WO2023034991A1 - Formulations de vaccin à arnm et leurs procédés d'utilisation - Google Patents

Formulations de vaccin à arnm et leurs procédés d'utilisation Download PDF

Info

Publication number
WO2023034991A1
WO2023034991A1 PCT/US2022/075944 US2022075944W WO2023034991A1 WO 2023034991 A1 WO2023034991 A1 WO 2023034991A1 US 2022075944 W US2022075944 W US 2022075944W WO 2023034991 A1 WO2023034991 A1 WO 2023034991A1
Authority
WO
WIPO (PCT)
Prior art keywords
rna
mrna
protein
cov
sars
Prior art date
Application number
PCT/US2022/075944
Other languages
English (en)
Inventor
Waithaka MWANGI
Huldah SANG
Robert K. Delong
Juergen A. Richt
Original Assignee
Kansas State University Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Kansas State University Research Foundation filed Critical Kansas State University Research Foundation
Publication of WO2023034991A1 publication Critical patent/WO2023034991A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5115Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/22Vectors comprising a coding region that has been codon optimised for expression in a respective host

Definitions

  • This disclosure relates to mRNA compositions, more particularly to mRNA vaccine formulations.
  • SARS-CoV-2 is a novel virus that belongs to the genus betacoronavirus and it is related to Severe Acute Respiratory Syndrome coronavirus (SARS-CoV), Middle East Respiratory Syndrome coronavirus (MERS-CoV) and some bat and pangolin coronaviruses. Similar to SARS-CoV-2 coronaviruses, SARS-CoV-2 utilizes the spike (S) protein to infect host cells by binding to human angiotensinconverting enzyme 2 (ACE2), but with much higher affinity.
  • SARS-CoV-2 utilizes the spike (S) protein to infect host cells by binding to human angiotensinconverting enzyme 2 (ACE2), but with much higher affinity.
  • the S protein which exists as a trimer displayed on the virus surface, contains ACE2 receptor binding domain [RBD]; this knowledge is relevant to vaccine development, since antibodies capable of blocking this interaction can neutralize the virus and stop the infection of cells.
  • Coronavirus virions exhibit the S glycoprotein that binds to host-cell receptors and mediates cell entry via fusion of the host cell and viral membranes.
  • S proteins are trimeric class I fusion glycoproteins that are expressed as a single polypeptide that is subsequently cleaved into S1 and S2 subunits by cellular proteases.
  • the S1 subunit contains the receptor-binding domain (RBD), which, in the case of SARS-CoV-2, recognizes the angiotensin-converting enzyme 2 (ACE2) receptor on the host-cell surface.
  • RBD receptor-binding domain
  • the S2 subunit mediates membrane fusion and contains an additional protease cleavage site, referred to as S2', that is adjacent to a hydrophobic fusion peptide. Binding of the RBD to ACE2 triggers S1 dissociation, allowing for a large rearrangement of S2 as it transitions from a metastable prefusion conformation to a highly stable postfusion conformation. During this rearrangement, the fusion peptide is inserted into the host-cell membrane after cleavage at S2', and two heptad repeats in each protomer associate to form a six-helix bundle that brings together the N- and C-termini of the S2 subunits as well as the viral and host-cell membranes.
  • Attachment and entry - both mediated by the S protein - are essential for the viral life cycle, making the S protein a primary target of neutralizing antibodies and a critical vaccine antigen- Stabilization of the prefusion S protein can be accomplished through a series of mutations that may increase the yield in recombinant expression systems and improve the induction of neutralizing antibodies.
  • Lipid nanoparticle (LNP) mRNA vaccine has been revolutionary for preventing severe COVID19 disease and may extend to treating other infectious diseases and cancer.
  • LNP-RNA temperature instability requiring storage at or below -20-0 °C for the RNA to retain activity. This cold chain requirement greatly limits efficacy and widespread deployment and is considered a contributing factor to the emergence of SARS-CoV-2 variants due to under vaccination.
  • the present disclosure addresses key unmet needs in the art, providing stabilized RNA and/or mRNA vaccines for SARS-CoV-2 that can be distributed and stored at higher temperatures for longer periods of time than those currently available.
  • RNA temperature-stabilization is achieved by binding RNA to zinc nanoparticle coated with protamine (ZNP).
  • ZNP- RNA was characterized by transmission electron microscopy, UV, dynamic light scatter and zeta potential analysis, with RNA payloads ranging from 20.1 -to-124 micrograms RNA per milligram ZNP.
  • Zinc oxide nanoparticle binding to RNA was confirmed by gel shift and best protected RNA from hydrolysis at physiological temperature (37 °C) in comparison to other inorganic nanoparticles commonly used for RNA delivery.
  • RNA structural stabilization was shown by circular dichroism (CD) spectroscopy, dependent on nanoparticle: RNA stoichiometry.
  • protamine coating enhanced expression in cell free lysates and cellular fluorescence after delivery of ZNP-mRNA encoding green fluorescent protein (GFP).
  • GFP green fluorescent protein
  • Temperature-stabilization was shown by differential scanning calorimetry (DSC) in three independent experiments, the RNA melting temperature increasing from 63.9-64.7 to 70.1 -71.6 °C in the presence of nanoparticle.
  • ZNP-RNA suspensions in phosphate buffered saline (PBS) could therefore be stored at 30, 40 and 50 °C for up to two weeks retaining intact RNA as shown by RNA agarose gel electrophoresis (RAGE).
  • PBS phosphate buffered saline
  • ZNP- mRNA (mCherry) incubated at these temperatures also retained expression activity in cell free isolates. ZNP delivery into HEK293 cells was shown for spike mRNA. RNA could also be loaded onto ZNP with cationic antiviral peptide LL37 albeit with less efficiency than protamine which is about 97% efficient and the RNA eluted in Tris-EDTA-urea-heparin buffer. These data suggest ZnO-protamine- RNA (ZNP-RNA) may be useful as an alternative to LNP to increase temperature resistance and enhance pharmaceutical performance of mRNA and potentially other therapeutic RNAs.
  • ZNP-RNA ZnO-protamine- RNA
  • this disclosure relates to a composition
  • a composition comprising a nucleic acid encoding a SARS-CoV-2 S protein; and a pharmaceutically acceptable carrier comprising a zinc oxide nanoparticle.
  • the composition further includes a further ingredient selected from the group consisting of a solvent, a dispersion media, a coating, a stabilizing agent, a diluent, a preservative, an antimicrobial agent, an antifungal agent, an isotonic agent, and an adsorption delaying agent.
  • the protein has at least 90% sequence homology with SEQ ID NO. 1.
  • the pharmaceutically-acceptable carrier comprises a stabilizing agent and/or a preservative and/or an antimicrobial agent.
  • the nucleic acid is at a concentration greater than 100 micrograms per milligram weight of the nanoparticle.
  • the zinc oxide nanoparticle comprises a cell penetrating peptide.
  • the zinc oxide nanoparticle comprises protamine.
  • the nucleic acid further comprises a sequence encoding a CD40L.
  • the nucleic acid encodes a tetrameric CD40L trimer.
  • the SARS-CoV-2 S protein and the CD40L are encoded by a polycistronic mRNA.
  • the nucleic acid encodes a SARS-CoV-2 S protein / CD40L fusion protein.
  • the vector includes an inserted sequence having at least 90% sequence homology with SEQ ID NO. 9 or 10.
  • the nucleic acid is present at a concentration of greater than 100 pg/mg of nanoparticle.
  • compositions described herein are provided. Such uses include reducing the incidence, severity, transmissibility of SARS-CoV-2 infection when administered to a subject in need thereof. Further information regarding these aspects and others are provided below. BRIEF DESCRIPTION OF THE DRAWINGS
  • Fig. 1 is an illustration of a preferred amino acid sequence of the SpikeFTm-2A- CD5 SPD-CD40L polypeptide
  • FIG. 2 is an illustration of a preferred amino acid sequence of the Spike-SPD- CD40L polypeptide
  • FIG. 3 is photograph illustrating protein expression by the four plasmid constructs provided above in transfected HEK 293 cells probed with anti-COVID-19 spike protein specific neutralizing mAbs;
  • FIG 4 is an illustration of the in vivo distribution.
  • Two mice were administered a 2 mg/kg single intravenous dose in 200 mL PBS of either ZnO-NP or cy5.5-ZnO-PEG NP and imaged directly in the bio-imager or sacrificed at 5 hours the brain, heart, lungs, liver, spleen and kidneys were removed and imaged in the bioimager (A).
  • the corresponding mice were sacrificed after 3 days for histopathological analysis (B).
  • B histopathological analysis
  • the tissues were removed and weighed, homogenized in PBS buffer and their relative fluorescence and zinc content per milligram tissue determined by fluorescence spectroscopy and ICP/MS analysis (C).
  • C fluorescence spectroscopy and ICP/MS analysis
  • D nude mouse reference standards
  • Figure 5 Uptake of cy5.5-labeled ZnO NP into Caco-2 3-D organoids (A), cy5.5-ASO delivery into human A375 melanoma cells by ZnO NP or Co3O4 NP relative to cy5.5-ASO control (B) with improved uptake by ZnO, Co3O4 or NiO NP confirmed by flow cytometry (C), finally splicing correction of the aberrant RBD transcript by nanoparticle delivery of ASO targeting that cryptic site (D).
  • FIG. 6 (A) Various nanoscale physiometacomposite materials were fabricated as described in the materials and methods and their nanoscale confirmed by TEM and NTA analysis (see supplemental data). The exact compositions of each type are provided in the summary table including all the oxide combinations, ZnS, FeZnS, MnZnS and MnZnS and others (B). The biocompatibility of the different compositions after 48 hour treatment of continuous exposure in serum containing media to NIH3T3 cells is also shown (C).
  • Figure 7 (A) PMC materials were spiked into PBS, serum, tumor or liver homogenates and their fluorescence versus concentration curves obtained, (B) Bioluminescence assays were conducted in the presence of FeZnS or MnZnS with/without Luciferase enzyme and substrate and the optimal excitation, emission and intensity 3-D plots obtained, (C) similarly these materials or MnZnSe were spiked into tissue slurries and homogenates and the 3-D fluorescence patterns obtained, (D and E) finally ex vivo slices of mouse brain, liver and lung were injected with MnZnS or MnZnSe and imaged directly in the bio-imager.
  • Figure 8 ZnO NP, CoZnO or NiZnO increase cy5.5-poly l:C labeling of 3-D tumor spheroid as shown by bio-imager relative to RNA alone or untreated controls (A), NiZnO or higher order PMC containing iron and manganese inhibited 3-D tumor spheroid growth and ablated these structures (B), and (C) ZnO or CoZnO PMC inhibited B16F10 cell invasion in the scratch assay, the PMC treated material still having the gap filled in by untreated or poly l:C negative controls.
  • Figure 9 (A) High throughput proteomic analysis of B16F10/BALB-C tumor, (b) RBD protein interference or (C) RBD or BCL-xL targeted ASO in B16F10, A375 or 132N1 , ERK/AKT RT-PCR after ZnO or Ni/ZnO (D) or treating drug-resistant canine mucosal melanoma cells with LL37 peptide or RBD ASO and aptamer complexes (F)
  • Figure 10 20 ug/ml inhibition of PMC inhibition of B-Gal biochemical activity by various PMC including MnZnS and FeZnS relative to ZnO NP or silver nanoparticle control.
  • FIG 11 illustrates A) Differential scanning calorimetry (DSC) analysis of poly l:C melting temperature increases from 63.9-64.1 degrees Celsius to greater than 70 deg C in the presence of ZnO NP.
  • DSC Differential scanning calorimetry
  • Figure 12 illustrates temperature-stability (physico-chemical stability) imparted to TY-RNA, when bound to ZnO NP via protamine, dried to a powder and resuspended in sterile PBS can be stored at elevated temperatures for prolonged periods of time (2 weeks). Luciferase mRNA formulations also show expression in vitro (data not shown).
  • Figure 13 illustrates protamine increases TY-RNA payload onto ZnO NP more than LL-37 antiviral peptide.
  • Figure 14 illustrates spike protein expression by mRNA. Expression and validity of mRNA-encoded SARS-CoV-2 spike protein was evaluated by immunocytometric analysis of HEK-293A transfected cells probed with anti-spike mAbs (R2F4 and CR3022). HEK293A cells transfected with the plasmid constructs used to generate mRNA served as positive controls, whereas non-transfected cells served as negative controls.
  • Figures 15A-E illustrate ZNP-RNA characterization by transmission electron microscopy (Fig. 15A), UV spectroscopy (Fig. 15B), nanoparticle size analysis (Fig. 15C), zeta potential (Fig. 15D) and payload (Fig 15E).
  • Figures 16A-D illustrate binding, stability and expression of ZNP-RNA and ZNP-mRNA as shown by RNA agarose gel electrophoresis (RAGE) (Fig. 16A), in vitro translation (Fig. 16B), cell free mRNA expression in lysates (Fig. 16C) and GFP mRNA cell delivery (Fig. 16D)
  • Figures 17A-B illustrate RNA Temperature-stabilization with Fig. 17A providing results from RAGE analysis of heat treated RNA , and Fig. 17B providing an expression analysis from heat treated ZNP-mRNA.
  • FIG. 18A-F illustrate different RNA biological activities with Fig. 18A illustrating loading efficiency and expression of SARS-CoV-2 spike mRNA ⁇ Figs. 18B-F illustrating protein expression by immunostaining of HEK293A cells transfected with SARS-CoV-2 spike mRNA formulated in/as:
  • Fig. 18B Zinc oxide nanoparticle-mRNA
  • Fig. 18C Protamine coated Zinc oxide nanoparticle
  • Fig. 18D Double protamine coated - one over the ZNP and other over mRNA (ZNP-protamine-mRNA- protamine)
  • Fig. 18A Zinc oxide nanoparticle-mRNA
  • FIG. 18E InvitrogenTM LipofectamineTM MessengerMAXTM transfection reagent (positive control); and (Fig. 18F) Mock-transfected cells which served as a negative control.
  • the cells were probed with a recombinant humanized anti-SARS-CoV- 2 spike-specific neutralizing monoclonal antibody.
  • the secondary antibody was antihuman IgG-Alkaline Phosphatase, and Fast Red was the substrate. Arrows show the cells expressing the spike protein.
  • Fig. 19 is a photograph illustrating that RNA elution from the particle is protamine-dependent using protamine high (1 :1 ), medium (1 :10), and low (1 :100);
  • Fig. 20 is a photograph illustrating the impact of buffer components on RNA elution from the particles
  • Fig. 21 is an illustration of the sequence alignment of low molecular weight protamine, LL37, and penetratin peptide
  • Fig. 22 is a graph illustrating the evidence for LL37 functioning as an RNA- binding peptide (RBP);
  • Fig. 23 is a photograph and a graph illustrating quantitative RNA agarose gel electrophoresis (RAGE) with from 0.093 to 3 micrograms RNA loaded per lane, stained, and analyzed as described in materials and methods.
  • RAGE quantitative RNA agarose gel electrophoresis
  • Sequence Identity refers to a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, namely a reference sequence and a given sequence to be compared with the reference sequence. Sequence identity is determined by comparing the given sequence to the reference sequence after the sequences have been optimally aligned to produce the highest degree of sequence similarity, as determined by the match between strings of such sequences. Upon such alignment, sequence identity is ascertained on a position- by-position basis, e.g., the sequences are “identical” at a particular position if at that position, the nucleotides or amino acid residues are identical.
  • Sequence identity can be readily calculated by known methods, including but not limited to, those described in Computational Molecular Biology, Lesk, A. N., ed., Oxford University Press, New York (1988), Biocomputing: Informatics and Genome Projects, Smith, D.W., ed., Academic Press, New York (1993); Computer Analysis of Sequence Data, Part I, Griffin, A.M., and Griffin, H. G., eds., Humana Press, New Jersey (1994); Sequence Analysis in Molecular Biology, von Heinge, G., Academic Press (1987); Sequence Analysis Primer, Gribskov, M.
  • Preferred methods to determine the sequence identity are designed to give the largest match between the sequences tested. Methods to determine sequence identity are codified in publicly available computer programs which determine sequence identity between given sequences. Examples of such programs include, but are not limited to, the GCG program package (Devereux, J., et al., Nucleic Acids Research, 12(1 ):387 (1984)), BLASTP, BLASTN and FASTA (Altschul, S. F.
  • BLASTX program is publicly available from NCBI and other sources (BLAST Manual, Altschul, S. et al., NCVI NLM NIH Bethesda, MD 20894, Altschul, S. F. et al., J. Molec. Biol., 215:403-410 (1990), the teachings of which are incorporated herein by reference). These programs optimally align sequences using default gap weights in order to produce the highest level of sequence identity between the given and reference sequences.
  • nucleotide sequence having at least, for example, 85%, preferably 90%, even more preferably 95% “sequence identity” to a reference nucleotide sequence it is intended that the nucleotide sequence of the given polynucleotide is identical to the reference sequence except that the given polynucleotide sequence may include up to 15, preferably up to 10, even more preferably up to 5 point mutations per each 100 nucleotides of the reference nucleotide sequence.
  • a polynucleotide having a nucleotide sequence having at least 85%, preferably 90%, even more preferably 95% identity relative to the reference nucleotide sequence up to 15%, preferably 10%, even more preferably 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 15%, preferably 10%, even more preferably 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence.
  • mutations of the reference sequence may occur at the 5’ or 3’ terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence.
  • a polypeptide having a given amino acid sequence having at least, for example, 85%, preferably 90%, even more preferably 95% sequence identity to a reference amino acid sequence it is intended that the given amino acid sequence of the polypeptide is identical to the reference sequence except that the given polypeptide sequence may include up to 15, preferably up to 10, even more preferably up to 5 amino acid alterations per each 100 amino acids of the reference amino acid sequence.
  • a given polypeptide sequence having at least 85%, preferably 90%, even more preferably 95% sequence identity with a reference amino acid sequence up to 15%, preferably up to 10%, even more preferably up to 5% of the amino acid residues in the reference sequence may be deleted or substituted with another amino acid, or a number of amino acids up to 15%, preferably up to 10%, even more preferably up to 5% of the total number of amino acid residues in the reference sequence may be inserted into the reference sequence.
  • These alterations of the reference sequence may occur at the amino or the carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in the one or more contiguous groups within the reference sequence.
  • Sequence homology refers to a method of determining the relatedness of two sequences. To determine sequence homology, two or more sequences are optimally aligned, and gaps are introduced if necessary. However, in contrast to “sequence identity”, conservative amino acid substitutions are counted as a match when determining sequence homology.
  • a polypeptide or polynucleotide having 95% sequence homology with a reference sequence 85%, preferably 90%, even more preferably 95% of the amino acid residues or nucleotides in the reference sequence must match or comprise a conservative substitution with another amino acid or nucleotide, or a number of amino acids or nucleotides up to 15%, preferably up to 10%, even more preferably up to 5% of the total amino acid residues or nucleotides, not including conservative substitutions, in the reference sequence may be inserted into the reference sequence.
