Vaccine Compositions For Preventing Coronavirus Disease

Abstract
The present disclosure provides Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) vaccines, recombinant vesicular stomatitis virus (VSV) vectors encoding the SARS-CoV-2 spike (S) protein or an immunogenic variant thereof, recombinant replicable VSV particles having a SARS-CoV-2 S protein or an immunogenic variant thereof on the surface of the particles, and immunogenic recombinant proteins comprising a SARS-CoV-2 S protein or a variant thereof. Immunogenic compositions comprising the SARS-CoV-2 vaccines, the recombinant VSV vectors, the recombinant replicable VSV particles and/or the immunogenic recombinant proteins may be used for inducing an immune response to the SARS-CoV-2, preventing infection by the SARS-CoV-2, vaccinating against the SARS-CoV-2 and/or producing adaptive mutants of the recombinant replicable VSV particles.
Description
FIELD

The present disclosure relates to recombinant vesicular stomatitis virus (VSV) for use as vaccines for infectious coronavirus disease COVID-19.


BACKGROUND

The recent outbreak of coronavirus disease COVID-19 is caused by a novel coronavirus 2019-nCoV, which is officially named Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) and sometimes referred to as the Wuhan coronavirus. Since the report of the first COVID-19 case in Wuhan, China, in November 2019, 27.9 million COVID-19 cases have been confirmed globally with more than 898,000 reported deaths as of Sep. 10, 2020. As a member of the large coronavirus family that causes sickness from the common cold to more severe diseases such as the Severe Acute Respiratory Syndrome (SARS) and the Middle East Respiratory Syndrome (MERS), the SARS-CoV-2 is a coronavirus that was not found in humans before this outbreak. Although infection with SARS-CoV-2 does not appear to be as deadly, with respect to case fatality rate, as infection with the SARS or MERS viruses, SARS-CoV-2 appears to be more infectious and able to be transmitted from infected people before the onset of symptoms. As a result, it is spreading far wider and more quickly than SARS or MERS and, as a result, is now responsible for the deaths of far more people worldwide. Some scientists have predicted that the SARS-CoV-2 may become endemic in human populations and potentially return every winter like other respiratory pathogens such as influenza. Many leading scientists believe that the infection will only be eradicated from human populations when a safe and effective vaccine to prevent SARS-CoV-2 infection is available.


The SARS-CoV-2 is genetically related to coronaviruses that infect bats. Like many other highly pathogenic viruses that originate from animal reservoirs, such as those that cause outbreaks of hemorrhagic fever like Ebola virus (EBOV), Lassa virus (LASV), and Marburg virus (MARV), the SARS-CoV-2 is an enveloped RNA virus. Viruses in this highly diverse category all have one or more multimeric glycoproteins exposed on their surface that play essential roles in infection of the host, notably during cell attachment and virus entry. Glycoproteins also are known to be the primary targets for protective immunity.


Sequences of more than seventy (70) SARS-CoV-2 variants have been published. Structural studies of these published sequences have been reported. It is believed that SARS-CoV-2 expresses a spike (S) protein that contains a receptor-binding domain (RBD), including receptor-binding motif (RBM), and interacts with host receptor angiotensin-converting enzyme 2 (ACE2). See Zhou, P. et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin, Nature 579:270-273 (published online 3 Feb. 2020); see also Wan, Y. et al. Receptor recognition by novel coronavirus from Wuhan: An analysis based on decade-long structural studies of SARS, J. Virol. 94(7): e00127-20, doi:10.1128/JVI.00127-20 (published online 29 Jan. 2020). The S protein comprises two subunits S1 (the surface unit, which binds the receptor) and S2 (the transmembrane unit, which facilitates viral fusion to cell membranes). The S protein is activated by cleavage at the spike S1/S2 site by host cell proteases and SARS-CoV-2 has a newly formed Furin cleavage site at the S1/S2 interface.


There remains a need to identify immunogenic antigens derived from the SARS-CoV-2 and develop vaccine compositions that are able to be produced at large scale using available manufacturing processes that stably express such immunogenic antigens for inducing relevant immune responses in vaccinated individuals to enable them to be protected against COVID-19 and that are able to be produced at large scale using available manufacturing processes.


SUMMARY

The present disclosure relates to vectors, virus particles, immunogenic recombinant proteins, and vaccines that relate to Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2). The disclosure includes multiple embodiments, including, but not limited to, the following embodiments.


Embodiment 1 is a recombinant VSV vector comprising:

    • a. at least a portion of the VSV genome comprising at least a VSV-N gene, VSV-P gene, VSV-M gene, and VSV-L gene, and
    • b. a nucleic acid sequence encoding a SARS-CoV-2 S protein or an immunogenic variant thereof.


Embodiment 2 is a recombinant VSV particle comprising the vector of embodiment 1 and displaying the S protein or an immunogenic variant thereof on the surface of the VSV particle.


Embodiment 3 is a recombinant VSV particle comprising:

    • a. at least a portion of the VSV genome comprising N, P, M, and L genes and
    • b. a nucleic acid sequence encoding a SARS-CoV-2 S protein or an immunogenic variant thereof.


Embodiment 4 is the recombinant VSV particle of embodiment 2 or 3, wherein the VSV particle is replicable.


Embodiment 5 is the recombinant VSV particle of any one of embodiments 3-4, wherein the VSV particle displays the S protein or an immunogenic variant thereof on the surface of the VSV particle.


Embodiment 6 is an immunogenic recombinant protein comprising a SARS-CoV-2 S protein or an immunogenic variant thereof expressed by the recombinant VSV vector of embodiment 1 or recombinant VSV particle of any one of embodiments 2-5.


Embodiment 7 is an immunogenic recombinant protein comprising a SARS-CoV-2 S protein or an immunogenic variant thereof and at least a fragment of a VSV glycoprotein (G).


Embodiment 8 is a SARS-CoV-2 vaccine comprising the recombinant VSV vector of embodiment 1, recombinant VSV particle of any one of embodiments 2-5, or immunogenic recombinant protein of embodiment 6 or 7.


Embodiment 9 is the recombinant VSV vector of embodiment 1, recombinant VSV particle of any one of embodiments 2-5, immunogenic recombinant protein of embodiment 6 or 7, or SARS-CoV-2 vaccine of embodiment 8, wherein the SARS-CoV-2 S protein or an immunogenic variant thereof comprises an amino acid sequence having a length of at least 1223, 1228, 1233, 1238, 1243, 1248, 1249, 1250, 1251, 1252, 1253, 1254, 1255, 1256, 1257, 1258, 1259, 1260, 1261, 1262, 1263, 1264, 1265, 1266, 1267, 1268, 1269, 1270, 1271, 1272, 1273, 1274, 1275, 1276, 1277 or 1278 amino acids and having homology over its own length of at least 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% homology to any one of SEQ ID NOS: 1, 4, 7, 10, 13, 16, 19, 22, 25-67, 153, or 158.


Embodiment 10 is the recombinant VSV vector of embodiment 1 or 9, recombinant VSV particle of any one of embodiments 2-5 or 9, or SARS-CoV-2 vaccine of embodiment 8 or 9, wherein the VSV genome comprises at least a fragment of a VSV-G gene.


Embodiment 11 is the recombinant VSV vector of any one of embodiments 1, 9, or 10, recombinant VSV particle of any one of embodiments 2-5, 9 or 10, immunogenic recombinant protein of any one of embodiments 6, 7, or 9, or SARS-CoV-2 vaccine of any one of embodiments 8-10, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein having a deletion at the C-terminal end of the SARS-CoV-2 S protein.


Embodiment 12 is the recombinant VSV vector of any one of embodiments 1, or 9-11, recombinant VSV particle of any one of embodiments 2-5 or 9-11, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11, or SARS-CoV-2 vaccine of any one of embodiments 8-11, wherein the immunogenic variant of the SARS-CoV-2 S protein comprises a fragment of the SARS-CoV-2 S protein having a deletion of from 5-25 or 9-23 amino acids at the C-terminal end of the SARS-CoV-2 S protein.


Embodiment 13 is the recombinant VSV vector of any one of embodiments 1, or 9-12, recombinant VSV particle of any one of embodiments 2-5 or 9-12, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11, or 12, or SARS-CoV-2 vaccine of any one of embodiments 8-12, wherein the immunogenic variant of the SARS-CoV-2 S protein comprises a fragment of the SARS-CoV-2 S protein having a deletion of 9, 13, 19, 21 or 23 amino acids at the C-terminal end of the SARS-CoV-2 S protein.


Embodiment 14 is the recombinant VSV vector of any one of embodiments 1 or 9-13, recombinant VSV particle of any one of embodiments 2-5 or 9-13, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-13, or SARS-CoV-2 vaccine of any one of embodiments 8-13, wherein (a) the portion of the VSV genome comprises a nucleic acid sequence encoding at least 21 amino acids at the C-terminal end of the VSV-G protein or (b) the immunogenic recombinant protein comprises at least 21 amino acids at the C-terminal end of the VSV-G protein.


Embodiment 15 is the recombinant VSV vector of any one of embodiments 1 or 9-14, recombinant VSV particle of any one of embodiments 2-5 or 9-14, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-14, or SARS-CoV-2 vaccine of any one of embodiments 8-14, wherein (a) the portion of the VSV genome comprises a nucleic acid sequence encoding at least 29 amino acids at the C-terminal end of the VSV-G protein or (b) the immunogenic recombinant protein comprises at least 29 amino acids at the C-terminal end of the VSV-G protein.


Embodiment 16 is the recombinant VSV vector of any one of embodiments 1 or 9-15, recombinant VSV particle of any one of embodiments 2-5 or 9-15, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-15, or SARS-CoV-2 vaccine of any one of embodiments 8-15, wherein the immunogenic variant of the SARS-CoV-2 S protein comprises at least one mutation in the exposed loop (a solvent-exposed loop that comprises the S1/S2 cleavage site) of the SARS-CoV-2 S protein.


Embodiment 17 is the recombinant VSV vector of any one of embodiments 1 or 9-16, recombinant VSV particle of any one of embodiments 2-5 or 9-16, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-16, or SARS-CoV-2 vaccine of any one of embodiments 8-16, wherein the immunogenic variant of the SARS-CoV-2 S protein comprises at least one mutation in the S1/S2 cleavage site (Furin).


Embodiment 18 is the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of embodiment 17, wherein the at least one mutation in the S1/S2 cleavage site (Furin) reduces or blocks S1/S2 cleavage of the immunogenic variant of the SARS-CoV-2 S protein as compared with that of wild-type SARS-CoV-2 S protein.


Embodiment 19 is the recombinant VSV vector of any one of embodiments 1 or 9-16, recombinant VSV particle of any one of embodiments 2-5 or 9-16, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-16, or SARS-CoV-2 vaccine of any one of embodiments 8-16, wherein the immunogenic variant of the SARS-CoV-2 protein comprises at least one mutation in the S2′ cleavage site (Cathepsin H, L).


Embodiment 20 is the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of embodiment 19, wherein the at least one mutation in the S2′ cleavage site (Cathepsin H, L) modulates S2′ cleavage of the immunogenic variant of the SARS-CoV-2 S protein as compared with that of wild-type SARS-CoV-2 S S2′.


Embodiment 21 is the recombinant VSV vector of any one of embodiments 1 or 9-20, recombinant VSV particle of any one of embodiments 2-5 or 9-20, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-20, or SARS-CoV-2 vaccine of any one of embodiments 8-20, wherein the immunogenic variant of the SARS-CoV-2 protein comprises one or more mutations relative to SEQ ID NO: 1 chosen from a mutation at residue 655, one or more mutations from residue 672 to residue 687, one or more mutations from residue 802 to residue 817, one or more mutations from residue 1233 to residue 1273, and combinations thereof.


Embodiment 22 is the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of embodiment 21, comprising a mutation at residues 655, wherein the mutation at residue 655 is H655Y.


Embodiment 23 is the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of embodiment 21 or 22, comprising one or more mutations from residue 672 to 687, wherein the one or more mutations from residue 672 to residue 687 are from residue 678 to residue 685.


Embodiment 24 is the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of any one of embodiments 21-23, comprising one or more mutations from residue 672 to residue 687, wherein the one or more mutations from residue 678 to residue 685 are chosen from T678I, P681S, R682K, R683G, R685G, and combinations thereof.


Embodiment 25 is the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of any one of embodiments 21-24, comprising one more mutations from residue 802 to residue 817, wherein the one or more mutations from residue 802 to residue 817 in SEQ ID NO: 1 are from residue 810 to residue 815.


Embodiment 26 is the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of any one of embodiments 21-25, comprising one or more mutations from residue 810 to residue 815, wherein the one or more mutations from residue 810 to residue 815 are chosen from P812R, S813R, S813F, and combinations thereof.


Embodiment 27 is the recombinant VSV vector of embodiment 1 or 9, recombinant VSV particle of any one of embodiments 2-5 or 9, or SARS-CoV-2 vaccine of embodiment 8 or 9, wherein:

    • a. the recombinant VSV vector is rVSVΔG-SARS-CoV-2 clone MB2, which has the nucleic acid sequence set forth in SEQ ID NO: 154; or
    • b. the recombinant VSV vector, recombinant VSV particle, or SARS-CoV-2 vaccine comprises nucleic acids encoding
      • i. a VSV L protein, which has the amino acid sequence set forth in SEQ ID NO: 152;
      • ii. a VSV N protein, which has the amino acid sequence set forth in SEQ ID NO: 155;
      • iii. a VSV P protein, which has the amino acid sequence set forth in SEQ ID NO: 156;
      • iv. a VSV M protein, which has the amino acid sequences set forth in SEQ ID NO: 157; and
      • v. a SARS-CoV-2 S protein, which has the amino acid sequence set forth in SEQ ID NO: 158.


Embodiment 28 is the recombinant VSV particle of any one of embodiments 2-5 or 9 or SARS-CoV-2 vaccine of embodiment 8 or 9, wherein the particle or vaccine comprises:

    • a. VSV L protein, which has the amino acid sequence set forth in SEQ ID NO: 152;
    • b. VSV N protein, which has the amino acid sequence set forth in SEQ ID NO: 155;
    • c. VSV P protein, which has the amino acid sequence set forth in SEQ ID NO: 156;
    • d. VSV M protein, which has the amino acid sequence set forth in SEQ ID NO: 157; and
    • e. SARS-CoV-2 S protein, which has the amino acid sequence set forth in SEQ ID NO: 158.


Embodiment 29 is the recombinant VSV vector of any one of embodiments 1, or 9-26, recombinant VSV particle of any one of embodiments 2-5 or 9-26, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-26, or SARS-CoV-2 vaccine of any one of embodiments 8-26, wherein the immunogenic variant of the S protein does not comprise the entire cytoplasmic tail.


Embodiment 30 is the recombinant VSV vector of any one of embodiments 1, 9-26, or 29, recombinant VSV particle of any one of embodiments 2-5, 9-26, or 29, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-26, or 29, or SARS-CoV-2 vaccine of any one of embodiments 8-26 or 29, wherein the immunogenic variant of the S protein does not comprise the endoplasmic reticulum retention sequence.


Embodiment 31 is the recombinant VSV vector of any one of embodiments 1, 9-26, 29, or 30, recombinant VSV particle of any one of embodiments 2-5, 9-26, 29, or 30, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-26, 29, or 30, or SARS-CoV-2 vaccine of any one of embodiments 8-26, 29, or 30, wherein the immunogenic variant of the S protein has a 23 amino acid deletion at the C-terminal domain relative to SEQ ID NO: 1.


Embodiment 32 is the recombinant VSV vector of any one of embodiments 1, 9-26, or 29-31, recombinant VSV particle of any one of embodiments 2-5, 9-26, or 29-31, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-26, or 29-31, or SARS-CoV-2 vaccine of any one of embodiments 8-26 or 29-31, wherein the immunogenic variant of the S protein has a deletion at position 1251 relative to SEQ ID NO: 1.


Embodiment 33 is the recombinant VSV vector of any one of embodiments 1, 9-26, or 29-32, recombinant VSV particle of any one of embodiments 2-5, 9-26, or 29-32, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-26, or 29-32, or SARS-CoV-2 vaccine of any one of embodiments 8-26 or 29-32, wherein the immunogenic variant of the S protein comprises an H655Y mutation relative to SEQ ID NO: 1.


Embodiment 34 is the recombinant VSV vector of any one of embodiments 1, 9-26, or 29-33, recombinant VSV particle of any one of embodiments 2-5, 9-26, or 29-33, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-26, or 29-33, or SARS-CoV-2 vaccine of any one of embodiments 8-26 or 29-33, wherein the immunogenic variant of the SARS-CoV-2 S protein comprises one or more mutations from residue 1233 to residue 1273 in SEQ ID NO: 1, wherein the one or more mutations from residue 1233 to residue 1273 are chosen from a deletion at the C-terminal end of the SEQ ID NO: 1, M1233K, and a combination thereof.


Embodiment 35 is the recombinant VSV vector of claim 1 or 9, recombinant VSV particle of any one of claims 2-5 or 9, or SARS-CoV-2 vaccine of claim 8 or 9, wherein:

    • a. the recombinant VSV vector is rVSVΔG-SARS-CoV-2 clone MB1, which has the nucleic acid sequence set forth in SEQ ID NO: 150; or
    • b. the recombinant VSV vector, recombinant VSV particle, or SARS-CoV-2 vaccine comprises nucleic acids encoding
      • i. VSV L protein, which has the amino acid sequence set forth in SEQ ID NO: 152;
      • ii. VSV N protein, which has the amino acid sequence set forth in SEQ ID NO: 155;
      • iii. VSV P protein, which has the amino acid sequence set forth in SEQ ID NO: 156;
      • iv. VSV M protein, which has the amino acid sequence set forth in SEQ ID NO: 153; and
      • v. SARS-CoV-2 S protein, which has the amino acid sequence set forth in SEQ ID NO: 151.


Embodiment 36 is the recombinant VSV particle of any one of claims 2-5 or 9 or SARS-CoV-2 vaccine of claim 8 or 9, wherein the particle or vaccine comprises VSV and SARS-CoV-2 proteins from MB1 comprising:

    • a. VSV L protein, which has the amino acid sequence set forth in SEQ ID NO: 152;
    • b. VSV N protein, which has the amino acid sequence set forth in SEQ ID NO: 155;
    • c. VSV P protein, which has the amino acid sequence set forth in SEQ ID NO: 156;
    • d. VSV M protein, which has the amino acid sequence set forth in SEQ ID NO: 151; and
    • e. SARS-CoV-2 S protein, which has the amino acid sequence set forth in SEQ ID NO: 153.


Embodiment 37 is the recombinant VSV vector of any one of embodiments 1, 9-28, or 30-34, recombinant VSV particle of any one of embodiments 2-5, 9-28, or 30-34, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-28, or 30-34, or SARS-CoV-2 vaccine of any one of embodiments 8-28 or 30-34, wherein the immunogenic variant of the S protein does not comprise the entire cytoplasmic tail (i.e., does not comprise the entire cytoplasmic tail of the S protein).


Embodiment 38 is the recombinant VSV vector of any one of embodiments 1, 9-29, 31-34, or 37, recombinant VSV particle of any one of embodiments 2-5, 9-29, 31-34, or 37, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-29, 31-34, or 37, or SARS-CoV-2 vaccine of any one of embodiments 8-29, 31-34, or 37, wherein the immunogenic variant of the S protein does not comprise the endoplasmic reticulum retention sequence (i.e., does not comprise the endoplasmic reticulum retention sequence of the SARS-CoV-2 S protein).


Embodiment 39 is the recombinant VSV vector of any one of embodiments 1, 9-30, 32-34, 37, or 38, recombinant VSV particle of any one of embodiments 2-5, 9-30, 32-34, 37, or 38, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-30, 32-34, 37, or 38, or SARS-CoV-2 vaccine of any one of embodiments 8-30, 32-34, 37, or 38, wherein the immunogenic variant of the S protein has a 23 amino acid deletion at the C-terminal domain.


Embodiment 40 is the recombinant VSV vector of any one of embodiments 1, 9-31, 33, 34, or 37-39, recombinant VSV particle of any one of embodiments 2-5, 9-31, 33, 34, or 37-39, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-31, 33, 34, or 37-39, or SARS-CoV-2 vaccine of any one of embodiments 8-31, 33, 34, or 37-39, wherein the immunogenic variant of the S protein has a deletion at position 1251.


Embodiment 41 is the recombinant VSV vector of any one of embodiments 1, 9-34, or 37-40, recombinant VSV particle of any one of embodiments 2-5, 9-34, or 37-40, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-34, or 37-40, or SARS-CoV-2 vaccine of any one of embodiments 8-34, or 37-40, wherein the immunogenic variant of the S protein comprises an R683G mutation relative to SEQ ID NO: 1.


Embodiment 42 is the recombinant VSV vector of any one of embodiments 1, 9-41, recombinant VSV particle of any one of embodiments 2-5, 9-41, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-41, or SARS-CoV-2 vaccine of any one of embodiments 8-41, wherein the immunogenic variant of the S protein does not comprise a mutation at R685 relative to SEQ ID NO: 1.


Embodiment 43 is the recombinant VSV vector of any one of embodiments 1, 9-42, recombinant VSV particle of any one of embodiments 2-5, 9-42, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-42, or SARS-CoV-2 vaccine of any one of embodiments 8-42, wherein the immunogenic variant of the S protein does not have a 24 amino acid deletion at the C-terminal domain.


Embodiment 44 is the recombinant VSV vector of any one of embodiments 1, 9-42, recombinant VSV particle of any one of embodiments 2-5, 9-42, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-42, or SARS-CoV-2 vaccine of any one of embodiments 8-42, wherein the immunogenic variant of the S protein does not comprise a 21 amino acid deletion at the C-terminal cytoplasmic domain.


Embodiment 45 is the recombinant VSV vector of any one of embodiments 1, 9-44, recombinant VSV particle of any one of embodiments 2-5, 9-44, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-44, or SARS-CoV-2 vaccine of any one of embodiments 8-44, wherein the immunogenic variant of the S protein has at least one mutation relative to SEQ ID NO: 1 along the length of the immunogenic variant.


Embodiment 46 is the recombinant VSV vector of any one of embodiments 1, 9-45, recombinant VSV particle of any one of embodiments 2-5, 9-45, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-45, or SARS-CoV-2 vaccine of any one of embodiments 8-45, wherein the immunogenic variant of the S protein comprises an S813F mutation relative to SEQ ID NO: 1.


Embodiment 47 is the recombinant VSV vector of any one of embodiments 1, 9-34, or 37-46, recombinant VSV particle of any one of embodiments 2-5, 9-34, or 37-46, immunogenic recombinant protein of any one of embodiments 6, 7, 9, 11-34, or 37-46, or SARS-CoV-2 vaccine of any one of embodiments 8-34 or 37-46, wherein the VSV-M protein comprises a Y61S mutation relative to SEQ ID NO: 1.


Embodiment 48 is the recombinant VSV vector of any one of embodiments 1 or 9-47, recombinant VSV particle of any one of embodiments 2-5 or 9-47, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-47, or SARS-CoV-2 vaccine of any one of embodiments 8-47, wherein the immunogenic variant of the SARS-CoV-2 S protein comprises at least one of the following mutations (either in isolation or in any combination): H655Y, R682K, R683G, N709S, S813F, N978K, S940G, D1118A, or D1163N relative to SEQ ID NO: 1.


Embodiment 49 is the recombinant VSV vector of any one of embodiments 1 or 9-48, recombinant VSV particle of any one of embodiments 2-5 or 9-48, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-48, or SARS-CoV-2 vaccine of any one of embodiments 8-48, wherein the immunogenic variant of the SARS-CoV-2 S protein comprises at least one mutation in SEQ ID NO: 1 chosen from F140V, Q321P, N715S, D1118A, and combinations thereof.


Embodiment 50 is the recombinant VSV vector of any one of embodiments 1 or 9-49, recombinant VSV particle of any one of embodiments 2-5 or 9-49, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-49, or SARS-CoV-2 vaccine of any one of embodiments 8-49, wherein the immunogenic variant of the SARS-CoV-2 S protein comprises one or more mutations relative to SEQ ID NO: 1 chosen from:

    • a. H655Y, R682K, and R685G;
    • b. H655Y, R682K, R685G, and a deletion of the 9 amino acids (Δ9) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • c. H655Y, R682K, R685G, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • d. Q321P, H655Y, T678I, and P812R;
    • e. Q321P, H655Y, T678I, P812R, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • f. E154D, S115L, T678I, R685G, and M1233K;
    • g. R685G, S813R, and a deletion of the 9 amino acids (Δ9) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • h. H655Y, R682K, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • i. R685G, S813F, and a deletion of the 9 amino acids (Δ9) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • j. R683G, S813F and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • k. R683G, S813F, R685G, and a deletion of 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • l. H655Y, P681S, R682K, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • m. R683G and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • n. H655Y and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • o. R683G, D1118A, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • p. R683G, N715S, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • q. H655Y, N709S, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • r. F140V, H655Y, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • s. a deletion of the 21 amino acids (Δ21) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • t. a deletion of the 19 amino acids (Δ19) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • u. E484D, H655Y, and a deletion of 21 amino acids (Δ21) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;
    • v. H665Y, R685G and a deletion of the 19 amino acids (Δ19) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1, wherein the SARS-CoV-2 S protein further comprises a fragment of at least 21 amino acids at the C-terminal end of a VSV-G protein;
    • w. D614G; or
    • x. D614N.


Embodiment 51 is the recombinant VSV vector of any one of embodiments 1 or 9-50, recombinant VSV particle of any one of embodiments 2-5 or 9-50, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-50, or SARS-CoV-2 vaccine of any one of embodiments 8-50, wherein the VSV-G protein comprises from 21 to 29 amino acids at the C-terminal end of a VSV-G protein.


Embodiment 52 is the recombinant VSV vector of any one of embodiments 1 or 9-51, recombinant VSV particle of any one of embodiments 2-5 or 9-51, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-51, or SARS-CoV-2 vaccine of any one of embodiments 8-51, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein that does not comprise any of the cytoplasmic tail.


Embodiment 53 is the recombinant VSV vector of any one of embodiments 1 or 9-52, recombinant VSV particle of any one of embodiments 2-5 or 9-52, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-52, or SARS-CoV-2 vaccine of any one of embodiments 8-52, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a transmembrane domain of the SARS-CoV-2 S protein.


Embodiment 54 is the recombinant VSV vector of any one of embodiments 1 or 9-53, recombinant VSV particle of any one of embodiments 2-5 or 9-53, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-53, or SARS-CoV-2 vaccine of any one of embodiments 8-53, wherein the fragment of the VSV-G gene encodes a cytoplasmic tail of a VSV-G protein.


Embodiment 55 is the recombinant VSV vector of any one of embodiments 1 or 9-54, recombinant VSV particle of any one of embodiments 2-5 or 9-54, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-54, or SARS-CoV-2 vaccine of any one of embodiments 8-54, wherein the fragment of the VSV-G gene encodes a transmembrane domain of a VSV-G protein.


Embodiment 56 is the recombinant VSV vector of any one of embodiments 1 or 9-55, recombinant VSV particle of any one of embodiments 2-5 or 9-55, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-55, or SARS-CoV-2 vaccine of any one of embodiments 8-56, wherein the at least a portion of the VSV genome comprises the VSV-N gene, VSV-P gene, VSV-M gene, and VSV-L gene arranged in sequence from 3′ to 5′.


Embodiment 57 is the recombinant VSV vector of any one of embodiments 1 or 9-56, recombinant VSV particle of any one of embodiments 2-5 or 9-56, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-56, or SARS-CoV-2 vaccine of any one of embodiments 8-56, wherein the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is 3′ of the VSV-N gene.


Embodiment 58 is the recombinant VSV vector of any one of embodiments 1 or 9-57, recombinant VSV particle of any one of embodiments 2-5 or 9-57, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-57, or SARS-CoV-2 vaccine of any one of embodiments 8-58, wherein the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is on the 3′ end of the VSV-N gene.


Embodiment 59 is the recombinant VSV vector of any one of embodiments 1 or 9-58, recombinant VSV particle of any one of embodiments 2-5 or 9-58, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-58, or SARS-CoV-2 vaccine of any one of embodiments 8-58, wherein the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is between the VSV-N gene and the VSV-P gene.


Embodiment 60 is the recombinant VSV vector of any one of embodiments 1 or 9-59, recombinant VSV particle of any one of embodiments 2-5 or 9-59, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-59, or SARS-CoV-2 vaccine of any one of embodiments 8-59, wherein the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is between the VSV-P gene and the VSV-M gene.


Embodiment 61 is the recombinant VSV vector of any one of embodiments 1 or 9-60, recombinant VSV particle of any one of embodiments 2-5 or 9-60, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-60, or SARS-CoV-2 vaccine of any one of embodiments 8-60, wherein the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is between the VSV-M gene and the VSV-L gene.


Embodiment 62 is the recombinant VSV vector of any one of embodiments 1 or 9-61, recombinant VSV particle of any one of embodiments 2-5 or 9-61, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-61, or SARS-CoV-2 vaccine of any one of embodiments 8-61, wherein the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is 5′ of the VSV-L gene.


Embodiment 63 is the recombinant VSV vector of any one of embodiments 1 or 9-62, recombinant VSV particle of any one of embodiments 2-5 or 9-62, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-62, or SARS-CoV-2 vaccine of any one of embodiments 8-62, wherein the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is on the 5′ end of the VSV-L gene.


Embodiment 64 is the recombinant VSV vector of any one of embodiments 1 or 9-63, recombinant VSV particle of any one of embodiments 2-5 or 9-63, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-63, or SARS-CoV-2 vaccine of any one of embodiments 8-63, the VSV genome does not comprise a VSV-G gene or a fragment thereof.


Embodiment 65 is the SARS-CoV-2 vaccine of any one of embodiments 8-64, further comprising a pharmaceutically acceptable excipient.


Embodiment 66 is the SARS-CoV-2 vaccine of any one of any one of embodiments 8-65, wherein the SARS-CoV-2 vaccine is formulated for oral, sublingual, intramuscular, intradermal, subcutaneous, intranasal, intraocular, rectal, transdermal, mucosal (including, but not limited to buccal), topical, or parenteral administration.


Embodiment 67 is the SARS-CoV-2 vaccine of any one of any one of embodiments 8-65, wherein the SARS-CoV-2 vaccine is formulated for oral administration.


Embodiment 68 is the SARS-CoV-2 vaccine of any one of any one of embodiments 8-65, wherein the SARS-CoV-2 vaccine is formulated for intranasal administration.


Embodiment 69 is the SARS-CoV-2 vaccine of any one of any one of embodiments 8-65, wherein the SARS-CoV-2 vaccine is formulated for oral mucosal administration and intranasal administration.


Embodiment 70 is a method for producing a recombinant VSV particle, comprising

    • a. introducing the recombinant VSV vector of any one of embodiments 1 or 9-64 into cells or infecting cells with the recombinant VSV particle of any one of embodiments 2-5 or 9-64; and
    • b. producing a recombinant VSV particle and optionally cells producing a recombinant S protein.


Embodiment 71 is the method of embodiment 70, wherein the recombinant VSV particle produced by the cells expresses a SARS-CoV-2 S protein or an immunogenic variant thereof on its surface.


Embodiment 72 is the method of embodiment 70 or 71, further comprising purifying the recombinant VSV particle from the cells.


Embodiment 73 is the method of any one of embodiments 70-72, further comprising purifying the SARS-CoV-2 S protein or an immunogenic variant thereof from the cells.


Embodiment 74 is the method of any one of embodiments 70-73, further comprising purifying the recombinant VSV vector from the cells.


Embodiment 75 is a recombinant cell comprising the recombinant VSV vector of any one of embodiments 1 or 9-64 or the recombinant VSV particle of any one of embodiments 2-5 or 9-64.


Embodiment 76 is the method of any one of embodiments 70-74 or the recombinant cell of embodiment 75, wherein the recombinant cell is a Vero cell.


Embodiment 77 is a method of generating an immune response against SARS-CoV-2 comprising

    • a. administering to a subject an effective amount of the VSV vector of any one of embodiments 1 or 9-64, recombinant VSV particle of any one of embodiments 2-5 or 9-64, immunogenic recombinant protein of any one of embodiments 6, 7, 9, or 11-64, or SARS-CoV-2 vaccine of any one of embodiments 8-69; and
    • b. generating an immune response to SARS-CoV-2.


Embodiment 78 is the method of embodiment 77, wherein the immune response comprises a humoral response.


Embodiment 79 is the method of embodiment 77 or 78, wherein the immune response comprises a cellular antigen-specific immune response.


Embodiment 80 is the method of any one of embodiments 77-79, wherein the immune response comprises production of antibodies by the vaccinated subject that block SARS-CoV-2 infection, the method further comprising harvesting the antibodies from the vaccinated subject.


Embodiment 81 is the method of any one of embodiments 77-80, wherein the antibodies are in the form of immune serum obtained from the vaccinated subject.


Embodiment 82 is the method of any one of embodiments 77-81, wherein the antibodies are monoclonal antibodies prepared from SARS-CoV-2-specific B cells obtained from the vaccinated subject.


Embodiment 83 is the method of any one of embodiments 77-82, further comprising mixing the harvested antibodies with a pharmaceutically acceptable excipient, whereby a pharmaceutical composition is prepared.


Embodiment 84 is the method of any one of embodiments 77-83, wherein administering the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises oral, sublingual, intramuscular, intradermal, subcutaneous, intranasal, intraocular, rectal, transdermal, mucosal (including, but not limited to buccal), topical, or parenteral administration.


Embodiment 85 is the method of any one of embodiments 77-83, wherein administering the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises intranasal administration.


Embodiment 86 is the method of any one of embodiments 77-83, wherein administering the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises oral administration.


Embodiment 87 is the method of any one of embodiments 77-83, wherein administering the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises oral mucosal administration.


Embodiment 88 is the method of any one of embodiments 77-87, wherein administering occurs after the subject is exposed to SARS-CoV-2.


Embodiment 89 is the method of any one of embodiments 77-88, further comprising preventing or inhibiting binding of SARS-CoV-2 to the receptor.


Embodiment 90 is the method of any one of embodiments 77-89, wherein the subject has a pre-existing medical condition.


Embodiment 91 is the method embodiment 90, wherein the pre-existing medical condition is chosen from asthma, blood or bone marrow transplant, cancer, cardiomyopathy, cerebrovascular disease, chronic kidney disease, chronic obstructive pulmonary disease, coronary artery disease, cystic fibrosis, diabetes (including type 1 diabetes mellitus and type 2 diabetes mellitus), heart disease, heart failure, HIV, hypertension (high blood pressure), immune deficiency, immunocompromised state from solid organ transplant, liver disease, lung disease, neurologic conditions, obesity, pregnancy, pulmonary fibrosis, sickle cell disease, smoking, thalassemia, use of corticosteroids, use of immune weakening medicines, and combinations thereof.


Embodiment 92 is the method of any one of embodiments 77-91, wherein a single dose of the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is administered.


Embodiment 93 is the method of any one of embodiments 77-92, wherein two or more doses of the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is administered.


Embodiment 94 is the method of any one of embodiments 77-93, wherein the dose of the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine ranges from 1.0×106 to 3.8×108.


Embodiment 95 is the method of any one of embodiments 77-94, wherein the dose of the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is 1.0×106, 3.8×106, 1.5×107, 5.6×107, or 3.8×108.


Embodiment 96 is the method of any one of embodiments 77-94, wherein the dose of the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is 5.0×105, 2.4×106, 1.15×107, or 5.55×107.


Embodiment 97 is the method of any one of embodiments 77-94, wherein the dose of the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is from 1.0×106 to 3.8×108, from 1.0×106 to 5.0×107, or from 2.0×106 to 2.0×107.


DESCRIPTION OF THE SEQUENCES

Table 1 provides a listing of certain sequences referenced herein. All sequences are written either N-to-C terminus or 5′ to 3′, for protein and nucleic acid sequences, respectively.









TABLE 1







Description of the Sequences











SEQ




ID


Description
Sequences
NO





Construct 1: wt S protein
Please see FIG. 1A
  1


amino acid sequence







Construct 1: nucleotide
Please see FIG. 1B
  2


sequence of wt S protein




to be added to Vector A







Construct 1: nucleotide
Please see FIG. 1C
  3


sequence of wt S protein




to be added to Vector B







Construct 2: S protein
Please see FIG. 1D
  4


with CT deletion







Construct 2: nucleotide
Please see FIG. 1E
  5


sequence of S protein




with CT deletion to be




added to Vector A







Construct 2: nucleotide
Please see FIG. 1F
  6


sequence of S protein




with CT deletion to be




added to Vector B







Construct 3: S protein with
Please see FIG. 1G
  7


VSVG-CT IND serotype







Construct 3: nucleotide
Please see FIG. 1H
  8


sequence of S protein with




VSVG-CT IND serotype to be




added to Vector A







Construct 3: nucleotide
Please see FIG. 1I
  9


sequence of S protein with




VSVG-CT IND serotype to




Vector B







Construct 4: S protein with
Please see FIG. 1J
 10


VSVG-TM-CT IND serotype







Construct 4: nucleotide
Please see FIG. 1K
 11


sequence of S protein with




VSVG-TM-CT IND serotype




to be added to Vector A







Construct 4: nucleotide
Please see FIG. 1L
 12


sequence of S protein with




VSVG-TM-CT IND serotype




to be added to Vector B







Construct 5: S protein with
Please see FIG. 1M
 13


MPER6aa and VSVG-TM-CT




IND serotype







Construct 5:
Please see FIG. 1N
 14


nucleotide sequence




of S protein with MPER6aa 




and VSVG-TM-CT IND serotype




To be added to Vector A







Construct 5:
Please see FIG. 1O
 15


nucleotide sequence




of S protein with MPER6aa




and VSVG-TM-CT IND serotype




To be added to Vector B







Construct 6: S protein with
Please see FIG. 1P
 16


dCt21







Construct 6: Nucleotide
Please see FIG. 1Q
 17


sequence of S protein with




dCt21 to be added Vector A







Construct 6: Nucleotide
Please see FIG. 1R
 18


sequence of S protein with




dCt21 to be added Vector B







Construct 7: S protein with
Please see FIG. 1S
 19


dCt19 and VSVGct







Construct 7: Nucleotide
Please see FIG. 1T
 20


sequence of S protein with




dCt19 and VSGct to be added




Vector A







Construct 7: Nucleotide
Please see FIG. 1U
 21


sequence of S protein with




dCt19 and VSGct to be added




Vector B







Construct 8: S protein with
Please see FIG. 1V
 22


dCt19 and VSVGct21







Construct 8: Nucleotide
Please see FIG. 1W
 23


sequence of S protein with




dCt19 and VSGct21 to be 




added Vector A







Construct 8: Nucleotide
Please see FIG. 1X
 24


sequence of S protein with




dCt19 and VSGct21 to be 




added Vector B







gb_QHW06059 (a full-length
Please see FIG. 2
 25


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHZ00379 (a full-length
Please see FIG. 2
 26


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHR84449 (a full-length
Please see FIG. 2
 27


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QIA20044 (a full-length
Please see FIG. 2
 28


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHZ00358 (a full-length
Please see FIG. 2
 29


SARS-CoV-2 S protein amino




acid variant sequence)







dbj_BCA25664 (a full-length
Please see FIG. 2
 30


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHZ87582 (a full-length
Please see FIG. 2
 31


SARS-CoV-2 S protein amino




acid variant sequence)







dbj_BCA25674 (a full-length
Please see FIG. 2
 32


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHZ00389 (a full-length
Please see FIG. 2
 33


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHZ87592 (a full-length
Please see FIG. 2
 34


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QIB84673 (a full-length
Please see FIG. 2
 35


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHR63260 (a full-length
Please see FIG. 2
 36


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QIA98606 (a full-length
Please see FIG. 2
 37


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QIA98596 (a full-length
Please see FIG. 2
 38


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHR63250 (a full-length
Please see FIG. 2
 39


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHR63280 (a full-length
Please see FIG. 2
 40


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHR63290 (a full-length
Please see FIG. 2
 41


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHR63270 (a full-length
Please see FIG. 2
 42


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHZ00399 (a full-length
Please see FIG. 2
 43


SARS-CoV-2 S protein amino




acid variant sequence)







dbj_BCA25644 (a full-length
Please see FIG. 2
 44


SARS-CoV-2 S protein amino




acid variant sequence)







dbj_BCA25654 (a full-length
Please see FIG. 2
 45


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHO62877 (a full-length
Please see FIG. 2
 46


SARS-CoV-2 S protein amino




acid variant sequence)







ref_YP_00972439
Please see FIG. 2
 47


(a full-length




SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHW06039 (a full-length
Please see FIG. 2
 48


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHO62112 (a full-length
Please see FIG. 2
 49


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHQ71973 (a full-length
Please see FIG. 2
 50


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHW06049 (a full-length
Please see FIG. 2
 51


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHO60594 (a full-length
Please see FIG. 2
 52


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHN73810 (a full-length
Please see FIG. 2
 53


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHO62107 (a full-length
Please see FIG. 2
 54


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHQ82464 (a full-length
Please see FIG. 2
 55


SARS-CoV-2 S protein amino




acid variant sequence)







dbj_BBW89517 (a full-length
Please see FIG. 2
 56


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHU79204 (a full-length
Please see FIG. 2
 57


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHQ71963 (a full-length
Please see FIG. 2
 58


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHU79194 (a full-length
Please see FIG. 2
 59


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHN73795 (a full-length
Please see FIG. 2
 60


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHU36864 (a full-length
Please see FIG. 2
 61


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHD43416 (a full-length
Please see FIG. 2
 62


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHU36854 (a full-length
Please see FIG. 2
 63


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHU36844 (a full-length
Please see FIG. 2
 64


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHU36834 (a full-length
Please see FIG. 2
 65


SARS-CoV-2 S protein amino




acid variant sequence)







gb_QHU36824 (a full-length
Please see FIG. 2
 66


SARS-CoV-2 S protein amino




acid variant sequence)







