 and 21 days after the first immunization showed early, dose-dependent immune activation against the S1 protein and the receptor binding domain (fig. 12). Sera obtained 14 and 21 days after immunization showed dose-dependent SARS-CoV-2 pseudovirus neutralizing activity (figure 13).

Immunogenicity Studies of LNP-formulated sarRNA (RBS004.2) encoding the viral S protein-V9 (SEQ ID NOS: 7, 9)

To investigate the efficacy of LNP formulated saRNA vaccine encoding V9, BALB/c mice were IM immunized once as shown in table 3. The immunogenicity of RNA vaccines was studied with a focus on antibody immune responses.

ELISA data available 7, 14 and 21 days after the first immunization showed early, dose-dependent immune activation against the S1 protein and the receptor binding domain (fig. 14). Sera obtained 14 and 21 days after immunization showed dose-dependent SARS-CoV-2 pseudovirus neutralizing activity (figure 15).

The above data demonstrate in vivo immune responses against the RBD with trimerization domain ("V5") and the mutated full-length S protein ("V8"/"V9") in all platforms tested, including the vaccines BNT162a1, BNT162b1, BNT162b2, and BNT162c 1. Antibody immune responses have been observed by ELISA at a very early time point (i.e., at day 7 post-immunization). Importantly, the induced antibodies were able to effectively neutralize SARS-COV-2 pseudovirus infection in vitro. Furthermore, when the modRNA platform (BNT162b1, BNT162b2) and saRNA platform (BNT162c1) were used, the antibody response induced with a very low immunization dose of 0.2 μ g/mouse indicated high efficacy of the vaccine candidate.

In mice, BNT162b2 induced higher antigen-specific titers than BNT162b1 encoded by the same RNA platform. As expected, immunogenicity to antigens in mice varied between RNA platforms. In mice, the most immunogenic platform based on antigen-specific antibody induction is modRNA followed by sarRNA. The uRNA platform induced the lowest antigen-specific antibody titers.

Example 3: selection of formulations

LNP delivery systems are often developed to efficiently and safely deliver therapeutic nucleic acids into the cytoplasm (cytosol) of various cell types following local administration in vivo. Early formulation work was performed with several promising LNP formulations and alternative RNAs encoding luciferase. The aim of the experiments was to correlate the effect of different ionizable cationic lipids on the efficacy of RNA delivery by LNP in vivo. Formulations were compared in terms of RNA encapsulation efficiency, apparent pKa, LNP size and polydispersity.

Among the screened cationic lipids, ALC-0315 showed suitable physical properties in terms of particle size, homogeneity and RNA encapsulation efficiency.

Based on this, ALC-0315/DSPC/CHOL/ALC-0159 prototype (prototype) was submitted for in vivo screening. The results shown in figure 16 summarize in vivo testing of two independent assay (pilot) batches using luciferase (Luc) RNA. The results demonstrate the improved efficacy of the ALC-0315 prototype compared to the internal baseline (ALC-0218). Based on these studies, ALC-0315 was identified as a highly potent cationic lipid and proposed for further product development studies.

The formulation screening procedure described above involves intravenous administration, resulting in delivery primarily to the liver. The mechanism of LNP uptake into hepatocytes is driven by endogenous apolipoprotein binding to LNP followed by receptor-mediated endocytosis (e.g., by low density lipoprotein receptor). To investigate whether intramuscular administration involves the same mechanism, LNP containing Luc RNA comprising ALC-0315 was injected intravenously (0.3mg/kg) and intramuscularly (0.2mg/kg) in ApoE knockout mice in the presence or absence of recombinant human ApoE 3. As a control, wild-type C57Bl/6 mice were also treated by different routes of administration. Prior to dosing, RNA-LNP was preincubated with recombinant human ApoE3(1mg of encapsulated mRNA with 1mg ApoE3) for 1 hour at Room Temperature (RT). Luc expression was monitored at 4, 24, 72 and 96 hours post-dose (fig. 17).

Luc expression was detected in wild-type C57Bl/6 mice when administered intravenously to the mice. In ApoE knockout mice, Luc expression was significantly reduced, however, when pre-incubated with exogenous ApoE, Luc expression was restored to similar expression levels as in wild type mice (fig. 18).

In vivo Luc expression experiments using a mouse model showed that, like intravenous administration, in the case of intramuscular administration, a similar mechanism is involved in the uptake of RNA-LNP, and this is not only for hepatocytes but also for local cells at the site of administration.

Finally, in vivo experiments following intramuscular administration of ALC-0315/DSPC/CHOL/ALC-0159 demonstrated minimal problems with respect to biodistribution, immunogenicity (vaccine activity) and tolerability (drainage).

Example 4: immunogenicity studies of coronavirus vaccine candidates

Use of IFN-gamma ELISpotPLUSThe kit (Mabtech, Cat. No.3321-4APT-2) detects a functional cellular immune response to a vaccine candidate by ELISpot assay. Briefly, spleens were removed from animals after sacrifice at day 28 post vaccination. The spleens were mechanically separated using a plunger of a syringe and a 70 μ M cell filter (Greiner Bio-One GmbH, Cat. No. 542070). Splenocytes were washed with excess volume of DPBS (Thermo Fisher Scientific, Cat. No. 14190-094), then centrifuged at 300 Xg for 6 minutes at RT and the supernatant discarded. The erythrocytes were then lysed with erythrocyte lysis buffer (154mM NH) at RT 4Cl,10mM KHCO30.1mM EDTA) for 5 minutes. The reaction was stopped with an excess volume of DPBS. After another washing step, the cells were resuspended in RPMI 1640 medium (Gibco, Cat. No.61870-010) supplemented with 10% FBS, 1% MEM non-essential amino acid solution (Gibco, Cat. No.11140-035), 1% sodium pyruvate (Gibco, Cat. No.11360-039), 0.5% penicillin/streptomycin (Gibco, Cat. No.15140-122), passed again through a 70 μm cell net and counted. According to the manufacturer's instructions, CD8a or CD4 was usedMicroBeads (Miltenyi Biotec, Cat. No. 130-117-. In parallel, 96-well ELISpot plates were washed with PBS and blocked with culture medium (RPMI 1640 medium supplemented with 10% FBS, 1% MEM nonessential amino acid solution, 1% sodium pyruvate, 0.5% penicillin/streptomycin matrix) for at least 30 minutes at 37 ℃. Followed by the addition of 50. mu.L of peptide solution (irrelevant control peptide AH1 (2. mu.g/mL; sequence: SPSYVYHQF), PepMix) in an IFN-. gamma.ELISpot assayTMSARS-CoV-2S-RBD (0.025. mu.g/mL per peptide; JPT, custom made) or PepMixTMSARS-CoV-2 spike glycoprotein (0.1. mu.g/mL per peptide; JPT, Cat. No. PM-WCPV-S-2) and 50. mu.L of autologous bone marrow-derived dendritic cells restimulated 1X 10 in 100. mu.L of medium 5CD8+ or CD4+ T cells. Each condition was tested in duplicate. The plates were treated with 5% CO2Incubated overnight in a humidified incubator at 37 ℃ and after approximately 18 hours (h), the cells were removed from the plates. IFN- γ spots were detected according to the manufacturer's protocol. After drying the plate under laminar flow for 2-3 hours, using an ELISpot microplate reader (plate reader) ((R))S6 Core Analyzer, CTL) were counted and the number of spots per well was analyzed.

In addition to the ELISpot assay, Luminex analysis was performed to understand the T of the T cell response detectedH1 or TH2 property. 5X 10 in 100. mu.L RPMI 1640 medium supplemented with 10% FBS, 1% MEM non-essential amino acid solution, 1% sodium pyruvate, 0.5% penicillin/streptomycin matrix5Splenocytes were transferred to 96-well flat-bottom cell culture plates. 100 μ L of an unrelated control peptide AH1(2 μ g/mL; sequence: SPSYVYHQF), or PepMix was addedTMSARS-CoV-2 spike glycoprotein (0.1. mu.g/mL per peptide; JPT, Cat. No. PM-WCPV-S-2). Plates were incubated for 48 hours and supernatants after harvesting were used for cytokine analysis (profiling). Use of bead-based T according to manufacturer's instructionsH1/TH2 Procartaplex immunoassay (Thermo Fisher Scientific, Cat. No. EPX110-20820- Cytokine concentrations in the re-stimulated splenocyte supernatants were determined. Fluorescence was measured using the Bioplex200 system (Biorad) and analyzed with ProcartaPlex Analyst 1.0 software (Thermo Fisher Scientific). The following analytes were measured: IFN-gamma; IL-12p 70; IL-13; IL-1 β (beta); IL-2; IL-4; IL-5; IL-6; TNF α (alpha); GM-CSF; IL-18.

For immunophenotypic analysis, flow cytometry analysis was performed. Briefly, erythrocytes from 50 μ L of fresh draw were lysed with ACK lysis buffer (Gibco) and stained with an immobilizable viability dye (eBioscience) and anti-CXCR 5 (rat IgG2a) antibody for 20 minutes at room temperature in flow buffer (dpbs (Gibco) supplemented with 2% FCS, 2mM EDTA (both from Sigma) and 0.01% sodium azide (morphist)) in the presence of Fc blocking solution (both from BD Bioscience). After staining with anti-rat IgG2a biotin for 20 minutes at 2-8 ℃ in flow Buffer, cells were stained extracellularly for 20 minutes at 2-8 ℃ with antibodies against CD3, CD4, CD8 α, CD38, CD44, PD-1, ICOS, CD62L, CXCR5, CD19 and streptavidin in Brilliant Stain Buffer Plus (BD Bioscience) diluted in flow Buffer. Cells were fixed with 2% rotihistofix (roth) for 15 min at room temperature. Cells were resuspended in Perm Buffer (FoxP3/Transcription Factor stabilizing Buffer Set, eBioscience) and incubated overnight at 2-8 ℃. Permeabilized cells were treated intracellularly with Fc blocking solution (block) at 2-8 ℃ for 10 minutes and stained with T-beta and GATA (BD bioscience) antibodies at 2-8 ℃ for 30 minutes. Cells were resuspended in flow buffer and harvested on a BD Symphony a3 flow cytometer (BD Bioscience) and analyzed with FlowJo 10.6.2.

For mouse B cell subtyping in draining lymph nodes, 2.5X10 was treated with Fc blocking fluid5Lymph node cells were stained for 15 min and extracellularly at 2-8 ℃ for 20 min with antibodies against CD19, CD45R/B220, IgD, CD138, IgM, CD38, CD95/FAS, IgG1, IgG2a, CD73, GR-1, F4/80, CD4, CD8 in Brilliant Stain Buffer (BD Bioscience). Cells were fixed with 2% RotiHistofix and incubated overnight at 2-8 ℃.

Immunogenicity Studies of BNT162b1(RBP020.3)

To investigate the efficacy of the LNP formulated modRNA vaccine encoding BNT162b1, BALB/c mice were IM immunized once as shown in table 3. The immunogenicity of RNA vaccines has been studied by focusing on cellular immune responses.

Both CD4+ and CD8+ T cells showed IFN- γ responses in the IFN- γ ELISpot assay after stimulation with S protein or RBD specific peptide pool (pool), but not with the unrelated peptide AH1 (fig. 22). In the Luminex assay, T is indicated byH1 driven immune response, confirmed cytokine production following peptide stimulation (figure 23).

Immunophenotypic analysis of blood 7 days post-immunization (fig. 24) showed a significant increase in circulating follicular helper T cells (Tfh) and activated T cells. On day 12 post-immunization, draining lymph nodes from immunized BALB/c mice were dissected and analyzed for B cell subsets (FIG. 25). A significant increase in B cells was found in lymph nodes with detectable numbers of plasma cells, class-switched B cells, and germinal center B cells positive for IgG1 or IgG2 a. Activation and maturation of the adaptive immune response was demonstrated in both blood and drainage lymph nodes.

Immunogenicity study of ModRNA (RBP020.1) encoding the viral P2-S protein V8 formulated in LNP

To investigate the efficacy of LNP formulated modRNA vaccine encoding RBP020.1, BALB/c mice were IM immunized once as shown in table 3. The immunogenicity of RNA vaccines has been studied by focusing on cellular immune responses.

Both CD4+ and CD8+ T cells showed IFN- γ responses in the IFN- γ ELISpot assay following stimulation with the S protein-specific peptide pool, but not with the irrelevant peptide AH1 (fig. 26). In the Luminex assay, T is indicated byH1 driven immune response, confirmed cytokine production following peptide stimulation (figure 27).

Immunogenicity Studies of LNP-formulated saRNA encoding the viral P2-S protein V9 (RBS004.2)

To investigate the efficacy of LNP formulated saRNA vaccine encoding RBS004.2, BALB/c mice were IM immunized once as shown in table 3. The immunogenicity of RNA was studied by focusing on cellular immune responses.

Both CD4+ and CD8+ T cells showed IFN- γ responses in the IFN- γ ELISpot assay following stimulation with the S protein-specific peptide pool, but not with the irrelevant peptide AH1 (fig. 28). In the Luminex assay, T is indicated byH1 driven immune response, confirmed cytokine production following peptide stimulation (figure 29).

Immunogenicity Studies of the BNT162b3 variant BNT162b3c and BNT162b3d

To understand the potential efficacy of transmembrane anchored RBD based vaccine antigens (schematic in figure 30; BNT162b3C (1) and BNT162b3d (2)), BALB/C mice were IM immunized once with 4 μ g of mRNA formulated in LNP-C12 or buffer used as a control. Non-clinical LNP-C12 formulated mRNA was used as a substitute for the BNT162b3 variants BNT162b3C and BNT162b3 d. Immunogenicity of RNA vaccines was investigated by focusing on antibody immune responses.

ELISA data at 6, 14 and 21 days after the first immunization showed early, dose-dependent immune activation against the S1 protein and the receptor binding domain (fig. 31). Sera obtained 6, 14 and 21 days after immunization showed high SARS-CoV-2 pseudovirus neutralization, associated with an increase in IgG antibody titers (figure 32).

Example 5: immunogenicity Studies of coronavirus vaccine candidates in non-human Primates (NHPs)

Six rhesus monkeys per group were immunized with 30 or 100 μ g BNT162b1 or with buffer IM on days 0 and 21. By 14 days after the first dose, antibodies bound to recombinant S1 were readily detectable, and by day 28 the level of antibodies bound to S1 exceeded the upper limit of quantitation determined (10,000U/mL). For comparison, 62 human COVID-19 convalescent sera obtained after symptom onset were analyzed for antibodies binding to S1. All time points in both NHP groups significantly exceeded the mean 422U/mL of human COVID-19 convalescent sera (fig. 33 a). By day 14 post-single immunization, VNT Geometric Mean Titers (GMTs) were detectable in sera of rhesus monkeys immunized with any dose level of BNT162b1 and reached geometric mean values of 768(30 μ g dose level) or 1,714(100 μ g) by day 28 (fig. 33 b). On day 35 (14 days post-boost), the neutralizing GMT was 282(30 μ g) and 975(100 μ g) (FIG. 33) b) In that respect CD4 on blood samples stimulated with the S-peptide mixture (mix) on day 42+Flow cytometry analysis of T cells showed TH1 significant secretion of the cytokines IFN γ, IL-2 and TNF α. In addition, IL-21 secretion was significantly increased. IL-21 is known to play a key role in B cell activation, expansion and plasma cell production as well as in the production of Tfh. In contrast, no significant amount of T was detectedHThe cytokine IL-4 (FIG. 33 c). In summary and in agreement with the results obtained in mice, BNT162b1 induced high antibody immunity with early affinity maturation with TH1 biased immune response.

In summary, we demonstrated that the methyl-nucleoside m1 Ψ -modified mRNA encoding the receptor binding domain of the trimeric SARS-CoV-2S protein plays a protective role in non-human primates (protective).

Example 6: phase 1/2 studies in healthy adults 18-55 years old to describe the safety, tolerability, and immunogenicity of COVID-19RNA vaccine candidates (BNT162b1)

We report the safety, tolerability and immunogenicity of blind observer dose escalation assays from placebo-controlled, observer-blind dose escalation tests performed in healthy adults who received 2 doses of either 10 μ g, 30 μ g or 100 μ g BNT162b1 at random, BNT162b1 being a Lipid Nanoparticle (LNP) -formulated, nucleoside-modified, mRNA vaccine candidate encoding the trimeric SARS-CoV-2 spike glycoprotein RBD antigen. Local reactions and systemic events were dose dependent, overall mild to moderate, and transient. The concentration of IgG that bound RBD and SARS-CoV-2 neutralizing titer increased with dose level and increased after the second dose. The geometric mean neutralization titer reaches 1.8-2.8 times of that of the human serum group in the COVID-19 convalescence stage.

The BNT162b1 vaccine candidate now undergoing clinical testing comprises the nucleoside 1-methyl-pseudouridine modified rna (modrna) and encodes the receptor binding domain of the critical target SARS-CoV-2 spike of neutralizing antibodies. The RBD antigen expressed by BNT162b1 was modified to enhance its immunogenicity by the addition of a "folder" trimerising domain derived from T4 secondary fibrin. Such RNA vaccine candidates are being tested in parallel in germany and in a coordinated study in the united states. Here, we present the data obtained in the us study.

Method

Study design and participants: this randomized, placebo-controlled, observer-blind phase 1/2 trial was conducted in the united states to evaluate the safety, tolerability, and immunogenicity of various BNT162 mRNA vaccine candidates at ascending dose levels. Evaluation of three dose levels (10, 30 or 100 μ g) of BNT162b1 candidate was performed at two sites in the united states. This study used a sentinel (sentinel) cohort design, with progression and dose escalation after reviewing the data for the sentinel cohort at each dose level. Healthy males between 18 and 55 years of age and non-pregnant females were enrolled. The key exclusion criteria include individuals known to be infected with human immunodeficiency virus, hepatitis c virus, or hepatitis b virus; immunocompromised individuals and individuals with a history of autoimmune disease; individuals at increased risk for severe COVID-19; a clinical or microbiological diagnosis of the previous COVID-19; receiving a drug intended to prevent COVID-19; any coronavirus vaccine previously vaccinated; and study SARS-CoV-2NAAT positive nasal swabs within 24 hours prior to vaccination.

The final protocol and informed consent documents were approved by the institutional review board of each study center participating in this study, and the study was conducted in accordance with all international conference on coordination (ICH) Good Clinical Practice (GCP) guidelines and ethical principles announced by helsinki. Prior to any activity on the study, an informed consent form is signed and dated.

End point: the primary endpoints of this study included: the proportion of participants reporting suggested local reactions, systemic events and use of antipyretics and/or analgesics within 7 days post-vaccination, AEs within 1 month post last dose, and SAEs within 6 months post-vaccination, and the proportion of pre-sentinel cohort participants with clinical laboratory abnormalities 1 week post-vaccination and grade changes in laboratory assessments between baseline and 1 and 7 days post dose 1, and between dose 2 and 7 days post dose 2. The secondary endpoints included: SARS-CoV-2 neutralization Geometric Mean Titer (GMT); geometric Mean Concentrations (GMC) of IgG binding to SARS-CoV-2S 1 and IgG binding to RBD 7 and 21 days after dose 1 and 7 and 14 days after dose 2; geometric mean fold increase (GMFR), > 4 fold increase from baseline and geometric mean ratio of SARS-CoV-2 serum neutralizing GMT to SARS-CoV-2 antigen binding antibody GMC at each time point (GMR).

The procedure is as follows: using a web-based interactive response technology system, study participants were randomly assigned to vaccine groups, each group consisting of 15 participants (12 active vaccine recipients and 3 placebo recipients). Participants received two 0.5mL doses of BNT162b1 or placebo administered by intramuscular injection into deltoid muscle.

BNT162b1 contains Good Manufacturing Process (GMP) -grade mRNA drug substance that encodes trimeric SARS-CoV-2 spike glycoprotein RBD antigen. mRNA is formulated with lipids as mRNA-LNP drug product. The vaccine was supplied as a buffered liquid solution for IM injection and stored at-80 ℃. Placebo was sterile saline solution for injection (0.9% sodium chloride injection, dose 0.5 mL).

Safety assessments for all participants included 4 hour observations after vaccination against the immediate AE (for the first 5 participants in each sentinel group) and 30 minute observations (for the remaining participants). Safety assessments also included self-reporting of suggested local reactions (injection site redness, swelling and pain), systemic events (fever, fatigue, headache, chills, vomiting, diarrhea, muscle pain and joint pain) for 7 days post-vaccination in an electronic diary (e-diary), and reporting of non-suggested AEs within 1 month post-vaccination with antipyretics and/or analgesics, and SAE within 6 months post-last vaccination. Hematological and chemical assessments were performed at screening, 1 and 7 days after dose 1, and 7 days after dose 2.

A safety stop rule is specified for all sentinel queue participants. Both the internal review board (IRC) and the external data review board (EDMC) reviewed all security data.

Immunogenicity testing: before each study vaccination, 7 and 21 days after dose 1, and 7, 14 days, 1 month, and 6 months after dose 2, 50mL of blood was collected for immunogenicity assessment (SARS-CoV-2 serum neutralization assay, SARS-CoV-2S 1 specific IgG direct Luminex immunoassay, SARS-CoV-2 RBD specific IgG direct Luminex immunoassay, and non-vaccine antigen (NVA) Ig direct Luminex immunoassay).

SARS-CoV-2 neutralization assay A previously described SARS-CoV-2 strain (USA _ WA1/2020) which has been rescued by reverse genetics and engineered by insertion of the mNeonGreen (mNG) gene in the open reading frame 7 of the viral genome WAs used. This reporter virus produced similar plaque morphology and indistinguishable growth curves as the wild-type virus. Serial dilutions of heat inactivated serum were incubated with reporter virus for 1 hour at 37 ℃ prior to inoculation with Vero E6 cell monolayers. Between 16 and 24 hours after inoculation, infected foci (foci) were detected by fluorescence by adding Hoechst 33342 solution and counted with a staining 7Cell Imaging Multi-Mode Reader.

Statistical analysis: the sample size of the sentinel cohort for this study was not based on statistical hypothesis testing. The primary safety objectives were evaluated statistically by descriptive summaries of local responses, systemic events, abnormal hematological and chemical laboratory parameters, and AEs and SAEs for each vaccine group. AE was summarized using the grade 3 (tier) method. The secondary immunogenic targets at each time point are summarized descriptively.

Results

Between 4 days 5/2020 and 19 days 6/2020, 76 subjects were screened and 45 participants were randomized and vaccinated. On days 0 and 21, 12 participants vaccinated with BNT162b1 at each dose level (10 μ g, 30 μ g, or 100 μ g) and 9 participants received placebo (fig. 34). The study population consisted of healthy male and non-pregnant female participants aged 18-55 years, with a mean age of 35.4 years (19 years minimum, 54 years maximum). Overall, 51.1% of participants were male and 48.9% were female. The majority of participants were whites (82.2%) and non-hispanic/non-hispanician (93.3%).

Safety and tolerability

Injection site pain was the most frequent local reaction within 7 days after either vaccination, 58.3% (7/12) in 10 μ g and 100.0% (each 12/12) in the 30 μ g and 100 μ g BNT162b1 groups after dose 1, and 22.2% (2/9) reports for placebo recipients, with 83.3% and 100.0% BNT162b1 recipients reported at 10 μ g and 30 μ g dose levels after dose 2, respectively. All local reactions were mild or moderate in severity, except for the severe pain reported after dose 1 of 100 μ g BNT162b 1.

In BNT162b1 and placebo recipients, the most common systemic events reported within 7 days post-vaccination were mild to moderate fatigue and headache. Systemic events increased with dose level and were reported in a greater number of subjects after the second dose (10 μ g and 30 μ g groups). After dose 1, 50.0% (6/12) of BNT162b1 recipients in the 100 μ g group and 8.3% (1/12) of each of the 10 μ g and 30 μ g groups reported a fever of ≧ 38.0 ℃. After dose 2, 8.3% (1/12) of the participants in the 10. mu.g group and 75.0% (9/12) of the participants in the 30. mu.g group reported fever ≧ 38.0 ℃. No grade 4 systemic events or fever were reported (fig. 35 and 36). Most local and systemic events peaked before day 2 post vaccination and resolved by day 7. Based on the reactogenicity profile, participants who received the initial 100 μ g dose did not receive the second vaccination.

50.0% (6/12) of participants who received 10 μ g or 30 μ g BNT162b1, 58.3% (7/12) of participants who received 100 μ g BNT162b1, and 11.1% (1/9) of placebo recipients reported adverse events. Two participants reported severe AE, one at a dose level of 30 μ g (grade 3 fever 2 days post vaccination) and one at a dose level of 100 μ g (sleep disturbance 1 day post vaccination). Related AEs were reported for 25% (3/12) to 50% (6/12) BNT162b1 recipients and 11.1% (1/6) of those receiving placebo. No serious adverse events were reported.

No grade 1 or higher changes were observed in routine clinical laboratory values or laboratory abnormalities for most subjects after either BNT162b1 vaccination. The most significant change was a decrease in lymphocyte count in 8.3% (1/12), 45.5% (5/11) and 50.0% (6/12) of the participants receiving 10 μ g, 30 μ g or 100 μ g of BNT162b1, respectively. There was a grade 3 lymphopenia in one participant at each of the 10 μ g (8.3%) and 30 μ g (9.1%) dose levels and in 4 participants at the 100 μ g dose level (33.3%). These hematological changes were noted in blood drawn 1-3 days after dose 1 and returned to normal 6-8 days after vaccination. Changes in laboratory values after vaccination were all independent of clinical findings. In addition, grade 2 neutropenia (neutropenia) was noted 6-9 days after the second dose of 10 μ g or 30 μ g BNT162b1, 1 participant each. The neutrophil counts of these two subjects were not repeated, however they were continuously tracked in the study and to date there was no report of adverse events or clinical manifestations of neutropenia.

Immunogenicity

The concentration of IgG that bound RBD and SARS-CoV-2 neutralizing titer were assessed in sera drawn at baseline and 7 and 21 days after the first dose and 7 days after the second dose (day 28) of BNT162b1 (figure 37 a). The Geometric Mean Concentration (GMC) of RBD-binding IgG was 534-1,778 units/mL by 21 days after the first dose (for all three dose levels) compared to 602 units/mL for a panel of COVID-19 convalescent human sera. By 7 days after the second dose (for 10. mu.g and 30. mu.g dose levels), the RBD-binding IgG GMC increased to 4,813-30,207 units/mL. Since the participants who received the first dose of 100 μ g BNT162b1 did not receive the second dose, the progress of the antibody response without the second dose could not be assessed, and the RBD-binding antibody concentration did not increase further in the participants of this dosing group beyond 21 days after the first dose. In participants receiving BNT162b1 at dose levels of 10 μ g and 30 μ g, highly elevated RBD-binding antibody concentrations continued up to day 35 (two weeks after the second dose).

A modest increase in the SARS-CoV-2 neutralization Geometric Mean Titer (GMT) was observed 21 days after dose 1 (FIG. 37 b). Significantly higher serum neutralization of GMT was achieved 7 days after the participants received the second 10 μ g or 30 μ g dose, reaching 168-267, compared to 94 in the COVID-19 convalescent serogroup.

Discussion of the related Art

The RNA-based vaccine candidate BNT162b1 is safe and well tolerated. All dose levels were shown to be consistent with the tolerance and safety profile (profile) previously observed for mRNA-based vaccines. Significant dose level responses were observed in adults 18-55 years of age after dose 1 and dose 2. The reactogenicity was generally higher after the second dose, but symptoms resolved within a few days after appearance. Participants who were randomly assigned to the 100 μ g group did not receive a second vaccination according to the tolerability profile of the first dose at the 100 μ g dose level. Transient lymphocyte depletion (grade 1-3) was observed within days after vaccination; however, lymphocyte counts in all participants returned to baseline within 6-8 days. These laboratory abnormalities were not associated with clinical findings. Lymphopenia (lymphopenia) after vaccination is most likely explained by transient migration of lymphocytes into tissues.

Robust immunogenicity was observed after vaccination with BNT162b 1. RBD-binding IgG concentrations were detected on day 21 and increased significantly 7 days after the booster dose was administered on day 21. IgG GMCs binding to RBD in vaccinated participants (10 μ g dose level) after the first dose were similar to that observed in a group of 38 sera from COVID-19 convalescent humans obtained 20-40 days after symptom onset and at least 14 days after the onset of the asymptomatic convalescent phase. The GMC was significantly higher in sera drawn from the 30 μ g and 100 μ g dose level cohorts than in the convalescent serogroup. After booster vaccination with 10 μ g or 30 μ g BNT162b1 (dose 2), IgG GMCs binding to RBD were 8.0-fold to 50-fold higher than those of the convalescent serogroup GMCs.

Sera from vaccinated participants were also tested in the SARS-CoV-2 neutralization assay. At day 21, the neutralizing titer was measurable at all dose levels. On day 28 (7 days after booster dose), a clear SARS-CoV-2 neutralizing titer was observed. Virus neutralization GMT after 10 μ g and 30 μ g booster vaccination (dose 2) was 1.8-fold and 2.8-fold, respectively, that of convalescent serogroups. Since the 100 μ g dose level cohort was not boosted, there was no corresponding data on immunogenicity after the second vaccination.

These clinical findings with respect to BNT162b1 vaccine candidates are very encouraging and provide strong evidence to support accelerated development and risk-oriented manufacturing (at-risk manufacturing) to maximize the opportunity to provide a prophylactic vaccine to prevent COVID-19 as soon as possible.

Example 7: concurrent antibody and T cell and cytokine responses elicited by the COVID-19RNA vaccine

In this example, we show characterization of antibody and T cell responses after vaccination with BNT162b1 from non-randomized open label phase I/II trials in healthy adults 18-55 years of age. Two doses of 1 μ g, 10 μ g, 30 μ g and 50 μ g BNT162b1 administered at 21 day intervals elicited concomitant (concomitant) antibodies and robust CD4+And CD8+T cell response. All subjects showed strong antibody responses with IgG concentrations significantly higher than that observed in human serum during COVID-19 recovery. Day 43, the SARS-CoV-2 serum neutralization geometric mean titers ranged from 0.7-fold (1 μ g) to 3.3-fold (50 μ g) compared to a panel of COVID-19 convalescent human sera and had broad activity against multiple (subve) SARS-CoV-2 spike variants. Interferon (IFN) gamma is an immunostimulatory cytokine with antiviral properties, and is encoded by high frequency RBD antigen-specific CD8 +T and a plurality of CD4+T cell production. Detection of enhanced T in RBD-stimulated immune cellsH1 immune cell profile of IL-12p 70. The robust RBD-specific antibodies, T cells and favorable cytokine responses of BNT162b1 mRNA vaccines suggest the potential for multiple beneficial protection mechanisms against COVID-19.

Materials and methods

Design of clinical trials

Study BNT162-01(NCT 04380701-german test) is an ongoing, first-person, phase I/II, open label dose exploration clinical trial to evaluate the safety, tolerability, and immunogenicity of various BNT162 mRNA vaccine candidates given increasing dose levels intramuscularly. Healthy men and non-pregnant women aged 18-55 years (corrected to an increase of 56-85 years) were eligible. Key exclusion criteria included prior clinical or microbiological diagnosis of COVID-19; receiving an agent that prevents COVID-19; prior vaccination with any coronavirus vaccine; serological type detection of SARS-CoV-2 IgM and/or IgG is positive at screening visit; and studying SARS-CoV-2NAAT positive nasal swab within 24 hours before vaccination; an increased risk of developing severe COVID-19; individuals with low immunity are known to be infected with HIV, hepatitis c virus or hepatitis b virus and have a history of autoimmune disease. The primary endpoints of the study were safety and immunogenicity.

In the study section reported herein, five dose levels (1 μ g, 10 μ g, 30 μ g, 50 μ g, or 60 μ g) of the BNT162b1 candidate were evaluated in one site in germany, with 12 healthy volunteers per dose level in a dose escalation and decline design. Sentinel dosing was performed in each dose escalation cohort. The progression and dose escalation of this queue requires the data to be reviewed by a security review board. Subjects received a BNT162b1 priming dose on day 1 and a booster dose on days 22 ± 2. Sera for antibody assays were obtained on days 1 (pre-prime), 8 + -1 (post-prime), 22 + -2 (pre-boost), 29 + -3 and 43 + -4 (post-boost). PBMCs were obtained for T cell studies on days 1 (pre-prime) and 29 ± 3 (post-boost). One subject at 10 μ g and one subject at the 50 μ g dose cohort left the study before boosting for consent and personal reasons.

The data presented only contained the cohort of BNT162b1 immunizations and based on a preliminary analysis of data with an extraction date of 2020, 7, month and 13, focused on an analysis of vaccine-induced immunogenicity (secondary endpoint) summarized descriptively at each time point. The immunogenicity assay included all participants with available data.

The test was carried out in Germany according to the declaration of Helsinki and good clinical practice guidelines and received an independent ethical Committee (Ethik-Kommision of theBaden-Huntteberg, Stuttgart, Germany) and the Bureau of administration (Paul-Ehrlich Institute, Langen, Germany). All subjects provided written informed consent.

Preparation of RNA

BNT162b1 contains Good Manufacturing Process (GMP) -grade mRNA drug substance that encodes trimeric SARS-CoV-2 spike glycoprotein RBD antigen. RNA was generated from DNA templates by in vitro transcription in the presence of 1-methylpseudouridine-5 '-triphosphate (m1 YTP; Thermo Fisher Scientific) instead of uridine-5' -triphosphate (UTP). Using trinucleotidesCap1 analog (trinucleatide cap1 analog) ((m)2 7,3’-O)Gppp(m2’-O) ApG, respectively; TriLink) for co-transcription capping. The RNA encoding the antigen contains sequence elements that increase RNA stability and translational efficiency in human dendritic cells (Holtkamp, S.et al, Blood 108, 4009-4017 (2006); Orlandini von Niessen, A. G.et al, mol.Ther.27, 824-836 (2019)). mRNA is formulated with lipids to obtain an RNA-LNP drug product. The vaccine was shipped and supplied in buffered liquid solution for IM injection and stored at-80 ℃.

Proteins and peptides

Pools of 15-membered (mer) peptides overlapping 11 amino acids and covering the full-length sequence of SARS-CoV-2 RBD encoded by BNT162b1 were used for ex vivo stimulation of PBMCs for flow cytometry, IFN γ ELISpot and cytokine analysis. CEF (CMV, EBV, influenza virus; HLA class I epitope Peptide pool) and CEFT (CMV, EBV, influenza virus, tetanus toxoid; HLA class II epitope Peptide pool) (both from JPT Peptide Technologies) were used as general T cell reactivity controls.

Human convalescent serum and PBMC panel

At least 14 days after PCR confirmed diagnosis, and when subjects were asymptomatic, human SARS-CoV-2 infection/covd-19 convalescent sera (n 38) were drawn from subjects 18-83 years old. Serum donors have symptomatic infection (n-35) or are hospitalized (n-1). Serum was obtained from Sanguine Biosciences (Sherman Oaks, Calif.), MT Group (Van Nuys, Calif.) and Pfizer Occupical Health and Wellness (Pearl River, NY). Human SARS-CoV-2 infection/codv-19 convalescent PBMC samples (n-6) were collected from subjects 41-79 years old when subjects were asymptomatic, 45-59 days after PCR confirmed. PBMC donors were asymptomatic/mildly infected (n ═ 4; clinical scores 1 and 2) or hospitalized (n ═ 2; clinical scores 4 and 5). Blood samples were obtained from Frankfurt (Frankfurt) university hospital (germany).

Cell culture and primary cell isolation

Vero cells (ATCC CCL-81) and Vero E6 cells (ATCC CRL-1586) were cultured in GlutaMAX supplemented with 10% Fetal Bovine Serum (FBS) (Sigma-Aldrich)TM(Gibco) in Dulbecco's Modified Eagle's Medium (DMEM). ReceivingThereafter, the cell lines were tested for mycoplasma contamination prior to expansion and cryopreservation. Peripheral Blood Mononuclear Cells (PBMC) were isolated by Ficoll-Hypaque (Amersham biosciences) density gradient centrifugation and cryopreserved prior to subsequent analysis.

RBD binding IgG antibody assay

Will contain a C-terminal AvitagTMRecombinant SARS-CoV-2 RBD from (Acro Biosystems) was bound to streptavidin-coated Luminex microspheres. Serum from heat-inactivated subjects was diluted at 1:500, 1:5,000, and 1:50,000. After overnight incubation at 2-8 ℃ while shaking, plates were washed in a solution containing 0.05% Tween-20. While shaking, a second goat anti-human polyclonal antibody fluorescently labeled (Jackson Labs) was added at room temperature for 90 minutes, and then the plate was washed again with a solution containing 0.05% Tween-20. Data were captured as Median Fluorescence Intensity (MFI) using a Luminex reader (reader) and converted to U/mL antibody concentrations, arbitrarily designated 100U/mL concentrations, using a reference standard curve, taking into account serum dilution factors. Three dilutions were used to increase the probability that at least one result of any sample falls within the range available for the standard curve. The results of the assay are reported as U/mL IgG. The final assay result is expressed as the geometric mean concentration of all sample dilutions that produce a valid assay result within the assay range.

SARS-CoV-2 neutralization assay

Neutralization assays use the previously described SARS-CoV-2 strain (USA _ WA1/2020), which has been rescued by reverse genetics and engineered by insertion of the meneongreen (mng) gene in the open reading frame 7 of the viral genome (Xie, x.et al., Cell Host Microbe 27,841-848.e3 (2020)). This reporter virus produced similar plaque morphology and indistinguishable growth curves as the wild-type virus. As described previously, the virus was allowed to stand for a main stock (2x 10)7PFU/mL) were grown in Vero E6 cells (Xie, x.et al., Cell Host Microbe 27,841-848.E3 (2020)). Serial dilutions of heat-inactivated serum were combined with reporter virus (2X 10 for a final multiplicity of infection (MOI) of 0.5) prior to inoculation with Vero CCL81 cell monolayers in 96-well plates (targeting 8,000-15,000 cells per well)4PFU per hole to produce about 1Vero monolayer infection rate of 0-30%) was incubated at 37 ℃ for 1 hour to allow accurate quantification of infected cells. The total number of cells per well was counted by nuclear staining (Hoechst 33342) and virus-infected foci were detected 16-24 hours after inoculation with a rotation 7Cell Imaging Multi-Mode Reader (Biotek) with Gen5 Image Prime version 3.09. In GraphPad Prism version 8.4.2, titers were calculated by generating a 4-parameter (4PL) logistic fit of percent neutralization at each serial dilution. 50% neutralization Titer (VNT) 50) Reported as the reciprocal interpolated dilution (interpolated reciprocal) that produced 50% reduction of fluorescent viral foci. VSV-SARS-CoV-2 spike variant pseudovirus neutralization assay

The production and neutralization assay of VSV-SARS-CoV-2-S pseudoparticles was performed as previously described (Baum, A.et. Science, eabd0831(2020). doi: 10.1126/Science. abd0831). Briefly, a human codon-optimized SARS-CoV-2 spike (GenBank: MN908947.3) (Genscript) was synthesized and cloned into an expression plasmid. The complete genomic sequence of SARS-CoV-2 was downloaded from the GISAID nucleotide database (https:// www.gisaid.org) (last visit 24/8/2020). Sequences were permuted and genetic diversity of the gene encoding the spikes was assessed across high quality genomic sequences using custom-made pipelines. Amino acid substitutions were cloned into spike expression plasmids using site-directed mutagenesis. HEK293T cells (ATCC CRL-3216) were inoculated (medium: DMEM high glucose (Life Technologies) supplemented with 10% heat-inactivated fetal bovine serum (FBS; Life Technologies) and penicillin/streptomycin/L-glutamine (Life Technologies)) and spike expression plasmid transfection was performed the next day using Lipofectamine LTX (Life Technologies) according to the manufacturer's protocol. 24 hours post-transfection, the cells were transfected with VSV Δ G diluted in Opti-MEM (Life technologies) at 37 ℃: the mNeon/VSV-G virus infects cells at a multiplicity of infection of 1. Cells were incubated at 37 ℃ for 1 hour, washed to remove residual input virus and overlaid infection medium (DMEM high glucose supplemented with 0.7% low IgG BSA (Sigma), sodium pyruvate (Life Technologies) and 0.5% gentamicin (Life Technologies)). After 24 hours, the supernatant containing the VSV-SARS-CoV-2-S pseudoparticles was collected at 37 ℃, clarified by centrifugation at 3000Xg for 5 minutes, and stored at-80 ℃ until further use.

For the pseudovirus neutralization assay, Vero cells (ATCC CCL-81) were seeded in culture media in 96-well plates and allowed to reach approximately 85% confluence before being used in the assay (after 24 hours). Starting from the 1: 40 dilution, the serum was serially diluted 1: 2 in the infection medium. Pseudo particles of VSV-SARS-CoV-2-S were prepared at a ratio of 1: 1 dilution in infection medium was used for a fluorescence focus unit (ffu) count of about 1000 in the assay. The serum dilutions were mixed with the mock particles at 1: 1 for 30 minutes at room temperature, then Vero cells were added and incubated for 24 hours at 37 ℃. The supernatant was removed and replaced with pbs (gibco) and foci quantified using a SpectraMax i3 plate reader (Molecular Devices) with MiniMax imaging cytometer. In GraphPad Prism version 8.4.2, neutralization titers were calculated by generating a 4-parameter (4PL) logistic fit of percent neutralization at each serial dilution. 50% pseudovirus neutralization Titers (VNT)50) Reported as the reciprocal interpolated dilution that produced a 50% reduction in fluorescent viral foci.

IFNγELISpot

Use of exhausted CD4+And enriched in CD8+T cell (CD 8)+Effector) or depleted CD8+And enriched in CD4+T cell (CD 4)+Effector) ex vivo IFN γ ELISpot assay (no further in vitro culture for amplification). One in duplicate and tested using a positive control (anti-CD 3 monoclonal antibody CD3-2 (1: 1,000; Mabtech)). Multiscreen filter plates (Merck Millipore) pre-coated with IFN γ -specific antibodies (elispot pro kit, Mabtech) were washed with PBS and blocked with X-VIVO 15 medium (Lonza) containing 2% human serum albumin (CSL-Behring) for 1-5 hours. Per well, 3.3x 10 stimulation with overlapping peptide pools representing the vaccine-encoded RBD 5Effector cells were 16-20 hours. Bound IFN γ was visualized with a secondary antibody conjugated directly to alkaline phosphatase, followed by incubation with BCIP/NBT substrate (ELISpotPro kit, Mabtech). The plates were scanned with an AID Classic Robot ELISPOT Reader and analyzed by ImmunoCapture V6.3(Cellular Technology Limited) or AID ELISPOT 7.0 software (AID Autoimmun Diagnostika). The spot count is shown as the average of each multiple wellAnd (4) average value. According to moodi et al (moodi, z., et al, j.immunol.methods 315,121-32 (2006); moodi, z.et al, Cancer immunol.immunoher.59, 1489-501 (2010)), based on two statistical tests (distribution-free re-sampling), an internal ELISpot data analysis tool (EDA) was used to compare the T cell response stimulated with the peptide to effectors incubated with medium alone as negative controls to maintain control of false positives while providing sensitivity.

To account for the variation in sample mass reflected in the number of spots in response to anti-CD 3 antibody stimulation, a normalization method was employed to achieve a direct comparison of spot counts/response intensities between individuals. This dependence is modeled in a log-linear fashion using a Bayesian model that includes a noise component (not published). For robust normalization, each normalization is sampled 1000 times from the model and the median value taken is taken as the normalized blob count value. Likelihood of the model: log lambda E=αlogλP+logβj+ σ ε, where λEIs the normalized spot count of the sample, and α is at all positive controls λPCommon stability factor (normal distribution), betajIs the specific component (normal distribution) of sample j, and σ ε is the noise component, where σ is Cauchy distributed and ε is Student's-t distributed. Beta is ajEnsure that each sample is treated as a different batch.

Flow cytometry

Cytokine-producing T cells were identified by intracellular cytokine staining. PBMCs thawed and resting for 4 hours in OpTsizer medium supplemented with 2. mu.g/mL DNAseI (Roche) were restimulated in the presence of GolgiPlug (BD) with a Peptide pool representing the vaccine-encoded SARS-CoV-2 RBD (2. mu.g/mL/Peptide; JPT Peptide Technologies) at 37 ℃ for 18 hours. Controls were treated with media containing DMSO. Cells were subjected to viability and staining of surface markers for 20 minutes at 4 ℃ in flow buffer ((DPBS (Gibco), supplemented with 2% FCS (Biochrom), 2mM EDTA (Sigma-Aldrich.) then samples were fixed and permeabilized using the Cytofix/Cytoperm kit (BD Biosciences) according to the manufacturer's instructionsrm/Wash buffer at 4 ℃ for 30 min. Samples were taken on an FACS VERSE instrument (BD Biosciences) and analyzed with FlowJo software version 10.5.3(FlowJo LLC, BD Biosciences). The background of RBD-specific cytokine production was corrected by subtracting the values obtained for the DMSO-containing medium. A negative value is set to 0. By calculating all CD4 positive for IFN gamma, IL-2 or IL-4 +The total number of fractions of T cells was calculated to calculate cytokine production in figure 42b, this total number was set to 100%, and the fraction of each subset in which a particular cytokine was produced was calculated.

Cytokine analysis (profiling)

Human PBMC were re-stimulated with SARS-CoV-2 RBD peptide pool (2. mu.g/mL final concentration per peptide) for 48 h. Stimulation with DMSO-containing medium served as negative control. Use of a bead-based, 11-plex T according to the manufacturer's instructionsH1/TH2 human ProcartaPlex immunoassay (Thermo Fisher Scientific) determined the concentration of TNF, IL-1 β and IL-12p70 in the supernatant. Fluorescence was measured using the Bioplex200 System (Bio-Rad) and analyzed using ProcartaPlex Analyst 1.0 software (Thermo Fisher Scientific). The background of RBD-specific cytokine production was corrected by subtracting the values obtained for the DMSO-containing medium. A negative value is set to 0.

Results

Study design and analysis setup

Between 23 days 4/2020 and 22 days 5/2020, 60 subjects were vaccinated with BNT162b 1. Every 1 μ g, 10 μ g, 30 μ g and 50 μ g dose level 12 participants received the first dose on day 1 and boosted on day 22, and 12 participants received a 60 μ g priming dose only on day 1 (fig. 43). The study population consisted of healthy males and non-pregnant females with an average age of 41 years (19-55 years) and equal gender distribution. The majority of participants were white (96.7%), one african american and one asian subject (1.7% each). Preliminary data analysis focused on immunogenicity (table 4).

TABLE 4 subject deployment and analysis settings

Antibody analysis: the values represent the number of subjects for which virus neutralization assays were performed. The values in parentheses represent the number of subjects for which RBD-binding IgG antibody assays were performed.

T cell analysis: values represent the number of subjects for which IFN γ ELISpot was performed. The values in parentheses represent the number of subjects for which flow cytometry was performed.

N/A: the sample is not yet available.

In short, no Serious Adverse Events (SAE), unexpected toxicity and withdrawal due to related AEs were observed. The majority of reported solicited (solicited) AEs were signs and symptoms of vaccine reactogenicity, usually with onset within the first 24 hours post immunization, such as systemic and injection site reactions, major symptoms of pain and sensitivity (fig. 44). Symptoms were mostly mild or moderate, with occasional severe (grade 3) AEs, such as fever, chills, headache, muscle and joint pain, and injection site reactions. All AEs regressed automatically, mostly within 24 hours after onset, and could be managed by simple measures (e.g. acetaminophen). Based on the reported reactogenicity of the first dose, participants who received the initial 60 μ g dose did not receive the second 60 μ g dose.

No relevant changes in routine clinical laboratory values occurred after BNT162b1 vaccination, but a transient increase in the inflammatory marker C-reactive protein (CRP) and a transient decrease in blood lymphocyte counts in a dose-dependent manner was observed in vaccinated subjects (fig. 45). Based on our previous clinical experience with RNA vaccines, the latter may be due to transient redistribution of lymphocytes associated with innate immune stimulation (Kamphuis, e., et al., Blood 108,3253-61 (2006)).

Vaccine induced antibody response

IgG concentrations bound to RBD and SARS-CoV-2 neutralizing titers were assessed at baseline, 7 and 21 days after the primary BNT162b1 dose (days 8 and 22) and 7 and 21 days after the booster dose (days 29 and 43), except for the 60 μ g cohort, which received only priming (figure 39).

All subjects, including those receiving a 1 μ g dose, showed strong, dose-dependent vaccine-induced antibody responses. The Geometric Mean Concentration (GMC) of IgG binding to RBD was dose-dependent 21 days after the priming dose (for four dose levels of 1-50. mu.g), approximately 265-1,672U/mL (FIG. 39). 7 days after the booster dose (day 29), the dose-dependent, strong increase of RBD-binding IgG GMC in subjects treated with 1-50 μ g BNT162b1 was to about 2,015-25,006U/mL. The antibody GMC that binds to RBD in individuals vaccinated with BNT162b1 was about 3,920-22,700U/mL on day 43 (21 days post-boost), compared to about 602U/mL measured in a panel of sera drawn from 38 SARS-CoV-2 infection convalescent patients (18-83 years) at least 14 days after PCR diagnosis. In the 60 μ g dose cohort using the priming dose alone, by day 29, the RBD-binding IgG GMC was about 1,058U/mL, indicating the necessity of a second dose to boost antibody titers.

The Geometric Mean Titer (GMT) of the SARS-CoV-2 neutralizing antibodies increased moderately (modestly) in a dose-dependent manner 21 days after the priming dose (FIG. 40 a). Significantly higher seroneutralization GMT was achieved 7 days after the booster dose, reaching about 36(1 μ g dose level), about 158(10 μ g dose level), about 308 (30 μ g dose level) and about 578(50 μ g dose level) compared to about 94 days in the convalescent serogroup. On day 43 (21 days post-boost), the neutralizing antibody GMT was further increased to about 62(1 μ g dose), relatively stable at about 126(10 μ g dose), or slightly decreased to about 157(30 μ g dose), and 309(50 μ g dose), depending on the dose level. The neutralizing antibody GMT was strongly associated with IgG GMC binding to RBD (fig. 40 b). In summary, neutralizing antibody titers were largely within the range of the previously reported us study using BNT162b 1.

Furthermore, by 7 days after the second dose, sera from vaccinated subjects showed broad neutralization activity of 17 SARS-CoV-2 spike variants of the entire (across) group, identified in the publicly available SARS-CoV-2 sequence, including 16 RBD mutants (Baum, A.et al, Science, eabd0831(2020). doi:10.1126/Science. abd0831) and dominant spike variant D614G (Baum, A.et al, Science, eabd0831(2020). doi:10.1126/Science. abd0831) (FIG. 40 c).

Vaccine induced T cell response

Characterization of CD4 in BNT162b1 immunized subjects using direct ex vivo IFN γ ELISPOT with PBMCs from 36 subjects across a 1 μ g to 50 μ g dose cohort before priming (day 1) and at 29 days post priming (7 days post booster vaccination)+And CD8+T cell responses (figure 41). In this assay, CD4 was used+Or CD8+The T cell effectors were re-stimulated overnight with overlapping peptides representing the full-length sequence of the vaccine-encoded RBD.

Of the 36 subjects, 34 (94.4%, including all subjects treated with. gtoreq.10. mu.g BNT162b 1) initiated RBD-specific CD4+T cell response. To varying degrees (magnitude) between individuals, the strongest CD 4T cell response was more than 10-fold the memory response observed in the same subject for a panel of Cytomegalovirus (CMV), Epstein Barr Virus (EBV), influenza virus, and tetanus toxoid-derived immunodominant (dominant) peptides (fig. 41 a-c). At baseline, CD4+T cell responses were undetectable except one subject had a small number of pre-existing RBD-reactive CD4+T cells, which increased significantly after vaccination (normalized mean plaque counts from 63 to 1,519 in the 50 μ g dose cohort). RBD-specific CD4 +The intensity of the T cell response was positively correlated with both IgG binding to RBD and SARS-CoV-2 neutralizing antibody titers (fig. 41d, fig. 46a), consistent with the concept of intramolecular help (intramolecular help) (set, a.et al, Immunity 28,847-58 (2008)). Lack of CD4+VNT of two subjects responding50Neither titer was detectable (fig. 41 d).

Vaccine induced CD8+T cell responses, most subjects mount some strong responses (29/36, 80.6%) (fig. 41a) and are comparable to memory responses against CMV, EBV, influenza virus and tetanus toxoid in the same subjects (fig. 41b, c). RBD-specific CD8+The intensity of the T cell response and vaccine induced CD4+T cell responses were positively correlated, but not significantly correlated with SARS-CoV-2 neutralizing antibody titers (FIGS. 46b, c).

Notably, although the immunogenicity rate at 1 μ g BNT162b1 was lower (6/8 responsive)Subject), vaccine-induced CD4 in some subjects+And CD8+The extent of T cells was almost as high as 50 μ g BNT162b1 (fig. 41 a). To assess the functionality and polarization of RBD-specific T cells, cytokines secreted in response to vaccine antigen stimulation were determined by intracellular staining (ICS) with IFN γ, IL-2 and IL-4 specific antibodies in PBMCs of 18 BNT162b1 immunized subjects before and after vaccination. RBD-specific CD4 +T cells secreted IFN γ, IL-2, or both, but not IL-4 (FIGS. 42 a-c). Similarly, a portion of the RBD-specific IFN γ+ CD8+T cells also secrete IL-2.

The average fraction of RBD-specific T cells in total circulating T cells obtained by BNT162b1 vaccination was significantly higher than that observed in six subjects recovered from COVID-19. RBD specific IFN gamma+CD8+The frequency (frequency) of T cells reached total peripheral blood CD8+Several percent of T cells (fig. 42 c). Analysis of the supernatant of PBMCs from a sub-population of 5 vaccinated subjects stimulated ex vivo with overlapping RBD peptides showed associated (cognate) release of the proinflammatory cytokines TNF, IL-1 β and IL-12 p70 (fig. 42 d).

Taken together, these findings indicate that BNT162b1 induces functional and pro-inflammatory CD4 in almost all subjects+/CD8+T cell response with TH1 polarization assisted (helper) response.

Discussion of the related Art

We observed concurrent production of neutralizing antibodies, virus-specific CD4+And CD8+Activation of T cells and robust release of immune regulatory cytokines, such as IFN γ, represent a synergistic immune response against viral invasion (for review see Vabret, n.et al, Immunity 52, 910-. IFN γ represents a critical cytokine for several antiviral responses. Indeed, patients with IFN γ gene polymorphisms associated with impaired IFN γ activity are shown to exhibit a 5-fold increase in susceptibility to SARS (Chong, w.p.et al, BMC infection.dis.6, 82 (2006)). In addition, IFN γ acts in concert with type I interferons to inhibit the replication of SARS-CoV-2 (Sainz, B., et al., Virology 329, 11- 7(2004)). IFN gamma from CD8+Robust production of T cells suggests a favorable immune response with antiviral and immune-augmenting (immune-augmenting) properties.

Importantly, the detection of IFN gamma, IL-2 and IL-12p70 but not IL-4 indicates a favorable TH1 spectrum and no potentially harmful TH2 immune response. CD4 as suggested in SARS-CoV-1 survivors+And CD8+T cells may confer long-term immunity against coronaviruses, of which CD8+T cell Immunity persists for 6-11 years (Vabret, N.et al., Immunity 52, 910-941 (2020); Ng, O. -W.et al., Vaccine 34,2008-14 (2016)).

Some examples of asymptomatic viral exposure are associated with a cellular immune response without seroconversion, indicating that SARS-Cov-2 specific T cells may be involved in disease control even in the absence of neutralizing antibodies (Gallis, F. et al (2020). doi: medRxiv: 10.1101/2020.06.21.20132449). Almost all vaccinated volunteers initiated RBD-specific T cell responses detected with an ex vivo ELISpot assay, which was performed without prior expansion of T cells, capturing only high intensity (magnitude) T cell responses. Although the intensity of the T cell response varied widely between subjects, we did not observe a significant dose dependence of the intensity of the T cell response in the dose range 1 μ g to 50 μ g, indicating that stimulation and robust expansion of T cells could be accomplished at the lowest mRNA-encoded immunogen level.

Studies demonstrated dose-dependence of IgG binding to RBD and neutralization response, reproducing our previous findings in the us assay at 10 and 30 μ g dose levels, and showing further increases in neutralizing antibody titers by a prime/boost regimen at 50 μ g.

Notably, two BNT162b1 injections at dose levels as low as 1 μ g were able to induce higher levels of IgG binding to RBD than those observed in convalescent sera and also increased serum neutralizing antibody titers by day 43. Given that the extent of protective neutralizing antibody titers is not known, and given the apparent T cell responses we observed in some subjects in the 1 μ g cohort, it means that a significant fraction of individuals can benefit from this lowest tested dose level.

It is believed that pure RBD-directed immunity is susceptible to viral escape through single amino acid changes in this small domain. However, neutralization of 17 pseudotyped viruses alleviates this potential concern, of which 16 use spike entry cells with different RBD variants found in the circulating strains, and one uses the dominant spike variant D614G.

Example 8: summary of safety and immunogenicity data from the COVID-19 vaccine BNT 1621 stage

This example provides additional safety and immunogenicity data for BNT162b1 and BNT162b2 vaccine candidates. These safety and tolerability data, as well as immunoglobulin g (igg) binding and severe acute respiratory syndrome coronavirus 2(SARS-CoV-2) neutralization titer data, were derived from the american participants in phase 1 U.S. studies of these vaccine candidates.

For BNT162b1, the following was observed:

for dose levels of 10 μ g to 30 μ g, reactogenicity (particularly systemic events) increased with increasing dose levels in participants aged 18-55 and 65-85 years. Reactogenicity (particularly systemic events) increased after dose 2 compared to dose 1.

For BNT162b2, the following was observed:

dose levels and dose number dependent increases in reactogenicity were minimal to mild (modest) for any age group. Based on all available data, the reactogenicity profile observed for BNT162b2 (and in particular SEQ ID NO:20) was quite favorable.

The immunogenicity data presented herein, focusing on SARS-CoV-2 neutralization response after dose 2, allows the following conclusions to be drawn:

for BNT162b1, on day 28 (7 days after dose 2):

the neutralizing antibody response elicited after immunization with doses of 10 μ g and 30 μ g in adults 18-55 years of age (where data is available in both age groups) was higher compared to 65-85 years of age. Neutralizing antibody responses were similar after 20 μ g and 30 μ g doses in the 65-85 year old group, although values were higher at the 20 μ g dose level.

For BNT162b2 (in particular, SEQ ID NO: 20), on day 28 (7 days after dose 2):

neutralizing antibody responses (data available in both age groups) were higher after a 20 μ g dose in the 18-55 year old group compared to the 65-85 year old group. Neutralizing antibody responses were higher after receiving 20 μ g compared to the 10 μ g dose level in the 18-55 year old group. The S1 IgG bound antibody data in figure 56, and responses after comparing dose 1 across dose levels, were highest at the 30 μ g dose level, suggesting that the neutralizing antibody level may be higher for the 30 μ g dose level after dose 2, since the level of bound antibody is very correlated with the neutralizing antibody level. In the 65-85 year old group, neutralizing antibody responses after 20 μ g and 30 μ g doses were higher at the 30 μ g dose level.

The data collectively show that the neutralizing antibody responses of BNT162b1 and BNT162b2 after dose 2 are similar.

Safety and tolerability of BNT162b1

Age group of 18-55 years old

All dose levels from dose 2 were available in this age group, except 20 μ g, which is currently part of the data available from dose 2 (and 100 μ g, where no second dose was administered under the recommendations of the internal examination committee (IRC)). The partial reaction is shown in FIG. 48. The systemic events are shown in figure 49.

Immunogenicity of BNT162b1

Age group 65-85 years old

All dose levels in this age group were available from immunogenicity data after dose 2. The Geometric Mean Concentration (GMC) of IgG bound to RBD is shown in FIG. 50. SARS-CoV-2 neutralization Geometric Mean Titer (GMT) is shown in FIG. 51.

Safety and tolerability of BNT162b2

Age group of 18-55 years old

All dose levels from dose 2 safety data were available for this age group. The partial reaction is shown in FIG. 52. The systemic events are shown in figure 53.

Age group 65-85 years old

All dose levels from the post-dose 2 safety data are available for this age group, however, the 10 μ g dose level data is only partial. The partial reaction is shown in FIG. 54. The systemic events are shown in figure 55.

Immunogenicity of BNT162b2

Age group of 18-55 years old

The age group's 30 μ g dose level was available from the immunogenicity data after dose 1, and the 10 μ g and 20 μ g dose levels after dose 2. IgG GMC binding to S1 is shown in FIG. 56. SARS-CoV-2 neutralization of GMT is shown in FIG. 57.

Age group 65-85 years old

The 20 μ g and 30 μ g dose levels for this age group were available from immunogenicity data after dose 2. IgG GMC binding to S1 is shown in FIG. 58. SARS-CoV-2 neutralization of GMT is shown in FIG. 59.

Conclusion

The local tolerance profiles of BNT162b1 and BNT162b2 (and in particular SEQ ID NO:20) and the immune response data of 2 candidates were similar. BNT162b2 (particularly SEQ ID NO:20) can show a favorable systemic reactogenicity profile (particularly in the 65-85 year old group). When selecting a dose level of BNT162b2 (particularly SEQ ID NO:20), the level of SARS-CoV-2 neutralizing antibody response in the 65-85 year old group may have a significant impact on maximizing neutralizing antibody response in this age group with the highest risk of severe disease. Comparing the neutralizing antibody levels in the 20 μ g and 30 μ g annual adult cohorts in this study, the 30 μ g dose level showed a higher level of neutralizing antibody than the 20 μ g cohort (fig. 59). GMT at the 30 μ g dose level was 1.6 times that of HCS compared to the neutralizing antibody level of the Human Convalescent Serogroup (HCS) with GMT 94; the GMT at the 20 μ g dose level was 0.9 times that of HCS. Thus, both showed neutralizing antibody titers at least comparable to the human convalescent serogroup. At least 14 days after PCR confirmed diagnosis, and when participants were asymptomatic, 38 human SARS-CoV-2 infection/COVID-19 convalescent sera were drawn from participants aged 18-83. Serum donors were mainly symptomatic (35/38) and one had been hospitalized. Serum was obtained from Sanguine Biosciences (Sherman Oaks, Calif.), MT Group (Van Nuys, Calif.) and Pfizer Occupical Health and Wellness (Pearl River, NY). In addition, the S1-IgG antibody binding concentration in the older (fig. 58, post dose 2) and younger (fig. 56, post dose 1) adult cohorts also biased towards selecting a 30 μ g dose level. Preliminary human T cell data being generated in german experiments using BNT162b2 demonstrated robust CD4+ and CD8+ expected for the RNA platform. In view of this, it was proposed to use BNT162b2 (in particular SEQ ID NO:20) at a dose level of 30 μ g for phase 2b/3, as this dose and construct provides the best combination of an advantageous reactogenicity profile and a robust immune response, possibly providing protection against covi-19 in young and older adults.

Example 9: immunology of COVID-19 vaccine BNT162

To support entry into 2/3 phase in 18-85 year old adults, non-clinical and clinical data are provided herein, summarizing T cell responses in mice following BNT162b2 immunization, as well as in humans involved in BNT162 trials. The following immunogenicity data are provided:

1. preliminary and unapproved mouse immunogenicity data: IFN γ ELISpot (fig. 60), intracellular cytokine staining and Luminex quantification of cytokines produced after BNT162b2 immunization.

2. From German test (BNT 162-01): BNT162b2 at a dose level of 10 μ g IFN γ ELISpot in participants 18-55 years old before the first dose and 7 days after dose 2 (fig. 61, 62, 63).

T cell response to BNT162b2 in mice

On day 0, four groups of eight female BALB/c mice were immunized with a dose of 0.2. mu.g, 1. mu.g or 5. mu.g BNT162b2 (in particular SEQ ID NO: 20) per animal, by Intramuscular (IM) injection, or with buffer only (control group). On days 12 and 28, spleens were harvested for splenocyte isolation and T cell response analysis using the IFN γ ELISpot assay. The Luminex assay and Intracellular Cytokine Staining (ICS) were performed to assess cytokine responses. At 12 and 28 days post-immunization, a majority of splenocytes of CD4+ and CD8+ T cell phenotypes were isolated from BNT162b2 immunized mice and, when restimulated ex vivo with the full-length S peptide mixture, produced strong antigen-specific IFN γ and IL-2 responses in ELISpot and flow cytometry assays (fig. 60a and b). Collected and used on day 28 Full-length S-peptide pool stimulated splenocytes produced high levels of TH1 cytokines IL-2 and IFN gamma, with corresponding minimum levels of T in multiplex immunoassaysH2 cytokines IL-4, IL-5 and IL-13 (FIG. 60 c).

T cell response in humans of BNT162b2 from germany studies

To assess the T cell phenotype elicited by immunization of humans with BNT162b2 (in particular SEQ ID NO:20), IFN γ ELISpot was performed on Peripheral Blood Mononuclear Cells (PBMCs) obtained from participants in the German study.

IFNγELISpot

Vaccine-elicited T cell responses were determined using CD4 or CD8 depleted PBMCs obtained from subjects before dose 1 and on day 29 (7 days after dose 2). IFN γ ELISpot data were generated on days 1 and 22 for 5 subjects immunized with 10 μ g BNT162b2 (in particular SEQ ID NO: 20). Spike-specific ex vivo CD4+ and CD8+ T cell responses were detected in 5/5 (100%), respectively, subjects post-vaccination. In the pre-vaccination samples, all responses were either very low or undetectable. The response was considered vaccine-induced (fig. 61, 62, 63).

The BNT162b2 vaccine stimulated by S peptide pool 1 (N-terminal part of the spike, which includes the receptor binding domain [ RBD ]) and S peptide pool 2 (C-terminal part of the spike) elicited antigen-specific CD8+ and CD4+ T cell responses comparable to or higher than memory responses against CMV, EBV, influenza virus and tetanus toxoid in the same subject (fig. 63).

Conclusion

These data for BNT162b2 vaccine candidates confirm results previously obtained in preclinical models and humans for modRNA (nucleoside modified) platform immunization. The data indicate that modRNA elicits significant Th 1-type CD4+ and CD8+ T cell responses.

Example 10: trimeric SARS-CoV-2 receptor binding domain RNA vaccines are highly immunogenic and protective in non-human primates

Here we report the design and non-clinical progress of BNT162b1 vaccine candidates. We demonstrate that nucleoside modifications encode structurally stable, trimerized SARS-CoV-2 receptor junctionsDomains (RBD) mRNA encapsulated in Lipid Nanoparticles (LNPs) for efficient intramuscular delivery in mice eliciting potent antibodies and TH1 dominant cellular immune response. Immunization of mice with a single dose of BNT162b1 elicited a significant dose-level dependent increase in the neutralizing titer of pseudovirus, as well as a strong IFN γ -positive CD4+And CD8+T cell response. Vaccination of rhesus monkeys with the BNT162b1 prime-boost vaccine elicited a reliable (authetic) neutralization geometric mean titer of SARS-CoV-2 that was 2.6-6.0 times that of the SARS-CoV-2 convalescent human serum group. Following the challenge of SARS-CoV-2 infection, the immunized rhesus monkey either had no viral RNA present in the nose and lungs or was more transient than the presence of the non-immunized control rhesus monkey viral RNA.

Materials and methods

Statement of ethics

All mouse studies were conducted in BioNTech SE and the protocol was approved by the local authorities (local Welfare Committee), conducted according to FELASA recommendations, and met German Animal Welfare Act and Instructions 2010/63/EU (Directive 2010/63/EU). Only animals with unconventional health conditions were selected for the test procedure.

Immunization for non-human primate (NHP) studies was performed at the University of Louisiana at Lafayette-New Iberia Research Center (NIRC), which has received approval from the Association for Association and authorization of Laboratory Animal health Care, AAC, Animal Association #: 000452). This work is in line with the guidelines of the USDA Animal Welfare Act and Regulations and NIH for studies involving recombinant DNA molecules and biosafety in microbiology and biomedical laboratories. All procedures performed on these animals were in compliance with regulatory and established guidelines and were reviewed and approved by the international animal care and use committee or by ethical review procedures. Infectious SARS-CoV-2 challenge for NHP studies was performed at the south-west National Primate Research Center (south west National primary Research Center). The Animal husbandry (Animal husbandry) follows the international AAALAC recommended standards and NIH guidelines for Laboratory Animal Care and Use (Care of Use of Laboratory Animals). This study was approved by the Texas Biomedical Research Institute Animal Care and Use Committee.

Protein and peptide reagents

Purified recombinant SARS-CoV-2 RBD fused to the constant region of mouse IgG1 was used as a target for western blotting, while recombinant SARS-CoV-2 RBD labeled with human Fc tag (both from Sino Biological) was used in ELISA to detect SARS-CoV-2S specific IgG. Purified recombinant rbd (nano biological) with histidine tag was used for Surface Plasmon Resonance (SPR) spectroscopy. Overlapping 15-membered peptide pools of S proteins were used for ELISpot, cytokine analysis and intracellular cytokine staining. A peptide control (SPSYVYHQF, derived from gp70 AH-1 (Slansky, J.E.et al., Immunity 13,529-538,2000)) was used as a control for the ELISpot assay. All peptides were obtained from JPT peptide Technologies.

Human convalescent serum

At least 14 days after PCR confirmed diagnosis, and when participants were asymptomatic, human COVID-19 convalescent sera were drawn from donors aged 18-83 (n-38). Serum donors were symptomatic infected (35/38) or hospitalized (1/38). Serum was obtained from Sanguine Biosciences (Sherman Oaks, Calif.), MT Group (Van Nuys, Calif.) and Pfizer Occupical Health and Wellness (Pearl River, NY).

Cell culture

Human Embryonic Kidney (HEK)293T/17 and Vero-76 cells (both from ATCC) were cultured with GlutaMAX supplemented with 10% fetal bovine serum (Sigma-Aldrich) TM(Gibco) in Dulbecco's Modified Eagle's Medium (DMEM). After receiving, the cell lines were tested for mycoplasma contamination prior to expansion and cryopreservation. Vero E6 and Vero CCL81 (both from ATCC) cells were cultured in DMEM (Gibco) containing 2% HyClone fetal bovine serum and 100U/mL penicillin/streptomycin (Gibco). Expi293FTMCells were grown at Expi293TMIn culture and using ExpifeacamineTM293 (all from Thermo Fisher Scientific).

Production of in vitro transcribed RNA

To generate a template for RNA synthesis, a DNA fragment encoding the fusion protein is cloned togetherIn a starting plasmid vector having backbone sequence elements (Orlandini von Niessen, A.G.et al, Mol Ther 27, 824-. The non-coding backbone element included a region from the T7 promoter to the 5 'and 3' UTR plus poly (a) tail (100 nucleotides), interrupted by a linker (a30LA70, 10 nucleotides). DNA was purified, spectrophotometrically quantified and analyzed for the presence of the trinucleotide cap1 analog ((m) 2 7,3’-O)Gppp(m2’-O) ApG, respectively; TriLink) and N in place of uridine-5' -triphosphate (UTP)1In vitro transcription with T7 RNA polymerase in the presence of methylpseuduridine-5' -triphosphate (m1 Ψ TP; Thermo Fisher Scientific) (Grudzien-Nogalska, E.et al, Methods in molecular biology (Clifton, N.J.)969, 55-72,2013). RNA was purified using magnetic particles (Berensmeier, s., appl. microbiol.biotechnol.73, 495-504,2006), integrity was assessed by microfluidic capillary electrophoresis (Agilent Fragment Analyser) and concentration, pH, osmolarity (osmolyte), endotoxin level and bioburden (bioburden) were determined.

Lipid nanoparticle formulations of RNA

Purified RNA was formulated into LNP using an ethanolic lipid cocktail of ionizable cationic lipids and transferred to an aqueous buffer system by diafiltration to produce an LNP composition similar to that previously described (Maier, M.A. et al, Molecular Therapy: the journal of the American Society of Gene Therapy 21, 1570-1578, 2013). BNT162b1 was stored at-70 ℃ at a concentration of 0.5 mg/mL.

mRNA transfection

HEK293T/17 cells were transfected with BNT162b1 RNA or BNT162b1 mixed with transfection reagents by incubation for 18 hours. non-LNP formulated mRNA (1. mu.g for Western blotting and flow cytometry, 2.5. mu.g for immunofluorescence) was diluted in Opti-MEM medium (Thermo Fisher Scientific) and mixed with transfection reagents according to the manufacturer's instructions (RiboJuice, Merck Millipore).