  • the homologous sequence comprises at least a stretch of 50, even more preferably 100, even more preferably 250, even more preferably 500 nucleotides.
  • a “conservative substitution” refers to the substitution of an amino acid residue or nucleotide with another amino acid residue or nucleotide having similar characteristics or properties including size, hydrophobicity, etc., such that the overall functionality does not change significantly.
  • the immunogenic compositions comprising any of the disclosed vaccine candidates as provided herewith are very effective in reducing the severity of or incidence of clinical signs associated with coronavirus infections including COVID-19 up to and including the prevention of such signs. Further, such immunogenic compositions reduce the transmissibility of COVID-19.
  • the immunogenic compositions described herein can further include one or more other immunomodulatory agents such as, e. g., interleukins, interferons, or other cytokines.
  • the immunogenic compositions can also include Gentamicin and Merthiolate.
  • the present disclosure contemplates vaccine compositions comprising from about 1 ug/ml to about 60 pg/ml of antibiotics, and more preferably less than about 30 pg/ml of antibiotics.
  • immunogenic protein refers to any amino acid sequence which elicits an immune response in a host against a pathogen comprising said immunogenic protein, immunogenic polypeptide or immunogenic amino acid sequence.
  • immunogenic fragment is meant a fragment of a protein which includes one or more epitopes and thus elicits the immunological response against the relevant pathogen.
  • Such fragments can be identified using any number of epitope mapping techniques, well known in the art. See, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66 (Glenn E. Morris, Ed., 1996) Humana Press, Totowa, New Jersey.
  • linear epitopes may be determined by e.g., concurrently synthesizing large numbers of peptides on solid supports, the peptides corresponding to portions of the protein molecule, and reacting the peptides with antibodies while the peptides are still attached to the supports.
  • Such techniques are known in the art and described in, e.g., U.S. Patent No. 4,708,871 ; Geysen et al.
  • conformational epitopes are readily identified by determining spatial conformation of amino acids such as by, e.g., x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols, supra.
  • Synthetic antigens are also included within the definition, for example, polyepitopes, flanking epitopes, and other recombinant or synthetically derived antigens. See, e.g., Bergmann et al. (1993) Eur. J. Immunol.
  • immunogenic proteins of the present disclosure include the S protein sequence, the F-Tm sequence, the SPD sequence, and the CD40L sequence.
  • polypeptide In the present description, the terms polypeptide, peptide and protein are interchangeable. [0044] In the present description, COVID-19 (the disease) and SARS-CoV2 (the causative agent) are used interchangeably.
  • any composition or vaccine candidate of the disclosure can include one or more pharmaceutical-acceptable or veterinary-acceptable carriers.
  • a pharmaceutical-acceptable carrier or “veterinary-acceptable carrier” includes any and all solvents, dispersion media, coatings, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, adsorption delaying agents, and the like.
  • the pharmaceutical or veterinary acceptable carrier is selected from the group consisting of a solvent, a dispersion media, a coating, a stabilizing agent, a preservative, an antimicrobial agent, an antifungal agent, an isotonic agent, an adsorption delaying agent, and any combination thereof.
  • the immunogenic compositions described herein can be administered to any animal susceptible to coronavirus infection including humans, dogs, cats, ferrets, bats, cattle, camels, hamsters, horses, chimps, gorillas, anteaters, dolphins, alligators, and sheep. Further, administration of any of the immunogenic compositions described herein will reduce the incidence of, severity of, and transmission of SARS-CoV2.
  • an “immunogenic or immunological composition” refers to a composition of matter that (i) comprises at least one antigen which elicits an immunological response in the host of a cellular and/ or antibody-mediated immune response to the composition or vaccine of interest, or (ii) comprises a nucleic acid such as a plasmid DNA or messenger RNA encoding such an antigen.
  • an “immunological response” includes but is not limited to one or more of the following effects: the production or activation of antibodies, B cells, helper T cells, suppressor T cells, and/or cytotoxic T cells and/or yS T cells, directed specifically to an antigen or antigens included in the composition or vaccine of interest.
  • the host will display either a therapeutic or protective immunological response such that resistance to new infection will be enhanced and/or the clinical severity of the disease reduced.
  • Such protection will be demonstrated by either a reduction in the severity or prevalence of, up to and including a lack of symptoms normally displayed by an infected host, a quicker recovery time and/or a lowered viral titer in the infected host.
  • a “reduction” in terms of incidence of symptoms, severity of symptoms, or transmissibility of infection is understood to encompass a comparison to a subject or group of subjects that has not received an administration of a composition of the present disclosure.
  • the reduction is at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 95, or even 100% in comparison to an animal that has not received an administration or dose of a composition described herein. It is understood that this percentage of reduction can be in terms of the number of symptoms, incidence of symptoms, severity of symptoms, and transmissibility of infection. Further, this can apply to individual animals or groups of animals wherein the reduction is in terms of a percentage of animals infected or showing symptoms vs uninfected or not showing symptoms.
  • adjuvants can include aluminum hydroxide and aluminum phosphate, saponins e.g., Quil A, QS-21 (Cambridge Biotech Inc., Cambridge MA), GPI-0100 (Galenica Pharmaceuticals, Inc., Birmingham, AL), water-in-oil emulsion, oil- in-water emulsion, water-in-oil-in-water emulsion.
  • the emulsion can be based in particular on light liquid paraffin oil (European Pharmacopea type); isoprenoid oil such as squalane or squalene oil resulting from the oligomerization of alkenes, in particular of isobutene or decene; esters of acids or of alcohols containing a linear alkyl group, more particularly plant oils, ethyl oleate, propylene glycol di-(caprylateZcaprate), glyceryl tri- (caprylate/caprate) or propylene glycol dioleate; esters of branched fatty acids or alcohols, in particular isostearic acid esters.
  • light liquid paraffin oil European Pharmacopea type
  • isoprenoid oil such as squalane or squalene oil resulting from the oligomerization of alkenes, in particular of isobutene or decene
  • esters of acids or of alcohols containing a linear alkyl group more
  • the oil is used in combination with emulsifiers to form the emulsion.
  • the emulsifiers are preferably nonionic surfactants, in particular esters of sorbitan, of mannide (e.g. anhydromannitol oleate), of glycol, of polyglycerol, of propylene glycol and of oleic, isostearic, ricinoleic or hydroxystearic acid, which are optionally ethoxylated, and polyoxypropylene-polyoxyethylene copolymer blocks, in particular the Pluronic products, especially L121 .
  • mannide e.g. anhydromannitol oleate
  • glycol of polyglycerol
  • propylene glycol and of oleic isostearic, ricinoleic or hydroxystearic acid, which are optionally ethoxylated, and polyoxypropylene-polyoxyethylene copolymer blocks, in particular the Pluronic
  • a further instance of an adjuvant is a compound chosen from the polymers of acrylic or methacrylic acid and the copolymers of maleic anhydride and alkenyl derivative.
  • Advantageous adjuvant compounds are the polymers of acrylic or methacrylic acid which are cross-linked, especially with polyalkenyl ethers of sugars or polyalcohols. These compounds are known by the term carbomer (Phameuropa Vol. 8, No. 2, June 1996). Persons skilled in the art can also refer to U. S. Patent No.
  • 2,909,462 which describes such acrylic polymers cross-linked with a polyhydroxylated compound having at least 3 hydroxyl groups, preferably not more than 8, the hydrogen atoms of at least three hydroxyls being replaced by unsaturated aliphatic radicals having at least 2 carbon atoms.
  • the preferred radicals are those containing from 2 to 4 carbon atoms, e.g. vinyls, allyls and other ethylenically unsaturated groups.
  • the unsaturated radicals may themselves contain other substituents, such as methyl.
  • the products sold under the name Carbopol; (BF Goodrich, Ohio, USA) are particularly appropriate. They are cross-linked with an allyl sucrose or with allyl pentaerythritol.
  • Carbopol 974P, 934P and 971 P there may be mentioned Carbopol 974P, 934P and 971 P.
  • the copolymers of maleic anhydride and alkenyl derivative the copolymers EMA (Monsanto) which are copolymers of maleic anhydride and ethylene.
  • the dissolution of these polymers in water leads to an acid solution that will be neutralized, preferably to physiological pH, in order to give the adjuvant solution into which the immunogenic, immunological or vaccine composition itself will be incorporated.
  • Suitable adjuvants include, but are not limited to, the RIBI adjuvant system (Ribi Inc.), Block co-polymer (CytRx, Atlanta GA), SAF-M (Chiron, Emeryville CA), monophosphoryl lipid A, Avridine lipid-amine adjuvant, heat-labile enterotoxin from E. coli (recombinant or otherwise), cholera toxin, IMS 1314 or muramyl dipeptide among many others.
  • a pharmaceutical-acceptable carrier or “veterinary- acceptable carrier” includes any and all solvents, dispersion media, coatings, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, adsorption delaying agents, and the like.
  • chemotherapeutic nanoparticle that can deliver targeted therapies to affected tissues is imperative.
  • SARS-COV-2 infects cells in the lung, but the virus is also detected and causes pathology in the liver, kidney, spleen and brain.
  • Zinc oxide (ZnO) nanoparticle (NP) is now considered a chemotherapeutic nanoparticle and its antimicrobial, antiviral and anticancer activity has now been well-characterized.
  • the inventors’ research group has studied the interaction mechanisms of zinc oxide nanoparticle (ZnO NP) to RNA extensively and patented 2-dimensional fluorescence difference spectroscopy as a quality control assay for RNA and protein bound ZnO NP.
  • ZnO NP zinc oxide nanoparticle
  • One area of particular interest among the inventors’ group has been the anticancer/antitumor properties of this material with more recent results as a RAS- targeted nanomedicine.
  • ZnO NP being capable of activating cellular and molecular immunity
  • RNA vaccines are under even greater such restrictions, we have begun to examine the effect of zinc-based nanoparticles on RNA stability, expression and activity. For example, we developed a scalable method to synthesize zinc-based physiometacomposite (PMC) nanoparticles finding that these have improved biocompatibility, fluorescence and bioluminescence characteristics. As an initial coronavirus model, porcine reproductive respiratory virus (PRRSV) was chosen, the nanoparticles screened, with MnZnS demonstrating significant inhibition (Fig. 5).
  • PRRSV porcine reproductive respiratory virus
  • RNA molecules we have used poly l:C or torula yeast RNA (TY-RNA) and examined the effect initially of ZnO NP on RNA structure and temperature stability by differential scanning calorimetry (DSC) and by circular dichroism spectroscopy (CD) (Fig. 6).
  • DSC differential scanning calorimetry
  • CD circular dichroism spectroscopy
  • Fig. 7 We next examined the temperaturestability imparted by binding protamine to ZnO NP prior to RNA attachment (Fig. 7) and its increase in particle-associated RNA relative to antiviral LL37 peptide (Fig. 8).
  • compositions including without limitation immunogenic compositions and/or vaccine compositions, comprising nucleic acids encoding a full-length or partial-length S protein of SARS-CoV- 2 and a pharmaceutically or veterinary acceptable carrier comprising a Zinc oxide (ZnO) nanoparticle (NP) or physiometacomposite (PMC).
  • ZnO Zinc oxide
  • NP nanoparticle
  • PMC physiometacomposite
  • nucleic acids used in these compositions are generally characterized in that they are selected from one or more of: a) a nucleotide sequence encoding a specific fragment of the sequence of SEQ ID No. 1 or one of its fragments; b) a nucleotide sequence homologous to a nucleotide sequence such as defined in a); c) a nucleotide sequence complementary to a nucleotide sequence such as defined in a) or b), and a nucleotide sequence of their corresponding RNA; d) a nucleotide sequence capable of hybridizing under stringent conditions with a sequence such as defined in a), b) or c); e) a nucleotide sequence comprising a sequence such as defined in a), b), c) or d); and f) a nucleotide sequence modified by a nucleotide sequence such as defined in a), b), c), d
  • Nucleotide, polynucleotide or nucleic acid sequence will be understood according to the present disclosure as meaning both a double-stranded or singlestranded DNA in the monomeric and dimeric (so-called in tandem) forms and the transcription products of said DNAs.
  • Nucleic acids of this disclosure can include, without limitation, RNAs such as messenger RNAs (mRNAs), DNAs such as plasmid DNAs (pDNAs), RNA/DNA chimeras and like nucleic acid forms known in the art. These nucleic acids optionally include backbone modifications (e.g., phosphorothioate or morpholino linkages) and/or nucleobase modifications.
  • the present disclosure does not relate to the genomic nucleotide sequences taken in their natural environment, that is to say in the natural state. It concerns sequences which it has been possible to isolate, purify or partially purify, starting from separation methods such as, for example, ion-exchange chromatography, by exclusion based on molecular size, or by affinity, or alternatively fractionation techniques based on solubility in different solvents, or starting from methods of genetic engineering such as amplification, cloning and subcloning, it being possible for the sequences of the disclosure to be carried by vectors. All genes and vectors in the present application were generated using synthetic biology.
  • Complementary nucleotide sequence of a sequence of the disclosure is understood as meaning any DNA whose nucleotides are complementary to those of the sequence of the disclosure, and whose orientation is reversed (antiparallel sequence).
  • Hybridization under conditions of stringency with a nucleotide sequence according to the disclosure is understood as meaning a hybridization under conditions of temperature and ionic strength chosen in such a way that they allow the maintenance of the hybridization between two fragments of complementary DNA.
  • nucleotide sequences according to the disclosure are those corresponding to S sequences, and coding for polypeptides, such as, for example, SEQ ID No. 1.
  • the nucleotide sequence fragments according to the disclosure can be obtained, for example, by specific amplification, such as PCR, or after digestion with appropriate restriction enzymes of nucleotide sequences according to the disclosure, these methods in particular being described in the work of Sambrook et al., 1989.
  • Said representative fragments can likewise be obtained by chemical synthesis when their size is not very large and according to methods well known to persons skilled in the art.
  • Modified nucleotide sequence will be understood as meaning any nucleotide sequence obtained by mutagenesis according to techniques well known to the person skilled in the art, and containing modifications with respect to the normal sequences according to the disclosure, for example mutations in the regulatory and/or promoter sequences of polypeptide expression, especially leading to a modification of the rate of expression of said polypeptide or to a modulation of the replicative cycle.
  • Modified nucleotide sequence will likewise be understood as meaning any nucleotide sequence coding for a modified polypeptide such as defined below.
  • the present disclosure relates to nucleotide sequences of SARS-CoV-2 according to the disclosure, characterized in that they are selected from the sequences encoding SEQ ID No. 1 or sequences having at least 80, 85, 88, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.6, 99.7, 99.8, 99.9, or even 100% sequence identity or homology to SEQ ID NO. 1 , or one of their fragments.
  • the disclosure likewise relates to nucleotide sequences characterized in that they comprise a nucleotide sequence selected from: a) a nucleotide sequence encoding SEQ ID No. 1 , or one of their fragments; b) a nucleotide sequence of a specific fragment of a sequence such as defined in a); c) a homologous nucleotide sequence having at least 80, 85, 88, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.6, 99.7, 99.8, 99.9, or even 100%% identity with a sequence such as defined in a) or b); d) a complementary nucleotide sequence or sequence of RNA corresponding to a sequence such as defined in a), b) or c); and e) a nucleotide sequence modified by a sequence such as defined in a), b), c) or d).
  • homologous, especially specific, sequences having a percentage identity with SEQ ID No.1 , or one of their fragments of at least 80, 85, 88, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.6, 99.7, 99.8, 99.9, or even 100% are preferred.
  • Said specific homologous sequences can comprise, for example, the sequences corresponding to S protein sequences of SARS-CoV-2.
  • these specific homologous sequences can correspond to variations linked to mutations within strains of SARS-CoV-2 and especially correspond to truncations, substitutions, deletions and/or additions of at least one nucleotide.
  • compositions according to this aspect of the disclosure generally comprise a nucleotide sequence that is (a) extracted or derived from the genome of SARS-CoV-2, and/or (b) encodes a SARS-CoV-2 gene product, for instance the portion encoding the SARS-CoV-2 S protein of SEQ ID NO. 1.
  • the nucleotide sequence encodes SARS-CoV-2 S protein that has at least 80, 85, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.6, 99.7, 99.8, 99.9, or even 100% sequence homology with SEQ ID NO.
  • the signal sequence is a F signal protein, preferably SEQ ID No. 2.
  • the linker is SEQ ID NO. 3.
  • a tag such as a His tag or FLAG tag is included, but not required.
  • the fusion protein transmembrane is SEQ ID NO. 4.
  • the 2A cleavable peptide is SEQ ID NO. 5.
  • the signal sequence is a CD5 signal sequence, preferably SEQ ID NO. 6.
  • the surfactant protein tetramerization domain is SEQ ID NO. 7.
  • the CD40L is SEQ ID NO. 8.
  • any SARS-CoV-2 S protein would be effective as the source of the SARS- CoV-2 S encoding sequence and/or polypeptide as used herein.
  • the nucleic acid encodes a polypeptide having at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.6, 99.7, 99.8, 99.9, or even 100% sequence homology or sequence identity with SEQ ID NO. 1.
  • a suitable SARS-CoV-2 S protein is that of SEQ ID NO. 1 , but it will be understood by those of skill in the art that this sequence could vary by as much as 10-20% in sequence homology and still retain the antigenic characteristics that render it useful in immunogenic compositions.
  • the antigenic characteristics of an immunological composition can be, for example, estimated by challenge experiments. Moreover, the antigenic characteristic of a modified antigen is still retained, when the modified antigen confers at least 70%, preferably 80%, more preferably 90, 91 , 92, 93, 94, 95, 96, 97, 98, or 99% of the protective immunity as compared to the SARS-CoV-2 S protein of SEQ ID NO. 1.