Consensus sequence
Please see FIG. 2
 67





1gVSVLeader_f (primer used
ACGAAGACAAACAAACCATTATTATCATTAAAAGG
 68


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







2gVSV_400f (primer used for 
AGCAGGGGATACAATCGGAATA
 69


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







3gVSV_800f (primer used for 
CACCTCTGCAAAATAACCGGAA
 70


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







4gVSV_1200f (primer used for
GCAGAGATGTGGTCGAATGGC
 71


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







5gVSV_1600f (primer used for
TGAAGGCTTTATACAGGGGC
 72


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







6gVSV_200r (primer used for 
TGGTAGACATATCCTCTTAG
 73


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







7gVSV_400r (primer used for 
GCTGGTTCTGGAAGCATCCG
 74


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







8gVSV_800r (primer used for 
CTGGAAGCATCATTTGGACCATTT
 75


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







9gVSV_1200r (primer used for
GCTCTTTTCGCATACTGCATCATA
 76


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







10gVSV_1600r (primer used for 
CCACTTCCTCATCTGCATAGTCAT
 77


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







11gVSV_1200f (primer used for 
GCAGAGATGTGGTCGAATGGC
 78


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







12gVSV_1600f (primer used for 
TGAAGGCTTTATACAGGGGC
 79


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







13gVSV_2000f (primer used for 
TCCATGACTTTCCAACCCAAGA
 80


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







14gVSV_2400f (primer used for 
CGAGATGGACACCTATGATCCGAA
 81


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







15gVSV_1200r (primer used for 
GCTCTTTTCGCATACTGCATCATA
 82


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







16gVSV_1600r (primer used for 
CCACTTCCTCATCTGCATAGTCAT
 83


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







17gVSV_2000r (primer used for 
CTCCGACAGAGATGAACTCTCCTC
 84


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







18gVSV_2400r (primer used for 
GCTGCCACATCTGAGTATGTTCTG
 85


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







19gVSV_2000f (primer used for 
TCCATGACTTTCCAACCCAAGA
 86


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







20gVSV_2400f (primer used for 
CGAGATGGACACCTATGATCCGAA
 87


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







21gVSV_2800f2 (primer used 
CTACGATGATGAGTCACTGG
 88


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







22gVSV_2400r (primer used for 
GCTGCCACATCTGAGTATGTTCTG
 89


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







23gVSV_2800r (primer used for 
CCTTTTTCTCGACAATCAGGCC
 90


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







24CoV_S_rvs1 (primer used for 
CTCTTTGTTCCATTGGTTCC
 91


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







25gVSV_2400f (primer used for 
CGAGATGGACACCTATGATCCGAA
 92


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







26CoV_S_fw1 (primer used for
AGTGGATCCTACTCGAGAGG
 93


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







27CoV_S_fw2 (primer used for
CATTAAAGTCTGTGAATTCC
 94


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







28CoV_S_fw3 (primer used for
AGCAGCTGCTTACTATGTTG
 95


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







29CoV_S_fw4 (primer used for
GTATATGCAGATTCCTTTGT
 96


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







30CoV_S_fw5 (primer used for
ACCTAAGAAGAGCACTAATCT
 97


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







31CoV_S_rvs1 (primer used for 
CTCTTTGTTCCATTGGTTCC
 98


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







32CoV_S_rvs2 (primer used for 
GATCTCTAACTAGGTTTATAGGTGT
 99


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







33CoV_S_rvs3 (primer used for 
AGTAGCATTGAAAACCTCTC
100


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







34CoV_S_rvs4 (primer used for 
CATTACACGGTGTGCTGCCC
101


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







35CoV_S_rvs5 (primer used for 
GATAGAGAACGGCAACTTGA
102


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







36CoV_S_rvs6 (primer used for 
GGAATCTCCACAGATGTACA
103


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







37CoV_S_fw5 (primer used for
ACCTAAGAAGAGCACTAATCT
104


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







38CoV_S_fw6 (primer used for
TGACATTCCAATCGGTGCCG
105


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







39CoV_S_fw7 (primer used for
CCATCAAGGATTTTGGAGGATTTAA
106


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







40CoV_S_fw8 (primer used for
GCTATAGGAAAGATTCAGGA
107


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







41CoV_S_fw9 (primer used for
ACATGTAACATATGTGCCCG
108


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







42CoV_S_fw10 (primer used for
TGATCGACCTTCAGGAGCTG
109


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







43CoV_S_rvs7 (primer used for 
TGTAATTGTACCAGCCAACA
110


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







44CoV_S_rvs8 (primer used for 
TGATGAGTTGTTGGGTGACG
111


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







45CoV_S_rvs9 (primer used for 
GGAATCGAGTTCAGGTTGTA
112


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







46CoV_S_rvs10 (primer used
AACGCGTAAACAGCTAGCCT
113


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







47gVSVGene4_f (primer used
TTAATGTTTGGCCTGATTGTCGAGA
114


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







48gVSVGene4_rev (primer
GCGCTCATCGGGATTCAGGAA
115


used for the Sanger 




sequencing of the rVSVΔG-




SARS-CoV-2 genome)







49gVSV_4800r (primer used for
CCACCACGTCAAATGTTATTTC
116


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







50CoV_S_fw10 (primer used for
ACATGTAACATATGTGCCCG
117


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







51gVSV_5300f (primer used for
GGGCAACAAACCAATTGAAT
118


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







52gVSV_5800f (primer used for
GCAAGAGAATCAAGGCCTTTAGT
119


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







53gVSV_6300f (primer used for
CCCATCGATAATATACTCTGACAAA
120


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







54gVSV_6800f (primer used for
TCCTTAATCGAGAGAACTCATGAA
121


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







55gVSV_4800r (primer used for
CCACCACGTCAAATGTTATTTC
122


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







56gVSV_5200r2 (primer used
ACTAAAGGCCTTGATTCTCTTGC
123


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







57gVSV_5200r (primer used for
GATGGGTCTAGTAAGTCGGGTATT
124


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







58gVSV_6200r2 (primer used
TTCATGAGTTCTCTCGATTAAGGA
125


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







59gVSV_6200r (primer used for 
CACTCGTGACCATCTCTTGG
126


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







60gVSV_6300f (primer used for 
CCCATCGATAATATACTCTGACAAA
127


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







61gVSV_6800f (primer used for 
TCCTTAATCGAGAGAACTCATGAA
128


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







62gVSV_7300f (primer used for 
TGACCAAATACCCACTTGTGC
129


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







63gVSV_7800f (primer used for 
CGAACTTGTTAAAGACTGAGGTTAA
130


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







64JKgVSV_8300f (primer used 
CGGGATCCATGACGTCTTTA
131


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







65gVSV_8800f (primer used for 
TTTGAGACCCATAGAAGAGATCAC
132


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







66gVSV_6200r (primer used for 
CACTCGTGACCATCTCTTGG
133


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







67gVSV_7200r2 (primer used
TTAACCTCAGTCTTTAACAAGTTCG
134


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







68gVSV_7200r (primer used for 
TGAACCCTGATGTGTTACATGG
135


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







69gVSV_8200r2 (primer used
GTGATCTCTTCTATGGGTCTCAAA
136


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







70gVSV_8200r (primer used for 
TCGTTTCTAATTCGTCTCTAATAGG
137


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







71gVSV_8300f (primer used for 
CGGGATCCATGACGTCTTTA
138


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







72gVSV_8800f (primer used for 
TTTGAGACCCATAGAAGAGATCAC
139


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







73gVSV_9300f (primer used for 
CTCCTATCCGACAAGCAACC
140


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







74gVSV_9800f (primer used for 
GAATCAGGTTGGGCCAATTA
141


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







75gVSV_10300f (primer used
GAGCGAAAAGAATGCAGTAACA
142


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







76gVSV_10800f (primer used
GCTTTTGGCTGAGTTTGATG
143


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







77JKVSVGene6_f (primer used
AAACACAGGAATGATTGAAT 
144


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







78gVSV_8200r (primer used for 
TCGTTTCTAATTCGTCTCTAATAGG
145


the Sanger sequencing of the




rVSVΔG-SARS-CoV-2 genome)







79gVSV_9200r2 (primer used
TAATTGGCCCAACCTGATTC
146


for the Sanger sequencing of




the rVSVΔG-SARS-CoV-2




genome)







80gVSV_9200r (primer
CGCTCTCACAAATATATGTTCCATA
147


used for the Sanger 




sequencing of the rVSVΔG-




SARS-CoV-2 genome)







81gVSV_10200r2 (primer 
CATCAAACTCAGCCAAAAGC
148


used  for the Sanger 




sequencing of the rVSVΔG-




SARS-CoV-2 genome)







82JRVSVTrai1er_r (primer 
ACGAAGACCACAAAACCAGATAAA
149


used for the Sanger 




sequencing of the rVSVΔG-




SARS-CoV-2 genome)















Nucleotide sequence for
    1
acgaagacaa acaaaccatt attatcatta aaaggctcag gagaaacttt aacagtaatc
150


rVSVΔG-SARS-CoV-2 clone
   61
aaaatgtctg ttacagtcaa gagaatcatt gacaacacag tcgtagttcc aaaacttcct



MB1
  121
gcaaatgagg atccagtgga atacccggca gattacttca gaaaatcaaa ggagattcct




  181
ctttacatca atactacaaa aagtttgtca gatctaagag gatatgtcta ccaaggcctc




  241
aaatccggaa atgtatcaat catacatgtc aacagctact tgtatggagc attaaaggac




  301
atccggggta agttggataa agattggtca agtttcggaa taaacatcgg gaaagcaggg




  361
gatacaatcg gaatatttga ccttgtatcc ttgaaagccc tggacggcgt acttccagat




  421
ggagtatcgg atgcttccag aaccagcgca gatgacaaat ggttgccttt gtatctactt




  481
ggcttataca gagtgggcag aacacaaatg cctgaataca gaaaaaagct catggatggg




  541
ctgacaaatc aatgcaaaat gatcaatgaa cagtttgaac ctcttgtgcc agaaggtcgt




  601
gacatttttg atgtgtgggg aaatgacagt aattacacaa aaattgtcgc tgcagtggac




  661
atgttcttcc acatgttcaa aaaacatgaa tgtgcctcgt tcagatacgg aactattgtt




  721
tccagattca aagattgtgc tgcattggca acatttggac acctctgcaa aataaccgga




  781
atgtctacag aagatgtaac gacctggatc ttgaaccgag aagttgcaga tgaaatggtc




  841
caaatgatgc ttccaggcca agaaattgac aaggccgatt catacatgcc ttatttgatc




  901
gactttggat tgtcttctaa gtctccatat tcttccgtca aaaaccctgc cttccacttc




  961
tgggggcaat tgacagctct tctgctcaga tccaccagag caaggaatgc ccgacagcct




 1021 
gatgacattg agtatacatc tcttactaca gcaggtttgt tgtacgctta tgcagtagga




 1081 
tcctctgccg acttggcaca acagttttgt gttggagata acaaatacac tccagatgat




 1141 
agtaccggag gattgacgac taatgcaccg ccacaaggca gagatgtggt cgaatggctc




 1201 
ggatggtttg aagatcaaaa cagaaaaccg actcctgata tgatgcagta tgcgaaaaga




 1261 
gcagtcatgt cactgcaagg cctaagagag aagacaattg gcaagtatgc taagtcagaa




 1321 
tttgacaaat gaccctataa ttctcagatc acctattata tattatgcta catatgaaaa




 1381 
aaactaacag atatcatgga taatctcaca aaagttcgtg agtatctcaa gtcctattct




 1441 
cgtctggatc aggcggtagg agagatagat gagatcgaag cacaacgagc tgaaaagtcc




 1501 
aattatgagt tgttccaaga ggatggagtg gaagagcata ctaagccctc ttattttcag




 1561 
gcagcagatg attctgacac agaatctgaa ccagaaattg aagacaatca aggcttgtat




 1621 
gcaccagatc cagaagctga gcaagttgaa ggctttatac aggggccttt agatgactat




 1681 
gcagatgagg aagtggatgt tgtatttact tcggactgga aacagcctga gcttgaatct




 1741 
gacgagcatg gaaagacctt acggttgaca tcgccagagg gtttaagtgg agagcagaaa




 1801 
tcccagtggc tttcgacgat taaagcagtc gtgcaaagtg ccaaatactg gaatctggca




 1861 
gagtgcacat ttgaagcatc gggagaaggg gtcattatga aggagcgcca gataactccg




 1921 
gatgtatata aggtcactcc agtgatgaac acacatccgt cccaatcaga agcagtatca




 1981 
gatgtttggt ctctctcaaa gacatccatg actttccaac ccaagaaagc aagtcttcag




 2041 
cctctcacca tatccttgga tgaattgttc tcatctagag gagagttcat ctctgtcgga




 2101 
ggtgacggac gaatgtctca taaagaggcc atcctgctcg gcctgagata caaaaagttg




 2161 
tacaatcagg cgagagtcaa atattctctg tagactatga aaaaaagtaa cagatatcac




 2221 
gatctaagtg ttatcccaat ccattcatca tgagttcctt aaagaagatt ctcggtctga




 2281 
aggggaaagg taagaaatct aagaaattag ggatcgcacc acccccttat gaagaggaca




 2341 
ctagcatgga gtatgctccg agcgctccaa ttgacaaatc ctattttgga gttgacgaga




 2401 
tggacaccta tgatccgaat caattaagat ctgagaaatt cttctttaca gtgaaaatga




 2461 
cggttagatc taatcgtccg ttcagaacat actcagatgt ggcagccgct gtatcccatt




 2521 
gggatcacat gtacatcgga atggcaggga aacgtccctt ctacaaaatc ttggcttttt




 2581 
tgggttcttc taatctaaag gccactccag cggtattggc agatcaaggt caaccagagt




 2641 
atcacgctca ctgcgaaggc agggcttatt tgccacatag gatggggaag acccctccca




 2701 
tgctcaatgt accagagcac ttcagaagac cattcaatat aggtctttac aagggaacga




 2761 
ttgagctcac aatgaccatc tacgatgatg agtcactgga agcagctcct atgatctggg




 2821 
atcatttcaa ttcttccaaa ttttctgatt tcagagagaa ggccttaatg tttggcctga




 2881 
ttgtcgagaa aaaggcatct ggagcgtggg tcctggactc tatcggccac ttcaaatgag




 2941 
ctagtctaac ttctagcttc tgaacaatcc ccggtttact cagtctcccc taattccagc




 3001 
ctctcgaaca actaatatcc tgtcttttct atccctatga aaaaaactaa cagagattga




 3061 
tctgtttacg cgctagtgga tcctactcga gaggagccac catgttcgtg ttcctggtgc




 3121 
tattacctct ggtttcgtct caatgcgtaa accttacaac tagaactcag cttcctccag




 3181 
catacacaaa ttccttcact cgcggagtgt attatcctga taaggtcttt cgatcatcag




 3241 
tgttgcattc cacccaggat ttgtttctcc cgttcttttc aaatgtaact tggttccatg




 3301 
ctatacatgt ttccggaacc aatggaacaa agagatttga taacccagtg ttaccattta




 3361 
acgacggagt ttatttcgca tcaactgaga aatccaatat cattagaggc tggattttcg




 3421 
gaacgaccct ggattctaaa acgcaatcct tgctgattgt taataatgca acaaatgtgg




 3481 
tcattaaagt ctgtgaattc caattttgca atgatccatt tctcggcgtc tattaccaca




 3541 
agaataacaa atcttggatg gagtcagagt tcagggttta tagttccgca aataattgta




 3601 
cttttgaata cgtttcccaa ccattcttaa tggacttgga gggaaaacag ggaaatttta




 3661 
agaatctaag agaattcgtc tttaagaata ttgatggata tttcaagatc tattcaaaac




 3721 
atacacctat aaacctagtt agagatctcc cgcaagggtt ttcagcccta gagccactag




 3781 
ttgacctgcc aattgggatc aacattacta gattccagac cctactcgct ctgcatcggt




 3841 
catatttgac accaggagat tcatcgtcag gatggaccgc tggagcagct gcttactatg




 3901 
ttgggtatct gcaacctaga acatttctcc taaagtataa tgaaaacggg actattacag




 3961 
acgcagtcga ttgcgcactg gatccactct cagagacaaa gtgcactcta aaatcattca




 4021 
ctgtcgagaa aggaatctat caaacatcaa atttcagggt ccagccaact gagagtattg




 4081 
tccggttccc taacataact aacttgtgcc ccttcggaga ggttttcaat gctactcggt




 4141 
tcgccagcgt ctacgcatgg aacagaaaga ggatttcaaa ctgtgtcgca gattatagcg




 4201 
tcctctataa ttcagcatca ttcagtacat ttaaatgcta tggtgtcagc cccaccaaac




 4261 
ttaatgactt atgttttacc aatgtatatg cagattcctt tgtaatcaga ggtgacgaag




 4321 
tgaggcaaat cgcacctgga cagaccggaa agattgctga ttataattat aaactccctg




 4381 
atgattttac cggatgtgtt attgcttgga acagcaataa cctcgatagt aaggtcggag




 4441 
gaaactataa ctatttgtac agactgttta gaaagtcgaa tttgaaacct tttgaaagag




 4501 
acatatccac cgagatttac caggcgggca gcacaccgtg taatggtgta gaaggattca




 4561 
attgttactt tcccctgcaa tcatatgggt ttcaaccaac caatggagtc ggatatcaac




 4621 
catatcgtgt cgtcgtcctt tccttcgagc tgcttcatgc accagctaca gtctgcggac




 4681 
ctaagaagag cactaatctt gtcaagaaca aatgtgtgaa ctttaatttt aatggattaa




 4741 
caggaaccgg agttttgacc gagagtaata agaagttctt gccgttccag caatttggac




 4801 
gagacattgc tgacaccaca gatgcggttc gtgacccgca aactttagag atcctagaca




 4861 
tcaccccatg ttcattcggt ggagtttccg ttattactcc tggaacgaat acaagcaatc




 4921 
aagttgccgt tctctatcaa gatgttaatt gtacagaagt gcctgtggcc attcatgcag




 4981 
atcaactaac accaacttgg agagtttaca gcactgggtc caatgtcttc caaacgcgcg




 5041 
ccggctgcct cattggtgca gaacatgtga ataactcata cgaatgtgac attccaatcg




 5101 
gtgccggcat atgcgcctct taccagactc agactaattc gccaagagga gccaggtctg




 5161 
tcgcaagtca gtcaattatt gcatacacaa tgtcgttagg agcagagaat agtgtagcat




 5221 
actcaaacaa ttctatagca atacctacca acttcactat atcagtaact acagaaatat




 5281 
tgccagtatc catgactaaa acaagtgtgg attgcaccat gtacatctgt ggagattcca




 5341 
cagaatgcag caatcttctc ttgcaatacg gatcattctg cacacaactg aatagggcac




 5401 
tgactggaat tgcagtcgag caggataaga acacacagga ggtgtttgcc caagtcaaac




 5461 
aaatatacaa aacaccaccc atcaaggatt ttggaggatt taatttctca caaatactcc




 5521 
ccgacccatc caagcccttc aaaaggagtt tcattgagga cctcttgttt aataaggtta




 5581 
ccttggcaga tgccgggttt attaagcagt acggcgactg tcttggagac atagcagcca




 5641 
gagatctaat ttgtgcccag aaattcaatg gactgacagt cctgcctccc ttattaactg




 5701 
atgagatgat agctcagtat acatcagcat tgttggctgg tacaattaca tctggatgga




 5761 
catttggtgc cggagcggca ttacaaatcc cttttgcaat gcaaatggcc tatagattta




 5821 
atggaatcgg agtaactcaa aatgttttat atgagaatca gaaattaatt gcaaatcaat




 5881 
tcaattcagc tataggaaag attcaggatt cactcagtag tacagcaagc gctctaggca




 5941 
aattacaaga cgtcgtcaat cagaatgcac aggcattaaa tacactggtg aagcaattga




 6001 
gttccaattt cggagcaatt tcatctgttc taaatgatat attgtcaaga ctggataaag




 6061 
tagaagccga ggtccaaatc gataggctga tcacaggaag acttcaatca ctacagacat




 6121 
acgtcaccca acaactcatc agagcagcag aaattagagc ctctgctaat ctagccgcaa




 6181 
caaagatgtc agagtgcgta ttgggacaat ctaagagggt cgacttttgt ggaaaggggt




 6241 
atcacttgat gtcctttcct caatctgcac cacacggagt tgtcttctta catgtaacat




 6301 
atgtgcccgc tcaagaaaag aatttcacta cagcacctgc aatatgtcat gacggaaaag




 6361 
cacattttcc tcgggaggga gttttcgttt ctaatggaac ccattggttc gtgacccaaa




 6421 
ggaactttta cgagcctcaa ataattacaa ctgataatac attcgtttct ggaaattgcg




 6481 
acgtagttat aggtattgta aataatactg tttatgaccc tttacaacct gaactcgatt




 6541 
ccttcaagga agaactcgac aaatatttta agaatcacac ctcaccggac gttgacttag




 6601 
gagacatttc cgggattaac gctagtgtag tcaatatcca aaaggagata gatagactga




 6661 
atgaggtagc aaagaatctt aatgaatctt tgatcgacct tcaggagctg gggaagtacg




 6721 
aacaatacat aaaatggcca tggtacattt ggctcgggtt tattgctgga ctaattgcaa




 6781 
tagtcatggt cactatcatg ctgtgttgta tgacatcgtg ctgctcatgc ctcaagggat




 6841 
gttgtagctg ttgatcttgt tgcaagttcg atgaggatga ttcagaacca gttttaaaag




 6901 
gagtaaagtt gcattacaca taaaggctag ctgtttacgc gttatccatg ctcaaagagg




 6961 
cctcaattat atttgagttt ttaattttta tgaaaaaaac taacagcaat catggaagtc




 7021 
cacgattttg agaccgacga gttcaatgat ttcaatgaag atgactatgc cacaagagaa




 7081 
ttcctgaatc ccgatgagcg catgacgtac ttgaatcatg ctgattacaa cctgaattct




 7141 
cctctaatta gtgatgatat tgacaattta atcaggaaat tcaattctct tccaattccc




 7201 
tcgatgtggg atagtaagaa ctgggatgga gttcttgaga tgttaacgtc atgtcaagcc




 7261 
aatcccatcc caacatctca gatgcataaa tggatgggaa gttggttaat gtctgataat




 7321 
catgatgcca gtcaagggta tagtttttta catgaagtgg acaaagaggc agaaataaca




 7381 
tttgacgtgg tggagacctt catccgcggc tggggcaaca aaccaattga atacatcaaa




 7441 
aaggaaagat ggactgactc attcaaaatt ctcgcttatt tgtgtcaaaa gtttttggac




 7501 
ttacacaagt tgacattaat cttaaatgct gtctctgagg tggaattgct caacttggcg




 7561 
aggactttca aaggcaaagt cagaagaagt tctcatggaa cgaacatatg caggattagg




 7621 
gttcccagct tgggtcctac ttttatttca gaaggatggg cttacttcaa gaaacttgat




 7681 
attctaatgg accgaaactt tctgttaatg gtcaaagatg tgattatagg gaggatgcaa




 7741 
acggtgctat ccatggtatg tagaatagac aacctgttct cagagcaaga catcttctcc




 7801 
cttctaaata tctacagaat tggagataaa attgtggaga ggcagggaaa tttttcttat




 7861 
gacttgatta aaatggtgga accgatatgc aacttgaagc tgatgaaatt agcaagagaa




 7921 
tcaaggcctt tagtcccaca attccctcat tttgaaaatc atatcaagac ttctgttgat




 7981 
gaaggggcaa aaattgaccg aggtataaga ttcctccatg atcagataat gagtgtgaaa




 8041 
acagtggatc tcacactggt gatttatgga tcgttcagac attggggtca tccttttata




 8101 
gattattaca ctggactaga aaaattacat tcccaagtaa ccatgaagaa agatattgat




 8161 
gtgtcatatg caaaagcact tgcaagtgat ttagctcgga ttgttctatt tcaacagttc




 8221 
aatgatcata aaaagtggtt cgtgaatgga gacttgctcc ctcatgatca tccctttaaa




 8281 
agtcatgtta aagaaaatac atggcccaca gctgctcaag ttcaagattt tggagataaa




 8341 
tggcatgaac ttccgctgat taaatgtttt gaaatacccg acttactaga cccatcgata




 8401 
atatactctg acaaaagtca ttcaatgaat aggtcagagg tgttgaaaca tgtccgaatg




 8461 
aatccgaaca ctcctatccc tagtaaaaag gtgttgcaga ctatgttgga cacaaaggct




 8521 
accaattgga aagaatttct taaagagatt gatgagaagg gcttagatga tgatgatcta




 8581 
attattggtc ttaaaggaaa ggagagggaa ctgaagttgg caggtagatt tttctcccta




 8641 
atgtcttgga aattgcgaga atactttgta attaccgaat atttgataaa gactcatttc




 8701 
gtccctatgt ttaaaggcct gacaatggcg gacgatctaa ctgcagtcat taaaaagatg




 8761 
ttagattcct catccggcca aggattgaag tcatatgagg caatttgcat agccaatcac




 8821 
attgattacg aaaaatggaa taaccaccaa aggaagttat caaacggccc agtgttccga




 8881
gttatgggcc agttcttagg ttatccatcc ttaatcgaga gaactcatga attttttgag




 8941
aaaagtctta tatactacaa tggaagacca gacttgatgc gtgttcacaa caacacactg




 9001
atcaattcaa cctcccaacg agtttgttgg caaggacaag agggtggact ggaaggtcta




 9061
cggcaaaaag gatggagtat cctcaatcta ctggttattc aaagagaggc taaaatcaga




 9121
aacactgctg tcaaagtctt ggcacaaggt gataatcaag ttatttgcac acagtataaa




 9181
acgaagaaat cgagaaacgt tgtagaatta cagggtgctc tcaatcaaat ggtttctaat




 9241
aatgagaaaa ttatgactgc aatcaaaata gggacaggga agttaggact tttgataaat




 9301
gacgatgaga ctatgcaatc tgcagattac ttgaattatg gaaaaatacc gattttccgt




 9361
ggagtgatta gagggttaga gaccaagaga tggtcacgag tgacttgtgt caccaatgac




 9421
caaataccca cttgtgctaa tataatgagc tcagtttcca caaatgctct caccgtagct




 9481
cattttgctg agaacccaat caatgccatg atacagtaca attattttgg gacatttgct




 9541
agactcttgt tgatgatgca tgatcctgct cttcgtcaat cattgtatga agttcaagat




 9601
aagataccag gcttgcacag ttctactttc aaatacgcca tgttgtattt ggacccttcc




 9661
attggaggag tgtcgggcat gtctttgtcc aggtttttga ttagagcctt cccagatccc




 9721
gtaacagaaa gtctctcatt ctggagattc atccatgtac atgctcgaag tgagcatctg




 9781
aaggagatga gtgcagtatt tggaaacccc gagatagcca agtttcgaat aactcacata




 9841
gacaagctag tagaagatcc aacctctctg aacatcgcta tgggaatgag tccagcgaac




 9901
ttgttaaaga ctgaggttaa aaaatgctta atcgaatcaa gacaaaccat caggaaccag




 9961
gtgattaagg atgcaaccat atatttgtat catgaagagg atcggctcag aagtttctta




10021
tggtcaataa atcctctgtt ccctagattt ttaagtgaat tcaaatcagg cacttttttg




10081
ggagtcgcag acgggctcat cagtctattt caaaattctc gtactattcg gaactccttt




10141
aagaaaaagt atcataggga attggatgat ttgattgtga ggagtgaggt atcctctttg




10201 
acacatttag ggaaacttca tttgagaagg ggatcatgta aaatgtggac atgttcagct




10261 
actcatgctg acacattaag atacaaatcc tggggccgta cagttattgg gacaactgta




10321 
ccccatccat tagaaatgtt gggtccacaa catcgaaaag agactccttg tgcaccatgt




10381 
aacacatcag ggttcaatta tgtttctgtg cattgtccag acgggatcca tgacgtcttt




10441 
agttcacggg gaccattgcc tgcttatcta gggtctaaaa catctgaatc tacatctatt




10501 
ttgcagcctt gggaaaggga aagcaaagtc ccactgatta aaagagctac acgtcttaga




10561 
gatgctatct cttggtttgt tgaacccgac tctaaactag caatgactat actttctaac




10621 
atccactctt taacaggcga agaatggacc aaaaggcagc atgggttcaa aagaacaggg




10681 
tctgcccttc ataggttttc gacatctcgg atgagccatg gtgggttcgc atctcagagc




10741 
actgcagcat tgaccaggtt gatggcaact acagacacca tgagggatct gggagatcag




10801 
aatttcgact ttttattcca agcaacgttg ctctatgctc aaattaccac cactgttgca




10861
agagacggat ggatcaccag ttgtacagat cattatcata ttgcctgtaa gtcctgtttg




10921
agacccatag aagagatcac cctggactca agtatggact acacgccccc agatgtatcc




10981 
catgtgctga agacatggag gaatggggaa ggttcgtggg gacaagagat aaaacagatc




11041 
tatcctttag aagggaattg gaagaattta gcacctgctg agcaatccta tcaagtcggc




11101 
agatgtatag gttttctata tggagacttg gcgtatagaa aatctactca tgccgaggac




11161
agttctctat ttcctctatc tatacaaggt cgtattagag gtcgaggttt cttaaaaggg




11221
ttgctagacg gattaatgag agcaagttgc tgccaagtaa tacaccggag aagtctggct




11281
catttgaaga ggccggccaa cgcagtgtac ggaggtttga tttacttgat tgataaattg




11341
agtgtatcac ctccattcct ttctcttact agatcaggac ctattagaga cgaattagaa




11401
acgattcccc acaagatccc aacctcctat ccgacaagca accgtgatat gggggtgatt




11461
gtcagaaatt acttcaaata ccaatgccgt ctaattgaaa agggaaaata cagatcacat




11521
tattcacaat tatggttatt ctcagatgtc ttatccatag acttcattgg accattctct




11581
atttccacca ccctcttgca aatcctatac aagccatttt tatctgggaa agataagaat




11641
gagttgagag agctggcaaa tctttcttca ttgctaagat caggagaggg gtgggaagac




11701
atacatgtga aattcttcac caaggacata ttattgtgtc cagaggaaat cagacatgct




11761
tgcaagttcg ggattgctaa ggataataat aaagacatga gctatccccc ttggggaagg




11821
gaatccagag ggacaattac aacaatccct gtttattata cgaccacccc ttacccaaag




11881
atgctagaga tgcctccaag aatccaaaat cccctgctgt ccggaatcag gttgggccaa




11941
ttaccaactg gcgctcatta taaaattcgg agtatattac atggaatggg aatccattac




12001
agggacttct tgagttgtgg agacggctcc ggagggatga ctgctgcatt actacgagaa




12061
aatgtgcata gcagaggaat attcaatagt ctgttagaat tatcagggtc agtcatgcga




12121
ggcgcctctc ctgagccccc cagtgcccta gaaactttag gaggagataa atcgagatgt




12181
gtaaatggtg aaacatgttg ggaatatcca tctgacttat gtgacccaag gacttgggac




12241
tatttcctcc gactcaaagc aggcttgggg cttcaaattg atttaattgt aatggatatg




12301
gaagttcggg attcttctac tagcctgaaa attgagacga atgttagaaa ttatgtgcac




12361
cggattttgg atgagcaagg agttttaatc tacaagactt atggaacata tatttgtgag




12421
agcgaaaaga atgcagtaac aatccttggt cccatgttca agacggtcga cttagttcaa




12481
acagaattta gtagttctca aacgtctgaa gtatatatgg tatgtaaagg tttgaagaaa




12541
ttaatcgatg aacccaatcc cgattggtct tccatcaatg aatcctggaa aaacctgtac




12601
gcattccagt catcagaaca ggaatttgcc agagcaaaga aggttagtac atactttacc




12661
ttgacaggta ttccctccca attcattcct gatccttttg taaacattga gactatgcta




12721
caaatattcg gagtacccac gggtgtgtct catgcggctg ccttaaaatc atctgataga




12781
cctgcagatt tattgaccat tagccttttt tatatggcga ttatatcgta ttataacatc




12841
aatcatatca gagtaggacc gatacctccg aaccccccat cagatggaat tgcacaaaat




12901
gtggggatcg ctataactgg tataagcttt tggctgagtt tgatggagaa agacattcca




12961
ctatatcaac agtgtttagc agttatccag caatcattcc cgattaggtg ggaggctgtt




13021
tcagtaaaag gaggatacaa gcagaagtgg agtactagag gtgatgggct cccaaaagat




13081
acccgaattt cagactcctt ggccccaatc gggaactgga tcagatctct ggaattggtc




13141
cgaaaccaag ttcgtctaaa tccattcaat gagatcttgt tcaatcagct atgtcgtaca




13201
gtggataatc atttgaaatg gtcaaatttg cgaagaaaca caggaatgat tgaatggatc




13261
aatagacgaa tttcaaaaga agaccggtct atactgatgt tgaagagtga cctacacgag




13321
gaaaactctt ggagagatta aaaaatcatg aggagactcc aaactttaag tatgaaaaaa




13381
actttgatcc ttaagaccct cttgtggttt ttatttttta tctggttttg tggtcttcgt













Amino acid sequence for VSV-
Please see FIG. 24D.
151


M protein in rVSVΔG-SARS-




CoV-2 clone MB1







Amino acid sequence for VSV-
Please see FIGS. 21F, 22E, 23E, and 24F.
152


L protein in rVSVΔG-SARS-




CoV-2 clone MB1, MB2, and




Vectors A and B







Amino acid sequence for
Please see FIG. 24E.
153


SARS-CoV-2 S protein in




rVSVΔG-SARS-CoV-2 clone




MB1















Nucleotide sequence for
    1
ACGAAGACAA ACAAACCATT ATTATCATTA AAAGGCTCAG GAGAAACTTT
154


