[go: up one dir, main page]

WO2024151612A1 - Compositions for inducing immune responses - Google Patents

Compositions for inducing immune responses Download PDF

Info

Publication number
WO2024151612A1
WO2024151612A1 PCT/US2024/010848 US2024010848W WO2024151612A1 WO 2024151612 A1 WO2024151612 A1 WO 2024151612A1 US 2024010848 W US2024010848 W US 2024010848W WO 2024151612 A1 WO2024151612 A1 WO 2024151612A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical agent
composition
spongilla
virus
inducing
Prior art date
Legal status (The legal status 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 status listed.)
Ceased
Application number
PCT/US2024/010848
Other languages
French (fr)
Inventor
Gerald Thomas PROEHL
Christopher Joseph NARDO
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Dermata Therapeutics Inc
Original Assignee
Dermata Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Dermata Therapeutics Inc filed Critical Dermata Therapeutics Inc
Publication of WO2024151612A1 publication Critical patent/WO2024151612A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/04Mycobacterium, e.g. Mycobacterium tuberculosis
    • 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
    • A61K35/56Materials from animals other than mammals
    • A61K35/655Aquatic animals other than those covered by groups A61K35/57 - A61K35/65
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration

Definitions

  • compositions comprising Spongilla for the intradermal delivery of vaccines.
  • Spongilla is a genus of freshwater sponge. Compositions derived from Spongilla can be used for a number of skin conditions, including acne. An important component of materials derived from Spongilla are the inorganic siliceous spicules that comprise the skeletal structure of Spongilla.
  • a topical mode of application may also permit an effective amount of the one or more vaccine antigens to penetrate the skin of the subject so that an effective amount of the one or more antigens is delivered to the desired site of action, such as the dermis of the subject.
  • Spongilla spicules can penetrate the stratum comeum and entire dermis of the skin of a subject during application and promote the penetration of topically-applied compositions, such as a hydrogel composed of polymers, copolymers, and cross-linked versions thereof into the skin of the subject, such as the dermis, to permit or improve the delivery of an effective amount of a vaccine formulation to the site of action necessary to effectively induce an immune response in the subject.
  • topically-applied compositions such as a hydrogel composed of polymers, copolymers, and cross-linked versions thereof into the skin of the subject, such as the dermis
  • the materials comprising Spongilla used herein may comprise all organic and/or inorganic compounds and materials that are part of the naturally-occurring Spongilla. or may include only a portion of the organic and/or inorganic compounds and materials that are part of the naturally-occurring Spongilla.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongilla,' and a second composition comprising one or more biological macromolecules.
  • the one or more biological macromolecules comprise a nucleic acid molecule, a protein molecule, or a combination thereof.
  • the nucleic acid molecule comprises an RNA or a DNA molecule encoding an antigenic protein or a fragment thereof.
  • the DNA molecule further comprises a promoter.
  • the promoter is a T7 promoter, a T3 promoter, or an SP6 promoter.
  • the RNA molecule is a self-replicating molecule.
  • the nucleic acid molecule further comprises a 5’ cap.
  • the nucleic acid molecule further comprises a 5’ UTR and/or a 3’ UTR.
  • the 3’ UTR comprises a poly(A) sequence.
  • the protein molecule is an antigenic protein or a fragment thereof.
  • the antigenic protein is a viral protein. In some embodiments, the viral protein originates from a human virus. [00015] In some embodiments, the antigenic protein is a bacterial protein.
  • the second composition comprises a vaccine against Mycobacterium tuberculosis.
  • the vaccine is the Bacillus Calmette- Guerin (BCG) vaccine.
  • the pharmaceutical agent further comprises a vaccine vector of murine origin.
  • the antigenic protein is a fungal protein. In some embodiments, the antigenic protein is a helminthic protein. In some embodiments, the antigenic protein is a protozoan protein. In some embodiments, the antigenic protein is a prion protein. In some embodiments, the antigenic protein is a cancer protein.
  • the pharmaceutical agent further comprises a whole cell cancer vaccine.
  • the protein molecule is an immunomodulatory protein.
  • the second composition comprises two or more biological macromolecules.
  • a first macromolecule of the two or more biological macromolecules comprises an antigenic protein or a fragment thereof, and a second macromolecule comprises an immunomodulatory protein.
  • the pharmaceutical agent further comprises a lipid formulation.
  • the lipid formulation is selected from a lipoplex, a liposome, a lipid nanoparticle, a polymer-based earner, an exosome, a lamellar body, a micelle, and an emulsion.
  • the lipid formulation further comprises a polyethylene glycol (PEG)-lipid conjugate.
  • the agent further comprises one or more cryoprotectants.
  • the one or more cryoprotectants are selected from sucrose, glycerol, or a combination of sucrose and glycerol.
  • the Spongilla is in the form of a powder. Tn some embodiments, the powder comprises particles that are substantially uniform in size.
  • not less than 90% of the particles comprising the Spongilla powder pass through a US 70-mesh screen.
  • the particles comprising the Spongilla powder have an average length of from about 50 pm to about 500 pm.
  • the particles comprising the Spongilla powder have an average diameter of from about 5 pm to about 50 pm.
  • the particles comprising the Spongilla powder have an aspect ratio of from about 1 to 100.
  • the particles comprising the Spongilla powder comprise only inorganic Spongilla spicules and any materials that are naturally associated with the spicules.
  • the second composition is a lyophilized or vacuum-dried composition. In some embodiments, the second composition is a liquid composition.
  • the first composition is in the form of a paste, gel, or topical cream.
  • the paste further comprises water, saline, or hydrogen peroxide.
  • the paste is prepared by mixing a powder comprising Spongilla and water or saline or hydrogen peroxide.
  • the first composition is packaged prior to use. In some embodiments, the first composition is prepared by treating with gamma radiation.
  • the Spongilla is Spongilla lacustris.
  • the first composition has a residual moisture content of not more than about 5%.
  • the amount of the first composition comprising Spongilla is from about 0.5 grams to about 50 grams.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongillcr, and a second composition comprising a vaccine against mycobacteria.
  • the vaccine against mycobacteria comprises BCG.
  • the present disclosure provides a method of delivering a vaccine to a subject in need thereof, comprising administering an effective amount of any of the pharmaceutical agents described herein to the subject.
  • the pharmaceutical agent is delivered intradermally.
  • the first composition is administered first, and the second composition is administered second. In some embodiments, the first composition and the second composition are administered together.
  • the second composition is lyophilized or vacuum-dried and is reconstituted prior to administration.
  • the present disclosure provides a method of preventing an infection in a subject in need thereof, comprising administering any of the pharmaceutical agents described herein to the subject.
  • the present disclosure provides a method of preventing cancer in a subject in need thereof, comprising administering any of the pharmaceutical agents described herein to the subject.
  • the present disclosure provides a method of inducing an immune response in a subject in need thereof comprising administering an effective amount of any of the pharmaceutical agents described herein to the subject.
  • the pharmaceutical agent is administered one time. In some embodiments, wherein the pharmaceutical agent is administered two or more times.
  • the pharmaceutical agent is applied to mucosal surfaces.
  • the pharmaceutical agent is administered intradermally.
  • the present disclosure provides a kit for inducing an immune response in a subject in need thereof, wherein the kit comprises a first composition comprising Spongilla and a second composition comprising one or more biological macromolecules.
  • the one or more biological macromolecules comprise a nucleic acid molecule, a protein molecule, or a combination thereof.
  • the kit may further comprise a reconstitution agent. In some embodiments, the kit may further comprise an antiseptic reagent.
  • the kit may further comprise instructions for use.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongilla, and a second composition comprising a whole cell cancer vaccine.
  • FIG. 1 shows the magnified surface of a t pical Spongilla composition.
  • a cell includes one or more cells, including mixtures thereof.
  • a and/or B is used herein to include all of the following alternatives: “A”, “B”, “A or B”, and “A and B”.
  • the term “about” means either within plus or minus 10% of the provided value, or rounded to the nearest significant figure, in all cases inclusive of the provided value. Where ranges are provided, they are inclusive of the boundary 7 values.
  • the terms “applied,” “applying,” “administration,” “administering,” “used,” or “deliver” means the delivery of a composition disclosed herein to a subject, in particular to the skin of the subject, by an administration route including, but not limited to, intradermal, subcutaneous, topical, or any combinations thereof.
  • the compositions disclosed herein are administered to the subject, in particular to the skin of the subject, by intradermal administration.
  • the term “‘aspect ratio” means with respect to the particles of Spongilla described herein, the ratio between the average length of the particles to the average diameter of the particles.
  • the terms “combination” and “in combination with” mean the application, use, or administration of one or more of the compositions disclosed herein, sequentially or simultaneously. It includes dosing simultaneously, or within minutes or hours of each other, or on the same day, or on alternating days, or using the compositions disclosed herein on a daily basis, or multiple days per week, or weekly basis, for example, while administering another composition on the same day or alternating days or weeks or on a periodic basis during a time simultaneous therewith or concurrent therewith, or at least a part of the time during which the composition disclosed herein is applied, used or administered.
  • Spongilla as used herein means a genus of freshwater sponges in the family Spongillidae, including, but not limited to, Spongillct lacustris, S. fragilis I.eidy. and Ephydatia fluviatilis.
  • Spongilla lacustris as used herein means a species of sponge of the freshwater sponge family Spongillidae.
  • FIG. 1 shows a scanning electron microscopy view of the structure of Spongillidae.
  • composition comprising Spongilla mean materials comprising Spongilla derived from raw Spongilla that is harvested and processed and may include all the various components of the Spongilla following harvest, including all organic and/or inorganic compounds and materials that are part of the naturally-occurring Spongilla, or may include only a portion of the organic and/or inorganic compounds and materials that are part of the naturally-occurring Spongilla.
  • the Spongilla materials comprise all or substantially all the organic and inorganic materials derived from the naturally occurring Spongilla.
  • the Spongilla materials comprise (a) only the inorganic spicules and any materials that are naturally associated with the spicules, or (b) substantially purified inorganic spicules and any materials that are naturally associated with the spicules, or (c) purified inorganic spicules and any materials that are naturally associated with the spicules that are a component part of naturally -occurring Spongilla.
  • any of the methods, compositions or kits disclosed herein wherein the Spongilla materials comprise only the inorganic spicules and any materials that are naturally associated with the spicules. In another aspect is provided any of the methods, compositions or kits disclosed herein, wherein the Spongilla materials comprise substantially purified inorganic spicules and any materials that are naturally associated with the spicules. In another aspect is provided any of the methods, compositions or kits disclosed herein, wherein the Spongilla materials comprise purified inorganic spicules and any materials that are naturally associated with the spicules that are a component part of naturally occurring Spongilla.
  • composition comprising one or more sponges/’ “powders comprising one or more sponges, “materials comprising one or more sponges,” “sponge in the form of a powder,” and the like, as used herein, mean materials derived from one or more sponges that is harvested and processed and may include all the various components of the sponge following harvest, including all organic and/or inorganic compounds and materials that are part of the naturally-occurring sponge, or any sponges that are specially grow n or adapted for use in the disclosed compositions, methods and/or kits, or may include only a portion of the organic and/or inorganic compounds and materials that are part of the naturally-occurring sponge.
  • the sponge materials comprise all or substantially all the organic and inorganic materials derived from the naturally occurring sponge.
  • the sponge materials comprise (a) only the spicules and any materials that are naturally associated with the spicules, or (b) substantially purified spicules and any materials that are naturally associated with the spicules, or (c) purified spicules and any materials that are naturally associated with the spicules that are a component part of naturally -occurring sponge.
  • any of the methods or kits disclosed herein wherein the sponge materials comprise only the spicules and any materials that are naturally associated with the spicules. In another aspect is provided any of the methods or kits disclosed herein, wherein the sponge materials comprise substantially purified spicules and any materials that are naturally associated with the spicules. In another aspect is provided any of the methods or kits disclosed herein, w herein the sponge materials comprise purified spicules and any materials that are naturally associated with the spicules that are a component part of naturally occurring sponge. These terms may be used herein in relation to materials derived from the phylum Porifera. In another aspect, the materials are derived from sponges of the class Demospongiae .
  • the materials are derived from sponges of the order Spongdilla. In another aspect, the materials are derived from sponges of the family Spongillidae. In another aspect, the materials are derived from sponges of the genus Spongilla. In another aspect, the materials are derived from sponges of the species Spongilla lacustris. In another aspect, the materials are derived from sponges of the order Haplosclerida. In another aspect, the materials are derived from sponges of the family Chalinidea. In another aspect, the materials are derived from sponges of the genus Halciona.
  • topical application means placing or spreading directly onto the outer skin, scalp, mucous membrane, or hair, for example, by using the hands or an applicator such as a wipe, roller, or spray.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongilla,' and a second composition comprising one or more biological macromolecules.
  • Spongilla is a genus of freshwater sponges with over 200 different species.
  • the organism has a leuconoid body form with a skeleton composed of siliceous spicules.
  • the Spongilla contains organic and inorganic compounds with a total lipid content of approximately 5% of the biomass of the dried sponge; and the protein is composed of spongin or sclerotized collagen.
  • the polysaccharides and N-acetyl-D-glucosamine (NAG) are part of chitin and chitosan that has been reported to be an important component within the skeletal fibers of Spongilla lacustris and detected 750 ⁇ 1 .5ug N-acetyl-D-glucosamine per 1 mg of spicule-free skeleton.
  • Chitin and chitosan are described as a family of linear polysaccharides consisting of varying amounts of a or
  • a-chitin the chains are arranged in sheets or stacks, the chains in any one sheet having the same direction or "sense’.
  • adjacent sheets along the c axis have the same direction; the sheets are parallel, while in a-chitin adjacent sheets along the c axis have the opposite direction, they are antiparallel.
  • Chitin is deacetylated into chitosan and can be further degraded into N- acetyl-D-glucosamine (NAG) units.
  • NAG N- acetyl-D-glucosamine
  • Chitosan can be used to prevent or treat wound and bum infections not only because of its intrinsic antimicrobial properties, but also by virtue of its ability to deliver extrinsic antimicrobial agents to wounds and bums.
  • Chitosan is water-insoluble and highly viscous in dilute acidic solutions. Soluble chitosan oligosaccharides were found to be instrumental in suppressing the LPS-induced nuclear factor kappa-light-chain-enhancer of activated B cell (NF-KB)- dependent inflammatory gene expression, and this was associated with reduced nuclear translocation of NF-KB. Chitosan has also been demonstrated to have an antimicrobial effect against various bacteria.
  • the Spongilla is in the form of a powder.
  • Spongilla. including Spongilla lacustris, and powders prepared from Spongilla that are utilized in the methods, uses, compositions for use as a medicament, and kits disclosed herein may be obtained, processed and characterized by methods known to those having ordinary skill in the art.
  • United States Patent No. 7,604,821 describes the harvest, processing and characterization of several species of Spongilla, including Spongilla lacustris. The disclosure of United States Patent No. 7,604,821 is incorporated herein by reference in its entirety.
  • Sponge materials may be collected using methods commonly known to those skilled in the art of marine biology.
  • sponges can be collected manually using basic under water diving techniques, or in deeper waters larger colonies are harvested using the Agassiz trawl (AGT) or epibenthic sledge (EBS).
  • AGT Agassiz trawl
  • EBS epibenthic sledge
  • Spongilla colonies occur in a thin crust-like carpet several meters across and must be collected manually, with fork-like tools, and nets.
  • the collected sponge mass is dried, cleaned of gross contamination, such as shells, stems, plants, rocks and other impurities, and is then washed to remove dirt, sand, silt and soluble impurities.
  • the cleaned sponge mass is weighed and dried using methods know n to those of ordinary skill in the art, such as air drying and the use of dryers that are used to dehydrate foods and pharmaceuticals.
  • the sponge mass is dried until residual moisture content is less than a desired value as further disclosed herein.
  • Residual moisture measurements can be performed using methods commonly known in the arts of food sciences, analytical chemistry or the pharmaceutical sciences. For example, 10 grams of dried material may be placed on a tared weighing boat and then weighed. The weighed material is then exposed to a heat source such as a drying oven or heat lamp operated at an appropriate temperature, the sample is then cooled in a desiccated chamber and re-weighed. Residual moisture is calculated as the percent difference between the sample weight before drying and the weight after cooling. Following drying, the sponge materials may be packaged in sealed containers, which optionally protect the materials from light, moisture and oxygen.
  • the materials may then be further tested for the presence of pathogens, coliform organisms and organisms that represent a bioburden.
  • the materials may be further heated or irradiated, as disclosed herein, to reduce any pathogens, coliform organisms or other organisms that represent a bioburden.
  • the materials may then be further processed using methods known to those having ordinary skill in the art to provide a powder comprising particles, or inorganic spicules having a desired size.
  • the sponge materials may be ground, and the resulting materials passed through one or more sieves of a defined size to provide a resulting material comprising particles, or inorganic spicules having a uniform, or substantially uniform, size.
  • the dried sponge material may be packaged in airtight moisture-proof containers and stored at an appropriate temperature, such as at about room or ambient temperature.
  • the powder comprises particles, or inorganic spicules that are substantially uniform in size. In embodiments, not less than about 50% of the particles, or inorganic spicules included in the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 60% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 70% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 75% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen.
  • not less than about 80% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 85% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 90% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 95% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen.
  • not less than about 96% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 97% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 98% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 99% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen.
  • not less than 90% of the particles, or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen.
  • the particles of Spongilla may be manufactured or produced from Spongilla materials that are harvested by procedures known to those of ordinary skill in the art, such as determining the appropriate harvest period, removal of foreign materials, drying, milling, grinding, and sieving using equipment known to those of ordinary skill in the art.
  • the particles comprising the Spongilla powder have an average length of from about 50 pm to about 500 pm. In some embodiments, the inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 500 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 300 pm.
  • the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 250 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 200 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 75 pm to about 500 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 75 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 80 pm to about 450 pm.
  • the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 80 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 85 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 85 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 90 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 90 pm to about 400 pm.
  • the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 90 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 300 pm.
  • the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 250 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 200 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 500 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 400 pm.
  • the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 300 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 250 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 200 pm.
  • the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 175 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 175 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 175 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 300 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 175 pm to about 250 pm.
  • the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 175 pm to about 200 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 50 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 75 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 80 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 85 pm.
  • the particles or inorganic spicules comprising the Spongilla powder have an average length of about 90 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 100 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 125 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 150 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 175 pm.
  • the particles or inorganic spicules comprising the Spongilla powder have an average length of about 200 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 225 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 250 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 300 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 350 pm.
  • the particles or inorganic spicules comprising the Spongilla powder have an average length of about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 500 pm.
  • the particles comprising the Spongilla powder may be manufactured or produced from Spongilla materials that are harvested by procedures known to those of ordinary skill in the art, such as milling and grinding using equipment known to those of ordinary 7 skill in the art.
  • the average length of particles or inorganic spicules including the Spongilla powder may be measured using analytical methods known to those of ordinary skill in the art, such as, for example, scanning electron microscopy (SEM) and sieve analysis. Sieve analysis may also be used to determine the particle size distribution of the particles or inorganic spicules comprising the Spongilla powder.
  • SEM scanning electron microscopy
  • Sieve analysis may also be used to determine the particle size distribution of the particles or inorganic spicules comprising the Spongilla powder.
  • the particles of the Spongilla powder have an average diameter of from about 5 pm to about 50 pm.
  • the inorganic spicules including the Spongilla powder have an average diameter of from about 5 pm to about 50 pm.
  • the particles or inorganic spicules including the Spongilla powder have an average diameter of from about 5 pm to about 45 pm, or from about 5 pm to about 40 pm, from about 5 pm to about 35 pm, from about 5 pm to about 30 pm. from about 5 pm to about 25 pm, from about 5 pm to about 20 pm.