  • immunogenic portions of a SARS-CoV-2 S protein are used as the antigenic component in the composition.
  • immunogenic portion refers to truncated and/or substituted forms, or fragments of SARS-CoV-2 S protein and/or polynucleotide, respectively.
  • such truncated and/or substituted forms, or fragments will comprise at least 6 contiguous amino acids from the full-length S polypeptide. More preferably, the truncated or substituted forms, or fragments will have at least 10, more preferably at least 15, and still more preferably at least 19 contiguous amino acids from the full-length S polypeptide. It is further understood that such sequences may be a part of larger fragments or truncated forms. Preferably, such truncated or substituted forms, or fragments will comprise at least 18 contiguous nucleotides from the full-length S nucleotide sequence, e.g. of SEQ ID NO. 1. More preferably, the truncated or substituted forms, or fragments will have at least 30, more preferably at least 45, and still more preferably at least 57 contiguous nucleotides the full-length S nucleotide sequence.
  • ZnO NPs are suitable for use in compositions according to this aspect of the disclosure, including without limitation NPs derived from zinc oxide nanopowder compositions characterized by average particle sizes ⁇ 100 nm or by specific surface areas of 15-25 m 2 /g sold by, e.g., Sigma-Aldrich (St. Louis, MO, USA). ZnO NPs be modified to incorporate one or more functional agents that impart cell- or tissue specificity, emit a detectable signal, etc.
  • the ZnO NPs in certain embodiments, further comprise a nucleic acid binding peptide such as a protamine such as recombinant human PRM1 or PRM2, or a pharmaceutically acceptable salt or other derivative thereof.
  • a nucleic acid binding peptide such as a protamine such as recombinant human PRM1 or PRM2, or a pharmaceutically acceptable salt or other derivative thereof.
  • an immunogenic composition effective for lessening the severity and/or reducing the incidence of clinical symptoms associated with SARS-CoV-2 infection, and/or reducing the transmissibility of SARS- CoV-2, comprising a nucleic acid encoding a SARS-CoV-2 S protein is provided.
  • the SARS-CoV-2 S protein encoded by the nucleic acid is selected from the group consisting of: 1 ) a polypeptide comprising the sequence of SEQ ID NO: 1 ; 2) any polypeptide that is at least 90% homologous to the polypeptide of 1 ); 3) any immunogenic portion of the polypeptides of 1 ) and/or 2); or 4) the immunogenic portion of 3), comprising at least 10 contiguous amino acids included in the sequence of SEQ ID NO: 1.
  • these immunogenic portions will have the immunogenic characteristics of SARS-CoV-2 S protein that has at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.6, 99.7, 99.8, 99.9, or even 100% sequence homology with SEQ ID NO. 1.
  • compositions of the present disclosure may incorporate known injectable, physiologically acceptable, sterile solutions.
  • aqueous isotonic solutions such as e.g. saline or corresponding plasma protein solutions are readily available.
  • the immunogenic and vaccine compositions of the present disclosure can include diluents, isotonic agents, stabilizers, or adjuvants. Diluents can include water, saline, dextrose, ethanol, glycerol, and the like.
  • Isotonic agents can include sodium chloride, dextrose, mannitol, sorbitol, and lactose, among others.
  • Stabilizers include albumin and alkali salts of ethylendiamintetracetic acid, among others. Suitable adjuvants, are those described above.
  • the immunogenic composition of the present disclosure further comprises a pharmaceutical acceptable salt, preferably a phosphate salt in physiologically acceptable concentrations.
  • a pharmaceutical acceptable salt preferably a phosphate salt in physiologically acceptable concentrations.
  • the pH of said immunogenic composition is adjusted to a physiological pH, meaning between about 6.5 and 7.5.
  • the immunogenic compositions described herein can further include one or more other immunomodulatory agents such as, e. g., interleukins, interferons, or other cytokines.
  • the immunogenic compositions can also include Gentamicin and Merthiolate.
  • the present disclosure contemplates vaccine compositions comprising from about 1 ug/ml to about 60 pg/ml of antibiotics, and more preferably less than about 30 pg/ml of antibiotics.
  • the immunogenic compositions comprising nucleic acids encoding recombinant SARS-CoV-2 S protein as provided herewith are very effective in reducing the severity of or incidence of clinical signs associated with SARS-CoV-2 infections up to and including the prevention of such signs. Additionally, the immunogenic compositions will be effective at reducing the transmissibility of SARS- CoV-2.
  • kits include a container comprising at least one dose of a vaccine composition protein as provided herewith, wherein one dose comprises a pharmaceutically effective quantity of an immunogenic composition comprising a nucleic acid encoding SARS-CoV-2 S protein.
  • Said container can comprise from 1 to 250 doses of the immunogenic composition.
  • the container contains 1 , 10, 25, 50, 100, 150, 200, or 250 doses of the immunogenic composition of nucleic acid encoding SARS- CoV-2 S protein. It may be advantageous, in the case of containers comprising more than one dose of the immunogenic composition, for such compositions to further comprise an anti-microbiological active agent.
  • one aspect of the present disclosure relates to a container that comprises from 1 to 250 doses of the immunogenic composition of a nucleic acid encoding SARS-CoV-2 S protein, wherein one dose comprises a pharmaceutically effective quantity of the immunogenic composition, and an antimicrobial selected from Gentamicin and/or Merthiolate, preferably from about 1 pg/ml to about 60 pg/ml of antibiotics, and more preferably less than about 30 pg/ml.
  • the kit also includes an instruction manual, including the information for the intramuscular application of at least one dose of the immunogenic composition of SARS-CoV-2 S protein into animals, to lessen the incidence and/or severity of clinical symptoms associated with SARS-CoV-2 infection.
  • said instruction manual comprises the information of a second or further administration(s) of at least one dose of the immunogenic composition of SARS-CoV-2 S, wherein the second administration or any further administration is at least 14 days beyond the initial or any former administration.
  • said instruction manual also includes the information, to administer an immune stimulant.
  • said immune stimulant shall be given at least twice.
  • the immune stimulant is given at least 10 days, preferably 15, even more preferably 20, and still even more preferably at least 22 days beyond the initial administration of the immunogenic composition of SARS-CoV-2 S protein.
  • any immune stimulant known to a person skilled in the art can also be used.
  • Immunogen means any agent or composition that can trigger a general immune response, preferably without initiating or increasing a specific immune response, for example the immune response against a specific pathogen. It is further instructed to administer the immune stimulant in a suitable dose.
  • the kit may also comprise a second container, including at least one dose of the immune stimulant.
  • a further aspect of the present disclosure relates to the kit as described above, comprising the immunogenic composition of SARS-CoV-2 S as provided herewith and the instruction manual, wherein the instruction manual further includes the information to administer the SARS-CoV-2 S immunogenic composition together, or around the same time as, with an immunogenic composition that comprises an additional antigen effective for reducing the severity of or incidence of clinical signs related to another mammalian pathogen.
  • the manual contains the information of when the SARS-CoV-2 S containing composition and the immunogenic composition that comprises an additional antigen are administered.
  • a further aspect relates to the use of any of the compositions provided herewith as a medicament, including as a veterinary medicament, even more preferably as a vaccine.
  • the present disclosure also relates to the use of any of the compositions described herein, for the preparation of a medicament for lessening the severity of clinical symptoms associated with SARS-CoV-2 infection.
  • the medicament is for the prevention of a SARS-CoV-2 infection in an animal susceptible to infection with SARS-CoV-2.
  • a further aspect relates to a method for (1 ) the prevention of an infection, or re-infection with SARS-CoV-2 or (2) the reduction in incidence or severity of or elimination of clinical symptoms caused by SARS-CoV-2 in a subject, comprising administering any of the immunogenic compositions provided herewith to a subject in need thereof.
  • the reduction is in comparison to a subject that has not received an administration of a composition of the present disclosure.
  • the reduction is at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 95, or even 100% in comparison to an animal that has not received an administration or dose of a composition described herein. It is understood that this percentage of reduction can be in terms of the number of symptoms, incidence of symptoms, or severity of symptoms.
  • one dose or two doses of the immunogenic composition is/are administered, wherein one dose preferably comprises at least about 2 pg SARS-CoV-2 S protein.
  • a further aspect relates to the method of treatment as described above, wherein a second application of the immunogenic composition is administered.
  • the second administration is done with the same immunogenic composition, preferably having the same amount of SARS-CoV-2 S protein.
  • the second administration is done at least 14 days beyond the initial administration, even more preferably at least 4 weeks beyond the initial administration.
  • the method is effective after just a single dose of the immunogenic composition and does not require a second or subsequent administration in order to confer the protective benefits upon the subject.
  • the present disclosure provides a multivalent combination vaccine which includes an immunological agent effective for reducing the incidence of or lessening the severity of SARS-CoV-2 infection, and at least one immunological active component against another disease-causing organism in mammals.
  • the immunological agent effective for reducing the incidence of or lessening the severity of SARS-CoV-2 infection is a SARS-CoV-2 antigen.
  • said SARS-CoV-2 antigen is a SARS-CoV-2 S protein as provided herewith, or any immunogenic composition as described above, that comprises SARS-CoV-2 S protein, such as a SARS-CoV-2 S protein that has at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.6, 99.7, 99.8, 99.9, or even 100% sequence homology with SEQ ID NO. 1.
  • an “immunological active component” as used herein means a component that induces or stimulates the immune response in an animal to which said component is administered. Said immune response may be directed to said component or to a microorganism comprising said component.
  • the immunological active component is an attenuated microorganism, including modified live virus (MLV), a killed-microorganism or at least an immunological active part of a microorganism.
  • Immunological active part of a microorganism means a protein-, sugar-, and or glycoprotein containing fraction of a microorganism that comprises at least one antigen that induces or stimulates the immune response in an animal to which said component is administered. According to a preferred embodiment, said immune response is directed to said immunological active part of a microorganism or to a microorganism comprising said immunological active part.
  • biologically functional plasmids, viral vectors and the like that contain the new recombinant nucleic acid molecules described herein, suitable host cells transfected by the vectors comprising the molecular DNA clones and the immunogenic polypeptide expression products.
  • immunogenic protein expression products will have the amino acid sequence set forth in SEQ ID NO: 1.
  • the biologically active variants thereof are further encompassed by the disclosure.
  • One of ordinary skill in the art would know how to modify, substitute, delete, etc., amino acid(s) from the polypeptide sequence and produce biologically active variants that retain the same, or substantially the same, activity as the parent sequence without undue effort.
  • the process may include in vitro transcription utilizing a DNA substrate such as a plasmid DNA.
  • a DNA substrate such as a plasmid DNA.
  • the process may include chemical synthesis of oligonucleotides using, e.g., phosphoram idite or other functionalized monomers.
  • the present disclosure relates to vaccines or immunogenic compositions comprising a pharmaceutically acceptable vehicle and a nucleic acid encoding a single polypeptide, wherein the single polypeptide consists of SEQ ID No. 1.
  • the present disclosure relates to methods of immunizing a mammal against SARS-CoV-2 comprising administering to a mammal an effective amount of a vaccine or immunogenic composition described above.
  • the present disclosure likewise encompasses the polypeptides encoded by a nucleotide sequence according to the disclosure, preferably a polypeptide whose sequence is represented by a fragment, especially a specific fragment, these six amino acid sequences corresponding to the polypeptides which can be encoded according to SEQ ID No. 1.
  • the disclosure also relates to the polypeptides, characterized in that they comprise a polypeptide selected from: a) a specific fragment of at least 5 amino acids of a polypeptide of an amino acid sequence according to the disclosure; b) a polypeptide homologous to a polypeptide such as defined in a); c) a specific biologically active fragment of a polypeptide such as defined in a) or b); and d) a polypeptide modified by a polypeptide such as defined in a), b) or c).
  • a polypeptide selected from: a) a specific fragment of at least 5 amino acids of a polypeptide of an amino acid sequence according to the disclosure; b) a polypeptide homologous to a polypeptide such as defined in a); c) a specific biologically active fragment of a polypeptide such as defined in a) or b); and d) a polypeptide modified by a polypeptide such as defined in a), b) or
  • polypeptides are also preferred, these polypeptides being especially capable of specifically recognizing the antibodies produced during infection by SARS- CoV-2. These polypeptides thus have epitopes specific for the SARS-CoV-2 and can thus be used in particular in the diagnostic field or as immunogenic agent to confer protection in an animal against infection by SARS-CoV-2.
  • polypeptide, peptide and protein are interchangeable.
  • polypeptides in natural form, that is to say that they are not taken in their natural environment but that they can be isolated or obtained by purification from natural sources, or else obtained by genetic recombination, or alternatively by chemical synthesis and that they can thus contain unnatural amino acids, as will be described below.
  • Polypeptide fragment according to the disclosure is understood as designating a polypeptide containing at least 5 consecutive amino acids, preferably 10 consecutive amino acids or 15 consecutive amino acids.
  • specific polypeptide fragment is understood as designating the consecutive polypeptide fragment encoded by a specific fragment nucleotide sequence according to the disclosure.
  • homologous polypeptide will be understood as designating the polypeptides having, with respect to the natural polypeptide, certain modifications such as, in particular, a deletion, addition or substitution of at least one amino acid, a truncation, a prolongation, a chimeric fusion, and/or a mutation.
  • those are preferred whose amino acid sequence has at least 80, 85, 88, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.6, 99.7, 99.8, 99.9, or even 100%, homology with the sequences of amino acids of polypeptides according to the disclosure.
  • homologous polypeptide will be understood as designating the homologous polypeptides such as defined above and having a specific fragment of polypeptide according to the disclosure.
  • amino acids are replaced by “equivalent” amino acids.
  • the expression “equivalent” amino acid is directed here at designating any amino acid capable of being substituted by one of the amino acids of the base structure without, however, essentially modifying the biological activities of the corresponding peptides and such that they will be defined by the following.
  • These equivalent amino acids can be determined either by depending on their structural homology with the amino acids which they substitute, or on results of comparative tests of biological activity between the different polypeptides, which are capable of being carried out.
  • the specific homologous polypeptides likewise correspond to polypeptides encoded by the specific homologous nucleotide sequences such as defined above and thus comprise in the present definition the polypeptides which are mutated or correspond to variants which can exist in SARS-CoV-2, and which especially correspond to truncations, substitutions, deletions and/or additions of at least one amino acid residue.
  • Specific biologically active fragment of a polypeptide according to the disclosure will be understood in particular as designating a specific polypeptide fragment, such as defined above, having at least one of the characteristics of polypeptides according to the disclosure, especially in that it is: capable of inducing an immunogenic reaction directed against a SARS-CoV-2; and/or capable of being recognized by a specific antibody of a polypeptide according to the disclosure; and/or capable of linking to a polypeptide or to a nucleotide sequence of SARS-CoV-2; and/or capable of exerting a physiological activity, even partial, such as, for example, a dissemination or structural (capsid) activity; and/or capable of modulating, of inducing or of inhibiting the expression of SARS-CoV-2 gene or one of its variants, and/or capable of modulating the replication cycle of SARS-CoV-2 in the cell and/or the host organism.
  • polypeptide fragments according to the disclosure can correspond to isolated or purified fragments naturally present in a SARS-CoV-2 or correspond to fragments which can be obtained by cleavage of said polypeptide by a proteolytic enzyme, such as trypsin or chymotrypsin or collagenase, or by a chemical reagent, such as cyanogen bromide (CNBr) or alternatively by placing said polypeptide in a very acidic environment, for example at pH 2.5.
  • a proteolytic enzyme such as trypsin or chymotrypsin or collagenase
  • CNBr cyanogen bromide
  • Such polypeptide fragments can likewise just as easily be prepared by chemical synthesis, from hosts transformed by an expression vector according to the disclosure containing a nucleic acid allowing the expression of said fragments, placed under the control of appropriate regulation and/or expression elements.
  • Modified polypeptide of a polypeptide according to the disclosure is understood as designating a polypeptide obtained by genetic recombination or by chemical synthesis as will be described below, having at least one modification with respect to the normal sequence. These modifications will especially be able to bear on amino acids at the origin of a specificity, of pathogenicity and/or of virulence, or at the origin of the structural conformation, and of the capacity of membrane insertion of the polypeptide according to the disclosure. It will thus be possible to create polypeptides of equivalent, increased or decreased activity, and of equivalent, narrower, or wider specificity. Among the modified polypeptides, it is necessary to mention the polypeptides in which up to 5 amino acids can be modified, truncated at the N- or C- terminal end, or even deleted or added.
