CORONAVIRUS VACCINE AND METHODS OF USE THEREOF

Information

  • Patent Application
  • 20210393769
  • Publication Number
    20210393769
  • Date Filed
    June 08, 2021
    3 years ago
  • Date Published
    December 23, 2021
    2 years ago
Abstract
An immunogenic composition effective for eliciting an immune response against cells that present coronavirus S protein and/or coronavirus M protein derived antigens on a virus-like-particle (VLP) system. In a method embodiment, the antigen presenting VLP is administered to a mammal, such as a human, to elicit an immune response against coronavirus S protein and/or coronavirus M protein. A preferred method embodiment may include at least one additional dose of immunogenic composition to enhance the immune response effectiveness of the coronavirus vaccine.
Description
SEQUENCE LISTING

This document incorporates by reference an electronic sequence listing text file, which was electronically submitted along with this document. The text file is named 15660001AA_ST25, is 53 kilobytes, and was created on Jun. 7, 2021.


FIELD OF THE INVENTION

The invention generally relates to a coronavirus vaccine and particularly to an immunogenic composition comprising polypeptide antigens that are derived from conserved C-terminal domains of coronavirus spike protein (S protein). The composition may further comprise at least one polypeptide antigen derived from the first Heptad Repeat (HR1) and/or the second Heptad Repeat (HR2) domain of S protein and/or coronavirus membrane protein (M protein) and/or coronavirus receptor binding domain (RBD). The present invention also contemplates a vaccine composition comprising coronavirus antigens presented on a virus-like-particle (VLP). In addition, the invention relates to a method of inducing an immune response to coronavirus antigens to protect a subject from acquiring COVID-19.


BACKGROUND

The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused untold devastation worldwide. Similar to other human coronaviruses, such as the Middle East respiratory syndrome coronavirus (MERS-CoV) and SARS-CoV-1, SARS-CoV-2 may have a zoonotic origin. To date, 160 million people have been infected and 3.3 million have died worldwide, with mortality and morbidity still rising in various parts of the world. Apart from the medical and public health havoc, the SARS-CoV-2 pandemic has also caused unprecedented social, economic and governmental upheaval unseen in modern times.


As a result, there has been a thrust towards rapid development of vaccines and therapeutics to tackle this urgent problem. The prophylactic vaccines are based on diverse platforms that include RNA technology, non-replicating viral vectors, inactivated virus, subunit vaccines, as well as DNA vaccines. A number of therapeutic approaches using monoclonal antibodies have also been reported. However, there are limited pre-clinical data published on the SARS-CoV-2 vaccines under development. Long range safety studies have not yet been reported with test vaccines against SARS-CoV-2. In the case of therapeutic antibodies, based on published research, focus has primarily been on isolation techniques, structural models and mechanism of action of therapeutic antibodies against SARS-CoV-2. Further investigation on the safety aspects of either vaccines or therapeutic antibodies against SARS-CoV-2 is needed.


SARS-CoV-2 pathophysiology shows that the COVID-19 disease clinically manifests in multiple forms. Mild to moderate disease has been reported in 80% of cases, severe disease in ˜15% cases and critical disease in ˜5% cases. Subjects with mild disease recover with little to no medical intervention while severe and critical disease require hospitalization and, in some cases, intensive care. Some patients may suffer from life-threatening acute respiratory distress syndrome (ARDS) with possible fatal outcomes. Severe disease is seen more often in males, particularly in those with co-morbidities such as cardiovascular disease and diabetes. Paradoxically, patients with severe disease have increased IgG response and higher titers of total antibodies against SARS-CoV-2, which are associated with worse outcomes. As such, the role of immune cells such as macrophages, dendritic cells and T cells in COVID-19 disease has not been systematically studied. Antibody dependent enhancement (ADE) of SARS-CoV-2 may result in potential side effects in those patients who produce undesirable non-neutralizing antibodies against the virus.


SARS-CoV-2 infects cells through the ACE2 receptor expressed on type II pneumocytes in the lung. However, ACE2 is also expressed in the heart, kidney, intestine as well as the vascular endothelium. The major determinant of the ACE2 cognate binding is the homotrimeric spike glycoprotein (S) expressed as 70-80 copies on the surface of SARS-CoV-2. The S protein consists of two domains—S1 and S2—that are linked by a fusion peptide FP. S1 domain contains the ACE2 receptor binding domain (RBD) while the S2 domain promotes fusion of the virus into the cell membrane using a polybasic furin cleavage site located within the S1-S2 boundary.


Majority of the vaccine candidates currently being clinically tested make use of the S protein of SARS-CoV-2 alone as the main immunogen, mostly in a non-specific manner. In the case of non-replicating virus vectors, the S protein is cloned and expressed in adenovirus vectors. Similarly, for RNA based vaccines, the S protein mRNA is encapsulated in lipid nanoparticles and used for intramuscular injections. The DNA based vaccine also encodes the S protein and is electroporated into subjects for vaccinations. Thus, while initial attempts at developing vaccines and therapeutic antibodies against SARS-CoV-2 focused solely on the use of S protein, it is increasingly becoming clear that without rational considerations into immunogen design, formulation and manufacturer, clinical trials could get entrapped in safety pitfalls, thereby jeopardizing global efforts at controlling the pandemic.


Three major areas of concerns regarding safety of current vaccines against SARS-CoV-2 have emerged. One of the concerns of the currently developed vaccines is that some of the vaccines may trigger antibody dependent enhancement (ADE) of viral infection. ADE is a phenomenon wherein antibodies that are elicited against a viral antigen actually enhance the uptake of the virus, causing more severe disease. The enhancement is typically the result of non-neutralizing or sub-neutralizing antibodies generated against viral antigen (Iwasaki et. al., 2020). While the role of ADE in SARS-CoV-2 has not been fully established, several lines of evidence suggest that it could potentially be of significant safety concern in the clinic (Kamikubo et. al., 2020).


A second challenge to developing a safe vaccine is the need to reduce an off-target pathological activation of the immune cells. There is emerging evidence that TH17 responses can direct certain cellular responses upon vaccination with inactivated vaccines and those based on viral vectors, and that the TH17 activation leads to up-regulation of the pro-inflammatory cytokines IL-6 and IL-1β (Blanco-Melo et. al., 2020, Liu et. al., 2020). IL-6 upregulation has been prominently observed in patients with severe COVID-19 disease (Herold et. al., 2020). In rhesus macaques vaccinated with a modified vaccinia Ankara (MVA) virus encoding the SARS-CoV-1 S glycoprotein, anti-spike antibodies promoted MCP1 and IL-8 production in alveolar macrophages causing acute lung injury (Liu et. al., 2019).


An exemplary mouse study showed that, when animals were challenged with SARS-CoV-1 after immunization with either an inactivated viral vaccine or rDNA-based vaccine with or without alum, the Th2-type pathology with prominent eosinophil infiltration with eosinophil scores were significantly lower for non-vaccine groups than for vaccine groups of across the tested mouse strains (Tseng et. al., 2012). Spleen atrophy and lymph node necrosis have also been reported to be deceased in COVID-19 patients suggesting immune mediated pathology in SARS-CoV-2 infections (Feng et. al., 2020).


A third concern regarding of some vaccines against SARS-CoV-2 is a possibility of triggering endothelium inflammation. Endothelial cells express ACE2, the receptor bound by SARS-CoV-2 to infect lung epithelial cells. Not surprisingly, in additional to pulmonary complications, additional clinical symptoms of COVID-19 include high blood pressure, thrombosis and pulmonary embolism. This raises the question of whether the endothelium is a key target organ of SARS-CoV-2 (Sardu et. al., 2020). Of note, supernatants from SARS-CoV-2 infected capillary organoid cultures could also infect Vero cells demonstrating that the production of viable progeny virus. Postmortem analysis of a SARS-CoV-2 patient with hypertension showed evidence of direct viral infection of the endothelial cell and diffused endothelial inflammation causing vascular dysfunction by shifting the equilibrium towards vasoconstriction, ischemia and a procoagulant state (Fox et. al., 2020; Tian et. al., 2020; Varga et. al., 2020).


Therapies such as afucosylated or defucosylated monoclonal antibodies have been proposed, however, in some instances, such therapies are pathogenic.


Additionally, there is a great need to quickly develop and verify efficacy and safety of a vaccine for the new emergent SARS-CoV-2 virus variants, and subsequently manufacture the vaccine on a very large scale, to meet immediate population demands. Use of shorter antigenic peptides that are developed from computer-based rational designs provide several advantages in comparison to conventional vaccines made of dead or attenuated pathogens or inactivated toxins. This version of polypeptide antigens may be synthesized rapidly and produced with much lower cost.


Thus, there is a need in the art for an improved SARS-CoV-2 vaccine with effective prophylactic properties and ability to control potential side effects to the lowest degree or to none. Further, there is a need for diversifying new SARS-CoV-2 vaccine targets and more specifically to new targets based on the conserved regions for being effective against other potential variants, in the most cost effective and safety-oriented manner.


SUMMARY OF THE INVENTION

The disclosure relates to the field of viral vaccines and methods of use of such vaccines to protect a subject from virus infection. In particular, the invention is an immunogenic composition comprising at least one antigenic polypeptide and methods of use to evoke an immune response to one or more coronavirus spike (S) protein and/or membrane (M) protein to protect an immunized subject from acquiring a viral-related disease such as COVID-19. The antigenic polypeptides or fragments derived from the S protein and/or M protein may be included in the composition separately or fused by a linker region. The antigens are presented on the surface of Virus-Like Particles (VLP) for inhibiting the fusion of coronavirus particles during the viral entry to a host cell. One of the main advantageous features of the present invention is to selectively target the viral fusion stage by introducing at least one antigenic polypeptide or fragment derived from at least one conserved domain of the S protein from SARS-CoV-2 or any other coronaviruses with the conserved domain, such as the first heptad repeat (HR1) and/or the second heptad repeat (HR2), in which the at least one antigenic polypeptide is presented on the surface of VLP as a path to develop a safe and affordable vaccine. By presenting rationally designed antigens on VLPs, host immune responses that block viral fusion will result in prevention of unwanted cytokine production and immunopathology. The S protein derived antigens may be manufactured in eukaryotic cells (e.g., mammalian cells, plant cells, fungal cells, etc.), more preferably in mammalian cells, to ensure proper folding and glycosylation of viral proteins as in the human system.


One aspect of the invention is a vaccine composition that is able to induce an immune response against a coronavirus, comprising a VLP presenting at least one polypeptide or fragment of the coronavirus as an immunogen, wherein the at least one antigenic polypeptide or fragment may be derived from HR1 domain and/or HR2 domain of S protein and/or M protein from the coronavirus. In some embodiments, the at least one antigenic polypeptide or fragment is derived from both HR1 and HR2 domains and/or the linker regions between the HR1 and HR2 domains. In other embodiments, the at least one antigenic polypeptide or fragment may be derived from N-terminal or C-terminal region of the coronavirus M protein.


Another aspect of the invention is a vaccine composition that is able to induce an immune response to coronavirus and to inhibit viral fusion, wherein the at least one antigenic polypeptide or fragment presented on VLP has a sequence selected from the group consisting of SEQ ID NOs:1 to 166. In some embodiments, the VLP has a sequence as set forth in SEQ ID NO: 167 or SEQ ID NO: 168. In some embodiments, the at least one antigenic polypeptide includes a plurality of polypeptides as set forth in SEQ ID Nos 1-7, 83-102, and 135-150


Another aspect of the invention is a method of inducing an immune response to at least one coronavirus antigen in a subject in need thereof, comprising the step of administering an immunogenic composition comprising a VLP presenting at least one antigenic polypeptide or fragment derived from S protein and/or M protein to the subject. In some embodiments, the method further comprises allowing a suitable period of time to elapse and administering at least one additional dose of the immunogenic composition. The immunogenic composition comprising the vaccine can be administered intramuscularly or intradermally with a hypodermic, transdermic or intradermal needle or with a needle-free device. In some embodiments, the immunogenic composition comprising the vaccine can be administered by intranasal and/or ocular delivery. In some embodiments, at least one adjuvant may be included in the vaccine composition. In other embodiments, 1-150 μg of the antigen presenting VLP is administered to the subject in each dose. In addition, a suitable period of time is defined as a time sufficient for producing antibodies against the immunogenic composition of the present invention in a subject.


Other features and advantages of the present invention will be set forth in the description of invention that follows, and in part will be apparent from the description or may be learned by practice of the invention. The invention will be realized and attained by the compositions and methods particularly pointed out in the written description and claims hereof.





BRIEF DESCRIPTION OF THE DRAWINGS

The accompanying drawings, which are incorporated in and constitute a part of this specification, illustrate embodiments of the invention and, together with a general description of the invention given above and the detailed description given below serve to explain the invention.



FIG. 1 shows a structural organization of various domains within SARS-CoV-2 S protein. SS, signal sequence; S2′, S2′ protease cleavage site; FP, fusion peptide; HR1, heptad repeat 1; CH, central helix CD, connector domain; HR2, heptad repeat 2; TM, transmembrane domain; CT, cytoplasmic tail. The S protein also contains a predicted set of 24 N-glycans and 4 O-glycan for each monomer of which 17 N-glycans have been confirmed using cryo-EM.



FIG. 2 shows a sequence variability within N-terminal domains of SARS-CoV-2 S protein.



FIG. 3A-B show a front view (A) and a top view (B) of sequence conservation within C-terminal domains of SARS-CoV-2 S protein.



FIG. 4A-B show a 6-helix bundle fusion complex formed between HR1 and HR2, front view (A) and top view (B). The long HR1 helixes are packed in the interior of the bundle whereas the short HR2 helices are located on the exterior.



FIG. 5A-D show a composition of the proposed antigens SC2SHR1 (A), SC2HSHRL (B), SC2HSHR2 (C) and SC2HSHR2L (D) in the cases where high-resolution structures of S or M protein are available. 6VXX.PDB, 6LXT.PDB and 6M3W.PDB are shown in the figure.





