WO2022011436A1 - Méthodes de traitement d'une infection à coronavirus avec du colostrum hyperimmun bovin - Google Patents

Méthodes de traitement d'une infection à coronavirus avec du colostrum hyperimmun bovin Download PDF

Info

Publication number
WO2022011436A1
WO2022011436A1 PCT/AU2021/050772 AU2021050772W WO2022011436A1 WO 2022011436 A1 WO2022011436 A1 WO 2022011436A1 AU 2021050772 W AU2021050772 W AU 2021050772W WO 2022011436 A1 WO2022011436 A1 WO 2022011436A1
Authority
WO
WIPO (PCT)
Prior art keywords
bovine
lps
etec
vaccine
hyperimmune colostrum
Prior art date
Application number
PCT/AU2021/050772
Other languages
English (en)
Inventor
Jerry Kanellos
Roger Aston
Peter ANASTASIOU
Original Assignee
Immuron Limited
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
Priority claimed from AU2020902486A external-priority patent/AU2020902486A0/en
Application filed by Immuron Limited filed Critical Immuron Limited
Publication of WO2022011436A1 publication Critical patent/WO2022011436A1/fr

Links

Classifications

    • 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
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/20Milk; Whey; Colostrum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0075Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a dry powder inhaler [DPI], e.g. comprising micronized drug mixed with lactose carrier particles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/04Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from milk
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1203Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1203Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
    • C07K16/1228Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • C07K16/1232Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia from Escherichia (G)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39508Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum from milk, i.e. lactoglobulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/12Immunoglobulins specific features characterized by their source of isolation or production isolated from milk