  • the particles or inorganic spicules including the Spongilla powder have an average diameter of about 5 pm, or about 10 pm, or about 15 pm, or about 20 pm, or about 25 pm, or about 30 pm, or about 35 pm, or about 40 pm, or about 45 pm, or about 50 pm.
  • the particles including the Spongilla powder may be manufactured or produced from Spongilla materials that are harvested by procedures known to those of ordinary’ skill in the art, such as milling and grinding using equipment known to those of ordinary skill in the art.
  • the average diameter of particles or inorganic spicules comprising the Spongilla powder may be measured using analytical methods known to those of ordinary' skill in the art, such as, for example, scanning electron microscopy (SEM) and sieve analysis. Sieve analysis may also be used to determine the particle size distribution of the particles or inorganic spicules comprising the Spongilla powder.
  • the particles comprising the Spongilla powder have an aspect ratio of from about 1 to 100.
  • the inorganic spicules comprising the Spongilla powder have an aspect ratio of from about 1 to 100.
  • the term ⁇ ‘aspect ratio” means with respect to the particles of Spongilla or the inorganic spicules described herein the ratio between the average length of the particles or inorganic spicules to the average diameter of the particles or inorganic spicules.
  • a specific aspect ratio is important for a more uniform final product so the spicules won't be too long or too thick, to ensure an ideal depth of penetration into the stratum comeum. Applicant discovered that an extended particle that is longer than it is wide produces a certain size opening in the skin that allows for subsequent delivery' of larger molecules.
  • the particles of the Spongilla powder have an aspect ratio of from about 1 to about 75.
  • the inorganic spicules including the Spongilla powder have an aspect ratio of from about 1 to about 75.
  • the particles or inorganic spicules including the Spongilla powder have an aspect ratio of from about 1 to about 50, or from about 1 to about 25, or from about 1 to about 20, or from about 1 to about 15, or from about 5 to about 100, or from about 5 to about 75, or from about 5 to about 50, or from about 5 to about 40, or from about 5 to about 35, or from about 5 to about 30, or from about 5 to about 25, or from about 5 to about 20, or from about 5 to about 15, or from about 7 to about 50, or from about 7 to about 45, or from about 7 to about 40.
  • the particles or inorganic spicules including the Spongilla powder have an aspect ratio of about 5, or about 6, or about 7, or about 8, or about 9, or about 10, or about 11, or about 12, or about 13, or about 14, or about 15, or about 16, or about 17, or about 18, or about 19, or about 20, or about 21, or about 22, or about 23, or about 24, or about 25. or about 26, or about 27. or about 28, or about 29. or about 30, or about 35. or about 40.
  • the particles including the Spongilla powder may be manufactured or produced from Spongilla materials that are harvested by procedures known to those of ordinary' skill in the art, such as milling and grinding using equipment known to those of ordinary skill in the art.
  • the aspect ratio of particles or inorganic spicules including the Spongilla powder may be measured using analytical methods known to those of ordinary skill in the art, such as, for example, scanning electron microscopy (SEM) and sieve analysis. Sieve analysis may also be used to determine the particle size distribution of the particles comprising the Spongilla pow der.
  • Materials comprising Spongilla may be processed and dried, using techniques known to those having ordinary skill in the art, such as the use of drying ovens, to provide materials having a desired residual moisture content.
  • the first composition including Spongilla has a residual moisture content of not more than about 20%.
  • the first composition has a residual moisture content of not more than about 15%, or not more than about 10%, or not more than about 9%, or not more than about 8%, or not more than about 7%, or not more than about 6%, or not more than about 5%, or not more than about 4%, or not more than about 3%. or not more than about 2%, or not more than 1%.
  • the first composition has a residual moisture content of not more than about 5%. In another embodiment, the first composition has a residual moisture content of not more than about 4%. In another embodiment, the first composition has a residual moisture content of not more than about 3%. In another embodiment, the first composition has a residual moisture content of not more than about 2%. In another embodiment, the first composition has a residual moisture content of not more than about 1%.
  • the moisture content of the Spongilla materials can be reduced by heating the raw Spongilla materials using methods known to those of ordinary skill in the art, such as by open-air drying, or by use of a conventional oven dryer or a vacuum dryer, using equipment know n to those of ordinary skill in the art.
  • raw Spongilla materials may be placed into a tray and heated in a dry ing oven at a temperature range from about 30°C to about 200°C, for example to about 70°C, for a period of time necessary to reduce the residual moisture content to the desired level.
  • the level of residual moisture of the materials may be measured using methods known to those of ordinary skill in the art such as those described in the United States Pharmacopeia methods USP ⁇ 731> (Loss on Drying) and USP ⁇ 921> (Water Determination).
  • Materials comprising Spongilla may be treated in order to reduce the bioburden, such as aerobic and anaerobic microbes, yeast and mold, Coliform bacteria. Salmonella, Pseudomonas aeruginosa, and Staphylococcus aureus, of the materials prior to their packaging and use. such as by use of ionizing irradiation, such as the use of gamma irradiation.
  • gamma irradiation may be performed on the raw Spongilla material prior to grinding to reduce the particle or inorganic spicule size, following grinding to reduce the particle or inorganic spicule size, the materials packaged in bulk and/or the materials following packaging in unit dose containers.
  • the materials may be treated with ionizing radiation, such as gamma radiation, using methods and equipment known to those of ordinary skill in the art. such as gamma irradiators or electron beam irradiators.
  • the first composition is prepared by treating with ionizing radiation, such as gamma radiation, to deliver an absorbed radiation dose between about 1 kGy and about 50 kGy prior to being packaged.
  • the first composition is prepared by treating with ionizing radiation, such as gamma radiation, to deliver an absorbed radiation dose between about 1 kGy and about 45 kGy, or between about 1 kGy and about 40 kGy .
  • the first composition is prepared by treating with ionizing radiation, such as gamma radiation, to deliver an absorbed radiation dose of about 1 kGy, or about 5 kGy, or about 10 kGy, 11 kGy, or about 12 kGy, or about 13 kGy, or about 14 kGy, or about 15 kGy, or about 16 kGy, or about 17 kGy, or about 18 kGy, or about 19 kGy.
  • ionizing radiation such as gamma radiation
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition comprising Spongilla has a combined yeast and mold content of not more than about 25 x 10 4 colony-forming units per gram (CFU/g).
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 5 x 10 4 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 1 x 10 4 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 5 x 10 3 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 1 x 10 4 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 10,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 7,500 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 5,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 2,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 2,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 1,500 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 1,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 250 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 100 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 75 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 25 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 10 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 1 CFU/g.
  • the combined yeast and mold content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or dry heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels.
  • the combined yeast and mold content of the Spongilla materials may be measured by methods known to those of ordinary' skill in the art, such as those described in the United States Pharmacopeia method USP ⁇ 61> (Microbial Enumeration Tests).
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 25 x 10 4 colony-forming units per gram (CFU/g). In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 5 x 10 4 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 1 x 10 4 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 5 x 10 3 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 1 x 10 4 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 10,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 7,500 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 5,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 2,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 2,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 1,500 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 1,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 250 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 100 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 75 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 25 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 10 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 1 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has no detectable Coliform bacterial content.
  • the Coliform bacteria content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or diy heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels.
  • the Coliform bacteria content of the Spongilla materials may be measured by methods known to those of ordinary skill in the art, such as those described in the United States Pharmacopeia method USP ⁇ 62> (Tests for Specified Microorganisms).
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 25 x 10 4 colony-forming units per gram (CFU/g). In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 5 x 10 4 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 1 x 10 4 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 5 x 10 3 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 1 x 10 4 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 10.000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 7,500 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 5,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 2,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 2,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 1,500 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 1,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 250 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 100 CFU/g. In another aspect is provided any of the methods, compositions for use as a medicament, drug products and kits disclosed herein, wherein the first composition has a Salmonella content of not more than about 75 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 25 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 10 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 1 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has no detectable Salmonella content.
  • the Salmonella content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or dry heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels.
  • the Salmonella content of the Spongilla materials may be measured by methods known to those of ordinary ⁇ skill in the art, such as those described in the United States Pharmacopeia method USP ⁇ 62> (Tests for Specified Microorganisms).
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 25 x 10 4 colony-forming units per gram (CFU/g). In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 5 x 10 4 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 1 x 10 4 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 5 x 10 3 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 1 x 10 4 CFU/g. Tn another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 10,000 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 7,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 5,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 2,500 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 2,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 1,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 1,000 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 250 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 100 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has Pseudomonas aeruginosa bacteria content of not more than about 75 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 25 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 10 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 1 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has no detectable Pseudomonas aeruginosa bacteria content.
  • the Pseudomonas aeruginosa bacteria content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or dry' heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels.
  • the Pseudomonas aeruginosa bacteria content of the Spongilla materials may be measured by methods known to those of ordinary skill in the art, such as those described in the United States Pharmacopeia method USP ⁇ 62> (Tests for Specified Microorganisms).
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 25 x 10 4 colony -forming units per gram (CFU/g). In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 5 x 10 4 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 1 x 10 4 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 5 x 10 3 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 1 x O 4 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 10,000 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 7,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 5,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 2,500 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 2.000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 1,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 1,000 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 250 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 100 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 75 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 25 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 10 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 1 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has no detectable Staphylococcus aureus bacteria content.
  • the Staphylococcus aureus bacteria content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or dry heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels.
  • the Staphylococcus aureus bacteria content of the Spongilla materials may be measured by methods known to those of ordinary skill in the art, such as those described in the United States Pharmacopeia method USP ⁇ 62> (Tests for Specified Microorganisms).
  • compositions disclosed herein may further comprise one or more conventional pharmaceutical carriers or excipients.
  • suitable pharmaceutical carriers and excipients include inert diluents, binders (such as saccharides and their derivatives, proteins, and synthetic polymers), fillers (such as colloidal silicon dioxide, sugars, including lactose, sucrose, mannitol, or sorbitol); and cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethylcellulose, or polyvinylpyrrolidone (PVP)), bulking agents, lubricants (such as magnesium stearate, sodium lauryl sulfate and talc), coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with diluent
  • PVP polyvinylpyrrolidone
  • compositions disclosed herein may further include preservatives like antioxidants, amino acids, citric acid, sodium citrate, and synthetic preservatives.
  • Antioxidants may include, but are not necessarily limited to vitamin A, vitamin E, vitamin C, retinyl palmitate, and selenium.
  • Amino acids suitable for preserving any of the pharmaceutical compositions described herein may include, but are not necessarily limited to cysteine and methionine.
  • Synthetic preservatives suitable for preserving any of the pharmaceutical compositions described herein may include, but are not necessarily limited to methyl paraben and propyl paraben.
  • the first composition comprising Spongilla and/or the second composition comprising one or more biological macromolecules may further comprise one or more adjuvants.
  • adjuvant means a substance added to a vaccine to increase a vaccine's immunogenicity. Some adjuvants are believed to enhance the immune response by slowly releasing the antigen, while other adjuvants are strongly immunogenic in their own right and are believed to function synergistically.
  • adjuvants function may include, but are not necessarily limited to, cellular infiltration against antigen-presenting cells (APCs), increased inflammation and trafficking to the injection site, enhancing the activity of APCs by up- regulating costimulatory signal or major histocompatibility gene complex (MHC) expression, enhanced antigen presentation, or induction of cytokine release for indirect effects.
  • APCs antigen-presenting cells
  • MHC major histocompatibility gene complex
  • the most appropriate adjuvant for a given vaccine immunogen will depend to a large extent on the type of immune response required for protective immunity. Adjuvant selection can be somewhat empirical.
  • Known vaccine adjuvants include, but are not limited to, oil and water emulsions (for example, complete Freund's adjuvant and incomplete Freund's adjuvant), toll-like receptor (TLR) agonists, monophosphoryl lipid A)(MPL®), synthetic lipid A, lipid A mimetics or analogs, aluminum salts, cytokines, saponins, muramyl dipeptide (MDP) derivatives, CpG oligos, lipopolysaccharide (LPS) of gram-negative bacteria.
  • oil and water emulsions for example, complete Freund's adjuvant and incomplete Freund's adjuvant
  • TLR toll-like receptor
  • MPL® monophosphoryl lipid A
  • synthetic lipid A lipid A mimetics or analogs
  • aluminum salts aluminum salts
  • cytokines saponins
  • MDP muramyl dipeptide
  • CpG oligos CpG oligos
  • LPS lipopoly
  • polyphosphazenes any water in oil emulsion, any oil in water emulsion that contains one or more of the following constituents: squalene or its analogues or any pharmaceutically acceptable oil, tween-80, sorbitantrioleate, alpha-tocopherol, cholecalciferol and aqueous buffer, or any of the analogues and derivatives of the molecules thereof, virosomes, cochleates, poly(lactide-co-glycolides) (PLG) microparticles, poloxamer particles, microparticles, endotoxins, for instance bacterial endotoxins, liposomes, Corynebacterium parvum, Bacillus Calmette Guerin, aluminum hydroxide, aluminum phosphate, aluminum sulphate phosphate, inulin, algammulin, resiquimod, poly(I:C), glucan, dextran sulfate, iron oxide, sodium alginate, Bacto-Adju
  • Spongilla spicules act as an adjuvant, for example, by irritating the site of administration to recruit relevant immune cells.
  • the particles comprising the Spongilla powder comprise only inorganic Spongilla spicules and any materials that are naturally associated with the spicules, as described above.
  • the first composition is in the form of a powder, paste, gel, liquid, or topical cream.
  • the powder, paste, gel, liquid, or topical cream further comprises water, saline, or hydrogen peroxide.
  • the paste, gel, liquid, or topical cream is prepared by mixing a powder comprising Spongilla and water or saline or hydrogen peroxide.
  • the first composition is applied to the skin of the subject in the form of a paste.
  • the paste further comprises water or saline.
  • the paste is prepared by mixing a composition comprising Spongilla and an aqueous solution comprising hydrogen peroxide.
  • the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 50% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 45% w/w.
  • the hydrogen peroxide is at a concentration of from about 0. 1 % w/w to about 40% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 35% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 30% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 25% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 20% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 0.1% w/w to about 20% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 0.1% w/w to about 20% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 0. 1 % w/w to
  • the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 10% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 9% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1 % w/w to about 7% w/w.
  • the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 6% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 5% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 4% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1 % w/w to about 3% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 2% w/w.
  • the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 1% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.5% w/w to about 45% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 45% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 40% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 35% w/w.
  • the hydrogen peroxide is at a concentration of from about 1% w/w to about 30% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 25% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 20% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 15% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1 % w/w to about 10% w/w.
  • the hydrogen peroxide is at a concentration of from about 1 % w/w to about 9% w/w. Tn some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 7% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 6% w/w.
  • the hydrogen peroxide is at a concentration of from about 1% w/w to about 5% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 4% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 3% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 2% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 45% w/w.
  • the hydrogen peroxide is at a concentration of from about 2% w/w to about 40% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 35% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 30% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 25% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 20% w/w.
  • the hydrogen peroxide is at a concentration of from about 2% w/w to about 15% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 10% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 9% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 7% w/w.
  • the hydrogen peroxide is at a concentration of from about 2% w/w to about 6% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 5% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 4% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 3% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 0. 1% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 0.5% w/w.
  • the hydrogen peroxide is at a concentration of about 1% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 2% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 3% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 4% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 5% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 6% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 7% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 8% w/w.
  • the hydrogen peroxide is at a concentration of about 9% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 10% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 15% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 20% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 25% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 30% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 35% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 40% w/w.
  • the hydrogen peroxide is at a concentration of about 45% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 50% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 3% w/w.
  • Aqueous hydrogen peroxide solutions that may be useful for the compositions and methods disclosed herein are commercially available or may be prepared by methods known to those of ordinary skill in the art.
  • the first composition is packaged prior to use.
  • the sponge materials may be packaged in sealed containers, which optionally protect the materials from light, moisture and oxygen.
  • the dried sponge material may be packaged in airtight moisture-proof containers and stored at an appropriate temperature, such as at about room or ambient temperature.
  • the one or more sponges may be marine sponges or freshwater sponges. In other embodiments, the one or more sponges is a marine sponge. In other embodiments, the sponge is a freshwater sponge. In other embodiments, the compositions are derived from sponges of the phylum Porifera. In other embodiments, the compositions are derived from sponges of the class Demospongiae. In other embodiments, the compositions are derived from sponges of the order Spongdilla. In other embodiments, the compositions are derived from sponges of the family Spongillidae . In other embodiments, the compositions are derived from sponges of the genus Spongilla.
  • the compositions are derived from sponges of the species Spongilla lacustris.
  • the type of sponge that can be used within the scope of the present disclosure is not necessarily limited to members of the genus Spongilla.
  • the compositions are derived from sponges of the order Haplosclerida.
  • the compositions are derived from sponges of the family Chalinidea.
  • the compositions are derived from sponges of the genus Halciona.
  • compositions described herein contain one or more biological macromolecules.
  • the biological macromolecules may be molecules that initiate an immune response when administered to a subject.
  • biological macromolecule refers to a biological molecule such as a nucleic acid, protein, antibody , carbohydrate, polysaccharide, lipid, and the like.
  • the one or more biological macromolecules may include a nucleic acid molecule, a protein molecule, or a combination thereof.
  • the nucleic acid molecule may comprise an RNA or a DNA molecule encoding an antigenic protein or a fragment thereof.
  • fragment when referring to a protein or nucleic acid, for example, means any shorter sequence than the full-length protein or nucleic acid. Accordingly, any sequence of a nucleic acid or protein other than the full-length nucleic acid or protein sequence can be a fragment.
  • a protein fragment includes an epitope. In other aspects, a protein fragment is an epitope.
  • the terms “functional variant” or “functional fragment” refer to a molecule, including a nucleic acid or protein, for example, that comprises a nucleotide and/or amino acid sequence that is altered by one or more nucleotides and/or amino acids compared to the nucleotide and/or amino acid sequences of the parent or reference molecule.
  • a functional variant is still able to function in a manner that is similar to the parent molecule.
  • the modifications in the amino acid and/or nucleotide sequence of the parent molecule do not significantly affect or alter the functional characteristics of the molecule encoded by the nucleotide sequence or containing the amino acid sequence.
  • the functional variant may have conservative sequence modifications including nucleotide and amino acid substitutions, additions and deletions. These modifications can be introduced by standard techniques known in the art, such as site-directed mutagenesis and random PCR- mediated mutagenesis.
  • Functional variants can also include, but are not limited to, derivatives that are substantially similar in primary structural sequence, but which contain, e.g., in vitro or in vivo modifications, chemical and/or biochemical, that are not found in the parent molecule.
  • Such modifications include, inter aha, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol.
  • cross-linking cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI-anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA-mediated addition of amino acids to proteins such as arginyl ati on, ubiquitination, and the like.
  • nucleic acid refers to any deoxyribonucleic acid (DNA) molecule, ribonucleic acid (RNA) molecule, or nucleic acid analogues.
  • a DNA or RNA molecule can be double-stranded or single-stranded and can be of any size.
  • Exemplary nucleic acids include, but are not limited to, chromosomal DNA, plasmid DNA, cDNA, cell- free DNA (cfDNA), mitochondrial DNA, chloroplast DNA.
  • viral DNA mRNA, tRNA, rRNA, long non-coding RNA. siRNA, micro RNA (miRNA or miR). hnRNA, and viral RNA.
  • nucleic analogues include peptide nucleic acid, morpholino- and locked nucleic acid, glycol nucleic acid, and threose nucleic acid.
  • nucleic acid molecule is meant to include fragments of nucleic acid molecules as well as any full- length or non-fragmented nucleic acid molecule, for example.
  • nucleic acid and nucleic acid molecule can be used interchangeably, unless context clearly indicates otherwise.
  • the DNA molecule may further comprise a promoter.
  • promoter refers to a regulatory sequence that initiates transcription.
  • a promoter can be operably linked to first and second polynucleotides of nucleic acid molecules provided herein.