Western blot analysis

Lysates of HEK293T/17 cells transfected with BNT162b1 RNA were analyzed by denaturing SDS-PAGE and protein blotting on a 10% Mini-Protean TGX pre-polyacrylamide gel (Bio-Rad). The Transfer to a nitrocellulose membrane (Carl Roth) was carried out using a semidry Transfer System (Trans-Blot Turbo Transfer System, Bio-Rad). Blotted proteins were detected with rabbit polyclonal primary antibody primed with recombinant S1 fragment of SARS-CoV S (SinoBiological) and horseradish peroxidase conjugated anti-rabbit secondary antibody (Sigma Aldrich). The stamp was developed with SuperSignal West Femto chemiluminescent substrate (Thermo Fisher Scientific) and imaged with Bio-Rad ChemiDoc system with Image Lab software version 5.0.

Immunofluorescence

Transfected HEK293T/17 cells were fixed in 4% Paraformaldehyde (PFA) and permeabilized in Phosphate Buffered Saline (PBS)/0.2% Triton X-100. The free binding sites were blocked and the cells were incubated with rabbit polyclonal antibodies and anti-rabbit IgG secondary antibodies (Jackson ImmunoResearch) recognizing the S1 subunit (SinoBiological) or labeled concanavalin a (invitrogen). DNA was stained with hoechst (Life technologies). Images were taken with a Leica SP8 confocal microscope.

Flow cytometry

Transfected HEK293T/17 cells were stained with an immobilizable viability dye (eBioscience). After Fixation (fire Buffer, Biolegend), cells were permeabilized (Perm Buffer, eBioscience) and stained with monoclonal SARS-CoV-2 spike S1 antibody (SinoBiological). Cells were harvested on a facscan II flow cytometer (BD Biosciences) using BD FACSDiva software version 8.0.1 and analyzed by FlowJo software version 10.6.2(FlowJo LLC, BD Biosciences).

For mouse T cell analysis in peripheral blood, red blood cells from 50 μ L freshly drawn blood were lysed (ACK lysing buffer, Gibco) and stained with fixable vital dye (eBioscience) and primary anti-stain cells in flow buffer (DPBS [ Gibco ] supplemented with 2% FCS, 2mM EDTA [ both from Sigma ] and 0.01% sodium azide [ morphine ]) in the presence of Fc blocking solution. After staining with biotin-conjugated secondary antibodies in flow Buffer, extracellular staining of cell surface markers was performed with directly labeled antibodies and streptavidin in Brilliant Stain Buffer Plus (BD Bioscience) diluted in flow Buffer. Cells were fixed with 2% RotiHistofix (Carl Roth) and then permeabilized (Perm buffer, FoxP 3/transcription factor staining buffer set, eBioscience) overnight. Permeabilized cells were treated intracellularly with Fc blocking solution and stained with antibodies to transcription factors in Perm buffer.

For mouse T cell analysis in lymphoid tissues, 1X 10 pairs of directly labeled antibodies were used6Lymph node cells and 4X 106Splenocytes were subjected to viability and extracellular antigen staining. Cells were washed with 2% RotiHistofix and then fixed (Fix/Perm buffer, FoxP 3/transcription factor staining buffer set, eBioscience) overnight. The intracellular staining method was performed according to the staining of blood T cells described above. For the analysis of B-cell subtypes in lymphoid tissues in mice, 2.5X 10 Fc blocking fluid was used for treatment5Lymph node cells and 1X 106Splenocytes, stained for viability and extracellular antigen as per the staining of blood T cells described above, then fixed with 2% RotiHistofix overnight. For intracellular cytokine staining in mouse T cells, 1X 10 mixtures of full-length S-peptides with a final concentration of 0.2. mu.g/mL per peptide were paired in the presence of GolgiStop and GolgiPlug (both BD bioscience)6Lymph nodes and 4X 106Splenocytes were stimulated for an additional 5 hours ex vivo. Cells were subjected to viability and extracellular antigen staining as described above for lymphoid T-cell staining. Cells were fixed with 2% RotiHistofix and then permeabilized overnight. Intracellular staining was performed according to the blood T cell staining method described above.

Mouse cells were obtained on a BD Symphony A3 or BD Celesta (B-cell subtype) flow cytometer (BD Biosciences) using BD FACSDiva software version 9.1 or 8.0.1.1, respectively, and analyzed with FlowJo 10.6 (FlowJo LLC, BD Biosciences).

Expression and purification of proteins

To express the RBD-folder encoded by BNT162b1 for biochemical and structural analysis, DNA corresponding to the RNA coding sequence was cloned into the pMCG1309 vector. Generates a code having a C-terminal His-10 and Avi-tagged plasmids of amino acids 1-615 of human ACE2 for transient expression of the ACE2 peptidase domain (ACE2 PD) in Expi293F cells. ACE2/B production by co-expression of two plasmids in Expi293F cells0AT1 complex, wherein one plasmid encodes ACE2 amino acids 1-17 followed by hemagglutinin tag, Strep II tag, and ACE2 amino acids 18-805, and the other contains methionine followed by FLAG tag and human B0Amino acids 2-634 of AT 1.

Secreted ACE2 PD was isolated from conditioned cell culture media using Nickel Excel resin (GE Healthcare) followed by gel filtration chromatography on a Superdex 20010/30 chromatography column (GE Healthcare) in PBS. 4% agarose beads per 1mL were covalently attached to approximately 5mg of purified ACE2 PD by amine coupling using AminoLink Plus resin (Thermo Fisher Scientific). RBD-trimer was purified from conditioned media by affinity capture with ACE2 PD cross-linked agarose and 3M MgCl2Eluting from the resin. After dialysis, concentration and purification of the protein was performed by gel filtration in HEPES Buffered Saline (HBS) with 10% glycerol using a Superdex 20010/300 chromatography column. The ACE2/B0AT1 complex was purified based on the method described previously (Yan, R.et al., Science (New York, N.Y.)367, 1444-. To form ACE2/B 0AT 1/RBD-trimer complex, ACE2/B0AT1 aliquots were diluted in ACE2/B0Purified RBD-foldon pool in AT1 size exclusion chromatography buffer (25mM Tris pH 8.0,150mM NaCl, 0.02% glyco diosgenin), molar ratio of RBD-trimer to ACE2 precursor 3: 1. after incubation at 4 ℃ for 30 min, the samples were concentrated and separated on a Superose 6 Increate 10/300 GL column. The peak fractions containing the complex were combined and concentrated.

Surface plasmon resonance spectroscopy

The binding kinetics of murine RBD-specific serum IgG was determined at 25 ℃ using a Biacore T200 instrument (Cytiva) with HBS-EP running buffer (BR100669, Cytiva). The carboxyl groups on the CM5 sensor chip matrix were activated by a mixture of 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) to form an active ester for reaction with amine groups. Anti-mouse Fc antibody (Jackson ImmunoResearch) was diluted in 10 mM sodium acetate buffer pH 5 (30 μ g/mL) for covalent coupling to an immobilization level of-10,000 Response Units (RU). The free N-hydroxysuccinimide ester on the sensor surface is deactivated with ethanolamine.

Mouse serum was diluted 1:50 in HBS-EP buffer and applied 10. mu.L/min to an active flow cell (flow cell) for 30 seconds to be captured by the immobilized antibody, while the reference flow cell was treated with buffer. The binding between the captured murine IgG antibody and RBD-His (nano Biological Inc.) was analyzed using a multi-cycle kinetic method at a concentration range of 1.5625 to 50 nM. The 180 second association period was followed by a 600 second dissociation period at a constant flow rate of 40 μ L/min, and a final regeneration step. Binding kinetics were calculated using a global kinetics fitting model (1:1 Langmuir, Biacore T200 Evaluation Software Version 3.1, Cytiva).

Interference of biological layer

Binding of the RBD-folder to ACE2-PD was measured by biolayer interferometry on Octet RED384 (Forte Bio) at 25 ℃ in a buffer containing 10mM HEPES pH 7.5, 150mM NaCl and 1mM EDTA. Human ACE2 PD with Avi tag was immobilized on streptavidin coated sensors. Binding data were collected for a series of concentrations of RBD-trimer, association phase 10 min, dissociation phase 15 min. Using Octet data analysis software v10.0 (Fortuer Bio), the data were subtracted by a reference value and fitted to R21 with a value greater than 0.96: 1 binding the model to determine the kinetics and affinity of binding. Dissociation rate (k) of interactiond) Below the measurement limit of the instrument and using the assumed dissociation rate kd 1×10-6s-1Estimation of binding affinity (K)D)。

Electron microscopy of negatively stained samples

mu.L of purified RBD-trimer protein was applied to a glow-discharge (glow-discharge) copper grid covered with Formvar and amorphous carbon (Ted Pella). According to the manufacturer's method, staining was performed using a Nano-W organotungstate (nanotungstate) stain (Nanoprobes), and the sample was imaged using a FEI TF-20 microscope run at 200k, with a magnification of 62000X and a defocus of-2.5 μm. The micrographs were corrected for Contrast Transfer Function (CTF) in RELION using CTFFIND-4.1 (Rohou, A. & Grigaroeff, N., Journal of structural biology 192, 216-. A 2D reference is generated with a small set of manually selected data for automatic selection. The resulting particles were collected in a RELION 3.0.6 for 2D classification (Zivanov, J.et al., eLife 7; 10.7554/eLife.42166 (2018).

Cryoelectron microscopy

mu.L of 6mg/mL purified ACE2/B0The AT 1/RBD-trimer complex was applied to a gold Quantifoil R1.2/1.3200 mesh grid and discharged with a Pelco Easilow glow discharge AT 20mA for 30 seconds in a thin gas (residual air). The samples were imprinted with Vitrobot Mark IV for 5 seconds at a force of-3 and then immersed in liquid ethane cooled by liquid nitrogen. A total of 7455 micrographs were acquired in super-resolution mode on a single grid of Titan Krios operating at 300keV, equipped with a Gatan K2 Summit direct electron detector, with a magnification of 165000x and a pixel size of large at the sample level ofTotal electron amountDivided into 40 frames and exposed for 6 secondsData were collected over a defocus range of-1.2 to-3.4 μm. Initial motion correction was performed in Warp (Tegunov, D.&Cramer, p., Nature methods 16, 1146-1152, 2019), during which the super-resolution data is binned to generateThe pixel size of (2). The corrected micrograph was imported into RELION 3.1-beta (Zivanov, J.et al., eLife 7; 10.7554/eLife.42166(2018)), and CTF (Rohou, A) was estimated using CTFFIND-4.1.&Gridiroreff, N.A., Journal of structural biology 192, 216-221, 2015). Use in RELIONThe existing LaPlacian-of-Gaussian particle selection algorithm selects particles and extracts the particles with a frame size of 450 pixels. The reference obtained by 2D classification was used for a second round of auto-selection based on the reference, resulting in a data set of 715,356 particles. Particle heterogeneity was filtered out with 2D and 3D sorting, mask size 280nm to filter out non-ACE 2 bound RBD copies per RBD-trimer, yielding a set of 87,487 particles optimized to C2 symmetry Subtracting micelle and B from particle0Optimization after AT1 Density, yieldImprovement of the figure. Rigid body fitting of an atomic model (Yan, R.et. al., Science (New York, N.Y.)367, 1444-1448, 2020) of PDB ID 6M17 toDensity, then manually constructed in Coot (Emsley, p.et., Acta crystalline medical section D, Biological crystalline medical section 66, 486-. Image acquisition was performed using the SerialEM software version 3.8.0 beta operating microscope (Mastronard, D.N., Journal of structural biology 152, 36-51, 2005). Biolayer interference Data was collected using Octet Data Acquisition software version 10.0.0.87 and processed with ForteBio Data Analysis software version 10.0.

Immunization

A mouse. Female BALB/c mice (Janvier; 8-12 weeks) were randomly grouped. BNT162b1 was diluted in PBS, 300mM sucrose or saline (0.9% NaCl) and injected IM into the calf muscle under isoflurane anesthesia in a volume of 20 μ Ι _.

Rhesus monkey (macaque). On days 0 and 21, male rhesus monkeys (2-4 years old) were randomly assigned to receive a volume of 0.5mL of BNT162b1 or saline placebo control administered by IM injection into the left quadriceps muscle. Serum and PBMCs were collected following the animal protocols 2017-8725-023 approved by the NIRC animal care and use committee. During blood sampling and immunization, animals were anesthetized with ketamine hydrochloride (10mg/kg, IM) and monitored closely for sedation.

SARS-CoV-2 challenge in rhesus monkeys

SARS-CoV-2 inoculum was prepared from 2.1X 10 previously prepared in the Texas Biomedical Research Institute (San Antonio, Texas)6PFU/mL stock was obtained, dispensed into single use vials, and stored at-70 ℃. Working virus stocks were generated from SARS-CoV-2USA-WA1/2020 isolate (4 th generation seed stock from BEI Resources; NR-52281) passaged twice in Vero E6 cells. The virus was confirmed to be SARS-CoV-2 by deep sequencing and to be identical to the published sequence (GenBank accession No. MN 985325.1).

Male rhesus monkeys (control) immunized with BNT162b1 (n ═ 6) and age-matched saline control (n ═ 6) were used at 1 × 106The plaque-forming unit SARS-CoV-2USA-WA1/2020 isolate WAs challenged by equally partitioning between the intranasal (IN, 0.2mL) and intratracheal (IT, 0.2mL) pathways as described previously (Singh, D.K.et., SARS-CoV-2infection leads to access infection with dynamic cells and infection fluorine IN the lung of non-human mammalian species, 2020). The challenge is carried out 41-48 days after the second immunization. Naive groups alone, uninmmunized age and gender matched animals (n ═ 3) received only DMEM supplemented with 10% FCS IN (0.2mL) and IT (0.2 mL). Approximately two weeks prior to challenge, animals were transferred to the animal biosafety level 3(ABSL-3) facility of the southwest national center for primate animal research (SNPRC; San Antonio, TX). The rectal temperature, body weight and physical examination of the animals were monitored periodically by a veterinarian clinician certified by the committee. Samples were collected under teletazolazepam (Telazol) anesthesia as described (Singh, D.K.et al, SARS-CoV-2 introduction leads to access introduction with dynamic cells and introduction fluorine in the long same ways as the sources of the vaccine drugs, 2020). Nasal swabs were collected from rhesus monkeys on days 0, 1, 3, and 6 post-inoculation to assess viral titers. At the top Bronchoalveolar lavage (BAL) was performed four times by instillation of 20mL saline one week before the battle and on days 3 and 6 after vaccination. The washes were combined, aliquoted and stored frozen at-70 ℃.

Quantitative reverse transcription polymerase chain reaction

For detection and quantification of SARS-CoV-2, viral RNA was extracted from nasal swabs and BAL samples as previously described (Mehra, S.et., The Journal of infectious diseases 207, 1115-1127, 2013; Gautam, U.S.et., Proceedings of The National Academy of science of The United States of America 115, E62-E71; 2018; Josten, S.A.et., PLoS Pathologens 6, E1000782,2010) and RT-qPCR assays (Single, D.K.et., SARS-CoV-2infection to nucleic acid infection with a nasal tissue and tissue infection, tissue culture 2020). Briefly, 10. mu.g of yeast tRNA and 1X 103PFU MS2 phage (E.coli phage MS2, ATCC) was added to each thawed sample and RNA was extracted using the NucleoMag Pathologen kit (Macherey-Nagel). SARS-CoV-2RT-qPCR was performed on the extracted RNA using a 2019-nCoV _ N1 assay developed by CDC on a QuantStaudio 3 instrument (Applied Biosystems). The positive cutoff (detection limit, LOD) was set at 10 Gene Equivalents (GE) per reaction (800 GE/mL). Samples were tested in duplicate. By repeated measurements, on day 6, one BAL sample from the control group and one nasal swab from day 1 from the BNT162b1 immunized group had viral RNA levels on each side of the LLOD. These samples were classified as indeterminate and excluded from the graphs and analysis.

Preparation of tissue

A mouse. Peripheral blood was collected from the retroorbital venous plexus or facial vein under isoflurane anesthesia. After centrifugation of blood at 16.000x g for 5 minutes, the serum was used immediately for downstream measurements or stored at-20 ℃. Splenic single cell suspensions were prepared in PBS by triturating the tissue with the plunger of a 3mL syringe (BD Biosciences) on the surface of a 70 μm cell filter (BD Falcon). Hypotonic lysis removes red blood cells. The popliteal, inguinal, and iliac lymph nodes were pooled, cut into small pieces, digested with collagenase D (1 mg/mL; Roche), and passed through a cell filter.

Rhesus monkey (macaque). Serum and PBMCs were collected following the Animal protocol 2017-.

IgG antibody assay for binding to RBD

For mouse sera, MaxiSorp plates (Thermo Fisher Scientific) were coated with recombinant RBD (100ng/100 μ L) in sodium carbonate buffer and bound IgG was detected using HRP-conjugated secondary antibody and TMB substrate (Biotrend). Data collection was performed with a BioTek Epoch reader and Gen5 software version 3.0.9. For concentration analysis, the signal for a particular sample is correlated with a standard curve for an isotype control. For rhesus monkeys and human serum, will contain a C-terminal Avitag TMRecombinant SARS-CoV-2 RBD from (Acro Biosystems) was bound to streptavidin-coated Luminex microspheres. Bound rhesus monkey or human anti-RBD antibodies present in the serum were detected with a fluorescently labeled goat anti-human polyclonal secondary antibody (Jackson ImmunoResearch). Data were captured as Median Fluorescence Intensity (MFI) using the Bioplex200 system (Bio-Rad) and converted to U/mL antibody concentrations using a reference standard curve consisting of 5 pooled human COVID-19 convalescent serum samples (PCR diagnostics)>Obtained 14 days), diluted in antibody-depleted human serum, arbitrarily assigned a concentration of 100U/mL, and taking into account serum dilution factors.

VSV-SARS-CoV-2 spike variant pseudovirus neutralization

Replication-defective recombinant Vesicular Stomatitis Virus (VSV) vectors, which encode GFP rather than VSV-G (VSV. DELTA.G-GFP), were pseudotyped with SARS-CoV-2S protein according to the published pseudotyping method (Berger Rentsch, M. & Zimmer, G., PLoS ONE 6, e25858,2011; Lester, S.et., Access Microbiology 1,20290,2019). Briefly, HEK293T/17 single layer cells transfected to express SARS-CoV-2S (SARS-CoV-2-S-C.DELTA.19) truncated by 19 amino acids of the C-terminal cytoplasm were inoculated with VSV.DELTA.G-GFP vector. After incubation at 37 ℃ for 1 hour, the inoculum was removed, the cells were washed with PBS, and then medium supplemented with anti-VSV-G antibody (clone 8G5F11, Kerafast Inc.) was added to neutralize residual input virus. 20 hours after inoculation, the medium containing the VSV/SARS-CoV-2 pseudovirus was harvested, filtered at 0.2 μm and stored at-80 ℃.

Vero-76 cells were seeded in 96-well plates. Serial dilutions of mouse serum samples were prepared and incubated with VSV/SARS-CoV-2 pseudovirus suspension (4.8X 10) at room temperature3Infectious unit [ IU]/mL) was preincubated for 10 minutes, and then the mixture was transferred to Vero-76 cells. The inoculated Vero-76 cells were incubated at 37 ℃ for 20 hours. Plates were placed in the IncuCyte viable cell assay system (Sartorius) and incubated for 30 minutes prior to analysis (IncuCyte 2019B Rev2 software). Full-well scans of bright field and GFP fluorescence were performed using a 4x objective. 50% of pseudoviruses were neutralized to titer (pVNT)50) Reported as the reciprocal of the first serum dilution that caused a 50% reduction in the number of GFP-positive infected cells per well compared to the average of serum-free pseudovirus-positive controls. Each serum sample dilution was tested in duplicate.

SARS-CoV-2 neutralization of human convalescent serum and rhesus serum

SARS-CoV-2 neutralization assay Using the previously described SARS-CoV-2 strain (USA _ WA1/2020), which has been rescued by reverse genetics and engineered by inserting the mNeonGreen (mNG) gene in the open reading frame 7 of the virome (Xie, X.et al., Cell host)&micro 27,841-848.e3,2020). This reporter virus produced similar plaque morphology and indistinguishable growth curves as the wild-type virus. The virus master stock was grown in Vero E6 cells as previously described (Lester, s.et al, Access Microbiology 1,20290,2019). When human convalescent serum samples are tested, the fluorescence neutralization assay produces results comparable to conventional plaque reduction neutralization assays. Serial dilutions of heat-inactivated serum were mixed with reporter virus (2X 10) prior to inoculation with Vero CCL81 cell monolayers in 96-well plates (targeting 15,000-cells per well) 4PFU per well to produce approximately 10-30% of Vero CCL81 monolayer infection rate) were incubated at 37 ℃ for 1 hour to allow accurate quantification of infected cells. Total Cell number per well was counted by nuclear staining (Hoechst 33342) and infection was detected with diseased using a staining 7Cell Imaging Multi-Mode Reader (Biotek) with Gen5 Image Prime version 3.09 16-24 hours after inoculationToxic fluorescent foci. In GraphPad Prism version 8.4.2, titers were calculated by a 4-parameter (4PL) logistic fit that generated a percent neutralization at each serial dilution of serum. 50% neutralization Titer (VNT)50) Reported as the reciprocal interpolated dilution that produced a 50% reduction in fluorescent viral foci.

IFNγELISpot.

Mouse IFN γ ELISpot was used according to the manufacturer's instructions (Mabtech)PLUSThe kit was used for ELISpot assay. With either full-length S-peptide cocktail (0.1. mu.g/mL final concentration per peptide, JPT) or control (gp70-AH1[ SPSYVYHQF ]](Slansky, J.E.et al., Immunity 13, 529-incorporated 538,2000), JPT, 4. mu.g/mL; concanavalin A (ConA), Sigma, 2. mu.g/mL) restimulation of a total of 5X 105Individual splenocytes ex vivo. streptavidin-ALP and BCIP/NBT-plus substrate were added and spots were counted using an ELISpot microplate reader (R) ((R))Core Analyzer, CTL). The number of spots was evaluated using ImmunoCapture Image acquisition software V7.0 and ImmunoSpot 7.0.17.0 Professional. For T cell subtyping, MACS MicroBeads (CD8a [ Ly-2 ] was used according to the manufacturer's instructions ]Miltenyi Biotec) isolation of CD8 from spleen cell suspensions+T cells. Flow-through for use as CD4+A source of T cells. Subsequent restimulation of CD8 with syngeneic bone marrow-derived dendritic cells+Or CD4+T cells, which are loaded with a full-length mixture of S-peptides (0.1. mu.g/mL final concentration per peptide) or medium as a control. Purity of the isolated T cell subsets was determined by flow cytometry to calculate per 1X 105An individual CD8+Or CD4+Number of spots of T cells.

Cytokine analysis

Mouse splenocytes were either re-stimulated with full-length S-peptide cocktail (0.2 μ g/mL final concentration per peptide) or culture medium alone for 48 hours. Use of 11-plex T based on beads according to the manufacturer's instructionsH1/TH2 mice ProcartaPlex multiplex immunoassay (Thermo Fisher Scientific) determined the concentration of IFN γ, IL-2, IL-4 and IL-5 in the supernatant. Using Bioplex200 System (Bio-Rad) and analyzed with ProcartaPlex Analyst 1.0 software (Thermo Fisher Scientific).

Statistics and repeatability

No statistical method is used to determine the group and sample sizes (n) in advance. All experiments were performed once. Use ofPROC gen mod of 9.4, determines the reported P-values for RT-qPCR analysis by performing a classification analysis on the treatment and day-long effects with a logit link on binomial responses (no viral RNA detected as successful after challenge and no viral RNA measurable as failed after challenge). The analysis contained samples from post challenge days (day 3 and day 6 for BAL; day 1, day 3 and day 6 for nasal swab). Uncertain results were excluded from the analysis. All remaining analyses were performed using GraphPad Prism 8.4.

Results

We designed a SARS-CoV-2 vaccine named BNT162b1 comprising LNP-encapsulated N1Methyl-pseudouridine (m1 Ψ) nucleoside modified mRNA encoding RBD fused at its C-terminus to the native trimerization domain (foldon) of T4 minor fibrin (Meier, S.et al, Journal of molecular biology 344, 1051-1069, 2004) (FIG. 64 a). The SARS-CoV-2S Signal Peptide (SP) allows ER to transport and secrete trimeric RBD. The m1 Ψ modification of RNA inhibits innate immune response and, together with optimized non-coding sequence elements, increases RNA translation in vivo (Orlandini von Niessen, A.G. et al, Mol Ther 27,824 836, 2019; Karik Shao, K.et al, Molecular Therapy: the journel of the American Society of Gene Therapy 16,1833 1840, 2008).

BNT162b1 RNA transcribed in vitro from plasmid DNA template by T7 polymerase had a single sharp microfluidic capillary electrophoresis peak spectrum, consistent with a calculated length of 1262 nucleotides, indicating its purity and integrity (data not shown). Western blot analysis of BNT162b1 RNA-transfected HEK293T/17 cell lysates showed that RBDs were expressed from RNA with an apparent molecular weight consistent with the calculated molecular weight of 29.46kDa (data not shown). Expression of the protein in transfected cells and localization of the endoplasmic reticulum on the secretory pathway were confirmed by flow cytometry and immunofluorescence microscopy, respectively (data not shown).

For structural characterization, trimeric RBDs were expressed in Expi293F cells using DNA sequences corresponding to the BNT162b1 RNA coding sequence and purified by ACE2 peptidase domain affinity capture immobilized on agarose beads. Trimeric RBDs with high affinity (5pM K)D) Binds to the human ACE2 Peptidase Domain (PD) at approximately 5.09nM K for the reported monomeric RBDD1000-fold higher, and consistent with the avidity effect of multimer binding (data not shown). Trimer binding valency (valency) of the RBD-folder and its flexibility were visualized by Electron Microscopy (EM) of negatively stained specimens, revealing a series of conformations (fig. 64 b). Although the flexible barrier of the RBD-folder allows high resolution direct structural analysis, when the complex is in the previously reported closed conformation (Yan, R. et al., Science (New York, N.Y.)367, 1444-1448, 2020), the RBD domain can bind to ACE2 and B by binding to0AT1 neutral amino acid transporter (ACE2 is a molecular chaperone) is immobilized. Pairing RBD-folder/ACE 2/B by cryo-electron microscopy (cryoEM)0The size and symmetry of the AT1 ternary complex were image reconstructed and the structure of the RBD domain in the complex was determined as Resolution (fig. 64 c). One copy of the RBD was resolved for each bound trimer. The binding interface between the resolved extracellular domains of RBD and ACE2 was identical to that previously reported and showed good agreement (He, Y.et al, Biochemical and Biophysical Research Communications 324, 773-&molecular immunology; 10.1038/s 41423-020-. The high affinity binding to ACE2 and the well resolved structure of the complex with ACE2 indicate that the recombinant RBD-folder does present many of the ACE2 binding sites targeted by the SARS-CoV-2 neutralizing antibody (Brouwer, P.J.M.et. Science (New York, N.Y.); 10.1126/science.abc5902 (2020); Zost, S.J.et. al., Nature media; 10.10.1038/s41591-020- 0998-x(2020))。

After a single Intramuscular (IM) immunization with 0.2, 1 or 5 μ g BNT162B1 or buffer only, respectively, a series of experiments in BALB/c mice characterized the B-and T-cell immune responses elicited by BNT162B 1. At all dose levels, RBD-specific serum IgG developed rapidly in a dose-dependent manner and stabilized around day 21 (1.63. + -. 0.13mg/mL for the 5. mu.g dose level; FIG. 65 a). Vaccine-induced IgG has high RBD binding affinity (geometric mean K) D48.0pM), high on-rate (geometric mean k)on 1.72x 106/Ms) and a low off-rate (geometric mean K)off 8.27x 10-5S; fig. 65 b). SARS-CoV-2 neutralization activity in mouse serum was measured using a SARS-CoV-2 pseudovirus neutralization assay based on Vesicular Stomatitis Virus (VSV). After immunization, 50% pseudovirus neutralization titers (pVNT) for dose levels of 0.2, 1 and 5. mu.g50) The mean values steadily increased to 102, 192 and 1056 on day 28, respectively (fig. 65 c).

High proportion of CD4 isolated from BNT162b1 immunized mice on days 12 and 28 post-immunization+And CD8+T cell phenotype of splenocytes, when restimulated ex vivo with the full length S peptide mixture, showed strong antigen-specific IFN γ responses in the ELISpot assay (fig. 65 d). Full-length S-peptide stimulated a large number of splenocytes and CD4 in flow cytokine release assays+And CD8+The subset also showed high IFN γ production and a significant IL-2 response at day 12, but the IL-4 response was much lower, indicating a THPhenotype 1 response (fig. 65 e). In multiplex immunoassays, THPhenotype 1 persists, with high levels of IL-2 and IFN γ, but T, produced by total splenocytes collected on day 28 and stimulated with full-length S peptide poolHThe amounts of 2 cytokines IL-4 and IL-5 were not detectable (FIG. 65 f).

In draining lymph nodes (dLN) and spleen obtained at day 12 after immunization of mice with BNT162B1 or buffer, much higher numbers of B cells (including plasma cells, class-switched IgG 1-positive and IgG2 a-positive B cells, and germinal center B cells) were observed in samples from mice receiving BNT162B1 (data not shown)Shown). In blood obtained 7 days after immunization, the number of circulating B cells was lower than in buffer-immunized mice, probably due to B cell homing to the lymph compartment (data not shown). dLN in mice immunized with BNT162b1 also showed an increase in T cell counts, especially follicular helper T (T)FH) The number of cells, including subsets with ICOS upregulation, was known to play a crucial role in germinal center formation (Hutloff, a., Oncotarget 6, 21785-21786, 2015) (data not shown). BNT162b 1-induced T was also detected in spleen and bloodFHCytosis (data not shown). Overall, these data indicate that BNT162b1 induces both robust SARS-CoV-2S specific neutralizing antibody titers and TH1 driven T cell response. The intramuscularly administered BNT162b1 appears to have been delivered to dLN as an immuno-educed site for effective vaccine priming (priming) whereby lymphocytes migrate from the blood to the lymph tissue to participate in the vaccine response.

BNT162b1 was next tested for immunogenicity in male rhesus monkeys between 2 and 4 years of age. Six groups of each were immunized with 30 or 100 μ g BNT162b1 or control saline IM on days 0 and 21. IgG bound to RBD could be readily detected at day 14 after a single immunization and levels further increased by day 21 (when booster doses were given) (fig. 66 a). 7 days after the second immunization (day 28), geometric mean concentration of RBD-bound IgG (GMC) was 20,962 units (U)/mL (30. mu.g dose level) and 48,575U/mL (100. mu.g dose level). For comparison, a group of 38 SARS-CoV-2 convalescent human sera had an RBD-binding IgG GMC of 602U/mL, which was significantly lower than the GMC of the immunized rhesus monkey after one or two doses. 50% neutralization Titer (VNT) determined by SARS-CoV-2 neutralization of authentic (authetic)50) Detectable in rhesus monkey serum at day 14 after a single immunization (Muruato, a.e.et al, bioRxiv: the print server for biology; 10.1101/2020.05.21.109546, 2020), and reached 768(30 μ g dose level) or 1714(100 μ g dose level) at 7 days post-boost (day 28, fig. 66 b). Robust neutralization of GMT for 247(30 μ g dose level) and 564(100 μ g dose level) continued for at least day 42 (most recent) Detection time point). For comparison, the 50% neutralization GMT of the human convalescent serogroup was 93.6.

Day 41-48 after the second immunization, a rhesus group (n-6) immunized with 100 μ g BNT162b1 or buffer control twice was used at 1 × 106Plaque-forming units of SARS-CoV-2 (strain USA-WA1/2020) were challenged and evenly distributed between the intranasal and intratracheal routes as described previously (Singh, D.K. et al. SARS-CoV-2 infection leads to access infection with dynamic cells and infection fluorine in the lung of vary varieties of access non-human mammalian species, 2020). Three non-immunized, age-matched male rhesus monkeys (sentinel) were challenged with cell culture media to simulate. At challenge, the SARS-CoV-2 neutralization titer was 208-1185 in BNT162b 1-immunized animals, but not detectable in control-immunized and sentinel groups of animals.

SARS-CoV-2RNA was measured in bronchoalveolar lavage (BAL) and nasal swab samples by quantitative reverse transcription-polymerase chain reaction (RT-qPCR). All BAL and nasal swab samples obtained prior to infection challenge and all samples obtained from sentinel animals lacked detectable SARS-CoV-2RNA (FIG. 67). Three days after the SARS-CoV-2 challenge, viral RNA was detected in BAL fluid from 5/6 control immunized animals and 2/6BNT162b1 immunized animals (FIG. 67 a). By 6 days post challenge, viral RNA was undetectable in the lungs of all six BNT162b 1-immunized rhesus monkeys; in control-immunized rhesus monkeys, three BAL solutions had high levels of viral RNA, two had cleared, and one RT-qPCR was inconclusive. At necropsy (7-23 days after challenge), viral RNA was undetectable in BAL fluid from any animal. Following SARS-CoV-2 challenge, viral RNA was detected in nasal swabs of control immunization groups at each time point: two animals on day 1 post challenge, three animals on day 3 and day 6 post challenge (fig. 67b), two animals at necropsy (not shown). In animals immunized with BNT162b1, all nasal swabs were negative or indeterminate at day 1 and negative at day 3 and necropsy; swabs from both animals were positive on day 6, indicating that detection of viral RNA is a more transient process than non-immunized rhesus monkeys. The difference in the proportion of animals with detectable viral RNA was statistically significant between BNT162b1 immunized animals and control immunized animals (BAL p 0.0037, nasal swab 0.0212). None of the challenged animals showed clinical or radiological evidence of significant disease, suggesting that the 2-4 year old male rhesus challenge model was primarily a model of infection with SARS-CoV-2, not a COVID-19 disease model.

Discussion of the related Art

We demonstrate that BNT162b1, an m1 Ψ nucleoside modified mRNA formulated in LNP encoding a trimeric RBD antigen, is highly immunogenic in mice and rhesus monkeys and limits infection in rhesus monkeys challenged with infectious SARS-CoV-2. The RBD-folder coding sequence directs the expression of a flexible trimeric protein that binds ACE2 with high affinity and has a structurally intact ACE2 receptor binding site. One key finding is that a single sub-microgram immunization rapidly induces high neutralizing antibody titers in mice that are within or above the range of recently reported SARS-CoV-2 vaccine candidates (van Doremalen, N.et al., bioRxiv: the preprint server for biology; 10.1101/2020.05.13.093195 (2020); Corbett, K.S.et al., bioRxiv: the preprint server for biology; 10.1101/2020.06.11.145920 (2020)). Mean tendency TH1-biased strong CD4+And stronger CD8+T cell response and TFHMay suggest a strong protective capacity induced by vaccine candidates (Pardi, N.et al, The Journal of Experimental Medicine 215, 1571-1588, 2018). T isFHProliferation at germinal centers is essential for the generation of adaptive B cell responses. In humans, T occurs in the circulation (circulation) after vaccination FHAssociated with high frequency of antigen-specific antibodies (Farooq, f.et al., Scientific reports 6,27944, 2016). Immunization with BNT162B1 triggered B cells and TFHRedistribution of cells from the blood to the lymphoid tissue where antigen presentation occurs.

Another important finding was that in rhesus monkeys, two doses of m1 Ψ nucleoside modified mRNA encoding the trimeric SARS-CoV-2S RBD-foldon elicited a neutralization of SARS-CoV-2 that was 8.2-18.2 times greater than the human serogroup GMT during the SARS-CoV-2 convalescence. The results in non-human primates demonstrate that the vaccine has high efficacy in preclinical models of acute SARS-CoV-2 infection and provides protection against SARS-CoV-2.

Example 11: RNA vaccines encoding pre-fusion stable SARS-CoV-2S with high immunogenicity in mice and non-human primates

Here, we report SARS-CoV-2 infection challenge in rhesus monkeys immunized with BNT162b2 vaccine.

Materials and methods

Preparation of in vitro transcribed RNA

To generate a template for RNA synthesis, a DNA fragment encoding the full-length SARS-CoV-2S protein (GenBank: MN908947) with amino acid exchanges of K986P and V987P was cloned into a starting plasmid vector with framework sequence elements for improved RNA stability and translation efficiency (Orlandini von Niessen, A.G.et al, Mol Ther 27, 824-836, 2019; Holtkamp, S.et al, Blood 108, 4009-4017, 2006). The non-coding backbone element included a region from the T7 promoter to the 5 'and 3' UTR plus poly (A) tail (100 nucleotides), interrupted by a linker (A30LA70, 10 nucleotides). DNA was purified, spectrophotometrically quantified and analyzed for the presence of the trinucleotide cap1 analog ((m) 2 7 ,3’-O)Gppp(m2’-O) ApG, respectively; TriLink) and N in place of uridine-5' -triphosphate (UTP)1In vitro transcription with T7 RNA polymerase in the presence of methylpseuduridine-5' -triphosphate (m1 Ψ TP; Thermo Fisher Scientific) (Grudzien-Nogalska, E.et. al., Methods in molecular biology (Clifton, N.J.)969, 55-72,2013). RNA was purified using magnetic particles (Berensmeier, s., appl. microbiol.biotechnol.73, 495-504, 2006), integrity was assessed by microfluidic capillary electrophoresis (Agilent Fragment Analyser), and concentration, pH, osmolality, endotoxin level and bioburden were determined.

Lipid nanoparticle formulations of RNA

Purified RNA was formulated into LNP using an ethanolic lipid cocktail of ionizable cationic lipids and transferred to an aqueous buffer system by diafiltration to produce an LNP composition similar to that previously described (Maier, M.A. et al, Molecular Therapy: the journal of the American Society of Gene Therapy 21, 1570-1578, 2013). BNT162b2 was stored at-70 ℃ at a concentration of 0.5 mg/mL.

Immunization

On days 0 and 21, male rhesus monkeys (2-4 years old) were randomly assigned to receive BNT162b2 or saline placebo control in a volume of 0.5mL administered by IM injection into the left quadriceps muscle. Serum and PBMCs were collected following the animal protocols 2017-8725-023 approved by the NIRC animal care and use committee. During blood sampling and immunization, animals were anesthetized with ketamine hydrochloride (10mg/kg, IM) and monitored closely for sedation.

SARS-CoV-2 challenge in rhesus monkeys

SARS-CoV-2 inoculum was prepared from 2.1X 10 previously prepared in the Texas Biomedical Research Institute (San Antonio, Texas)6PFU/mL stock was obtained, dispensed into single use vials, and stored at-70 ℃. Working virus stocks were generated from SARS-CoV-2USA-WA1/2020 isolate (4 th generation seed stock from BEI Resources; NR-52281) passaged twice in Vero E6 cells. The virus was confirmed to be SARS-CoV-2 by deep sequencing and to be identical to the published sequence (GenBank accession No. MN 985325.1).

Male rhesus monkeys (control group) immunized with BNT162b1 (n ═ 6) and age-matched saline control (n ═ 6) were used at 1 × 106The plaque-forming unit SARS-CoV-2USA-WA1/2020 isolate WAs challenged by equally partitioning between the intranasal (IN, 0.2mL) and intratracheal (IT, 0.2mL) pathways as previously described (Singh, D.K.et., SARS-CoV-2infection leads to access infection with dynamic cells and infection fluorine IN the lung of non-human mammalian species, 2020). Challenges were made 41-48 days after the second immunization. Separate sentinel groups of non-immunized age and gender matched animals (n ═ 3) received DMEM only supplemented with 10% FCS IN (0.2mL) and IT (0.2 mL). Approximately two weeks prior to challenge, animals were transferred to the animal biosafety level 3(ABSL-3) facility of the southwest national center for primate research (SNPRC; San Antonio, TX). The animals were monitored periodically for rectal temperature, body weight and physical examination by a committee-certified veterinary clinician. As described in (Singh, D.K.et. al., SARS-CoV-2infection leads to access infection with dynamic cells and infection fluorine in the long that vary across different non-human patient samples, 2020) samples were taken under tiletamine zolazepam (Telazol) anesthesia. Nasal swabs were collected from rhesus monkeys on days 0, 1, 3, and 6 post-inoculation to assess viral titers. Bronchoalveolar lavage (BAL) was performed four times by instillation of 20mL saline one week before challenge and on days 3 and 6 after vaccination. The washes were pooled, collected, aliquoted and stored frozen at-70 ℃.

Quantitative reverse transcription polymerase chain reaction

SARS-CoV-2 was detected and quantified in NHP essentially as described in example 10.

Results

The results indicate that after a single dose, the immunogenicity of the COVID-19mRNA vaccine BNT162b2 elicits an IgG response, which is enhanced by a second dose. These also showed a dose response. At 30 μ g BNT162, the neutralization geometric mean titer was compared to that observed in convalescent plasma of SARS CoV-2 human patients, and was found to be about 8-fold higher 7 days after the dose, yielding a higher excess of about 18-fold over 7 days after the higher dose of 100 μ g, and remained 3.3-fold of this baseline five weeks after the last immunization. In monkeys, the response was also characterized by a predominance of Th1 for IFN-. gamma.and IL-2, but no IL-4 response. CD4 and CD8 positive cellular responses were also observed in monkeys. This cellular immune response is characterized by a strong Th1 biased CD4 +T cell responses, with interferon-gamma (IFN-. gamma.) + CD8+ T cell responses.

Day 41-48 after the second immunization, the rhesus monkey group (n-6) immunized with 100 μ g BNT162b2 or buffer control twice was immunized with 1 × 106The plaque forming unit of SARS-CoV-2 (strain USA-WA1/2020) WAs challenged by equally dividing between the intranasal and intratracheal routes as described previously (Singh, D.K. et al. SARS-CoV-2 infection leads to access infection with dynamic cells and infection fluorine in the lung of vary varieties across non-human matrices, 2020). Three non-immunized, age-matched male rhesus monkeys (sentinel) were simulated with cell culture mediumAnd (5) challenging. At challenge, the SARS-CoV-2 neutralization titer was 204-938 in the BNT162b 2-immunized animal group, but not detectable in the control-immunized and sentinel group animals.

Measurement of SARS-CoV-2RNA in bronchoalveolar lavage (BAL) and nasal swab samples by quantitative reverse transcription-polymerase chain reaction (RT-qPCR). All BAL and nasal swab samples obtained prior to infection challenge and all samples obtained from sentinel animals lacked detectable SARS-CoV-2RNA (FIG. 68). Three postnatals of SARS-CoV-2 challenge, viral RNA was detected in BAL fluid from 5/6 control-immunized animals and 2/6BNT162b 2-immunized animals (FIG. 68). By 6 days post challenge, viral RNA was undetectable in the lungs of all six BNT162b 2-immunized rhesus monkeys; in control-immunized rhesus monkeys, three BAL solutions had high levels of viral RNA, two had cleared, and one RT-qPCR was inconclusive. After the SARS-CoV-2 challenge, viral RNA was detected in the nasal swab of the control immunised group at each time point: two animals on day 1 post challenge, three animals on day 3 and day 6 post challenge (fig. 68). In animals immunized with BNT162b2, all nasal swabs were negative on days 3 and 6.

In lung tissue, increased computed tomography scores indicated that control monkeys had evidence of lung disease, with scores at day 10 lower than day 3 suggesting recovery; in contrast, monkeys given the COVID-19mRNA vaccine BNT162b2 scored lower. Microscopic analysis of lung tissue showed that lung inflammation was similar between control and BNT162b2 immunized monkeys, with no evidence of exacerbation of respiratory disease.

Discussion of the related Art

Results in non-human primates demonstrate the efficacy (potency) and ability of BNT162b2 (LNP formulated m1 Ψ nucleoside modified mRNA encoding S antigen captured in the pre-fusion conformation) to provide protection against SARS-CoV-2 challenge in preclinical models of acute SARS-CoV-2 infection.

Example 12: storage, transport and dose preparation

This example illustrates the storage, transport and dose preparation of multi-dose vial BNT162b2 concentrate for injection.

As shown in fig. 69, in the initial packaging stage, 2ml type 1 glass preservative free multi-dose vials (MDV) were used, with 0.45ml of frozen liquid drug product, 5 doses per vial. In the secondary packaging stage, a single tray holds 195 vials, for example 975 doses per tray. The size of the tray (white box) was 229X40 mm. At least 1 tray (975 doses) (or up to 5 trays (up to 4875 doses)) is stacked in the loading bin in three packaging stages. The loading bin was submerged in 23Kg of dry ice pellets (10mm-16mm pellets). The dimensions of the insulated transport (thermal spreader) are as follows: internal dimensions: 245mm X245 mm X241 mm; external dimensions: 400mm X400 mm X560 mm. The total weight of the heat-insulating transportation tool is 35 Kg.

Ultra-low temperature (ULT) freezers of different sizes are available on the market. Fig. 70 is an example of small volume storage (about 90 liters; about 30K dose (left)) and large volume storage (about 500 liters; about 200K dose (right)). If stored without unsealing at a temperature of 15 ℃ to 25 ℃, the insulated carrier may maintain a ULT (e.g., -90 ℃ to-60 ℃) for up to 10 days, and the storage period may be further extended by continually refilling the top of the container with dry ice. Once received and opened, the box should be refilled with dry ice (23Kg dry ice pellets (10 mm-to 16 mm)) within 24 hours, the insulated vehicle is refrozen every 5 days, it is recommended that the insulated vehicle be opened no more than twice a day, the insulated vehicle should be closed within 1 minute (or less) after opening, the vaccine can be stored at 2 ℃ to 8 ℃ for up to 2 days, or thawed at room temperature for no more than 2 hours, and the time period for use after dilution is 6 hours.

Figure 71 shows exemplary dose preparation of BNT162b 25 dose vials containing frozen concentrated solution without preservative that must be thawed and diluted prior to administration. The preparation steps are as follows:

a 5-dose vial of BNT162b2 concentrate for injection is removed from the cryopreserved box and thawed at room temperature (e.g., up to 25 ℃) for about 30 minutes. In some embodiments, such BNT162b2 multi-dose vials can be thawed and stored in a refrigerator (e.g., 2 ℃ -8 ℃), for example, for up to 5 days. Vials thawed at room temperature must be diluted within 2 hours or transferred to a refrigerator. The undiluted vials can be stored in the refrigerator for up to 48 hours. The thawed vials were not refrozen. During storage, exposure to indoor light is minimized and exposure to direct sunlight and ultraviolet light is avoided. The thawed vials can be processed under room light conditions.

After thawing and before use, the vials were allowed to equilibrate to room temperature and mixed by gently inverting 10 times. Do not shake.

Using aseptic technique, the vial stopper was cleaned with a disposable sterile cotton swab, and then the vial of melted BNT162b2 was diluted by adding 1.8mL of 0.9% sodium chloride injection (USP) to the vial. A 21 gauge (gauge) needle or a narrower needle is recommended. However, those skilled in the art will appreciate that in some embodiments, a wider needle may be used. For example, in some embodiments, needles 20, 19, 18, 17, 16, 15 or wider needles may be used.

When the diluent is added, pressure in the vial may be experienced. Before removing the needle from the vial, pressure equalization in the vial was ensured by drawing 1.8mL of air into the empty diluent syringe.

The diluted vial was gently inverted 10 times for mixing. Do not shake.

The date and time of dilution was recorded at the appropriate location on the vial label of BNT162b 2. The shelf life after dilution was 6 hours. The diluted multi-dose vials were stored between 2 ℃ and 25 ℃. Do not freeze. If frozen, discarded.

Using aseptic technique, the vial stopper was cleaned with a disposable sterile cotton swab, and 0.3mL of diluted dosage solution was withdrawn using a novel sterile metered dose syringe with a needle suitable for intramuscular injection. The syringe should be adjusted to remove air bubbles while the needle is still in the vial to avoid loss of dosage solution. It is recommended to aspirate and administer the drug using the same needle as much as possible. If a second needle is required for administration, the syringe plunger is pulled rearwardly before the first needle is removed until a small amount of air enters the syringe to avoid losing the dose of solution when the needle is replaced. Care was taken when priming the needle to prevent loss of dose.

For each additional dose, a new sterile syringe and needle are used, and the vial stopper is ensured to be cleaned with the antimicrobial agent prior to each draw. The prepared syringe should be used immediately. If administration is not immediate, it must be within 6 hours of the initial dilution of the vial. Before administration, a final injection volume of 0.3mL in the syringe was ensured.

Example 13: vaccine candidates against COVID-19 were successful in the first interim analysis of phase 3 studies

To date, more than 43,000 participants have been enrolled in phase 3 clinical trials of BNT162b2, with nearly 39,000 of them having received a second dose of vaccine candidate by 11, 8 days of 2020. Approximately 42% of global participants and 30% of american participants have diverse backgrounds of race (e.g., including white, black or african american, american indian or alaska former, asian, hawaii former or other pacific islands, multi-races) and ethnic (e.g., including hispanic/hispanic (Latino) and non-hispanic/non-hispanic). The clinical trial was continuing to recruit and a final analysis was expected to accumulate to a total of 164 confirmed COVID-19 cases.

Vaccine candidate BNT162b2 succeeded in the first interim analysis of the phase 3 study. In the first intermediate efficacy assay, vaccine candidates were found to be more than 90% effective at preventing COVID-19 in participants with no evidence of prior SARS-CoV-2 infection. Analysis 94 confirmed COVID-19 cases among the trial participants were evaluated. No serious potential safety hazard is found.

A panel of results from the phase 3 covd-19 vaccine trial collected 11, 4 days ago 2020 provides evidence that BNT162b2 is able to prevent covd-19. The division of the disease between vaccinated and placebo-received individuals showed that the effective rate of the vaccine was over 90% 7 days after the second dose. In particular, early analysis of results showed that more than 90% of subjects receiving two vaccine injections three weeks apart exhibited symptomatic COVID-19 than subjects receiving placebo. This confirms that protection is achieved 28 days after the start of vaccination consisting of a 2 dose schedule.

Preliminary such data include the following table:

TABLE 5 vaccine efficacy-first development of COVID-19 from 7 days after dose 2-subjects without evidence of infection before 7 days after dose 2-assessable efficacy population (7 days) -metaphase assay 1

Abbreviations: n-binding to SARS-CoV-2 nucleoprotein (nucleoprotein); NAAT ═ nucleic acid amplification assay; SARS-CoV-2 ═ severe acute respiratory syndrome coronavirus 2; VE ═ vaccine potency.

Note: the assay included subjects who had no serological or virological evidence of prior SARS-CoV-2 infection (7 days after receiving the last dose) (i.e., N-binding antibody [ serum ] was negative at visit 1, no SARS-CoV-2 was detected by NAAT [ nasal swab ] at visits 1 and 2), and NAAT was negative at any unplanned visit 7 days after dose 2.

Note: data including undiagnosed subjects 7 days after dose 2 were analyzed to fully display all data reported and/or to facilitate calculation of total monitoring time, but may vary with additional follow-up.

a.N number of subjects in the indicated group.

N 1-number of subjects who meet the endpoint definition.

Total monitoring time for specific endpoints of 1000 human-year, all subjects within each endpoint risk group. The time period for COVID-19 case accumulation ranged from 7 days after dose 2 to the end of the monitoring period.

N 2-number of subjects at endpoint risk.

e. The confidence interval for VE is calculated using a beta-binomial model adjusted for the monitoring time.

f. The posterior probability (Pr) is calculated using a beta-binomial model adjusted for the monitoring time. In the interim analysis, the probability must be at least 99.5% to conclude that the vaccine is effective.

TABLE 6 Severe COVID-19 occurring after dose 1-dose 1 all available efficacy populations-metaphase assay 1

Note: data including undiagnosed subjects 7 days after dose 2 were analyzed to fully display all data reported and/or to facilitate calculation of total monitoring time, but may vary with additional follow-up.

a.N number of subjects in the indicated group.

b.n number of subjects who meet the endpoint definition.

Example 14 evaluation of potency and immunogenicity

Efficacy results

The first interim analysis, performed after at least 62 cases accumulated after protocol and SAP, demonstrated the vaccine efficacy of BNT162b2 against COVID-19 in participants who had no evidence of prior SARS-CoV-2 infection. The primary efficacy results presented in this section are from this interim analysis.

This interim analysis only analyzed and presented the vaccine efficacy of BNT162b2 for the first primary efficacy endpoint (based on COVID-19 incidence in central laboratories or locally determined NAAT participants who had no serological or virological evidence of previous SARS-CoV-2 infection prior to 7 days after receiving the second dose).

First primary efficacy endpoint

Among the participants included in the efficacy-assessed population, 32,279 participants (16,061 in BNT162b2 and 16,218 in the placebo group) had no evidence of SARS-CoV-2 infection up to (through)7 days after the second dose. There were 4 cases of COVID-19 in BNT162b2, compared to 90 cases of COVID-19 reported in the placebo group. These data provide an estimated vaccine potency of 95.5% for BNT162b 2. A posterior (posterior) probability of > 99.99% meets the pre-specified mid-term analysis success criteria of > 99.5% (table 7). The 95% confidence interval for vaccine efficacy was 88.8% -98.4%, indicating a 95% probability of true VE in this interval according to the currently observed data. In addition, note that the posterior probability of true VE > 86.0% is 99.5%, and the posterior probability of VE > 88.8% is 97.5%.

Based on all available efficacy populations, the vaccine efficacy of BNT162b2 for the same primary efficacy endpoint was 95.7%, with 4 and 93 in BNT162b2 and placebo, respectively.

Within the cohort of dose 2 evaluable efficacy, no clinically significant differences in VE were observed by age group, country, ethnicity, gender or ethnicity, with VE estimates ranging from 91.2% to 100.0% (table 8).

Severe COVID-19 cases

By the date of data expiration of the first interim analysis by day 11, month 4 of 2020, a total of 7 participants reported severe COVID-19 cases in phase 3, all of which were placebo. 5 of these cases were reported between dose 1 and dose 2, no reports were reported for less than 7 days after dose 2, and 2 were reported at least 7 days after dose 2.

Conclusion on efficacy

The first primary efficacy target meets the success criteria. In participants with no evidence of infection up to 7 days after dose 2, BNT162b2 vaccine potency reached 95.5%, with a bilateral (2-sized) 95% confidence interval of 88.8% -98.4%, and a posterior probability of > 99.99% for true vaccine potency of greater than 30% under available data.

By the expiration date of the interim analysis, a total of 7 severe COVID-19 cases were observed in the placebo group (dose 1 later).

Immunogenic results

Stage 1

This phase 1 intermediate Clinical Study Report (CSR) presents immunogenicity results for two adult age groups up to 1 month after dose 2 of BNT162b1 and BNT162b2 vaccine candidates at dose levels of 10 μ g, 20 μ g, and 30 μ g, and up to 7 weeks after dose 1 of BNT162b1 at dose levels of 100 μ g (light annual group only).

Only the results 7 days after the dose 1 time point in the young group (18-55 years) of 10 μ g and 30 μ g BNT162b1 were analyzed and presented.

SARS-CoV-2 neutralization titer-1 phase

GMT

Overall, SARS-CoV-250% neutralization of GMT increased moderately by day 21 after dose 1 and significantly by day 7 after dose 2 for BNT162b1 and BNT162b2 recipients in both age groups. In general, at most points in time, the GMTs for the older group were lower than the GMTs for the younger group for the BNT162b1 and BNT162b2 recipients.

BNT162b1

In the young group, SARS-CoV-250% neutralized GMT increased moderately by day 21 after dose 1 and significantly by day 7 after dose 2 of BNT162b1 (day 28), with higher GMT observed in the 30 μ g dose group compared to the 10 μ g and 20 μ g dose groups (figure 72). GMT increased 14 days after dose 2 (day 35) for all dose groups and, although GMT decreased 1 month after dose 2 (day 52), GMT remained significantly higher at day 52 than at earlier time points after dose 1.

In the 100 μ g dose group, the SARS-CoV-250% neutralization GMT increased moderately on day 21 after dose 1 of BNT162b1, and decreased to near baseline by day 52.

A substantially similar trend was observed in the older groups, with higher GMTs observed in the 20 μ g and 30 μ g dose groups of BNT162b1 compared to the 10 μ g dose group (figure 73).

A similar trend was also observed for SARS-CoV-290% neutralization of GMT.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

RCDC at SARS-CoV-250% and 90% neutralization titers after BNT162b1 administration in the young and old groups indicated that most participants responded 7 days after dose 2 to BNT162b 1.

BNT162b2

In the young group, SARS-CoV-250% neutralized GMT to BNT162b2 increased by day 21 after dose 1 and significantly increased by day 7 after dose 2 (day 28), with higher GMT observed in the 20 μ g and 30 μ g dose groups compared to the 10 μ g dose group (figure 74). GMT declined at 14 days after dose 2 (day 35) and 1 month after dose 2 (day 52) of BNT162b2, but GMT remained significantly higher than the GMT at the earlier time point after dose 1.

A generally similar trend was observed in the older groups, with higher GMT observed in the 30 μ g dose group compared to the 20 μ g and 10 μ g dose groups (figure 75). 7 days after dose 2, SARS-CoV-250% neutralized GMT increased and was similar in the 10. mu.g and 20. mu.g dose groups, and higher in the 30. mu.g dose group. The GMT remained significantly higher 1 month after dose 2 than at the earlier time point after dose 1. In the older group, SARS-CoV-250% neutralized GMT was generally lower than that in the younger group.

A similar trend was also observed for SARS-CoV-290% neutralization of GMT.

The results of all available immunogenic populations in the young and old populations are similar to those observed for the evaluable immunogenic population.

RCDC at SARS-CoV-250% and 90% neutralization titers in the young and old groups indicated that most participants responded 7 days after dose 2 to BNT162b 2.

GMFR

Overall, for BNT162b1 and BNT162b2 recipients, and in both age groups, the GMFR of SARS-CoV-250% neutralization titer from pre-vaccination to 7 days after dose 2 (day 28) was significantly higher than the GMFR of each after dose 1. For BNT162b1 and BNT162b2 recipients, GMFR in the senior group is typically lower than the GMFR of the senior group.

BNT162b1

For all dose groups, GMFR neutralization titers of SARS-CoV-250% were significantly higher from pre-vaccination to 7 days after BNT162b1 dose 2 (day 28) compared to GMFR at earlier time points after BNT162b1 dose 1 in the young group, and GMFR was highest in the 30 μ g dose group. 1 month after dose 2, the GMFR remained higher than that at the earlier time point after dose 1.

In the 100 μ g dose group, there was no significant increase in GMFR for SARS-CoV-250% neutralization titer by day 52 after BNT162b1 dose 1.

For the 20 μ g and 30 μ g dose groups, GMFR neutralization titers of SARS-CoV-250% were significantly higher in the older group from before vaccination to 7 days after BNT162b1 dose 2 (day 28) compared to GMFR at an earlier time point after BNT162b1 dose 1, and GMFR was highest in the 20 μ g dose group. GMFR 1 month after BNT162b1 dose 2 (day 52) remained high in the 20 μ g and 30 μ g dose groups compared to GMFR at an earlier time point after dose 1.

A similar trend was also observed for the SARS-CoV-290% neutralizing titer GMFR in the young and old groups.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

BNT162b2

For all dose groups, GMFR was significantly higher in SARS-CoV-250% neutralizing titers from pre-vaccination to 7 days after BNT162b2 dose 2 (day 28) compared to GMFR at an earlier time point after dose 1 of BNT162b2 in the young group, and GMFR was similar and highest in the 20 μ g and 30 μ g dose groups. GMFR remained high by 1 month after dose 2 of BNT162b2 compared to GMFR 21 days after dose 1 of BNT162b 2.

For all dose groups, GMFR neutralization titers of SARS-CoV-250% were significantly higher from pre-vaccination to 7 days after dose 2 of BNT162b2 (day 28) in the older group compared to GMFR at an earlier time point after dose 1 of BNT162b2, and GMFR was highest in the 30 μ g dose group. GMFR remained high by 1 month after dose 2 of BNT162b2 compared to GMFR 21 days after dose 1 of BNT162b 2.

A similar trend was also observed for the SARS-CoV-290% neutralizing titer GMFR in the young and old groups.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

Number of participants (%)

Overall, for BNT162b1 and BNT162b2 recipients, and in both age groups, from pre-vaccination to 7 days post-dose 2, most participants achieved > 4-fold increase in SARS-CoV-250% neutralization titer (rise), except for the older participants in the 10 μ g BNT162b1 dose group.

BNT162b1

In the young group, no participants were present in the 10. mu.g dose group from before vaccination to 21 days after dose 1 of BNT162b1, and < 3 participants in the 20. mu.g and 30. mu.g dose groups achieved > 4-fold increase in the neutralization titer of SARS-CoV-250%. From before vaccination to 7 days after dose 2 of BNT162b1 and 1 month, most or all of the participants in the 10 μ g, 20 μ g and 30 μ g dose groups achieved > 4-fold increase in the SARS-CoV-250% neutralization titer.

In the older group, only 1 participant in the 30 μ g dose group achieved a > 4-fold increase in SARS-CoV-250% neutralization titer from pre-vaccination to 21 days after dose 1 of BNT162b 1. From before vaccination until 7 days after dose 2 of BNT162b1 and 1 month, 2 participants in the 10. mu.g group and 9-11 participants in the 20. mu.g and 30. mu.g dose groups achieved a 4-fold increase in the SARS-CoV-250% neutralization titer.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

BNT162b2

In the young group, 2 (18.2%) participants in the 10. mu.g dose group, 3 (25.0%) participants in the 20. mu.g dose group, and none of the participants in the 30. mu.g dose group achieved a 4-fold increase in SARS-CoV-250% neutralization titer from pre-vaccination to 21 days after dose 1 for BNT162b 2. From before vaccination to 7 days after dose 2 of BNT162b2, all participants achieved a > 4-fold increase in the SARS-CoV-250% neutralization titer, which was maintained up to 1 month after dose 2 of BNT162b 2.

In the older groups, none of the participants in any dose group achieved a > 4-fold increase in SARS-CoV-250% neutralization titer from pre-vaccination to 21 days after dose 1 of BNT162b 2. 10 (83.3%), 9 (81.8%) and 10 (90.9%) participants achieved > 4-fold increase in SARS-CoV-250% neutralization titer in the 10. mu.g, 20. mu.g and 30. mu.g dose groups from pre-vaccination to 7 days after dose 2 of BNT162b 2. From before vaccination up to 1 month after dose 2 of BNT162b2, 9 (75.0%), 6 (54.5%) and 11 (100.0%) participants achieved > 4-fold increase in SARS-CoV-250% neutralization titer in the 10, 20, and 30 μ g dose groups, respectively.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

SARS-CoV-2 antigen specific binding antibody level-1 phase

Vaccine candidate BNT162b1 encodes the RBD of SARS-CoV-2. This section describes the RBD-binding IgG response of the candidate at each dose level and age group. The RBD binding IgG level of candidate BNT62b2 encoding P2S of SARS-CoV-2 was also assessed.

Vaccine candidate BNT162b2 encodes P2S of SARS-CoV-2. This section describes the S1 binding IgG response for each dose level and age group of the candidate. The level of S1 binding IgG of BNT62b1, a candidate for an RBD encoding SARS-CoV-2, was also assessed.

GMC

Overall, for BNT162b1 and BNT162b2 recipients, and in both age groups, by day 21 post dose 1, RBD and S1 bound GMC increased significantly and further increased by day 7 post dose 2. The response was maintained by day 52. GMCs in the older group were generally lower than those in the younger group, except for RBD and S1 binding IgG levels at day 28 of the 20 μ g BNT162b1 dose group.

BNT162b1

In the young group, the RBD bound GMC increased significantly on day 21 after dose 1 to BNT162b1 and further increased 7 days (day 28) after dose 2 to BNT162b1, with higher GMC observed in the 30 μ g dose group compared to the 10 μ g and 20 μ g dose groups (fig. 76). At 1 month after dose 2 (day 52), GMC remained significantly higher than at earlier time points after dose 1.

In the 100 μ g BNT162b1 group, RBD bound GMC increased significantly by day 21 after BNT162b1 and remained at a higher GMC by day 52 than day 7.

In the older group, the RBD bound GMC increased significantly on day 21 after dose 1 to BNT162b1 and further increased 7 days (day 28) after dose 2 to BNT162b1, with higher GMC observed in the 20 and 30 μ g dose groups compared to the 10 μ g group (fig. 77). At 1 month after dose 2 (day 52), GMC remained significantly higher than at earlier time points after dose 1.

In the young (fig. 78) and older (fig. 79) groups, and in the 100 μ g BNT162b1 group, S1 of BNT162b1 bound IgG GMC with results similar to those observed for RBD bound IgG GMC.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

RCDC of RBD and S1 bound IgG levels showed that most participants responded 21 days after dose 1 to BNT162b 1.

BNT162b2

In the young group, S1 bound GMC increased significantly by day 21 after BNT162b2 dose 1 and by day 7 after BNT162b2 dose 2 (day 28), with higher GMC observed in the 20 μ g and 30 μ g dose groups compared to the 10 μ g group (figure 80). 1 month after dose 2 (day 52), the GMC remained significantly higher than the GMC at the earlier time point after dose 1.

A similar trend was observed in the older groups, with higher S1 binding to GMC observed in the 30 μ g dose group compared to the 10 μ g and 20 μ g dose groups (figure 81).

RBD binding IgG GMC results for BNT162b2 were similar to S1 binding IgG GMC results observed in the young (fig. 82) and old groups (fig. 83).

The results of all available immunogenic populations in the young and old groups were similar to those observed in the evaluable immunogenic population.

RCDC of RBD and S1 bound IgG levels after BNT162b2 showed that most participants responded 21 days after BNT162b2 dose 1.

GMFR

Overall, for BNT162b1 and BNT162b2 recipients, and in both age groups, GMFR at RBD bound IgG levels and GMFR at S1 bound IgG levels were significantly higher from pre-vaccination to 21 days post-dose 1, with higher GMFR observed from pre-vaccination to 7 days post-dose 2.

BNT162b1

For the 10 μ g, 20 μ g and 30 μ g doses, GMFR levels of RBD-bound IgG were significantly higher in the young and old groups from before vaccination to day 21 after BNT162b1 dose 1 (before dose 2), with higher GMFR observed from before vaccination to day 7 after BNT162b1 dose 2 (day 28). For both age groups, GMFR remained significantly higher in the 10, 20, and 30 μ g BNT162b1 groups from pre-vaccination to 1 month after dose 2 compared to the earlier time point after dose 1.

In the 100 μ g BNT162b1 group, the level of RBD-bound IgG GMFR was significantly higher from before vaccination to 21 days after BNT162b1 vaccination and remained higher than day 7 GMFR by day 52.

A similar trend was observed for S1 binding IgG levels of GMFR for BNT162b 1.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

BNT162b2

For the 10 μ g, 20 μ g and 30 μ g dose groups, GMFR levels of S1 bound IgG were significantly higher from pre-vaccination to day 21 after dose 1 of BNT162b2 (before dose 2) in the young and old groups, with higher GMFR observed from pre-vaccination to day 7 after dose 2 of BNT162b2 (day 28). For both age groups, all BNT162b2 groups maintained significantly higher GMFR from pre-vaccination to 1 month post-dose 2 compared to the earlier time point after dose 1.

A similar trend was observed for BNT162b2 for GMFR at RBD binding IgG levels.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

Number of participants (%)

Overall, for the BNT162b1 and BNT162b2 recipients, and in both age groups, all participants achieved > 4-fold increases in S1 and RBD bound IgG levels from pre-vaccination to 7 days post-dose 2, except for 1 participant in the young 20 μ g BNT162b1 group.

BNT162b1

In the young group, all participants (except 1 in the 20 μ g dose group) in all dose groups achieved a > 4-fold increase in RBD-bound IgG levels from before vaccination to 21 days after dose 1 of BNT162b 1. All participants in the 20 μ g dose group achieved a > 4-fold increase in RBD-bound IgG levels from pre-vaccination to 14 days post-dose 2 (day 35).

In the older group, all participants in the 20 μ g and 30 μ g dose groups and 8 (72.7%) participants in the 10 μ g dose group achieved a 4-fold increase in RBD-bound IgG levels from pre-vaccination to 21 days after dose 1 of BNT162b 1. All participants in the 10 μ g dose group achieved a > 4-fold increase in RBD-bound IgG levels from pre-vaccination to 7 days post-dose 2 (day 28).

With respect to BNT162b1, a similar trend was generally observed for participants achieving > 4-fold increase in S1 binding IgG levels.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

BNT162b2

In the young groups, all participants in each dose group achieved a > 4-fold increase in S1 bound IgG levels from pre-vaccination to 21 days after dose 1 of BNT162b 2.

In the older group, all participants in the 10. mu.g and 30. mu.g dose groups and 11 (91.7%) participants in the 20. mu.g dose group achieved a 4-fold increase in S1 bound IgG levels from before vaccination to 21 days after dose 1 of BNT162b 2. All participants in the 20 μ g dose group achieved a > 4-fold increase in S1 bound IgG levels from pre-vaccination to 7 days post-dose 2 (day 28).

A similar trend was generally observed for BNT162b2 for participants achieving an increase in RBD binding IgG levels of ≧ 4-fold.

The results for all available immunogenic populations were similar to those observed for the evaluable immunogenic populations in the young and old groups.

SARS-CoV-2 neutralization titer GMR over SARS-CoV-2 antigen-specific binding antibody levels

Overall, for BNT162b1 and BNT162b2 recipients, GMR with SARS-CoV-250% neutralizing titers compared to RBD or S1 binding IgG levels showed more robust RBD or S1 binding levels relative to neutralizing titers, which were similar in each age group.

BNT162b1

21 days after dose 1 of 10. mu.g, 20. mu.g or 30. mu.g, GMR with a SARS-CoV-250% neutralization titer vs. RBD-binding IgG level is < 0.035 in the young group and < 0.183 in the older group. GMR was ≦ 0.032 for the young group and ≦ 0.018 for the older group 14 days after dose 2.

For the 100 μ g dose group, GMR was 0.018 at 21 days after dose 1 and 0.014 at 35 days after dose 1.

After BNT162b1, the GMR with SARS-CoV-250% neutralization titer versus S1 bound IgG level was similar to the GMR with SARS-CoV-250% neutralization titer versus RBD bound IgG level in the young and old groups.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

BNT162b2

21 days after dose 1, the SARS-CoV-250% neutralization titer vs. S1 binding IgG level GMR was ≦ 0.035 in the young group and ≦ 0.124 in the older group. GMR in the young group is 0.040 or less and GMR in the older group is 0.037 or less 14 days after dose 2.

The results for all available immunogenic populations in the young and old groups were similar to those observed for the evaluable immunogenic population.

GMR evaluation of BNT162b1 and BNT162b2

In the young group 21 days after dose 1, GMR with SARS-CoV-250% neutralization titer vs. RBD binding IgG level was ≦ 0.035 after BNT162b1 and ≦ 0.054 after BNT162b 2. After 14 days of dose 2, GMR after BNT162b1 was ≤ 0.032, and GMR after BNT162b2 was ≤ 0.046.

In the older group 21 days after dose 1, GMR with a SARS-CoV-250% neutralization titer vs. RBD-bound IgG level was ≦ 0.183 after BNT162b1 and ≦ 0.196 after BNT162b 2. After 14 days after dose 2, GMR after BNT162b1 was ≦ 0.018, and after BNT162b2 was ≦ 0.043.

In the young group 21 days after dose 1, the SARS-CoV-250% neutralization titer vs. S1 GMR bound IgG level was ≦ 0.061 after BNT162b1 and ≦ 0.035 after BNT162b 2. After 14 days of dose 2, GMR after BNT162b1 was ≤ 0.035, and GMR after BNT162b2 was ≤ 0.040.

In the older group 21 days after dose 1, the SARS-CoV-250% neutralization titer vs. S1 bound IgG level GMR was ≦ 0.328 after BNT162b1 and ≦ 0.124 after BNT162b 2. After 14 days of dose 2, GMR after BNT162b1 was ≦ 0.022, and after BNT162b2 was ≦ 0.037.