Definitions

  • This invention relates to compositions and methods for the treatment and/or prevention of COVID-19 and COVID-19 associated disease.
  • the global COVID-19 vaccine R&D landscape includes 115 vaccine candidates, of which 78 are confirmed as active and 37 are unconfirmed (development status cannot be determined from publicly available or proprietary information sources); of these candidates there is a focus on live attenuated viruses, inactivated viruses, non-replicating viral vectors, replicating viral vectors, recombinant proteins, peptide based vaccines, virus like particles, and DNA and RNA vaccines.
  • the present invention provides a method of treating and/or preventing human coronavirus infection in a subject comprising administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of reducing viral shedding from a subject infected with a human coronavirus comprising administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of treating and/or preventing human coronavirus-associated diarrhoea in a subject comprising administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of inhibiting coronavirus mediated cytotoxicity comprising contacting one or more human cells with an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of improving cell viability comprising contacting one or more human cells with an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • the present invention provides a method of maintaining gastrointestinal cell viability comprising contacting one or more human cells with an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method as described herein, wherein the vaccine comprising ETEC LPS comprises one or more LPS O serotypes selected from the group consisting of 078, 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159.
  • the present invention provides a method as described herein, wherein the vaccine comprising ETEC LPS comprises two or more LPS 0 serotypes selected from the group consisting of 078, 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159.
  • the present invention provides a method as described herein, wherein the vaccine comprising ETEC LPS comprises LPS 0 serotypes 078, 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159.
  • the present invention provides a method as described herein, wherein the vaccine comprising ETEC LPS comprises the lipid A core and O-polysaccharide regions of the LPS.
  • the present invention provides a method as described herein, wherein the composition comprising bovine-hyperimmune colostrum comprises bovine immunoglobulin that binds to ETEC LPS.
  • the present invention provides a method as described herein, wherein the composition comprising bovine-hyperimmune colostrum comprises bovine immunoglobulin that binds to one or more LPS 0 serotypes selected from the group consisting of 078, 06, 08, 015, 025, 027, 063, 071 , 0114, 0115, 0117, 0128, 0148, 0153, and 0159, 0167.
  • the present invention provides a method as described herein, wherein the composition comprising bovine-hyperimmune colostrum comprises bovine immunoglobulin that binds to one or more LPS serotypes of a gram negative bacteria selected from the group consisting of Enteropathogenic Escherichia coli strain E2348/69, Klebsiella pneumoniae strain ATCC 26, Pseudomonas aeruginosa strain ATCC 27853, Salmonella typhimurium strain ATCC 14028, Vibrio cholerae strain 6239, Yersinia enterocolitica strain 67R, C. jejuni, Vibrio cholera Ogawa, V. cholera Inaba, and V.
  • a gram negative bacteria selected from the group consisting of Enteropathogenic Escherichia coli strain E2348/69, Klebsiella pneumoniae strain ATCC 26, Pseudomonas aeruginosa strain ATCC 27853, Salmonella typhimurium strain AT
  • the present invention provides a method as described herein, wherein the composition comprising bovine-hyperimmune colostrum is prepared using a first vaccine comprising ETEC LPS and a second vaccine comprising ETEC LPS.
  • the present invention provides a method as described herein, wherein the composition comprising bovine-hyperimmune colostrum is prepared using a first vaccine comprising ETEC LPS and a second vaccine comprising a bovine coronavirus antigen and/or a bovine coronavirus.
  • the present invention provides a method as described herein, wherein the composition comprising bovine-hyperimmune colostrum is prepared using a vaccine comprising ETEC LPS administered to an animal sequentially- or co-administered a vaccine comprising a bovine coronavirus antigen and/or a bovine coronavirus.
  • the vaccine comprising a bovine coronavirus is an attenuated bovine coronavirus.
  • the present invention provides a method as described herein, wherein the composition comprising bovine-hyperimmune colostrum is prepared using a vaccine comprising ETEC LPS administered to an animal producing antibodies to bovine coronavirus.
  • the present invention provides a method as described herein, wherein the composition comprising bovine-hyperimmune colostrum comprises bovine immunoglobulin that binds to bovine coronavirus.
  • the present invention provides a method as described herein, wherein the human coronavirus is SARS-COV-2.
  • the present invention provides a method as described herein, wherein the hyperimmune colostrum is prepared by administering a bovine animal with a vaccine comprising ETEC LPS and collecting hyperimmune colostrum from the animal.
  • the present invention provides a method as described herein, wherein the hyperimmune colostrum is prepared by administering one bovine animal with a first vaccine and a second bovine animal with a second vaccine, and subsequently combining the hyperimmune colostrum collected from the animals.
  • the present invention provides a method as described herein, wherein the hyperimmune colostrum comprises one or more immunoglobulin classes selected from the group of IgG, IgM and IgA.
  • the present invention provides a method as described herein, wherein the hyperimmune colostrum further comprises a component selected from oligosaccharides, beta lactoglobulin, alpha-lactalbumin, lactoferrin, bovine serum albumin, growth factors, lactoperoxidase, plasm in, lipoprotein lipase, esterase, ribonucleases A, B, C, D and 11-1 , Lysozyme, enzyme inhibitors, cytokines, and lipids.
  • oligosaccharides selected from oligosaccharides, beta lactoglobulin, alpha-lactalbumin, lactoferrin, bovine serum albumin, growth factors, lactoperoxidase, plasm in, lipoprotein lipase, esterase, ribonucleases A, B, C, D and 11-1 , Lysozyme, enzyme inhibitors, cytokines, and lipids.
  • the present invention provides a method as described herein, wherein the effective amount of a composition comprising bovine- hyperimmune colostrum is administered to the subject orally, nasally or using pulmonary administration.
  • the present invention provides a method as described herein, wherein the contacting is performed in vivo in a human.
  • the present invention provides a method as described herein, wherein a symptom of coronavirus infection is treated and/or prevented.
  • the present invention provides a method as described herein, wherein the symptom of coronavirus infection is selected from the group consisting of a secondary bacterial infection, sepsis, bowel pain, nausea, vomiting and diarrhoea.
  • the present invention provides a method as described herein, wherein the symptom of coronavirus infection is the load of coronavirus in stool.
  • the present invention provides a method of treating and/or preventing coronavirus infection in one or more human subjects comprising applying to a surface selected from air filters, PPE, room surfaces and respiratory mucosal membranes an effective amount of a composition comprising bovine- hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • composition comprising bovine-hyperimmune colostrum is adsorbed on a particulate carrier of a size in the range from 50 to 120 microns.
  • composition comprising bovine-hyperimmune colostrum is adsorbed onto a solid carrier for administration by dry powder inhaler.
  • composition comprising bovine-hyperimmune colostrum is administered as an aerosol.
  • composition comprising bovine-hyperimmune colostrum is in dried form.
  • the present invention provides a method of decreasing levels of LPS in the blood of a subject, comprising administering to the subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of treating and/or preventing sepsis in a subject, comprising administering to the subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of decreasing inflammation in a subject, comprising administering to the subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of treating and/or preventing cytokine storm in a subject comprising administering to the subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • the present invention provides a method of treating and/or preventing a symptom associated with coronavirus infection in a subject comprising administering to the subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the symptom associated with coronavirus infection is selected from the group consisting of loss of gastrointestinal flora, increased inflammation, elevated ALT levels, elevated AST levels, pathologic change, LPS in the blood, and cytokine storm, human coronavirus-associated diarrhoea.
  • the symptom associated with coronavirus infection is secondary to SARS-CoV-2 infection.
  • the pathologic change is gastrointestinal tract pathology, or lung pathology.
  • the present invention provides a method as described herein wherein the increased inflammation is increased inflammation in the lungs, increased inflammation in the liver, and/or systemic inflammation.
  • the present invention provides a method of reducing viral shedding from a subject infected with a human coronavirus comprising administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the subject is a subject with SARS-CoV-2 infection.
  • Figure 1 A shows LPS extracted from ETEC strains in the ETEC vaccine, run on 15% Tris-tricine SDS-PAGE and stained using LPS specific silver stain. 0- polysaccharide chains are shown as ladders and lipid A core region is marked with an arrow.
  • Figure 1 B shows western transfer to PVDF membrane and blotted with Bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS, made from a pool of colostrum from over 1000 cows.
  • ETEC enterotoxigenic E. coli
  • FIG. 2A shows a silver stained 15% T ris-tricine gel showing LPS extracted from ETEC serotypes not included in the ETEC vaccine. Shown are LPS extracted from 2 - ETEC strain M452C1 [serotype 020: H-]; 3 - ETEC strain T0225-C4 [075: H4]; 4 - ETEC strain 83-552 [0126: FI-]; 5 - ETEC strain G33 [0126: H 12]; 6 - ETEC strain M145C2 [0128:H(NT)]; 7 - ETEC strain E23477/0/A [0139:H25]; 8 - ETEC strain ND782 [0141 :H4]; 9 - ETEC strain ND748 [O149:H10]; and 10 - ETEC strain
  • FIG. 2B shows western transfer to PVDF membrane and blotted with Bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS, made from a pool of colostrum from over 1000 cows.
  • ETEC enterotoxigenic E. coli
  • Figure 3A shows a silver stained 15% T ris-tricine gel showing LPS extracted from pathogens not included in the ETEC vaccine. Shown are LPS extracted from 2 - Enterobacter aerogenes strain ATCC 13048; 3 - Enteropathogenic Escherichia coli strain E2348/69; 4 - Klebsiella pneumoniae strain ATCC 26; 5 - Pseudomonas aeruginosa strain ATCC 27853; 6 - Salmonella typhimurium strain ATCC 14028; 7 - Vibrio cholerae strain 6239; 8 - Yersinia enterocolitica strain 67R; 9 - Citrobacter rodentium strain DSB100; and 10 - ETEC strain E11881A [025: H42] (+ control).
  • FIG. 3B shows western transfer to PVDF membrane and blotted with Bovine- hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS, made from a pool of colostrum from over 1000 cows.
  • ETEC enterotoxigenic E. coli
  • FIG 4 shows hyperimmune colostrum powder Batch # 1710002659 had an EC50 of 44.7 ug/ml (left hand curve, raw data points “+”) and a CC50 of 1085.3 ug/ml (right hand curve, raw data points “°”).
  • This batch of colostrum inhibits viral replication at a dose at which there is no cell toxicity, and at a dose that inhibits 90% of viral replication there is no cell toxicity (an EC90 of 53.15 ug/ml).
  • FIG. 5 shows hyperimmune colostrum powder Batch # 1710002660 had an EC50 of 91 .9 7 ug/ml (left hand curve, raw data points “+”) and a CC50 of 1790.6 ug/ml (right hand curve, raw data points “°”).
  • This batch of colostrum inhibits viral replication at a dose at which there is no cell toxicity, and at a dose that inhibits 90% of viral replication there is no cell toxicity (an EC90 of 155.39 ug/ml).
  • FIG. 6 shows hyperimmune colostrum powder Batch # 1710002662 had an EC50 of 40.5 7 ug/ml (bottom curve, raw data points “+”) and a CC50 of 2053.3 ug/ml (top/right hand curve, raw data points “°”).
  • This batch of colostrum inhibits viral replication at a dose at which there is no cell toxicity, and at a dose that inhibits 90% of viral replication there is no cell toxicity (an EC90 of 45.17 ug/ml).
  • FIG. 7 shows hyperimmune colostrum powder Batch # 1710002859 had an EC50 of 42.6 7 ug/ml (left hand curve, raw data points “+”) and a CC50 of 2506.6 ug/ml (right hand curve, raw data points “°”).
  • This batch of colostrum inhibits viral replication at a dose at which there is no cell toxicity, and at a dose that inhibits 90% of viral replication there is no cell toxicity (an EC90 of 48.69 ug/ml).
  • Figure 8 shows pro-milk powder Batch # 6068 N50 (control) had an EC50 of > the CC50 (EC50 - bottom curve, raw data points “+”), and a CC50 of 4818.6 ug/ml (top curve, raw data points “°”). Accordingly, this control milk powder inhibits viral replication at a dose >25000 ug/ml, and importantly does not inhibit viral replication at the doses at which it is cytotoxic to cells.
  • Figure 9 shows the positive control, remdesivir, has an EC50 of 1 16pM.
  • Figure 10 shows a composition prepared according to Example 1 decreases levels of the liver enzymes ASL and ALT in human subjects when administered orally.
  • Figure 11 shows a composition prepared according to Example 1 decreases LPS in the serum of human subjects when administered orally.
  • the present inventors have developed a composition comprising bovine- hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS that inhibits SARS-CoV-2 infection of cells.
  • ETEC enterotoxigenic E. coli
  • Example 2 shows hyperimmune colostrum powder compositions of the present invention, such as those prepared in Example 1 , inhibit viral replication at a dose at which there is no cell toxicity.
  • the compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS described herein are stable at room temperature, have a desirable safety profile, and/or do not require the induction of a humoral immune response in subjects administered.