  • promoters included in DNA molecules provided herein include promoters for in vitro transcription (IVT). Any suitable promoter for in vitro transcription can be included in DNA molecules provided herein, such as a T7 promoter, a T3 promoter, an SP6 promoter, and others.
  • the promoter may be a T7 promoter, a T3 promoter, or an SP6 promoter.
  • the RNA molecule is a self-replicating molecule.
  • RNA self-replicating RNA
  • self-transcribing and selfreplicating RNA self-amplifying RNA (saRNA),” and “replicon” may be used interchangeably, unless context clearly indicates otherwise.
  • replicon or “viral replicon” refers to a self-replicating subgenomic RNA derived from a viral genome that includes viral genes encoding non-structural proteins important for viral replication and that lacks viral genes encoding structural proteins.
  • a self-replicating RNA can encode further subgenomic RNAs that are not able to self-replicate.
  • the nucleic acid molecule further comprises a 5’ cap.
  • Any 5’ cap can be included in RNA molecules provided herein, including 5' caps having a Cap 1 structure, a Cap 1 (m6A) structure, a Cap 2 structure, a Cap 0 structure, or any combination thereof.
  • RNA molecules provided herein include a 5’ cap having Cap 1 structure.
  • the nucleic acid molecule further comprises a 5’ UTR and/or a 3’ UTR. In some embodiments, the nucleic acid molecule further comprises a 5’ UTR. In some embodiments, the nucleic acid molecule further comprises a 3’ UTR. In some embodiments, the nucleic acid molecule further comprises a 5’ UTR and a 3‘ UTR. In some embodiments, the 3’ UTR may comprise a poly(A) sequence.
  • protein refers to any polymeric chain of amino acids.
  • peptide and “polypeptide” can be used interchangeably with the term protein, unless context clearly indicates otherwise, and can also refer to a polymeric chain of amino acids.
  • protein encompasses native or artificial proteins, protein fragments and polypeptide analogs of a protein sequence.
  • a protein may be monomeric or polymeric.
  • protein encompasses fragments and variants (including fragments of variants) thereof, unless otherwise contradicted by context.
  • said one or more biological macromolecules is selected from a recombinant protein, a fusion protein, an antibody, a monoclonal antibody, a humanized monoclonal antibody, a bivalent antibody, an antibody fragment, an antibodydrug conjugate, a Fc fragment, a Fab fragment, a Fab' fragment, a (Fab')2 fragment, a Fv fragment, and a scFv fragment.
  • the one or more biological macromolecules is a recombinant protein.
  • the one or more biological macromolecules is a fusion protein.
  • the one or more biological macromolecules is an antibody.
  • the one or more biological macromolecules is a monoclonal antibody . In another embodiment, the one or more biological macromolecules is a humanized monoclonal antibody. In another embodiment, the one or more biological macromolecules is a bivalent antibody. In another embodiment, the one or more biological macromolecules is an antibody fragment. In another embodiment, the one or more biological macromolecules is an antibody-drug conjugate. In another embodiment, the one or more biological macromolecules is a Fc fragment. In another embodiment, the one or more biological macromolecules is a Fab fragment. In another embodiment, the one or more biological macromolecules is a Fab' fragment. In another embodiment, the one or more biological macromolecules is a (Fab')2 fragment. In another embodiment, the one or more biological macromolecules is a Fv fragment. In another embodiment, the one or more biological macromolecules is a scFv fragment.
  • the biological macromolecule is a drug selected from a T- cell co-stimulation modulator, an antagonist of one or more interleukin receptors, an antagonist of one or more interleukins, an interferon-gamma antagonist, a tissue necrosis factor-alpha antagonist, and a transforming growth factor-beta agonist.
  • said drug is a T-cell co-stimulation modulator.
  • said drug is an antagonist of one or more interleukin receptors.
  • said drug is an antagonist of one or more interleukins.
  • said drug is an interferon-gamma antagonist.
  • said drug is a tissue necrosis factoralpha antagonist.
  • said drug is a transforming growth factor-beta agonist.
  • protein molecule encoded by the nucleic acid molecule, or the protein molecule itself is an antigenic protein or a fragment thereof.
  • the antigenic protein is a viral protein.
  • the viral protein originates from a virus that infects humans.
  • the virus may be a DNA virus, a RNA virus, or a retrovirus.
  • viruses useful with the present invention include, but are not limited to Ebola virus, measles virus, SARS-Coronaviruses, Chikungunya virus, hepatitis viruses, Marburg virus, yellow fever virus, MERS-Coronaviruses, Dengue viruses, Lassa virus, influenza viruses, rhabdovirus or HIV.
  • a hepatitis virus may include hepatitis A, hepatitis B, or hepatitis C virus.
  • An influenza virus may include, for example, influenza A or influenza B.
  • An HIV may include HIV 1 or HIV 2.
  • the antigenic viral sequence may be from a human respiratory syncytial virus, Sudan ebola virus, Bundibugyo virus.
  • Canine distemper virus Canaine pneumovirus, Cedar virus, Cell fusing agent virus, Cetacean morbillivirus, Chandipura virus, Chaoyang virus, Chapare mammarenavirus, Chikungunya virus, Colobus monkey papillomavirus, Colorado tick fever virus, Cowpox virus, Crimean-Congo hemorrhagic fever virus.
  • Culex flavivirus Cupixi mammarenavirus, Dengue virus. Dobrava- Belgrade virus, Donggang virus, Dugbe virus, Duvenhage virus.
  • Eastern equine encephalitis virus Entebbe bat virus, Enterovirus A-D, European bat lyssavirus 1-2, Eyach virus, Feline morbillivirus, Fer-de-Lance paramyxovirus, Fitzroy River virus, Flaviviridae virus, Flexal mammarenavirus, GB virus C. Gairo virus, Gemy circularvirus, Goose paramyoxiviurs SF02, Great Island virus, Guanarito mammarenavirus, Hantaan virus.
  • Hantavirus Z10 Heartland virus, Hendra virus, Hepatitis A/B/C/E, Hepatitis delta virus, Human bocavirus, Human coronavirus, Human endogenous retrovirus K, Human enteric coronavirus, Human genital- associated circular DNA virus-1, Human herpesvirus 1-8, Human immunodeficiency virus 1/2, Huan mastadenovirus A-G, Human papillomavirus, Human parainfluenza virus 1-4.
  • Human paraechovirus Human picobimavirus, Human smacovirus, Ikoma lyssavirus, Ilheus virus, Influenza A-C, Ippy mammarenavirus, Irkut virus, J-virus, JC polyomavirus, Japanese encephalitis virus, Junin mammarenavirus, KI polyomavirus, Kadipiro virus, Kamiti River virus, Kedougou virus, Khujand virus, Kokobera virus, Kyasanur forest disease virus, Lagos bat virus, Langat virus.
  • Lassa mammarenavirus Latino mammarenavirus, Leopards Hill virus, Liao ning virus, Ljungan virus, Lloviu virus, Louping ill virus, Lujo mammarenavirus, Luna mammarenavirus, Lunk virus, Lymphocytic choriomeningitis mammarenavirus.
  • Louis encephalitis virus Louis encephalitis virus, Sunshine virus, TTV-like mini virus, Tacaribe mammarenavirus, Taila virus, Tamana bat virus, Tamiami mammarenavirus, Tembusu virus, Thogoto virus, Thottapalayam virus, Tick-bome encephalitis virus, Tioman virus, Togaviridae virus, Torque teno canis virus, Torque teno douroucouli virus, Torque teno felis virus.
  • RNA vimses that may be detected include one or more of (or any combination of) Coronaviridae virus, a Picomaviridae vims, a Caliciviridae virus, a Flaviviridae vims, a Togaviridae vims, a Bomaviridae, a Filoviridae, a Paramyxoviridae, a Pneumoviridae, a Rhabdoviridae, an Arenaviridae, a Bunyaviridae, an Orthomyxoviridae, or a Deltavirus.
  • the vims is Coronavims, SARS, Poliovirus, Rhinovims, Hepatitis A, Norwalk vims, Yellow fever vims. West Nile vims, Hepatitis C virus, Dengue fever virus, Zika vims, Rubella virus, Ross River vims, Sindbis vims, Chikungunya vims, Boma disease vims, Ebola virus, Marburg vims, Measles vims, Mumps virus, Nipah virus, Hendra vims, Newcastle disease virus. Human respiratory syncytial vims, Rabies virus, Lassa vims, Hantavims, Crimean-Congo hemorrhagic fever vims. Influenza, or Hepatitis D virus.
  • the antigenic protein is a bacterial protein.
  • This may include, but is not necessarily limited to, proteins from gram positive bacteria, gram negative bacteria, or other bacteria, such as Bacillus (e.g., Bacillus anthracis), Mycobacterium (e.g., Mycobacterium tuberculosis, Mycobacterium Leprae), Shigella (e.g., Shigella sonnet, Shigella dysenteriae, Shigella flexneri), Helicobacter (e.g., Helicobacter pylori), Salmonella (e.g., Salmonella enterica, Salmonella typhi. Salmonella typhimurium).
  • Bacillus e.g., Bacillus anthracis
  • Mycobacterium e.g., Mycobacterium tuberculosis, Mycobacterium Leprae
  • Shigella e.g., Shigella sonnet, Shigella dysenteriae, Shigella flexneri
  • Helicobacter e.g
  • Neisseria gonorrhoeae Neisseria meningitidis
  • Moraxella e.g., Moraxella catarrhalis
  • Haemophilus e.g., Haemophilus influenzae
  • Klebsiella e.g., Klebsiella pneumoniae
  • Legionella e.g., Legionella pneumophila
  • Pseudomonas e.g., Pseudomonas aeruginosa
  • Acinetobacter e.g., Acinetobacter baumannii
  • Listeria e.g., Listeria monocytogenes.
  • Bordetella e.g., Bordetella pertussis
  • Enterococcus e.g.. Enterococcus faecalis, Enterococcus faecum
  • Vibrio e.g., Vibrio cholerae
  • Yersinia e.g., Yersinia pestis
  • Burkholderia e.g., Burkholderia cepacia complex
  • Coxiella e.g., Coxiella burnetii
  • Francisella e.g., Francisella tularensis
  • Escherichia e.g., enterotoxigenic, enterohemorrhagic or Shiga toxin-producing E.
  • the antigenic protein is from a eukaryotic organism, including protists and fungi, such as Plasmodium (e.g., Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale, Plasmodium malariae, Plasmodium diarrhea), Candida (e.g., Candida albicans), Aspergillus (e.g., Aspergillus fumigatus), Cryptococcus (e.g., Cryptococcus neoformans), Histoplasma (e.g., Histoplasma capsulatum), Pneumocystis (e.g., Pneumocystis jirovecii), and Coccidiodes (e.g., Coccidiodes immitis).
  • Plasmodium e.g., Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale, Plasmodium malariae, Plasmodium diarrhea
  • Candida e.
  • the second composition comprises a vaccine against Mycobacterium tuberculosis .
  • the vaccine is the Bacillus Calmette- Guerin (BCG) vaccine.
  • the second composition comprises a vaccine against Mpox virus (formerly known as monkeypox virus). In some embodiments, the second composition comprises a vaccine against rabies virus.
  • the pharmaceutical agent further compnses a vaccine vector.
  • the vaccine vector is an adenovirus vector, an adeno- associated virus vector, a herpesvirus vector, a retrovirus vector, a poxvirus vector, a baculovirus vector, a rabies virus vector, or a Sendai virus vector.
  • the herpesvirus vector may include, but is not necessarily limited to, an Epstein-Barr virus vector.
  • the retrovirus vector may include, but is not necessarily limited to, a lentivirus vector or a Moloney Murine Leukemia Virus (MMLV) vector.
  • MMLV Moloney Murine Leukemia Virus
  • the pharmaceutical agent further comprises a vaccine vector of murine origin, such as, but not necessarily limited to, a MMLV vector.
  • the antigenic protein may be a fungal protein.
  • the fungi may include, but are not necessarily limited to, one or more of (or any combination of), Aspergillus, Blastomyces, Candidiasis, Coccidiodomycosis, Cryptococcus neoformans, Cryptococcus gatti, Histoplasma sp. (such as Histoplasma capsulatum), Pneumocystis sp.
  • the fungal protein is a yeast protein.
  • fungal antigenic proteins or nucleic acids coding for these proteins include without limitation proteins from one or more of (or any combination of), Aspergillus species (such as Aspergillus fumnigatus, Aspergillus flavus and Aspergillus clavatus), Cryptococcus sp.
  • a Geotrichum species such as Cryptococcus neoformans, Cryptococcus gattii. Cryptococcus laurentii and Cryptococcus albidus
  • a Geotrichum species such as Cryptococcus neoformans, Cryptococcus gattii. Cryptococcus laurentii and Cryptococcus albidus
  • a Geotrichum species such as Cryptococcus neoformans, Cryptococcus gattii. Cryptococcus laurentii and Cryptococcus albidus
  • Saccharomyces species such as Hansenula species
  • a Candida species such as Candida albicans
  • Kluyveromyces species such as Candida albicans
  • Debaryomyces species such as Pichia species, or combination thereof.
  • the fungus is a mold.
  • Example molds include, but are not limited to, a Penicillium species, a Cladosporium species, a Byssochlamys species, or a combination thereof.
  • the antigenic protein is a helminthic protein.
  • the helminths may include, but are not necessarily limited to, Nematodes, Cestodes, and Trematodes.
  • the antigenic protein is a protozoan protein.
  • protozoan antigenic proteins that can be used in accordance with the disclosed compositions and methods include, but are not necessarily limited to, any one or more of (or any combination of), proteins from Euglenozoa, Heterolobosea, Vaccinonadida, Amoebozoa, Blastocystic, and Apicomplexa.
  • Example Euglenoza include, but are not limited to, Trypanosoma cruzi (Chagas disease), T. brucei gambiense, T. brucei rhodesiense.
  • Example Heterolobosea include, but are not limited to, Naegleria fowleri.
  • Example Vaccinonadid include, but are not limited to, Giardia intestinalis (G. lamblia, G. duodenalis) .
  • Example Amoebozoa include, but are not limited to, Acanthamoeba castellanii, Balamuthia madrillaris, Entamoeba histolytica.
  • Example Blastocystis include, but are not limited to, Blastocystic hominis.
  • Example Apicomplexa include, but are not limited to, Babesia microti, Cryptosporidium parvum, Cyclospora cayetanensis, Plasmodium falciparum. P. vivax, P. ovale, P. malariae, and Toxoplasma gondii.
  • the antigenic protein may be a prion protein, such as misfolded forms of PrP, which are known to cause Creutzfeldt-Jakob disease or other neurodegenerative diseases in humans.
  • the antigenic protein is a tumor-associated antigen (TAA).
  • TAA tumor-associated antigen
  • the terms "tumor associated antigen” or “TAA” are used to describe proteins that are significantly over-expressed in cancer compared to normal cells.
  • the antigenic protein is a neoantigen.
  • neoantigen is used to describe newly formed antigens that have not been previously recognized by the immune system. Neoantigens can arise from altered tumor proteins formed as a result of tumor mutations, or from viral proteins.
  • cancer refers to human cancers and carcinomas, sarcomas, adenocarcinomas, lymphomas, leukemias, etc., including solid and lymphoid cancers, kidney, breast, lung, non-small cell lung, bladder, colon, ovarian, prostate, pancreas, stomach, brain, head and neck, skin, uterine, testicular, glioma, esophagus, and liver cancer, including hepatocarcinoma, lymphoma, including B-acute lymphoblastic lymphoma, nonHodgkin's lymphomas (e.g., Burkitt’s, Small Cell, and Large Cell lymphomas), Hodgkin’s lymphoma, leukemia (including AML, ALL, and CML), or multiple myeloma.
  • solid and lymphoid cancers including solid and lymphoid cancers, kidney, breast, lung, non-small cell lung, bladder, colon, ovarian, prostate, pancreas, stomach,
  • Additional examples include cancer of the thyroid, endocrine system, cervix, melanoma, mesothelioma, sarcoma, medulloblastoma, colorectal cancer, neuroblastoma, glioblastoma multiforme, rhabdomyosarcoma, primary' thrombocytosis, primary' macroglobulinemia, primary' brain tumors, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, lymphomas, thyroid cancer, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenal cortical cancer, neoplasms of the endocrine or exocrine pancreas, medullary thyroid cancer, medullary' thyroid carcinoma, melanoma, colorectal cancer, papillary' thyroid cancer, or hepatocellular carcinoma.
  • leukemia refers broadly to progressive, malignant diseases of the blood-forming organs and is generally characterized by a distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow. Leukemia is generally clinically classified on the basis of (1) the duration and character of the diseaseacute or chronic; (2) the type of cell involved; myeloid (myelogenous), lymphoid (lymphogenous), or monocytic; and (3) the increase or non-increase in the number abnormal cells in the blood-leukemic or aleukemic (subleukemic).
  • Exemplary' leukemias that may be prevented or treated with a compound or method provided herein include, for example, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, acute granulocytic leukemia, chronic granulocytic leukemia, acute promyelocytic leukemia, adult T-cell leukemia, aleukemic leukemia, a leukocythemic leukemia, basophylic leukemia, blast cell leukemia, bovine leukemia, chronic myelocytic leukemia, leukemia cutis, embryonal leukemia, eosinophilic leukemia.
  • lymphoma refers to a group of cancers affecting hematopoietic and lymphoid tissues. It begins in lymphocytes, the blood cells that are found primarily in lymph nodes, spleen, thymus, and bone marrow. Two main types of lymphoma are non-Hodgkin lymphoma and Hodgkin's disease. Hodgkin's disease represents approximately 15% of all diagnosed lymphomas. This is a cancer associated with Reed- Sternberg malignant B lymphocytes. Non-Hodgkin’s lymphomas (NHL) can be classified based on the rate at which cancer grows and the type of cells involved.
  • B-cell lymphomas that may be prevented or treated with a compound or method provided herein include, but are not limited to, small lymphocytic lymphoma, Mantle cell lymphoma, follicular lymphoma, marginal zone lymphoma, extranodal (MALT) lymphoma, nodal (monocytoid B-cell) lymphoma, splenic lymphoma, diffuse large cell B-lymphoma, Burkitt’s lymphoma, lymphoblastic lymphoma, immunoblastic large cell lymphoma, or precursor B-lymphoblastic lymphoma.
  • small lymphocytic lymphoma Mantle cell lymphoma
  • follicular lymphoma marginal zone lymphoma
  • MALT extranodal lymphoma
  • nodal lymphoma nodal lymphocytoid B-cell lymphoma
  • splenic lymphoma diffuse large cell B-lymphoma
  • Exemplary T- cell lymphomas that may be prevented or treated with a compound or method provided herein include, but are not limited to, cutaneous T-cell lymphoma, peripheral T-cell lymphoma, anaplastic large cell lymphoma, mycosis fungoides, and precursor T-lymphoblastic lymphoma.
  • sarcoma generally refers to a tumor which is made up of a substance like the embryonic connective tissue and is generally composed of closely packed cells embedded in a fibrillar or homogeneous substance.
  • Sarcomas that may be prevented or treated with a compound or method provided herein include a chondrosarcoma, fibrosarcoma, lymphosarcoma, melanosarcoma, myxosarcoma, osteosarcoma, Abernethy's sarcoma, adipose sarcoma, liposarcoma, alveolar soft part sarcoma, ameloblastic sarcoma, botryoid sarcoma, chloroma sarcoma, chorio carcinoma, embryonal sarcoma, Wilms' tumor sarcoma, endometrial sarcoma, stromal sarcoma, Ewing's sarcoma, fascial s
  • Jensen's sarcoma Kaposi's sarcoma, Kupffer cell sarcoma, angiosarcoma, leukosarcoma, malignant mesenchymoma sarcoma, parosteal sarcoma, reticulocytic sarcoma, Rous sarcoma, serocystic sarcoma, synovial sarcoma, or telangiectaltic sarcoma.