Phase 1 summary of the results in assessing immunogenicity of BNT162b1 and BNT162b2

Generally, after the first dose, a moderate neutralizing immune response was observed in the young and old groups. In the early and senior groups, at all dose levels, a much more robust immune response was observed 7 days after the second dose of BNT162b1 or BNT162b 2. The antibody levels tested at the final time points were still significantly higher than the baseline levels.

In the young group:

7 days after dose 2, BNT162b2 recipients were higher than BNT162b1 recipients for SARS-CoV-250% neutralized GMT in the 20 μ g and 30 μ g dose groups. GMTs were similar in the 10 μ g dose group for both recipients. For BNT162b1 and BNT162b2 recipients, GMT remained significantly higher than at earlier time points after dose 1 by 1 month (day 52) after dose 2.

From pre-vaccination to 7 days post dose 2, the neutralizing titer of GMFR for SARS-CoV-250% was significantly high for BNT162b1 and BNT162b2 recipients at the 30 μ g dose level.

All participants receiving BNT162b1 or BNT162b2 achieved a > 4-fold increase in SARS-CoV-250% neutralization titer at a dose level of 30 μ g from pre-vaccination to 7 days post dose 2.

In the senior group:

7 days after dose 2, BNT162b2 recipients were higher than BNT162b1 recipients for SARS-CoV-250% neutralized GMT in the 30 μ g dose group. 1 month after dose 2 (day 52), the BNT162b1 and BNT162b2 recipients neutralized GMT similarly in SARS-CoV-250% in the 30 μ g dose group.

The neutralizing titer of GMFR for SARS-CoV-250% was significantly high in BNT162b1 and BNT162b2 recipients at the 30 μ g dose level from pre-vaccination to 7 days post dose 2.

At a dose level of 30 μ g, most participants receiving BNT162b1 or BNT162b2 achieved a > 4-fold increase in the SARS-CoV-250% neutralization titer from pre-vaccination to 7 days post-dose 2.

Phase 1 conclusions on immunogenicity

7 days after dose 2, both BNT162b1 and BNT162b2 elicited robust SARS-CoV-2 neutralizing antibody responses in young and older adults based on GMT, GMFR, proportion of participants achieving an increase of > 4-fold neutralization titer, and RCDC. Neutralizing antibody responses were maintained by day 52 and were similar in the corresponding age and dose groups of the candidates.

7 days after dose 2, both BNT162b1 and BNT162b2 caused a significant increase in antigen-bound IgG levels based on GMT, GMFR and the proportion of elevated participants that achieved ≧ 4-fold IgG-antigen specific binding. Responses were maintained by day 52.

In the 100 μ g dose group, the SARS-CoV-2 neutralizing antibody response increased moderately by 3 weeks after dose 1 of BNT162b1, but by 7 weeks after dose 1, the neutralizing antibody response returned to a level similar to baseline.

These data support the need for a 2-dose vaccination series.

Stage 2

Immunogenicity was the exploratory endpoint of the phase 2 part of the study.

Stage 3

Immunogenicity is a secondary and exploratory endpoint in the phase 3 portion of the study (12-15 years of age compared to 16-25 years of age).

Example 15: evaluation of safety

In this mid-stage CSR, all participants in stage 1 and the first 6610 participants in stage 2/3 (including 360 participants in stage 2) used an electronic diary to report local reactions and systemic events. 2/3A total of 1125 participants were identified as baseline SARS-CoV-2 positive, defined as having a positive result for the N-binding antibody test or a positive result for the Nucleic Acid Amplification Test (NAAT) on the day of dose 1; of these, 545 received BNT162b2 and 580 received placebo.

Stage 1

Safety data was obtained by date of data expiration (24/8/2020) and summarized at various time points relative to dose 1 or dose 2. Up to 1 month (or data expiration date) after dose 2 of vaccine candidates BNT162b1 and BNT162b2 at dose levels of 10 μ g, 20 μ g, and 30 μ g, the stage 1 safety results for two adult age groups are presented. Based on the data expiration date, the safety results of BNT162b1 at the 100 μ g dose level were shown up to three weeks after dose 1 or up to dose 2 in the young group. Note that, as determined by IRC, 18-55 year old participants who received 100 μ g BNT162b1 did not receive the second dose of 100 μ g BNT162b 2.

Local reaction-1 phase

Overall, injection site pain was the most frequent local reaction for BNT162b1 and BNT162b2 recipients, and in both age groups. Redness and swelling occurred less frequently in the BNT162b2 group and the BNT162b1 group. In BNT162b1 and BNT162b2, the frequency of local reactions was lower in the older group compared to the younger group, and there was a tendency for the frequency of local reactions to be higher with increasing dose.

BNT162b1

In the young group, injection site pain was the most frequently reported local response within 7 days after dose 1 of BNT162b 1. An increase in the frequency of pain at the injection site was observed as the dose level increased from 10 μ g to 30 μ g (58.3% to 100.0%, with 7 and 12 participants, respectively), whereas none was observed in the placebo group (figure 84). Red swelling was reported by 2 (16.7%) participants in the 30 μ g dose group, 3 (25.0%) participants in the 20 μ g dose group and 2 (16.7%) participants in the 30 μ g dose group. In the 100 μ g dose group, injection site pain (12 [ 100.0% ] participants), swelling (5 [ 41.7% ] participants) and redness (4 [ 33.3% ] participants) were reported, and 1 [ 8.3% ] participant had severe injection site pain (note: dose 2 was later given to the participants at the 10 μ g dose level, as determined by IRC).

In the young group, injection site pain was still the most frequently reported local response within 7 days after dose 2 of BNT162b1, with 12 (100.0%) participants in the 30 μ g dose group compared to the placebo group (2 [ 22.2% ] participants), whereas in the 30 μ g dose group the highest proportion of participants with redness (2 [ 16.7% ] participants) and swelling (3 [ 25.0% ] participants) (fig. 84). No redness or swelling was reported in the placebo group.

In the older group, within 7 days after dose 1 of BNT162b1, injection site pain was the most frequently reported local response in the 20 μ g and 30 μ g dose groups (11 [ 91.7% ] participants each) compared to the placebo group (1 [ 11.1% ] participants) (figure 85). No redness was reported and the frequency of swelling (2 [ 16.7% ] participants) was highest in the 30 μ g group. No redness or swelling was reported in the placebo group.

In the older group, injection site pain was the most frequently reported local response in the 20 μ g and 30 μ g dose groups (9 [ 75.0% ] participants each) within 7 days after dose 2 of BNT162b 1. The frequency of swelling (3 [ 25.0% ] participants) was highest at 30 μ g, and redness was reported for the 20 μ g and 30 μ g dose groups (1 [ 8.3% ] participant each). No redness or swelling was reported in the placebo group.

In both age groups, most of the local responses were mild or moderate in severity after the first and second dose, and no grade 4 local response was reported.

Overall, injection site pain was the most frequent local reaction (58.3% -100.0%) with redness (0% -16.7%) and swelling (0% -25.0%) occurring less frequently in BNT162b1 recipients, and in both age groups. It is worth noting that the local response frequency is lower in the older group compared to the younger group, and there is a trend for higher local response frequency with increasing dose.

In the young group, the median day of onset of pain at the injection site was day 1.0 (day of vaccination) after any dose of BNT162b1 between 10 μ g and 30 μ g and after dose 1 of 100 μ g BNT162b 1. The median day of onset of redness and swelling was between days 1.0 and 3.0 in all dose groups.

In the older group, the median day of onset of pain at the injection site was day 1.0 (day of vaccination) after dose 1 of BNT162b1 for all dose groups and after dose 2 for the 20 μ g and 30 μ g dose groups (day 1.5 after dose 2, median day of onset for the 10 μ g dose group). Median onset days for all other regional responses reported for redness or swelling in all dose groups were between days 1.0 and 3.0, except for the 4 th day (20 μ g dose group) and 5 th day (30 μ g dose group) of 1 participant after dose 2.

At all dose levels, the local response subsided in the young and older groups with median duration between 1.0 and 4.0 days.

BNT162b2

In the young group, injection site pain was the most frequently reported local response within 7 days after dose 1, which was the most in the 30 μ g dose group (11 [ 91.7% ] participants) (fig. 86). One [ 8.3% ] participant had severe injection site pain after dose 1 of 30 μ g. Most participants reported no swelling and redness. Following dose 2, injection site pain was still the most frequently reported local response in the 20 μ g and 30 μ g dose groups (83.3%, 10 participants each) compared to the placebo group (2 [ 22.2% ] participants). In any dose group, including the placebo group, no participants reported redness and swelling.

In the older group, injection site pain was reported in all dose groups within 7 days after dose 1 of BNT162b2, and up to 30 μ g dose groups (75.0%, 9 participants), while redness and swelling were not reported in all groups (fig. 87). No local response was reported in the placebo group. Injection site pain was reported in the 30 μ g dose group after dose 2 (8 [ 66.7% ] participants) compared to the placebo group (9 [ 11.1% ] participants); no redness and swelling were reported by participants receiving BNT162b2 or placebo.

After the first and second dose, and in both age groups, the majority of local responses were mild or moderate in severity, and no grade 4 local response was reported.

Overall, injection site pain was the most frequent local reaction (33.3% -91.7%) with less redness (0% -8.3%) and swelling (0% -16.7%) for BNT162b2 recipients, and in both age groups. The local response frequency was lower in the older group compared to the younger group, and there was a tendency for the local response frequency to be higher with increasing dose.

In the young group, the median day of onset of local response occurred between day 1.0 (day of vaccination) and day 2.0 after any dose of BNT162b2 at any dose level. In the senior group, the median day of onset of local response occurred between day 1.0 (vaccination day) and day 2.0 after any dose of BNT162b2 at any dose level. At all dose levels, the local response usually subsided in the young and older groups with median duration between 1.0 and 2.0 days.

Systemic event phase-1

Overall, fatigue is usually the most frequently reported systemic event in the young and old BNT162b1 group and the old BNT162b2 group within 7 days after dose 1; while headache and fatigue were most frequently reported in the young BNT162b2 dose group. Overall, within 7 days after dose 2, headache was the most frequently reported systemic event in the young and old BNT162b1 group, while fatigue was the most frequently reported systemic event in the young and old BNT162b2 group. After dose 2, chills were reported generally more frequently, and the frequency was higher in BNT162b1 than in BNT162b 2. After dose 2, fever was reported more frequently in the young BNT162b1 group than the older BNT162b2 group. For BNT162b1 and BNT162b2 recipients, after the first dose and the second dose, and in both age groups, the severity of most systemic events was mild or moderate, and grade 4 systemic events were not reported.

BNT162b1

Fatigue was the most frequently reported systemic event within 7 days after BNT162b1 dose 1 in the young group, with 4 (33.3%), 8 (66.7%) and 6 (50.0%) participants reporting in the 10 μ g, 20 μ g and 30 μ g dose groups, respectively, compared to the placebo group (2 [ 22.2% ] participants) (fig. 88). Headache (6 [ 50.0% ] participants) and chills (7 [ 58.3% ] participants) were reported in the 30 μ g dose group, with < 1 (8.1%) participants reporting fever in each 30 μ g group. In the placebo group, headache (1 [ 11.1% ] participant) was reported, and no fever or chills were reported. The reporting frequency was higher for the 100 μ g dose group compared to the 30 μ g dose group: fatigue (10 [ 83.3% ] participants), headache (9 [ 75.0% ] participants), chills (10 [ 83.3% ] participants) and fever (6 [ 50.0% ] participants).

Within 7 days after dose 2 of BNT162b1, headache was the most frequently reported systemic event in the young group, reported in all 12 (100.0%) participants in the 30 μ g dose group, none in the placebo group, and 10 (83.3%) and 8 (66.7%) participants in the 30 μ g dose group reported fatigue and chills, respectively. Fever was reported in 17% and 75% of participants in the 20 μ g and 30 μ g dose groups, respectively. In the placebo group, 2 (22.2%) participants reported fatigue, and none reported fever and chills.

Fatigue was the most frequently reported systemic event within 7 days after dose 1 of BNT162b1 in the older group, with 7 (58.3%) and 6 (50.0%) participants reported fatigue in the 20 μ g and 30 μ g dose groups, respectively (fig. 89), compared to 4 (44.4%) participants in the placebo group. In the 30 μ g dose group, headache (6 [ 50.0% ] participants) and chills (2 [ 16.7% ] participants) were reported, while fever was only reported in the 30 μ g dose group (3 [ 25.0% ] participants). Chills (2 [ 22.2% ] participants) were reported in the placebo group, while none reported headache or fever. One participant each reported severe muscle pain (20 μ g dose group) and severe headache (30 μ g dose group) (the former was pain associated with shingles episodes).

In the older group, within 7 days after dose 2 of BNT162b1, headache was the most frequently reported systemic event in the 20 μ g and 30 μ g dose groups (9 [ 75% ] participants each) compared to the placebo group (1 [ 11.1% ] participant). In the 20 μ g and 30 μ g dose groups, 7 (58.3%) and 4 (33.3%) participants reported chills, respectively. Fever was reported in the 20 μ g dose group with 6 (50.0%) participants and in the 30 μ g dose group with 4 (33.3%) participants, of which 1 participant had a fever >38.9 ℃ -40.0 ℃. In the placebo group, fatigue was reported (2 [ 22.2% ] participants), and none reported fever and chills.

After the first and second dose, most systemic events were mild or moderate in severity in both age groups, and no grade 4 systemic events were reported. After each dose, systemic events were suggested to a lesser extent and less frequently in the older group compared to the younger group.

The 100 μ g dose group reported the highest frequency and/or highest severity of systemic events following dose 1. The use of antipyretic/analgesic drugs in both age groups also increased with increasing dose levels and number of doses. For these reasons, IRC decided that participants in the young group should not receive the second dose of 100 μ g BNT162b 1.

In the young group, the median day of onset for most systemic events was between days 1.0 and 2.0 after any dose of BNT162b1 between the 10 μ g-30 μ g doses, and after dose 1 of 100 μ g BNT162b 1. Most systemic events usually resolve with median duration between 1.0 and 2.0 days. For fatigue, median duration was 4.0 days in the 10 μ g dose group after dose 1 compared to 2.0 days in the 30 μ g dose group.

In the senior group, the median onset day of most systemic events was between days 1.0 and 3.5 following any dose of BNT162b1 in any dose group. Most systemic events usually resolve with a median duration of between 1.0 and 3.0 days.

BNT162b2

In the young group, headache (4 [ 33.3% ] to 6 [ 50.0% ] participants) and fatigue (3 [ 25.0% ] to 5 [ 41.7% ] participants) were the most frequently reported systemic events within 7 days after BNT162b2 dose 1, compared to placebo (3 [ 33.3% ] participants each) (figure 90). Fever (2 [ 16.7% ] participants) and chills (4 [ 33.3% ] participants) were reported only in the 30 μ g dose group. On day 7 after dose 1, one participant with a history of migraine in the 30 μ g dose group reported severe migraine.

In the young group, fatigue was the most frequently reported systemic events in the 20 μ g and 30 μ g dose groups (7 [ 58.3% ] and 9 [ 75.0% ] participants, respectively) compared to the placebo group (5 [ 55.6% ] participants) within 7 days after dose 2 of BNT162b 2. Headache (8 [ 66.7% ] participants), chills (7 [ 58.3% ] participants), muscle pain (7 [ 58.3% ] participants) and fever (2 [ 16.7% ] participants) were reported in the 30 μ g dose group. Of these events, fatigue (5 [ 55.6% ] participants), headache (1 [ 11.1% ] participants) and chills (1 [ 11.1% ] participants) were reported in the placebo group, but none reported muscle pain.

In the older group, the most frequently reported systemic event in the 20 μ g and 30 μ g dose groups (4 [ 33.3% ] participants and 3 [ 25.0% ] participants, respectively) was fatigue within 7 days after dose 1 of BNT162b2, compared to the placebo group (2 [ 22.2% ] participants) (fig. 91). Headache (3 [ 25.0% ] participants), chills (2 [ 16.7% ] participants) and muscle pain (1 [ 8.3% ] participants) were the most in the 20 μ g dose group. Among these events, only headache (1 [ 11.1% ] participant) and muscle pain (2 [ 22.2% ] participants) were reported in the placebo group. No fever was reported.

In the older group, fatigue remained the most frequently reported systemic event in the 20 μ g and 30 μ g dose groups (6 [ 50.0% ] and 5 [ 41.7% ] participants, respectively) within 7 days after dose 2 of BNT162b2, compared to the placebo group (1 [ 11.1% ] participant). Headache was reported in the 20 μ g and 30 μ g dose groups (4 [ 33.3% ] and 3 [ 25.0% ] participants, respectively), while muscle pain and chills were reported in the 30 μ g dose group (3 [ 25.0% ] and 2 [ 16.7% ] participants, respectively). Fever was reported in the 30 μ g dose group (1 [ 8.3% ] participant). Among these events, headache and muscle pain were reported in the placebo group (1 [ 11.1% ] participants each).

After the first and second dose, most systemic events were mild or moderate in severity in both age groups, and no grade 4 systemic events were reported.

In the young group, the median day of onset for most systemic events was between days 1.0 and 4.0 after any dose of BNT162b 2. Most systemic events usually resolve with a median duration of between 1.0 and 2.5 days.

In the older group, median onset days for any systemic event were between day 1.5 and day 2.0 after any dose of BNT162b2 at any dose level, except for the systemic event after dose 1 in the 10 μ g dose group, where median onset day was day 5.5. Most systemic events usually resolve with a median duration of between 1.0 and 3.0 days.

Adverse Event (AE) -phase 1

Summary of adverse events phase-1

This summary includes all AEs from dose 1 to data cutoff date 2020, 8, and 24 days for all dose levels and age groups of each vaccine candidate, except the BNT162b 1100 μ g group, where AEs from dose 1 to before dose 2 were summarized.

Overall, fewer participants reported at least 1 AE in the older BNT162b2 group (8.3% -25.0%) compared to the younger (41.7% -50.0%) and older (25.0% -58.3%) BNT162b1 groups and the younger BNT162b2 group (33.3% -41.7%) after dose 1.

BNT162b1

In the young group, 5 (41.7%) to 6 (50%) participants reported at least 1 AE in the dose group up to 30 μ g after dose 1 of BNT162b1, while 2 (22.2%) participants in the placebo group. The associated ae (related ae) increased with increasing BNT162b1 dose levels (25.0% -50.0%); at least 1 relevant AE was reported by 6 (50%) participants in the 30 μ g dose group. Severe AE (fever) was reported in 1 (8.3%) of the 30 μ g dose groups.

In the 100 μ g dose group, 8 (66.7%) participants reported at least 1 AE after dose 1 to before dose 2 of BNT162b1 compared to 1 (33.3%) participant in the placebo group. 6 (50.0%) participants had at least 1 related AE, and 1 (8.3%) participant reported a severe AE (sleep disorder).

In the senior group, 3 (25.0%) participants (30 μ g dose group) and 7 (58.3%) participants (10 μ g and 20 μ g dose group) each reported at least 1 AE after dose 1 of BNT162b1 compared to 4 (44.4%) participants in the placebo group. At least 1 relevant AE was reported by 2 (16.7%) to 4 (33.3%) participants, with the highest frequency in the 20 μ g dose group. Severe AE was reported in 1 participant each in the 20 μ g (shingles) and 30 μ g (fatigue) dose groups.

No SAE, AE causing withdrawal, or death was reported by any age group.

BNT162b2

In the young group, 4 (33.3%) to 5 (41.7%) participants reported at least 1 AE after dose 1 of BNT162b2 compared to 2 (22.2%) participants in the placebo group. At least 1 relevant AE was reported by 2 (16.7%) to 4 (33.3%) participants, with the highest frequency in the 20 μ g dose group. Severe AE (migraine) was reported in 1 participant in the 30 μ g dose group.

In the senior group, 1 (8.3%) to 3 (25.0%) participants reported at least 1 AE after dose 1 of BNT162b2 compared to 2 (22.2%) participants in the placebo group. Only 1 participant (8.3%) reported at least 1 relevant AE (20 μ g dose group). Severe AEs were reported in 1 each of the 30 μ g dose group (muscle spasm) and the placebo group (radiculopathy).

No SAE, AE causing withdrawal, or death was reported by any age group.

Analysis of adverse events-phase 1

Adverse events by System Organ Class (SOC) and Preferred Term (PT) -phase 1

The section summarizes AEs by SOC and PT, including AEs from dose 1 to 1 month after dose 2 for all groups except BNT162b 1100 μ g group, where AEs from dose 1 to 3 weeks after dose 1 or from dose 1 to before dose 2 are summarized.

In the older BNT162b1 group and the younger BNT162b2 group, systemic disease and various reactions at the site of administration are the most commonly reported SOCs. The most commonly reported SOCs are gastrointestinal disease for the young BNT162b1 group and neurological disease for the older BNT162b2 group. In general, most of the PT was reported by ≦ 2 participants per dose group.

BNT162b1

In the young group, gastrointestinal disease (2 [ 16.7% ] participants per dose group) was the most frequently reported SOC in BNT162b1 group of up to 30 μ g from dose 1 to 1 month after dose 2 of BNT162b 1. Paresthesia (3 names [ 25.0% ]) was the most frequent AE per PT in the 20 μ g dose group only. All other AEs were reported by ≦ 2 participants in each dose group, including in the placebo group.

In the 100 μ g dose group, from dose 1 of BNT162b1 to 3 weeks after dose 1, psychiatric disorders were the most frequently reported SOCs (3 [ 25.0% ] participants), while sleep disorders (3 [ 25% ] participants) were the most common AEs per PT. All other AEs were reported by ≦ 2 participants/dose groups, including in the placebo group.

In the older group, from dose 1 to 1 month after dose 2 of BNT162b1, systemic disease and various responses at the site of administration were the most frequently reported SOC in BNT162b1 group, for a total of 6 participants: the 10 μ g dose group had 1 (8.3%) participant, the 20 μ g dose group had 2 (16.7%) participants and the 30 μ g dose group had 3 (25.0%) participants. No more than 1 participant in each dose group reported any AE per PT.

BNT162b2

In the young group, systemic disease and various responses at the site of administration are the most frequently reported SOCs. These events included injection site pain and injection site erythema. No more than 1 participant in each dose group reported any AE per PT.

In the older group, neurological disease was the most frequently reported SOC, with 1 participant in each of the 30 μ g dose group (sciatica) and placebo group (radiculopathy). No more than 1 participant in each dose group reported any AE per PT.

Associated adverse event phase-1

Overall, systemic disease and various responses at the site of administration are the most frequently reported SOCs for the young and older BNT162b1 group and the young BNT162b2 group. In the senior BNT162b2 group, 1 (8.3%) participant reported nausea, which is the only relevant AE.

BNT162b1

In the young group, systemic disease and various reactions at the site of administration were the most frequently reported SOCs (pain at the injection site, fever, chills, fatigue and swelling at the injection site). Two participants (16.7%) in the 30 μ g dose group reported related AEs of tachycardia and fever. All other relevant AEs were reported by ≦ 2 participants/dose group.

In the 100 μ g BNT162b1 group, psychiatric disease (psychiatric disorder) is the most frequently reported SOC. Sleep disorders were reported as psychiatric disorders in 3 (25.0%) participants. All other relevant AEs were reported by ≦ 2 participants for each dose group.

In the older group, systemic diseases and various reactions at the site of administration are the most common SOC (fatigue, spots at the site of injection, pain at the site of injection and swelling around). No more than 1 participant in each dose group reported AE per PT.

BNT162b2

In the young group, systemic disease and various reactions at the site of administration are the most frequently reported SOCs (injection site pain and injection site erythema). No more than 1 participant in each dose group reported any relevant AEs by PT, including in the placebo group.

In the elderly group, only 1 (8.3%) of the participants in the 20 μ g dose group reported related AEs in nausea.

Immediate adverse event phase-1

BNT162b1

In the young group, 1 participant reported an immediate AE of paresthesia after dose 1 of 20 μ g BNT162b 1. In the 100 μ g dose group, no participants reported an immediate AE after dose 1.

In the senior group, 1 participant reported an immediate AE of ocular paresthesia after dose 1 of 10 μ g BNT162b 1.

After dose 2 of BNT162b1, none of the participants reported any immediate AE in any of the age groups.

BNT162b2

In the young group, 3 participants reported immediate AEs after dose 1 of BNT162b 2: injection site erythema (erythema) (10 μ g dose group), taste loss (geusia) (20 μ g dose group) and injection site pain (30 μ g dose group). After dose 2 of BNT162b2, 1 participant reported an immediate AE (20 μ g dose group) of dysgeusia (tast disorder).

In the senior group, none of the participants reported any immediate AE after any dose of BNT162b 2.

Severe adverse event stage-1

BNT162b1

In the young group, 2 days after dose 2(30 μ g dose group), 1 participant reported a severe AE of fever (pyrexia) (102.4 ° F), and 1 day after dose 1 (100 μ g dose group), 1 participant reported a severe AE of sleep disturbance. Both AEs were identified by the investigator as being relevant to study intervention.

In the senior group, 2 participants reported severe AE: shingles occurred 2 days after dose 1 (20 μ g dose group, considered unrelated to BNT162b 1), and fatigue 1 day after dose 2 (30 μ g dose group, considered related to BNT162b 1).

BNT162b2

In the young group, 1 participant with a history of migraine reported severe migraine at 7 days post-dose 1 (30 μ g dose group, considered unrelated to BNT162b 2). In the senior group, 2 participants reported severe AE: muscle spasm 2 days after dose 2 (30 μ g dose group, considered unrelated to BNT162b 2), and radiculopathy 3 days after dose 1 (placebo), considered unrelated to study intervention.

Death, serious adverse events, withdrawal of safety-related participants, and other major adverse events-stage 1 death-stage 1

In this mid-stage CSR, no phase 1 participants died by the data expiration date of 2020, 8-24 days.

Severe adverse event stage-1

During this period of medium-term CSR coverage, no SAE were reported in phase 1 participants.

Security-related participant exit-1 phase

In this mid-term CSR, no phase 1 participants were withdrawn by any AE by the data expiration date of 2020, 8, 24.

Other major adverse events phase 1

For phase 1 of the study, no AE of particular interest was defined.

Other safety evaluation period-1

Severe COVID-19 disease stage-1

No COVID-19 cases were reported in phase 1 participants by the date of data expiration of 2020, 8 months and 24 days.

Stage 1 of pregnancy

No pregnancy was reported in the phase 1 participants by the date of data expiration of 2020, 8, 24.

Analysis and discussion of deaths, serious adverse events, safety-related participant withdrawal, and other major adverse events-stage 1

During this period of medium-term CSR coverage, no SAE, AE causing withdrawal, or death was reported by any age group.

Evaluation in clinical laboratory-stage 1

Overall, in the young and old BNT162b1 and BNT162b2 groups, transient decreases (<0.8 × LLN) of lymphocytes occurred 1 to 3 days after dose 1, which returned to normal 6-8 days after dose 1. The majority of the changes were a decrease in lymphocyte counts from normal or grade 1 to grade 1, 2 or 3, which returned to normal 6-8 days after dose 1 and was observed in all age and dose groups. A reduced change in neutrophils from normal to grade 1 (young BNT162b1 group) or grade 2 (older BNT162b2 group) was also observed, but less frequently.

Overall, other clinical chemistry abnormalities or changes in laboratory results are reported less frequently. The incidence of decreased lymphocyte counts was lower in BNT162b2 recipients compared to BNT162b1 recipients. These laboratory abnormalities were not associated with clinical findings.

BNT162b1

In the young group, laboratory abnormalities of transient lymphocyte reduction (<0.8 × LLN) were observed in 1 (8.3%), 4 (33.3%) and 6 (54.5%) participants 1-3 days after dose 1 of 10 μ g, 20 μ g or 30 μ g BNT162b1, respectively, which returned to normal 6-8 days after dose 1. A change in lymphocyte counts from normal to grade 3 reduction was observed in each of 1 participant in the 10 μ g and 30 μ g dose groups, while 2 (16.7%) were in the 20 μ g dose group. No grade 3 reduction in lymphocyte count was observed 6-8 days after dose 1. After dose 1, a change from normal to grade 2 neutropenia was observed in 1 (11.1%) of the participants in the placebo group, which was no longer observed 19-23 days after dose 1. Changes in neutropenia were observed in 1 participant in each of the 10 μ g dose group (grade 1 to grade 2) and the 30 μ g dose group (normal to grade 2) 6-8 days after dose 2. Approximately 1 month after dose 2, both were shifted to grade 1 at the time of the non-planned visit.

In the 100 μ g BNT162b1 group, 1-3 days after dose 1, laboratory abnormalities of transient lymphocyte reduction (<0.8 × LLN) were observed in 9 (75.0%) participants, which returned to normal 6-8 days after dose 1. A change in lymphocyte counts from normal to grade 3 reduction was observed in 4 (33.3%) participants 1-3 days after dose 1, which returned to normal 6-8 days after dose 1. A change in lymphocyte counts from normal to grade 1 reduction was observed in 3 (25.0%) participants 6-8 days after dose 1, which returned to normal 19-23 days after dose 1.

In the older group, laboratory abnormalities of transient lymphocyte reduction (<0.8 × LLN) were observed in 1 (8.3%), 3 (25.0%) and 2 (16.7%) participants 1-3 days after dose 1 of 10, 20 or 30 μ g BNT162b1, respectively, which returned to normal 6-8 days after dose 1. A change in lymphocyte counts from normal to grade 3 or grade 4 decrease was observed in 1 (8.3%) of each of the participants in the 30 μ g and 10 μ g dose groups 1-3 days after dose 1 of BNT162b1, and both returned to normal 6-8 days after dose 1.

Overall, other clinical chemistry abnormalities or changes in laboratory results are reported less frequently. These abnormalities are not related to clinical findings.

BNT162b2

In the young group, laboratory abnormalities of transient reduction (<0.8 × LLN) of lymphocytes were observed in 1 (8.3%) of each of the participants in the 20 μ g and 30 μ g dose groups 1-3 days after dose 1 of BNT162b2, which returned to normal 6-8 days after dose 1. 1-3 days after dose 1 of BNT162b2, a change in lymphocyte count reduction from normal to grade 1 was observed in 3 (25.0%), 2 (16.7%) and 4 (33.3%) participants of the 10. mu.g, 20. mu.g and 30. mu.g dose groups, respectively, and a change in lymphocyte count reduction from normal to grade 2 was observed in 1 (8.3%) participants of each of the 20. mu.g and 30. mu.g dose groups. No grade 2 or 3 reduction in lymphocyte count was observed 6-8 days after dose 1.

In the older group, laboratory abnormalities of transient lymphocyte reduction (<0.8 × LLN) were observed in 1 (8.3%) of participants 1-3 days after dose 1 of BNT162b2 at all dose levels, which returned to normal 6-8 days after dose 1. After dose 1, a change in lymphocyte counts decreasing from normal to grade 3 (10 μ g dose group) and from grade 1 to grade 3 (30 μ g dose group) was observed in each of 1 (8.3%) participants. A change from normal to grade 2 neutropenia was observed in 2 (16.7%) participants in the 20 μ g dose group 1-3 days after dose 1, to no shift to grade 2 observed 6-8 days after dose 1. A change from normal to grade 2 neutropenia was observed in 1 (8.3%) of the participants in the 10 μ g dose group 6-8 days after dose 1. By day 19-23 after dose 1, no change to grade 2 neutropenia was observed in any of the dose groups.

Overall, other clinical chemistry abnormalities or changes in laboratory results are reported less frequently. The incidence of decreased lymphocyte counts was lower in BNT162b2 recipients compared to BNT162b1 recipients. These laboratory abnormalities were not associated with clinical findings.

Physical examination result-1 stage

Overall, physical examination after BNT162b2 found fewer abnormalities than after BNT162b1 in both age groups. Abnormalities are usually observed 1-3 days after dose 1 and are mostly limb, musculoskeletal system or skin abnormalities.

BNT162b1

In the young group, no abnormalities were found during the baseline physical examination. Overall, most abnormalities were observed 1-3 days after dose 1 (9 [ 20.0% ] participants) and 6-8 days after dose 2 (7 [ 15.6% ] participants) for 10 μ g, 20 μ g, or 30 μ g BNT162b1 after randomization. In the 30 μ g dose group, 1-3 days after dose 1, up to 6 (50.0%) participants had abnormalities, and the majority were limb abnormalities.

In the 100 μ g dose group, only 1 (8.3%) of the participants had abnormalities at baseline. From dose 1 to 3 weeks after dose 1, 9 (75.0%) participants had abnormalities 1-3 days after BNT162b1, and most were limb abnormalities.

In the older group, 5 (11.1%) participants were found to have abnormalities during the baseline physical examination, and 2 or less in any dose group. Overall, most abnormalities (15 [ 33.3% ] participants) were observed 1-3 days after dose 1 of BNT162b1 following randomized cohort. 1-3 days after dose 1, 6 (50.0%) and 4 (33.3%) of the participants in the 20 μ g and 30 μ g dose groups, respectively, had abnormalities, and most of the abnormalities involved in the musculoskeletal system or limbs.

No clinically significant findings were derived from the physical examination.

BNT162b2

In the young group, 5 (11.1%) participants were found to have abnormalities during the baseline physical examination, and 2 or less in any dose group. Overall, after randomization, most abnormalities were observed 1-3 days after dose 1 (5 [ 11.1% ] participants) and 6-8 days after dose 2 (4 [ 8.9% ] participants) for 10 μ g, 20 μ g, or 30 μ g BNT162b2, and most were limb or skin abnormalities.

In the senior group, 1 (8.3%) of the participants in the 30 μ g dose group was found to have abnormalities during the baseline physical examination. After random grouping, the physical examination during any visit window had ≦ 2 participants with abnormalities in any dose group overall.

No clinically significant findings were made from the baseline physical examination.

Phase 1 summary of safety results for evaluation of BNT162b1 and BNT162b2

Overall, reactogenic events were well tolerated and transient (median duration 1.0-4.0 days). All participants returned to receiving the second dose. All reactogenic events resulted in regression of AE with no sequelae.

For local responses in both age groups, BNT162b1 recipients reported injection site pain (58.3% -100.0%), redness (0% -16.7%) and swelling (0% -25.0%), more frequently than BNT162b2 recipients: pain (33.3% -91.7%), redness (0% -8.3%) and swelling (0% -16.7%) at the injection site. In general, local responses are observed more frequently as the dose level increases.

The local reaction frequency was lower in the older group compared to the younger group. The frequency of injection site pain (which is the most frequently reported local response) in the older group after 30 μ g of BNT162b1 (91.7% and 75.0% for dose 1 and dose 2, respectively) and 30 μ g of BNT162b2 (75.0% and 66.7% for dose 1 and dose 2, respectively) was lower than in the younger group after 30 μ g of BNT162b1 (100% for dose 1 and dose 2, respectively) and 30 μ g of BNT162b2 (91.7% and 83.3% for dose 1 and dose 2, respectively).

The BNT162b2 recipients in the senior group reported less frequent local reactions than BNT162b1 recipients in the senior group. The injection site pain frequency after dose 1 (75.0%) and dose 2 (66.7%) was lower in the annual 30 μ g BNT162b2 group than in the annual 30 μ g BNT162b1 group after dose 1 (91.7%) and dose 2 (75.0%).

In both age groups, common systemic events after dose 1 or dose 2 included fatigue (16.7% -83.3%), headache (25.0% -100%), chills (8.3% -66.7%), fever (0% -75.0%) and muscle pain (8.3% -75.0%) for BNT162b1 recipients up to 30 μ g, whereas BNT162b2 recipients up to 30 μ g were less frequent compared: fatigue (8.3% -75.0%), headache (0% -66.7%), chills (0% -58.3%), fever (0% -16.7%) and muscle pain (0% -58.3%). In general, systemic events are observed more frequently with increasing dose levels.

The frequency of systemic events was lower in the older group compared to the younger group. The frequency of fatigue in the older group after 30 μ g BNT162b1 (50.0% and 66.7% for dose 1 and dose 2, respectively) and 30 μ g BNT162b2 (25.0% and 41.7% for dose 1 and dose 2, respectively) was lower than in the younger group after 30 μ g BNT162b1 (50.0% and 83.3% for dose 1 and dose 2, respectively) and 30 μ g BNT162b2 (41.7% and 75.0% for dose 1 and dose 2, respectively).

The BNT162b2 recipients in the senior group reported less frequent systemic events than the BNT162b1 recipients in the senior group. The frequency of fatigue in the 30 μ g annually BNT162b2 group (25.0% and 41.7% for dose 1 and dose 2, respectively) was lower than in the 30 μ g annually BNT162b1 group (50.0% and 66.7% for dose 1 and dose 2, respectively).

Most AEs were mild or moderate in severity. Most related AEs were similar to solicited reactogenicity events reported in electronic diaries. Severe AEs were reported rarely and were considered unrelated to study intervention.

There was no SAE, death or withdrawal due to AE.

In all age and dose groups, a transient decrease in lymphocytes was observed 1-3 days after dose 1, which regressed 6-8 days after dose 1.

No clinically significant findings were derived from the physical examination.

BNT162b2 demonstrated an advantageous reactogenicity and safety profile compared to BNT162b1, facilitating selection of BNT162b2 for stage 2/3 development.

Phase 1 safety conclusions

All tested doses (10 μ g, 20 μ g and 30 μ g) of BNT162b1 and BNT162b2 were safe and well tolerated except for 100 μ g BNT162b1 (terminated after the first dose due to its reactogenicity profile).

The reactogenicity after dose 2 is generally higher than after dose 1.

The frequency of local and systemic reactogenicity for BNT162b2 was generally lower compared to BNT162b1, especially after the second dose.

After each dose of BNT162b1 and BNT162b2, reactogenic events were less severe and less frequent in older adults than in younger adults. Most reactogenic events are mild or moderate in severity.

Most AEs were mild or moderate. There is no SAE or termination due to AE.

Overall, participants who received BNT162b2 experienced fewer AEs than participants who received BNT162b1, while BNT162b2 experienced the least number of participants in the older group. After BNT162b2, severe AEs were rarely observed in the older group and were all considered unrelated to study intervention.

Clinical laboratory evaluations showed that after dose 1, a transient decrease in lymphocytes was observed in all ages and dose groups, which resolved within a few days, which was not associated with any other clinical sequelae and was considered clinically irrelevant.

30 μ g of BNT162b2 was selected for entry into the 2/3 phase portion of the study because the dose and construct provided the best combination of favorable reactogenicity profile and robust immune response.

Stage 2

Security data up to the date of expiration of the data (9.2.2020) is available and is summarized to the date expiration of 360 participants in 2. All participants in phase 2 used an electronic diary to report local reactions and systemic events.

Local reaction-2 phase

Following the first and second doses of BNT162b2, the severity of most local reactions was mild or moderate in both age groups, and grade 4 (potentially life-threatening) local reactions were not reported.

In BNT162b2 group, the young group (fig. 92) reported that injection site pain was more frequent than the older group (fig. 93), and the frequency after dose 1 compared to after dose 2 of BNT162b2 was similar in the young (85.2% and 80.2%, respectively) and older (70.7% and 72.5%, respectively). In the placebo group, the frequency of reported injection site pain was similar (7.8% -10.2%) in the young and old groups after dose 1 and dose 2.

Redness and swelling were similar in the young and old groups after dose 1 in BNT162b2 group. After dose 2, the frequency of redness and swelling was slightly higher in the older group (7.7% and 12.1%, respectively) than in the younger group (3.5% and 3.5%, respectively). In the placebo group, only 1 participant in the senior group reported redness after dose 1, but no swelling.

In BNT162b2 group (older group) 1 participant reported severe injection site pain after dose 1, while in the younger group 1 participant reported severe injection site pain after dose 2. Severe redness was reported after dose 2 in 1 participant in BNT162b2 group (older group).

Overall, injection site pain was the most frequently reported local response in all age groups, with no increase following dose 2, and the frequency of redness and swelling was generally similar after dose 1 and dose 2.

The median onset of local response after any dose of BNT162b2 group was between days 1.0 (day 1.0 was the day of vaccination) and 3.0 in all age groups, and the range was generally similar in the young and old groups. In all age groups, local response declined after a median duration of 1.0-3.0 days after any dose of BNT162b2, which was generally similar in the young and older groups.

Systemic event-phase 2

In BNT162b2 group, systemic events in the young group (fig. 94) were reported more frequently and more severe than in the older group (fig. 95), and frequency and severity increased with increasing number of doses (dose 1vs dose 2). Emesis and diarrhea are exceptions, with emesis being uncommon and similar in both age groups, and emesis and diarrhea similar after each dose. In the young and old BNT162b2 group (dose 1vs dose 2), the frequency of systemic events was as follows:

fatigue: younger group (50.0% vs 59.3%), compared to older group (35.9% vs 52.7%)

Headache: younger group (31.8% vs 51.2%), compared to older group (27.2% vs 36.3%)

Muscle pain: younger group (23.9% vs 45.3%), compared to older group (14.1% vs 28.6%)

And (3) chills: younger group (9.1% vs 40.7%), compared to older group (7.6% vs 20.9%)

Arthralgia pain: younger group (9.1% vs 17.4%), compared to older group (4.3% vs 16.5%)

Fever is caused: younger group (3.4% vs 17.4%), compared to older group (0.0% vs 11.0%)

Vomiting: in both age groups and after any dose were similar.

Diarrhea: fewer reports were reported in the senior group and similar after any dose.

The frequency of reporting systemic events in the placebo group was generally lower than in the BNT162b2 group for both age groups and doses, with some exceptions. In the young group, fever, headache, chills, vomiting and diarrhea were reported after dose 1 and vomiting frequency was similar after dose 2 in the placebo and BNT162b2 groups (fig. 94). In the older group, the frequency of reporting emesis, diarrhea, muscle pain, and joint pain after dose 1 and emesis and diarrhea after dose 2 was similar in the placebo group and BNT162b2 group (fig. 95).

After two doses, the frequency of use of antipyretic/analgesic drugs was slightly lower in the older groups, but overall, in both age groups, increased after dose 2 compared to after dose 1. The frequency of use of antipyretic/analgesic drugs was lower in the placebo group compared to the BNT162b2 group.

After the first and second dose, most systemic events were mild or moderate in severity in both age groups, and no grade 4 (potentially life-threatening) systemic events were reported. In all age groups, severe systemic events including fever (1.1%), fatigue (4.0%), headache (2.8%), chills (2.3%) and muscle pain (1.7%) were generally reported only after dose 2 of BNT162b 2.

The median day of onset of the systemic event after two doses of BNT162b2 was between days 2.0 and 3.0 (day 1.0 is the day of vaccination) in all age groups, and the range was similar in the young and old groups. In all age groups, the systemic events resolved in this group after either dose, with a median duration of 1 day, which was similar in the young and older groups. The duration of the systemic events that occurred after dose 1 was not significantly different from that which occurred after dose 2.

Adverse event period-2

Summary of adverse events phase 2

The number of participants reporting at least 1 AE in BNT162b2 group was similar to that of the placebo group, and in the 2 vaccine groups of the young and old groups, it was also generally similar (table 10 and table 11, respectively).

BNT162b2 2 participants in the young group reported two severe events: myalgia (myalgia) (AE) and gastric adenocarcinoma (SAE). SAE development of gastric adenocarcinoma occurred 23 days after receiving dose 1. Both events were evaluated by the investigator as being unrelated to study intervention.

No other participants reported any AEs from 7 days post dose 2 to the data expiration date (9/2 days 2020).

Analysis of adverse events-phase 2

Associated adverse event by systemic organ type and preferred language-stage 2

Table 12 presents the number of participants reporting at least 1 AE per SOC and PT from dose 1 to 7 days after dose 2.

The number of participants reporting at least 1 AE in BNT162b2 group was similar to the placebo group from dose 1 to 7 days after dose 2.

In the young group, 8 (9.1%) and 10 (11.1%) participants reported at least 1 AE in BNT162b2 and placebo, respectively. In the senior group, 4 (4.3%) and 8 (8.9%) participants reported at least 1 AE in BNT162b2 and placebo, respectively.

Overall, by 7 days after dose 2, most AEs reported were in SOC of gastrointestinal disease (3 [ 1.7% ] in BNT162b2 group, 2[ 1.1% ] in placebo group), systemic disease and various reactions at the site of administration (3 [ 1.7% ] in BNT162b2 group, 7[ 3.9% ] in placebo group, and musculoskeletal and connective tissue disease (3 [ 1.7% ] in BNT162b2 group, 1[ 0.6% ]inplacebo group).

The most frequently reported AE per PT was injection site pain in the young BNT162b2 group (3 out [ 3.4% ]), all of which occurred during the reported period of local response on the vaccination day of dose 1. 2 events resolved within 3 days and 1 event resolved after 11 days. Less than 2 participants in each vaccine group reported all other AEs per PT.

1 participant in the senior BNT162b2 group had contusion AE in the left upper arm deltoid region, which was evaluated by the investigator as being relevant to study intervention.

Associated adverse event by systemic organ type and preferred language-stage 2

From dose 1 to 7 days after dose 2, the frequency of participants evaluated by the investigators as AE associated with study intervention was lower and similar in BNT162b2 and placebo groups. In BNT162b2 group, a similar proportion of participants in the young and older groups reported related AEs. The related AEs evaluated by most investigators were reactogenic events in SOC of various reactions at systemic disease and administration site, reported by a similar proportion of participants in the BNT162b2 group overall, compared to the placebo group, where injection site pain was the most frequently reported PT and occurred only in the BNT162b2 young group.

Immediate adverse event-phase 2

After either dose of BNT162b 230 μ g or placebo, there was no immediate AE.

Severe or life threatening adverse event-phase 2

Both participants (both in the young group of BNT162b 2) reported severe events of myalgia (AE) and gastric adenocarcinoma (SAE). Participants reporting myalgia had scapular pain, which started 2 days after dose 2 and was still on hold at the data cut-off. Both events were evaluated by the investigator as being unrelated to study intervention.

Death, serious adverse events, withdrawal of safety-related participants, and other major adverse events-stage 2 death-stage 2

In this mid-stage CSR, no phase 2 participants died by the data expiration date of 2020, 9/2.

Severe adverse event stage-2

1 participant had SAE from dose 1 to 7 days after dose 2 (table 13). 1 participant in the young BNT162b2 group had SAE of gastric adenocarcinoma 23 days after dose 1, which was evaluated by the investigator as not related to study intervention (table 13). The SAE continued at the data cutoff and due to SAE, the participant exited the study.

No other participants reported any SAE from 7 days after dose 2 to the data expiration date (9/2 days 2020).

Security-related participant exit-2 phase

Participants in gastric adenocarcinoma SAE were reported in the young group BNT162b2, and were withdrawn from the study on day 23 after dose 1 of BNT162b 2.

Narrative-2 phase of security-related participant exit

A description of phase 2 participants who withdrew from the study for SAE by date of data expiration (9/2/2020) is provided.

Other major adverse events-phase 2

No AE of particular interest was defined for phase 2 of the study; however, the target medical event is monitored throughout the study.

Analysis and discussion of deaths, serious adverse events, safety-related participant withdrawal, and other major adverse events-phase 2

By the date of data expiration of 2020, 9/2, 1 participant in the young group (BNT162b2 group) was withdrawn from the study due to gastric adenocarcinoma SAE, which was evaluated by the investigator as not being relevant to study intervention.

Phase 2 safety conclusions

The frequency of local responses after each dose was generally similar in all age groups, and the frequency and severity of systemic events after dose 2 was generally increased compared to after dose 1. Both local and systemic reactogenic events are well tolerated and transient.

After each dose of BNT162b2, reactogenic events were less frequent and less frequent in older adults than those observed in younger adults. Most reactogenic events are mild or moderate in severity. No level 4 events are reported.

The frequency of AEs in participants was low, and the severity of most AEs was mild or moderate. No SAE or investigator evaluated as relevant because of termination of AE.

The reactogenicity and AE profile after BNT162b 230 μ g evaluated in 360 participants was consistent with the safety profile observed after BNT162b 230 μ g in phase 1.

By 7 days post dose 2, 30 μ g BNT162b2 was safe and well tolerated.

Stage 2/3

In this mid-term CSR, the phase 3 safety results included 36,855 older adolescents and adult participants (16-91 years) by the expiration date of the safety data, 10/6/2020. The AE summary includes any AEs reported, regardless of whether the participant completed the visit at 1 month after dose 2. The first 6610 adult participants (18-85 years old, including 360 participants in phase 2) reported local reactions and systemic events using an electronic diary and summarized safety data at least 1 month after dose 2.

During the 2/3 phase portion of the study, if a unilateral probability of equal or more extreme adverse severe case splitting (split) of 5% or less is observed, a stopping rule of theoretical concern for vaccine-enhanced disease will be triggered, and if this probability is less than 11%, a warning criterion should be triggered, given the same true incidence of vaccine and placebo recipients. It should also be noted that 18,000 per arm, the probability of detecting at least 1 adverse event in this study was greater than 83%.

Local reaction period-2/3

In BNT162b2 group, injection site pain was reported more frequently in the young group (fig. 96) than in the older group (fig. 97), and the frequency after dose 1 compared to after dose 2 of BNT162b2 was similar in the young (85.3% and 79.5%, respectively) and older (71.7% and 66.6%, respectively). In the placebo group, injection site pain after dose 1 and dose 2 was reported slightly more frequently in the young group (13.8% and 11.9%, respectively) than in the older group (8.8% and 7.7%, respectively).

In BNT162b2 group, the frequency of redness and swelling was similar in the young and old groups after dose 1 and dose 2. The frequency of redness after dose 1 compared to after dose 2 for BNT162b2 was similar in the young (4.3% and 5.4%, respectively) and older (4.5% and 6.6%, respectively). Dose 1 of BNT162b2 the frequency of swelling after dose 2 was similar in the young (5.5% and 5.9%, respectively) and older (6.5% and 7.0%, respectively). In the placebo group, little redness and swelling was reported in the young (< 0.8%) and older (< 1.3%) groups after dose 1 and dose 2.

Overall, there was no increase in injection site pain after dose 2 and the frequency of redness and swelling was generally similar after dose 1 and dose 2 in all age groups. In BNT162b2 group, after any dose, severe localized reactions (< 0.8%) were reported less overall, but occurred more frequently in the younger group. After the first and second dose, most of the local reactions were mild or moderate in severity in both age groups, and no grade 4 local reactions were reported.

Subgroup analysis

No clinically significant differences in local response were observed depending on country, gender, race or ethnicity.

The median day of onset of local response for BNT162b2 group was between days 1.0-3.0 (day 1.0 is the day of vaccination) in all age groups after any dose, and the range was generally similar in the young and older groups. In all age groups, the local response subsided after any dose, with median duration between 1.0 and 2.0 days, which was similar in the young and older groups.

Systemic event-phase 2/3

The frequency and severity of systemic events generally increased in the young group (fig. 98) compared to the older group (fig. 99), with frequency and severity increasing with the number of doses (dose 1vs dose 2). Emesis and diarrhea are exceptions, and reports of emesis are similar and rare in both age groups, and emesis and diarrhea are similar after each dose. The frequency of systemic events (dose 1vs dose 2) in the young and old BNT162b2 groups was as follows:

fatigue: younger group (49.0% vs 61.6%), compared to older group (34.3% vs 51.2%)

Headache: younger group (42.9% vs 53.1%), compared to older group (25.4% vs 39.5%)

Muscle pain: younger group (22.0% vs. 38.6%), compared to older group (14.0% vs. 28.5%)

And (3) chills: younger group (14.4% vs 36.5%), compared to older group (6.2% vs 22.8%)

Arthralgia pain: younger group (10.9% vs 22.4%), compared to older group (8.3% vs 18.9%)

Fever is caused: younger group (3.7% vs. 16.6%), compared to older group (1.4% vs. 11.5%)

Vomiting: in both age groups and after any dose were similar

Diarrhea: fewer reports were reported in the senior group, similar after each dose.

The frequency of reporting systemic events in the placebo group was generally lower than in the BNT162b2 group for both age groups and doses, with some exceptions. In the young group, the reporting frequency of fever and joint pain (after dose 1) as well as vomiting and diarrhea (after dose 1 and dose 2) was similar in the placebo and BNT162b2 groups (fig. 98). In the senior group, the reporting frequency of fever and joint pain (after dose 1) as well as emesis and diarrhea (after dose 1 and dose 2) was similar in the placebo and BNT162b2 groups (fig. 99).

After two doses, the frequency of use of antipyretic/analgesic drugs was slightly lower in the older group (20.1% -37.4%) than in the younger group (28.1% -45.8%), and in both age groups, drug use increased after dose 2 compared to after dose 1. The frequency of antipyretic/analgesic drug use was lower in the placebo group than in the BNT162b2 group and similar after dose 1 and after dose 2 (9.8% -13.7%) in the young and older placebo groups.

After dose 1 of BNT162b2, the frequency of severe systemic events was generally lower for all age groups than after dose 2: fever (0.1% vs 0.8%), fatigue (0.8% vs 3.7%), headache (0.5% vs 1.9%), chills (0.2% vs 1.7%), muscle pain (0.3% vs 1.6%) and joint pain (0.1% vs 0.6%). The frequency of diarrhea and vomiting is generally similar.

In the placebo group, the frequency of reporting severe fever after dose 1 and dose 2 was similar (0.1%). Only on day 2 post dose 2, 1 participant in the young BNT162b2 group reported a fever of 41.2 ℃, and no fever on all other days of the reporting period.

After the first and second dose, most systemic events were mild or moderate in severity in both age groups, and grade 4 (potentially life-threatening) systemic events were not reported, except that 1 participant in BNT162b2 developed a fever (41.2 ℃) for only 1 day.

Subgroup analysis

No clinically significant differences in systemic events were observed depending on country, gender, race or ethnicity.

The median day of onset for most systemic events was day 2.0 (day 1.0 is the day of vaccination) after any dose of BNT162b2 in all age groups, and the range was generally similar in the young and older groups. In all age groups, all systemic events resolved, with a median duration of 1.0 day, which is generally similar in the young and older groups.

Adverse event period-2/3

In this mid-term CSR, the first 6610 adult participants (including 360 participants in stage 2) aggregated safety data for at least 1 month post dose 2. A summary of AEs for all 36,855 participants by the expiration date (10/6/2020) included any AEs reported, regardless of whether the participants completed the visit at 1 month after dose 2. By the date of the data expiration, a small percentage of participants (< 0.7%) had at least 1 unencoded term (unencoded term).

Summary of adverse events-2/3 phase

First 6610 participants, stage 2/3

Table 14 shows a summary of the first 6610 participants reporting at least 1 AE from dose 1 to 1 month after dose 2.

The number of participants in BNT162b2 reporting at least 1 AE was similar compared to placebo. In both groups, the reporting rates for severe AEs, SAEs, and AEs leading to withdrawal were ≦ 1.1%, 0.5%, and 0.2%, respectively.

In the young and old groups, the number of participants reporting at least 1 AE was similar in BNT162b2 group and the corresponding placebo group from dose 1 to 1 month after dose 2. The reporting rates (rates) of related AEs, severe AEs, SAEs, and AEs leading to withdrawal were also similar in the young and older groups to the corresponding placebo groups.

From dose 1 to the date of data expiration, the first 6610 participants in BNT162b2 group and placebo group that reported at least 1 AE were similar to those in the corresponding group 1 month after dose 2 (table 14). From 1 month after dose 2 to the date of data expiration, at least 1 AE was reported by 4 participants in the young group (3 in BNT162b2 and 1 in the placebo group) and 10 participants in the older group (3 in BNT162b2 and 7 in the placebo group). No other related AEs, severe AEs, SAEs, or AEs leading to withdrawal were reported in any of the groups.

All participants stage 2/3

From dose 1 to the data expiration date, more total participants reported in BNT162b2 from at least 1 AE than placebo. In both groups, the reporting rates for severe AE, SAE and the AE leading to withdrawal were ≦ 0.8%, 0.3% and 0.1%, respectively. Termination due to associated AE was reported by 6 participants in BNT162b2 group and 4 participants in placebo group.

3 stage 3 participants died: BNT162b2 group 1 participant and placebo group 2 participants. Participants who died in BNT162b2 group experienced an SAE in arteriosclerosis, which was evaluated by the investigator as not related to study intervention.

In the young group, BNT162b2 and the placebo group reported at least 1920 (18.1%) and 880 (8.3%) participants with 1 AE, respectively. In the senior group, BNT162b2 and the placebo group reported at least 1 AE as 1166 (14.9%) and 582 (7.4%), respectively.

Analysis of adverse events-phase 2/3

Adverse event by systemic organ type and preferred language-2/3 stage

First 6610 participants, stage 2/3

No grade 1 (tier) AE was identified in this project.

No grade 2 AE was reported from dose 1 to 1 month after dose 2 (event rate ≧ 1.0% defined as any vaccine group [ PT level ]).

Most AEs reported up to 1 month after dose 2 were generally reactogenic and variously responsive in systemic disease and dosing sites (81 [ 2.4% ] in BNT162b2 group, 57[ 1.7% ] in placebo group), musculoskeletal and connective tissue diseases (81 [ 2.4% ] in BNT162b2 group, 56[ 1.7% ] in placebo group), infection and infestation (56 [ 1.7% ] in BNT162b2 group, 48[ 1.5% ] in placebo group), and in SOC of gastrointestinal tract diseases (54[ 1.6% ] in BNT162b2 group, 41[ 1.2% ]inplacebo group) (table 15). In the young BNT162b2 group, the AE rates in these SOCs are: systemic diseases and various reactions at the site of administration (54[ 3.0% ]), musculoskeletal and connective tissue diseases (53[ 3.0% ]), infections and infections (31[ 1.7% ]), and gastrointestinal diseases (32[ 1.8% ]). In the older BNT162b2 group, the AE rates in these SOCs are: systemic diseases and various reactions at the site of administration (27[ 1.8% ]), musculoskeletal and connective tissue diseases (28 [ 1.8% ]), infections and infections (25[ 1.6% ]), and gastrointestinal diseases (22[ 1.4% ]).

In the BNT162b2 group, the most frequently reported AEs per PT were injection site pain (30[ 0.9% ]), headache (30[ 0.9% ]), and fatigue (27[ 0.8% ]) (table 15), and during this period (from dose 1 to 1 month after dose 2), most of these AEs were reported within the 1 week reporting period of the electronic diary. Most of these PTs were reported in the young group: headache (21[ 1.2% ]) and fatigue (17[ 1.0% ]). The frequency of injection site pain reporting was similar in the young (16[ 0.9% ]) and older (14[ 0.9% ]) groups.

In BNT162b2 group, 10 (0.3%) participants reported lymphadenopathy AE: 6 in the young group and 4 in the older group, but not in the placebo group; 1 (0.1%) was male and 9 (0.5%) was female. The AE of lymphadenopathy occurred in the arm and neck regions (axilla, left paraclavicular, left supraclavicular, bilateral cervix or unspecified lymph nodes). Most lymphadenopathy events were reported within 2 to 4 days after vaccination (2 events were reported 8 days after vaccination). Wherein 5 events last ≦ 4 days, 3 events last 12-16 days, and 2 events are still persisting at the data expiration.

In the young group, 13 days after dose 1, 1 participant each (BNT162b2 group) reported angioedema AE (both eyes) and hypersensitivity (allergic onset [ no other information at this report ], not related to study intervention) and 1 participant (placebo group) reported drug hypersensitivity (oral penicillin reaction) AE. These practices were evaluated by the investigator as being unrelated to study intervention. 3 participants in the young BNT162b2 group reported appendicitis, while 1 participant in the older placebo group had perforated appendicitis; these were all assessed by the investigator as being independent of study intervention.

All participants stage 2/3

By the date of data expiration, there were at least 1 unencoded term for a total of 121 (0.7%) participants in the BNT162b2 group and 51 (0.3%) participants in the placebo group for all 36,855 participants. As a result, there are also unencoded terms in other AE tables that are summarized according to SOC and PT.

From dose 1 to the data expiration date, the BNT162b2 group reported a greater total number of participants with AEs of at least 1 (3086[ 16.8% ]) compared to the placebo group (1462[ 7.9% ]). From dose 1 to the data expiration date, most AEs reported by all participants were reactogenic and in SOC of systemic disease and various reactions at the site of administration (1941 [ 10.5% for BNT162b2 group, 438[ 2.4% for placebo group), musculoskeletal and connective tissue diseases (742 [ 4.0% for BNT162b2 group, 227[ 1.2% for placebo group), and neurological diseases (567 [ 3.1% for BNT162b2 group, 251[ 1.4% for placebo group).

In the BNT162b2 group, the most frequently reported AEs to PT were injection site pain (1222[ 6.6% ]), fever (504[ 2.7% ]), fatigue (481[ 2.6% ]), headache (470[ 2.6% ]), chills (458[ 2.5% ]), and myalgia (454-out [ 2.5% ]). Most of these PTs were reported in the young group: pain (787[ 7.4% ]), fever (351 [ 3.3% ]), fatigue (309[ 2.9% ]), headache (303[ 2.9% ]), chills (316[ 3.0% ]) and myalgia (304[ 2.9% ]) at the injection site.

Except for the first 6610 participants, events related to reactogenicity were no longer reported using an electronic diary, but were reported as AEs. Therefore, post hoc analysis was performed to assess the AE imbalance observed in the population of participants from dose 1 to the date of data expiration, which was not observed in the first 6610 participants from dose 1 to 1 month after dose 2, whether due to reactogenic events. The analysis examined the AE reported within 7 days after each dose, which represents the reactogenicity reporting period. The reason that this time period was chosen was that many AEs were reported in SOC of systemic diseases and various reactions at the site of administration, musculoskeletal and connective tissue diseases and neurological diseases, including AEs consistent with reactogenic events, and could be attributed only to reactogenicity if they occurred within this time period rather than at most one month after each dose.

From dose 1 to 7 days after dose 1 (data expiration date), 1494 (8.1%) of the BNT162b2 groups reported at least 1 AE, which was about half of the total of 3086 [ 16.8% ] participants who reported at least 1 AE by the data expiration date. In the placebo group, 555 (3.0%) participants reported at least 1 AE from dose 1 to 7 days after dose 1, while a total of 1462 (7.9%) participants reported at least 1 AE by the date of data expiration.

From dose 2 to 7 days after dose 2 (data expiration date), 1165 (6.3%) of the BNT162b2 groups reported at least 1 AE, which accounted for 3086 [ 16.8% ] of the total number of participants reporting at least 1 AE by the data expiration date about 38%. From dose 2 to 7 days after dose 2, fewer participants reported AEs in the placebo group than in the BNT162b2 group. In the placebo group, from dose 2 to 7 days after dose 2, 268 (1.5%) participants reported at least 1 AE, while a total of at least 1 AE was reported by the date of data expiration as 1462 (7.9%) participants.

AEs in SOC of systemic disease and various reactions at the site of administration were reported from dose 1 to 7 days after dose 1 (1127 [ 6.1% ] in BNT162b2 group, 251[ 1.4% ] in placebo group) which accounted for more than half of the total number of participants reporting AEs in this SOC by at least 1 by the data cutoff date (1941 [ 10.5% ] in BNT162b2 group, 438[ 2.4% ]inplacebo group). Musculoskeletal and connective tissue diseases (252 [ 1.4% ] in BNT162b2 group, 76[ 0.4% ] in placebo group) and neurological diseases (220 [ 1.2% ] in BNT162b2 group, 115 [ 0.6% ] in placebo group) were also frequently reported, accounting for a small proportion of the total number of participants reporting AEs in these SOCs.

In the BNT162b2 group, the most frequently reported AEs at PT from dose 1 to 7 days after dose 1 were injection site pain (881[ 4.8% ]), fatigue (231[ 1.3% ]), headache (181[ 1.0% ]), myalgia (147[ 0.8% ]), fever (110[ 0.6% ]), and chills (100[ 0.5% ]). Most of these PTs were reported in the young group: pain (566 [ 5.3% ]), fatigue (153[ 1.4% ]), headache (118[ 1.1% ]), myalgia (99[ 0.9% ]), fever (82[ 0.8% ]), and chills (75[ 0.7% ]) at the injection site. From dose 1 to 7 days after dose 1, the reported injection site pain (881[ 4.8% ]) accounted for a large proportion of the total number of participants reporting AE for this PT (1222[ 6.6% ]).

From dose 2 to 7 days after dose 2, AEs in SOC were reported for systemic disease and various reactions at the site of administration (828 [ 4.5% ] in BNT162b2 group, 93[ 0.5% ] in placebo group), musculoskeletal and connective tissue diseases (377 [ 2.0% ] in BNT162b2 group, 38[ 0.2% ]inplacebo group), and neurological diseases (294 [ 1.6% ] in BNT162b2 group, 40[ 0.2% ]inplacebo group). From dose 2 to 7 days after dose 2, musculoskeletal and connective tissue diseases and neurological diseases reported accounted for at least half of the total number of participants reporting at least 1 AE in these SOCs.

In the BNT162b2 group, the most frequently reported AEs at PT from dose 2 to 7 days after dose 2 were fever (375[ 2.0% ]), chills (327[ 1.8% ]), injection site pain (313[ 1.7% ]), myalgia (282[ 1.5% ]), headache (258[ 1.4% ]), and fatigue (227[ 1.2% ]). Most of these PTs were reported in the young group: fever (251[ 2.4% ]), chills (216[ 2.0% ]), myalgia (185[ 1.7% ]), injection site pain (183[ 1.7% ]), headache (154[ 1.5% ]) and fatigue (134[ 1.3% ]). From dose 2 to 7 days after dose 2, the majority of AEs reported for these PTs accounted for at least half of the total number of participants reporting AEs for these PTs: fever (504[ 2.7% ]), chills (458[ 2.5% ]), myalgia (454 [ 2.5% ]), headache (470[ 2.6% ]) and fatigue (481[ 2.6% ]).

Overall, the reported AEs were largely due to reactogenic events from dose 1 to 7 days post dose 1, and from dose 2 to 7 days post dose 2. This observation provides a reasonable explanation for the overall higher observed AE rates for BNT162b2 group than placebo group.

From dose 1 to the data expiration date, the BNT162b2 group had 44 (0.2%) participants reporting the AE for lymphadenopathy, including the report in the first 6610 participants (10 [ 0.3% ]). By the date of data expiration, 34 other participants in BNT162b2 group and 4 other participants in the placebo group reported an AE for lymphadenopathy. In BNT162b2 group, 34 (0.3%) participants in the young group and 10 (0.1%) participants in the older group reported lymphadenopathy, while 4 (0.0%) in the placebo group (3 in the young group and 1 in the older group). Lymphadenopathy occurs primarily in the arm and neck regions, with most events reported in the left axillary lymph nodes. Most lymphadenopathy events occurred after dose 2, with < 3 days after dose 1 or dose 2, with grade 1 or grade 2 severity, which resolved by 32 of the events by the data expiration date 48. In the young BNT162b2 group, grade 1 lymphadenopathy (right axillary lymphadenectasis) was reported by 1 participant as an immediate AE, occurring after dose 1 and continuing at the date of data expiration.

In the young group, 1 participant in each of BNT162b2 group (SAE) and placebo reported an AE suspected of being COVID-19.

In the BNT162b2 group, 6 participants reported an immune response assessed to be associated with study intervention (vaccine response or systemic vaccine response [ no other information until this report at present ]). In addition to 1 participant in the placebo group reporting drug hypersensitivity, 3 participants in the BNT162b2 group also reported drug hypersensitivity. Drug hypersensitivity (anaphylaxis) was assessed as being associated in 1 participant in group BNT162b2, while drug hypersensitivity (drug allergy or anaphylaxis to dipyridamole) in 2 participants in group BNT162b2 was considered to be unrelated to study intervention.

At least 1 vaccine complication (mostly a description of reactogenic events) was reported in 19 (0.1%) participants in BNT162b2 group (14 in the young and 5 in the older group), and not in the placebo group. All these were evaluated as relevant to the study intervention and included: myalgia, fever, physical soreness, headache, chills, nausea, adverse reactions, arthralgia (arthralgia), fatigue, soreness (ache), muscle soreness, weakness (malaise) and pain in the left shoulder (sore soft shoulder) after vaccination. Most events were grade 1, starting within 3 days of vaccination and lasting 1-3 days.

Of the first 6610 participants, an additional 3 participants in BNT162b2 reported appendicitis (including 1 participant with perforated appendicitis) for all participants from dose 1 to the date of data expiration, except 4 participants with appendicitis (including 1 participant with perforated appendicitis) in the placebo group. Thus, 6 participants in the BNT162b2 group reported appendicitis (including 1 perforated appendicitis), with 4 in the young group and 2 in the senior group, and 1 in the placebo group (senior group) reported appendicitis (perforated). All events were severe or life-threatening, but were evaluated as unrelated to study intervention.

Adverse event by systemic organ type and preferred language-2/3 stage

First 6610 participants, stage 2/3

From dose 1 to 1 month after dose 2, 135 (4.1%) of the BNT162b2 groups reported at least 1 AE evaluated as relevant by the investigator and 68 (2.1%) of the participants reported at least 1 relevant AE in the placebo group. Most of the related AEs were reactogenic events and were in SOC (69 [ 2.1% ] in BNT162b2 group and 40[ 1.2% ] in placebo group) of systemic disease and various reactions at the site of administration.

Of the 10 participants, 8 reported lymphadenopathy AEs were evaluated by the investigator as being relevant to study intervention.

All participants stage 2/3

From dose 1 to the data expiration date, 2303 participants (12.5%) in BNT162b2 group and 593 participants (3.2%) in the placebo group reported related AEs evaluated by the investigators for at least 1, including the first 6610 participants. Most of the related AEs were reactogenic events and were in SOC of various reactions at systemic disease and site of administration (1869 [ 10.1% ] in BNT162b2 group and 365[ 2.0% ] in placebo group).

AEs for reported lymphadenopathy were evaluated by investigators as being related to study intervention in 30 of 44 participants in BNT162b2 group and 2 of 4 participants in placebo group.

In the BNT162b2 group, according to all the information currently available at the time of this report:

the immune response (vaccine response or systemic vaccine response) evaluated as being associated with study intervention was reported by 6 participants. In most participants, the immune response occurred 1 or 2 days after dose 2, lasting 2 or 3 days (1 participant was recovering at the date of data expiration), with a severity of grade 1 or 2. In 1 of the participants, the immune response (systemic vaccine response) occurred 2 days (grade 1) and lasted 2 days after dose 1, and 1 day (grade 3) and lasted 4 days after dose 2.

AE reported by 1 participant for drug hypersensitivity (anaphylaxis), urticaria (anaphylaxis) and headache, all on level 2, and were evaluated by the investigator to be associated with study intervention. AE of both drug hypersensitivity and urticaria occurred within 1 day after dose 1 and resolved on the same day. AE for headache occurred the next day after vaccination and continued for 4 days.

Immediate adverse event period-2/3

First 6610 participants, stage 2/3

After dose 1, 0.3% or less of the participants reported an immediate AE. Most immediate AEs are in systemic disease and SOC of various reactions at the site of administration and are events associated with injection site reactions (injection site pain, injection site erythema and injection site swelling).

After dose 2, 0.1% of participants in each group reported immediate AE. Most immediate AEs are in systemic disease and SOC of various reactions at the site of administration and are events associated with injection site reactions (injection site pain, injection site hypersensitivity (peraesthesia) and injection site itching).

After any dose of BNT162b2, none of the participants reported an immediate allergic response to the vaccine.

All participants stage 2/3

After dose 1, 0.3% of participants in each group reported immediate AE. Most immediate AEs were in SOC of systemic disease and various responses at the site of administration, and most events were associated with injection site reactions, with injection site pain being most frequently reported (40 [ 0.2% ] participants in BNT162b2 group and 27 (0.1%) participants in placebo group). Immediate AE of lymphadenopathy occurred in 1 participant after dose 1. All other immediate AEs were each reported by ≦ 3 participants in the BNT162b2 group.

After dose 2, 0.1% of participants in each group reported immediate AE. Most immediate AEs were in SOC of systemic disease and various responses at the site of administration, and most events were injection site reactions, with injection site pain most frequently reported (10 [ 0.1% ] participants in BNT162b2 group and 7 [ 0.0% ] participants in placebo group). All other immediate AEs were reported by ≦ 2 participants each.

After any dose of BNT162b2, none of the participants reported an immediate allergic response to the vaccine.