  • the present invention provides a method of treating and/or preventing human coronavirus infection in a subject comprising administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the subject is a subject with impaired liver function.
  • the subject with impaired liver function has decreased AST and/or ALT levels.
  • the method includes a step of selecting a subject with impaired liver function prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the method includes a step of selecting a subject with decreased AST and/or ALT levels prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the subject is a subject with human coronavirus- associated diarrhoea.
  • the method includes a step of selecting a subject with human coronavirus-associated diarrhoea prior to administering an effective amount of a composition comprising bovine- hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the subject is a subject with sepsis.
  • the method includes a step of selecting a subject with sepsis prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the subject is a subject with increased inflammation.
  • the increased inflammation is increased inflammation in the lungs, increased inflammation in the liver, and/or systemic inflammation.
  • the method includes a step of selecting a subject with increased inflammation prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the method includes a step of selecting a subject with increased inflammation in the lungs prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the method includes a step of selecting a subject with increased inflammation in the liver prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the method includes a step of selecting a subject with increased systemic inflammation prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the subject is a subject with LPS-induced inflammation. In one embodiment the subject has elevated levels of LPS in the serum of the subject.
  • the method includes a step of selecting a subject with LPS-induced inflammation prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the method includes a step of selecting a subject with elevated levels of LPS in the serum prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the subject is a subject with cytokine storm.
  • the method includes a step of selecting a subject with cytokine storm prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the subject is a subject with loss of gastrointestinal flora.
  • the method includes a step of selecting a subject with loss of gastrointestinal flora prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the subject is a subject with increased pathologic change.
  • the pathologic change is gastrointestinal tract pathology, or lung pathology.
  • the method includes a step of selecting a subject with increased pathologic change prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the method includes a step of selecting a subject with increased gastrointestinal tract pathology prior to administering an effective amount of a composition comprising bovine- hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the method includes a step of selecting a subject with increased lung pathology prior to administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • human coronavirus includes severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2): SARS-CoV-2 is the virus strain that causes coronavirus disease 2019 (COVID-19), a respiratory illness. It is colloquially known as the coronavirus and was previously referred to by its provisional name 2019 novel coronavirus (2019-nCoV). SARS-CoV-2 is a positive-sense single- stranded RNA virus. It is contagious in humans, and the World Health Organization (WHO) has designated the ongoing pandemic of COVID-19 a Public Health Emergency of International Concern.
  • WHO World Health Organization
  • SARS-CoV-2 is a strain of Severe acute respiratory syndrome-related coronavirus (SARS-CoV). It is believed to have zoonotic origins and has close genetic similarity to bat coronaviruses, suggesting it emerged from a bat-borne virus. An intermediate animal reservoir such as a pangolin is also thought to be involved in its introduction to humans. The virus shows little genetic diversity, indicating that the spillover event introducing SARS-CoV-2 to humans is likely to have occurred in late 2019. Epidemiological studies estimate each infection results in 1.4 to 3.9 new ones when no members of the community are immune and no preventive measures taken. The virus is primarily spread between people through close contact and via respiratory droplets produced from coughs or sneezes. It mainly enters human cells by binding to the receptor angiotensin converting enzyme 2 (ACE2).
  • ACE2 receptor angiotensin converting enzyme 2
  • the human coronavirus is SARS-COV-2.
  • subject is used interchangeably with “patient”, and refers to a human infected with, or at risk of infection with a human coronavirus.
  • effective amount includes a “therapeutically effective amount” and a “prophylactically effective amount”.
  • a “therapeutically effective amount” refers to the amount of the composition comprising bovine-hyperimmune colostrum which when administered alone or in combination to a subject for treating Covid-19, or at least one of the clinical symptoms of Covid-19, is sufficient to affect such treatment of the disease, or symptom.
  • the therapeutically effective amount can vary depending, for example, on the formulation of the composition, the infection, and/or symptoms of the infection, severity of the infection, and/or symptoms of the infection, the age, weight, and/or health of the patient to be treated, and the judgment of the prescribing physician.
  • a therapeutically effective amount in any given instance may be ascertained by those skilled in the art or capable of determination by routine experimentation.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody are outweighed by the beneficial effects.
  • a “prophylactically effective amount” is any amount of the antibody that, when administered alone or in combination to a patient, inhibits or delays the onset or recurrence of Covid-19, or at least one of the clinical symptoms of Covid-19. In some embodiments, the prophylactically effective amount prevents the onset or recurrence of Covid-19 infection entirely. "Inhibiting" the onset means either lessening the likelihood of the infection's onset or preventing the onset entirely. The term includes preventing the onset of the symptoms of the disorder in a subject at risk of developing the disorder.
  • Each dose form may, for example, comprise the colostrum equivalent of up to 20g per day but preferably less than 1200 mg (dry weight basis), preferably less than 800 mg, preferably less than 400 mg, more preferably less than 200 mg.
  • Colostrum equivalent refers to the amount of raw colostrum, howsoever purified, which is processed to provide the contents of a dose form, for example an oral, a nasal, or a pulmonary dose form.
  • the oral dose form may comprise 5 mg to 800 mg bovine colostrum powder (BCP) (dry weight basis), e.g. 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 125, 150, 175, 200, 225, 250, 500, 550, 600, 650, 700, 750 or 800 mg.
  • BCP bovine colostrum powder
  • Suitable dosage ranges are, e.g. from about 30mg to about 20000mg/day, preferably 50mg to about 5000mg/day, more preferably 500mg to about 5000mg/day, or most preferably 1500mg to about 2000mg/day BCP (dry weight basis). In one preferred embodiment, the dose is 1800mg/day BCP (dry weight basis).
  • the composition is formulated for administration at a dose of about 30 mg to about 10000 mg per day, or formulated for administration at a dose of about 1800 mg per day.
  • the composition is administered at a dose of about 30 mg to about 10000 mg per day, or administered at a dose of about 1800 mg per day.
  • the antibodies are present in the composition for administration in an amount sufficient to provide at least 30% by dry weight of the composition of IgG.
  • the oral dose form may comprise 1 .5 mg to 240 mg IgG, e.g. 1.5, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 4045, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 125, 150, 175, 200, 240, 400, 600, 800 or 1000 mg IgG.
  • the nasal dose form may comprise 1.5 mg to 240 mg IgG, e.g. 1.5, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 125, 150, 175, 200, 240, 400, 600, 800 or 1000 mg IgG.
  • the pulmonary dose form may comprise 1.5 mg to 240 mg IgG, e.g. 1.5, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 4045, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 125, 150, 175, 200, 240, 400, 600, 800 or 1000 mg IgG.
  • Suitable dosage ranges are, e.g. from about 10 to about 3333 mg/day, preferably 20 to about 4000 mg/day, more preferably 200 to about 2000mg/day, or most preferably 600 mg to about 800 mg/day IgG. In one preferred embodiment, the dose is 600 mg/day IgG.
  • the antibodies that bind to the antigen are present in the composition for administration in an amount sufficient to provide at least 5% specific IgG of the weight of IgG.
  • the oral dose form may comprise 0.075 mg to 12 mg specific IgG, e.g. 0.075, 0.1 , 0.125, 0.15, 0.175, 0.2, 0.4, 0.6, 0.8, 1 , 1.2,
  • the nasal dose form may comprise 0.075 mg to 12 mg specific IgG, e.g. 0.075, 0.1 , 0.125, 0.15, 0.175, 0.2, 0.4, 0.6, 0.8, 1 , 1.2, 1.4, 1.6, 1.8, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10.0, 11.0 or 12 mg specific IgG.
  • the pulmonary dose form may comprise 0.075 mg to 12 mg specific IgG, e.g. 0.075, 0.1 , 0.125, 0.15, 0.175, 0.2, 0.4, 0.6, 0.8, 1 , 1.2, 1.4, 1.6, 1.8, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0,
  • the antibodies that bind to the antigen are present in the composition for administration in an amount sufficient to provide at least 10% specific IgG of the weight of IgG.
  • Suitable dosage ranges are, e.g. from about 0.5 to about 167 mg/day, preferably 10 to about 150 mg/day, more preferably 15 to about 100mg/day, or most preferably 30 mg to about 100 mg/day specific IgG. In one preferred embodiment, the dose is 30 mg/day specific IgG.
  • the dose form may be administered for about 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30 or 31 days.
  • the composition is administered for 30 days.
  • the magnitude of prophylactic or therapeutic dose of the active ingredients can, of course, vary with the nature of the severity of the condition to be treated. It can also vary according to the age, weight and response of the individual patient, and may be administered in subject in single or divided doses. On the other hand, it may be necessary to use dosages outside the ranges provided herein in some cases.
  • the term “prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS” refers to bovine-hyperimmune colostrum prepared by administering to a bovine animal a vaccine comprising enterotoxigenic E. coli (ETEC) LPS and collecting colostrum from the bovine animal.
  • the hyperimmune colostrum is prepared by administering a bovine animal with a vaccine comprising ETEC LPS and collecting hyperimmune colostrum from the animal.
  • the hyperimmune colostrum is prepared by administering one bovine animal with a first vaccine and a second bovine animal with a second vaccine, and subsequently combining the hyperimmune colostrum collected from the animals.
  • the terms "antibody”, “antibodies” and the like include any monospecific molecule comprising a portion of the light chain variable region and/or the heavy chain variable region to effect binding to the epitope to which the antibody has binding specificity.
  • Exemplary antibodies and fragments thereof that may be prepared according to this aspect of the invention include intact immunoglobulin molecules, substantially intact immunoglobulin molecules and fragments that contain a paratope.
  • Fragments typically include a portion of an antibody molecule that retains the ability to specifically bind to an antigen (e.g., a SARS-CoV-2 and/or LPS antigen) and include, but are not limited to, Fab, Fab', F(ab')2 and F(v).
  • Antibody fragments may be obtained from antibodies such as described above by methods such as digestion by enzymes, such as pepsin or papain and/or by cleavage of the disulfide bridges by chemical reduction. Single chain antibodies are also intended to be encompassed within the term "fragment”.
  • the bovine hyperimmune colostrum comprises antibodies (e.g. “hyperimmune material-derived antibodies”) that bind to LPS.
  • the bovine hyperimmune colostrum comprises antibodies (e.g. “hyperimmune material-derived antibodies”) that bind to LPS, and antibodies (e.g. “hyperimmune material-derived antibodies”) that bind to a human coronavirus or antigen thereof.
  • the bovine hyperimmune colostrum comprises antibodies (e.g. “hyperimmune material-derived antibodies”) may comprise monomeric, dimeric or multimeric immunoglobulin selected from the group consisting of IgG, IgA and IgM and any fragments thereof.
  • antibodies e.g. “hyperimmune material-derived antibodies”
  • the principal compositional difference between colostrum and mature milk is the very high content of colostral immunoglobulin, of which IgG class makes up 80-90%.
  • the hyperimmune colostrum comprises one or more immunoglobulin classes selected from the group of IgG, IgM and IgA.
  • the hyperimmune material- derived antibodies mainly comprise IgG, specifically, lgG1 and lgG2.
  • IgG immunoglobulin G
  • IgG immunoglobulin G
  • lgG2a and lgG2b are the most pro-inflammatory IgG molecules showing a greater activity than mouse IgG 1 and lgG3 in many in vivo model systems.
  • hyperimmune material-derived antibodies may comprise a secretory antibody, specifically, slgA.
  • IgA and IgM are secreted by a number of exocrine tissues.
  • IgA is the predominant secretory immunoglobulin present in colostrum, milk, saliva, tears, bronchial secretions, nasal mucosa, prostatic fluid, vaginal secretions, and mucous secretions from the small intestine.
  • IgA output exceeds that of all other immunoglobulins, making it the major antibody produced by the body daily and is the major immunoglobulin found in human milk, whey and colostrum.
  • IgM secretion is less abundant but can increase to compensate for deficiencies in IgA secretion.
  • IgA J chain containing IgA is produced and secreted by plasma B immunocytes located in the lamina basement membrane of exocrine cells.
  • IgA has a typical immunoglobulin four-chain structure (Mr 160,000) made up of two heavy chains (Mr 55,000) and two light chains (Mr 23,000).
  • IgAI immunoglobulin four-chain structure