  • melanoma is taken to mean a tumor arising from the melanocytic system of the skin and other organs.
  • Melanomas that may be prevented or treated with a compound or method provided herein include, for example, acral-lentiginous melanoma, amelanotic melanoma, benign juvenile melanoma, Cloudman's melanoma, S91 melanoma.
  • carcinoma refers to a malignant new growth made up of epithelial cells tending to infiltrate the surrounding tissues and give rise to metastases.
  • exemplary carcinomas that may be prevented or treated with a compound or method provided herein include, for example, medullary thyroid carcinoma, familial medullary thyroid carcinoma, acinar carcinoma, acinous carcinoma, adenocystic carcinoma, adenoid cystic carcinoma, carcinoma adenomatosum.
  • carcinoma of adrenal cortex alveolar carcinoma, alveolar cell carcinoma, basal cell carcinoma, carcinoma basocellulare, basaloid carcinoma, basosquamous cell carcinoma, bronchioalveolar carcinoma, bronchiolar carcinoma, bronchogenic carcinoma, cerebriform carcinoma, cholangiocellular carcinoma, chorionic carcinoma, colloid carcinoma, comedo carcinoma, corpus carcinoma, cribriform carcinoma, carcinoma en cuirasse, carcinoma cutaneum, cylindrical carcinoma, cylindrical cell carcinoma, duct carcinoma, carcinoma durum, embryonal carcinoma, encephaloid carcinoma, epiermoid carcinoma, carcinoma epitheliale adenoides, exophytic carcinoma, carcinoma ex ulcere, carcinoma fibrosum, gelatinifomi carcinoma, gelatinous carcinoma, giant cell carcinoma, carcinoma gigantocellulare, glandular carcinoma, granulosa cell carcinoma, hair-matrix carcinoma, hematoid carcinoma, hepatocellular carcinoma, Hurthle cell carcinoma, hyaline carcinoma, hypemephroid carcinoma, infantile embryonal carcinoma,
  • carcinoma mucocellulare mucoepidermoid carcinoma, carcinoma mucosum, mucous carcinoma, carcinoma myxomatodes, nasopharyngeal carcinoma, oat cell carcinoma, carcinoma ossificans, osteoid carcinoma, papillary carcinoma, periportal carcinoma, preinvasive carcinoma, prickle cell carcinoma, pultaceous carcinoma, renal cell carcinoma of kidney, reserve cell carcinoma, carcinoma sarcomatodes, Schneiderian carcinoma, scirrhous carcinoma, carcinoma scroti, signet-ring cell carcinoma, carcinoma simplex, small-cell carcinoma, solanoid carcinoma, spheroidal cell carcinoma, spindle cell carcinoma, carcinoma spongiosum, squamous carcinoma, squamous cell carcinoma, string carcinoma, carcinoma telangiectaticum, carcinoma telangiectodes, transitional cell carcinoma, carcinoma tuberosum, tuberous carcinoma, verrucous carcinoma, or carcinoma villosum.
  • the pharmaceutical agent further comprises a whole cell cancer vaccine.
  • whole tumor cells used as a vaccine rather than a specific protein or peptide tumor antigen are that the cells provide a source of all potential antigens, eliminating the need to identify the most optimal antigen to target in a particular type of cancer. This way, multiple tumor antigens can be targeted at once, generating immune responses to more than one tumor antigen, thereby bypassing issues of tumor antigen loss.
  • immunized lymphocyte and serologic responses can be exploited to identify novel tumor antigens or categorize the importance of a response to a particular tumor antigen through the comparison of immune responses pre- and post- vaccination and by correlating responses with prognosis.
  • An autologous approach employs tumor cells from the same patient to be treated and ensures that patients are vaccinated with cells containing the same tumor antigens that their tumor expresses.
  • Vaccines made from allogeneic cells circumvent the issue of individualizing each patient's therapy and by using several cell lines derived from different tumors in the vaccine, there is an increased likelihood that the patient's tumor will share antigens (e g., neoantigens or TAAs) expressed by the vaccine cells, including important tumor antigens overexpressed or mutated in a high percentage of that particular cancer.
  • antigens e g., neoantigens or TAAs
  • HLA mismatch between vaccinating cell lines and the patient will result in a response directed against foreign HLA molecules rather than tumor antigens. While anti-HLA responses do develop, they have not been shown to inhibit the tumor antigen response and have actually been associated with clinical response to whole cell vaccination.
  • the second composition comprises two or more biological macromolecules.
  • a first macromolecule of the two or more biological macromolecules comprises an antigenic protein or a fragment thereof
  • a second macromolecule comprises an immunomodulatory protein.
  • the immunomodulatory protein may include, but is not necessarily limited to, a cytokine, a chemokine, or an interleukin.
  • the pharmaceutical agent further compnses a lipid formulation.
  • the lipid formulation is selected from a lipoplex, a liposome, a lipid nanoparticle, a polymer-based earner, an exosome, a lamellar body, a micelle, and an emulsion.
  • the lipid formulation is a liposome selected from a cationic liposome, a nanoliposome, a proteoliposome, a unilamellar liposome, a multilamellar liposome, a ceramide-containing nanoliposome, and a multivesicular liposome.
  • the lipid formulation is a lipid nanoparticle.
  • the lipid nanoparticle has a size of less than about 200 nm. In some aspects, the lipid nanoparticle has a size of less than about 150 nm. In some aspects, the lipid nanoparticle has a size of less than about 100 nm. In some aspects, the lipid nanoparticle has a size of about 55 nm to about 90 nm.
  • the lipid formulation comprises polymer-based carriers such as polyethyleneimine (PEI), lipid nanoparticles and liposomes, nanoliposomes, ceramide- containing nanoliposomes, multivesicular liposomes, proteoliposomes, both natural and synthetically-derived exosomes, natural, synthetic and semi-synthetic lamellar bodies, nanoparticulates, micelles, and emulsions.
  • PEI polyethyleneimine
  • lipid nanoparticles and liposomes such as polyethyleneimine (PEI)
  • nanoliposomes such as lipid nanoliposomes, ceramide- containing nanoliposomes, multivesicular liposomes, proteoliposomes, both natural and synthetically-derived exosomes, natural, synthetic and semi-synthetic lamellar bodies, nanoparticulates, micelles, and emulsions.
  • Conventional liposomes are vesicles that consist of at least one bilayer and an internal aque
  • Bilayer membranes of liposomes are typically formed by amphiphilic molecules, such as lipids of synthetic or natural origin that comprise spatially- separated hydrophilic and hydrophobic domains (Lasic. Trends Biotechnol., 16: 307-321, 1998). Bilayer membranes of the liposomes can also be formed by amphiphilic polymers and surfactants (e.g., polymerosomes, niosomes, etc.). They generally present as spherical vesicles and can range in size from 20 nm to a few microns. Liposomal formulations can be prepared as a colloidal dispersion or they can be lyophilized to reduce stability risks and to improve the shelf-life for liposome-based drugs. Methods of preparing liposomal compositions are known in the art and w ould be within the skill of an ordinary artisan.
  • Liposomes that have only one bilayer are referred to as being unilamellar, and those having more than one bilayer are referred to as multilamellar.
  • the most common types of liposomes are small unilamellar vesicles (SUV), large unilamellar vesicle (LUV), and multilamellar vesicles (MLV).
  • lysosomes, micelles, and reversed micelles are composed of monolay ers of lipids.
  • a liposome is thought of as having a single interior compartment, however some formulations can be multivesicular liposomes (MVL), which consist of numerous discontinuous internal aqueous compartments separated by several nonconcentric lipid bilayers.
  • MDL multivesicular liposomes
  • Liposomes have long been perceived as drug delivery vehicles because of their superior biocompatibility, given that liposomes are basically analogs of biological membranes, and can be prepared from both natural and synthetic phospholipids (Int J Nanomedicine. 2014; 9: 1833-1843).
  • a liposome has an aqueous solution core surrounded by a hydrophobic membrane, hydrophilic solutes dissolved in the core cannot readily pass through the bilayer, and hydrophobic compounds will associate with the bilayer.
  • a liposome can be loaded with hydrophobic and/or hydrophilic molecules.
  • a liposome is used to cany- a nucleic acid such as RNA, the nucleic acid will be contained within the liposomal compartment in an aqueous phase.
  • lipid nanoparticles In contrast to liposomes and cationic liposomes, lipid nanoparticles (LNP) have a structure that includes a single monolayer or bilayer of lipids that encapsulates a compound in a solid phase. Thus, unlike liposomes, lipid nanoparticles do not have an aqueous phase or other liquid phase in its interior, but rather the lipids from the bilayer or monolayer shell are directly complexed to the internal compound thereby encapsulating it in a solid core. Lipid nanoparticles are typically spherical vesicles having a relatively uniform dispersion of shape and size.
  • lipid nanoparticle can have a diameter in the range of from 10 nm to 1000 nm. However, more commonly they are considered to be smaller than 120 nm or even 100 nm.
  • the lipid shell is formulated to include an ionizable cationic lipid which can complex to and associate with the negatively charged backbone of the nucleic acid core.
  • Ionizable cationic lipids with apparent pKa values below about 7 have the benefit of providing a cationic lipid for complexing with the nucleic acid’s negatively charged backbone and loading into the lipid nanoparticle at pH values below the pKa of the ionizable lipid where it is positively charged.
  • the lipid nanoparticle can adopt a relatively neutral exterior allowing for a significant increase in the circulation half-lives of the particles following i.v. administration.
  • lipid nanoparticles offer many advantages over other lipid- based nucleic acid delivery systems including high nucleic acid encapsulation efficiency, potent transfection, improved penetration into tissues to deliver therapeutics, and low levels of cytotoxicity and immunogenicity.
  • the lipid formulation is selected from a lipoplex, a liposome, a lipid nanoparticle, a polymer-based earner, an exosome, a lamellar body, a micelle, and an emulsion.
  • the lipid formulation further comprises a polyethylene glycol (PEG)-lipid conjugate.
  • PEG polyethylene glycol
  • PEGylation has been used to stabilize lipid formulations and their payloads through physical, chemical, and biological mechanisms.
  • Detergent-like PEG lipids e.g., PEG-DSPE
  • PEG-DSPE can enter the lipid formulation to form a hydrated layer and steric barrier on the surface.
  • the surface layer can be generally divided into two types, brush-like and mushroom-like layers.
  • PEG-DSPE-stabilized formulations PEG will take on the mushroom conformation at a low degree of PEGylation (usually less than 5 mol%) and will shift to brush conformation as the content of PEG-DSPE is increased past a certain level (Journal of Nanomaterials. 2011 ;2011 : 12).
  • PEGylation leads to a significant increase in the circulation half-life of lipid formulations (Annu. Rev. Biomed. Eng. 2011 Aug 15; 13():507-30; J. Control Release. 2010 Aug 3; 145(3): 178-81).
  • PEG-lipids include, but are not limited to, PEG coupled to dialkyloxypropyls (PEG-DAA), PEG coupled to diacylglycerol (PEG-DAG), PEG coupled to phospholipids such as phosphatidylethanolamine (PEG-PE), PEG conjugated to ceramides, PEG conjugated to cholesterol or a derivative thereof, and mixtures thereof.
  • PEG-DAA dialkyloxypropyls
  • PEG-DAG PEG coupled to diacylglycerol
  • PEG coupled to phospholipids such as phosphatidylethanolamine (PEG-PE)
  • PEG conjugated to ceramides PEG conjugated to cholesterol or a derivative thereof, and mixtures thereof.
  • PEG is a linear, water-soluble polymer of ethylene PEG repeating units with two terminal hydroxyl groups.
  • PEGs are classified by their molecular weights and include the following: monomethoxypolyethylene glycol (MePEG-OH), monomethoxypolyethylene glycol- succinate (MePEG-S), monomethoxypolyethylene glycol-succinimidyl succinate (MePEG-S-NHS), monomethoxypolyethylene glycol-amine (MePEG-NH2), monomethoxypolyethylene glycol-tresylate (MePEG-TRES), monomethoxypolyethylene glycol-imidazolyl-carbonyl (MePEG-IM), as well as such compounds containing a terminal hydroxyl group instead of a terminal methoxy group (e.g., HO-PEG-S, HO-PEG-S-NHS, HO-PEG-NH2).
  • MePEG-OH monomethoxypoly
  • the pharmaceutical agent further comprises one or more cryoprotectants.
  • the cryoprotectant comprises glycerol.
  • the composition or compositions comprise between about 5% and about 50% glycerol.
  • the storage medium comprises about 20% glycerol.
  • the cryoprotectant comprises sucrose.
  • the composition comprises between about 5% and about 30% sucrose.
  • the composition comprises about 10% sucrose.
  • the cryoprotectant comprises dimethylsulfoxide (DMSO).
  • the DMSO is at a concentration of between 2% and 10%.
  • the one or more cryoprotectants are selected from sucrose, glycerol, or a combination of sucrose and glycerol.
  • the second composition is a liquid, semi-liquid, solid, frozen, lyophilized, or vacuum-dried formulation.
  • the second composition is in a liquid formulation.
  • the second composition is in a semi-liquid formulation.
  • the second composition is in a solid formulation.
  • the second composition is in a frozen formulation.
  • the second composition is in a lyophilized formulation.
  • the second composition is a vacuum-dried composition.
  • the first composition and/or the second composition may be used in combination with a gel or cream.
  • the gel or cream may or may not further comprise hydrogen peroxide.
  • the gel or cream does not further comprise hydrogen peroxide.
  • the gel or cream further comprises hydrogen peroxide.
  • Such gels or creams are generally commercially available any may contain from about 0.5% w/w to about 50% w/w hydrogen peroxide.
  • a gel containing about 1% w/w, or about 2% w/w, or about 3% w/w, or about 4% w/w, or about 6% w/w, or about 7% w/w, or about 8% w/w, or about 9% w/w, or about 10% w/w, or about 15% w/w, or about 20% w/w, or about 25% w/w, or about 30% w/w, or about 40% w/w, or about 45% w/w, or about 50% w/w hydrogen peroxide may be used in any of the methods disclosed herein in combination with the first composition and the second composition.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongillcr, and a second composition comprising a vaccine against mycobacteria.
  • the vaccine against mycobacteria is BCG.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongillcr, and a second composition comprising a vaccine against Mpox virus (formerly known as monkeypox virus).
  • the agent comprises a first composition comprising Spongillcr, and a second composition comprising a vaccine against Mpox virus (formerly known as monkeypox virus).
  • the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongillc, and a second composition comprising a vaccine against rabies virus.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the agent further comprises a T-cell co-stimulation modulator, an antagonist of one or more interleukin receptors, an antagonist of one or more interleukins, an interferon-gamma antagonist, a tissue necrosis factor-alpha antagonist, or a transforming growth factor-beta agonist.
  • the agent further comprises an antagonist of one or more interleukin receptors.
  • said interleukin receptor is interleukin 1 receptor (IL-1R).
  • IL-5R IL-6R
  • IL-10R IL-12R
  • IL-13R IL-17R
  • IL-17RA IL-21R
  • IL-22R IL-23R
  • IL-31R IL-35R
  • IL-35R IL-35R
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the agent further comprises an antagonist of one or more interleukins.
  • said interleukin is IL-1, IL-1A, IL-1B, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, IL-13, IL-17, IL-17A, IL-21, IL-22, IL-23, IL-31, IL-35, or a combination of these.
  • the present disclosure provides a second composition comprising a live-attenuated virus or other pathogen, an inactivated virus or other pathogen, virus-like particles, a subunit vaccine, a conjugate vaccine, and/or a dendritic cell vaccine.
  • Live-attenuated vaccines comprise attenuated forms of a pathogen that is still infectious but exhibits reduced virulence while still stimulating a strong and effective immune response.
  • Inactivated vaccines comprise pathogenic entities or organisms that have either been killed or inactivated so that they are no longer infectious but are still able to induce an immune response in a recipient.
  • Virus-like particles are molecules that closely resemble viruses, but are non-infectious because they contain no viral genetic material. The components of virus-like particles are able to induce an effective immune response.
  • subunit vaccines do not contain the whole pathogen, but contain only the antigenic parts such as proteins, polysaccharides, or peptides.
  • Conjugate vaccines are a ty pe of subunit vaccine which combines a weak antigen with a strong antigen as a carrier so that the immune system has a stronger response to the weak antigen. Most vaccines contain a single antigen; however, some antigens do not elicit a strong immune response. Conjugate vaccines are great for invoking immune responses against weak antigens because the weak antigen is covalently attached to a strong antigen, thereby eliciting a stronger immunological response to the weak antigen.
  • Dendritic cells are professional antigen-presenting cells that process antigen material and present it on their surface to T cells, which then carry out effector functions of the adaptive immune system. Dendritic cells can be isolated from a patient, loaded with an antigen of choice, and then returned to the patient to induce the desired immune response. Methods for generating any or all of these types of vaccines are well known in the art.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 25 x 10 4 colony-forming units per gram (CFU/g).
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 10 x 10 4 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 5 x 10 4 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 1 x 10 4 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 5 x 10 3 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 1 x 10 4 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 10,000 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 7,500 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 5,000 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 2.500 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 2,000 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 1,500 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 1,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 500 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 250 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 150 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 100 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 75 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 50 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 25 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 10 CFU/g.
  • the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 1 CFU/g.
  • the combined aerobic and anaerobic microbial content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or dry heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels.
  • the combined aerobic and anaerobic microbial content of the Spongilla materials may be measured by methods known to those of ordinary skill in the art, such as those described in the United States Pharmacopeia method USP ⁇ 61> (Microbial Enumeration Tests).
  • the pharmaceutical compositions disclosed herein may be in unit dosage forms suitable for single administration of precise dosages.
  • any of the methods or kits disclosed herein wherein the unit dosage forms of the first composition and/or the second composition are suitable for two administrations, three administrations, four administrations, five administrations, six administrations, seven administrations, eight administrations, 9 administrations, 10 administrations, 11 administrations, 12 administrations, 13 administrations, 14 administrations, 15 administrations, 16 administrations, 17 administrations, 18 administrations, 19 administrations, 20 administrations, 21 administrations, 22 administrations, 23 administrations, 24 administrations, 25 administrations, 26 administrations, 27 administrations. 28 administrations.
  • 29 administrations 30 administrations, administrations for two months, administrations for three months, administrations for four months, administrations for five months, administrations for six months, administrations for seven months, administrations for eight months, administrations for nine months, administrations for ten months, administrations for eleven months, or administrations for 12 months.
  • compositions disclosed herein may vary according to the composition being used, the mode of administration, and the particular site of administration. Those skilled in the art using conventional dosagedetermination tests in view of the experimental data for a given composition may ascertain optimal dosages for a given set of conditions.
  • compositions and formulations disclosed herein including activity, pharmacokinetics, pharmacodynamics, and bioavailability thereof), the physiological condition of the subject (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication) or cells, the nature of the pharmaceutically acceptable carrier mg/kg or carriers in the formulation, and the route of administration.
  • an effective or therapeutically effective amount may vary depending on whether the one or more compositions and formulations disclosed herein is administered alone or in combination with other drug(s), other therapy/therapies or other therapeutic method(s) or modality/ modalities.
  • Dosage regimens using the first composition and the second composition may be adjusted to provide the optimum desired response.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects; each unit containing a predetermined quantity of the compositions disclosed herein, calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the compositions disclosed herein are dictated by and directly dependent on (a) the characteristics of the composition and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such a composition for the prophylactic or therapeutic treatment a particular condition in a subject.
  • the dose and dosing regimen using the compositions disclosed herein may be adjusted in accordance with methods well-known in the prophylactic and therapeutic arts. That is, the maximum tolerable dose can be readily established, and the effective amount providing a detectable therapeutic benefit to a subject may also be determined, as can the temporal requirements for administering each agent to provide a detectable prophylactic and/or therapeutic benefit to the subject. Accordingly, while certain dose and administration regimens are exemplified herein, these examples in no way limit the dose and administration regimen that may be provided to a subject in practicing the presently disclosed methods.