  • the modifications of the polypeptide will especially have as objective: to render it capable of modulating, of inhibiting or of inducing the expression of at least one SARS-CoV-2 gene and/or capable of modulating the replication cycle of SARS-CoV-2 in the cell and/or the host organism, of allowing its incorporation into vaccine compositions, and/or of modifying its bioavailability as a compound for therapeutic use.
  • Chemical synthesis likewise has the advantage of being able to use unnatural amino acids, or nonpeptide bonds.
  • unnatural amino acids for example in D form, or else amino acid analogs, especially sulfur-containing forms, for example.
  • nucleotide sequences coding for a polypeptide according to the disclosure are likewise part of the disclosure.
  • the disclosure likewise relates to nucleotide sequences utilizable as a primer or probe, characterized in that said sequences are selected from the nucleotide sequences according to the disclosure.
  • the disclosure additionally relates to the use of a nucleotide sequence according to the disclosure as a primer or probe for the detection and/or the amplification of nucleic acid sequences.
  • the nucleotide sequences according to the disclosure can thus be used to amplify nucleotide sequences, especially by the PCR technique (polymerase chain reaction) (Erlich, 1989; Innis et al., 1990; Rolfs et al., 1991 ; and White et al., 1997).
  • PCR technique polymerase chain reaction
  • oligodeoxyribonucleotide or oligoribonucleotide primers advantageously have a length of at least 8 nucleotides, preferably of at least 12 nucleotides, and even more preferentially at least 20 nucleotides.
  • Other amplification techniques of the target nucleic acid can be advantageously employed as alternatives to PCR.
  • the nucleotide sequences of the disclosure can likewise be employed in other procedures of amplification of a target nucleic acid, such as: the TAS technique (Transcription-based Amplification System), described by Kwoh et al. in 1989; the 3SR technique (Self- Sustained Sequence Replication), described by Guatelli et al. in 1990; the NASBA technique (Nucleic Acid Sequence Based Amplification), described by Kievitis et al. in 1991 ; the SDA technique (Strand Displacement Amplification) (Walker et al., 1992); the TMA technique (Transcription Mediated Amplification).
  • the polynucleotides of the disclosure can also be employed in techniques of amplification or of modification of the nucleic acid serving as a probe, such as: the LCR technique (Ligase Chain Reaction), described by Landegren et al. in 1988 and improved by Barany et al. in 1991 , which employs a thermostable ligase; the RCR technique (Repair Chain Reaction), described by Segev in 1992; the CPR technique (Cycling Probe Reaction), described by Duck et al. in 1990; the amplification technique with Q- beta replicase, described by Miele et al. in 1983 and especially improved by Chu et al. in 1986, Lizardi et al. in 1988, then by Burg et al. as well as by Stone et al. in 1996.
  • LCR technique Liigase Chain Reaction
  • RCR technique Repair Chain Reaction
  • CPR technique Cycling Probe Reaction
  • the amplification technique with Q- beta replicase described by
  • the target polynucleotide to be detected is possibly an RNA, for example an mRNA
  • an enzyme of reverse transcriptase type in order to obtain a cDNA from the RNA contained in the biological sample.
  • the cDNA obtained will thus serve as a target for the primer(s) or the probe(s) employed in the amplification or detection procedure according to the disclosure.
  • the detection probe will be chosen in such a manner that it hybridizes with the target sequence or the amplicon generated from the target sequence.
  • a probe will advantageously have a sequence of at least 12 nucleotides, in particular of at least 20 nucleotides, and preferably of at least 100 nucleotides.
  • the disclosure also comprises the nucleotide sequences utilizable as a probe or primer according to the disclosure, characterized in that they are labeled with a radioactive compound or with a nonradioactive compound.
  • the unlabeled nucleotide sequences can be used directly as probes or primers, although the sequences are generally labeled with a radioactive element ( 32 P, 35 S, 3 H, 125 l) or with a nonradioactive molecule (biotin, acetylaminofluorene, digoxigenin, 5-bromodeoxyuridine, fluorescein) to obtain probes which are utilizable for numerous applications.
  • a radioactive element 32 P, 35 S, 3 H, 125 l
  • a nonradioactive molecule biotin, acetylaminofluorene, digoxigenin, 5-bromodeoxyuridine, fluorescein
  • nonradioactive labeling of nucleotide sequences are described, for example, in French Patent No. 78.10975 or by Urdea et al. or by Sanchez-Pescador et al. in 1988. In the latter case, it will also be possible to use one of the labeling methods described in patents FR-2
  • the hybridization technique can be carried out in various manners (Matthews et al., 1988).
  • the most general method consists in immobilizing the nucleic acid extract of cells on a support (such as nitrocellulose, nylon, polystyrene) and in incubating, under well-defined conditions, the immobilized target nucleic acid with the probe. After hybridization, the excess of probe is eliminated and the hybrid molecules formed are detected by the appropriate method (measurement of the radioactivity, of the fluorescence or of the enzymatic activity linked to the probe).
  • the disclosure likewise comprises the nucleotide sequences according to the disclosure, characterized in that they are immobilized on a support, covalently or noncovalently.
  • nucleotide sequences According to another advantageous mode of employing nucleotide sequences according to the disclosure, the latter can be used immobilized on a support and can thus serve to capture, by specific hybridization, the target nucleic acid obtained from the biological sample to be tested. If necessary, the solid support is separated from the sample and the hybridization complex formed between said capture probe and the target nucleic acid is then detected with the aid of a second probe, a so-called detection probe, labeled with an easily detectable element.
  • Another subject of the present disclosure is a vector for the cloning and/or expression of a sequence, characterized in that it contains a nucleotide sequence according to the disclosure.
  • the vectors according to the disclosure characterized in that they contain the elements allowing the expression in a determined host cell, are likewise part of the disclosure.
  • the vector must then contain a promoter, signals of initiation and termination of translation, as well as appropriate regions of regulation of transcription. It must be able to be maintained stably in the host cell and can optionally have particular signals specifying the secretion of the translated protein. These different elements are chosen as a function of the host cell used.
  • the nucleotide sequences according to the disclosure can be inserted into autonomous replication vectors within the chosen host, or integrated vectors of the chosen host.
  • vectors will be prepared according to the methods currently used by the person skilled in the art, and it will be possible to introduce the clones resulting therefrom into an appropriate host by standard methods, such as, for example, lipofection, electroporation and thermal shock.
  • the vectors according to the disclosure are, for example, vectors of plasmid or viral origin.
  • the preparation procedures employing a vector, and/or a cell transformed by said vector and/or a transgenic animal comprising one of said transformed cells, containing a nucleotide sequence according to the disclosure coding for a polypeptide of SARS-CoV-2, are preferred.
  • the recombinant polypeptides obtained as indicated above can just as well be present in glycosylated form as in nonglycosylated form and can or cannot have the natural tertiary structure.
  • the disclosure relates to a procedure for preparation of a polypeptide of the disclosure comprising the following steps: a) culture of transformed cells under conditions allowing the expression of a recombinant polypeptide by a nucleotide sequence according to the disclosure; b) if need be, recovery of said recombinant polypeptide.
  • the disclosure also relates to a polypeptide which is capable of being obtained by a procedure of the disclosure such as described previously.
  • the disclosure also comprises a procedure for preparation of a synthetic polypeptide, characterized in that it uses a sequence of amino acids of polypeptides according to the disclosure.
  • the disclosure likewise relates to a synthetic polypeptide obtained by a procedure according to the disclosure.
  • polypeptides according to the disclosure can likewise be prepared by techniques which are conventional in the field of the synthesis of peptides. This synthesis can be carried out in homogeneous solution or in solid phase. For example, reference can be made to the technique of synthesis in homogeneous solution described by Houben-Weyl in 1974.
  • This method of synthesis consists in successively condensing, two by two, the successive amino acids in the order required, or in condensing amino acids and fragments formed previously and already containing several amino acids in the appropriate order, or alternatively several fragments previously prepared in this way, it being understood that it will be necessary to protect beforehand all the reactive functions carried by these amino acids or fragments, with the exception of amine functions of one and carboxyls of the other or vice-versa, which must normally be involved in the formation of peptide bonds, especially after activation of the carboxyl function, according to the methods well known in the synthesis of peptides.
  • recourse will be made to the technique described by Merrifield.
  • the disclosure additionally relates to hybrid polypeptides having at least one polypeptide according to the disclosure, and a sequence of a polypeptide capable of inducing an immune response in man or animals.
  • the antigenic determinant is such that it is capable of inducing a humoral and/or cellular response. It will be possible for such a determinant to comprise a polypeptide according to the disclosure in glycosylated form used with a view to obtaining immunogenic compositions capable of inducing the synthesis of antibodies directed against multiple epitopes. Said polypeptides or their glycosylated fragments are likewise part of the disclosure.
  • hybrid molecules can be formed, in part, of a polypeptide carrier molecule or of fragments thereof according to the disclosure, associated with a possibly immunogenic part, in particular an epitope of the diphtheria toxin, the tetanus toxin, a surface antigen of the hepatitis B virus (patent FR 79 21811 ), the VP1 antigen of the poliomyelitis virus or any other viral or bacterial toxin or antigen.
  • the procedures for synthesis of hybrid molecules encompass the methods used in genetic engineering for constructing hybrid nucleotide sequences coding for the polypeptide sequences sought. It will be possible, for example, to refer advantageously to the technique for obtainment of genes coding for fusion proteins described by Minton in 1984.
  • hybrid nucleotide sequences coding for a hybrid polypeptide as well as the hybrid polypeptides according to the disclosure characterized in that they are recombinant polypeptides obtained by the expression of said hybrid nucleotide sequences are likewise part of the disclosure.
  • the disclosure likewise comprises the vectors characterized in that they contain one of said hybrid nucleotide sequences.
  • the host cells transformed by said vectors, the transgenic animals comprising one of said transformed cells as well as the procedures for preparation of recombinant polypeptides using said vectors, said transformed cells and/or said transgenic animals are, of course, likewise part of the disclosure.
  • the disclosure relates to the polypeptides according to the disclosure, labeled with the aid of an adequate label such as of the enzymatic, fluorescent or radioactive type.
  • Such methods comprise, for example, the following steps: 1 ) deposition of determined quantities of a polypeptide composition according to the disclosure in the wells of a microtiter plate; 2) introduction into said wells of increasing dilutions of serum, or of a biological sample other than that defined previously, having to be analyzed,; 3) incubation of the microplate; and 4) introduction into the wells of the microtiter plate of labeled antibodies directed against mammal immunoglobulins, the labeling of these antibodies having been carried out with the aid of an enzyme selected from those which are capable of hydrolyzing a substrate by modifying the absorption of the radiation of the latter, at least at a determined wavelength, for example at 550 nm, detection, by comparison with a control test, of the quantity of hydrolyzed substrate.
  • the polypeptides according to the disclosure allow monoclonal or polyclonal antibodies to be prepared which are characterized in that they specifically recognize the polypeptides according to the disclosure. It will advantageously be possible to prepare the monoclonal antibodies from hybridomas according to the technique described by Kohler and Milstein in 1975. It will be possible to prepare the polyclonal antibodies, for example, by immunization of an animal, in particular a mouse, with a polypeptide or a DNA, according to the disclosure, associated with an adjuvant of the immune response, and then purification of the specific antibodies contained in the serum of the immunized animals on an affinity column on which the polypeptide which has served as an antigen has previously been immobilized.
  • the polyclonal antibodies according to the disclosure can also be prepared by purification, on an affinity column on which a polypeptide according to the disclosure has previously been immobilized, of the antibodies contained in the serum of animals infected by a SARS-CoV-2.
  • the disclosure likewise relates to a pharmaceutical composition comprising a compound selected from the following compounds: a) a nucleotide sequence according to the disclosure; b) a polypeptide according to the disclosure; c) a vector, a viral particle or a cell transformed according to the disclosure; d) an antibody according to the disclosure; and e) a compound capable of being selected by a selection method according to the disclosure; possibly in combination with a pharmaceutically acceptable carrier and, if need be, with one or more adjuvants of the appropriate immunity.
  • the disclosure further relates to a nanomaterial that is capable of operating as a pharmaceutically acceptable carrier and/or as a polypeptide/nucleic acid binding, stabilization, and delivery enhancing agent.
  • the disclosure also relates to an immunogenic and/or vaccine composition, characterized in that it comprises a compound selected from the following compounds: a) a nucleotide sequence according to the disclosure; b) a polypeptide according to the disclosure; c) a vector or a viral particle according to the disclosure; and d) a cell according to the disclosure.
  • the vaccine composition according to the disclosure is characterized in that it comprises a mixture of at least two of said compounds a), b), c) and d) above and in that one of the two said compounds is related to the SARS-CoV-2.
  • the vaccine composition is characterized in that it comprises at least one compound a), b), c), or d) above which is related to SARS-CoV-2.
  • the vaccine composition is characterized in that it comprises at least one compound a), b), c), or d) above which is related to SARS-CoV-2 S protein.
  • a compound related to the SARS-CoV-2 is understood here as respectively designating a compound obtained from the genomic sequence of the SARS-CoV-2 and/or S protein of SARS-CoV-2.
  • the disclosure is additionally aimed at an immunogenic and/or vaccine composition, characterized in that it comprises at least one of the following compounds: 1 ) a nucleotide sequence encoding SEQ ID No. 1 or one of its fragments or homologues; 2) a polypeptide of sequence SEQ ID No. 1, SEQ ID No. 9, or SEQ ID No. 10 or one of their fragments, or a modification thereof; 3) a vector or a viral particle comprising a nucleotide sequence encoding SEQ ID No. 1 or one of its fragments or homologues; 4) a transformed cell capable of expressing a polypeptide of sequence SEQ ID No. 1 , or one of its fragments, or a modification thereof; or 5) a mixture of at least two of said compounds.
  • the disclosure also comprises an immunogenic and/or vaccine composition according to the disclosure, characterized in that it comprises said mixture of at least two of said compounds as a combination product for simultaneous, separate or protracted use for the prevention or the treatment of infection by a SARS-CoV-2.
  • the disclosure is likewise directed at a pharmaceutical composition according to the disclosure, for the prevention or the treatment of an infection by a SARS-CoV-2.
  • prevention includes the complete prevention of infection by a SARS-CoV-2, but also encompasses a reduction in the severity of or incidence of clinical signs associated with or caused by SARS-CoV-2 infection. Such prevention is also referred to herein as a protective effect.
  • the disclosure likewise concerns the use of a composition according to the disclosure, for the preparation of a medicament intended for the prevention or the treatment of infection by a SARS-CoV-2.
  • the disclosure relates to a vector, a viral particle or a cell according to the disclosure, for the treatment and/or the prevention of a disease by gene therapy.
  • the disclosure comprises the use of a vector, of a viral particle or of a cell according to the disclosure for the preparation of a medicament intended for the treatment and/or the prevention of a disease by gene therapy.
  • polypeptides of the disclosure entering into the immunogenic or vaccine compositions according to the disclosure can be selected by techniques known to the person skilled in the art such as, for example, depending on the capacity of said polypeptides to stimulate the T cells, which is translated, for example, by their proliferation or the secretion of interleukins, and which leads to the production of antibodies directed against said polypeptides.
  • compositions according to the disclosure will contain an effective quantity of the compounds of the disclosure, that is to say in sufficient quantity of said compound(s) allowing the desired effect to be obtained, such as, for example, the modulation of the cellular replication of SARS-CoV-2.
  • the desired effect such as, for example, the modulation of the cellular replication of SARS-CoV-2.
  • the person skilled in the art will know how to determine this quantity, as a function, for example, of the age and of the weight of the individual to be treated, of the state of advancement of the pathology, of the possible secondary effects and by means of a test of evaluation of the effects obtained on a population range, these tests being known in these fields of application.
  • said vaccine combinations will preferably be combined with a pharmaceutically or veterinary acceptable carrier and, if need be, with one or more adjuvants of the appropriate immunity.
  • This type of vaccination is carried out with a particular plasmid originating from a plasmid of E. coli which does not replicate in vivo and which codes uniquely for the vaccinating protein. Animals have been immunized by simply injecting the naked plasmid DNA into the muscle. This technique leads to the expression of the vaccine protein in situ and to an immune response of cellular type (CTL) and of humoral type (antibody). This double induction of the immune response is one of the principal advantages of the vaccination technique with naked DNA.