  • lgA2 two subclasses of IgA.
  • IgA can occur as monomers, dimers, trimers or multimers.
  • 10% of the total IgA is polymeric while the remaining 90% is monomeric.
  • the secreted IgA binds to a Mr 100,000 poly-lg receptor positioned in the basolateral surface of most mucosal cells.
  • the receptor-lgA complex is next translocated to the apical surface where IgA is secreted.
  • the binding of dimeric IgA to the poly-lg receptor is completely dependent upon the presence of a J chain. Monomeric IgA will not bind to the receptor.
  • IgG The difference in function of IgG and IgA, includes the position where the molecules operate. IgA is found mainly on mucosal surfaces where there is little in the way of tissue fluid to carry immune cells and chemicals. Therefore, IgA (often as a dimer) would be preferably used for physical neutralisation of pathogens and may be too effective at other immune functions. IgGs are present in the tissue fluid and blood where there is the full collection of leukocytes, complement system, macrophages etc. may physically neutralize a pathogen effectively and are also more effective in a communication/presentation role than IgA, i.e. , they tend to induce better opsonisation by phagocytes (e.g., Killer T cells and macrophages) and switch on the complement system better.
  • phagocytes e.g., Killer T cells and macrophages
  • the hyperimmune material-derived antibodies of the invention may be obtained from any one of colostrum, colostrum fractions hyperimmunised milk or colostrum, colostrum whey (either cheese or casein), cheese or casein whey, directly from skim milk, whole milk, or a reconstituted form of such streams.
  • the present invention provides a method of preparing hyperimmune colostrum that binds to human coronavirus, said method comprising a step of administering a bovine animal with a vaccine comprising ETEC LPS and collecting hyperimmune colostrum from the animal.
  • hyperimmune material within the composition of the invention may be any fraction of colostrum.
  • colostrum where used herein includes colostral milk, processed colostral-milk such as colostral milk processed to partly or completely removes one or more of fat, cellular debris, lactose and casein.
  • the colostrum, or milk, containing antibodies that bind to LPS and optionally, containing the antibodies that bind to a human coronavirus or antigen thereof, the antigen-specific antibodies may be preferably collected by milking the animal colostrum or milk thus collected can either be used directly, may be further processed, for instance to purify anti-antigen antibodies and optionally, antigen-specific antibodies. Methods for the (partial) purification of antibodies from colostrum or milk are present in the art.
  • Example 1 The present inventors have demonstrated in Example 1 the preparation of compositions comprising bovine-hyperimmune colostrum using a vaccine comprising enterotoxigenic LPS from ETEC strain B2C [serotype 06]; ETEC strain C55 3/3c3 [serotype 08]; ETEC strain PE 595 [serotype 015]; ETEC strain E11881 A [serotype 025]; ETEC strain C1064-77 [serotype 027]; ETEC strain PE 672 [serotype 063]; ETEC strain E20738/0 [serotype 0114]; ETEC strain PE 724 [serotype 0115]; ETEC strain El 37-2 [serotype 0128]; ETEC strain B7A [serotype 0148]; ETEC strain E8772/0 [serotype 0153]; and ETEC strain PE 768 [serotype 0159], and LPS extracted from ETEC strain H 10407 [078: H11]
  • the vaccine comprising ETEC LPS comprises one or more LPS 0 serotypes selected from the group consisting of 078, 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159.
  • the vaccine comprising ETEC LPS comprises LPS 0 serotypes 078, 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159.
  • LPS refers to Lipopolysaccharide, which comprises a lipid and a polysaccharide composed of O-antigen, core oligosaccharide (e.g. outer and inner core) joined by a covalent bond, and lipid A.
  • the vaccine comprising ETEC LPS comprises the lipid A, core and O-polysaccharide regions of the LPS.
  • compositions of the present invention are prepared using the methods described in PCT/AU2004/000277, hereby incorporated by reference in its entirely.
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic LPS comprise bovine immunoglobulin that binds to ETEC LPS.
  • the present invention provides a method as described herein wherein the composition comprising bovine-hyperimmune colostrum comprises bovine immunoglobulin that binds to ETEC LPS.
  • the present invention provides a method as described herein wherein the composition comprising bovine-hyperimmune colostrum comprises bovine immunoglobulin that binds to one or more LPS 0 serotypes selected from the group consisting of 078, 06, 08, 015, 025, 027, 063, 071, 0114, 0115, 0117, 0128, 0148, 0153, and 0159, 0167.
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic LPS from ETEC strain B2C [serotype 06]; ETEC strain C55 3/3c3 [serotype 08]; ETEC strain PE 595 [serotype 015]; ETEC strain E11881 A [serotype 025]; ETEC strain C1064-77 [serotype 027]; ETEC strain PE 672 [serotype 063]; ETEC strain E20738/0 [serotype 0114]; ETEC strain PE 724 [serotype 0115]; ETEC strain El 37-2 [serotype 0128]; ETEC strain B7A [serotype 0148]; ETEC strain E8772/0 [serotype 0153]; and ETEC strain PE 768 [serotype 0159], and LPS extracted from ETEC strain H10407 [078:H11] comprise bovine immunosorbent
  • the present invention provides a method as described herein wherein the composition comprising bovine-hyperimmune colostrum comprises bovine immunoglobulin that binds to one or more LPS serotypes of a gram negative bacteria selected from the group consisting of Enteropathogenic Escherichia coli strain E2348/69, Klebsiella pneumoniae strain ATCC 26, Pseudomonas aeruginosa strain ATCC 27853, Salmonella typhimurium strain ATCC 14028, Vibrio cholerae strain 6239, Yersinia enterocolitica strain 67R, C. jejuni, Vibrio cholera Ogawa, V. cholera Inaba, and V.
  • a gram negative bacteria selected from the group consisting of Enteropathogenic Escherichia coli strain E2348/69, Klebsiella pneumoniae strain ATCC 26, Pseudomonas aeruginosa strain ATCC 27853, Salmonella typhimurium strain ATCC
  • a bovine mammal is co-administered with more than one vaccine, for example a first vaccine comprising one or more LPS from a one or more strains of ETEC and a second vaccine comprising one or more LPS from a one or more strains of ETEC.
  • the present invention provides a method as described herein wherein the composition comprising bovine-hyperimmune colostrum is prepared using a first vaccine comprising ETEC LPS and a second vaccine comprising ETEC LPS.
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) inhibit SARS-CoV-2 replication, and inhibit SARS-CoV- 2 replication at doses that are not cytotoxic to cells.
  • ETEC enterotoxigenic E. coli
  • a bovine mammal infected or previously infected with bovine coronavirus is administered a vaccine comprising one or more LPS from a one or more strains of ETEC.
  • Bovine coronavirus BCoV is a widely distributed pathogen, causing disease and economic losses in the cattle industry worldwide.
  • the present inventors propose that administration of vaccine comprising one or more LPS from a one or more strains of ETEC to a bovine animal infected or previously infected with bovine coronavirus results in bovine-hyperimmune colostrum that can inhibit SARS-CoV-2 replication of human cells.
  • Cross-protection against a human enteric coronavirus and a virulent bovine enteric coronavirus in gnotobiotic calves has been demonstrated, and the present inventors propose administration of a vaccine comprising one or more LPS from a one or more strains of ETEC can enhance titres of anti-coronavirus antibodies.
  • a bovine mammal is co-administered with more than one vaccine, for example a first vaccine comprising one or more LPS from a one or more strains of ETEC and second vaccine comprising a coronavirus antigen and/or a coronavirus.
  • a first vaccine comprising one or more LPS from a one or more strains of ETEC
  • second vaccine comprising a coronavirus antigen and/or a coronavirus.
  • the present invention provides a method as described herein wherein the composition comprising bovine-hyperimmune colostrum is prepared using a first vaccine comprising ETEC LPS and a second vaccine comprising a coronavirus antigen and/or a coronavirus.
  • the composition comprising bovine-hyperimmune colostrum is prepared using a vaccine comprising ETEC LPS administered to an animal sequentially- or co-administered a vaccine comprising a coronavirus antigen and/or a coronavirus.
  • the vaccine comprising a coronavirus is an attenuated coronavirus.
  • composition comprising bovine-hyperimmune colostrum is prepared using a vaccine comprising ETEC LPS administered to an animal producing antibodies to coronavirus.
  • composition comprising bovine-hyperimmune colostrum comprises bovine immunoglobulin that binds to coronavirus.
  • the coronavirus antigen is a human or bovine coronavirus antigen.
  • the coronavirus is a human or bovine coronavirus.
  • any adjuvants may be added to the compositions of the invention.
  • Appropriate adjuvants therefore may be any antigen, antibody, glycosphingolipids, proteins, cytokines, adhesion molecules, and component that can activate or alter the function of antigen presenting cell or of any other cell related to the immune system in a direct and indirect manner.
  • antibodies that bind to LPS and/or antibodies that bind to a human coronavirus or antigen thereof may be an affinity purified antibody or any fragment thereof.
  • antibody is meant to include both intact molecules as well as fragments thereof, such as, for example, Fab and F(ab')2, which are capable of binding antigen.
  • Fab and F(ab')2 fragments lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody. It will be appreciated that Fab and F(ab')2 and other fragments of the antibodies useful in the present invention may be used for immuno-modulation, according to the methods disclosed herein for intact antibody molecules. Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
  • the composition of the invention may optionally further comprise colostrum component/s such as for example, alarmins, defensins, colostrinin, and any other colostrum or milk derived carbohydrates, glycolipids or any other molecules or components that may further enhance or inhibit modulation of an immune response, or any preparations, mixtures or combinations thereof.
  • the composition of the invention may comprise any additional adjuvant.
  • Appropriate adjuvants therefore may be any antigen, antibody, glycosphingolipids, proteins, cytokines, adhesion molecules, and component that can activate or alter the function of antigen presenting cell or of any other cell related to the immune system in a direct and indirect manner.
  • the hyperimmune colostrum further comprises a component selected from oligosaccharides, beta lactoglobulin, alpha-lactalbumin, lactoferrin, bovine serum albumin, growth factors, lactoperoxidase, plasm in, lipoprotein lipase, esterase, ribonucleases A, B, C, D and 11-1 , Lysozyme, enzyme inhibitors, cytokines, and lipids.
  • oligosaccharides selected from oligosaccharides, beta lactoglobulin, alpha-lactalbumin, lactoferrin, bovine serum albumin, growth factors, lactoperoxidase, plasm in, lipoprotein lipase, esterase, ribonucleases A, B, C, D and 11-1 , Lysozyme, enzyme inhibitors, cytokines, and lipids.
  • the hyperimmune colostrum further comprises an effective amount of a therapeutic agent, for example, an antiviral agent.
  • a therapeutic agent for example, an antiviral agent.
  • the effective amount of a composition comprising bovine-hyperimmune colostrum is administered to the subject orally, nasally or using pulmonary administration.
  • the composition is formulated for oral administration.
  • composition of the invention may be suitable for mucosal administration, for example, pulmonary, buccal, nasal, intranasal, sublingual, rectal, vaginal administration and any combination thereof.
  • any other route of administration may be applicable, for example, intravenous, intravenous, intramuscular, subcutaneous, intraperitoneal, parenteral, intravaginal, intranasal, mucosal, sublingual, topical, rectal or subcutaneous administration, or any combination thereof.
  • any of the compositions of the invention may be administered orally or by inhalation as an aerosol or by intravenous, intramuscular, subcutaneous, intraperitoneal, parenteral, transdermal, intravaginal, intranasal, mucosal, sublingual, topical, rectal or subcutaneous administration, or any combination thereof.
  • compositions described herein may be administered in dosage formulations containing conventional non-toxic acceptable carriers and may also include one or more acceptable additives, including acceptable salts, polymers, solvents, buffers, excipients, bulking agents, diluents, excipients, suspending agents, lubricating agents, adjuvants, vehicles, deliver systems, emulsifiers, disintegrants, absorbents, preservatives, surfactants, colorants, flavourants or sweeteners.
  • An optional dosage form of the present invention may be a powder for incorporation into beverages, pills, syrup, capsules, tablets, granules, beads, chewable lozenges or food additives, using techniques known in the art.
  • immuno-modulating composition of the invention may be administered in a form selected from the group consisting of orally-active powders, pills, capsules, teas, extracts, dried extracts, sublinguals, sprays, dispersions, solutions, suspensions, emulsions, foams, syrups, lotions, ointments, gels, pastes, dermal patches, injectables, creams and suppositories.
  • Therapeutic formulations may be administered in any conventional dosage formulation.
  • Formulations typically comprise at least one active ingredient, as defined above, together with one or more acceptable carriers thereof.
  • Each carrier should be both pharmaceutically and physiologically acceptable in the sense of being compatible with the other ingredients and not injurious to the patient.
  • Formulations include those suitable for oral, rectal, nasal, or parenteral (including subcutaneous, intramuscular, intravenous and intradermal or by inhalation) administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The nature, availability and sources, and the administration of all such compounds including the effective amounts necessary to produce desirable effects in a subject are well known in the art and need not be further described herein.