  • intradermal vaccination offers a dose-sparing benefit, which reduces the volume of vaccine by 60-80% compared to intramuscular vaccination (Blumberg, L.H.(2018) Rabies vaccines: WHO position paper - April 2018 Vaccine 36:5500-5503).
  • Dose-sparing is beneficial to immunization programs because it reduces the costs of purchase, distribution, and storage of vaccines, thereby leading to increased vaccine availability and effectiveness.
  • dosage values may vary 7 with the type and severity of the condition to be alleviated or prevented and may include single or multiple doses. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary 7 only and are not intended to limit the scope or practice of the claimed composition. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamic parameters, which may include clinical effects such as toxic effects and/or laboratory values. The embodiments disclosed herein are intended to encompass intrasubject dose-escalation as determined by the skilled artisan. Determining appropriate dosages and regimens for administration of the prophylactic or therapeutic agent are well-known in the relevant art and would be understood to be encompassed by the skilled artisan once provided the teachings disclosed herein.
  • the present disclosure provides a method of delivering a vaccine to a subj ect in need thereof, comprising administering an effective amount of any of the pharmaceutical agents described herein to the skin or the dermis of the subject.
  • the term '‘in need of treatment’’ as used herein refers to a judgment made by a caregiver (e.g., physician, nurse, nurse practitioner, or individual in the case of humans; veterinarian in the case of animals, including non-human mammals) that a subject requires or will benefit from a therapeutic or prophylactic treatment. This judgment is made based on a variety of factors that are in the realm of a caregiver’s expertise, but that include the knowledge that the subject is ill, or will be ill, as the result of a condition that is preventable or treatable by the compositions of the invention.
  • a caregiver e.g., physician, nurse, nurse practitioner, or individual in the case of humans; veterinarian in the case of animals, including non-human mammals
  • the term “subject” or “patient” refers to a human or non-human animal.
  • the subject is a human.
  • the subject or patient is in need of prevention or treatment with the composition as described herein, e g., is at risk of becoming infected with an infectious agent or may develop cancer.
  • the subject is a mammal, including but not limited to humans, dogs, cats, cattle, sheep, and goats.
  • the subject is a dog.
  • the subject is a cat.
  • the subject is a cow.
  • the subject is a sheep.
  • the subject is a goat.
  • the term "effective amount” or “therapeutically effective amount” refers to that amount of a Spongilla particle, protein, nucleic acid molecule, composition, or pharmaceutical composition described herein that is sufficient to effect the intended application, including but not limited to inducing an immune response and/or disease treatment, as defined herein.
  • the therapeutically effective amount may vary’ depending upon the intended application (e.g.. inducing an immune response, treatment, application in vivo), or the subject or patient and disease condition being prevented or treated, e g., the weight and age of the subject, the species, the severity' of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary' skill in the art.
  • the term also applies to a dose that will induce a particular response in a target cell.
  • the specific dose will vary depending on the particular Spongilla particle, protein, nucleic acid molecule, composition, or pharmaceutical composition chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • the pharmaceutical agent is delivered intradermally.
  • the amount of the first composition comprising Spongilla that is administered to the subject is from about 0.5 grams to about 50 grams. In some embodiments, the amount of the first composition is measured as a dry' weight. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 40 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 35 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 30 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 25 grams.
  • the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 20 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 15 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 10 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.75 grams to about 20 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.75 grams to about 15 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.75 grams to about 10 grams.
  • the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 20 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 15 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 10 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 9 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 8 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subj ect is from about 1 gram to about 7 grams.
  • the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 6 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 5 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 4 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 3 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to 2 grams. In some embodiments, the amount of Spongilla administered, such as those disclosed above, are each measured as a dry weight.
  • the amount of the first composition including Spongilla applied to the skin of the subject is about 0.5 grams, or about 0.75 grams, or about 1 gram, or about 1.25 grams, or about 1.5 grams, or about 1.75 grams, or about 2 grams, or about 2.25 grams, or about 2.5 grams, or about 2.75 grams, or about 3 grams, or about 3.25 grams, or about 3.5 grams, or about 3.75 grams, or about 4 grams, or about 4.25 grams, or about 4.5 grams, or about 4.75 grams, or about 5 grams, or about 5.25 grams, or about 5.5 grams, or about 5.75 grams, or about 6 grams, or about 6.25 grams, or about 6.5 grams, or about 7 grams, or about 7.25 grams, or about 7.5 grams, or about 7.75 grams, or about 8 grams, or about 8.25 grams, or about 8.5 grams, or about 8.75 grams, or about 9 grams, or about 9.25 grams, or about 9.5 grams, or about 9.75 grams, or about 10 grams, or about 11 grams, or about 12 grams, or
  • the first composition is administered first, and the second composition is administered second.
  • the second composition is administered first, and the first composition is administered second.
  • the first and second composition are each provided separately in separate containers.
  • the first and second composition are each provided in lyophilized, vacuum- dried, or liquid form in their separate containers.
  • the first composition is lyophilized and the second composition is vacuum-dried or vice versa.
  • the first composition is lyophilized and the second composition is in liquid form or vice versa.
  • the first composition is vacuum-dried and the second composition is in liquid form or vice versa. Any other combination of the above is also envisioned.
  • the first composition and the second composition are administered together. In some embodiments, the first composition and the second composition are administered at the same time. In some embodiments, the first composition and the second composition are administered as a single composition. For example, the first composition and the second composition may be combined prior to administration, by the healthcare professional doing the administering.
  • the first composition and the second composition may be a single composition.
  • the single composition comprising the first and second composition together is provided in one container.
  • the single composition is provided in lyophilized, vacuum-dried, or liquid form.
  • the second composition is lyophilized or vacuum-dried and is reconstituted prior to administration.
  • the reconstitution reagent may comprise saline, buffered saline, or a hydrogel.
  • the present disclosure provides a method of preventing an infection in a subject in need thereof, comprising administering any of the pharmaceutical agents described herein by applying an effective amount of the agent to the skin of the subject.
  • the term “prevent” refers to a decrease in the occurrence of disease symptoms in a patient.
  • the prevention may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment.
  • the treatment may prevent severe disease, hospitalization and/or death.
  • the present disclosure provides a method of preventing or treating cancer in a subject in need thereof, comprising administering any of the pharmaceutical agents described herein by applying an effective amount of the agent to the skin of the subject.
  • the present disclosure provides a method of inducing an immune response in a subject in need thereof comprising administering an effective amount of any of the pharmaceutical agents described herein to the skin of the subject.
  • immune response can be induced using the methods provided herein, including adaptive and innate immune responses.
  • immune responses induced using the methods provided herein include an antibody response, a cellular immune response, or both an antibody response and a cellular immune response.
  • the pharmaceutical agent is administered one time. In some embodiments, the pharmaceutical agent is administered two or more times. In some embodiments, the pharmaceutical agents and/or compositions described herein are administered two or more times at three week intervals, four week intervals, five week intervals, six week intervals, seven week intervals, eight week intervals, nine week interv als, ten week intervals, eleven week intervals, or twelve week intervals.
  • the pharmaceutical agents and/or compositions described herein are administered two or more times at one month intervals, two month interv als, three month intervals, four month intervals, five month intervals, six month intervals, seven month intervals, eight month intervals, nine month intervals, ten month intervals, eleven month intervals, or twelve month intervals.
  • the pharmaceutical agent is applied to mucosal surfaces.
  • the skin of the subject is cleaned using a non-comedogenic cleanser, water, or a combination of a non-comedogenic cleanser and water following application of the first composition comprising Spongilla.
  • the skin of the subject is cleaned using a non-comedogenic cleanser, water, or a combination of a non- comedogenic cleanser and water following application of the second composition comprising one or more drugs to the skin of the subject.
  • Non-comedogenic cleansers are those formulated not to cause blocked pores in the skin of subjects to which such cleansers are applied.
  • the compositions may be used in combination with one or more additional compositions.
  • the one or more additional compositions may be administered sequentially or simultaneously with the first composition and/or the second composition disclosed herein.
  • the additional compositions is administered to a subject prior to, at the same time as, or following administration of the first composition and/or the second composition disclosed herein.
  • the additional composition is administered to the subject prior to the administration of the first composition and/or the second composition disclosed herein.
  • the additional composition is administered to the subject at the same time the first composition and/or the second composition disclosed herein are administered to the subject.
  • the additional composition is administered to the subject following to the administration of the first composition and/or the second composition disclosed herein.
  • the present disclosure provides a kit for inducing an immune response in a subject in need thereof, wherein the kit comprises a first composition comprising Spongilla and a second composition comprising one or more biological macromolecules.
  • the kit or compositions described herein may include a second composition comprising one or more drugs selected from a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials.
  • said one or more drugs is a biological macromolecule.
  • said one or more drugs is a chemical compound.
  • the one or more drugs is a mixture of chemical compounds.
  • the one or more drugs is a biological macromolecule.
  • the one or more drugs is an extract made from biological materials.
  • the one or more biological macromolecules comprise a nucleic acid molecule, a protein molecule, or a combination thereof, as described in detail above.
  • the kit further comprises a reconstitution agent.
  • the reconstitution agent may include, but is not necessarily limited to, saline, buffered saline, or a hydrogel.
  • the kit further comprises one or more antiseptic reagents.
  • the one or more antiseptic reagents is a hydrogen peroxide solution, a hydrogen peroxide-containing hydrogel, a non-comedogenic cleanser, a solution of isopropyl alcohol.
  • the one or more antiseptic reagents is any acceptable skin antiseptic agent commonly used in surgery, including, but not limited to, chlorhexidine, chlorhexidine-gluconate, iodine, hexachlorophine, benzalkonium chloride, and any reconstitutions thereof.
  • the kit further comprises instructions for use.
  • compositions and kits described herein do not require cold chain storage.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oceanography (AREA)
  • Zoology (AREA)
  • Communicable Diseases (AREA)
  • Pulmonology (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present disclosure is drawn to compositions, methods, and kits for inducing immune responses in a subject in need thereof. In some embodiments, the disclosure is drawn to a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongilla; and a second composition comprising one or more biological macromolecules. In some embodiments, the second composition comprises a vaccine agent to treat or prevent infectious disease. In some embodiments, the second composition comprises a vaccine agent to treat or prevent cancer.

Description

COMPOSITIONS FOR INDUCING IMMUNE RESPONSES
CROSS-REFERENCES TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application No. 63/479.897, filed January 13, 2023, which is incorporated herein by reference in its entirety and for all purposes.
FIELD
[0002] The present disclosure relates to the use of compositions comprising Spongilla for the intradermal delivery of vaccines.
BACKGROUND
[0003] Spongilla is a genus of freshwater sponge. Compositions derived from Spongilla can be used for a number of skin conditions, including acne. An important component of materials derived from Spongilla are the inorganic siliceous spicules that comprise the skeletal structure of Spongilla.
[0004] Traditional delivery' methods for vaccines via needle injection have a variety of drawbacks, including causing anxiety in patients with a fear of needles, injection site pain, and injection site reactions. Therefore, there is a need for improved methods for delivering vaccines, such as by topical application to the skin of the subject, which avoids the drawbacks of needle injection that can lead to undesired side effects or adverse events. A topical mode of application may also permit an effective amount of the one or more vaccine antigens to penetrate the skin of the subject so that an effective amount of the one or more antigens is delivered to the desired site of action, such as the dermis of the subject.
SUMMARY
[0005] Surprisingly, it has been discovered that Spongilla spicules can penetrate the stratum comeum and entire dermis of the skin of a subject during application and promote the penetration of topically-applied compositions, such as a hydrogel composed of polymers, copolymers, and cross-linked versions thereof into the skin of the subject, such as the dermis, to permit or improve the delivery of an effective amount of a vaccine formulation to the site of action necessary to effectively induce an immune response in the subject. It was further discovered that the spicules derived from Spongilla are useful in facilitating and permitting certain therapeutic and prophylactic compounds and compositions to penetrate into the skin of subjects to which the spicules are applied, which compounds and compositions would otherwise not be able to penetrate the skin of the subject in order to reach their targets and treat and/or prevent certain infections and/or cancer. The materials comprising Spongilla used herein may comprise all organic and/or inorganic compounds and materials that are part of the naturally-occurring Spongilla. or may include only a portion of the organic and/or inorganic compounds and materials that are part of the naturally-occurring Spongilla.
[0006] In one aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongilla,' and a second composition comprising one or more biological macromolecules.
[0007] In some embodiments, the one or more biological macromolecules comprise a nucleic acid molecule, a protein molecule, or a combination thereof.
[0008] In some embodiments, the nucleic acid molecule comprises an RNA or a DNA molecule encoding an antigenic protein or a fragment thereof.
[0009] In some embodiments, the DNA molecule further comprises a promoter. In some embodiments, the promoter is a T7 promoter, a T3 promoter, or an SP6 promoter.
[00010] In some embodiments, the RNA molecule is a self-replicating molecule.
[00011] In some embodiments, the nucleic acid molecule further comprises a 5’ cap.
[00012] In some embodiments, the nucleic acid molecule further comprises a 5’ UTR and/or a 3’ UTR. In some embodiments, the 3’ UTR comprises a poly(A) sequence.
[00013] In some embodiments, the protein molecule is an antigenic protein or a fragment thereof.
[00014] In some embodiments, the antigenic protein is a viral protein. In some embodiments, the viral protein originates from a human virus. [00015] In some embodiments, the antigenic protein is a bacterial protein.
[00016] In some embodiments, the second composition comprises a vaccine against Mycobacterium tuberculosis. In some embodiments, the vaccine is the Bacillus Calmette- Guerin (BCG) vaccine.
[00017] In some embodiments, the pharmaceutical agent further comprises a vaccine vector of murine origin.
[00018] In some embodiments, the antigenic protein is a fungal protein. In some embodiments, the antigenic protein is a helminthic protein. In some embodiments, the antigenic protein is a protozoan protein. In some embodiments, the antigenic protein is a prion protein. In some embodiments, the antigenic protein is a cancer protein.
[00019] In some embodiments, the pharmaceutical agent further comprises a whole cell cancer vaccine.
[00020] In some embodiments, the protein molecule is an immunomodulatory protein.
[00021] In some embodiments, the second composition comprises two or more biological macromolecules.
[00022] In some embodiments, a first macromolecule of the two or more biological macromolecules comprises an antigenic protein or a fragment thereof, and a second macromolecule comprises an immunomodulatory protein.
[00023] In some embodiments, the pharmaceutical agent further comprises a lipid formulation. In some embodiments, the lipid formulation is selected from a lipoplex, a liposome, a lipid nanoparticle, a polymer-based earner, an exosome, a lamellar body, a micelle, and an emulsion. In some embodiments, the lipid formulation further comprises a polyethylene glycol (PEG)-lipid conjugate.
[00024] In some embodiments, the agent further comprises one or more cryoprotectants. In some embodiments, the one or more cryoprotectants are selected from sucrose, glycerol, or a combination of sucrose and glycerol. [00025] In some embodiments, the Spongilla is in the form of a powder. Tn some embodiments, the powder comprises particles that are substantially uniform in size.
[00026] In some embodiments, not less than 90% of the particles comprising the Spongilla powder pass through a US 70-mesh screen.
[00027] In some embodiments, the particles comprising the Spongilla powder have an average length of from about 50 pm to about 500 pm.
[00028] In some embodiments, the particles comprising the Spongilla powder have an average diameter of from about 5 pm to about 50 pm.
[00029] In some embodiments, the particles comprising the Spongilla powder have an aspect ratio of from about 1 to 100.
[00030] In some embodiments, the particles comprising the Spongilla powder comprise only inorganic Spongilla spicules and any materials that are naturally associated with the spicules.
[00031] In some embodiments, the second composition is a lyophilized or vacuum-dried composition. In some embodiments, the second composition is a liquid composition.
[00032] In some embodiments, the first composition is in the form of a paste, gel, or topical cream. In some embodiments, the paste further comprises water, saline, or hydrogen peroxide. In some embodiments, the paste is prepared by mixing a powder comprising Spongilla and water or saline or hydrogen peroxide.
[00033] In some embodiments, the first composition is packaged prior to use. In some embodiments, the first composition is prepared by treating with gamma radiation.
[00034] In some embodiments, the Spongilla is Spongilla lacustris.
[00035] In some embodiments, the first composition has a residual moisture content of not more than about 5%.
[00036] In some embodiments, the amount of the first composition comprising Spongilla is from about 0.5 grams to about 50 grams. [00037] In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongillcr, and a second composition comprising a vaccine against mycobacteria.
[00038] In some embodiments, the vaccine against mycobacteria comprises BCG.
[00039] In yet another aspect, the present disclosure provides a method of delivering a vaccine to a subject in need thereof, comprising administering an effective amount of any of the pharmaceutical agents described herein to the subject.
[00040] In some embodiments, the pharmaceutical agent is delivered intradermally.
[00041] In some embodiments, the first composition is administered first, and the second composition is administered second. In some embodiments, the first composition and the second composition are administered together.
[00042] In some embodiments, the second composition is lyophilized or vacuum-dried and is reconstituted prior to administration.
[00043] In yet another aspect, the present disclosure provides a method of preventing an infection in a subject in need thereof, comprising administering any of the pharmaceutical agents described herein to the subject.
[00044] In yet another aspect, the present disclosure provides a method of preventing cancer in a subject in need thereof, comprising administering any of the pharmaceutical agents described herein to the subject.
[00045] In yet another aspect, the present disclosure provides a method of inducing an immune response in a subject in need thereof comprising administering an effective amount of any of the pharmaceutical agents described herein to the subject.
[00046] In some embodiments, the pharmaceutical agent is administered one time. In some embodiments, wherein the pharmaceutical agent is administered two or more times.
[00047] In some embodiments, the pharmaceutical agent is applied to mucosal surfaces.
[00048] In some embodiments, the pharmaceutical agent is administered intradermally. [00049] In yet another aspect, the present disclosure provides a kit for inducing an immune response in a subject in need thereof, wherein the kit comprises a first composition comprising Spongilla and a second composition comprising one or more biological macromolecules.
[00050] In some embodiments, the one or more biological macromolecules comprise a nucleic acid molecule, a protein molecule, or a combination thereof.
[00051] In some embodiments, the kit may further comprise a reconstitution agent. In some embodiments, the kit may further comprise an antiseptic reagent.
[00052] In some embodiments, the kit may further comprise instructions for use.
[00053] In yet another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongilla, and a second composition comprising a whole cell cancer vaccine.
BRIEF DESCRIPTION OF THE DRAWINGS
[00054] FIG. 1 shows the magnified surface of a t pical Spongilla composition.
DETAILED DESCRIPTION
[00055] The singular form "a”, "an”, and “the” include plural references unless the context clearly dictates otherwise. For example, the term “a cell” includes one or more cells, including mixtures thereof. “A and/or B” is used herein to include all of the following alternatives: “A”, “B”, “A or B”, and “A and B”.
[00056] As used herein, the term “about” means either within plus or minus 10% of the provided value, or rounded to the nearest significant figure, in all cases inclusive of the provided value. Where ranges are provided, they are inclusive of the boundary7 values.
[00057] As used herein, the terms “applied,” “applying,” “administration,” “administering,” “used,” or “deliver” means the delivery of a composition disclosed herein to a subject, in particular to the skin of the subject, by an administration route including, but not limited to, intradermal, subcutaneous, topical, or any combinations thereof. In some embodiments disclosed herein, the compositions disclosed herein are administered to the subject, in particular to the skin of the subject, by intradermal administration.
[00058] As used herein, the term "‘aspect ratio” means with respect to the particles of Spongilla described herein, the ratio between the average length of the particles to the average diameter of the particles.