DETAILED DESCRIPTION

Embodiments of the invention relate to an immunogenic composition comprising at least one antigenic polypeptide or fragment thereof, wherein the at least one polypeptide or fragment antigen thereof is derived from coronavirus spike (S) protein and/or membrane (M) protein. In particular, HR1 domain and/or HR2 domain of SARS-CoV-2 S protein is contemplated as the protein in which at least one antigenic polypeptide or fragment thereof is derived from. When incorporated together, the polypeptide or fragment derived from HR1 and HR2 may be fused, chemical and/or bio-conjugated to each other by a linker region and presented on the surface of Virus-Like-Particles (VLP) for inhibiting the fusion of SARS-CoV-2 virus during the viral entry to a host cell. The disclosure also relates to a method of use of such immunogenic composition to protect a subject from COVID-19 infection.


The vast majority of the vaccines that are currently being developed target the spike (S) protein of SARS-CoV-2 in a non-specific manner, opening the possibilities of unwanted immune responses and pathologies clinically reported in severe and critical cases of COVID-19. In particular, acute lung injury, immunopathology and endothelial dysfunction have been described in deceased patients, raising questions about the fundamental mechanism of COVID-19 etiology. In the present disclosure, an alternate strategy based on rational design that selectively targets blockade of viral fusion as a path to develop a safe and affordable vaccine against SARS-CoV-2 is described. The present invention displays antigenic polypeptides or fragments that are derived from specific, membrane-proximal, conserved domains within the SARS-CoV-2 S protein as well as from membrane glycoprotein as immunogens. By presenting rationally designed antigens on VLP, host immune responses that block viral fusion will result in prevention of unwanted cytokine production and immunopathology. Manufacture in mammalian cells ensures proper folding and glycosylation of viral proteins as in the human system, further strengthening vaccine safety. Combined with proven, scalable, cost-effective manufacturing technologies, the present invention may provide a shelf life and lower cost of goods for people living in low-income countries.


In preferred embodiments, a vaccine composition of the present invention induces an immune response to coronavirus and inhibits viral fusion. The immunogenic composition comprises at least one antigenic polypeptide or fragment thereof. In preferred embodiments, one or more antigens are presented on VLP. In other embodiments the antigens may be displayed or included in a VLP by a plurality of methods described below. In some embodiments, the antigenic polypeptide or fragment has 80% or more, preferably 90% or more, more preferably 95% or more sequence identity with a sequence selected from the group consisting of SEQ ID NOs:1 to 166. In some embodiments, the VLP has 80% or more, preferably 90% or more, more preferably 95% or more sequence identity with a sequence selected from the group consisting of SEQ ID NOs:167-168.


For each cDNA sequence presented herein, the invention includes the mRNA equivalent of the cDNA, meaning that the invention includes each cDNA sequence wherein each T is replaced by U. Exemplary antigenic peptide sequences include:


Amino Acids 1-31 Covering Amino Terminus of M Protein:











(SEQ ID NO: 151)



MADSNGTITVEELKKLLEQWNLVIGFLFLTW






Corresponding DNA Sequence:









(SEQ ID NO: 152)


ATGGCTGATTCTAATGGGACAATTACAGTCGAAGAGCTTAAGAAATC





GCTGGAGCAATGGAATCTTGTTATTGGCTTTCTCTTCCTGACCTGG







Amino Acids 130-160 from Carboxy Terminus of M Protein:











(SEQ ID NO: 153)



TRPLLESELVIGAVILRGHLRIAGHHLGRCD






Corresponding DNA Sequence:









(SEQ ID NO: 154)


ACCCGACCCCTCCTTGAATCCGAACTTGTTATTGGCGCTGTCATTCT





CCGCGGACACCTTAGAATTGCTGGACATCACCTTGGACGCTGTGAT






Amino Acids 913-984 Covering the First Heptad Repeat HR1 of S Protein:











(SEQ ID NO: 155)



QNVLYENQKLIANQFNSAIGKIQDSLSSTASALGKL







QDVVNQNAQALNTLVKQLSSNFGAISSVLNDILSRL






Corresponding DNA Sequence:











(SEQ ID NO: 156)



CAGAATGTCCTTTACGAAAATCAGAAA







CTCATCGCAAATCAATTCAACTCAGCA







ATCGGAAAAATTCAAGATTCCCTCTCT







TCTACCGCATCTGCTCTTGGCAAGCTT







CAAGATGTCGTCAATCAAAATGCCCAA







GCTCTCAACACTCTCGTTAAACAACTC







TCTTCTAACTTTGGTGCCATATCCTCC







GTCCTCAATGATATCCTTTCCCGCCTG






Amino Acids 1147-1170 Covering the Linker Region Connecting HR1 and HR2:











(SEQ ID NO: 157)



SFKEELDKYFKNHTSPDVDLGDIS






Corresponding DNA Sequence:











(SEQ ID NO: 158)



AGTTTCAAAGAAGAACTTGACAAATACT






TTAAAAATCATACCTCCCCTGACGTGG






ACCTTGGCGATATCTCC






Amino Acids 1171-1212 Covering the Second Heptad Repeat HR2 of S Protein:











(SEQ ID NO: 159)



GINASVVNIQKEIDRLNEVAKNLNESLIDLQELGKYEQYIKW






Corresponding DNA Sequence:











(SEQ ID NO: 160)



GGAATTAACGCATCCGTGGTTAACATA






CAGAAAGAAATTGACAGACTGAACGAA






GTGGCCAAGAACCTTAACGAATCTCTC






ATAGACCTTCAAGAGCTGGGAAAGTAC






GAACAATACATAAAATGG







Amino acids 1147-1212 covering both the linker as well as HR2 regions:











(SEQ ID NO: 161)



SFKEELDKYFKNHTSPDVDLGDLSGINASVVNIQ






KEIDRLNEVAKNLNESLIDLQELGKYEQYIKW






Corresponding DNA Sequence:











(SEQ ID NO: 162 )



AGTTTTAAGGAAGAACTGGACAAATATTTCAAGAATCATA






CATCTCCAGACGTGGACCTGGGCGACATTTCTGGCATTAA






CGCATCCGTGGTTAACATTCAAAAAGAAATTGATAGACTG






AACGAAGTGGCTAAAAATCTGAACGAGTCCCTTATCGATC






TGCAGGAATTGGGAAAATACGAGCAATACATCAAATGG






Amino Acids 318-541 Representing RBD of S Protein:











(SEQ ID NO: 163)



GFRVQPTESIVRFPNITNLCPPGEVFNATRFASVYAWNR






KRISNCVADYSVLYNSASFSTFKCYGVSPTKLNDLCFTN






VYADSFVIRGDEVRQIAPGQTGKIADYNYKLPDDFTGCV






IAWNSNNLDSKVGGNYNYLYRLFRKSNLKPFERDISTEI






YQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVV






VLSFELLHAPATVCGPKKSTNLVKNFCCVNFNFNGLTGT






GVLTESNKKFLPFQQFGRDIADTTDAVRDPQT






Corresponding DNA Sequence:











(SEQ ID NO: 164)



GGCTTCAGAGTCCAACCAACAGAGTCCATCGTGAGGTTC






CCCAATATTACTAATCTGTGCCCCTTTGGTGAGGTGTTT






AATGCTACCAGATTTGCCTCTGTCTATGCATGGAATCGG






AAGCGGATTAGTAACTGCGTCGCCGACTATAGTGTTCTC






TATAATTCCGCTAGTTTCTCTACGTTCAAATGCTATGGC






GTCTCCCCGACAAAGCTCAATGACTTGTGTTTCACTAAC






GTCTACGCTGATTCTTTCGTGATCCGCGGTGATGAAGTG






CGCCAGATCGCCCCAGGACAAACCGGAAAAATCGCTGAT






TACAATTACAAACTCCCCGACGACTTCACCGGCTGCGTT






ATTGCCTGGAACTCTAACAATCTGGACAGCAAGGTTGGC






GGCAATTATAACTATCTGTACCGCCTGTTTCGGAAGTCA






AATCTCAAACCATTCGAACGCGATATTAGTACAGAAATC






TATCAGGCTGGCAGCACCCCCTGTAACGGTGTTGAAGGG






TTTAATTGTTATTTCCCTCTCCAATCATACGGTTTCCAG






CCCACAAACGGCGTTGGGTACCAACCATACCGAGTCGTT






GTTCTGTCTTTTGAACTTCTCCATGCTCCAGCTACTGTT






TGTGGACCGAAGAAGAGCACCAATCTTGTCAAAAATAAA






TGCGTGAATTTTAACTTCAATGGTCTTACAGGTACCGGC






GTGCTTACCGAAAGTAACAAAAAATTTCTCCCTTTTCAG






CAGTTCGGACGAGACATTGCAGATACCACCGACGCCGTG






AGAGATCCACAGACC







Amino Acids 318-541 Representing RBD of S Protein with K417N/E484K/N501Y Mutations:











(SEQ ID NO: 165)



GFRVQPTESIVRFPNITNLCPFGEVFNATRFASVYAWNR






KRISNCVADYSVLYNSASFSTFKCYGVSPTKLNDLCFTN






VYADSFVIRGDEVRQIAPGQTGNIADYNYKLPDDFTGCV






IAWNSNNLDSKVGGNYNYLYRLFRKSNLKPFERDISTEI






YQAGSTPCNGVKGFNCYFPLQSYGFQPTYGVGYQPYRVV






VLSFELLHAPATVCGPKKSTNLVKNKCVNFNFNGLTGTG






VLTESNKKFLPFQQFGRDIADTTDAVRDPQT






Corresponding DNA Sequence:











(SEQ ID NO: 166)



GGCTTCAGAGTCCAACCAACAGAGTCCATCGTGAGGTTC






CCCAATATTACTAATCTGTGCCCCTTTGGTGAGGTGTTT






AATGCTACCAGATTTGCCTCTGTCTATGCATGGAATCGG






AAGCGGATTAGTAACTGCGTCGCCGACTATAGTGTTCTC






TATAATTCCGCTAGTTTCTCTACGTTCAAATGCTATGGC






GTCTCCCCGACAAAGCTCAATGACTTGTGTTTCACTAAC






GTCTACGCTGATTCTTTCGTGATCCGCGGTGATGAAGTG






CGCCAGATCGCCCCAGGACAAACCGGAAATATCGCTGAT






TACAATTACAAACTCCCCGACGACTTCACCGGCTGCGTT






ATTGCCTGGAACTCTAACAATCTGGACAGCAAGGTTGGC






GGCAATTATAACTATCTGTACCGCCTGTTTCGGAAGTCA






AATCTCAAACCATTCGAACGCGATATTAGTACAGAAATC






TATCAGGCTGGCAGCACCCCCTGTAACGGTGTTAAAGGG






TTTAATTGTTATTTCCCTCTCCAATCATACGGTTTCCAG






CCCACATACGGCGTTGGGTACCAACCATACCGAGTCGTT






GTTCTGTCTTTTGAACTTCTCCATGCTCCAGCTACTGTT






TGTGGACCGAAGAAGAGCACCAATCTTGTCAAAAATAAA






TGCGTGAATTTTAACTTCAATGGTCTTACAGGTACCGGC






GTGCTTACCGAAAGTAACAAAAAATTTCTCCCTTTTCAG






CAGTTCGGACGAGACATTGCAGATACCACCGACGCCGTG






AGAGATCCACAGACC






Exemplary VLP Amino Acid Sequence:











(SEQ ID NO: 167)



GGGGGGMENITSGFLGPLLVLQAGFFLLTRILTIPQSLDSW






WTSLNFLGGSPVCLGQNSQSPTSNHSPTSCPPICPGYRWMC






RRRFIIFLFILLLCLIFLLVLLDYQGMLPVCPLIPGSTTTS






TGPCKTCTTPAQGNSMFPSCCCTKPTDGNCTCIPIPSSWAF






AKYLWEWASVRFSWVSLLVPFVQWFVGLSPTVWLSAIWMMW






YWGPSLYSIVSPFIPLLPILFCLWVYI






Exemplary VLP DNA Sequence:











(SEQ ID NO: 168)



GGCGGCGGCGGCGGAGGCATGGAAAATATCACTTCTGGATT






TCTGGGGCCCCTCCTGGTTCTGCAGGCAGGCTTTTTCCTTT






TGACACGCATCCTGACTATCCCACAATCCCTTGACTCATGG






TGGACATCACTGAACTTCCTTGGCGGTTCTCCCGTTTGCCT






TGGCCAGAATTCCCAGTCACCCACTTCTAATCATTCTCCCA






CATCTTGCCCTCCTATCTGCCCAGGCTACCGATGGATGTGC






AGAAGACGCTTCATTATCTTCCTGTTCATTTTGCTGCTGTG






TCTGATCTTTCTCTTGGTCTTGCTTGATTATCAAGGCATGT






TGCCCGTGTGTCCCCTCATTCCAGGATCAACAACGACTTCC






ACAGGCCCCTGCAAAACGTGCACCACACCAGCCCAAGGAAA






TAGCATGTTCCCCTCTTGCTGTTGCACTAAACCTACGGACG






GCAACTGTACCTGTATCCCGATACCCTCTTCTTGGGCTTTT






GCTAAATATCTCTGGGAATGGGCTTCCGTCAGATTCTCTTG






GGTGAGCCTTCTTGTCCCCTTCGTGCAATGGTTCGTTGGAC






TCAGTCCTACCGTTTGGCTCAGCGCAATCTGGATGATGTGG






TACTGGGGACCATCTCTCTACAGCATTGTTTCACCCTTTAT






CCCCCTGCTTCCAATCTTGTTCTGCTTGTGGGTTTATATAT






AAACGCGT






Coronaviruses constitute the subfamily Orthocoronavirinae, in the family Coronaviridae, order Nidovirales, and realm Riboviria. Coronaviruses have four genera: alpha-, beta-, gamma-, and delta-coronaviruses. They are enveloped viruses with a positive-sense single-stranded RNA genome and a nucleocapsid of helical symmetry. The genome size of coronaviruses ranges from approximately 26 to 32 kilobases. Exemplary coronaviruses that may be treated with the compositions of the disclosure include, but are not limited to, SARS-Cov, SARS-Cov-2, MERS-Cov, HCoV-OC43, HCoV-HKU1, HCoV-229E, and HCoV-NL63.