  • compositions are well known in the art and has been described in many articles and textbooks, see e.g., Remington's Pharmaceutical Sciences, Gennaro A. R. ed., Mack Publishing Co., Easton, PA, 1990, and especially pp. 1521-1712 therein, fully incorporated herein by reference.
  • compositions of the invention generally comprise a buffering agent, an agent that adjusts the osmolarity thereof, and optionally, one or more pharmaceutically acceptable carriers, excipients and/or additives as known in the art.
  • Supplementary active ingredients can also be incorporated into the compositions.
  • the carrier can be solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents and the like.
  • the use of such media and agents for pharmaceutical active substances is well known in the art. Except as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic composition is contemplated.
  • the active components can be combined with a non-toxic pharmaceutically acceptable inert carrier such as lactose, starch, sucrose, glucose, modified sugars, modified starches, methylcellulose and its derivatives, dicalcium phosphate, calcium sulfate, mannitol, sorbitol, and other reducing and non-reducing sugars, magnesium stearate, stearic acid, sodium stearyl fumarate, glyceryl behenate, calcium stearate and the like.
  • a non-toxic pharmaceutically acceptable inert carriers such as ethanol, glycerol, water and the like.
  • suitable binders, lubricants, disintegrating agents and colouring and flavouring agents can also be incorporated into the mixture.
  • Stabilizing agents such as antioxidants, propyl gallate, sodium ascorbate, citric acid, calcium metabisulphite, hydroquinone, and 7-hydroxycoumarin can also be added to stabilize the dosage forms.
  • Other suitable compounds can include gelatin, sweeteners, natural and synthetic gums such as acacia, tragacanth, or alginates, carboxymethylcellulose, polyethylene, glycol, waxes and the like.
  • the present invention provides a method of treating and/or preventing coronavirus infection in one or more human subjects comprising applying to a surface selected from air filters, PPE, room surfaces and respiratory mucosal membranes an effective amount of a composition comprising bovine- hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the surface is a filter on which the immune material is adsorbed.
  • the filter may be used to inhibit infection of people with SARS-CoV-2 which may be present in the airstream.
  • the antibodies or fragments are dissolved or dispersed in a liquid which is then used in a nebulising device or to provide an antibody aerosol which is released into the atmosphere of a hospital ward or sick room or other atmosphere containing SARS-CoV-2 virus.
  • the antibody aerosol is directed into an air stream comprising polluted air which is then passed over or through a surface, for example a porous surface or a hairy surface or some other surface having an extended surface area.
  • a surface for example a porous surface or a hairy surface or some other surface having an extended surface area.
  • the role of the surface is to capture both SARS-CoV-2 viruses or virions and also antibody moieties, leading to at least partial neutralisation of the polluted air stream.
  • the antibodies or fragments are deposited on the surface of a face mask, or within the fabric of a face mask.
  • the personal protective equipment and air filters include marks or personal air filters such as may be worn over the nose and mouth of the subjects or the filters may be building or air conditioner filters in buildings inhabited by the subjects.
  • the surface is any surface in a room frequented by subjects with Covid-19, for example a hospital room.
  • composition comprising bovine-hyperimmune colostrum is adsorbed on a particulate carrier of a size in the range from 50 to 120 microns
  • composition comprising bovine-hyperimmune colostrum is adsorbed onto a solid carrier for administration by dry powder inhaler.
  • the antibodies or fragments thereof are provided in the form of an inhalational dose.
  • composition comprising bovine-hyperimmune colostrum is administered as an aerosol.
  • composition comprising bovine-hyperimmune colostrum in dried form.
  • compositions may be provided in milled particulate form of the immune material wherein the average particle size is less than 20 microns, preferably less than 10 microns, more preferably less than 5 microns. Once small particles have been produced, the micronized substance may be blended with an excipient.
  • suitable carriers may include one or more carbohydrate, such as fructose, glucose, galactose, sucrose, lactose, trehalose, raffinose, melezitose; alditols, such as mannitol and xylitol; maltodextrins, dextrans, cyclodextrins, amino acids, such as glycine, arginine, lysine, aspartic acid, glutamic acid and polypeptides, such as human serum albumin and gelatin.
  • flavouring particles containing maltodextrin and peppermint oil may be incorporated into dry powder formulations. Large sized particles increase mouth deposition and reduce lung deposition. Lactose is a particularly preferred carrier.
  • the carrier particles are typically relatively large such as on the order of 50 to 120 microns, or approximately 50 times bigger than the milled particles containing the bovine hyperimmune material. These carrier particles help to facilitate the dispersion of the small particles and allow precise filling into the dry powder inhaler (DPI) powder storage system in a reproducible manner.
  • Milled bovine hyperimmune colostrum is blended with lactose at concentrations ranging from ⁇ 1 to 50% by weight. Finally, the blend is filled into the powder storage systems of an inhaler at weights ranging from approximately 3-25 mg. Preferred powder storage systems are shown in U.S. Pat. Nos.
  • Size reduction may be accomplished by a variety of techniques including spray drying, precipitation from supercritical fluids, and jet milling or micronization. Preferably, jet milling is used. This technique uses high pressure, high velocity gas to cause particle to particle attrition to generate small particles at high efficiencies.
  • Multidose DPIs may be used with disposable cassettes or foil blister disks, strips or unit dose blisters to deliver many doses, contributing to the cost effectiveness of this approach as compared with syringes, particularly single use syringes. These DPI's may be provided with disposable mouthpieces that can be used in mass dosing campaigns. Alternatively, unit dose DPIs with vaccine sealed in the aerosolization chamber can be used.
  • the inhaler may have a body, a mouthpiece and an airflow passage.
  • a restrictor plate may be used having flow control openings in the airflow passage opposite from the mouthpiece.
  • a dose of dry powder vaccine sealed by a foil strip or capsule is received within the body of the inhaler.
  • the foil strip or capsule is pulled out or back, peeling or breaking open blister formed around the dose or a capsule is received in a chamber provided with means such as a sharp object for piercing the capsule to release the powder which may occur in response to actuation by the user to urge the sharp object into the capsule or blister by means of a trigger lever, button or the like.
  • the subject inhales on the mouthpiece, and the dose is drawn into the lungs.
  • the inhaler may be provided with means for actively generating an air flow in response to actuation by manual operation of the user or commencement of the inhalation process by the user.
  • composition is administered so as to come in contact with an airway surface of a human subject in the upper respiratory tract, preferably an airway surface within 3 or 4 branch points of the trachea.
  • the composition optionally further comprises colostrum, milk or milk products component/s and any adjuvant/s, preferably, alarmins, defensins, colostrinin and any preparation, mixture or combination thereof.
  • the composition of the invention may comprise any additional adjuvant.
  • Appropriate adjuvants therefore may be any antigen, antibody, glycosphingolipids, proteins, cytokines, adhesion molecules, and component that can activate or alter the function of antigen presenting cell or of any other cell related to the immune system in a direct and indirect manner.
  • the present invention further provides the use of colostrum or any colostrum-derived preparations in the combined compositions of the invention for enhancing an immunomodulatory effect of an immunomodulatory therapeutic agent.
  • alarmin denotes an array of structurally diverse multifunctional host proteins that are rapidly released during infection or tissue damage, and that have mobilizing and activating effects on receptor-expressing cells engaged in host defence and tissue repair.
  • Innate-immune mediators that have alarmin function include defensins, eosinophil-derived neurotoxin, cathelicidins and HMGB1.
  • Defensins are small (15-20 residue) cysteine-rich cationic proteins found in both vertebrates and invertebrates. They are active against bacteria, fungi and enveloped viruses. They consist of 15-20 amino acids including six to eight conserved cysteine residues.
  • Cells of the immune system contain these peptides to assist in killing phagocytized bacteria, for example in neutrophil granulocytes and almost all epithelial cells. Most defensins function by penetrating the microbes cell membrane by way of electrical attraction, and once embedded, forming a pore in the membrane which allows efflux.
  • Colostrinin refers to a polypeptide which, in its natural form, is obtained from mammalian colostrum. Colostrinin is sometimes known as “colostrinine", and has a molecular weight in the range 16,000 to 26,000 Daltons. Colostrinin may form a dimer or trimer of sub-units (each having a molecular weight in the range 5,000 to 10,000 Daltons, preferably 6,000 Daltons), and contains mostly proline (the amount of proline is greater than the amount of any other single amino acid).
  • Colostrinin is characterized in that it stimulates the production of cytokines, especially gamma interferon (IFN-y), tumor necrosis factor TNF-a), interleukins (e.g. IL-6 and IL-10) and various growth factors.
  • cytokines especially gamma interferon (IFN-y), tumor necrosis factor TNF-a), interleukins (e.g. IL-6 and IL-10) and various growth factors.
  • ARDS Acute respiratory distress syndrome
  • ARDS pathogenesis involves inflammatory injury to the alveolo-capillary membrane, which results in increased lung permeability and the exudation of protein- rich pulmonary edema fluid into the airspaces, leading in the end to respiratory insufficiency.
  • increased circulating levels of pro-inflammatory cytokines eg, Interferon y, interleukin (IL-) 1 B, IL-6, IL-12
  • chemokines CXCL10, and CCL2
  • Th2-immune-oriented cytokines such as IL-4 and IL-10, whose main effect is to suppress inflammation.
  • ARDS is the ultimate result of a cytokine storm.
  • the release by immune effector cells of large amounts of pro-inflammatory cytokines IFNa, IFNy, IL-1 b, IL-6, IL-12, IL-18, IL-33, TNFa, TGFP
  • chemokines CXCL10, CXCL8, CXCL9, CCL2, CCL3, CCL5
  • the cytokine storm is readily followed by the immune system “attacking” the body, which in turn will cause ARDS and multiple organ failure, the final result being death, at least in the most severe cases of SARS-CoV-2 infection.
  • LPS Lipopolysaccharide
  • Example 4 The present inventors have demonstrated in Example 4 that the bovine hyperimmune colostrum compositions described herein can decrease levels of LPS in the serum of patients with inflammation. Without wishing to be bound by theory, the present inventors propose that the bovine hyperimmune colostrum compositions described herein can be used to treat and/or prevent one or more symptoms associated with Covid-19, including cytokine storm.
  • the present invention provides methods of decreasing levels of LPS in the blood of a subject comprising administering to the subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides methods of treating and/or preventing sepsis in a subject comprising administering to the subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides methods of decreasing inflammation in a subject comprising administering to the subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • the present invention provides methods of treating and/or preventing cytokine storm in a subject comprising administering to the subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the cytokine storm is LPS-associated cytokine storm.
  • the subject is a patient infected with a coronavirus.
  • the coronavirus is SARS-Cov-2.
  • decreasing toxic cytokine release or toxic cytokine levels comprises decreasing or inhibiting production of toxic cytokine levels in a subject, or inhibiting or reducing the incidence of cytokine release syndrome or a cytokine storm in a subject by decreasing LPS in the serum of the subject.
  • toxic cytokine levels are reduced during cytokine storm.
  • decreasing or inhibiting the production of toxic cytokine levels comprises treating cytokine storm.
  • decreasing or inhibiting the production of toxic cytokine levels comprises preventing cytokine storm.
  • decreasing or inhibiting the production of toxic cytokine levels comprises alleviating a cytokine storm.
  • decreasing or inhibiting the production of toxic cytokine levels comprises ameliorating a cytokine storm.
  • the toxic cytokines comprise pro-inflammatory cytokines.
  • pro-inflammatory cytokines comprise IL-6.
  • pro inflammatory cytokines comprise IL-1.
  • pro-inflammatory cytokines comprise TNF-a.
  • pro-inflammatory cytokines comprise IL-6, IL- 13, or TNF-a, or any combination thereof.
  • cytokine storm is characterized by elevated levels of several inflammatory cytokines and adverse physical reactions in a subject such as low blood pressure, high fever and shivering.
  • inflammatory cytokines comprise IL-6, IL-1 , and TNF-a.
  • cytokine storm is characterized by elevated levels of IL-6, IL- 13, or TNF-a, or any combination thereof.
  • cytokine storm is characterized by elevated levels of IL-8, or IL-13, or any combination thereof.
  • a cytokine storm is characterized by increases in TNF-alpha, IFN-gamma, IL-lbeta, IL-2, IL-6, IL-8, IL- 10, IL-13, GM CSF, IL-5, CX3CL1 , or a combination thereof or a subset thereof.
  • IL-6 comprises a marker of cytokine storm.
  • IFN-y comprises a marker of cytokine storm.
  • patients with coronavirus infection have higher incidence and severity of cytokine storm.
  • bovine hyperimmune colostrum compositions described herein can decrease markers of inflammation in human subjects.
  • the present inventors propose that the bovine hyperimmune colostrum compositions described herein can be used to treat and/or prevent one or more symptoms associated with inflammation, such as immune pathology associated with CARS-CoV-2 infection.