Severe or life threatening adverse event-period 2/3

First 6610 participants, stage 2/3

Severe AE were reported from 35 (1.1%) participants in BNT162b2 group and 19 (0.6%) in placebo group from dose 1 to 1 month after dose 2.

From dose 1 to 1 month after dose 2, 4 (0.1%) participants in BNT162b2 group and 7 (0.2%) participants in placebo group had at least 1 life-threatening AE. These events were evaluated by the investigator as being independent of study intervention.

In BNT162b2 set:

stage 2, 1 participant had a severe gastric adenocarcinoma event (SAE), which was discussed in the previous section.

2 participants had a severe appendicitis event: one event started 9 days after dose 1 and another started 15 days after dose 2 (SAE), which was evaluated by the investigator as being irrelevant for study intervention.

2 life-threatening AEs (all SAE) with appendicitis and celiac abscess in 1 participant 7 days after dose 1; these 2 events were all evaluated by the investigator as unrelated to study intervention.

1 participant had 8 serious events: anemia, congestive heart failure, abdominal adhesions (abdominal adhesion), sepsis, hypokalemia, mental state changes, acute kidney injury, and acute respiratory failure (all SAE). These events were evaluated by the investigator as being unrelated to study intervention.

All participants stage 2/3

Severe AEs reported by date expiration, including those discussed for the first 6610 participants, were reported by 142 [ 0.8% ] participants in the BNT162b2 group and 70 (0.4%) participants in the placebo group. Other events include:

there were 2 participants in BNT162b2 group who had a severe event of appendicitis: one event started 17 days after dose 1 and another started 11 days after dose 1 (SAE), which was evaluated by the investigator as being irrelevant for study intervention.

In group BNT162b2, 1 participant had a severe event of perforated appendicitis (SAE) on the same day following dose 1, which was assessed by the investigator as not related to study intervention.

From dose 1 to the data expiration date, 9 participants (0.0%) in BNT162b2 group and 12 participants (0.1%) in the placebo group reported at least 1 life-threatening AE, including those discussed for the first 6610 participants. These events were evaluated by the investigator as being unrelated to study intervention.

Death, serious adverse events, withdrawal of safety-related participants, and other major adverse events-2/3 stage death-2/3 stage

By date of data expiration of 2020, 10/6, 3 of the phase 3 participants (1 in BNT162b2 group and 2 in placebo group) died. None of these deaths occurred in the first 6610 participants (table 14) and were all evaluated by the investigator as unrelated to study intervention.

1 participant in the older BNT162b2 group experienced a grade 4 SAE of arteriosclerosis 4 days after dose 1 and died 15 days after dose 1.

1 participant in the young placebo group experienced a grade 4 SAE (of unknown origin [ no other information so far as reported) that failed to evaluate the event 8 days after dose 1 and died on the same day.

1 participant in the senior placebo group experienced a grade 4 SAE of hemorrhagic stroke 15 days after dose 2 and died 35 days after dose 2.

Description of death

Narratives were provided for participants who died by the date of data expiration (10/6/2020).

Severe adverse event stage 2/3

First 6610 participants, stage 2/3

From dose 1 to 1 month after dose 2, at least 1 participant count of SAE was reported to be similar in BNT162 group (18 [ 0.5% ]) and placebo group (17 [ 0.5% ]) (table 16). These SAEs were all evaluated by the investigator as being independent of study intervention. Most of the SAE per PT were reported by only 1 participant (3 participants reported SAE for appendicitis).

From dose 1 to 1 month after dose 2, it was reported that the number of participants of SAE was similar in the young group and the old group from at least 1.

In BNT162b2 set:

2 participants had SAE of appendicitis: one starting day 9 days after dose 1 and another starting day 15 days after dose 2.

7 days after dose 1, 1 participant had SAE with appendicitis and celiac abscess, respectively, which was considered life-threatening. Both events lasted 17 days.

17 days after dose 1, 1 participant had 8 SAEs: anemia, congestive heart failure, abdominal adhesions, sepsis, hypokalemia, changes in mental status, acute kidney injury, and acute respiratory failure (all severe). The SAEs of abdominal adhesions and acute respiratory failure lasted 2 and 14 days, respectively. All other SAEs lasted 19 days.

At 9 days after dose 2, 1 participant had SAE with an allergic response due to bee stinging, which was considered life-threatening. The event subsided on the same day.

In the placebo group, 1 participant had SAE (both severe) with perforated appendicitis and peritonitis 13 and 15 days after dose 2, respectively. These two events lasted 4 days and 5 days, respectively.

No other SAE were reported by the first 6610 participants from 1 month after dose 2 to the date of data expiration.

All participants stage 2/3

From dose 1 to the data expiration date, including those discussed for the first 6610 participants, the number of participants reporting SAE from at least 1 was similar in BNT162b2 group (63 [ 0.3% ]) and placebo group (49 [ 0.3% ]) (table 17). Other events include:

in BNT162b2 group, the respective SAE among 2 participants in the young group were evaluated by the investigator as being related to study intervention:

13 days after dose 1, 1 participant reported SAE of lymphadenopathy (right axilla) that did not resolve by the time the data was over. The participants were 48 year old women with a history of eczema and related topical use of crisabonole, given BNT162b2 vaccine in their left deltoid and had right axillary pain and lymphadenopathy. Her right arm was not injured, had no fever, and had no history of similar events. Her WBCs were normal, lymphocyte counts were normal, and right axillary ultrasound showed 4 enlarged lymph nodes (max 2.5 × 1.1 × 2.4 cm). Biopsies were performed and reported as normal, without markers for lymphoma or other cancers. A follow-up of the oncology (and possibly repeated ultrasound examination) is planned for 3 months.

1 participant had a shoulder-injured SAE (SIRVA, erroneously administered into or near the shoulder capsule) associated with vaccine administration after dose 2, which was recovering at the data cutoff.

From dose 1 to the data expiration date, a total of 6 participants in the BNT162b2 group reported SAE for appendicitis. Three of these SAE of appendicitis occurred in the first 6610 participants. An additional 3 appendicitis SAEs are described below, as well as other designated SAEs evaluated as unrelated to the study intervention of BNT162b2 group:

2 participants had SAE of appendicitis: 1 event started 17 days after dose 1 and lasted 3 days (young group), and another event started 11 days after dose 1 and lasted 5 days (older group).

On the same day after dose 1, 1 participant in the older group had SAE of perforated appendicitis, which resolved at the data expiration.

On the same day after dose 2, 1 participant in the young group had an SAE suspected of being COVID-19, which lasted 6 days. The nasal swab results were negative.

Narrative of Severe adverse events-2/3 stage

A statement is provided regarding phase 3 participants who reported SAE assessed by the investigator as being relevant to study intervention and completed the visit from 1 month after dose 2 to the date of data expiration (10 months and 6 days of 2020).

Security-related participant exit-2/3 phase

First 6610 participants, stage 2/3

From dose 1 to 1 month after dose 2, 6 (0.2%) participants in BNT162b2 group and 5 (0.2%) participants in placebo group were withdrawn by AE (table 18), and no other withdrawal of these participants was reported from 1 month after dose 2 to the data expiration date.

Exit of interest in BNT162b2 group:

2 participants were withdrawn by the investigator as an AE related to study intervention. 8 days after dose 1, 1 participant in the young group had an AE of myalgia, which was recovering at the data cutoff. 2 days after dose 1, 1 participant in the older group had an AE of cutaneous pruritus and an AE of tachycardia; both events were 1 day in duration and were severe.

Each of the 3 participants had SAE and exited the study: young (gastric adenocarcinoma) and older (coronary artery disease and coronary dissection).

Withdrawal of interest in the placebo group:

2 days after dose 1, 1 participant (young group) was withdrawn due to AE of vaccine allergy (study intervention) and AE of erythematous rash; both AEs resolved after 18 days and were evaluated by the investigator as being relevant to study intervention.

1 participant in the older group had an SAE (coronary occlusion) that was evaluated as irrelevant by the investigator and exited the study.

10 days after dose 1, 1 participant in the older group was withdrawn from the study due to AE in urticaria. The event resolved on the day and was evaluated by the investigator as not being relevant to study intervention.

All participants stage 2/3

From dose 1 to the data expiration date, 18 (0.1%) participants in BNT162b2 group and 14 (0.1%) participants in the placebo group were withdrawn from the study by AE. In addition to the exits discussed for the first 6610 participants, other exits include:

13 days after dose 1, 1 participant in the young BNT162b2 group had an SAE evaluated by the investigator as lymphadenopathy associated with study outcome (right axilla) and exited, which did not resolve by the time the data was cut off.

There were 3 participants in the young group (1 BNT162b2 and 2 placebo) who were pregnant and withdrawn after dose 1.

At day 39 after dose 1, pregnancy test results were positive (exposure during pregnancy) and dropped out for 1 participant in the young placebo group.

Narrative of exit of Security-related participants-2/3

Narratives provided to the data expiration date (10/6/2020) concerning participants at stage 2/3 with any AEs that led to withdrawal from the study.

Other major adverse event-period 2/3

No AE of particular interest was defined for phase 2/3 of the study; however, the target medical event is monitored throughout the study.

Other safety evaluation period-2/3

Severe COVID-19 disease-2/3 stage

At mid-efficacy analysis by 2020, 11/4 days, all 7 severe COVID-19 cases were reported in the placebo group.

Pregnancy period-2/3

By date of data expiration of 2020, 10, 6, 5 stage 3 participants reported pregnancy: 1 participant in BNT162b2 group and 4 participants in the placebo group. Incomplete spontaneous abortion (incomplete spontaneous abortions) occurred in 1 participant in the placebo group.

A description of pregnancy is provided.

Analysis and discussion of deaths, serious adverse events, safety-related participant withdrawal, and other major adverse events stage 2/3

By the data expiration date of 2020, day 6/10, the number of SAEs in BNT162b2 group (63[ 0.3% ]) and placebo group (49 [ 0.3% ]) were similar. There were 2 participants in the BNT162b2 group who reported SAE assessed by the investigator as being relevant to the study intervention.

Very few participants in BNT162b2 group (18 [ 0.1% ]) and placebo group (14 [ 0.1% ]) were withdrawn by AE.

There were 3 deaths (1 in BNT162b2 group and 2 in placebo group); these deaths were all rated by the investigator as not being relevant to study intervention.

2/3 phase safety conclusion

The frequency of local responses after each dose was generally similar in all age groups, and the frequency and severity of systemic events generally increased after dose 2 compared to after dose 1. Both local and systemic reactogenic events are well tolerated and transient (median duration of 1.0-2.0 days).

After each dose of BNT162b2, the reactogenic events were generally milder and less frequent in older adults than those observed in younger adults. Most reactogenic events are mild or moderate in severity. No grade 4 events were reported, except for fever in 1 participant in BNT162b2 group, which started 1 day after dose 2 and continued for 1 day.

The reactogenicity and AE profile after BNT162b 230 μ g evaluated in 6610 participants was consistent with the safety profile observed after BNT162b 230 μ g in phase 1 and phase 2.

AEs were reported in 16.8% of participants in BNT162b2 group, and most AEs were mild or moderate in severity. At the data expiration date, there were more participants with AEs in BNT162b2 group compared to placebo group (7.9%), after analysis due to AEs reported as reactogenic events within 7 days after each dose.

At the date of data expiration, there were 2 related SAEs in BNT162b2 group (lymphadenopathy and shoulder injury associated with vaccination (SIRVA, incorrectly administered into or near the shoulder joint capsule), and 6 end-offs due to the associated AE 1 death (arteriosclerosis) in BNT162b2 group and 2 deaths in placebo group were evaluated as not relevant to study intervention.

Overall, 30 μ g BNT162b2 was well tolerated according to the dosing schedule, when measured up to 1 month after dose 2.

Example 16: conclusion of the COVID-19 vaccine phase 3 study, all major efficacy endpoints were reached

After the final efficacy analysis of the ongoing phase 3 study was performed, the mRNA-based COVID-19 vaccine BNT162b2 reached all the primary efficacy endpoints of the study. Analysis of the data showed that the vaccine efficacy rate was 95% (p <0.0001) in participants who had not previously been infected with SARS-CoV-2 (first primary target) and in participants who had and had not previously been infected with SARS-CoV-2 (second primary target), each measured 28 days after the first dose and 7 days after the second dose. The first major objective analysis was based on 170 COVID-19 cases, of which 162 COVID-19 cases were observed in the placebo group and 8 COVID-19 cases were observed in the BNT162b2 group. Efficacy is consistent in age, gender, race and ethnic demographics. Efficacy of over 94% was observed in adults over 65 years of age.

10 severe COVID-19 cases were observed in the trial, 9 of which occurred in the placebo group and 1 in the BNT162b2 vaccination group. No serious safety issues associated with vaccines have been reported. Review of the non-blind reactogenicity data from the final analysis (consisting of a random subset of at least 8,000 participants aged 18 and older in the 2/3 phase study) indicated that the vaccine was well tolerated and that most of the adverse events solicited resolved soon after vaccination. The only grade 3 (severe) adverse events with a frequency greater than or equal to 2% following either the first or second dose were fatigue at 3.8% and headache at 2.0% following dose 2. Consistent with previously shared results, older adults tend to report fewer and less demanding adverse events after vaccination. The local reactogenicity profile of the SARS-CoV-2 positive participant is consistent with the overall reactogenicity subset; similarly, comparing AE data to that of "all subjects", there is no indication of a poor safety profile in baseline positive participants. Indeed, there is no indication of a poor safety profile in baseline positive participants; thus, BNT162b2 can be used regardless of the history of COVID-19 or the serological status of SARS-CoV-2.

In addition, the U.S. Food and Drug Administration (FDA) has reached the safety milestone required for Emergency Use Authorization (EUA).

This first global trial to reach the final efficacy analysis marker indicates that a high protection rate against COVID-19 can be achieved quickly after the first 30 μ g dose, highlighting the potential of BNT162 to provide early protection.

To summarize:

preliminary efficacy analysis showed that BNT162b2 was 95% effective against COVID-19 starting 28 days after the first dose; the evaluation was carried out in 170 diagnosed COVID-19 cases, 162 were observed in the placebo group and 8 in the vaccine group

Efficacy is consistent in age, gender, race and ethnic demographics; efficacy observed in adults over 65 years of age of 94% or more

Has reached the safety data milestone required by the U.S. Food and Drug Administration (FDA) for Emergency Use Authorization (EUA)

The data indicate that the vaccine is well tolerated in all populations, with over 43,000 participants; no serious safety issues were observed; the only grade 3 adverse events with a frequency of more than 2% were fatigue (3.8%) and headache (2.0%).

Example 17: all confirmed COVID-19 cases after dose 1

Some cases of diagnosed COVID-19 disease were not captured in the first major endpoint analysis on the evaluable efficacy population because they occurred less than 7 days after dose 2, or because they occurred in participants who were excluded from the evaluable efficacy population or had evidence of infection prior to or during the vaccination regimen.

All reports of COVID-19 at any time after dose 1 are considered in table 19, which provides a summary of all enrollee cases in the dose 1 population for all available potencies (modified intent-to-treat), regardless of evidence of infection prior to or during the vaccination regimen. Of these participants, 50 codv-19 in BNT162b2 group occurred after dose 1 compared to 275 in placebo group (table 19). Notably, in the BNT162b2 group, most cases occurred prior to dose 2. The estimated VE for the confirmed diagnostic COVID-19 occurring after dose 1 was 82% (two-sided 95% CI: 75.6%, 86.9%), and the estimated VE for the confirmed diagnostic COVID-19 occurring after dose 1 but before dose 2 was 52.4% (two-sided 95% CI: 29.5%, 68.4%).

Early protection can be readily seen in the graph 100, which shows the cumulative incidence of first-occurring COVID-19 after dose 1 in all vaccinated participants based on dose 1 in all available efficacy (improved intent-to-treat) populations. For BNT162b2 and placebo, the disease onset appeared to track each other (track together) until approximately 14 days after dose 1, at which point the curve deviated, and cases steadily accumulated in the placebo group, while remaining nearly flat in BNT162b2 group.

In both placebo and vaccine recipients, the cumulative incidence of COVID-19 cases began to deviate over time by 14 days after dose 1, with an estimated median incubation period of 5 days approximately 9 days after, indicating the early appearance of partial protective efficacy of immunization. In the interval between dose 1 and dose 2, the vaccine efficacy observed was 52%, and in the first 7 days after dose 2, it was 91%, reaching full efficacy for COVID-19, which had occurred at least 7 days after dose 2.

Example 18: secondary efficacy results-Final analysis

Final analysis of vaccine efficacy for COVID-19 development within 14 days after dose 2

Participants who had no evidence of infection prior to vaccination

For this efficacy endpoint, participants with positive or unknown NAAT results at any visit 14 days after dose 2 were not included in the efficacy assessment.

In participants who had no evidence of SARS-CoV-2 infection before and during the vaccination regimen, the VE for the confirmed COVID-19 that occurred at least 14 days after dose 2 was 94.2%, 8 and 139 cases in BNT162b2 and the placebo group, respectively. The posterior probability of a true VE greater than 30% is > 99.99%, meeting the pre-specified success criteria for this endpoint > 98.6%. The 95% confidence interval for vaccine efficacy is 88.7% to 97.2%, indicating a true VE of at least 88.7%, considering the probability of available data as 97.5%.

Participants with or without evidence of infection prior to vaccination

Among the participants with or without evidence of SARS-CoV-2 infection before and during the vaccination regimen, the definitive diagnosis of COVID-19 occurred at least 14 days after dose 2 had 94.4% VE, with 8 and 144 cases for BNT162b2 and the placebo group, respectively. The posterior probability of a true VE greater than 30% is > 99.99%, meeting the pre-specified success criteria for this endpoint > 98.6%. The 95% confidence interval for vaccine efficacy is 89.1% to 97.3%, indicating a true VE of at least 89.1%, considering the probability of 97.5% for all available data.

Vaccine efficacy-Final analysis for severe COVID-19 cases

Potency against severe COVID-19 (7 days after dose 2)

Participants with no evidence of infection before and during vaccination protocols

For this efficacy endpoint, participants with positive or unknown NAAT results at any visit 7 days after dose 2 were not included in the efficacy assessment.

In participants who had no evidence of severe SARS-CoV-2 infection before and during the vaccination regimen, the estimated VE for severe COVID-19 occurring at least 7 days after dose 2 was 66.4%, with 1 and 3 cases in BNT162b2 and the placebo group, respectively. The posterior probability of true vaccine efficacy of greater than 30% is 74.29%, failing to meet the pre-specified success criteria for this endpoint > 98.6%, due to the small number of severe cases observed after dose 2 in this study.

Participants with and without evidence of infection before and during vaccination protocols

Of the participants with or without evidence of severe SARS-CoV-2 infection 7 days after dose 2, VE was 66.3% for severe COVID-19 occurring at least 7 days after dose 2, with 1 and 3 cases for BNT162b2 and the placebo group, respectively. The posterior probability of true vaccine efficacy greater than 30% is 74.19%.

All Severe COVID-19 cases confirmed after dose 1-all available populations

Of the participants in all available efficacy groups, 1 COVID-19 occurred after dose 1 in BNT162b2 group compared to 9 in placebo group. The estimated VE for severe COVID-19 occurring after dose 1 was 88.9% (two-sided 95% CI: 20.1%, 99.7%), and the estimated VE for severe COVID-19 occurring at least 7 days after dose 2 was 75.0%.

Efficacy against severe COVID-19 (more than or equal to 14 days after dose 2)

Participants with no evidence of infection before and during the vaccination regimen (14 days) -Severe

In participants who had no evidence of severe SARS-CoV-2 infection before and during the vaccination regimen, the estimated VE for severe COVID-19 occurring at least 14 days after dose 2 was 66.4%, with 1 and 3 cases in BNT162b2 and the placebo group, respectively. The posterior probability of true vaccine efficacy greater than 30% is 74.32%.

Participants with or without evidence of infection (14 days) before and during the vaccination regimen-severe

Of the participants with or without evidence of severe SARS-CoV-2 infection before and during the vaccination period, the VE against severe COVID-19 occurring at least 14 days after dose 2 was 66.3%, with 1 and 3 cases in BNT162b2 and the placebo group, respectively. The posterior probability of true vaccine efficacy greater than 30% is 74.18%.

Vaccine efficacy in COVID-19 cases-Final analysis, as defined by CDC

Efficacy against COVID-19 (7 days after dose 2) based on CDC-defined symptoms

Participants with no evidence of infection before and during the vaccination protocol-7 days defined by CDC

Among participants who had no evidence of SARS-CoV-2 infection before and during the vaccination regimen, the VE for COVID-19 defined for CDC occurring at least 7 days after dose 2 was 95.1% (two-sided 95% CI: 90.2%, 97.9%), 8 and 165 cases in BNT162b2 and the placebo group, respectively.

Participants with and without evidence of infection before and during vaccination protocol-7 days defined by CDC

Among participants with and without evidence of SARS-CoV-2 infection before and during the vaccination regimen, the COVID-19 defined for CDC occurring at least 7 days after dose 2 had a VE of 94.7% (both sides 95% CI: 89.8% -97.6%), 9 and 172 cases in BNT162b2 and the placebo group, respectively.

Efficacy against COVID-19 symptoms defined based on CDC ≧ 14 days after dose 2

In participants who did not and with or without evidence of SARS-CoV-2 infection before and during the vaccination regimen, the VE for CDC defined COVID-19 that occurred at least 14 days after dose 2 was similar to that which occurred at least 7 days after dose 2.

Example 19: efficacy conclusions-Final analysis

In the final efficacy analysis, the VE of the definitive diagnosis COVID-19, which occurred at least 7 days after dose 2, was 95.0% in participants who had no evidence of SARS-CoV-2 infection before and during the vaccination regimen, with 8 COVID-19 cases in the BNT162b2 group compared to 162 COVID-19 cases in the placebo group. The 95% confidence interval for vaccine efficacy is 90.3% -97.6%. For the second primary endpoint, the VE for COVID-19 was 94.6% for the definitive diagnosis that occurred at least 7 days after dose 2 in participants with and without evidence of SARS-CoV-2 infection before and during the vaccination regimen, and 9 and 169 in BNT162b2 and placebo groups, respectively. The posterior probability of a true VE greater than 30% is > 99.99%, meeting the pre-specified success criteria for this endpoint > 98.6%. The 95% confidence interval for vaccine efficacy is 89.9% -97.3%, indicating a true VE of at least 89.9%, with a probability of 97.5% considering all available data.

The VE observed for the first primary efficacy endpoint was very high in all age, gender, race/ethnicity and national subgroups, as VE > 93% in all subgroups, except for "all other" race group (89.3% VE) and brazil (87.7% VE).

A total of 10 severe COVID-19 cases occurred after dose 1, with 1 in BNT162b2 compared to 9 in the placebo group.

In all participants (whether evidence of infection before or during the vaccination regimen), 50 cases of covd-19 occurred after dose 1 in the BNT162b2 group compared to 275 cases in the placebo group, indicating an estimated VE of 82% (95% CI: 75.6%, 86.9%) for the confirmation of the occurrence of covd-19 after dose 1.

Early protection is readily seen to occur from the cumulative incidence curve, which shows that for BNT162b2 and placebo, until about 14 days after dose 1, the disease onset trajectories correlate (track conjoint), the curves diverge at this point, the cases steadily accumulate in the placebo group, and remain nearly flat after BNT162b 2.

In summary, the final efficacy results show that 30 μ g of BNT162b2 provides protection against COVID-19 in participants who have no evidence of prior SARS-CoV-2 infection, including a subset across the population, with severe cases observed primarily in the placebo group.

Tables 20 and 21 below show details of the demographic groups evaluated.

Example 20: some observations of the response of the adolescent population to immunization with BNT162b2

In the clinical trials described in examples 13-19, the following were observed in the adolescent population.

Local reactions in adolescents

Adolescents between 12 and 15 years of age (N100; 49 in BNT162b2 group, 51 in placebo group) provided preliminary data on the reactogenicity subgroup and were analyzed separately. In this age group, pain at the injection site was the most frequent local reaction in the BNT162b2 group, and 71.4% of participants reported after dose 1 compared to 17.6% in the placebo group. After dose 2, the incidence of pain was reduced (to 58.7% vs 8.7%) in BNT162b2 and placebo groups. Redness was reported in BNT162b2 group by 1 participant after dose 1 and 2 participants after dose 2, but not after any dose in the placebo group. Swelling was reported in BNT162b2 group 2 participants after dose 1 and 3 participants after dose 2, whereas in placebo group swelling was reported in 1 participant after dose 1 and not after dose 2. Most local reactions are mild to moderate in severity. Two severe reactions were reported, both in BNT162b2 group: severe redness and severe pain at the injection site.

Systemic reactions in adolescents

Adolescents between 12 and 15 years of age (N100; 49 in BNT162b2 group, 51 in placebo group) provided preliminary data on the reactogenicity subgroup and were analyzed separately. Most systemic events (except emesis and diarrhea, which occur at low rates in all groups) were reported at a higher rate in BNT162b2 than in placebo. However, there was no significant tendency for the incidence or severity to increase after dose 1 compared to after dose 2. In this age group, the most common systemic events after dose 1 (dose 1vs dose 2) compared to dose 2 were:

fatigue: BNT162b2 (49.0% vs 50.0%), vs placebo (25.5% vs 6.5%)

Headache: BNT162b2 (42.9% vs 45.7%), vs placebo (35.3% vs 21.7%)

Muscle pain: BNT162b2 (22.4% vs 30.4%), vs placebo (13.7% vs 4.3%)

Chills: BNT162b2 (30.6% vs 28.3%), vs placebo (7.8% vs 8.7%)

Joint pain: BNT162b2 (12.2% vs 17.4%), vs placebo (9.8% vs 6.5%)

Fever: BNT162b2 (14.3% vs 19.6%), vs placebo (0% vs 0%)

Emesis: the frequency of reporting was similar in both groups and similar after each dose

Diarrhea: the frequency of reporting was similar in both groups and similar after each dose.

The severity of most systemic events in adolescents is mild to moderate. The frequency of severe events was lower in both groups and occurred in no more than 1 or 2 participants after any dose.

The use of antipyretic/analgesic drugs in the adolescent group was slightly increased after dose 2 compared to dose 1 (30.6% vs 41.3%) and higher than in the placebo group (9.8% vs 13%).

In summary, as observed in the elderly age group (e.g., greater than 16 years, such as 16-85 years), reactogenicity is primarily mild to moderate and is short lived following dosing to adolescents of 12-15 years of age, and the spectrum of adverse events does not suggest any serious safety issues.

The following examples 21-24 further demonstrate that neutralizing antibody responses and/or cell-mediated immune responses can be achieved with the mRNA compositions described herein (including, e.g., BNT162b1 and BNT162b2) according to various dosing regimens described herein (e.g., dosing regimens including one or more doses of 30 μ g or less, such as 20 μ g, 10 μ g, 3 μ g, etc.). In addition, the data provided in examples 21-24 further demonstrate that administration of certain mRNA compositions described herein (including, e.g., BNT162b1 and BNT162b2) with one or more doses of 3 μ g or more induces an immune response against SARS-CoV-2 (e.g., as described herein).

As will be understood by those skilled in the art upon reading this disclosure, it is demonstrated that administration of various mRNA compositions described herein can induce an immune response, including neutralizing antibodies against SARS-CoV-2; it has also been demonstrated that certain such compositions (i.e., which induce neutralizing antibodies and/or induce cell-mediated immune responses, such as T cell responses) are capable of inducing protective immune responses that reduce the incidence of SARS-CoV-2 infection and/or COVID19 disease in organisms, including in particular primate organisms (where they have induced such neutralizing antibodies and/or cell-mediated immune responses), and also including humans. In some embodiments, it is also demonstrated that certain such compositions (e.g., as described herein) do not significantly induce vaccine-mediated disease enhancement, for example, as demonstrated by only one of 10 cases of severe COVID-19 observed after the first dose. Indeed, the present disclosure demonstrates that such compositions are effective for vaccinating humans (see, e.g., the clinical trial results shown in examples 13-19), e.g., against severe COVID-19 disease.

Example 21: immunogenicity Studies of functional antibody responses

In the clinical trial described in example 7, following immunization with BNT162b1 or BNT162b2, the following was observed in healthy young adults (18-55 years) and older adults (56-85 years). In young adults, two doses of 1. mu.g, 3. mu.g, 10. mu.g, 20. mu.g or 30. mu.g are administered 21 days apart. In older adults, two doses of 20 μ g are administered 21 days apart. Functional antibody data for the young adult cohort were determined up to day 50 after the initial dose was administered for dose groups 1 μ g and 3 μ g, and up to day 85 after the initial dose was administered for dose groups 10, 20 and 30 μ g. For older adults dosed with BNT162b2, data was available until day 29 after the initial dose was administered.

Virus neutralizing antibody GMT (neutralizing GMT) and 95% confidence intervals for participants aged 18-55 years after BNT162b1 dosing, see figure 40.

Virus neutralizing antibody GMT (neutralizing GMT) and 95% confidence intervals for young participants aged 18-55 years and older participants aged 56-85 years after BNT162b2 dosing, see figure 101 (50% neutralization titer).

Geometric Mean Fold Increase (GMFI) from baseline for functional antibody titer data is shown in figure 102(BNT162b1) and figure 103(BNT162b 2).

Participants dosed with BNT162b1 showed strong, dose-dependent antibody responses. At day 22, 21 days after dose 1, the virus neutralizing antibody GMT increased in a dose dependent manner for the 1, 10, 30 and 50 μ g dose groups. At day 29 (7 days after dose 2), neutralizing GMT showed a strong, dose-level dependent booster response. In the single 60 μ g dose group, neutralizing GMT remained low, indicating that a booster dose was required to increase functional antibody titers.

On day 43 (21 days after dose 2 of BNT162b 1), GMT reduction was neutralized (except for 1 μ g dose level). At day 43, the virus neutralized GMT was 0.7-fold (1. mu.g) to 3.6-fold (50. mu.g) that of the COVID-19HCS group.

The COVID-19HCS group comprises 38 human COVID-19HCS sera from individuals aged 18-85 years at least 14 days after diagnosis (d) and obtained when the individual is asymptomatic. Serodonors were mainly symptomatic (35/38) and 1 had been hospitalized. Serum was obtained from Sanguine Biosciences (Sherman Oaks, Calif.), MT Group (Van Nuys, Calif.) and Pfizer Occupical Health and Wellness (Pearl River, NY).

Participants dosed with BNT162b2 showed strong antibody responses induced by BNT162b 2. Virus neutralization of GMT was detected 21 days after dose 1 (day 22) and by 7 days after dose 2 (day 29), it was significantly increased in young participants (aged 18-55 years) immunized with ≥ 3 μ g of BNT162b2 and in older participants (aged 56-85 years) immunized with 20 μ g of BNT162b 2. At day 29, virus neutralization between the 20 μ g dose level cohorts for young and older adults was comparable to GMT. The lowest therapeutic dose of 1 μ g BNT162b2 elicited the least neutralizing response in participants aged 18-55 years.

At day 43 (21 days after dose 2 of BNT162b2), virus neutralization GMT in the cohort of young adults declined for dose levels of 3, 20, and 30 μ g. Then, for the young adult dose groups 10, 20 and 30 μ g, the neutralizing GMT remained stable between day 29 and day 43 until day 85 (63 days after dose 2) and was 1.3-fold to 1.9-fold that of the COVID-19HCS group.

Seroconversion in this context is defined as the minimum 4-fold increase in antibody GMT compared to baseline. The frequencies of participants with serum turnover are shown in graph 104(BNT162b1) and graph 105(BNT162b 2).

Seroconversion occurred 7 days or 21 days (day 29 or 43) after dose 2 in all participants dosed with more than or equal to 30 μ g BNT162b1 or BNT162b2 at dose 1. All participants dosed with 30 μ g BNT162b2 subsequently remained seropositive until day 85.

Example 22: immunogenicity Studies of bound antibody concentrations

In the clinical trial described in example 7, following immunization with BNT162b1 or BNT162b2, the following was observed in healthy young adults (18-55 years) and older adults (56-85 years). Dosing BNT162b1 young participants aged 18-55 years were dosed with 1, 10, 30, 50 or 60 μ g on day 1 (all dose levels) and day 22 (all dose levels except 60 μ g) (n-12/group) with antibody concentration data available until day 43.

Participants dosed with BNT162b2 data were available for dosing 1, 3, 10, 20 or 30 μ g of young participants aged 18-55 years and 20 μ g of older participants aged 56-85 years on days 1 and 22 (n-12/group). For the young participant dose groups, data for bound antibody concentrations of 1 μ g and 3 μ g for the dose groups were available up to day 50, and data for bound antibody concentrations of 10, 20, and 30 μ g for the dose groups were available up to day 85. For older participants dosed with BNT162b2, data was available until day 29.

The fold increase in bound antibody concentration from baseline after dosing with BNT162b1 and BNT162b2 is shown in figure 106 and figure 107, respectively.

On day 21 (day 22) after dose 1, participants dosed with BNT162b1 exhibited a strong, dose-dependent antibody response against the S1 subunit of the SARS-CoV-2 spike (S) protein. Immunoglobulin (IgG) GMC bound to S1 showed a strong, dose-dependent booster response 7 days after dose 2 (day 29). In the 60 μ g dose group, which was dosed only once, IgG GMC bound to S1 remained low, indicating that a booster dose was required to increase antibody concentration.

At 21 days 2 (day 43) after dose 2 of BNT162b1, IgG GMC binding to S1 decreased (except for the 1 μ g dose group), but clearly above that of the COVID-19HSC group for all doses tested.

At 21 days after dose 1 (day 22), participants dosed with BNT162b2 exhibited a strong BNT162b 2-induced IgG response binding to S1, as reflected by a dose-dependent response only between 1 μ g and 10 μ g dose levels. By 7 days post dose 2 (day 29), IgG GMCs bound to S1 showed a clear booster response. At day 29, IgG GMCs that bound S1 were comparable between young and older participants at a 20 μ g dose level.

On day 85 (63 days post dose 2, 10-30 μ g dose levels), antibody levels decreased with time in all dose level cohorts, but the S1-binding antibody GMC was well above that observed in the COVID-19HCS group (figure 107).

For the S1-binding antibody response, almost all participants immunized with BNT162b1 and BNT162b2 seroconverted 21 days (day 22) as early as dose 1. The frequency of participants with seroconversion after dosing with BNT162b1 is shown in figure 108, and BNT162b2 is shown in figure 109. A similar situation was found using only the RBD domain as the target antigen.

Example 23: exemplary cell-mediated immune responses: SARS-CoV-2 specific CD4+And CD8+T cell response

In the clinical trial described in example 7, following immunization with BNT162b1 or BNT162b2, the following was observed in healthy young adults (18-55 years) and older adults (56-85 years). CD4 +And CD8+T cell response data were available from 97 study participants receiving BNT162b1 and 76 participants receiving BNT162b2, with 70 young participants at a dose level of 1, 3, 10, 20, 30, 50, or 60 μ g (note: dose 2 was not given in the 60 μ g dose group) and 27 older participants at a dose level of 10, 20, or 30 μ g and participants receiving BNT162b2 at a dose level of 1, 3, 10, 20, or 30 μ g (47 young participants at a dose level of 10, 20, or 30 μ g)Person) or 10, 20 or 30 μ g (elderly participants).

BNT162b1 was administered at most dose 1 and dose 2 (86 out of 88 [ 97.7%]) Medium-induced strong RBD-specific CD4+T cell responses, including all older participants (27 out of 27 [ 100%]) (ii) a CD8 was induced in 47 of 61 young participants (77.0%) and 21 of 27 older participants (77.7%)+And (6) responding. In contrast, T cell responses were less detectable and to a lesser extent in the 60 μ g dose group of 9 young participants who received only dose 1, indicating the importance of the booster dose.

BNT162b2 Young or older participants (76 [ 100% in 76%) dosed at all doses]) Medium induction strong SARS-CoV-2S protein specificity CD4 +A T cell response; induction of CD8 in 45/47 (95.7%) young participants and 24/29 (82.8%) older participants+T cell response. Albeit CD8 among older participants+The immunogenicity rate was slightly lower, but the extent of response induced by BNT162b2 was comparable to that induced in young participants who received 30 μ g of BNT162b 2. These T cell responses were directed against different portions of the antigen, including non-RBD sequences, indicating that BNT162b2 induced a multi-epitope response in all age groups.

Dosing BNT162b1 or BNT162b2 twice resulted in a significant increase in the incidence and magnitude of T cell responses in both age groups of BNT162b1 and at all dose levels. Although BNT162b2 induced CD4+The magnitude of the T cell response was similar at different dose levels, but CD8+The magnitude of the T cell response was highest at the 30 μ g dose level. With the strongest CD4+The memory response in participants of the T cell response was more than 10-fold greater than the memory response observed in the same participants for immunodominant peptides of cytomegalovirus, Epstein Barr virus, influenza virus and tetanus toxoid. The same participants also had strong CD8+A T cell response comparable to a memory response against the viral antigens described above.

After vaccination, de novo induction of BNT162b1 was observed in 97.5% of the participants, de novo induction of BNT162b2 was observed in 100% of the participants,RBD and S protein specific CD4+T cell response. Following vaccination, de novo induction of BNT162b1 was observed in 95.5% of participants, de novo induction of BNT162b2, RBD and S protein-specific CD8 was observed in 96.6% of participants+T cell response.

Example 24: exemplary cell-mediated immune responses: functional and pro-inflammatory CD4+/CD8+T cell response

In the clinical trial described in example 7, the following was observed in healthy young adults (18-55 years) and older adults (56-85 years) vaccinated with BNT162b1 or BNT162b 2. De novo induction of T cells against SARS-CoV-2S protein or RBD protein was confirmed using Intracellular Cytokine Staining (ICS). Similar cell-mediated immune responses as described below were also observed with BNT162b2 as described for BNT162b1 in example 7.

For example, both BNT162b1 and BNT162b2 robustly induced IFN γ -producing CD4+ and CD8+ T cells against SARS-CoV-2S protein or RBD. No significant dose dependence was observed for both BNT162b1 and BNT162b 2. The cytokine response elicited after dosing BNT162b1 or BNT162b2 in older participants was nearly identical in pattern and intensity to that of younger participants.

BNT162b1 and BNT162 induced a multifunctional and pro-inflammatory CD4+/CD8+ T cell response in almost all participants. Detection of Interferon (IFN) γ, Interleukin (IL) -2, but no IL-4, indicates a favorable Th1 profile and no potentially harmful Th2 immune response.

With respect to BNT162b2, fractions of Peripheral Blood Mononuclear Cells (PBMCs) isolated from blood of study participants collected at baseline (before dose 1) and 29 ± 3 days after dose 1 of BNT162b2 were analyzed. This included data for a total of 74 study participants:

young participants aged 18-55 years per dose group: 1 μ g (n-8), 3 μ g (n-9), 10 μ g (n-10), 20 μ g (n-9), 30 μ g (n-10).

Aged participants at the age of 56-85 years per dose group: 10 μ g (n-11), 20 μ g (n-8), 30 μ g (n-9).

The functionality and polarization of vaccine-induced SARS-CoV-2S-specific T cells was assessed by intracellular accumulation of cytokines IFN-. gamma. (gamma), IL-2 and IL-4 responses, which represent overlapping peptide stimulations of the vaccine-encoded full-length RBD sequence and wild-type SARS CoV-2 protein, respectively. As a control, PMBC from 18 virally diagnosed COVID-19 convalescent patients were used.

In both age groups analyzed, two doses of BNT162b2 (dose range 1-30 μ g) induced vaccine-specific T cell responses (fig. 110 and 111). The assay for the T cell response specific for the SARS-CoV-2S protein was performed with two different peptide pools, S pool 1 containing overlapping peptides (not identical to domains) of the N-terminal region of the S protein and S pool 2 containing the C-terminal region of the S protein. S-specific CD4 analyzed in participants dosed with 74 BNT162b2 +The T cell response is characterized by a Th1 cytokine profile that secretes IFN- γ or IL-2 or both.

In response to stimulation with the S-peptide subpool, almost no T cells secreting the Th2 cytokine IL-4 could be detected (average score: 20 and 30 μ g adult cohort antigen-specific circulating CD 4)+T cells were 0.01% and 0.02%, respectively; stimulation with S protein subpool 1 and subpool 2, respectively). Of 61 of 74 participants analyzed (40 of 46 participants, 40 of 46, and 21 of 28 of older adults), S-specific CD8+T cells secrete IFN gamma, and IL-2-secreting CD8 can also be detected+T cells. S-specific IFN gamma targeting the N-terminal domain of the S protein+CD8+The fraction of T cells reached peripheral blood CD8 in the dose groups of 20 and 30 μ g young participants+1% of the total T cells reached 2.4% in the 30 μ g older participant dose group. Pre-existing CD8 was detected against the C-terminal region of S protein in 17 of 74 dosed participants+T cell response (range: 0.07% -5.59% IFN γ producing CD8+T cells). In 6 of 17 participants, these pre-existing responses were slightly amplified following BNT162b2 dosing.

In general, S-specific CD4 +And CD8+The average fraction of T cells was found to be 18 from CSignificantly higher observed in patients with OVID-19 recovery (e.g., S protein pool 1IFN γ CD8 in participants dosed at 30 μ g+Response 12.5 times higher). Importantly, for the 30 μ g dose group of clinical interest, cytokine responses elicited in the older participants were nearly identical in pattern and intensity to the responses of the younger participants following BNT162b2 vaccination.

BNT162b 2-induced T cell response, in particular to CD8+T cells, not only restricted to RBD, but also observed a clearly strong T cell recognition of the non-RBD region of the S protein.

BNT162b2 induced multifunctionality and proinflammatory CD4 in almost all participants+/CD8+T cell response. Th1 polarization of helper responses is characterized by robust IFN γ/IL-2 production upon antigen-specific (wild-type SARS CoV 2S protein peptide pool) restimulation and by production of only small amounts of IL-4.

Example 25: certain T cell responses induced by BNT162b2

In addition to examples 23 and 24, which describe certain T cell responses observed in Germany trials (study BNT 162-01; NCT04380701) induced by immunization with BNT162b2, this example further demonstrates the immunogenicity of prime-boost vaccination with 1, 10, 20 and 30 μ g BNT162b2 in participants aged 19-55, including detailed characterization of T cell responses, e.g., the first identification of CD8 induced by the COVID-19 vaccine described herein +Epitopes recognized by T cells. Without wishing to be bound by any particular theory, it is noted that the identity of the epitope that elicits the response in the subject and/or the extent of the response to a particular epitope or combination of epitopes may affect one or more characteristics (e.g., effectiveness and/or duration) of the immune response and/or immune protection provided by the administered vaccine. In some embodiments, a dosing regimen may involve one or more steps of monitoring one or more characteristics of an immune response, including, for example, the presence and/or level of a response (e.g., T cells and/or antibodies) that recognizes one or more particular epitopes. In some embodiments, the need, timing, and/or extent of subsequent doses may be determined based on such monitoring.

As followsDescribed in one step, this example shows, in part, that several BNT162b2 induce CD8 when presented on frequent (frequency) MHC alleles+Epitopes recognized by T cells were identified with peptide MHC multimers; and CD8+T cells were shown to have early differentiated effector memory phenotype, monospecific to circulating CD8+0.01-3% of T cells. Without wishing to be bound by any particular theory, it is noted that cells exhibiting an "effector memory" phenotype can provide longer term protection.

This example also recorded that some participants who received BNT162b2 had pre-existing T cell responses. Thus, this example demonstrates that the compositions described herein, and in particular BNT162b2, can be well-administered to a subject who has even been exposed to one or more related viruses, including potentially the same virus-i.e., SARS-CoV-2 and/or exposed to an antigen thereof, or another antigen sharing one or more epitopes with the SARS-CoV-2 spike protein.

Dominance and extent of vaccine-induced T cell response

T cell responses of 37 participants immunized with BNT162b2 sufficient Peripheral Blood Mononuclear Cells (PBMCs) were analyzed before vaccination (day 1) and 7 days after booster dose (day 29) by direct ex vivo IFN γ enzyme-linked immunosorbent spot (ELISpot) assay (fig. 112 and 113). One of ordinary skill in the art will appreciate that SARS-CoV-2S protein includes a signal peptide (aa 1-13), an N-terminal S1 protease fragment (aa 14-685) and a C-terminal S2 protease fragment (aa 686-; and S1 contains an RBD (aa 319) -541) that binds to host receptors and S2 mediates fusion between the viral envelope and cell membrane. To resolve (deconvolute) reactivity to S protein, CD4 was stimulated with overlapping peptides representing different portions of the SARS-CoV-2S wild-type sequence +Or CD8+T cell effectors overnight, N-terminal pools "S pool 1" (aa 1-643) and "RBD" (aa 1-16 fused to aa 327-528 of S), and C-terminal "S pool 2" (aa 633-1273).

Robust amplified SARS-CoV-2S-specific CD4 was detectable in all 37 participants 7 days post-boost with BNT162b2 at any of the doses indicated+T cells (FIG. 112a, FIG. 113 a). Of 34 of the participants, comparison with pre-vaccinated PBMC was possible. 30 of these 34 subjects (88.2%) had de novo (absent at baseline) CD4 for both S pool 1 and S pool 2 of SARS-CoV-2+T cell response. One participant only responds from the beginning to pool 2. The remaining 3 participants had a head-to-head response to S pool 1, and a small amount of pre-existing S pool 2 reactive CD4+T cells. In 2 of these 3 participants, the pre-existing response to S pool 2 was amplified by vaccination (from 91 and 188 spots/10, respectively, prior to vaccination)6From one cell to 1391 and 965 spots post vaccination), while in 1 of 3 participants, the pre-existing response to S pool 2 remained stable (53 to 140 spots/10)6Individual cells). These data demonstrate that two doses of BNT162b2 induced multiple epitopes CD4 in 94.1% (32/34) of participants +T cell responses (de novo or amplified), directed to the N-terminal and C-terminal portions of S, and thus to epitopes outside the RBD (fig. 113 b).

Although for dosage levels of ≧ 10 μ g, CD4+The extent of T cell response does not appear to be dose dependent, but varies from individual to individual. Among the strongest responders, S-specific CD4+T cell responses were more than 10-fold higher than individual memory responses to common viral and recall antigens (from cytomegalovirus, Epstein Barr virus, influenza virus and tetanus toxoid) (figure 112b, c).

Vaccine-induced S-specific CD8 was detected in 34 (91.9%) of 37 vaccinated participants+T cell response. The majority were strong responses (fig. 112a, fig. 113a), comparable to individual memory responses to Cytomegalovirus (CMV), Epstein Barr Virus (EBV) and influenza virus (fig. 112b, c). Induction of de novo S-specific CD8 in 33 participants+T cell responses directed to either of the two S pools (22 participants), or one of the S pools (S pool 1 in 10 participants, S pool 2 in 2 participants), indicating that multi-epitope responses, including non-RBD S-specific T cells, predominate (figure 113 b). Of the 7 participants, a pre-existing CD8 was detected for S pool 2 +The response of the T-cell is,it was not further enhanced by vaccination. 6 of these 7 participants had simultaneous de novo responses to pool 1 of S that were not significantly different in intensity from those observed in individuals without pre-existing responses to pool 2 of S (fig. 113 c). Notably, in the presence of detectable CD8+Of the 34 participants in the T cell response, the strongest (above the third quartile) response to S pool 1 was observed in participants with no pre-existing S pool 2-specific response.

S-specific CD4+The extent of T cell responses was positively correlated with S1 binding IgG (fig. 114a) and conformed to the concept of intramolecular help (e.g., CD4 response to one epitope in an antigen could support the development of CD8 response to an epitope in the same antigen) and to S-specific CD8+The intensity of the T cell response (fig. 114 b). S-specific CD8+T cell responses were also positively correlated with S1 binding IgG (fig. 114c), indicating convergent development of humoral and cellular adaptive immunity.

Polarization of vaccine-induced T cell responses

To assess the functionality and polarization of S-specific T cells, the cellular factors secreted in response to stimulation of S-pool 1, S-pool 2 and RBD-pool were determined by intracellular staining (ICS) of IFN γ, IL-2 and IL-4 specific responses in PBMCs before and after vaccination of 37 participants immunized with different doses of BNT162b 2. Vaccine-induced, S-specific CD4 +A significant proportion of T cells secrete IFN γ, IL-2 or both, and T cells secreteH2 cytokines IL-4T cells were barely detectable (figure 115a-c, figure 113 d-e). Vaccine induced S-specific CD8+T cells secrete primarily IFN γ and lower levels of IL-2 in response to S pool 1 and S pool 2 stimulation. IFN gamma specific for S pool 1+CD8+Fraction of T cells constitutes peripheral blood CD8+Up to about 1% of the total number of T cells (fig. 115 d). Notably, of the participants analyzed, several (n-3 in the 20 μ g dose cohort and n-3 in the 30 μ g dose cohort) showed pre-existing S-pool 2-specific CD8+T cell responses, 5 of which were not further amplified after vaccination in 6 participants. In one participant(20 μ g dose cohort) Strong pre-existing S pool 2-specific IFN γ could be detected+CD4+T cell responses (figure 115 c).

In two assay systems, CD4+And CD8+The multiepitopic character of the T cell response elicited by BNT162b2 was further confirmed by the cytokine production by T cells in response to the peptide pool comprising the complete SARS-CoV-2S over the response to the RBD peptide pool. BNT162b 2-induced S-specific IFN γ in total circulating T cells+Or IL-2+CD4+And CD8+The average score of T cells was higher than that detected in 18 control subjects (HCS) recovered from COVID-19 (figure 115c, d).

CD8 was observed following BNT162b2 immunization+Epitope specificity and phenotype of T cells

Of the three participants, CD8 was characterized at the antigen level+T cell response, the participants were immunized with a 2 dose regimen with 2 doses (e.g., 10 μ g/dose or 30 μ g/dose) at 21 day intervals.

Peripheral Blood Mononuclear Cells (PBMCs) collected from participants were stained with the personalized peptide/MHC multimer staining mixture for flow cytometry analysis before and after vaccination. A diverse set of peptide/MHC allele pairs of 23 pairs (4 for HLA-B0702, 19 for HLA-a 2402), 14 pairs (HLA-B3501) and 23 pairs (7 for HLA-B4401 and 16 for HLA-a 0201) were used for participants 1, 2 and 3 respectively, thus probing a selected set of potential reactivities, rather than extensively capturing multi-epitope T cell responses. For each participant, de novo induced CD8 was identified against multiple epitopes+T cell reactivity, with a total of 8 different epitope/MHC pairs distributed throughout the full length of the S protein (fig. 116a, c). The extent of epitope-specific T cell response was peripheral CD8+0.01-3.09% of T cells, and was observed against HLA-A0201 YLQPRTFLL (CD 8)+3.09% multimer +)、 HLA-A*2402QYIKWPWYI(CD8+1.27% multimer+) And HLA-B3501 QPTESIVRF (CD 8)+0.17% multimer+) Is most deeply extended. Determined by ELISpot and intracellular staining (ICS), andmajority of (bulk) IFN γ against the complete S protein in some individuals+CD8+Comparison of T cell responses showed that pMHC technology may be more useful for assessing the true extent of cellular immune responses (fig. 113 f).

Identified pMHC multimers that undergo S antigen+CD8 (1)+T cell-specific phenotypic analysis revealed an early differentiated effector memory phenotype characterized by low expression of CCR7 and CD45RA and high expression of the co-stimulatory molecules CD28 and CD 27. CD8+T cells also expressed markers associated with the associated activation, such as CD38, HLA-DR, and PD-1 (FIG. 116 b).

Discussion of the related Art

Effectors of the adaptive immune system have a complementary role in the defense against viral infections. Although neutralizing antibodies were the first line of defense, CD8+Cytotoxic T Lymphocytes (CTLs) help to clear virus from intracellular compartments that are inaccessible to neutralizing antibodies. Antigen-specific CD4+T cells have immune coordination functions and are B cells and CD8+T cells provide associative help, support memory generation, and cytotoxic activity either indirectly (e.g., via IFN γ) or directly (against MHC class II expressing target cells).

This example shows that vaccination with BNT162b2 induces the induction of CD4 with SARS-CoV-2S-specific neutralizing antibodies (as described in the other examples), CD4+T cell, CD8+T cells and immune regulatory cytokines such as IFN gamma synergistic immune response.

All participants vaccinated with BNT162b2 presented de novo S-specific CD4 as detected by the ex vivo ELISpot assay+T cell responses and almost 92% of participants presented with CD8+T cell response. The extent of T cell response varies from person to person and is not significantly dose dependent. Even with the lowest dose of 1. mu.g BNT162b2, most vaccinated participants showed CD4+And CD8+Robust expansion of T cells. T cell responses were directed against the RBD, S1 and S2 regions of the S protein, indicating immune recognition of multiple independent MHC I and II epitopes.

BNT162b 2-induced CD4+T cellsExpresses IFN gamma and IL-2 but only low levels of IL-4, indicating a TH1 profile and no potentially harmful TH2 immune response.

Although directed against all CD8 of the S1 subunit of the S protein+T cell responses were all de novo and were not detected at baseline, but pre-existing immune responses to the S2 subunit were found in several individuals. The sequence similarity of the S1 fragment to the corresponding seasonal coronavirus sequence is less compared to the S2 fragment; without wishing to be bound by theory, it is believed that this finding suggests that pre-existing cross-reactive CD8 may have been detected +T cells.

Peptide MHC (pMHC) multimer technology enables the identification of vaccine-induced CD8+T cells recognize the S protein epitope and direct quantification of the respective epitope-specific T cells. The cumulative T cell frequency in each participant exceeded the overall T cell response measured in the ELISpot and ICS assays, indicating that these assays underestimate the true extent of the polyepitopic response. One skilled in the art will recognize that single peptide assays are known to produce higher T cell frequencies than functional T cell assays with numerous immunogenic epitope competitions stimulated with peptide pools. High proportion of induced CD8+T cells are early differentiated effector memory cells. This favorable phenotype has the potential for a rapid response, but has limited ability to produce IFN γ and is therefore unlikely to be detected in a functional T cell assay. Although the infected individual generated CD8 for it+Epitopes in SARS-CoV-2S of T cells have been identified and are known in the art (see, e.g., Shomuradova et al, Immunity (2020) doi:10.1016/j. immunity.2020.11.004; and Peng et al Nat. immunity.21, 1336-1345(2020)), the data provided herein demonstrate for the first time that COVID-19 vaccines induce epitopes that T cells recognize. Notably, the immunodominant HLA-a 02 identified in this study: the 01-restricted peptide YLQPRTFLL was previously described in convalescent COVID-19 patients (supra).

Materials and methods

Proteins and peptides

Two pools of 15-membered peptides overlapping by 11 amino acids (aa), collectively covering the entire sequence of wild-type SARS-CoV-2S (S pool 1 characterized by aa 1-643, S pool 2 characterized by aa 633-. CEF (CMV, EBV, influenza virus; human leukocyte antigen [ HLA ] class I epitope peptide pool) and CEFT (CMV, EBV, influenza virus, tetanus toxoid; HLA class II epitope peptide pool) were used as controls for general T cell responsiveness and to calibrate (benchmark) the extent of memory T cell responses. All peptides were obtained from JPT Peptide Technologies.

Human convalescent serum and PBMC panel

At least 14 days after PCR confirmed diagnosis, and when participants were asymptomatic, human SARS-CoV-2 infection/COVID-19 convalescent sera (n 38) were drawn from donors aged 18-83 years. The average age of the donors was 45 years. Neutralization GMTs for the donor subgroups were as follows: symptomatic infection 90(n ═ 35); asymptomatic infection 156 (n-3); hospitalization 618(n ═ 1). Serum was obtained from Sanguine Biosciences (Sherman Oaks, Calif.), MT Group (Van Nuys, Calif.) and Pfizer Occupical Health and Wellness (Pearl River, NY). PBMC samples of human SARS-CoV-2 infection/COVID-19 convalescent phase (n ═ 18) were collected from donors aged 22-79 years 30-62 days after PCR confirmed when donors were asymptomatic. PBMC donors were asymptomatic or mildly infected (n ═ 16, clinical scores 1 and 2), or hospitalized (n ═ 2, clinical scores 4 and 5). Blood samples were obtained from frankfurt university hospital.

Isolation of Primary cells

By Ficoll-PaqueTMPBMCs were separated by PLUS (Cytiva) density gradient centrifugation and stored frozen prior to analysis.

IFNγELISpot

With exhausted CD4+And enriched in CD8+T cell (CD 8)+Effector) or depleted CD8+And enriched in CD4+T cell (CD 4)+Effector) ex vivo IFN γ ELISpot assay (without further in vitro culture for amplification). One-way two-part testing was performed and positive controls (anti-CD 3 monoclonal antibody CD3-2[1:1,000; Mabtech)]). The IFN γ -specific antibody (ELISpotPro kit,mabtech) precoated Multiscreen filter plates (Merck Millipore) and blocked with X-VIVO 15 medium (Lonza) containing 2% human serum albumin (CSL-Behring) for 1-5 hours. Each well was filled with three overlapping peptide pools representing different portions of the SARS-CoV-2S wild-type sequence (N-terminal pool S pool 1[ aa 1-643)]And RBD [ aa 1-16 fused to aa 327-528]And C terminal S pool 2[ aa 633-]) Stimulation 3.3X 105And each effector cell lasts for 16-20 hours. Bound IFN γ was visualized using a secondary antibody directly coupled to alkaline phosphatase, followed by incubation with 5-bromo-4-chloro-3' -indolylphosphate (BCIP)/Nitro Blue Tetrazolium (NBT) substrate (ELISpotPro kit, Mabtech). AID Classic Robot ELISPOT Reader scanner plates were used and analyzed by AID ELISPOT 7.0 software (AID Autoimmun diagnostic). The spot counts are shown as the average for each replicate well. Using an internal ELISpot data analysis tool (EDA), based on two statistical tests (no partition resampling), the T cell response stimulated with peptide was compared to effectors incubated with medium alone as negative controls to provide sensitivity while maintaining control over false positives.

To account for the variation in sample mass reflected in the number of spots in response to anti-CD 3 antibody stimulation, a normalization method was employed to achieve direct comparison of spot counts and response intensities between individuals. This dependence is modeled in a log-linear fashion using a Bayesian model that includes a noise component (not disclosed). For robust normalization, each normalization is sampled 1000 times from the model and the median taken is taken as the normalized blob count value. Likelihood of the model: log lambdaE=αlogλP+logβj+ α ε, where λEIs the normalized spot count of the sample, and α is at all positive controls λPCommon stability factor (normal distribution), betajIs the specific component (normal distribution) of sample j, and σ ε is the noise component, where σ is Cauchy distributed and ε is Student's-t distributed. Beta is ajEnsure that each sample is treated as a different batch.

Flow cytometry

Cytokine-producing T cells were identified by intracellular cytokine staining. PBMCs thawed and resting for 4 hours in OpTsizer medium supplemented with 2. mu.g/mL DNAseI (Roche) in the presence of GolgiPlug (BD) were restimulated with different portions of the wild-type sequence of SARS-CoV-2S in a Peptide pool as described in the ELISpot section (2. mu.g/mL/Peptide; JPT Peptide Technologies) at 37 ℃ for 18 hours. Controls were treated with DMSO-containing medium. Cells were stained for viability and surface markers (CD3 BV421,1: 250; CD4 BV480,1: 50; CD8 BB515,1: 100; both from BD Biosciences) in flow buffer (DPBS [ Gibco ], supplemented with 2% FCS [ Biochrom ], 2mM ethylenediaminetetraacetic acid [ EDTA; Sigma-Aldrich ]) at 4 ℃ for 20 minutes. Thereafter, the samples were fixed and permeabilized using the Cytofix/Cytoperm kit according to the manufacturer's instructions (BD Biosciences). Intracellular staining (CD3 BV421,1: 250; CD4 BV480,1: 50; CD8 BB515,1: 100; IFN γ PE-Cy7, 1:50[ for HCS ]; IFN γ BB700, 1:250[ for participants ]; IL-2PE, 1: 10; IL-4 APC, 1: 500; both from BD Biosciences) was performed in Perm/Wash buffer at 4 ℃ for 30 min. Samples were collected on a Fluorescence Activated Cell Sorter (FACS) VERSE instrument (BD Biosciences) and analyzed using FlowJo software version 10.6.2(FlowJo LLC, BD Biosciences). The background of production of S-and RBD-specific cytokines was corrected by subtracting the values obtained with dimethyl sulfoxide (DMSO) -containing medium. Negative values are set to zero. Cytokine production in figure 116b was calculated by counting the total number of fractions of all CD4+ T cells positive for IFN γ, IL-2 or IL-4, setting this total to 100%, and calculating the fraction of each subset in which a particular cytokine was produced. The pseudo-color axis is on the scale of log 10.

peptide/MHC multimer staining

To select class I MHC epitopes for multimer analysis, mass-based binding and presentation predictors (e.g., as described in Abelin et al, Immunity 46,315-326 (2017); and Poran et al, Genome Med.12,70 (2020)) were applied to 8-12 amino acid long peptide sequences of spike glycoproteins derived from SARS-CoV-2 (access: NC-045512.2, https:// www.ncbi.nlm.nih.gov/nuccore/NC-045512) and paired with 18 MHC-class I alleles with frequencies greater than 5% in the European population. By passingThe threshold was set to combine the percentage rating (< 1%) with the presentation score (> 10)-2.2) Taking into account the purity of synthesis>90% of the peptides, the highest ranked predicted epitope can be identified. Using easYmer technique (kit, ImmuneAware Aps), refolding pMHC complexes and verifying complex formation in bead-based flow cytometry assays according to the manufacturer's instructions. Combinatorial labeling was used to study the antigen specificity of T cells, using a two-color combination of 5 different fluorescent labels, enabling detection of up to 10 different T cell populations per sample. For tetramerization, Streptavidin (SA) -fluorescein conjugate was added: SA BV421, SA BV711, SA PE-Cy7, SA APC (all from BD Biosciences). For three participants in the BNT162b2 vaccination, the individualized pMHC multimeric staining mixture contained up to 10 pMHC complexes, each encoded by a unique two-color combination. PBMC (2X 10) 6) Each pMHC multimer mixture was mixed with Brilliant Staining Buffer Plus (BSB Plus [ BD Horizon ] at room temperatureTM]) At a final concentration of 4nM, ex vivo staining was performed for 20 min. In flow buffer (DPBS [ Gibco ]) supplemented with BSB Plus]With 2% FBS [ Biochrom]、2mM EDTA[Sigma- Aldrich]) Surface and viability staining was performed at 4 ℃ for 30 min (CD3 BUV395, 1: 50; CD45RA BUV563, 1: 200; CD27 BUV737, 1: 200; CD8 BV480, 1: 200; CD279 BV650, 1: 20; CD197 BV786, 1: 15; CD4 BB515, 1: 50; CD28 BB700, 1: 100; CD38 PE-CF594, 1: 600; HLA-DR APC-R700, 1: 150; both from BD Biosciences; DUMP channel: CD14 APC-eFluor780, 1: 100; CD16 APC-eFluor780, 1: 100; CD19 APC-eFluor780, 1: 100; a fixable vigor dye eFluor780, 1: 1667; all from ThermoFisher Scientific). Cells were fixed in 1 × Stable fixative (BD) at 4 deg.C for 15 min in FACSYSPhonyTMA3 were collected on a flow cytometer (BD Biosciences) and analyzed with FlowJo software version 10.6.2(FlowJo LLC, BD Biosciences). CD8 when an aggregated population was observed that was color-labeled with only two pMHC multimers+T cellsReactivity was considered positive.

Example 26: evidence indicating the possibility of reinfection

The primary endpoint was evaluated in individuals without prior evidence of COVID-19 disease, and there were few confirmed cases of COVID-19 in participants with evidence of infection prior to vaccination (although more cases occurred in the placebo group compared to the vaccine group). However, as shown in tables 22-23, the available data, while limited, indicate that previously infected individuals may be at risk for COVID-19 (i.e., reinfection) and may benefit from vaccination.

TABLE 22 vaccine efficacy-first development of COVID-19 from 7 days post dose 2, efficacy (7 days) population can be evaluated by test status-subjects with or without evidence of infection before 7 days post dose 2

TABLE 23 vaccine efficacy-first development of COVID-19 from 7 days after dose 2, by test State-subjects with or without evidence of infection before 7 days after dose 2-all available efficacy populations

Example 27: pharmacokinetic (PK) and absorption, distribution, metabolism and excretion (ADME) analysis of certain lipid excipients

This example describes various evaluated properties (e.g., PK/ADME profiles) of lipids used in the vaccine compositions described herein. Without wishing to be bound by any particular theory, it is noted that these characteristics of the lipid component may contribute to relevant characteristics (e.g., distribution, expression, etc.) of the administered vaccine, including efficacy in general and/or under particular circumstances (e.g., according to a particular protocol and/or when administered to a particular population, etc.).

Absorption of

After Intravenous (IV) bolus injection of the nanoparticle formulation in rats, single dose PK studies were performed on ALC-0315 and ALC-0319 to assess PK and metabolism of lipid excipients ALC-0315 and ALC-0159. This study used LNP (same lipid composition as BNT162b 2) containing alternative luciferase RNA to study the in vivo disposition of ALC-0159 and ALC-0315.

In this 2 week study, the concentration of ALC-0159 in plasma and liver decreased by about 8000-fold and>250 times. For ALC-0315, the rate of removal of molecules from plasma and liver is slow, but plasma and liver concentrations decrease by about 7000-fold and 4-fold over two weeks, respectively. Overall, the apparent terminal t of plasma and liver1/2Similarity in both tissues was 2-3 days and 6-8 days for ALC-0159 and ALC-0315, respectively. The apparent end t1/2 in plasma may represent redistribution of individual lipids back into plasma from the tissues to which they were distributed as LNPs, where they are cleared.

Metabolism

The in vitro metabolism of ALC-0315 and ALC-0159 was evaluated in blood, liver microsomes, S9 fractions and hepatocytes of mice, rats, monkeys and humans. Rat plasma, urine, feces and liver samples were examined for in vivo metabolism in PK studies. In vitro and in vivo, metabolism of ALC-0315 and ALC-0159 appears to occur relatively slowly. It is clear that ALC-0315 and ALC-0159 are metabolized by the hydrolytic metabolism of the ester and amide functional groups, respectively, and this hydrolytic metabolism is observed in all species evaluated.

Excretion device

Voiding studies appear to demonstrate that 50% of ALC-0159 is cleared unchanged in feces and that metabolism plays a role in clearing ALC-0315, as little or no unchanged material is detected in urine or feces.

Urine, stool, and plasma studies from rat PK studies identified a series of ester cleavage products of ALC-0315. Without wishing to be bound by any particular theory, it is believed that this may represent the primary clearance mechanism acting on the molecule in vivo.

In vitro, ALC-0159 is slowly metabolized by hydrolytic metabolism of amide functionality.

Example 28: profiling of administered vaccine compositions

The in vivo biodistribution of the COVID-19 mRNA vaccine BNT162b2 was evaluated using mice as a model system and evaluating luciferase expression as a surrogate reporter. Protein expression was demonstrated at the injection site and to a lesser extent, and more transiently in the liver, after IM injection of luciferase-encoding RNA in a bnp formulation similar to BNT162b2 in mice. Luciferase expression was identified at the injection site 6 hours after injection and reduced to near baseline levels on day 9. Expression was also present in the liver 6 hours post injection, and was not detected 48 hours post injection. Except for liver, all other tissues evaluated contained equal to or less than 1% of the dose.

Example 29: repeated dose toxicity study for various dosing regimens

GLP-compliant repeat dose studies were performed in rats to assess the immunogenicity and toxicity of COVID-19mRNA vaccines (including BNT162b 2).

In certain studies, male and female Wistar Han rats were administered a vaccine composition as described herein; compositions based on various RNA platforms (e.g., BNT162b2) were tested for IM injection into the hind limb 3 times each every other week (days 1, 8 and 15 dosing). Different doses (10, 30 and 100 μ g) were tested; the lower dose was given as a single injection of 20-70 μ l, while the highest dose (100 μ g) and controls were given as two injections, 100 μ l each (once per hind limb). The control was phosphate buffered saline/300 mM sucrose, corresponding to the storage buffer of the vaccine product. Each group had 18 male and 18 female rats, assigned 10 as the primary study, and recovered 5 groups, and 3 additional animals were used for cytokine analysis. The recovery period was 3 weeks after the last dose. Necropsies were performed on day 17 of the study, approximately 48 hours after the last dose and after 3 weeks recovery.

No unexpected death was observed.

Dosing was considered well tolerated without showing any signs of systemic toxicity; the body temperature increased slightly within a few hours after the dose administration and the body weight was reduced in the same period, but these levels were not considered to be undesirable.

Local inflammatory reactions were observed at the site of intramuscular injection. Changes in injection site were edema, erythema and induration (induration), more severe and more frequent after the second and/or third dose compared to the first dose; however, these resolved before the subsequent dose was administered and recovered completely at the end of the 3-week recovery period.

Macroscopic findings at the injection site included induration or thickening (inching), occasionally accompanied by incrustation, which was observed in almost all rats. Under the microscope, this is associated with inflammation and variable fibrosis, edema and myofibrosis. Inflammation at the injection site is accompanied by elevation of circulating leukocytes and acute phase proteins (fibrinogen, alpha-2 macroglobulin, and alpha-1 acid glycoprotein).

Occasionally, inflammation occurs and spreads to tissues near the injection site. At the end of dosing, there was significant drainage (iliac) lymph node enlargement. This is associated with increased cell type at the germinal center and increased plasma cells in draining (iliac) lymph nodes and is the expected immune response to the administered vaccine.

Under the microscope, enlargement of the spleen and increase in spleen weight are also associated with increased hematopoiesis, which is also evident in the bone marrow. These findings may be the result of an immune/inflammatory response secondary to the vaccine.

At the end of the recovery period, the injection site was normal and was relieved by clinical pathology and macroscopic observation, and microscopically evidence suggests that inflammation at the injection site was recovered.

After the dosing phase, there are microscopic vacuoles of portal hepatocytes. After the recovery period, this observation disappeared. Alanine aminotransferase (ALAT) was not elevated. All vaccinated rats had elevated gamma-glutamyl transferase (GGT), but there were no macroscopic or microscopic findings consistent with cholestasis or hepatobiliary damage to explain the increased gamma-GT activity, which had completely disappeared at the end of the 3-week recovery period. Vacuolization may be related to the hepatic distribution of pegylated lipids in LNP. No change in serum cytokine concentration was observed.

No effect on ophthalmic and auditory evaluation, nor on appearance or behavior; in particular, gait is normal, which means that the observed changes do not affect the activity of the rat. Serum cytokine concentrations were not subject to vaccine-related changes.

Immunogenicity testing has shown that COVID-19mRNA vaccines (including, for example, BNT162b2, such as BNT162b2 v8) elicit specific IgG antibody responses to SARS CoV-2 spike protein against S1 fragment and receptor binding domain. Neutralizing antibody responses of the vaccine were also observed in the pseudovirus neutralization assay.

The results of the ELISA assay are shown in figures 117 and 118 (as shown, from day 17 or day 10), where the uppermost trace (trace) is the trace of the COVID-19mRNA vaccine BNT162b2, and the other traces are the traces of other COVID-19mRNA vaccines described herein using different constructs: similar results were shown on day 38 (not shown here). These were converted to neutralizing activity as seen in the VSV/SARS-CoV-2-S pseudovirus neutralization assay using Vero 76 cells (figure 119): the results at day 38 were similar (not shown here). Of all the vaccines tested in this study, those with higher antigen-specific antibody titers also had more pronounced virus neutralization.

The COVID-19mRNA vaccine (e.g., BNT162b2) is well tolerated, produces inflammatory changes at the injection site and draining lymph nodes, increases hematopoiesis in the bone marrow and spleen, and the clinical pathology results from immune responses or inflammation at the injection site.

One skilled in the art reading this disclosure will appreciate that the findings in this example may be considered typical of those expected from dosing various mRNA constructs and/or lipid nanoparticles as described herein.

Example 30: toxicity and immunogenicity studies with a three dose regimen

A study was conducted to evaluate toxicity in rats given a COVID-19mRNA vaccine (e.g., BNT162b 2). This study met good laboratory specifications.