  • CTL cellular type
  • antibody humoral type
  • the constitutive nucleotide sequence of the vaccine composition according to the disclosure can be injected into the host after having been coupled to compounds which favor the penetration of this polynucleotide into the interior of the cell or its transport to the cell nucleus.
  • the resultant conjugates can be encapsulated in polymeric microparticles, as described in the international application No. WO 94/27238 (Medisorb Technologies International).
  • the nucleotide sequence preferably a DNA
  • the nucleotide sequence is complexed with DEAE- dextran (Pagano et al., 1967) or with nuclear proteins (Kaneda et al., 1989), with lipids (Feigner et al., 1987) or encapsulated in liposomes (Fraley et al., 1980) or else introduced in the form of a gel facilitating its transfection into the cells (Midoux et al., 1993, Pastore et al., 1994).
  • the polynucleotide or the vector according to the disclosure can also be in suspension in a buffer solution or be combined with liposomes.
  • such a vaccine will be prepared according to the technique described by Tacson et al. or Huygen et al. in 1996 or alternatively according to the technique described by Davis et al. in the international application No. WO 95/11307.
  • Such a vaccine can likewise be prepared in the form of a composition containing a vector according to the disclosure, placed under the control of regulation elements allowing its expression in man or animal. It will be possible, for example, to use, by way of in vivo expression vector of the polypeptide antigen of interest, the plasmid pcDNA3 or the plasmid pcDNA1/neo, both marketed by Invitrogen (R&D Systems, Abingdon, United Kingdom). It is also possible to use the plasmid V1 Jns.tPA, described by Shiver et al. in 1995. Such a vaccine will advantageously comprise, apart from the recombinant vector, a saline solution, for example a sodium chloride solution.
  • these can comprise adjuvants of the appropriate immunity which are known to the person skilled in the art, such as, for example, those described above.
  • these compounds can be administered by the systemic route, in particular by the intravenous route, by the intramuscular, intradermal or subcutaneous route, or by the oral route.
  • the vaccine composition comprising polypeptides according to the disclosure will be administered by the intramuscular route, through the food or by nebulization several times, staggered over time.
  • Their administration modes, dosages and optimum pharmaceutical forms can be determined according to the criteria generally taken into account in the establishment of a treatment adapted to an animal such as, for example, the age or the weight, the seriousness of its general condition, the tolerance to the treatment and the secondary effects noted.
  • the vaccine of the present disclosure is administered in an amount that is protective or provides a protective effect against SARS-CoV-2 infection.
  • the polypeptide will be administered one time or several times, spread out over time, directly or by means of a transformed cell capable of expressing the polypeptide, in an amount of about 0.1 to 10 pg per kilogram weight of the animal, preferably about 0.2 to about 5 pg/kg, more preferably about 0.5 to about 2 pg/kg for a dose.
  • the present disclosure likewise relates to the use of nucleotide sequences of SARS-CoV-2 according to the disclosure for the construction of autoreplicative retroviral vectors and the therapeutic applications of these, especially in the field of gene therapy in vivo.
  • the principle of gene therapy is to deliver a functional gene, called a gene of interest, of which the RNA or the corresponding protein will produce the desired biochemical effect in the targeted cells or tissues.
  • a gene of interest of which the RNA or the corresponding protein will produce the desired biochemical effect in the targeted cells or tissues.
  • the insertion of genes allows the prolonged expression of complex and unstable molecules such as RNAs or proteins which can be extremely difficult or even impossible to obtain or to administer directly.
  • the controlled insertion of the desired gene into the interior of targeted specific cells allows the expression product to be regulated in defined tissues. For this, it is necessary to be able to insert the desired therapeutic gene into the interior of chosen cells and thus to have available a method of insertion capable of specifically targeting the cells or the tissues chosen.
  • Some preferred genes of interest for the present disclosure are those that encode the S protein.
  • cell-specific viral vectors having a tissue-specific tropism, and whose gene of interest can be translated adequately by the target cells, is realizable, for example, by fusing a specific ligand of the target host cells to the N- terminal part of a surface protein of the envelope of SARS-CoV-2.
  • retroviral particles having the CD4 molecule on the surface of the envelope so as to target the human cells infected by the HIV virus, viral particles having a peptide hormone fused to an envelope protein to specifically infect the cells expressing the corresponding receptor or else alternatively viral particles having a fused polypeptide capable of immobilizing on the receptor of the epidermal growth factor (EGF).
  • EGF epidermal growth factor
  • single-chain fragments of antibodies directed against surface antigens of the target cells are inserted by fusion with the N-terminal part of the envelope protein.
  • a gene of interest in use in the disclosure can be obtained from a eukaryotic or prokaryotic organism or from a virus by any conventional technique. It is, preferably, capable of producing an expression product having a therapeutic effect and it can be a product homologous to the cell host or, alternatively, heterologous.
  • a gene of interest can code for an (1 ) intracellular or (2) membrane product present on the surface of the host cell or (3) secreted outside the host cell. It can therefore comprise appropriate additional elements such as, for example, a sequence coding for a secretion signal. These signals are known to the person skilled in the art.
  • a gene of interest can code for a protein corresponding to all or part of a native protein as found in nature. It can likewise be a chimeric protein, for example arising from the fusion of polypeptides of various origins or from a mutant having improved and/or modified biological properties. Such a mutant can be obtained, by conventional biological techniques, by substitution, deletion and/or addition of one or more amino acid residues.
  • the disclosure thus relates to the vectors characterized in that they comprise a nucleotide sequence of SARS-CoV-2 according to the disclosure, and in that they additionally comprise a gene of interest (“GOI”).
  • GOI gene of interest
  • the present disclosure likewise relates to viral particles generated from said vector according to the disclosure. It additionally relates to methods for the preparation of viral particles according to the disclosure, characterized in that they employ a vector according to the disclosure, including viral pseudoparticles (VLP, virus-like particles).
  • VLP viral pseudoparticles
  • the disclosure likewise relates to animal cells transfected by a vector according to the disclosure.
  • animal cells especially mammalian, infected by a viral particle according to the disclosure.
  • Additional genetically engineered vaccines which are desirable in the present disclosure, are produced by techniques known in the art. Such techniques involve, but are not limited to, further manipulation of recombinant DNA, modification of or substitutions to the amino acid sequences of the recombinant proteins and the like. Genetically engineered vaccines based on recombinant DNA technology are made, for instance, by identifying alternative portions of the viral gene encoding proteins responsible for inducing a stronger immune or protective response in animals (e.g., proteins derived from the S). Such identified genes or immuno-dominant fragments can be cloned into standard protein expression vectors, such as the baculovirus vector, and used to infect appropriate host cells. The host cells are cultured, thus expressing the desired vaccine proteins, which can be purified to the desired extent and formulated into a suitable vaccine product.
  • standard protein expression vectors such as the baculovirus vector
  • the clones retain any undesirable natural abilities of causing disease, it is also possible to pinpoint the nucleotide sequences in the viral genome responsible for any residual virulence, and genetically engineer the virus avirulent through, for example, site-directed mutagenesis.
  • Site-directed mutagenesis is able to add, delete or change one or more nucleotides.
  • An oligonucleotide is synthesized containing the desired mutation and annealed to a portion of single stranded viral DNA. The hybrid molecule, which results from that procedure, is employed to transform bacteria.
  • doublestranded DNA which is isolated containing the appropriate mutation, is used to produce full-length DNA by ligation to a restriction fragment of the latter that is subsequently transfected into a suitable cell culture.
  • Ligation of the genome into the suitable vector for transfer may be accomplished through any standard technique known to those of ordinary skill in the art.
  • Transfection of the vector into host cells for the production of viral progeny may be done using any of the conventional methods such as calciumphosphate or DEAE-dextran mediated transfection, electroporation, protoplast fusion and other well-known techniques.
  • the cloned virus then exhibits the desired mutation.
  • two oligonucleotides can be synthesized which contain the appropriate mutation. These may be annealed to form double-stranded DNA that can be inserted in the viral DNA to produce full-length DNA.
  • Genetically engineered proteins useful in vaccines, for instance, may be expressed in insect cells, yeast cells or mammalian cells.
  • the genetically engineered proteins which may be purified or isolated by conventional methods, can be directly inoculated into animals to confer protection against viral infection caused by SARS- CoV-2.
  • An insect cell line (like HI-FIVE) can be transformed with a transfer vector containing nucleic acid molecules obtained from the virus or copied from the viral genome which encodes one or more of the immuno-dominant proteins of the virus.
  • the transfer vector includes, for example, linearized baculovirus DNA and a plasmid containing the desired polynucleotides.
  • the host cell line may be co-transfected with the linearized baculovirus DNA and a plasmid in order to make a recombinant baculovirus.
  • An immunologically effective amount of one of the vaccines or immunogenic compositions of the present disclosure is administered to an animal in need of protection against viral infection, pneumonia, fever, cough, and loss of taste and/or smell.
  • the immunologically effective amount or the immunogenic amount that inoculates the animal can be easily determined or readily titrated by routine testing.
  • An effective amount is one in which a sufficient immunological response to the vaccine is attained to protect the animal exposed to SARS-CoV-2.
  • the animal receiving a dose of a vaccine or immunogenic composition according to this disclosure is protected to an extent in which one to all of the adverse physiological symptoms or effects of the viral disease are significantly reduced, ameliorated or totally prevented.
  • the vaccine can be administered in a single dose or in repeated doses with single doses being preferred.
  • Single dose vaccines provide protection after a single dose without the need for any booster or subsequent dosages. Protection can include the complete prevention of clinical signs of infection, or a lessening of the severity, duration, or likelihood of the manifestation of one or more clinical signs of infection.
  • the vaccine is administered to a human or animal not yet exposed to the SARS-CoV-2 virus.
  • the present vaccine may be prepared in the form of an aqueous solution, syrup, an elixir, a tincture and the like.
  • Such formulations are known in the art and are typically prepared by dissolution of the antigen and other typical additives in the appropriate carrier or solvent systems.
  • Suitable carriers or solvents include, but are not limited to, water, saline, ethanol, ethylene glycol, glycerol, etc.
  • Typical additives are, for example, certified dyes, flavors, sweeteners and antimicrobial preservatives such as thimerosal (sodium ethylmercurithiosalicylate). Such solutions may be stabilized, for example, by addition of partially hydrolyzed gelatin, sorbitol or cell culture medium, and may be buffered by conventional methods using reagents known in the art, such as sodium hydrogen phosphate, sodium dihydrogen phosphate, potassium hydrogen phosphate, potassium dihydrogen phosphate, a mixture thereof, and the like. [0173] Liquid formulations also may include suspensions and emulsions that contain suspending or emulsifying agents in combination with other standard co-formulants.
  • Fig. 12 illustrates the temperature stability for ZnO-Protamine-TYRNA that had been stored as a suspension in PBS in a closed eppendorf tube for 2 weeks at the specified temperatures at a high (H) concentration of protamine coating.
  • Parenteral formulations designed for injection into body fluid systems, require proper isotonicity and pH buffering to the corresponding levels of body fluids. Isotonicity can be appropriately adjusted with sodium chloride and other salts as needed. Suitable solvents, such as ethanol or propylene glycol, can be used to increase the solubility of the ingredients in the formulation and the stability of the liquid preparation. Further additives that can be employed in the present vaccine include, but are not limited to, dextrose, conventional antioxidants and conventional chelating agents such as ethylenediamine tetraacetic acid (EDTA). Parenteral dosage forms must also be sterilized prior to use.
  • EDTA ethylenediamine tetraacetic acid
  • Another aspect of the present disclosure is the preparation of the combination vaccine(s) or immunogenic compositions.
  • Such combinations can be between the different vaccine components described herein.
  • a vaccine of the present disclosure can include both protein portions and DNA portions of SARS-CoV-2, as described herein, which are administered concurrently or separately. Additionally, the combinations can be between the SARS-CoV-2 vaccine components described herein and antigens of other disease-causing organisms, such as those described above.
  • the vaccine or immunogenic composition is first dehydrated. If the composition is first lyophilized or dehydrated by other methods, then, prior to vaccination, said composition is rehydrated in aqueous (e.g. saline, PBS (phosphate buffered saline)) or non-aqueous solutions (e.g. oil emulsion (mineral oil, or vegetable/metabolizable oil based/single or double emulsion based), aluminum-based, carbomer based adjuvant).
  • aqueous e.g. saline, PBS (phosphate buffered saline)
  • non-aqueous solutions e.g. oil emulsion (mineral oil, or vegetable/metabolizable oil based/single or double emulsion based), aluminum-based, carbomer based adjuvant.
  • compositions according to the disclosure may be applied orally, though compositions of similar type are most frequently delivered parenterally, particularly intravenously, intramuscularly, transdermally, intradermally, intratracheally, intravaginally or intranasally.
  • parenterally particularly intravenously, intramuscularly, transdermally, intradermally, intratracheally, intravaginally or intranasally.
  • it can prove advantageous to apply the pharmaceutical compositions as described above via an intravenous injection or by direct injection into target tissues.
  • the intravenous, intravascular, intramuscular, intranasal, intraarterial, intraperitoneal, oral, or intrathecal routes are preferred.
  • a more local application can be effected subcutaneously, intradermally, intracutaneously, intracardially, intralobally, intramedullarly, intrapulmonarily or directly in or near the tissue to be treated (connective-, bone-, muscle-, nerve-, epithelial tissue).
  • the compositions according to the disclosure may be administered once or several times, also intermittently, for instance on a daily basis for several days, weeks or months, and in different dosages.
  • a method for protecting humans from SARS-CoV-2 infection transmitted by animals comprises administering at least one dose of a composition comprising the S protein from SARS-CoV-2 to a susceptible animal.
  • a composition comprising the S protein from SARS-CoV-2 to a susceptible animal.
  • Preferred susceptible animals are provided above.
  • Such administration prevents or reduces the transmission from an infected animal to humans.
  • the transmission to humans will be reduced by at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 95, or even 100%.
  • Such a method removes a potential host or reservoir for SARS-CoV-2 and reduces the overall circulation of SARS- CoV-2.
  • Preferred compositions are selected from those disclosed herein.
  • a method of inducing an immunological response against SARS-CoV-2 comprises the steps of administering a composition of the present disclosure, to an animal in need of prevention or treatment of SARS-CoV-2 associated disease.
  • the S protein has at least 90% sequence homology with SEQ ID NO. 1.
  • the pharmaceutical-acceptable carrier comprises a stabilizing agent and/or a preservative and/or an antimicrobial agent.
  • the protein is present in the final composition in an amount from 0.2 to about 400 pg/ml.
  • the composition further comprises an immune stimulant.
  • the composition further comprises at least one immunological active component against another diseasecausing organism.
  • the animal in need thereof is selected from the group consisting of humans, dogs, cats, ferrets, bats, cattle, camels, hamsters, horses, chimps, gorillas, anteaters, dolphins, alligators, and sheep.
  • the composition is administered a first time and a second time. In some forms, the second time is at least 10 days after the first time.
  • Immunogenic compositions of the present disclosure exhibit improved stability relative to currently available mRNA vaccines, advantageously permitting distribution without the need for an unbroken cold-chain, permitting higher temperature storage of vaccine compositions once distributed, and potentially extending the useful shelf life of multi-dosage vaccine and immunogenic composition forms both before and after they are opened.
  • Fig. 4 the in vivo distribution
  • Fig. 6 panel b, c the zinc-based compositions studied so far and biocompatibility
  • Figures 1 , 12 and 13 relate to the temperature-stability conferred to RNA by binding to ZnO or ZnO-protamine.
  • protamine in addition to being a condensing agent for nucleic acids is considered a cell penetrating peptide (CPP) itself. Accordingly, when it complexes to RNA it can form RNA-protein nanoparticles that have some ability to carry nucleic acids into cells even in the absence of a core metal nanoparticle. Thus in addition to a stabilizing element, protamine increases binding to the nanoparticle (Fig. 13) but also importantly likely influences intracellular delivery of the nucleic acid to some extent.
  • CPP cell penetrating peptide
  • the LL37 peptide while less effective at loading RNA onto zinc oxide (Fig. 13), there is some evidence to suggest this peptide’s antiviral activity and/or more recently role in COVID-19 immunology.
  • the peptide component can contribute to the activity of the formulation, more so than just stabilizing the nucleic acid, increasing the RNA particle association, or enhancing delivery, it can actually contribute to the immunology of the vaccine and/or its antiviral activity.