rVSVΔG-SARS-CoV-2 clone
   51 
AACAGTAATC AAAATGTCTG TTACAGTCAA GAGAATCATT GACAACACAG



MB2
  101
TCGTAGTTCC AAAACTTCCT GCAAATGAGG ATCCAGTGGA ATACCCGGCA




  151
GATTACTTCA GAAAATCAAA GGAGATTCCT CTTTACATCA ATACTACAAA




  201
AAGTTTGTCA GATCTAAGAG GATATGTCTA CCAAGGCCTC AAATCCGGAA




  251
ATGTATCAAT CATACATGTC AACAGCTACT TGTATGGAGC ATTAAAGGAC




  301
ATCCGGGGTA AGTTGGATAA AGATTGGTCA AGTTTCGGAA TAAACATCGG




  351
GAAAGCAGGG GATACAATCG GAATATTTGA CCTTGTATCC TTGAAAGCCC




  401
TGGACGGCGT ACTTCCAGAT GGAGTATCGG ATGCTTCCAG AACCAGCGCA




  451
GATGACAAAT GGTTGCCTTT GTATCTACTT GGCTTATACA GAGTGGGCAG




  501
AACACAAATG CCTGAATACA GAAAAAAGCT CATGGATGGG CTGACAAATC




  551
AATGCAAAAT GATCAATGAA CAGTTTGAAC CTCTTGTGCC AGAAGGTCGT




  601
GACATTTTTG ATGTGTGGGG AAATGACAGT AATTACACAA AAATTGTCGC




  651
TGCAGTGGAC ATGTTCTTCC ACATGTTCAA AAAACATGAA TGTGCCTCGT




  701
TCAGATACGG AACTATTGTT TCCAGATTCA AAGATTGTGC TGCATTGGCA




  751
ACATTTGGAC ACCTCTGCAA AATAACCGGA ATGTCTACAG AAGATGTAAC




  801
GACCTGGATC TTGAACCGAG AAGTTGCAGA TGAAATGGTC CAAATGATGC




  851
TTCCAGGCCA AGAAATTGAC AAGGCCGATT CATACATGCC TTATTTGATC




  901
GACTTTGGAT TGTCTTCTAA GTCTCCATAT TCTTCCGTCA AAAACCCTGC




  951
CTTCCACTTC TGGGGGCAAT TGACAGCTCT TCTGCTCAGA TCCACCAGAG




 1001
CAAGGAATGC CCGACAGCCT GATGACATTG AGTATACATC TCTTACTACA




 1051
GCAGGTTTGT TGTACGCTTA TGCAGTAGGA TCCTCTGCCG ACTTGGCACA




 1101
ACAGTTTTGT GTTGGAGATA ACAAATACAC TCCAGATGAT AGTACCGGAG




 1151
GATTGACGAC TAATGCACCG CCACAAGGCA GAGATGTGGT CGAATGGCTC




 1201
GGATGGTTTG AAGATCAAAA CAGAAAACCG ACTCCTGATA TGATGCAGTA




 1251
TGCGAAAAGA GCAGTCATGT CACTGCAAGG CCTAAGAGAG AAGACAATTG




 1301
GCAAGTATGC TAAGTCAGAA TTTGACAAAT GACCCTATAA TTCTCAGATC




 1351
ACCTATTATA TATTATGCTA CATATGAAAA AAACTAACAG ATATCATGGA




 1401
TAATCTCACA AAAGTTCGTG AGTATCTCAA GTCCTATTCT CGTCTGGATC




 1451
AGGCGGTAGG AGAGATAGAT GAGATCGAAG CACAACGAGC TGAAAAGTCC




 1501
AATTATGAGT TGTTCCAAGA GGATGGAGTG GAAGAGCATA CTAAGCCCTC




 1551
TTATTTTCAG GCAGCAGATG ATTCTGACAC AGAATCTGAA CCAGAAATTG




 1601
AAGACAATCA AGGCTTGTAT GCACCAGATC CAGAAGCTGA GCAAGTTGAA




 1651
GGCTTTATAC AGGGGCCTTT AGATGACTAT GCAGATGAGG AAGTGGATGT




 1701
TGTATTTACT TCGGACTGGA AACAGCCTGA GCTTGAATCT GACGAGCATG




 1751
GAAAGACCTT ACGGTTGACA TCGCCAGAGG GTTTAAGTGG AGAGCAGAAA




 1801
TCCCAGTGGC TTTCGACGAT TAAAGCAGTC GTGCAAAGTG CCAAATACTG




 1851
GAATCTGGCA GAGTGCACAT TTGAAGCATC GGGAGAAGGG GTCATTATGA




 1901
AGGAGCGCCA GATAACTCCG GATGTATATA AGGTCACTCC AGTGATGAAC




 1951
ACACATCCGT CCCAATCAGA AGCAGTATCA GATGTTTGGT CTCTCTCAAA




 2001
GACATCCATG ACTTTCCAAC CCAAGAAAGC AAGTCTTCAG CCTCTCACCA




 2051
TATCCTTGGA TGAATTGTTC TCATCTAGAG GAGAGTTCAT CTCTGTCGGA




 2101
GGTGACGGAC GAATGTCTCA TAAAGAGGCC ATCCTGCTCG GCCTGAGATA




 2151
CAAAAAGTTG TACAATCAGG CGAGAGTCAA ATATTCTCTG TAGACTATGA




 2201
AAAAAAGTAA CAGATATCAC GATCTAAGTG TTATCCCAAT CCATTCATCA




 2251
TGAGTTCCTT AAAGAAGATT CTCGGTCTGA AGGGGAAAGG TAAGAAATCT




 2301
AAGAAATTAG GGATCGCACC ACCCCCTTAT GAAGAGGACA CTAGCATGGA




 2351
GTATGCTCCG AGCGCTCCAA TTGACAAATC CTATTTTGGA GTTGACGAGA




 2401
TGGACACCTA TGATCCGAAT CAATTAAGAT ATGAGAAATT CTTCTTTACA




 2451
GTGAAAATGA CGGTTAGATC TAATCGTCCG TTCAGAACAT ACTCAGATGT




 2501
GGCAGCCGCT GTATCCCATT GGGATCACAT GTACATCGGA ATGGCAGGGA




 2551
AACGTCCCTT CTACAAAATC TTGGCTTTTT TGGGTTCTTC TAATCTAAAG




 2601
GCCACTCCAG CGGTATTGGC AGATCAAGGT CAACCAGAGT ATCACGCTCA




 2651
CTGCGAAGGC AGGGCTTATT TGCCACATAG GATGGGGAAG ACCCCTCCCA




 2701
TGCTCAATGT ACCAGAGCAC TTCAGAAGAC CATTCAATAT AGGTCTTTAC




 2751
AAGGGAACGA TTGAGCTCAC AATGACCATC TACGATGATG AGTCACTGGA




 2801
AGCAGCTCCT ATGATCTGGG ATCATTTCAA TTCTTCCAAA TTTTCTGATT




 2851
TCAGAGAGAA GGCCTTAATG TTTGGCCTGA TTGTCGAGAA AAAGGCATCT




 2901
GGAGCGTGGG TCCTGGACTC TATCGGCCAC TTCAAATGAG CTAGTCTAAC




 2951
TTCTAGCTTC TGAACAATCC CCGGTTTACT CAGTCTCCCC TAATTCCAGC




 3001
CTCTCGAACA ACTAATATCC TGTCTTTTCT ATCCCTATGA AAAAAACTAA




 3051
CAGAGATCGA TCTGTTTACG CGCTAGTGGA TCCTACTCGA GAGGAGCCAC




 3101
CATGTTCGTG TTCCTGGTGC TATTACCTCT GGTTTCGTCT CAATGCGTAA




 3151
ACCTTACAAC TAGAACTCAG CTTCCTCCAG CATACACAAA TTCCTTCACT




 3201
CGCGGAGTGT ATTATCCTGA TAAGGTCTTT CGATCATCAG TGTTGCATTC




 3251
CACCCAGGAT TTGTTTCTCC CGTTCTTTTC AAATGTAACT TGGTTCCATG




 3301
CTATACATGT TTCCGGAACC AATGGAACAA AGAGATTTGA TAACCCAGTG




 3351
TTACCATTTA ACGACGGAGT TTATTTCGCA TCAACTGAGA AATCCAATAT




 3401
CATTAGAGGC TGGATTTTCG GAACGACCCT GGATTCTAAA ACGCAATCCT




 3451
TGCTGATTGT TAATAATGCA ACAAATGTGG TCATTAAAGT CTGTGAATTC




 3501
CAATTTTGCA ATGATCCATT TCTCGGCGTC TATTACCACA AGAATAACAA




 3551
ATCTTGGATG GAGTCAGAGT TCAGGGTTTA TAGTTCCGCA AATAATTGTA




 3601
CTTTTGAATA CGTTTCCCAA CCATTCTTAA TGGACTTGGA GGGAAAACAG




 3651
GGAAATTTTA AGAATCTAAG AGAATTCGTC TTTAAGAATA TTGATGGATA




 3701
TTTCAAGATC TATTCAAAAC ATACACCTAT AAACCTAGTT AGAGATCTCC




 3751
CGCAAGGGTT TTCAGCCCTA GAGCCACTAG TTGACCTGCC AATTGGGATC




 3801
AACATTACTA GATTCCAGAC CCTACTCGCT CTGCATCGGT CATATTTGAC




 3851
ACCAGGAGAT TCATCGTCAG GATGGACCGC TGGAGCAGCT GCTTACTATG




 3901
TTGGGTATCT GCAACCTAGA ACATTTCTCC TAAAGTATAA TGAAAACGGG




 3951
ACTATTACAG ACGCAGTCGA TTGCGCACTG GATCCACTCT CAGAGACAAA




 4001
GTGCACTCTA AAATCATTCA CTGTCGAGAA AGGAATCTAT CAAACATCAA




 4051
ATTTCAGGGT CCAGCCAACT GAGAGTATTG TCCGGTTCCC TAACATAACT




 4101
AACTTGTGCC CCTTCGGAGA GGTTTTCAAT GCTACTCGGT TCGCCAGCGT




 4151
CTACGCATGG AACAGAAAGA GGATTTCAAA CTGTGTCGCA GATTATAGCG




 4201
TCCTCTATAA TTCAGCATCA TTCAGTACAT TTAAATGCTA TGGTGTCAGC




 4251
CCCACCAAAC TTAATGACTT ATGTTTTACC AATGTATATG CAGATTCCTT




 4301
TGTAATCAGA GGTGACGAAG TGAGGCAAAT CGCACCTGGA CAGACCGGAA




 4351
AGATTGCTGA TTATAATTAT AAACTCCCTG ATGATTTTAC CGGATGTGTT




 4401
ATTGCTTGGA ACAGCAATAA CCTCGATAGT AAGGTCGGAG GAAACTATAA




 4451
CTATTTGTAC AGACTGTTTA GAAAGTCGAA TTTGAAACCT TTTGAAAGAG




 4501
ACATATCCAC CGAGATTTAC CAGGCGGGCA GCACACCGTG TAATGGTGTA




 4551
GAAGGATTCA ATTGTTACTT TCCCCTGCAA TCATATGGGT TTCAACCAAC




 4601
CAATGGAGTC GGATATCAAC CATATCGTGT CGTCGTCCTT TCCTTCGAGC




 4651
TGCTTCATGC ACCAGCTACA GTCTGCGGAC CTAAGAAGAG CACTAATCTT




 4701
GTCAAGAACA AATGTGTGAA CTTTAATTTT AATGGATTAA CAGGAACCGG




 4751
AGTTTTGACC GAGAGTAATA AGAAGTTCTT GCCGTTCCAG CAATTTGGAC




 4801
GAGACATTGC TGACACCACA GATGCGGTTC GTGACCCGCA AACTTTAGAG




 4851
ATCCTAGACA TCACCCCATG TTCATTCGGT GGAGTTTCCG TTATTACTCC




 4901
TGGAACGAAT ACAAGCAATC AAGTTGCCGT TCTCTATCAA GATGTTAATT




 4951
GTACAGAAGT GCCTGTGGCC ATTCATGCAG ATCAACTAAC ACCAACTTGG




 5001
AGAGTTTACA GCACTGGGTC CAATGTCTTC CAAACGCGCG CCGGCTGCCT




 5051
CATTGGTGCA GAATATGTGA ATAACTCATA CGAATGTGAC ATTCCAATCG




 5101
GTGCCGGCAT ATGCGCCTCT TACCAGACTC AGACTAATTC GCCAAGAAGA




 5151
GCCAGGTCTG TCGCAAGTCA GTCAATTATT GCATACACAA TGTCGTTAGG




 5201
AGCAGAGAAT AGTGTAGCAT ACTCAAACAA TTCTATAGCA ATACCTACCA




 5251
ACTTCACTAT ATCAGTAACT ACAGAAATAT TGCCAGTATC CATGACTAAA




 5301
ACAAGTGTGG ATTGCACCAT GTACATCTGT GGAGATTCCA CAGAATGCAG




 5351
CAATCTTCTC TTGCAATACG GATCATTCTG CACACAACTG AATAGGGCAC




 5401
TGACTGGAAT TGCAGTCGAG CAGGATAAGA ACACACAGGA GGTGTTTGCC




 5451
CAAGTCAAAC AAATATACAA AACACCACCC ATCAAGGATT TTGGAGGATT




 5501
TAATTTCTCA CAAATACTCC CCGACCCATC CAAGCCCTCC AAAAGGAGTT




 5551
TCATTGAGGA CCTCTTGTTT AATAAGGTTA CCTTGGCAGA TGCCGGGTTT




 5601
ATTAAGCAGT ACGGCGACTG TCTTGGAGAC ATAGCAGCCA GAGATCTAAT




 5651
TTGTGCCCAG AAATTCAATG GACTGACAGT CCTGCCTCCC TTATTAACTG




 5701
ATGAGATGAT AGCTCAGTAT ACATCAGCAT TGTTGGCTGG TACAATTACA




 5751
TCTGGATGGA CATTTGGTGC CGGAGCGGCA TTACAAATCC CTTTTGCAAT




 5801
GCAAATGGCC TATAGATTTA ATGGAATCGG AGTAACTCAA AATGTTTTAT




 5851
ATGAGAATCA GAAATTAATT GCAAATCAAT TCAATTCAGC TATAGGAAAG




 5901
ATTCAGGATT CACTCAGTAG TACAGCAAGC GCTCTAGGCA AATTACAAGA




 5951
CGTCGTCAAT CAGAATGCAC AGGCATTAAA TACACTGGTG AAGCAATTGA




 6001
GTTCCAATTT CGGAGCAATT TCATCTGTTC TAAATGATAT ATTGTCAAGA




 6051
CTGGATAAAG TAGAAGCCGA GGTCCAAATC GATAGGCTGA TCACAGGAAG




 6101
ACTTCAATCA CTACAGACAT ACGTCACCCA ACAACTCATC AGAGCAGCAG




 6151
AAATTAGAGC CTCTGCTAAT CTAGCCGCAA CAAAGATGTC AGAGTGCGTA




 6201
TTGGGACAAT CTAAGAGGGT CGACTTTTGT GGAAAGGGGT ATCACTTGAT




 6251
GTCCTTTCCT CAATCTGCAC CACACGGAGT TGTCTTCTTA CATGTAACAT




 6301
ATGTGCCCGC TCAAGAAAAG AATTTCACTA CAGCACCTGC AATATGTCAT




 6351
GACGGAAAAG CACATTTTCC TCGGGAGGGA GTTTTCGTTT CTAATGGAAC




 6401
CCATTGGTTC GTGACCCAAA GGAACTTTTA CGAGCCTCAA ATAATTACAA




 6451
CTGATAATAC ATTCGTTTCT GGAAATTGCG ACGTAGTTAT AGGTATTGTA




 6501
AATAATACTG TTTATGACCC TTTACAACCT GAACTCGATT CCTTCAAGGA




 6551
AGAACTCGAC AAATATTTTA AGAATCACAC CTCACCGGAC GTTGACTTAG




 6601
GAGACATTTC CGGGATTAAC GCTAGTGTAG TCAATATCCA AAAGGAGATA




 6651
GATAGACTGA ATGAGGTAGC AAAGAATCTT AATGAATCTT TGATCGACCT




 6701
TCAGGAGCTG GGGAAGTACG AACAATACAT AAAATGGCCA TGGTACATTT




 6751
GGCTCGGGTT TATTGCTGGA CTAATTGCAA TAGTCATGGT CACTATCATG




 6801
CTGTGTTGTA TGACATCGTG CTGCTCATGC CTCAAGGGAT GTTGTAGCTG




 6851
TTGATCTTGT TGCAAGTTCG ATGAGGATGA TTCAGAACCA GTTTTAAAAG




 6901
GAGTAAAGTT GCATTACACA TAAAGGCTAG CTGTTTACGC GTTATCCATG




 6951
CTCAAAGAGG CCTCAATTAT ATTTGAGTTT TTAATTTTTA TGAAAAAAAC




 7001
TAACAGCAAT CATGGAAGTC CACGATTTTG AGACCGACGA GTTCAATGAT




 7051
TTCAATGAAG ATGACTATGC CACAAGAGAA TTCCTGAATC CCGATGAGCG




 7101
CATGACGTAC TTGAATCATG CTGATTACAA CCTGAATTCT CCTCTAATTA




 7151
GTGATGATAT TGACAATTTA ATCAGGAAAT TCAATTCTCT TCCAATTCCC




 7201
TCGATGTGGG ATAGTAAGAA CTGGGATGGA GTTCTTGAGA TGTTAACGTC




 7251
ATGTCAAGCC AATCCCATCC CAACATCTCA GATGCATAAA TGGATGGGAA




 7301
GTTGGTTAAT GTCTGATAAT CATGATGCCA GTCAAGGGTA TAGTTTTTTA




 7351
CATGAAGTGG ACAAAGAGGC AGAAATAACA TTTGACGTGG TGGAGACCTT




 7401
CATCCGCGGC TGGGGCAACA AACCAATTGA ATACATCAAA AAGGAAAGAT




 7451
GGACTGACTC ATTCAAAATT CTCGCTTATT TGTGTCAAAA GTTTTTGGAC




 7501
TTACACAAGT TGACATTAAT CTTAAATGCT GTCTCTGAGG TGGAATTGCT




 7551
CAACTTGGCG AGGACTTTCA AAGGCAAAGT CAGAAGAAGT TCTCATGGAA




 7601
CGAACATATG CAGGATTAGG GTTCCCAGCT TGGGTCCTAC TTTTATTTCA




 7651
GAAGGATGGG CTTACTTCAA GAAACTTGAT ATTCTAATGG ACCGAAACTT




 7701
TCTGTTAATG GTCAAAGATG TGATTATAGG GAGGATGCAA ACGGTGCTAT




 7751
CCATGGTATG TAGAATAGAC AACCTGTTCT CAGAGCAAGA CATCTTCTCC




 7801
CTTCTAAATA TCTACAGAAT TGGAGATAAA ATTGTGGAGA GGCAGGGAAA




 7851
TTTTTCTTAT GACTTGATTA AAATGGTGGA ACCGATATGC AACTTGAAGC




 7901
TGATGAAATT AGCAAGAGAA TCAAGGCCTT TAGTCCCACA ATTCCCTCAT




 7951
TTTGAAAATC ATATCAAGAC TTCTGTTGAT GAAGGGGCAA AAATTGACCG




 8001
AGGTATAAGA TTCCTCCATG ATCAGATAAT GAGTGTGAAA ACAGTGGATC




 8051
TCACACTGGT GATTTATGGA TCGTTCAGAC ATTGGGGTCA TCCTTTTATA




 8101
GATTATTACA CTGGACTAGA AAAATTACAT TCCCAAGTAA CCATGAAGAA




 8151
AGATATTGAT GTGTCATATG CAAAAGCACT TGCAAGTGAT TTAGCTCGGA




 8201
TTGTTCTATT TCAACAGTTC AATGATCATA AAAAGTGGTT CGTGAATGGA




 8251
GACTTGCTCC CTCATGATCA TCCCTTTAAA AGTCATGTTA AAGAAAATAC




 8301
ATGGCCCACA GCTGCTCAAG TTCAAGATTT TGGAGATAAA TGGCATGAAC




 8351
TTCCGCTGAT TAAATGTTTT GAAATACCCG ACTTACTAGA CCCATCGATA




 8401
ATATACTCTG ACAAAAGTCA TTCAATGAAT AGGTCAGAGG TGTTGAAACA




 8451
TGTCCGAATG AATCCGAACA CTCCTATCCC TAGTAAAAAG GTGTTGCAGA




 8501
CTATGTTGGA CACAAAGGCT ACCAATTGGA AAGAATTTCT TAAAGAGATT




 8551
GATGAGAAGG GCTTAGATGA TGATGATCTA ATTATTGGTC TTAAAGGAAA




 8601
GGAGAGGGAA CTGAAGTTGG CAGGTAGATT TTTCTCCCTA ATGTCTTGGA




 8651
AATTGCGAGA ATACTTTGTA ATTACCGAAT ATTTGATAAA GACTCATTTC




 8701
GTCCCTATGT TTAAAGGCCT GACAATGGCG GACGATCTAA CTGCAGTCAT




 8751
TAAAAAGATG TTAGATTCCT CATCCGGCCA AGGATTGAAG TCATATGAGG




 8801
CAATTTGCAT AGCCAATCAC ATTGATTACG AAAAATGGAA TAACCACCAA




 8851
AGGAAGTTAT CAAACGGCCC AGTGTTCCGA GTTATGGGCC AGTTCTTAGG




 8901
TTATCCATCC TTAATCGAGA GAACTCATGA ATTTTTTGAG AAAAGTCTTA




 8951
TATACTACAA TGGAAGACCA GACTTGATGC GTGTTCACAA CAACACACTG




 9001
ATCAATTCAA CCTCCCAACG AGTTTGTTGG CAAGGACAAG AGGGTGGACT




 9051
GGAAGGTCTA CGGCAAAAAG GATGGAGTAT CCTCAATCTA CTGGTTATTC




 9101
AAAGAGAGGC TAAAATCAGA AACACTGCTG TCAAAGTCTT GGCACAAGGT




 9151
GATAATCAAG TTATTTGCAC ACAGTATAAA ACGAAGAAAT CGAGAAACGT




 9201
TGTAGAATTA CAGGGTGCTC TCAATCAAAT GGTTTCTAAT AATGAGAAAA




 9251
TTATGACTGC AATCAAAATA GGGACAGGGA AGTTAGGACT TTTGATAAAT




 9301
GACGATGAGA CTATGCAATC TGCAGATTAC TTGAATTATG GAAAAATACC




 9351
GATTTTCCGT GGAGTGATTA GAGGGTTAGA GACCAAGAGA TGGTCACGAG




 9401
TGACTTGTGT CACCAATGAC CAAATACCCA CTTGTGCTAA TATAATGAGC




 9451
TCAGTTTCCA CAAATGCTCT CACCGTAGCT CATTTTGCTG AGAACCCAAT




 9501
CAATGCCATG ATACAGTACA ATTATTTTGG GACATTTGCT AGACTCTTGT




 9551
TGATGATGCA TGATCCTGCT CTTCGTCAAT CATTGTATGA AGTTCAAGAT




 9601
AAGATACCAG GCTTGCACAG TTCTACTTTC AAATACGCCA TGTTGTATTT




 9651
GGACCCTTCC ATTGGAGGAG TGTCGGGCAT GTCTTTGTCC AGGTTTTTGA




 9701
TTAGAGCCTT CCCAGATCCC GTAACAGAAA GTCTCTCATT CTGGAGATTC




 9751
ATCCATGTAC ATGCTCGAAG TGAGCATCTG AAGGAGATGA GTGCAGTATT




 9801
TGGAAACCCC GAGATAGCCA AGTTTCGAAT AACTCACATA GACAAGCTAG




 9851
TAGAAGATCC AACCTCTCTG AACATCGCTA TGGGAATGAG TCCAGCGAAC




 9901
TTGTTAAAGA CTGAGGTTAA AAAATGCTTA ATCGAATCAA GACAAACCAT




 9951
CAGGAACCAG GTGATTAAGG ATGCAACCAT ATATTTGTAT CATGAAGAGG




10001
ATCGGCTCAG AAGTTTCTTA TGGTCAATAA ATCCTCTGTT CCCTAGATTT




10051
TTAAGTGAAT TCAAATCAGG CACTTTTTTG GGAGTCGCAG ACGGGCTCAT




10101
CAGTCTATTT CAAAATTCTC GTACTATTCG GAACTCCTTT AAGAAAAAGT




10151
ATCATAGGGA ATTGGATGAT TTGATTGTGA GGAGTGAGGT ATCCTCTTTG




10201
ACACATTTAG GGAAACTTCA TTTGAGAAGG GGATCATGTA AAATGTGGAC




10251
ATGTTCAGCT ACTCATGCTG ACACATTAAG ATACAAATCC TGGGGCCGTA




10301
CAGTTATTGG GACAACTGTA CCCCATCCAT TAGAAATGTT GGGTCCACAA




10351
CATCGAAAAG AGACTCCTTG TGCACCATGT AACACATCAG GGTTCAATTA




10401
TGTTTCTGTG CATTGTCCAG ACGGGATCCA TGACGTCTTT AGTTCACGGG




10451
GACCATTGCC TGCTTATCTA GGGTCTAAAA CATCTGAATC TACATCTATT




10501
TTGCAGCCTT GGGAAAGGGA AAGCAAAGTC CCACTGATTA AAAGAGCTAC




10551
ACGTCTTAGA GATGCTATCT CTTGGTTTGT TGAACCCGAC TCTAAACTAG




10601
CAATGACTAT ACTTTCTAAC ATCCACTCTT TAACAGGCGA AGAATGGACC




10651
AAAAGGCAGC ATGGGTTCAA AAGAACAGGG TCTGCCCTTC ATAGGTTTTC




10701
GACATCTCGG ATGAGCCATG GTGGGTTCGC ATCTCAGAGC ACTGCAGCAT




10751
TGACCAGGTT GATGGCAACT ACAGACACCA TGAGGGATCT GGGAGATCAG




10801
AATTTCGACT TTTTATTCCA AGCAACGTTG CTCTATGCTC AAATTACCAC




10851
CACTGTTGCA AGAGACGGAT GGATCACCAG TTGTACAGAT CATTATCATA




10901
TTGCCTGTAA GTCCTGTTTG AGACCCATAG AAGAGATCAC CCTGGACTCA




10951
AGTATGGACT ACACGCCCCC AGATGTATCC CATGTGCTGA AGACATGGAG




11001
GAATGGGGAA GGTTCGTGGG GACAAGAGAT AAAACAGATA TATCCTTTAG




11051
AAGGGAATTG GAAGAATTTA GCACCTGCTG AGCAATCCTA TCAAGTCGGC




11101
AGATGTATAG GTTTTCTATA TGGAGACTTG GCGTATAGAA AATCTACTCA




11151
TGCCGAGGAC AGTTCTCTAT TTCCTCTATC TATACAAGGT CGTATTAGAG




11201
GTCGAGGTTT CTTAAAAGGG TTGCTAGACG GATTAATGAG AGCAAGTTGC




11251
TGCCAAGTAA TACACCGGAG AAGTCTGGCT CATTTGAAGA GGCCGGCCAA




11301
CGCAGTGTAC GGAGGTTTGA TTTACTTGAT TGATAAATTG AGTGTATCAC




11351
CTCCATTCCT TTCTCTTACT AGATCAGGAC CTATTAGAGA CGAATTAGAA




11401
ACGATTCCCC ACAAGATCCC AACCTCCTAT CCGACAAGCA ACCGTGATAT




11451
GGGGGTGATT GTCAGAAATT ACTTCAAATA CCAATGCCGT CTAATTGAAA




11501
AGGGAAAATA CAGATCACAT TATTCACAAT TATGGTTATT CTCAGATGTC




11551
TTATCCATAG ACTTCATTGG ACCATTCTCT ATTTCCACCA CCCTCTTGCA




11601
AATCCTATAC AAGCCATTTT TATCTGGGAA AGATAAGAAT GAGTTGAGAG




11651
AGCTGGCAAA TCTTTCTTCA TTGCTAAGAT CAGGAGAGGG GTGGGAAGAC




11701
ATACATGTGA AATTCTTCAC CAAGGACATA TTATTGTGTC CAGAGGAAAT




11751
CAGACATGCT TGCAAGTTCG GGATTGCTAA GGATAATAAT AAAGACATGA




11801
GCTATCCCCC TTGGGGAAGG GAATCCAGAG GGACAATTAC AACAATCCCT




11851
GTTTATTATA CGACCACCCC TTACCCAAAG ATGCTAGAGA TGCCTCCAAG




11901
AATCCAAAAT CCCCTGCTGT CCGGAATCAG GTTGGGCCAA TTACCAACTG




11951
GCGCTCATTA TAAAATTCGG AGTATATTAC ATGGAATGGG AATCCATTAC




12001
AGGGACTTCT TGAGTTGTGG AGACGGCTCC GGAGGGATGA CTGCTGCATT




12051
ACTACGAGAA AATGTGCATA GCAGAGGAAT ATTCAATAGT CTGTTAGAAT




12101
TATCAGGGTC AGTCATGCGA GGCGCCTCTC CTGAGCCCCC CAGTGCCCTA




12151
GAAACTTTAG GAGGAGATAA ATCGAGATGT GTAAATGGTG AAACATGTTG




12201
GGAATATCCA TCTGACTTAT GTGACCCAAG GACTTGGGAC TATTTCCTCC




12251
GACTCAAAGC AGGCTTGGGG CTTCAAATTG ATTTAATTGT AATGGATATG




12301
GAAGTTCGGG ATTCTTCTAC TAGCCTGAAA ATTGAGACGA ATGTTAGAAA




12351
TTATGTGCAC CGGATTTTGG ATGAGCAAGG AGTTTTAATC TACAAGACTT




12401
ATGGAACATA TATTTGTGAG AGCGAAAAGA ATGCAGTAAC AATCCTTGGT




12451
CCCATGTTCA AGACGGTCGA CTTAGTTCAA ACAGAATTTA GTAGTTCTCA




12501
AACGTCTGAA GTATATATGG TATGTAAAGG TTTGAAGAAA TTAATCGATG




12551
AACCCAATCC CGATTGGTCT TCCATCAATG AATCCTGGAA AAACCTGTAC




12601
GCATTCCAGT CATCAGAACA GGAATTTGCC AGAGCAAAGA AGGTTAGTAC




12651
ATACTTTACC TTGACAGGTA TTCCCTCCCA ATTCATTCCT GATCCTTTTG




12701
TAAACATTGA GACTATGCTA CAAATATTCG GAGTACCCAC GGGTGTGTCT




12751
CATGCGGCTG CCTTAAAATC ATCTGATAGA CCTGCAGATT TATTGACCAT




12801
TAGCCTTTTT TATATGGCGA TTATATCGTA TTATAACATC AATCATATCA




12851
GAGTAGGACC GATACCTCCG AACCCCCCAT CAGATGGAAT TGCACAAAAT




12901
GTGGGGATCG CTATAACTGG TATAAGCTTT TGGCTGAGTT TGATGGAGAA




12951
AGACATTCCA CTATATCAAC AGTGTTTAGC AGTTATCCAG CAATCATTCC




13001
CGATTAGGTG GGAGGCTGTT TCAGTAAAAG GAGGATACAA GCAGAAGTGG




13051
AGTACTAGAG GTGATGGGCT CCCAAAAGAT ACCCGAATTT CAGACTCCTT




13101
GGCCCCAATC GGGAACTGGA TCAGATCTCT GGAATTGGTC CGAAACCAAG




13151
TTCGTCTAAA TCCATTCAAT GAGATCTTGT TCAATCAGCT ATGTCGTACA




13201
GTGGATAATC ATTTGAAATG GTCAAATTTG CGAAGAAACA CAGGAATGAT




13251
TGAATGGATC AATAGACGAA TTTCAAAAGA AGACCGGTCT ATACTGATGT




13301
TGAAGAGTGA CCTACACGAG GAAAACTCTT GGAGAGATTA AAAAATCATG




13351
AGGAGACTCC AAACTTTAAG TATGAAAAAA ACTTTGATCC TTAAGACCCT




13401
CTTGTGGTTT TTATTTTTTA TCTGGTTTTG TGGTCTTCGT













Amino acid sequence for VSV-
Please see FIGS. 21B, 22B, 23B, and 24B.
155


N protein in rVSVΔG-SARS-




CoV-2 clones MB1 and MB2,




and Vectors A and B







Amino acid sequence for VSV-
Please see FIGS. 21C, 22C, 23C, and 24C.
156


P protein in rVSVΔG-SARS-




CoV-2 clones MB1 and MB2,




and Vectors A and B







Amino acid sequence for VSV-
Please see FIGS. 21D, 22D, and 23D.
157


M protein in rVSVΔG-SARS-




CoV-2 clone MB2, and Vectors




A and B







Amino acid sequence for
Please see FIG. 21E.
158


SARS-CoV-2 S protein in




rVSVΔG-SARS-CoV-2 clone




MB2















Nucleotide sequence for
    1 
acgaagacaa acaaaccatt attatcatta aaaggctcag gagaaacttt aacagtaatc
159


rVSVΔG-SARS-CoV-2 with
   61 
aaaatgtctg ttacagtcaa gagaatcatt gacaacacag tcgtagttcc aaaacttcct



codon-optimized, wild-type
  121 
gcaaatgagg atccagtgga atacccggca gattacttca gaaaatcaaa ggagattcct



S protein
  181 
ctttacatca atactacaaa aagtttgtca gatctaagag gatatgtcta ccaaggcctc




  241 
aaatccggaa atgtatcaat catacatgtc aacagctact tgtatggagc attaaaggac




  301 
atccggggta agttggataa agattggtca agtttcggaa taaacatcgg gaaagcaggg




  361 
gatacaatcg gaatatttga ccttgtatcc ttgaaagccc tggacggcgt acttccagat




  421 
ggagtatcgg atgcttccag aaccagcgca gatgacaaat ggttgccttt gtatctactt




  481 
ggcttataca gagtgggcag aacacaaatg cctgaataca gaaaaaagct catggatggg




  541 
ctgacaaatc aatgcaaaat gatcaatgaa cagtttgaac ctcttgtgcc agaaggtcgt




  601
gacatttttg atgtgtgggg aaatgacagt aattacacaa aaattgtcgc tgcagtggac




  661
atgttcttcc acatgttcaa aaaacatgaa tgtgcctcgt tcagatacgg aactattgtt




  721
tccagattca aagattgtgc tgcattggca acatttggac acctctgcaa aataaccgga




  781
atgtctacag aagatgtaac gacctggatc ttgaaccgag aagttgcaga tgaaatggtc




  841
caaatgatgc ttccaggcca agaaattgac aaggccgatt catacatgcc ttatttgatc




  901
gactttggat tgtcttctaa gtctccatat tcttccgtca aaaaccctgc cttccacttc




  961
tgggggcaat tgacagctct tctgctcaga tccaccagag caaggaatgc ccgacagcct




 1021 
gatgacattg agtatacatc tcttactaca gcaggtttgt tgtacgctta tgcagtagga




 1081 
tcctctgccg acttggcaca acagttttgt gttggagata acaaatacac tccagatgat




 1141 
agtaccggag gattgacgac taatgcaccg ccacaaggca gagatgtggt cgaatggctc




 1201 
ggatggtttg aagatcaaaa cagaaaaccg actcctgata tgatgcagta tgcgaaaaga




 1261 
gcagtcatgt cactgcaagg cctaagagag aagacaattg gcaagtatgc taagtcagaa




 1321 
tttgacaaat gaccctataa ttctcagatc acctattata tattatgcta catatgaaaa




 1381 
aaactaacag atatcatgga taatctcaca aaagttcgtg agtatctcaa gtcctattct




 1441 
cgtctggatc aggcggtagg agagatagat gagatcgaag cacaacgagc tgaaaagtcc




 1501 
aattatgagt tgttccaaga ggatggagtg gaagagcata ctaagccctc ttattttcag




 1561 
gcagcagatg attctgacac agaatctgaa ccagaaattg aagacaatca aggcttgtat




 1621 
gcaccagatc cagaagctga gcaagttgaa ggctttatac aggggccttt agatgactat




 1681 
gcagatgagg aagtggatgt tgtatttact tcggactgga aacagcctga gcttgaatct




 1741 
gacgagcatg gaaagacctt acggttgaca tcgccagagg gtttaagtgg agagcagaaa




 1801 
tcccagtggc tttcgacgat taaagcagtc gtgcaaagtg ccaaatactg gaatctggca




 1861 
gagtgcacat ttgaagcatc gggagaaggg gtcattatga aggagcgcca gataactccg




 1921 
gatgtatata aggtcactcc agtgatgaac acacatccgt cccaatcaga agcagtatca




 1981 
gatgtttggt ctctctcaaa gacatccatg actttccaac ccaagaaagc aagtcttcag




 2041 
cctctcacca tatccttgga tgaattgttc tcatctagag gagagttcat ctctgtcgga




 2101 
ggtgacggac gaatgtctca taaagaggcc atcctgctcg gcctgagata caaaaagttg




 2161 
tacaatcagg cgagagtcaa atattctctg tagactatga aaaaaagtaa cagatatcac




 2221 
gatctaagtg ttatcccaat ccattcatca tgagttcctt aaagaagatt ctcggtctga




 2281 
aggggaaagg taagaaatct aagaaattag ggatcgcacc acccccttat gaagaggaca




 2341 
ctagcatgga gtatgctccg agcgctccaa ttgacaaatc ctattttgga gttgacgaga




 2401 
tggacaccta tgatccgaat caattaagat atgagaaatt cttctttaca gtgaaaatga




 2461 
cggttagatc taatcgtccg ttcagaacat actcagatgt ggcagccgct gtatcccatt




 2521 
gggatcacat gtacatcgga atggcaggga aacgtccctt ctacaaaatc ttggcttttt




 2581 
tgggttcttc taatctaaag gccactccag cggtattggc agatcaaggt caaccagagt




 2641 
atcacgctca ctgcgaaggc agggcttatt tgccacatag gatggggaag acccctccca




 2701 
tgctcaatgt accagagcac ttcagaagac cattcaatat aggtctttac aagggaacga




 2761 
ttgagctcac aatgaccatc tacgatgatg agtcactgga agcagctcct atgatctggg




 2821 
atcatttcaa ttcttccaaa ttttctgatt tcagagagaa ggccttaatg tttggcctga




 2881 
ttgtcgagaa aaaggcatct ggagcgtggg tcctggactc tatcggccac ttcaaatgag




 2941 
ctagtctaac ttctagcttc tgaacaatcc ccggtttact cagtctcccc taattccagc




 3001 
ctctcgaaca actaatatcc tgtcttttct atccctatga aaaaaactaa cagagatcga




 3061 
tctgtttacg cgctagtgga tcctactcga gaggagccac catgttcgtg ttcctggtgc




 3121 
tattacctct ggtttcgtct caatgcgtaa accttacaac tagaactcag cttcctccag




 3181 
catacacaaa ttccttcact cgcggagtgt attatcctga taaggtcttt cgatcatcag




 3241 
tgttgcattc cacccaggat ttgtttctcc cgttcttttc aaatgtaact tggttccatg




 3301 
ctatacatgt ttccggaacc aatggaacaa agagatttga taacccagtg ttaccattta




 3361 
acgacggagt ttatttcgca tcaactgaga aatccaatat cattagaggc tggattttcg




 3421 
gaacgaccct ggattctaaa acgcaatcct tgctgattgt taataatgca acaaatgtgg




 3481 
tcattaaagt ctgtgaattc caattttgca atgatccatt tctcggcgtc tattaccaca




 3541 
agaataacaa atcttggatg gagtcagagt tcagggttta tagttccgca aataattgta




 3601 
cttttgaata cgtttcccaa ccattcttaa tggacttgga gggaaaacag ggaaatttta




 3661 
agaatctaag agaattcgtc tttaagaata ttgatggata tttcaagatc tattcaaaac




 3721 
atacacctat aaacctagtt agagatctcc cgcaagggtt ttcagcccta gagccactag




 3781 
ttgacctgcc aattgggatc aacattacta gattccagac cctactcgct ctgcatcggt




 3841 
catatttgac accaggagat tcatcgtcag gatggaccgc tggagcagct gcttactatg




 3901 
ttgggtatct gcaacctaga acatttctcc taaagtataa tgaaaacggg actattacag




 3961 
acgcagtcga ttgcgcactg gatccactct cagagacaaa gtgcactcta aaatcattca




 4021 
ctgtcgagaa aggaatctat caaacatcaa atttcagggt ccagccaact gagagtattg




 4081 
tccggttccc taacataact aacttgtgcc ccttcggaga ggttttcaat gctactcggt




 4141 
tcgccagcgt ctacgcatgg aacagaaaga ggatttcaaa ctgtgtcgca gattatagcg




 4201 
tcctctataa ttcagcatca ttcagtacat ttaaatgcta tggtgtcagc cccaccaaac




 4261 
ttaatgactt atgttttacc aatgtatatg cagattcctt tgtaatcaga ggtgacgaag




 4321 
tgaggcaaat cgcacctgga cagaccggaa agattgctga ttataattat aaactccctg




 4381 
atgattttac cggatgtgtt attgcttgga acagcaataa cctcgatagt aaggtcggag




 4441 
gaaactataa ctatttgtac agactgttta gaaagtcgaa tttgaaacct tttgaaagag




 4501 
acatatccac cgagatttac caggcgggca gcacaccgtg taatggtgta gaaggattca




 4561 
attgttactt tcccctgcaa tcatatgggt ttcaaccaac caatggagtc ggatatcaac




 4621 
catatcgtgt cgtcgtcctt tccttcgagc tgcttcatgc accagctaca gtctgcggac




 4681 
ctaagaagag cactaatctt gtcaagaaca aatgtgtgaa ctttaatttt aatggattaa




 4741 
caggaaccgg agttttgacc gagagtaata agaagttctt gccgttccag caatttggac




 4801 
gagacattgc tgacaccaca gatgcggttc gtgacccgca aactttagag atcctagaca




 4861 
tcaccccatg ttcattcggt ggagtttccg ttattactcc tggaacgaat acaagcaatc




 4921 
aagttgccgt tctctatcaa gatgttaatt gtacagaagt gcctgtggcc attcatgcag




 4981 
atcaactaac accaacttgg agagtttaca gcactgggtc caatgtcttc caaacgcgcg




 5041 
ccggctgcct cattggtgca gaacatgtga ataactcata cgaatgtgac attccaatcg




 5101 
gtgccggcat atgcgcctct taccagactc agactaattc gccaagaaga gccaggtctg




 5161 
tcgcaagtca gtcaattatt gcatacacaa tgtcgttagg agcagagaat agtgtagcat




 5221 
actcaaacaa ttctatagca atacctacca acttcactat atcagtaact acagaaatat




 5281 
tgccagtatc catgactaaa acaagtgtgg attgcaccat gtacatctgt ggagattcca




 5341 
cagaatgcag caatcttctc ttgcaatacg gatcattctg cacacaactg aatagggcac




 5401 
tgactggaat tgcagtcgag caggataaga acacacagga ggtgtttgcc caagtcaaac




 5461 
aaatatacaa aacaccaccc atcaaggatt ttggaggatt taatttctca caaatactcc




 5521 
ccgacccatc caagccctcc aaaaggagtt tcattgagga cctcttgttt aataaggtta




 5581 
ccttggcaga tgccgggttt attaagcagt acggcgactg tcttggagac atagcagcca




 5641 
gagatctaat ttgtgcccag aaattcaatg gactgacagt cctgcctccc ttattaactg




 5701 
atgagatgat agctcagtat acatcagcat tgttggctgg tacaattaca tctggatgga




 5761 
catttggtgc cggagcggca ttacaaatcc cttttgcaat gcaaatggcc tatagattta




 5821 
atggaatcgg agtaactcaa aatgttttat atgagaatca gaaattaatt gcaaatcaat




 5881 
tcaattcagc tataggaaag attcaggatt cactcagtag tacagcaagc gctctaggca




 5941 
aattacaaga cgtcgtcaat cagaatgcac aggcattaaa tacactggtg aagcaattga




 6001 
gttccaattt cggagcaatt tcatctgttc taaatgatat attgtcaaga ctggataaag




 6061 
tagaagccga ggtccaaatc gataggctga tcacaggaag acttcaatca ctacagacat




 6121 
acgtcaccca acaactcatc agagcagcag aaattagagc ctctgctaat ctagccgcaa




 6181 
caaagatgtc agagtgcgta ttgggacaat ctaagagggt cgacttttgt ggaaaggggt




 6241 
atcacttgat gtcctttcct caatctgcac cacacggagt tgtcttctta catgtaacat




 6301 
atgtgcccgc tcaagaaaag aatttcacta cagcacctgc aatatgtcat gacggaaaag




 6361 
cacattttcc tcgggaggga gttttcgttt ctaatggaac ccattggttc gtgacccaaa




 6421 
ggaactttta cgagcctcaa ataattacaa ctgataatac attcgtttct ggaaattgcg




 6481 
acgtagttat aggtattgta aataatactg tttatgaccc tttacaacct gaactcgatt




 6541 
ccttcaagga agaactcgac aaatatttta agaatcacac ctcaccggac gttgacttag




 6601 
gagacatttc cgggattaac gctagtgtag tcaatatcca aaaggagata gatagactga




 6661 
atgaggtagc aaagaatctt aatgaatctt tgatcgacct tcaggagctg gggaagtacg




 6721 
aacaatacat aaaatggcca tggtacattt ggctcgggtt tattgctgga ctaattgcaa




 6781 
tagtcatggt cactatcatg ctgtgttgta tgacatcgtg ctgctcatgc ctcaagggat




 6841 
gttgtagctg tggatcttgt tgcaagttcg atgaggatga ttcagaacca gttttaaaag




 6901 
gagtaaagtt gcattacaca taaaggctag ctgtttacgc gttatccatg ctcaaagagg




 6961 
cctcaattat atttgagttt ttaattttta tgaaaaaaaa ctaacagcaa tcatggaagt




 7021 
ccacgatttt gagaccgacg agttcaatga tttcaatgaa gatgactatg ccacaagaga




 7081 
attcctgaat cccgatgagc gcatgacgta cttgaatcat gctgattaca acctgaattc




 7141 
tcctctaatt agtgatgata ttgacaattt aatcaggaaa ttcaattctc ttccaattcc




 7201 
ctcgatgtgg gatagtaaga actgggatgg agttcttgag atgttaacgt catgtcaagc




 7261 
caatcccatc ccaacatctc agatgcataa atggatggga agttggttaa tgtctgataa




 7321 
tcatgatgcc agtcaagggt atagtttttt acatgaagtg gacaaagagg cagaaataac




 7381 
atttgacgtg gtggagacct tcatccgcgg ctggggcaac aaaccaattg aatacatcaa




 7441 
aaaggaaaga tggactgact cattcaaaat tctcgcttat ttgtgtcaaa agtttttgga




 7501 
cttacacaag ttgacattaa tcttaaatgc tgtctctgag gtggaattgc tcaacttggc




 7561 
gaggactttc aaaggcaaag tcagaagaag ttctcatgga acgaacatat gcaggattag




 7621 
ggttcccagc ttgggtccta cttttatttc agaaggatgg gcttacttca agaaacttga




 7681 
tattctaatg gaccgaaact ttctgttaat ggtcaaagat gtgattatag ggaggatgca




 7741 
aacggtgcta tccatggtat gtagaataga caacctgttc tcagagcaag acatcttctc




 7801 
ccttctaaat atctacagaa ttggagataa aattgtggag aggcagggaa atttttctta




 7861 
tgacttgatt aaaatggtgg aaccgatatg caacttgaag ctgatgaaat tagcaagaga




 7921 
atcaaggcct ttagtcccac aattccctca ttttgaaaat catatcaaga cttctgttga




 7981 
tgaaggggca aaaattgacc gaggtataag attcctccat gatcagataa tgagtgtgaa




 8041 
aacagtggat ctcacactgg tgatttatgg atcgttcaga cattggggtc atccttttat




 8101 
agattattac actggactag aaaaattaca ttcccaagta accatgaaga aagatattga




 8161 
tgtgtcatat gcaaaagcac ttgcaagtga tttagctcgg attgttctat ttcaacagtt




 8221 
caatgatcat aaaaagtggt tcgtgaatgg agacttgctc cctcatgatc atccctttaa




 8281 
aagtcatgtt aaagaaaata catggcccac agctgctcaa gttcaagatt ttggagataa




 8341 
atggcatgaa cttccgctga ttaaatgttt tgaaataccc gacttactag acccatcgat




 8401 
aatatactct gacaaaagtc attcaatgaa taggtcagag gtgttgaaac atgtccgaat




 8461 
gaatccgaac actcctatcc ctagtaaaaa ggtgttgcag actatgttgg acacaaaggc




 8521 
taccaattgg aaagaatttc ttaaagagat tgatgagaag ggcttagatg atgatgatct




 8581 
aattattggt cttaaaggaa aggagaggga actgaagttg gcaggtagat ttttctccct




 8641 
aatgtcttgg aaattgcgag aatactttgt aattaccgaa tatttgataa agactcattt




 8701 
cgtccctatg tttaaaggcc tgacaatggc ggacgatcta actgcagtca ttaaaaagat




 8761 
gttagattcc tcatccggcc aaggattgaa gtcatatgag gcaatttgca tagccaatca




 8821 
cattgattac gaaaaatgga ataaccacca aaggaagtta tcaaacggcc cagtgttccg




 8881 
agttatgggc cagttcttag gttatccatc cttaatcgag agaactcatg aattttttga




 8941 
gaaaagtctt atatactaca atggaagacc agacttgatg cgtgttcaca acaacacact




 9001 
gatcaattca acctcccaac gagtttgttg gcaaggacaa gagggtggac tggaaggtct




 9061 
acggcaaaaa ggatggagta tcctcaatct actggttatt caaagagagg ctaaaatcag




 9121 
aaacactgct gtcaaagtct tggcacaagg tgataatcaa gttatttgca cacagtataa




 9181 
aacgaagaaa tcgagaaacg ttgtagaatt acagggtgct ctcaatcaaa tggtttctaa




 9241 
taatgagaaa attatgactg caatcaaaat agggacaggg aagttaggac ttttgataaa




 9301 
tgacgatgag actatgcaat ctgcagatta cttgaattat ggaaaaatac cgattttccg




 9361 
tggagtgatt agagggttag agaccaagag atggtcacga gtgacttgtg tcaccaatga




 9421 
ccaaataccc acttgtgcta atataatgag ctcagtttcc acaaatgctc tcaccgtagc




 9481 
tcattttgct gagaacccaa tcaatgccat gatacagtac aattattttg ggacatttgc




 9541 
tagactcttg ttgatgatgc atgatcctgc tcttcgtcaa tcattgtatg aagttcaaga




 9601 
taagatacca ggcttgcaca gttctacttt caaatacgcc atgttgtatt tggacccttc




 9661 
cattggagga gtgtcgggca tgtctttgtc caggtttttg attagagcct tcccagatcc




 9721
cgtaacagaa agtctctcat tctggagatt catccatgta catgctcgaa gtgagcatct




 9781
gaaggagatg agtgcagtat ttggaaaccc cgagatagcc aagtttcgaa taactcacat




 9841
agacaagcta gtagaagatc caacctctct gaacatcgct atgggaatga gtccagcgaa




 9901
cttgttaaag actgaggtta aaaaatgctt aatcgaatca agacaaacca tcaggaacca




 9961
ggtgattaag gatgcaacca tatatttgta tcatgaagag gatcggctca gaagtttctt




10021
atggtcaata aatcctctgt tccctagatt tttaagtgaa ttcaaatcag gcactttttt




10081
gggagtcgca gacgggctca tcagtctatt tcaaaattct cgtactattc ggaactcctt




10141
taagaaaaag tatcataggg aattggatga tttgattgtg aggagtgagg tatcctcttt




10201
gacacattta gggaaacttc atttgagaag gggatcatgt aaaatgtgga catgttcagc




10261
tactcatgct gacacattaa gatacaaatc ctggggccgt acagttattg ggacaactgt




10321
accccatcca ttagaaatgt tgggtccaca acatcgaaaa gagactcctt gtgcaccatg




10381
taacacatca gggttcaatt atgtttctgt gcattgtcca gacgggatcc atgacgtctt




10441
tagttcacgg ggaccattgc ctgcttatct agggtctaaa acatctgaat ctacatctat




10501
tttgcagcct tgggaaaggg aaagcaaagt cccactgatt aaaagagcta cacgtcttag




10561
agatgctatc tcttggtttg ttgaacccga ctctaaacta gcaatgacta tactttctaa




10621
catccactct ttaacaggcg aagaatggac caaaaggcag catgggttca aaagaacagg




10681
gtctgccctt cataggtttt cgacatctcg gatgagccat ggtgggttcg catctcagag




10741
cactgcagca ttgaccaggt tgatggcaac tacagacacc atgagggatc tgggagatca




10801
gaatttcgac tttttattcc aagcaacgtt gctctatgct caaattacca ccactgttgc




10861
aagagacgga tggatcacca gttgtacaga tcattatcat attgcctgta agtcctgttt




10921
gagacccata gaagagatca ccctggactc aagtatggac tacacgcccc cagatgtatc




10981
ccatgtgctg aagacatgga ggaatgggga aggttcgtgg ggacaagaga taaaacagat




11041
ctatccttta gaagggaatt ggaagaattt agcacctgct gagcaatcct atcaagtcgg




11101
cagatgtata ggttttctat atggagactt ggcgtataga aaatctactc atgccgagga




11161
cagttctcta tttcctctat ctatacaagg tcgtattaga ggtcgaggtt tcttaaaagg




11221
gttgctagac ggattaatga gagcaagttg ctgccaagta atacaccgga gaagtctggc




11281
tcatttgaag aggccggcca acgcagtgta cggaggtttg atttacttga ttgataaatt




11341
gagtgtatca cctccattcc tttctcttac tagatcagga cctattagag acgaattaga




11401
aacgattccc cacaagatcc caacctccta tccgacaagc aaccgtgata tgggggtgat




11461
tgtcagaaat tacttcaaat accaatgccg tctaattgaa aagggaaaat acagatcaca




11521
ttattcacaa ttatggttat tctcagatgt cttatccata gacttcattg gaccattctc




11581
tatttccacc accctcttgc aaatcctata caagccattt ttatctggga aagataagaa




11641
tgagttgaga gagctggcaa atctttcttc attgctaaga tcaggagagg ggtgggaaga




11701
catacatgtg aaattcttca ccaaggacat attattgtgt ccagaggaaa tcagacatgc




11761
ttgcaagttc gggattgcta aggataataa taaagacatg agctatcccc cttggggaag




11821
ggaatccaga gggacaatta caacaatccc tgtttattat acgaccaccc cttacccaaa




11881
gatgctagag atgcctccaa gaatccaaaa tcccctgctg tccggaatca ggttgggcca




11941
attaccaact ggcgctcatt ataaaattcg gagtatatta catggaatgg gaatccatta




12001
cagggacttc ttgagttgtg gagacggctc cggagggatg actgctgcat tactacgaga




12061
aaatgtgcat agcagaggaa tattcaatag tctgttagaa ttatcagggt cagtcatgcg




12121
aggcgcctct cctgagcccc ccagtgccct agaaacttta ggaggagata aatcgagatg




12181
tgtaaatggt gaaacatgtt gggaatatcc atctgactta tgtgacccaa ggacttggga




12241
ctatttcctc cgactcaaag caggcttggg gcttcaaatt gatttaattg taatggatat




12301
ggaagttcgg gattcttcta ctagcctgaa aattgagacg aatgttagaa attatgtgca




12361
ccggattttg gatgagcaag gagttttaat ctacaagact tatggaacat atatttgtga




12421
gagcgaaaag aatgcagtaa caatccttgg tcccatgttc aagacggtcg acttagttca




12481
aacagaattt agtagttctc aaacgtctga agtatatatg gtatgtaaag gtttgaagaa




12541
attaatcgat gaacccaatc ccgattggtc ttccatcaat gaatcctgga aaaacctgta




12601
cgcattccag tcatcagaac aggaatttgc cagagcaaag aaggttagta catactttac




12661
cttgacaggt attccctccc aattcattcc tgatcctttt gtaaacattg agactatgct




12721
acaaatattc ggagtaccca cgggtgtgtc tcatgcggct gccttaaaat catctgatag




12781
acctgcagat ttattgacca ttagcctttt ttatatggcg attatatcgt attataacat




12841
caatcatatc agagtaggac cgatacctcc gaacccccca tcagatggaa ttgcacaaaa




12901
tgtggggatc gctataactg gtataagctt ttggctgagt ttgatggaga aagacattcc




12961
actatatcaa cagtgtttag cagttatcca gcaatcattc ccgattaggt gggaggctgt




13021
ttcagtaaaa ggaggataca agcagaagtg gagtactaga ggtgatgggc tcccaaaaga




13081
tacccgaatt tcagactcct tggccccaat cgggaactgg atcagatctc tggaattggt




13141
ccgaaaccaa gttcgtctaa atccattcaa tgagatcttg ttcaatcagc tatgtcgtac




13201
agtggataat catttgaaat ggtcaaattt gcgaagaaac acaggaatga ttgaatggat




13261
caatagacga atttcaaaag aagaccggtc tatactgatg ttgaagagtg acctacacga




13321
ggaaaactct tggagagatt aaaaaatcat gaggagactc caaactttaa gtatgaaaaa




13381
aactttgatc cttaagaccc tcttgtggtt tttatttttt atctggtttt gtggtcttcg




13441
t













Nucleotide sequence for
Please see FIG. 22F.
160


Vector A, which is a VSV




vector without the VSV-G




gene.