As used herein, the term “severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)”, “2019 novel coronavirus (2019-nCoV)”, “human coronavirus 2019 (HCoV-19)” or “severe acute respiratory syndrome-related coronavirus (SARSr-CoV)” refers to virus comprising a virion with 50-200 nanometers in diameter and a genomic size of about 30 kilobases, encoding multiple structural proteins, such as the S (spike), E (envelope), M (membrane) and N (nucleocapsid), and non-structural proteins. Coronaviruses are a group of related RNA viruses that cause diseases in mammals and birds. In humans and birds, they cause respiratory tract infections that can range from mild to lethal. Mild illnesses in humans include some cases of the common cold (which is also caused by other viruses, predominantly rhinoviruses), while more lethal varieties can cause SARS, MERS, and COVID-19.


Since SARS-CoV-2 shares 80% sequence homology with SARS-CoV-1, the antibodies against S protein in SARS-CoV-2 may also trigger immune response against SARS-Cov-1. Thus, other virus types such as SARS-CoV (i.e., SARS-CoV-1) and MERS-CoV that are similar in virion structure may also be subjected to the present invention. As used herein, the term “S protein” or “Spike protein” is used to refer to a knoblike structured (i.e., spikes) peplomer, which is composed of glycoprotein to project from the lipid bilayer of the surface envelope of an enveloped virus. The “spike protein” or “S protein” is interchangeably referred to a protein and/or a glycoprotein. Furthermore, the sequences encoding the SARS-CoV-2 glycoprotein may also be referred to as a peptide or amino acid sequence.


As used herein, the terms “polypeptide”, “short protein”, “fragment of protein”, “polypeptide or fragment”, “antigenic polypeptide or fragment” and “peptide” are used interchangeably and refer to chains of amino acids comprising between 2, or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10, or 11, or 12, or 13, or 14, or 15 and 10, or 11, or 12, or 13, or 14, or 15, or 20, or 25, or 30, or 35, or 40, or 45, or 50 or 55 or 60 or 100 or 150 or 200 or 300 or 350 or 400 or 500 amino acids. As used herein, the term “epitope” or “T cell epitope” refers to a sequence of contiguous amino acids contained within a protein antigen that possess a binding affinity to a T cell receptor when presented on the surface of antigen presenting cells. An epitope is antigen-specific but not individual specific. An epitope, a T cell epitope, a polypeptide, a fragment of a polypeptide or a composition comprising a polypeptide or a fragment thereof is “immunogenic” for a specific human individual if it is capable of inducing an immune cell response in that individual. In some embodiments, an “immune response”, “T cell response” or “immunogenic response” are used interchangeably and may further include an antibody response. As used historically, the term “antigen” is used to designate an entity that is bound by an antigen-specific antibody or B-cell antigen receptor.


As used herein, the term “antigens”, “proteins”, “peptides”, “polypeptides”, “fragments”, or “epitopes” may be used interchangeably. In particular, an “antigen” refers to a molecule containing one or more epitopes (either linear, conformational or both) that will stimulate a host's immune-system to make a humoral and/or cellular antigen-specific response. The term is used interchangeably with the term “immunogen”. Further, antigenic polypeptide or fragment “derived from” a particular viral protein or protein domain refers to a full-length or near full-length viral protein or domain, as well as a fragment thereof, or a viral protein with internal deletions. Accordingly, the polypeptide may comprise the full-length sequence, fragments, truncated and partial sequences, as well as analogs and precursor forms of the reference molecule. In addition, the term “derived” may refer to construction of a peptide based on the knowledge of a representative protein domain sequence using any one of several suitable means, including, by way of example, isolation or synthesis. Thus, the term includes variations of the specified polypeptide.


In some embodiments, the antigenic peptides as described herein are 5 to 150 residues in length, e.g. 10 to 100 residues. The antigenic peptides may include consecutive or nonconsecutive sequences from the coronavirus viral domains.


The antigenic peptides described herein may comprise epitopes, i.e. amino acids that bind to an antibody generated in response to such sequence. An epitope for use in the subject invention is not limited to a polypeptide having the exact sequence of the portion of the parent protein from which it is derived. Indeed, viral genomes are in a state of constant flux and contain several variable domains which exhibit relatively high degrees of variability between isolates. Thus, the term “epitope” encompasses sequences identical to the native sequence, as well as modifications to the native sequence, such as deletions, additions, and substitutions (generally conservative in nature).


As used herein, the term “conformational epitope” refers to a recombinant epitope having structural features native to the amino acid sequence encoding the epitope within the full-length natural protein. Native structural features include, but are not limited to, glycosylation and three-dimensional structure. The length of the epitope-defining sequence can be subject to wide variations as these epitopes are believed to be formed by the three-dimensional shape of the antigen (e.g., folding). Thus, amino acids defining the epitope can be relatively few in number, but widely dispersed along the length of the molecule (or even on different molecules in the case of dimers, etc.), being brought into correct epitope conformation via folding. The portions of the antigen between the residues defining the epitope may not be critical to the conformational structure of the epitope. For example, deletion or substitution of these intervening sequences may not affect the conformational epitope provided sequences critical to epitope conformation are maintained (e.g., cysteines involved in disulfide bonding, glycosylation sites, etc.).


As used herein, the term “Virus-Like Particle (VLP)” refers to molecules that closely resembles viruses but are lacking viral genetic materials. Use of VLPs derived from the Hepatitis B virus (HBV) and composed of the small HBV derived surface antigen (HBsAg) are well known in the art. A plurality of detailed VLP formation and release methods are described in U.S. Pat. Nos. 7,951,384 and 9,352,031, herein incorporated by reference. In some embodiments, the present invention includes VLP presenting S protein derived antigens on its surface. In other embodiments, the VLP displays S protein and M protein derived antigens. In other embodiments, the VLP displays two or more domains of S protein (i.e., HR1 and HR2 domains and the linker region between the HR1 and HR2 domains) and M protein derived antigens.


In order to increase the immunogenicity of the composition, in some embodiments, the immunogenic compositions comprise one or more adjuvants and/or cytokines. Suitable adjuvants include an aluminum salt such as aluminum hydroxide or aluminum phosphate, but may also be a salt of calcium, iron or zinc, or may be an insoluble suspension of acylated tyrosine, or acylated sugars, or may be cationically or anionically derivatized saccharides, polyphosphazenes, biodegradable microspheres, monophosphoryl lipid A (MPL), lipid A derivatives (e.g. of reduced toxicity), 3-O-deacylated MPL [3D-MPL], quil A, Saponin, QS21, Freund's Incomplete Adjuvant (Difco Laboratories, Detroit, Mich.), Merck Adjuvant 65 (Merck and Company, Inc., Rahway, N.J.), AS-2 (Smith-Kline Beecham, Philadelphia, Pa.), CpG oligonucleotides, bioadhesives and mucoadhesives, microparticles, liposomes, polyoxyethylene ether formulations, polyoxyethylene ester formulations, muramyl peptides or imidazoquinolone compounds (e.g. imiquamod and its homologues). Human immunomodulators suitable for use as adjuvants in the disclosure include cytokines such as interleukins (e.g. IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF), granulocyte, macrophage colony stimulating factor (GM-CSF) may also be used as adjuvants.


In some embodiments, the compositions comprise an adjuvant selected from the group consisting of Montanide ISA-51 (Seppic, Inc., Fairfield, N.J., United States of America), QS-21 (Aquila Biopharmaceuticals, Inc., Lexington, Mass., United States of America), GM-CSF, cyclophosamide, bacillus Calmette-Guerin (BCG), corynbacterium parvum, levamisole, azimezone, isoprinisone, dinitrochlorobenezene (DNCB), keyhole limpet hemocyanins (KLH), Freunds adjuvant (complete and incomplete), mineral gels, aluminum hydroxide (Alum), lysolecithin, pluronic polyols, polyanions, oil emulsions, dinitrophenol, diphtheria toxin (DT). In some embodiments, the adjuvant is Montanide adjuvant. It is expected that an adjuvant or cytokine can be added in an amount of about 0.01 mg to about 10 mg per dose, preferably in an amount of about 0.2 mg to about 5 mg per dose. Alternatively, the adjuvant or cytokine may be at a concentration of about 0.01 to 50%, preferably at a concentration of about 2% to 30%.


In certain aspects, the immunogenic compositions of the disclosure are prepared by physically mixing the adjuvant and/or cytokine with peptides described herein under appropriate sterile conditions in accordance with known techniques to produce the final product. The dose may be determined according to various parameters, especially according to the substance used; the age, weight and condition of the individual to be treated; the route of administration; and the required regimen. The amount of antigen in each dose is selected as an amount which induces an immune response. A physician will be able to determine the required route of administration and dosage for any particular individual. The dose may be provided as a single dose or may be provided as multiple doses, for example taken at regular intervals, for example 2, 3 or 4 doses administered weekly. Typically, peptides, polynucleotides or oligonucleotides are typically administered in the range of 1 pg to 1 mg, more typically 1 pg to 10 μg for particle mediated delivery and 1 μg to 1 mg, and more typically 1-150 μg. Generally, it is expected that each dose will comprise 0.01-3 mg of antigen. An optimal amount for a particular vaccine can be ascertained by studies involving observation of immune responses in individuals.


In preferred embodiments, the immunogenic composition contains a SARS-CoV-2 antigen VLP. Alternatively, in other embodiments, HR1 and HR2 domains of S protein and/or M protein derived antigens may exist as nucleic acids and may be formulated as a DNA vaccine. In some embodiments, DNA vaccines, or gene vaccines, comprise a plasmid with a promoter and appropriate transcription and translation control elements and a nucleic acid sequence encoding one or more polypeptides of the disclosure. In some embodiments, the plasmids also include sequences to enhance, for example, expression levels, intracellular targeting, or proteasomal processing. In some embodiments, DNA vaccines comprise a viral vector containing a nucleic acid sequence encoding one or more polypeptides of the disclosure. In additional aspects, the compositions disclosed herein comprise one or more nucleic acids encoding peptides determined to have immunoreactivity with a biological sample. For example, in some embodiments, the compositions comprise one or more nucleotide sequences encoding 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more peptides comprising a polypeptide or a fragment that is a fusion antigen derived from S protein and M protein of SARS-CoV-2 coronavirus. The DNA or gene vaccine also encodes immunomodulatory molecules to manipulate the resulting immune responses, such as enhancing the potency of the vaccine, stimulating the immune system or reducing immunosuppression. Strategies for enhancing the immunogenicity of DNA or gene vaccines include encoding of xenogeneic versions of antigens, fusion of antigens to molecules that activate T cells or trigger associative recognition, priming with DNA vectors followed by boosting with viral vector, and utilization of immunomodulatory molecules. In some embodiments, the DNA vaccine is introduced by a needle, a gene gun, an aerosol injector, with patches, via microneedles, by abrasion, among other forms. In some forms the DNA vaccine is incorporated into liposomes or other forms of nanobodies. In some embodiments, the DNA vaccine includes a delivery system selected from the group consisting of a transfection agent; protamine; a protamine liposome; a polysaccharide particle; a cationic nanoemulsion; a cationic polymer; a cationic polymer liposome; a cationic nanoparticle; a cationic lipid and cholesterol nanoparticle; a cationic lipid, cholesterol, and PEG nanoparticle; a dendrimer nanoparticle. In some embodiments, the DNA vaccines is administered by inhalation or ingestion. In some embodiments, the DNA vaccine is introduced into the blood, the thymus, the pancreas, the skin, the muscle, a tumor, or other sites.


In yet other embodiments, the composition may be prepared as an RNA vaccine. In some embodiments, the RNA is non-replicating mRNA or virally derived, self-amplifying RNA. In some embodiments, the non-replicating mRNA encodes the peptides disclosed herein and contains 5′ and 3′ untranslated regions (UTRs). In some embodiments, the virally derived, self-amplifying RNA encodes not only the peptides disclosed herein but also the viral replication machinery that enables intracellular RNA amplification and abundant protein expression. In some embodiments, the RNA is directly introduced into the individual. In some embodiments, the RNA is chemically synthesized or transcribed in vitro. In some embodiments, the mRNA is produced from a linear DNA template using a T7, a T3, or a Sp6 phage RNA polymerase, and the resulting product contains an open reading frame that encodes the peptides disclosed herein, flanking UTRs, a 5′ cap, and a poly(A) tail. In some embodiments, various versions of 5′ caps are added during or after the transcription reaction using a vaccinia virus capping enzyme or by incorporating synthetic cap or anti-reverse cap analogues. In some embodiments, an optimal length of the poly(A) tail is added to mRNA either directly from the encoding DNA template or by using poly(A) polymerase. In some embodiments, the fragments are derived from an antigen that is expressed within the sequence of S and/or M proteins of SARS-CoV-2. In some embodiments, the RNA includes signals to enhance stability and translation. In some embodiments, the RNA also includes unnatural nucleotides to increase the half-life or modified nucleosides to change the immunostimulatory profile.


In some embodiments, the RNA is introduced by a needle, a gene gun, an aerosol injector, with patches, via microneedles, by abrasion, among other forms. In some forms the RNA vaccine is incorporated into liposomes or other forms of nanobodies that facilitate cellular uptake of RNA and protect it from degradation. In some embodiments, the RNA vaccine includes a delivery system selected from the group consisting of a transfection agent; protamine; a protamine liposome; a polysaccharide particle; a cationic nanoemulsion; a cationic polymer; a cationic polymer liposome; a cationic nanoparticle; a cationic lipid and cholesterol nanoparticle; a cationic lipid, cholesterol, and PEG nanoparticle; a dendrimer nanoparticle; and/or naked mRNA; naked mRNA with in vivo electroporation; protamine-complexed mRNA; mRNA associated with a positively charged oil-in-water cationic nanoemulsion; mRNA associated with a chemically modified dendrimer and complexed with polyethylene glycol (PEG)-lipid; protamine-complexed mRNA in a PEG-lipid nanoparticle; mRNA associated with a cationic polymer such as polyethylenimine (PEI); mRNA associated with a cationic polymer such as PEI and a lipid component; mRNA associated with a polysaccharide (for example, chitosan) particle or gel; mRNA in a cationic lipid nanoparticle (for example, 1,2 dioleoyloxy 3 trimethylammoniumpropane (DOTAP) or dioleoylphosphatidylethanolamine (DOPE) lipids); mRNA complexed with cationic lipids and cholesterol; or mRNA complexed with cationic lipids, cholesterol and PEG-lipid. In some embodiments, the RNA vaccine is administered by inhalation or ingestion. In some embodiments, the RNA is introduced into the blood, the thymus, the pancreas, the skin, the muscle, a tumor, or other sites, and/or by an intradermal, intramuscular, subcutaneous, intranasal, intranodal, intravenous, intrasplenic, intratumoral or other delivery route.