[00059] As used herein, the terms “combination” and “in combination with” mean the application, use, or administration of one or more of the compositions disclosed herein, sequentially or simultaneously. It includes dosing simultaneously, or within minutes or hours of each other, or on the same day, or on alternating days, or using the compositions disclosed herein on a daily basis, or multiple days per week, or weekly basis, for example, while administering another composition on the same day or alternating days or weeks or on a periodic basis during a time simultaneous therewith or concurrent therewith, or at least a part of the time during which the composition disclosed herein is applied, used or administered.
[00060] The term "Spongilla" as used herein means a genus of freshwater sponges in the family Spongillidae, including, but not limited to, Spongillct lacustris, S. fragilis I.eidy. and Ephydatia fluviatilis. The term “Spongilla lacustris” as used herein means a species of sponge of the freshwater sponge family Spongillidae. FIG. 1 shows a scanning electron microscopy view of the structure of Spongillidae.
[00061] The terms “composition comprising Spongilla, ” “powders comprising Spongilla," “materials comprising Spongilla," “Spongilla in the form of a powder,” and the like, as used herein, mean materials comprising Spongilla derived from raw Spongilla that is harvested and processed and may include all the various components of the Spongilla following harvest, including all organic and/or inorganic compounds and materials that are part of the naturally-occurring Spongilla, or may include only a portion of the organic and/or inorganic compounds and materials that are part of the naturally-occurring Spongilla. In one aspect is provided any of the methods, compositions or kits disclosed herein, wherein the Spongilla materials comprise all or substantially all the organic and inorganic materials derived from the naturally occurring Spongilla. In another aspect is provided any of the methods, compositions or kits disclosed herein, wherein the Spongilla materials comprise (a) only the inorganic spicules and any materials that are naturally associated with the spicules, or (b) substantially purified inorganic spicules and any materials that are naturally associated with the spicules, or (c) purified inorganic spicules and any materials that are naturally associated with the spicules that are a component part of naturally -occurring Spongilla. In another aspect is provided any of the methods, compositions or kits disclosed herein, wherein the Spongilla materials comprise only the inorganic spicules and any materials that are naturally associated with the spicules. In another aspect is provided any of the methods, compositions or kits disclosed herein, wherein the Spongilla materials comprise substantially purified inorganic spicules and any materials that are naturally associated with the spicules. In another aspect is provided any of the methods, compositions or kits disclosed herein, wherein the Spongilla materials comprise purified inorganic spicules and any materials that are naturally associated with the spicules that are a component part of naturally occurring Spongilla.
[00062] The terms “composition comprising one or more sponges/’ “powders comprising one or more sponges, “materials comprising one or more sponges,” “sponge in the form of a powder,” and the like, as used herein, mean materials derived from one or more sponges that is harvested and processed and may include all the various components of the sponge following harvest, including all organic and/or inorganic compounds and materials that are part of the naturally-occurring sponge, or any sponges that are specially grow n or adapted for use in the disclosed compositions, methods and/or kits, or may include only a portion of the organic and/or inorganic compounds and materials that are part of the naturally-occurring sponge. In one aspect is provided any of the methods or kits disclosed herein, wherein the sponge materials comprise all or substantially all the organic and inorganic materials derived from the naturally occurring sponge. In another aspect is provided any of the methods or kits disclosed herein, wherein the sponge materials comprise (a) only the spicules and any materials that are naturally associated with the spicules, or (b) substantially purified spicules and any materials that are naturally associated with the spicules, or (c) purified spicules and any materials that are naturally associated with the spicules that are a component part of naturally -occurring sponge. In another aspect is provided any of the methods or kits disclosed herein, wherein the sponge materials comprise only the spicules and any materials that are naturally associated with the spicules. In another aspect is provided any of the methods or kits disclosed herein, wherein the sponge materials comprise substantially purified spicules and any materials that are naturally associated with the spicules. In another aspect is provided any of the methods or kits disclosed herein, w herein the sponge materials comprise purified spicules and any materials that are naturally associated with the spicules that are a component part of naturally occurring sponge. These terms may be used herein in relation to materials derived from the phylum Porifera. In another aspect, the materials are derived from sponges of the class Demospongiae . In another aspect, the materials are derived from sponges of the order Spongdilla. In another aspect, the materials are derived from sponges of the family Spongillidae. In another aspect, the materials are derived from sponges of the genus Spongilla. In another aspect, the materials are derived from sponges of the species Spongilla lacustris. In another aspect, the materials are derived from sponges of the order Haplosclerida. In another aspect, the materials are derived from sponges of the family Chalinidea. In another aspect, the materials are derived from sponges of the genus Halciona.
[00063] As used herein, "topical application" means placing or spreading directly onto the outer skin, scalp, mucous membrane, or hair, for example, by using the hands or an applicator such as a wipe, roller, or spray.
COMPOSITIONS
[00064] In one aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongilla,' and a second composition comprising one or more biological macromolecules.
Spongilla
[00065] Spongilla is a genus of freshwater sponges with over 200 different species. The organism has a leuconoid body form with a skeleton composed of siliceous spicules. The Spongilla contains organic and inorganic compounds with a total lipid content of approximately 5% of the biomass of the dried sponge; and the protein is composed of spongin or sclerotized collagen. The polysaccharides and N-acetyl-D-glucosamine (NAG) are part of chitin and chitosan that has been reported to be an important component within the skeletal fibers of Spongilla lacustris and detected 750 ± 1 .5ug N-acetyl-D-glucosamine per 1 mg of spicule-free skeleton. Chitin and chitosan are described as a family of linear polysaccharides consisting of varying amounts of a or |3 (1-4) linked residues of N-acetyl-2 amino-2-deoxy-D-glucose and 2-amino-2- deoxy -D glucose residues. In a-chitin, the chains are arranged in sheets or stacks, the chains in any one sheet having the same direction or "sense’. In P-chitin. adjacent sheets along the c axis have the same direction; the sheets are parallel, while in a-chitin adjacent sheets along the c axis have the opposite direction, they are antiparallel. Chitin is deacetylated into chitosan and can be further degraded into N- acetyl-D-glucosamine (NAG) units. Chitosan preparations are classified into native chitosan, chitosan formulations, complexes and derivatives with other substances. Chitosan can be used to prevent or treat wound and bum infections not only because of its intrinsic antimicrobial properties, but also by virtue of its ability to deliver extrinsic antimicrobial agents to wounds and bums. Chitosan is water-insoluble and highly viscous in dilute acidic solutions. Soluble chitosan oligosaccharides were found to be instrumental in suppressing the LPS-induced nuclear factor kappa-light-chain-enhancer of activated B cell (NF-KB)- dependent inflammatory gene expression, and this was associated with reduced nuclear translocation of NF-KB. Chitosan has also been demonstrated to have an antimicrobial effect against various bacteria.
[00066] In some embodiments, the Spongilla is in the form of a powder. Spongilla. including Spongilla lacustris, and powders prepared from Spongilla that are utilized in the methods, uses, compositions for use as a medicament, and kits disclosed herein may be obtained, processed and characterized by methods known to those having ordinary skill in the art. For example, United States Patent No. 7,604,821 describes the harvest, processing and characterization of several species of Spongilla, including Spongilla lacustris. The disclosure of United States Patent No. 7,604,821 is incorporated herein by reference in its entirety. Sponge materials may be collected using methods commonly known to those skilled in the art of marine biology. For example, sponges can be collected manually using basic under water diving techniques, or in deeper waters larger colonies are harvested using the Agassiz trawl (AGT) or epibenthic sledge (EBS). Under certain environmental conditions, Spongilla colonies occur in a thin crust-like carpet several meters across and must be collected manually, with fork-like tools, and nets. The collected sponge mass is dried, cleaned of gross contamination, such as shells, stems, plants, rocks and other impurities, and is then washed to remove dirt, sand, silt and soluble impurities. The cleaned sponge mass is weighed and dried using methods know n to those of ordinary skill in the art, such as air drying and the use of dryers that are used to dehydrate foods and pharmaceuticals. The sponge mass is dried until residual moisture content is less than a desired value as further disclosed herein. Residual moisture measurements can be performed using methods commonly known in the arts of food sciences, analytical chemistry or the pharmaceutical sciences. For example, 10 grams of dried material may be placed on a tared weighing boat and then weighed. The weighed material is then exposed to a heat source such as a drying oven or heat lamp operated at an appropriate temperature, the sample is then cooled in a desiccated chamber and re-weighed. Residual moisture is calculated as the percent difference between the sample weight before drying and the weight after cooling. Following drying, the sponge materials may be packaged in sealed containers, which optionally protect the materials from light, moisture and oxygen. The materials may then be further tested for the presence of pathogens, coliform organisms and organisms that represent a bioburden. The materials may be further heated or irradiated, as disclosed herein, to reduce any pathogens, coliform organisms or other organisms that represent a bioburden. The materials may then be further processed using methods known to those having ordinary skill in the art to provide a powder comprising particles, or inorganic spicules having a desired size. For example, the sponge materials may be ground, and the resulting materials passed through one or more sieves of a defined size to provide a resulting material comprising particles, or inorganic spicules having a uniform, or substantially uniform, size. After final processing and sizing processes are completed, the dried sponge material may be packaged in airtight moisture-proof containers and stored at an appropriate temperature, such as at about room or ambient temperature.
[00067] In some embodiments, the powder comprises particles, or inorganic spicules that are substantially uniform in size. In embodiments, not less than about 50% of the particles, or inorganic spicules included in the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 60% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 70% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 75% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 80% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 85% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 90% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 95% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 96% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 97% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 98% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In embodiments, not less than about 99% of the particles or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. In specific embodiments, not less than 90% of the particles, or inorganic spicules comprising the Spongilla powder pass through a US 70-mesh screen. The particles of Spongilla may be manufactured or produced from Spongilla materials that are harvested by procedures known to those of ordinary skill in the art, such as determining the appropriate harvest period, removal of foreign materials, drying, milling, grinding, and sieving using equipment known to those of ordinary skill in the art.
[00068] In some embodiments, the particles comprising the Spongilla powder have an average length of from about 50 pm to about 500 pm. In some embodiments, the inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 500 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 300 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 250 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 50 pm to about 200 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 75 pm to about 500 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 75 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 80 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 80 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 85 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 85 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 90 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 90 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 90 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 300 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 250 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 200 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 500 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 100 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 300 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 250 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 150 pm to about 200 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 175 pm to about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 175 pm to about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 175 pm to about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 300 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 175 pm to about 250 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of from about 175 pm to about 200 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 50 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 75 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 80 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 85 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 90 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 100 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 125 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 150 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 175 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 200 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 225 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 250 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 300 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 350 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 400 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 450 pm. In some embodiments, the particles or inorganic spicules comprising the Spongilla powder have an average length of about 500 pm. The particles comprising the Spongilla powder may be manufactured or produced from Spongilla materials that are harvested by procedures known to those of ordinary skill in the art, such as milling and grinding using equipment known to those of ordinary7 skill in the art. The average length of particles or inorganic spicules including the Spongilla powder may be measured using analytical methods known to those of ordinary skill in the art, such as, for example, scanning electron microscopy (SEM) and sieve analysis. Sieve analysis may also be used to determine the particle size distribution of the particles or inorganic spicules comprising the Spongilla powder.
[00069] In some embodiments, the particles of the Spongilla powder have an average diameter of from about 5 pm to about 50 pm. In some embodiments, the inorganic spicules including the Spongilla powder have an average diameter of from about 5 pm to about 50 pm. In some embodiments, the particles or inorganic spicules including the Spongilla powder have an average diameter of from about 5 pm to about 45 pm, or from about 5 pm to about 40 pm, from about 5 pm to about 35 pm, from about 5 pm to about 30 pm. from about 5 pm to about 25 pm, from about 5 pm to about 20 pm. from about 10 pm to about 45 pm, from about 10 pm to about 40 pm, from about 10 pm to about 35 pm, from about 10 pm to about 30 pm, from about 10 pm to about 25 pm, from about 10 pm to about 20 pm, and from about 10 pm to about 15 pm. In some embodiments, the particles or inorganic spicules including the Spongilla powder have an average diameter of about 5 pm, or about 10 pm, or about 15 pm, or about 20 pm, or about 25 pm, or about 30 pm, or about 35 pm, or about 40 pm, or about 45 pm, or about 50 pm. The particles including the Spongilla powder may be manufactured or produced from Spongilla materials that are harvested by procedures known to those of ordinary’ skill in the art, such as milling and grinding using equipment known to those of ordinary skill in the art. The average diameter of particles or inorganic spicules comprising the Spongilla powder may be measured using analytical methods known to those of ordinary' skill in the art, such as, for example, scanning electron microscopy (SEM) and sieve analysis. Sieve analysis may also be used to determine the particle size distribution of the particles or inorganic spicules comprising the Spongilla powder.
[00070] In some embodiments, the particles comprising the Spongilla powder have an aspect ratio of from about 1 to 100. In some embodiments, the inorganic spicules comprising the Spongilla powder have an aspect ratio of from about 1 to 100. As used herein, the term ■‘aspect ratio” means with respect to the particles of Spongilla or the inorganic spicules described herein the ratio between the average length of the particles or inorganic spicules to the average diameter of the particles or inorganic spicules. A specific aspect ratio is important for a more uniform final product so the spicules won't be too long or too thick, to ensure an ideal depth of penetration into the stratum comeum. Applicant discovered that an extended particle that is longer than it is wide produces a certain size opening in the skin that allows for subsequent delivery' of larger molecules.
[00071] In some embodiments, the particles of the Spongilla powder have an aspect ratio of from about 1 to about 75. In some embodiments, the inorganic spicules including the Spongilla powder have an aspect ratio of from about 1 to about 75. In some embodiments, the particles or inorganic spicules including the Spongilla powder have an aspect ratio of from about 1 to about 50, or from about 1 to about 25, or from about 1 to about 20, or from about 1 to about 15, or from about 5 to about 100, or from about 5 to about 75, or from about 5 to about 50, or from about 5 to about 40, or from about 5 to about 35, or from about 5 to about 30, or from about 5 to about 25, or from about 5 to about 20, or from about 5 to about 15, or from about 7 to about 50, or from about 7 to about 45, or from about 7 to about 40. or from about 7 to about 35, or from about 7 to about 30, or from about 7 to about 25, or from about 10 to about 50, or from about 10 to about 45, or from about 10 to about 40, or from about 10 to about 35, or from about 10 to about 30, or from about 10 to about 25, or from about 10 to about 15. In some embodiments, the particles or inorganic spicules including the Spongilla powder have an aspect ratio of about 5, or about 6, or about 7, or about 8, or about 9, or about 10, or about 11, or about 12, or about 13, or about 14, or about 15, or about 16, or about 17, or about 18, or about 19, or about 20, or about 21, or about 22, or about 23, or about 24, or about 25. or about 26, or about 27. or about 28, or about 29. or about 30, or about 35. or about 40. or about 45, or about 50. or about 75, or about 100. The particles including the Spongilla powder may be manufactured or produced from Spongilla materials that are harvested by procedures known to those of ordinary' skill in the art, such as milling and grinding using equipment known to those of ordinary skill in the art. The aspect ratio of particles or inorganic spicules including the Spongilla powder may be measured using analytical methods known to those of ordinary skill in the art, such as, for example, scanning electron microscopy (SEM) and sieve analysis. Sieve analysis may also be used to determine the particle size distribution of the particles comprising the Spongilla pow der.
[00072] Materials comprising Spongilla may be processed and dried, using techniques known to those having ordinary skill in the art, such as the use of drying ovens, to provide materials having a desired residual moisture content. In some embodiments, the first composition including Spongilla has a residual moisture content of not more than about 20%. In another embodiment, the first composition has a residual moisture content of not more than about 15%, or not more than about 10%, or not more than about 9%, or not more than about 8%, or not more than about 7%, or not more than about 6%, or not more than about 5%, or not more than about 4%, or not more than about 3%. or not more than about 2%, or not more than 1%. In another embodiment, the first composition has a residual moisture content of not more than about 5%. In another embodiment, the first composition has a residual moisture content of not more than about 4%. In another embodiment, the first composition has a residual moisture content of not more than about 3%. In another embodiment, the first composition has a residual moisture content of not more than about 2%. In another embodiment, the first composition has a residual moisture content of not more than about 1%. The moisture content of the Spongilla materials can be reduced by heating the raw Spongilla materials using methods known to those of ordinary skill in the art, such as by open-air drying, or by use of a conventional oven dryer or a vacuum dryer, using equipment know n to those of ordinary skill in the art. For example, raw Spongilla materials may be placed into a tray and heated in a dry ing oven at a temperature range from about 30°C to about 200°C, for example to about 70°C, for a period of time necessary to reduce the residual moisture content to the desired level. The level of residual moisture of the materials may be measured using methods known to those of ordinary skill in the art such as those described in the United States Pharmacopeia methods USP <731> (Loss on Drying) and USP <921> (Water Determination).
[00073] Materials comprising Spongilla may be treated in order to reduce the bioburden, such as aerobic and anaerobic microbes, yeast and mold, Coliform bacteria. Salmonella, Pseudomonas aeruginosa, and Staphylococcus aureus, of the materials prior to their packaging and use. such as by use of ionizing irradiation, such as the use of gamma irradiation. For example, gamma irradiation may be performed on the raw Spongilla material prior to grinding to reduce the particle or inorganic spicule size, following grinding to reduce the particle or inorganic spicule size, the materials packaged in bulk and/or the materials following packaging in unit dose containers. The materials may be treated with ionizing radiation, such as gamma radiation, using methods and equipment known to those of ordinary skill in the art. such as gamma irradiators or electron beam irradiators. In other embodiments, the first composition is prepared by treating with ionizing radiation, such as gamma radiation, to deliver an absorbed radiation dose between about 1 kGy and about 50 kGy prior to being packaged. In other embodiments, the first composition is prepared by treating with ionizing radiation, such as gamma radiation, to deliver an absorbed radiation dose between about 1 kGy and about 45 kGy, or between about 1 kGy and about 40 kGy . between about 1 kGy and about 35 kGy, between about 1 kGy and about 30 kGy, or between about 1 kGy and about 25 kGy or between about 5 kGy and about 50 kGy. or between about 5 kGy and about 45 kGy, or between about 5 kGy and about 40 kGy, or between about 5 kGy and about 35 kGy, or between about 5 kGy and about 30 kGy, or between about 5 kGy and about 25 kGy, or between about 10 kGy and about 50 kGy, or between about 10 kGy and about 45 kGy, or between about 10 kGy and about 40 kGy, or between about 10 kGy and about 35 kGy, or between about 10 kGy and about 30 kGy, or between about 10 kGy and about 25 kGy. or between about 15 kGy and about 50 kGy, or between about 15 kGy and about 45 kGy, or between about 15 kGy and about 40 kGy, or between about 15 kGy and about 35 kGy, or between about 15 kGy and about 30 kGy, or between about 15 kGy and about 25 kGy. In other embodiments, the first composition is prepared by treating with ionizing radiation, such as gamma radiation, to deliver an absorbed radiation dose of about 1 kGy, or about 5 kGy, or about 10 kGy, 11 kGy, or about 12 kGy, or about 13 kGy, or about 14 kGy, or about 15 kGy, or about 16 kGy, or about 17 kGy, or about 18 kGy, or about 19 kGy. or about 20 kGy, or about 21 kGy, or about 22 kGy, or about 23 kGy, or about 24 kGy, or about 25 kGy, or about 26 kGy, or about 27 kGy, or about 28 kGy, or about 29 kGy, or about 30 kGy, or about 31 kGy, or about 32 kGy, or about 33 kGy, or about 34 kGy, or about 35 kGy, or about 36 kGy, or about 37 kGy, or about 38 kGy, or about 39 kGy, or about 40 kGy, or about 41 kGy, or about 42 kGy', or about 43 kGy', or about 44 kGy, or about 45 kGy, or about 46 kGy, or about 47 kGy, or about 48 kGy, or about 49 kGy, or about 50 kGy.