Nucleotide sequence for
Please see FIG. 23F.
161


Vector B, which is a VSV




vector without the VSV-G




gene.







Amino acid sequence of the
mfvflvllplvssqc
162


signal sequence (SS) of SEQ




ID NOS: 1, 4, 7, 10, 13, 16,




19, and 22







Amino acid sequence of the
yeqyikwpwyiw
163


membrane-proximal external




region (MPER) of SEQ ID




NOS: 1, 4, 7, 10, and 16







Amino acid sequence of the
lgfiagliaivmvtiml
164


transmembrane region (TM) of




SEQ ID NOS: 1, 4, 7, and 16







Amino acid sequence of the
ccmtsccsclkgccscgscckfdeddsepvlkgvklhyt
165


cytoplasmic tail (CT) of SEQ




ID NO: 1







Kozak sequence of SEQ ID
aggagccacc
166


NOS: 2, 3, 5, 6, 8, 9, 11,




12, 14, 15, 17, 18, 20, 21,




23, and 24







Nucleotide sequence encoding
atgttcgtgttcctggtgctattacctctggtttcgtctcaatgc
167


the SS of SEQ ID NOS: 2, 3,




5, 6, 8, 9, 11, 12, 14, 15,




17, 18, 20, 21, 23, and 24







Nucleotide sequence encoding
tacgaacaatacataaaatggccatggtacatttgg
168


the MPER of SEQ ID NOS: 2,




3, 5, 6, 8, 9, 11, 12, 17,




and 18







Nucleotide sequence encoding
ctcgggtttattgctggactaattgcaatagtcatggtcactatcatgctg
169


the TM of SEQ ID NOS: 2, 3,




5, 6, 8, 9, 17, and 18







Nucleotide sequence encoding
tgttgtatgacatcgtgctgctcatgcctcaagggatgttgtagctgtggatcttgttgcaagttcg
170


the CT of SEQ ID NOS: 2 and 
atgaggatgattcagaaccagttttaaaaggagtaaagttgcattacaca



3







Amino acid sequence of the
ccmtsccs
171


CT of SEQ ID NO: 4







Nucleotide sequence encoding
tgttgtatgacatcgtgctgctca
172


the CT of SEQ ID NOS: 5 and




6







Amino acid sequence of the
rvgiylciklkhtkkrqiytdiemnrlgk
173


Vector B VSV-G-CT IND




serotype of SEQ ID NOS: 7,




10, 13, and 19







Nucleotide sequence encoding
cgagttggtatttatctttgcattaaattaaagcacaccaagaaaagacagatttatacagacatag
174


the Vector B VSV-G-CT IND
agatgaaccgacttggaaag



serotype sequence of SEQ ID




NOS: 8, 9, 11, 12, 14, 15,




20, and 21







Amino acid sequence of the
ssiasfffiigliiglflvl
175


Vector B VSV-G-TM IND




serotype of SEQ ID NOS: 10




and 13







Nucleotide sequence encoding
agctctattgcctcttttttctttatcatagggttaatcattggactattcttggttctc
176


the Vector B VSV-G-TM IND




serotype sequence of SEQ ID




NOS: 11, 12, 14, and 15







Amino acid sequence of the
yeqyik
177


MPER of SEQ ID NOS: 13, 19,




and 22







Nucleotide sequence encoding
tacgaacaatacataaaa
178


the MPER of SEQ ID NOS: 14




and 15







Amino acid sequence of the
ccmtsccsclkgccscgs
179


CT of SEQ ID NO: 16







Nucleotide sequence encoding
tgttgtatgacatcgtgctgctcatgcctcaagggatgttgtagctgtggatct
180


the CT of SEQ ID NOS: 17 and




18







Amino acid sequence of the
WPWYIWLGFIAGLIAIVMVTIML
181


TM of SEQ ID NOS: 19 and 22







Amino acid sequence of the
CCMTSCCSCLKGCCSCGSCC
182


CT of SEQ ID NOS: 19 and 22







Nucleotide sequence encoding
tggccatggtacatttggctcgggtttattgctggactaattgcaatagtcatggtcactatcatgctg
183


the TM of SEQ ID NOS: 20,




21, 23, and 24







Nucleotide sequence encoding
tgttgtatgacatcgtgctgctcatgcctcaagggatgttgtagctgtggatcttgttgc
184


the CT of SEQ ID NOS: 20, 21,




23, and 24







Amino acid sequence for the
KLKHTKKRQIYTDIEMNRLGK
185


Vector B VSV-G-CT IND




serotype of SEQ ID NO: 22







Nucleotide sequence encoding
aaattaaagcacaccaagaaaagacagatttatacagacatagagatgaaccgacttggaaag
186


the Vector B VSV-G-CT IND




serotype sequence of SEQ ID




NO: 23 and 24







VSV-G-TM-CT segment of
SSIASFFFII GLIIGLFLVL RVGIHLCIKL KHTKKRQIYT DIEMNRLGK
187


Vector A







VSV-G-TM-CT segment of
SSIASFFFII GLIIGLFLVL RVGIYLCIKL KHTKKRQIYT DIEMNRLGK
188


Vector B







Leader sequence 1 
NEVAKNLNESLIDLGELGK
189





Leader sequence 2 
TLFFGDTGLSKNPIEFVEGWFSSWK
190





A segment of VSV-G
TLFFGDTGLSKNPIEFVEGWFSSWKSSIASFFFTIGLIIGLFLVLRVGIYLCIKLKHTKKRQIYTDIEM
191



NRLGK






Nucleotide sequence encoding 
tacgaacaatacataaaa
192


the MPER of SEQ ID NOS: 20




and 23







A portion of the C-terminus 
GSCCKFDEDDSEPVLKGVKLHYT
193


of SARS-CoV-2 S protein







A SNP at position 6993 
gaaaaaaaac
194












BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A-1Y show amino acid and nucleotide sequences. 1A-1X show amino acid sequences and nucleotide sequences of Constructs 1-8 which are as follows: Construct 1: wild-type (wt) SARS-CoV-2 S protein (S_WILD-TYPE) (FIGS. 1A-C); Construct 2: SARS-CoV-2 S protein with CT deletion (S_ACT31) (FIGS. 1D-F); Construct 3: SARS-CoV-2 S protein with VSVG-CT Indiana (IND) serotype (S-VSV-Gct) (FIGS. 1G-I); Construct 4: SARS-CoV-2 S protein with VSVG-TM-CT IND serotype (S_VSV-Gtmct) (FIGS. 1J-L); Construct 5: SARS-CoV-2 S protein with MPER6aa and VSVG-TM-CT IND serotype (S_VSV_Gtm-ctamper6) (FIGS. 1M-O); Construct 6: SARS-CoV-2 S protein with dCt21 (S_Δ21) (FIGS. 1P-R); Construct 7: SARS-CoV-2 S protein with dCt19 and VSVGct (S_ΔCT19_Gct) (FIGS. 1S-U); and Construct 8: SARS-CoV-2 S protein with dCt19 and VSVGct21 FIG. 1 (V-X). In FIG. 1G, the cytoplasmic tail of SARS-CoV-2 S protein is swapped with the VSV-G cytoplasmic tail. In FIG. 1J, the cytoplasmic tail and transmembrane domain of SARS-CoV-2 S protein are swapped with the cytoplasmic tail and transmembrane domain of VSV-G. FIG. 1Y shows comparison of Vector A and Vector B sequences for the VSVG-TM-CT segment. Vector A is based on GenBank Accession number EA043260. Vector B is based on GenBank MO746802 and was developed by modifying sequences in GenBank Accession number EF197793. While both Vectors A and B are of the VSV IND serotype, they have different amino acids at position 25. Vector A VSVG-TM-CT (SEQ ID NO: 187) comprises His25 (underlined) while Vector B VSVG-TM-CT (SEQ ID NO: 188) comprises Tyr25 (underlined). Vector B VSV-G-TM-CT (SEQ ID NO: 188) may also be found at amino acid position number 1218 to amino acid position number 1266 of SEQ ID NO: 10, and at amino acid position number 1212 to amino acid position number 1260 of SEQ ID NO: 13. WT, wild type; SS, signal peptide sequence; MPER, membrane-proximal external region; TM, transmembrane region; CT, cytoplasmic tail. The Kozak sequence is an additional 10 nucleotides before (i.e., 5′ of) the start codon which may help control translation of the protein. Sequences are written from N-to-C terminus or 5′ to 3′, for amino acid or nucleic acid sequences, respectively.



FIG. 2 shows alignment of 42 full-length SARS-CoV-2 S protein amino acid variant sequences. A consensus sequence is shown at the bottom of the alignment. The number on the left of each sequence is the GenBank Accession Number for the corresponding SARS-CoV-2 S protein. Sequences are written from N-to-C terminus. The nucleotide sequences for SARS-CoV-2 S protein variants shown here are incorporated by reference from Genbank. Point mutations within each variant are shown with black text and white highlight. Point mutations in the consensus sequence are shown in lowercase underlined. QHO60594 is the source sequence for SARS-CoV-2 S protein and is identified with an underline.



FIG. 3 is a phylogenic tree showing the country of origin for each corresponding SARS-CoV-2 in the alignment in FIG. 2. QHO60594 is the source sequence for SARS-CoV-2 S protein and is identified with an underline.



FIG. 4 illustrates rational design of a VSVΔG-SARS-CoV-2 recombinant viral particle, wherein the VSV glycoprotein (VSV-G) is replaced by SARS-CoV-2 S protein or variants thereof. S adaptive refers to SARS-CoV-2 S proteins that comprise adaptive mutations after passaging in the cells.



FIG. 5 shows SARS-CoV-2 S proteins inserted in the VSV genomic clone to produce Constructs 1-8 of FIG. 1. In Construct 1 SEQ ID NO: 1, the sequences of Leader Sequence 1 SEQ ID NO: 189, MPER SEQ ID NO: 163, TM SEQ ID NO: 164, and CT SEQ ID NO: 165 span from amino acid position nos. 1187-1273 of SEQ ID NO: 1. In Construct 2 SEQ ID NO: 4, the sequences of Leader Sequence 1 SEQ ID NO: 189, MPER SEQ ID NO: 163, TM SEQ ID NO: 164, and CT SEQ ID NO: 171 span from amino acid position nos. 1187-1242 of SEQ ID NO: 4. In Construct 3 SEQ ID NO: 7, the sequences of Leader Sequence 1 SEQ ID NO: 189, MPER SEQ ID NO: 163, TM SEQ ID NO: 164, and SEQ ID NO: 173 span from amino acid position nos. 1187-1263 of SEQ ID NO: 7. In Construct 4 SEQ ID NO: 10, the sequences of Leader Sequence 1 SEQ ID NO: 189, MPER SEQ ID NO: 163, SEQ ID NO: 175, and SEQ ID NO: 173 span from amino acid position nos. 1187-1266 of SEQ ID NO: 10. In Construct 5 SEQ ID NO: 13, the sequences of Leader Sequence 1 SEQ ID NO: 189, MPER SEQ ID NO: 177, SEQ ID NO: 175, and SEQ ID NO: 173 span from amino acid position nos. 1187-1260 of SEQ ID NO: 13. In Construct 6 SEQ ID NO: 16, the sequences of Leader Sequence 1 SEQ ID NO: 189, MPER SEQ ID NO: 163, TM SEQ ID NO: 164, and CT SEQ ID NO: 179 span from amino acid position nos. 1187-1252 of SEQ ID NO: 16. In Construct 7 SEQ ID NO: 19, the sequences of Leader Sequence 1 SEQ ID NO: 189, MPER SEQ ID NO: 177, TM SEQ ID NO: 181, CT SEQ ID NO: 182, and SEQ ID NO: 173 span from amino acid position nos. 1187-1283 of SEQ ID NO: 19. In Construct 8 SEQ ID NO: 22, the sequences of Leader Sequence 1 SEQ ID NO: 189, MPER SEQ ID NO: 177, TM SEQ ID NO: 181, CT SEQ ID NO: 182, and SEQ ID NO: 185 span from amino acid position nos. 1187-1275 of SEQ ID NO: 22. A segment of VSV-G with the sequence set forth in SEQ ID NO: 191 is also shown. In this VSV-G segment, the sequences of Leader Sequence 1, SEQ ID NO: 175, and SEQ ID NO: 173 span from amino acid position nos. 1-74 of SEQ ID NO: 191. Also in this VSV-G segment, the sequence of SEQ ID NO: 185 spans from amino acid position nos. 54-74 of SEQ ID NO: 191. SS, signal sequence; MPER, membrane-proximal external region; TM, transmembrane region; CT, cytoplasmic tail. Sequences in bold indicate inserted sequences that originate from VSV-G. S1 and S2 domains are shown, as well as the Furin cleavage site. Leader Sequence 1 is SEQ ID NO: 189. Leader Sequence 2 is SEQ ID NO: 190.



FIG. 6 illustrates recombinant VSVΔG-SARS-CoV-2 rescue from plasmid DNA. To initiate recovery of infectious virus from plasmid DNA, a blend of seven plasmids (contained in the box at the top of the figure) is electroporated into Vero cells that have been qualified for vaccine production. Witko et al., An efficient helper-virus free method for rescue of recombinant paramyxoviruses and rhadoviruses from a cell line suitable for vaccine development, J. Virol. Methods (2006); doi:10.1016/j.jviromet.2006.02.006. One of the plasmids is the VSVΔG-SARS-CoV-2 genomic clone that can be transcribed by the phage T7 RNA polymerase to generate viral RNA genomes. The remaining 6 supporting plasmids are controlled by the human cytomegalovirus (hCMV) promoter that is recognized by cellular RNA polymerase II in the nucleus. These supporting plasmids are designed to express proteins needed to initiate the virus life cycle, including the T7 bacteriophage RNA polymerase and the five VSV structural proteins (VSV-N, nucleocapsid protein; VSV-P, phosphoprotein; VSV-M, matrix protein; VSV-G, glycoprotein; and VSV-L, or large protein, which is the catalytic subunit of the viral RNA-dependent RNA polymerase; Dietzgen et al., The family Rhabdoviridae: mono- and bipartite negative-sense RNA viruses with diverse genome organization and common evolutionary origins, Virus Res. (2017); doi:10.1016/j.viruses.2016.10.010). Following electroporation, the cells are cultured for approximately 72 hours during which time the plasmids are transcribed, viral proteins are synthesized and viral genomes are encapsidated with VSV-N to form a nucleocapsid. The nucleocapsid can then be transcribed and replicated by the viral RNA polymerase allowing initiation of virus replication and assembly and release of infectious progeny virions that can be used later to infect fresh cultures of cells.



FIG. 7 shows virus infectivity titers of Construct 1 (rescued using a plasmid encoding wild-type SARS-CoV-2 S protein) improved over passages. Construct 1 (wild-type SARS-CoV-2 S protein) is adapted to the Vero cell culture by serial passages. Titers improved from 4.00E+05 pfu/mL to 2.00E+07 pfu/mL.



FIGS. 8A-B show Vero cells infected by Construct 1 (wild-type SARS-CoV-2 S protein with adaptive mutations evolving over multiple passages): passage 4 (P4) 52 hours post infection (FIG. 8A) and passage 12 (P12) 24 hours post infection (FIG. 8B). After serial passage, Construct 1 (wild-type SARS-CoV-2 S protein) was adapted to the Vero cell culture. At P12, more lytic cytopathic effect replaced the earlier syncytia at P4. The infection progressed more rapidly at the higher passages.



FIG. 9 shows SARS-CoV 2 S protein detected in the infected Vero cell lysate by Western blot. Vero cells were infected by clone isolates A*1 to A*12. Clone isolates with reduction of cleaved S protein had mutations at the Furin cleavage site, for example, R682K, R685G, S813R or S813F. Full length SARS-CoV-2 S protein (S) and S2 subunit (S2) are shown.



FIGS. 10A-C shows SARS-CoV S protein detected in the infected Vero cell lysate by Western blot. Monomeric (S) and dimeric (dimer) full-length SARS-CoV-2 S protein, as well as the S1 (S1) and S2 (S2) subunits are indicated. Vero cells were infected with clone isolates. In FIG. 10A, clone isolates SF1 and SF2 showed reduction of S protein post-translational processing, indicative of a mutations, e.g., R683G, at the Furin cleavage site. FIGS. 10A-B show western blots for isolates cloned in serum-free medium. In FIG. 10B, the arrow marks the control lanes. FIG. 10C shows a western blot for isolates cloned in serum-containing medium.



FIG. 11 shows titers of is rVSVΔG-SARS-CoV-2 clones SC1, SC3, SC7, and MB2 that were amplified in serum-free medium (VP-SFM). Titers of each clone were measured at indicating time points.



FIG. 12 shows titers of two vaccine candidates, rVSVΔG-SARS-CoV-2 clones MB1 and MB2, in the cell culture medium after infection. These two vaccine candidates have been further tested in non-human primates (NHP) and small animal models. See Example 15 below. rVSVΔG-SARS-CoV-2 clone MB1 reached to higher titers than rVSVΔG-SARS-CoV-2 clone MB2 at all selected time points. For virus production purpose, the harvest time should occur from 54 to 65 hours post infection rVSVΔG-SARS-CoV-2 clone MB1 and 72 h post infection for rVSVΔG-SARS-CoV-2 clone MB2.



FIGS. 13A-B shows cytopathic effect on Vero cells at the time of harvest of rVSVΔG-SARS-CoV-2 clone MB1(FIG. 13A) and rVSVΔG-SARS-CoV-2 clone MB2 (FIG. 13B).



FIGS. 14A-B shows Vero cells infected by Construct 6. FIG. 14A shows passage 4 (P4) at 43 hours post infection. FIG. 14B shows passage 11 (P11) at 48 hours post infection. Infectious titer was improved when the virus was adapted to the Vero cell culture by serial passages. The titers were 6.50E+05 pfu/mL at P4 and 2.70E+07 pfu/mL at P11, respectively. Mutations at E484D, H655Y in SARS-CoV-2 S protein, E213K in VSV-M and V411M in VSV-L were identified in the higher passage P11.



FIG. 15 shows the domains, mutations, and cleavage sites of SARS-CoV-2 S protein. MB1 and MB2 variants of the rVSVΔG-SARS-CoV-2 S protein are also shown.



FIG. 16 shows post-vaccination anti-SARS-CoV-2 spike protein-specific immunoglobulin (IgG) enzyme-linked immunosorbent assay (ELISA) titers through day 26 from Study HAM-CoV-2-001 Part A. The immunogenicity of rVSVΔG-SARS-CoV-2 clone MB1 and its ability to protect against SARS-CoV-2 challenge was studied in Golden Syrian hamster model of SARS-CoV-2 infection. Hamsters were administered rVSVΔG-SARS-CoV-2 clone MB1 via intramuscular (IM) or oral mucosal (OM) routes, or received a vaccine consisting of the SARS-CoV-2 S protein adjuvanted with aluminum phosphate (AdjuPhos) as described in Tables 10 and 11. We evaluated the ability of rVSVΔG-SARS-CoV-2 clone MB1 to protect against SARS-CoV-2-induced body weight loss. Serum samples were collected on Days 7, 17, and 26 post-immunization, and on Day 14 post-challenge. Serum samples were analyzed for SARS-CoV-2-S protein-specific IgG titers on Days 7, 14, and 26 in an ELISA. rVSVΔG-SARS-CoV-2 clone MB1 did not demonstrate anti-S protein-specific IgG titers after OM administration at Day 7, 14, or 26 post-vaccination, with titers similar to non-vaccinated animals.



FIGS. 17A-B show post-vaccination SARS-CoV-2 pseudoneutralization titers (PRNT) assay from Study HAM-CoV-2-001 Part A. The immunogenicity of rVSVΔG-SARS-CoV-2 clone MB1 and its ability to protect against SARS-CoV-2 challenge was studied in Golden Syrian hamster model of SARS-CoV-2 infection. Hamsters were administered rVSVΔG-SARS-CoV-2 clone MB1 via intramuscular (IM) or oral mucosal (OM) routes, or received a vaccine consisting of the SARS-CoV-2 S protein adjuvanted with aluminum phosphate (AdjuPhos) as described in Tables 10 and 11. In FIG. 17A, serum samples were collected on Days 7, 17, and 26 post-immunization and analyzed for SARS-CoV-2-specific PRNT titers. rVSVΔG-SARS-CoV-2 clone MB1 initiated immunogenicity after IM administration similarly to vaccination with S protein as shown by Day 7 and Day 26 post-vaccination SARS-CoV-2 neutralization assay titers. rVSVΔG-SARS-CoV-2 clone MB1 did not demonstrate SARS-CoV-2 neutralizing titers after OM administration at Day 7 or Day 26 post-vaccination. These titers were similar to titers in unvaccinated animals, with the exception of one animal with a high titer on Day 26. The dotted line indicates the limit of detection. FIG. 17B shows day 14 PRNT neutralizing titers from assay conducted with SARS-CoV-2. Serum samples were collected on Day 14 post-immunization and analyzed for SARS-CoV-2-specific PRNT titers. rVSVΔG-SARS-CoV-2 clone MB1 initiated immunogenicity after IM administration similarly to vaccination with S protein as shown by Day 14 post-vaccination SARS-CoV-2 neutralization assay titers, as determined by BIOQUAL. rVSVΔG-SARS-CoV-2 clone MB1 did not demonstrate SARS-CoV-2 neutralizing titers after OM administration at Day 14 post-vaccination, with titers similar to non-vaccinated animals. The dotted line indicates the limit of detection.



FIG. 18 shows post-vaccination SARS-CoV-2 titers (a PRNT assay) on days 7 and 14 from Study HAM-CoV-2-001 Part A.



FIGS. 19A-C show that rVSVΔG-SARS-CoV-2 clones MB1 and MB2 protect against COVID-19 body weight loss in the Golden Syrian hamster model of SARS-CoV-2 infection (Study Part A). The immunogenicity of rVSVΔG-SARS-CoV-2 clones MB1 and MB2 and its ability to protect against SARS-CoV-2 challenge was studied in Golden Syrian hamster model of SARS-CoV-2 infection. Hamsters were administered rVSVΔG-SARS-CoV-2 clone MB1 or MB2 via intramuscular (IM) or oral mucosal (OM) routes, or received a vaccine consisting of the SARS-CoV-2 S protein adjuvanted with aluminum phosphate (Spike Protein/AdjuPhos) as described in Tables 10 and 11. We evaluated the ability of rVSVΔG-SARS-CoV-2 clones MB1 and MB2 to protect against SARS-CoV-2-induced body weight loss. FIG. 19A shows body weight change over the course of 14 days due to MB1 administration via IM or OM administration. FIGS. 19B-C show body weight change over the course of 5 days and 14 days, respectively, due to rVSVΔG-SARS-CoV-2 clone MB1 or 2 MB2 via IM or OM administration. Both rVSVΔG-SARS-CoV-2 clones MB1 and MB2 protected animals against SARS-CoV-2-induced body weight loss after IM administration similarly to protection by vaccination with adjuvanted SARS-CoV-2 S protein (Spike Protein/AdjuPhos) in the Golden Syrian hamster model. Both rVSVΔG-SARS-CoV-2 clones MB1 and MB2 did not protect animals from SARS-CoV-2-induced body weight loss, similarly to unvaccinated animals.



FIG. 20 shows a sequence alignment comparing the nucleotide sequence of the S gene in rVSVΔG-SARS-CoV-2 clone MB1 (SEQ ID NO: 150; the subject sequence (shown as “sbjct”) in the alignment) with the codon-optimized, wild-type S gene in VSVΔG-SARS-CoV-2 (SEQ ID NO: 159; the query sequence (shown as “query) in the alignment).



FIGS. 21A-G show amino acid and nucleotide sequences for VSVΔG-SARS-CoV-2 clone MB2. FIG. 21A shows the order of genes in the genome of VSVΔG-SARS-CoV-2 clone MB2 from 5′ to 3′: VSV-N, VSV-P, VSV-M, codon-optimized SARS-CoV-2 S gene with H655Y and G1251* mutations, and VSV-L. Also shown are the XhoI and NheI sites. FIG. 21B shows the amino acid sequence for VSV-N(SEQ ID NO: 155). FIG. 21C shows the amino acid sequence for VSV-P (SEQ ID NO: 156). FIG. 21D shows the amino acid sequence for VSV-M (SEQ ID NO: 157). FIG. 21E shows the amino acid sequence for SARS-CoV-2 S protein with H665Y and G1251* mutations (SEQ ID NO: 158). The mutations are underlined. FIG. 21F shows the amino acid sequence for VSV-L with an I1343I silent mutation (SEQ ID NO: 152). The mutation is underlined. FIG. 21G shows the nucleotide sequence for the genome of VSVΔG-SARS-CoV-2 clone MB2, which has the nucleic acid sequence set forth in SEQ ID NO: 154. VSVΔG-SARS-CoV-2 clone MB2 encodes the proteins represented by the amino acid sequences in FIGS. 21B-F. The coding regions for VSV-N (nucleotides 64-1332), VSV-P (nucleotides 1396-2193), VSV-M (nucleotides 2250-2939), SARS-CoV-2 S protein (nucleotides 3102-6854; nucleotides that code for the H665Y and G1251* mutations are shown with black highlights and white text), and VSV-L (nucleotides 7011-13341; nucleotides that code for the I1343I silent mutation are shown with black highlights and white text) are underlined.



FIGS. 22A-F show amino acid and nucleotide sequences for Vector A, which is a VSV vector without the VSV-G gene. FIG. 22A shows the order of genes in Vector A from 5′ to 3′: VSV-N, VSV-P, VSV-M, and VSV-L. Also shown are the XhoI and NheI sites. FIG. 22B shows the amino acid sequence for Vector A VSV-N(SEQ ID NO: 155). FIG. 22C shows the amino acid sequence for Vector A VSV-P (SEQ ID NO: 156). FIG. 22D shows the amino acid sequence for Vector A VSV-M (SEQ ID NO: 157). FIG. 22E shows the amino acid sequence for Vector A VSV-L (SEQ ID NO: 152). FIG. 22F shows the nucleotide sequence for the genome of Vector A (without VSV-G gene) (SEQ ID NO: 160). The coding regions for VSV-N (nucleotides 64-1332), VSV-P (nucleotides 1396-2193), VSV-M (nucleotides 2250-2939), and VSV-L (nucleotides 3178-9507) are underlined. Vector A encodes the proteins represented by the amino acid sequences in FIGS. 22B-E.



FIGS. 23A-F show amino acid and nucleotide sequences for Vector B, which is a VSV vector without the VSV-G gene. FIG. 23A shows the order of genes in Vector B from 5′ to 3′: VSV-N, VSV-P, VSV-M, and VSV-L. Also shown are the BstBI and PacI sites. FIG. 23B shows the amino acid sequence for Vector B VSV-N(SEQ ID NO: 155). FIG. 23C shows the amino acid sequence for Vector B VSV-P (SEQ ID NO: 156). FIG. 23D shows the amino acid sequence for Vector B VSV-M (SEQ ID NO: 157). FIG. 23E shows the amino acid sequence for Vector B VSV-L (SEQ ID NO: 152). FIG. 23F shows the nucleotide sequence for the genome of Vector B (without VSV-G gene) (SEQ ID NO: 161). The coding regions for VSV-N (nucleotides 64-1332), VSV-P (nucleotides 1396-2193), VSV-M (nucleotides 2253-2942), and VSV-L (nucleotides 3062-9391) are underlined. Vector B encodes the proteins represented by the amino acid sequences in FIGS. 23B-E.



FIGS. 24A-G show amino acid and nucleotide sequences for VSVΔG-SARS-CoV-2 clone MB1. FIG. 24A shows the order of genes in VSVΔG-SARS-CoV-2 clone MB1 from 5′ to 3′: VSV-N (nucleotides 64 to 1332), VSV-P (nucleotides 1396 to 2193), VSV-M (nucleotides 2250 to 2939), codon-optimized SARS-CoV-2 S protein-optimized gene (nucleotides 3102 to 6854), and VSV-L (nucleotides 7012 to 13341). FIG. 24B shows the amino acid sequence for VSV-N(SEQ ID NO: 155). FIG. 24C shows the amino acid sequence for VSV-P (SEQ ID NO: 156). FIG. 24D shows the amino acid sequence for VSV-M (SEQ ID NO: 151) which comprises an amino acid Y61S mutation. FIG. 24E shows the amino acid sequence of SARS-CoV-2 S protein (SEQ ID NO: 153) which comprises R683G, S813F, and G1251* mutations. The G1251* mutation results in early termination of the SARS-CoV-2 S protein with the deletion of the last 23 amino acids. FIG. 24F shows the amino acid sequence for VSV-L (SEQ ID NO: 152). FIG. 24G shows the nucleotide sequence for VSVΔG-SARS-CoV-2 clone MB1 (SEQ ID NO: 150). VSVΔG-SARS-CoV-2 clone MB1 encodes the proteins represented by the amino acid sequences in FIGS. 24B-F (SEQ ID NOS: 155, 156, 151, 153, and 152, respectively). The following nucleotide and amino changes were observed relative to the wild-type sequence and are shown with black highlights and white text: (a) VSV-M—nucleotide A2431C; amino acid Y61S; (b) non-coding sequence—nucleotide C3058T; (c) SARS-CoV-2 S protein-nucleotide A5148G; amino acid R683G; (d) SARS-CoV-2 S protein—nucleotide C5539T; amino acid S813F; (e) SARS-CoV-2 S protein—nucleotide G6852T; amino acid G1251* (which results in early termination of the SARS-CoV-2 S protein with the deletion of the last 23 amino acids).



FIG. 25 shows NT50 (inverse of the dilution of the serum at which 50% of input virus is prevented from infecting cells) and VC average plaque counts for PRNT assays of clone isolates. VC=virus control.



FIGS. 26A-B show flow virometry data for HeLa cells expressing ACE2 (hAVE2 cells) that were infected with recombinant virus of clone isolates. FIG. 26A shows data for clones isolated in serum-containing medium. FIG. 26B shows data for clones isolated in serum-free medium. Anti-VSV-N antibody staining is shown in the y-axis. Anti-SARS-CoV-2 S protein neutralizing antibody staining (nAB) is shown in the x-axis.



FIG. 27 shows binding of recombinant virus of clones SC1, SC3, SC7, and MB2 to anti-hACE2 Ig antibody. Anti-VSV-N antibody staining is shown in the y-axis. Anti-huACE2 Ig antibody staining is shown in the x-axis.



FIG. 28 shows PRNT neutralizing titers at Days 14 and 28 after vaccination.



FIGS. 29A-D show viral loads as determined by TCID50 in the lung and the nose at Day 4 after challenge. FIG. 29A shows IM titration lung titers. FIG. 29B shows IM titration nose titers. FIG. 29C shows oral dosing lung titers. FIG. 29D shows oral dosing nose titers. IM, intramuscular; OM, oral mucosal; OMx2, two oral mucosal doses; PO, oral; IN+OM, intranasal+oral mucosal.



FIG. 30 shows the SARS-CoV-2 S protein IgG levels in the serum of rhesus macaques vaccinated with VSVΔG-SARS-CoV-2 strain Fp11. IN, intranasal; OM, oral mucosal; IN+OM, intranasal+oral mucosal.





DETAILED DESCRIPTION

The present disclosure relates to vaccine compositions and uses thereof for preventing COVID-19 caused by the pandemic coronavirus Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2). The present disclosure describes vaccine compositions for delivering a SARS-CoV-2 spike (S) protein or its immunogenic variant using replication-competent vesicular stomatitis virus (VSV) chimeric virus technology (VSVΔG). This technology allows immunization with the SARS-CoV-2 S protein or its immunogenic variant presented on the surface of infected cells and recombinant VSV particles in the context of immune stimulation by a viral infection. Different VSVΔG chimeras expressing the SARS-CoV-2 S protein or its immunogenic variant may be generated using different recombinant VSV backbones, for example, the one used to generate the FDA and EMA-licensed Merck Zaire Ebola virus (ZEBOV) vaccine (ERVEBO®). VSV (Indiana serotype) genetic backgrounds can be used but also other vesiculoviruses can be considered for use such as VSV (New Jersey serotype) or related vesiculoviruses such as Maraba virus, Carajas virus, Alagoas virus, Cocal virus, or Isfahan virus. Additionally, genetically modified variants of VSV (Indiana serotype) or the other vesculovirsues can be used. Moreover, the present disclosure describes adaptive mutants of the recombinant replicable VSV particle having better growth in a cell culture. These mutants have one or more mutations in, for example, the exposed loop (a solvent-exposed loop that comprises the S1/S2 cleavage site), S1/S2 cleavage site, S2′ (previously referenced as S″) cleavage site and/or cytoplasmic tail of the SARS-CoV-2 S protein. The vaccine compositions may be administered to subjects in one or two doses to induce protective immunity within days, for example, as little as 10 days. Vaccination up to 1-3 days after exposure to SARS-CoV-2 may be effective for preventing COVID-19. Upon vaccination of subjects, virus-neutralizing antibodies (nAbs) against the SARS-CoV-2 S protein may be induced in the subjects and harvested to make a pharmaceutical composition for preventing or treating COVID-19.


VSV typically grows very rapidly and robustly in cell culture. Generation of a chimeric virus by replacing VSV glycoprotein (VSV-G) with a heterologous glycoprotein like the SARS-CoV-2 S protein can substantially diminish the replicative capacity of the recombinant virus. In addition to using rational design of SARS-CoV-2 S protein variants to achieve VSVΔG-SARS-CoV-2 chimera for replication needed to support vaccine manufacturing as well as effective immunization (FIG. 4), the present disclosure describes that the evolutionary potential of VSV can also be used to identify novel SARS-CoV-2 S protein modifications that enhance virus growth.


Viruses like VSV with RNA genomes are known to have the capacity to mutate and evolve when faced with new circumstances that are not optimal virus growth (Novella IS. 2003. Contributions of vesicular stomatitis virus to the understanding of RNA virus evolution. Curr Opin Microbiol 6:399-405). These circumstances can be environmental factors like changes in temperature or alternative host cells, or they can be caused by deleterious modifications to the virus, such as replacement of the VSV-G with the glycoprotein from a heterologous virus. As VSV replicates under conditions in which replication is impaired, it will evolve mutations that contribute to restoration of its replicative capacity. Thus, this ability of VSV to generate favorable adaptive mutations can be used to identify amino acid substitutions or other types of modifications in the SARS-CoV-2 spike that enable improved replication of a VSVΔG-SARS-CoV-2 chimera. In the laboratory, evolution of favorable mutations can be accomplished by serial passage of the virus in cultured cells while monitoring for emergence of mutant strains that grows more robustly. Adaptive mutations responsible for improved growth can then be identified by determining the nucleotide sequence of the new virus strain.


I. Definitions

The term “antibody” includes intact molecules as well as any fragments thereof that are capable of specifically binding the epitope of the antibody, such as, but not limited to, Fab, F(ab′)2, Fv and scFv which are capable of binding the epitope determinant. These antibody fragments retain some ability to selectively bind with its antigen or receptor and include, for example: (i) Fab, the fragment which contains a monovalent antigen-binding fragment of an antibody molecule can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain; (ii) Fab′, the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the 20 heavy chain; two Fab′ fragments are obtained per antibody molecule; (iii) F(ab′)2, the fragment of the antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction; F(ab′)2 is a dimer of two Fab′ fragments held together by two disulfide bonds; (iv) scFv, including a genetically engineered fragment containing the variable region of a heavy and a light chain as a fused single chain molecule.


As used herein, the terms “antigen” or “immunogen” are used interchangeably to refer to a substance, typically a protein, which is capable of inducing an immune response in a subject. The term also refers to proteins that are immunologically active in the sense that once administered to a subject (either directly or by administering to the subject a nucleotide sequence or vector that encodes the protein) is able to evoke an immune response of the humoral and/or cellular type directed against that protein.


The term “derived from” used herein refers to an origin or source, and may include naturally occurring, recombinant, unpurified or purified molecules. The molecules of the present disclosure may be derived from viral or non-viral molecules. A protein or polypeptide derived from an original protein or polypeptide may comprise the original protein or polypeptide, in part or in whole, and may be a fragment or variant of the original protein or polypeptide. A nucleic acid molecule or polynucleotide derived from an original nucleic acid molecule or polynucleotide may comprise the original nucleic acid molecule or polynucleotide, in part or in whole, and may be a fragment or variant of the original nucleic acid molecule or polynucleotide.


The term “foreign gene” used herein refers to a gene of an origin or source different from that of a vector, into which the foreign gene is inserted. For example, the vector may be derived from a vesicular stomatitis virus (VSV) while the foreign gene may be derived from a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2).


The term “fragment” of a protein as used herein refers to a polypeptide having an amino acid sequence that is the same as a part, but not all, of the amino acid sequence of the protein. A fragment may be a functional fragment of a protein that retains the same function as the protein.


The terms “nucleic acid molecule” and “polynucleotide” are used interchangeably herein to refer to a polymer of nucleic acids of any length and having a “nucleic acid sequence” or “nucleotide sequence.” As used herein the terms “nucleotide sequence” and “nucleic acid sequence” refer to a deoxyribonucleic acid (DNA) sequence, a ribonucleic acid (RNA) sequence or a combination thereof, including, without limitation, messenger RNA (mRNA), DNA/RNA hybrids, or synthetic nucleic acids. The “nucleic acid molecule” and “polynucleotide” can be single-stranded, or partially or completely double-stranded (duplex), which can be homoduplex or heteroduplex.


For the proteins of the present disclosure to be expressed, the protein coding sequence should be “operably linked” to regulatory or nucleic acid control sequences that direct transcription and translation of the protein. As used herein, a coding sequence and a nucleic acid control sequence or promoter are said to be “operably linked” when they are covalently linked in such a way as to place the expression or transcription and/or translation of the coding sequence under the influence or control of the nucleic acid control sequence. The “nucleic acid control sequence” can be any nucleic acid element, such as, but not limited to promoters, enhancers, IRES, introns, and other elements described herein that direct the expression of a nucleic acid sequence or coding sequence that is operably linked thereto. The expression of the transgenes of the present disclosure can be under the control of a constitutive promoter or of an inducible promoter, which initiates transcription when exposed to some particular external stimulus, such as, without limitation, antibiotics such as tetracycline, hormones such as ecdysone, or heavy metals. The promoter can also be specific to a particular cell-type, tissue or organ. Many suitable promoters and enhancers are known in the art, and any such suitable promoter or enhancer may be used for expression of the transgenes of the disclosure. For example, suitable promoters and/or enhancers can be selected from the Eukaryotic Promoter Database (EPDB).


The term “percent homology” refers to the amount of sequence identity between two sequences. If a variant is a fragment of the wild-type SARS-CoV-2 S protein, % homology is measured over the length of the fragment instead of the length of the wild-type SARS-CoV-2 S protein. In some embodiments, the full-length variant or fragment variant comprises at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% homology with the wild-type SARS-CoV-2 S protein. In some embodiments, the full-length variant or fragment variant comprises 100% homology with the wild-type SARS-CoV-2 S protein (SEQ ID NO: 1).


The terms “protein”, “peptide”, “polypeptide”, and “amino acid sequence” are used interchangeably herein to refer to polymers of amino acid residues of any length. The polymer may be linear or branched, it may comprise modified amino acids or amino acid analogs, and it may be interrupted by chemical moieties other than amino acids. The terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling or bioactive component.


As used herein the term “transgene” may be used to refer to “recombinant” nucleotide sequences that may be derived from any of the nucleotide sequences encoding the proteins of the present disclosure. The term “recombinant” means a nucleotide sequence that has been manipulated “by man” and which does not occur in nature, or is linked to another nucleotide sequence or found in a different arrangement in nature. It is understood that manipulated “by man” means manipulated by some artificial means, including by use of machines, codon optimization, restriction enzymes, etc.


For example, in one embodiment the nucleotide sequences may be mutated such that the activity of the encoded proteins in vivo is abrogated.


As regards codon optimization, the nucleic acid molecules of the disclosure have a nucleotide sequence that encodes the antigens of the disclosure and can be designed to employ codons that are used in the genes of the subject in which the antigen is to be produced. Many viruses, including coronaviruses, use a large number of rare codons and, by altering these codons to correspond to codons commonly used in the desired subject, enhanced expression of the antigens can be achieved. In some embodiments, the codons used are “humanized” codons, i.e., the codons are those that appear frequently in highly expressed human genes (Andre et al., J. Virol. 72:1497-1503, 1998) instead of those codons that are frequently used by a coronavirus. Such codon usage provides for efficient expression of the transgenic coronavirus proteins in human cells. Any suitable method of codon optimization may be used. Such methods, and the selection of such methods, are well known to those of skill in the art. In addition, there are several companies that will optimize codons of sequences, such as Geneart (geneart.com). Thus, the nucleotide sequences of the disclosure can readily be codon optimized.


The disclosure further provides nucleotide sequences encoding functionally and/or antigenically equivalent variants and derivatives of the antigens of the disclosure and functionally equivalent fragments thereof, for example, the SARS-CoV-2 S protein or an immunogenic variant thereof. These functionally equivalent variants, derivatives, and fragments display the ability to retain antigenic activity. For instance, changes in a DNA sequence that do not change the encoded amino acid sequence, as well as those that result in conservative substitutions of amino acid residues, one or more than one amino acid deletions or additions, and substitution of amino acid residues by amino acid analogs are those which will not significantly affect properties of the encoded polypeptide. In one embodiment, the variants have at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, 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% or at least 99% homology or identity to the antigen, epitope, immunogen, peptide or polypeptide of interest.