  • ZnO NP zinc oxide nanoparticles
  • the data illustrates that zinc oxide nanoparticles (ZnO NP) interacts with RNA, protects RNA from hydrolysis and nuclease-RNase mediated degradation, and also contributes to potentiating the immune response of RNA or peptide.
  • the disclosure provides a composition comprising a nucleic acid encoding a SARS-CoV-2 S protein and a pharmaceutically acceptable carrier comprising a zinc oxide nanoparticle.
  • the pharmaceutically acceptable carrier further comprises a solvent, a dispersion media, a coating, a stabilizing agent, a diluent, a preservative, an antimicrobial agent, an antifungal agent, an isotonic agent, an adsorption delaying agent, and any combination thereof.
  • the protein has at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or even 100% sequence homology with SEQ ID NO. 1.
  • the pharmaceutical-acceptable carrier comprises a stabilizing agent and/or a preservative and/or an antimicrobial agent.
  • the nucleic acid is present at a concentration greater than 100 micrograms per milligram weight of nanoparticle.
  • the zinc nanoparticle comprises or further comprises a cell penetrating peptide.
  • the zinc nanoparticle comprises or further comprises protamine.
  • the nucleic acid further comprises a sequence encoding a CD40L.
  • the nucleic acid encodes a tetrameric CD40L trimer.
  • the SARS-CoV-2 S protein and/or the CD40L are encoded by a polycistronic mRNA.
  • the nucleic acid encodes a SARS- CoV-2 S protein / CD40L fusion protein.
  • the vector includes an inserted sequence having at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or even 100%% sequence homology with SEQ ID NO. 9 or 10.
  • the nucleic acid is present at a concentration of greater than 100 pg/mg of nanoparticle.
  • the nucleic acid is RNA or mRNA.
  • the nucleic acid is a component in a lipid nanoparticle immunogenic composition.
  • the lipid nanoparticle immunogenic composition is an mRNA immunogenic composition.
  • a method of temperature stabilizing a lipid nanoparticle RNA or mRNA immunogenic composition comprises the step of binding the RNA or mRNA to a zinc nanoparticle. In some forms, the method further comprises the step of coating the zinc nanoparticle with protamine. In some forms, the RNA or mRNA encodes a SARS-CoV-2 S protein. In some forms, the SARS-CoV-2 S protein has at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or even 100%% sequence homology with SEQ ID NO. 1. In some forms, the immunogenic composition further comprises a pharmaceutical-acceptable carrier.
  • the pharmaceutical-acceptable carrier comprises a stabilizing agent and/or a preservative and/or an antimicrobial agent.
  • the RNA or mRNA is present in said immunogenic composition at a concentration greater than 100 micrograms per milligram weight of nanoparticle.
  • the zinc nanoparticle comprises a cell penetrating peptide.
  • the zinc nanoparticle comprises protamine.
  • the RNA or mRNA further comprises a sequence encoding a CD40L.
  • the RNA or mRNA further comprises a nucleic acid encoding a tetrameric CD40L trimer.
  • the SARS-CoV-2 S protein is encoded by a polycistronic mRNA.
  • the CD40L protein is encoded by a polycistronic mRNA.
  • the RNA or mRNA includes a nucleic acid encoding a SARS- CoV-2 S protein / CD40L fusion protein.
  • the RNA or mRNA includes an inserted sequence having at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or even 100%% sequence homology with SEQ ID NO. 9 or 10.
  • the RNA or mRNA is present at a concentration of greater than 100 pg/mg of nanoparticle.
  • the temperature stabilization provides an increase in intact RNA or mRNA after at least two weeks of storage at a temperature above freezing when compared to RNA or mRNA that is not bound to a zinc nanoparticle. In some forms, the temperature stabilization provides an increase in expression activity of the RNA or mRNA after at least two weeks of storage at a temperature above freezing when compared to RNA or mRNA that is not bound to a zinc nanoparticle.
  • a method of increasing the amount of intact RNA or mRNA during storage at a temperature above freezing comprises the step of binding the RNA or mRNA to a zinc nanoparticle, wherein the increase of the amount of intact RNA or mRNA is in comparison to RNA or mRNA that is not bound to a zinc nanoparticle.
  • the method further comprises the step of coating the zinc nanoparticle with protamine.
  • the RNA or mRNA encodes a SARS-CoV-2 S protein.
  • the SARS-CoV-2 S protein has at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or even 100%% sequence homology with SEQ ID NO. 1.
  • the immunogenic composition further comprises a pharmaceutical-acceptable carrier.
  • the pharmaceutical-acceptable carrier comprises a stabilizing agent and/or a preservative and/or an antimicrobial agent.
  • the RNA or mRNA is present in said immunogenic composition at a concentration greater than 100 micrograms per milligram weight of nanoparticle.
  • the zinc nanoparticle comprises a cell penetrating peptide.
  • the zinc nanoparticle comprises protamine.
  • the RNA or mRNA further comprises a sequence encoding a CD40L.
  • the nucleic acid encodes a tetrameric CD40L trimer.
  • the SARS-CoV-2 S protein is encoded by a polycistronic mRNA.
  • the CD40L protein is encoded by a polycistronic mRNA.
  • the RNA or mRNA includes a nucleic acid encoding a SARS- CoV-2 S protein / CD40L fusion protein.
  • the RNA or mRNA includes an inserted sequence having at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or even 100%% sequence homology with SEQ ID NO. 9 or 10.
  • the RNA or mRNA is present at a concentration of greater than 100 pg/mg of nanoparticle.
  • the storage is for a period of at least 2 weeks.
  • the disclosure provides a method of increasing the expression activity of RNA or mRNA during storage at a temperature above freezing.
  • the method comprises the step of binding the RNA or mRNA to a zinc nanoparticle, wherein the increase in expression activity of the RNA or mRNA is in comparison to RNA or mRNA that is not bound to a zinc nanoparticle.
  • the method further comprises the step of coating the zinc nanoparticle with protamine.
  • the RNA or mRNA encodes a SARS-CoV-2 S protein.
  • the SARS-CoV-2 S protein has at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or even 100%% sequence homology with SEQ ID NO. 1.
  • the immunogenic composition further comprises a pharmaceutical-acceptable carrier.
  • the pharmaceutical-acceptable carrier comprises a stabilizing agent and/or a preservative and/or an antimicrobial agent.
  • the RNA or mRNA is present in said immunogenic composition at a concentration greater than 100 micrograms per milligram weight of nanoparticle.
  • the zinc nanoparticle comprises a cell penetrating peptide.
  • the zinc nanoparticle comprises protamine.
  • the RNA or mRNA further comprises a sequence encoding a CD40L. In some forms, the RNA or mRNA further comprises a nucleic acid encoding a tetrameric CD40L trimer. In some forms, the SARS-CoV-2 S protein is encoded by a polycistronic mRNA. In some forms, the CD40L protein is encoded by a polycistronic mRNA. In some forms, the RNA or mRNA includes a nucleic acid encoding a SARS- CoV-2 S protein / CD40L fusion protein.
  • the RNA or mRNA includes an inserted sequence having at least 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or even 100%% sequence homology with SEQ ID NO. 9 or 10.
  • the RNA or mRNA is present at a concentration of greater than 100 pg/mg of nanoparticle.
  • the ZnO-protamine-RNA likely has several other characteristics which will make it a successful RNA and/or mRNA vaccine formulation including high RNA loading, delivery, and immunological activity.
  • SARS-COV-2 infects lung and is detected in kidney, liver and brain and metastasis of drug-resistant cancer to these organs can cause significant suffering and lead to rapid physiological decline in humans, companion animals and many other organisms.
  • Antimicrobial, antiviral and anticancer activity has been previously reported for zinc oxide nanoparticle (ZnO NP), but its in vivo characteristics have been insufficiently described.
  • ZnO NP zinc oxide nanoparticle
  • FIG. 4 shows ZnO NP or its cy5.5 fluorophore conjugate tissue distribution to liver, kidney, spleen, lungs and brain with no overt toxicity in these organs and blood counts in the normal range relative to untreated or reference mice.
  • ZnO tissue uptake was in the following order; liver>spleen>kidney>lung>heart>brain. At 5 hours or 3 days after intravenous administration no tissue damage or change in blood cell count was observed relative to control PBS injected mice or reference standards.
  • cy5.5-ZnO is readily taken into 3-D organoid tissues (2a) and increases the uptake and intracellular distribution of cy5.5-ASO into melanoma cells (2b) as shown by confocal fluorescence microscopy with apparent nuclear ASO location for CO3O4 NP. Increased ASO uptake stimulated by the NP delivery was confirmed by flow cytometry, where the ZnO, CO3O4 or NiO NP complexed to cy5.5- ASO had a 3-log order shift in cellular fluorescence compared to cy5.5-ASO oligo only control (2c).
  • Fig. 6 shows the outstanding biocompatibility of the Zn-based PMC nanoscale materials, particularly the MnZnSe, MnZnS or alternatively FeZnS. Very little cytotoxicity is seen for MnZnS and FeZnS or MnZnSe after 48 hour continuous exposure to untransformed highly sensitive NIH3T3 cells at 10, 20 or 25 microgr/ml concentration (2c). The MgZnO material was relatively toxic at the two higher doses, either 20 or 25 microgr/ml whereas the FeZnS and NiZnO were intermediate and better tolerated.
  • Fig. 7 shows the unquenchable fluorescent yield of the PMCs in serum, liver and tumor homogenate (7a). FeZnS and MnZnS also potentiate bioluminescence causing a red-shifted signal at 10 4 -10 4 relative light units above background (7b).
  • the PMC show some tissue-specificity for example MnZnSe fluorescence shifts in the kidney slurry and the MnZnS fluorescence is enhanced on the liver homogenate (7c). FeZnS also red-shifts in the kidney homogenate to emission at 725 nm. Dose-dependent signal intensity is also enhanced in ex vivo brain, liver (7d) and lung tissue (7e), especially for MnZnSe and MnZnS.
  • 2-D FDS 2-dimensional fluorescence difference spectroscopy
  • PMC are able to load and label 3-D spheroids with cy5.5- poly l:C (8a).
  • 3-D tumor spheroids treated with PMC break apart and many of the cells within the dense interior dye as shown by green/red intravtial staining (8b).
  • the PMCs or ZnO NP control also inhibit cancer cell invasion in the scratch assay (9c).
  • ANTICANCER ACTIVITY OF PMCs Anticancer activity of the PMC derivations and their ASO and aptamer complexes was tested next. First melanoma tumor at the time of metastasis was harvested and subjected to high throughput proteomics analysis confirming the import of proteins in the RAS/ERK/AKT pathways and secondarily BCL apoptosis pathways. To address this melanoma or model glioblastoma line (132N1 ) were treated with NP delivered RBD decoy protein interference or ASO targeting RBD or BCL-xL demonstrating outstanding inhibition (Fig. 9).
  • proteomics analysis revealed multiple proteins in the RAS/ERK/AKT associated with metastatic melanoma (9a, c). This could be targeted wither by PMC delivery of RBD decoy as a protein interference approach for CoZnO or CoFeZnO ternary or quaternary composites (9b), or by ASO targeting RBD or BCL-xL. Dogs are considered an excellent comparative oncology model for melanoma especially for rarer drug resistant forms, the canine mucosal melanoma are activated in the ERK/AKT pathway and are considered an excellent preclinical model for nanomedicine testing. These cells could be greatly inhibited by NiZnO even at early timepoints, but ironically in concert with RBD targeted ASO or aptamer no enhanced anticancer activity was seen, except for ZnO or MnZnS (9e).
  • MnZnS and MnZnSe in particular gave enhanced fluorescence and could be used to image these tissues based on their outstanding red-shifted fluorescence yield when exposed to ex vivo tissues, or their homogenates or slurries, notably including tumor, lung and brain slices or tissues of significance for both COVID and cancer metastasis.
  • PMC RBD or RBD targeted ASO or aptamer showed dose-dependent cancer cell inhibition and could be used to inhibit drug-resistant rarer forms of melanoma such as canine mucosal melanoma (M5) line used here.
  • Zn-based physiometacomposite nanoparticles are biocompatible and indigenously fluorescent. Importantly PMC have antiviral activity and can achieve significant RNA payloads and impart structural retention and temperature stabilization to RNA. PMC materials are thus of interest for pre-clinical applications of RNA vaccines and RNA-based therapeutics.
  • Nanomaterials and Reagents [0205] All nanoparticles used were obtained pure from Sigma-Aldrich or PlasmaChem GmbH (Berlin, Germany), except for MnZnSe was provided by Dr. Emily McLaurin formerly in the Department of Chemistry Kansas State University, MnZnS and FeZnS or NiZnO were provided by Dr. Garry Glaspell US Army Corps of Engineers, Cobalt Zinc Oxide (CoZnO) cobalt ferrite PMC were synthesized by Dr. KC Ghosh’s laboratory (Missouri State University).
  • Poly inosinic:poly cytidylic acid [poly(l:C)] was obtained from Sigma-Aldrich (Cat# P958250MG'). Cy5.5-labelled SSO (sequence: 3- CCUCUUACCUCAGUUACA-5) was obtained from Trilink Biotechnologies. Clinical- grade LL-37 peptide was obtained from our collaborator Dr. Cheng Kao Indiana University. NIH3T3 and A375 cells for cytotoxicity studies were obtained from ATCC. All NP and RNA were precipitated from 70% alcohol/H2O washed once with 100% alcohol, air dried in the biosafety cabinet prior to RNA and protein complexation, cell or animal administration. Copper (Cu) was purchased from PlasmaChem 10-100 nm in size..
  • Manganese Zinc Sulfide (MnZnS (1 , 3, 5%)), 50/50% Nickel Zinc Oxide (NiZnO), 10/90% NiZnO and Iron Zinc Sulfide (FeZnS) were all synthesized by Dr. G Glaspell’s laboratory (Virginia Commonwealth University). Cobalt Oxide (CO3O4), Nickel Oxide (NiO), 2/98% Scalable MnZnS and FeZnS , pure powders were physically mixed, heated to a flux, allowed to cool in an oxygen purged atmosphere and jet ball milled to nanoscale confirmed by transmission electron microscopy and nanoparticle tracking analysis [see supplemental data].
  • Cobalt Oxide CO3O4
  • NiO Nickel Oxide
  • FeZnS Iron Zinc Sulfide
  • NPs were washed with double-distilled water, 70% ethanol/water, ethanol, and were stored dry prior to use.
  • Costar (Coming, NY, USA) 96-well black, clear bottom assay plates were used for the assays.
  • Luciferase enzyme (Photinus pyralis, >10x10 1 ° (units/mg protein) was obtained from Sigma Aldrich and diluted it to a 0.2% solution [1 :500 dilution with PBS buffer], PBS buffer at 10X concentration was diluted to a 10% solution with de-ionized water [ddH20], Luciferase enzyme substrate buffer (ATP, Mg) was diluted to a 1 :1 vol/vol ratio with PBS buffer.
  • ATP Luciferase enzyme substrate buffer
  • ⁇ -Galactosidase from Aspergillus oryzae was obtained from Sigma Aldrich (>8.0 units/mg solid, Louis, Ml, USA) and was diluted to a 1 mg/kg solution in spectral grade H2O.
  • B-Gal substrate, Resorufin [3-D-galactopyranoside was purchased in a 10 g vial from Marker Gene Technologies (Eugene, OR, USA) and was diluted down into ten 10 mg/kg aliquots in spectral grade H2O and re-suspended into a 1 mg/kg solution for experimentation. Fluorescence emission, excitation and intensities were determined by Spectramax Paradigm.
  • HNOs 70% nitric acid
  • Histopathology and hematology analyses were performed at the Veterinary Medical Diagnostic Laboratory, KSU. The remainder of the collected tissues were fixed in 10% neutral buffered formalin. Sections of fixed tissue were routinely processed on a Sakura Tissue-TEK VIP 6 Processor prior to paraffin embedding. Slides were cut at and routinely stained with hematoxilin and eosin on a Leica Autostainer XL ST5010. Representative images at 10x magnification were captured on an Olympus LC20 camera mounted on an Olympus BX53F2 light microscope with CellSens (Olympus Corporation).
  • 3D Spheroid Culture of Caco2 cells Human Caco2 cells (ATCC®, passage 30) were seeded onto a 35mm sterile glass-bottomed cell culture dish (FluoroDishTM- World Precision Instruments) to form 3D spheroids in a thin layer of 10% Matrigel (Corning® Matrigel® Basement Membrane Matrix, LDEV-free). Culture medium was comprised of 1X Minimum Essential Media (MEM, L-glutamine free), 10% Fetal bovine serum, 1 % L-glutamine, and 1 % Pen/Strep. Caco2 spheroids were in culture for approximately 24-hours prior to imaging.
  • MEM Minimum Essential Media
  • Fetal bovine serum 1 % L-glutamine
  • Pen/Strep Pen/Strep
  • ZnO-PEG-Cy5.5 (ZnOCy5.5) nanoparticles (NPs) to human Caco2 spheroids: Immediately prior to delivery, ZnOCy5.5 NPs were diluted to a stock concentration in Ham’s F12 medium (160 pg/mL) sonicated for 60 seconds at room temperature (Fisher Scientific 60 Sonic Dismembrator Model F60 Cell Disrupter). Caco2 Matrigel-embedded spheroids were exposed to 20 pg/mL of ZnOCy5.5 NPs in culture media overnight in a 5% CO2 humidified incubator at 37 °C.
  • B16F10 cells were incubated with cy5.5-ASO control or complexes with cy5.5-ZnO-NP cy5.5-ASO:Co3O4 NP incubated for 24 h, rinsed with PBS and imaged by confocal microscopy.
  • NIH3T3 cells were treated with Cas9-GFP fusion protein or the MgO-NP control or MgO-Cas9 NP and imaged by fluorescence microscopy.
  • B16F10 melanoma cells were incubated with cy5.5-ASO control or the complexes with ZnO, CO3O4 or NiO NP and 24 h later, rinsed with PBS, trypsinized and analyzed for cellular fluorescence by flow cytometry (K-State VDL core lab).
  • B16F10 melanoma spheroids were allowed to establish for several days and treated with cy5.5-poly l:C viral mimetic RNA, the ZnO NP or cobalt (Co/ZnO) or nickel (Ni/ZnO) composites and imaged by fluorescence microscopy.
  • Engineered human melanoma cells we previously reported under the control of an ASO inducible luciferase expression and delivery in the 5 replicate wells quantified by relative luminescence per well.
  • Mouse tumor was isolated at the time of metastasis, 20 mg samples lysed, the proteins extracted (2-3 tumors were pooled) and standardized to A280 (Molecular Devices Spectramax i3x, Sunnyvale, CA, USA). Slides were incubated with Cy3- Streptavidin (Sigma Aldrich, St. Louis, MO, USA), dried by centrifugation and stored under dark conditions and imaged using Molecular Devices Genepix 4000B (Sunnyvale, CA, USA). Nanobio interaction was confirmed by CD, FT-IR and zeta potential and payload estimated as previously described. The molecular dynamics simulation was performed with the peptide in the water far above the surface and observed the adsorption process (2a).
  • Fluorescent microscopy was conducted on an Olympus IX73 within poly-D-lysine coated 8 chamber slide, cells inoculum density (5x10 4 ) after o/n adherence exposed to 20 ug/ml NP with maximal payload of Cas9-GFP (Applied Biological Materials Inc. Richmond, BC, Canada) or cy5.5-SSO versus cy5.5-SSO control (200 nM) assayed in the Texas Red/Rhodamine filter/channel.
  • 3-D spheroids were formed within Insphero Corp plates, 48 h later the media was changed with NP as above containing Rhoda-poly l:C or stained with Invitrogen live/dead stain and imaged in the bio-imager (Licor Pearl Trilogy) or on the fluorescence microscope respectively.
  • Bio-luminescence and fluorescent readings were taken on PerkinElmer (Caliper) LifeSciences IVIS Lumina II imager. Fluorescent readings were set to sixty second exposure, medium binning, 1 F/stop, dsRed emission filter, and intensity viewed through Rainbow setting. 200 ⁇ l of NP (FeZnS and 3% MnZnS) ⁇ luciferase were added to an all-black FB brand 9-well microtiter plate in PBS solution (1 :1 vol/vol). Doseresponse assay utilized NPs that elicited high relative light units (RLUs) in earlier experiments for further analysis of the effects of increasing NP dose when incubated with ⁇ -Gal.