BNT162b2 was administered to hind limbs of male and female Wistar Han rats 3 times with one week intervals (dosing on days 1, 8, 15) by IM injection. Necropsies were performed on day 17 of the study, about 48 hours after the last dose, and after 3 weeks recovery. The COVID-19mRNA vaccine BNT162b2 was supplied at 0.5mg/ml in a dose volume of 60. mu.l, given at 30. mu.g per dose. Control rats received physiological saline.

Blood was collected at various time points during the evaluation period, before and during dosing and during recovery, and antibody responses to the vaccine components were evaluated.

All rats given the COVID-19mRNA vaccine (e.g., BNT162b2) survived to the scheduled necropsy time: no change in clinical signs was observed, nor was body weight. The food intake was reduced on days 4 and 11 (0.83 times of the control group), and there was an increase in average body temperature on days 1 (up to 0.54 ℃), 8 (up to 0.98 ℃) and 15 (up to 1.03 ℃) after dosing compared to the control group.

Edema and erythema appeared at the injection site on day 1 (minimal edema and minimal erythema), day 8 (minimal edema and minimal erythema), and day 15 (minimal edema and minimal erythema), which completely disappeared, and were not found before dosing on days 8 and 15.

Hematology tests showed that leukocytes were high (up to 2.95-fold of control), mainly involving neutrophils (up to 6.80-fold of control), monocytes (up to 3.30-fold of control) and large unstained cells LUC (up to 13.2-fold of control), and slightly higher eosinophils and basophils on days 4 and 17. Leukocytes were higher at day 17 compared to day 4. There was a transient decrease in reticulocytes in both sexes at day 4 (0.27-fold over control), while there were higher reticulocytes in females only at day 17 (up to 1.31-fold over control). On days 4 and 17, there was a lower red blood cell count parameter (to 0.90-fold control). On days 4 and 17, there was a lower A: G ratio (up to 0.82 fold). Higher fibrinogen (up to 2.49 fold) was observed on day 17 compared to control, consistent with an acute phase response. Acute phase proteins alpha-1-acid glycoprotein (up to 39 fold on day 17) and alpha-2 macroglobulin (up to 71 fold on day 17) were elevated on days 4 and 17, with higher concentrations in males. There was no change in urinalysis parameters.

After necropsy, the absolute and relative weights of the spleens of vaccinated rats were high (up to 1.42 times for males and 1.62 times for females). There were no other changes in organ weight. Macroscopic findings included enlargement of draining lymph nodes and pale/dark firm injection sites (firm) in a few vaccinated rats. Dosing was tolerated without inducing any systemic toxicity, all changes were consistent with inflammatory responses and immune activation: it was found to be consistent with that normally associated with dosing lipid nanoparticle-encapsulated mRNA vaccines.

Example 31: reproductive toxicity

A study was conducted to evaluate the reproductive toxicity in female rats administered with COVID-19mRNA vaccine (including BNT162b 2). Female rats are given the COVID-19mRNA vaccine (e.g., BNT162b2) twice before mating begins and a human clinical dose (e.g., 30 μ gRNA/dose day) twice during pregnancy. The COVID-19mRNA vaccine was administered Intramuscularly (IM) to F0 female Wistar rats 21 days and 14 days prior to the onset of mating (M-21 and M-14, respectively), followed by 4 doses on Gestational Days (GD)9 days and GD20 days. One subgroup was terminated at GD21, while the other (litter) group was terminated on the postpartum day (PND) 21. SARS-CoV-2 neutralizing antibody titers were found in most females pre-mating (M-14), most females and late gestation fetuses (GD21), and most late lactation offspring (PND 21). There was a transient decrease in body weight gain and food consumption after each dose. No effect on the oestrus cycle or fertility index was observed. Although pre-implantation losses increased (by a factor of about 2) (compared to controls), the percentage of pre-implantation losses observed in the vaccinated group were within the historical control data (5.1% -11.5%). In the fetus (total n ═ 21 mothers/fetuses), the incidence of abdominal fissures, mouth/jaw abnormalities, right aortic arch and/or cervical spine abnormalities is very low. For the bone study, the exposure group's sacral anterior arch (anterior vertebral arch), suprarenal short rib (suprarenal lumbar rib), supralumbar short rib (supralumbar lumbar short rib), and caudal vertebra number <5) were found to be comparable to the control group level. There was no indication of adverse effects on postpartum pups (terminated at PND 21).

This study showed that there was no significant adverse effect on fertility and early embryogenesis.

Example 32: safety and immunogenicity of the SARS-COV-2BNT162b1 mRNA vaccine in young and older adults in china: randomized, placebo-controlled, observer-blind phase I study

This example reports the preliminary results of a phase I trial testing BNT162b1 in 144 healthy chinese participants. BNT162b1 encodes the SARS-CoV-2 spike glycoprotein Receptor Binding Domain (RBD) and is one of several RNA-based SARS-CoV-2 vaccine candidates described herein.

This example specifically reports the observed safety profile, with fever >39 ℃ being the only reported adverse event of grade 3. Prime-boost vaccination with 10 μ g or 30 μ g BNT162b1 induced robust antibody and T cell responses in young (18-59 years) and older (65-85 years) adults in china. Both dose levels induced serum turnover after 41 days: the geometric mean titers of SARS-CoV-2 serum neutralizing antibodies in young participants of the 10. mu.g and 30. mu.g dose groups were 1.9 and 2.1 times higher than the convalescent serum of the restored COVID-19 patients, respectively; 0.7 and 1.3 times in older participants. Interferon-gamma T cell responses to RBD antigen challenge were significantly higher in participants receiving BNT162b1 than in placebo.

In young and older adults in china, increased reactogenicity and more favorable vaccine-elicited virus neutralization responses were associated with a dose of 30 μ g of BNT162b 1.

The safety and immunogenicity data for BNT162b1 provided in this example, particularly with respect to vaccination of healthy, young and elderly chinese participants, indicate that at least 28 days after prime-boost vaccination, a dose level of 10 μ g and 30 μ g of BNT162b1 vaccine induced strong humoral and cellular immunogenic responses in both 18-55 years old young adults and 65-85 years old older adults, with robust RBD-specific antibody and T cell responses seen in both young and elderly participants. Certain findings in this study further confirmed the tolerability profile of BNT162b1, as also observed in the us and german populations, for example.

Method

Randomized, placebo-controlled, observer-blind phase 1 trials were conducted in 144 healthy young adults aged 18-59 years and older adults 65-85 years of age, Taizhou, Jiangsu province, China. Eligible participants were randomized to receive two doses of either 10 μ g or 30 μ g of BNT162b1 or placebo, administered by intramuscular injection, 21 days apart. Study participants, researchers, and laboratory workers were blind to treatment management. The main safety endpoints are local or systemic adverse reactions at the injection site within 14 days after vaccination, and adverse events occurring up to 28 days after receiving booster vaccination. At predetermined time points, the immunogenic endpoints of the vaccine-elicited virus neutralizing antibodies as well as antigen-specific binding antibodies and cellular immune responses are measured.

Results

Study design and analysis setup

296 adults aged 18-55 years or 65-85 years were screened in the clinical research center for vaccines in thazhou city, Jiangsu province, China. 144 eligible participants agreed to participate in the trial and were randomized to 1:1 prime and boost doses at 21 day intervals, or two placebo doses, to receive 10 μ g or 30 μ g of BNT162b 1. After the priming dose, two participants aged 65-85 years withdrew booster doses (one at 10 μ g and one at 30 μ g). Table 24 shows the demographic characteristics of the participants. In the treatment groups, the average age of the young participants was 37.9-42.0 years, the average age of the older participants was 68.5-70.7 years, and the gender distribution was equal between the treatment groups. The medical history or current underlying disease of participants was similar between treatment groups, except for hypertension, which was present in older participants at baseline.

Observed safety and tolerability data

Within 14 days after completion of dosing, 21 (88%) young participants in the 10 μ g BNT162B1 dose group and 24 (100%) young participants in the 30 μ g BNT162B1 dose group reported at least one adverse reaction solicited compared to 17% of the young participants in the placebo group (tables 25A-25B). Reactogenicity was dose level dependent and was most evident in the 30 μ g BNT162b1 dose group. The most common solicited adverse reactions reported were pain at the injection site, fever, headache, fatigue, weakness, joint pain, muscle pain, chills. Adverse events were transient, managed with simple standard of care, or resolved spontaneously (resolve). The severity of most of the adverse effects reported were mild or moderate and resolved within the first 7 days after each dose of BNT162b 1. No injection site reactions were graded as severe (grade 3). All grade 3 systemic adverse reactions associated with vaccination were fever, observed mainly in the young participants. One male participant in the senior group experienced a sporadic grade 3 fever with injection site pain and itching following administration of a 30 μ g dose of the priming BNT162b1, and selectively withdrew from booster vaccine administration.

During the course of the study, the pre-specified trial stop rule was not reached. Only one 67 year old participant reported a serious adverse event (humeral fracture due to car accident, rendering the participants unacceptable to booster doses) considered unrelated to the vaccine or study procedure. The overall frequency of injection site adverse reactions following immunization was comparable after the priming and boosting doses of BNT162b 1. In young adults, some systemic adverse effects, such as fever, headache, fatigue and fatigue, are more common after a BNT162b1 booster dose than after a prime dose. No increase in reactogenicity occurred in older participants after the boost dose of BNT162b1 compared to younger participants.

No blood pressure and respiratory rate changes were reported in participants in the different treatment groups before and after BNT162b1 administration. A transient increase in body temperature and pulse rate was found in young and old participants, especially in the 30 μ g dose group, 24 hours after vaccination. The most common abnormalities in laboratory values compared to baseline are a transient decrease in lymphocyte and platelet counts and an increase in C-reactive protein. All laboratory abnormalities were self-limited (self-limited) and resolved within a short time with no clinical presentation. These data are consistent with certain findings reported in other populations (e.g., as described elsewhere herein).

Vaccine induced antibody response

All participants were seronegative at baseline (day 1, before vaccination) and seroconversion was monitored by analysis of SARS-CoV-2 neutralizing antibodies and RBD and S1 protein binding antibodies on days 8, 22, 29 and 43. The antibody response induced by BNT162b1 in the vaccinated participants was compared to a panel of human covd-19 convalescent sera obtained from 28 covd-19 patients at least 14 days after PCR confirmation. The highest neutralization titers were observed at day 43 (i.e., 21 days post-BNT 162b1 booster), indicating a continuing upward trend in neutralization titers in this asian group of participants after day 29, which appeared to increase further in neutralization titers in the older participants at day 43 compared to reports from other groups, indicating that peak titers appeared earlier in this subject population and subsequently declined. On day 43, the 10 μ g and 30 μ g BNT162b1 dose levels induced significant virus neutralizing antibody responses after the BNT162b1 priming dose, boosted with the second dose of BNT162b1, with Geometric Mean Titers (GMTs) of 232.9 (95% CI 151.3-358.5) and 254.0 (184.6-349.4) for the young participants and 80.0(49.2-130.2) and 160.0 (96.7-264.6) for the older participants in the 10 μ g and 30 μ g dose groups, respectively (figure 120). Viral neutralization responses of the mild aged participants in the 10. mu.g and 30. mu.g dose groups were 1.9 and 2.1 times greater than the GMT of one group of convalescent sera (GMT, 119.9; 95% CI, 70.4-203.9). The corresponding ratio in the 10 μ g and 30 μ g dose groups was 0.7 and 1.3 times in the older participants, respectively. All young recipients developed a positive seroconversion at day 43 and in older recipients at day 43, the seroconversion was 91% at the 10 μ g dose and 96% at the 30 μ g dose. Participants receiving a 30 μ g dose appeared to have a higher virus neutralizing antibody response than participants receiving a 10 μ g dose. However, older participants aged 65-85 generally exhibited slower virus neutralization and lower peak responses than younger participants aged 18-55.

Similarly, both doses of BNT162b1 induced high levels of S1-and RBD-binding IgG in the participants following the prime-boost regimen. Levels of S1-and RBD-bound IgG in vaccine recipients were highly correlated with neutralization titers at all time points post-vaccination, with correlation coefficients of 0.85 and 0.79(P <0.0001), regardless of age and dose group, respectively.

Vaccine induced T cell response

Induction of CD8 against vaccine in individuals immunized with BNT162b1 before (day 1), day 29 (7 days after booster vaccination), and day 43 (21 days after booster vaccination) with Peripheral Blood Mononuclear Cells (PBMC) by direct ex vivo IFN γ enzyme linked immunosorbent spot (ELISpot) assay+T cell responses were characterized. On day 29, the specific IFN- γ ELISpot response to the Sp1 peptide pool (overlaid RBD) was significantly higher in participants receiving BNT162b1 than in the placebo group (fig. 121). The average spot-forming cells receiving 10. mu.g of vaccination among young participants at 18-55 years of age were 227.5 (95% CI, 146.5-308.5)/105PBMC with an average spot-forming cell size of 223.5(181.2-265.9)/10 after 30. mu.g vaccination5And (5) PBMCs. In the older participants 65-85 years of age, spot-forming cells were slightly lower after vaccination in both dose groups, with an average of 156.5(84.1-229.0) and 171.9 (113.4-230.3). On day 43, young participants receiving the prime-boost BNT162b1 regimen tended to decline slightly in S1 specific IFN- γ ELISpot responses compared to those seen on day 29; no blood samples were collected from the elderly participants at this time point because the data was not available. No difference in IFN- γ ELISpot response to the Sp2 peptide pool (which did not include the peptide of the RBD encoded by BNT162b 1) was observed between BNT162b1 and the placebo group, and to CD8 in both dose groups +Small non-specific responses of T cells.

Discussion of the related Art

The experiments described in this example were performed in parallel with other BNT162 vaccine candidates in multiple regions in China14. One focus of the study was to establish data on the safety and immunogenicity of mRNA vaccines in asian populations. This example reports the first evaluation of the safety and immunogenicity profiles of this mRNA vaccine in the chinese population, as well as the safety and immunogenicity profiles of the chinese population, which is the young and old of china.

This is a preliminary report of a clinical trial of a modified RNA-based SARS-CoV-2 vaccine candidate BNT162b1, BNT162b1 encoding SARS-CoV-2 RBD, administered to a healthy chinese adult population. BNT162b1, like BNT162b2 (modRNA encoding S protein, derived from the same nucleoside modification platform), induced strong vaccine-induced antibody responses and strong T cell responses. The clinical safety and immunogenicity of BNT162b1 and BNT162b2 candidates have been evaluated in a population of healthy adults in Germany (young adults; BNT162-01) and in the United states (young adults and older adults 65-85 years of age; BNT 162-02). In the young adult group, there were fewer severe local reactogenicity AEs within 7 days for the us study (BNT162-02) and the present study (BNT162-03) compared to the german study (BNT 162-01). Systemic reactogenicity AE over 7 days were approximately similar in each study. Systemic AE (independent of correlation) in BNT162-02 and BNT162-03 studies was slightly higher within 28 days after dose 2 compared to the BNT162-01 study.

In the older adult group, the severe local reactogenicity AE within 7 days were similar in each study. The systemic reactogenicity AE in the BNT162-03 study was slightly lower within 7 days compared to the BNT162-01 and BNT162-02 studies. Systemic AEs (not related to correlation) were slightly higher in the BNT162-03 study over 28 days after dose 2, whereas severe AEs were lower compared to the BNT162-02 study. In summary, comparative analysis of the BNT162-01, BNT162-02 and BNT162-03 studies BNT162b1 safety profiles at 30 μ g showed a substantially comparable profile, and in the systemic reactogenicity/annual population, the safety profile in the asian population was even better than the non-asian population. Therefore, in clinical trials conducted in Germany and the United states7,8,15The findings reported herein further complement andreport of extended BNT162b1 and other RNA-based vaccine candidates.

The rationale for this study was to assess whether the intrinsic and extrinsic differences between the german and chinese populations had any effect on the tolerance or immune response of this novel vaccine. In local and systemic reactions7,15In terms of severe reactogenicity, the safety profile of the vaccine candidate BNT162b1 observed in our study appeared to be better in healthy chinese adults than reported in other populations. Body habits, endogenous antibody repertoires (repotoreies) may have an effect. The reactogenicity of BNT162b1 was dose dependent. An increased frequency of adverse events was observed following administration of booster immunizations compared to after primary immunization, especially in young participants. Older adults have a lower incidence of adverse effects than younger participants. Grade 3 fever was reported in 17% of the young participants and 8% of the older participants who received a 30 μ g dose. Almost all severe fever reactions are transient and self-limiting. 1 subject withdrew from booster vaccination in response to the priming dose, had occasional fever or cold intolerance, had or had no body temperature recordings, with pain, itching and itching at the injection site, for more than two weeks, and resolved after administration of hydrocortisone butyrate ointment. A transient decrease in lymphocyte count, which is a pharmacodynamic marker, was observed predominantly in young recipients at a dose level of 30 μ g of BNT162b1, which correlates with the redistribution of lymphocytes into lymphoid tissues by innate immune stimulation 16

Both doses of the vaccine candidate BNT162b1 were effective in eliciting specific humoral and cellular immune responses, with the second vaccination having a significant boosting effect on the antibody titers found in young and older adults. Administration of BNT162b1 at a dose of 30 μ g following a prime-boost regimen induced an optimal immune response level against virus neutralizing antibodies to SARS-CoV-2 that was higher than the level in a panel of human convalescent serum samples, regardless of age. The humoral response in chinese participants showed a unique temporal pattern and peaked at day 43 in both age groups. Although the number of participants is small and differences in the measurement methodology that may occur may affect the observed results, the findings reported here suggest that there may be population differences in the response to the vaccine.

Since the vaccine candidate BNT162b1 is a modified RNA vaccine encoding the trimeric form of RBD, the vaccine recipients of the study reported in this example showed a significant specific T cell response to the S1 peptide pool (containing 166 15-membered S1 peptides from human SARS-CoV-2 virus), but not to the S2 peptide pool. The results indicate that the cellular response elicited by BNT162b1 is antigen-specific. In contrast, the vaccine candidate BNT162b2 profile (spectrum) is distinct from other RNA-based SARS-CoV-2 vaccines, inducing a T cell response that is able to recognize the S1 and S2 peptide pools 15. However, the data show that BNT162b1 at a dose of 30 μ g is highly immunogenic and is capable of eliciting strong humoral and cell-mediated responses in healthy chinese adults.

Those skilled in the art recognize that small sample sizes and age limits of 18 years and above may limit the severity of the conclusions observed in this example. In any event, since the prophylactic RNA vaccines described herein represent a novel vaccination approach, safety assessments, including those in specific populations (e.g., children and adolescent populations) are particularly valuable. Likewise, while comparison of the seroneutralization response elicited by the vaccine candidates described herein to the human convalescent serogroup provides meaningful vaccine assessment, the serological level of immunity required to provide protection against COVID-19 has not been rigorously established17. Those skilled in the art also recognize that human convalescent serogroups that have been used in different trials have not been standardized between laboratories and thus may have different patient characteristic distributions and acquisition time points, such that direct comparison of results (e.g., characterizing different vaccine candidates and/or characterizing vaccine candidates relative to different convalescent sera) may not provide useful information.

In summary, the results described in this example demonstrate the dose-dependent safety and good immunogenicity of RNA-based SARS-CoV-2 vaccine candidate BNT162b1, and further extend the previous findings of BNT162b1 in Germany and United states experiments7,8,15. At a young ageAnd in older adults, increased reactogenicity and more favorable vaccine-elicited virus neutralization responses were found to be associated with 30 μ g of BNT162b 1. In contrast, another vaccine candidate BNT162b2 produced from the same platform showed a more favorable safety profile8. And BNT162b2 encoding the full-length spike immunogen18In contrast, BNT162b1 encodes a relatively small RBD immunogen, which is likely to induce a narrow range of neutralizing antibodies, which is less robust against the potential antigenic drift of SARS-CoV-2. Notably, the candidate BNT162b2 was found to be more than 95% effective at preventing COVID-19 in participants, without a decrease in efficacy in the population over 65 years of age19

Method

Study design and participants

This randomized, placebo-controlled, observer-blind phase I trial was conducted in Taizhou, Jiangsu province, China, in healthy young adults 18-59 years of age and older adults 65-85 years of age. The overall good health of the participants was determined by screening the visit for medical history, physical examination, and laboratory tests. Both men and women were included and agreed to contraceptive measures during the trial. We excluded participants in pregnancy or lactation. Participants who tested positive for SARS-CoV-2 by the IgM/IgG antibody commercial Rapid diagnostic kit for SARS-CoV-2 (manufactured by Livzon diagnostics inc., Zhuhai, China) or by the pharyngeal swab nucleic acid diagnostic test (manufactured by Fosun pharma, Shanghai, China) were excluded. The appearance of COVID-19 in breast CT scans is a further exclusion criterion. Participants with severe cardiovascular disease or chronic disease such as uncontrolled diabetes and hypertension, human immunodeficiency virus, hepatitis b and hepatitis c were excluded. Written informed consent was obtained from each participant prior to the start of the study.

The study was conducted according to the Declaration of Helsinki and Good Clinical Practice (Good Clinical Practice). The test protocol was approved by the review committees of National medicine Products Administration (China) and the National Center for Prevention and Control of Disease in Jiangsu province (Jiangsu province Center of Disease Control and preservation).

Randomization and blinding

Eligible participants aged 18-55 were enrolled in the younger group, and older participants aged greater than or equal to 65 years and less than or equal to 85 years were enrolled in the older group. Participants were randomized in a 1:1:1 ratio to receive either low dose BNT126b1 or high dose BNT126b1 or placebo. Participants are ranked by gender using a Web-based Interactive Response Technology (IRT) system. The masked randomized list was generated by an independent statistician using SAS software (version 9.4).

An authorized non-blind pharmacist prepares a vaccine or a placebo according to the distribution situation of the participants through the IRT system, and a nurse manages the participants for study products. The non-blind staff did not participate further in the trial and prohibited disclosure of the assignment information to others. All other researchers, participants, lab workers, and sponsors were blind throughout the experiment.

Vaccines and vaccination

BNT162b1 was administered consisting of Good Manufacturing Practice (GMP) grade mRNA drug substance encoding the trimeric SARS-CoV-2 spike glycoprotein RBD antigen formulated with lipids to obtain the RNA-LNP drug product. As described herein, the vaccine is shipped and provided as a buffered liquid solution for intramuscular injection and stored at-80 ℃.

The low-dose and high-dose BNT126b1 contained 10 μ g and 30 μ g of active ingredient, respectively, while the placebo was a commercial saline solution. Each participant received a prime-boost dosing regimen of 10 μ g/0.5ml or 30 μ g/0.5ml of the vaccine candidate BNT162b1 or 0.5ml of placebo administered to the deltoid muscle at 21 day intervals.

Safety and immunogenicity monitoring

Each participant was asked to remain on the study site at least six hours after vaccine administration to donate blood samples 24 hours before and after the primary immunization and before and after the booster vaccinationBlood samples were re-donated 8 days later for laboratory testing. Vital signs including body temperature, blood pressure, pulse and respiratory rate were measured at baseline and one, three and six hours after vaccination. Participants used a log to record any adverse events following vaccination until day 28 after booster dosing. Young group participants were enrolled and received vaccination first. Enrollment in the older groups was initiated after evaluation of the first 14 days of preliminary safety data after prime vaccination in the young groups. The severity of adverse events and laboratory abnormalities were determined by the Chinese State Food and Drug Administration 20And the United states Food and Drug Administration (FDA)21Published specifications (scale) are ranked.

Serum and PBMCs were collected prior to vaccination, at day 8 and/or day 22 post booster dose to facilitate measurement of specific IgG antibody responses to RBD and spike glycoprotein S1, neutralizing antibodies to SARS-CoV-2, and T cell responses.

Researchers reviewed all reported adverse events. Adverse events are classified as likely, likely or positively associated with vaccine candidates.

Human convalescent serum

The neutralization titer is the reciprocal of the highest sample dilution that protects at least 50% of the cells from cytopathic effects. A set of 24 convalescent human serum samples was obtained from donors recovered from SARS-CoV-2 infection between 18 and 70 years of age (mean age 45.8 years); samples were obtained at least 14 days after confirmation of diagnosis by polymerase chain reaction and resolution of symptoms. The severity of the disease in these patients varies from asymptomatic (n-3, 13%), mild (n-8, 33%), moderate (n-10, 42%) or severe (n-3, 13%).

The geometric mean titer of neutralization (GMT) in the donor subgroup was as follows: 40 for 3 asymptomatic infected donors; 91.9 for 8 mildly infected donors; 160 in moderate infected patients; 226.3 were used as 3 severely infected donors. Each serum sample in the group was from a different donor. Thus, most serum samples were obtained from persons with moderate Covid-19. Convalescent serum samples were tested in parallel comparisons with serum samples obtained from participants in the test.

ELISA

We used an enzyme-linked immunosorbent assay (ELISA) to assess the binding antibody response against SARS-CoV-2 RBD and S1.

Micro-neutralization assay

We detected SARS-CoV-2 specific neutralizing antibodies in serum by the micro-neutralization assay based on cytopathic effects observed by SARS-CoV-2 strain BetaCoV/JS02/human/2020(EPI _ ISL _411952) in the Biosafety laboratory grade 3 (BSL-3).

ELISpot

Specific T cell responses to peptides were assessed by using a commercial ex vivo interferon-gamma (INF-gamma) enzyme linked immunospot (ELISpot) kit manufactured by Mabtech (Nacka Strand, Stockholm, Sweden)22. PBMCs were isolated from fresh blood samples and stimulated with different overlapping peptide pools prior to measurement. Used in this study were: s1 peptide pool covering the N-terminal half of the SARS-CoV-2 spike, including RBD; and S2 peptide pool covering the C-terminus of the SARS-CoV-2 spike, excluding RBD23. Peptide pool (CEF peptide pool) consisting of 32 MHC class I restricted viral peptides from human cytomegalovirus, Epstein-Barr virus and influenza virus for stimulation of CD8+T cells to assess general T cell reactivity (not specific for SARS-CoV-2)24)。

Results

The primary and secondary objective of this trial was to evaluate the safety and immunogenicity of the vaccine candidate BNT162b1 in healthy chinese adults. The primary endpoints of safety assessments are the incidence of solicited local or systemic adverse reactions at the injection site 14 days after vaccination, and adverse events 28 days after immunization until receiving booster doses. Any clinical laboratory abnormalities from baseline to 24 hours or 7 days post-vaccination, as well as any Serious Adverse Events (SAE) that occurred, were also recorded.

Secondary endpoints of immunogenicity were Geometric Mean Titer (GMT), seroconversion and fold increase of virus neutralizing antibodies, as well as ELISA IgG antibodies binding to S1 or RBD measured at day 8, 22 after each vaccination. Seroconversion is defined as a four-fold or greater increase in antibody titer compared to baseline, or a lower limit if the baseline titer is below the detection limit. Serum dilutions for ELISA were performed from 1: 100 starts and serum dilution for the microneutralization assay starts at 1: 10.

As exploratory endpoints, the concentrations at 1X 10 on days 8 and 22 after the booster dose were investigated5(ii) cellular immune response to the number of positive cells secreting interferon gamma (IFN-. gamma.) in PBMC per well.

Statistical analysis

The total sample size for this study was 144 participants, with each treatment group including 24 participants in each age group. The probability of at least one event observed in 24 participants per dose group was 86.5%, based on the assumption that the incidence of adverse reactions was 8% post-vaccination.

All randomized participants who received at least one dose of the investigational vaccine were included in the safety analysis. The safety endpoint is described as the frequency (%) of the 95% Confidence Interval (CI) for the adverse reaction or event during the observation. We compared the proportion of participants with adverse reactions or events for each group using Chi-square or Fisher's exact method (eaxct). All participants who received at least one vaccination and had serological measurements before or after vaccination were included in the immunogenicity analysis. The immunological endpoints were summarized descriptively at the indicated time points and compared among groups using ANOVA for logarithmic (log) transformed antibody titers and Wilcoxon rank sum test (rank-sum test) for non-normal data. The neutralizing antibody responses of the participants in each dose group were compared to those of patients who had been PCR diagnosed with SARS-CoV-2 infection. Any serological value below the lower limit of detection was set to half this value (1: 50 for ELISA and 1:5 for microneutralization assay), while values above the highest dilution titer were assigned the highest diluted value for calculation. Pearson correlation analysis of RBD or S1 specific ELISA antibodies and neutralizing antibodies was performed to assess the relationship between the responses of the different assays.

Reference to the literature

1.Gudbjartsson,D.F.,et al.Spread of SARS-CoV-2in the Icelandic Population.The New England journal of medicine 382,2302-2315(2020);see also WHO factsheet: https://covid19.who.int/.

2.Huang,C.,et al.Clinical features of patients infected with 2019novel coronavirus in Wuhan,China.Lancet(London,England)395,497-506(2020).

3.Jackson,L.A.,et al.An mRNA Vaccine against SARS-CoV-2-Preliminary Report. The New England journal of medicine 383,1920-1931(2020).

4.World Health Organization.DRAFT landscape of COVID-19candidate vaccines–3 November 2020.Vol.2020(2020).

5.Corbett,K.S.,et al.SARS-CoV-2mRNA vaccine design enabled by prototype pathogen preparedness.Nature 586,567-571(2020).

6.Anderson,E.J.,et al.Safety and Immunogenicity of SARS-CoV-2mRNA-1273 Vaccine in Older Adults.The New England journal of medicine(2020).

7.Mulligan,M.J.,et al.Phase I/II study of COVID-19RNA vaccine BNT162b1 in adults.Nature 586,589-593(2020).

8.Walsh,E.E.,et al.Safety and Immunogenicity of Two RNA-Based Covid-19 Vaccine Candidates.The New England journal of medicine(2020).

9.Wrapp,D.,et al.Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation.Science(New York,N.Y.)367,1260-1263(2020).

10.Güthe,S.,et al.Very fast folding and association of a trimerization domain from bacteriophage T4 fibritin.Journal of molecular biology 337,905-915(2004).

11.Krammer,F.SARS-CoV-2vaccines in development.Nature 586,516-527(2020).

12.O'Callaghan,K.P.,Blatz,A.M.&Offit,P.A.Developing a SARS-CoV-2Vaccine at Warp Speed.Jama 324,437-438(2020).

13.Poland,G.A.,Ovsyannikova,I.G.&Kennedy,R.B.SARS-CoV-2immunity:review and applications to phase 3vaccine candidates.Lancet(London,England)396,1595- 1606(2020).

14.Deming,M.E.,Michael,N.L.,Robb,M.,Cohen,M.S.&Neuzil,K.M.Accelerating Development of SARS-CoV-2Vaccines-The Role for Controlled Human Infection Models.The New England journal of medicine 383,e63(2020).

15.Sahin,U.,et al.COVID-19vaccine BNT162b1 elicits human antibody and T(H)1 T cell responses.Nature 586,594-599(2020).

16.Kamphuis,E.,Junt,T.,Waibler,Z.,Forster,R.Kalinke,U.Type I interferons directly regulate lymphocyte recirculation and cause transient blood lymphopenia.Blood 108, 3253–61(2006).

17.Hodgson,S.H.,et al.What defines an efficacious COVID-19vaccineA review of the challenges assessing the clinical efficacy of vaccines against SARS-CoV-2.The Lancet.Infectious diseases(2020).

18.Lan,J.,et al.Structure of the SARS-CoV-2spike receptor-binding domain bound to the ACE2 receptor.Nature 581,215-220(2020).

19.Pfizer Inc.Pfizer and BioNTech Announce Vaccine Candidate Against COVID-19 Achieved Success in First Interim Analysis from Phase 3Study.Vol.2020(2020).

20.Center For Drug Evaluation,NMPA.Guidances for grading adverse reactions in clinical trials of preventive vaccines.Vol.2020(2008).

21.US Food and Drug Administration.Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials.Vol.2020 (2007).

22.Slota,M.,Lim,J.B.,Dang,Y.&Disis,M.L.ELISpot for measuring human immune responses to vaccines.Expert review of vaccines 10,299-306(2011).

23.Buchan,S.A.,et al.Effectiveness of Live Attenuated vs Inactivated Influenza Vaccines in Children During the 2012-2013Through 2015-2016Influenza Seasons in Alberta,Canada:A Canadian Immunization Research Network(CIRN)Study.JAMA pediatrics 172,e181514(2018).

24.Victor,J.C.,et al.Efficacy of a Russian-backbone live attenuated influenza vaccine among children in Senegal:a randomised,double-blind,placebo-controlled trial.The Lancet.Global health 4,e955-e965(2016).

TABLE 24 Baseline characteristics of participants, by age group

Data are mean (SD) or n (%). "other" includes tonsillitis, helicobacter infection, human papilloma virus infection, periodontitis, electrocardiographic hypertension, lymphadenopathy, anemia, hepatic cyst, oropharyngeal discomfort, hyperthyroidism, noninfectious gingivitis, hyperlipidemia, benign prostatic hyperplasia, prostatitis, unilateral blindness, cerebral infarction, limb injury, spinal deformity, lithangiuria, and lymphadenopathy.

Table 25A: solicited adverse reactions within 14 days post-vaccination, up to non-solicited adverse reactions on day 43, by age group

Table 25B (modified) adverse reactions solicited within 14 days post-vaccination, and non-solicited adverse reactions up to day 43, age group by age group, including "placebo corrected" AE rates

Data is shown as the number of participants with the event (%). Grade 3 is a severe response (i.e. blocking activity). SAE is a serious adverse event. Within a particular response category, one participant only counted once, and more than once, for adverse reactions. Only two or more participants reported non-solicited adverse reactions. Participants with fever were ranked according to the guidelines of the U.S. Food and Drug Administration (FDA). Fever is also graded according to the grading guideline for adverse events of vaccine clinical trials issued by the national drug administration (NMPA), wherein grade 3 fever is defined as axillary temperature being more than or equal to 38.5 ℃. The subject experienced grade 3 fever after the priming dose, with pain, itching and itching at the injection site, and selectively withdrew from booster vaccination.

Example 33: serum elicited by BNT162b2 vaccine neutralizes the SARS-CoV-2 lineage B.1.1.7 pseudovirus

SARS-CoV-2 variant b.1.1.7 was detected in the uk at 9 months 2020, and its prevalence subsequently increased, showed enhanced transmission, and spread to other countries and continents. B1.1.7 has a series of mutations in its spike protein: Δ H69/V70, Δ Y144, N501Y, A570D, D614G, P681H, T716I, S982A and D1118H. Of these, N501Y is of particular interest because it is located at the receptor binding site; spikes with this mutation bind more tightly to their cellular receptor ACE-2; and the host range of viruses having such mutations is increased, including mice. Immunization of sera with BNT162b2 neutralized 19 pseudoviruses, each with SARS-CoV-2S with different mutations found in the circulating strain, as effective as the non-mutant pseudoviruses. The following studies show that viruses with a full set of mutations in british variant spikes can also be effectively neutralized by BNT162b 2-immune sera.

We generated VSV-SARS-CoV-2-S pseudovirus with either the Wuhan reference strain or the lineage B.1.1.7 strain spike protein. Sera from 16 participants of the previously reported trial (Sahin U.et al, medRxiv 2020.12.09.20245175; doi: https:// doi.org/10.1101/2020.12.09.20245175) were drawn from 8 young (18-55 years) and 8 older adults (56-85 years) 21 days after boosting with 30 μ g BNT162b2 and determined by 50% pseudovirus neutralization (pVNT) 50(ii) a FIG. 122), tested for neutralization of VSV against SARS-CoV-2 Wuhan and lineage B.1.1.7 spike-pseudotyped. The ratio of 50% neutralizing GMT of serum against SARS-CoV-2 lineage b.1.1.7 spike pseudotype VSV to 50% neutralizing GMT against wuhan reference spike pseudotype VSV was 0.79, indicating no significant biological difference in neutralizing activity against both pseudoviruses.

The neutralization of b.1.1.7 spike-bearing pseudoviruses by BNT162b 2-immune sera was retained, indicating that british variant viruses cannot escape the BNT162b 2-mediated protection. Furthermore, the use of a non-replicating pseudoviral system is not expected to be a potential limitation of work due to the good agreement between pseudotype neutralization and SARS-CoV-2 neutralization assays.

Materials and methods

Generation of VSV-SARS-CoV-2S variant pseudovirus

According to the published pseudotyped packaging method, a recombinant replication defective Vesicular Stomatitis Virus (VSV) vector encoding a Green Fluorescent Protein (GFP) and a luciferase instead of VSV-glycoprotein (VSV-G) is pseudotyped with SARS-CoV-2 spike (S) pseudotyped packaging (FIG. 123) (PMID: 21998709) derived from Wuhan reference strain (NCBI Ref:43740568) or variant of interest (VOC) -202012/01 (also known as SARS-CoV-2 lineage B.1.1.7). Briefly, HEK293T/17 monolayers transfected with VSV-G complementary VSV Δ G vectors to express SARS-CoV-2S were inoculated. After incubation at 37 ℃ for 1 hour, the inoculum was removed. Cells were washed with PBS and then medium supplemented with anti-VSV-G antibody (clone 8G5F11, Kerafast Inc.) was added to neutralize residual VSV-G complementation input virus. The medium containing the VSV-SARS-CoV-2-S pseudotype was harvested 20 hours after inoculation, filtered at 0.2 μm and stored at-80 ℃. Prior to use in neutralization tests, pseudovirus batches were titrated on Vero 76 cells and the percentage of infected cells was determined by flow cytometry (figure 124). Individual titers were calculated in Transduction Units (TU) per ml. Similar titers were obtained by generating VSV-SARS-CoV-2-S pseudovirus with the spike protein of the Wuhan reference strain or the panel B.1.1.7 strain (Table 26).

TABLE 26 Titers of SARS-CoV-2 Wuhan reference Strain and lineage B.1.1.7 Taurushi VSV, Transduction Units (TU)/mL

Serum samples and neutralization assays

Figure 125 schematically illustrates an immunization and serum collection protocol. For measurement of neutralization titers, each serum was serially diluted 2-fold in culture medium, the first dilution being 1: 20 (dilution range 1: 20-1: 2560). VSV-SARS-CoV-2-S particles were diluted in medium to obtain 100TU in the assay. Serum dilutions were mixed with pseudovirus at 1:1 for 30 min at room temperature, then added to Vero 76 cell monolayers in 96-well plates and incubated for 24 h at 37 ℃. The supernatant was removed and the cells were lysed with luciferase reagent (Promega). Fluorescence was recorded and neutralization titers were calculated by 4-parameter logistic (4PL) fitting that generated percent neutralization at each serial serum dilution in GraphPad Prism version 9. 50% pseudovirus neutralization Titers (pVNT)50) Reported as the reciprocal interpolated dilution giving 50% reduction in fluorescence. A table of neutralization titers is provided (table 27). The pVNT of each serum against SARS-CoV-2 lineage B.1.1.7 and Wuhan reference strain spike-pseudotyped VSV is plotted in FIG. 12650The ratio of (a) to (b).

TABLE 27.16 pVNT of post-BNT 162b2 immunized sera against SARS-CoV-2 Wuhan reference strain spike-pseudotyped and lineage B.1.1.7 spike-pseudotyped VSV 50Value of

Example 34: exemplary protocol for administering SARS-CoV-2RNA vaccine in pregnant women

This example describes an exemplary regimen for administering a SARS-CoV-2RNA vaccine described herein (specifically, in this example, BNT162b2) in a pregnant woman (e.g., a healthy pregnant woman aged 18 years and older).

Pregnant women are at risk of contracting SARS-CoV-2 infection and COVID-19. Pregnancy may lead to an increased risk of developing severe covi-19, since physiological changes during pregnancy may increase the susceptibility to respiratory infections and subsequently rapidly progress to respiratory failure. Furthermore, it has been reported that pregnant women with COVID-19 have a higher proportion of premature births, caesarean sections, fetal distress and infants in need of neonatal intensive care.

This example describes certain protocols according to which BNT162b2 can be administered to pregnant women and/or infants born to these pregnant women, and also describes certain assessments that can be made and/or results that can be achieved. For example, the present example describes a study that will evaluate the safety of BNT162b2 in pregnant women and their infants; BNT162b2 was also evaluated for immunogenicity in pregnant women, transfer of antibodies to their infants, and kinetics of antibody transfer in infants.

In other aspects, this example describes a study that will evaluate the safety and tolerability of prophylactic BNT162b2 when administered to vaccinated maternal participants at or above 18 years of age between 24-34 weeks of gestation. Without wishing to be bound by any particular theory, the present example suggests that starting vaccination during this period of time may provide particular advantages. This example describes a particular protocol in which a pregnant mother receives a first dose of vaccine between about 24 and about 34 weeks of gestation, or in some embodiments between about 27 and about 34 weeks of gestation, and after about 21 days, ideally before delivery of the infant, recognizing that Vaccination is advised at any time during Pregnancy (see, e.g., "Israel Recommendations in All classes of Pregnancy, and" Haaretz February 1,2021 ") and avoiding Vaccination during Pregnancy (see, e.g., WHO Strategical advisogenic Group recommendation).

Without wishing to be bound by any particular theory, the present example suggests that vaccination according to this approach may e.g. reduce the risk to the fetus, which e.g. may be exposed to an immune maternal immune response early in pregnancy. Furthermore, while not wishing to be bound by any particular theory, the present example proposes that the vaccination schedule provided may provide particular benefits when at least two doses are administered prior to delivery of the infant. Furthermore, the present embodiments propose that the provided approach may provide a particularly beneficial risk/benefit balance. Furthermore, the present disclosure teaches that immunity can be conferred to an infant by immunizing the pregnant mother, particularly by performing such immunization according to the protocols described in this example, which in some embodiments can be carried by delivery (which reduces the need for immunization of the infant for at least a period of days, weeks, months, or years post-partum (e.g., 1,2, 3, 4, 5, 6, 7, 8 weeks or more, or 1,2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 months or more, or 1,2, 3, 4, or 5 years). Thus, in some embodiments, as described herein, an infant born to a mother who is vaccinated during pregnancy, e.g., according to particular protocols described herein, may not require further vaccination for a period of time after birth (e.g., as described herein), or may require reduced vaccination (e.g., lower doses and/or fewer dosing times-e.g., boost-and/or lower overall exposure over a given period of time).

For example, among maternal participants who received at least 1 dose of study intervention per vaccine group, the percentage of maternal participants reporting the following will be evaluated: (i) local response up to 7 days after each dose; (ii) systemic events up to 7 days after each dose; (iii) AE 1 month after dose 1 to dose 2; (iv) SAE from dose 1 to 1 month postnatal. Alternatively or additionally, in maternal participants who met certain key agreement criteria (evaluable maternal participants) and who had no serological or virological evidence of past SARS-CoV-2 infection (up to 1 month after receiving the second dose): (v) GMR can be assessed as estimated by the ratio of the geometric mean of the SARS-CoV-2 neutralization titer in pregnant women 1 month after dose 2 to the geometric mean of the SARS-CoV-2 neutralization titer in non-pregnant women.

Still further alternatively or additionally, in maternal participants who met key agreement criteria (participants who can be evaluated) and/or who had or had not (e.g., those with and without, respectively, or independently) serological or virological evidence of past SARS-CoV-2 infection (7 days after receiving dose 2): (vi) can evaluate 100 × (1-IRR) [ ratio of active vaccine to placebo ].

Still further alternatively or additionally, one or more of the following may be evaluated:

among maternal participants from each vaccine group that meet key agreement criteria (evaluable maternal participants): (a) GMC/gmt at baseline (before dose 1), 2 weeks after dose 2, 1 month after dose 2 and 6 months post partum (delivery) (b) GMFR from baseline to 2 weeks after dose 2, 1 month after dose 2 and 6 months post partum;

-the percentage of infants among infants born by maternal participants who received at least 1 dose of study intervention per vaccine group, of: (a) specific birth results; (b) AE from birth to 1 month old; (c) SAE and AESI (major congenital malformations, developmental delay) by 6 months of age.

-in the infants born by evaluable maternal participants of each group of vaccines: (a) GMC and GMFR at birth and 6 months postpartum;

in the maternal participants who received BNT162b2 (at initial randomization and one month post partum): (a) incidence of follow-up every 1,000 people-year;

among maternal participants who received BNT162b2 at initial randomization and had no evidence of previous SARS-CoV-2 infection: (a) incidence of follow-up every 1,000 people-year

-in each subset of evaluable maternal participants for each vaccine group: (a) diagnosed COVID-19; (b) diagnosed severe COVID-19; (c) SARS-CoV-2 infection but no definitive diagnosis of COVID-19; (d) GMC/GMT and GMFR at baseline, 1 month after dose 2 and 6 months postpartum:

-in evaluable mother participants: (a) GMC/GMT at baseline and before dose 2; (b) GMFR from baseline to before dose 2;

-based on the breastfeeding status, in the infant born to the mother participant of each vaccine group: (a) GMC and GMFR at birth and 6 months after birth;

-based on breast-feeding status, among the infants born by the maternal participants who received at least 1 dose of study intervention per vaccine group, the percentage of infants that were: (a) AE from birth to 1 month old; (b) SAE and AESI (major congenital malformations, developmental delay) from 6 months old;

-in the infant born to the mother participant of each vaccine group: (a) the incidence of infant participants who have confirmed COVID-19;

-in the infant born to the mother participant of each vaccine group: (a) incidence of MIS-C.

In some embodiments, the first dose is administered during the 27-34 cycle of pregnancy of the pregnant woman, followed by the second dose after about 21 days. In some embodiments, the first dose is administered during the 24-34 cycle of pregnancy of the pregnant woman, followed by administration of the second dose after about 21 days. In some embodiments, the participant mothers are evaluated for up to about 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18 months or longer (e.g., after study initiation, after administration of the first dose, after administration of the second dose, and/or after birth of the infant).

In some embodiments, an infant born to a mother who has been administered one or more (e.g., two) vaccine doses (e.g., two doses administered during pregnancy) is evaluated for a period of up to about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 months or longer (e.g., after study initiation, after administration of the first dose, after administration of the second dose, and/or after birth of the infant).

In some embodiments, the dose will be 30 μ g of BNT162b2, as described herein.

In some embodiments, the assessment of vaccine performance is performed in a population of pregnant women at any age or within a particular age range (e.g., equal to or above 18 years of age). In some embodiments, the assessment of vaccine performance is performed in a population of women carrying a single birth.

In some embodiments, gestational age is assessed by one or more of the last menstrual period, ultrasonography, physical examination, and/or a combination thereof. In some embodiments, the gestational age is determined by ultrasound examination. In some embodiments, the gestational age is determined by considering two or more assessments (e.g., two or more ultrasound exams performed at different times of pregnancy, such as at different gestational periods).

In some embodiments, the assessment of vaccine performance is performed in a population having one or more of the following characteristics: ultrasound examinations performed at least 18 weeks gestation, and no significant fetal abnormalities were observed (e.g., assessed by a licensed researcher); (ii) the recorded HIV, syphilis and/or HBV detection or combination thereof is negative; BMI before pregnancy is less than or equal to 40kg/m2

In some embodiments, the assessment of vaccine performance is performed in a population that does not include subjects with one or more of the following characteristics: a medical or psychiatric condition with a risk of potentially increasing vaccination, or otherwise, the subject is deemed to be unsuitable for receiving the vaccine according to the sound judgment of the licensed researcher; a previous clinical or microbiological diagnosis is COVID-19; a history of severe adverse reactions associated with the vaccine and/or a history of severe allergic reactions to any component of the vaccine (e.g. anaphylaxis); a known or suspected immunodeficiency; hemorrhagic constitution or disease conditions associated with long term bleeding, gestational hypertension or preeclampsia-eclampsia, placental abnormalities, polyhydramnios or oligohydramnios, significant bleeding or coagulation disorders, gestational diabetes mellitus, current pregnancy with one or more signs of preterm birth, or current pregnancy with continued intervention (medical/surgical) to prevent preterm birth, prior death or neonatal death, prior low birth weight or preterm birth, prior history of at least 3 abortions, prior pregnancy times more than 5, or prior infants with known genetic disease or major congenital malformation, previously vaccinated with any coronavirus vaccine, received a drug intended to prevent COVID-19, received or scheduled to receive (1 exception, anti-D immunoglobulin (e.g., RhoGAM) by parturition (ivedely) 60 days before study intervention administration (which may be given at any time) blood/plasma preparations or immunoglobulins, subjects currently under treatment with alcohol abuse or illicit use of drugs, receiving immunosuppressive therapy (including cytotoxic agents or systemic corticosteroids, e.g., for cancer or autoimmune diseases, or scheduled for post-vaccination blood withdrawal) are enrolled in additional studies involving study intervention within 28 days prior to enrollment and/or during enrollment, are enrolled in additional studies involving study intervention with LNP, are currently suffering from febrile disease, are currently symptomatic of COVID-19 infection, are enrolled in any seasonal or pandemic influenza vaccine within the last 14 days, are eligible to receive any seasonal or pandemic influenza vaccine within 7 days after study intervention, are eligible to receive a vaccine containing tetanus, diphtheria and/or pertussis within the last 14 days, and are eligible to receive an intervention containing tetanus, diphtheria, pertussis within 7 days after study administration, Diphtheria and/or pertussis vaccines, receive short (<14 days) systemic corticosteroids (allowing for inhalation/nebulization, intra-articular, intracapsular or topical (skin or eye) corticosteroids) less than 28 days prior to dosing.

In some embodiments, a vaccinated mother as described herein may be taking or may begin taking, for example, an antipyretic or other analgesic to treat the symptoms associated with vaccination. Alternatively or additionally, in some embodiments, the mother may be taking or may begin taking medications required to treat a pre-existing stable disease condition and/or inhaling, topically or locally injecting corticosteroids.

In some embodiments, a vaccinated mother as described herein may be administered one or more prenatal corticosteroids, particularly if the pregnancy is at risk of preterm birth. In some embodiments, the corticosteroid is a glucocorticoid. In some embodiments, the corticosteroid is betamethasone or progesterone, or mixtures thereof.

In some embodiments, vaccination as described in this example reduces the incidence of covd-19 disease (and/or recorded SARS-CoV-2 infection) or severe covd-19 disease in mothers and/or their born infants, e.g., relative to the incidence observed in a comparable unvaccinated (e.g., placebo-receiving) population. In some embodiments, a mother is considered to have a COVID-19 disease if at least 1 symptom of the COVID-19 disease is present (fever; new or increased cough; new or increased breathlessness, new or increased muscle pain; loss of taste or smell novelty, sore throat; diarrhea; vomiting; and/or, in certain embodiments, fatigue, headache, nasal congestion or discharge, nausea) and a SARS-CoV-2NAAT positive test is obtained in a central laboratory or local testing facility (using an acceptable test) during or before or after the symptom period. In some embodiments, a mother is considered to have severe COVID-19 disease if the mother has diagnosed COVID-19 and one or more of the following are present:

Clinical signs at rest suggest severe systemic disease (RR > 30 breaths/min, HR > 125 breaths/min, SpO 2< 93% in air in the sea level room, or PaO2/FiO2<300 mmHg);

respiratory failure (defined as the need for high flow of oxygen, non-invasive ventilation, mechanical ventilation or ECMO);

evidence of shock (SBP <90mmHg, DBP <60mmHg, or need for vasopressors);

-overt acute renal, hepatic or neurological dysfunction;

-entering an ICU;

-death.

In some embodiments, an infant is considered to have COVID-19 disease if there is at least one symptom (fever; new or increased cough; new or increased breathlessness; diarrhea; vomiting; and/or in certain embodiments, one or more of nasal congestion or discharge, poor appetite or poor feeding, abdominal pain/colic) and a positive test result for SARS-COV-2NAAT is obtained at a central laboratory or local testing facility (using an acceptable test) during or 4 days before or after the symptomatic phase. In some embodiments, an infant is considered to have severe covi-19 disease if the infant has been diagnosed with covi-19 and one or more of the following are present:

(i) Clinical signs at rest suggest severe systemic disease:

-RR (breaths/min): greater than 50 from birth to 1 week, greater than or equal to 40 from 1 week to 1 month, greater than or equal to 34 from 1 month to 6 months;

HR (times/min): > 180;

SpO2 ≦ 92% or > 50% FiO2 in indoor air to maintain ≥ 92%, or PaO2/FiO2<300mmHg 24;

(ii) respiratory failure (defined as a need for high flow of oxygen, including nasal CPaP/BiPaP, non-invasive ventilation, mechanical ventilation, or ECMO);

(iii) there is evidence of shock or heart failure:

sbp (mm hg) (< 5 th percentile of age);

+ from birth to <65 for 1 week of age, from 1 week to 75 for 1 month of age, from 1 month to 6 months of age < 100;

or

-the need to use vasoactive drugs to maintain BP in the normal range;

(iv) evident acute renal failure: serum creatinine > 2-fold increase in ULN at age or baseline creatinine 2-fold;

(v) overt GI/liver failure: total bilirubin >4mg/dL or ALT is a 2 times aged ULN;

(vi) apparent neurological dysfunction: glasgow Coma Scale score <11, or acute change in mental status, Glasgow Coma Scale score was decreased by > 3 points from abnormal baseline.

(vii) Entering an ICU;

(viii) and death.

In some embodiments, the incidence of multisystemic inflammatory syndrome is not significantly increased in infants that are vaccinated by the mother as described herein (e.g., relative to a comparable population that is not vaccinated by the mother and/or not vaccinated according to the protocol described herein). In some embodiments, the infant is considered to have multisystemic inflammatory syndrome when:

-fever in infants (38.0 ℃ C. for 24 hours or more, or subjective fever reported for 24 hours or more); and

laboratory evidence of inflammation (based on local laboratory scale), including but not limited to one or more of the following: CRP, ESR, fibrinogen, procalcitonin, D-dimer, ferritin, LDH or IL-6 are elevated, neutrophils are elevated, lymphocytes are reduced, albumin is low; and

evidence suggests that clinically severe disease requires hospitalization (as defined above for severe disease), and that multiple systems (. gtoreq.2) are involved:

the heart (e.g. shock, elevated troponin, elevated BNP, echocardiographic abnormalities, arrhythmia);

omicron (e.g., acute kidney injury or renal failure);

the respiratory system (e.g. pneumonia, ARDS, pulmonary embolism);

haematology (e.g. elevated D-dimer, thrombophilia (thrombophilia) or thrombocytopenia);

omicron GI/liver (e.g. elevated bilirubin, elevated liver enzymes, or diarrhea);

omicron skin diseases (e.g. rashes, mucocutaneous lesions);

omicron (e.g. CVA, aseptic meningitis, encephalopathy); and

-no alternative trusted diagnosis; and

-the infant is determined to be positive for the current or recent infection with SARS-CoV-2 by RT-PCR, serology or antigen testing; or

Infants had COVID-19 exposure within 4 weeks before symptoms appeared.

In some embodiments, maternal vaccination as described herein does not substantially increase the incidence of preterm birth in infants.

In some embodiments, the incidence of COVID-19 disease (and/or recorded SARS-CoV-2 infection) in a mother's vaccinated infant as described herein is reduced relative to the incidence of a mother's unsupervised infant. In some embodiments, the incidence of COVID-19 disease (and/or recorded SARS-CoV-2 infection) in a mother vaccinated infant as described herein is comparable to the incidence in a post-partum directly vaccinated infant.

In some embodiments, vaccination as described herein achieves one or more of the following primary or secondary outcome measures.

Measurement of main results:

1. reporting percentage of maternal participants in local reactions

Electronic diary self-documented injection site pain, redness and swelling

[ time frame: 7 days after dose 1 and dose 2 ]

2. Percentage of maternal participants reporting systemic events

Fever, fatigue, headache, chills, vomiting, diarrhea, new or increased muscle pain and new or increased joint pain self-documented by electronic diary

[ time frame: 7 days after dose 1 and dose 2 ]

3. Percentage of maternal participants reporting adverse events

Elicited by the staff of the investigation point

[ time frame: from dose 1 to 1 month after dose 2 ]

4. Percentage of maternal participants reporting severe adverse events

Elicited by the staff of the investigation point

[ time frame: from dose 1 to 6 months postpartum

5. Demonstrates a comparable immune response in pregnant women compared to the non-pregnant female participants of the C4591001 study without evidence of prior SARS-CoV-2 infection.

GMR, estimated by the ratio of geometric mean of SARS CoV 2 neutralization titers of pregnant versus non-pregnant female participants.

[ time frame: 1 month after dose 2 ]

6. Demonstrates a comparable immune response in pregnant women compared to the non-pregnant female participants of the C4591001 study with or without evidence of previous SARS-CoV-2 infection.

GMR, estimated by the ratio of geometric mean of SARS CoV 2 neutralization titers of pregnant versus non-pregnant female participants.

[ time frame: 1 month after dose 2 ]

Secondary outcome measurement:

7. evaluation of efficacy of COVID 19 diagnosed in participants against non-evidence of infection prior to vaccination

Follow-up visit for 1000 people.

[ time frame: 7 days after dose 2 ]

8. Efficacy was assessed against codv 19 diagnosed in participants with no evidence of prior infection.

Follow-up visit of 1000 people

[ time frame: 7 days after dose 2 ]

Example 35: human serum elicited by BNT162b2 vaccine neutralized SARS-CoV-2 lineage B.1.1.298 (Danish strain; also known as SARS-CoV-2/hu/DK/CL-5/1 (cluster 5)) and B.1.351 (south African strain; also known as 20H/501Y.V2(501.V2)) pseudoviruses

Sera from 12 young adult participants in the previously reported German 1/2 phase trial (drawn 7 or 21 days after booster immunization with 30 μ g BNT162b 2) were tested by a 50% neutralization assay (pVNT50) for neutralization of SARS-CoV-2 Wuhan Hu-1 (ref), south African lineage B.1.351(SA strain) and Danish mink (Danish mink) associated lineage B.1.1.298(DNK strain) spike protein pseudotype VSV. Compared to wuhan reference, the SA strain spike protein carries the following amino acid changes: L18F, D80A, D215G, delta L242-244, R246I, K417N, E484K, N501Y, D614G and A701V. Compared to the wuhan reference, the DNK strain spike protein carries the following amino acid changes: Y453F, D614G, I692V, M1229I.

The neutralization of DNK strain pseudovirus by BNT162b2 immune serum was almost as effective as SARS-CoV-2 Wuhan Hu-1 pseudotype reference. The measured neutralizing titer against SARS-CoV-2 lineage B.1.351 pseudovirus was reduced (5-fold) compared to the titer against the Wuhan Hu-1 pseudotype reference. Importantly, all BNT162b2 immune sera tested were still able to neutralize, with no complete escape found (fig. 127).

Materials and methods:

recombinant replication-defective VSV vectors were ligated with: Wuhan-Hu-1 isolate SARS-CoV-2 spike (S) (GenBank: QHD 43416.1); a variant carrying 4 mutations found in the S protein of the danish mink related lineage b.1.1.298 (Y453F, D614G, I692V, M1229I); or a variant carrying 10 mutations (L18F, D80A, D215G, R246I, Δ 242/243/244, K417N, E484K, N501Y, D614G, A701V) pseudotyped packages found in the S protein of south African lineage B.1.351, which vectors encode Green Fluorescent Protein (GFP) and luciferase (Luc) instead of VSV-glycoprotein (VSV-G). Briefly, HEK293T/17 monolayers transfected to express individual SARS-CoV-2S (SARS-CoV-2-S (C.DELTA.19)) truncated by the C-terminal cytosolic 19 amino acids were inoculated with VSV.DELTA.G-GFP/Luc vector. After incubation for 1 hour at 37 ℃, the inoculum was removed, the cells were washed with PBS, and then medium supplemented with anti-VSV-G antibody (clone 8G5F11, Kerafast) was added to neutralize residual input virus. The culture medium containing VSV-SARS-CoV-2 pseudovirus was collected 20 hours after inoculation, filtered at 0.2- μm and stored at-80 ℃.

For the pseudovirus neutralization assay, 40,000 Vero 76 cells were seeded per 96 wells. The serum was diluted 1: 10 in series 1: 2 in the medium (dilution range 1: 10-1: 2, 560). VSV-SARS-CoV-2-S pseudoparticles were diluted in culture medium and used for a fluorescence focus unit (ffu) count of approximately 1,000TU in the assay. Serum dilutions were mixed with pseudoviruses at 1:1 for 30 min at room temperature, then added to Vero 76 cell monolayers in 96-well plates and incubated for 24 h at 37 ℃. The supernatant was removed and the cells were lysed with luciferase reagent (Promega). Fluorescence was recorded and neutralization titers were calculated by 4-parameter logistic (4PL) fitting that generated percent neutralization at each serial serum dilution in GraphPad Prism version 9. The 50% pseudovirus neutralization titer (pVNT50) was reported as the reciprocal interpolated value of the dilution that produced 50% fluorescence reduction.

Example 36: serum-neutralizing N501Y mutant SARS-CoV-2 elicited by BNT162b2 vaccine

A rapidly spreading SARS-CoV-2 variant (Volz E.et al. report 42-Transmission of SARS-CoV-2 Lineare B.1.1.7in England: insight from linking epigenetic and genetic data. https:// www.imperial.ac.uk/mrc-global-infection-disease-analysis/covid-19/report-42-SARS-CoV-2-variant/; Tegay H.et al. Emergeneand Rapid spread of a new segment acid metabolism syndrome-related codon 2(SARS-CoV-2) linkage with multiple functions in South Africa. rv. 19/36) appeared in the United kingdom and South Africa. These variants have multiple mutations in their S glycoprotein, which is a key target for virus neutralizing antibodies. These rapidly propagating variants share spike N501Y substitutions. This mutation is of particular interest because it is located in the binding site of the viral receptor for entry into the cell, increases binding to the receptor (angiotensin converting enzyme 2), and allows the virus to expand its host range to infect mice (Gu H.et al.Adaptation of SARS-CoV-2in BALB/c microorganism for stimulating vaccine effect. science 2020; 369: 1603-7; Chan K.K.et al.Aningirected receptor for SARS-CoV-2 branched binding proteins S sequence variants. Cold Spring Harbor Laboratory 2020.doi: 10.1101/2020.10.18.344622).

We generated isogenic Y501 SARS-CoV-2 on the genetic background of N501 clinical strain USA-WA1/2020, which also provided the genetic background of the spike antigen encoded by BNT162b 2. Sera from 20 participants in the previously reported trial (drawn 2 or 4 weeks after immunization with Two 30 μ g doses of BNT162b2 spaced 3 weeks apart) (Walsh E.E.et al.safety and Immunogenicity of Two RNA-Based Covid-19Vaccine Candidates. N Engl J Med 2020; Polack F.P.et al.safety and efficacy of the BNT162b2 mRNA Covid-19Vaccine. N Eng. J. Med. 2020: DOI: 10.1056/NEJMoa2034577) were tested by a 50% plaque reduction neutralization assay for neutralization of N501 and Y501 viruses (PRNT) and50(ii) a Fig. 128). The ratio of 50% neutralized GMT of serum against Y501 virus to 50% neutralized GMT against N501 virus was 1.46, indicating no reduction in neutralizing activity against viruses with Y501 spikes.

Materials and methods

Construction of isogenic (isogenic) viruses

We prepared an isogenic pair of SARS-CoV-2 containing either the N501 or Y501 spike proteins (FIG. 129).The N501Y mutation WAs made by making an A to T substitution at nucleotide 23,063 of the viral genome using an Infectious cDNA Clone of clinical strain WA1(2019-nCoV/USA _ WA1/2020) (Xie X.et al. an Infectious cDNA Clone of SARS-CoV-2.Cell Host Microbe 2020; 27: 841-8e 3). According to the mutagenesis method previously reported (plant J.A.et al.spike mutation D614G alters SARS-CoV-2 fisness.Nature 2020), we recovered the N501 and Y501 viruses at titers of >107Plaque Forming Units (PFU)/ml. Both viruses developed similar plaque morphologies on Vero E6 cells (figure 130).

Serum samples and neutralization assays

Figure 131 schematically illustrates an immunization and serum collection protocol. For measurement of neutralization titers, each serum was serially diluted 2-fold in culture medium, the first dilution being at a degree of 1:40 (dilution range 1: 40-1: 1280). The diluted serum was incubated with 100 PFU of N501 or Y501 virus at 37 ℃ for 1 hour before plating the serum-virus mixture onto a Vero E6 cell monolayer in 6-well plates. Conventional (non-fluorescent) plaque reduction neutralization assays were performed to quantify serum-mediated viral inhibition as previously reported (Muruto A.E.et al.A. high-throughput neutral antibody assay for COVID-19 diagnostics and vaccine evaluation. Nat Commun 2020; 11: 4059). Suppression of>50% of the minimal serum dilution of viral plaques was defined as PRNT50. A table of neutralization titers is provided (table 28). FIG. 132 is a graph depicting PRNT of each serum against N501 and Y501 viruses50The ratio of (a) to (b).

TABLE 28.20 PRNT in BNT162b2 post-immune sera against N501 and Y501 SARS-CoV-250Value of

Example 37: serum neutralization of spike 69/70 deletion, E484K and N501Y SARS-CoV-2 by BNT162b2 vaccine

Rapid spread of SARS-CoV-2 variants (volatile E.et al CMe. report 42-Transmission of SARS-CoV-2line B.1.1.7in England: instruments from linking and genetic data. https: wWimpierialacuk/mrc-global-interaction-release-analysis/covid-19/report-42-SARS-CoV-2-variant/2021; volatile H.et al. expression and radial transformed of a new segment acid yield-related complex 2 (SARS-CoV-2) line/2020.12.21.20248640). These variants have multiple mutations in their spike glycoproteins, which are key targets for virus neutralizing antibodies. Emerging spike mutations raise concerns about the efficacy of vaccines against these new strains. The objective of this study was to investigate the effect of several key spike mutations of the UK and SA strains on the neutralization elicited by the BNT162b2 vaccine.

We designed three SARS-CoV-2 containing key spike mutations from the newly emerging british (UK) and South Africa (SA) variants: N501Y from UK and SA; 69/70 deletion + N501Y + D614G from UK; and E484K + N501Y + D614G from SA. The geometric neutralization mean titer (GMT) of twenty BTN162b2 vaccinated human sera against the three mutant viruses was 0.81-1.46 times the GMT against the parent virus, indicating that neutralization of sera elicited by two BNT162b2 doses was minimally affected by the mutation.

Using an Infectious cDNA Clone of SARS-CoV-2 (Xie X. et al. an infection cDNA Clone of SARS-CoV-2.Cell Host Microbe 2020; 27:841-8e 3), we designed three spike mutant viruses in the genetic background of clinical strain USA-WA1/2020 (FIG. 133). (i) Mutant N501Y virus contained the N501Y mutation shared by the UK and SA variants. The mutation is located in the Receptor Binding Domain (RBD) of the virus for entry into the Cell, increases binding to the receptor (angiotensin converting enzyme 2), and allows the virus to expand its Host range to infect mice (Xie X. et al. an infection cDNA Clone of SARS-CoV-2.Cell Host Microbe 2020; 27:841-8e 3; Wrapp D. et al. Cryo-EM structure of the 2019-nCoV spike in the fusion formation. Science 2020; 367: 1260-3). (ii) The mutant Δ 69/70+ N501Y + D614G virus contains two additional changes from the UK variant: amino acid 69 and 70 deletions (. DELTA. 69/70) and D614G substitutions. Amino acids 69 and 70 are located in the N-terminal domain of the spike S1 fragment, and deletion of these residues may alter the conformation of the spike (Alternally) (Wrapp D.et al Cryo-EM structure of the 2019-nCoV spike in the prefusion compatibility. Science 2020; 367: 1260-3). The D614G mutation is predominant in circulating strains throughout the world (plant JA et al. Spike mutation D614G variants SARS-CoV-2 fire. Nature 2020; Korber B.et al. tracking Changes in SARS-CoV-2 Spike: Evidence that D614G incomes infection of the CODV-19 Virus. cell 2020). (iii) Mutant E484K + N501Y + D614G virus contained an E484K substitution, which was also located in the virus RBD. The E484K substitution alone confers resistance to several monoclonal antibodies (Ku Z. et al. molecular determinants and mechanisms for antibody coding tail prediction SARS-CoV-2 expression. Nat Commun 2021; 12: 469; Baum A. et al. antibody coding tail to SARS-CoV-2spike protein prediction polypeptide mutated polypeptide. science 2020; 369: 1014-8). The three mutant viruses exhibited similar plaque morphology on Vero E6 cells compared to the wild type USA-WA1/2020 strain (FIG. 134).

We tested a panel of 20 participants in a previously reported clinical trial (drawn at 2 or 4 weeks post-immunization with Two 30 μ g doses of BNT162b2 spaced 3 weeks apart) (Walsh EE et al. safety and Immunogenicity of Two RNA-Based Covid-19 Vaccine peptides. N Engl J Med 2020; Polack FP et al. safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med 2020) (FIG. 135). Neutralization of the wild type USA-WA1/2020 strain and three mutant viruses (PRNT) WAs tested for each serum by a 50% plaque reduction neutralization assay50(ii) a Tables 29 and 30).

TABLE 29.20 PRNT in BNT162b2 post-immunization sera against wild type (USA-WA1/2020) and mutant N501Y SARS-CoV-250

TABLE 30.20 parts of BNT162b2 post-immunization sera against PRNT50 of wild type (USA-WA1/2020), Δ 69/70+ N501Y + D614G and E484K + N501Y + D614G SARS-CoV-2

All sera showed comparable neutralizing titers between wild-type and mutant viruses, with differences ≦ 4-fold (FIG. 136). Notably, 10 of 20 sera had twice their neutralizing titer against mutant Δ 69/70+ N501Y + D614G virus as against wild type virus (fig. 136b), while 6 of 20 sera had half their neutralizing titer against mutant E484K + N501Y + D614G virus as against wild type virus (fig. 136 c). The ratio of the neutralizing GMT of the sera against N501Y, Δ 69/70+ N501Y + D614G and E484K + N501Y + D614G viruses to its neutralizing GMT against USA-WA1/2020 viruses WAs 1.46, 1.41 and 0.81, respectively (fig. 137).

Consistent with recent other reports on neutralization of the neutralizing or convalescent phase of the variant SARS-CoV-2 or post-immunization sera to the corresponding pseudovirus (Wibmer CK et al. SARS-CoV-2501Y. V2 escapes neutralization by South African CODV-19 donor plasmid. bioRxiv 2021: doi: https:// doi.org/10.1101/2021.01.18.427166; Wang Z.et al. mRNA vaccine-mutated antibodies to SARS-CoV-2and circulating variants. bioRxiv 1: https:// doi.org/10.1101/2021.01.15.426911), serogroups had a lower neutralization to GMT of the virus with three mutations from the SA variant (E35484 + N501Y + D614 + D20226) than to GMT of the virus with three mutations from the N variant (E3528 + N501 + D20223 + 31) or had mutations from the virus with Δ 493 + K + Y (E.501 + N501 + D + 31). However, the degree of difference in neutralizing GMT for any virus in this study was small compared to the 4-fold difference in hemagglutination inhibition titers that is potentially required to identify strain changes in influenza vaccines (Smith DJ et al, Mapping the antigenic and genetic evaluation of influenza virus 2004; 305: 371-6).

Method

And (5) constructing isogenic viruses. Three recombinant SARS-CoV-2 mutants (N501Y, Delta 69/70+ N501Y + D614G, E484K + N501Y + D614G in spike protein) were prepared in the genetic background of an Infectious cDNA Clone derived from clinical strain WA1 (2019-nCoV/USA _ WA1/2020) by a previously reported PCR-based mutagenesis method (plant JA et al. spike mutagenesis D614G clones of SARS-CoV-2.Cell Host Microbe 2020; 27: 841-8E 3). The full-length infectious cDNA is ligated in vitro and used as a template to transcribe the full-length viral RNA. After electroporation of the in vitro RNA transcripts, mutant viruses were recovered from Vero E6 cells on day 2 (P0). The P1 virus was harvested as stock (stock) by passaging the P0 virus once on Vero E6 cells. Titers of P1 virus were determined by plaque assay on Vero E6. The genomic sequence of the P1 virus was verified by Sanger sequencing. Detailed procedures were as recently reported (Xie X.et al. engineering SARS-CoV-2using a reverse genetic system. Nature Protocols 2021: https:// doi.org/10.1038/s 41596-021-.