For the purposes of the present disclosure, sequence identity or homology is determined by comparing the sequences when aligned so as to maximize overlap and identity while minimizing sequence gaps. In particular, sequence identity may be determined using any of a number of mathematical algorithms. A nonlimiting example of a mathematical algorithm used for comparison of two sequences is the algorithm of Karlin & Altschul, Proc. Natl. Acad. Sci. USA 1990; 87: 2264-2268, modified as in Karlin & Altschul, Proc. Natl. Acad. Sci. USA 1993; 90: 5873-5877.


Another example of a mathematical algorithm used for comparison of sequences is the algorithm of Myers & Miller, CABIOS 1988; 4: 11-17. Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. Yet another useful algorithm for identifying regions of local sequence similarity and alignment is the FASTA algorithm as described in Pearson & Lipman, Proc. Natl. Acad. Sci. USA 1988; 85: 2444-2448.


Advantageous for use according to the present disclosure is the WU-BLAST (Washington University BLAST) version 2.0 software. WU-BLAST version 2.0 executable programs for several UNIX platforms can be downloaded. This program is based on WU-BLAST version 1.4, which in turn is based on the public domain NCBI-BLAST version 1.4 (Altschul & Gish, 1996, Local alignment statistics, Doolittle ed., Methods in Enzymology 266: 460-480; Altschul et al., Journal of Molecular Biology 1990; 215: 403-410; Gish & States, 1993; Nature Genetics 3: 266-272; Karlin & Altschul, 1993; Proc. Natl. Acad. Sci. USA 90: 5873-5877; all of which are incorporated by reference herein).


The various recombinant nucleotide sequences and proteins of the disclosure are made using standard recombinant DNA and cloning techniques. Such techniques are well known to those of skill in the art. See for example, “Molecular Cloning: A Laboratory Manual”, second edition (Sambrook et al. 1989).


The term “variant” of a protein refers to a polypeptide having an amino acid sequence that is the same as that of the protein except having at least one amino acid modified, for example, deleted, inserted, or replaced, so long as the variant functions like the protein (even if the amount or characteristics of the function differ from the protein). The amino acid replacement may be a conservative amino acid substitution. The amino acid replacement may be a non-essential amino acid residue in the protein. A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains are known in the art. For example, amino acids are generally divided into four families:

    • (1) acidic—aspartatic acid and glutamic acid;
    • (2) basic—lysine, arginine, histidine;
    • (3) non-polar—alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan (with valine, isoleucine, and leucine seen as particularly related); and
    • (4) uncharged polar—glycine, asparagine, glutamine, cysteine, serine threonine, tyrosine (with asparagine and glutamine seen as particularly related).


      Conservative amino acid substitutions may be made within these families.


Phenylalanine, tryptophan, and tyrosine are sometimes classified as aromatic amino acids with hydrophobic side chains and may be substituted for each other. Glycine may also be substituted for alanine and vice versa. Serine, threonine, and methionine may also be substituted for each other.


For example, it is reasonably predictable that an isolated replacement of leucine with isoleucine or valine, or vice versa; an aspartate with a glutamate or vice versa; a threonine with a serine or vice versa; or a similar conservative replacement of an amino acid with a structurally related amino acid, will not have a major effect on the biological activity. Proteins having substantially the same amino acid sequence as the sequences illustrated and described but possessing minor amino acid substitutions that do not substantially affect the immunogenicity of the protein are, therefore, within the scope of the disclosure. Other immunogenic variants are also encompassed by this disclosure.


The terms “virus” and “viral particle” are used herein interchangeably and refer to a replicable virus having its genome enclosed by a protein coat. The term “replication capacity” used herein refers to how quickly a virus or viral particle reproduces (i.e., replicates). Where the virus or viral particle is recombinant and carries a foreign gene and/or foreign protein, the replication capacity of the virus or viral particle may be reduced.


II. Vectors and Viral Particles

A recombinant VSV vector or recombinant VSV viral particle comprises at least a portion of the VSV genome comprising the N, P, M, and L genes and a nucleic acid sequence encoding the SARS-CoV-2 S protein or an immunogenic variant thereof. This allows generation of a recombinant VSV particle comprising this vector and displaying the S protein or an immunogenic variant thereof on the surface of the VSV particle, specifically the S protein or immunogenic variant thereof displayed on the surface of the VSV particle is the S protein encoded by the vector or by the nucleic acids of the recombinant VSV particle.


In certain embodiments the VSV particle is replicable.


In some embodiments the vectors or particles encode an immunogenic recombinant protein comprising a SARS-CoV-2 S protein or an immunogenic variant thereof. In some embodiments, the vectors or particles encode an immunogenic recombinant protein encoding both a SARS-CoV-2 S protein or an immunogenic variant thereof and at least a fragment of the VSV glycoprotein (G).


These vectors or particles, or the immunogenic proteins encoded by them, may be used as vaccines.


Any vector that allows expression of the proteins of the present disclosure, namely, the SARS-CoV-2 S protein or an immunogenic variant thereof, may be used in accordance with the present disclosure. The vectors may contain a suitable gene regulatory region, such as a promoter or enhancer, such that the proteins and viral particles of the present disclosure can be produced.


When the aim, for example, is to express the proteins of the disclosure in vitro, or in cultured cells, or in any prokaryotic or eukaryotic system for the purpose of producing the protein(s), then any suitable vector can be used depending on the application. For example, plasmids, viral vectors, bacterial vectors, protozoal vectors, insect vectors, baculovirus expression vectors, yeast vectors, mammalian cell vectors, and the like, can be used. Suitable vectors can be selected by the skilled artisan taking into consideration the characteristics of the vector and the requirements for expressing the proteins under the identified circumstances.


In certain embodiments, the antigens and/or antibodies of the present disclosure may be used in vitro (such as using cell-free expression systems) and/or in cultured cells grown in vitro in order to produce the encoded SARS-CoV-2-antigens and/or antibodies which may then be used for various applications such as in the production of proteinaceous vaccines. For such applications, any vector that allows expression of the antigens and/or antibodies in vitro and/or in cultured cells may be used.


When the aim is to express the proteins or viral particles of the disclosure in vivo in a subject, for example, in order to generate an immune response against a SARS-CoV-2 antigen and/or generate protective immunity against SARS-CoV-2, expression vectors that are suitable for expression on that subject, and that are safe for use in vivo, should be chosen. Methods on generating an immune response against SARS-CoV-2 are described in detail in Section VII below.


For applications where it is desired that the proteins be expressed in vivo, for example, when the transgenes of the disclosure are used in DNA or DNA-containing vaccines, any vector that allows for the expression of the proteins of the present disclosure and is safe for use in vivo may be used. In some embodiments the vectors used are safe for use in humans, mammals and/or laboratory animals.


In some embodiments it may be desirable to express and isolate the proteins or viral particles of the present disclosure, such as from animal subjects or cells (such as bacterial cells, yeast, insect cells, and mammalian cells) grown in culture, for preclinical testing of the immunogenic recombinant proteins or vaccines of the disclosure. Any suitable transfection, transformation, or gene delivery methods can be used and such methods are well known by those skilled in the art. Examples of methods known in the art can be chosen from transfection, transformation, microinjection, infection, electroporation, lipofection, and liposome-mediated delivery. The proteins and viral particles of the disclosure can also be expressed using methods known in the art, including in vitro transcription/translation systems.


In other embodiments, it may be desirable to express the proteins or viral particles of the disclosure in human subjects, such as in clinical trials and for actual clinical use of the immunogenic recombinant proteins or vaccine of the present disclosure. In some embodiments, vectors used for these in vivo applications are attenuated to vector from amplifying in the subject. For example, if plasmid vectors are used, they may lack an origin of replication that functions in the subject so as to enhance safety for in vivo use in the subject. If viral vectors are used, they may be attenuated or replication-defective in the subject, again, so as to enhance safety for in vivo use in the subject.


In some embodiments of the present disclosure, viral vectors are used. The present disclosure relates to recombinant vesicular stomatitis (VSV) vectors, however, other vectors may be contemplated in other embodiments of the disclosure such as, but not limited to, prime boost administration which may comprise administration of a recombinant VSV vector in combination with another recombinant vector expressing one or more SARS-CoV-2 epitopes. Viral expression vectors are well known to those skilled in the art and include, for example, viruses such as adenoviruses, adeno-associated viruses (AAV), alphaviruses, herpesviruses, retroviruses and poxviruses, including avipox viruses, attenuated poxviruses, vaccinia viruses, and particularly, the modified vaccinia Ankara virus (MVA; ATCC Accession No. VR-1566). Such viruses, when used as expression vectors are innately non-pathogenic in the selected subjects such as humans or have been modified to render them non-pathogenic in the selected subjects. For example, replication-defective adenoviruses and alphaviruses are well known and can be used as gene delivery vectors.


The nucleotide sequences of the present disclosure may be inserted into “vectors.” The term “vector” is widely used and understood by those of skill in the art, and as used herein the term “vector” is used consistent with its meaning to those of skill in the art. For example, the term “vector” is commonly used by those skilled in the art to refer to a vehicle that allows or facilitates the transfer of nucleic acid molecules from one environment to another or that allows or facilitates the manipulation of a nucleic acid molecule.


A. Recombinant VSV Vectors and Recombinant VSV Particles


In some embodiments, the vector is a recombinant vesicular stomatitis virus (VSV) vector. The term “VSV vector” refers to a vector of the Rhabdoviridae family of enveloped viruses that can be used to deliver genetic material to a host cell, and the VSV vector can be maintained by replication within the host cell. VSVs contain a single-stranded, nonsegmented, negative-sense RNA genome. Because the VSV naturally infects livestock and is known to infect humans producing mild illness or no symptoms of infection, the VSV has been modified to be a human vaccine vector for producing recombinant replicable VSV particles presenting foreign immunogenic antigens to induce protective immune response to a pathogen in a subject. VSV vectors are practical, safe, and immunogenic vectors that are used for conducting animal studies, and an attractive candidate for developing vaccines for use in humans.


A VSV vector may comprise any combination of the VSV genes N, P, M, G, and L (VSV-N, VSV-P, VSV-M, VSV-G, and VSV-L), wherein some or all of these genes may be present or absent, and each of these genes may be present or absent in part or in its entirety. The VSV genes are described in Section II.C below. Additional protein coding genes or open reading frames (ORFs) may be expressed from a VSV vector.


A recombinant VSV vector may comprise a VSV genome, which may provide a recombinant VSV particle. A recombinant VSV particle may also comprise a VSV genome. In some embodiments, the recombinant VSV vector and/or recombinant VSV particle is replicable.


1. Modified VSV Genome


In many embodiments, the recombinant VSV vector or recombinant VSV particle may comprise a modified VSV genome wherein at least a portion of the VSV genome is present. In some embodiments, the recombinant VSV vector or recombinant VSV particle comprises the VSV-N, VSV-P, VSV-M, and VSV-L genes, or any combination thereof. The VSV-N, VSV-P, VSV-M, and VSV-L genes are described in Section II.C.1 below. In some embodiments, the VSV genome further comprises at least a fragment of the VSV-G gene. In other embodiments, the VSV genome may exclude the VSV-G gene in its entirety. The VSV-G is described in Section II.C.2 below.


2. Modified VSV Genome and a Foreign Gene


In some embodiments, the modified VSV genome may further comprise a foreign gene, thereby giving rise to expression of a foreign epitope. In some embodiments, the foreign gene is derived from SARS-CoV-2 and the foreign epitope is a SARS-CoV-2 epitope, for example, a SARS-CoV-2 S protein or an immunogenic variant thereof. Therefore, a recombinant VSV vector or recombinant VSV particle may comprise a modified VSV genome and a nucleic acid sequence encoding the SARS-CoV-2 S protein or an immunogenic variant thereof. FIGS. 4 and 5 show exemplary nucleotide sequences encoding VSVΔG-SARS-CoV-2 S proteins inserted in a VSV genomic background, encoding the SARS-CoV-2 S protein or variant thereof. As such, in some embodiments, a recombinant VSV vector or recombinant VSV particle may comprise modified VSV genome and a nucleotide sequence encoding a SARS-CoV-2 S protein. The SARS-CoV-2 S protein and variants thereof are described in Section II.B below, while immunogenic proteins are described below in Section III below. Exemplary recombinant VSV vectors comprising modified VSV genome and a SARS-CoV-2 S protein variant are described in Section II.F below. In some embodiments, the recombinant VSV particle displays the SARS-CoV-2 S protein or variant thereof on its surface.


B. SARS-CoV-2 S Proteins


A recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine may comprise any epitope recognized by an anti-SARS-CoV-2 antibody. For example, the SARS-CoV-2 S protein of any coronavirus that causes COVID-19, including any coronavirus that may be isolated from a COVID-19 patient, may be used. In many embodiments, a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of the present disclosure may comprise a SARS-CoV-2 S protein or variant thereof.


The SARS-CoV-2 S protein binds to and enters the host cell by recognizing the receptor ACE2. (Li, W. et al., Nature, 426, 46965 (2003): 450-454.) The SARS-CoV-2 S protein has 1273 amino acids and contains S1 and S2 subunits (amino acids 14-685 and 686-1273, respectively). (Huang, Y. et al., Acta Pharmacologica Sinica, 41, 9 (2020):1141-1149.) The S1/S2 cleavage site lies (SPRRARSV, which are amino acids 680-687 in SEQ ID NO: 1, wherein cleavage occurs between R685 and S686) within a solvent-exposed loop Furin (Wrapp, D. et al., Science, 367, 6483 (2020):1260-1263; amino acids 673-691 in SEQ ID NO: 1) and is cleaved by proteases such as Furin that recognize basic amino acid sequences like 682-RRAR-685 found in the SARS-CoV-2 S precursor protein. Cleavage at S1/S2 enhances viral infectivity. (Tang et al., Coronavirus membrane fusion mechanism offers a potential target for antiviral development, Antiviral Res (2020), doi:10.1016/j.antiviral.2020.104792.) The S2′ cleavage site (previously referenced as S″; SKPSKRSF, which are amino acids 810-817 in SEQ ID NO: 1, wherein cleavage occurs between 815R and 816S) lies within a solvent-exposed loop (Wrapp, D. et al., Science, 367, 6483 (2020):1260-1263; amino acids 673-691 in SEQ ID NO: 1) and is joined to the N-terminus of the fusion peptide. Cleavage at the S2′ site at amino acid R815 by proteases like Cathepsin L or TMPRSS2 triggers fusion peptide activity that allows the S2 subunit to direct fusion between the viral and cellular membranes. Cleavage at the S2′ site is related to virus infection. (Tang et al., Coronavirus membrane fusion mechanism offers a potential target for antiviral development, Antiviral Res (2020), doi:10.1016/j.antiviral.2020.104792.)


All numbering of the SARS-CoV-2 S protein in the disclosure and claims is relative to SEQ ID NO: 1. This includes both position numbers and numbers of amino acids deleted from certain truncation mutations; all are relative to SEQ ID NO: 1. Unless otherwise specified by referring to a particular variant or by specifically referring to wildtype, all references in the disclosure and claims to SARS-CoV-2 S protein or S protein include both the wildtype and immunogenic variant forms of the S protein (including immunogenic variants that have substitution mutations, insertion mutations, deletion mutations (including fragments, which are shorter in length than wildtype), and/or modifications).


The SARS-CoV-2 S protein further comprises the following domains: signal peptide (amino acids 1-13 in Huang et al.; amino acids 1-15 in SEQ ID NO: 1), N-terminal domain (NTD; amino acids 14-305 in Huang et al.), receptor-binding domain (RBD; amino acids 319-541 in Huang et al.), fusion peptide (FP; amino acids 788-806 in Huang et al.), heptad repeat 1 (HR1; amino acids 912-984 in Huang et al.), heptad repeat 2 (HR2; amino acids 1163-1213 in Huang et al.), transmembrane domain (TMD; amino acids 1213-1237 in Huang et al.; amino acids 1218-1234 in SEQ ID NO: 1), and cytoplasmic tail (CT; amino acids 1237-1273 in Huang et al.; amino acids 1235-1273 in SEQ ID NO: 1) (Huang, Y. et al., Acta Pharmacologica Sinica, 41, 9 (2020):1141-1149). Within the HR2 domain, lies a membrane-proximal external region (MPER; amino acids 1205-1213 in Zhu et al.; amino acids 1206-1217 in SEQ ID NO: 1). (Zhu, W. et al., Journal of Virology, 94, 14 (2020):1-12.) Within the CT lies an endoplasmic reticulum retention sequence (ERRS; amino acids 1251-1255 in Ujike et al.). (Ujike, M. et al., Journal of General Virology, 97, 8 (2016):1853-1864.)


The SARS-CoV-2 virus binds to the host cell receptor ACE2 via the RBD in the S1 subunit. (Huang, Y. et al., Acta Pharmacologica Sinica, 41, 9 (2020):1141-1149.) This binding promotes the formation of endosomes, which initiates viral fusion activity under low pH. The S2 subunit, which comprises the FP, HR1, HR2, TMD and CT, is responsible for viral fusion and entry. (Huang, Y. et al., Acta Pharmacologica Sinica, 41, 9 (2020):1141-1149.) FP is a 15-20 stretch of mainly hydrophobic amino acids (such as glycine or alanine) and has been shown to disrupt and connect lipid bilayers of the host cell membrane. (Huang, Y. et al., Acta Pharmacologica Sinica, 41, 9 (2020):1141-1149.) HR1 and HR2 comprise repeats of a heptapeptide, and the two HR domains come together to form a six-helical bundle that fuses the host cell membrane with the viral membrane. (Huang, Y. et al., Acta Pharmacologica Sinica, 41, 9 (2020):1141-1149.) Following entry, viral RNA is released into the cytoplasm and their replication and transcription begin. (Song, Z. et al., Viruses, 11, 59 (2019):1-58.)


The SARS-CoV-2 virus assembles by packaging genomic RNA and nucleocapsid (N) protein from the cytoplasm, followed by budding into the lumen of the endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC). (Song, Z. et al., Viruses, 11, 59 (2019):1-58.) The ERRS is thought to facilitate accumulation of SARS-CoV-2 S proteins at the SARS-CoV-2 budding site and SARS-CoV-2 S protein incorporation into viral particles. (Ujike, M. et al., Journal of General Virology, 97, 8 (2016):1853-1864.)


In many embodiments, any variant of the SARS-CoV-2 S protein may be used in the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of the present disclosure. In some embodiments, the variant may be of equal (i.e., full-length), shorter, or longer length when compared to the wild-type SARS-CoV-2 S protein. In some embodiments, the variant comprises truncations or fragments of the wild-type SARS-CoV-2 S protein. In some embodiments, the variant comprises point mutations, silent mutations, synonymous mutations, nonsynonymous mutations, nonsense mutations, deletions, and/or insertions. In some embodiments, the variant comprises at least one mutation and/or truncation relative to the wild-type SARS-CoV-2 S protein (SEQ ID NO: 1). In some embodiments, the SARS-CoV-2 S protein comprises an amino acid sequence having a length of at least 1223, 1228, 1233, 1238, 1243, 1248, 1249, 1250, 1251, 1252, 1253, 1254, 1255, 1256, 1257, 1258, 1259, 1260, 1261, 1262, 1263, 1264, 1265, 1266, 1267, 1268, 1269, 1270, 1271, 1272, 1273, 1274, 1275, 1276, 1277 or 1278 amino acids and having homology over its own length of at least 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% homology to any one of SEQ ID NOS: 1, 4, 7, 10, 13, 16, 19, 22, 25-67, 153, or 158. In some embodiments, the SARS-CoV-2 S protein or immunogenic variant thereof (or the nucleotide sequence encoding it) comprises any one of SEQ ID NOS: 1, 4, 7, 10, 13, 16, 19, 22, 25-67, 153, or 158.


Advantageously, codons may be optimized for the SARS-CoV-2 S gene so it has the codon bias that is characteristic of VSV. This also results in a relatively low Guanine+Cytosine content of 40-45%. See, e.g., Rabinovich et al., PLoS One. 2014 Sep. 12; 9(9):e106597. doi: 10.1371/journal.pone.0106597. eCollection 2014. Adaptive mutations may also be used to identify variants of the SARS-CoV-2 S protein gene capable of improving replication capacity of a recombinant VSV comprising the SARS-CoV-2 S gene.


The nucleotide sequence encoding a SARS-CoV-2 S protein may comprise any of the nucleotide sequences of SEQ ID NOS: 2, 3, 5, 6, 8, 9, 11, 12, 14, 15, 17, 18, 20, 21, 23, and 24, as shown in FIG. 1B, 1C, 1E, 1F, 1H, 1I, 1K, 1L, 1N, 1O, IQ, 1R, 1T, 1U, 1W or 1X; any nucleotide sequence encoding a SARS-CoV-2 S protein chosen from SARS-CoV-2 genome sequences that are readily available to a skilled artisan at Genbank, including but not limited to LR757997, MT072688, MN996527, MT039873, MN938384, MT020781, MN996529, MN996530, MN996531, MT044258, LR757998, LR757996, MT066175, MT066176, LR757995, LC522972, LC522973, LC522974, LC522975, MT039887, MN988668, MN988669, MT044257, MT039888, MN997409, MN985325, MN994467, MT027062, MT027063, MT027064, MT020880, MT020881, MN988713, MT019533, MN994468, MT093571, MT019530, MT019532, MN975262, MN996528, MT007544, MT019529, MT019531, MT049951, MT039890, MN908947, NC_045512, MT093631; or any of the amino acid sequences of SEQ ID NOS: 1, 4, 7, 10, 13, 16, 19, 22, 25-67, 153, or 158. The SARS-CoV-2 S protein may comprise any of the amino acid sequences of SEQ ID NOS: 1, 4, 7, 10, 13, 16, 19, 22, 25-67, 153, or 158. The SARS-CoV-2 S protein may comprise any amino acid sequence encoded by all or part of the nucleic acid sequences referenced herein.


In many embodiments, the SARS-CoV-2 S protein may have an amino acid sequence of at least 70%, 80%, 90%, 95%, 99%, or 100% identical to the amino acid sequence of any of the specific SARS-CoV-2 S proteins referenced herein. In many embodiments, the SARS-CoV-2 S protein of the present disclosure may comprise a nucleotide sequence that is a fragment of the wild-type SARS-CoV-2 S protein nucleotide sequence or a nucleotide sequence of a SARS-CoV-2 S protein nucleotide sequence having at least one modification (e.g., deletion, addition, substitution). The SARS-CoV-2 S protein may comprise at least one mutation relative to SEQ ID NO: 1.


In many embodiments, the SARS-CoV-2 S protein is an immunogenic variant that is capable of inducing an immune response in a subject administered the immunogenic variant. General descriptions on immunogenic recombinant proteins as well as details that relate to immunogenic SARS-CoV-2 S protein variants are described in Section III below.


1. Optional Deletions to the SARS-CoV-2 S Protein


A variety of C-terminal deletions have been observed in variants of the SARS-CoV-2 S protein (see Example 8 below), which may provide a benefit by improving viral stability. In many embodiments, the SARS-CoV-2 S protein comprises a C-terminal deletion. The SARS-CoV-2 S protein may be a fragment of the wild-type SARS-CoV-2 S protein. SARS-CoV-2 S proteins with optional deletion(s) are provided herein. In some embodiments, the SARS-CoV-2 S protein is the same length as or is a fragment of the full-length of the SARS-CoV-2 S protein. In some embodiments, the deleted SARS-CoV-2 S protein is an immunogenic recombinant protein.


In some embodiments, distinct domains or subunits of the SARS-CoV-2 S protein are removed to produce the immunogenic variant. Distinct domains or subunits that are removed from the SARS-CoV-2 S protein may be chosen from S1 subunit, S2 subunit, S1/S2 cleavage site, protease cleavage site, signal peptide, NTD, RBD, FP, HR1, HR2, and CT.


In some embodiments, deletions of the SARS-CoV-2 S protein are made starting from the C-terminus of SEQ ID NO: 1 or any of the other variants of the S protein described herein. The full-length SARS-CoV-2 S protein comprises 1273 amino acids. A fragment length of SARS-CoV-2 S protein is calculated by subtracting the number of deleted amino acids from 1273. In some embodiments, the SARS-CoV-2 S protein is 1223, 1228, 1233, 1238, 1243, 1248, 1249, 1250, 1251, 1252, 1253, 1254, 1255, 1256, 1257, 1258, 1259, 1260, 1261, 1262, 1263, 1264, 1265, 1266, 1267, 1268, 1269, 1270, 1271, 1272, 1273, 1274, 1275, 1276, 1277 or 1278 amino acids. In some embodiments, the SARS-CoV-2 S protein may comprise a fragment of the SARS-CoV-2 S protein having a deletion at the C-terminal end of the SARS-CoV-2 S protein. In some embodiments, the SARS-CoV-2 S protein comprises a deletion of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, or 50 amino acids from the C-terminal end of an S protein described herein with the deletion measured relative to the length of SEQ ID NO: 1.


In some embodiments, the SARS-CoV-2 S protein may have a deletion of from 1 to 30, 5 to 25, or 9 to 23 amino acids from the C-terminal end of the SARS-CoV-2 S protein. The SARS-CoV-2 S protein may comprise a fragment of the SARS-CoV-2 S protein having a deletion of 9 amino acids (Δ9), 13 amino acids (Δ13), 19 amino acids (Δ19), 21 amino acids (Δ21), or 23 amino acids (Δ23) at the C-terminal end of the SARS-CoV-2 S protein. In some embodiments, the SARS-CoV-2 S protein comprises a deletion of 1 to 30, 5 to 25, or 9 to 23 amino acids from the C-terminal end of SEQ ID NO: 1. In some embodiments, the SARS-CoV-2 S protein comprises a deletion of 9 amino acids (Δ9), 13 amino acids (Δ13), 19 amino acids (Δ19), 21 amino acids (Δ21) or 23 amino acids (Δ23) from the C-terminal end of SEQ ID NO: 1 or a relative length of another variant described herein.


The full-length SARS-CoV-2 S protein comprises a cytoplasmic tail and a transmembrane domain at the C-terminal end. In some embodiments, the SARS-CoV-2 S protein may comprise a fragment of the SARS-CoV-2 S protein lacking the cytoplasmic tail of the SARS-CoV-2 S protein. In some embodiments, the SARS-CoV-2 S protein does not comprise the entire cytoplasmic tail. In some embodiments, the SARS-CoV-2 S protein may lack a cytoplasmic tail in part or in its entirety.


In some embodiments, the SARS-CoV-2 S protein may lack the transmembrane domain of the SARS-CoV-2 S protein. The SARS-CoV-2 S protein may comprise a fragment of the SARS-CoV-2 S protein lacking the cytoplasmic tail and the transmembrane domain of the SARS-CoV-2 S protein or a variant thereof.


An ERRS (endoplasmic reticulum retention sequence (amino acids 1251-1255 of the SARS-CoV-2 S protein disclosed in Ujike, M. et al., Journal of General Virology, 97, 8 (2016):1853-1864) lies within the cytoplasmic tail of the full-length SARS-CoV-2 S protein. In some embodiments, the SARS-CoV-2 S protein does not comprise ERRS in part or in its entirety.


In some embodiments, a deletion of the SARS-CoV-2 S protein occurs at position 1251 relative to SEQ ID NO: 1. In some of these embodiments, the codon encoding amino acid 1251 was changed to a stop codon, which causes amino acid 1251 and subsequent amino acids to be absent. In these embodiments, amino acid 1251 is deleted. In some embodiments, the SARS-CoV-2 S protein has a 23 amino acid deletion at the C-terminal cytoplasmic tail. In some of these embodiments, the 23 amino acid deletion includes deletion of amino acid at position 1251 relative to SEQ ID NO: 1. In some embodiments, the following amino acids are deleted from the C-terminus of SARS-CoV-2 S protein: GSCCKFDEDDSEPVLKGVKLHYT (SEQ ID NO: 193).


In some embodiments, the SARS-CoV-2 of the S protein does not comprise a 24 amino acid deletion at the C-terminal cytoplasmic tail. In some embodiments, the SARS-CoV-2 S protein does not comprise a 21-amino acid deletion at the C-terminal cytoplasmic tail.


In some embodiments, the SARS-CoV-2 S gene does not comprise a TGC to TGA mutation at nucleotide 3759. In some embodiments, the SARS-CoV-2 S protein does not comprise a cysteine to stop mutation at amino acid 1253. In some embodiments, the SARS-CoV-2 S protein does not comprise an alanine mutation at amino acid 1269 and/or 1271.


2. Optional Fusion of SARS-CoV-2 S Protein with Virus Glycoprotein


In some embodiments, the SARS-CoV-2 S protein comprises (1) a cytoplasmic tail and a transmembrane domain of a virus glycoprotein protein; and (2) a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail and a transmembrane domain. In these embodiments, the cytoplasmic tail and the transmembrane domain of the SARS-CoV-2 S protein are replaced by the cytoplasmic tail and transmembrane domain of the virus glycoprotein protein. In some embodiments, the SARS-CoV-2 S protein may lack a SARS-CoV-2 S protein cytoplasmic tail and/or a SARS-CoV-2 S protein transmembrane domain in part or in its entirety. When expressed, the cytoplasmic tail and the transmembrane domain of the virus glycoprotein protein and the SARS-CoV-2 S protein lacking its cytoplasmic tail and transmembrane domain may form a fusion protein, wherein the cytoplasmic tail and the transmembrane domain of the SARS-CoV-2 S protein are replaced by the cytoplasmic tail and transmembrane domain of the virus glycoprotein protein. In some embodiments, the virus glycoprotein is VSV-G. Details on VSV-G are provided in Section II.C.2 below. The SARS-CoV-2 S protein may comprise the amino acid sequence in FIG. 1D, 1G, 1J, 1M, 1P, 1S or 1V (SEQ ID NO: 4, 7, 10, 13, 16, 19 or 22, respectively).


3. Optional Substitution Mutations to the SARS-CoV-2 S Protein


In many embodiments, the SARS-CoV-2 S protein may comprise at least one substitution mutation. In some embodiments, the at least one mutation relative to SEQ ID NO: 1 is along the length of the immunogenic variant.


Point mutations have been observed in the Furin cleavage site of SARS-CoV-2 S protein following passage in Vero cells, which may indicate that these point mutations are adaptive mutations to the Vero host cell line. The at least one mutation may be in the exposed loop (a solvent-exposed loop (amino acids 673-691 in SEQ ID NO: 1) that comprises the S1/S2 cleavage site (SPRRARSV, which are amino acids 680-687 in SEQ ID NO: 1, wherein cleavage occurs between R685 and S686)) of the SARS-CoV-2 S protein. Further, the at least one mutation may reduce or block S1/S2 cleavage of the SARS-CoV-2 S protein by, for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, as compared with that of a wild-type SARS-CoV-2 S protein without the at least one mutation. The at least one mutation may be in the S2′ cleavage site (Cathepsin H, L; amino acids 810-817 in SEQ ID NO: 1, wherein cleavage occurs between 815R and 816S) of the SARS-CoV-2 S protein and such at least one mutation may modulate S2′ cleavage of the SARS-CoV-2 S protein by, for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, as compared with that of a wild-type SARS-CoV-2 S protein without the at least one mutation. The at least one mutation may be in the C-terminal end of the SARS-CoV-2 S protein.


In some embodiments, the at least one mutation in the SARS-CoV-2 S protein or SEQ ID NO: 1 may be chosen from a mutation at residue 655, at least one mutation from residue 672 to residue 687, at least one mutation from residue 802 to residue 817, at least one mutation from residue 1233 to residue 1273, and combinations thereof. In some embodiments, the mutation at residue 655 in SEQ ID NO: 1 may be H655Y. In some embodiments, the at least one mutation between residues 672 and 687 in SEQ ID NO: 1 may be from residue 678 to residue 685, which may be chosen from T678I, P681S, R682K, R683G, R685G and combinations thereof. In some embodiments, the at least one mutation from residue 802 to residue 817 in SEQ ID NO: 1 may be from residue 810 to residue 815, which may be chosen from P812R, S813R, S813F and combinations thereof. In some embodiments, the at least one mutation from residue 1233 to residue 1273 in SEQ ID NO: 1 may be chosen from a deletion at the C-terminal end of the SEQ ID NO: 1, M1233K and a combination thereof.


In many embodiments, the at least one mutation in SEQ ID NO: 1, either in isolation or in any combination, comprises H655Y, R682K, R683G, N709S, S813F, N978K, S940G, D1118A, and/or D1163N.


In some embodiments, the SARS-CoV-2 S protein may further comprise at least one mutation in SEQ ID NO: 1 chosen from F140V, Q321P, N715S, D1118A and combinations thereof.


In some embodiments, the SARS-CoV-2 S protein may further comprise at least one mutation in SEQ ID NO: 1 chosen from E154D, S115L, D614G, D614N, R685G, or combinations thereof.


In some embodiments, the at least one mutation in SEQ ID NO: 1 does not comprise a mutation at R685.


C. VSV Components


1. VSV N, P, M, and L Genes


The VSV genome is composed of 5 genes encoding a VSV nucleoprotein (VSV-N), a VSV phosphoprotein (VSV-P), a VSV matrix protein (VSV-M), a VSV glycoprotein (VSV-G) and a VSV polymerase (VSV-L) and arranged sequentially 3′-N-P-M-G-L-5′. Each gene encodes a polypeptide found in mature virions


In many embodiments, a recombinant VSV vector, recombinant VSV particle, or SARS-CoV-2 vaccine may comprise a modified VSV genome or at least a portion of the VSV genome. In some embodiments, the VSV genome comprises the VSV-N, VSV-P, VSV-M, and VSV-L genes or any combination thereof. In some embodiments, the VSV genome does not comprise the VSV-G gene. In some embodiments, the VSV genome may comprise at least a fragment of the VSV-G gene. VSV-G and variants thereof are described in Section II.C.2 below.


In some embodiments, the VSV genome comprises at least one mutation in at least one of the VSV genes. In many embodiments, any variant of the VSV genes or VSV proteins may be used in the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of the present disclosure. In some embodiments, the variant may be of equal (i.e., full-length), shorter, or longer length when compared to a wild-type protein sequence of a VSV gene. In some embodiments, the variant comprises deletions or fragments of the wild-type protein sequence of a VSV gene. In some embodiments, the variant comprises point mutations, silent mutations, synonymous mutations, nonsynonymous mutations, nonsense mutations, deletions, and/or insertions.


In some embodiments, the variant comprises at least one mutation and/or deletion relative to the wild-type VSV-M protein (SEQ ID NO: 157). In some embodiments, the VSV-M protein comprises an amino acid sequence having homology over its own length of at least 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% homology to SEQ ID NO: 151 or 157. In some embodiments, the VSV-M protein comprises Y61S. In some embodiments, the VSV-M protein or immunogenic variant thereof comprises SEQ ID NO: 151.


In some embodiments, the variant comprises at least one mutation and/or deletion relative to the wild-type VSV-L protein (SEQ ID NO: 152). In some embodiments, the VSV-L protein comprises an amino acid sequence having homology over its own length of at least 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% homology to SEQ ID NO: 152 or 158. In some embodiments, the VSV-L protein comprises a I1343I silent mutation.


In some embodiments, the modified VSV genome may comprise a foreign gene encoding the SARS-CoV-2 S protein or an immunogenic variant thereof. SARS-CoV-2 S protein and variants thereof are described in Section II.B above. Immunogenic recombinant proteins are described in Section III below.


All numbering of the VSV proteins in the disclosure and claims is relative to wildtype versions. This includes both position numbers and numbers of amino acids deleted from certain deletion mutations. Unless otherwise specified by referring to a particular variant or by specifically referring to wildtype, all references in the disclosure and claims to the VSV proteins include both the wildtype and mutant forms of the VSV proteins (including variants that have substitution mutations, insertion mutations, deletion mutations (including fragments, which are shorter in length than wildtype), and/or modifications).


2. Optional Mutations with VSV Glycoprotein


VSV-G is a transmembrane polypeptide that is present in the viral envelope as a homotrimer, and like the envelope protein, mediates cell attachment and infection. VSV-G comprises a cytoplasmic tail and a transmembrane domain at the C-terminal end. In some embodiments, the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of the present disclosure does not comprise a VSV-G.


In other embodiments, the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises a VSV-G gene/protein or at least a fragment thereof. In some embodiments, the VSV-G protein or fragment thereof is covalently linked to a SARS-CoV-2 S protein. Details relating to recombinant VSV vectors and recombinant VSV particles are provided in Section II.A above. Details relating to immunogenic recombinant proteins are provided in Section III below. Details relating to vaccines are provided in Section IV.A below. The VSV-G variant may be a fragment of the VSV-G nucleotide sequence or a fragment of the VSV-G amino acid sequence, wherein the nucleotide or amino acid sequence may have at least one modification (e.g., deletion, addition, or substitution). In some embodiments, the cytoplasmic tail is absent, in part or in its entirety, from the VSV-G. In some embodiments, the cytoplasmic tail and the transmembrane domain are absent, in part or in their entirety, from the VSV-G.


In some embodiments, the VSV-G gene encodes a cytoplasmic tail of a VSV-G protein and/or a transmembrane domain of a VSV-G protein. In some embodiments, the VSV-G protein comprises a fragment of at least 21 amino acids at the C-terminal end of full-length VSV-G protein; or a fragment of at least 29 amino acids at the C-terminal end of full-length VSV-G protein. In some embodiments, the fragment of the VSV-G protein may comprise from 21 to 29 amino acids at the C-terminal end of the full-length VSV-G protein.


In some embodiments, the recombinant VSV vector, recombinant VSV particle, or SARS-CoV-2 vaccine comprises a fusion gene wherein a fragment of VSV-G gene is operably linked to nucleotide sequence encoding a fragment of SARS-CoV-2 S protein. In related embodiments, the recombinant VSV particle or SARS-CoV-2 vaccine comprises a fusion protein, wherein the VSV-G protein is expressed as a fragment that is covalently linked to a SARS-CoV-2 S protein fragment. In these embodiments, as described in detail in Section II.B.2 above, the SARS-CoV-2 S protein fragment lacks a SARS-CoV-2 S protein cytoplasmic tail and a SARS-CoV-2 S protein transmembrane domain, and has a VSV-G cytoplasmic tail and VSV-G transmembrane domain instead.


D. Arrangement of Elements in a Recombinant VSV Vector


In the recombinant VSV vector of the present disclosure, the modified VSV genome may comprise VSV genes, such as the VSV-N, VSV-P, VSV-M, and VSV-L genes or any combination thereof. These VSV-N, VSV-P, VSV-M, and VSV-L genes may be arranged in sequence from 3′ end to 5′ end. The foreign gene may be at any location in the modified VSV genome. For example, the foreign gene may be inserted on the 3′ end of the VSV-N gene, between the VSV-N gene and the VSV-P gene, between the VSV-P gene and the VSV-M gene, between the VSV-M gene and the VSV-L gene, or on the 5′ end of the VSV-L gene.


In some embodiments, the foreign gene is a nucleotide sequence encoding the SARS-CoV-2 S protein or immunogenic variant thereof. In a recombinant VSV vector, the VSV genes and the SARS-CoV-2 S protein or immunogenic variant thereof may be arranged in a variety of ways. In some embodiments, the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is 3′ of the VSV-N gene. In some embodiments, the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is on the 3′ end of the VSV-N gene. In some embodiments, the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is between the VSV-N gene and the VSV-P gene. In some embodiments, the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is between the VSV-P gene and the VSV-M gene. In some embodiments, the SARS-COV-2 S protein or an immunogenic variant thereof is between the VSV-M gene and the VSV-L gene. In some embodiments, the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is 5′ of the VSV-L gene. In some embodiments, the nucleic acid sequence encoding the SARS-COV-2 S protein or an immunogenic variant thereof is on the 5′ end of the VSV-L gene.


E. Exemplary Variants of the SARS-CoV-2 S Protein and VSV Proteins


The recombinant VSV vectors, recombinant VSV particles, immunogenic recombinant proteins, and SARS-CoV-2 vaccines of the present disclosure may comprise one of the following exemplary mutants of the SARS-CoV-2 S protein (SEQ ID NO: 1):


(1) H655Y, R682K, and R685G;


(2) H655Y, R682K, R685G, and a deletion of the 9 amino acids (Δ9) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(3) H655Y, R682K, R685G, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(4) Q321P, H655Y, T678I, and P812R;


(5) Q321P, H655Y, T678I, P812R, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(6) E154D, S155L, T678I, R685G, and M1233K;


(7) R685G, S813R, and a deletion of the 9 amino acids (Δ9) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(8) H655Y, R682K, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(9) R685G, S813F, and a deletion of the 9 amino acids (Δ9) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(10) R683G, S813F and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(11) R683G, S813F, R685G, and a deletion of 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(12) H655Y, P681S, R682K, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(13) R683G and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(14) H655Y and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(15) R683G, D1118A, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(16) R683G, N715S, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(17) H655Y, N709S, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(18) F140V, H655Y and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(19) a deletion of the 21 amino acids (Δ21) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(20) a deletion of the 19 amino acids (Δ19) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1, wherein the SARS-CoV-2 S protein further comprises a fragment of at least 21 amino acids at the C-terminal end of a VSV-G protein;


(21) E484D, H655Y, and a deletion of 21 amino acids (Δ21) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;


(22) H665Y, R685G and a deletion of the 19 amino acids (Δ19) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1, wherein the SARS-CoV-2 S protein further comprises a fragment of at least 21 amino acids at the C-terminal end of a VSV-G protein;


(23) D614G; or


(24) D614N.


In addition to an exemplary mutant of SARS-CoV-2 S protein, the recombinant VSV vectors, recombinant VSV particles, immunogenic recombinant proteins, and SARS-CoV-2 vaccines of the present disclosure may comprise any or all of the following exemplary mutants of the VSV proteins:


(1) Y61S in VSV-M;


(2) E213K in VSV-M; and/or


(3) V411M in VSV-L.


F. Exemplary Recombinant VSV Vectors


In many embodiments, the recombinant VSV vectors, recombinant VSV particles, and SARS-CoV-2 vaccines of the present disclosure may comprise a modified VSV genome and a nucleotide sequence encoding SARS-CoV-2 S protein or immunogenic variant thereof, arranged in a variety of ways on a VSV vector backbone. Details and variants relating to modified VSV genomes and VSV components; and SARS-CoV-2 S proteins are described in Sections II.A.1 and II.C; and II.B, respectively. In some embodiments, the recombinant VSV vectors, recombinant VSV particles, and SARS-CoV-2 vaccines do not comprise VSV-G. In other embodiments, the recombinant VSV vectors, VSV particles, and SARS-CoV-2 vaccines may comprise VSV-G or a fragment thereof.


In some embodiments, the recombinant VSV vector, recombinant VSV particle, or SARS-CoV-2 vaccine comprises R683G, S813F and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1; and Y61S in VSV-M. In some embodiments, the recombinant VSV vector is rVSVΔG-SARS-CoV-2 clone MB1 (which has the nucleic acid sequence SEQ ID NO: 150; amino acid sequences SEQ ID NOS: 151-153, 155, and 156; and FIGS. 24A-G). The recombinant VSV vector rVSVΔG-SARS-CoV-2 clone MB1 does not comprise VSV-G. In some embodiments with these mutations, the recombinant VSV vector, particle, or vaccine does not comprise VSV-G in its entirety, but comprises it in part, such as the 21 amino acids of the cytoplasmic tail.