  • the present invention provides methods of treating and/or preventing a symptom associated with coronavirus infection. in a subject comprising administering to the subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the symptom associated with coronavirus infection is a cytokine storm, LPS-associated cytokine storm, inflammation, pathologic change, LPS in the blood, elevated ALT, ASL levels.
  • the bovine hyperimmune colostrum compositions described herein can decrease AST and ALT levels in patents with inflammation.
  • the compositions of the invention significantly decreased the serum levels of ALT and AST. Therefore, according to one embodiment, the composition of the invention leads to at least one of a decrease in the serum levels of ALT and AST in a subject with inflammation.
  • Decrease, reduction, inhibition, as used herein refers to a reduction of about 5% to 99% of the serum level of ALT or AST in a subject.
  • such reduction may be a reduction of about, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% and over 99%, as compared to the levels prior to the treatment, or the levels of untreated control.
  • the pathologic change is gastrointestinal tract pathology, or lung pathology.
  • the inflammation is systemic inflammation, inflammation in the lungs or inflammation in the liver.
  • the subject is a patient infected with a coronavirus.
  • the coronavirus is SARS-Cov-2.
  • the subject is an elderly subject.
  • cytokine includes cytokines (e.g., interferon gamma, granulocyte macrophage colony stimulating factor, tumor necrosis factor alpha), chemokines (e.g., MIP 1 alpha, MIP 1 beta, RANTES), and other soluble mediators of inflammation, such as reactive oxygen species and nitric oxide.
  • cytokines e.g., interferon gamma, granulocyte macrophage colony stimulating factor, tumor necrosis factor alpha
  • chemokines e.g., MIP 1 alpha, MIP 1 beta, RANTES
  • other soluble mediators of inflammation such as reactive oxygen species and nitric oxide.
  • cytokine storm is characterized by any or all of the following symptoms: fever with or without rigors, malaise, fatigue, anorexia, myalgias, arthalgias, nausea, vomiting, headache, skin rash, nausea, vomiting, diarrhea, Tachypnea, hypoxemia Cardiovascular Tachycardia, widened pulse pressure, hypotension, increased cardiac output (early), potentially diminished cardiac output (late), Elevated D-dimer, hypofibrinogenemia with or without bleeding, Azotemia Hepatic Transaminitis, hyperbilirubinemia, mental status changes, confusion, delirium, word finding difficulty or frank aphasia, hallucinations, tremor, dysmetria, altered gait, seizures.
  • a cytokine storm is characterized by IL-2 release and lymphoproliferation.
  • cytokine storm leads to potentially life-threatening complications including cardiac dysfunction, adult respiratory distress syndrome, neurologic toxicity, renal and/or hepatic failure, and disseminated intravascular coagulation.
  • measurement of cytokine levels or concentration, as an indicator of cytokine storm may be expressed as -fold increase, percent (%) increase, net increase or rate of change in cytokine levels or concentration.
  • absolute cytokine levels or concentrations above a certain level or concentration may be an indication of a subject undergoing or about to experience a cytokine storm.
  • absolute cytokine levels or concentration at a certain level or concentration for example a level or concentration normally found in a control subject not undergoing CAR-T cell therapy, may be an indication of a method for inhibiting or reducing the incidence of a cytokine storm in a subject undergoing CAR T-cell.
  • cytokine level may encompass a measure of concentration, a measure of fold change, a measure of percent (%) change, or a measure of rate change.
  • cytokine level may encompass a measure of concentration, a measure of fold change, a measure of percent (%) change, or a measure of rate change.
  • IL-6 levels may be used as a common measure of cytokine storm and/or as a common measure of the effectiveness of a treatment for cytokine storms.
  • cytokines may be used as markers of a cytokine storm, for example any of TNF-a, IL- 6, IL-8, IL-13, or INF-y, or any combination above may be used as a marker a cytokine storm.
  • assay methods for measuring cytokines are well known in the art.
  • treatment refers to the reduction or elimination of the severity of a symptom of the disease, the frequency with which such a symptom is exhibited, or both.
  • the term includes an action that occurs while a patient is suffering from Covid-19 or associated disorder that reduces the severity of one or more symptoms or effects of Covid-19 or a Covid-19 associated disease or symptom.
  • prophylaxis refers to completely or partially preventing or inhibiting a symptom of the disease or the frequency with which such a symptom is exhibited, or a reduction of the risk of acquiring a given symptom, of Covid- 19.
  • Prophylaxis includes inhibiting Covid-19 associated diarrhea, preventing Covid- 19 associated diarrhea, decreasing the severity of Covid-19 associated diarrhea or improving signs and symptoms related to having Covid-19 associated diarrhea.
  • Prophylaxis includes inhibiting, preventing, or decreasing the severity a symptom of Covid-19 as described herein.
  • the term includes action that occurs before a patient begins to suffer from Covid-19 or associated disorder, such as but not limited to bowel or gastrointestinal disorder, that delays the onset of, and/or inhibits or reduces the severity of Covid-19 or Covid-19 associated disease or symptom.
  • Covid-19 or associated disorder such as but not limited to bowel or gastrointestinal disorder, that delays the onset of, and/or inhibits or reduces the severity of Covid-19 or Covid-19 associated disease or symptom.
  • the present invention provides a method as described herein wherein a symptom of coronavirus infection is treated and/or prevented.
  • SARS-Cov-2 primarily causes lung infection through binding of ACE2 receptors present on the alveolar epithelial cells, yet it was recently reported that SARS-CoV-2 RNA was found in the faeces of infected patients. Interestingly, the intestinal epithelial cells particularly the enterocytes of the small intestine also express ACE2 receptors. The role of the gut microbiota in influencing lung disease is well known.
  • SARS-CoV-2 is shed in the faeces of infected individuals. Reducing SARS- CoV-2 shedding includes decreasing the number of SARS-CoV-2 virions in the faeces of an infected individual.
  • the at least one symptom of Covid-19 is selected from the group consisting of diarrhea, abdominal pain, bowel pain, fever, loss of appetite, SARS-CoV-2 viral shedding, nausea, vomiting, and Covid-19 associated mortality.
  • ACE2 mainly enters human cells by binding to the receptor angiotensin converting enzyme 2 (ACE2) also expressed by gastrointestinal epithelial cells. Accordingly, in another embodiment, the at least one symptom of Covid-19 is SARS-CoV-2 infection in the gastrointestinal tract.
  • ACE2 receptor angiotensin converting enzyme 2
  • compositions comprising enterotoxigenic E. coli (ETEC) that inhibit SARS-CoV-2 replication improve cell viability.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of improving cell viability comprising contacting one or more human cells with an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of maintaining gastrointestinal cell viability comprising contacting one or more human cells with an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • the contacting is performed in vivo.
  • the present invention provides a method of improving cell viability in a subject comprising to a subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of maintaining gastrointestinal cell viability in a subject comprising to a subject an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the subject is a subject with a SARS-CoV-2 infection.
  • the present inventors propose that infection of gastrointestinal epithelial cells with SARS-CoV-2 and the resultant pathology allows secondary bacterial infections in the gastrointestinal tract to flourish. Furthermore, the extensive use of antibiotics for acute respiratory infection can cause a loss of gastrointestinal flora resulting in an increased susceptibility to secondary bacterial infections in the gastrointestinal tract.
  • the at least one symptom of Covid-19 is loss of gastrointestinal flora secondary to SARS-CoV-2 infection.
  • the at least one symptom of Covid-19 is bacterial sepsis associated with SARS-CoV-2 infection or bacterial sepsis secondary to SARS- CoV-2 infection.
  • the at least one symptom of Covid-19 is cytokine storm associated with SARS-CoV-2 infection or increased severity of a gastrointestinal disease associated with to SARS-CoV-2 infection.
  • the present invention provides a method of reducing viral shedding from a subject infected with a human coronavirus comprising administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • the present invention provides a method of treating and/or preventing human coronavirus-associated diarrhoea in a subject comprising administering an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the present invention provides a method of inhibiting coronavirus mediated cytotoxicity comprising contacting one or more human cells with an effective amount of a composition comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS.
  • ETEC enterotoxigenic E. coli
  • the contacting is performed in vivo in a human.
  • the symptom of coronavirus infection is selected from the group consisting of nausea, vomiting and diarrhoea.
  • the symptom of coronavirus infection is the load of coronavirus in stool.
  • the symptom of coronavirus infection is bowel pain.
  • the present disclosure is illustrated by the following non-limiting Examples.
  • Example 1 Bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS binds to other pathogens.
  • ETEC enterotoxigenic E. coli
  • Bovine-hyperimmune colostrum is prepared using the methods described in PCT/AU2004/000277, hereby incorporated by reference in its entirely.
  • a composition comprising bovine-hyperimmune colostrum is prepared by immunising cows with a vaccine comprising ETEC LPS of LPS O serotypes 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159, and a vaccine comprising 078, wherein the LPS comprises the lipid A, core and O-polysaccharide regions of the LPS.
  • Cytokines / hormones / growth factors detected in the composition comprising bovine-hyperimmune colostrum is prepared by immunising cows with a vaccine comprising ETEC LPS of LPS 0 serotypes 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159, and a vaccine comprising 078, wherein the LPS comprises the lipid A, core and O-polysaccharide regions of the LPS by ELISA included in batches tested TNF-alpha, IGF-1 , Lactoferrin, and Bovine Growth Hormone, with lgG1 making up between 65% and 90% of total immunoglobulins, TGFP2 in the range of 0.3 - 0.7 pg/gl, and lactoferrin in the range of 1.4 - 3.8 mg/g.
  • Figure 1A shows LPS extracted from ETEC strains in the ETEC vaccine, run on 15% Tris-tricine SDS-PAGE and stained using LPS specific silver stain. O-polysaccharide chains are shown as ladders and lipid A core region is marked with an arrow.
  • Figure 1 B shows western transfer to PVDF membrane and blotted with Bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS, made from a pool of colostrum from over 1000 cows.
  • ETEC enterotoxigenic E. coli
  • hyperimmune colostrum from cattle immunised with LPS 0 serotypes 078, 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159 comprises bovine immunoglobulin that binds LPS 0 serotypes selected from the group consisting of 078, 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159, and lipid A cores of 078, 06, 08, 015, 025, 027, 063, 0114, 0115, 0128, 0148, 0153 and 0159 LPS.
  • FIG. 2B shows western transfer to PVDF membrane and blotted with Bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS, made from a pool of colostrum from over 1000 cows.
  • ETEC enterotoxigenic E. coli
  • Figure 3A shows a silver stained 15% Tris-tricine gel showing LPS extracted from pathogens not included in the ETEC vaccine. Shown are LPS extracted from 2 - Enterobacter aerogenes strain ATCC 13048; 3 - Enteropathogenic Escherichia coli strain E2348/69; 4 - Klebsiella pneumoniae strain ATCC 26; 5 - Pseudomonas aeruginosa strain ATCC 27853; 6 - Salmonella typhimurium strain ATCC 14028; 7 - Vibrio cholerae strain 6239; 8 - Yersinia enterocolitica strain 67R; 9 - Citrobacter rodentium strain DSB100; and 10 - ETEC strain E11881A [025: H42] (+ control)
  • Figure 3B shows western transfer to PVDF membrane and blotted with Bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS, made from a pool of colostrum from over 1000 cows.
  • ETEC enterotoxigenic E. coli
  • HAI Hemagglutination Inhibition Assay with ETEC of the colonization factor (CF) types above was performed.
  • the IgG adheres to the CF of the ETEC strain preventing bovine MRHA.
  • the bovine- hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS exhibited the strongest inhibitory effect of MRHA by ETEC expressing CFA/I, CS2, and CS3 as shown in the Table 2.
  • the HAI titer is the dilution of the the bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E.
  • ETEC ETEC LPS stock solution (140 mg/ml) which inhibited MRHA by the ETEC strain.
  • the 140 mg/ml stock concentration corresponds to the concentration of 50 mg/ml IgG in the the bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS colostrum.
  • the inhibitory concentration of the bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS IgG refers to the amount of IgG needed to bind to the CF to prevent adherence of the bacteria to the bovine red blood cells resulting in no agglutination.
  • Promilk 85 a product containing powdered milk proteins, was used as a negative control at the 140 mg/ml concentration. Promilk did not inhibit hemagglutination by any of the ETEC strains tested.
  • Table 2 Hemagglutination titers and inhibitory concentrations resulting from the bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS combined with ETEC expressing common CF types.
  • ETEC enterotoxigenic E. coli
  • the ETEC bacterial suspensions used in the HAI were electrophoresed on SDS PAGE and immunoblotted.
  • the blots were immunodetected with the bovine- hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS also or Promilk 85.
  • Promilk is a negative control containing powdered milk proteins.