[00074] In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition comprising Spongilla has a combined yeast and mold content of not more than about 25 x 104 colony-forming units per gram (CFU/g). In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 5 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 1 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 5 x 103 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 1 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 10,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 7,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 5,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 2,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 2,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 1,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 1,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 250 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 100 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 75 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 25 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 10 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined yeast and mold content of not more than about 1 CFU/g. The combined yeast and mold content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or dry heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels. The combined yeast and mold content of the Spongilla materials may be measured by methods known to those of ordinary' skill in the art, such as those described in the United States Pharmacopeia method USP <61> (Microbial Enumeration Tests).
[00075] In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 25 x 104 colony-forming units per gram (CFU/g). In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 5 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 1 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 5 x 103 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 1 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 10,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 7,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 5,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 2,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 2,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Coliform bacteria in the first composition is not more than about 1,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 1,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 250 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 100 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 75 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 25 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 10 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Coliform bacteria content of not more than about 1 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has no detectable Coliform bacterial content. The Coliform bacteria content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or diy heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels. The Coliform bacteria content of the Spongilla materials may be measured by methods known to those of ordinary skill in the art, such as those described in the United States Pharmacopeia method USP <62> (Tests for Specified Microorganisms).
[00076] In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 25 x 104 colony-forming units per gram (CFU/g). In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 5 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 1 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 5 x 103 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 1 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 10.000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 7,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 5,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 2,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 2,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Salmonella in the first composition is not more than about 1,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 1,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 250 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 100 CFU/g. In another aspect is provided any of the methods, compositions for use as a medicament, drug products and kits disclosed herein, wherein the first composition has a Salmonella content of not more than about 75 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 25 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 10 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Salmonella content of not more than about 1 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has no detectable Salmonella content. The Salmonella content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or dry heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels. The Salmonella content of the Spongilla materials may be measured by methods known to those of ordinary^ skill in the art, such as those described in the United States Pharmacopeia method USP <62> (Tests for Specified Microorganisms).
[00077] In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 25 x 104 colony-forming units per gram (CFU/g). In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 5 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 1 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 5 x 103 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 1 x 104 CFU/g. Tn another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 10,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 7,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 5,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 2,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 2,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Pseudomonas aeruginosa bacteria in the first composition is not more than about 1,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 1,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 250 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 100 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has Pseudomonas aeruginosa bacteria content of not more than about 75 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 25 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 10 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Pseudomonas aeruginosa bacteria content of not more than about 1 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has no detectable Pseudomonas aeruginosa bacteria content. The Pseudomonas aeruginosa bacteria content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or dry' heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels. The Pseudomonas aeruginosa bacteria content of the Spongilla materials may be measured by methods known to those of ordinary skill in the art, such as those described in the United States Pharmacopeia method USP <62> (Tests for Specified Microorganisms).
[00078] In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 25 x 104 colony -forming units per gram (CFU/g). In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 5 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 1 x 104 CFU/g. Tn another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 5 x 103 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 1 x O4 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 10,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 7,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 5,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 2,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 2.000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the amount of Staphylococcus aureus bacteria in the first composition is not more than about 1,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 1,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 250 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 100 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 75 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 25 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 10 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a Staphylococcus aureus bacteria content of not more than about 1 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has no detectable Staphylococcus aureus bacteria content. The Staphylococcus aureus bacteria content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or dry heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels. The Staphylococcus aureus bacteria content of the Spongilla materials may be measured by methods known to those of ordinary skill in the art, such as those described in the United States Pharmacopeia method USP <62> (Tests for Specified Microorganisms).
[00079] The compositions disclosed herein, such as the first composition comprising Spongilla and/or the second composition comprising one or more biological macromolecules, may further comprise one or more conventional pharmaceutical carriers or excipients. Suitable pharmaceutical carriers and excipients include inert diluents, binders (such as saccharides and their derivatives, proteins, and synthetic polymers), fillers (such as colloidal silicon dioxide, sugars, including lactose, sucrose, mannitol, or sorbitol); and cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethylcellulose, or polyvinylpyrrolidone (PVP)), bulking agents, lubricants (such as magnesium stearate, sodium lauryl sulfate and talc), coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with diluents such as water, saline, ethanol, hydrogen peroxide, propylene glycol, glycerin, or combinations thereof. Excipients may also include, but are not necessarily limited to, antiadherents like magnesium stearate or hydrated magnesium silicate.
[00080] In some embodiments, the compositions disclosed herein may further include preservatives like antioxidants, amino acids, citric acid, sodium citrate, and synthetic preservatives. Antioxidants may include, but are not necessarily limited to vitamin A, vitamin E, vitamin C, retinyl palmitate, and selenium. Amino acids suitable for preserving any of the pharmaceutical compositions described herein may include, but are not necessarily limited to cysteine and methionine. Synthetic preservatives suitable for preserving any of the pharmaceutical compositions described herein may include, but are not necessarily limited to methyl paraben and propyl paraben.
[00081] In some embodiments, the first composition comprising Spongilla and/or the second composition comprising one or more biological macromolecules may further comprise one or more adjuvants. As used herein, the term “adjuvant” means a substance added to a vaccine to increase a vaccine's immunogenicity. Some adjuvants are believed to enhance the immune response by slowly releasing the antigen, while other adjuvants are strongly immunogenic in their own right and are believed to function synergistically. Additional mechanisms by which adjuvants function may include, but are not necessarily limited to, cellular infiltration against antigen-presenting cells (APCs), increased inflammation and trafficking to the injection site, enhancing the activity of APCs by up- regulating costimulatory signal or major histocompatibility gene complex (MHC) expression, enhanced antigen presentation, or induction of cytokine release for indirect effects. The most appropriate adjuvant for a given vaccine immunogen will depend to a large extent on the type of immune response required for protective immunity. Adjuvant selection can be somewhat empirical. Known vaccine adjuvants include, but are not limited to, oil and water emulsions (for example, complete Freund's adjuvant and incomplete Freund's adjuvant), toll-like receptor (TLR) agonists, monophosphoryl lipid A)(MPL®), synthetic lipid A, lipid A mimetics or analogs, aluminum salts, cytokines, saponins, muramyl dipeptide (MDP) derivatives, CpG oligos, lipopolysaccharide (LPS) of gram-negative bacteria. polyphosphazenes, any water in oil emulsion, any oil in water emulsion that contains one or more of the following constituents: squalene or its analogues or any pharmaceutically acceptable oil, tween-80, sorbitantrioleate, alpha-tocopherol, cholecalciferol and aqueous buffer, or any of the analogues and derivatives of the molecules thereof, virosomes, cochleates, poly(lactide-co-glycolides) (PLG) microparticles, poloxamer particles, microparticles, endotoxins, for instance bacterial endotoxins, liposomes, Corynebacterium parvum, Bacillus Calmette Guerin, aluminum hydroxide, aluminum phosphate, aluminum sulphate phosphate, inulin, algammulin, resiquimod, poly(I:C), glucan, dextran sulfate, iron oxide, sodium alginate, Bacto-Adjuvant, certain synthetic polymers such as poly amino acids and co-polymers of amino acids, saponin, “REGRESSIN’? (Vetrepharm, Athens, Ga.), “AVRIDINE” (N,N-dioctadecyl-N',N'-bis(2-hydroxyethyl)-propanediamine), paraffin oil, muramyl dipeptide and the like. In some embodiments, the Spongilla spicules act as an adjuvant, for example, by irritating the site of administration to recruit relevant immune cells.
[00082] In some embodiments, the particles comprising the Spongilla powder comprise only inorganic Spongilla spicules and any materials that are naturally associated with the spicules, as described above.
[00083] In some embodiments, the first composition is in the form of a powder, paste, gel, liquid, or topical cream. In some embodiments, the powder, paste, gel, liquid, or topical cream further comprises water, saline, or hydrogen peroxide. In some embodiments, the paste, gel, liquid, or topical cream is prepared by mixing a powder comprising Spongilla and water or saline or hydrogen peroxide.
[00084] In another aspect is provided any of the methods disclosed herein, wherein the first composition is applied to the skin of the subject in the form of a paste. In another aspect is provided any of the methods disclosed herein, wherein the paste further comprises water or saline. In another aspect is provided any of the methods disclosed herein, wherein the paste is prepared by mixing a composition comprising Spongilla and an aqueous solution comprising hydrogen peroxide. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 50% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 45% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1 % w/w to about 40% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 35% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 30% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 25% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 20% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 15% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 10% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 9% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1 % w/w to about 7% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 6% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 5% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1% w/w to about 4% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0. 1 % w/w to about 3% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 2% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.1% w/w to about 1% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 0.5% w/w to about 45% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 45% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 40% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 35% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 30% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 25% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 20% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 15% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1 % w/w to about 10% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1 % w/w to about 9% w/w. Tn some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 7% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 6% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 5% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 4% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 3% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 2% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 45% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 40% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 35% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 30% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 25% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 20% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 15% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 10% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 9% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 1% w/w to about 7% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 6% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 5% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 4% w/w. In some embodiments, the hydrogen peroxide is at a concentration of from about 2% w/w to about 3% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 0. 1% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 0.5% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 1% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 2% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 3% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 4% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 5% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 6% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 7% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 8% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 9% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 10% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 15% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 20% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 25% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 30% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 35% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 40% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 45% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 50% w/w. In some embodiments, the hydrogen peroxide is at a concentration of about 3% w/w. Aqueous hydrogen peroxide solutions that may be useful for the compositions and methods disclosed herein are commercially available or may be prepared by methods known to those of ordinary skill in the art.
[00085] In some embodiments, the first composition is packaged prior to use. As described in detail above, the sponge materials may be packaged in sealed containers, which optionally protect the materials from light, moisture and oxygen. The dried sponge material may be packaged in airtight moisture-proof containers and stored at an appropriate temperature, such as at about room or ambient temperature.
[00086] In some embodiments, the one or more sponges may be marine sponges or freshwater sponges. In other embodiments, the one or more sponges is a marine sponge. In other embodiments, the sponge is a freshwater sponge. In other embodiments, the compositions are derived from sponges of the phylum Porifera. In other embodiments, the compositions are derived from sponges of the class Demospongiae. In other embodiments, the compositions are derived from sponges of the order Spongdilla. In other embodiments, the compositions are derived from sponges of the family Spongillidae . In other embodiments, the compositions are derived from sponges of the genus Spongilla. In other embodiments, the compositions are derived from sponges of the species Spongilla lacustris. However, in some embodiments, the type of sponge that can be used within the scope of the present disclosure is not necessarily limited to members of the genus Spongilla. In other embodiments, the compositions are derived from sponges of the order Haplosclerida. In other embodiments, the compositions are derived from sponges of the family Chalinidea. In other embodiments, the compositions are derived from sponges of the genus Halciona.
Biological Macromolecules
[00087] In embodiments, the compositions described herein contain one or more biological macromolecules. The biological macromolecules may be molecules that initiate an immune response when administered to a subject.
[00088] The term “biological macromolecule” as used herein refers to a biological molecule such as a nucleic acid, protein, antibody , carbohydrate, polysaccharide, lipid, and the like. In some embodiments, the one or more biological macromolecules may include a nucleic acid molecule, a protein molecule, or a combination thereof.
[00089] In some embodiments, the nucleic acid molecule may comprise an RNA or a DNA molecule encoding an antigenic protein or a fragment thereof.
[00090] As used herein, the term “fragment,” when referring to a protein or nucleic acid, for example, means any shorter sequence than the full-length protein or nucleic acid. Accordingly, any sequence of a nucleic acid or protein other than the full-length nucleic acid or protein sequence can be a fragment. In some aspects, a protein fragment includes an epitope. In other aspects, a protein fragment is an epitope.
[00091] As used herein, the terms “functional variant” or “functional fragment” refer to a molecule, including a nucleic acid or protein, for example, that comprises a nucleotide and/or amino acid sequence that is altered by one or more nucleotides and/or amino acids compared to the nucleotide and/or amino acid sequences of the parent or reference molecule. For a protein, a functional variant is still able to function in a manner that is similar to the parent molecule. In other words, the modifications in the amino acid and/or nucleotide sequence of the parent molecule do not significantly affect or alter the functional characteristics of the molecule encoded by the nucleotide sequence or containing the amino acid sequence. The functional variant may have conservative sequence modifications including nucleotide and amino acid substitutions, additions and deletions. These modifications can be introduced by standard techniques known in the art, such as site-directed mutagenesis and random PCR- mediated mutagenesis. Functional variants can also include, but are not limited to, derivatives that are substantially similar in primary structural sequence, but which contain, e.g., in vitro or in vivo modifications, chemical and/or biochemical, that are not found in the parent molecule. Such modifications include, inter aha, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol. cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI-anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA-mediated addition of amino acids to proteins such as arginyl ati on, ubiquitination, and the like.
[00092] As used herein, the term "‘nucleic acid” refers to any deoxyribonucleic acid (DNA) molecule, ribonucleic acid (RNA) molecule, or nucleic acid analogues. A DNA or RNA molecule can be double-stranded or single-stranded and can be of any size. Exemplary nucleic acids include, but are not limited to, chromosomal DNA, plasmid DNA, cDNA, cell- free DNA (cfDNA), mitochondrial DNA, chloroplast DNA. viral DNA, mRNA, tRNA, rRNA, long non-coding RNA. siRNA, micro RNA (miRNA or miR). hnRNA, and viral RNA. Exemplary' nucleic analogues include peptide nucleic acid, morpholino- and locked nucleic acid, glycol nucleic acid, and threose nucleic acid. As used herein, the term “nucleic acid molecule’' is meant to include fragments of nucleic acid molecules as well as any full- length or non-fragmented nucleic acid molecule, for example. As used herein, the terms “nucleic acid” and “nucleic acid molecule” can be used interchangeably, unless context clearly indicates otherwise.
[00093] In some embodiments, the DNA molecule may further comprise a promoter. As used herein, the term “promoter” refers to a regulatory sequence that initiates transcription. A promoter can be operably linked to first and second polynucleotides of nucleic acid molecules provided herein. Generally, promoters included in DNA molecules provided herein include promoters for in vitro transcription (IVT). Any suitable promoter for in vitro transcription can be included in DNA molecules provided herein, such as a T7 promoter, a T3 promoter, an SP6 promoter, and others. In some embodiments, the promoter may be a T7 promoter, a T3 promoter, or an SP6 promoter.
[00094] In some embodiments, the RNA molecule is a self-replicating molecule.
[00095] As used herein, the terms “self-replicating RNA,” “self-transcribing and selfreplicating RNA.” “self-amplifying RNA (saRNA),” and “replicon” may be used interchangeably, unless context clearly indicates otherwise. Generally, the term “replicon” or “viral replicon” refers to a self-replicating subgenomic RNA derived from a viral genome that includes viral genes encoding non-structural proteins important for viral replication and that lacks viral genes encoding structural proteins. A self-replicating RNA can encode further subgenomic RNAs that are not able to self-replicate.
[00096] In some embodiments, the nucleic acid molecule further comprises a 5’ cap. Any 5’ cap can be included in RNA molecules provided herein, including 5' caps having a Cap 1 structure, a Cap 1 (m6A) structure, a Cap 2 structure, a Cap 0 structure, or any combination thereof. In one aspect, RNA molecules provided herein include a 5’ cap having Cap 1 structure.
[00097] In some embodiments, the nucleic acid molecule further comprises a 5’ UTR and/or a 3’ UTR. In some embodiments, the nucleic acid molecule further comprises a 5’ UTR. In some embodiments, the nucleic acid molecule further comprises a 3’ UTR. In some embodiments, the nucleic acid molecule further comprises a 5’ UTR and a 3‘ UTR. In some embodiments, the 3’ UTR may comprise a poly(A) sequence.
[00098] As used herein, the term “protein” refers to any polymeric chain of amino acids. The terms “peptide” and “polypeptide” can be used interchangeably with the term protein, unless context clearly indicates otherwise, and can also refer to a polymeric chain of amino acids. The term “protein” encompasses native or artificial proteins, protein fragments and polypeptide analogs of a protein sequence. A protein may be monomeric or polymeric. The term “protein” encompasses fragments and variants (including fragments of variants) thereof, unless otherwise contradicted by context.
[00099] In another embodiment, said one or more biological macromolecules is selected from a recombinant protein, a fusion protein, an antibody, a monoclonal antibody, a humanized monoclonal antibody, a bivalent antibody, an antibody fragment, an antibodydrug conjugate, a Fc fragment, a Fab fragment, a Fab' fragment, a (Fab')2 fragment, a Fv fragment, and a scFv fragment. In another embodiment, the one or more biological macromolecules is a recombinant protein. In another embodiment, the one or more biological macromolecules is a fusion protein. In another embodiment, the one or more biological macromolecules is an antibody. In another embodiment, the one or more biological macromolecules is a monoclonal antibody . In another embodiment, the one or more biological macromolecules is a humanized monoclonal antibody. In another embodiment, the one or more biological macromolecules is a bivalent antibody. In another embodiment, the one or more biological macromolecules is an antibody fragment. In another embodiment, the one or more biological macromolecules is an antibody-drug conjugate. In another embodiment, the one or more biological macromolecules is a Fc fragment. In another embodiment, the one or more biological macromolecules is a Fab fragment. In another embodiment, the one or more biological macromolecules is a Fab' fragment. In another embodiment, the one or more biological macromolecules is a (Fab')2 fragment. In another embodiment, the one or more biological macromolecules is a Fv fragment. In another embodiment, the one or more biological macromolecules is a scFv fragment.
[000100] In some embodiments, the biological macromolecule is a drug selected from a T- cell co-stimulation modulator, an antagonist of one or more interleukin receptors, an antagonist of one or more interleukins, an interferon-gamma antagonist, a tissue necrosis factor-alpha antagonist, and a transforming growth factor-beta agonist. In another embodiment, said drug is a T-cell co-stimulation modulator. In another embodiment, said drug is an antagonist of one or more interleukin receptors. In another embodiment, said drug is an antagonist of one or more interleukins. In another embodiment, said drug is an interferon-gamma antagonist. In another embodiment, said drug is a tissue necrosis factoralpha antagonist. In another embodiment, said drug is a transforming growth factor-beta agonist.