In some embodiments, the recombinant VSV vector, recombinant VSV particle, or SARS-CoV-2 vaccine comprises H655Y and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1. In some embodiments, the recombinant VSV vector is rVSVΔG-SARS-CoV-2 clone MB2 (which has the nucleic acid sequence SEQ ID NO: 154; amino acid sequences SEQ ID NOS: 152, 155, 156, 157, and 158; FIGS. 21A-G). The recombinant VSV vector rVSVΔG-SARS-CoV-2 clone MB2 does not comprise VSV-G. In some embodiments with these mutations, the recombinant VSV vector, particle, or vaccine does not comprise VSV-G in its entirety, but comprises it in part, such as the 21 amino acids of the cytoplasmic tail.


III. Immunogenic Recombinant Proteins

The recombinant VSV vectors, recombinant VSV particles, immunogenic recombinant proteins, or SARS-CoV-2 vaccines of the present disclosure may produce or comprise an immunogenic recombinant protein. An immunogenic recombinant protein is capable of inducing an immune response in a subject administered the immunogenic recombinant protein. In some embodiments, the immunogenic recombinant protein may induce an immune response capable of blocking at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% of SARS-CoV-2 infection in a subject administered the immunogenic recombinant protein as compared with a control subject not administered the immunogenic recombinant protein. The term “immunogenic variant” refers to a variant of an immunogenic protein that induces at least a portion of the immune response of the wild-type protein. In some embodiments, the variant is an immunogenic variant that induces a relevant immune response in vaccinated individuals to enable protection against COVID-19. In some embodiments, the immunogenic variant may induce an immune response capable of blocking at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% of a SARS-CoV-2 infection in a subject administered with the immunogenic variant as compared with a control subject administered the SARS-CoV-2 S protein. In some embodiments, the variant comprises a sequence that shares homology with the wild-type SARS-CoV-2 S protein (SEQ ID NO: 1), wherein homology is at least 90% similarity with the variant as the reference protein sequence.


In some embodiments, the immunogenic recombinant protein is a variant that may have an amino acid length that is the same as or a fragment of the full-length of the immunogenic recombinant protein. In many embodiments, the immunogenic recombinant protein may comprise a full-length SARS-CoV-2 S protein, fragments thereof, or mutant variants thereof, that are expressed by the recombinant VSV vector or recombinant VSV particle of the present disclosure. In embodiments where the immunogenic recombinant protein is not the full-length or wild-type SARS-CoV-2 S protein, the immunogenic recombinant protein is an immunogenic variant of the SARS-CoV-2 S protein. Details on full-length SARS-CoV-2 S protein, fragments thereof, and mutants thereof are provided in Section II.B above. Details on recombinant VSV vectors and recombinant VSV particles are provided in Section II.A above.


In some embodiments, the immunogenic variant is a fragment of the SARS-CoV-2 S protein having a deletion at the C-terminal end. In some embodiments, distinct domain of the SARS-CoV-2 S protein are removed to produce an immunogenic variant. In other embodiments, specific fragment lengths are deleted from the SARS-CoV-2 S protein. The distinct domains and specific fragments are discussed in Section II.B above.


The immunogenic variant of the SARS-CoV-2 S protein may comprise a fragment of the SARS-CoV-2 S protein or a variant thereof (the SARS-CoV-2 S protein fragment), and a portion of a VSV-G or a variant thereof (the VSV-G protein portion). In some embodiments, the SARS-CoV-2 S protein fragment lacks the cytoplasmic tail of the SARS-CoV-2 S protein, and the VSV-G protein portion comprises the cytoplasmic tail of the VSV-G protein. In some embodiments, the SARS-CoV-2 S protein may lack a cytoplasmic tail in part or in its entirety. In another embodiment, the SARS-CoV-2 S protein fragment lacks the cytoplasmic tail and the transmembrane domain of the SARS-CoV-2 S protein, and the VSV-G protein portion comprises the cytoplasmic tail and the transmembrane domain of the VSV-G protein. Details on the VSV-G protein are provided above in Section II.C.2.


In some embodiments, the immunogenic recombinant protein comprises VSV and SARS-CoV-2 proteins in rVSVΔG-SARS-CoV-2 clone MB1 (specifically amino acid SEQ ID NOS: 151-153, 155, and 156) or VSV and SARS-CoV-2 protein in rVSVΔG-SARS-CoV-2 clone MB2 (specifically amino acid SEQ ID NOS: 152 and 155-158).


In some embodiments, the SARS-CoV-2 vaccine comprises rVSVΔG-SARS-CoV-2 clone MB1 (nucleic acid sequence SEQ ID NO: 150; or amino acid sequences SEQ ID NO: 151-153, 155, and 156:) or rVSVΔG-SARS-CoV-2 clone MB2 (nucleic acid sequence SEQ ID NO: 154; or amino acid sequences SEQ ID NO: 155-158, and 152). Details on rVSVΔG-SARS-CoV-2 clones MB1 and MB2 are provided in Section II.F above.


IV. Pharmaceutical Compositions

The present disclosure further provides pharmaceutical compositions that may comprise any recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of the present disclosure. Recombinant VSV vectors and VSV particles are described in Section II.A above while the VSV components are described in Section II.C above. The SARS-CoV-2 S protein and variants thereof are described in Section II.B above, while immunogenic variants are described in Section III above.


The recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine may be produced according to the methods disclosed herein and provided to a pharmaceutical composition. In particular, methods for producing recombinant VSV particles are described in Section V below.


In some embodiments, the pharmaceutical composition comprises an effective amount of a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, and/or SARS-CoV-2 vaccine to generate an immune response against SARS-CoV-2 in a subject. In some embodiments, the pharmaceutical composition is a vaccine composition. In many embodiments, the pharmaceutical composition, including a vaccine composition, comprises at least one pharmaceutical excipient.


The pharmaceutical composition, including a vaccine composition, may be formulated for oral (including oral mucosal, buccal, and/or gastrointestinal) sublingual, intramuscular, intradermal, subcutaneous, intranasal (including nasal mucosal), intraocular, rectal, transdermal, mucosal, topical, intravenous, or parenteral administration.


The pharmaceutical compositions of the disclosure may be injectable suspensions, solutions, sprays, lyophilized powders, syrups, elixirs and the like. Any suitable form of pharmaceutical composition may be used. To prepare such a pharmaceutical composition, a nucleic acid molecule or vector of the disclosure, having the desired degree of purity, is mixed with one or more pharmaceutically acceptable carriers and/or excipients. The carriers and excipients must be “acceptable” in the sense of being compatible with the other ingredients of the composition. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to, water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, or combinations thereof, buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or nonionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG).


The pharmaceutical composition may be an immunogenic or immunological composition. The pharmaceutical composition can be formulated in the form of an oil-in-water emulsion. The oil-in-water emulsion can be based, for example, on light liquid paraffin oil (European Pharmacopea type); isoprenoid oil such as squalane, squalene, EICOSANE™ or tetratetracontane; oil resulting from the oligomerization of alkene(s), e.g., isobutene or decene; esters of acids or of alcohols containing a linear alkyl group, such as plant oils, ethyl oleate, propylene glycol di(caprylate/caprate), glyceryl tri(caprylate/caprate) or propylene glycol dioleate; esters of branched fatty acids or alcohols, e.g., isostearic acid esters. The oil advantageously is used in combination with emulsifiers to form the emulsion. The emulsifiers can be nonionic surfactants, such as esters of sorbitan, mannide (e.g., anhydromannitol oleate), glycerol, polyglycerol, propylene glycol, and oleic, isostearic, ricinoleic, or hydroxystearic acid, which are optionally ethoxylated, and polyoxypropylene-polyoxyethylene copolymer blocks, such as the Pluronic® products, e.g., L121.


The pharmaceutical composition of the disclosure can contain additional substances, such as wetting or emulsifying agents, buffering agents, or adjuvants to enhance the effectiveness of the vaccines (Remington's Pharmaceutical Sciences, 18th edition, Mack Publishing Company, (ed.) 1980).


Adjuvants may also be included. As used herein, the term “adjuvant” refers to a composition or compound that is capable of enhancing the immune response against an antigen of interest. Adjuvants are substances or combinations of substances that are used in conjunction with a vaccine antigen to enhance (e.g., increase, accelerate, prolong and/or possibly target) or modulate to a different type (e.g., switch a Th1 immune response to a Th2 response, or a humoral response to a cytotoxic T cell response) the specific immune response to the vaccine antigen in order to enhance the clinical effectiveness of the vaccine. In some embodiments, the adjuvant may modify (Th1/Th2) the immune response. In some embodiments, the adjuvant may boost the strength and longevity of the immune response. In some embodiments, the adjuvant may broaden the immune response to a concomitantly administered antigen. In some embodiments, the adjuvant may be capable of inducing strong antibody and T cell responses. In some embodiments, the adjuvant may be capable of increasing the polyclonal ability of the induced antibodies. In some embodiments, the adjuvant may be used to decrease the amount of antigen necessary to provoke the desired immune response and provide protection against the disease. In some embodiments, the adjuvant may be used to decrease the number of injections needed in a clinical regimen to induce a durable immune response and provide protection against the disease. Adjuvant containing formulations described herein may demonstrate enhancements in humoral and/or cellular immunogenicity of vaccine antigens, for example, subunit vaccine antigens. Adjuvants of the present invention are not used to deliver antigens, antibodies, APIs, or VLPs.


Adjuvants include, but are not limited to, mineral salts (e.g., AlK(SO4)2, AlNa(SO4)2, AlNH(SO4)2, silica, alum, Al(OH)3, Ca3(PO4)2, kaolin, or carbon), polynucleotides with or without immune stimulating complexes (ISCOMs) (e.g., CpG oligonucleotides, such as those described in Chuang, T. H. et al, (2002) J. Leuk. Biol. 71(3): 538-44; Ahmad-Nejad, P. et al (2002) Eur. J. Immunol. 32(7): 1958-68; poly IC or poly AU acids, polyarginine with or without CpG (also known in the art as IC31; see Schellack, C. et al (2003) Proceedings of the 34th Annual Meeting of the German Society of Immunology; Lingnau, K. et al (2002) Vaccine 20(29-30): 3498-508), JuvaVax™ (U.S. Pat. No. 6,693,086), certain natural substances (e.g., wax D from Mycobacterium tuberculosis, substances found in Cornyebacterium parvum, Bordetella pertussis, or members of the genus Brucella), flagellin (Toll-like receptor 5 ligand; see McSorley, S. J. et al (2002) J. Immunol. 169(7): 3914-9), saponins such as QS21, QS17, and QS7 (U.S. Pat. Nos. 5,057,540; 5,650,398; 6,524,584; 6,645,495), monophosphoryl lipid A, in particular, 3-de-O-acylated monophosphoryl lipid A (3D-MPL), imiquimod (also known in the art as IQM and commercially available as Aldara®; U.S. Pat. Nos. 4,689,338; 5,238,944; Zuber, A. K. et al (2004) 22(13-14): 1791-8), and the CCR5 inhibitor CMPD167 (see Veazey, R. S. et al (2003) J. Exp. Med. 198: 1551-1562). The adjuvant can be a mixture of emulsifier(s), micelle-forming agent, and oil such as that which is commercially available under the name Provax® (IDEC Pharmaceuticals, San Diego, Calif.).


Aluminum hydroxide or phosphate (alum) are commonly used at 0.05 to 0.1% solution in phosphate buffered saline. Other adjuvants that can be used with DNA vaccine, are cholera toxin, CTA1-DD/ISCOMs (see Mowat, A. M. et al (2001) J. Immunol. 167(6): 3398-405), polyphosphazenes (Allcock, H. R. (1998) App. Organometallic Chem. 12(10-11): 659-666; Payne, L. G. et al (1995) Pharm. Biotechnol. 6: 473-93), cytokines such as, but not limited to, IL-2, IL-4, GM-CSF, IL-12, IL-15 IGF-1, IFN-α, IFN-β, and IFNγ (Boyer et al., (2002) J. Liposome Res. 121:137-142; WO01/095919), immunoregulatory proteins such as CD40L (ADX40; see, for example, WO03/063899), and the CD1a ligand of natural killer cells (also known as CRONY or α-galactosyl ceramide; see Green, T. D. et al, (2003) J. Virol. 77(3): 2046-2055), immunostimulatory fusion proteins such as IL-2 fused to the Fc fragment of immunoglobulins (Barouch et al., Science 290:486-492, 2000) and co-stimulatory molecules B7.1 and B7.2 (Boyer), all of which can be administered either as proteins or in the form of DNA, on the same expression vectors as those encoding the antigens of the disclosure or on separate expression vectors.


In an advantageous embodiment, the adjuvants may be lecithin is combined with an acrylic polymer (Adjuplex-LAP), lecithin coated oil droplets in an oil-in-water emulsion (Adjuplex-LE) or lecithin and acrylic polymer in an oil-in-water emulsion (Adjuplex-LAO) (Advanced BioAdjuvants (ABA)).


The compositions can be designed to introduce the nucleic acid molecules or expression vectors to a desired site of action and release them at an appropriate and controllable rate. Methods of preparing controlled-release formulations are known in the art. For example, controlled release preparations can be produced by the use of polymers to complex or absorb the immunogen and/or immunogenic composition. A controlled-release formulation can be prepared using appropriate macromolecules (for example, polyesters, polyamino acids, polyvinyl, pyrrolidone, ethylenevinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate) known to provide the desired controlled release characteristics or release profile. Another possible method to control the duration of action by a controlled-release preparation is to incorporate the active ingredients into particles of a polymeric material such as, for example, polyesters, polyamino acids, hydrogels, polylactic acid, polyglycolic acid, copolymers of these acids, or ethylene vinylacetate copolymers. Alternatively, instead of incorporating these active ingredients into polymeric particles, it is possible to entrap these materials into microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacrylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in New Trends and Developments in Vaccines, Voller et al. (eds.), University Park Press, Baltimore, Md., 1978 and Remington's Pharmaceutical Sciences, 16th edition.


A. SARS-CoV-2 Vaccine


The vectors and viral particles of the present disclosure are useful for providing vaccines for delivering the nucleic acids encoding the proteins of the disclosure to a subject, such as a human, such that the proteins are then expressed in the subject to elicit an immune response. A vaccine may comprise recombinant VSV vector, VSV particle, a SARS-CoV-2 S protein, and/or an immunogenic recombinant variant thereof. Recombinant VSV vectors and VSV particles are described in Section II.A above while the VSV components are described in Section II.C above. The SARS-CoV-2 S protein and variants thereof are described in Section II.B above, while immunogenic variants are described in Section III above. Details on methods of administration are provided in Section VII.A below. Immunogenic compositions are also encompassed by the use of the term vaccine throughout the disclosure and claims.


V. Methods for Producing a Recombinant VSV Particles

Methods for producing a recombinant VSV particle may comprise introducing into cells a recombinant VSV vector of present disclosure such that a recombinant VSV particle is produced. Alternatively, the methods may comprise infecting cells with a recombinant VSV particle of the present disclosure, and then producing a recombinant VSV particle. In some embodiments, the methods further comprise expressing a SARS-CoV-2 S protein or an immunogenic variant thereof, wherein the SARS-CoV-2 S protein or immunogenic variant thereof may be presented on the surface of the recombinant VSV particle. Details on the recombinant VSV particle are provided in Section II.A.


The cells that may be used in a variety of embodiments are Vero cells, human 293 cells, MRC-5 cells, Vero E6 cells, Huh7 cells, DBT cells, Calu-3 2B4 cells or primary human airway epithelial cells. In some embodiments, the cell is a Vero cell.


The methods may further comprise purifying the recombinant VSV particle from the cells. The methods may further comprise purifying the SARS-CoV-2 S protein or immunogenic variant thereof from the cells. The methods may also comprise purifying the recombinant VSV vector from the cells.


A method of producing an adaptive mutant of the recombinant VSV particle of the present disclosure is provided. The term “adaptive mutation” used herein refers to a process that produces mutations in a virus or viral particle in response to one or more nonlethal selections and the resulting adaptive mutant virus or viral particle may have an improved biological property (e.g., replication capacity). For example, one or more mutations in a recombinant VSV particle (e.g., VSVΔG-SARS-CoV-2 chimera) may occur due to a change in one or more environmental factors (e.g., temperature, host cells and culture medium), which may reduce replication capacity of the recombinant VSV particle. The adaptive mutations may include at least one amino acid modified, for example, deleted, inserted, or replaced, and can be detected by conventional technologies. An adaptive mutant of the recombinant VSV may exhibit greater replication capacity than the recombinant VSV by, for example, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150% or 200%.


The method of producing an adaptive mutant may comprise replicating the recombinant VSV particle in a cell culture, changing a condition of the cell culture, identifying a mutant of the recombinant VSV particle exhibiting a better desirable property than the recombinant VSV particle. As result, an adaptive mutant of the recombinant VSV particle is obtained. The desirable property may be increased immunogenicity, as evidenced by, for example, greater replication capacity and/or increased expression of the SARS-CoV-2 S protein or an immunogenic fragment thereof. The condition may be chosen from of temperature, culture medium and cell substrates. For example, the temperature of the cell culture may be changed from a range of 30-36° C. to a range of 38-40° C. The culture medium may be changed from complete medium containing 10% fetal bovine serum to complete medium containing lower quantities of serum like 2% or to alternative mediums like serum-free growth medium. Where the desirable property is replication capacity, the adaptive mutant may exhibit at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150% or 200% greater replication capacity than the recombinant VSV particle. The adaptive method may further comprise subjecting the recombinant VSV particle to mutagenesis prior to growing the recombinant VSV particle in the cell culture. The mutagenesis may be chemical.


VI. Recombinant Cells

In many embodiments, a recombinant cell comprises a recombinant VSV vector and/or a recombinant VSV particle. In some embodiments, the recombinant cell produces a recombinant VSV particle. A SARS-CoV-2 S protein or a variant thereof is expressed on the surface of the recombinant VSV particle. Cells that may be used as recombinant cells are chosen from Vero cells, human 293 cells, MRC-5 cells, Vero E6 cells, Huh7 cells, DBT cells, Calu-3 2B4 cells, and primary human airway epithelial cells. In many embodiments, the recombinant cell is a Vero cell.


VII. Methods of Generating an Immune Response Against SARS-CoV-2

In many embodiments, an effective amount of a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of the present disclosure is administered in vivo, for example, to produce an immune response in a subject. The recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine may be used as one or more components of a prophylactic or therapeutic vaccine against SARS-CoV-2 for the prevention, amelioration or treatment of COVID-19. The term “immune response” used herein refers to any immune response, including but not limited to a humoral response, a cellular antigen-specific immune response, or a combination thereof. A “subject” in the context of the present disclosure may be any animal, for example, a human. In some embodiments the subject is a human, for example, a human that is infected with, or is at risk of infection with, SARS-CoV-2.


In some embodiments, the immune response may block infection by SARS-CoV-2 of a subject administered the SARS-CoV-2 S protein or an immunogenic variant thereof by, for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or 100%, as compared with that of a control subject not administered the SARS-CoV-2 S protein or an immunogenic variant thereof. Generation of an immune response includes both immune responses generated after one dose, as well as immune responses generated after two or more doses. Thus, an immune response may occur after administration of a first vaccine or a first vaccine and a second vaccine.


Methods of generating an immune response against SARS-CoV-2 may comprise (1) administering to the subject an effective amount of the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of the present disclosure; and (2) generating an immune response to SARS-CoV-2. In some embodiments, the immune response is generated in the subject without causing a disease or symptom associated with the SARS-CoV-2 and the subject is vaccinated. A disease or symptom associated with the SARS-CoV-2 may be fever, cough, shortness of breath, or gastrointestinal symptoms.


The immune response may comprise a humoral response, a cellular antigen-specific immune response or a combination thereof. Anti-SARS-CoV-2 antibodies may be isolated from a COVID-19 or produced by immunizing a subject with a SARS-CoV-2 epitope. Where the immune response comprises production of antibodies by the subject that block SARS-CoV-2 infection, the methods may further comprise harvesting the antibodies from the vaccinated subject, and optionally mixing the harvested antibodies with a pharmaceutically acceptable excipient to make a pharmaceutical composition. In some embodiments, the antibodies are in the form of immune serum obtained from the vaccinated subject. The antibodies may be in the form of immune serum obtained from vaccinated subjects or monoclonal antibodies prepared from SARS-CoV-2-specific B cells obtained from vaccinated subject.


When provided prophylactically, the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine of the present disclosure are ideally administered to a subject in advance of SARS-CoV-2 infection or evidence of SARS-CoV-2 infection, or in advance of any symptom due to COVID-19, including high-risk subjects. The prophylactic administration of the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine can serve to provide protective immunity of a subject against SARS-CoV-2 infection or to prevent or attenuate the progression of COVID-19 in a subject already infected with SARS-CoV-2.


When provided therapeutically (i.e., after the subject is exposed to SARS-CoV-2 and either before or after the onset of symptoms), the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine can serve to ameliorate and treat COVID-19 symptoms and are advantageously used as soon after infection as possible. In some embodiments, the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine are provided before appearance of any symptoms of COVID-19 but may also be used at (or after) the onset of the disease symptoms. In some embodiments, the method further comprises preventing or inhibiting binding of SARS-CoV-2 to the receptor. In some embodiments, the receptor is angiotensin-converting enzyme 2 (ACE2).


In some embodiments, the methods may comprise administering the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine to a tissue in the subject that expresses a receptor for SARS-CoV-2, that may comprise ACE2. These methods may prevent or inhibit SARS-CoV-2 from binding to the receptor after the subject is exposed to the SARS-CoV-2. The target tissue may be in the gastrointestinal tract, such as the small intestine, of the subject.


A. Methods of Administration


Any suitable method may be used to administer the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine, including but not limited to for oral (including oral mucosal, buccal, and/or gastrointestinal) sublingual, intramuscular, intradermal, subcutaneous, intranasal (including nasal mucosal), intraocular, rectal, transdermal, mucosal, topical, intravenous, or parenteral delivery methods. Other delivery methods of DNA to animal tissue have been achieved by cationic liposomes (Watanabe et al., (1994) Mol. Reprod. Dev. 38:268-274; and WO 96/20013), direct injection of naked DNA into animal muscle tissue (Robinson et al., (1993) Vaccine 11:957-960; Hoffman et al., (1994) Vaccine 12: 1529-1533; Xiang et al., (1994) Virology 199: 132-140; Webster et al., (1994) Vaccine 12: 1495-1498; Davis et al., (1994) Vaccine 12: 1503-1509; and Davis et al., (1993) Hum. Mol. Gen. 2: 1847-1851), or intradermal injection of DNA using “gene gun” technology (Johnston et al., (1994) Meth. Cell Biol. 43:353-365). Alternatively, delivery routes can be oral, intranasal or by any other suitable route. Delivery also be accomplished via a mucosal surface such as the anal, vaginal, or oral mucosa.


In some embodiments, the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is administered orally or oral mucosally. Oral or oral mucosal administration of the vaccine would greatly simplify the production, formulation, packaging and delivery of the vaccine and thus greatly facilitate the implementation and reach of vaccination programs worldwide. Notably, this method can simplify mass vaccination by allowing use of needle-free devices. In addition, the vaccine compositions of the present disclosure administered mucosally require less extensive purification simplifying vaccine manufacturing. Lastly, mucosal vaccine composition of the present disclosure is known to stimulate greater protective immunity at mucosal barriers where most infectious pathogens gains access to the host. Accordingly, one embodiment of the present disclosure is a vaccine composition that is suitable for oral or oral mucosal vaccination. Oral or oral mucosal administration of this vaccine is supported by the high-level expression of ACE2 and Furin (which facilitate viral infection) at mucosal surfaces including those of the oropharyngeal cavity. In yet another embodiment, the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is administered orally or oral mucosally so that may target gastrointestinal mucosa.


In some embodiments, the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is formulated for oral administration. In some embodiments, the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is formulated for intranasal administration. In some embodiments, the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine formulated for oral mucosal administration and intranasal administration combined (a dual route of administration).


In some embodiments, administering the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises intranasal administration. In some embodiments, administering the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises oral administration. In some embodiments, administering the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises oral mucosal and intranasal administration combined (a dual route of administration).


B. Subjects


The subject may be a male or female. The subject may be of all ages. The subject may be younger than 10, 10 or older than 10, 20, 30, 40, 50, 60, 70 or 80 years old. The subject may be 0-10, 10-20, 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90 or 90-100 years old. The subject may have a pre-existing medical condition, including without limitation, serious chronic medical condition. The pre-existing medical condition may be chosen from asthma, blood or bone marrow transplant, cancer, cardiomyopathy, cerebrovascular disease, chronic kidney disease, chronic obstructive pulmonary disease, coronary artery disease, cystic fibrosis, diabetes (including type 1 diabetes mellitus and type 2 diabetes mellitus), heart disease, heart failure, HIV, hypertension (high blood pressure), immune deficiency, immunocompromised state from solid organ transplant, liver disease, lung disease, neurologic conditions, obesity, pregnancy, pulmonary fibrosis, sickle cell disease, smoking, thalassemia, use of corticosteroids, use of immune weakening medicines, and combinations thereof.


C. Immunization Doses, Schedules and Regimens


In some embodiments, a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is provided in an effective amount for inducing an immune response in a subject wherein at least one dose is administered. In some embodiments, a vaccine comprises a recombinant VSV vector, recombinant VSV particle, or immunogenic recombinant protein in an effective amount for inducing an immune response against the SARS-CoV-2 S protein in a subject wherein at least one dose is administered.


As used herein, the term “dose” means a quantity (of, for example, a VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine) taken or recommended to be taken at a particular time. As used herein, the term “single dose” refers requiring only one administration to induce an immune response. As used herein, the term “multiple doses” refers requiring more than one dose to induce an immune response. In some embodiments, the dose or doses provide protection from disease. Suitable doses may be selected based on studies on safety, tolerability, immunogenicity, animal challenge, dose-response, and/or literature-based modeling.


While dose can vary depending on the route of administration and the size of the subject, suitable doses can be determined by those of skill in the art, for example, by measuring the immune response of a subject, such as a laboratory animal, using conventional immunological techniques, and adjusting the dosages as appropriate. Such techniques for measuring the immune response of the subject include but are not limited to, chromium release assays, tetramer binding assays, IFN-γ ELISPOT assays, IL-2 ELISPOT assays, intracellular cytokine assays, and other immunological detection assays, e.g., as detailed in the text “Antibodies: A Laboratory Manual” by Ed Harlow and David Lane.


In some embodiments, a dose may comprise an effective amount in the range of 1×103 to 1×109 plaque forming units as measured by standard methods. One example of an effective amount could include approximately 1×104, 1×105, 1×106, 1×107, or 1×108 plaque-forming units.


In some embodiments, dose levels for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine may range from 1.0×106 to 3.8×108 plaque forming units. In some embodiments, a lowest dose level for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine (which may comprise rVSVΔG-SARS-CoV-2 clone MB1 or rVSVΔG-SARS-CoV-2 clone MB2) is 1.0×106. In some embodiments, a lower middle dose level for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine (which may comprise rVSVΔG-SARS-CoV-2 clone MB1 or rVSVΔG-SARS-CoV-2 clone MB2) is 3.8×106. In some embodiments, an upper middle dose level for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine (which may comprise rVSVΔG-SARS-CoV-2 clone MB1 or rVSVΔG-SARS-CoV-2 clone MB2) is 1.5×107. In some embodiments, a higher dose level for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine (which may comprise rVSVΔG-SARS-CoV-2 clone MB1) is 5.6×107. In some embodiments, a highest dose level for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine (which may comprise rVSVΔG-SARS-CoV-2 clone MB1) is 3.8×108.


In some embodiments, dose levels for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine (which may comprise rVSVΔG-SARS-CoV-2 clone MB1 or rVSVΔG-SARS-CoV-2 clone MB2) may range from 1.0×106 to 3.8×108, from 1.0×106 to 5.0×107, or from 2.0×106 to 2.0×107 plaque forming units.


In some embodiments, dose levels for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine (which may comprise rVSVΔG-SARS-CoV-2 clone MB1 or rVSVΔG-SARS-CoV-2 clone MB2) may range from 1.0×106 to 3.8×106 plaque forming units. In some embodiments, dose levels for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine (which may comprise rVSVΔG-SARS-CoV-2 clone MB1 or rVSVΔG-SARS-CoV-2 clone MB2) may range from 3.8×106 to 1.5×107 plaque forming units. In some embodiments, dose levels for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine (which may comprise rVSVΔG-SARS-CoV-2 clone MB1 or rVSVΔG-SARS-CoV-2 clone MB2) may range from 1.5×107 to 5.6×107 plaque forming units. In some embodiments, dose levels for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine (which may comprise rVSVΔG-SARS-CoV-2 clone MB1) may range from 5.6×107 to 3.8×108 plaque forming units.


In some embodiments, a dose level for a recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine is chosen from 5.0×105, 2.4×106, 1.15×107, and 5.55×107 plaque forming units.


The recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine may be administered in a single dose, in two doses, or in multiple doses. The immunization regimes may comprise 1 to 6 doses, but may have as few as one or 2 or 4. Two or multiple doses may be administered sequentially with days, weeks, months or years apart. In some embodiments, a set time interval between doses may range from 10 days to several weeks, and may be 2, 4, 6 or 8 weeks. For example, two doses may be administered at approximately 1, 2, 3, 4 weeks, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 months, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 years apart. For humans, the interval is typically from 2 to 6 weeks.


The first dose may be for a priming immunization and the second dose or follow-up dose(s) may be a boosting immunization. Each dose shall have an effective amount of the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine as described herein.


Immunizations can also include administration of an adjuvant with the VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine. In some embodiments, the methods comprise annual, biannual or other long interval (e.g., 5-10 years) booster immunizations which serve to supplement the initial immunization protocol.


Immunizations may include a variety of prime-boost regimens, for example, DNA prime-Adenovirus boost regimens. In many embodiments, one or more priming immunizations are followed by one or more boosting immunizations. The vaccine or pharmaceutical composition can be the same or different for each immunization and the type of immunogenic composition (e.g., containing recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine), the route, and formulation of the immunogens can also be varied. For example, if an expression vector is used for the priming and boosting steps, it can either be of the same or different type (e.g., DNA or bacterial or viral expression vector). One useful prime-boost regimen provides for two priming immunizations, four weeks apart, followed by two boosting immunizations at 4 and 8 weeks after the last priming immunization. It should also be readily apparent to one of skill in the art that there are several permutations and combinations that are encompassed using the DNA, bacterial and viral expression vectors of the disclosure to provide priming and boosting regimens.


In some embodiments, at least one priming dose is administered and at least one boosting dose is administered, wherein the doses can be the same or different, provided that at least one of the doses comprises recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine, and wherein a sufficient amount was administered in each dose to induce a SARS-CoV-2-specific immune response in the subject. The SARS-CoV-2-specific immune response can include a SARS-CoV-2-specific T-cell immune response or a SARS-CoV-2-specific B-cell immune response. In some embodiments, the immunizations can be done at intervals, such as at least 0-29 or more weeks.


In instances where at least a portion of the VSV-G protein is retained, the prime-boost regimen can also include VSV vectors that derive their G protein from different serotype vesicular stomatitis viruses (Rose N F, Roberts A, Buonocore L, Rose J K. Glycoprotein exchange vectors based on vesicular stomatitis virus allow effective boosting and generation of neutralizing antibodies to a primary isolate of human immunodeficiency virus type 1. J Virol. 2000 December; 74(23):10903-10). The VSV vectors used in these examples contain a G protein derived from the Indiana serotype of VSV. Vectors can also be constructed to express epitopes in the context of G molecules derived from other VSV serotypes (i.e. vesicular stomatitis New Jersey virus or vesicular stomatitis Alagoas virus) or other vesiculoviruses (i.e. Chandipura virus, Cocal virus, Isfahan virus).


The recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine can be administered alone, or can be co-administered, or sequentially administered, with other SARS-CoV-2 immunogens and/or SARS-CoV-2 immunogenic compositions, e.g., with “other” immunogenic, antigenic or vaccine or therapeutic compositions thereby providing multivalent or “cocktail” or combination compositions of the disclosure and methods of employing them. Again, the ingredients and manner (sequential or co-administration) of administration, as well as dosages can be determined taking into consideration such factors as the age, sex, weight, species and condition of the particular subject, and the route of administration.


EXAMPLES
Example 1. VSVΔG-SARS-CoV-2 Genomic Clones

VSVΔG-SARS-CoV-2 (also referred to as rVSVΔG-SARS-CoV-2) genomic clones have been generated by replacing VSV-G gene in VSV with a nucleotide sequence encoding wild-type SARS-CoV-2 S protein or a variation thereof. The resulting VSVΔG-SARS-CoV-2 chimeric viruses exhibited substantially diminished replicative capacity. To obtain replication-competent VSVΔG-SARS-CoV-2 chimeric viruses, adaptive mutants having rescued replicative capacity were obtained by infecting cells with the VSVΔG-SARS-CoV-2 chimeric viruses. The replication-competent VSVΔG-SARS-CoV-2 clone isolates have been found to contain novel mutations in the SARS-CoV-2 S protein and used to select two vaccine candidates: rVSVΔG-SARS-CoV-2 clones MB1 and MB2.


A gene encoding a wild-type SARS-CoV-2 S protein or a variant thereof was cloned into the rVSV genetic background as described by Lawson et al. (PNAS 92, 4477-4481 (1995)) used for development of the rVSVΔG-ZEBOV-GP chimera as described by Garbutt et al. (J Virol 78, 5458-5465 (2004)) and Jones et al. (Nature medicine 11, 786-790 (2005)) (FIG. 4).


The nucleotide sequence for the SARS-CoV-2 S protein, wild-type or a variant thereof (FIG. 5), was synthesized at GenScript, Inc. and cloned into a VSV genomic plasmid in the position normally occupied by the VSV-G gene. The nucleotide sequences of the resulting VSVΔG-SARS-CoV-2 genomic clones were confirmed prior to their use for virus rescue.


For example, the wild-type SARS-CoV-2 S protein sequence was from SARS-CoV-2 isolate USA-WA1 (accession number MN985325). The resulting VSVΔG-SARS-CoV-2 genomic clone, Construct 1 with wild-type S protein, was successfully used in cell culture to rescue replication-competent rVSVΔG-SARS-CoV-2 having adaptive mutations in the SARS-CoV-2 S protein.


Example 2. Cell Culture

Vero cells (WHO 10-87) were used to cultivate VSVΔG-SARS-CoV-2 vectors as described in this application. The Vero Working Cell Bank (WCB Lot 117-10004) was produced under GMP conditions at our manufacturer, SAFC Pharma (Carlsbad, Calif.).


Vero cells generated from the WCB were cultured in DMEM (#12430-047) supplemented with 10% heat-inactivated Fetal bovine serum (SAFC #12103C) plus 2 mM L-glutamine, 1 mM sodium pyruvate, and 0.1 mM MEM nonessential amino acids (all obtained from Thermo Fisher unless specified). The cells were maintained in incubators at 37° C., 5% CO2 and 85% Humidity.


The cell density at the time of infection was 1.20E+07 viable cells in T175 flask.


Example 3. Virus Rescue

rVSVΔG-SARS-CoV-2 rescue was conducted using Vero cells as illustrated in FIG. 6. In brief, Vero cells were electroporated with one of the rVSVΔG-SARS-CoV-2 genomic clones plus a blend of six ‘supporting’ plasmids that express the five VSV structural proteins needed to initiate the virus replication cycle [nucleoprotein (N); phosphoprotein (P); matrix (M); glycoprotein (G); large protein (L) or viral RNA-dependent RNA polymerase] as well as bacteriophage T7 RNA polymerase (Witko et al., J Virol Methods 135, 91-101 (2006); Rabinovich et al., PLoS ONE 9, e106597 (2014)). Conditions for electroporation with BTX830 apparatus was described by Rabinovich et al. (PLoS ONE 9, e106597 (2014)).


After electroporation, the cells were plated in the medium of 10% FBS to rest in the incubator at 37° C. with 5% CO2, 85% Humidity for two hours after which the cultures were subjected to heat shock at 42° C., 5% CO2 and 85% Humidity for another 2 hours. Following the heat shock, the cells were incubated for three days at 37° C. with 5% CO2, 85% humidity.


Example 4. Serial Passages in Vero Cells to Derive a Recombinant Virus that Propagates to Higher Titers

Rescue of rVSVΔG-SARS-CoV-2 virus is challenging because growth of a virus expressing an exogenous S protein is substantially attenuated. To overcome this barrier, supernatant collected from a rescue flask containing the rVSVΔG-SARS-CoV-2 virus was passaged one time on the Vero cells transiently expressing VSV-G to enhance the initial growth of recombinant virus. Following this first round of virus amplification, the recombinant virus was passaged additional times in Vero cell monolayers without providing VSV-G complementation. As mentioned above, adaptation by serial passage allows isolation of a genetically stable virus with improved growth characteristics (FIGS. 7, 8 and 14).


Vero cell propagation and infection were conducted using growth medium containing 10% FBS. Following the additional serial passages described above, virus harvest in the medium was used for clonal isolation by plaque purification. All samples are characterized by assays listed on Table 2.









TABLE 2





Characterization Assays
















1
Infectivity titer on Vero cells by plaque assay


2
Whole genome sequence determined by Sanger Sequencing


3
S glycoprotein expression by Western blot


4
Modeling of sequence variants on SARS-CoV-2 S protein structure


5
SARS-CoV-2 pseudoneutralization (PRNT) assay


6
hACE2-human cell line infectivity assay


7
Ig-ACE2 flow virometry









Example 5. Plaque Purification and Characterization

Plaque purification was executed by picking multiple well-isolated plaques after which each plaque was amplified in T-25 flasks to generate virus stocks. Each plaque isolate was then analyzed by the characterization assays described in Table 2.


Plaque isolation was performed using two related procedures. In the first procedure, plaques were picked from monolayers cultured in growth medium supplemented with FBS after which the virus was amplified in the presence of serum. In the second procedure, the Vero cell monolayer was cultured in serum-free medium (VP-SFM). rVSVΔG-SARS-CoV-2 clone MB1 (nucleic acid sequence SEQ ID NO: 150 or amino acid sequences SEQ ID NOS: 151-153, 155, and 156) is a virus clone isolated using VP-SFM. rVSVΔG-SARS-CoV-2 clone MB2 (nucleic acid sequence SEQ ID NO: 154 or amino acid sequences SEQ ID NOS: 152, 155-158) is a virus clone prepared using medium supplemented with FBS (described in Example 7 below).


Example 6. Amplification of rVSVΔG-SARS-CoV-2 Clones MB1 and MB2 to Generate Two Leading Vaccine Candidates

Two clone isolates, rVSVΔG-SARS-CoV-2 clone MB1 (nucleic acid SEQ ID NO: 150 or amino acid sequences SEQ ID NOS: 151-153, 155, and 156) and rVSVΔG-SARS-CoV-2 clone MB2 (nucleic acid SEQ ID NO: 154 or amino acid sequences SEQ ID NOS: 152 and 155-158), were selected as vaccine candidates. Expansion of these two clone isolates was conducted using Vero cells cultured in VP-SFM. Both pre-master virus seed (pre-MVS) vaccine candidates were passaged in Vero cells to ensure no further genomic changes occurred during subsequent processing. rVSVΔG-SARS-CoV-2 clone MB1 was passaged an additional five passages. rVSVΔG-SARS-CoV-2 clone MB2 was passaged an additional three passages. rVSVΔG-SARS-CoV-2 clone MB1 was prioritized for GMP manufacturing of the Master Virus Seed (MVS) (FIGS. 12 and 13A-B).


Example 7. Virus Infectivity Titers Measured by Plaque Assay

We set out to identify at least one clone that provided a high virus titer by testing over 44 clone isolates for infectivity. A higher titer will enable higher doses, which may enable better infectivity and a more robust immune response in a subject. Virus infectivity titers of clone isolates were measured over time after cell infection by a standard immunoplaque assay. The clones were isolated and grown at serum-containing medium or in serum-free medium. Pre-master seed viral stocks were generated for each clone. Vero cells were used to provide data that will support vaccine manufacturing. Ten-fold dilutions of the viral stock were prepared using 2% FBS as diluent and 0.5 mL of the serial dilution was inoculated on the Vero cell monolayer in each well of 6-well plate. After one-hour incubation, the inoculums were removed, and the Vero cells were overlayed with 0.8% Agarose plus 2% FBS. Plaques became visible from 24 to 72 hours post infection. Cells were then fixed with 7% formaldehyde adding on top of the agarose for 30 minutes. After the cells were fixed, the agarose overlays were removed to allow the monolayer to be stained by Crystal violet. The infectivity titers were expressed as plaque-forming units per ml (PFU/mL) and calculated by the average of at least two replicates. Using this first method, we measured infectivity over time for Construct 1 (rescued using a plasmid encoding the wild-type S protein); infectivity titers improved from 4.00E+05 pfu/mL to 2.00E+07 PFU/mL with increasing rounds of passaging (FIG. 7).


A second method was also used to measure infectivity titers. Vero cells were plated, incubated at 37±1° C. and 5±1% CO2, and later checked for confluency. A dilution box was prepared as shown in Table 3. Samples were thawed at ambient temperature in a Biosafety Cabinet and the positive control was thawed at 37±1° C. in a water bath. Samples and the positive control were diluted according to Table 3.









TABLE 3







Dilution Box Set-Up for Drug Substance and PC Sample










Dilutions
Dilution Scheme







1st Dilution
1:10-900 μL dilution medium +




100 μL sample



2nd Dilution
1:100-900 μL dilution medium +




100 μL previous dilution (10)



3rd Dilution
1:4000-975 μL dilution medium +




25 μL previous dilution (100)



4th Dilution
1:8000-500 μL dilution medium +




500 μL previous dilution (4K)



5th Dilution
1:16000-500 μL dilution medium +




500 μL previous dilution (8K)



6th Dilution
1:32000-500 μL dilution medium +




500 μL previous dilution (16K)










Plates of Vero cells were removed from the 37±1° C., 5+1% CO2 incubator for media removal. Media was removed by gentle decanting. We used an absorbent mat to remove residual media by gently bringing the mat in contact with the plate. To avoid drying the monolayer of Vero cells, the plates of cells were decanted only one at a time. Each well of cells was inoculated with 150 μL of each dilution in the plate. Each plate was rocked ensure that each inoculum covered the entire well. Plates were placed back into the 37±1° C., 5±1% CO2 incubator for approximately 1 hour to allow for attachment. In instances where the number of plates being assayed is very large, the plates were assayed in batches. During the attachment time period, plates were rocked manually at 30±5 minute intervals to ensure even distribution of the samples in the wells and to prevent the cells from drying out.


After the 1 hour attachment time period, the plates were removed from the incubator and 1 mL of overlay was added to each well. Plates were handled very gently and with caution to avoid disturbing the monolayer of Vero cells, such as touching pipet tips of the multichannel pipet to the side of each well while pipetting so as to avoid unnecessary turbulence. The plates were left to cool in the Biosafety Cabinet for 15 to 30 minutes or until the overlay had firmed. After that, the plates were placed back in the incubator for 24±1 hours until staining.


For staining, plates were removed from the incubator and 200 μL of formalin was added to each well. The cells were incubated with formalin at room temperature for 60±10 minutes. The wells were decanted by gently inverting the plates and tapping to remove plugs. 200 μL of 1% crystal violet solution was applied to each well. After 3 to 5 minutes, the plates were submerged in water for rinsing. The plates were submerged three times or more times, until we observed no more stain coming off in the rinse. The plates were then placed on absorbent pads and left to dry at ambient temperature. The plaques were then counted to determine titer. The titer for each clone is shown in Table 4.