  • RLUs relative light units
  • Mouse specimens were provided by Comparative Medicine Group. Lung sections were removed and were evenly divided into 2 sections. Ex-vivo imaging was performed in the Pearl® Trilogy Bioimaging system. 1 mg/ml of MnZnSe was diluted to a 1 :3 with HPLC water and injected into individual sample and then imaged under white light, 700, and 800 nm filter, with 85 pm resolution and “0” focus. Increasing volumes (pl) were injected (1 -20) with increasing fluorescent output. Tissue slurry/homogenate preparation consisted of heart, liver, kidney, brain, spleen, and lung from three different mice. Tissues were weighed on the XS204 Mettler Toledo (Columbus, OH, USA) analytical balance.
  • Sectioned samples 100 mg per ml were then placed in sterile 10% PBS buffer, and homogenized via Vibra-Cell Processor VCX 130 (Newton, CT, USA) for 2 minutes, with 10 second pulses and 5 seconds rest.
  • Slurry composition contained stromal tissue homogenized in with the sample (liver and kidney); homogenate composition had stroma removed via centrifugation and removal of supernatant to a separate tube (lung, heart, small intestine, liver, kidney and spleen).
  • PMC’s (1 mg/ml) were spiked into tissue slurry/homogenate and then placed into BRAND® 96-well black bottomed plates (CAT# 781668). 200 ⁇ l of PMC/tissue slurry/homogenate was assayed via SpectraMax i3x by Molecular Devices (San Jose, CA, USA) for 2-Dimensional Fluorescence Difference Spectroscopy (2D-FDS). For NP- Luc 2-D FDS bioluminescent readings, an all-black, FB brand 96-well titer plate was loaded with 200 ⁇ l of NP (FeZnS and MnZnS) in a 1 :1 vol/vol solution.
  • Luc was added (10 pl) with 200 ⁇ l of NP and spun down at 140 RPM for one minute and re-suspended and added to the microtiter plate.
  • the wavelength settings were set to unknown, spin before read, and optimization settings were set at excitation (250-830 nm) and emission (270-850 nm).
  • MTT Assay For cytotoxicity (MTT) assay, NIH3T3 fibroblast cells were seeded on a 96- well plate with 5,000 cells/well and allowed to grow for 24 h in DMEM with 10% FBS and 1 % penicillin/streptomycin. After 24 h, the medium was replaced with PMCs (5% NiZnO, MnZnS, 5% MgZnO, FeZnS and MnZnSe) dissolved in DMEM at 10, 20 and 25 pg/ml. Each treatment was tested on four wells. Four wells containing DMEM alone served as the blank and four wells with untreated cells served as the control. The cells were monitored daily for visible cytotoxicity using a light microscope.
  • MTT cytotoxicity
  • PRRSV-GFP Green fluorescence protein
  • MARC-145 cells derived from African green monkey kidney cells.
  • Assays to determine the antiviral activity of MnZnS nanoparticles (NP) were similar to previous work investigating the in vitro efficacy of fatty acid and formaldehyde based additives on reducing the titer of African swine fever virus (Niederwerder et al. 2020).
  • MnZnS NP Dilutions of MnZnS NP (100 pg/ml, 50 pg/ml, 20 pg/ml, 10 pg/ml) were prepared in Minimum Essential Medium (Corning® Eagle's MEM; Fisher Scientific) supplemented with fetal bovine serum, antibiotics, and anti-mycotics. Each dilution of MnZnS NP was mixed with an equal volume of PRRSV-GFP (titer 10 6 50% tissue culture infectious dose per ml, TCIDso/ml) for testing anti-viral activity. Positive controls included PRRSV-GFP mixed with an equal volume of MEM. Cytotoxicity controls included MnZnS NP mixed with an equal volume of MEM.
  • mRNAs were synthesized in vitro using mMESSAGE mMACHINETM T7 ULTRA Transcription Kit (AM1345, Thermofisher). Briefly, pcDNA3.1 construct encoding spike protein was linearized using Xbal restriction enzyme and used as template to synthesize mRNA. After synthesis, ⁇ 100 nt long poly(A) tail was added. mRNA recovery was performed by LiCI precipitation method. Lipofectamine MessengerMAX (LMRNA001 ) was used to transfect mRNA into HEK 293A cells. Briefly, 3pL of transfection reagent and 700ng of mRNA was used and 36 hrs.
  • LMRNA001 Lipofectamine MessengerMAX
  • FIG. 1 provides the amino acid sequence of the SpikeFTm-2A-cD5SPD-CD40L polypeptide.
  • the SARS-CoV-2 spike has a Fusion [F] protein leader sequence at the N-terminus, a flexible linker, and the transmembrane domain of an F protein at the C terminus.
  • a 2A autocleavable sequence was added to allow co-expression of human CD40L using human surfactant protein D [SPD] tetramerization motif.
  • This cassette is designed to allow optimal expression of Spike antigen on the surface of transfected cells and concurrent secretion of the CD40L cytokine.
  • Figure 2 provides an amino acid sequence of the Spike-SPD-CD40L polypeptide, which has a CD5 secretory signal followed by the SARS-CoV-2 spike, a flexible linker, SPD, and the CD40L functional domain.
  • This cassette is designed to allow secretion of the spike-CD40L chimeric protein.
  • the F Signal is a Fusion [F] protein signal sequence
  • Spike is a SARS-CoV-2 S protein
  • Flexible linker is a (G4S)3 linker sequence that allows the spike protein to fold
  • F Tm is a Fusion [F] protein transmembrane domain for expression of the spike on the surface for optimal recognition by B cells
  • 2A peptide is a 2A autocleavable peptide to allow multicistronic expression of the surface displayed spike antigen and secreted tetrameric CD40L molecular adjuvant
  • CD5 signal sequence is a CD5 secretory signal sequence
  • SPD is a Surfactant protein D tetramerization domain
  • CD40L is a molecular adjuvant/B cell agonist.
  • pCDNA3 FLp Spike FTM -2A- CD5 SPD- human CD40L [designated plasmid TX001 : pTX001 ]
  • the pCDNA3.1 vector has a T7 promoter that allows mRNA synthesis using the constructs above and a commercial mRNA miniprep synthesis kit.
  • the above mentioned pTX001 [and pTX003 which has hamster CD40L instead of human] constructs have been used to generate miniprep mRNA [ ⁇ 40 ⁇ g] for preliminary studies and showed that the generated mRNA is expressing spike antigen as judged by immunocytometric analyses using SARS-CoV-2 neutralizing mAbs (Fig. 14).
  • Formulation preparation The samples were made by weighing out 1.5mg of Zinc oxide nanoparticles (ZnO) (Sigma Alrich 100 nm) or (PlasmaChem 14 nm) and 1.5mg of protamine (Pr). ZnO was first washed with 1ml of 70% isopropyl, microcentrifuged at 5000 rpm for 5 minutes, and the supernatant was aspirated. The protamine was prepared by dissolving it in 1ml of milli-q water (Prot-hi).
  • ZnO Zinc oxide nanoparticles
  • Pr protamine
  • 3 different dilutions of protamine were created by taking 100 ⁇ I of Protamine solution and adding to another tube and adding 900pl of milli-q water to make a dilution of 1 :10 (Prot-med). Another dilution of 1 :100 was made from taking 100 ⁇ I from 1 :10 and adding 900pl of milli-q water (Prot-Lo) Once all 3 dilutions of Pr were created 33pl were added to the ZnO and mixed. These were microcentrifuged at 5000 rpm for 5 minutes and supernatant was aspirated. The RNA solution was made from 3 mg to 1 ml of milli-q water.
  • RNA 3 mg/ml was added to the ZnO and Pr sample, mixed, and washed with 1 ml of 100% cold alcohol, ethanol or isopropanol. The sample was microcentrifuged at 5000 rpm at 5 minutes and supernatant was aspirated. The samples were left to dry overnight in their certain temperatures of either 20, 30, 40, or 50 °C. To re-suspend the sample, 1 ml of PBS was added to the samples. 100 ⁇ I was added into another tube and 1 ml of cold alcohol was added for reprecipitation, then microcentrifuged at 5000 rpm for 5 minutes and supernatant was aspirated. Samples were air dried briefly prior to RNA elutuion and RAGE analysis.
  • RNA elution An initial elution buffer combined saturated heparin and poly- acrylic acid with 10X PBS and 50X TAE and 10% SDS at 1 :1 :1 :1 vol: vol. In subsequent experiments we noted elution could also be accomplished with a second lot of saturated Heparin buffered again by TAE/PBS with adjustment to 0.1 % SDS. Saturated urea in the presence of TAE provided a high intensity RNA band from a formulation prepared as above eluted and analyzed by RAGE analysis. Final elution buffer consisted of a 1 :1 ratio of 1X TAE and 1X PBS and was saturated with Urea.
  • Stability analyses Dry powder or PBS re-suspended formulations were maintained in stability chambers (30, 40 and 50 °C) for one to two days, one to two weeks or the formulations or the RNA stock solution stored in the refrigerator for the 4°C samples. In the hydrolysis experiment, 1 mg/ml nanoparticle was exposed to 1 mg/ml RNA overnight in a hot plate set to 37 °C, the samples removed and analyzed by RAGE.
  • NIH 3T3 cells were seeded in 24-well plates at a density of 30,000 cells per square centimeter. 7.4 mg of protamine sulfate were dissolved in 5 milliliters of deionized water autoclaved water. The solution was sonicated for 1 min, and the solution was divided into 1.5 mg of protamine. Each vial was centrifuged, and the supernatant was discarded. The precipitate was re-suspended in 50 mcl of cold 75 % ethanol and centrifuged. The alcohol was removed. 50 mcl of DI autoclaved water was added to all vials, and 7 mcl of 1.5 mg/mL protamine solution was added.
  • each vial was vortexed and centrifuged at 5000 rpm for 5 min. After the removal of the supernatant, 50 mcl of DD water was added to all vials. Then, green fluorescent protein was added to one vial and green wfluorescent protein mRNA was added to the other two vials. 7 mcl of protamine solution was added to one of the vials containing green fluorescent protein mRNA. 100 mcl of cold 75% ethanol were added to all vials to further sterilize the samples; ethanol was removed by centrifugation. Each formulation was suspended in 2 ml of DMEM, all samples were dispersed readily into solution.
  • mCherry mRNA cell lysate expression 10pg/ml ZnO solution was prepared using RNase free water. Protamine solution was also prepared using RNase free water and the solute to solvent ratio was 1 :100. 10OpI of 3T3 cell suspension (sonicated) was added in 4 wells of a 96 well plate with a clear bottom. 3pl of RNase free water was added in the first well with 3T3, 2pl of RNase free water and 1 ⁇ l of mCherry RNA was added in the 2nd well. In the third well, 1 ⁇ l of mCherry RNA, 1 ⁇ l RNase free water and 1 ⁇ l of ZnO solution was added.
  • fluorometric readings were taken on a spectroscope with the settings of fluorescence spectrum, fixed excitation at 550 nm and 587 nm and emission at 570-650 and 607-700 nm in 10 nm steps while the temperature was 37 degrees Celsius. Three readings were taken at the time periods of 0, 3 hrs, and 6 hrs.
  • Spike mRNA synthesis mMESSAGE mMACHINETM T7 ULTRA Transcription Kit (AM1345) was used to perform mRNA synthesis.
  • a pcDNA3.1 construct encoding spike protein was linearized with Xbal restriction enzyme and used as a template DNA for mRNA synthesis.
  • a 100 nt poly(A) tail was added after mRNA synthesis.
  • the LiCI precipitation method was used to recover mRNA. The recovered mRNA was quantified and stored at -80C for further use.
  • Spike mRNA transfection HEK293A cell monolayers were used to transfect mRNA on a 12-well plate format.
  • the spike mRNA, zinc oxide nanoparticle, with or without protamine formulation was mixed with HEK293A cell culture media and overlaid on cells in their respective wells.
  • the positive control was transfected with 3uL of lipofectamine MessengerMAX (LMRNA001 ) transfection reagent and 700ng of spike mRNA. Mock-transfected cells served as a negative control. To assess spike protein expression, the cells were fixed 24 hours after transfection and probed with a recombinant humanized anti-SARS-CoV-2 spike-specific neutralizing monoclonal antibody.
  • LMRNA001 lipofectamine MessengerMAX
  • Fig. 15A shows the lighter biomolecular RNA-protamine complexes associated at the nanoparticles surface.
  • the loss of RNA from the supernatant analysis also confirming RNA association to the protamine coated zinc nanoparticle (ZNP) is shown in Fig. 15B.
  • the nanoparticle size shifts as a consequence of protamine coating and RNA loading as shown in Fig. 15C.
  • Zeta potential measures the effective surface charge of nanoparticle.
  • RNA payloads with and without protamine coating was compared for several different singlestranded RNA types ranging from as low as only a few milligrams without protamine to more than one hundred for protamine coated TY-RNA, based on RNA loss from supernatant UV analysis (Fig. 15E).
  • RNA stability and expression activity Binding of RNA to zinc or protamine coated zinc nanoparticle (ZNP) was shown by gel shift and circular dichroism (CD) spectroscopy, in vitro translation, mRNA expression and cell delivery was studied next (Figs. 16A-D). Binding, stability and expression of ZNP-RNA and ZNP-mRNA shown by RNA agarose gel electrophoresis (RAGE) is shown in Fig. 16A, circular dichroism is shown in Fig. 11 B, in vitro translation is shown in Fig. 16B, cell free mRNA expression in lysates is shown in Fig. 16C and GFP mRNA cell delivery is shown in Fig. 16E.
  • ZNP zinc or protamine coated zinc nanoparticle
  • RNA stability after incubating torula yeast RNA (TY- RNA) with the nanoparticles in water at 37 °C followed by RNA agarose gel electrophoresis (RAGE) analysis was in the following order: ZnO > Fe3O4 > Ag > MSN ⁇ Cu > CNT ZnO NP (ZNP) and to a lesser extent Fe3O4, caused a slight gel shift consistent with their RNA interaction, but importantly retained RNA band staining intensity. In these cases, the band is tighter in comparison to the control RNA incubated over-night in water at 4 °C indicating some hydrolysis occurs in the control RNA but is protected in the ZnO NP and Fe3O4 samples.
  • RNA stain intensity or a smear pattern consistent with RNA degradation was seen with MSN, copper (Cu) NP and CNT, and silver (Ag) was intermediate.
  • the CD data shown in Fig. 11 B shows the typical two peak CD pattern for poly I and poly C which is well-known to form an A-form RNA double helix. This is stabilized with peak enhancement when ZNP is complexed to it.
  • Structural stability was a function of the stoichiometric ratio of poly l:C:ZnO-NP.
  • Fig. 16B although some variability was observed with two different batches of Luc mRNA used, both batches of ZNPe-mRNA formulations supported in vitro translation.
  • mRNA expression was independent of protamine ratio with 1 :1 (high), 1 :10 (medium) and 1 :100 (low), all supporting translation, elution of RNA from the particles was protamine-dependent (Fig. 19).
  • mCherry mRNA expression of the ZnO-protamine-mRNA was higher than ZnO-mRNA alone in both NIH3T3 and RAW264.7 cell extracts decreasing from three to six hours (Fig. 16C).
  • mRNA cell delivery experiments were conducted with green fluorescent protein (GFP). mRNA was compared to GFP protein control and protamine enhancing cell fluorescence (Fig. 16D).
  • RNA temperature stabilization was demonstrated next (Figs. 17A-B).
  • Figures 17A-B RNA Temperature-stabilization. Three independent DSC experiments (Fig. 11 A), RAGE analysis of heat treated RNA (Fig. 17B) and expression analysis from heat treated ZNP-mRNA (Fig. 17C).
  • RNA was then allowed to bind, the preparations were then sedimented from alcohol, air dried and then re-suspended in phosphate buffered saline (PBS) and stored for two weeks at 30, 40 or 50 °C, versus material that was left out on the bench (20 °C) or in the refrigerator (4 °C) for 2 weeks.
  • PBS phosphate buffered saline
  • RNA band staining intensity was slightly higher for 14 nm in comparison to 100 nm, as expected. Whereas some slight band broadening occurred more noticeably at the higher temperatures, intact RNA was clearly present after two weeks of storage even at room or above physiological temperature comigrating with control RNA. Importantly, this accelerated stability and retention of RNA integrity was here maintained by liquid formulations, even after high temperature exposure for two weeks. No lyophilization, concentration or dry powder step was required in order to demonstrate this stability. Finally expression of the heat treated mRNA samples in NIH3T3 cell extracts was protected by ZNP-mRNA (Fig. 17B).
  • RNA loading with protamine (ZNP) versus another cationic antiviral peptide, cathalecidin (LL-37)21 was evaluated next (Figs. 18A- F).
  • FIG. 18A-F RNA biological activity. Loading efficiency (Fig. 18A) and expression of SARS-CoV-2 spike mRNA: Protein expression was evaluated by immunostaining of HEK293A cells transfected with SARS-CoV-2 spike mRNA formulated in/as: (Fig. 18B) Zinc oxide nanoparticle-mRNA); (Fig. 18C) Protamine coated Zinc oxide nanoparticle (ZNP- mRNA) (Fig. 18D) Double protamine coated - one over the ZNP and other over mRNA (ZNP-protamine-mRNA-protamine) (Fig. 18E) InvitrogenTM LipofectamineTM MessengerMAXTM transfection reagent (positive control); and Fig.
  • 18(F) Mock-transfected cells which served as a negative control.
  • the cells were probed with a recombinant humanized anti-SARS-CoV-2 spike-specific neutralizing monoclonal antibody.
  • the secondary antibody was anti-human IgG-Alkaline Phosphatase, and Fast Red was the substrate. Arrows showing the cells expressing the spike protein.
  • Figs. 18A-F shows loading efficiency of RNA (TY-RNA) onto ZnO, ZNP or LL37 coated ZnO (ZN-LL37) (Fig. 18A) which was in the order; ZNP > ZN-LL37 > ZnO.
  • Figures 18A-F also demonstrate spike mRNA expression in cells consistent with the data shown above. Spike mRNA expression comparable to commercial transfection reagent was observed both for protamine underlayer (Fig. 18B) or overlayer coating (Fig. 18C) demonstrating the activity of the formulation. Clearly the cells grew and divided much more in the case of ZNP treated cells comparing the cell density in each well to that of lipofectamine (Fig. 18D), transfection demonstrated even in the presence of a dense cell mat, indicating some toxicity of the lipid-based formulation.
  • RNA temperature-stabilization of the ZnO-protamine-RNA (ZNP-RNA) formulation Stability-indicating methods used here were RAGE, CD and DSC, each method independently verifying the stability conferred to RNA by interaction to ZnO or ZnO-protamine (ZNP).
  • ZNP ZnO-protamine
  • structural stability is imparted to RNA by binding to ZnO nanoparticle as shown by CD which significantly increases RNA melting temperature as shown by DSC.
  • An early report suggested RNA temperature stabilization by surface-functionalized mesoporous silica nanoparticles (MSN) allowing brief storage of RNA at 4 °C.
  • RNA binding peptide (RBP) (Fig. 20) known for its cell membrane and antiviral activity. Alignment of low molecular weight protamine to the FK16 fragment of LL37 that is thought to have this membrane cell surface activity has some interesting similarities to cell penetrating peptide (penetratin) (Fig. 21 ). This suggests that peptide sequences which balance RNA loading with release and intracellular delivery may be of some interest in the future.
  • Zahmatkeshan, Mohammad Farahmand, Davod Javanmard Seyed Jalal Kiani, Maryam Esghaei, Vahid Pirhajati-Mahabadi, Seyed Hamidreza Monavari , Angila Ataei-Pirkooh J Biomed Sci. 2019 Sep 10;26(1 ):70. doi: 10.1186/s12929-019-0563-4.
  • RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Mycology (AREA)
  • Nanotechnology (AREA)
  • Inorganic Chemistry (AREA)
  • Plant Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Optics & Photonics (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne des compositions stabilisées et des procédés se rapportant à l'augmentation de la stabilité, de l'activité d'expression et de la quantité d'acide nucléique intact pendant le stockage. Cette invention est particulièrement applicable à des compositions immunogènes comprenant de l'ARNm et peut être incorporée dans des compositions pour réduire la gravité, l'incidence et/ou la transmissibilité du SARS-CoV-2.
PCT/US2022/075944 2021-09-02 2022-09-02 Formulations de vaccin à arnm et leurs procédés d'utilisation WO2023034991A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163260859P 2021-09-02 2021-09-02
US63/260,859 2021-09-02