  • the bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS colostrum reacted with many whole cell lysate proteins in each of the HAI strains tested.
  • ETEC enterotoxigenic E. coli
  • the immune-reactive protein bands are similar among the 4 isolates of S. boydii; two prominent immune-reactive bands are uniquely shown between 37 - 75 kD. Unlike others, the isolate# JH13-575 (S. boydii 18) has an additional prominent band between 37 - 50 kD.
  • the western blot immune-reactive bands of S. boydii isolates from Thailand and Bhutan are similar. For S. dysenteriae, the immune-reactive protein bands are slightly different among the 3 isolates of S.
  • dysenteriae one prominent immune-reactive band between 50 - 75 kD is uniquely shown in all isolates and the prominent bands between 37 - 50 kD have different immune-reactive pattern among the 3 isolates, and two bands are detected in the isolates# KRC-131 (S. dysenteriae-4) and JPC13-737 (S. dysenteriae-12).
  • the immune-reactive protein bands are similar among the 15 isolates of S. flexneri.
  • Two prominent immune-reactive bands are shown between 50 - 75 kD and 37 - 50 kD.
  • the isolate# TBFI-01-0001 (S. flexneri 2a) has an additional prominent immune-reactive band at 37 kD.
  • the western blot immune-reactive bands of 15 S. flexneri isolates from Bhutan, Cambodia, Nepal and Thailand are similar. All isolates of S. flexneri 3a, S. flexneri 4, S. flexneri 4av, and S. flexneri 6 have two prominent immune-reactive bands shown between 50 - 75 kD. For S. sonnei, all S. sonnei isolates showed similar pattern of immunoreactive bands. There is one prominent band between 37 to 50 kD, and several bands between 50 to 75 with one prominent band.
  • Example 2 Bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS inhibits SARS-CoV-2 viral replication.
  • ETEC enterotoxigenic E. coli
  • In-vitro susceptibility of viruses to an antiviral agent is usually assessed using a quantitative assay to measure virus replication in the presence of increasing concentrations of the product compared to replication in the absence of the product.
  • the effective concentration is the concentration of product at which virus replication is inhibited by 50 percent (EC50 for cell-based assays).
  • Assays that evaluate antiviral activity include, but are not limited to, virus inactivation assays, plaque reduction assays, cytopathic effect inhibition assays, peripheral blood mononuclear cell (PBMC) assays, and binding and fusion assays.
  • bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) LPS to inhibit SARS-CoV-2 replication was examined.
  • ETEC enterotoxigenic E. coli
  • five batches of colostrum powder were prepared as per Example 1 , and these bovine-hyperimmune colostrum preparations (“Test Compounds”) were tested in a cytopathic effect inhibition assay against SARS-CoV-2 hCoV- 19/AustraliaA/IC01/2020 according to the following protocol.
  • SARS-CoV-2 hCoV-19/Australia/VIC01/2020 was a gift from Melbourne’s Peter Doherty Institute for Infection and Immunity (Melbourne, Australia). Documentation received with the parent stock indicated that prior to receipt, the virus had been passaged as follows: two passage in Vero cells. A working stock was generated at 360biolabs by two further passages in Vero cells in virus growth media, which comprised Minimal Essential Medium without L-glutamine supplemented with 1 % (w/v) L-glutamine 1.0pg/mL of TPCK-Trypsin, 0.2% BSA, 1x Pen/Strep, and 1 % Insulin Transferrin Selenium (ITS).
  • Minimal Essential Medium without L-glutamine supplemented with 1 % (w/v) L-glutamine 1.0pg/mL of TPCK-Trypsin, 0.2% BSA, 1x Pen/Strep, and 1 % Insulin Transferrin Selenium (ITS).
  • African Green Monkey Kidney (Vero E6) cells (ATCC-CRL1586) were sub cultured to generate cell bank stocks in cell growth medium, which comprised Minimal Essential Medium without L-glutamine supplemented with 10% (v/v) heat-inactivated Fetal Bovine Serum and 1 % (w/v) L-glutamine. Cell stocks were frozen at -80°C overnight and then transferred to liquid nitrogen.
  • Vero E6 cells were passaged for a maximum of 13 passages, after which a new working cell bank stock was retrieved from liquid nitrogen for further use.
  • Vero cells were seeded into 96-well plates at 2x104 cells/well in 100pL seeding media (Minimal Essential Medium supplemented with 1 % (w/v) L-glutamine, 1 % ITS, 0.2% BSA). Plates were incubated overnight at 37°C, 5% C02.
  • seeding media Minimal Essential Medium supplemented with 1 % (w/v) L-glutamine, 1 % ITS, 0.2% BSA. Plates were incubated overnight at 37°C, 5% C02.
  • Products were prepared on the day of assay. Powders were dissolved in PBS (pH 7.4), with shaking at room temperature for 2 hours. Supernatants were subsequently collected after centrifugation at 4,000 X g for 10 minutes and used in the assay.
  • Remdesivir was prepared as a 10mM stock in DMSO and stored at -20°C.
  • a DMSO dilution series of Remdesivir was generated by addition of 15pL (1 OmM) compound to column 3 and 10pL DMSO to columns 2, 4-11 .
  • Remdesivir was serially diluted 1 :3 by transfer of 5pL compound from column 3 to column 4, column 4 to column 5 and continued to column 10 and then discarded.
  • An intermediate dilution series was prepared by addition of 4pL from columns 2-11 to 496pL virus growth media.
  • a 50pL volume from the intermediate dilution series was added to rows B-G of the assay control plate containing pre-seeded cells in 100pL seeding media. Virus was then added as outlined below.
  • viable cells were determined by staining with MTT.
  • a 100pL volume of a 3mg/ml_ solution of MTT was added to plates and incubated for 4 hours at 37°C in a 5% C02 incubator.
  • Wells were aspirated to dryness using a multichannel manifold attached to a vacuum chamber and formazan crystals solubilised by the addition of 200pL 100% 2-Propanol at room temperature for 30 minutes. Absorbance was measured at 540 - 650nm on a plate reader.
  • Percent cell protection ([ODt]virus - [ODc]virus / [ODc]mock - [ODc]virus) x 100
  • [ODt]virus the optical density measured in a well examining the effect of a given concentration of test article or positive control on virus-infected cells.
  • [ODc]virus the optical density measured in a well examining the effect of the negative control on virus-infected cells.
  • EC50 values were calculated from the percent cell protection results by non-linear regression analysis using the Hill (sigmoid Emax) formula as shown below:
  • x test or control article concentration
  • y percent cell protection
  • Min minimum
  • Max maximum
  • D slope coefficient
  • the 50% cytotoxic concentration is defined as the concentration of the test compound that reduces the absorbance of the mock infected cells by 50% of the control value.
  • the CC50 value was calculated as the ratio of (ODt)mock/(ODc)mock.
  • IDBS XLFit4 Excel Add-in (ID Business Solutions Inc., Alameda, CA) was used to perform the above calculations.
  • FIG 4 shows hyperimmune colostrum powder Batch # 1710002659 had an EC50 of 44.7 ug/ml and a CC50 of 1085.3 ug/ml.
  • This batch of colostrum inhibits viral replication at a dose at which there is no cell toxicity, and at a dose that inhibits 90% of viral replication there is no cell toxicity (an EC90 of 53.15 ug/ml).
  • Figure 5 shows hyperimmune colostrum powder Batch # 1710002660had an EC50 of 91.9 7 ug/ml and a CC50 of 1790.6 ug/ml.
  • This batch of colostrum inhibits viral replication at a dose at which there is no cell toxicity, and at a dose that inhibits 90% of viral replication there is no cell toxicity (an EC90 of 155.39 ug/ml).
  • FIG. 6 shows hyperimmune colostrum powder Batch # 1710002662 had an EC50 of 40.5 7 ug/ml and a CC50 of 2053.3 ug/ml. This batch of colostrum inhibits viral replication at a dose at which there is no cell toxicity, and at a dose that inhibits 90% of viral replication there is no cell toxicity (an EC90 of 45.17 ug/ml).
  • Figure 7 shows hyperimmune colostrum powder Batch # 1710002859 had an EC50 of 42.6 7 ug/ml and a CC50 of 2506.6 ug/ml. This batch of colostrum inhibits viral replication at a dose at which there is no cell toxicity, and at a dose that inhibits 90% of viral replication there is no cell toxicity (an EC90 of 48.69 ug/ml).
  • Figure 8 shows pro-milk powder Batch # 6068 N50 (control) had an EC50 of > the CC50, and a CC50 of 4818.6 ug/ml. Accordingly, this control milk powder inhibits viral replication at a dose >25000 ug/ml, and importantly does not inhibit viral replication at the doses at which it is cytotoxic to cells.
  • Figure 9 shows the positive control, remdesivir, has an EC50 of 1 16pM.
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) inhibit SARS-CoV-2 replication, and inhibit SARS-CoV-2 replication at doses that are not cytotoxic to cells.
  • ETEC enterotoxigenic E. coli
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) that inhibit SARS-CoV-2 replication.
  • ETEC enterotoxigenic E. coli
  • Figure 4 shows hyperimmune colostrum powder Batch # 1710002659 improved cell viability by 260% relative to control, at 102.8 ug/ml.
  • Figure 5 shows hyperimmune colostrum powder Batch # 1710002660 improved cell viability by 212% relative to control, at 102.8 ug/ml.
  • Figure 6 shows hyperimmune colostrum powder Batch # 1710002662 improved cell viability by 186% relative to control, at 102.8 ug/ml.
  • Figure 7 shows hyperimmune colostrum powder Batch # 1710002859 improved cell viability by 218% relative to control, at 102.8 ug/ml.
  • Figure 8 shows pro-milk powder Batch # 6068 N50 (control) did not improve cell relative to control.
  • Figure 9 shows the positive control, remdesivir, did not improve cell relative to control.
  • ETEC enterotoxigenic E. coli
  • Example 3 Compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) that inhibit SARS-CoV-replication improve liver inflammation.
  • ETEC enterotoxigenic E. coli
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) that inhibit SARS- CoV-replication to alter liver function were examined.
  • ETEC enterotoxigenic E. coli
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) that inhibit SARS-CoV-replication to alter liver function
  • ETEC enterotoxigenic E. coli
  • 133 biopsy proven NASH patients were administered colostrum powder prepared as per Example 1.
  • a multinational, randomized, double-blind study comparing 2 doses of colostrum powder prepared as per Example 1 were compared to placebo in adults with any stage biopsy- proven NASH.
  • the trial enrolled 133 patients across 25 clinical sites in Australia (6), Israel (2) and the USA (17).
  • the trial had 12 scheduled visits over a 28-week study duration, with 24 weeks of treatment and four weeks of follow-up and screened a total of 237 patients.
  • the patients were randomized into three arms: placebo, high dose (1200mg), and low dose (600mg). Endpoints included changes in ASL and ALT as well as other liver enzymes and metabolic markers of liver inflammation.
  • Figure 10 shows that patients administered with colostrum powder prepared as per Example 1 have a statistically significant improvement in both AST and ALT levels (e.g. a 15% decrease in AST and ALT or a 30% decrease in AST and ALT) relative to placebo.
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) that inhibit SARS-CoV-2 replication, improve liver function.
  • ETEC enterotoxigenic E. coli
  • Example 4 Compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) that inhibit SARS-CoV-replication decrease levels of LPS in the serum of patients with inflammatory disease.
  • ETEC enterotoxigenic E. coli
  • pathogenesis of COVID-19 is believed to involve viral-induced suppression of innate pathogen surveillance systems.
  • PPR pathogen pattern recognition receptors
  • IFN interferon
  • genomic studies conducted on the original severe acute respiratory syndrome coronavirus (SARS- CoV) demonstrate that the virus encodes for proteins that serve as innate immune antagonists by suppressing the expression of IFN and promoting evasion of viral RNA from host defense mechanisms, independent of pro-inflammatory cytokine release.
  • cytokines i.e. , TNFa, IL-6, IP10 or CXCL10, etc.
  • coronaviruses pathogenesis involves the delayed release of IFN and an accumulation of monocyte/macrophages together with an inappropriate T-cell response.
  • severe cases of COVID- 19 appear to present with dysregulated T-cell counts and elevated inflammatory cytokine levels.
  • this disruption leads to a condition described as a “cytokine storm”, in which excessive amounts of pro-inflammatory cytokines are produced and may contribute to morbidity and mortality in these patients.
  • TLR4 is expressed on a wide range of immune cells, which specifically recognize bacterial lipopolysaccharide, and activation of TLR4 leads to the synthesis of proinflammatory cytokines and chemokine with the ultimate goal of identifying and destroying the pathogen.
  • activation of the innate immune system occasionally leads to host tissue collateral damage, resulting in organ dysfunction and death.
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) that inhibit SARS- CoV-replication to decrease serum levels of LPS, a cytokine storm inducing molecule, was examined.
  • ETEC enterotoxigenic E. coli
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) that inhibit SARS-CoV-replication to alter serum LPS levels
  • ETEC enterotoxigenic E. coli
  • 133 biopsy proven NASH patients were administered colostrum powder prepared as per Example 1.
  • a multinational, randomized, double-blind study comparing 2 doses of colostrum powder prepared as per Example 1 were compared to placebo in adults with any stage biopsy- proven NASH.
  • the trial enrolled 133 patients across 25 clinical sites in Australia (6), Israel (2) and the USA (17).
  • the trial had 12 scheduled visits over a 28-week study duration, with 24 weeks of treatment and four weeks of follow-up and screened a total of 237 patients.
  • the patients were randomized into three arms: placebo, high dose (1200mg), and low dose (600mg). Endpoints included serum LPS levels.
  • Figure 11 shows that patients administered with colostrum powder prepared as per Example 1 have an improvement in serum LPS levels relative to placebo, at all cut-offs, and statistically significantly improved. These differences are significant for cut-offs of at least a 15% decrease, 10%, 5% or any decrease. Furthermore, these data demonstrate that there is less deterioration in the 1200mg arm than placebo, significant at all cut-offs.
  • compositions comprising bovine-hyperimmune colostrum prepared using a vaccine comprising enterotoxigenic E. coli (ETEC) that inhibit SARS-CoV-2 replication, decrease serum LPS.
  • ETEC enterotoxigenic E. coli