In some embodiments, the polynucleotide or oligonucleotide components are naked nucleotide sequences or be in combination with cationic lipids, polymers or targeting systems. They may be delivered by any available technique. For example, the polynucleotide or oligonucleotide may be introduced by needle injection, preferably intradermally, subcutaneously or intramuscularly. Alternatively, the polynucleotide or oligonucleotide may be delivered directly across the skin using a delivery device such as particle-mediated gene delivery. The polynucleotide or oligonucleotide may be administered topically to the skin, or to mucosal surfaces for example by intranasal, oral, or intrarectal administration. Uptake of polynucleotide or oligonucleotide constructs may be enhanced by several known transfection techniques, for example those including the use of transfection agents. Examples of these agents include cationic agents, for example, calcium phosphate and DEAE-Dextran and lipofectants, for example, lipofectam and transfectam. The dosage of the polynucleotide or oligonucleotide to be administered can be altered.


The immunogenic compositions or vaccines described herein comprise, in addition to one or more peptides, nucleic acids or vectors, a pharmaceutically acceptable excipient, carrier, diluent, buffer, stabilizer, preservative, adjuvant or other materials well known to those skilled in the art. Such materials are preferably non-toxic and preferably do not interfere with the pharmaceutical activity of the active ingredient(s). The pharmaceutical carrier or diluent may be, for example, water containing solutions. The precise nature of the carrier or other material may depend on the route of administration, e.g., oral, intravenous, cutaneous or subcutaneous, nasal, intramuscular, intradermal, and intraperitoneal routes. In some embodiments, the pharmaceutical compositions of the disclosure comprise one or more “pharmaceutically acceptable carriers”. These are typically large, slowly metabolized macromolecules such as proteins, saccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, sucrose (Paoletti et. al., 2001, Vaccine, 19:2118-2126), trehalose (WO 00/56365), lactose and lipid aggregates (such as oil droplets or liposomes). Such carriers are well known to those of ordinary skill in the art. In some embodiments, the pharmaceutical compositions contain diluents, such as water, saline, glycerol, etc. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present. Sterile pyrogen-free, phosphate buffered physiologic saline is a typical carrier (Gennaro et. al., 2000, Remington: The Science and Practice of Pharmacy, 20th edition, ISBN:0683306472).


In some embodiments, the immunogenic compositions of the disclosure are lyophilized or in aqueous form, i.e., solutions or suspensions. Liquid formulations of this type allow the compositions to be administered direct from their packaged form, without the need for reconstitution in an aqueous medium, and are thus ideal for injection. In some embodiments, the immunogenic compositions are presented in vials, or they may be presented in ready filled syringes. The syringes may be supplied with or without needles. A syringe will include a single dose, whereas a vial may include a single dose or multiple doses. Liquid formulations of the disclosure are also suitable for reconstituting other medicaments from a lyophilized form. Where a immunogenic composition is to be used for such extemporaneous reconstitution, the disclosure provides a kit, which may comprise two vials, or may comprise one ready-filled syringe and one vial, with the contents of the syringe being used to reconstitute the contents of the vial prior to injection.


In some embodiments, the immunogenic compositions of the disclosure include an antimicrobial, particularly when packaged in a multiple dose format. Antimicrobials may be used, such as 2-phenoxyethanol or parabens (methyl, ethyl, propyl parabens). Any preservative is preferably present at low levels. Preservative may be added exogenously and/or may be a component of the bulk antigens which are mixed to form the composition (e.g., present as a preservative in pertussis antigens).


In some embodiments, the immunogenic compositions of the disclosure comprise detergent e.g., Tween (polysorbate), DMSO (dimethyl sulfoxide), DMF (dimethylformamide). Detergents are generally present at low levels, e.g., <0.01%, but may also be used at higher levels, e.g., 0.01-50%. In some embodiments, the immunogenic compositions of the disclosure include sodium salts (e.g., sodium chloride) and free phosphate ions in solution (e.g., by the use of a phosphate buffer).


In some embodiments, the immunogenic compositions are encapsulated in a suitable vehicle either to deliver the peptides into antigen presenting cells or to increase the stability. As will be appreciated by a skilled artisan, a variety of vehicles are suitable for delivering a immunogenic composition of the disclosure. Non-limiting examples of suitable structured fluid delivery systems may include nanoparticles, liposomes, microemulsions, micelles, dendrimers and other phospholipid-containing systems. Methods of incorporating immunogenic compositions into delivery vehicles are known in the art.


Other aspects of the present invention relate to methods of inducing an immune response to at least one coronavirus polypeptide or fragment antigen as described above in a subject in need thereof, comprising the steps of: administering to the subject an effective amount of the immunogenic composition of antigenic polypeptide or fragments derived from coronavirus S protein, coronavirus M protein or combinations thereof; allowing a suitable period of time to elapse; and optionally administering at least one additional dose of the immunogenic composition. A “suitable period of time” is defined herein as a sufficient time for a subject to produce antibodies against the administered antigens described herein. A sufficient time for a subject to acquire ability to produce antibodies may be days (e.g., 2, 3, 4, 5, 6 or 7 days), weeks (e.g., 1, 2, 3 or 4 weeks), months (e.g., 1, 2, 3, 4, 5, or 6 months), or years (e.g. 1, 2, 3, 4, or 5 years) after a first, second or third dose of the immunogenic composition is administered.


As used herein, the term “effective amount” refers to an amount of VLPs necessary or sufficient to realize a desired biologic effect. An effective amount of the composition would be the amount that achieves a selected result, and such an amount could be determined as a matter of routine experimentation by a person skilled in the art. For example, an effective amount for preventing, treating and/or ameliorating an infection could be that amount necessary to cause activation of the immune system, resulting in the development of an antigen specific immune response upon exposure to VLPs of the invention. The term is also synonymous with “sufficient amount” or “therapeutically effective amount”.


In some embodiments, the coronavirus polypeptide or fragment antigen has a sequence identity selected from the group consisting of SEQ ID NOs:1 to 166.


In some embodiments, the method of treatment comprises administration to an individual of more than one peptide, polynucleic acid in a VLP or vector. These may be administered together/simultaneously and/or at different times or sequentially. The use of combinations of different peptides, optionally targeting different antigens, may be important to overcome the challenges of viral or individual heterogeneity. The use of peptides of the disclosure in combination expands the group of individuals who can experience clinical benefit from vaccination. Multiple immunogenic compositions, manufactured for use in one regimen, may define a drug product. In some cases, different peptides, polynucleic acids or vectors of a single treatment may be administered to the individual within a period of, for example, 1 year, or 6 months, or 3 months, or 60 or 50 or 40 or 30 days.


Preferably, the sequence employed to form the VLP immunogenic composition exhibits between about 60-80% sequence identity to a naturally occurring coronavirus polynucleotide or polypeptide sequence or conformational epitope sequence and more preferably the sequences exhibit between about 80-100% sequence identity, e.g., at least about 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to a naturally occurring polynucleotide or polypeptide sequence or a sequence as disclosed herein. In addition, the sequences described herein can be operably linked to each other in any combinations. For example, one or more sequences may be expressed from the same promoter and/or from different promoters.


Routes of administration include but are not limited to ocular, intranasal, oral, subcutaneous, intradermal, and intramuscular. The subcutaneous administration is particularly preferred. Subcutaneous administration may for example be by injection into the abdomen, lateral and anterior aspects of upper arm or thigh, scapular area of back, or upper ventrodorsal gluteal area. In some embodiments, the compositions of the disclosure are administered in one, or more doses, as well as, by other routes of administration. For example, such other routes include, intracutaneously, intravenously, intravascularly, intraarterially, intraperitnoeally, intrathecally, intratracheally, intracardially, intralobally, intramedullarly, intrapulmonarily, and intravaginally. Depending on the desired duration of the treatment, the compositions according to the disclosure may be administered once or several times, also intermittently, for instance on a monthly basis for several months or years and in different dosages. Solid dosage forms for oral administration include capsules, tablets, caplets, pills, powders, pellets, and granules. In such solid dosage forms, the active ingredient is ordinarily combined with one or more pharmaceutically acceptable excipients, examples of which are detailed above. Oral preparations may also be administered as aqueous suspensions, elixirs, or syrups. For these, the active ingredient may be combined with various sweetening or flavoring agents, coloring agents, and, if so desired, emulsifying and/or suspending agents, as well as diluents such as water, ethanol, glycerin, and combinations thereof.


In some embodiments, the compositions of the disclosure are administered, or the methods and uses for treatment according to the disclosure are performed, alone or in combination with other pharmacological compositions or treatments, for example other immunotherapy, vaccine or anti-viral. In some embodiments, the other therapeutic compositions or treatments are administered either simultaneously or sequentially with (before or after) the composition(s) or treatment of the disclosure.


The following descriptions and examples illustrate some exemplary embodiments of the disclosed invention in detail. Those of the skill in the art will recognize that there are numerous variations and modifications of this invention that are encompassed by its scope. Accordingly, the description of a certain exemplary embodiment should not be deemed to limit the scope of the present invention.


Example 1

The primary objective of designing a safer vaccine against SARS-CoV-2 using a rational approach is to minimize the possibility of inducing ADE upon vaccination, to reduce or eliminate off target penetration of the virus into immune cells and to prevent endothelial damage and thereby minimize the risk of vascular dysfunction in infected patients. A secondary objective is to create a stable formulation and affordable manufacture of the vaccine by using appropriate adjuvants and vaccine stabilization techniques.


The receptor binding domain (RBD) in the spike protein is the most variable part of the virus genome (Anderson et. al., 2020). Blue spheres in FIG. 2 depict the variability of the RBD as well as the N-terminal domain (NTD) of S protein monomer based on sequence analysis of approximately 60 clinical isolates (Wrapp et. al., 2020). This RBD variability is thought to allows SARS-CoV-2 gain antigen drift, thereby permitting immune escape, while still allowing high-affinity binding to ACE2 receptor (Yang et. al., 2005). Surprisingly, convalescent plasma of COVID-19 patients who recovered without hospitalization do not contain high levels of neutralizing antibodies against RBD (Robbiani et. al., 2020). As discussed above, should variability in regions of S protein produce non-neutralizing or sub-neutralizing antibodies, that might prove detrimental through induction of ADE.


In contrast, the C-terminal region of the S protein is relatively conserved, not only among distinct SARS-CoV-2 strains isolated till date, but also across other members of the sarbecovirus family members as shown below (FIG. 3, Wec et. al., 2020). The SARS-CoV-2 HR1 domain (aa 908-986) has 7 amino acids different with SARS-CoV-1 or bat coronavirus RaTG13, while HR2 (aa 1107-1175) is 100% identical at amino acid level between the three. The CH domain (aa 986-1035) is also 100% conserved whereas the CD domain (aa 1076-1141) has 9 amino acid changes against the SARS-CoV-1 or bat coronavirus RaTG1 sequences (Ceraolo et. al., 2020).


The S protein is activated by proteolytic cleavage at two sites, S1/S2 and S2′ (FIG. 1) by both TMPRSS2 as well as the proprotein convertase, furin (Bestle et. al., 2020; Shang et. al., 2020). The S1 subunit contains the RBD while the S2 subunit contains the hydrophobic heptad repeats HR1 and HR2 that are essential for membrane fusion. Once the RBD binds to the ACE2 receptor on target cells, the HR1 and HR2 domains interact with one another forming a six-helix bundle complex. The connector domain between HR1 and HR2 forms a loop inducing a conformational change that allows the fusion peptide on the virus insert into the target cell membrane causing virus-cell fusion.


Membrane protein (M) is a type III transmembrane glycoprotein and is the most abundant protein in SARS-CoV-2. The M protein has a short amino terminus domain located outside the virus, three transmembrane domains and a carboxy terminus domain located inside the viral envelope. On the viral surface, M protein is juxtaposed alongside the S protein and plays a role in the budding process (Alsaadi et. al., 2019). In alpha coronavirus, the M protein has been shown to participate in viral entry into host cells by facilitating membrane fusion (Naskalska et. al., 2019). While the M protein is highly conserved within SARS-CoV-2 family, there is sequence heterogeneity at the N-terminus of the protein where an insertion of a serine residue is unique to SARS-CoV-2 compared to bat or pangolin homologs (Bianchi et. al., 2020). Such mutations at the amino terminus region of the M protein could probably play a role in the host cell interactions.


One aim of the present disclosure is to prevent viral fusion with cell membrane by targeting a vaccine immune response against conserved C-terminal membrane-proximal fragments of SARS-CoV-2, specifically the HR1 and HR2 regions and the spacer in between.


Such a targeted approach has several advantages. First, by limiting the exposure of the host immune system, unwanted antibodies against undesirable regions of the S protein are eliminated. This in turn proves advantageous since virus bound, non-neutralizing antibodies cannot be taken up in immune effector cells such as macrophages, dendritic cells and T-cells, thereby diminishing the possibility of unwanted cytokine production and immunopathology. Second, by presenting short, rationally designed antigens to block viral fusion, the off-target entry of virus into immune cells or vascular endothelial cells can be prevented. Third, simple antigens lend themselves to simpler, cost-effective production with the possibility of easier scale up but without the need of BSL3 manufacturing and testing facilities, thereby lowering cost of goods. Fourth, antigen design will allow facile use of established adjuvants such as alum to skew host response away from a Th2 type which is linked to eosinophil-derived immunopathology (Chen et. al., 2020). The simpler, rationally designed antigens will be possessing a much better shelf life, enabling easy storage and distribution of vaccine with the need of complex logistics that are poorly available in low-income countries.


Human antibodies isolated from transgenic mice against the SARS-CoV-1 S2 domain have been shown to successfully neutralize pseudo-typed viruses expressing different S proteins of various clinical isolates on SARS-CoV-1 in an RBD-independent manner (Elshabrawy et. al., 2012). In addition, monoclonal antibodies raised against SARS-CoV-1 S protein were found to neutralize viral infection by inhibiting virus entry into Vero E6 cells, a mechanism distinct from virus-ACE2 receptor binding blockade, demonstrating the feasibility of preventing viral fusion as a viable vaccine strategy (Lai et. al., 2005, Lip et. al., 2006). Similarly, anti-M protein monoclonal antibodies have been identified from lymphocytes of convalescent patients infected with SARS-CoV-1 that potently neutralize viral entry into Vero-E6 cells (Liang et. al., 2005). In addition, sera from SARS-CoV-1 have been shown to bind both amino as well as carboxy terminus of M protein (Hu et. al., 2003; Vob et. al., 2009).