Publications (1)

Publication Number Publication Date
WO2023034991A1 true WO2023034991A1 (fr) 2023-03-09

Family

ID=85411634

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/075944 WO2023034991A1 (fr) 2021-09-02 2022-09-02 Formulations de vaccin à arnm et leurs procédés d'utilisation

Country Status (1)

Country Link
WO (1) WO2023034991A1 (fr)

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080226562A1 (en) * 2005-12-22 2008-09-18 Kevin Groves Biocompatible fluorescent metal oxide nanoparticles
US20170131272A1 (en) * 2015-11-05 2017-05-11 Kansas State University Research Foundation Two-dimensional fluorescence difference spectroscopy characterization of nanoparticles and their interactions
US20180223260A1 (en) * 2011-10-21 2018-08-09 Stemgenics, Inc. Functionalized nanoparticles for the intracellular delivery of biologically active molecules and methods for their manufacture and use
CN111358953A (zh) * 2020-03-25 2020-07-03 上海市公共卫生临床中心 一种高效诱导机体体液免疫应答的疫苗载体、其制备方法及用途
WO2020185628A1 (fr) * 2019-03-08 2020-09-17 Obsidian Therapeutics, Inc. Compositions de cd40l et procédés de régulation accordable
US20200360513A1 (en) * 2019-05-17 2020-11-19 Sila Nanotechnologies Inc. Nanofiber compositions for a vaccine adjuvant, porous scaffold or porous membrane
US20210085619A1 (en) * 2019-09-23 2021-03-25 Northeastern University Nanoencapsulated Combination Drug Formulations
WO2021159040A2 (fr) * 2020-02-07 2021-08-12 Modernatx, Inc. Vaccins à domaine arnm anti sars-cov-2
WO2021163438A1 (fr) * 2020-02-14 2021-08-19 University Of Washington Polypeptides, compositions et leur utilisation pour traiter ou limiter le développement d'une infection
WO2022178543A1 (fr) * 2021-02-18 2022-08-25 Kansas State University Research Foundation Nanoparticules de stabilisation d'arn

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080226562A1 (en) * 2005-12-22 2008-09-18 Kevin Groves Biocompatible fluorescent metal oxide nanoparticles
US20180223260A1 (en) * 2011-10-21 2018-08-09 Stemgenics, Inc. Functionalized nanoparticles for the intracellular delivery of biologically active molecules and methods for their manufacture and use
US20170131272A1 (en) * 2015-11-05 2017-05-11 Kansas State University Research Foundation Two-dimensional fluorescence difference spectroscopy characterization of nanoparticles and their interactions
WO2020185628A1 (fr) * 2019-03-08 2020-09-17 Obsidian Therapeutics, Inc. Compositions de cd40l et procédés de régulation accordable
US20200360513A1 (en) * 2019-05-17 2020-11-19 Sila Nanotechnologies Inc. Nanofiber compositions for a vaccine adjuvant, porous scaffold or porous membrane
US20210085619A1 (en) * 2019-09-23 2021-03-25 Northeastern University Nanoencapsulated Combination Drug Formulations
WO2021159040A2 (fr) * 2020-02-07 2021-08-12 Modernatx, Inc. Vaccins à domaine arnm anti sars-cov-2
WO2021163438A1 (fr) * 2020-02-14 2021-08-19 University Of Washington Polypeptides, compositions et leur utilisation pour traiter ou limiter le développement d'une infection
CN111358953A (zh) * 2020-03-25 2020-07-03 上海市公共卫生临床中心 一种高效诱导机体体液免疫应答的疫苗载体、其制备方法及用途
WO2022178543A1 (fr) * 2021-02-18 2022-08-25 Kansas State University Research Foundation Nanoparticules de stabilisation d'arn

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
PROMPETCHARA ID EAKACHAI, KETLOY ID CHUTITORN, THARAKHET KITTIPAN, KAEWPANG PAPATSARA, BURANAPRADITKUN SUPRANEE, TECHAWIWATTANABOO: "DNA vaccine candidate encoding SARS-CoV-2 spike proteins elicited potent humoral and Th1 cell-mediated immune responses in mice", 22 March 2021 (2021-03-22), pages 1 - 16, XP055825107 *

Similar Documents

Publication Publication Date Title
CN112076315A (zh) 新冠病毒s蛋白和铁蛋白亚基融合的纳米抗原颗粒、新冠疫苗及其制备方法和应用
CN107098974B (zh) 一种融合蛋白及其应用
CN112439056B (zh) 基于自组装铁蛋白纳米抗原颗粒及由其制备的o型口蹄疫疫苗和应用
CN111996216B (zh) 腺相关病毒介导的新型冠状病毒抗体诱导物及疫苗组合物
WO2008087434A1 (fr) Polynucléotides et leurs utilisations
US20170290905A1 (en) Vaccine
US20230167159A1 (en) Sars-coronavirus 2 (sars-cov-2) subunit vaccine candidates
CN114277039B (zh) 呼吸道合胞病毒mRNA疫苗及其制备方法和应用
US20230174588A1 (en) A vaccine against sars-cov-2 and preparation thereof
JP2023543623A (ja) 融合関連小膜貫通タンパク質が配合されたプロテオリピド小胞
Bhuyan et al. The construction of recombinant Lactobacillus casei expressing BVDV E2 protein and its immune response in mice
TW201502136A (zh) 疫苗
CN110638787B (zh) 一种防治鼻咽癌的亚单位纳米疫苗及其制备方法
WO2023034991A1 (fr) Formulations de vaccin à arnm et leurs procédés d'utilisation
WO2022233287A1 (fr) Réactif vaccinal et méthode de vaccination
EP3511417B1 (fr) Glycoprotéines gn et gc du virus de la fièvre de la vallée du rift, et leur utilisation
Tang et al. A ferritin nanoparticle vaccine based on the hemagglutinin extracellular domain of swine influenza A (H1N1) virus elicits protective immune responses in mice and pigs
CN113087777B (zh) 抗SARS-CoV-2感染的蛋白以及用该蛋白制备的疫苗
CN115960180A (zh) 2019-nCoV S蛋白的突变体及其基因工程化的mRNA和疫苗组合物
KR102400570B1 (ko) 톡소포자충 정점막 항원 1을 포함하는 바이러스-유사입자 및 이를 이용한 백신
CN113801206A (zh) 利用受体识别域诱导抗新冠病毒中和抗体的方法
Liu et al. Rabbit hemorrhagic disease virus VP60 protein expressed in recombinant swinepox virus self-assembles into virus-like particles with strong immunogenicity in rabbits
WO2022246449A1 (fr) Vaccins à sous-unité de protéine de spicule de sars-coronavirus 2 (sars-cov-2)
TWI853485B (zh) 核酸-脂質奈米粒子及使用其之方法
KR102536927B1 (ko) 말라리아 원충의 메로조이트 표면 단백질-8을 포함하는 바이러스-유사입자 및 이를 이용한 백신

Legal Events

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

Ref document number: 22865858

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22865858

Country of ref document: EP

Kind code of ref document: A1