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Virology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Otolaryngology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pulmonology (AREA)
  • Zoology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Physiology (AREA)
  • Nutrition Science (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L'invention concerne une méthode de traitement et/ou de prévention d'une infection à coronavirus humain chez un sujet, comprenant l'administration d'une quantité efficace d'une composition comprenant du colostrum hyperimmun bovin préparé à l'aide d'un vaccin comprenant un LPS d'E. coli entérotoxinogène (ETEC).
PCT/AU2021/050772 2020-07-17 2021-07-17 Méthodes de traitement d'une infection à coronavirus avec du colostrum hyperimmun bovin WO2022011436A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2020902486 2020-07-17
AU2020902486A AU2020902486A0 (en) 2020-07-17 Methods and compositions

Publications (1)

Publication Number Publication Date
WO2022011436A1 true WO2022011436A1 (fr) 2022-01-20

Family

ID=79555867

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2021/050772 WO2022011436A1 (fr) 2020-07-17 2021-07-17 Méthodes de traitement d'une infection à coronavirus avec du colostrum hyperimmun bovin

Country Status (1)

Country Link
WO (1) WO2022011436A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0930316A1 (fr) * 1998-01-19 1999-07-21 Medipharm CZ, s.r.o. Produit oral pour la prévention et le traitement des gastroentérites infectieuses des veaux
WO2004078209A1 (fr) * 2003-03-04 2004-09-16 Anadis Ltd Composition et methode pour le traitement et la prevention d'infections bacteriennes enteriques
WO2008124870A1 (fr) * 2007-04-11 2008-10-23 Anadis Ltd Administration d'anticorps antigrippaux au niveau de surfaces en contact avec l'air
WO2012023051A2 (fr) * 2010-08-17 2012-02-23 Immuron Ltd. Préparation d'immunoglobuline enrichie en anti-lps, destinée à être utilisée dans le traitement et/ou la prophylaxie d'un trouble pathologique

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0930316A1 (fr) * 1998-01-19 1999-07-21 Medipharm CZ, s.r.o. Produit oral pour la prévention et le traitement des gastroentérites infectieuses des veaux
WO2004078209A1 (fr) * 2003-03-04 2004-09-16 Anadis Ltd Composition et methode pour le traitement et la prevention d'infections bacteriennes enteriques
WO2008124870A1 (fr) * 2007-04-11 2008-10-23 Anadis Ltd Administration d'anticorps antigrippaux au niveau de surfaces en contact avec l'air
WO2012023051A2 (fr) * 2010-08-17 2012-02-23 Immuron Ltd. Préparation d'immunoglobuline enrichie en anti-lps, destinée à être utilisée dans le traitement et/ou la prophylaxie d'un trouble pathologique

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ULFMAN LAURIEN H., LEUSEN JEANETTE H. W., SAVELKOUL HUUB F. J., WARNER JOHN O., VAN NEERVEN R. J. JOOST: "Effects of Bovine Immunoglobulins on Immune Function, Allergy, and Infection", FRONTIERS IN NUTRITION, vol. 5, no. 52, pages 1 - 20, XP055899645, DOI: 10.3389/fnut.2018.00052 *

Similar Documents

Publication Publication Date Title
Serrano et al. Liposomal lactoferrin as potential preventative and cure for COVID-19
CN115605508A (zh) 用于治疗冠状病毒感染和所产生的炎症诱导的肺损伤的方法
Peng et al. A comprehensive summary of the knowledge on COVID-19 treatment
JP5970465B2 (ja) ペプチドおよびウイルス・ノイラミニダーゼ阻害剤を含んでなる組成物
ITTO950112A1 (it) Composizione e procedimemto per prevenire e trattare infiammazioni con immunoglobina a.
JP2016516801A (ja) 免疫微量栄養
Padayachee et al. The nose as a route for therapy. Part 2 immunotherapy
CA2764257A1 (fr) Agent pharmaceutique
KR20190132891A (ko) 잣나무 부산물 추출물을 함유하는 헬리코박터파일로리균 제균 효과를 갖는 조성물
WO2022011436A1 (fr) Méthodes de traitement d'une infection à coronavirus avec du colostrum hyperimmun bovin
US20100196495A1 (en) Delivery of flu antibodies to surfaces in contact with air
JP2020506882A (ja) 炎症性障害の治療療法
US20040242700A1 (en) Anti-influenzal agent
US20230346899A1 (en) PHARMACEUTICAL COMBINATION THERAPY AND PREVENTION WITH APROTININ + NIRMATRELVIR OF SARS-CoV-2 AND/OR DISEASE ASSOCIATED WITH THIS INFECTION, INCLUDING COVID-19
JP2008513468A (ja) Hiv侵入阻害剤及び局所殺菌剤としてのデンプン−ザクロジュース複合体
JP7302795B2 (ja) コロナウイルスの感染防御方法
AU2021247258A1 (en) Anti-CD6 antibody compositions and methods for treating and reducing negative effects of a coronavirus including COVID-19
US11491197B2 (en) Multiple viral antigen COVID vaccine and therapeutic
US10464998B2 (en) Anti-LPS enriched immunoglobulin for use in treatment and/or prophylaxis of fibrosis
US20240150441A1 (en) Monoclonal antibodies against sars-cov-2 and variants
CN116712480B (zh) 一种用于预防流行性感冒的药物组合物及其制剂和应用
Ahmed et al. Comparative study of the possible gastroprotective effect of probiotic (lactobacillus plantarum) and ranitidine on indomethacin induced gastric ulcer in adult male albino rat
US11517611B2 (en) Methods of treating viral infection with a composition comprising IL-18 and IL-22
Joshi et al. Role of Vitamin-C, Vitamin-D and Zinc in Covid-19 Pandemic
US20220025019A1 (en) Methods and compositions for preventing or treating acute exacerbations with polyclonal immunoglobulin

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: 21842834

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: 21842834

Country of ref document: EP

Kind code of ref document: A1