Example 2
Antigen Design

One of the important aspects of the antigen design is to represent the prefusion state for the S protein. This is because conversion of the S protein to post-fusion state involves a significant conformational change wherein the HR1 and HR2 domains pack against one another to form an anti-parallel six-helix bundle. This rearrangement triggers fusion of the viral membrane with the host cell. Thus, the rational design strategy is to block the virus in the prefusion state and prevent conversion into the post fusion state. In such a case, the viral entry and spread can be stopped irrespective of the cell the virus infects. Indeed, it has been shown that the SARS-CoV-1 HR2 domain forms a coiled coil structure in solution consisting of three helices folded as a parallel trimer, as in prefusion state (Hakkanson et. al., 2006). Thus, even targeting HR2 as a standalone antigen may be possible.


Seven distinct antigens were designed based on M protein and the heptad repeat structure within the prefusion SARS-CoV-2 S protein as follows:


SC2MN1: Amino acids 1-31 covering amino terminus of M protein


SC2MC1: Amino acids 130-160 from carboxy terminus of M protein


SC2SHR1: Amino acids 913-984 covering the first heptad repeat HR1 of S protein


SC2HSHRL: Amino acids 1147-1170 covering the linker region connecting HR1 and HR2


SC2HSHR2: Amino acids 1171-1212 covering the second heptad repeat HR2 of S protein


SC2HSHR2L: Amino acids 1147-1212 covering both the linker as well as HR2 regions


SC2RBD (control antigen): Amino acids 318-541 representing RBD of S protein


In reference to FIG. 2, the composition of the proposed antigens in those cases where high-resolution structures of S or M protein are available (6VXX.PDB, 6LXT.PDB, 6M3W.PDB). These antigens were created in the form of fusion protein expressed on virus like particles (VLP) using the Hepatitis B surface antigen S protein HBV-S. Presenting viral antigens on VLPs will result in enhanced quality of host immune response because VLPs can present antigen in draining lymph nodes and efficiently engage B cell receptors due to their repetitive structure resulting in a higher abundance of neutralizing antibodies (Coleman et. al., 2014). The HBV-S core protein is a very well-established system for VLP generation (Martini et. al., 2019).


A widely used approach for presenting antigens on VLPs is through the use of genetic fusion. However, while this technique can result in virus-like configuration of the antigen, longer peptides or those with charge or significant hydrophobicity can interfere with the actual assembly of the VLPs. To circumvent this problem, a mosaic approach was undertaken to construct VLPs (Ramasamy et. al., 2018). Specifically, the HBV S protein was coexpressed with proposed antigens in a fixed stoichiometry. In some embodiments, the ratio of HBV S protein to a SARS-CoV-2 polypeptide may be 6:1, preferably 5:1, more preferably 4:1. Microscopic as well as immunological assessment were carried out to ensure bona fide VLP formation. Because the HR1, linker and HR2 regions are conserved between SARS CoV-1 and SAR-CoV-2, monoclonal antibodies previously made using SARS-CoV-1 were used as analytical and immunological tools to ensure proper VLP assembly. Each of the antigens will be produced and tested, in vitro and in vivo.


Example 3
Production of Immunogens

VLPs contained within formulations of approved vaccines such as those against human papillomavirus (HPV) or hepatitis B virus (HBV) are produced in yeast cells. These vaccines have proven to be safe and effective, and their manufacturing processes are very well established. Separately, VLPs expressed in recombinant baculovirus systems covering multi-component antigens such as HA and matrix 1 for influenza vaccine have also been established. The antigens proposed in our strategy are glycosylated at multiple sites and it becomes important to ensure glycosylation in antigen preparation as close to the human system as possible (Wu et. al., 2010). Therefore, immunogen production in Vero cells is proposed (Ammerman et. al., 2008).


Vero cells are derived from African green monkeys and are a close approximation to human cells. Vero cell lines have been approved in the US for production of licensed viral vaccines such as those against rotavirus, smallpox and inactivated poliovirus. Worldwide, Vero cells have also been used for the production of vaccines against Rabies virus, Reovirus and Japanese encephalitis virus. Commercially, Vero cells have been scaled up to 660 m2 with cell density approaching 2.3×105 cells/cm3. As such Vero cell technology, is cheap, scalable and well-established vaccine production technology worldwide.


Example 4
Epitope Prediction for SARS-CoV-2 M and S Antigens

1: M protein (UniProt: P59596)


The results presented below for T cell epitopes (Table 1) are derived from experimental data deposited at the IEDB ImmunoBrowser (Vita et al., 2014). Due to the unavailability of a structure for the M protein, a sequence-based algorithm for B cell epitope prediction was used (BepiPred 2.0, Table 2; Jespersen et al., 2017).









TABLE 1







T cell epitope prediction for the N (top) and C terminus (bottom)


of SARS-CoV-2 M protein based on experimental data available


at the IEDB. Epitopes tested immediately downstream or


upstream of these sequences did not give positive responses.














SEQ

Mapped
Mapped



















ID

Start
End
Subjects
Subjects
Assay
Assay
Response


NO
Sequence
Position
Position
Tested
Responded
Positive
Negative
Freq.


















1
MADNGTIT
1
15
4
1
1
1
0.25



VEELKQL












2
MADNGTIT
1
16
40
10
1
0
0.25



VEELKQLL












3
MADNGTIT
1
18
1
1
1
0
1



VEELKQL










LEQ












4
MADNGTIT
1
20
32
1
1
1
0.03



VEELKQLL










EQWN












5
MADNGTI
1
31
40
40
1
0
1



TVEELK










QLLEQW










NLVIGF










LFLAWI












6
ITVEEL
7
14
4
3
1
0
0.75



KQ












7
ITVEELKQ
7
23
40
28
1
0
0.7



LLEQWNLV










I












8
KLNTDHA
204
221
1
1
1
0
1



GSNDNIAL










LVQ












9
TDHAGSND
207
221
48
27
1
1
0.56



NIALLVQ










Sequence-based B cell epitope prediction using the BepiPred 2.0 server









TABLE 2







B cell epitope prediction for the N (left) and C terminus (center, right)


of SARS-CoV-2M protein based on the BepiPred algorithm, a server for


sequence-based epitope prediction (cbs.dtu.dk/services/BepiPred/).


Residues immediately downstream or upstream of these were not identified


as predicted epitopes (default threshold of 0.5).










Position
AminoAcid
Exposed/Buried
EpitopeProbability













5
G
E
0.501


6
T
E
0.522


7
I
B
0.535


8
T
E
0.524


9
V
B
0.531


10
E
E
0.539


11
E
E
0.538


12
L
B
0.518


13
K
B
0.519


14
Q
E
0.541


15
L
B
0.541


16
L
B
0.530


17
E
E
0.540


18
Q
E
0.521


180
L
B
0.519


181
G
E
0.530


182
A
E
0.549


183
S
E
0.572


184
Q
E
0.587


185
R
E
0.595


186
V
B
0.600


187
G
E
0.598


188
T
E
0.576


189
D
E
0.555


190
S
B
0.540


191
G
B
0.522


192
F
B
0.501


195
Y
B
0.505


196
N
B
0.509


197
R
B
0.531


198
Y
E
0.549


199
R
E
0.560


200
I
B
0.571


201
G
E
0.567


202
N
E
0.564


203
Y
B
0.563


204
K
E
0.563


205
L
B
0.580


206
N
E
0.594


207
T
B
0.606


208
D
E
0.613


209
H
E
0.621


210
A
E
0.630


211
G
E
0.636


212
S
E
0.626


213
N
E
0.617


214
D
E
0.594


215
N
E
0.568


216
I
B
0.538


217
A
E
0.506









2: S Protein (UniProt: P59594)

The results presented below for T cell epitopes (Tables 3 and 5) are derived from experimental data available at the IEDB ImmunoBrowser (Vita et al., 2014). Structure-based B cell epitope predictions (Tables 4 and 6) were generated with SEPPA 3.0 (Zhou et al., 2019). Table 7 shows a sequence-based epitope prediction using BepiPred 2.0 (Jespersen et al., 2017) for amino acids 1104-1184 as the structures available did not span that region.









TABLE 3







T cell epitope prediction for the receptor binding domain of SARS-CoV-1 S protein


(residues 306-527) with peptides spanning the receptor-binding motif (424-494) highlighted in


blue. Response frequency is based on experimental data available at the IEDB. Tested epitopes


that did not give positive responses are absent.















SEQ

Mapped
Mapped







ID

Start
End
Subjects
Subjects
Assay
Assay
Response


NO
Sequence
Position
Position
Tested
Responded
Positive
Negative
Freq.


