Serum samples and neutralization assays. Figure 135 schematically illustrates an immunization and serum collection protocol. Conventional (non-fluorescent) plaque reduction neutralization assays were performed to quantify serum-mediated viral inhibition as previously reported (Muruato AE et al. A high-throughput neutral anti-viral assay for COVID-19diagnosis and vaccine evaluation. Nat Commun 2020; 11: 4059). Briefly, each serum was serially diluted 2-fold in culture medium, the first dilution being 1:40 (dilution range 1:40-1: 1280). Diluted sera were incubated with 100 plaque forming units of wild-type or mutant virus at 37 ℃ for 1 hour (h) before seeding the serum-virus mixture onto Vero E6 cell monolayers in 6-well plates. After 1 hour of infection at 37 ℃, 2ml of 2% seaopaque agar (Lonza) in Dulbecco' S modified Eagle medium (DMEM) containing 2% Fetal Bovine Serum (FBS) and 1% penicillin/streptomycin (P/S) was added to the cells. After 2 days of incubation, 2ml of 2% seaopaque agar (Lonza) in DMEM containing 2% FBS, 1% P/S and 0.01% neutral red (Sigma) was added to the top of the first layer. Further incubation at 37 ℃ for 16 hAfter time, plaque numbers were counted. The minimum serum dilution that inhibited 50% plaque count was defined as the 50% Plaque Reduction Neutralization Titer (PRNT) 50). Each serum was tested in duplicate. PRNT were performed in a BioSafty level 3 laboratory of the University of Texas Medical Branch50And (4) measuring.

Example 38: BNT162b 2-elicited neutralization Activity of serum

New highly contagious SARS-CoV-2 variants with mutations in the S gene, first tested in the United kingdom (B.1.1.7 lineage), south Africa (B.1.351 lineage) and Brazil (P.1 lineage), are spreading worldwide. To analyze the effect of neutralization elicited by BNT162b2, we designed S mutations from each of the three new lineages into USA-WA1/2020, an earlier isolate of the virus at 1 month 2020 (FIG. 138). We subsequently prepared 5 recombinant viruses. The first had all mutations found in the S gene in the B.1.1.7 lineage (B.1.1.7-spikes), the second had all mutations found in the S gene in the P.1 lineage (P.1-spikes), the third had all mutations found in the S gene in the B.1.351 lineage (B.1.351-spikes), the fourth had N-terminal domain deletions found in the B.1.351 lineage and a worldwide dominant D614G substitution (B.1.351-. DELTA.242-244 + D614G), and the fifth had three mutations from the B.1.351 lineage (K36417, E484K and N501Y) at the receptor binding site and a D614G substitution (B.1.351-RBD + D59614). The amino acid residues mutated in the b.1.351-RBD + D614G virus are also among those mutated in the p.1 lineage virus, but in the p.1 lineage virus, K417 is mutated to threonine instead of asparagine. All mutant viruses produced infectious virus titers in excess of 10 7Plaque forming units/ml. B.1.1.7-spike and B.1.351-spike viruses formed plaques smaller than other viruses (FIG. 139).

2 or 4 weeks after administration of a second dose of 30 μ g BNT162b2 (3 weeks after the first immunization), we performed a 50% Plaque Reduction Neutralization Test (PRNT) using 20 serum samples obtained from 15 participants in a critical test50)(Polack FP et al.Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine.N Engl J Med 2020;383:2603-15;Walsh EE et al.Safety and immunological of two RNA-based Covid-19vaccine candidates N Engl J Med 2020; 383:2439-50) (FIG. 140). All serum samples neutralized USA-WA1/2020 effectively and most of them had titers above 1: 40. The geometric mean neutralization titers against USA-WA1/2020, B.1.1.7-spike, P.1-spike, B.1.351- Δ 242-244+ D614G and B.1.351-RBD + D614G viruses were 532, 663, 437, 194, 485 and 331, respectively (FIG. 141 and Table 31). Thus, neutralization of B.1.1.7-spike and P.1-spike viruses WAs substantially equivalent, while neutralization of B.1.351-spike viruses WAs still robust, but 2.7-fold lower, compared to neutralization of USA-WA 1/2020. Our data are consistent with neutralizing titers against viruses with the full set of B.1.351-spike mutations lower than those with any subset of mutations and indicate that mutations in the receptor binding sites (K417N, E484K, and N501Y) have a greater effect on neutralization than the 242-244 deletion in the N-terminal domain of the spike protein.

Since the neutralization of b.1.1.7-spike and p.1-spike viruses by BNT162b2 primed sera WAs substantially equal to that of USA-WA 1/2020, the neutralization data strongly supports that BNT162b2 will continue to provide protection against the first detected variant in UK or brazil. Protection against B.1.351 lineage viruses is also expected, given that, when strong potency has been observed in the critical C4591001 potency assay (Polack FP et al. safety and efficacy of the BNT162b2 mRNA Covid-19vaccine. N Engl J Med 2020; 383: 2603-15; Walsh et al. safety and immunogenicity of two RNA-based Covid-19vaccine details. N Engl J Med 2020; 383: 2439-50; Sahin U et al. BNT162b2 antigens SARS-CoV-2-neutral antibodies and T cells of human sum. December 11,2020 (httwhtw. predxiv-org-10. clamp-10. 12. pre-10. 12. J. Med. J. Med. 2020. 22-50; Sahin U.B.N.T. BNB.2-2-neutral antibodies and T cells. 20. 12. December. 3683. 9. pre. 3-3. 1. 3. 50; Sahin. Tk. Tp. 10. 1. 1.10. 1. T10. 1. T. 1. T. 1. T10. T. 1. T. N. 1. N. 1. N. 1. N. 1. N. 1. N. 2. N. p, although the neutralizing titer was lower for this variant, it was still robust and well above the barely detected titer observed after one dose of BNT162b 2. In addition, T cell immunity (Liao M et al. Single-cell landscapes of branched immunological cells in tissues with COVID-19.Nature Medicine 2020) can also be involved in protection, and BNT162b2 immunity elicits a CD8+ T cell response that recognizes multiple variants (Skelly DT et al. vaccine-induced immunological pathologies more than nuclear robust nutritional immunity to infectious SARS-CoV-2variants of cancer. research Square 2021).

Materials and methods

And (5) constructing isogenic viruses. All recombinant SARS-CoV-2 with spike mutations were prepared in the genetic background of Infectious cNDA clones derived from clinical strain USA-WA1/2020 (Xie X et al. an infection cDNA Clone of SARS-CoV-2.Cell Host Microbe 2020; 27:841-8e3) (FIG. 138). Mutations were introduced into the spike gene using previously reported PCR-based mutagenesis methods (plant JA et al. spike mutation D614G alternates SARS-CoV-2 fitness. Nature 2020.doi:10.1038/s 41586-020-2895-3; Xie X et al. mutagenesis of SARS-CoV-2 spike 69/70deletion, E484K and N501Y variants by BNT162b2 vaccine-associated sera. nat. Med 2021.doi:10.1038/s 41591-021-01270-4). The full-length infectious cDNA is ligated and used as a template to transcribe full-length viral RNA in vitro. Following electroporation of in vitro transcribed RNA, the original virus stock was recovered from Vero E6 cells on day 2 (P0). P0 virus was propagated on Vero E6 cells for another round to produce P1 virus for neutralization assays. The titer of P1 virus was measured by plaque assay on Vero E6 cells (fig. 139). The complete spike sequence of the P1 virus WAs determined by Sanger sequencing, with only the expected nucleotide changes to the sequence from USA-WA 1/2020. A detailed method of the above experiment has recently been reported (Xie X et al. engineering SARS-CoV-2 using a reverse genetic system. Nature Protocols 2021: https:// doi. org/10.1038/s 41596-021-.

Serum samples and neutralization assays. Figure 140 shows immunization and serum collection protocols. A conventional 50% Plaque Reduction Neutralization Test (PRNT) was performed as previously reported (Muruato AE et al. A high-throughput neutral anti-plaque assay for COVID-19 diagnosis and vaccine evaluation. Nat Commun 2020; 11:4059)50) To quantify serum-mediated viral inhibition. Briefly, each serum was serially diluted 2-fold in culture medium at an initial dilution of 1:40 (dilution range 1:40-1: 1280). The diluted serum WAs mixed with 100PFU of USA-WA1/2020 or Takara ShuzoVariant SARS-CoV-2 incubation. After incubation for 1 hour at 37 ℃, the serum-virus mixture was seeded on a monolayer of Vero E6 cells pre-seeded on a 6-well plate the day before. Suppression of>A50% minimum serum dilution of viral plaques was defined as PRNT50. The neutralization titers are shown in table 31.

TABLE 31.20 parts BNT162b2 post-immunization sera against PRNT in USA-WA1/2020 and mutant SARS-CoV-250

Data from USA-WA1/2020 were from 3 experiments; data for the B.1.1.7-spike, B.1.351-. DELTA.242-244 + D614G, and B.1.351-RBD-D614G viruses were each from one experiment; data for P.1-spike and B.1.351-spike viruses were from 2 experiments. For each independent experiment, the respective PRNT 50Values are geometric mean of duplicate plaque assays; no difference was observed between duplicate assays.

Geometric mean neutralization titer.

#95% confidence interval (95% CI) for GMT.

Example 39: persistence of BNT162b 2-induced CD4+ and CD8+ T cell responses

Samples collected on day 85 and day 184 (9 and 23 weeks post boost, respectively) were analyzed in a subset of 24 subjects spanning dose levels of 10-30 μ g to determine the persistence of T cell responses induced by BNT162b 2. CD4 in most individuals on day 184 and after initial contraction+And CD8+T cell responses were detectable across the 3 dose levels tested. CD4 observed in 4 older adult subjects vaccinated with 10 μ g BNT162b2+And CD8+The kinetics of the response were comparable to younger adult subjects and in all 4 subjects 23 weeks after booster vaccination, S protein specific CD4+T cellsIs still detectable. BNT162b 2-induced CD4+And CD8+The response was higher than or within the range of recall antigen memory responses (fig. 142).

Example 40: BNT162b 2-induced MHC-I binding epitope recognized by CD 8T cells

Using MHC class I multimer technology, several epitopes distributed across the entire length of the S protein and presented by a combination of common HLA-a and HLA-B alleles were identified for CD8+ T cell recognition induced by BNT162-B2 (measured 7 days post booster vaccination). Some peptide/HLA combinations were found in more than one subject.

TABLE 32 MHC-I binding epitopes recognized by BNT162b 2-induced CD 8T cells

Example 41: histological findings after administration of BNT162b2

Classical chromogenic Immunohistochemistry (IHC) and chromogenic double IHC-ISH (in situ hybridization experiments) were performed to study the biodistribution of BNT162b2 in mouse tissues 6 hours and 6 days after injection.

Method

After harvesting, the tissues were fixed at 4% RotiHistofix at 4 ℃ overnight and dehydrated in a Leica Tissue dehydrator (Tissue Processor) and embedded in paraffin. Color development IHC was performed. The Spike protein was detected with an anti-Spike 2 mouse monoclonal antibody (Genetex). The dual IHC-ISH assay was performed according to a self-established method, based on the document MK 51-149, Advanced Cell Diagnostics, using company kits and reagents. The BNT162b2 probe of ISH (modV9) was customized by Advanced Cell Diagnostics based on the sequences provided by TRON. IHC methods for immune Cell markers CD11c (Cell Signaling), CD19(Cell Signaling), CD169(Thermo Fisher) and F4/80(Cell Signaling) are at TRON and are adapted to the dual IHC-ISH assay of the project. Images were obtained using a Vectra Polaris Multispectral Slide Scanner microscope (Akoya Bioscience) and analyzed with PhenoChart software (Akoya Bioscience).

Results

As can be seen in fig. 143, in the dual IHC-ISH assay, a specific vaccine mRNA signal (red) was detected in the lymph nodes 6 hours after injection using the modV9 probe. The vaccine was mainly localized to subcapsular sinus (LN in positions 9 and 5) and B cell follicles (LN in positions 12 and 1). Dendritic cells were visualized by CD11c staining (blue-green, upper panel) and only some of them had absorbed vaccine. Most of the CD169+ macrophages (tunica basalis sinus macrophages, blue-green, middle panel) were positive for the vaccine. B cells (CD19+, blue-green, lower panel) were the second major population exhibiting vaccine signaling.

In the dual IHC-ISH assay, specific vaccine mRNA signals were still detectable in LN, albeit in very small amounts, using the modV9 probe 6 days after injection (data not shown). Some CD11c + DCs and subcapsular sinus macrophages were positive for the vaccine. Most of the vaccine signals detected were in B cells (CD19 +).

As can be seen in figure 144, in the dual IHC-ISH assay, a specific vaccine mRNA signal (red) was detected in the spleen 6 hours post injection using the modV9 probe. The major vaccine signal was detected in the white marrow. Dendritic cells were visualized by CD11c staining (blue-green, upper panel) and only some of them absorbed vaccine. A small fraction of F4/80+ macrophages (blue-green, middle panel) absorbed the vaccine. B cells (CD19+, blue-green, lower panel) are the predominant population exhibiting vaccine signaling.

In the dual IHC-ISH assay, specific vaccine mRNA signals were still detectable in the spleen, albeit in very small amounts, using the modV9 probe 6 days post injection (data not shown). The vaccine signal detected was only in B cells (CD19 +). None of the DCs and macrophages showed vaccine signals 6 days after injection.

After 6 days, we detected signals in the muscle, particularly in some muscle fibers and connective tissue fascial membrane, using the mouse anti-S2 mouse monoclonal antibody. In LN, we detected cells expressing spike proteins in the T cell region.

After 6 days, no spike expression was detected in muscle using the mouse anti-S2 mouse monoclonal antibody. In contrast, LN was filled with cells expressing the vaccine (data not shown).

In the chromogenic IHC experiment, no non-specific staining was detected with the S2 mouse monoclonal antibody.

Summary of the invention

Very strong vaccine signals were observed in draining LN and spleen 6 hours after injection. In LN, the vaccine is mainly detected in B-cell follicles and subcapsular sinus, and some signals are also located in the T-cell zone. By means of the double IHC-ISH we show that B cells (CD19+) and subcapsular sinus macrophages (CD169+) are indeed the main cells for vaccine uptake.

Dendritic cells (CD11c +) and the central sinuses (intermedia sinuses) in the T cell region also take up the vaccine. After 6 days, some vaccine mRNA was still observed in draining LN. The signal observed in the T cell region after 6 days was in dendritic cells (CD11c +). Some B cells and LN macrophages still have some vaccine at this stage.

Analysis of spleens collected 6 hours after injection showed that the vaccine had reached the spleen within 6 hours, primarily probably through blood circulation. The signal is located in the white marrow where B cells and T cells form the main population and antigen presentation to T cells occurs in the white marrow. By a dual IHC-ISH assay, we show that most B cells take up the vaccine. Many DCs surrounding B cells (CD11c +) were also positive. After 6 days, the signal was restricted to B cells.

The IHC method was set up to detect spike protein expression on cell pellets treated/not with vaccine using an anti-spike S2 mouse monoclonal antibody. Specific signals were detected only in cells treated with BNT162b 2. At the beginning of the testNo non-specific staining was observed in the tissues. In muscle, 6 hours after injection, spike expression was detected in muscle fibers and connective tissue fascial membrane. After 6 days, no staining was detected in the muscle. In contrast, 6 days after injection, a large spike surface was observed in LN Particularly in the T cell region.

Example 42: stability study

The stability evaluation of BNT162b2 formulations was performed at various concentrations (e.g., 0.5mg/mL, 1mg/mL, and 2mg/mL) and includes the evaluation of compositions stored at various temperatures (e.g., -70 ℃ (e.g., -70 ± 10 ℃), -20 ℃ (e.g., -20 ± 5 ℃), +5 ℃ (e.g., 5 ± 3 ℃), or +25 ℃ (e.g., 25 ± 2 ℃)) and/or for various time periods (e.g., 0.5 month, 1 month, 2 months, 3 months, 4 months, and, in some cases, one or more intermediate time points (e.g., 1.5 months, 2.5 months, etc.).

In an exemplary study, mice (one leg) were injected with 20 μ L of the relevant formulation on day 0. Blood was collected and serum was produced after 14, 21 and 28 days post-dose; spleens were isolated on day 28.

ELISA was performed to detect the presence of antibodies in serum that bind to S1 protein or specifically bind to RBD domain. Figure 145 shows exemplary S1 ELISA results obtained from 28-day sera of mice injected with a specified formulation that has been stored under specified temperature conditions for a specified period of time. Referring to fig. 145, it can be seen that all stored samples performed well and were reasonably comparable after 1 month of storage. Some reduction in activity was observed for samples stored at +25 ℃ after 2 or 3 months of storage, but significant performance was maintained for samples stored at-70 ℃, -20 ℃, +5 ℃.

At some point in time, one or more parameters such as appearance, RNA content, RNA integrity, RNA encapsulation, lipid content (whole and/or individual components and/or ratios thereof), particle size, particle polydispersity index, in vitro expressibility, etc. are evaluated; additional or alternative parameters may or may have been evaluated.

Exemplary observations include that a period of time of about 2 weeks or more at +25 ℃ is not recommended, and preferably no more than about 1 week; in addition, a significant decrease in RNA integrity was observed. It was also observed that, in at least some cases, a significant ability to induce antibodies was maintained, even in cases where the in vitro expressibility was significantly reduced. It was observed that the change in polydispersity index of formulations stored at +5 ℃ or above was greater than formulations stored at lower temperatures, especially after about 3 months or 4 months of storage.

Sequence listing

<110> biotechnological RNA pharmaceuticals Ltd

<120> coronavirus vaccine

<130> I21DE176487C

<150> PCT/EP2020/061239

<151> 2020-04-22

<150> PCT/EP2020/066968

<151> 2020-06-18

<150> PCT/EP2020/068174

<151> 2020-06-26

<150> PCT/EP2020/069805

<151> 2020-07-13

<150> PCT/EP2020/071733

<151> 2020-07-31

<150> PCT/EP2020/071839

<151> 2020-08-03

<150> PCT/EP2020/073668

<151> 2020-08-24

<150> PCT/EP2020/081544

<151> 2020-11-09

<150> PCT/EP2020/081981

<151> 2020-11-12

<150> PCT/EP2020/082601

<151> 2020-11-18

<150> PCT/EP2020/082989

<151> 2020-11-20

<150> PCT/EP2020/083435

<151> 2020-11-25

<150> PCT/EP2020/084342

<151> 2020-12-02

<150> PCT/EP2020/085145

<151> 2020-12-08

<150> PCT/EP2020/085653

<151> 2020-12-10

<150> PCT/EP2020/087844

<151> 2020-12-23

<150> PCT/EP2021/050027

<151> 2021-01-04

<150> PCT/EP2021/050874

<151> 2021-01-15

<150> PCT/EP2021/050875

<151> 2021-01-15

<150> PCT/EP2021/051772

<151> 2021-01-26

<150> PCT/EP2021/052572

<151> 2021-02-03

<150> PCT/EP2021/052716

<151> 2021-02-04

<150> PCT/EP2021/054622

<151> 2021-02-24

<160> 57

<170> PatentIn version 3.5

<210> 1

<211> 1273

<212> PRT

<213> Artificial sequence

<220>

<223> S protein

<400> 1

Met Phe Val Phe Leu Val Leu Leu Pro Leu Val Ser Ser Gln Cys Val

1 5 10 15

Asn Leu Thr Thr Arg Thr Gln Leu Pro Pro Ala Tyr Thr Asn Ser Phe

20 25 30

Thr Arg Gly Val Tyr Tyr Pro Asp Lys Val Phe Arg Ser Ser Val Leu

35 40 45

His Ser Thr Gln Asp Leu Phe Leu Pro Phe Phe Ser Asn Val Thr Trp

50 55 60

Phe His Ala Ile His Val Ser Gly Thr Asn Gly Thr Lys Arg Phe Asp

65 70 75 80

Asn Pro Val Leu Pro Phe Asn Asp Gly Val Tyr Phe Ala Ser Thr Glu

85 90 95

Lys Ser Asn Ile Ile Arg Gly Trp Ile Phe Gly Thr Thr Leu Asp Ser

100 105 110

Lys Thr Gln Ser Leu Leu Ile Val Asn Asn Ala Thr Asn Val Val Ile

115 120 125

Lys Val Cys Glu Phe Gln Phe Cys Asn Asp Pro Phe Leu Gly Val Tyr

130 135 140

Tyr His Lys Asn Asn Lys Ser Trp Met Glu Ser Glu Phe Arg Val Tyr

145 150 155 160

Ser Ser Ala Asn Asn Cys Thr Phe Glu Tyr Val Ser Gln Pro Phe Leu

165 170 175

Met Asp Leu Glu Gly Lys Gln Gly Asn Phe Lys Asn Leu Arg Glu Phe

180 185 190

Val Phe Lys Asn Ile Asp Gly Tyr Phe Lys Ile Tyr Ser Lys His Thr

195 200 205

Pro Ile Asn Leu Val Arg Asp Leu Pro Gln Gly Phe Ser Ala Leu Glu

210 215 220

Pro Leu Val Asp Leu Pro Ile Gly Ile Asn Ile Thr Arg Phe Gln Thr

225 230 235 240

Leu Leu Ala Leu His Arg Ser Tyr Leu Thr Pro Gly Asp Ser Ser Ser

245 250 255

Gly Trp Thr Ala Gly Ala Ala Ala Tyr Tyr Val Gly Tyr Leu Gln Pro

260 265 270

Arg Thr Phe Leu Leu Lys Tyr Asn Glu Asn Gly Thr Ile Thr Asp Ala

275 280 285

Val Asp Cys Ala Leu Asp Pro Leu Ser Glu Thr Lys Cys Thr Leu Lys

290 295 300

Ser Phe Thr Val Glu Lys Gly Ile Tyr Gln Thr Ser Asn Phe Arg Val

305 310 315 320

Gln Pro Thr Glu Ser Ile Val Arg Phe Pro Asn Ile Thr Asn Leu Cys

325 330 335

Pro Phe Gly Glu Val Phe Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala

340 345 350

Trp Asn Arg Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser Val Leu

355 360 365

Tyr Asn Ser Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro

370 375 380

Thr Lys Leu Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe

385 390 395 400

Val Ile Arg Gly Asp Glu Val Arg Gln Ile Ala Pro Gly Gln Thr Gly

405 410 415

Lys Ile Ala Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys

420 425 430

Val Ile Ala Trp Asn Ser Asn Asn Leu Asp Ser Lys Val Gly Gly Asn

435 440 445

Tyr Asn Tyr Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe

450 455 460

Glu Arg Asp Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys

465 470 475 480

Asn Gly Val Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly

485 490 495

Phe Gln Pro Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val Val Val

500 505 510

Leu Ser Phe Glu Leu Leu His Ala Pro Ala Thr Val Cys Gly Pro Lys

515 520 525

Lys Ser Thr Asn Leu Val Lys Asn Lys Cys Val Asn Phe Asn Phe Asn

530 535 540

Gly Leu Thr Gly Thr Gly Val Leu Thr Glu Ser Asn Lys Lys Phe Leu

545 550 555 560

Pro Phe Gln Gln Phe Gly Arg Asp Ile Ala Asp Thr Thr Asp Ala Val

565 570 575

Arg Asp Pro Gln Thr Leu Glu Ile Leu Asp Ile Thr Pro Cys Ser Phe

580 585 590

Gly Gly Val Ser Val Ile Thr Pro Gly Thr Asn Thr Ser Asn Gln Val

595 600 605

Ala Val Leu Tyr Gln Asp Val Asn Cys Thr Glu Val Pro Val Ala Ile

610 615 620

His Ala Asp Gln Leu Thr Pro Thr Trp Arg Val Tyr Ser Thr Gly Ser

625 630 635 640

Asn Val Phe Gln Thr Arg Ala Gly Cys Leu Ile Gly Ala Glu His Val

645 650 655

Asn Asn Ser Tyr Glu Cys Asp Ile Pro Ile Gly Ala Gly Ile Cys Ala

660 665 670

Ser Tyr Gln Thr Gln Thr Asn Ser Pro Arg Arg Ala Arg Ser Val Ala

675 680 685

Ser Gln Ser Ile Ile Ala Tyr Thr Met Ser Leu Gly Ala Glu Asn Ser

690 695 700

Val Ala Tyr Ser Asn Asn Ser Ile Ala Ile Pro Thr Asn Phe Thr Ile

705 710 715 720

Ser Val Thr Thr Glu Ile Leu Pro Val Ser Met Thr Lys Thr Ser Val

725 730 735

Asp Cys Thr Met Tyr Ile Cys Gly Asp Ser Thr Glu Cys Ser Asn Leu

740 745 750

Leu Leu Gln Tyr Gly Ser Phe Cys Thr Gln Leu Asn Arg Ala Leu Thr

755 760 765

Gly Ile Ala Val Glu Gln Asp Lys Asn Thr Gln Glu Val Phe Ala Gln

770 775 780

Val Lys Gln Ile Tyr Lys Thr Pro Pro Ile Lys Asp Phe Gly Gly Phe

785 790 795 800

Asn Phe Ser Gln Ile Leu Pro Asp Pro Ser Lys Pro Ser Lys Arg Ser

805 810 815

Phe Ile Glu Asp Leu Leu Phe Asn Lys Val Thr Leu Ala Asp Ala Gly

820 825 830

Phe Ile Lys Gln Tyr Gly Asp Cys Leu Gly Asp Ile Ala Ala Arg Asp

835 840 845

Leu Ile Cys Ala Gln Lys Phe Asn Gly Leu Thr Val Leu Pro Pro Leu

850 855 860

Leu Thr Asp Glu Met Ile Ala Gln Tyr Thr Ser Ala Leu Leu Ala Gly

865 870 875 880

Thr Ile Thr Ser Gly Trp Thr Phe Gly Ala Gly Ala Ala Leu Gln Ile

885 890 895

Pro Phe Ala Met Gln Met Ala Tyr Arg Phe Asn Gly Ile Gly Val Thr

900 905 910

Gln Asn Val Leu Tyr Glu Asn Gln Lys Leu Ile Ala Asn Gln Phe Asn

915 920 925

Ser Ala Ile Gly Lys Ile Gln Asp Ser Leu Ser Ser Thr Ala Ser Ala

930 935 940

Leu Gly Lys Leu Gln Asp Val Val Asn Gln Asn Ala Gln Ala Leu Asn

945 950 955 960

Thr Leu Val Lys Gln Leu Ser Ser Asn Phe Gly Ala Ile Ser Ser Val

965 970 975

Leu Asn Asp Ile Leu Ser Arg Leu Asp Lys Val Glu Ala Glu Val Gln

980 985 990

Ile Asp Arg Leu Ile Thr Gly Arg Leu Gln Ser Leu Gln Thr Tyr Val

995 1000 1005

Thr Gln Gln Leu Ile Arg Ala Ala Glu Ile Arg Ala Ser Ala Asn

1010 1015 1020

Leu Ala Ala Thr Lys Met Ser Glu Cys Val Leu Gly Gln Ser Lys

1025 1030 1035

Arg Val Asp Phe Cys Gly Lys Gly Tyr His Leu Met Ser Phe Pro

1040 1045 1050

Gln Ser Ala Pro His Gly Val Val Phe Leu His Val Thr Tyr Val

1055 1060 1065

Pro Ala Gln Glu Lys Asn Phe Thr Thr Ala Pro Ala Ile Cys His

1070 1075 1080

Asp Gly Lys Ala His Phe Pro Arg Glu Gly Val Phe Val Ser Asn

1085 1090 1095

Gly Thr His Trp Phe Val Thr Gln Arg Asn Phe Tyr Glu Pro Gln

1100 1105 1110

Ile Ile Thr Thr Asp Asn Thr Phe Val Ser Gly Asn Cys Asp Val

1115 1120 1125

Val Ile Gly Ile Val Asn Asn Thr Val Tyr Asp Pro Leu Gln Pro

1130 1135 1140

Glu Leu Asp Ser Phe Lys Glu Glu Leu Asp Lys Tyr Phe Lys Asn

1145 1150 1155

His Thr Ser Pro Asp Val Asp Leu Gly Asp Ile Ser Gly Ile Asn

1160 1165 1170

Ala Ser Val Val Asn Ile Gln Lys Glu Ile Asp Arg Leu Asn Glu

1175 1180 1185

Val Ala Lys Asn Leu Asn Glu Ser Leu Ile Asp Leu Gln Glu Leu

1190 1195 1200

Gly Lys Tyr Glu Gln Tyr Ile Lys Trp Pro Trp Tyr Ile Trp Leu

1205 1210 1215

Gly Phe Ile Ala Gly Leu Ile Ala Ile Val Met Val Thr Ile Met

1220 1225 1230

Leu Cys Cys Met Thr Ser Cys Cys Ser Cys Leu Lys Gly Cys Cys

1235 1240 1245

Ser Cys Gly Ser Cys Cys Lys Phe Asp Glu Asp Asp Ser Glu Pro

1250 1255 1260

Val Leu Lys Gly Val Lys Leu His Tyr Thr

1265 1270

<210> 2

<211> 3819

<212> RNA

<213> Artificial sequence

<220>

<223> coding sequence

<400> 2

auguuugugu uucuugugcu gcugccucuu gugucuucuc agugugugaa uuugacaaca 60

agaacacagc ugccaccagc uuauacaaau ucuuuuacca gaggagugua uuauccugau 120

aaaguguuua gaucuucugu gcugcacagc acacaggacc uguuucugcc auuuuuuagc 180

aaugugacau gguuucaugc aauucaugug ucuggaacaa auggaacaaa aagauuugau 240

aauccugugc ugccuuuuaa ugauggagug uauuuugcuu caacagaaaa gucaaauauu 300

auuagaggau ggauuuuugg aacaacacug gauucuaaaa cacagucucu gcugauugug 360

aauaaugcaa caaauguggu gauuaaagug ugugaauuuc aguuuuguaa ugauccuuuu 420

cugggagugu auuaucacaa aaauaauaaa ucuuggaugg aaucugaauu uagaguguau 480

uccucugcaa auaauuguac auuugaauau gugucucagc cuuuucugau ggaucuggaa 540

ggaaaacagg gcaauuuuaa aaaucugaga gaauuugugu uuaaaaauau ugauggauau 600

uuuaaaauuu auucuaaaca cacaccaauu aauuuaguga gagaucugcc ucagggauuu 660

ucugcucugg aaccucuggu ggaucugcca auuggcauua auauuacaag auuucagaca 720

cugcuggcuc ugcacagauc uuaucugaca ccuggagauu cuucuucugg auggacagcc 780

ggagcugcag cuuauuaugu gggcuaucug cagccaagaa cauuucugcu gaaauauaau 840

gaaaauggaa caauuacaga ugcuguggau ugugcucugg auccucuguc ugaaacaaaa 900

uguacauuaa aaucuuuuac aguggaaaaa ggcauuuauc agacaucuaa uuuuagagug 960

cagccaacag aaucuauugu gagauuucca aauauuacaa aucugugucc auuuggagaa 1020

guguuuaaug caacaagauu ugcaucugug uaugcaugga auagaaaaag aauuucuaau 1080

uguguggcug auuauucugu gcuguauaau agugcuucuu uuuccacauu uaaauguuau 1140

ggagugucuc caacaaaauu aaaugauuua uguuuuacaa auguguaugc ugauucuuuu 1200

gugaucagag gugaugaagu gagacagauu gcccccggac agacaggaaa aauugcugau 1260

uacaauuaca aacugccuga ugauuuuaca ggauguguga uugcuuggaa uucuaauaau 1320

uuagauucua aagugggagg aaauuacaau uaucuguaca gacuguuuag aaaaucaaau 1380

cugaaaccuu uugaaagaga uauuucaaca gaaauuuauc aggcuggauc aacaccuugu 1440

aauggagugg aaggauuuaa uuguuauuuu ccauuacaga gcuauggauu ucagccaacc 1500

aauggugugg gauaucagcc auauagagug guggugcugu cuuuugaacu gcugcaugca 1560

ccugcaacag uguguggacc uaaaaaaucu acaaauuuag ugaaaaauaa augugugaau 1620

uuuaauuuua auggauuaac aggaacagga gugcugacag aaucuaauaa aaaauuucug 1680

ccuuuucagc aguuuggcag agauauugca gauaccacag augcagugag agauccucag 1740

acauuagaaa uucuggauau uacaccuugu ucuuuugggg gugugucugu gauuacaccu 1800

ggaacaaaua caucuaauca gguggcugug cuguaucagg augugaauug uacagaagug 1860

ccaguggcaa uucaugcaga ucagcugaca ccaacaugga gaguguauuc uacaggaucu 1920

aauguguuuc agacaagagc aggaugucug auuggagcag aacaugugaa uaauucuuau 1980

gaaugugaua uuccaauugg agcaggcauu ugugcaucuu aucagacaca gacaaauucc 2040

ccaaggagag caagaucugu ggcaucucag ucuauuauug cauacaccau gucucuggga 2100

gcagaaaauu cuguggcaua uucuaauaau ucuauugcua uuccaacaaa uuuuaccauu 2160

ucugugacaa cagaaauuuu accugugucu augacaaaaa caucugugga uuguaccaug 2220

uacauuugug gagauucuac agaauguucu aaucugcugc ugcaguaugg aucuuuuugu 2280

acacagcuga auagagcuuu aacaggaauu gcuguggaac aggauaaaaa uacacaggaa 2340

guguuugcuc aggugaaaca gauuuacaaa acaccaccaa uuaaagauuu uggaggauuu 2400

aauuuuagcc agauucugcc ugauccuucu aaaccuucua aaagaucuuu uauugaagau 2460

cugcuguuua auaaagugac acuggcagau gcaggauuua uuaaacagua uggagauugc 2520

cugggugaua uugcugcaag agaucugauu ugugcucaga aauuuaaugg acugacagug 2580

cugccuccuc ugcugacaga ugaaaugauu gcucaguaca caucugcuuu acuggcugga 2640

acaauuacaa gcggauggac auuuggagcu ggagcugcuc ugcagauucc uuuugcaaug 2700

cagauggcuu acagauuuaa uggaauugga gugacacaga auguguuaua ugaaaaucag 2760

aaacugauug caaaucaguu uaauucugca auuggcaaaa uucaggauuc ucugucuucu 2820

acagcuucug cucugggaaa acugcaggau guggugaauc agaaugcaca ggcacugaau 2880

acucugguga aacagcuguc uagcaauuuu ggggcaauuu cuucugugcu gaaugauauu 2940

cugucuagac uggauaaagu ggaagcugaa gugcagauug auagacugau cacaggaaga 3000

cugcagucuc ugcagacuua ugugacacag cagcugauua gagcugcuga aauuagagcu 3060

ucugcuaauc uggcugcuac aaaaaugucu gaaugugugc ugggacaguc aaaaagagug 3120

gauuuuugug gaaaaggaua ucaucugaug ucuuuuccac agucugcucc acauggagug 3180

guguuuuuac augugacaua ugugccagca caggaaaaga auuuuaccac agcaccagca 3240

auuugucaug auggaaaagc acauuuucca agagaaggag uguuuguguc uaauggaaca 3300

cauugguuug ugacacagag aaauuuuuau gaaccucaga uuauuacaac agauaauaca 3360

uuugugucag gaaauuguga uguggugauu ggaauuguga auaauacagu guaugaucca 3420

cugcagccag aacuggauuc uuuuaaagaa gaacuggaua aauauuuuaa aaaucacaca 3480

ucuccugaug uggauuuagg agauauuucu ggaaucaaug caucuguggu gaauauucag 3540

aaagaaauug auagacugaa ugaaguggcc aaaaaucuga augaaucucu gauugaucug 3600

caggaacuug gaaaauauga acaguacauu aaauggccuu gguacauuug gcuuggauuu 3660

auugcaggau uaauugcaau ugugauggug acaauuaugu uauguuguau gacaucaugu 3720

uguucuuguu uaaaaggaug uuguucuugu ggaagcuguu guaaauuuga ugaagaugau 3780

ucugaaccug uguuaaaagg agugaaauug cauuacaca 3819

<210> 3

<211> 218

<212> PRT

<213> Artificial sequence

<220>

<223> S protein RBD fusion

<400> 3

Met Phe Val Phe Leu Val Leu Leu Pro Leu Val Ser Ser Gln Cys Val

1 5 10 15

Val Arg Phe Pro Asn Ile Thr Asn Leu Cys Pro Phe Gly Glu Val Phe

20 25 30

Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala Trp Asn Arg Lys Arg Ile

35 40 45

Ser Asn Cys Val Ala Asp Tyr Ser Val Leu Tyr Asn Ser Ala Ser Phe

50 55 60

Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro Thr Lys Leu Asn Asp Leu

65 70 75 80

Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe Val Ile Arg Gly Asp Glu

85 90 95

Val Arg Gln Ile Ala Pro Gly Gln Thr Gly Lys Ile Ala Asp Tyr Asn

100 105 110

Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys Val Ile Ala Trp Asn Ser

115 120 125

Asn Asn Leu Asp Ser Lys Val Gly Gly Asn Tyr Asn Tyr Leu Tyr Arg

130 135 140

Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe Glu Arg Asp Ile Ser Thr

145 150 155 160

Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys Asn Gly Val Glu Gly Phe

165 170 175

Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly Phe Gln Pro Thr Asn Gly

180 185 190

Val Gly Tyr Gln Pro Tyr Arg Val Val Val Leu Ser Phe Glu Leu Leu

195 200 205

His Ala Pro Ala Thr Val Cys Gly Pro Lys

210 215

<210> 4

<211> 654

<212> RNA

<213> Artificial sequence

<220>

<223> coding sequence

<400> 4

auguuugugu uucuugugcu gcugccucuu gugucuucuc aguguguggu gagauuucca 60

aauauuacaa aucugugucc auuuggagaa guguuuaaug caacaagauu ugcaucugug 120

uaugcaugga auagaaaaag aauuucuaau uguguggcug auuauucugu gcuguauaau 180

agugcuucuu uuuccacauu uaaauguuau ggagugucuc caacaaaauu aaaugauuua 240

uguuuuacaa auguguaugc ugauucuuuu gugaucagag gugaugaagu gagacagauu 300

gcccccggac agacaggaaa aauugcugau uacaauuaca aacugccuga ugauuuuaca 360

ggauguguga uugcuuggaa uucuaauaau uuagauucua aagugggagg aaauuacaau 420

uaucuguaca gacuguuuag aaaaucaaau cugaaaccuu uugaaagaga uauuucaaca 480

gaaauuuauc aggcuggauc aacaccuugu aauggagugg aaggauuuaa uuguuauuuu 540

ccauuacaga gcuauggauu ucagccaacc aauggugugg gauaucagcc auauagagug 600

guggugcugu cuuuugaacu gcugcaugca ccugcaacag uguguggacc uaaa 654

<210> 5

<211> 266

<212> PRT

<213> Artificial sequence

<220>

<223> S protein RBD fusion

<400> 5

Met Phe Val Phe Leu Val Leu Leu Pro Leu Val Ser Ser Gln Cys Val

1 5 10 15

Val Arg Phe Pro Asn Ile Thr Asn Leu Cys Pro Phe Gly Glu Val Phe

20 25 30

Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala Trp Asn Arg Lys Arg Ile

35 40 45

Ser Asn Cys Val Ala Asp Tyr Ser Val Leu Tyr Asn Ser Ala Ser Phe

50 55 60

Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro Thr Lys Leu Asn Asp Leu

65 70 75 80

Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe Val Ile Arg Gly Asp Glu

85 90 95

Val Arg Gln Ile Ala Pro Gly Gln Thr Gly Lys Ile Ala Asp Tyr Asn

100 105 110

Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys Val Ile Ala Trp Asn Ser

115 120 125

Asn Asn Leu Asp Ser Lys Val Gly Gly Asn Tyr Asn Tyr Leu Tyr Arg

130 135 140

Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe Glu Arg Asp Ile Ser Thr

145 150 155 160

Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys Asn Gly Val Glu Gly Phe

165 170 175

Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly Phe Gln Pro Thr Asn Gly

180 185 190

Val Gly Tyr Gln Pro Tyr Arg Val Val Val Leu Ser Phe Glu Leu Leu

195 200 205

His Ala Pro Ala Thr Val Cys Gly Pro Lys Gly Ser Pro Gly Ser Gly

210 215 220

Ser Gly Ser Gly Tyr Ile Pro Glu Ala Pro Arg Asp Gly Gln Ala Tyr

225 230 235 240

Val Arg Lys Asp Gly Glu Trp Val Leu Leu Ser Thr Phe Leu Gly Arg

245 250 255

Ser Leu Glu Val Leu Phe Gln Gly Pro Gly

260 265

<210> 6

<211> 798

<212> RNA

<213> Artificial sequence

<220>

<223> coding sequence

<400> 6

auguuugugu uucuugugcu gcugccucuu gugucuucuc aguguguggu gagauuucca 60

aauauuacaa aucugugucc auuuggagaa guguuuaaug caacaagauu ugcaucugug 120

uaugcaugga auagaaaaag aauuucuaau uguguggcug auuauucugu gcuguauaau 180

agugcuucuu uuuccacauu uaaauguuau ggagugucuc caacaaaauu aaaugauuua 240

uguuuuacaa auguguaugc ugauucuuuu gugaucagag gugaugaagu gagacagauu 300

gcccccggac agacaggaaa aauugcugau uacaauuaca aacugccuga ugauuuuaca 360

ggauguguga uugcuuggaa uucuaauaau uuagauucua aagugggagg aaauuacaau 420

uaucuguaca gacuguuuag aaaaucaaau cugaaaccuu uugaaagaga uauuucaaca 480

gaaauuuauc aggcuggauc aacaccuugu aauggagugg aaggauuuaa uuguuauuuu 540

ccauuacaga gcuauggauu ucagccaacc aauggugugg gauaucagcc auauagagug 600

guggugcugu cuuuugaacu gcugcaugca ccugcaacag uguguggacc uaaaggcucc 660

cccggcuccg gcuccggauc ugguuauauu ccugaagcuc caagagaugg gcaagcuuac 720

guucguaaag auggcgaaug gguauuacuu ucuaccuuuu uaggccgguc ccuggaggug 780

cuguuccagg gccccggc 798

<210> 7

<211> 1273

<212> PRT

<213> Artificial sequence

<220>

<223> S protein variants

<400> 7

Met Phe Val Phe Leu Val Leu Leu Pro Leu Val Ser Ser Gln Cys Val

1 5 10 15

Asn Leu Thr Thr Arg Thr Gln Leu Pro Pro Ala Tyr Thr Asn Ser Phe

20 25 30

Thr Arg Gly Val Tyr Tyr Pro Asp Lys Val Phe Arg Ser Ser Val Leu

35 40 45

His Ser Thr Gln Asp Leu Phe Leu Pro Phe Phe Ser Asn Val Thr Trp

50 55 60

Phe His Ala Ile His Val Ser Gly Thr Asn Gly Thr Lys Arg Phe Asp

65 70 75 80

Asn Pro Val Leu Pro Phe Asn Asp Gly Val Tyr Phe Ala Ser Thr Glu

85 90 95

Lys Ser Asn Ile Ile Arg Gly Trp Ile Phe Gly Thr Thr Leu Asp Ser

100 105 110

Lys Thr Gln Ser Leu Leu Ile Val Asn Asn Ala Thr Asn Val Val Ile

115 120 125

Lys Val Cys Glu Phe Gln Phe Cys Asn Asp Pro Phe Leu Gly Val Tyr

130 135 140

Tyr His Lys Asn Asn Lys Ser Trp Met Glu Ser Glu Phe Arg Val Tyr

145 150 155 160

Ser Ser Ala Asn Asn Cys Thr Phe Glu Tyr Val Ser Gln Pro Phe Leu

165 170 175

Met Asp Leu Glu Gly Lys Gln Gly Asn Phe Lys Asn Leu Arg Glu Phe

180 185 190

Val Phe Lys Asn Ile Asp Gly Tyr Phe Lys Ile Tyr Ser Lys His Thr

195 200 205

Pro Ile Asn Leu Val Arg Asp Leu Pro Gln Gly Phe Ser Ala Leu Glu

210 215 220

Pro Leu Val Asp Leu Pro Ile Gly Ile Asn Ile Thr Arg Phe Gln Thr

225 230 235 240

Leu Leu Ala Leu His Arg Ser Tyr Leu Thr Pro Gly Asp Ser Ser Ser

245 250 255

Gly Trp Thr Ala Gly Ala Ala Ala Tyr Tyr Val Gly Tyr Leu Gln Pro

260 265 270

Arg Thr Phe Leu Leu Lys Tyr Asn Glu Asn Gly Thr Ile Thr Asp Ala

275 280 285

Val Asp Cys Ala Leu Asp Pro Leu Ser Glu Thr Lys Cys Thr Leu Lys

290 295 300

Ser Phe Thr Val Glu Lys Gly Ile Tyr Gln Thr Ser Asn Phe Arg Val

305 310 315 320

Gln Pro Thr Glu Ser Ile Val Arg Phe Pro Asn Ile Thr Asn Leu Cys

325 330 335

Pro Phe Gly Glu Val Phe Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala

340 345 350

Trp Asn Arg Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser Val Leu

355 360 365

Tyr Asn Ser Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro

370 375 380

Thr Lys Leu Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe

385 390 395 400

Val Ile Arg Gly Asp Glu Val Arg Gln Ile Ala Pro Gly Gln Thr Gly

405 410 415

Lys Ile Ala Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys

420 425 430

Val Ile Ala Trp Asn Ser Asn Asn Leu Asp Ser Lys Val Gly Gly Asn

435 440 445

Tyr Asn Tyr Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe

450 455 460

Glu Arg Asp Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys

465 470 475 480

Asn Gly Val Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly

485 490 495

Phe Gln Pro Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val Val Val

500 505 510

Leu Ser Phe Glu Leu Leu His Ala Pro Ala Thr Val Cys Gly Pro Lys

515 520 525

Lys Ser Thr Asn Leu Val Lys Asn Lys Cys Val Asn Phe Asn Phe Asn

530 535 540

Gly Leu Thr Gly Thr Gly Val Leu Thr Glu Ser Asn Lys Lys Phe Leu

545 550 555 560

Pro Phe Gln Gln Phe Gly Arg Asp Ile Ala Asp Thr Thr Asp Ala Val

565 570 575

Arg Asp Pro Gln Thr Leu Glu Ile Leu Asp Ile Thr Pro Cys Ser Phe

580 585 590

Gly Gly Val Ser Val Ile Thr Pro Gly Thr Asn Thr Ser Asn Gln Val

595 600 605

Ala Val Leu Tyr Gln Asp Val Asn Cys Thr Glu Val Pro Val Ala Ile

610 615 620

His Ala Asp Gln Leu Thr Pro Thr Trp Arg Val Tyr Ser Thr Gly Ser

625 630 635 640

Asn Val Phe Gln Thr Arg Ala Gly Cys Leu Ile Gly Ala Glu His Val

645 650 655

Asn Asn Ser Tyr Glu Cys Asp Ile Pro Ile Gly Ala Gly Ile Cys Ala

660 665 670

Ser Tyr Gln Thr Gln Thr Asn Ser Pro Arg Arg Ala Arg Ser Val Ala

675 680 685

Ser Gln Ser Ile Ile Ala Tyr Thr Met Ser Leu Gly Ala Glu Asn Ser

690 695 700

Val Ala Tyr Ser Asn Asn Ser Ile Ala Ile Pro Thr Asn Phe Thr Ile

705 710 715 720

Ser Val Thr Thr Glu Ile Leu Pro Val Ser Met Thr Lys Thr Ser Val

725 730 735

Asp Cys Thr Met Tyr Ile Cys Gly Asp Ser Thr Glu Cys Ser Asn Leu

740 745 750

Leu Leu Gln Tyr Gly Ser Phe Cys Thr Gln Leu Asn Arg Ala Leu Thr

755 760 765

Gly Ile Ala Val Glu Gln Asp Lys Asn Thr Gln Glu Val Phe Ala Gln

770 775 780

Val Lys Gln Ile Tyr Lys Thr Pro Pro Ile Lys Asp Phe Gly Gly Phe

785 790 795 800

Asn Phe Ser Gln Ile Leu Pro Asp Pro Ser Lys Pro Ser Lys Arg Ser

805 810 815

Phe Ile Glu Asp Leu Leu Phe Asn Lys Val Thr Leu Ala Asp Ala Gly

820 825 830

Phe Ile Lys Gln Tyr Gly Asp Cys Leu Gly Asp Ile Ala Ala Arg Asp

835 840 845

Leu Ile Cys Ala Gln Lys Phe Asn Gly Leu Thr Val Leu Pro Pro Leu

850 855 860

Leu Thr Asp Glu Met Ile Ala Gln Tyr Thr Ser Ala Leu Leu Ala Gly

865 870 875 880

Thr Ile Thr Ser Gly Trp Thr Phe Gly Ala Gly Ala Ala Leu Gln Ile

885 890 895

Pro Phe Ala Met Gln Met Ala Tyr Arg Phe Asn Gly Ile Gly Val Thr

900 905 910

Gln Asn Val Leu Tyr Glu Asn Gln Lys Leu Ile Ala Asn Gln Phe Asn

915 920 925

Ser Ala Ile Gly Lys Ile Gln Asp Ser Leu Ser Ser Thr Ala Ser Ala

930 935 940

Leu Gly Lys Leu Gln Asp Val Val Asn Gln Asn Ala Gln Ala Leu Asn

945 950 955 960

Thr Leu Val Lys Gln Leu Ser Ser Asn Phe Gly Ala Ile Ser Ser Val

965 970 975

Leu Asn Asp Ile Leu Ser Arg Leu Asp Pro Pro Glu Ala Glu Val Gln

980 985 990

Ile Asp Arg Leu Ile Thr Gly Arg Leu Gln Ser Leu Gln Thr Tyr Val

995 1000 1005

Thr Gln Gln Leu Ile Arg Ala Ala Glu Ile Arg Ala Ser Ala Asn

1010 1015 1020

Leu Ala Ala Thr Lys Met Ser Glu Cys Val Leu Gly Gln Ser Lys

1025 1030 1035

Arg Val Asp Phe Cys Gly Lys Gly Tyr His Leu Met Ser Phe Pro

1040 1045 1050

Gln Ser Ala Pro His Gly Val Val Phe Leu His Val Thr Tyr Val

1055 1060 1065

Pro Ala Gln Glu Lys Asn Phe Thr Thr Ala Pro Ala Ile Cys His

1070 1075 1080

Asp Gly Lys Ala His Phe Pro Arg Glu Gly Val Phe Val Ser Asn

1085 1090 1095

Gly Thr His Trp Phe Val Thr Gln Arg Asn Phe Tyr Glu Pro Gln

1100 1105 1110

Ile Ile Thr Thr Asp Asn Thr Phe Val Ser Gly Asn Cys Asp Val

1115 1120 1125

Val Ile Gly Ile Val Asn Asn Thr Val Tyr Asp Pro Leu Gln Pro

1130 1135 1140

Glu Leu Asp Ser Phe Lys Glu Glu Leu Asp Lys Tyr Phe Lys Asn

1145 1150 1155

His Thr Ser Pro Asp Val Asp Leu Gly Asp Ile Ser Gly Ile Asn

1160 1165 1170

Ala Ser Val Val Asn Ile Gln Lys Glu Ile Asp Arg Leu Asn Glu

1175 1180 1185

Val Ala Lys Asn Leu Asn Glu Ser Leu Ile Asp Leu Gln Glu Leu

1190 1195 1200

Gly Lys Tyr Glu Gln Tyr Ile Lys Trp Pro Trp Tyr Ile Trp Leu

1205 1210 1215

Gly Phe Ile Ala Gly Leu Ile Ala Ile Val Met Val Thr Ile Met

1220 1225 1230

Leu Cys Cys Met Thr Ser Cys Cys Ser Cys Leu Lys Gly Cys Cys

1235 1240 1245

Ser Cys Gly Ser Cys Cys Lys Phe Asp Glu Asp Asp Ser Glu Pro

1250 1255 1260

Val Leu Lys Gly Val Lys Leu His Tyr Thr

1265 1270

<210> 8

<211> 3819

<212> RNA

<213> Artificial sequence

<220>

<223> coding sequence

<400> 8

auguuugugu uucuugugcu gcugccucuu gugucuucuc agugugugaa uuugacaaca 60

agaacacagc ugccaccagc uuauacaaau ucuuuuacca gaggagugua uuauccugau 120

aaaguguuua gaucuucugu gcugcacagc acacaggacc uguuucugcc auuuuuuagc 180

aaugugacau gguuucaugc aauucaugug ucuggaacaa auggaacaaa aagauuugau 240

aauccugugc ugccuuuuaa ugauggagug uauuuugcuu caacagaaaa gucaaauauu 300

auuagaggau ggauuuuugg aacaacacug gauucuaaaa cacagucucu gcugauugug 360

aauaaugcaa caaauguggu gauuaaagug ugugaauuuc aguuuuguaa ugauccuuuu 420

cugggagugu auuaucacaa aaauaauaaa ucuuggaugg aaucugaauu uagaguguau 480

uccucugcaa auaauuguac auuugaauau gugucucagc cuuuucugau ggaucuggaa 540

ggaaaacagg gcaauuuuaa aaaucugaga gaauuugugu uuaaaaauau ugauggauau 600

uuuaaaauuu auucuaaaca cacaccaauu aauuuaguga gagaucugcc ucagggauuu 660

ucugcucugg aaccucuggu ggaucugcca auuggcauua auauuacaag auuucagaca 720

cugcuggcuc ugcacagauc uuaucugaca ccuggagauu cuucuucugg auggacagcc 780

ggagcugcag cuuauuaugu gggcuaucug cagccaagaa cauuucugcu gaaauauaau 840

gaaaauggaa caauuacaga ugcuguggau ugugcucugg auccucuguc ugaaacaaaa 900

uguacauuaa aaucuuuuac aguggaaaaa ggcauuuauc agacaucuaa uuuuagagug 960

cagccaacag aaucuauugu gagauuucca aauauuacaa aucugugucc auuuggagaa 1020

guguuuaaug caacaagauu ugcaucugug uaugcaugga auagaaaaag aauuucuaau 1080

uguguggcug auuauucugu gcuguauaau agugcuucuu uuuccacauu uaaauguuau 1140

ggagugucuc caacaaaauu aaaugauuua uguuuuacaa auguguaugc ugauucuuuu 1200

gugaucagag gugaugaagu gagacagauu gcccccggac agacaggaaa aauugcugau 1260

uacaauuaca aacugccuga ugauuuuaca ggauguguga uugcuuggaa uucuaauaau 1320

uuagauucua aagugggagg aaauuacaau uaucuguaca gacuguuuag aaaaucaaau 1380

cugaaaccuu uugaaagaga uauuucaaca gaaauuuauc aggcuggauc aacaccuugu 1440

aauggagugg aaggauuuaa uuguuauuuu ccauuacaga gcuauggauu ucagccaacc 1500

aauggugugg gauaucagcc auauagagug guggugcugu cuuuugaacu gcugcaugca 1560

ccugcaacag uguguggacc uaaaaaaucu acaaauuuag ugaaaaauaa augugugaau 1620

uuuaauuuua auggauuaac aggaacagga gugcugacag aaucuaauaa aaaauuucug 1680

ccuuuucagc aguuuggcag agauauugca gauaccacag augcagugag agauccucag 1740

acauuagaaa uucuggauau uacaccuugu ucuuuugggg gugugucugu gauuacaccu 1800

ggaacaaaua caucuaauca gguggcugug cuguaucagg augugaauug uacagaagug 1860

ccaguggcaa uucaugcaga ucagcugaca ccaacaugga gaguguauuc uacaggaucu 1920

aauguguuuc agacaagagc aggaugucug auuggagcag aacaugugaa uaauucuuau 1980

gaaugugaua uuccaauugg agcaggcauu ugugcaucuu aucagacaca gacaaauucc 2040

ccaaggagag caagaucugu ggcaucucag ucuauuauug cauacaccau gucucuggga 2100

gcagaaaauu cuguggcaua uucuaauaau ucuauugcua uuccaacaaa uuuuaccauu 2160

ucugugacaa cagaaauuuu accugugucu augacaaaaa caucugugga uuguaccaug 2220

uacauuugug gagauucuac agaauguucu aaucugcugc ugcaguaugg aucuuuuugu 2280

acacagcuga auagagcuuu aacaggaauu gcuguggaac aggauaaaaa uacacaggaa 2340

guguuugcuc aggugaaaca gauuuacaaa acaccaccaa uuaaagauuu uggaggauuu 2400

aauuuuagcc agauucugcc ugauccuucu aaaccuucua aaagaucuuu uauugaagau 2460

cugcuguuua auaaagugac acuggcagau gcaggauuua uuaaacagua uggagauugc 2520

cugggugaua uugcugcaag agaucugauu ugugcucaga aauuuaaugg acugacagug 2580

cugccuccuc ugcugacaga ugaaaugauu gcucaguaca caucugcuuu acuggcugga 2640

acaauuacaa gcggauggac auuuggagcu ggagcugcuc ugcagauucc uuuugcaaug 2700

cagauggcuu acagauuuaa uggaauugga gugacacaga auguguuaua ugaaaaucag 2760

aaacugauug caaaucaguu uaauucugca auuggcaaaa uucaggauuc ucugucuucu 2820

acagcuucug cucugggaaa acugcaggau guggugaauc agaaugcaca ggcacugaau 2880

acucugguga aacagcuguc uagcaauuuu ggggcaauuu cuucugugcu gaaugauauu 2940

cugucuagac uggauccucc ugaagcugaa gugcagauug auagacugau cacaggaaga 3000

cugcagucuc ugcagacuua ugugacacag cagcugauua gagcugcuga aauuagagcu 3060

ucugcuaauc uggcugcuac aaaaaugucu gaaugugugc ugggacaguc aaaaagagug 3120

gauuuuugug gaaaaggaua ucaucugaug ucuuuuccac agucugcucc acauggagug 3180

guguuuuuac augugacaua ugugccagca caggaaaaga auuuuaccac agcaccagca 3240

auuugucaug auggaaaagc acauuuucca agagaaggag uguuuguguc uaauggaaca 3300

cauugguuug ugacacagag aaauuuuuau gaaccucaga uuauuacaac agauaauaca 3360

uuugugucag gaaauuguga uguggugauu ggaauuguga auaauacagu guaugaucca 3420

cugcagccag aacuggauuc uuuuaaagaa gaacuggaua aauauuuuaa aaaucacaca 3480

ucuccugaug uggauuuagg agauauuucu ggaaucaaug caucuguggu gaauauucag 3540

aaagaaauug auagacugaa ugaaguggcc aaaaaucuga augaaucucu gauugaucug 3600

caggaacuug gaaaauauga acaguacauu aaauggccuu gguacauuug gcuuggauuu 3660

auugcaggau uaauugcaau ugugauggug acaauuaugu uauguuguau gacaucaugu 3720

uguucuuguu uaaaaggaug uuguucuugu ggaagcuguu guaaauuuga ugaagaugau 3780

ucugaaccug uguuaaaagg agugaaauug cauuacaca 3819

<210> 9

<211> 3819

<212> RNA

<213> Artificial sequence

<220>

<223> coding sequence

<400> 9

auguucgugu uccuggugcu gcugccucug guguccagcc agugugugaa ccugaccacc 60

agaacacagc ugccuccagc cuacaccaac agcuuuacca gaggcgugua cuaccccgac 120

aagguguuca gauccagcgu gcugcacucu acccaggacc uguuccugcc uuucuucagc 180

aacgugaccu gguuccacgc cauccacgug uccggcacca auggcaccaa gagauucgac 240

aaccccgugc ugcccuucaa cgacggggug uacuuugcca gcaccgagaa guccaacauc 300

aucagaggcu ggaucuucgg caccacacug gacagcaaga cccagagccu gcugaucgug 360

aacaacgcca ccaacguggu caucaaagug ugcgaguucc aguucugcaa cgaccccuuc 420

cugggcgucu acuaccacaa gaacaacaag agcuggaugg aaagcgaguu ccggguguac 480

agcagcgcca acaacugcac cuucgaguac gugucccagc cuuuccugau ggaccuggaa 540

ggcaagcagg gcaacuucaa gaaccugcgc gaguucgugu uuaagaacau cgacggcuac 600

uucaagaucu acagcaagca caccccuauc aaccucgugc gggaucugcc ucagggcuuc 660

ucugcucugg aaccccuggu ggaucugccc aucggcauca acaucacccg guuucagaca 720

cugcuggccc ugcacagaag cuaccugaca ccuggcgaua gcagcagcgg auggacagcu 780

ggugccgccg cuuacuaugu gggcuaccug cagccuagaa ccuuccugcu gaaguacaac 840

gagaacggca ccaucaccga cgccguggau ugugcucugg auccucugag cgagacaaag 900

ugcacccuga aguccuucac cguggaaaag ggcaucuacc agaccagcaa cuuccgggug 960

cagcccaccg aauccaucgu gcgguucccc aauaucacca aucugugccc cuucggcgag 1020

guguucaaug ccaccagauu cgccucugug uacgccugga accggaagcg gaucagcaau 1080

ugcguggccg acuacuccgu gcuguacaac uccgccagcu ucagcaccuu caagugcuac 1140

ggcguguccc cuaccaagcu gaacgaccug ugcuucacaa acguguacgc cgacagcuuc 1200

gugauccggg gagaugaagu gcggcagauu gccccuggac agacaggcaa gaucgccgac 1260

uacaacuaca agcugcccga cgacuucacc ggcuguguga uugccuggaa cagcaacaac 1320

cuggacucca aagucggcgg caacuacaau uaccuguacc ggcuguuccg gaaguccaau 1380

cugaagcccu ucgagcggga caucuccacc gagaucuauc aggccggcag caccccuugu 1440

aacggcgugg aaggcuucaa cugcuacuuc ccacugcagu ccuacggcuu ucagcccaca 1500

aauggcgugg gcuaucagcc cuacagagug guggugcuga gcuucgaacu gcugcaugcc 1560

ccugccacag ugugcggccc uaagaaaagc accaaucucg ugaagaacaa augcgugaac 1620

uucaacuuca acggccugac cggcaccggc gugcugacag agagcaacaa gaaguuccug 1680

ccauuccagc aguuuggccg ggauaucgcc gauaccacag acgccguuag agauccccag 1740

acacuggaaa uccuggacau caccccuugc agcuucggcg gagugucugu gaucaccccu 1800

ggcaccaaca ccagcaauca gguggcagug cuguaccagg acgugaacug uaccgaagug 1860

cccguggcca uucacgccga ucagcugaca ccuacauggc ggguguacuc caccggcagc 1920

aauguguuuc agaccagagc cggcugucug aucggagccg agcacgugaa caauagcuac 1980

gagugcgaca uccccaucgg cgcuggaauc ugcgccagcu accagacaca gacaaacagc 2040

ccucggagag ccagaagcgu ggccagccag agcaucauug ccuacacaau gucucugggc 2100

gccgagaaca gcguggccua cuccaacaac ucuaucgcua uccccaccaa cuucaccauc 2160

agcgugacca cagagauccu gccugugucc augaccaaga ccagcgugga cugcaccaug 2220

uacaucugcg gcgauuccac cgagugcucc aaccugcugc ugcaguacgg cagcuucugc 2280

acccagcuga auagagcccu gacagggauc gccguggaac aggacaagaa cacccaagag 2340

guguucgccc aagugaagca gaucuacaag accccuccua ucaaggacuu cggcggcuuc 2400

aauuucagcc agauucugcc cgauccuagc aagcccagca agcggagcuu caucgaggac 2460

cugcuguuca acaaagugac acuggccgac gccggcuuca ucaagcagua uggcgauugu 2520

cugggcgaca uugccgccag ggaucugauu ugcgcccaga aguuuaacgg acugacagug 2580

cugccuccuc ugcugaccga ugagaugauc gcccaguaca caucugcccu gcuggccggc 2640

acaaucacaa gcggcuggac auuuggagca ggcgccgcuc ugcagauccc cuuugcuaug 2700

cagauggccu accgguucaa cggcaucgga gugacccaga augugcugua cgagaaccag 2760

aagcugaucg ccaaccaguu caacagcgcc aucggcaaga uccaggacag ccugagcagc 2820

acagcaagcg cccugggaaa gcugcaggac guggucaacc agaaugccca ggcacugaac 2880

acccugguca agcagcuguc cuccaacuuc ggcgccauca gcucugugcu gaacgauauc 2940

cugagcagac uggacccucc ugaggccgag gugcagaucg acagacugau cacaggcaga 3000

cugcagagcc uccagacaua cgugacccag cagcugauca gagccgccga gauuagagcc 3060

ucugccaauc uggccgccac caagaugucu gagugugugc ugggccagag caagagagug 3120

gacuuuugcg gcaagggcua ccaccugaug agcuucccuc agucugcccc ucacggcgug 3180

guguuucugc acgugacaua ugugcccgcu caagagaaga auuucaccac cgcuccagcc 3240

aucugccacg acggcaaagc ccacuuuccu agagaaggcg uguucguguc caacggcacc 3300

cauugguucg ugacacagcg gaacuucuac gagccccaga ucaucaccac cgacaacacc 3360

uucgugucug gcaacugcga cgucgugauc ggcauuguga acaauaccgu guacgacccu 3420

cugcagcccg agcuggacag cuucaaagag gaacuggaca aguacuuuaa gaaccacaca 3480

agccccgacg uggaccuggg cgauaucagc ggaaucaaug ccagcgucgu gaacauccag 3540

aaagagaucg accggcugaa cgagguggcc aagaaucuga acgagagccu gaucgaccug 3600

caagaacugg ggaaguacga gcaguacauc aaguggcccu gguacaucug gcugggcuuu 3660

aucgccggac ugauugccau cgugaugguc acaaucaugc uguguugcau gaccagcugc 3720

uguagcugcc ugaagggcug uuguagcugu ggcagcugcu gcaaguucga cgaggacgau 3780

ucugagcccg ugcugaaggg cgugaaacug cacuacaca 3819

<210> 10

<211> 41

<212> PRT

<213> Artificial sequence

<220>

<223> folder subsequence

<400> 10

Gly Ser Gly Tyr Ile Pro Glu Ala Pro Arg Asp Gly Gln Ala Tyr Val

1 5 10 15

Arg Lys Asp Gly Glu Trp Val Leu Leu Ser Thr Phe Leu Gly Arg Ser

20 25 30

Leu Glu Val Leu Phe Gln Gly Pro Gly

35 40

<210> 11

<211> 123

<212> RNA

<213> Artificial sequence

<220>

<223> coding sequence

<400> 11

ggaucugguu auauuccuga agcuccaaga gaugggcaag cuuacguucg uaaagauggc 60

gaauggguau uacuuucuac cuuuuuaggc cggucccugg aggugcuguu ccagggcccc 120

ggc 123

<210> 12

<211> 47

<212> RNA

<213> Artificial sequence

<220>

<223> 5'-UTR

<400> 12

aacuaguauu cuucuggucc ccacagacuc agagagaacc cgccacc 47

<210> 13

<211> 278

<212> RNA

<213> Artificial sequence

<220>

<223> 3'-UTR

<400> 13

cugguacugc augcacgcaa ugcuagcugc cccuuucccg uccuggguac cccgagucuc 60

ccccgaccuc gggucccagg uaugcuccca ccuccaccug ccccacucac caccucugcu 120

aguuccagac accucccaag cacgcagcaa ugcagcucaa aacgcuuagc cuagccacac 180

ccccacggga aacagcagug auuaaccuuu agcaauaaac gaaaguuuaa cuaagcuaua 240

cuaaccccag gguuggucaa uuucgugcca gccacacc 278

<210> 14

<211> 110

<212> RNA

<213> Artificial sequence

<220>

<223> A30L70

<400> 14

aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa gcauaugacu aaaaaaaaaa aaaaaaaaaa 60

aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 110

<210> 15

<211> 4282

<212> RNA

<213> Artificial sequence

<220>

<223> RBL063.1

<400> 15

gggcgaacua guauucuucu gguccccaca gacucagaga gaacccgcca ccauguuugu 60

guuucuugug cugcugccuc uugugucuuc ucagugugug aauuugacaa caagaacaca 120

gcugccacca gcuuauacaa auucuuuuac cagaggagug uauuauccug auaaaguguu 180

uagaucuucu gugcugcaca gcacacagga ccuguuucug ccauuuuuua gcaaugugac 240

augguuucau gcaauucaug ugucuggaac aaauggaaca aaaagauuug auaauccugu 300

gcugccuuuu aaugauggag uguauuuugc uucaacagaa aagucaaaua uuauuagagg 360

auggauuuuu ggaacaacac uggauucuaa aacacagucu cugcugauug ugaauaaugc 420

aacaaaugug gugauuaaag ugugugaauu ucaguuuugu aaugauccuu uucugggagu 480

guauuaucac aaaaauaaua aaucuuggau ggaaucugaa uuuagagugu auuccucugc 540

aaauaauugu acauuugaau augugucuca gccuuuucug auggaucugg aaggaaaaca 600

gggcaauuuu aaaaaucuga gagaauuugu guuuaaaaau auugauggau auuuuaaaau 660

uuauucuaaa cacacaccaa uuaauuuagu gagagaucug ccucagggau uuucugcucu 720

ggaaccucug guggaucugc caauuggcau uaauauuaca agauuucaga cacugcuggc 780

ucugcacaga ucuuaucuga caccuggaga uucuucuucu ggauggacag ccggagcugc 840

agcuuauuau gugggcuauc ugcagccaag aacauuucug cugaaauaua augaaaaugg 900

aacaauuaca gaugcugugg auugugcucu ggauccucug ucugaaacaa aauguacauu 960

aaaaucuuuu acaguggaaa aaggcauuua ucagacaucu aauuuuagag ugcagccaac 1020

agaaucuauu gugagauuuc caaauauuac aaaucugugu ccauuuggag aaguguuuaa 1080

ugcaacaaga uuugcaucug uguaugcaug gaauagaaaa agaauuucua auuguguggc 1140

ugauuauucu gugcuguaua auagugcuuc uuuuuccaca uuuaaauguu auggaguguc 1200

uccaacaaaa uuaaaugauu uauguuuuac aaauguguau gcugauucuu uugugaucag 1260

aggugaugaa gugagacaga uugcccccgg acagacagga aaaauugcug auuacaauua 1320

caaacugccu gaugauuuua caggaugugu gauugcuugg aauucuaaua auuuagauuc 1380

uaaaguggga ggaaauuaca auuaucugua cagacuguuu agaaaaucaa aucugaaacc 1440

uuuugaaaga gauauuucaa cagaaauuua ucaggcugga ucaacaccuu guaauggagu 1500

ggaaggauuu aauuguuauu uuccauuaca gagcuaugga uuucagccaa ccaauggugu 1560

gggauaucag ccauauagag ugguggugcu gucuuuugaa cugcugcaug caccugcaac 1620

agugugugga ccuaaaaaau cuacaaauuu agugaaaaau aaauguguga auuuuaauuu 1680

uaauggauua acaggaacag gagugcugac agaaucuaau aaaaaauuuc ugccuuuuca 1740

gcaguuuggc agagauauug cagauaccac agaugcagug agagauccuc agacauuaga 1800

aauucuggau auuacaccuu guucuuuugg gggugugucu gugauuacac cuggaacaaa 1860

uacaucuaau cagguggcug ugcuguauca ggaugugaau uguacagaag ugccaguggc 1920

aauucaugca gaucagcuga caccaacaug gagaguguau ucuacaggau cuaauguguu 1980

ucagacaaga gcaggauguc ugauuggagc agaacaugug aauaauucuu augaauguga 2040

uauuccaauu ggagcaggca uuugugcauc uuaucagaca cagacaaauu ccccaaggag 2100

agcaagaucu guggcaucuc agucuauuau ugcauacacc augucucugg gagcagaaaa 2160

uucuguggca uauucuaaua auucuauugc uauuccaaca aauuuuacca uuucugugac 2220

aacagaaauu uuaccugugu cuaugacaaa aacaucugug gauuguacca uguacauuug 2280

uggagauucu acagaauguu cuaaucugcu gcugcaguau ggaucuuuuu guacacagcu 2340

gaauagagcu uuaacaggaa uugcugugga acaggauaaa aauacacagg aaguguuugc 2400

ucaggugaaa cagauuuaca aaacaccacc aauuaaagau uuuggaggau uuaauuuuag 2460

ccagauucug ccugauccuu cuaaaccuuc uaaaagaucu uuuauugaag aucugcuguu 2520

uaauaaagug acacuggcag augcaggauu uauuaaacag uauggagauu gccuggguga 2580

uauugcugca agagaucuga uuugugcuca gaaauuuaau ggacugacag ugcugccucc 2640

ucugcugaca gaugaaauga uugcucagua cacaucugcu uuacuggcug gaacaauuac 2700

aagcggaugg acauuuggag cuggagcugc ucugcagauu ccuuuugcaa ugcagauggc 2760

uuacagauuu aauggaauug gagugacaca gaauguguua uaugaaaauc agaaacugau 2820

ugcaaaucag uuuaauucug caauuggcaa aauucaggau ucucugucuu cuacagcuuc 2880

ugcucuggga aaacugcagg auguggugaa ucagaaugca caggcacuga auacucuggu 2940

gaaacagcug ucuagcaauu uuggggcaau uucuucugug cugaaugaua uucugucuag 3000

acuggauccu ccugaagcug aagugcagau ugauagacug aucacaggaa gacugcaguc 3060

ucugcagacu uaugugacac agcagcugau uagagcugcu gaaauuagag cuucugcuaa 3120

ucuggcugcu acaaaaaugu cugaaugugu gcugggacag ucaaaaagag uggauuuuug 3180

uggaaaagga uaucaucuga ugucuuuucc acagucugcu ccacauggag ugguguuuuu 3240

acaugugaca uaugugccag cacaggaaaa gaauuuuacc acagcaccag caauuuguca 3300

ugauggaaaa gcacauuuuc caagagaagg aguguuugug ucuaauggaa cacauugguu 3360

ugugacacag agaaauuuuu augaaccuca gauuauuaca acagauaaua cauuuguguc 3420

aggaaauugu gaugugguga uuggaauugu gaauaauaca guguaugauc cacugcagcc 3480

agaacuggau ucuuuuaaag aagaacugga uaaauauuuu aaaaaucaca caucuccuga 3540

uguggauuua ggagauauuu cuggaaucaa ugcaucugug gugaauauuc agaaagaaau 3600

ugauagacug aaugaagugg ccaaaaaucu gaaugaaucu cugauugauc ugcaggaacu 3660

uggaaaauau gaacaguaca uuaaauggcc uugguacauu uggcuuggau uuauugcagg 3720

auuaauugca auugugaugg ugacaauuau guuauguugu augacaucau guuguucuug 3780

uuuaaaagga uguuguucuu guggaagcug uuguaaauuu gaugaagaug auucugaacc 3840

uguguuaaaa ggagugaaau ugcauuacac augaugacuc gagcugguac ugcaugcacg 3900

caaugcuagc ugccccuuuc ccguccuggg uaccccgagu cucccccgac cucggguccc 3960

agguaugcuc ccaccuccac cugccccacu caccaccucu gcuaguucca gacaccuccc 4020

aagcacgcag caaugcagcu caaaacgcuu agccuagcca cacccccacg ggaaacagca 4080

gugauuaacc uuuagcaaua aacgaaaguu uaacuaagcu auacuaaccc caggguuggu 4140

caauuucgug ccagccacac ccuggagcua gcaaaaaaaa aaaaaaaaaa aaaaaaaaaa 4200

aagcauauga cuaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 4260

aaaaaaaaaa aaaaaaaaaa aa 4282

<210> 16

<211> 4282

<212> RNA

<213> Artificial sequence

<220>

<223> RBL063.2

<400> 16

gggcgaacua guauucuucu gguccccaca gacucagaga gaacccgcca ccauguucgu 60

guuccuggug cugcugccuc ugguguccag ccagugugug aaccugacca ccagaacaca 120

gcugccucca gccuacacca acagcuuuac cagaggcgug uacuaccccg acaagguguu 180

cagauccagc gugcugcacu cuacccagga ccuguuccug ccuuucuuca gcaacgugac 240

cugguuccac gccauccacg uguccggcac caauggcacc aagagauucg acaaccccgu 300

gcugcccuuc aacgacgggg uguacuuugc cagcaccgag aaguccaaca ucaucagagg 360

cuggaucuuc ggcaccacac uggacagcaa gacccagagc cugcugaucg ugaacaacgc 420

caccaacgug gucaucaaag ugugcgaguu ccaguucugc aacgaccccu uccugggcgu 480

cuacuaccac aagaacaaca agagcuggau ggaaagcgag uuccgggugu acagcagcgc 540

caacaacugc accuucgagu acguguccca gccuuuccug auggaccugg aaggcaagca 600

gggcaacuuc aagaaccugc gcgaguucgu guuuaagaac aucgacggcu acuucaagau 660

cuacagcaag cacaccccua ucaaccucgu gcgggaucug ccucagggcu ucucugcucu 720

ggaaccccug guggaucugc ccaucggcau caacaucacc cgguuucaga cacugcuggc 780

ccugcacaga agcuaccuga caccuggcga uagcagcagc ggauggacag cuggugccgc 840

cgcuuacuau gugggcuacc ugcagccuag aaccuuccug cugaaguaca acgagaacgg 900

caccaucacc gacgccgugg auugugcucu ggauccucug agcgagacaa agugcacccu 960

gaaguccuuc accguggaaa agggcaucua ccagaccagc aacuuccggg ugcagcccac 1020

cgaauccauc gugcgguucc ccaauaucac caaucugugc cccuucggcg agguguucaa 1080

ugccaccaga uucgccucug uguacgccug gaaccggaag cggaucagca auugcguggc 1140

cgacuacucc gugcuguaca acuccgccag cuucagcacc uucaagugcu acggcguguc 1200

cccuaccaag cugaacgacc ugugcuucac aaacguguac gccgacagcu ucgugauccg 1260

gggagaugaa gugcggcaga uugccccugg acagacaggc aagaucgccg acuacaacua 1320

caagcugccc gacgacuuca ccggcugugu gauugccugg aacagcaaca accuggacuc 1380

caaagucggc ggcaacuaca auuaccugua ccggcuguuc cggaagucca aucugaagcc 1440

cuucgagcgg gacaucucca ccgagaucua ucaggccggc agcaccccuu guaacggcgu 1500

ggaaggcuuc aacugcuacu ucccacugca guccuacggc uuucagccca caaauggcgu 1560

gggcuaucag cccuacagag ugguggugcu gagcuucgaa cugcugcaug ccccugccac 1620

agugugcggc ccuaagaaaa gcaccaaucu cgugaagaac aaaugcguga acuucaacuu 1680

caacggccug accggcaccg gcgugcugac agagagcaac aagaaguucc ugccauucca 1740

gcaguuuggc cgggauaucg ccgauaccac agacgccguu agagaucccc agacacugga 1800

aauccuggac aucaccccuu gcagcuucgg cggagugucu gugaucaccc cuggcaccaa 1860

caccagcaau cagguggcag ugcuguacca ggacgugaac uguaccgaag ugcccguggc 1920

cauucacgcc gaucagcuga caccuacaug gcggguguac uccaccggca gcaauguguu 1980

ucagaccaga gccggcuguc ugaucggagc cgagcacgug aacaauagcu acgagugcga 2040

cauccccauc ggcgcuggaa ucugcgccag cuaccagaca cagacaaaca gcccucggag 2100

agccagaagc guggccagcc agagcaucau ugccuacaca augucucugg gcgccgagaa 2160

cagcguggcc uacuccaaca acucuaucgc uauccccacc aacuucacca ucagcgugac 2220

cacagagauc cugccugugu ccaugaccaa gaccagcgug gacugcacca uguacaucug 2280

cggcgauucc accgagugcu ccaaccugcu gcugcaguac ggcagcuucu gcacccagcu 2340

gaauagagcc cugacaggga ucgccgugga acaggacaag aacacccaag agguguucgc 2400

ccaagugaag cagaucuaca agaccccucc uaucaaggac uucggcggcu ucaauuucag 2460

ccagauucug cccgauccua gcaagcccag caagcggagc uucaucgagg accugcuguu 2520

caacaaagug acacuggccg acgccggcuu caucaagcag uauggcgauu gucugggcga 2580

cauugccgcc agggaucuga uuugcgccca gaaguuuaac ggacugacag ugcugccucc 2640

ucugcugacc gaugagauga ucgcccagua cacaucugcc cugcuggccg gcacaaucac 2700

aagcggcugg acauuuggag caggcgccgc ucugcagauc cccuuugcua ugcagauggc 2760

cuaccgguuc aacggcaucg gagugaccca gaaugugcug uacgagaacc agaagcugau 2820

cgccaaccag uucaacagcg ccaucggcaa gauccaggac agccugagca gcacagcaag 2880

cgcccuggga aagcugcagg acguggucaa ccagaaugcc caggcacuga acacccuggu 2940

caagcagcug uccuccaacu ucggcgccau cagcucugug cugaacgaua uccugagcag 3000

acuggacccu ccugaggccg aggugcagau cgacagacug aucacaggca gacugcagag 3060

ccuccagaca uacgugaccc agcagcugau cagagccgcc gagauuagag ccucugccaa 3120

ucuggccgcc accaagaugu cugagugugu gcugggccag agcaagagag uggacuuuug 3180

cggcaagggc uaccaccuga ugagcuuccc ucagucugcc ccucacggcg ugguguuucu 3240

gcacgugaca uaugugcccg cucaagagaa gaauuucacc accgcuccag ccaucugcca 3300

cgacggcaaa gcccacuuuc cuagagaagg cguguucgug uccaacggca cccauugguu 3360

cgugacacag cggaacuucu acgagcccca gaucaucacc accgacaaca ccuucguguc 3420

uggcaacugc gacgucguga ucggcauugu gaacaauacc guguacgacc cucugcagcc 3480

cgagcuggac agcuucaaag aggaacugga caaguacuuu aagaaccaca caagccccga 3540

cguggaccug ggcgauauca gcggaaucaa ugccagcguc gugaacaucc agaaagagau 3600

cgaccggcug aacgaggugg ccaagaaucu gaacgagagc cugaucgacc ugcaagaacu 3660

ggggaaguac gagcaguaca ucaaguggcc cugguacauc uggcugggcu uuaucgccgg 3720

acugauugcc aucgugaugg ucacaaucau gcuguguugc augaccagcu gcuguagcug 3780

ccugaagggc uguuguagcu guggcagcug cugcaaguuc gacgaggacg auucugagcc 3840

cgugcugaag ggcgugaaac ugcacuacac augaugacuc gagcugguac ugcaugcacg 3900

caaugcuagc ugccccuuuc ccguccuggg uaccccgagu cucccccgac cucggguccc 3960

agguaugcuc ccaccuccac cugccccacu caccaccucu gcuaguucca gacaccuccc 4020

aagcacgcag caaugcagcu caaaacgcuu agccuagcca cacccccacg ggaaacagca 4080

gugauuaacc uuuagcaaua aacgaaaguu uaacuaagcu auacuaaccc caggguuggu 4140

caauuucgug ccagccacac ccuggagcua gcaaaaaaaa aaaaaaaaaa aaaaaaaaaa 4200

aagcauauga cuaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 4260

aaaaaaaaaa aaaaaaaaaa aa 4282

<210> 17

<211> 1261

<212> RNA

<213> Artificial sequence

<220>

<223> RBL063.3

<400> 17

gggcgaacua guauucuucu gguccccaca gacucagaga gaacccgcca ccauguuugu 60

guuucuugug cugcugccuc uugugucuuc ucagugugug gugagauuuc caaauauuac 120

aaaucugugu ccauuuggag aaguguuuaa ugcaacaaga uuugcaucug uguaugcaug 180

gaauagaaaa agaauuucua auuguguggc ugauuauucu gugcuguaua auagugcuuc 240

uuuuuccaca uuuaaauguu auggaguguc uccaacaaaa uuaaaugauu uauguuuuac 300

aaauguguau gcugauucuu uugugaucag aggugaugaa gugagacaga uugcccccgg 360

acagacagga aaaauugcug auuacaauua caaacugccu gaugauuuua caggaugugu 420

gauugcuugg aauucuaaua auuuagauuc uaaaguggga ggaaauuaca auuaucugua 480

cagacuguuu agaaaaucaa aucugaaacc uuuugaaaga gauauuucaa cagaaauuua 540

ucaggcugga ucaacaccuu guaauggagu ggaaggauuu aauuguuauu uuccauuaca 600

gagcuaugga uuucagccaa ccaauggugu gggauaucag ccauauagag ugguggugcu 660

gucuuuugaa cugcugcaug caccugcaac agugugugga ccuaaaggcu cccccggcuc 720

cggcuccgga ucugguuaua uuccugaagc uccaagagau gggcaagcuu acguucguaa 780

agauggcgaa uggguauuac uuucuaccuu uuuaggccgg ucccuggagg ugcuguucca 840

gggccccggc ugaugacucg agcugguacu gcaugcacgc aaugcuagcu gccccuuucc 900

cguccugggu accccgaguc ucccccgacc ucggguccca gguaugcucc caccuccacc 960

ugccccacuc accaccucug cuaguuccag acaccuccca agcacgcagc aaugcagcuc 1020

aaaacgcuua gccuagccac acccccacgg gaaacagcag ugauuaaccu uuagcaauaa 1080

acgaaaguuu aacuaagcua uacuaacccc aggguugguc aauuucgugc cagccacacc 1140

cuggagcuag caaaaaaaaa aaaaaaaaaa aaaaaaaaaa agcauaugac uaaaaaaaaa 1200

aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 1260

a 1261

<210> 18

<211> 250

<212> PRT

<213> Artificial sequence

<220>

<223> S protein RBD fusion

<400> 18

Val Arg Phe Pro Asn Ile Thr Asn Leu Cys Pro Phe Gly Glu Val Phe

1 5 10 15

Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala Trp Asn Arg Lys Arg Ile

20 25 30

Ser Asn Cys Val Ala Asp Tyr Ser Val Leu Tyr Asn Ser Ala Ser Phe

35 40 45

Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro Thr Lys Leu Asn Asp Leu

50 55 60

Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe Val Ile Arg Gly Asp Glu

65 70 75 80

Val Arg Gln Ile Ala Pro Gly Gln Thr Gly Lys Ile Ala Asp Tyr Asn

85 90 95

Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys Val Ile Ala Trp Asn Ser

100 105 110

Asn Asn Leu Asp Ser Lys Val Gly Gly Asn Tyr Asn Tyr Leu Tyr Arg

115 120 125

Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe Glu Arg Asp Ile Ser Thr

130 135 140

Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys Asn Gly Val Glu Gly Phe

145 150 155 160

Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly Phe Gln Pro Thr Asn Gly

165 170 175

Val Gly Tyr Gln Pro Tyr Arg Val Val Val Leu Ser Phe Glu Leu Leu

180 185 190

His Ala Pro Ala Thr Val Cys Gly Pro Lys Gly Ser Pro Gly Ser Gly

195 200 205

Ser Gly Ser Gly Tyr Ile Pro Glu Ala Pro Arg Asp Gly Gln Ala Tyr

210 215 220

Val Arg Lys Asp Gly Glu Trp Val Leu Leu Ser Thr Phe Leu Gly Arg

225 230 235 240

Ser Leu Glu Val Leu Phe Gln Gly Pro Gly

245 250

<210> 19

<211> 4283

<212> RNA

<213> Artificial sequence

<220>

<223> RBP020.1

<400> 19

agaauaaacu aguauucuuc ugguccccac agacucagag agaacccgcc accauguuug 60

uguuucuugu gcugcugccu cuugugucuu cucagugugu gaauuugaca acaagaacac 120

agcugccacc agcuuauaca aauucuuuua ccagaggagu guauuauccu gauaaagugu 180

uuagaucuuc ugugcugcac agcacacagg accuguuucu gccauuuuuu agcaauguga 240

caugguuuca ugcaauucau gugucuggaa caaauggaac aaaaagauuu gauaauccug 300

ugcugccuuu uaaugaugga guguauuuug cuucaacaga aaagucaaau auuauuagag 360

gauggauuuu uggaacaaca cuggauucua aaacacaguc ucugcugauu gugaauaaug 420

caacaaaugu ggugauuaaa gugugugaau uucaguuuug uaaugauccu uuucugggag 480

uguauuauca caaaaauaau aaaucuugga uggaaucuga auuuagagug uauuccucug 540

caaauaauug uacauuugaa uaugugucuc agccuuuucu gauggaucug gaaggaaaac 600

agggcaauuu uaaaaaucug agagaauuug uguuuaaaaa uauugaugga uauuuuaaaa 660

uuuauucuaa acacacacca auuaauuuag ugagagaucu gccucaggga uuuucugcuc 720

uggaaccucu gguggaucug ccaauuggca uuaauauuac aagauuucag acacugcugg 780

cucugcacag aucuuaucug acaccuggag auucuucuuc uggauggaca gccggagcug 840

cagcuuauua ugugggcuau cugcagccaa gaacauuucu gcugaaauau aaugaaaaug 900

gaacaauuac agaugcugug gauugugcuc uggauccucu gucugaaaca aaauguacau 960

uaaaaucuuu uacaguggaa aaaggcauuu aucagacauc uaauuuuaga gugcagccaa 1020

cagaaucuau ugugagauuu ccaaauauua caaaucugug uccauuugga gaaguguuua 1080

augcaacaag auuugcaucu guguaugcau ggaauagaaa aagaauuucu aauugugugg 1140

cugauuauuc ugugcuguau aauagugcuu cuuuuuccac auuuaaaugu uauggagugu 1200

cuccaacaaa auuaaaugau uuauguuuua caaaugugua ugcugauucu uuugugauca 1260

gaggugauga agugagacag auugcccccg gacagacagg aaaaauugcu gauuacaauu 1320

acaaacugcc ugaugauuuu acaggaugug ugauugcuug gaauucuaau aauuuagauu 1380

cuaaaguggg aggaaauuac aauuaucugu acagacuguu uagaaaauca aaucugaaac 1440

cuuuugaaag agauauuuca acagaaauuu aucaggcugg aucaacaccu uguaauggag 1500

uggaaggauu uaauuguuau uuuccauuac agagcuaugg auuucagcca accaauggug 1560

ugggauauca gccauauaga gugguggugc ugucuuuuga acugcugcau gcaccugcaa 1620

cagugugugg accuaaaaaa ucuacaaauu uagugaaaaa uaaaugugug aauuuuaauu 1680

uuaauggauu aacaggaaca ggagugcuga cagaaucuaa uaaaaaauuu cugccuuuuc 1740

agcaguuugg cagagauauu gcagauacca cagaugcagu gagagauccu cagacauuag 1800

aaauucugga uauuacaccu uguucuuuug gggguguguc ugugauuaca ccuggaacaa 1860

auacaucuaa ucagguggcu gugcuguauc aggaugugaa uuguacagaa gugccagugg 1920

caauucaugc agaucagcug acaccaacau ggagagugua uucuacagga ucuaaugugu 1980

uucagacaag agcaggaugu cugauuggag cagaacaugu gaauaauucu uaugaaugug 2040

auauuccaau uggagcaggc auuugugcau cuuaucagac acagacaaau uccccaagga 2100

gagcaagauc uguggcaucu cagucuauua uugcauacac caugucucug ggagcagaaa 2160

auucuguggc auauucuaau aauucuauug cuauuccaac aaauuuuacc auuucuguga 2220

caacagaaau uuuaccugug ucuaugacaa aaacaucugu ggauuguacc auguacauuu 2280

guggagauuc uacagaaugu ucuaaucugc ugcugcagua uggaucuuuu uguacacagc 2340

ugaauagagc uuuaacagga auugcugugg aacaggauaa aaauacacag gaaguguuug 2400

cucaggugaa acagauuuac aaaacaccac caauuaaaga uuuuggagga uuuaauuuua 2460

gccagauucu gccugauccu ucuaaaccuu cuaaaagauc uuuuauugaa gaucugcugu 2520

uuaauaaagu gacacuggca gaugcaggau uuauuaaaca guauggagau ugccugggug 2580

auauugcugc aagagaucug auuugugcuc agaaauuuaa uggacugaca gugcugccuc 2640

cucugcugac agaugaaaug auugcucagu acacaucugc uuuacuggcu ggaacaauua 2700

caagcggaug gacauuugga gcuggagcug cucugcagau uccuuuugca augcagaugg 2760

cuuacagauu uaauggaauu ggagugacac agaauguguu auaugaaaau cagaaacuga 2820

uugcaaauca guuuaauucu gcaauuggca aaauucagga uucucugucu ucuacagcuu 2880

cugcucuggg aaaacugcag gaugugguga aucagaaugc acaggcacug aauacucugg 2940

ugaaacagcu gucuagcaau uuuggggcaa uuucuucugu gcugaaugau auucugucua 3000

gacuggaucc uccugaagcu gaagugcaga uugauagacu gaucacagga agacugcagu 3060

cucugcagac uuaugugaca cagcagcuga uuagagcugc ugaaauuaga gcuucugcua 3120

aucuggcugc uacaaaaaug ucugaaugug ugcugggaca gucaaaaaga guggauuuuu 3180

guggaaaagg auaucaucug augucuuuuc cacagucugc uccacaugga gugguguuuu 3240

uacaugugac auaugugcca gcacaggaaa agaauuuuac cacagcacca gcaauuuguc 3300

augauggaaa agcacauuuu ccaagagaag gaguguuugu gucuaaugga acacauuggu 3360

uugugacaca gagaaauuuu uaugaaccuc agauuauuac aacagauaau acauuugugu 3420

caggaaauug ugauguggug auuggaauug ugaauaauac aguguaugau ccacugcagc 3480

cagaacugga uucuuuuaaa gaagaacugg auaaauauuu uaaaaaucac acaucuccug 3540

auguggauuu aggagauauu ucuggaauca augcaucugu ggugaauauu cagaaagaaa 3600

uugauagacu gaaugaagug gccaaaaauc ugaaugaauc ucugauugau cugcaggaac 3660

uuggaaaaua ugaacaguac auuaaauggc cuugguacau uuggcuugga uuuauugcag 3720

gauuaauugc aauugugaug gugacaauua uguuauguug uaugacauca uguuguucuu 3780

guuuaaaagg auguuguucu uguggaagcu guuguaaauu ugaugaagau gauucugaac 3840

cuguguuaaa aggagugaaa uugcauuaca caugaugacu cgagcuggua cugcaugcac 3900

gcaaugcuag cugccccuuu cccguccugg guaccccgag ucucccccga ccucgggucc 3960

cagguaugcu cccaccucca ccugccccac ucaccaccuc ugcuaguucc agacaccucc 4020

caagcacgca gcaaugcagc ucaaaacgcu uagccuagcc acacccccac gggaaacagc 4080

agugauuaac cuuuagcaau aaacgaaagu uuaacuaagc uauacuaacc ccaggguugg 4140

ucaauuucgu gccagccaca cccuggagcu agcaaaaaaa aaaaaaaaaa aaaaaaaaaa 4200

aaagcauaug acuaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 4260

aaaaaaaaaa aaaaaaaaaa aaa 4283

<210> 20

<211> 4283

<212> RNA

<213> Artificial sequence

<220>

<223> RBP020.2

<400> 20

agaauaaacu aguauucuuc ugguccccac agacucagag agaacccgcc accauguucg 60

uguuccuggu gcugcugccu cuggugucca gccagugugu gaaccugacc accagaacac 120

agcugccucc agccuacacc aacagcuuua ccagaggcgu guacuacccc gacaaggugu 180

ucagauccag cgugcugcac ucuacccagg accuguuccu gccuuucuuc agcaacguga 240

ccugguucca cgccauccac guguccggca ccaauggcac caagagauuc gacaaccccg 300

ugcugcccuu caacgacggg guguacuuug ccagcaccga gaaguccaac aucaucagag 360

gcuggaucuu cggcaccaca cuggacagca agacccagag ccugcugauc gugaacaacg 420

ccaccaacgu ggucaucaaa gugugcgagu uccaguucug caacgacccc uuccugggcg 480

ucuacuacca caagaacaac aagagcugga uggaaagcga guuccgggug uacagcagcg 540

ccaacaacug caccuucgag uacguguccc agccuuuccu gauggaccug gaaggcaagc 600

agggcaacuu caagaaccug cgcgaguucg uguuuaagaa caucgacggc uacuucaaga 660

ucuacagcaa gcacaccccu aucaaccucg ugcgggaucu gccucagggc uucucugcuc 720

uggaaccccu gguggaucug cccaucggca ucaacaucac ccgguuucag acacugcugg 780

cccugcacag aagcuaccug acaccuggcg auagcagcag cggauggaca gcuggugccg 840

ccgcuuacua ugugggcuac cugcagccua gaaccuuccu gcugaaguac aacgagaacg 900

gcaccaucac cgacgccgug gauugugcuc uggauccucu gagcgagaca aagugcaccc 960

ugaaguccuu caccguggaa aagggcaucu accagaccag caacuuccgg gugcagccca 1020

ccgaauccau cgugcgguuc cccaauauca ccaaucugug ccccuucggc gagguguuca 1080

augccaccag auucgccucu guguacgccu ggaaccggaa gcggaucagc aauugcgugg 1140

ccgacuacuc cgugcuguac aacuccgcca gcuucagcac cuucaagugc uacggcgugu 1200

ccccuaccaa gcugaacgac cugugcuuca caaacgugua cgccgacagc uucgugaucc 1260

ggggagauga agugcggcag auugccccug gacagacagg caagaucgcc gacuacaacu 1320

acaagcugcc cgacgacuuc accggcugug ugauugccug gaacagcaac aaccuggacu 1380

ccaaagucgg cggcaacuac aauuaccugu accggcuguu ccggaagucc aaucugaagc 1440

ccuucgagcg ggacaucucc accgagaucu aucaggccgg cagcaccccu uguaacggcg 1500

uggaaggcuu caacugcuac uucccacugc aguccuacgg cuuucagccc acaaauggcg 1560

ugggcuauca gcccuacaga gugguggugc ugagcuucga acugcugcau gccccugcca 1620

cagugugcgg cccuaagaaa agcaccaauc ucgugaagaa caaaugcgug aacuucaacu 1680

ucaacggccu gaccggcacc ggcgugcuga cagagagcaa caagaaguuc cugccauucc 1740

agcaguuugg ccgggauauc gccgauacca cagacgccgu uagagauccc cagacacugg 1800

aaauccugga caucaccccu ugcagcuucg gcggaguguc ugugaucacc ccuggcacca 1860

acaccagcaa ucagguggca gugcuguacc aggacgugaa cuguaccgaa gugcccgugg 1920

ccauucacgc cgaucagcug acaccuacau ggcgggugua cuccaccggc agcaaugugu 1980

uucagaccag agccggcugu cugaucggag ccgagcacgu gaacaauagc uacgagugcg 2040

acauccccau cggcgcugga aucugcgcca gcuaccagac acagacaaac agcccucgga 2100

gagccagaag cguggccagc cagagcauca uugccuacac aaugucucug ggcgccgaga 2160

acagcguggc cuacuccaac aacucuaucg cuauccccac caacuucacc aucagcguga 2220

ccacagagau ccugccugug uccaugacca agaccagcgu ggacugcacc auguacaucu 2280

gcggcgauuc caccgagugc uccaaccugc ugcugcagua cggcagcuuc ugcacccagc 2340

ugaauagagc ccugacaggg aucgccgugg aacaggacaa gaacacccaa gagguguucg 2400

cccaagugaa gcagaucuac aagaccccuc cuaucaagga cuucggcggc uucaauuuca 2460

gccagauucu gcccgauccu agcaagccca gcaagcggag cuucaucgag gaccugcugu 2520

ucaacaaagu gacacuggcc gacgccggcu ucaucaagca guauggcgau ugucugggcg 2580

acauugccgc cagggaucug auuugcgccc agaaguuuaa cggacugaca gugcugccuc 2640

cucugcugac cgaugagaug aucgcccagu acacaucugc ccugcuggcc ggcacaauca 2700

caagcggcug gacauuugga gcaggcgccg cucugcagau ccccuuugcu augcagaugg 2760

ccuaccgguu caacggcauc ggagugaccc agaaugugcu guacgagaac cagaagcuga 2820

ucgccaacca guucaacagc gccaucggca agauccagga cagccugagc agcacagcaa 2880

gcgcccuggg aaagcugcag gacgugguca accagaaugc ccaggcacug aacacccugg 2940

ucaagcagcu guccuccaac uucggcgcca ucagcucugu gcugaacgau auccugagca 3000

gacuggaccc uccugaggcc gaggugcaga ucgacagacu gaucacaggc agacugcaga 3060

gccuccagac auacgugacc cagcagcuga ucagagccgc cgagauuaga gccucugcca 3120

aucuggccgc caccaagaug ucugagugug ugcugggcca gagcaagaga guggacuuuu 3180

gcggcaaggg cuaccaccug augagcuucc cucagucugc cccucacggc gugguguuuc 3240

ugcacgugac auaugugccc gcucaagaga agaauuucac caccgcucca gccaucugcc 3300

acgacggcaa agcccacuuu ccuagagaag gcguguucgu guccaacggc acccauuggu 3360

ucgugacaca gcggaacuuc uacgagcccc agaucaucac caccgacaac accuucgugu 3420

cuggcaacug cgacgucgug aucggcauug ugaacaauac cguguacgac ccucugcagc 3480

ccgagcugga cagcuucaaa gaggaacugg acaaguacuu uaagaaccac acaagccccg 3540

acguggaccu gggcgauauc agcggaauca augccagcgu cgugaacauc cagaaagaga 3600

ucgaccggcu gaacgaggug gccaagaauc ugaacgagag ccugaucgac cugcaagaac 3660

uggggaagua cgagcaguac aucaaguggc ccugguacau cuggcugggc uuuaucgccg 3720

gacugauugc caucgugaug gucacaauca ugcuguguug caugaccagc ugcuguagcu 3780

gccugaaggg cuguuguagc uguggcagcu gcugcaaguu cgacgaggac gauucugagc 3840

ccgugcugaa gggcgugaaa cugcacuaca caugaugacu cgagcuggua cugcaugcac 3900

gcaaugcuag cugccccuuu cccguccugg guaccccgag ucucccccga ccucgggucc 3960

cagguaugcu cccaccucca ccugccccac ucaccaccuc ugcuaguucc agacaccucc 4020

caagcacgca gcaaugcagc ucaaaacgcu uagccuagcc acacccccac gggaaacagc 4080

agugauuaac cuuuagcaau aaacgaaagu uuaacuaagc uauacuaacc ccaggguugg 4140

ucaauuucgu gccagccaca cccuggagcu agcaaaaaaa aaaaaaaaaa aaaaaaaaaa 4200

aaagcauaug acuaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 4260

aaaaaaaaaa aaaaaaaaaa aaa 4283

<210> 21

<211> 1262

<212> RNA

<213> Artificial sequence

<220>

<223> RBP020.3

<400> 21

agaauaaacu aguauucuuc ugguccccac agacucagag agaacccgcc accauguuug 60

uguuucuugu gcugcugccu cuugugucuu cucagugugu ggugagauuu ccaaauauua 120

caaaucugug uccauuugga gaaguguuua augcaacaag auuugcaucu guguaugcau 180

ggaauagaaa aagaauuucu aauugugugg cugauuauuc ugugcuguau aauagugcuu 240

cuuuuuccac auuuaaaugu uauggagugu cuccaacaaa auuaaaugau uuauguuuua 300

caaaugugua ugcugauucu uuugugauca gaggugauga agugagacag auugcccccg 360

gacagacagg aaaaauugcu gauuacaauu acaaacugcc ugaugauuuu acaggaugug 420

ugauugcuug gaauucuaau aauuuagauu cuaaaguggg aggaaauuac aauuaucugu 480

acagacuguu uagaaaauca aaucugaaac cuuuugaaag agauauuuca acagaaauuu 540

aucaggcugg aucaacaccu uguaauggag uggaaggauu uaauuguuau uuuccauuac 600

agagcuaugg auuucagcca accaauggug ugggauauca gccauauaga gugguggugc 660

ugucuuuuga acugcugcau gcaccugcaa cagugugugg accuaaaggc ucccccggcu 720

ccggcuccgg aucugguuau auuccugaag cuccaagaga ugggcaagcu uacguucgua 780

aagauggcga auggguauua cuuucuaccu uuuuaggccg gucccuggag gugcuguucc 840

agggccccgg cugaugacuc gagcugguac ugcaugcacg caaugcuagc ugccccuuuc 900

ccguccuggg uaccccgagu cucccccgac cucggguccc agguaugcuc ccaccuccac 960

cugccccacu caccaccucu gcuaguucca gacaccuccc aagcacgcag caaugcagcu 1020

caaaacgcuu agccuagcca cacccccacg ggaaacagca gugauuaacc uuuagcaaua 1080

aacgaaaguu uaacuaagcu auacuaaccc caggguuggu caauuucgug ccagccacac 1140

ccuggagcua gcaaaaaaaa aaaaaaaaaa aaaaaaaaaa aagcauauga cuaaaaaaaa 1200

aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 1260

aa 1262

<210> 22

<211> 1879

<212> PRT

<213> Artificial sequence

<220>

<223> viral proteins

<400> 22

Met Glu Lys Val His Val Asp Ile Glu Glu Asp Ser Pro Phe Leu Arg

1 5 10 15

Ala Leu Gln Arg Ser Phe Pro Gln Phe Glu Val Glu Ala Lys Gln Val

20 25 30

Thr Asp Asn Asp His Ala Asn Ala Arg Ala Phe Ser His Leu Ala Ser

35 40 45

Lys Leu Ile Glu Thr Glu Val Asp Pro Ser Asp Thr Ile Leu Asp Ile

50 55 60

Gly Ser Ala Pro Ala Arg Arg Met Tyr Ser Lys His Lys Tyr His Cys

65 70 75 80

Ile Cys Pro Met Arg Cys Ala Glu Asp Pro Asp Arg Leu Tyr Lys Tyr

85 90 95

Ala Thr Lys Leu Lys Lys Asn Cys Lys Glu Ile Thr Asp Lys Glu Leu

100 105 110

Asp Lys Lys Met Lys Glu Leu Ala Ala Val Met Ser Asp Pro Asp Leu

115 120 125

Glu Thr Glu Thr Met Cys Leu His Asp Asp Glu Ser Cys Arg Tyr Glu

130 135 140

Gly Gln Val Ala Val Tyr Gln Asp Val Tyr Ala Val Asp Gly Pro Thr

145 150 155 160

Ser Leu Tyr His Gln Ala Asn Lys Gly Val Arg Val Ala Tyr Trp Ile

165 170 175

Gly Phe Asp Thr Thr Pro Phe Met Phe Lys Asn Leu Ala Gly Ala Tyr

180 185 190

Pro Ser Tyr Ser Thr Asn Trp Ala Asp Glu Thr Val Leu Thr Ala Arg

195 200 205

Asn Ile Gly Leu Cys Ser Ser Asp Val Met Glu Arg Ser Arg Arg Gly

210 215 220

Met Ser Ile Leu Arg Lys Lys Tyr Leu Lys Pro Ser Asn Asn Val Leu

225 230 235 240

Phe Ser Val Gly Ser Thr Ile Tyr His Glu Lys Arg Asp Leu Leu Arg

245 250 255

Ser Trp His Leu Pro Ser Val Phe His Leu Arg Gly Lys Gln Asn Tyr

260 265 270

Thr Cys Arg Cys Glu Thr Ile Val Ser Cys Asp Gly Tyr Val Val Lys

275 280 285

Arg Ile Ala Ile Ser Pro Gly Leu Tyr Gly Lys Pro Ser Gly Tyr Ala

290 295 300

Ala Thr Met His Arg Glu Gly Phe Leu Cys Cys Lys Val Thr Asp Thr

305 310 315 320

Leu Asn Gly Glu Arg Val Ser Phe Pro Val Cys Thr Tyr Val Pro Ala

325 330 335

Thr Leu Cys Asp Gln Met Thr Gly Ile Leu Ala Thr Asp Val Ser Ala

340 345 350

Asp Asp Ala Gln Lys Leu Leu Val Gly Leu Asn Gln Arg Ile Val Val

355 360 365

Asn Gly Arg Thr Gln Arg Asn Thr Asn Thr Met Lys Asn Tyr Leu Leu

370 375 380

Pro Val Val Ala Gln Ala Phe Ala Arg Trp Ala Lys Glu Tyr Lys Glu

385 390 395 400

Asp Gln Glu Asp Glu Arg Pro Leu Gly Leu Arg Asp Arg Gln Leu Val

405 410 415

Met Gly Cys Cys Trp Ala Phe Arg Arg His Lys Ile Thr Ser Ile Tyr

420 425 430

Lys Arg Pro Asp Thr Gln Thr Ile Ile Lys Val Asn Ser Asp Phe His

435 440 445

Ser Phe Val Leu Pro Arg Ile Gly Ser Asn Thr Leu Glu Ile Gly Leu

450 455 460

Arg Thr Arg Ile Arg Lys Met Leu Glu Glu His Lys Glu Pro Ser Pro

465 470 475 480

Leu Ile Thr Ala Glu Asp Val Gln Glu Ala Lys Cys Ala Ala Asp Glu

485 490 495

Ala Lys Glu Val Arg Glu Ala Glu Glu Leu Arg Ala Ala Leu Pro Pro

500 505 510

Leu Ala Ala Asp Val Glu Glu Pro Thr Leu Glu Ala Asp Val Asp Leu

515 520 525

Met Leu Gln Glu Ala Gly Ala Gly Ser Val Glu Thr Pro Arg Gly Leu

530 535 540

Ile Lys Val Thr Ser Tyr Ala Gly Glu Asp Lys Ile Gly Ser Tyr Ala

545 550 555 560

Val Leu Ser Pro Gln Ala Val Leu Lys Ser Glu Lys Leu Ser Cys Ile

565 570 575

His Pro Leu Ala Glu Gln Val Ile Val Ile Thr His Ser Gly Arg Lys

580 585 590

Gly Arg Tyr Ala Val Glu Pro Tyr His Gly Lys Val Val Val Pro Glu

595 600 605

Gly His Ala Ile Pro Val Gln Asp Phe Gln Ala Leu Ser Glu Ser Ala

610 615 620

Thr Ile Val Tyr Asn Glu Arg Glu Phe Val Asn Arg Tyr Leu His His

625 630 635 640

Ile Ala Thr His Gly Gly Ala Leu Asn Thr Asp Glu Glu Tyr Tyr Lys

645 650 655

Thr Val Lys Pro Ser Glu His Asp Gly Glu Tyr Leu Tyr Asp Ile Asp

660 665 670

Arg Lys Gln Cys Val Lys Lys Glu Leu Val Thr Gly Leu Gly Leu Thr

675 680 685

Gly Glu Leu Val Asp Pro Pro Phe His Glu Phe Ala Tyr Glu Ser Leu

690 695 700

Arg Thr Arg Pro Ala Ala Pro Tyr Gln Val Pro Thr Ile Gly Val Tyr

705 710 715 720

Gly Val Pro Gly Ser Gly Lys Ser Gly Ile Ile Lys Ser Ala Val Thr

725 730 735

Lys Lys Asp Leu Val Val Ser Ala Lys Lys Glu Asn Cys Ala Glu Ile

740 745 750

Ile Arg Asp Val Lys Lys Met Lys Gly Leu Asp Val Asn Ala Arg Thr

755 760 765

Val Asp Ser Val Leu Leu Asn Gly Cys Lys His Pro Val Glu Thr Leu

770 775 780

Tyr Ile Asp Glu Ala Phe Ala Cys His Ala Gly Thr Leu Arg Ala Leu

785 790 795 800

Ile Ala Ile Ile Arg Pro Lys Lys Ala Val Leu Cys Gly Asp Pro Lys

805 810 815

Gln Cys Gly Phe Phe Asn Met Met Cys Leu Lys Val His Phe Asn His

820 825 830

Glu Ile Cys Thr Gln Val Phe His Lys Ser Ile Ser Arg Arg Cys Thr

835 840 845

Lys Ser Val Thr Ser Val Val Ser Thr Leu Phe Tyr Asp Lys Lys Met

850 855 860

Arg Thr Thr Asn Pro Lys Glu Thr Lys Ile Val Ile Asp Thr Thr Gly

865 870 875 880

Ser Thr Lys Pro Lys Gln Asp Asp Leu Ile Leu Thr Cys Phe Arg Gly

885 890 895

Trp Val Lys Gln Leu Gln Ile Asp Tyr Lys Gly Asn Glu Ile Met Thr

900 905 910

Ala Ala Ala Ser Gln Gly Leu Thr Arg Lys Gly Val Tyr Ala Val Arg

915 920 925

Tyr Lys Val Asn Glu Asn Pro Leu Tyr Ala Pro Thr Ser Glu His Val

930 935 940

Asn Val Leu Leu Thr Arg Thr Glu Asp Arg Ile Val Trp Lys Thr Leu

945 950 955 960

Ala Gly Asp Pro Trp Ile Lys Thr Leu Thr Ala Lys Tyr Pro Gly Asn

965 970 975

Phe Thr Ala Thr Ile Glu Glu Trp Gln Ala Glu His Asp Ala Ile Met

980 985 990

Arg His Ile Leu Glu Arg Pro Asp Pro Thr Asp Val Phe Gln Asn Lys

995 1000 1005

Ala Asn Val Cys Trp Ala Lys Ala Leu Val Pro Val Leu Lys Thr

1010 1015 1020

Ala Gly Ile Asp Met Thr Thr Glu Gln Trp Asn Thr Val Asp Tyr

1025 1030 1035

Phe Glu Thr Asp Lys Ala His Ser Ala Glu Ile Val Leu Asn Gln

1040 1045 1050

Leu Cys Val Arg Phe Phe Gly Leu Asp Leu Asp Ser Gly Leu Phe

1055 1060 1065

Ser Ala Pro Thr Val Pro Leu Ser Ile Arg Asn Asn His Trp Asp

1070 1075 1080

Asn Ser Pro Ser Pro Asn Met Tyr Gly Leu Asn Lys Glu Val Val

1085 1090 1095

Arg Gln Leu Ser Arg Arg Tyr Pro Gln Leu Pro Arg Ala Val Ala

1100 1105 1110

Thr Gly Arg Val Tyr Asp Met Asn Thr Gly Thr Leu Arg Asn Tyr

1115 1120 1125

Asp Pro Arg Ile Asn Leu Val Pro Val Asn Arg Arg Leu Pro His

1130 1135 1140

Ala Leu Val Leu His His Asn Glu His Pro Gln Ser Asp Phe Ser

1145 1150 1155

Ser Phe Val Ser Lys Leu Lys Gly Arg Thr Val Leu Val Val Gly

1160 1165 1170

Glu Lys Leu Ser Val Pro Gly Lys Met Val Asp Trp Leu Ser Asp

1175 1180 1185

Arg Pro Glu Ala Thr Phe Arg Ala Arg Leu Asp Leu Gly Ile Pro

1190 1195 1200

Gly Asp Val Pro Lys Tyr Asp Ile Ile Phe Val Asn Val Arg Thr

1205 1210 1215

Pro Tyr Lys Tyr His His Tyr Gln Gln Cys Glu Asp His Ala Ile

1220 1225 1230

Lys Leu Ser Met Leu Thr Lys Lys Ala Cys Leu His Leu Asn Pro

1235 1240 1245

Gly Gly Thr Cys Val Ser Ile Gly Tyr Gly Tyr Ala Asp Arg Ala

1250 1255 1260

Ser Glu Ser Ile Ile Gly Ala Ile Ala Arg Gln Phe Lys Phe Ser

1265 1270 1275

Arg Val Cys Lys Pro Lys Ser Ser Leu Glu Glu Thr Glu Val Leu

1280 1285 1290

Phe Val Phe Ile Gly Tyr Asp Arg Lys Ala Arg Thr His Asn Pro

1295 1300 1305

Tyr Lys Leu Ser Ser Thr Leu Thr Asn Ile Tyr Thr Gly Ser Arg

1310 1315 1320

Leu His Glu Ala Gly Cys Ala Pro Ser Tyr His Val Val Arg Gly

1325 1330 1335

Asp Ile Ala Thr Ala Thr Glu Gly Val Ile Ile Asn Ala Ala Asn

1340 1345 1350

Ser Lys Gly Gln Pro Gly Gly Gly Val Cys Gly Ala Leu Tyr Lys

1355 1360 1365

Lys Phe Pro Glu Ser Phe Asp Leu Gln Pro Ile Glu Val Gly Lys

1370 1375 1380

Ala Arg Leu Val Lys Gly Ala Ala Lys His Ile Ile His Ala Val

1385 1390 1395

Gly Pro Asn Phe Asn Lys Val Ser Glu Val Glu Gly Asp Lys Gln

1400 1405 1410

Leu Ala Glu Ala Tyr Glu Ser Ile Ala Lys Ile Val Asn Asp Asn

1415 1420 1425

Asn Tyr Lys Ser Val Ala Ile Pro Leu Leu Ser Thr Gly Ile Phe

1430 1435 1440

Ser Gly Asn Lys Asp Arg Leu Thr Gln Ser Leu Asn His Leu Leu

1445 1450 1455

Thr Ala Leu Asp Thr Thr Asp Ala Asp Val Ala Ile Tyr Cys Arg

1460 1465 1470

Asp Lys Lys Trp Glu Met Thr Leu Lys Glu Ala Val Ala Arg Arg

1475 1480 1485

Glu Ala Val Glu Glu Ile Cys Ile Ser Asp Asp Ser Ser Val Thr

1490 1495 1500

Glu Pro Asp Ala Glu Leu Val Arg Val His Pro Lys Ser Ser Leu

1505 1510 1515

Ala Gly Arg Lys Gly Tyr Ser Thr Ser Asp Gly Lys Thr Phe Ser

1520 1525 1530

Tyr Leu Glu Gly Thr Lys Phe His Gln Ala Ala Lys Asp Ile Ala

1535 1540 1545

Glu Ile Asn Ala Met Trp Pro Val Ala Thr Glu Ala Asn Glu Gln

1550 1555 1560

Val Cys Met Tyr Ile Leu Gly Glu Ser Met Ser Ser Ile Arg Ser

1565 1570 1575

Lys Cys Pro Val Glu Glu Ser Glu Ala Ser Thr Pro Pro Ser Thr

1580 1585 1590

Leu Pro Cys Leu Cys Ile His Ala Met Thr Pro Glu Arg Val Gln

1595 1600 1605

Arg Leu Lys Ala Ser Arg Pro Glu Gln Ile Thr Val Cys Ser Ser

1610 1615 1620

Phe Pro Leu Pro Lys Tyr Arg Ile Thr Gly Val Gln Lys Ile Gln

1625 1630 1635

Cys Ser Gln Pro Ile Leu Phe Ser Pro Lys Val Pro Ala Tyr Ile

1640 1645 1650

His Pro Arg Lys Tyr Leu Val Glu Thr Pro Pro Val Asp Glu Thr

1655 1660 1665

Pro Glu Pro Ser Ala Glu Asn Gln Ser Thr Glu Gly Thr Pro Glu

1670 1675 1680

Gln Pro Pro Leu Ile Thr Glu Asp Glu Thr Arg Thr Arg Thr Pro

1685 1690 1695

Glu Pro Ile Ile Ile Glu Glu Glu Glu Glu Asp Ser Ile Ser Leu

1700 1705 1710

Leu Ser Asp Gly Pro Thr His Gln Val Leu Gln Val Glu Ala Asp

1715 1720 1725

Ile His Gly Pro Pro Ser Val Ser Ser Ser Ser Trp Ser Ile Pro

1730 1735 1740

His Ala Ser Asp Phe Asp Val Asp Ser Leu Ser Ile Leu Asp Thr

1745 1750 1755

Leu Glu Gly Ala Ser Val Thr Ser Gly Ala Thr Ser Ala Glu Thr

1760 1765 1770

Asn Ser Tyr Phe Ala Lys Ser Met Glu Phe Leu Ala Arg Pro Val

1775 1780 1785

Pro Ala Pro Arg Thr Val Phe Arg Asn Pro Pro His Pro Ala Pro

1790 1795 1800

Arg Thr Arg Thr Pro Ser Leu Ala Pro Ser Arg Ala Cys Ser Arg

1805 1810 1815

Thr Ser Leu Val Ser Thr Pro Pro Gly Val Asn Arg Val Ile Thr

1820 1825 1830

Arg Glu Glu Leu Glu Ala Leu Thr Pro Ser Arg Thr Pro Ser Arg

1835 1840 1845

Ser Val Ser Arg Thr Ser Leu Val Ser Asn Pro Pro Gly Val Asn

1850 1855 1860

Arg Val Ile Thr Arg Glu Glu Phe Glu Ala Phe Val Ala Gln Gln

1865 1870 1875

Gln

<210> 23

<211> 613

<212> PRT

<213> Artificial sequence

<220>

<223> viral proteins

<400> 23

Arg Phe Asp Ala Gly Ala Tyr Ile Phe Ser Ser Asp Thr Gly Gln Gly

1 5 10 15

His Leu Gln Gln Lys Ser Val Arg Gln Thr Val Leu Ser Glu Val Val

20 25 30

Leu Glu Arg Thr Glu Leu Glu Ile Ser Tyr Ala Pro Arg Leu Asp Gln

35 40 45

Glu Lys Glu Glu Leu Leu Arg Lys Lys Leu Gln Leu Asn Pro Thr Pro

50 55 60

Ala Asn Arg Ser Arg Tyr Gln Ser Arg Lys Val Glu Asn Met Lys Ala

65 70 75 80

Ile Thr Ala Arg Arg Ile Leu Gln Gly Leu Gly His Tyr Leu Lys Ala

85 90 95

Glu Gly Lys Val Glu Cys Tyr Arg Thr Leu His Pro Val Pro Leu Tyr

100 105 110

Ser Ser Ser Val Asn Arg Ala Phe Ser Ser Pro Lys Val Ala Val Glu

115 120 125

Ala Cys Asn Ala Met Leu Lys Glu Asn Phe Pro Thr Val Ala Ser Tyr

130 135 140

Cys Ile Ile Pro Glu Tyr Asp Ala Tyr Leu Asp Met Val Asp Gly Ala

145 150 155 160

Ser Cys Cys Leu Asp Thr Ala Ser Phe Cys Pro Ala Lys Leu Arg Ser

165 170 175

Phe Pro Lys Lys His Ser Tyr Leu Glu Pro Thr Ile Arg Ser Ala Val

180 185 190

Pro Ser Ala Ile Gln Asn Thr Leu Gln Asn Val Leu Ala Ala Ala Thr

195 200 205

Lys Arg Asn Cys Asn Val Thr Gln Met Arg Glu Leu Pro Val Leu Asp

210 215 220

Ser Ala Ala Phe Asn Val Glu Cys Phe Lys Lys Tyr Ala Cys Asn Asn

225 230 235 240

Glu Tyr Trp Glu Thr Phe Lys Glu Asn Pro Ile Arg Leu Thr Glu Glu

245 250 255

Asn Val Val Asn Tyr Ile Thr Lys Leu Lys Gly Pro Lys Ala Ala Ala

260 265 270

Leu Phe Ala Lys Thr His Asn Leu Asn Met Leu Gln Asp Ile Pro Met

275 280 285

Asp Arg Phe Val Met Asp Leu Lys Arg Asp Val Lys Val Thr Pro Gly

290 295 300

Thr Lys His Thr Glu Glu Arg Pro Lys Val Gln Val Ile Gln Ala Ala

305 310 315 320

Asp Pro Leu Ala Thr Ala Tyr Leu Cys Gly Ile His Arg Glu Leu Val

325 330 335

Arg Arg Leu Asn Ala Val Leu Leu Pro Asn Ile His Thr Leu Phe Asp

340 345 350

Met Ser Ala Glu Asp Phe Asp Ala Ile Ile Ala Glu His Phe Gln Pro

355 360 365

Gly Asp Cys Val Leu Glu Thr Asp Ile Ala Ser Phe Asp Lys Ser Glu

370 375 380

Asp Asp Ala Met Ala Leu Thr Ala Leu Met Ile Leu Glu Asp Leu Gly

385 390 395 400

Val Asp Ala Glu Leu Leu Thr Leu Ile Glu Ala Ala Phe Gly Glu Ile

405 410 415

Ser Ser Ile His Leu Pro Thr Lys Thr Lys Phe Lys Phe Gly Ala Met

420 425 430

Met Lys Ser Gly Met Phe Leu Thr Leu Phe Val Asn Thr Val Ile Asn

435 440 445

Ile Val Ile Ala Ser Arg Val Leu Arg Glu Arg Leu Thr Gly Ser Pro

450 455 460

Cys Ala Ala Phe Ile Gly Asp Asp Asn Ile Val Lys Gly Val Lys Ser

465 470 475 480

Asp Lys Leu Met Ala Asp Arg Cys Ala Thr Trp Leu Asn Met Glu Val

485 490 495

Lys Ile Ile Asp Ala Val Val Gly Glu Lys Ala Pro Tyr Phe Cys Gly

500 505 510

Gly Phe Ile Leu Cys Asp Ser Val Thr Gly Thr Ala Cys Arg Val Ala

515 520 525

Asp Pro Leu Lys Arg Leu Phe Lys Leu Gly Lys Pro Leu Ala Ala Asp

530 535 540

Asp Glu His Asp Asp Asp Arg Arg Arg Ala Leu His Glu Glu Ser Thr

545 550 555 560

Arg Trp Asn Arg Val Gly Ile Leu Ser Glu Leu Cys Lys Ala Val Glu

565 570 575

Ser Arg Tyr Glu Thr Val Gly Thr Ser Ile Ile Val Met Ala Met Thr

580 585 590

Thr Leu Ala Ser Ser Val Lys Ser Phe Ser Tyr Leu Arg Gly Ala Pro

595 600 605

Ile Thr Leu Tyr Gly

610

<210> 24

<211> 11917

<212> RNA

<213> Artificial sequence

<220>

<223> RBS004.1

<400> 24

gaugggcggc gcaugagaga agcccagacc aauuaccuac ccaaaaugga gaaaguucac 60

guugacaucg aggaagacag cccauuccuc agagcuuugc agcggagcuu cccgcaguuu 120

gagguagaag ccaagcaggu cacugauaau gaccaugcua augccagagc guuuucgcau 180

cuggcuucaa aacugaucga aacggaggug gacccauccg acacgauccu ugacauugga 240

agugcgcccg cccgcagaau guauucuaag cacaaguauc auuguaucug uccgaugaga 300

ugugcggaag auccggacag auuguauaag uaugcaacua agcugaagaa aaacuguaag 360

gaaauaacug auaaggaauu ggacaagaaa augaaggagc ucgccgccgu caugagcgac 420

ccugaccugg aaacugagac uaugugccuc cacgacgacg agucgugucg cuacgaaggg 480

caagucgcug uuuaccagga uguauacgcg guugacggac cgacaagucu cuaucaccaa 540

gccaauaagg gaguuagagu cgccuacugg auaggcuuug acaccacccc uuuuauguuu 600

aagaacuugg cuggagcaua uccaucauac ucuaccaacu gggccgacga aaccguguua 660

acggcucgua acauaggccu augcagcucu gacguuaugg agcggucacg uagagggaug 720

uccauucuua gaaagaagua uuugaaacca uccaacaaug uucuauucuc uguuggcucg 780

accaucuacc acgaaaagag ggacuuacug aggagcuggc accugccguc uguauuucac 840

uuacguggca agcaaaauua cacaugucgg ugugagacua uaguuaguug cgacggguac 900

gucguuaaaa gaauagcuau caguccaggc cuguauggga agccuucagg cuaugcugcu 960

acgaugcacc gcgagggauu cuugugcugc aaagugacag acacauugaa cggggagagg 1020

gucucuuuuc ccgugugcac guaugugcca gcuacauugu gugaccaaau gacuggcaua 1080

cuggcaacag augucagugc ggacgacgcg caaaaacugc ugguugggcu caaccagcgu 1140

auagucguca acggucgcac ccagagaaac accaauacca ugaaaaauua ccuuuugccc 1200

guaguggccc aggcauuugc uaggugggca aaggaauaua aggaagauca agaagaugaa 1260

aggccacuag gacuacgaga uagacaguua gucauggggu guuguugggc uuuuagaagg 1320

cacaagauaa caucuauuua uaagcgcccg gauacccaaa ccaucaucaa agugaacagc 1380

gauuuccacu cauucgugcu gcccaggaua ggcaguaaca cauuggagau cgggcugaga 1440

acaagaauca ggaaaauguu agaggagcac aaggagccgu caccucucau uaccgccgag 1500

gacguacaag aagcuaagug cgcagccgau gaggcuaagg aggugcguga agccgaggag 1560

uugcgcgcag cucuaccacc uuuggcagcu gauguugagg agcccacucu ggaagccgau 1620

gucgacuuga uguuacaaga ggcuggggcc ggcucagugg agacaccucg uggcuugaua 1680

aagguuacca gcuacgcugg cgaggacaag aucggcucuu acgcugugcu uucuccgcag 1740

gcuguacuca agagugaaaa auuaucuugc auccacccuc ucgcugaaca agucauagug 1800

auaacacacu cuggccgaaa agggcguuau gccguggaac cauaccaugg uaaaguagug 1860

gugccagagg gacaugcaau acccguccag gacuuucaag cucugaguga aagugccacc 1920

auuguguaca acgaacguga guucguaaac agguaccugc accauauugc cacacaugga 1980

ggagcgcuga acacugauga agaauauuac aaaacuguca agcccagcga gcacgacggc 2040

gaauaccugu acgacaucga caggaaacag ugcgucaaga aagagcuagu cacugggcua 2100

gggcucacag gcgagcuggu cgauccuccc uuccaugaau ucgccuacga gagucugaga 2160

acacgaccag ccgcuccuua ccaaguacca accauagggg uguauggcgu gccaggauca 2220

ggcaagucug gcaucauuaa aagcgcaguc accaaaaaag aucuaguggu gagcgccaag 2280

aaagaaaacu gugcagaaau uauaagggac gucaagaaaa ugaaagggcu ggacgucaau 2340

gccagaacug uggacucagu gcucuugaau ggaugcaaac accccguaga gacccuguau 2400

auugacgagg cuuuugcuug ucaugcaggu acucucagag cgcucauagc cauuauaaga 2460

ccuaaaaagg cagugcucug cggagauccc aaacagugcg guuuuuuuaa caugaugugc 2520

cugaaagugc auuuuaacca cgagauuugc acacaagucu uccacaaaag caucucucgc 2580

cguugcacua aaucugugac uucggucguc ucaaccuugu uuuacgacaa aaaaaugaga 2640

acgacgaauc cgaaagagac uaagauugug auugacacua ccggcaguac caaaccuaag 2700

caggacgauc ucauucucac uuguuucaga ggguggguga agcaguugca aauagauuac 2760

aaaggcaacg aaauaaugac ggcagcugcc ucucaagggc ugacccguaa agguguguau 2820

gccguucggu acaaggugaa ugaaaauccu cuguacgcac ccaccucaga acaugugaac 2880

guccuacuga cccgcacgga ggaccgcauc guguggaaaa cacuagccgg cgacccaugg 2940

auaaaaacac ugacugccaa guacccuggg aauuucacug ccacgauaga ggaguggcaa 3000

gcagagcaug augccaucau gaggcacauc uuggagagac cggacccuac cgacgucuuc 3060

cagaauaagg caaacgugug uugggccaag gcuuuagugc cggugcugaa gaccgcuggc 3120

auagacauga ccacugaaca auggaacacu guggauuauu uugaaacgga caaagcucac 3180

ucagcagaga uaguauugaa ccaacuaugc gugagguucu uuggacucga ucuggacucc 3240

ggucuauuuu cugcacccac uguuccguua uccauuagga auaaucacug ggauaacucc 3300

ccgucgccua acauguacgg gcugaauaaa gaaguggucc gucagcucuc ucgcagguac 3360

ccacaacugc cucgggcagu ugccacuggu agagucuaug acaugaacac ugguacacug 3420

cgcaauuaug auccgcgcau aaaccuagua ccuguaaaca gaagacugcc ucaugcuuua 3480

guccuccacc auaaugaaca cccacagagu gacuuuucuu cauucgucag caaauugaag 3540

ggcagaacug uccugguggu cggggaaaag uuguccgucc caggcaaaau gguugacugg 3600

uugucagacc ggccugaggc uaccuucaga gcucggcugg auuuaggcau cccaggugau 3660

gugcccaaau augacauaau auuuguuaau gugaggaccc cauauaaaua ccaucacuau 3720

cagcagugug aagaccaugc cauuaagcua agcauguuga ccaagaaagc augucugcau 3780

cugaaucccg gcggaaccug ugucagcaua gguuaugguu acgcugacag ggccagcgaa 3840

agcaucauug gugcuauagc gcggcaguuc aaguuuuccc gaguaugcaa accgaaaucc 3900

ucacuugagg agacggaagu ucuguuugua uucauugggu acgaucgcaa ggcccguacg 3960

cacaauccuu acaagcuauc aucaaccuug accaacauuu auacagguuc cagacuccac 4020

gaagccggau gugcacccuc auaucaugug gugcgagggg auauugccac ggccaccgaa 4080

ggagugauua uaaaugcugc uaacagcaaa ggacaaccug gcggaggggu gugcggagcg 4140

cuguauaaga aauucccgga aaguuucgau uuacagccga ucgaaguagg aaaagcgcga 4200

cuggucaaag gugcagcuaa acauaucauu caugccguag gaccaaacuu caacaaaguu 4260

ucggagguug aaggugacaa acaguuggca gaggcuuaug aguccaucgc uaagauuguc 4320

aacgauaaca auuacaaguc aguagcgauu ccacuguugu ccaccggcau cuuuuccggg 4380

aacaaagauc gacuaaccca aucauugaac cauuugcuga cagcuuuaga caccacugau 4440

gcagauguag ccauauacug cagggacaag aaaugggaaa ugacucucaa ggaagcagug 4500

gcuaggagag aagcagugga ggagauaugc auauccgacg auucuucagu gacagaaccu 4560

gaugcagagc uggugagggu gcaucccaag aguucuuugg cuggaaggaa gggcuacagc 4620

acaagcgaug gcaaaacuuu cucauauuug gaagggacca aguuucacca ggcggccaag 4680

gauauagcag aaauuaaugc cauguggccc guugcaacgg aggccaauga gcagguaugc 4740

auguauaucc ucggagaaag caugagcagu auuaggucga aaugccccgu cgaggagucg 4800

gaagccucca caccaccuag cacgcugccu ugcuugugca uccaugccau gacuccagaa 4860

agaguacagc gccuaaaagc cucacgucca gaacaaauua cugugugcuc auccuuucca 4920

uugccgaagu auagaaucac uggugugcag aagauccaau gcucccagcc uauauuguuc 4980

ucaccgaaag ugccugcgua uauucaucca aggaaguauc ucguggaaac accaccggua 5040

gacgagacuc cggagccauc ggcagagaac caauccacag aggggacacc ugaacaacca 5100

ccacuuauaa ccgaggauga gaccaggacu agaacgccug agccgaucau caucgaagaa 5160

gaagaagaag auagcauaag uuugcuguca gauggcccga cccaccaggu gcugcaaguc 5220

gaggcagaca uucacgggcc gcccucugua ucuagcucau ccugguccau uccucaugca 5280

uccgacuuug auguggacag uuuauccaua cuugacaccc uggagggagc uagcgugacc 5340

agcggggcaa cgucagccga gacuaacucu uacuucgcaa agaguaugga guuucuggcg 5400

cgaccggugc cugcgccucg aacaguauuc aggaacccuc cacaucccgc uccgcgcaca 5460

agaacaccgu cacuugcacc cagcagggcc ugcuccagaa ccagccuagu uuccaccccg 5520

ccaggcguga auagggugau cacuagagag gagcucgaag cgcuuacccc gucacgcacu 5580

ccuagcaggu cggucuccag aaccagccug gucuccaacc cgccaggcgu aaauagggug 5640

auuacaagag aggaguuuga ggcguucgua gcacaacaac aaugacgguu ugaugcgggu 5700

gcauacaucu uuuccuccga caccggucaa gggcauuuac aacaaaaauc aguaaggcaa 5760

acggugcuau ccgaaguggu guuggagagg accgaauugg agauuucgua ugccccgcgc 5820

cucgaccaag aaaaagaaga auuacuacgc aagaaauuac aguuaaaucc cacaccugcu 5880

aacagaagca gauaccaguc caggaaggug gagaacauga aagccauaac agcuagacgu 5940

auucugcaag gccuagggca uuauuugaag gcagaaggaa aaguggagug cuaccgaacc 6000

cugcauccug uuccuuugua uucaucuagu gugaaccgug ccuuuucaag ccccaagguc 6060

gcaguggaag ccuguaacgc cauguugaaa gagaacuuuc cgacuguggc uucuuacugu 6120

auuauuccag aguacgaugc cuauuuggac augguugacg gagcuucaug cugcuuagac 6180

acugccaguu uuugcccugc aaagcugcgc agcuuuccaa agaaacacuc cuauuuggaa 6240

cccacaauac gaucggcagu gccuucagcg auccagaaca cgcuccagaa cguccuggca 6300

gcugccacaa aaagaaauug caaugucacg caaaugagag aauugcccgu auuggauucg 6360

gcggccuuua auguggaaug cuucaagaaa uaugcgugua auaaugaaua uugggaaacg 6420

uuuaaagaaa accccaucag gcuuacugaa gaaaacgugg uaaauuacau uaccaaauua 6480

aaaggaccaa aagcugcugc ucuuuuugcg aagacacaua auuugaauau guugcaggac 6540

auaccaaugg acagguuugu aauggacuua aagagagacg ugaaagugac uccaggaaca 6600

aaacauacug aagaacggcc caagguacag gugauccagg cugccgaucc gcuagcaaca 6660

gcguaucugu gcggaaucca ccgagagcug guuaggagau uaaaugcggu ccugcuuccg 6720

aacauucaua cacuguuuga uaugucggcu gaagacuuug acgcuauuau agccgagcac 6780

uuccagccug gggauugugu ucuggaaacu gacaucgcgu cguuugauaa aagugaggac 6840

gacgccaugg cucugaccgc guuaaugauu cuggaagacu uaggugugga cgcagagcug 6900

uugacgcuga uugaggcggc uuucggcgaa auuucaucaa uacauuugcc cacuaaaacu 6960

aaauuuaaau ucggagccau gaugaaaucu ggaauguucc ucacacuguu ugugaacaca 7020

gucauuaaca uuguaaucgc aagcagagug uugagagaac ggcuaaccgg aucaccaugu 7080

gcagcauuca uuggagauga caauaucgug aaaggaguca aaucggacaa auuaauggca 7140

gacaggugcg ccaccugguu gaauauggaa gucaagauua uagaugcugu ggugggcgag 7200

aaagcgccuu auuucugugg aggguuuauu uugugugacu ccgugaccgg cacagcgugc 7260

cguguggcag acccccuaaa aaggcuguuu aagcuaggca aaccucuggc agcagacgau 7320

gaacaugaug augacaggag aagggcauug caugaggagu caacacgcug gaaccgagug 7380

gguauucuuu cagagcugug caaggcagua gaaucaaggu augaaaccgu aggaacuucc 7440

aucauaguua uggccaugac uacucuagcu agcaguguua aaucauucag cuaccugaga 7500

ggggccccua uaacucucua cggcuaaccu gaauggacua cgacauaguc uaguccgcca 7560

agacuaguau guuuguguuu cuugugcugc ugccucuugu gucuucucag ugugugaauu 7620

ugacaacaag aacacagcug ccaccagcuu auacaaauuc uuuuaccaga ggaguguauu 7680

auccugauaa aguguuuaga ucuucugugc ugcacagcac acaggaccug uuucugccau 7740

uuuuuagcaa ugugacaugg uuucaugcaa uucauguguc uggaacaaau ggaacaaaaa 7800

gauuugauaa uccugugcug ccuuuuaaug auggagugua uuuugcuuca acagaaaagu 7860

caaauauuau uagaggaugg auuuuuggaa caacacugga uucuaaaaca cagucucugc 7920

ugauugugaa uaaugcaaca aaugugguga uuaaagugug ugaauuucag uuuuguaaug 7980

auccuuuucu gggaguguau uaucacaaaa auaauaaauc uuggauggaa ucugaauuua 8040

gaguguauuc cucugcaaau aauuguacau uugaauaugu gucucagccu uuucugaugg 8100

aucuggaagg aaaacagggc aauuuuaaaa aucugagaga auuuguguuu aaaaauauug 8160

auggauauuu uaaaauuuau ucuaaacaca caccaauuaa uuuagugaga gaucugccuc 8220

agggauuuuc ugcucuggaa ccucuggugg aucugccaau uggcauuaau auuacaagau 8280

uucagacacu gcuggcucug cacagaucuu aucugacacc uggagauucu ucuucuggau 8340

ggacagccgg agcugcagcu uauuaugugg gcuaucugca gccaagaaca uuucugcuga 8400

aauauaauga aaauggaaca auuacagaug cuguggauug ugcucuggau ccucugucug 8460

aaacaaaaug uacauuaaaa ucuuuuacag uggaaaaagg cauuuaucag acaucuaauu 8520

uuagagugca gccaacagaa ucuauuguga gauuuccaaa uauuacaaau cuguguccau 8580

uuggagaagu guuuaaugca acaagauuug caucugugua ugcauggaau agaaaaagaa 8640

uuucuaauug uguggcugau uauucugugc uguauaauag ugcuucuuuu uccacauuua 8700

aauguuaugg agugucucca acaaaauuaa augauuuaug uuuuacaaau guguaugcug 8760

auucuuuugu gaucagaggu gaugaaguga gacagauugc ccccggacag acaggaaaaa 8820

uugcugauua caauuacaaa cugccugaug auuuuacagg augugugauu gcuuggaauu 8880

cuaauaauuu agauucuaaa gugggaggaa auuacaauua ucuguacaga cuguuuagaa 8940

aaucaaaucu gaaaccuuuu gaaagagaua uuucaacaga aauuuaucag gcuggaucaa 9000

caccuuguaa uggaguggaa ggauuuaauu guuauuuucc auuacagagc uauggauuuc 9060

agccaaccaa ugguguggga uaucagccau auagaguggu ggugcugucu uuugaacugc 9120

ugcaugcacc ugcaacagug uguggaccua aaaaaucuac aaauuuagug aaaaauaaau 9180

gugugaauuu uaauuuuaau ggauuaacag gaacaggagu gcugacagaa ucuaauaaaa 9240

aauuucugcc uuuucagcag uuuggcagag auauugcaga uaccacagau gcagugagag 9300

auccucagac auuagaaauu cuggauauua caccuuguuc uuuugggggu gugucuguga 9360

uuacaccugg aacaaauaca ucuaaucagg uggcugugcu guaucaggau gugaauugua 9420

cagaagugcc aguggcaauu caugcagauc agcugacacc aacauggaga guguauucua 9480

caggaucuaa uguguuucag acaagagcag gaugucugau uggagcagaa caugugaaua 9540

auucuuauga augugauauu ccaauuggag caggcauuug ugcaucuuau cagacacaga 9600

caaauucccc aaggagagca agaucugugg caucucaguc uauuauugca uacaccaugu 9660

cucugggagc agaaaauucu guggcauauu cuaauaauuc uauugcuauu ccaacaaauu 9720

uuaccauuuc ugugacaaca gaaauuuuac cugugucuau gacaaaaaca ucuguggauu 9780

guaccaugua cauuugugga gauucuacag aauguucuaa ucugcugcug caguauggau 9840

cuuuuuguac acagcugaau agagcuuuaa caggaauugc uguggaacag gauaaaaaua 9900

cacaggaagu guuugcucag gugaaacaga uuuacaaaac accaccaauu aaagauuuug 9960

gaggauuuaa uuuuagccag auucugccug auccuucuaa accuucuaaa agaucuuuua 10020

uugaagaucu gcuguuuaau aaagugacac uggcagaugc aggauuuauu aaacaguaug 10080

gagauugccu gggugauauu gcugcaagag aucugauuug ugcucagaaa uuuaauggac 10140

ugacagugcu gccuccucug cugacagaug aaaugauugc ucaguacaca ucugcuuuac 10200

uggcuggaac aauuacaagc ggauggacau uuggagcugg agcugcucug cagauuccuu 10260

uugcaaugca gauggcuuac agauuuaaug gaauuggagu gacacagaau guguuauaug 10320

aaaaucagaa acugauugca aaucaguuua auucugcaau uggcaaaauu caggauucuc 10380

ugucuucuac agcuucugcu cugggaaaac ugcaggaugu ggugaaucag aaugcacagg 10440

cacugaauac ucuggugaaa cagcugucua gcaauuuugg ggcaauuucu ucugugcuga 10500

augauauucu gucuagacug gauccuccug aagcugaagu gcagauugau agacugauca 10560

caggaagacu gcagucucug cagacuuaug ugacacagca gcugauuaga gcugcugaaa 10620

uuagagcuuc ugcuaaucug gcugcuacaa aaaugucuga augugugcug ggacagucaa 10680

aaagagugga uuuuugugga aaaggauauc aucugauguc uuuuccacag ucugcuccac 10740

auggaguggu guuuuuacau gugacauaug ugccagcaca ggaaaagaau uuuaccacag 10800

caccagcaau uugucaugau ggaaaagcac auuuuccaag agaaggagug uuugugucua 10860

auggaacaca uugguuugug acacagagaa auuuuuauga accucagauu auuacaacag 10920

auaauacauu ugugucagga aauugugaug uggugauugg aauugugaau aauacagugu 10980

augauccacu gcagccagaa cuggauucuu uuaaagaaga acuggauaaa uauuuuaaaa 11040

aucacacauc uccugaugug gauuuaggag auauuucugg aaucaaugca ucugugguga 11100

auauucagaa agaaauugau agacugaaug aaguggccaa aaaucugaau gaaucucuga 11160

uugaucugca ggaacuugga aaauaugaac aguacauuaa auggccuugg uacauuuggc 11220

uuggauuuau ugcaggauua auugcaauug ugauggugac aauuauguua uguuguauga 11280

caucauguug uucuuguuua aaaggauguu guucuugugg aagcuguugu aaauuugaug 11340

aagaugauuc ugaaccugug uuaaaaggag ugaaauugca uuacacauga ugacucgagc 11400

ugguacugca ugcacgcaau gcuagcugcc ccuuucccgu ccuggguacc ccgagucucc 11460

cccgaccucg ggucccaggu augcucccac cuccaccugc cccacucacc accucugcua 11520

guuccagaca ccucccaagc acgcagcaau gcagcucaaa acgcuuagcc uagccacacc 11580

cccacgggaa acagcaguga uuaaccuuua gcaauaaacg aaaguuuaac uaagcuauac 11640

uaaccccagg guuggucaau uucgugccag ccacaccgcg gccgcaugaa uacagcagca 11700

auuggcaagc ugcuuacaua gaacucgcgg cgauuggcau gccgccuuaa aauuuuuauu 11760

uuauuuuuuc uuuucuuuuc cgaaucggau uuuguuuuua auauuucaaa aaaaaaaaaa 11820

aaaaaaaaaa aaaaaaagca uaugacuaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 11880

aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaa 11917

<210> 25

<211> 11917

<212> RNA

<213> Artificial sequence

<220>

<223> RBS004.2

<400> 25

gaugggcggc gcaugagaga agcccagacc aauuaccuac ccaaaaugga gaaaguucac 60

guugacaucg aggaagacag cccauuccuc agagcuuugc agcggagcuu cccgcaguuu 120

gagguagaag ccaagcaggu cacugauaau gaccaugcua augccagagc guuuucgcau 180

cuggcuucaa aacugaucga aacggaggug gacccauccg acacgauccu ugacauugga 240

agugcgcccg cccgcagaau guauucuaag cacaaguauc auuguaucug uccgaugaga 300

ugugcggaag auccggacag auuguauaag uaugcaacua agcugaagaa aaacuguaag 360

gaaauaacug auaaggaauu ggacaagaaa augaaggagc ucgccgccgu caugagcgac 420

ccugaccugg aaacugagac uaugugccuc cacgacgacg agucgugucg cuacgaaggg 480

caagucgcug uuuaccagga uguauacgcg guugacggac cgacaagucu cuaucaccaa 540

gccaauaagg gaguuagagu cgccuacugg auaggcuuug acaccacccc uuuuauguuu 600

aagaacuugg cuggagcaua uccaucauac ucuaccaacu gggccgacga aaccguguua 660

acggcucgua acauaggccu augcagcucu gacguuaugg agcggucacg uagagggaug 720

uccauucuua gaaagaagua uuugaaacca uccaacaaug uucuauucuc uguuggcucg 780

accaucuacc acgaaaagag ggacuuacug aggagcuggc accugccguc uguauuucac 840

uuacguggca agcaaaauua cacaugucgg ugugagacua uaguuaguug cgacggguac 900

gucguuaaaa gaauagcuau caguccaggc cuguauggga agccuucagg cuaugcugcu 960

acgaugcacc gcgagggauu cuugugcugc aaagugacag acacauugaa cggggagagg 1020

gucucuuuuc ccgugugcac guaugugcca gcuacauugu gugaccaaau gacuggcaua 1080

cuggcaacag augucagugc ggacgacgcg caaaaacugc ugguugggcu caaccagcgu 1140

auagucguca acggucgcac ccagagaaac accaauacca ugaaaaauua ccuuuugccc 1200

guaguggccc aggcauuugc uaggugggca aaggaauaua aggaagauca agaagaugaa 1260

aggccacuag gacuacgaga uagacaguua gucauggggu guuguugggc uuuuagaagg 1320

cacaagauaa caucuauuua uaagcgcccg gauacccaaa ccaucaucaa agugaacagc 1380

gauuuccacu cauucgugcu gcccaggaua ggcaguaaca cauuggagau cgggcugaga 1440

acaagaauca ggaaaauguu agaggagcac aaggagccgu caccucucau uaccgccgag 1500

gacguacaag aagcuaagug cgcagccgau gaggcuaagg aggugcguga agccgaggag 1560

uugcgcgcag cucuaccacc uuuggcagcu gauguugagg agcccacucu ggaagccgau 1620

gucgacuuga uguuacaaga ggcuggggcc ggcucagugg agacaccucg uggcuugaua 1680

aagguuacca gcuacgcugg cgaggacaag aucggcucuu acgcugugcu uucuccgcag 1740

gcuguacuca agagugaaaa auuaucuugc auccacccuc ucgcugaaca agucauagug 1800

auaacacacu cuggccgaaa agggcguuau gccguggaac cauaccaugg uaaaguagug 1860

gugccagagg gacaugcaau acccguccag gacuuucaag cucugaguga aagugccacc 1920

auuguguaca acgaacguga guucguaaac agguaccugc accauauugc cacacaugga 1980

ggagcgcuga acacugauga agaauauuac aaaacuguca agcccagcga gcacgacggc 2040

gaauaccugu acgacaucga caggaaacag ugcgucaaga aagagcuagu cacugggcua 2100

gggcucacag gcgagcuggu cgauccuccc uuccaugaau ucgccuacga gagucugaga 2160

acacgaccag ccgcuccuua ccaaguacca accauagggg uguauggcgu gccaggauca 2220

ggcaagucug gcaucauuaa aagcgcaguc accaaaaaag aucuaguggu gagcgccaag 2280

aaagaaaacu gugcagaaau uauaagggac gucaagaaaa ugaaagggcu ggacgucaau 2340

gccagaacug uggacucagu gcucuugaau ggaugcaaac accccguaga gacccuguau 2400

auugacgagg cuuuugcuug ucaugcaggu acucucagag cgcucauagc cauuauaaga 2460

ccuaaaaagg cagugcucug cggagauccc aaacagugcg guuuuuuuaa caugaugugc 2520

cugaaagugc auuuuaacca cgagauuugc acacaagucu uccacaaaag caucucucgc 2580

cguugcacua aaucugugac uucggucguc ucaaccuugu uuuacgacaa aaaaaugaga 2640

acgacgaauc cgaaagagac uaagauugug auugacacua ccggcaguac caaaccuaag 2700

caggacgauc ucauucucac uuguuucaga ggguggguga agcaguugca aauagauuac 2760

aaaggcaacg aaauaaugac ggcagcugcc ucucaagggc ugacccguaa agguguguau 2820

gccguucggu acaaggugaa ugaaaauccu cuguacgcac ccaccucaga acaugugaac 2880

guccuacuga cccgcacgga ggaccgcauc guguggaaaa cacuagccgg cgacccaugg 2940

auaaaaacac ugacugccaa guacccuggg aauuucacug ccacgauaga ggaguggcaa 3000

gcagagcaug augccaucau gaggcacauc uuggagagac cggacccuac cgacgucuuc 3060

cagaauaagg caaacgugug uugggccaag gcuuuagugc cggugcugaa gaccgcuggc 3120

auagacauga ccacugaaca auggaacacu guggauuauu uugaaacgga caaagcucac 3180

ucagcagaga uaguauugaa ccaacuaugc gugagguucu uuggacucga ucuggacucc 3240

ggucuauuuu cugcacccac uguuccguua uccauuagga auaaucacug ggauaacucc 3300

ccgucgccua acauguacgg gcugaauaaa gaaguggucc gucagcucuc ucgcagguac 3360

ccacaacugc cucgggcagu ugccacuggu agagucuaug acaugaacac ugguacacug 3420

cgcaauuaug auccgcgcau aaaccuagua ccuguaaaca gaagacugcc ucaugcuuua 3480

guccuccacc auaaugaaca cccacagagu gacuuuucuu cauucgucag caaauugaag 3540

ggcagaacug uccugguggu cggggaaaag uuguccgucc caggcaaaau gguugacugg 3600

uugucagacc ggccugaggc uaccuucaga gcucggcugg auuuaggcau cccaggugau 3660

gugcccaaau augacauaau auuuguuaau gugaggaccc cauauaaaua ccaucacuau 3720

cagcagugug aagaccaugc cauuaagcua agcauguuga ccaagaaagc augucugcau 3780

cugaaucccg gcggaaccug ugucagcaua gguuaugguu acgcugacag ggccagcgaa 3840

agcaucauug gugcuauagc gcggcaguuc aaguuuuccc gaguaugcaa accgaaaucc 3900

ucacuugagg agacggaagu ucuguuugua uucauugggu acgaucgcaa ggcccguacg 3960

cacaauccuu acaagcuauc aucaaccuug accaacauuu auacagguuc cagacuccac 4020

gaagccggau gugcacccuc auaucaugug gugcgagggg auauugccac ggccaccgaa 4080

ggagugauua uaaaugcugc uaacagcaaa ggacaaccug gcggaggggu gugcggagcg 4140

cuguauaaga aauucccgga aaguuucgau uuacagccga ucgaaguagg aaaagcgcga 4200

cuggucaaag gugcagcuaa acauaucauu caugccguag gaccaaacuu caacaaaguu 4260

ucggagguug aaggugacaa acaguuggca gaggcuuaug aguccaucgc uaagauuguc 4320

aacgauaaca auuacaaguc aguagcgauu ccacuguugu ccaccggcau cuuuuccggg 4380

aacaaagauc gacuaaccca aucauugaac cauuugcuga cagcuuuaga caccacugau 4440

gcagauguag ccauauacug cagggacaag aaaugggaaa ugacucucaa ggaagcagug 4500

gcuaggagag aagcagugga ggagauaugc auauccgacg auucuucagu gacagaaccu 4560

gaugcagagc uggugagggu gcaucccaag aguucuuugg cuggaaggaa gggcuacagc 4620

acaagcgaug gcaaaacuuu cucauauuug gaagggacca aguuucacca ggcggccaag 4680

gauauagcag aaauuaaugc cauguggccc guugcaacgg aggccaauga gcagguaugc 4740

auguauaucc ucggagaaag caugagcagu auuaggucga aaugccccgu cgaggagucg 4800

gaagccucca caccaccuag cacgcugccu ugcuugugca uccaugccau gacuccagaa 4860

agaguacagc gccuaaaagc cucacgucca gaacaaauua cugugugcuc auccuuucca 4920

uugccgaagu auagaaucac uggugugcag aagauccaau gcucccagcc uauauuguuc 4980

ucaccgaaag ugccugcgua uauucaucca aggaaguauc ucguggaaac accaccggua 5040

gacgagacuc cggagccauc ggcagagaac caauccacag aggggacacc ugaacaacca 5100

ccacuuauaa ccgaggauga gaccaggacu agaacgccug agccgaucau caucgaagaa 5160

gaagaagaag auagcauaag uuugcuguca gauggcccga cccaccaggu gcugcaaguc 5220

gaggcagaca uucacgggcc gcccucugua ucuagcucau ccugguccau uccucaugca 5280

uccgacuuug auguggacag uuuauccaua cuugacaccc uggagggagc uagcgugacc 5340

agcggggcaa cgucagccga gacuaacucu uacuucgcaa agaguaugga guuucuggcg 5400

cgaccggugc cugcgccucg aacaguauuc aggaacccuc cacaucccgc uccgcgcaca 5460

agaacaccgu cacuugcacc cagcagggcc ugcuccagaa ccagccuagu uuccaccccg 5520

ccaggcguga auagggugau cacuagagag gagcucgaag cgcuuacccc gucacgcacu 5580

ccuagcaggu cggucuccag aaccagccug gucuccaacc cgccaggcgu aaauagggug 5640

auuacaagag aggaguuuga ggcguucgua gcacaacaac aaugacgguu ugaugcgggu 5700

gcauacaucu uuuccuccga caccggucaa gggcauuuac aacaaaaauc aguaaggcaa 5760

acggugcuau ccgaaguggu guuggagagg accgaauugg agauuucgua ugccccgcgc 5820

cucgaccaag aaaaagaaga auuacuacgc aagaaauuac aguuaaaucc cacaccugcu 5880

aacagaagca gauaccaguc caggaaggug gagaacauga aagccauaac agcuagacgu 5940

auucugcaag gccuagggca uuauuugaag gcagaaggaa aaguggagug cuaccgaacc 6000

cugcauccug uuccuuugua uucaucuagu gugaaccgug ccuuuucaag ccccaagguc 6060

gcaguggaag ccuguaacgc cauguugaaa gagaacuuuc cgacuguggc uucuuacugu 6120

auuauuccag aguacgaugc cuauuuggac augguugacg gagcuucaug cugcuuagac 6180

acugccaguu uuugcccugc aaagcugcgc agcuuuccaa agaaacacuc cuauuuggaa 6240

cccacaauac gaucggcagu gccuucagcg auccagaaca cgcuccagaa cguccuggca 6300

gcugccacaa aaagaaauug caaugucacg caaaugagag aauugcccgu auuggauucg 6360

gcggccuuua auguggaaug cuucaagaaa uaugcgugua auaaugaaua uugggaaacg 6420

uuuaaagaaa accccaucag gcuuacugaa gaaaacgugg uaaauuacau uaccaaauua 6480

aaaggaccaa aagcugcugc ucuuuuugcg aagacacaua auuugaauau guugcaggac 6540

auaccaaugg acagguuugu aauggacuua aagagagacg ugaaagugac uccaggaaca 6600

aaacauacug aagaacggcc caagguacag gugauccagg cugccgaucc gcuagcaaca 6660

gcguaucugu gcggaaucca ccgagagcug guuaggagau uaaaugcggu ccugcuuccg 6720

aacauucaua cacuguuuga uaugucggcu gaagacuuug acgcuauuau agccgagcac 6780

uuccagccug gggauugugu ucuggaaacu gacaucgcgu cguuugauaa aagugaggac 6840

gacgccaugg cucugaccgc guuaaugauu cuggaagacu uaggugugga cgcagagcug 6900

uugacgcuga uugaggcggc uuucggcgaa auuucaucaa uacauuugcc cacuaaaacu 6960

aaauuuaaau ucggagccau gaugaaaucu ggaauguucc ucacacuguu ugugaacaca 7020

gucauuaaca uuguaaucgc aagcagagug uugagagaac ggcuaaccgg aucaccaugu 7080

gcagcauuca uuggagauga caauaucgug aaaggaguca aaucggacaa auuaauggca 7140

gacaggugcg ccaccugguu gaauauggaa gucaagauua uagaugcugu ggugggcgag 7200

aaagcgccuu auuucugugg aggguuuauu uugugugacu ccgugaccgg cacagcgugc 7260

cguguggcag acccccuaaa aaggcuguuu aagcuaggca aaccucuggc agcagacgau 7320

gaacaugaug augacaggag aagggcauug caugaggagu caacacgcug gaaccgagug 7380

gguauucuuu cagagcugug caaggcagua gaaucaaggu augaaaccgu aggaacuucc 7440

aucauaguua uggccaugac uacucuagcu agcaguguua aaucauucag cuaccugaga 7500

ggggccccua uaacucucua cggcuaaccu gaauggacua cgacauaguc uaguccgcca 7560

agacuaguau guucguguuc cuggugcugc ugccucuggu guccagccag ugugugaacc 7620

ugaccaccag aacacagcug ccuccagccu acaccaacag cuuuaccaga ggcguguacu 7680

accccgacaa gguguucaga uccagcgugc ugcacucuac ccaggaccug uuccugccuu 7740

ucuucagcaa cgugaccugg uuccacgcca uccacguguc cggcaccaau ggcaccaaga 7800

gauucgacaa ccccgugcug cccuucaacg acggggugua cuuugccagc accgagaagu 7860

ccaacaucau cagaggcugg aucuucggca ccacacugga cagcaagacc cagagccugc 7920

ugaucgugaa caacgccacc aacgugguca ucaaagugug cgaguuccag uucugcaacg 7980

accccuuccu gggcgucuac uaccacaaga acaacaagag cuggauggaa agcgaguucc 8040

ggguguacag cagcgccaac aacugcaccu ucgaguacgu gucccagccu uuccugaugg 8100

accuggaagg caagcagggc aacuucaaga accugcgcga guucguguuu aagaacaucg 8160

acggcuacuu caagaucuac agcaagcaca ccccuaucaa ccucgugcgg gaucugccuc 8220

agggcuucuc ugcucuggaa ccccuggugg aucugcccau cggcaucaac aucacccggu 8280

uucagacacu gcuggcccug cacagaagcu accugacacc uggcgauagc agcagcggau 8340

ggacagcugg ugccgccgcu uacuaugugg gcuaccugca gccuagaacc uuccugcuga 8400

aguacaacga gaacggcacc aucaccgacg ccguggauug ugcucuggau ccucugagcg 8460

agacaaagug cacccugaag uccuucaccg uggaaaaggg caucuaccag accagcaacu 8520

uccgggugca gcccaccgaa uccaucgugc gguuccccaa uaucaccaau cugugccccu 8580

ucggcgaggu guucaaugcc accagauucg ccucugugua cgccuggaac cggaagcgga 8640

ucagcaauug cguggccgac uacuccgugc uguacaacuc cgccagcuuc agcaccuuca 8700

agugcuacgg cguguccccu accaagcuga acgaccugug cuucacaaac guguacgccg 8760

acagcuucgu gauccgggga gaugaagugc ggcagauugc cccuggacag acaggcaaga 8820

ucgccgacua caacuacaag cugcccgacg acuucaccgg cugugugauu gccuggaaca 8880

gcaacaaccu ggacuccaaa gucggcggca acuacaauua ccuguaccgg cuguuccgga 8940

aguccaaucu gaagcccuuc gagcgggaca ucuccaccga gaucuaucag gccggcagca 9000

ccccuuguaa cggcguggaa ggcuucaacu gcuacuuccc acugcagucc uacggcuuuc 9060

agcccacaaa uggcgugggc uaucagcccu acagaguggu ggugcugagc uucgaacugc 9120

ugcaugcccc ugccacagug ugcggcccua agaaaagcac caaucucgug aagaacaaau 9180

gcgugaacuu caacuucaac ggccugaccg gcaccggcgu gcugacagag agcaacaaga 9240

aguuccugcc auuccagcag uuuggccggg auaucgccga uaccacagac gccguuagag 9300

auccccagac acuggaaauc cuggacauca ccccuugcag cuucggcgga gugucuguga 9360

ucaccccugg caccaacacc agcaaucagg uggcagugcu guaccaggac gugaacugua 9420

ccgaagugcc cguggccauu cacgccgauc agcugacacc uacauggcgg guguacucca 9480

ccggcagcaa uguguuucag accagagccg gcugucugau cggagccgag cacgugaaca 9540

auagcuacga gugcgacauc cccaucggcg cuggaaucug cgccagcuac cagacacaga 9600

caaacagccc ucggagagcc agaagcgugg ccagccagag caucauugcc uacacaaugu 9660

cucugggcgc cgagaacagc guggccuacu ccaacaacuc uaucgcuauc cccaccaacu 9720

ucaccaucag cgugaccaca gagauccugc cuguguccau gaccaagacc agcguggacu 9780

gcaccaugua caucugcggc gauuccaccg agugcuccaa ccugcugcug caguacggca 9840

gcuucugcac ccagcugaau agagcccuga cagggaucgc cguggaacag gacaagaaca 9900

cccaagaggu guucgcccaa gugaagcaga ucuacaagac cccuccuauc aaggacuucg 9960

gcggcuucaa uuucagccag auucugcccg auccuagcaa gcccagcaag cggagcuuca 10020

ucgaggaccu gcuguucaac aaagugacac uggccgacgc cggcuucauc aagcaguaug 10080

gcgauugucu gggcgacauu gccgccaggg aucugauuug cgcccagaag uuuaacggac 10140

ugacagugcu gccuccucug cugaccgaug agaugaucgc ccaguacaca ucugcccugc 10200

uggccggcac aaucacaagc ggcuggacau uuggagcagg cgccgcucug cagauccccu 10260

uugcuaugca gauggccuac cgguucaacg gcaucggagu gacccagaau gugcuguacg 10320

agaaccagaa gcugaucgcc aaccaguuca acagcgccau cggcaagauc caggacagcc 10380

ugagcagcac agcaagcgcc cugggaaagc ugcaggacgu ggucaaccag aaugcccagg 10440

cacugaacac ccuggucaag cagcuguccu ccaacuucgg cgccaucagc ucugugcuga 10500

acgauauccu gagcagacug gacccuccug aggccgaggu gcagaucgac agacugauca 10560

caggcagacu gcagagccuc cagacauacg ugacccagca gcugaucaga gccgccgaga 10620

uuagagccuc ugccaaucug gccgccacca agaugucuga gugugugcug ggccagagca 10680

agagagugga cuuuugcggc aagggcuacc accugaugag cuucccucag ucugccccuc 10740

acggcguggu guuucugcac gugacauaug ugcccgcuca agagaagaau uucaccaccg 10800

cuccagccau cugccacgac ggcaaagccc acuuuccuag agaaggcgug uucgugucca 10860

acggcaccca uugguucgug acacagcgga acuucuacga gccccagauc aucaccaccg 10920

acaacaccuu cgugucuggc aacugcgacg ucgugaucgg cauugugaac aauaccgugu 10980

acgacccucu gcagcccgag cuggacagcu ucaaagagga acuggacaag uacuuuaaga 11040

accacacaag ccccgacgug gaccugggcg auaucagcgg aaucaaugcc agcgucguga 11100

acauccagaa agagaucgac cggcugaacg agguggccaa gaaucugaac gagagccuga 11160

ucgaccugca agaacugggg aaguacgagc aguacaucaa guggcccugg uacaucuggc 11220

ugggcuuuau cgccggacug auugccaucg ugauggucac aaucaugcug uguugcauga 11280

ccagcugcug uagcugccug aagggcuguu guagcugugg cagcugcugc aaguucgacg 11340

aggacgauuc ugagcccgug cugaagggcg ugaaacugca cuacacauga ugacucgagc 11400

ugguacugca ugcacgcaau gcuagcugcc ccuuucccgu ccuggguacc ccgagucucc 11460

cccgaccucg ggucccaggu augcucccac cuccaccugc cccacucacc accucugcua 11520

guuccagaca ccucccaagc acgcagcaau gcagcucaaa acgcuuagcc uagccacacc 11580

cccacgggaa acagcaguga uuaaccuuua gcaauaaacg aaaguuuaac uaagcuauac 11640

uaaccccagg guuggucaau uucgugccag ccacaccgcg gccgcaugaa uacagcagca 11700

auuggcaagc ugcuuacaua gaacucgcgg cgauuggcau gccgccuuaa aauuuuuauu 11760

uuauuuuuuc uuuucuuuuc cgaaucggau uuuguuuuua auauuucaaa aaaaaaaaaa 11820

aaaaaaaaaa aaaaaaagca uaugacuaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 11880

aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaa 11917

<210> 26

<211> 8896

<212> RNA

<213> Artificial sequence

<220>

<223> RBS004.3

<400> 26

gaugggcggc gcaugagaga agcccagacc aauuaccuac ccaaaaugga gaaaguucac 60

guugacaucg aggaagacag cccauuccuc agagcuuugc agcggagcuu cccgcaguuu 120

gagguagaag ccaagcaggu cacugauaau gaccaugcua augccagagc guuuucgcau 180

cuggcuucaa aacugaucga aacggaggug gacccauccg acacgauccu ugacauugga 240

agugcgcccg cccgcagaau guauucuaag cacaaguauc auuguaucug uccgaugaga 300

ugugcggaag auccggacag auuguauaag uaugcaacua agcugaagaa aaacuguaag 360

gaaauaacug auaaggaauu ggacaagaaa augaaggagc ucgccgccgu caugagcgac 420

ccugaccugg aaacugagac uaugugccuc cacgacgacg agucgugucg cuacgaaggg 480

caagucgcug uuuaccagga uguauacgcg guugacggac cgacaagucu cuaucaccaa 540

gccaauaagg gaguuagagu cgccuacugg auaggcuuug acaccacccc uuuuauguuu 600

aagaacuugg cuggagcaua uccaucauac ucuaccaacu gggccgacga aaccguguua 660

acggcucgua acauaggccu augcagcucu gacguuaugg agcggucacg uagagggaug 720

uccauucuua gaaagaagua uuugaaacca uccaacaaug uucuauucuc uguuggcucg 780

accaucuacc acgaaaagag ggacuuacug aggagcuggc accugccguc uguauuucac 840

uuacguggca agcaaaauua cacaugucgg ugugagacua uaguuaguug cgacggguac 900

gucguuaaaa gaauagcuau caguccaggc cuguauggga agccuucagg cuaugcugcu 960

acgaugcacc gcgagggauu cuugugcugc aaagugacag acacauugaa cggggagagg 1020

gucucuuuuc ccgugugcac guaugugcca gcuacauugu gugaccaaau gacuggcaua 1080

cuggcaacag augucagugc ggacgacgcg caaaaacugc ugguugggcu caaccagcgu 1140

auagucguca acggucgcac ccagagaaac accaauacca ugaaaaauua ccuuuugccc 1200

guaguggccc aggcauuugc uaggugggca aaggaauaua aggaagauca agaagaugaa 1260

aggccacuag gacuacgaga uagacaguua gucauggggu guuguugggc uuuuagaagg 1320

cacaagauaa caucuauuua uaagcgcccg gauacccaaa ccaucaucaa agugaacagc 1380

gauuuccacu cauucgugcu gcccaggaua ggcaguaaca cauuggagau cgggcugaga 1440

acaagaauca ggaaaauguu agaggagcac aaggagccgu caccucucau uaccgccgag 1500

gacguacaag aagcuaagug cgcagccgau gaggcuaagg aggugcguga agccgaggag 1560

uugcgcgcag cucuaccacc uuuggcagcu gauguugagg agcccacucu ggaagccgau 1620

gucgacuuga uguuacaaga ggcuggggcc ggcucagugg agacaccucg uggcuugaua 1680

aagguuacca gcuacgcugg cgaggacaag aucggcucuu acgcugugcu uucuccgcag 1740

gcuguacuca agagugaaaa auuaucuugc auccacccuc ucgcugaaca agucauagug 1800

auaacacacu cuggccgaaa agggcguuau gccguggaac cauaccaugg uaaaguagug 1860

gugccagagg gacaugcaau acccguccag gacuuucaag cucugaguga aagugccacc 1920

auuguguaca acgaacguga guucguaaac agguaccugc accauauugc cacacaugga 1980

ggagcgcuga acacugauga agaauauuac aaaacuguca agcccagcga gcacgacggc 2040

gaauaccugu acgacaucga caggaaacag ugcgucaaga aagagcuagu cacugggcua 2100

gggcucacag gcgagcuggu cgauccuccc uuccaugaau ucgccuacga gagucugaga 2160

acacgaccag ccgcuccuua ccaaguacca accauagggg uguauggcgu gccaggauca 2220

ggcaagucug gcaucauuaa aagcgcaguc accaaaaaag aucuaguggu gagcgccaag 2280

aaagaaaacu gugcagaaau uauaagggac gucaagaaaa ugaaagggcu ggacgucaau 2340

gccagaacug uggacucagu gcucuugaau ggaugcaaac accccguaga gacccuguau 2400

auugacgagg cuuuugcuug ucaugcaggu acucucagag cgcucauagc cauuauaaga 2460

ccuaaaaagg cagugcucug cggagauccc aaacagugcg guuuuuuuaa caugaugugc 2520

cugaaagugc auuuuaacca cgagauuugc acacaagucu uccacaaaag caucucucgc 2580

cguugcacua aaucugugac uucggucguc ucaaccuugu uuuacgacaa aaaaaugaga 2640

acgacgaauc cgaaagagac uaagauugug auugacacua ccggcaguac caaaccuaag 2700

caggacgauc ucauucucac uuguuucaga ggguggguga agcaguugca aauagauuac 2760

aaaggcaacg aaauaaugac ggcagcugcc ucucaagggc ugacccguaa agguguguau 2820

gccguucggu acaaggugaa ugaaaauccu cuguacgcac ccaccucaga acaugugaac 2880

guccuacuga cccgcacgga ggaccgcauc guguggaaaa cacuagccgg cgacccaugg 2940

auaaaaacac ugacugccaa guacccuggg aauuucacug ccacgauaga ggaguggcaa 3000

gcagagcaug augccaucau gaggcacauc uuggagagac cggacccuac cgacgucuuc 3060

cagaauaagg caaacgugug uugggccaag gcuuuagugc cggugcugaa gaccgcuggc 3120

auagacauga ccacugaaca auggaacacu guggauuauu uugaaacgga caaagcucac 3180

ucagcagaga uaguauugaa ccaacuaugc gugagguucu uuggacucga ucuggacucc 3240

ggucuauuuu cugcacccac uguuccguua uccauuagga auaaucacug ggauaacucc 3300

ccgucgccua acauguacgg gcugaauaaa gaaguggucc gucagcucuc ucgcagguac 3360

ccacaacugc cucgggcagu ugccacuggu agagucuaug acaugaacac ugguacacug 3420

cgcaauuaug auccgcgcau aaaccuagua ccuguaaaca gaagacugcc ucaugcuuua 3480

guccuccacc auaaugaaca cccacagagu gacuuuucuu cauucgucag caaauugaag 3540

ggcagaacug uccugguggu cggggaaaag uuguccgucc caggcaaaau gguugacugg 3600

uugucagacc ggccugaggc uaccuucaga gcucggcugg auuuaggcau cccaggugau 3660

gugcccaaau augacauaau auuuguuaau gugaggaccc cauauaaaua ccaucacuau 3720

cagcagugug aagaccaugc cauuaagcua agcauguuga ccaagaaagc augucugcau 3780

cugaaucccg gcggaaccug ugucagcaua gguuaugguu acgcugacag ggccagcgaa 3840

agcaucauug gugcuauagc gcggcaguuc aaguuuuccc gaguaugcaa accgaaaucc 3900

ucacuugagg agacggaagu ucuguuugua uucauugggu acgaucgcaa ggcccguacg 3960

cacaauccuu acaagcuauc aucaaccuug accaacauuu auacagguuc cagacuccac 4020

gaagccggau gugcacccuc auaucaugug gugcgagggg auauugccac ggccaccgaa 4080

ggagugauua uaaaugcugc uaacagcaaa ggacaaccug gcggaggggu gugcggagcg 4140

cuguauaaga aauucccgga aaguuucgau uuacagccga ucgaaguagg aaaagcgcga 4200

cuggucaaag gugcagcuaa acauaucauu caugccguag gaccaaacuu caacaaaguu 4260

ucggagguug aaggugacaa acaguuggca gaggcuuaug aguccaucgc uaagauuguc 4320

aacgauaaca auuacaaguc aguagcgauu ccacuguugu ccaccggcau cuuuuccggg 4380

aacaaagauc gacuaaccca aucauugaac cauuugcuga cagcuuuaga caccacugau 4440

gcagauguag ccauauacug cagggacaag aaaugggaaa ugacucucaa ggaagcagug 4500

gcuaggagag aagcagugga ggagauaugc auauccgacg auucuucagu gacagaaccu 4560

gaugcagagc uggugagggu gcaucccaag aguucuuugg cuggaaggaa gggcuacagc 4620

acaagcgaug gcaaaacuuu cucauauuug gaagggacca aguuucacca ggcggccaag 4680

gauauagcag aaauuaaugc cauguggccc guugcaacgg aggccaauga gcagguaugc 4740

auguauaucc ucggagaaag caugagcagu auuaggucga aaugccccgu cgaggagucg 4800

gaagccucca caccaccuag cacgcugccu ugcuugugca uccaugccau gacuccagaa 4860

agaguacagc gccuaaaagc cucacgucca gaacaaauua cugugugcuc auccuuucca 4920

uugccgaagu auagaaucac uggugugcag aagauccaau gcucccagcc uauauuguuc 4980

ucaccgaaag ugccugcgua uauucaucca aggaaguauc ucguggaaac accaccggua 5040

gacgagacuc cggagccauc ggcagagaac caauccacag aggggacacc ugaacaacca 5100

ccacuuauaa ccgaggauga gaccaggacu agaacgccug agccgaucau caucgaagaa 5160

gaagaagaag auagcauaag uuugcuguca gauggcccga cccaccaggu gcugcaaguc 5220

gaggcagaca uucacgggcc gcccucugua ucuagcucau ccugguccau uccucaugca 5280

uccgacuuug auguggacag uuuauccaua cuugacaccc uggagggagc uagcgugacc 5340

agcggggcaa cgucagccga gacuaacucu uacuucgcaa agaguaugga guuucuggcg 5400

cgaccggugc cugcgccucg aacaguauuc aggaacccuc cacaucccgc uccgcgcaca 5460

agaacaccgu cacuugcacc cagcagggcc ugcuccagaa ccagccuagu uuccaccccg 5520

ccaggcguga auagggugau cacuagagag gagcucgaag cgcuuacccc gucacgcacu 5580

ccuagcaggu cggucuccag aaccagccug gucuccaacc cgccaggcgu aaauagggug 5640

auuacaagag aggaguuuga ggcguucgua gcacaacaac aaugacgguu ugaugcgggu 5700

gcauacaucu uuuccuccga caccggucaa gggcauuuac aacaaaaauc aguaaggcaa 5760

acggugcuau ccgaaguggu guuggagagg accgaauugg agauuucgua ugccccgcgc 5820

cucgaccaag aaaaagaaga auuacuacgc aagaaauuac aguuaaaucc cacaccugcu 5880

aacagaagca gauaccaguc caggaaggug gagaacauga aagccauaac agcuagacgu 5940

auucugcaag gccuagggca uuauuugaag gcagaaggaa aaguggagug cuaccgaacc 6000

cugcauccug uuccuuugua uucaucuagu gugaaccgug ccuuuucaag ccccaagguc 6060

gcaguggaag ccuguaacgc cauguugaaa gagaacuuuc cgacuguggc uucuuacugu 6120

auuauuccag aguacgaugc cuauuuggac augguugacg gagcuucaug cugcuuagac 6180

acugccaguu uuugcccugc aaagcugcgc agcuuuccaa agaaacacuc cuauuuggaa 6240

cccacaauac gaucggcagu gccuucagcg auccagaaca cgcuccagaa cguccuggca 6300

gcugccacaa aaagaaauug caaugucacg caaaugagag aauugcccgu auuggauucg 6360

gcggccuuua auguggaaug cuucaagaaa uaugcgugua auaaugaaua uugggaaacg 6420

uuuaaagaaa accccaucag gcuuacugaa gaaaacgugg uaaauuacau uaccaaauua 6480

aaaggaccaa aagcugcugc ucuuuuugcg aagacacaua auuugaauau guugcaggac 6540

auaccaaugg acagguuugu aauggacuua aagagagacg ugaaagugac uccaggaaca 6600

aaacauacug aagaacggcc caagguacag gugauccagg cugccgaucc gcuagcaaca 6660

gcguaucugu gcggaaucca ccgagagcug guuaggagau uaaaugcggu ccugcuuccg 6720

aacauucaua cacuguuuga uaugucggcu gaagacuuug acgcuauuau agccgagcac 6780

uuccagccug gggauugugu ucuggaaacu gacaucgcgu cguuugauaa aagugaggac 6840

gacgccaugg cucugaccgc guuaaugauu cuggaagacu uaggugugga cgcagagcug 6900

uugacgcuga uugaggcggc uuucggcgaa auuucaucaa uacauuugcc cacuaaaacu 6960

aaauuuaaau ucggagccau gaugaaaucu ggaauguucc ucacacuguu ugugaacaca 7020

gucauuaaca uuguaaucgc aagcagagug uugagagaac ggcuaaccgg aucaccaugu 7080

gcagcauuca uuggagauga caauaucgug aaaggaguca aaucggacaa auuaauggca 7140

gacaggugcg ccaccugguu gaauauggaa gucaagauua uagaugcugu ggugggcgag 7200

aaagcgccuu auuucugugg aggguuuauu uugugugacu ccgugaccgg cacagcgugc 7260

cguguggcag acccccuaaa aaggcuguuu aagcuaggca aaccucuggc agcagacgau 7320

gaacaugaug augacaggag aagggcauug caugaggagu caacacgcug gaaccgagug 7380

gguauucuuu cagagcugug caaggcagua gaaucaaggu augaaaccgu aggaacuucc 7440

aucauaguua uggccaugac uacucuagcu agcaguguua aaucauucag cuaccugaga 7500

ggggccccua uaacucucua cggcuaaccu gaauggacua cgacauaguc uaguccgcca 7560

agacuaguau guuuguguuu cuugugcugc ugccucuugu gucuucucag ugugugguga 7620

gauuuccaaa uauuacaaau cuguguccau uuggagaagu guuuaaugca acaagauuug 7680

caucugugua ugcauggaau agaaaaagaa uuucuaauug uguggcugau uauucugugc 7740

uguauaauag ugcuucuuuu uccacauuua aauguuaugg agugucucca acaaaauuaa 7800

augauuuaug uuuuacaaau guguaugcug auucuuuugu gaucagaggu gaugaaguga 7860

gacagauugc ccccggacag acaggaaaaa uugcugauua caauuacaaa cugccugaug 7920

auuuuacagg augugugauu gcuuggaauu cuaauaauuu agauucuaaa gugggaggaa 7980

auuacaauua ucuguacaga cuguuuagaa aaucaaaucu gaaaccuuuu gaaagagaua 8040

uuucaacaga aauuuaucag gcuggaucaa caccuuguaa uggaguggaa ggauuuaauu 8100

guuauuuucc auuacagagc uauggauuuc agccaaccaa ugguguggga uaucagccau 8160

auagaguggu ggugcugucu uuugaacugc ugcaugcacc ugcaacagug uguggaccua 8220

aaggcucccc cggcuccggc uccggaucug guuauauucc ugaagcucca agagaugggc 8280

aagcuuacgu ucguaaagau ggcgaauggg uauuacuuuc uaccuuuuua ggccgguccc 8340

uggaggugcu guuccagggc cccggcugau gacucgagcu gguacugcau gcacgcaaug 8400

cuagcugccc cuuucccguc cuggguaccc cgagucuccc ccgaccucgg gucccaggua 8460

ugcucccacc uccaccugcc ccacucacca ccucugcuag uuccagacac cucccaagca 8520

cgcagcaaug cagcucaaaa cgcuuagccu agccacaccc ccacgggaaa cagcagugau 8580

uaaccuuuag caauaaacga aaguuuaacu aagcuauacu aaccccaggg uuggucaauu 8640

ucgugccagc cacaccgcgg ccgcaugaau acagcagcaa uuggcaagcu gcuuacauag 8700

aacucgcggc gauuggcaug ccgccuuaaa auuuuuauuu uauuuuuucu uuucuuuucc 8760

gaaucggauu uuguuuuuaa uauuucaaaa aaaaaaaaaa aaaaaaaaaa aaaaaagcau 8820

augacuaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 8880

aaaaaaaaaa aaaaaa 8896

<210> 27

<211> 9079

<212> RNA

<213> Artificial sequence

<220>

<223> RBS004.4

<400> 27

gaugggcggc gcaugagaga agcccagacc aauuaccuac ccaaaaugga gaaaguucac 60

guugacaucg aggaagacag cccauuccuc agagcuuugc agcggagcuu cccgcaguuu 120

gagguagaag ccaagcaggu cacugauaau gaccaugcua augccagagc guuuucgcau 180

cuggcuucaa aacugaucga aacggaggug gacccauccg acacgauccu ugacauugga 240

agugcgcccg cccgcagaau guauucuaag cacaaguauc auuguaucug uccgaugaga 300

ugugcggaag auccggacag auuguauaag uaugcaacua agcugaagaa aaacuguaag 360

gaaauaacug auaaggaauu ggacaagaaa augaaggagc ucgccgccgu caugagcgac 420

ccugaccugg aaacugagac uaugugccuc cacgacgacg agucgugucg cuacgaaggg 480

caagucgcug uuuaccagga uguauacgcg guugacggac cgacaagucu cuaucaccaa 540

gccaauaagg gaguuagagu cgccuacugg auaggcuuug acaccacccc uuuuauguuu 600

aagaacuugg cuggagcaua uccaucauac ucuaccaacu gggccgacga aaccguguua 660

acggcucgua acauaggccu augcagcucu gacguuaugg agcggucacg uagagggaug 720

uccauucuua gaaagaagua uuugaaacca uccaacaaug uucuauucuc uguuggcucg 780

accaucuacc acgaaaagag ggacuuacug aggagcuggc accugccguc uguauuucac 840

uuacguggca agcaaaauua cacaugucgg ugugagacua uaguuaguug cgacggguac 900

gucguuaaaa gaauagcuau caguccaggc cuguauggga agccuucagg cuaugcugcu 960

acgaugcacc gcgagggauu cuugugcugc aaagugacag acacauugaa cggggagagg 1020

gucucuuuuc ccgugugcac guaugugcca gcuacauugu gugaccaaau gacuggcaua 1080

cuggcaacag augucagugc ggacgacgcg caaaaacugc ugguugggcu caaccagcgu 1140

auagucguca acggucgcac ccagagaaac accaauacca ugaaaaauua ccuuuugccc 1200

guaguggccc aggcauuugc uaggugggca aaggaauaua aggaagauca agaagaugaa 1260

aggccacuag gacuacgaga uagacaguua gucauggggu guuguugggc uuuuagaagg 1320

cacaagauaa caucuauuua uaagcgcccg gauacccaaa ccaucaucaa agugaacagc 1380

gauuuccacu cauucgugcu gcccaggaua ggcaguaaca cauuggagau cgggcugaga 1440

acaagaauca ggaaaauguu agaggagcac aaggagccgu caccucucau uaccgccgag 1500

gacguacaag aagcuaagug cgcagccgau gaggcuaagg aggugcguga agccgaggag 1560

uugcgcgcag cucuaccacc uuuggcagcu gauguugagg agcccacucu ggaagccgau 1620

gucgacuuga uguuacaaga ggcuggggcc ggcucagugg agacaccucg uggcuugaua 1680

aagguuacca gcuacgcugg cgaggacaag aucggcucuu acgcugugcu uucuccgcag 1740

gcuguacuca agagugaaaa auuaucuugc auccacccuc ucgcugaaca agucauagug 1800

auaacacacu cuggccgaaa agggcguuau gccguggaac cauaccaugg uaaaguagug 1860

gugccagagg gacaugcaau acccguccag gacuuucaag cucugaguga aagugccacc 1920

auuguguaca acgaacguga guucguaaac agguaccugc accauauugc cacacaugga 1980

ggagcgcuga acacugauga agaauauuac aaaacuguca agcccagcga gcacgacggc 2040

gaauaccugu acgacaucga caggaaacag ugcgucaaga aagagcuagu cacugggcua 2100

gggcucacag gcgagcuggu cgauccuccc uuccaugaau ucgccuacga gagucugaga 2160

acacgaccag ccgcuccuua ccaaguacca accauagggg uguauggcgu gccaggauca 2220

ggcaagucug gcaucauuaa aagcgcaguc accaaaaaag aucuaguggu gagcgccaag 2280

aaagaaaacu gugcagaaau uauaagggac gucaagaaaa ugaaagggcu ggacgucaau 2340

gccagaacug uggacucagu gcucuugaau ggaugcaaac accccguaga gacccuguau 2400

auugacgagg cuuuugcuug ucaugcaggu acucucagag cgcucauagc cauuauaaga 2460

ccuaaaaagg cagugcucug cggagauccc aaacagugcg guuuuuuuaa caugaugugc 2520

cugaaagugc auuuuaacca cgagauuugc acacaagucu uccacaaaag caucucucgc 2580

cguugcacua aaucugugac uucggucguc ucaaccuugu uuuacgacaa aaaaaugaga 2640

acgacgaauc cgaaagagac uaagauugug auugacacua ccggcaguac caaaccuaag 2700

caggacgauc ucauucucac uuguuucaga ggguggguga agcaguugca aauagauuac 2760

aaaggcaacg aaauaaugac ggcagcugcc ucucaagggc ugacccguaa agguguguau 2820

gccguucggu acaaggugaa ugaaaauccu cuguacgcac ccaccucaga acaugugaac 2880

guccuacuga cccgcacgga ggaccgcauc guguggaaaa cacuagccgg cgacccaugg 2940

auaaaaacac ugacugccaa guacccuggg aauuucacug ccacgauaga ggaguggcaa 3000

gcagagcaug augccaucau gaggcacauc uuggagagac cggacccuac cgacgucuuc 3060

cagaauaagg caaacgugug uugggccaag gcuuuagugc cggugcugaa gaccgcuggc 3120

auagacauga ccacugaaca auggaacacu guggauuauu uugaaacgga caaagcucac 3180

ucagcagaga uaguauugaa ccaacuaugc gugagguucu uuggacucga ucuggacucc 3240

ggucuauuuu cugcacccac uguuccguua uccauuagga auaaucacug ggauaacucc 3300

ccgucgccua acauguacgg gcugaauaaa gaaguggucc gucagcucuc ucgcagguac 3360

ccacaacugc cucgggcagu ugccacuggu agagucuaug acaugaacac ugguacacug 3420

cgcaauuaug auccgcgcau aaaccuagua ccuguaaaca gaagacugcc ucaugcuuua 3480

guccuccacc auaaugaaca cccacagagu gacuuuucuu cauucgucag caaauugaag 3540

ggcagaacug uccugguggu cggggaaaag uuguccgucc caggcaaaau gguugacugg 3600

uugucagacc ggccugaggc uaccuucaga gcucggcugg auuuaggcau cccaggugau 3660

gugcccaaau augacauaau auuuguuaau gugaggaccc cauauaaaua ccaucacuau 3720

cagcagugug aagaccaugc cauuaagcua agcauguuga ccaagaaagc augucugcau 3780

cugaaucccg gcggaaccug ugucagcaua gguuaugguu acgcugacag ggccagcgaa 3840

agcaucauug gugcuauagc gcggcaguuc aaguuuuccc gaguaugcaa accgaaaucc 3900

ucacuugagg agacggaagu ucuguuugua uucauugggu acgaucgcaa ggcccguacg 3960

cacaauccuu acaagcuauc aucaaccuug accaacauuu auacagguuc cagacuccac 4020

gaagccggau gugcacccuc auaucaugug gugcgagggg auauugccac ggccaccgaa 4080

ggagugauua uaaaugcugc uaacagcaaa ggacaaccug gcggaggggu gugcggagcg 4140

cuguauaaga aauucccgga aaguuucgau uuacagccga ucgaaguagg aaaagcgcga 4200

cuggucaaag gugcagcuaa acauaucauu caugccguag gaccaaacuu caacaaaguu 4260

ucggagguug aaggugacaa acaguuggca gaggcuuaug aguccaucgc uaagauuguc 4320

aacgauaaca auuacaaguc aguagcgauu ccacuguugu ccaccggcau cuuuuccggg 4380

aacaaagauc gacuaaccca aucauugaac cauuugcuga cagcuuuaga caccacugau 4440

gcagauguag ccauauacug cagggacaag aaaugggaaa ugacucucaa ggaagcagug 4500

gcuaggagag aagcagugga ggagauaugc auauccgacg auucuucagu gacagaaccu 4560

gaugcagagc uggugagggu gcaucccaag aguucuuugg cuggaaggaa gggcuacagc 4620

acaagcgaug gcaaaacuuu cucauauuug gaagggacca aguuucacca ggcggccaag 4680

gauauagcag aaauuaaugc cauguggccc guugcaacgg aggccaauga gcagguaugc 4740

auguauaucc ucggagaaag caugagcagu auuaggucga aaugccccgu cgaggagucg 4800

gaagccucca caccaccuag cacgcugccu ugcuugugca uccaugccau gacuccagaa 4860

agaguacagc gccuaaaagc cucacgucca gaacaaauua cugugugcuc auccuuucca 4920

uugccgaagu auagaaucac uggugugcag aagauccaau gcucccagcc uauauuguuc 4980

ucaccgaaag ugccugcgua uauucaucca aggaaguauc ucguggaaac accaccggua 5040

gacgagacuc cggagccauc ggcagagaac caauccacag aggggacacc ugaacaacca 5100

ccacuuauaa ccgaggauga gaccaggacu agaacgccug agccgaucau caucgaagaa 5160

gaagaagaag auagcauaag uuugcuguca gauggcccga cccaccaggu gcugcaaguc 5220

gaggcagaca uucacgggcc gcccucugua ucuagcucau ccugguccau uccucaugca 5280

uccgacuuug auguggacag uuuauccaua cuugacaccc uggagggagc uagcgugacc 5340

agcggggcaa cgucagccga gacuaacucu uacuucgcaa agaguaugga guuucuggcg 5400

cgaccggugc cugcgccucg aacaguauuc aggaacccuc cacaucccgc uccgcgcaca 5460

agaacaccgu cacuugcacc cagcagggcc ugcuccagaa ccagccuagu uuccaccccg 5520

ccaggcguga auagggugau cacuagagag gagcucgaag cgcuuacccc gucacgcacu 5580

ccuagcaggu cggucuccag aaccagccug gucuccaacc cgccaggcgu aaauagggug 5640

auuacaagag aggaguuuga ggcguucgua gcacaacaac aaugacgguu ugaugcgggu 5700

gcauacaucu uuuccuccga caccggucaa gggcauuuac aacaaaaauc aguaaggcaa 5760

acggugcuau ccgaaguggu guuggagagg accgaauugg agauuucgua ugccccgcgc 5820

cucgaccaag aaaaagaaga auuacuacgc aagaaauuac aguuaaaucc cacaccugcu 5880

aacagaagca gauaccaguc caggaaggug gagaacauga aagccauaac agcuagacgu 5940

auucugcaag gccuagggca uuauuugaag gcagaaggaa aaguggagug cuaccgaacc 6000

cugcauccug uuccuuugua uucaucuagu gugaaccgug ccuuuucaag ccccaagguc 6060

gcaguggaag ccuguaacgc cauguugaaa gagaacuuuc cgacuguggc uucuuacugu 6120

auuauuccag aguacgaugc cuauuuggac augguugacg gagcuucaug cugcuuagac 6180

acugccaguu uuugcccugc aaagcugcgc agcuuuccaa agaaacacuc cuauuuggaa 6240

cccacaauac gaucggcagu gccuucagcg auccagaaca cgcuccagaa cguccuggca 6300

gcugccacaa aaagaaauug caaugucacg caaaugagag aauugcccgu auuggauucg 6360

gcggccuuua auguggaaug cuucaagaaa uaugcgugua auaaugaaua uugggaaacg 6420

uuuaaagaaa accccaucag gcuuacugaa gaaaacgugg uaaauuacau uaccaaauua 6480

aaaggaccaa aagcugcugc ucuuuuugcg aagacacaua auuugaauau guugcaggac 6540

auaccaaugg acagguuugu aauggacuua aagagagacg ugaaagugac uccaggaaca 6600

aaacauacug aagaacggcc caagguacag gugauccagg cugccgaucc gcuagcaaca 6660

gcguaucugu gcggaaucca ccgagagcug guuaggagau uaaaugcggu ccugcuuccg 6720

aacauucaua cacuguuuga uaugucggcu gaagacuuug acgcuauuau agccgagcac 6780

uuccagccug gggauugugu ucuggaaacu gacaucgcgu cguuugauaa aagugaggac 6840

gacgccaugg cucugaccgc guuaaugauu cuggaagacu uaggugugga cgcagagcug 6900

uugacgcuga uugaggcggc uuucggcgaa auuucaucaa uacauuugcc cacuaaaacu 6960

aaauuuaaau ucggagccau gaugaaaucu ggaauguucc ucacacuguu ugugaacaca 7020

gucauuaaca uuguaaucgc aagcagagug uugagagaac ggcuaaccgg aucaccaugu 7080

gcagcauuca uuggagauga caauaucgug aaaggaguca aaucggacaa auuaauggca 7140

gacaggugcg ccaccugguu gaauauggaa gucaagauua uagaugcugu ggugggcgag 7200

aaagcgccuu auuucugugg aggguuuauu uugugugacu ccgugaccgg cacagcgugc 7260

cguguggcag acccccuaaa aaggcuguuu aagcuaggca aaccucuggc agcagacgau 7320

gaacaugaug augacaggag aagggcauug caugaggagu caacacgcug gaaccgagug 7380

gguauucuuu cagagcugug caaggcagua gaaucaaggu augaaaccgu aggaacuucc 7440

aucauaguua uggccaugac uacucuagcu agcaguguua aaucauucag cuaccugaga 7500

ggggccccua uaacucucua cggcuaaccu gaauggacua cgacauaguc uaguccgcca 7560

agacuaguau guuuguguuu cuugugcugc ugccucuugu gucuucucag ugugugguga 7620

gauuuccaaa uauuacaaau cuguguccau uuggagaagu guuuaaugca acaagauuug 7680

caucugugua ugcauggaau agaaaaagaa uuucuaauug uguggcugau uauucugugc 7740

uguauaauag ugcuucuuuu uccacauuua aauguuaugg agugucucca acaaaauuaa 7800

augauuuaug uuuuacaaau guguaugcug auucuuuugu gaucagaggu gaugaaguga 7860

gacagauugc ccccggacag acaggaaaaa uugcugauua caauuacaaa cugccugaug 7920

auuuuacagg augugugauu gcuuggaauu cuaauaauuu agauucuaaa gugggaggaa 7980

auuacaauua ucuguacaga cuguuuagaa aaucaaaucu gaaaccuuuu gaaagagaua 8040

uuucaacaga aauuuaucag gcuggaucaa caccuuguaa uggaguggaa ggauuuaauu 8100

guuauuuucc auuacagagc uauggauuuc agccaaccaa ugguguggga uaucagccau 8160

auagaguggu ggugcugucu uuugaacugc ugcaugcacc ugcaacagug uguggaccua 8220

aaggcucccc cggcuccggc uccggaucug guuauauucc ugaagcucca agagaugggc 8280

aagcuuacgu ucguaaagau ggcgaauggg uauuacuuuc uaccuuuuua ggaagcggca 8340

gcggaucuga acaguacauu aaauggccuu gguacauuug gcuuggauuu auugcaggau 8400

uaauugcaau ugugauggug acaauuaugu uauguuguau gacaucaugu uguucuuguu 8460

uaaaaggaug uuguucuugu ggaagcuguu guaaauuuga ugaagaugau ucugaaccug 8520

uguuaaaagg agugaaauug cauuacacau gaugacucga gcugguacug caugcacgca 8580

augcuagcug ccccuuuccc guccugggua ccccgagucu cccccgaccu cgggucccag 8640

guaugcuccc accuccaccu gccccacuca ccaccucugc uaguuccaga caccucccaa 8700

gcacgcagca augcagcuca aaacgcuuag ccuagccaca cccccacggg aaacagcagu 8760

gauuaaccuu uagcaauaaa cgaaaguuua acuaagcuau acuaacccca ggguugguca 8820

auuucgugcc agccacaccg cggccgcaug aauacagcag caauuggcaa gcugcuuaca 8880

uagaacucgc ggcgauuggc augccgccuu aaaauuuuua uuuuauuuuu ucuuuucuuu 8940

uccgaaucgg auuuuguuuu uaauauuuca aaaaaaaaaa aaaaaaaaaa aaaaaaaaag 9000

cauaugacua aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 9060

aaaaaaaaaa aaaaaaaaa 9079

<210> 28

<211> 327

<212> PRT

<213> Artificial sequence

<220>

<223> vaccine antigens

<400> 28

Met Phe Val Phe Leu Val Leu Leu Pro Leu Val Ser Ser Gln Cys Val

1 5 10 15

Val Arg Phe Pro Asn Ile Thr Asn Leu Cys Pro Phe Gly Glu Val Phe

20 25 30

Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala Trp Asn Arg Lys Arg Ile

35 40 45

Ser Asn Cys Val Ala Asp Tyr Ser Val Leu Tyr Asn Ser Ala Ser Phe

50 55 60

Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro Thr Lys Leu Asn Asp Leu

65 70 75 80

Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe Val Ile Arg Gly Asp Glu

85 90 95

Val Arg Gln Ile Ala Pro Gly Gln Thr Gly Lys Ile Ala Asp Tyr Asn

100 105 110

Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys Val Ile Ala Trp Asn Ser

115 120 125

Asn Asn Leu Asp Ser Lys Val Gly Gly Asn Tyr Asn Tyr Leu Tyr Arg

130 135 140

Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe Glu Arg Asp Ile Ser Thr

145 150 155 160

Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys Asn Gly Val Glu Gly Phe

165 170 175

Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly Phe Gln Pro Thr Asn Gly

180 185 190

Val Gly Tyr Gln Pro Tyr Arg Val Val Val Leu Ser Phe Glu Leu Leu

195 200 205

His Ala Pro Ala Thr Val Cys Gly Pro Lys Gly Ser Pro Gly Ser Gly

210 215 220

Ser Gly Ser Gly Tyr Ile Pro Glu Ala Pro Arg Asp Gly Gln Ala Tyr

225 230 235 240

Val Arg Lys Asp Gly Glu Trp Val Leu Leu Ser Thr Phe Leu Gly Ser

245 250 255

Gly Ser Gly Ser Glu Gln Tyr Ile Lys Trp Pro Trp Tyr Ile Trp Leu

260 265 270

Gly Phe Ile Ala Gly Leu Ile Ala Ile Val Met Val Thr Ile Met Leu

275 280 285

Cys Cys Met Thr Ser Cys Cys Ser Cys Leu Lys Gly Cys Cys Ser Cys

290 295 300

Gly Ser Cys Cys Lys Phe Asp Glu Asp Asp Ser Glu Pro Val Leu Lys

305 310 315 320

Gly Val Lys Leu His Tyr Thr

325

<210> 29

<211> 311

<212> PRT

<213> Artificial sequence

<220>

<223> BNT162b3c

<400> 29

Met Phe Val Phe Leu Val Leu Leu Pro Leu Val Ser Ser Gln Cys Val

1 5 10 15

Asn Leu Thr Val Arg Phe Pro Asn Ile Thr Asn Leu Cys Pro Phe Gly

20 25 30

Glu Val Phe Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala Trp Asn Arg

35 40 45

Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser Val Leu Tyr Asn Ser

50 55 60

Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro Thr Lys Leu

65 70 75 80

Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe Val Ile Arg

85 90 95

Gly Asp Glu Val Arg Gln Ile Ala Pro Gly Gln Thr Gly Lys Ile Ala

100 105 110

Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys Val Ile Ala

115 120 125

Trp Asn Ser Asn Asn Leu Asp Ser Lys Val Gly Gly Asn Tyr Asn Tyr

130 135 140

Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe Glu Arg Asp

145 150 155 160

Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys Asn Gly Val

165 170 175

Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly Phe Gln Pro

180 185 190

Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val Val Val Leu Ser Phe

195 200 205

Glu Leu Leu His Ala Pro Ala Thr Val Cys Gly Pro Lys Gly Ser Pro

210 215 220

Gly Ser Gly Ser Gly Ser Gly Tyr Ile Pro Glu Ala Pro Arg Asp Gly

225 230 235 240

Gln Ala Tyr Val Arg Lys Asp Gly Glu Trp Val Leu Leu Ser Thr Phe

245 250 255

Leu Gly Ser Gly Ser Gly Ser Glu Gln Tyr Ile Lys Trp Pro Trp Tyr

260 265 270

Ile Trp Leu Gly Phe Ile Ala Gly Leu Ile Ala Ile Val Met Val Thr

275 280 285

Ile Met Leu Cys Cys Met Thr Ser Cys Cys Ser Cys Leu Lys Gly Cys

290 295 300

Cys Ser Cys Gly Ser Cys Cys

305 310

<210> 30

<211> 1397

<212> RNA

<213> Artificial sequence

<220>

<223> BNT162b3c

<400> 30

agaauaaacu aguauucuuc ugguccccac agacucagag agaacccgcc accauguuug 60

uguuucuugu gcugcugccu cuugugucuu cucagugugu gaauuugaca gugagauuuc 120

caaauauuac aaaucugugu ccauuuggag aaguguuuaa ugcaacaaga uuugcaucug 180

uguaugcaug gaauagaaaa agaauuucua auuguguggc ugauuauucu gugcuguaua 240

auagugcuuc uuuuuccaca uuuaaauguu auggaguguc uccaacaaaa uuaaaugauu 300

uauguuuuac aaauguguau gcugauucuu uugugaucag aggugaugaa gugagacaga 360

uugcccccgg acagacagga aaaauugcug auuacaauua caaacugccu gaugauuuua 420

caggaugugu gauugcuugg aauucuaaua auuuagauuc uaaaguggga ggaaauuaca 480

auuaucugua cagacuguuu agaaaaucaa aucugaaacc uuuugaaaga gauauuucaa 540

cagaaauuua ucaggcugga ucaacaccuu guaauggagu ggaaggauuu aauuguuauu 600

uuccauuaca gagcuaugga uuucagccaa ccaauggugu gggauaucag ccauauagag 660

ugguggugcu gucuuuugaa cugcugcaug caccugcaac agugugugga ccuaaaggcu 720

cccccggcuc cggcuccgga ucugguuaua uuccugaagc uccaagagau gggcaagcuu 780

acguucguaa agauggcgaa uggguauuac uuucuaccuu uuuaggaagc ggcagcggau 840

cugaacagua cauuaaaugg ccuugguaca uuuggcuugg auuuauugca ggauuaauug 900

caauugugau ggugacaauu auguuauguu guaugacauc auguuguucu uguuuaaaag 960

gauguuguuc uuguggaagc uguuguugau gacucgagcu gguacugcau gcacgcaaug 1020

cuagcugccc cuuucccguc cuggguaccc cgagucuccc ccgaccucgg gucccaggua 1080

ugcucccacc uccaccugcc ccacucacca ccucugcuag uuccagacac cucccaagca 1140

cgcagcaaug cagcucaaaa cgcuuagccu agccacaccc ccacgggaaa cagcagugau 1200

uaaccuuuag caauaaacga aaguuuaacu aagcuauacu aaccccaggg uuggucaauu 1260

ucgugccagc cacacccugg agcuagcaaa aaaaaaaaaa aaaaaaaaaa aaaaaaagca 1320

uaugacuaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 1380

aaaaaaaaaa aaaaaaa 1397

<210> 31

<211> 314

<212> PRT

<213> Artificial sequence

<220>

<223> BNT162b3d

<400> 31

Met Asp Trp Ile Trp Arg Ile Leu Phe Leu Val Gly Ala Ala Thr Gly

1 5 10 15

Ala His Ser Gln Met Gln Val Arg Phe Pro Asn Ile Thr Asn Leu Cys

20 25 30

Pro Phe Gly Glu Val Phe Asn Ala Thr Arg Phe Ala Ser Val Tyr Ala

35 40 45

Trp Asn Arg Lys Arg Ile Ser Asn Cys Val Ala Asp Tyr Ser Val Leu

50 55 60

Tyr Asn Ser Ala Ser Phe Ser Thr Phe Lys Cys Tyr Gly Val Ser Pro

65 70 75 80

Thr Lys Leu Asn Asp Leu Cys Phe Thr Asn Val Tyr Ala Asp Ser Phe

85 90 95

Val Ile Arg Gly Asp Glu Val Arg Gln Ile Ala Pro Gly Gln Thr Gly

100 105 110

Lys Ile Ala Asp Tyr Asn Tyr Lys Leu Pro Asp Asp Phe Thr Gly Cys

115 120 125

Val Ile Ala Trp Asn Ser Asn Asn Leu Asp Ser Lys Val Gly Gly Asn

130 135 140

Tyr Asn Tyr Leu Tyr Arg Leu Phe Arg Lys Ser Asn Leu Lys Pro Phe

145 150 155 160

Glu Arg Asp Ile Ser Thr Glu Ile Tyr Gln Ala Gly Ser Thr Pro Cys

165 170 175

Asn Gly Val Glu Gly Phe Asn Cys Tyr Phe Pro Leu Gln Ser Tyr Gly

180 185 190

Phe Gln Pro Thr Asn Gly Val Gly Tyr Gln Pro Tyr Arg Val Val Val

195 200 205

Leu Ser Phe Glu Leu Leu His Ala Pro Ala Thr Val Cys Gly Pro Lys

210 215 220

Gly Ser Pro Gly Ser Gly Ser Gly Ser Gly Tyr Ile Pro Glu Ala Pro

225 230 235 240

Arg Asp Gly Gln Ala Tyr Val Arg Lys Asp Gly Glu Trp Val Leu Leu

245 250 255

Ser Thr Phe Leu Gly Ser Gly Ser Gly Ser Glu Gln Tyr Ile Lys Trp

260 265 270

Pro Trp Tyr Ile Trp Leu Gly Phe Ile Ala Gly Leu Ile Ala Ile Val

275 280 285

Met Val Thr Ile Met Leu Cys Cys Met Thr Ser Cys Cys Ser Cys Leu

290 295 300

Lys Gly Cys Cys Ser Cys Gly Ser Cys Cys

305 310

<210> 32

<211> 1406

<212> RNA

<213> Artificial sequence

<220>

<223> BNT162b3d

<400> 32

agaauaaacu aguauucuuc ugguccccac agacucagag agaacccgcc accauggauu 60

ggauuuggag aauccuguuc cucgugggag ccgcuacagg agcccacucc cagaugcagg 120

ugagauuucc aaauauuaca aaucuguguc cauuuggaga aguguuuaau gcaacaagau 180

uugcaucugu guaugcaugg aauagaaaaa gaauuucuaa uuguguggcu gauuauucug 240

ugcuguauaa uagugcuucu uuuuccacau uuaaauguua uggagugucu ccaacaaaau 300

uaaaugauuu auguuuuaca aauguguaug cugauucuuu ugugaucaga ggugaugaag 360

ugagacagau ugcccccgga cagacaggaa aaauugcuga uuacaauuac aaacugccug 420

augauuuuac aggaugugug auugcuugga auucuaauaa uuuagauucu aaagugggag 480

gaaauuacaa uuaucuguac agacuguuua gaaaaucaaa ucugaaaccu uuugaaagag 540

auauuucaac agaaauuuau caggcuggau caacaccuug uaauggagug gaaggauuua 600

auuguuauuu uccauuacag agcuauggau uucagccaac caauggugug ggauaucagc 660

cauauagagu gguggugcug ucuuuugaac ugcugcaugc accugcaaca guguguggac 720

cuaaaggcuc ccccggcucc ggcuccggau cugguuauau uccugaagcu ccaagagaug 780

ggcaagcuua cguucguaaa gauggcgaau ggguauuacu uucuaccuuu uuaggaagcg 840

gcagcggauc ugaacaguac auuaaauggc cuugguacau uuggcuugga uuuauugcag 900

gauuaauugc aauugugaug gugacaauua uguuauguug uaugacauca uguuguucuu 960

guuuaaaagg auguuguucu uguggaagcu guuguugaug acucgagcug guacugcaug 1020

cacgcaaugc uagcugcccc uuucccgucc uggguacccc gagucucccc cgaccucggg 1080

ucccagguau gcucccaccu ccaccugccc cacucaccac cucugcuagu uccagacacc 1140

ucccaagcac gcagcaaugc agcucaaaac gcuuagccua gccacacccc cacgggaaac 1200

agcagugauu aaccuuuagc aauaaacgaa aguuuaacua agcuauacua accccagggu 1260

uggucaauuu cgugccagcc acacccugga gcuagcaaaa aaaaaaaaaa aaaaaaaaaa 1320

aaaaaagcau augacuaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 1380

aaaaaaaaaa aaaaaaaaaa aaaaaa 1406

<210> 33

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> joint

<400> 33

Gly Ser Pro Gly Ser Gly Ser Gly Ser

1 5

<210> 34

<211> 6

<212> PRT

<213> Artificial sequence

<220>

<223> joint

<400> 34

Gly Ser Gly Ser Gly Ser

1 5

<210> 35

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> AH1

<400> 35

Ser Pro Ser Tyr Val Tyr His Gln Phe

1 5

<210> 36

<211> 20

<212> PRT

<213> Artificial sequence

<220>

<223> HuIgGk Signal peptide

<400> 36

Met Glu Thr Pro Ala Gln Leu Leu Phe Leu Leu Leu Leu Trp Leu Pro

1 5 10 15

Asp Thr Thr Gly

20

<210> 37

<211> 18

<212> PRT

<213> Artificial sequence

<220>

<223> IgE heavy chain epsilon-1 signal peptide

<400> 37

Met Asp Trp Thr Trp Ile Leu Phe Leu Val Ala Ala Ala Thr Arg Val

1 5 10 15

His Ser

<210> 38

<211> 24

<212> PRT

<213> Artificial sequence

<220>

<223> Japanese encephalitis PRM Signal sequence

<400> 38

Met Leu Gly Ser Asn Ser Gly Gln Arg Val Val Phe Thr Ile Leu Leu

1 5 10 15

Leu Leu Val Ala Pro Ala Tyr Ser

20

<210> 39

<211> 17

<212> PRT

<213> Artificial sequence

<220>

<223> VSVG protein Signal sequence

<400> 39

Met Lys Cys Leu Leu Tyr Leu Ala Phe Leu Phe Ile Gly Val Asn Cys

1 5 10 15

Ala

<210> 40

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 40

Tyr Leu Gln Pro Arg Thr Phe Leu Leu

1 5

<210> 41

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 41

Arg Leu Gln Ser Leu Gln Thr Tyr Val

1 5

<210> 42

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 42

Gln Tyr Ile Lys Trp Pro Trp Tyr Ile

1 5

<210> 43

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 43

Asn Tyr Asn Tyr Leu Tyr Arg Leu Phe

1 5

<210> 44

<211> 10

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 44

Lys Trp Pro Trp Tyr Ile Trp Leu Gly Phe

1 5 10

<210> 45

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 45

Gln Pro Thr Glu Ser Ile Val Arg Phe

1 5

<210> 46

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 46

Ile Pro Phe Ala Met Gln Met Ala Tyr

1 5

<210> 47

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 47

Leu Pro Phe Asn Asp Gly Val Tyr Phe

1 5

<210> 48

<211> 15

<212> DNA

<213> Artificial sequence

<220>

<223> nucleic acid fragment

<400> 48

cccactaatg gtgtt 15

<210> 49

<211> 5

<212> PRT

<213> Artificial sequence

<220>

<223> protein fragment

<400> 49

Pro Thr Asn Gly Val

1 5

<210> 50

<211> 15

<212> DNA

<213> Artificial sequence

<220>

<223> nucleic acid fragment

<400> 50

cccacttatg gtgtt 15

<210> 51

<211> 5

<212> PRT

<213> Artificial sequence

<220>

<223> protein fragment

<400> 51

Pro Thr Tyr Gly Val

1 5

<210> 52

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 52

Gly Val Tyr Phe Ala Ser Thr Glu Lys

1 5

<210> 53

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 53

Cys Val Ala Asp Tyr Ser Val Leu Tyr

1 5

<210> 54

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 54

Lys Cys Tyr Gly Val Ser Pro Thr Lys

1 5

<210> 55

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 55

Phe Gln Pro Thr Asn Gly Val Gly Tyr

1 5

<210> 56

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 56

Gly Thr His Trp Phe Val Thr Gln Arg

1 5

<210> 57

<211> 9

<212> PRT

<213> Artificial sequence

<220>

<223> epitope

<400> 57

Val Tyr Asp Pro Leu Gln Pro Glu Leu

1 5

612页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:冠状病毒疫苗

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!