TABLE 4







Titers from Immunoplaque (Infectivity) Assay











Source
Clone
Geomean Infectious Titer















Round 1
1
1.6E+06




2
1.5E+06




3
1.6E+06




4
1.9E+06




5
2.0E+06




6
2.1E+06




7
1.5E+06




8
1.9E+06




9
2.1E+06




10
2.3E+06




11
2.1E+06




12
1.9E+06




13
2.4E+06




14
2.4E+06




15
2.0E+06




16
1.3E+06




17
2.2E+06




18
2.1E+06




19
2.0E+06




20
1.7E+06



Plaque isolates
SC1
4.5E+05



from serum-
SC3
4.3E+05



containing
SC7
2.7E+05



medium
MB2
1.6E+05



Plaque isolates
SF1
7.4E+04



from serum-free
SF2
3.24E+03 



medium
SF3
4.95E+05 




SF4
1.95E+03 




SF5
3.25E+04 




SF6
9.97E+04 




SF7
5.80E+04 




SF8
6.99E+03 




SF9
1.4E+04




SF10
2.85E+04 




SF11
1.05E+06 




SF12
3.91E+05 




SF13
3.58E+05 




SF14
1.42E+05 




SF15
1.26E+05 




SF16
1.51E+05 




SF17
9.62E+03 




SF18
8.51E+04 




SF19
5.51E+04 




MB1
1.80E+06 










We then measured infectivity over time for clones SC1, SC3, SC7, and MB2 from the serum-containing medium and clone MB1 from the serum-free medium. PFU/mL for clones SC1, SC3, SC7, and MB2 at 53 and 67 hours are shown in FIG. 11. PFU/mL for clones MB1 and MB2 at 28, 42, 54, 65, and 72 hours are shown in FIG. 12. All five clones also showed improved infectivity titers over time (FIGS. 11 and 12).


Example 8. Whole Genome Sequencing

A. Materials and methods


We set out to determine the purity and stability of each clone, which included determining that there were no changes to attenuating mutations in VSV. The whole genome of virus clone isolates were sequenced.


The Sanger sequencing method was used to determine the nucleotide sequences of the clone isolates. Viral RNA of each clone isolate was isolated by RNA extraction kit from Qiagen (#52906). One-step RT-PCR (SuperScript III) reactions were conducted to reverse transcribe and amplify the cDNAs cross the VSV genome. Eight overlapping cDNA fragments were generated to cover the 14 kb RNA genome. Each cDNA was 2 to 3 kb in length for further Sanger sequencing. Data was analyzed using DNASTAR SeqMan Pro, MegAlign, Vector NTi and ABI Seq Scanner2. Table 5 lists the primers used for sequencing the eight overlapping cDNA fragments in RT-PCR reactions N, P, M, S1, S2, L1, L2 and L3. Table 6 lists the primers used for the Sanger sequencing of the whole genome of rVSVΔG-SARS-CoV-2 clone isolates.









TABLE 5







List of Primers for One-step RT-PCR










RT-PCR reaction name
Forward primer name
Reverse primer name
Expected Size(kb)













N
1gVSVLeader_f
10gVSV_1600r
1.7


P
11gVSV_1200f
18gVSV_2400r
1.3


M
19gVSV_2000f
24CoV_S_rvs1
1.1


S1
25gVSV_2400f
36CoV_S_rvs6
2.5


S2
37CoV_S_fw5
49gVSV_4800r
2.5


L1
50CoV_S_fw10
59gVSV_6200r
2.7


L2
60gVSV_6300f
70gVSV_8200r
3.0


L3
71gVSV_8300f
82JRVSVTrailer_r
3.0
















TABLE 6







List of Primers Used for the Sanger


Sequencing of the rVSVΔG-SARS-CoV-2 Genome













SEQ



Primer name
Sequence
ID NO:






1gVSVLeader_f
ACGAAGACAAACAAACCATT
 68




ATTATCATTAAAAGG







2gVSV_400f
AGCAGGGGATACAATCGGAA
 69




TA







3gVSV_800f
CACCTCTGCAAAATAACCGG
 70




AA







4gVSV_1200f
GCAGAGATGTGGTCGAATGG
 71




C







5gVSV_1600f
TGAAGGCTTTATACAGGGGC
 72






6gVSV_200r
TGGTAGACATATCCTCTTAG
 73






7gVSV_400r
GCTGGTTCTGGAAGCATCCG
 74






8gVSV_800r
CTGGAAGCATCATTTGGACC
 75




ATTT







9gVSV_1200r
GCTCTTTTCGCATACTGCAT
 76




CATA







10gVSV_1600r
CCACTTCCTCATCTGCATAG
 77




TCAT







11gVSV_1200f
GCAGAGATGTGGTCGAATGG
 78




C







12gVSV_1600f
TGAAGGCTTTATACAGGGGC
 79






13gVSV_2000f
TCCATGACTTTCCAACCCAA
 80




GA







14gVSV_2400f
CGAGATGGACACCTATGATC
 81




CGAA







15gVSV_1200r
GCTCTTTTCGCATACTGCAT
 82




CATA







16gVSV_1600r
CCACTTCCTCATCTGCATAG
 83




TCAT







17gVSV_2000r
CTCCGACAGAGATGAACTCT
 84




CCTC







18gVSV_2400r
GCTGCCACATCTGAGTATGT
 85




TCTG







19gVSV_2000f
TCCATGACTTTCCAACCCAA
 86




GA







20gVSV_2400f
CGAGATGGACACCTATGATC
 87




CGAA







21gVSV_2800f2
CTACGATGATGAGTCACTGG
 88






22gVSV_2400r
GCTGCCACATCTGAGTATGT
 89




TCTG







23gVSV_2800r
CCTTTTTCTCGACAATCAGG
 90




CC







24CoV_S_rvs1
CTCTTTGTTCCATTGGTTCC
 91






25gVSV_2400f
CGAGATGGACACCTATGATC
 92




CGAA







26CoV_S_fw1
AGTGGATCCTACTCGAGAGG
 93






27CoV_S_fw2
CATTAAAGTCTGTGAATTCC
 94






28CoV_S_fw3
AGCAGCTGCTTACTATGTTG
 95






29CoV_S_fw4
GTATATGCAGATTCCTTTGT
 96






30CoV_S_fw5
ACCTAAGAAGAGCACTAATC
 97




T







31CoV_S_rvs1
CTCTTTGTTCCATTGGTTCC
 98






32CoV_S_rvs2
GATCTCTAACTAGGTTTATA
 99




GGTGT







33CoV_S_rvs3
AGTAGCATTGAAAACCTCTC
100






34CoV_S_rvs4
CATTACACGGTGTGCTGCCC
101






35CoV_S_rvs5
GATAGAGAACGGCAACTTGA
102






36CoV_S_rvs6
GGAATCTCCACAGATGTACA
103






37CoV_S_fw5
ACCTAAGAAGAGCACTAATC
104




T







38CoV_S_fw6
TGACATTCCAATCGGTGCCG
105






39CoV_S_fw7
CCATCAAGGATTTTGGAGGA
106




TTTAA







40CoV_S_fw8
GCTATAGGAAAGATTCAGGA
107






41CoV_S_fw9
ACATGTAACATATGTGCCCG
108






42CoV_S_fw10
TGATCGACCTTCAGGAGCTG
109






43CoV_S_rvs7
TGTAATTGTACCAGCCAACA
110






44CoV_S_rvs8
TGATGAGTTGTTGGGTGACG
111






45CoV_S_rvs9
GGAATCGAGTTCAGGTTGTA
112






46CoV_S_rvs10
AACGCGTAAACAGCTAGCCT
113






47gVSVGene4_f
TTAATGTTTGGCCTGATTGT
114




CGAGA







48gVSVGene4_rev
GCGCTCATCGGGATTCAGGA
115




A







49gVSV_4800r
CCACCACGTCAAATGTTATT
116




TC







50CoV_S_fw10
ACATGTAACATATGTGCCCG
117






51gVSV_5300f
GGGCAACAAACCAATTGAAT
118






52gVSV_5800f
GCAAGAGAATCAAGGCCTTT
119




AGT







53gVSV_6300f
CCCATCGATAATATACTCTG
120




ACAAA







54gVSV_6800f
TCCTTAATCGAGAGAACTCA
121




TGAA







55gVSV_4800r
CCACCACGTCAAATGTTATT
122




TC







56gVSV_5200r2
ACTAAAGGCCTTGATTCTCT
123




TGC







57gVSV_5200r
GATGGGTCTAGTAAGTCGGG
124




TATT







58gVSV_6200r2
TTCATGAGTTCTCTCGATTA
125




AGGA







59gVSV_6200r
CACTCGTGACCATCTCTTGG
126






60gVSV_6300f
CCCATCGATAATATACTCTG
127




ACAAA







61gVSV_6800f
TCCTTAATCGAGAGAACTCA
128




TGAA







62gVSV_7300f
TGACCAAATACCCACTTGTG
129




C







63gVSV_7800f
CGAACTTGTTAAAGACTGAG
130




GTTAA







64JKgVSV_8300f
CGGGATCCATGACGTCTTTA
131






65gVSV_8800f
TTTGAGACCCATAGAAGAGA
132




TCAC







66gVSV_6200r
CACTCGTGACCATCTCTTGG
133






67gVSV_7200r2
TTAACCTCAGTCTTTAACAA
134




GTTCG







68gVSV_7200r
TGAACCCTGATGTGTTACAT
135




GG







69gVSV_8200r2
GTGATCTCTTCTATGGGTCT
136




CAAA







70gVSV_8200r
TCGTTTCTAATTCGTCTCTA
137




ATAGG







71gVSV_8300f
CGGGATCCATGACGTCTTTA
138






72gVSV_8800f
TTTGAGACCCATAGAAGAGA
139




TCAC







73gVSV_9300f
CTCCTATCCGACAAGCAACC
140






74gVSV_9800f
GAATCAGGTTGGGCCAATTA
141






75gVSV_10300f
GAGCGAAAAGAATGCAGTAA
142




CA







76gVSV_10800f
GCTTTTGGCTGAGTTTGATG
143






77JKVSVGene6_f
AAACACAGGAATGATTGAAT
144






78gVSV_8200r
TCGTTTCTAATTCGTCTCTA
145




ATAGG







79gVSV_9200r2
TAATTGGCCCAACCTGATTC
146






80gVSV_9200r
CGCTCTCACAAATATATGTT
147




CCATA







81gVSV_10200r2
CATCAAACTCAGCCAAAAGC
148






82JRVSVTrailer_r
ACGAAGACCACAAAACCAGA
149




TAAA









The Illumina Next-Generation Sequencing method was also used to determine the nucleotide sequences of the clones. A cDNA library was constructed using the KAPA RNA HyperPrep for Kit Illumina Platforms (Roche KiK8541) with KAPA Unique Dual-Indexed Adapters (Roche KiK8727) as per the manufacturer's instructions. The quality of the library was evaluated with the 4200 TapeStation system (Agilent G2991AA) and concentration determined using the Qubit dsDNA HS Assay Kit (Q32851) on the Qubit Fluorometer. The samples were pooled and sequencing was performed on the Illumina NextSeq 500 Platform using the NextSeq 500/550 High Output v2.5 kit, 300 cycles (2_151 read length). Files were demultiplexed and adapters removed (bclfastq version 2.17.1.4).


B. Results


The sequencing results confirmed that (1) there were minimal to no sequence changes in the VSV genes; (2) the clones were pure and there was no contamination; and (3) the sequence variation in each clone was fully penetrant. The sequencing results identified variations from the template genome that was present in each clone. Sequencing results are shown in Table 7.









TABLE 7







Mutations Present in the Sequence of Each Clone









Source
Clone
Sequence variation












Round 1
1
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*non-coding a6993- VSV-L R201W



2
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*non-coding g6992GG VSV-L R201W



3
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201WVSV-L A326T



4
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



5
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



6
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



7
VSV-M A216GCoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L




D28EVSV-L R201WVSV-L Q1986R



8
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201WVSV-L A326T



9
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



10
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



11
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



12
CoV2-Swt V622ACoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



13
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



14
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt Silent K933CoV2-Swt S1261*VSV-L R201W



15
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



16
VSV-M A216GCoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



17
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



18
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



19
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W



20
CoV2-Swt H655YCoV2-Swt R682KCoV2-Swt S1261*VSV-L R201W


Plaque
SC1
non-coding c3058T CoV2-Swt R683GCoV2-Swt G1251*


isolates
SC3
CoV2-Swt H655YCoV2-Swt Silent L864CoV2-Swt G1251*VSV-L K189Qnon-coding


from serum-
SC7
c13439A


containing
MB2
CoV2-Swt H655YCoV2-Swt S1261*VSV-L Silent E2VSV-L T561A


medium

CoV2-Swt H655YCoV2-Swt G1251*VSV-L Silent I1343


Plaque
SF1
VSV-M E213KCoV2-Swt S1261*


isolates
SF2
VSV-M E213KCoV2-Swt N978KCoV2-Swt S1261*


from serum-
SF3
VSV-P E67DCoV2-Swt H655YCoV2-Swt S940GCoV2-Swt G1251*VSV-L Silent


free

V1701VSV-L N1849D


medium
SF4
non-coding c3058T CoV2-Swt R683GCoV2-Swt G1251*



SF5
VSV-M A186Snon-coding c3058T CoV2-Swt R683GCoV2-Swt G1251*



SF6
VSV-M E213KCoV2-Swt N709SCoV2-Swt G1251*



SF7
VSV-M E213KCoV2-Swt N709SCoV2-Swt G1251*VSV-L Y218HVSV-L Silent




D1141VSV-L Silent S1734



SF8
VSV-M E213KCoV2-Swt N709SCoV2-Swt G1251*



SF9
VSV-M E213KCoV2-Swt Silent S46CoV2-Swt N709SCoV2-Swt D1163NCoV2-Swt




G1251*non-coding a6990T VSV-L Silent N58



SF10
VSV-P R228KVSV-M G112WSV-M A219Vnon-coding c3058T CoV2-Swt




R683GCoV2-Swt D1118ACoV2-Swt G1251*



SF11
VSV-N Silent F222VSV-M Y61Snon-coding c3058T CoV2-Swt R683GCoV2-Swt




S813FCoV2-Swt G1251*



SF12
VSV-M E213KCoV2-Swt Silent Y248CoV2-Swt H655YCoV2-Swt N709SCoV2-Swt




G1251*VSV-L A1283WSV-L Silent H2067



SF13
CoV2-Swt H655YCoV2-Swt S1261*VSV-L Silent I1992non-coding t13403C



SF14
VSV-M E213KCoV2-Swt N709SCoV2-Swt G1251*



SF15
VSV-M E213KCoV2-Swt N709SCoV2-Swt G1251*



SF16
VSV-M E213KCoV2-Swt N709SCoV2-Swt G1251*



SF17
non-coding c3058T CoV2-Swt R683GCoV2-Swt D1118ACoV2-Swt G1251*non-coding




g13407T



SF18
VSV-M E213Knon-codmg t3074C CoV2-Swt N709SCoV2-Swt G1251*VSV-L C1076Y



SF19
VSV-M E213KCoV2-Swt N709SCoV2-Swt G1251*VSV-L L285S



MB1
VSV-M Y61Snon-coding c3058T CoV2-Swt R683GCoV2-Swt S813FCoV2-Swt G1251*









We did not proceed with clones that comprised the E213K mutation in the VSV-M protein as this mutation is associated with VSV attenuation. Clone MB1 comprised a Y61S mutation in the VSV-M protein, four mutations in the SARS-CoV-2 S protein—a non-coding nucleotide mutation C3058T; two amino acid substitutions R683G and S813F; and a premature stop codon at nucleotide G6852T (amino acid G1251*). The full nucleotide and amino acid sequences for clone MB1 are shown in FIGS. 24A-B. Clone MB2 comprised two mutations in the SARS-CoV-2 S protein, H655Y and G1251, and a silent I1343I mutation in the VSV-L protein. The full nucleotide and amino acid sequences for clone MB2 are shown in FIGS. 21A-G.


Example 9. Modeling Sequence Variants on SARS-CoV-2 S Protein Structure to Ensure Changes Did not Impact Critical Functional Epitopes

Given that mutations were found in the SARS-CoV-2 S protein of MB1 and MB2, we used modeling to predict the impact of the identified sequence variations on the SARS-CoV-2 S protein structure. The findings from our modeling efforts for MB1 and MB2 are described in Table 8.









TABLE 8







Modeling of Sequence Variations on SARS-CoV-2 S Protein













Surface
Type of



Mutation
Protein
exposure
change
Notes










Sequence variations in MB1











R683G
SARS-CoV-2
Disordered
Basic to
Reduces S1/S2 cleavage



S protein

nonpolar



Furin site


S813F
SARS-CoV-2
Disordered
Polar to
Very close to the S2′ cleavage



S protein

nonpolar
site (R815) where cleavage



S2/HR1


exposed the fusion peptide


Y61S
M
Yes
Nonpolar
Y61 forms a hydrogen bond





to polar
with K215; K215 mutation






has been found to make the






virus temperature sensitive







Sequence variations in MB2











H655Y
SARS-CoV-2
Yes
Basic to
Expected to promote more



S protein SD2

polar
efficient glycosylation









We found no predicted impact on the structural stability of SARS-CoV-2 S protein. There was also minimal to no sequence variation in the receptor binding domain of SARS-CoV-2 S protein or known binding sites for neutralizing antibodies to SARS-CoV-2 S protein.


The H655Y substitution occurs adjacent to a consensus site (NxS/T where x is any amino acid) in the SARS-CoV-2 S protein that can be modified by N-linked glycosylation. The H655Y substitution generates a sequence context that is expected to promote more efficient glycosylation at this location.


Example 10. rVSVΔG-SARS-CoV-2 S Protein Expression as Detected by Western Blot

We analyzed the expression of SARS-CoV-2 S protein in Vero cells were infected with the clone isolates (FIGS. 9 and 10A-C). Cells were harvested from 48 to 72-hour post infection. Cell lysates were prepared from infected cells using CelLytic M buffer (Sigma #C2978) supplemented with 1% protease inhibitors. Cell lysates were clarified by centrifugation at 13000 rpm for 5 min. To denature, sample was mixed with 4XLSD loading buffer (Thermofisher #NP0007) and 10× reducing buffer (ThermoFisher #NP0009), boiled at 80° C. for 10 minutes.


The denatured samples were subjected to SDS polyacrylamide gel electrophoresis, transferred to nictrocellulose membrane and incubate with blocking buffer (Thermofisher #37539) for 2 hours at room temperature. Monoclonal antibody recognizing the S2 subunit (GeneTex Cat #GTX632604) was used to probe the full-length S protein and the post-translationally cleaved S2 subunit respectively. Secondary antibody conjugated to HRP was used to allow the chemiluminescence detection of the specific bands.


Mutations in the Furin cleavage sites affected the full-length S protein processing to S1 and S2 subunits which could be visualized on the Western (FIGS. 9 and 10A-C). For example, both clone isolates SF1 and SF2 had the R683G mutation (FIG. 10A). The amount of the S2 protein in clone isolates SF1 and SF2 were reduced compared to the clone isolate SF3 without such mutation. In FIG. 10B, clone MB1 had full-length S protein remaining as a result of reduced S1/S2 cleavage, indicative of the R683G mutation confirmed by sequencing (see Example 8 above). In FIG. 10C clone MB2, in contrast, had complete cleavage of full-length S protein.


Example 11. PRNT Assay to Evaluate Effect of Mutations in the rVSVΔG-SARS-CoV-2 S Protein

We conducted a PRNT assay to determine if any of the mutations found in the rVSVΔG-SARS-CoV-2 S protein affected the ability of monoclonal antibodies (mAbs) in convalescent sera to neutralize the recombinant virus; convalescent sera were collected from individuals who were infected with SARS-CoV-2. The PRNT assay is described below in Example 15, Sections I and K. Effective neutralization of the candidate premaster seed virus was viewed was evidence that the recombinant virus was expressing SARS-CoV-2 S protein in an immunologically relevant conformation. The titers of each of the clones identified in the three groups (i.e., Round 1, serum-containing medium, and serum-free medium) are shown in FIG. 25. All of the recombinant viruses tested were neutralized by convalescent sera to varying degrees, including clones MB1 and MB2.


Example 12. Ability of Recombinant VSV Particles to Infect hACE2-expressing Cells

Several clone isolates were tested to ensure that the mutations in VSVΔG-SARS-CoV-2 S protein do not prevent infection of human cells. HeLa cells expressing human ACE2 (hACE2 cells) were infected with a subset of nine clones (four isolated in serum-containing medium and five identified isolated in serum-free medium; see Example 7 above). Each infected cell line was analyzed for the level viral of infection by flow virometry (i.e., viral particles were detected by flow cytometry; FIGS. 26A and B). hACE2 cells infected with rVSVΔG-SARS-CoV-2 were visualized with anti-VSV-N antibody and anti-SARS-CoV-2 S protein antibody. Double positive cells (in the upper right hand quarter of each plot) showed the relative number of positive cells. All nine clones that were tested infected hAVE2 cells to varying degrees, with clones MB1 and SF11 having the most number of infected hACE2 cells (FIG. 26B). Sequencing results confirmed that MB1 and SF11 were genetically identical.


Example 13. Ability of Recombinant VSV Particles to Bind to hACE2 and Monoclonal Antibodies

In order to infect cells, the SARS-CoV-2 S protein is required to bind to hACE2. Four clones previously identified in serum-free medium, clones SC1, SC3, SC7, and MB2 (see Example 7 above), were tested for binding to hACE2 to ensure that any adaptive mutations in VSVΔG-SARS-CoV-2 do not disrupt hACE2 binding. Recombinant virus of clones SC1, SC3, SC7, and MB2 were analyzed by flow virometry. Viral particles were visualized with anti-VSV-N antibody and anti-huACE2 Ig antibody. All four clones bound to bound to the anti-huACE2 Ig antibody, regardless of whether the Furin cleavage site was intact or not (FIG. 27). These results indicate that the recombinant viruses of clones SC1, SC3, SC7, and MB2 may infect human cells.


Example 14. Generation of rVSVΔG-SARS-CoV-2 S Clones MB1 and MB2

The process of generating rVSVΔG-SARS-CoV-2 S protein variants comprised serial passaging and nucleotide sequencing of transfected cells at key passages. Mutations may facilitate the adaptation of rVSVΔG-SARS-CoV-2 S protein to replicate in Vero E6 cells.


The variant rVSVΔG-SARS-CoV-2 clone MB1 was isolated in serum-free media and has a 23-amino acid C-terminal deletion. Due to the deletion, an endoplasmic reticulum retention sequence within the cytoplasmic domain of rVSVΔG-SARS-CoV-2 clone MB1 has been removed. rVSVΔG-SARS-CoV-2 clone MB1 also comprises the following mutations: R683G and S813F in the SARS-CoV-2 S protein, and Y61S in the VSV matrix protein. The clone MB1 sequence does not have a R685G or a 24-amino acid C-terminal deletion in the S protein.


The variant rVSVΔG-SARS-CoV-2 clone MB2 was isolated in serum-containing media and, like MB1, has a 23-amino acid C-terminal deletion. Due to the deletion, an endoplasmic reticulum retention sequence within the cytoplasmic domain of rVSVΔG-SARS-CoV-2 clone MB2 has been removed. rVSVΔG-SARS-CoV-2 clone MB2 also comprises the H655Y mutation the SARS-CoV-2 S protein, and Y61S in the VSV matrix protein. The clone MB2 sequence does not have a R685G or a 24-amino acid C-terminal deletion in the SARS-CoV-2 S protein.


Example 15. Immunogenicity of rVSVΔG-SARS-CoV-2 Clones MB1 and MB2 in a Hamster Model of SARS-CoV-2 Infection

This example describes a nonclinical pharmacology study to evaluate the immunogenicity rVSVΔG-SARS-CoV-2 clones MB1 and MB2 and to determine if it protects against SARS-CoV-2 challenge. The Golden Syrian hamster model is suitable for the assessment as SARS-CoV-2 replicates in the hamster and aspects of COVID-19 are recapitulated post-infection (Chan J F, Zhang A J, Yuan S, Poon V K, Chan C C, Lee A C, et al. Simulation of the clinical and pathological manifestations of Coronavirus Disease 2019 (COVID-19) in golden Syrian hamster model: implications for disease pathogenesis and transmissibility. epub ahead of print: Clin Infect Dis. 2020 Mar. 26; ciaa325). The study was conducted in two parts to evaluate the ability of rVSVΔG-SARS-CoV-2 clone MB1 to protect against: (A) SARS-CoV-2-induced body weight loss; and (B) viral replication. The study also evaluated both the intramuscular (IM) injection route of administration and the oral-mucosal route of administration.


A. Test Article: rVSVΔG-SARS-CoV-2 Clones MB1 and MB2


rVSVΔG-SARS-CoV-2 clone MB1 comprises (from 5′ to 3′) VSV-N, VSV-P, VSV-M with Y61S, SARS-CoV-2 S protein with R683G and S813F mutations, and VSV-L.


rVSVΔG-SARS-CoV-2 clone MB2 comprises (from 5′ to 3′) VSV-N, VSV-P, VSV-M, SARS-CoV-2 S protein with H655Y and G1251* mutations, and VSV-L.


Descriptions and sequences for rVSVΔG-SARS-CoV-2 clones MB1 and MB2 are provided in Section II.F above; and FIGS. 21A-G and 24A-B.


81 The formulation for rVSVΔG-SARS-CoV-2 clone MB1 comprised 10 mM Tromethamine (Tris) and 2.5 mg/mL rice-derived recombinant human serum albumin (rHSA).


B. Control Article: SARS-CoV-2 S Protein


The control article is the SARS-CoV-2 S protein (SARS-CoV-2 pre-S-His; no tag). The protein is adjuvanted with aluminum phosphate (2%) (AdjuPhos). The formulation comprised HEPES buffer and 2% aluminum phosphate.


C. Test Animals


Table 9 provides details on test animals used in the study. Procedures involving the care and use of animals in the study were reviewed and approved by the Institutional Animal Care and Use Committee at Merck Research Laboratories. During the study, the care and use of animals were conducted in accordance with the principles outlined in the guidance of the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC), the Animal Welfare Act, the American Veterinary Medical Association (AVMA) Euthanasia Panel on Euthanasia, and the Institute for Laboratory Animal Research (ILAR) Guide to the Care and Use of Laboratory Animals. Animals were assigned to study groups randomly, with an even gender distribution each study group.









TABLE 9





Test animals.
















Species

Mesoaicetus auratus (Golden Syrian hamster)



Sex
Male and female


Source
Envigo, Indianapolis, Indiana, USA


Number of animals used
54


Age of animals at experiment initiation
6 to 8 weeks old









D. Group Designation, Dose Levels, and Dosing Schedule


Group designation, dose levels, and dosing schedule for Part A of the study are presented in Table 10 and for Part B of the study in Table 11. The study included both IM and oral-mucosal routes of administration.









TABLE 10







Group Designation, Dose Levels, and Dosing Schedule


for Part A, Merck Study HAM-CoV-2-001














No. of

Dose

Vol.
Challenge Dose


Group
animals
Treatment
level a
ROA
(mL)
(Day) b
















1
5
None
NA
NA
NA
2 × 104








PFU SARS-








CoV-2








(Day 28 post-








vaccination)


2
5
SARS-CoV-2 S
10 μg
IM
0.1
2 × 104




Protein
(40 μg


PFU SARS-




(adjuvanted
adjuvant)


CoV-2




with



(Day 28 post-




aluminum



vaccination)




phosphate;




AdjuPhos)


3
6
rVSVΔG-SARS-
2.97 × 106
IM
0.1
2 × 104




CoV-2 clone
PFU


PFU SARS-




MB1



CoV-2








(Day 28 post-








vaccination)


4
6
rVSVΔG-SARS-
2.97 × 105
OM
0.01
2 × 104




CoV-2 clone
PFU


PFU SARS-




MB1



CoV-2








(Day 28 post-








vaccination)





IM = intramuscular(ly); μg = microgram(s); mL = milliliter(s); NA = not applicable; No. = number; OM = oral mucosal; PFU = plaque-forming unit(s); ROA = route of administration; SARS = Severe acute respiratory syndrome; SARS-CoV-2 = SARS-associated coronavirus-2.



a Nominal dose.




b The challenge inoculum was administered 0.1 mL per animal, 0.05 mL per nostril.














TABLE 11







Group Designation, Dose Levels, and Dosing Schedule


for Part B, Merck Study HAM-CoV-2-001














No. of

Dose

Vol.
Challenge Dose


Group
animals
Treatment
level a
ROA
(mL)
(Day) b
















7
4
None
NA
NA
NA
2 × 104








PFU SARS-








CoV-2








(Day 28 post-








vaccination)


8
4
rVSVΔG-
2.97 × 106
IM
0.1
2 × 104




SARS-CoV-2
PFU


PFU SARS-




clone MB1



CoV-2








(Day 28 post-








vaccination)


9
4
rVSVΔG-
2.97 × 105
OM
0.01
2 × 104




SARS-CoV-2
PFU


PFU SARS-




clone MB1



CoV-2








(Day 28 post-








vaccination)





IM = intramuscular(ly); μg = microgram(s); mL = milliliter(s); NA = not applicable; No. = number; OM = oral mucosal; PFU = plaque-forming unit(s); ROA = route of administration; SARS = Severe acute respiratory syndrome; SARS-CoV-2 = SARS-associated coronavirus-2.



a Nominal dose.




b The challenge inoculum was administered 0.1 mL per animal, 0.05 mL per nostril.







E. Study Measurements


Serum was collected on Days 7, 14, and 26 post-vaccination in both Part A and Part B, and in Part A, on Day 42 (Day 14 post-challenge). Serum was assayed for anti-SARS-CoV-2 S protein-specific IgG titers and for anti-SARS-CoV-2 PRNT titers.


In Part B, scheduled necropsies were carried out for nares and lung tissue collection on Day 32 (Day 4 post-challenge). Processed samples were assayed for viral load.


In both Part A and Part B, body weights and clinical observations were made daily post-challenge. Clinical assessments included monitoring for signs of COVID-19, i.e., ruffled fur, hunched posture, labored breathing.


F. ELISA for Anti-SARS-CoV-2 S Protein-Specific IgG Titers


An ELISA was developed for the detection of anti-SARS-CoV-2 S protein-specific IgG antibodies in serum. Briefly, the endpoint titration method utilizes an indirect capture ELISA format in which the S1 SARS-CoV-2 S protein is used as the solid-phase immobilized antigen and horseradish peroxidase-labeled anti-hamster IgG is used for detection. The primary assay endpoint is reported as the reciprocal of the highest dilution above the assay cut point.


To coat plates, diluted SARS-CoV-2 S protein RBD-His recombinant protein, was added at 50 μL per well into half-area 96 well ELISA plates, covered, and incubated at 4° C. overnight. Plates were washed with PBST (1×PBS, pH 7.4, 0.05% Tween-20) and blocked by adding 150 μL per well of blocking buffer (PBST, 3% w/v milk) and allowed to incubate at 37° C. for 1.5 hours. A 1:100 dilution of each test serum sample was prepared using dilution solution (PBST, 2% w/v milk) in a 96-well U-bottom plate. Blocked assay plates were then washed with PBST. After adding 50 μL per well of dilution solution to the wells in columns 2-12, 75 μL per well of diluted test serum samples was added from the U-bottom plate into wells of column 1 and a 3-fold serial dilution was performed across the plate until column 11, leaving the wells in column 12 as the blank. The test serum sample-filled assay plates were then incubated at 37° C. for 1 hour. After washing assay plates with PBST, 50 μL per well of diluted secondary antibody HRP-conjugated goat anti-Golden Syrian Hamster IgG antibody (1:6,000 in dilution solution) was added to each well and incubated at 37° C. for 1 hour. After washing plates with PBST, 50 μL per well of TMB was added per well and incubated at room temperature for 10 minutes; the enzymatic reaction was stopped with addition of 50 μL per well of stop solution (2 N sulfuric acid). Absorbance at OD at 450 nm was determined using a VersaMax™ microplate reader (Molecular Devices, San Jose, Calif., USA) with SoftMax Pro GxP Data Acquisition software. Endpoint titers were determined using GraphPad Prism software, version 8 (GraphPad Software, San Diego, Calif., USA).


G. SARS-CoV-2 S Protein-Specific Pseudoneutralization Assay (PRNT) (Merck and BIOQUAL Assays)


To measure SARS-CoV-2 S protein-specific neutralizing antibodies, a pseudoneutralization assay was conducted at Merck Research Laboratories. Briefly, serial dilutions of heat inactivated test sera were mixed with an equal volume of rVSVΔG-SARS-CoV-2 clone MB1 and neutralization was allowed to proceed for 1 hour at 37° C., 5% CO2. Vero African Green Monkey Kidney cells were infected with the serum/virus mixture and incubated at 37° C. for 1 hour. A methylcellulose overlay was then placed on the cells and the plates were incubated at 37° C. for 2 days. Following incubation, the methylcellulose layer was removed, plates were fixed, blocked overnight, and immunostained using a rabbit anti-SARS-CoV-2 (2019-nCoV) S protein RBD polyclonal antibody and an AlexaFluor 488-labeled goat anti-rabbit secondary antibody. Immunostained plaques were visualized and counted using the EnSight™ (PerkinElmer, Waltham, Mass., USA). The primary assay endpoint is reported as the NT50 or the inverse of the dilution of serum at which 50% of the input virus is prevented from infecting cells. The assay LOD is 20.


H. Determination of Viral Load Using Quantitative Polymerase Chain Reaction (RT-qPCR)


Respiratory tract tissues were assessed for vaccine virus by RT-qPCR using standard molecular biology methods. Total RNA was isolated and reverse transcribed into cDNA. rVSVΔG-SARS-CoV-2 clone MB1 virus was quantified using PCR.


I. Statistical Analyses


For the ELISA, endpoint titers were determined as the reciprocal of the highest dilution above the assay cut point using GraphPad Prism software version 8 (GraphPad Software).


For the pseudoneutralization assay, neutralizing titers, i.e., NT50 values, were determined by 4-parameter curve fit using GraphPad Prism software v.8.1.1 (GraphPad Software) by plotting the log transformed sample dilution (x-axis) by the percent neutralization (y-axis). Percent neutralization was calculated by the following equation:





% Neutralization=(1−[(sample plaque count−average cell control count)/(average virus control−average cell control])×100


The geometric mean titer ±950% confidence interval was then determined with GraphPad Prism software.


J. Materials


Materials used in this study are provided in Table 12.









TABLE 12







List of Materials









Material
Vendor
Catalog no./reference





SARS-CoV-2 challenge stock, isolate
BEI Resources
NR-52281


USA-WA1/2020







ELISA









SARS-CoV-2 (COVID-19) S Protein
SinoBiological
Cat. no. 40592-V08H


RBD-His Recombinant Protein


Goat Anti-Golden Syrian Hamster
Rockland
Cat. no. 607-1302


IgG(H + L) Antibody HRP conjugated


Assay control: SARS-CoV/ SARS-CoV-2
GeneTex
Cat. no. GTX632604


(COVID-19) S protein S2 antibody [1A9]


Assay control: SARS-CoV/SARS-CoV-2
SinoBiological
Cat. no. 40150-D001


S protein antibody, Chimeric Mab


Tween-20
Sigma Aldrich
Cat. no. P9416


Blotting-Grade Blocker (300 g, nonfat dry
Fisher Scientific
Cat. no. 1706404


milk)


Dulbecco's phosphate-buffered saline, 1X
Corning
Cat. no. 21-031-CV


without calcium and magnesium


Pierce 1-Step Ultra ® TMB-ELISA
ThermoScientific
Cat. no. 34029


ELISA Coating Buffer (5X)
BioLegend
Cat. no. 421701


Sulfuric acid, 2N
Fisher Scientific
Cat. no. MK-H381-1


Costar ® Assay Plate, 96 well, Clear, Flat
Corning
Cat. no. 3690


Bottom, Half-area, High-Binding,


Polystyrene, Non-Sterile


Falcon Tissue Culture Plate, 96 well, U-
BD Biosciences
Cat. no. 353227


Bottom with Low Evaporation Lid







PENT Assay









rVSVΔG-SARS-CoV-2 clone MB1 (1st lot);
Merck Research
NB-Barrcoll-5030085-0024


titer = 5.7 × 107 PFU/mL
Laboratories
NB-hartmame-5031563-0019


VERO cells (African green monkey
ATCC
CCL-81


kidney epithelial cells)


Fixation solution: 3.7% Formaldehyde
Polysciences, Inc.
Cat. no. 04018


(Methanol Free) 10% UltraPure EM


Grade) diluted in PBS


Primary Antibody: SARS-CoV-2 (2019-
SinoBiological
Cat. no. 40592-T62


nCoV) S protein RBD Antibody, Rabbit


PAb, Antigen Affinity Purified


Secondary Antibody: AlexaFluor 488 goat
Life Technologies
Cat. no. A1 1008


anti-rabbit IgG (H7L)


BIOQUAL assay:
BEI Resources
NR-52281


SARS-CoV-2 isolate USA-WA1/2020





COVID-19 = coronavirus disease of 2019; ELISA = enzyme-linked immunosorbent assay; H + L = heavy and light chain; His = histidine; IgG = immunoglobulin G; no. = number; MAb = monoclonal antibody; PAb = polyclonal antibody; PBS = phosphate-buffered saline; SARS-CoV-2 = Severe Acute Respiratory Syndrome Coronavirus 2; TMB = 3,3′,5,5′-tetramethylbenzidine; USA = United States of America.






K. Results: Evaluation of Immunogenicity


Post-vaccination ELISA titers for Part A are shown in FIG. 16 though Day 26. Post-vaccination PRNT neutralizing titers for Part A are shown in FIG. 17A for Days 7 and 26 (Merck), and in FIG. 17B for Day 14. Preliminary data through Day 26 suggest immunogenicity responses were initiated post-vaccination for rVSVΔG-SARS-CoV-2 clone MB1 by the IM route of administration.


PRNT neutralizing titers after 7 and 14 days following a single immunization are shown in FIG. 18. MB1 and MB2 showed comparable immunogenicity following IM administration. Results: Part A—Assessment of Protection Against Cody Weight Loss After SARS-CoV-2 Challenge


Body weight loss is a sign of COVID-19 in the hamster model of SARS-CoV-2 infection (Chan et al, 2020). Body weight (B3W) loss was assessed for 5 days post-challenge in Part A of the study. As shown in FIGS. 19A-C, IM administration of rVSVΔG-SARS-CoV-2 clones MB1 and MB2 protected animals from BW loss post-challenge. OM administration of rVSVΔG-SARS-CoV-2 either clone did not offer protection against BW loss, with loss similar to non-vaccinated animals.


L. Conclusions


Data through Day 26 suggest immunogenicity responses were initiated post-vaccination for rVSVΔG-SARS-CoV-2 clones MB1 (nucleic acid SEQ ID NO: 150 or amino acid sequences SEQ ID NOS: 151-153, 155, and 156) and MB2 (nucleic acid SEQ ID NO: 154 or amino acid sequences 152 and 155-158) by the IM route of administration as seen by anti-SARS-CoV-2 S protein IgG titers and SARS-CoV-2 PRNT titers.


Single-dose IM administration of rVSVΔG-SARS-CoV-2 clone MB1 protected animals from BW loss post-challenge, suggesting a protection against COVID-19.


Single-dose OM administration of rVSVΔG-SARS-CoV-2 clone MB1 did not protect animals from BW loss post-challenge, suggesting there was inadequate protection against COVID-19. This aligns with the low immunogenicity responses observed in the ELISA and PRNT assays.


Example 16. Single-Nucleotide Polymorphisms (SNPs) in rVSVΔG-SARS-CoV-2 Clone MB1 (Nucleic Acid Sequence SEQ ID NO: 150 or Amino Acid Sequences SEQ ID NOS: 151-153, 155, and 156)

rVSVΔG-SARS-CoV-2 clone MB1, SEQ ID NO: 150, comprises a variant of the codon-optimized nucleotide sequence of rVSVΔG-SARS-CoV-2 S protein, SEQ ID NO: 160. Tables 13 and 14 summarize SNPs for rVSVΔG-SARS-CoV-2 clone MB1 (nucleic acid sequence SEQ ID NO: 150 or amino acid sequences SEQ ID NOS: 151-153, 155, and 156) relative to the expected nucleic acid SEQ ID NO: 150.









TABLE 13







SNP summary for rVSVΔG-SARS-CoV-2 clone MB1






















Variant call
Variant call
Is the variant







Total
Variant
disbalance
entropy
call major or
Confidence




Variant call

coverage
call
(F/R, closer
(closer to 1
minor
in variant


Position
Ref
freq, pct
Variant
depth
depth
to 1 is better)
is better)
variant?
call



















6852
G
99.9
T
608675
608317
0.95
0.82
major variant
High


5064
C
99.9
T
468358
467908
0.90
0.85
major variant
High


11041
C
99.9
A
435606
435140
1.12
0.85
major variant
High


13438
T
54.4
C
195
106
0.93
0.62
major variant
High





Single nucleotide variant (SNV) calls ≥5% frequency with confidence calculations (indels excluded).













TABLE 14







Summary for position 6993-A










Position 6993-A
Sample 24






HIVE coverage at 6993
171326






Ref gaaaaaaaac (SEQ ID NO: 194)
  7781






Var g-aaaaaaac
141410






Var-A frequency
     0.83









Example 17. SNPs in rVSVΔG-SARS-CoV-2 Clone MB2 (Nucleic Acid Sequence SEQ ID NO: 154 or Amino Acid Sequences SEQ ID NOS: 152 and 155-158)

rVSVΔG-SARS-CoV-2 clone MB2, SEQ ID NO: 154 comprises a variant of the codon-optimized rVSVΔG-SARS-CoV-2 S protein gene, SEQ ID NO: 158. The rVSVΔG-SARS-CoV-2 clone MB2 was identified after one round of clonal isolation. Tables 15 and 16 summarize SNPs for rVSVΔG-SARS-CoV-2 clone MB2 (nucleic acid sequence SEQ ID NO: 154 or amino acid sequences SEQ ID NOS: 152 and 155-158) relative to the expected wildtype SEQ ID NO: 159 for the S protein.









TABLE 15







SNP summary for rVSVΔG-SARS-CoV-2 clone MB2










Position
Ref
Var
Plaque 13













5064
C
T
1.00


6852
G
T
1.00


6993
A

*


11041
C
A
1.00


11098
C
A
0.05


13434
G
A
0.12


13436
C
A
0.33
















TABLE 16







Summary for position 6993










Position 6993
Plaque 13






gaaaaaaaac (SEQ ID NO: 194)
392






gaaaaaaac
 16






var frequency*
  0.04









Example 18. Immunogenicity of rVSVΔG-SARS-CoV-2 Clone MB1 in a Hamster Model of SARS-CoV-2 Infection Following Mucosal Application

This example describes a nonclinical pharmacology study to evaluate the immunogenicity pf rVSVΔG-SARS-CoV-2 clone MB1 to determine if it protects against SARS-CoV-2 challenge. The Golden Syrian hamster model used here is as described in Example 15 above. The study was conducted in two parts to evaluate the ability of mucosal administration of rVSVΔG-SARS-CoV-2 clone MB1-A) to generate a neutralizing antibody response against SARS-CoV-2 pseudovirus; and B) to protect against SARS-CoV-2 replication in the nose and lung of challenged hamsters. The study evaluated both the oral-mucosal (OM), oral (PO), intranasal+oral-mucosal (IN+OM) and intramuscular (IM) injection routes of administration.


A. Test Article: rVSVΔG-SARS-CoV-2 Clone MB1


The composition and formulation of test article rVSVΔG-SARS-CoV-2 clone MB1 were as described in Example 15.A above.


Description and sequence for rVSVΔG-SARS-CoV-2 clone MB1 is provided in Section II.F above, and FIGS. 21A-G and 24A-B.


B. Test Animals


Details on test animals were as described in Example 15.C and Table 9 above.