10
EIDKGIYQTS
294
310
42
21
1
0
0.5



NFRVVPS












11
KGIYQTSNFR
297
316
1
1
1
0
1



VVPSGDVVR










F












12
KGIYQTSN
297
304
4
2
1
0
0.5





13
IYQTSNFRVV
299
316
3
1
1
2
0.33



PSGDVVRF












14
TSNFRVVPS
302
318
42
20
1
0
0.48



GDVVRFPN












15
SGDVVRFPNI
310
326
42
13






TNLCPFG




1
0
0.31





16
VRFPNITNLC
314
330
42
10
1
0
0.24



PFGEVFN












17
LCPFGEVFN
322
338
42
11
1
0
0.26



ATKFPSVY












18
FGEVFNAT
325
332
4
2
1
0
0.5





19
FNATKFPSV
329
345
3
2






YAWERKKI




1
0
0.67



N330, A331,
330
485
1
1
1
0
1



T332, K333,










F360, W423,










N424, N427,










I428, A430,










T431, S432,










N435, N437,










T485












20
TKFPSVYAW
332
351
1
1
1
0
1



ERKKISNCV










AD












21
PSVYAWERK
335
351
42
12
1
0
0.29



KISNCVAD












22
SVYAWERK
336
352
3
1






KISNCVADY




1
2
0.33





23
VYAWERKKI










SNCVADYSV
337
360
1
1
1
0
1



LYNSTF










K343, K344,
343
428
1
1
1
0
1



1345, S346,










N347, C348,










V349, A350,










D351, Y352,










S353, V354,










L355, Y356,










N357, S358,










T359, F360,










F361, S362,










T363, F364,










K365, C366,










Y367, K373,










L374, N375,










D376, L377,










C378, F379,










S380, N381,










V382, Y383,










A384, D385,










S386, F387,










V388, V389,










K390, K411,










L412, P413,










D414, D415,










F416, M417,










G418, C419,










V420, L421,










A422, W423,










N424, T425,










R426, N427,










I428












24
KKISNCVAD
343
360
5
1
1
2
0.2



YSVLYNSTF












25
KKISNCVAD
343
359
42
5
1
0
0.12



YSVLYNST










K344, F360,
344
487
1
1
1
0
1



Y442, L472,










D480, T487












26
CVADYSVLY
348
356
11
3
2
0
0.27





27
YSVLYNSTFF
352
368
42
12
1
0
0.29



STFKCYG










Y356, N357,
356
494
1
1
1
0
1



S358, T359,










F361, S362,










T363, F364,










K365, C366,










A371, T372,










G391, R395,










N424, 1489,










Y494










Y356, N357,
356
417
1
1
1
0
1



S362, T363,










F364, K365,










C366, Y367,










G368, V369,










S370, A371,










T372, K373,










D376, L377,










R395, D415,










M417












28
STFFSTFKCY
358
374
4
2
2
2
0.5



GVSATKL










T359, S362,
359
494
1
1
1
0
1



G391, D392,










N424, R426,










N427, T486,










T487, G488,










1489, G490,










Y491, Q492,










Y494










T359, T363,
359
494
2
1
1
1
0.5



K365, K390,










G391, D392,










R395, R426,










Y436, G482,










Y484, T485,










T486, T487,










G488, 1489,










G490, Y491,










Q492, Y494












29
FFSTFKCYG
360
376
42
5
1
0
0.12



VSATKLND












30
KCYGVSATK
365
374
4
3
3
1
0.75



L












31
CYGVSATKL
366
374
3
3
3
0
1





32
CYGVSATKL
366
382
42
13
1
0
0.31



NDLCFSNV












33
YGVSATKL
367
374
1
1
1
0
1





34
KLNDLCFSN
373
389
42
8
1
0
0.19



VYADSFVV












35
SNVYADSFV
380
399
1
1
1
0
1



VKGDDVRQI










AP












36
NVYADSFVV
381
397
43
13






KGDDVRQI




1
1
0.3



D392, V394
392
394
1
1
1
0
1



D392, V394,
392
454
1
1
1
0
1



D414, F416,










R441, D454












37
IAPGQTGVIA
397
413
42
10






DYNYKLP




1
0
0.24





38
IADYNYKLP
405
421
42
5






DDFMGCVL




1
0
0.12





39
KLPDDFMGC
411
427
42
9






VLAWNTRN




1
0
0.21





40
KLPDDFMGC
411
420
20
4






V




1
1
0.2





41
CVLAWNTR
419
435
42
8
1
0
0.19



NIDATSTGN












42
NTRNIDATST
424
435
8
8
1
0
1



GN










R426, S432,
426
492
1
1
1
0
1



T433, Y436,










N437, K439,










Y440, Y442,










P469, P470,










A471, L472,










N473, C474,










Y475, W476,










L478, N479,










D480, Y481,










G482, Y484,










T485, T486,










T487, G488,










1489, Y491,










Q492












43
NIDATSTGN
427
444
3
1
1
2
0.33



YNYKYRYLR












44
NIDATSTGN
427
443
42
11
1
0
0.26



YNYKYRYL










I428, A430,
428
439
1
1






K439




1
0
1



D429, R441,
429
454
1
1






D454




1
0
1



D429, R441,
429
454
1
1






E452, D454




1
0
1



D429, R441,
429
454
1
1






D454




1
0
1



D429, R441,
429
463
1
1






D454, D463




1
0
1



D429, R441,
429
454
1







E452, D454



1
1
0
1



T431, S432,
431
446
1
1






K439, G446




1
0
1





45
TSTGNYNYK
431
445
4
1
1
1
0.25



YRYLRH












46
GNYNYKYR
434
448
1
1
1
0
1



YLRHGKL












47
GNYNYKYR
434
453
1
1
1
0
1



YLRHGKLRP










FER












48
NYNYKYRYL
435
444
3
2
2
1
0.67



R












49
NYNYKYRYL
435
451
46
12
3
2
0.26



RHGKLRPF












50
YNYKYRYL
436
443
2
2
2
0
1





51
YNYKYRYLR
436
455
1
1
1
0
1



HGKLRPFER










DI












52
HNYKYRYL
436
443
18
12
3
0
0.67





53
YNYKYRYLR
436
450
4
1






HGKLRP




1
1
0.25





54
NYKYRYLRH
437
459
1
1






GKLRPFERDI










SNVP




1
0
1





55
KYRYLRHGK
439
454
1
1






LRPFERD




1
0
1



K439, G446,
439
463
1
1






S461, D463




1
0
1



R441
441
441
2
2
2
0
1



Y442
442
442
1
1
1
0
1



Y442, A834
442
834
1
1
1
0
1



Y442, D757
442
757
1
1
1
0
1





56
YLRHGKLRP
442
458
4
2
2
2
0.5



FERDISNV












57
YLRHGKLRP
442
459
4
2
1
0
0.5



FERDISNVP












58
LRHGKLRPF
443
459
3
2
1
0
0.67



ERDISNVP










G446, P462,
446
475
1
1
1
0
1



D463, Y475












59
KLRPFERDI
447
455
1
1
1
0
1





60
KLRPFERDIS
447
458
1
1
1
0
1



NV












61
RPFERDISNV
449
458
2
2
2
0
1





62
RPFERDISNV
449
461
4
2
1
0
0.5



PFS












63
RPFERDISNV
449
465
44
11
1
2
0.25



PFSPDGK










D454
454
454
1
1
1
0
1





64
SNVPFSPDG
456
472
44
11
1
2
0.25



KPCTPPAL












65
PFSPDGKPCT
459
470
8
8
1
0
1



PP












66
FSPDGKPCTP
460
476
1
1
1
0
1



PALNCYW












67
SPDGKPCTPP
461
477
42
5
1
0
0.12



ALNCYWP










P462
462
462
1
1
1
0
1





68
CTPPALNCY
467
483
42
8
1
0
0.19



WPLNDYGF












69
ALNCYWPLN
471
503
1
1
1
0
1



DYGFYTTTGI










GYQPYRVVV










LSFEL












70
ALNCYWPLN
471
503
42
30
1
0
0.71



DYGFYTTTGI










GYQPYRVVV










LSFEL












71
ALNCYW
471
476
4
1
1
0
0.25





72
CYWPLNDY
474
490
42
7
1
0
0.17



GFYTTTGIG










N479
479
479
1
1
1
0
1



D480, Y484
480
484
1
1
1
0
1





73
GFYTTTGIGY
482
498
42
8
1
0
0.19



QPYRVVV










Y484, T487
484
487
1
1
1
0
1





74
TTGIGYQ
486
492
1
1
1
0
1





75
GYQPYRVVV
490
510
2
2
2
0
1



LSFELLNAPA










TV












76
GYQPYRVVV
490
506
44
9
2
1
0.2



LSFELLNA












77
QPYRVVVLS
492
501
4
1
1
0
0.25



F












78
VVLSFELLN
497
514
2
1
1
1
0.5



APATVCGPK












79
VLSFELLNAP
498
514
42
8
1
0
0.19



ATVCGPK












80
NAPATVCGP
505
521
44
7
1
2
0.16



KLSTDLIK












81
CGPKLSTDLI
511
527
42
7
1
0
0.17



KNQCVNF










Sequence-based B cell epitope prediction using the SEPPA 3.0 server and the atomic coordinates deposited as 5X58.pdb.









TABLE 4







B cell epitope prediction for the receptor binding domain of SARS-CoV-2


S protein (residues 306-527) with peptides spanning the receptor-binding


motif (424-494) highlighted in blue. The server SEPPA 3.0 was used with


the default threshold of 0.064. Absent residues did not rank as


epitope residues.










resS eq
resName
score
location





314
VAL
0.100
surface


315
ARG
0.081
surface


316
PHE
0.152
surface


317
PRO
0.101
surface


318
ASN
0.155
surface


331
ALA
0.115
surface


332
THR
0.158
surface


334
PHE
0.097
surface


335
PRO
0.110
surface


336
SER
0.083
surface


337
VAL
0.073
surface


338
TYR
0.077
surface


347
ASN
0.127
surface


378
CYS
0.179
surface


379
PHE
0.104
surface


380
SER
0.258
surface


381
ASN
0.072
surface


382
VAL
0.070
surface


391
GLY
0.073
surface


400
GLY
0.071
surface


401
GLN
0.068
surface


403
GLY
0.071
surface


405
ILE
0.085
surface


406
ALA
0.073
surface


407
ASP
0.079
surface


408
TYR
0.097
surface


409
ASN
0.080
surface


411
LYS
0.083
surface


424
ASN
0.066
surface


425
THR
0.072
surface


426
ARG
0.162
surface


427
ASN
0.078
surface


428
ILE
0.084
surface


429
ASP
0.083
surface


430
ALA
0.122
surface


431
THR
0.174
surface


432
SER
0.222
surface


433
THR
0.145
surface


434
GLY
0.202
surface


435
ASN
0.166
surface


436
TYR
0.220
surface


437
ASN
0.096
surface


438
TYR
0.101
surface


439
LYS
0.181
surface


440
TYR
0.081
surface


441
ARG
0.071
surface


442
TYR
0.093
surface


443
LEU
0.105
surface


444
ARG
0.087
surface


445
HIS
0.118
surface


446
GLY
0.073
surface


447
LYS
0.075
surface


448
LEU
0.066
surface


451
PHE
0.067
surface


454
ASP
0.083
surface


456
SER
0.114
surface


457
ASN
0.117
surface


458
VAL
0.116
surface


459
PRO
0.110
surface


460
PHE
0.130
surface


461
SER
0.171
surface


462
PRO
0.165
surface


463
ASP
0.173
surface


464
GLY
0.185
surface


465
LYS
0.174
surface


466
PRO
0.178
surface


467
CYS
0.161
surface


468
THR
0.195
surface


469
PRO
0.189
surface


470
PRO
0.225
surface


471
ALA
0.233
surface


472
LEU
0.242
surface


473
ASN
0.186
surface


474
CYS
0.193
surface


475
TYR
0.116
surface


476
TRP
0.106
surface


478
LEU
0.179
surface


479
ASN
0.172
surface


480
ASP
0.100
surface


481
TYR
0.144
surface


482
GLY
0.255
surface


483
PHE
0.138
surface


484
TYR
0.236
surface


485
THR
0.206
surface


486
THR
0.203
surface


487
THR
0.238
surface


488
GLY
0.210
surface


489
ILE
0.115
surface


490
GLY
0.105
surface


491
TYR
0.117
surface


492
GLN
0.096
surface


494
TYR
0.070
surface


501
PHE
0.072
surface


502
GLU
0.254
surface


503
LEU
0.261
surface


504
LEU
0.434
surface


505
ASN
0.324
surface


506
ALA
0.362
surface


507
PRO
0.441
surface


508
ALA
0.259
surface


509
THR
0.147
surface










Heptad repeat 1 (902-952), Heptad repeat 2 (1145-1184) and a linker T cell epitopes based on data available at the IEDB immunoBrowser.









TABLE 5







T cell epitope prediction for heptad repeat 1, linker and heptad repeat 2 of SARS-


CoV-2 S protein (residues 902-1184). Response frequency is based on experimental data


available at the IEDB. Tested epitopes that did not give positive responses are absent.















SEQ

Mapped
Mapped







ID

Start
End
Subjects
Subjects
Assay
Assay
Response


NO
Sequence
Position
Position
Tested
Responded
Positive
Negative
Freq.


















82
VLYENQK
897
913
43
20
1
1
0.47



QIANQFNK










AI












83
QIANQFNK
904
920
43
5
1
1
0.12



AISQIQESL












84
KAISQIQES
911
927
43
6
1
1
0.14



LTTTSTAL










Q917, E918,
917
940
1
1
1
0
1



S919, T921,










T922, S924,










T925, A926,










G928, K929,










Q931, D932,










V933, N935,










Q936, A938,










Q939, A940












85
ESLTTTSTA
918
934
45
13
2
1
0.29



LGKLQDV










V










T921, T922,
921
944
1
1
1
0
1



T923, T925,










A926, G928,










K929, L930,










D932, V933,










N935, Q936,










N937, Q939,










A940, N942,










T943, L944












86
TALGKLQD
925
941
43
8
1
1
0.19



VVNQNAQ










AL












87
DVVNQNA
932
948
43
9
1
1
0.21



QALNTLVK










QL












88
QALNTLVK
939
955
46
17
2
2
0.37



QLSSNFGAI












89
ALNTLVKQ
940
948
52
3
3
4
0.06



L












90
KQLSSNFG
946
962
3
1
1
0
0.33



AISSVLNDI












91
AISSVLNDI
954
971
1
1
1
0
1



LSRLDKVE










A












92
AISSVLNDI
954
970
42
11
1
0
0.26



LSRLDKVE












93
SVLNDILSR
957
965
13
1
1
1
0.08





94
VLNDILSR
958
966
166
12
6
3
0.07



L












95
ILSRLDKV
962
978
3
1
1
0
0.33



EAEVQIDR










L












96
RLDKVEAE
965
973
41
5
2
2
0.12



V












97
EAEVQIDR
970
986
43
6
1
1
0.14



LITGRLQSL












98
AEVQIDRLI
971
979
5
1
1
0
0.2



RLITGRLQS












99
LQTYVTQQ
977
993
42
19
1
0
0.45





100
LITGRLAA
978
986
2
1
1
1
0.5



L












101
LITGRLQSL
978
986
67
5
5
5
0.07





102
RLQSLQTY
982
990
63
18
4
3
0.29



V












103
SLQTYVTQ
985
1001
42
16
1
0
0.38



QLIRAAEIR












104
QLIRAAEIR
993
1010
4
3
3
0
0.75



ASANLAAT










K












105
QLIRAAEIR
993
1009
42
8
1
0
0.19



ASANLAAT












106
RASANLAA
1001
1017
42
7
1
0
0.17



TKMSECVL










G












107
AATKMSEC
1007
1023
42
13
1
0
0.31



VLGQSKRV










D












108
VLGQSKRV
1015
1031
3
1
1
0
0.33



DFCGKGY










HL












109
RVDFCGKG
1021
1029
36
2
1
0
0.06



Y












110
DFCGKGY
1023
1039
42
11
1
0
0.26



HLMSFPQA










AP












111
LMSFPQAA
1031
1047
44
14
2
1
0.32



PHGVVFLH










V












112
APHGVVFL
1038
1047
13
3
1
0
0.23



HV












113
PHGVVFLH
1039
1055
42
11
1
0
0.26



VTYVPSQE










R












114
GVVFLHVT
1041
1049
2
1
1
0
0.5



Y












115
VVFLHVTY
1042
1050
37
4
4
2
0.11



V












116
VTYVPSQE
1047
1063
42
10
1
0
0.24



RNFTTAPAI












117
ERNFTTAP
1054
1071
2
1
1
1
0.5



AICHEGKA










YF












118
RNFTTAPAI
1055
1071
42
7
1
0
0.17



CHEGKAYF












119
PAICHEGK
1061
1093
15
12
1
0
0.8



AYFPREGV










FVFNGTSW










FITQRNFFS












120
HEGKAYFP
1065
1076
8
8
1
0
1



REGV












121
AYFPREGV
1069
1085
42
2
1
0
0.05



FVFNGTSW










F












122
FVFNGTSW
1077
1093
42
2
1
0
0.05



FITQRNFFS












123
GTSWFITQ
1081
1095
3
1
1
0
0.33



RNFFSPQ












124
SWFITQRN
1083
1097
5
3
2
1
0.6



FFSPQII












125
NFFSPQIIT
1090
1106
3
1
1
0
0.33



TDNTFVSG












126
FFSPQIITT
1091
1130
1
1
1
0
1



DNTFVSGN












127
CDVVIGIIN
1096
1104
18
1
1
0
0.06



NTVYDPLQ










PELDSF










IITTDNTFV












128
ITTDNTFVS
1097
1113
3
1
1
0
0.33



GNCDVVIG












129
FVSGNCDV
1103
1120
2
1
1
1
0.5



VIGIINNTV










Y












130
SGNCDVVI
1105
1121
42
6
1
0
0.14



GIINNTVY










D












131
VIGIINNTV
1111
1130
2
2
2
0
1



YDPLQPEL










DSF












132
VIGIINNTV
1111
1127
44
9
2
1
0.2



YDPLQPEL












133
TVYDPLQP
1118
1134
43
2
1
1
0.05



ELDSFKEE










L












134
PELDSFKE
1125
1141
4
1
1
1
0.25



ELDKYFKN










H












135
DSFKEELD
1128
1159
2
1
1
1
0.5



KYFKNHTS










PDVDLGDI










SGINASVV












136
DSFKEELD
1128
1137
36
1
1
0
0.03



KY












137
EELDKYFK
1132
1148
4
1
1
1
0.25



NHTSPDVD










L












138
DKYFKNHT
1135
1150
1
1
1
0
1



SPDVDLGD












139
KNHTSPDV
1139
1155
42
4
1
0
0.1



DLGDISGIN












140
SPDVDLGD
1143
1157
65
2
2
2
0.03



ISGINAS












141
VDLGDISGI
1146
1154
1
1
1
0
1





142
DLGDISGIN
1147
1163
43
6
1
1
0.14



ASVVNIQK












143
LGDISGINA
1148
1162
17
1
1
0
0.06



SVVNIQ












144
ISGINASVV
1151
1170
1
1
1
0
1



NIQKEIDRL










NE












145
ISGINASVV
1151
1192
1
1
1
0
1



NIQKEIDRL










NEVAKNL










NESLIDLQE










LGKYEQYI










N1155,
1155
1178
1
1
1
0
1



A1156,










S1157,










V1159,










N1160,










Q1162,










K1163,










E1164,










D1166,










R1167,










N1169,










E1170,










V1171,










K1173,










N1174,










N1176,










E1177,










S1178










Q1162,
1162
1185
1
1
1
0
1



K1163,










E1164,










D1166,










R1167,










N1169,










E1170,










V1171,










K1173,










N1174,










N1176,










E1177,










S1178,










11180,










D1181,










Q1183,










E1184,










L1185












146
KEIDRLNE
1163
1179
3
1
1
0
0.33



VAKNLNES










L












147
EIDRLNEV
1164
1191
1
1
1
0
1



AKNLNESL










IDLQELGK










YEQY












148
EIDRLNEV
1164
1191
42
28
1
0
0.67



AKNLNESL










IDLQELGK










YEQY












149
RLNEVAKN
1167
1175
246
17
7
6
0.07



L












150
EVAKNLNE
1170
1186
42
6
1
0
0.14



SLIDLQELG










Structure-based B cell epitope prediction using the SEPPA 3.0 server and the atomic coordinates deposited as 5X58.pdb: For amino acids downstream of 1104, a sequence-based method (BepiPred 2.0) was used due to the lack of a solved structure encompassing that region.









TABLE 6







B cell epitope prediction for heptad repeat 1 (residues 902-952) and linker


up to residue 1104 of SARS-CoV-2 S protein (remaining linker region and


heptad repeat 2 are absent from the atomic coordinates of 5X58.pdb). The


server SEPPA 3.0 was used with the default threshold of 0.064.


Absent residues did not rank as epitope residues.










resSeq
resName
score
location













902
GLN
0.702
surface


903
LYS
0.515
surface


904
GLN
0.486
surface


906
ALA
0.430
surface


907
ASN
0.313
surface


908
GLN
0.302
surface


909
PHE
0.146
surface


910
ASN
0.136
surface


912
ALA
0.089
surface


913
ILE
0.122
surface


914
SER
0.104
surface


915
GLN
0.098
surface


916
ILE
0.068
surface


917
GLN
0.073
surface


918
GLU
0.099
surface


919
SER
0.109
surface


920
LEU
0.084
surface


921
THR
0.104
surface


941
LEU
0.097
surface


942
ASN
0.134
surface


943
THR
0.134
surface


944
LEU
0.107
surface


945
VAL
0.113
surface


947
GLN
0.174
surface


948
LEU
0.222
surface


949
SER
0.151
surface


950
SER
0.157
surface


952
PHE
0.236
surface


953
GLY
0.183
surface


954
ALA
0.172
surface


955
ILE
0.097
surface


956
SER
0.080
surface


957
SER
0.117
surface


958
VAL
0.089
surface


959
LEU
0.258
surface


960
ASN
0.115
surface


961
ASP
0.111
surface


962
ILE
0.115
surface


963
LEU
0.090
surface


964
SER
0.147
surface


965
ARG
0.144
surface


966
LEU
0.133
surface


967
ASP
0.135
surface


968
LYS
0.208
surface


969
VAL
0.177
surface


970
GLU
0.194
surface


972
GLU
0.236
surface


973
VAL
0.366
surface


974
GLN
0.237
surface


975
ILE
0.395
surface


976
ASP
0.401
surface


977
ARG
0.283
surface


978
LEU
0.270
surface


979
ILE
0.413
surface


980
THR
0.432
surface


981
GLY
0.360
surface


982
ARG
0.240
surface


983
LEU
0.288
surface


984
GLN
0.344
surface


985
SER
0.176
surface


987
GLN
0.098
surface


988
THR
0.070
surface


1028
GLY
0.149
surface


1029
TYR
0.255
surface


1050
VAL
0.513
surface


1051
PRO
0.432
surface


1052
SER
0.479
surface


1053
GLN
0.427
surface


1054
GLU
0.159
surface


1058
THR
0.073
surface


1059
THR
0.107
surface


1060
ALA
0.135
surface


1061
PRO
0.131
surface


1062
ALA
0.261
surface


1063
ILE
0.245
surface


1064
CYS
0.146
surface


1065
HIS
0.230
surface


1066
GLU
0.249
surface


1067
GLY
0.237
surface


1068
LYS
0.211
surface


1069
ALA
0.268
surface


1070
TYR
0.218
surface


1071
PHE
0.274
surface


1072
PRO
0.297
surface


1073
ARG
0.269
surface


1074
GLU
0.214
surface


1075
GLY
0.201
surface


1076
VAL
0.085
surface


1079
PHE
0.091
surface


1080
ASN
0.093
surface


1082
THR
0.090
surface


1083
SER
0.103
surface


1084
TRP
0.076
surface


1085
PHE
0.080
surface


1086
ILE
0.137
surface


1087
THR
0.170
surface


1088
GLN
0.147
surface


1089
ARG
0.168
surface


1090
ASN
0.253
surface


1091
PHE
0.398
surface


1092
PHE
0.136
surface


1093
SER
0.169
surface


1094
PRO
0.105
surface


1095
GLN
0.130
surface


1096
ILE
0.146
surface


1097
ILE
0.210
surface


1098
THR
0.233
surface


1099
THR
0.328
surface


1100
ASP
0.376
surface


1101
ASN
0.273
surface


1102
THR
0.321
surface


1103
PHE
0.322
surface


1104
VAL
0.320
surface
















TABLE 7







B cell epitope prediction for amino acids 1105-1184 of SARS-CoV-2 S


comprising the heptad repeat 2 and part of the upstream linker protein


(based on the BepiPred algorithm, a server for sequence-based analysis


(cbs.dtu.dk/services/BepiPred/). Residues immediately downstream or


upstream of these were not identified as predicted epitopes (default


threshold of 0.5).










Position
AminoAcid
Exposed/Buried
EpitopeProbability





1115
I
B
0.515


1116
N
E
0.522


1117
N
E
0.548


1118
T
E
0.567


1119
V
E
0.575


1120
Y
B
0.586


1121
D
E
0.589


1122
P
E
0.582


1123
L
B
0.569


1124
Q
B
0.557


1125
P
E
0.559


1126
E
E
0.541


1127
L
E
0.529


1128
D
B
0.521


1129
S
E
0.531


1130
F
B
0.515


1131
K
E
0.508


1132
E
E
0.517


1133
E
E
0.527


1134
L
B
0.519


1135
D
E
0.544


1136
K
E
0.555


1137
Y
B
0.569


1138
F
B
0.576


1139
K
E
0.587


1140
N
E
0.595


1141
H
B
0.601


1142
T
E
0.604


1143
S
E
0.617


1144
P
E
0.601


1145
D
E
0.606


1146
V
B
0.607


1147
D
E
0.590


1148
L
B
0.600


1149
G
E
0.593


1150
D
E
0.572


1151
I
B
0.574


1152
S
E
0.564


1153
G
E
0.560


1154
I
B
0.535


1155
N
E
0.516


1156
A
B
0.517


1177
E
E
0.501


1180
I
B
0.508


1181
D
B
0.510


1182
L
B
0.511


1183
Q
E
0.517


1184
E
E
0.509









Example 5
Biochemical and Cell Based Assays

Immune sera from vaccinated animals will be tested using ELISA. Here, the binding of polyclonal sera derived from animals is tested against VLP antigen immobilized on wells using serial dilutions. Binding of ELISA positive sera will be tested against full length SARS-COV-2 S protein. This will be done in two ways. Using ELISA, binding of anti-sera will be verified against the ectodomain of S protein recombinantly-expressed in 293HEK cells and purified to homogeneity using affinity chromatography. Further, binding will be characterized against full length S protein transiently-expressed in CHO or COS-7 cells using flow cytometry. These assays address whether anti-sera from immunized animals bind prefusion, glycosylated and intact S protein. Finally, neutralizing activity of antisera will be verified using live virus neutralization assays wherein SARS-CoV-2 virus together with antisera are used to infect Vero cells followed by detection of internalized virus using commercially available, anti-S protein antibody such as CR3022 (Pinto et. al., 2020).


Example 6
Animal Studies

Mouse studies will be conducted to screen the antigen panel and identify a lead and backup vaccine candidate. Up to 3 dose strengths will be tested for each vaccine construct. All vaccine formulations will be alum adjuvanted. Groups of mice (n=10, 6-8 week old female Balb/c) will be vaccinated with VLP vaccine constructs intramuscularly followed by a booster at 14 day intervals. Sera will be collected prior to immunization as well after 7 days after each immunization cycle. 10 days after the booster, animals will be challenged intranasal with SARS-CoV-2 virus. Animals will be monitored for signs of disease. Animals will be euthanized at the end of the study and lung tissue are harvested for pathology and immunohistochemical staining.


Non-Human Primate (NHP) Studies:

NHP studies will be carried out on the lead and backup vaccine candidates. The lead and/or the backup mixture of more than one of the vaccine constructs, including the RBD construct, will be also considered for the study. Up to 2 doses (high dose and low dose) for each vaccine construct will be tested. All vaccine formulations will be alum adjuvated. Adult rhesus macaques (n=4 for each group, M/F) will be vaccinated be intramuscularly followed by a booster 14 days later. Blood samples will be collected before and after vaccination. Seven days following the booster, animals will be challenged with live SARS-CoV-2 virus intratracheally, intranasally, orally and ocularly. Clinical exams will be performed at regular intervals and BAL fluid are collected on periodic days. At the end of the study, animals will be euthanized and necropsy are performed.


Vaccine Safety Evaluation:

Safety evaluation of the lead vaccine candidate will be conducted in macaques. Groups of animals (n=5 M/F) will be immunized with high dose of the lead vaccine. A total of 3 intramuscular injections will be made (immunization plus two boosters). Safety evaluation include clinical observations as well as gross pathology on lung, heart, spleen, liver, kidney and brain.


Example 7
Process Development

Upstream and downstream processes for vaccine manufacture will be established according to standard industry norms in a cGMP grade BSL2 facility (Alvim et. al., 2019). Vero cell lines initially grown in tissue culture flasks at the research scale will be scaled-up in 5 L benchtop, stirred-tank bioreactors (Knowles et. al., 2013). Microcarriers will be used to enhance attachment of the adherent Vero cells. The culture will be maintained in a fed-batch mode with medium replenished on days 3 and 5 with fresh media to boost cellular productivity. VLPs secreted during the cell culture fermentation will be separated from cell mass by tangential-flow filtration (TFF) and downstream purification are undertaken. Concentration and buffer exchange will be conducted using additional TFF steps. This is followed by hydrophobic interaction chromatography and an ion-exchange step to purify the VLP fraction. Purified VLPs will be formulated and sterile filtered as described below (Aravelo et. al., 2016).


Process robustness and reproducibility will be established by running at least 3 lots of VLP production for both USP and DSP steps. Analytical method development includes tests for purity and potency in addition to validation of physical characteristics (appearance, particle size, pH) and identity (ELISA or Western Blot). Purity assessment will be done by SDS-PAGE densitometry as well as total protein content measured by BCA assay. Potency assessment will be conducted by use of validated ELISA methods using antibodies specific to VLP antigens. Impurity testing includes host protein and DNA quantitation.


Vaccines will be tested using a panel of buffers and excipients to identify a stable formulation. Histidine Tris and PBS buffers will be mixed with excipients such as Polysorbate 80, amino acids, sucrose and trehalose. In addition, vaccines will be adsorbed on alum adjuvant for increased stability (as well as enhanced immunogenicity). Formulated vaccine will be sterile filtered and filled in 2 mL single dose glass vials. Multi dose vials will be tested once vaccine PoC was established. An initial goal is to establish a formulation compatible with a vaccine vial monitor (VMM) 14 label. To do so, formal stability studies will be conducted as described below.


Accelerated stability studies will be conducted at 4° C., 25° C., 37° C. and 40° C. followed by potency assessment allowed selection of the lead formulation from the panel above. For formal stability studies, a minimum of three lots of lead vaccine formulation will be placed on stability study for 3 to 6 months. Stability-indicating assays including pH, physical appearance, potency and sterility will be conducted.


It is to be understood that this invention is not limited to any particular embodiment described herein and may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.


Where a range of values is provided, it is understood that each intervening value between the upper and lower limit of that range (to a tenth of the unit of the lower limit) is included in the range and encompassed within the invention, unless the context or description clearly dictates otherwise. In addition, smaller ranges between any two values in the range are encompassed, unless the context or description clearly indicates otherwise.


Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Representative illustrative methods and materials are herein described; methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention.


All publications and patents cited in this specification are herein incorporated by reference as if each individual publication or patent were specifically and individually indicated to be incorporated by reference, and are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual dates of public availability and may need to be independently confirmed.


It is noted that, as used herein and in the appended claims, the singular forms “a”, “an”, and “the” include plural referents unless the context clearly dictates otherwise. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as support for the recitation in the claims of such exclusive terminology as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative” limitations, such as “wherein [a particular feature or element] is absent”, or “except for [a particular feature or element]”, or “wherein [a particular feature or element] is not present (included, etc.) . . . ”.


As will be apparent to those of skill in the art upon reading this disclosure, each of the individual embodiments described and illustrated herein has discrete components and features which may be readily separated from or combined with the features of any of the other several embodiments without departing from the scope or spirit of the present invention. Any recited method can be carried out in the order of events recited or in any other order which is logically possible.


REFERENCES

Each of the references cited herein is expressly incorporated herein by reference in its entirety.