C. Group Designation, Dose Levels, and Dosing Schedule


Group designation, dose levels, and dosing schedule for the study are shown in Table 17.









TABLE 17







Group Designation, Dose Levels and Dosing Schedule













No. of
Treatment


Challenge Dose


Group
animals
(SD0)
Route
Dose
(Day) a















1
10
PBS
IM
NA
2 × 104







PFU SARS-







CoV-2







(Day 28 post-







vaccination)


2
10
MB1
IM
3.00E+06
2 × 104







PFU SARS-







CoV-2







(Day 28 post-







vaccination)


3
10
MB1
IM
1.00E+05
2 × 104







PFU SARS-







CoV-2







(Day 28 post-







vaccination)


4
10
MB1
IM
1.00E+03
2 × 104







PFU SARS-







CoV-2







(Day 28 post-







vaccination)


5
10
MB1
OM
3.00E+05
2 × 104







PFU SARS-







CoV-2







(Day 28 post-







vaccination)


6
10
MB1
PO
3.00E+06
2 × 104







PFU SARS-







CoV-2







(Day 28 post-







vaccination)


7
10
MB1
IN + OM
3.00E+05
2 × 104







PFU SARS-







CoV-2







(Day 28 post-







vaccination)


8
10
MB1
OM
3.00E+05
2 × 104







PFU SARS-







CoV-2







(Day 28 post-







vaccination)





IM = intramuscular; OM = oral mucosal; PO = oral; IN+OM = intranasal + oral mucosal; NA = not applicable; No. = number; PFU = plaque-fbrming unit(s); SD0 = Study Day Zero (0); SARS = Severe acute respiratory syndrome; SARS-CoV- 2 = SARS-associated coronavirus-2.


The challenge inoculum was administered 0.1 mL per animal, 0.05 mL per nostril.






D. Study Measurements


Serum was collected on Days 7, 14, and 28 post-vaccination. Serum was assayed for anti-SARS-CoV-2 S protein-specific IgG titers and for anti-SARS-CoV-2 PRNT titers.


Scheduled necropsies were carried out for nares and lung tissue collection on Day 32 (Day 4 post-challenge). Processed samples were assayed for viral load.


E. ELISA for Anti-SARS-CoV-2 S Protein-Specific IgG Titers


The ELISA assays were as described in Example 15.F above.


F. SARS-CoV-2 S Protein-Specific Pseudoneutralization Assay (PRNT) (Merck and BIOQUAL Assays)


The pseudoneutralization assays were as described in Example 15.G above.


G. Determination of Viral Load Using TCID50


A TCID50 assay was conducted by BIOQUAL using their SOP BV-008. Vero E6 cells (ATCC cat. no. CRL-1586) were plated at 25,000 cells/well in DMEM+10% FBS+Gentamicin and the cultures were incubated at 37° C., 5.0% CO2. Cells should be 80% to 100% confluent the following day. Medium was aspirated and replaced with 180 μL of DMEM+2% FBS+gentamicin. Twenty (20) μL of sample was added to top row in quadruplicate and mixed using a P200 pipettor 5 times. Using the pipettor, 20 μL was transferred to the next row, and repeated down the plate (columns A-H) representing 10-fold dilutions. The tips were disposed for each row and repeated until the last row. Positive (virus stock of known infectious titer in the assay) and negative (medium only) control wells were included in each assay set-up. The plates were incubated at 37° C., 5.0% CO2 for 4 days. The cell monolayers were visually inspected for cytopathic effects (CPE). Non-infected wells had a clear confluent cell layer while infected cells have cell rounding. The presence of CPE was marked on the lab form as a “+” and absence of CPE as “−”. The assay was controlled by obtaining a TCID50 value of the positive control within 2-fold of the expected value. The TCID50 value was calculated using the Read-Muench formula and bar graphed using GraphPad Prism software version 8 (GraphPad Software)


H. Statistical Analyses


Statistical analyses methods for ELISA and the pseudoneutralization assay were as described in Example 15.1 above.


I. Determination of Viral Load or Serum Neutralizing Capacity Using TCID50 Assay (BIOQUAL)


The viral load in respiratory tract tissue samples and in serum samples was determined using the TCID50 assay and was conducted by BIOQUAL, Inc. per BIOQUAL procedure 100011.00.006.


The TCID50 assay quantifies viral infectious units by determining the cytopathic effect of samples on Vero E6 cells. Vero E6 cells have been subcloned for high expression of ACE2.


Briefly, Vero E6 cells were plated at 25,000 cells per well in DMEM containing 10% FBS and gentamicin, and incubated at 37° C., 5% carbon dioxide until cells were 80 to 100% confluent. Media was replaced in each well with 180 μL DMEM containing 2% FBS and gentamicin. Each sample was added as 20 μL in quadruplicate wells. Solutions in each well were mixed by trituration, and samples were then serially diluted ten-fold across the plate by transferring 20 μL into the next well, changing tips after each transfer. Sample plates were then incubated at 37° C., 5% carbon dioxide for 4 days. After 4 days each well was visually inspected for cytopathic effect. Wells with no sample, i.e., non-infected wells, were confirmed as having a clear confluent cell layer. The presence of cytopathic effect was noted and the TCID50 calculated using the Reed-Muench formula (Reed and Muench, 1938). The TCID50 of the positive control was confirmed to be within 2-fold of the expected value.


The limit of detection was determined to be 250 TCID50 per 0.2 g tissue. Serum capacity to prevent infection was determined using the TCID50 assay and expressed as IC90 and IC50 values.


J. Materials


Materials used in this study were as provided in Example 15.J and Table 12 above.


K. Results: Evaluation of Immunogenicity


PRNT neutralizing titers after 14 and 28 days following a single immunization are shown in FIG. 28. MB1 showed comparable immunogenicity following IM administration.


L. Assessment of Viral Load after Challenge


SARS-CoV-2 viral load was measured in lung and nose 4 days after challenge. Levels of replicating SARS-CoV-2 quantified by TCID50 are shown in FIGS. 29A-D. Following IM administration, the 3×10≢pfu dose of MB1 led to a ˜3.5 log drop in SARS-CoV-2 in hamster lung (FIG. 29A) and a ˜1.5 log drop in SARS-CoV-2 in the nose (FIG. 29B). The 1×10≡pfu dose led to a smaller drop in viral titer and the 1×10{circumflex over ( )}3 pfu dose of MB1 was not notably different than no vaccine (FIGS. 29A-B). Following a single OM vaccination, 2/5 animals showed no evidence of SARS-CoV-2 in nose or lung, while the remaining animals showed lower to no protection from challenge (FIGS. 29C and 29D). Following PO vaccination, 3/5 animals showed no evidence of SARS-CoV-2 in nose or lung (FIGS. 29C and 29D). Following IN+OM vaccination, 5/5 animals had no evidence of SARS-CoV-2 in lung, and 1/5 animals had a slightly higher than background level of SARS-CoV-2 in nose.


M. Conclusions


Data through Day 26 suggest immunogenicity responses were initiated post-vaccination for rVSVΔG-SARS-CoV-2 clones MB1 (nucleic acid SEQ ID NO: 150 or amino acid sequences SEQ ID NOS: 151-153, 155, and 156) by the IM, OM, PO, and IN+OM routes of administration as seen by anti-SARS-CoV-2 S protein IgG titers and SARS-CoV-2 PRNT titers.


Single-dose IM administration of rVSVΔG-SARS-CoV-2 clone MB1 protected animals from SARS-CoV-2 in lung and nose post-challenge, suggesting a protection against COVID-19.


Single-dose OM administration of rVSVΔG-SARS-CoV-2 clone MB1 provided protection from SARS-CoV-2 in lung and nose in 3/10 hamsters, suggesting there was inadequate protection against COVID-19. This aligns with the lower immunogenicity responses observed in the ELISA and PRNT assays.


Single-dose PO administration of rVSVΔG-SARS-CoV-2 clone MB1 protected 3/5 animals from SARS-CoV-2 in lung and nose post-challenge, suggesting partial protection against COVID-19. This aligns with the immunogenicity responses observed in the ELISA and PRNT assays.


Single-dose IN+OM administration of rVSVΔG-SARS-CoV-2 clone MB1 protected animals from SARS-CoV-2 in lung and nose post-challenge, suggesting a protection against COVID-19.


Example 19. Mucosal Vaccination

A. Vaccination by Applying VSVΔG-SARS-CoV-2 to the Mucosal Surfaces in the Nasal and/or Oral Cavities


In preclinical studies, SARS-CoV-2 infection has been demonstrated by applying virus directly to the mucosal surfaces in the nasal or oral cavities. Lee et al., Cell Rep Med, 1:100121 (2020). Consistent with these findings, studies have shown that the ACE2 used by the SARS-CoV-2 S protein for cell attachment is expressed by cells in these mucosal tissues. Ziegler et al., Cell, 181:1016-1035 e19 (2020); Suresh et al., Frontiers in Pharmacology, 11 (2020); Xu et al., Int J Oral Sci, 12:8 (2020). The susceptibility of these mucosal tissues to SARS-CoV-2 infection suggested that vaccination with VSVΔG-SARS-CoV-2 could be conducted by applying the vaccine virus directly to mucosal surfaces in the nasal or oral cavity. Vaccination with VSVΔG-SARS-CoV-2 by these routes may be advantageous because it has the potential to induce both systemic and local mucosal barrier immunities that can prevent severe disease in the lower respiratory tract as well as reduce SARS-CoV-2 replication in the upper airways, thereby reducing the risk of virus transmission. Gallo et al., Mucosal Immunol, doi:10.1038/s41385-020-00359-2 (2020).


To investigate the feasibility of mucosal vaccination with a VSVΔG-SARS-CoV-2 vaccine, rhesus macaques were vaccinated by three different methods (4 animals per method) using an investigational vaccine strain, Fp11, that differed from VSVΔG-SARS-CoV-2 MB1 as described in Table 18 below. In the first vaccination method, anesthetized animals were vaccinated by the intranasal route (IN) using a spray device (mucosal atomizer device; MAD™ from Teleflex Inc.) that delivered 1×107 PFUs per nostril. In the second method, virus was delivered to the oral mucosa (OM) by applying 2×107 PFUs dropwise to the top surface of the tongue of anesthetized macaques. The final method was a combination of both intranasal and oral mucosal approaches (IN+OM) with the total dose of 2×107 PFUs split equally across the two nostrils and the tongue. Vaccinations were conducted at week 0 and week 8.









TABLE 18







Mutations in MB1 and Strain Fp11












Affected





Donor
amino acid


sequence
position
Wild-type
MB1
Strain Fp11





SARS-CoV-
C-terminus
No deletion
23 amino acids deleted
21 amino acids


2 S protein


from the C-terminus
deleted from the C-






terminus



484
E
E
D



655
H
H
Y



683
R
G
R



813
S
F
S


VSV Matrix
61
Y
S
Y


VSV Matrix
213
E
E
K









B. ELISA


Materials used for ELISA in this example are provided in Table 19.









TABLE 19







List of Materials









Material
Vendor
Catalog no.





Adhesive Film for Microplates
VWR
60941-064


Costar ® assay plate, 96 well, clear,
Corning
3690


flat bottom, half-area,


high-binding, polystyrene, non-sterile


ELISA coating buffer (5X)
Biolegend
421701


Coating protein: SARS-CoV-2 spike protein prefusion-
Lake Pharma
46328


stabilized ectodomain, C-term His tag,


with furin cleavage


site removed, trimerization stabilized


Secondary antibody: horseradish peroxidase (HRP)
Jackson
109-035-098


conjugated goat anti-human IgG
ImmunoResearch


Tween-20
Sigma Aldrich
P9416


2N H2SO4
Fisher Scientific
MK-H381-1


DPBS, 1X without calcium and magnesium
Corning
21-031-CV


1-Step Ultra ® TMB-ELISA at room temperature
Thermo Scientific
34029


Tissue Culture Plate, 96 well, U-bottom with low
BD Falcon
353227


evaporation lid


Sterile pipets
VWR


Cordless pipet-aid
Drummond


Sterile barrier pipet tips
Ranin, BioHit,



Molecular



Bioproducts


Pipettors
VWR, Ranin, BioHit


Distilled or reverse osmosis purified water at room


temperature


ELISA coating buffer 1X diluted with distilled/reverse
Biolegend


osmosis purified water


Plate wash buffer, PBST (1X PBS pH 7.4, 0.05% Tween-


20) at room temperature


Blocking buffer solution (3% bovine serum albumin (BSA


pH 7.4) at room temperature


Dilution solution (1% BSA in 1X PBS pH 7.4) at room


temperature


Stop solution (2N H2SO4) at room temperature


ELx405 select deep well microplate washer
Biotek


Versa Max microplate reader and SoftMax Pro GxP data
Molecular Devices


acquisition software


GraphPad Prism 9 software









The ELISA method was used to detect anti-SARS-CoV-2 S protein-specific IgG antibodies in the serum of vaccinated animals. The protein coating used for the ELISA method was a soluble SARS-CoV-2 S protein in which the S1 and S2 subunits were covalently linked.


96 well ELISA plates were coated with 50 μL/well of SARS-CoV-2 antigen (1 ug/ml) diluted in 1× coating buffer. Only half of the wells in each plate were used. Each plate was covered with microplate adhesive film and incubated overnight at 4° C. The plates were washed using a plate washer. Then, the plates were blocked with 150 μL/well of blocking buffer and incubated at 37° C. for 1 hour 30 minutes.


Meanwhile, test serum was diluted in a 96-well U-bottom plate at 1:100 using dilution solution and set aside.


Back to the blocked plates, the blocked plates were washed using a plate washer. Wells in columns 2-12 were filled with 50 μL/well of dilution solution. For incubation with serum, 75 ul of diluted serum from the U-bottom plate was transferred into each well of column 1. A 3-fold serial dilution was performed across plate from column 2 through column 11, leaving the wells in column 12 as the blank. Plates were incubated at 37° C. for 1 hours.


The serum-treated plates were washed using a plate washer. For antibody labeling, 50 μL of diluted secondary antibody (1:5,000 in dilution solution) was added to each well. The plates were incubated at 37° C. for 1 hour.


The antibody-labeled plates were washed using a plate washer. 50 μL of 1-Step Ultra® TMB was added to each well at room temperature. The incubation period was 10 mins. 50 μL of Stop Solution was added to stop development. Plates were read using a plate reader at 450 nm.


C. Results


As shown in FIG. 30, the study results demonstrated that most macaques vaccinated with VSVΔG-SARS-CoV-2 strain Fp11 by the IN or IN+OM method developed detectable serum antibodies against the SARS-CoV-2 S protein by week 4 and all were positive by week 8. Animals vaccinated only by the OM method did not respond. To evaluate the effect of a second vaccination, macaques were vaccinated with identical dose and method at week 8. At two weeks following the second vaccination, there was a pronounced increase in serum antibody titers in animals vaccinated by the IN or IN+OM methods indicating the initial vaccination established immunologic memory that allowed a robust recall response following the second vaccination.


Example 20. Embodiments

The following numbered items provide a description of certain embodiments herein.


Item 1. A recombinant vesicular stomatitis virus (VSV) vector, comprising a modified VSV genome, wherein the modified VSV genome comprises a foreign gene derived from a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2).


Item 2. A recombinant vesicular stomatitis virus (VSV) vector, comprising a modified VSV genome, wherein the modified VSV genome comprises a foreign gene encoding a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or an immunogenic variant thereof.


Item 3. A recombinant vesicular stomatitis virus (VSV) vector, comprising a modified VSV genome, wherein the modified VSV genome comprises a portion of a VSV glycoprotein (G) gene and a foreign gene encoding an immunogenic variant of a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein.


Item 4. The recombinant VSV vector of any one of the preceding items, wherein the SARS-CoV-2 S protein consists of the amino acid sequence as shown in FIG. 1A or an amino acid sequence having GenBank Accession number gb_QHW06059, gb_QHZ00379, gb_QHR84449, gb_QIA20044, gb_QHZ00358, dbj_BCA25664, gb_QHZ87582, dbj_BCA25674, gb_QHZ00389, gb_QHZ87592, gb_QIB84673, gb_QHR63260, gb_QIA98606, gb_QIA98596, gb_QHR63250, gb_QHR63280, gb_QHR63290, gb_QHR63270, gb_QHZ00399, dbj_BCA25644, dbj_BCA25654, gb_QHO62877, ref_YP_00972439, gb_QHW06039, gb_QHO62112, gb_QHQ71973, gb_QHW06049, gb_QHO60594, gb_QHN73810, gb_QHO62107, gb_QHQ82464, dbj_BBW89517, gb_QHU79204, gb_QHQ71963, gb_QHU79194, gb_QHN73795, gb_QHU36864, gb_QHD43416, gb_QHU36854, gb_QHU36844, gb_QHU36834 or gb_QHU36824 as shown in FIG. 2.


1 Item 5. The recombinant VSV vector of any one items 1-3, wherein the immunogenic variant of the SARS-CoV-2 S protein consists of an amino acid sequence at least 90% identical to that of the SARS-CoV-2 S protein.


Item 6. The recombinant VSV vector of any one items 1-3, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein having a deletion at the C-terminal end of the SARS-CoV-2 S protein.


Item 7. The recombinant VSV vector of item 1 or 2, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail of the SARS-CoV-2 S protein.


Item 8. The recombinant VSV vector of item 1 or 2, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail and a transmembrane domain of the SARS-CoV-2 S protein.


Item 9. The recombinant VSV vector of any one items 1-3, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail of the SARS-CoV-2 S protein, and wherein the portion of the VSV-G gene encodes a cytoplasmic tail of a VSV-G protein.


Item 10. The recombinant VSV vector of any one items 1-3, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail and a transmembrane domain of the SARS-CoV-2 S protein, and wherein the portion of the VSV-G gene encodes a cytoplasmic tail and a transmembrane domain of a VSV-G protein.


Item 11. The recombinant VSV vector of any one of the preceding items, wherein the modified VSV genome further comprises a VSV nucleoprotein (N) gene, a VSV phosphoprotein (P) gene, a VSV matrix protein (M) gene, and a VSV polymerase (L) gene arranged in sequence from 3′ end to 5′ end.


Item 12. The recombinant VSV vector of any one items 1-10, wherein the foreign gene is inserted on the 3′ end of the VSV-N gene.


Item 13. The recombinant VSV vector of any one items 1-10, wherein the foreign gene is between the VSV-N gene and the VSV-P gene.


Item 14. The recombinant VSV vector of any one items 1-10, wherein the foreign gene is between the VSV-P gene and the VSV-M gene.


Item 15. The recombinant VSV vector of any one items 1-10, wherein the foreign gene is between the VSV-M gene and the VSV-L gene.


Item 16. The recombinant VSV vector of any one items 1-10, wherein the foreign gene is on the 5′ end of the VSV-L gene.


Item 17. The recombinant VSV vector of item 1 or 2, wherein the foreign gene encodes the SARS-CoV-2 S protein, and wherein the modified VSV genome does not comprise a VSV glycoprotein (G) gene or a portion thereof.


Item 18. A recombinant replicable vesicular stomatitis virus (VSV) particle, comprising a modified VSV genome and a Severe Acute Respiratory Syndrome coronavirus 2 (SARSCoV-2) spike (S) protein or an immunogenic variant thereof on the surface of the recombinant replicable VSV particle, wherein the modified VSV genome comprises a foreign gene encoding the SARS-CoV-2 S protein or the immunogenic variant thereof.


Item 19. A recombinant replicable vesicular stomatitis virus (VSV) particle, comprising a modified VSV genome and an immunogenic variant of a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein on the surface of the recombinant replicable VSV particle, wherein the modified VSV genome comprises a portion of a VSV glycoprotein (G) gene and a foreign gene encoding the immunogenic variant of the SARSCoV-2 S protein.


Item 20. The recombinant replicable VSV particle of item 18 or 19, wherein the SARS10 CoV-2 S protein consists of the amino acid sequence as shown in FIG. 1A or an amino acid sequence having GenBank Accession number gb_QHW06059, gb_QHZ00379, gb_QHR84449, gb_QIA20044, gb_QHZ00358, dbj_BCA25664, gb_QHZ87582, dbj_BCA25674, gb_QHZ00389, gb_QHZ87592, gb_QIB84673, gb_QHR63260, gb_QIA98606, gb_QIA98596, gb_QHR63250, gb_QHR63280, gb_QHR63290, gb_QHR63270, gb_QHZ00399, dbj_BCA25644, dbj_BCA25654, gb_QHO62877, ref_YP_00972439, gb_QHW06039, gb_QHO62112, gb_QHQ71973, gb_QHW06049, gb_QHO60594, gb_QHN73810, gb_QHO62107, gb_QHQ82464, dbj_BBW89517, gb_QHU79204, gb_QHQ71963, gb_QHU79194, gb_QHN73795, gb_QHU36864, gb_QHD43416, gb_QHU36854, gb_QHU36844, gb_QHU36834 or gb_QHU36824 as shown in FIG. 2.


Item 21. The recombinant replicable VSV particle of item 18 or 19, wherein the immunogenic variant of the SARS-CoV-2 S protein consists of an amino acid sequence at least 90% identical to that of the SARS-CoV-2 S protein.


Item 22. The recombinant replicable VSV particle of item 18 or 19, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein having a deletion at the C-terminal end of the SARS-CoV-2 S protein.


Item 23. The recombinant replicable VSV particle of item 18, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail of the SARS-CoV-2 S protein.


Item 24. The recombinant replicable VSV particle of item 18, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail and a transmembrane domain of the SARS-CoV-2 S protein.


Item 25. The recombinant replicable VSV particle of item 18 or 19, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail of the SARS-CoV-2 S protein, and wherein the portion of the VSV-G gene encodes a cytoplasmic tail of a VSV-G protein.


Item 26. The recombinant replicable VSV particle of item 18 or 19, wherein the immunogenic variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail and a transmembrane domain of the SARS-CoV-2 S protein, and wherein the portion of the VSV-G gene encodes a cytoplasmic tail and a transmembrane domain of a VSV-G protein.


Item 27. The recombinant replicable VSV particle of any one of items 18-26, wherein the modified VSV genome further comprises a VSV nucleoprotein (N) gene, a VSV phosphoprotein (P) gene, a VSV matrix protein (M) gene, and a VSV polymerase (L) gene arranged in sequence from 3′ end to 5′ end.


Item 28. The recombinant replicable VSV particle of any one of items 18-27, wherein the foreign gene is inserted on the 3′ end of the VSV-N gene.


Item 29. The recombinant replicable VSV particle of any one of items 18-27, wherein the foreign gene is between the VSV-N gene and the VSV-P gene.


Item 30. The recombinant replicable VSV particle of any one of items 18-27, wherein the foreign gene is between the VSV-P gene and the VSV-M gene.


Item 31. The recombinant replicable VSV particle of any one of items 18-27, wherein the foreign gene is between the VSV-M gene and the VSV-L gene.


Item 32. The recombinant replicable VSV particle of any one of items 18-27, wherein the foreign gene is on the 5′ end of the VSV-L gene.


Item 33. The recombinant replicable VSV particle of item 18, wherein the foreign gene encodes the SARS-CoV-2 S protein, and wherein the modified VSV genome does not comprise a VSV glycoprotein (G) gene or a portion thereof.


Item 34. An immunogenic recombinant protein comprising a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof derived from the recombinant VSV vector of any one of items 1-17.


Item 35. An immunogenic recombinant protein comprising a variant of a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein and a portion of a VSV glycoprotein (G).


Item 36. The immunogenic recombinant protein of item 34 or 35, wherein the SARSCoV-2 S protein consists of the amino acid sequence as shown in FIG. 1A or an amino acid sequence having GenBank Accession number gb_QHW06059, gb_QHZ00379, gb_QHR84449, gb_QIA20044, gb_QHZ00358, dbj_BCA25664, gb_QHZ87582, dbj_BCA25674, gb_QHZ00389, gb_QHZ87592, gb_QIB84673, gb_QHR63260, gb_QIA98606, gb_QIA98596, gb_QHR63250, gb_QHR63280, gb_QHR63290, gb_QHR63270, gb_QHZ00399, dbj_BCA25644, dbj_BCA25654, gb_QHO62877, ref_YP_00972439, gb_QHW06039, gb_QHO62112, gb_QHQ71973, gb_QHW06049, gb_QHO60594, gb_QHN73810, gb_QHO62107, gb_QHQ82464, dbj_BBW89517, gb_QHU79204, gb_QHQ71963, gb_QHU79194, gb_QHN73795, gb_QHU36864, gb_QHD43416, gb_QHU36854, gb_QHU36844, gb_QHU36834 or gb_QHU36824 as shown in FIG. 2.


Item 37. The immunogenic recombinant protein of item 34 or 35, wherein the variant of the SARS-CoV-2 S protein consists of an amino acid sequence at least 90% identical to that of the SARS-CoV-2 S protein.


Item 38. The immunogenic recombinant protein of item 34 or 35, wherein the variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein having a deletion at the C-terminal end of the SARS-CoV-2 S protein.


Item 39. The immunogenic recombinant protein of item 34, wherein the variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail of the SARS-CoV-2 S protein.


Item 40. The immunogenic recombinant protein of item 34, wherein the variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail and a transmembrane domain of the SARS-CoV-2 S protein.


Item 41. The immunogenic recombinant protein of item 34 or 35, wherein the variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail of the SARS-CoV-2 S protein, and wherein the portion of the VSV-G gene encodes a cytoplasmic tail of a VSV-G protein.


Item 42. The immunogenic recombinant protein of item 34 or 35, wherein the variant of the SARS-CoV-2 S protein is a fragment of the SARS-CoV-2 S protein lacking a cytoplasmic tail and a transmembrane domain of the SARS-CoV-2 S protein, and wherein the portion of the VSV-G gene encodes a cytoplasmic tail and a transmembrane domain of a VSV-G protein. Item 43. A recombinant nucleic acid molecule comprising a nucleotide sequence encoding a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof.


Item 44. The recombinant nucleic acid molecule of item 43, wherein the nucleotide sequence is the sequence shown in FIG. 1B, 1C, 1E, 1F, 1H, 1I, 1K, 1L, 1N, 1O, 1Q, 1R, 1T, 1U, 1W or 1X (SEQ ID NO: 2, 3, 5, 6, 8, 9, 11, 12, 14, 15, 17, 18, 20, 21, 23 or 24, respectively) or a nucleotide sequence encoding a SARS-CoV-2 S protein derived from the SARS-CoV-2 genome sequence having GenBank Accession Number LR757997, MT072688, MN996527, MT039873, MN938384, MT020781, MN996529, MN996530, MN996531, MT044258, LR757998, LR757996, MT066175, MT066176, LR757995, LC522972, LC522973, LC522974, LC522975, MT039887, MN988668, MN988669, MT044257, MT039888, MN997409, MN985325, MN994467, MT027062, MT027063, MT027064, MT020880, MT020881, MN988713, MT019533, MN994468, MT093571, MT019530, MT019532, MN975262, MN996528, MT007544, MT019529, MT019531, MT049951, MT039890, MN908947, NC_045512 or MT093631.


Item 45. A method for producing a recombinant replicable VSV particle, comprising (a) introducing the recombinant VSV vector of any one of items 1-17 into cells, whereby a recombinant replicable VSV particle is produced, and (b) expressing a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or an immunogenic variant thereof on the surface of the recombinant replicable VSV particle.


Item 46. The method of item 45, wherein the cells are Vero cells.


Item 47. The method of item 45 or 46, further comprising purifying the recombinant replicable VSV particle from the cells.


Item 48. The method of item 45 or 46, further comprising purifying the SARS-CoV-2 S protein or an immunogenic variant thereof from the cells.


Item 49. The method of item 45 or 46, further comprising purifying the recombinant VSV vector from the cells.


Item 50. A recombinant cell comprising the recombinant VSV vector of any one of items 1-17 and producing a recombinant replicable VSV particle, wherein a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof is expressed on the surface of the recombinant replicable VSV particle.


Item 52. A pharmaceutical composition comprising the recombinant VSV vector of any one of items 1-17, the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49.


Item 53. The pharmaceutical composition of item 52, further comprising a pharmaceutically acceptable excipient.


Item 54. The pharmaceutical composition of item 52 or 53, wherein the pharmaceutical composition is a vaccine composition.


Item 55. The pharmaceutical composition of any one of items 52-54, wherein the pharmaceutical composition is formulated for oral, sublingual, intramuscular, intradermal, subcutaneous, intranasal, intraocular, rectal, transdermal, mucosal, topical or parenteral administration.


Item 56. An oral or oral mucosal composition, comprising an effective amount of the recombinant VSV vector of any one of items 1-17, the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49.


Item 57. The oral or oral mucosal composition of item 56, wherein the oral or oral mucosal composition is formulated for buccal delivery.


Item 58. The oral or oral mucosal composition of item 56, wherein the oral or oral mucosal composition is formulated for gastrointestinal delivery.


Item 59. An intranasal composition, comprising an effective amount of the recombinant VSV vector of any one of items 1-17, the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49.


Item 60. The intranasal composition of item 59, wherein the intranasal composition is formulated for nasal mucosal delivery.


Item 61. A vaccine composition comprising the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49.


Item 62. The vaccine composition of item 61, wherein the vaccine composition is formulated for oral, sublingual, intramuscular, intradermal, subcutaneous, intranasal, intraocular, rectal, transdermal, mucosal, topical or parenteral administration.


Item 63. An oral or oral mucosal vaccine composition, comprising an effective amount of the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49.


Item 64. The oral or oral mucosal vaccine composition of item 63, wherein the oral or oral mucosal vaccine composition is formulated for buccal delivery.


Item 65. The oral or oral mucosal vaccine composition of item 63, wherein the oral or oral mucosal vaccine composition is formulated for gastrointestinal delivery.


Item 66. An intranasal vaccine composition, comprising an effective amount of the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49.


Item 67. A method of inducing an immunological response to a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) in a subject, comprising administering to the subject an effective amount of the pharmaceutical composition of any one of items 52-55, whereby an immunological response is stimulated in the subject without causing a disease or symptom associated with the SARS-CoV-2 and the subject is vaccinated.


Item 68. The method of item 67, wherein the immunogenic response comprises a humoral response, a cellular antigen-specific immune response or a combination thereof.


Item 69. The method of item 67 or 68, wherein the immunological response comprises production of antibodies by the vaccinated subject that block SARS-CoV-2 infection, the method further comprising harvesting the antibodies from the vaccinated subject.


Item 70. The method of any one of items 67-69, wherein the antibodies are in the form of immune serum obtained from the vaccinated subject.


Item 71. The method of any one of items 67-69, wherein the antibodies are monoclonal antibodies prepared from SARS-CoV-2-specific B cells obtained from the vaccinated subject.


Item 72. The method of any one of items 69-71, further comprising mixing the harvested antibodies with a pharmaceutically acceptable excipient, whereby a pharmaceutical composition is prepared.


Item 73. A method of preventing infection of a subject by a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), comprising administering to the subject an effective amount of the pharmaceutical composition of item 52 or 53, wherein the subject does not suffer a disease or symptom associated with the SARS-CoV-2.


Item 74. A method of vaccinating a subject against a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), comprising administering to the subject an effective amount of the pharmaceutical composition of any one of items 52-55, wherein the subject does not suffer a disease or symptom associated with the SARS-CoV-2 and has not been exposed to the SARS-CoV-2.


Item 75. The method of any one of items 67-74, further comprising administering the pharmaceutical composition to a tissue in the subject that expresses a receptor for the SARS-CoV-2, and preventing or inhibiting binding of the SARS-CoV-2 to the receptor.


Item 76. The method of item 75, wherein the receptor is angiotensin-converting enzyme 2 (ACE2).


Item 77. The method of item 75 or 76, wherein the tissue is in the gastrointestinal (GI) tract of the subject.


Item 78. The method of any one of items 67-76, wherein the pharmaceutical composition is administered orally, sublingually, intramuscularly, intradermally, subcutaneously, intranasally, intraocularly, rectally, transdermally, mucosally, topically or parenterally.


Item 79. The method of any one of items 67-76, wherein the pharmaceutical composition is administered orally.


Item 80. The method of any one of items 67-76, wherein the pharmaceutical composition is administered intramuscularly.


Item 81. The method of any one of items 67-76, wherein the pharmaceutical composition is administered intranasally.


Item 82. A method of vaccinating a subject against a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), comprising administering to the subject an effective amount of an oral or oral mucosal vaccine composition, wherein the oral or oral mucosal vaccine composition comprises the recombinant VSV vector of any one of items 1-17, the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49, wherein the subject does not suffer a disease or symptom associated with the SARS-CoV-2 and has not been exposed to the SARS-CoV-2.


Item 83. The method of item 82, further comprising delivering to a tissue in the subject the recombinant VSV vector of any one of items 1-17, the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARSCoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49, wherein the tissue is buccal mucosa.


Item 84. The method of item 82, further comprising delivering to a tissue in the subject the recombinant VSV vector of any one of items 1-17, the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARSCoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49, wherein the tissue is gastrointestinal mucosa.


Item 85. A method of vaccinating a subject against a Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), comprising administering to the subject an effective amount of an intranasal vaccine composition, wherein the intranasal mucosal vaccine composition comprises the recombinant VSV vector of any one of items 1-17, the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49, wherein the subject does not suffer a disease or symptom associated with the SARS-CoV-2 and has not been exposed to the SARS-CoV-2.


Item 86. The method of item 85, further comprising delivering to a tissue in the subject the recombinant VSV vector of any one of items 1-17, the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARSCoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49, wherein the tissue is intranasal mucosa.


Item 87. The method of item 82 or 85, further comprising delivering to a tissue in the subject the recombinant VSV vector of any one of items 1-17, the recombinant replicable VSV particle of any one of items 18-33, the immunogenic recombinant protein of any one of items 34-42, the recombinant replicable VSV particle produced according to the method of any one of items 45-49, the Severe Acute Respiratory Syndrome coronavirus 2 (SARSCoV-2) spike (S) protein or a variant thereof purified according to the method of item 48, or the recombinant VSV vector purified according to the method of item 49, wherein the tissue expresses a receptor for the SARS-CoV-2.


Item 88. The method of any one of items 82, 85, or 87, wherein the receptor is angiotensin-converting enzyme 2 (ACE2).


Item 89. The method of any one of items 82, 85, 87, or 88, further comprising preventing or inhibiting binding of the SARS-CoV-2 to the receptor after the subject is exposed to the SARS-CoV-2.


Item 90. The method of any one of items 67-89, wherein the subject is a male.


Item 91. The method of any one of items 67-89, wherein the subject is a female.


Item 92. The method of any one of items 67-91, wherein the subject has a preexisting medical condition.


Item 93. The method of item 92, wherein the pre-existing medical condition is selected from the group consisting of high blood pressure, a heart disease, diabetes, a lung disease and combinations thereof.


Item 94. The method of any one of items 67-93, wherein the pharmaceutical composition is administered in a single dose.


Item 95. The method of any one of items 67-93, wherein the pharmaceutical composition is administered in two or multiple doses.


Item 96. The method of item 95, wherein the first dose is for a priming immunization and the second dose is a boosting immunization.


Item 97. A method of producing an adaptive mutant of the recombinant replicable VSV particle of any one of items 18-33, comprising growing the recombinant replicable VSV particle in a cell culture, changing a condition of the cell culture, identifying a mutant of the recombinant replicable VSV particle exhibiting greater replication capacity than the recombinant replicable VSV particle, whereby an adaptive mutant of the recombinant replicable VSV particle is obtained.


Item 98. The method of item 97, wherein the condition is selected from the group consisting of temperature, culture medium and cell substrate.


Item 99. The method of item 97 or 98, wherein the adaptive mutant exhibits at least 50% greater replication capacity than the recombinant replicable VSV particle. All documents, books, manuals, papers, patents, published patent applications, guides, abstracts, and other references cited herein are incorporated by reference in their entirety. Other embodiments will be apparent to those skilled in the art from consideration and practice of the disclosure. It is intended that the specification and examples be considered as exemplary and embodiments should be construed in accordance with the appended claims and any equivalents thereof.

Claims
  • 1. A recombinant VSV vector comprising: a. at least a portion of a VSV genome comprising at least a VSV-N gene, VSV-P gene, VSV-M gene, and VSV-L gene, andb. a nucleic acid sequence encoding a SARS-CoV-2 S protein or an immunogenic variant thereof.
  • 2. A recombinant VSV particle comprising the vector of claim 1 and displaying the S protein or immunogenic variant thereof on the surface of the VSV particle.
  • 3. A recombinant VSV particle comprising: a. at least a portion of a VSV genome comprising N, P, M, and L genes andb. a nucleic acid sequence encoding a SARS-CoV-2 S protein or an immunogenic variant thereof.
  • 4. The recombinant VSV particle of claim 2, wherein the VSV particle is replicable.
  • 5. The recombinant VSV particle of claim 3, wherein the VSV particle displays the S protein or an immunogenic variant thereof on the surface of the VSV particle.
  • 6. An immunogenic recombinant protein comprising a SARS-CoV-2 S protein or an immunogenic variant thereof expressed by the recombinant VSV vector of claim 1.
  • 7. An immunogenic recombinant protein comprising a SARS-CoV-2 S protein or an immunogenic variant thereof and at least a fragment of a VSV glycoprotein (G).
  • 8. A SARS-CoV-2 vaccine comprising the recombinant VSV vector of claim 1.
  • 9. The recombinant VSV vector of claim 1, wherein the SARS-CoV-2 S protein or an immunogenic variant thereof comprises an amino acid sequence having a length of at least 1223, 1228, 1233, 1238, 1243, 1248, 1249, 1250, 1251, 1252, 1253, 1254, 1255, 1256, 1257, 1258, 1259, 1260, 1261, 1262, 1263, 1264, 1265, 1266, 1267, 1268, 1269, 1270, 1271, 1272, 1273, 1274, 1275, 1276, 1277 or 1278 amino acids and having homology over its own length of at least 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% homology to any one of SEQ ID NOS: 1, 4, 7, 10, 13, 16, 19, 22, 25-67, 153, or 158.
  • 10. The recombinant VSV vector of claim 1, wherein the immunogenic variant of the SARS-CoV-2 S protein comprises a fragment of the SARS-CoV-2 S protein having a deletion of 9, 13, 19, 21 or 23 amino acids at the C-terminal end of the SARS-CoV-2 S protein.
  • 11. The recombinant VSV vector of claim 1, wherein: a. the recombinant VSV vector is rVSVΔG-SARS-CoV-2 clone MB2 (SEQ ID NO: 154); orb. the recombinant VSV vector, recombinant VSV particle, or SARS-CoV-2 vaccine comprises nucleic acids encoding i. VSV L protein (SEQ ID NO: 152);ii. VSV N protein (SEQ ID NO: 155);iii. VSV P protein (SEQ ID NO: 156);iv. VSV M protein (SEQ ID NO: 157); andv. SARS-CoV-2 S protein (SEQ ID NO: 158).
  • 12. The recombinant VSV particle of claim 2, wherein the particle or vaccine comprises VSV and SARS-CoV-2 proteins from MB2 comprising: a. VSV L protein (SEQ ID NO: 152);b. VSV N protein (SEQ ID NO: 155);c. VSV P protein (SEQ ID NO: 156);d. VSV M protein (SEQ ID NO: 157); ande. SARS-CoV-2 S protein (SEQ ID NO: 158).
  • 13. The recombinant VSV vector of claim 1, wherein: a. the recombinant VSV vector is rVSVΔG-SARS-CoV-2 clone MB1 (SEQ ID NO: 150); orb. the recombinant VSV vector, recombinant VSV particle, or SARS-CoV-2 vaccine comprises nucleic acids encoding i. VSV L protein (SEQ ID NO: 152);ii. VSV N protein (SEQ ID NO: 155);iii. VSV P protein (SEQ ID NO: 156);iv. VSV M protein (SEQ ID NO: 153); andv. SARS-CoV-2 S protein (SEQ ID NO: 151).
  • 14. The recombinant VSV particle of claim 2, wherein the particle or vaccine comprises VSV and SARS-CoV-2 proteins from MB1 comprising: a. VSV L protein (SEQ ID NO: 152);b. VSV N protein (SEQ ID NO: 155);c. VSV P protein (SEQ ID NO: 156);d. VSV M protein (SEQ ID NO: 151); ande. SARS-CoV-2 S protein (SEQ ID NO: 153).
  • 15. The recombinant VSV vector of claim 1, wherein the VSV-M protein comprises a Y61S mutation relative to SEQ ID NO: 1.
  • 16. The recombinant VSV vector of claim 1, wherein the immunogenic variant of the SARS-CoV-2 S protein comprises one or more mutations relative to SEQ ID NO: 1 chosen from: a. H655Y, R682K, and R685G;b. H655Y, R682K, R685G, and a deletion of the 9 amino acids (Δ9) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;c. H655Y, R682K, R685G, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;d. Q321P, H655Y, T678I, and P812R;e. Q321P, H655Y, T678I, P812R, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;f. E154D, S115L, T678I, R685G, and M1233K;g. R685G, S813R, and a deletion of the 9 amino acids (Δ9) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;h. H655Y, R682K, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;i. R685G, S813F, and a deletion of the 9 amino acids (Δ9) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;j. R683G, S813F and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;k. R683G, S813F, R685G, and a deletion of 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;l. H655Y, P681S, R682K, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;m. R683G and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;n. H655Y and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;o. R683G, D1118A, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;p. R683G, N715S, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;q. H655Y, N709S, and a deletion of the 13 amino acids (Δ13) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;r. F140V, H655Y, and a deletion of the 23 amino acids (Δ23) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;s. a deletion of the 21 amino acids (Δ21) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;t. a deletion of the 19 amino acids (Δ19) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;u. E484D, H655Y, and a deletion of 21 amino acids (Δ21) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1;v. H665Y, R685G and a deletion of the 19 amino acids (Δ19) at the C-terminal end of the amino acid sequence of SEQ ID NO: 1, wherein the SARS-CoV-2 S protein further comprises a fragment of at least 21 amino acids at the C-terminal end of a VSV-G protein;w. D614G; andx. D614N.
  • 17. The SARS-CoV-2 vaccine of claim 8, wherein the SARS-CoV-2 vaccine is formulated for oral administration.
  • 18. The SARS-CoV-2 vaccine of claim 8, wherein the SARS-CoV-2 vaccine is formulated for intranasal administration.
  • 19. The SARS-CoV-2 vaccine of claim 8, wherein the SARS-CoV-2 vaccine is formulated for oral mucosal administration and intranasal administration.
  • 20. A method for producing a recombinant VSV particle, comprising a. introducing the recombinant VSV vector of claim 1 into cells andb. producing a recombinant VSV particle and optionally cells producing a recombinant S protein.
  • 21. A recombinant cell comprising the recombinant VSV vector of claim 1.
  • 22. A method of generating an immune response against SARS-CoV-2 comprising a. administering to a subject an effective amount of the VSV vector of claim 1 andb. generating an immune response to SARS-CoV-2.
  • 23. The method of claim 22, wherein administering the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises intranasal administration.
  • 24. The method of claim 22, wherein administering the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises oral administration.
  • 25. The method of claim 22, wherein administering the recombinant VSV vector, recombinant VSV particle, immunogenic recombinant protein, or SARS-CoV-2 vaccine comprises oral mucosal administration.
GOVERNMENT RIGHTS IN INVENTION

This invention was made with Government support under HHSO100201600031C awarded by Assistant Secretary of Preparedness and Response, Biomedical Advanced Research and Development Authority (ASPR-BARDA). The Government has certain rights in the invention.

Provisional Applications (5)
Number Date Country
63143522 Jan 2021 US
63079944 Sep 2020 US
63005717 Apr 2020 US
62989128 Mar 2020 US
62979949 Feb 2020 US