  • Alsaadi, E. A. J., “Membrane binding proteins of coronaviruses” FutureVirol 14:4 (275-286) 2019.
  • Alvim, R. G. F., “Zika virus-like particles (VLPs): Stable cell lines and continuous perfusion processes as a new potential vaccine manufacturing platform”, Vaccine 37 (6970-6977) 2019.
  • Ammerman, N. C., “Growth and Maintenance of Vero Cell Lines”, Curr Protoc Microbiol. November 2008 Andersen, K. G., “The proximal origin of SARS-CoV-2”, Nature Medicine, 26 (450-455) April 2020 Arevalo, M. T., “Expression and Purification of Virus-like Particles for Vaccination”, Journal of Visualized Experiments June 2016|112|e54041|.
  • Bestle, D., “TMPRSS2 and furin are both essential for proteolytic activation and spread of SARS-CoV-2 in human airway epithelial cells and provide promising drug targets”, bioRxiv preprint doi: doi.org/10.1101/2020.04.15.042085.
  • Bianchi, M. “SARS-CoV-2 Envelope and Membrane proteins: differences from closely related proteins linked to cross-species transmission?” doi:10.20944/preprints202004.0089.v1 Apr. 2020.
  • Blanco-Melo, D., “Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19”, Cell 181 (1-10) May 2020.
  • Burton, D. R. “Rational Vaccine Design in the Time of COVID-19”, Cell Host & Microbe 27, May 13, 2020 Cao, X., “COVID-19: immunopathology and its implications for therapy”, Nature Reviews Immunology, 20 (260) May 2020.
  • Ceraolo, C., “Genomic variance of the 2019-nCoV coronavirus”, J Med Virol. 92 (522-528) 2020 Chakraborti, S. “Symptomatic SARS-CoV-2 infections display specific IgG Fc structures”, medRxiv preprint doi: doi.org/10.1101/2020.05.15.20103341.
  • Chen, W. H., “Yeast-Expressed SARS-CoV Recombinant Receptor-Binding Domain (RBD219-N1) Formulated with Alum Induces Protective Immunity and Reduces Immune Enhancement”, bioRxiv preprint doi: doi.org/10.1101/2020.05.15.098079.
  • Coleman, C. M., “Purified coronavirus spike protein nanoparticles induce coronavirus neutralizing antibodies in mice”, Vaccine 32 (3169-3174) 2014.
  • Doremalen, N. V., “ChAdOx1 nCoV-19 vaccination prevents SARS-CoV-2 pneumonia in rhesus macaques”, doi.org/10.1101/2020.05.13.093195.
  • Ejemel, M., “IgA MAb blocks SARS-CoV-2 Spike-ACE2 interaction providing mucosal immunity”, bioRxiv preprint doi: doi.org/10.1101/2020.05.15.096719.
  • Elshabrawy, H. A., “Human Monoclonal Antibodies against Highly Conserved HR1 and HR2 Domains of the SARS-CoV Spike Protein Are More Broadly Neutralizing”, PLOS ONE 7:11, e50366, November 2012 Feng Z., “The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directly decimates human spleens and lymph nodes” medRxiv doi: 10.1101/2020.03.27.20045427 2020.
  • Fleming, A. B., “Current studies of convalescent plasma therapy for COVID-19 may underestimate risk of antibody-T dependent enhancement”, J Clin Virol 127, 104388 (2020).
  • Fox, S. E., “Pulmonary and Cardiac Pathology in Covid-19: The First Autopsy Series from New Orleans”, medRxiv preprint doi: doi.org/10.1101/2020.04.06.20050575.
  • Gao, Q., “Rapid development of an inactivated vaccine for SARS-CoV-2”, bioRxiv preprint doi: doi.org/10.1101/2020.04.17.046375.
  • Graham, B. S., “Rapid COVID-19 vaccine development”, Science 10, 1126 (2020).
  • Hakkanson-McReynolds, S., “Solution Structure of the Severe Acute Respiratory Syndrome-Coronavirus Heptad Repeat 2 Domain in the Prefusion State” J Biol Chem 281:17 (2006).
  • Herold, T., “Level of IL-6 predicts respiratory failure in hospitalized symptomatic COVID-19 patients” medRxiv preprint doi: doi.org/10.1101/2020.04.01.20047381 2020.
  • Hotez, P. J., “COVID-19 vaccine design: the Janus face of immune enhancement”, Nature Reviews Immunology.
  • Huang, C., “Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China” Lancet 395 (497-506) 2020.
  • Iwasaki, A., “The potential danger of suboptimal antibody responses in COVID-19”, Nature Reviews Immunology.
  • Jaume, M., “Anti-Severe Acute Respiratory Syndrome Coronavirus Spike Antibodies Trigger Infection of Human Immune Cells via a pH- and Cysteine Protease-Independent FcgR Pathway” J Virol, (10582-10597) 2011.
  • Jespersen, et al. BepiPred-2.0: improving sequence-based B-cell epitope prediction using conformational epitopes. Nucleic Acids Res. 45, W24-W29, 2017.
  • Jiang, S., “Don't rush to deploy COVID-19 vaccines and drugs”, Nature 579 (321) 2020.
  • Kamikubo, Y., “Paradoxical dynamics of SARS-CoV-2 by herd immunity and antibody-dependent enhancement” Cambridge Open Engage doi:10.33774/coe-2020-fsnb3 2020.
  • Katzelnick, L. C., “Antibody-dependent enhancement of severe dengue disease in humans”, Science 358 (929-932) 2017.
  • Knowles, S., “Linear scalability of virus production in the integrity@ iCELLis® single-use fixed-bed bioreactors from bench to industrial scale”, BMC Proceedings 7:6 (60) 2013.
  • Liang, M. F., “SARS Patients-derived Human Recombinant Antibodies to S and M Proteins Efficiently Neutralize SARS-Coronavirus Infectivity” Biomed & Env Sci 18, (363-374) 2005.
  • Lip, K. M., “Monoclonal Antibodies Targeting the HR2 Domain and the Region Immediately Upstream of the HR2 of the S Protein Neutralize In Vitro Infection of Severe Acute Respiratory Syndrome Coronavirus”, J Virol (941-950) 2006.
  • Liu, L., “Anti-spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection”, JCI Insight. 4:4 (e123158) 2019.
  • Liu, Y., “2019-novel coronavirus (2019-nCoV) infections trigger an exaggerated cytokine response aggravating lung injury” ChinaXiv.
  • Martini, A., “A Universal Plug-and-Display Vaccine Carrier Based on HBsAg VLP to Maximize Effective Antibody Response”, Frontiers in Immunology 10 (2931) 2019.
  • Monteil, V., “Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2”, Cell 181 (1-9) 2020.
  • Naskalsak, A. “Membrane Protein of Human Coronavirus NL63 Is Responsible for Interaction with the Adhesion Receptor” J Virol 93:19 (1-14) 2019.
  • Pinto, D., “Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody”, Nature doi.org/10.1038/s41586-020-2349-y, April 2020.
  • Ramasamy, V. “A tetravalent virus-like particle vaccine designed to display domain III of dengue envelope proteins induces multi-serotype neutralizing antibodies in mice and macaques which confer protection against antibody dependent enhancement in AG129 mice”, PLOS Neglected Tropical Diseases|doi.org/10.1371/joumal.pntd.0006191 2018.
  • Richardson, S., “Presenting Characteristics, Comorbidities, and Outcomes Among 5700 Patients Hospitalized With COVID-19 in the New York City Area”, JAMA doi:10.1001/jama.2020.6775, Ricke, D. O., “Medical Countermeasures Analysis of 2019-nCoV and Vaccine Risks for Antibody-dependent Enhancement (ADE)”.
  • Robianni, D. F., “Convergent Antibody Responses to SARS-CoV-2 Infection in Convalescent Individuals”, bioRxiv preprint doi: doi.org/10.1101/2020.05.13.092619.
  • Sardu, C., “Is COVID-19 an endothelial disease? Clinical and basic evidence”, doi:10.20944/preprints202004.0204.v1.
  • Shang, J., “Cell entry mechanisms of SARS-CoV-2”, Proc Natl Acad Sci (USA) 117: 21 (11727-11734) 2020 Tan, L., “Lymphopenia predicts disease severity of COVID-19: a descriptive and predictive study”, Signal Transduction and Targeted Therapy 5 (33) 2020.
  • Tetro, J., “Is COVID-19 receiving ADE from other coronaviruses?”, Microbes and Infection 22:72 (e73) 20.
  • Tian, S., “Pathological study of the 2019 novel coronavirus disease (COVID-19) through postmortem core biopsies”, Modern Pathology doi.org/10.1038/s41379-020-0536-x.
  • Tseng, C. T., “Immunization with SARS Coronavirus Vaccines Leads to Pulmonary Immunopathology on Challenge with the SARS Virus”, PLoS ONE 7: 4 (e35421) 2012.
  • Varga, Z., “Endothelial cell infection and endotheliitis in COVID-19” Lancet doi.org/10.1016/S0140-6736(20)30937-5.
  • Vita et. al., The immune epitope database (IEDB) 3.0. Nucleic Acids Res. 43, D405-D412, 2014.
  • Walls, A. C., “Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein” Cell 180 (281-292) 2020.
  • Wang, Q., “Immunodominant SARS Coronavirus Epitopes in Humans Elicited both Enhancing and Neutralizing Effects on Infection in Non-human Primates” ACS Infect. Dis. 2 (361-376) 2016.
  • Wang, D., “Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China” JAMA, doi:10.1001/jama.2020.1585 2020.
  • Wang, C., “A human monoclonal antibody blocking SARS-CoV-2 infection” NATURE COMMUNICATIONS doi.org/10.1038/s41467-020-16256-y, 2020 bioRxiv preprint doi: doi.org/10.1101/2020.05.15.096511.
  • Wec, A. Z., “Broad sarbecovirus neutralizing antibodies define a key site of vulnerability on the SARS-CoV-2 spike protein” bioRxiv preprint doi: doi.org/10.1101/2020.05.20.103325.
  • Wrapp, D., “Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation” Science 367 (1260-1263) 2020.
  • Wu, F., “Neutralizing antibody responses to SARS-CoV-2 in a COVID-19 recovered patient cohort and their implications”, medRxiv preprint doi: doi.org/10.1101/2020.03.30.20047365.
  • Wu, F., “A new coronavirus associated with human respiratory disease in China” Nature 579 (12) 2020 Wu, C. Y., “Mammalian Expression of Virus-Like Particles for Advanced Mimicry of Authentic Influenza Virus” PLoS ONE 5:3 (e9784) 2010.
  • Xia, S., “Inhibition of SARS-CoV-2 (previously 2019-nCoV) infection by a highly potent pan-coronavirus fusion inhibitor targeting its spike protein that harbors a high capacity to mediate membrane fusion” Cell Research 30 (343-355) 2020.
  • Xu, Z., “Pathological findings of COVID-19 associated with acute respiratory distress syndrome” Lancet Respir Med 8 (420-422) 2020.
  • Yang, Z. Y., “Evasion of antibody neutralization in emerging severe acute respiratory syndrome coronaviruses” Proc Natl Acad Sci (USA) 102:3 (797-801) 2005.
  • Yip, M. S., “Antibody-dependent infection of human macrophages by severe acute respiratory syndrome coronavirus”, Virology Journal 11 (82) 2014.
  • Yuan, M., “A highly conserved cryptic epitope in the receptor binding domains of SARS-CoV-2 and SARS-CoV” Science 368 (630-633) 2020.
  • Zhao, J., et al “Antibody responses to SARS-CoV-2 in patients of novel coronavirus disease 2019” doi.org/10.1101/2020.03.02.20030189, 2020.
  • Zhou, et al. SEPPA 3.0—enhanced spatial epitope prediction enabling glycoprotein antigens. Nucleic Acids Res. 47, W388-W394, 2019.

Claims
  • 1. An immunogenic composition comprising a virus-like particle (VLP) and at least one antigenic polypeptide displayed on the surface of the VLP, wherein the at least one antigenic polypeptide is derived from one or more domains of a coronavirus spike (S) protein, a coronavirus membrane (M) protein or combinations thereof.
  • 2. The immunogenic composition of claim 1, wherein the one or more domains of the S protein include a heptad repeat 1 (HR1) domain.
  • 3. The immunogenic composition of claim 1, wherein the one or more domains of the S protein include a heptad repeat 2 (HR2) domain.
  • 4. The immunogenic composition of claim 1, wherein the one or more domains of the S protein include a linker region between the HR1 and HR2 domains.
  • 5. The immunogenic composition of claim 1, wherein the one or more domains of the S protein include a receptor binding domain (RBD).
  • 6. The immunogenic composition of claim 1, wherein the at least one antigenic polypeptide includes one or more polypeptides having at least 95% sequence identity to at least one of SEQ ID NO: 151, SEQ ID NO: 153, SEQ ID NO: 155, SEQ ID NO: 157, SEQ ID NO: 159, SEQ ID NO: 161, SEQ ID NO: 163, and SEQ ID NO: 165.
  • 7. The immunogenic composition of claim 1, wherein the at least one antigenic polypeptide includes one or more polypeptides having at least 95% sequence identity to at least one of SEQ ID Nos 1-150.
  • 8. The immunogenic composition of claim 1, wherein the at least one antigenic polypeptide includes a plurality of polypeptides as set forth in SEQ ID Nos 1-7, 83-102, and 135-150.
  • 9. The immunogenic composition of claim 1, wherein the at least one antigenic polypeptide is linked to one or more of: an amino acid linker; a signal sequence; a stop transfer sequence; a transmembrane domain; a protein purification ligand; a heterologous protein; or one or more additional antigenic polypeptides comprising any one of SEQ ID NOs: 1-150.
  • 10. The immunogenic composition of claim 1, wherein the VLP has at least 95% sequence identity to SEQ ID NO: 167.
  • 11. The immunogenic composition of claim 1, further comprising at least one pharmaceutically acceptable adjuvant that increases immunogenicity of the VLP.
  • 12. The immunogenic composition of claim 1, wherein the coronavirus is a betacoronavirus.
  • 13. The immunogenic composition of claim 12, wherein the coronavirus is SARS-CoV-2.
  • 14. The immunogenic composition of claim 1, wherein the VLP is formed from a hepatitis B surface antigen S protein (HBV-S).
  • 15. A method of inducing an immune response to at least one coronavirus antigen in a subject in need thereof, comprising administering an effective amount of the immunogenic composition of claim 1 to the subject.
  • 16. The method of claim 15, further comprising the steps of: allowing a suitable period of time to elapse; andadministering at least one additional dose of the immunogenic composition of claim 1 to the subject.
  • 17. An immunogenic composition comprising a nucleotide sequence encoding a VLP; andat least one antigenic polypeptide displayed on the surface of the VLP, wherein the at least one antigenic polypeptide is derived from one or more domains of a coronavirus spike (S) protein, a coronavirus membrane (M) protein or combinations thereof.
  • 18. The immunogenic composition of claim 17, wherein the nucleotide sequence is a DNA sequence.
  • 19. The immunogenic composition of claim 17, wherein the nucleotide sequence is a RNA sequence.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority to U.S. Provisional Application 63/037,367 filed on Jun. 10, 2020. The complete content thereof is herein incorporated by reference.

Provisional Applications (1)
Number Date Country
63037367 Jun 2020 US