[000101] In specific embodiments, protein molecule encoded by the nucleic acid molecule, or the protein molecule itself, is an antigenic protein or a fragment thereof. In some embodiments, the antigenic protein is a viral protein. In some embodiments, the viral protein originates from a virus that infects humans. [000102] The virus may be a DNA virus, a RNA virus, or a retrovirus. Non-limiting example of viruses useful with the present invention include, but are not limited to Ebola virus, measles virus, SARS-Coronaviruses, Chikungunya virus, hepatitis viruses, Marburg virus, yellow fever virus, MERS-Coronaviruses, Dengue viruses, Lassa virus, influenza viruses, rhabdovirus or HIV. A hepatitis virus may include hepatitis A, hepatitis B, or hepatitis C virus. An influenza virus may include, for example, influenza A or influenza B. An HIV may include HIV 1 or HIV 2. In certain example embodiments, the antigenic viral sequence may be from a human respiratory syncytial virus, Sudan ebola virus, Bundibugyo virus. Tai Forest ebola virus. Reston ebola virus, Achimota. Aedes flavivirus, Aguacate virus, Akabane virus, Alethinophid reptarenavirus, Allpahuayo mammarenavirus, Amapari mmarenavirus, Andes virus, Apoi virus, Aravan virus, Aroa virus, Arumwot virus, Atlantic salmon paramyoxivirus, Australian bat lyssavirus. Avian bomavirus, Avian metapneumovirus, Avian paramyoxviruses, penguin or Falkland Islandsvirus, BK polyomavirus, Bagaza virus, Banna virus, Bat hepevirus, Bat sapovirus. Bear Canon mammarenavirus, Beilong virus, Betacoronoavirus, Betapapillomavirus 1-6, Bhanja virus, Bokeloh bat lyssavirus, Boma disease virus, Bourbon virus, Bovine hepacivirus, Bovine parainfluenza virus 3, Bovine respiratory’ syncytial virus, Brazoran virus, Bunyamwere virus, Caliciviridae virus. California encephalitis virus, Candiru virus. Canine distemper virus, Canaine pneumovirus, Cedar virus, Cell fusing agent virus, Cetacean morbillivirus, Chandipura virus, Chaoyang virus, Chapare mammarenavirus, Chikungunya virus, Colobus monkey papillomavirus, Colorado tick fever virus, Cowpox virus, Crimean-Congo hemorrhagic fever virus. Culex flavivirus. Cupixi mammarenavirus, Dengue virus. Dobrava- Belgrade virus, Donggang virus, Dugbe virus, Duvenhage virus. Eastern equine encephalitis virus, Entebbe bat virus, Enterovirus A-D, European bat lyssavirus 1-2, Eyach virus, Feline morbillivirus, Fer-de-Lance paramyxovirus, Fitzroy River virus, Flaviviridae virus, Flexal mammarenavirus, GB virus C. Gairo virus, Gemy circularvirus, Goose paramyoxiviurs SF02, Great Island virus, Guanarito mammarenavirus, Hantaan virus. Hantavirus Z10, Heartland virus, Hendra virus, Hepatitis A/B/C/E, Hepatitis delta virus, Human bocavirus, Human coronavirus, Human endogenous retrovirus K, Human enteric coronavirus, Human genital- associated circular DNA virus-1, Human herpesvirus 1-8, Human immunodeficiency virus 1/2, Huan mastadenovirus A-G, Human papillomavirus, Human parainfluenza virus 1-4. Human paraechovirus, Human picobimavirus, Human smacovirus, Ikoma lyssavirus, Ilheus virus, Influenza A-C, Ippy mammarenavirus, Irkut virus, J-virus, JC polyomavirus, Japanese encephalitis virus, Junin mammarenavirus, KI polyomavirus, Kadipiro virus, Kamiti River virus, Kedougou virus, Khujand virus, Kokobera virus, Kyasanur forest disease virus, Lagos bat virus, Langat virus. Lassa mammarenavirus, Latino mammarenavirus, Leopards Hill virus, Liao ning virus, Ljungan virus, Lloviu virus, Louping ill virus, Lujo mammarenavirus, Luna mammarenavirus, Lunk virus, Lymphocytic choriomeningitis mammarenavirus. Lyssavirus Ozemoe, MSSI2V225 virus, Machupo mammarenavirus, Mamastrovirus 1, Manzanilla virus, Mapuera virus, Marburg virus, Mayaro virus, Measles virus, Menangle virus, Mercadeo virus, Merkel cell polyomavirus, Middle East respiratory’ syndrome coronavirus, Mobala mammarenavirus, Modoc virus, Moijang virus, Mokolo virus, Monkeypox virus, Montana myotis leukoenchalitis virus, Mopeia lassa virus reassortant 29, Mopeia mammarenavirus, Morogoro virus, Mossman virus, Mumps virus, Murine pneumonia virus, Murray Valley encephalitis virus, Nariva virus, Newcastle disease virus, Nipah virus, Norwalk virus, Norway rat hepacivirus, Ntaya virus. O'nyong-nyong virus, Oliveros mammarenavirus, Omsk hemorrhagic fever virus, Oropouche virus, Parainfluenza virus 5, Parana mammarenavirus, Parramatta River virus, Peste-des-petits-ruminants virus, Pichande mammarenavirus, Picomaviridae virus, Pirital mammarenavirus, Piscihepevirus A, Porcine parainfluenza virus 1, porcine rubulavirus, Powassan virus. Primate T-lymphotropic virus 1-2, Primate erythroparvovirus 1, Punta Toro virus. Puumala virus. Quang Binh virus, Rabies virus, Razdan virus. Reptile bomavirus 1, Rhinovirus A-B, Rift Valley fever virus, Rinderpest virus, Rio Bravo virus, Rodent Torque Teno virus, Rodent hepacivirus, Ross River virus, Rotavirus A-I. Royal Farm virus, Rubella virus, Sabia mammarenavirus. Salem virus, Sandfly fever Naples virus, Sandfly fever Sicilian virus, Sapporo virus, Sathuperi virus, Seal anellovirus, Semliki Forest virus, Sendai virus, Seoul virus, Sepik virus, Severe acute respiratory syndrome-related coronavirus, Severe fever with thrombocytopenia syndrome virus. Shamonda virus, Shimoni bat virus, Shuni virus, Simbu virus, Simian torque teno virus, Simian virus 40-41, Sin Nombre virus, Sindbis virus, Small anellovirus, Sosuga virus, Spanish goat encephalitis virus, Spondweni virus, St. Louis encephalitis virus, Sunshine virus, TTV-like mini virus, Tacaribe mammarenavirus, Taila virus, Tamana bat virus, Tamiami mammarenavirus, Tembusu virus, Thogoto virus, Thottapalayam virus, Tick-bome encephalitis virus, Tioman virus, Togaviridae virus, Torque teno canis virus, Torque teno douroucouli virus, Torque teno felis virus. Torque teno midi virus, Torque teno sus virus, Torque teno tamarin virus, Torque teno virus, Torque teno zalophus virus, Tuhoko virus, Tula virus, Tupaia paramyxovirus, Usutu virus, Uukuniemi virus, Vaccinia virus, Variola virus, Venezuelan equine encephalitis virus, Vesicular stomatitis Indiana virus, WU Polyomavirus, Wesselsbron virus, West Caucasian bat vims, West Nile vims, Western equine encephalitis vims, Whitewater Arroyo mammarenavims, Yellow fever virus, Yokose virus, Yug Bogdanovac virus, Zaire ebolavirus, Zika virus, or Zygosaccharomyces bailii vims Z viral sequence. Examples of RNA vimses that may be detected include one or more of (or any combination of) Coronaviridae virus, a Picomaviridae vims, a Caliciviridae virus, a Flaviviridae vims, a Togaviridae vims, a Bomaviridae, a Filoviridae, a Paramyxoviridae, a Pneumoviridae, a Rhabdoviridae, an Arenaviridae, a Bunyaviridae, an Orthomyxoviridae, or a Deltavirus. In certain example embodiments, the vims is Coronavims, SARS, Poliovirus, Rhinovims, Hepatitis A, Norwalk vims, Yellow fever vims. West Nile vims, Hepatitis C virus, Dengue fever virus, Zika vims, Rubella virus, Ross River vims, Sindbis vims, Chikungunya vims, Boma disease vims, Ebola virus, Marburg vims, Measles vims, Mumps virus, Nipah virus, Hendra vims, Newcastle disease virus. Human respiratory syncytial vims, Rabies virus, Lassa vims, Hantavims, Crimean-Congo hemorrhagic fever vims. Influenza, or Hepatitis D virus.
[000103] In some embodiments, the antigenic protein is a bacterial protein. This may include, but is not necessarily limited to, proteins from gram positive bacteria, gram negative bacteria, or other bacteria, such as Bacillus (e.g., Bacillus anthracis), Mycobacterium (e.g., Mycobacterium tuberculosis, Mycobacterium Leprae), Shigella (e.g., Shigella sonnet, Shigella dysenteriae, Shigella flexneri), Helicobacter (e.g., Helicobacter pylori), Salmonella (e.g., Salmonella enterica, Salmonella typhi. Salmonella typhimurium). Neisseria (e.g..
Neisseria gonorrhoeae. Neisseria meningitidis), Moraxella (e.g., Moraxella catarrhalis), Haemophilus (e.g., Haemophilus influenzae), Klebsiella (e.g., Klebsiella pneumoniae), Legionella (e.g., Legionella pneumophila), Pseudomonas (e.g., Pseudomonas aeruginosa). Acinetobacter (e.g., Acinetobacter baumannii), Listeria (e.g., Listeria monocytogenes). Staphylococcus (e.g., Staphylococcus aureus), Streptococcus (e.g.. Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus agcdactiae), Corynebacterium (e.g., Corynebacterium diphtheria), Clostridium (e.g., Clostridium botulinum, Clostridium tetani, Clostridium difficile), Chlamydia (e.g., Chlamydia pneumonia, Chlamydia trachomatis), Caphylobacter (e.g.. Caphylobacter jejuni), Bordetella (e.g., Bordetella pertussis). Enterococcus (e.g.. Enterococcus faecalis, Enterococcus faecum), Vibrio (e.g., Vibrio cholerae), Yersinia (e.g., Yersinia pestis), Burkholderia (e.g., Burkholderia cepacia complex), Coxiella (e.g., Coxiella burnetii), Francisella (e.g., Francisella tularensis), and Escherichia (e.g., enterotoxigenic, enterohemorrhagic or Shiga toxin-producing E. coin such as ETEC, EHEC, EPEC, EIEC, and EAEC)). In another aspect, the antigenic protein is from a eukaryotic organism, including protists and fungi, such as Plasmodium (e.g., Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale, Plasmodium malariae, Plasmodium diarrhea), Candida (e.g., Candida albicans), Aspergillus (e.g., Aspergillus fumigatus), Cryptococcus (e.g., Cryptococcus neoformans), Histoplasma (e.g., Histoplasma capsulatum), Pneumocystis (e.g., Pneumocystis jirovecii), and Coccidiodes (e.g., Coccidiodes immitis).
[000104] In some embodiments, the second composition comprises a vaccine against Mycobacterium tuberculosis . In some embodiments, the vaccine is the Bacillus Calmette- Guerin (BCG) vaccine.
[000105] In some embodiments, the second composition comprises a vaccine against Mpox virus (formerly known as monkeypox virus). In some embodiments, the second composition comprises a vaccine against rabies virus.
[000106] In some embodiments, the pharmaceutical agent further compnses a vaccine vector. In some embodiments, the vaccine vector is an adenovirus vector, an adeno- associated virus vector, a herpesvirus vector, a retrovirus vector, a poxvirus vector, a baculovirus vector, a rabies virus vector, or a Sendai virus vector. In some embodiments, the herpesvirus vector may include, but is not necessarily limited to, an Epstein-Barr virus vector. In some embodiments, the retrovirus vector may include, but is not necessarily limited to, a lentivirus vector or a Moloney Murine Leukemia Virus (MMLV) vector.
[000107] In specific embodiments, the pharmaceutical agent further comprises a vaccine vector of murine origin, such as, but not necessarily limited to, a MMLV vector.
[000108] In some embodiments, the antigenic protein may be a fungal protein. In some embodiments, the fungi may include, but are not necessarily limited to, one or more of (or any combination of), Aspergillus, Blastomyces, Candidiasis, Coccidiodomycosis, Cryptococcus neoformans, Cryptococcus gatti, Histoplasma sp. (such as Histoplasma capsulatum), Pneumocystis sp. (such as Pneumocystis jirovecii), Stachybotrys (such as Stachybotrys chartarum), Mucroymcosis, Sporothrix, Exserohilum, Cladosporium, or various fungi causing eye infections. In certain example embodiments, the fungal protein is a yeast protein. Examples of fungal antigenic proteins or nucleic acids coding for these proteins that can be used in accordance with the disclosed compositions and methods include without limitation proteins from one or more of (or any combination of), Aspergillus species (such as Aspergillus fumnigatus, Aspergillus flavus and Aspergillus clavatus), Cryptococcus sp. (such as Cryptococcus neoformans, Cryptococcus gattii. Cryptococcus laurentii and Cryptococcus albidus), a Geotrichum species, a Saccharomyces species, a Hansenula species, a Candida species (such as Candida albicans), a Kluyveromyces species, a Debaryomyces species, a Pichia species, or combination thereof.
[000109] In certain example embodiments, the fungus is a mold. Example molds include, but are not limited to, a Penicillium species, a Cladosporium species, a Byssochlamys species, or a combination thereof.
[000110] In some embodiments, the antigenic protein is a helminthic protein. In certain example embodiments, the helminths may include, but are not necessarily limited to, Nematodes, Cestodes, and Trematodes.
[000111] In some embodiments, the antigenic protein is a protozoan protein. Examples of protozoan antigenic proteins that can be used in accordance with the disclosed compositions and methods include, but are not necessarily limited to, any one or more of (or any combination of), proteins from Euglenozoa, Heterolobosea, Diplomonadida, Amoebozoa, Blastocystic, and Apicomplexa. Example Euglenoza include, but are not limited to, Trypanosoma cruzi (Chagas disease), T. brucei gambiense, T. brucei rhodesiense.
Leishmania braziliensis, L. infantum, L. mexicana, L. major, L. tropica, and L. donovani. Example Heterolobosea include, but are not limited to, Naegleria fowleri. Example Diplomonadid include, but are not limited to, Giardia intestinalis (G. lamblia, G. duodenalis) . Example Amoebozoa include, but are not limited to, Acanthamoeba castellanii, Balamuthia madrillaris, Entamoeba histolytica. Example Blastocystis include, but are not limited to, Blastocystic hominis. Example Apicomplexa include, but are not limited to, Babesia microti, Cryptosporidium parvum, Cyclospora cayetanensis, Plasmodium falciparum. P. vivax, P. ovale, P. malariae, and Toxoplasma gondii.
[000112] In some embodiments, the antigenic protein may be a prion protein, such as misfolded forms of PrP, which are known to cause Creutzfeldt-Jakob disease or other neurodegenerative diseases in humans. [000113] In some embodiments, the antigenic protein is a tumor-associated antigen (TAA). As used herein, the terms "tumor associated antigen” or “TAA” are used to describe proteins that are significantly over-expressed in cancer compared to normal cells.
[000114] In some embodiments, the antigenic protein is a neoantigen. As used herein, the term “neoantigen” is used to describe newly formed antigens that have not been previously recognized by the immune system. Neoantigens can arise from altered tumor proteins formed as a result of tumor mutations, or from viral proteins.
[000115] In some embodiments, “cancer” refers to human cancers and carcinomas, sarcomas, adenocarcinomas, lymphomas, leukemias, etc., including solid and lymphoid cancers, kidney, breast, lung, non-small cell lung, bladder, colon, ovarian, prostate, pancreas, stomach, brain, head and neck, skin, uterine, testicular, glioma, esophagus, and liver cancer, including hepatocarcinoma, lymphoma, including B-acute lymphoblastic lymphoma, nonHodgkin's lymphomas (e.g., Burkitt’s, Small Cell, and Large Cell lymphomas), Hodgkin’s lymphoma, leukemia (including AML, ALL, and CML), or multiple myeloma. Additional examples include cancer of the thyroid, endocrine system, cervix, melanoma, mesothelioma, sarcoma, medulloblastoma, colorectal cancer, neuroblastoma, glioblastoma multiforme, rhabdomyosarcoma, primary' thrombocytosis, primary' macroglobulinemia, primary' brain tumors, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, lymphomas, thyroid cancer, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenal cortical cancer, neoplasms of the endocrine or exocrine pancreas, medullary thyroid cancer, medullary' thyroid carcinoma, melanoma, colorectal cancer, papillary' thyroid cancer, or hepatocellular carcinoma.
[000116] The term "leukemia" refers broadly to progressive, malignant diseases of the blood-forming organs and is generally characterized by a distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow. Leukemia is generally clinically classified on the basis of (1) the duration and character of the diseaseacute or chronic; (2) the type of cell involved; myeloid (myelogenous), lymphoid (lymphogenous), or monocytic; and (3) the increase or non-increase in the number abnormal cells in the blood-leukemic or aleukemic (subleukemic). Exemplary' leukemias that may be prevented or treated with a compound or method provided herein include, for example, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, acute granulocytic leukemia, chronic granulocytic leukemia, acute promyelocytic leukemia, adult T-cell leukemia, aleukemic leukemia, a leukocythemic leukemia, basophylic leukemia, blast cell leukemia, bovine leukemia, chronic myelocytic leukemia, leukemia cutis, embryonal leukemia, eosinophilic leukemia. Gross' leukemia, hairy-cell leukemia, hemoblastic leukemia, hemocytoblastic leukemia, histiocytic leukemia, stem cell leukemia, acute monocytic leukemia, leukopenic leukemia, lymphatic leukemia, lymphoblastic leukemia, lymphocytic leukemia, lymphogenous leukemia, lymphoid leukemia, lymphosarcoma cell leukemia, mast cell leukemia, megakaryocytic leukemia, micromyeloblastic leukemia, monocytic leukemia, myeloblastic leukemia, myelocytic leukemia, myeloid granulocytic leukemia, myelomonocytic leukemia, Naegeli leukemia, plasma cell leukemia, multiple myeloma, plasmacytic leukemia, promyelocytic leukemia, Rieder cell leukemia, Schilling's leukemia, stem cell leukemia, subleukemic leukemia, or undifferentiated cell leukemia.
[000117] As used herein, the term “lymphoma” refers to a group of cancers affecting hematopoietic and lymphoid tissues. It begins in lymphocytes, the blood cells that are found primarily in lymph nodes, spleen, thymus, and bone marrow. Two main types of lymphoma are non-Hodgkin lymphoma and Hodgkin's disease. Hodgkin's disease represents approximately 15% of all diagnosed lymphomas. This is a cancer associated with Reed- Sternberg malignant B lymphocytes. Non-Hodgkin’s lymphomas (NHL) can be classified based on the rate at which cancer grows and the type of cells involved. There are aggressive (high grade) and indolent (low grade) types of NHL. Based on the type of cells involved, there are B-cell and T-cell NHLs. Exemplary B-cell lymphomas that may be prevented or treated with a compound or method provided herein include, but are not limited to, small lymphocytic lymphoma, Mantle cell lymphoma, follicular lymphoma, marginal zone lymphoma, extranodal (MALT) lymphoma, nodal (monocytoid B-cell) lymphoma, splenic lymphoma, diffuse large cell B-lymphoma, Burkitt’s lymphoma, lymphoblastic lymphoma, immunoblastic large cell lymphoma, or precursor B-lymphoblastic lymphoma. Exemplary T- cell lymphomas that may be prevented or treated with a compound or method provided herein include, but are not limited to, cutaneous T-cell lymphoma, peripheral T-cell lymphoma, anaplastic large cell lymphoma, mycosis fungoides, and precursor T-lymphoblastic lymphoma.
[000118] The term "sarcoma" generally refers to a tumor which is made up of a substance like the embryonic connective tissue and is generally composed of closely packed cells embedded in a fibrillar or homogeneous substance. Sarcomas that may be prevented or treated with a compound or method provided herein include a chondrosarcoma, fibrosarcoma, lymphosarcoma, melanosarcoma, myxosarcoma, osteosarcoma, Abernethy's sarcoma, adipose sarcoma, liposarcoma, alveolar soft part sarcoma, ameloblastic sarcoma, botryoid sarcoma, chloroma sarcoma, chorio carcinoma, embryonal sarcoma, Wilms' tumor sarcoma, endometrial sarcoma, stromal sarcoma, Ewing's sarcoma, fascial sarcoma, fibroblastic sarcoma, giant cell sarcoma, granulocytic sarcoma, Hodgkin's sarcoma, idiopathic multiple pigmented hemorrhagic sarcoma, immunoblastic sarcoma of B cells, lymphoma, immunoblastic sarcoma of T-cells. Jensen's sarcoma, Kaposi's sarcoma, Kupffer cell sarcoma, angiosarcoma, leukosarcoma, malignant mesenchymoma sarcoma, parosteal sarcoma, reticulocytic sarcoma, Rous sarcoma, serocystic sarcoma, synovial sarcoma, or telangiectaltic sarcoma.
[000119] The term "melanoma" is taken to mean a tumor arising from the melanocytic system of the skin and other organs. Melanomas that may be prevented or treated with a compound or method provided herein include, for example, acral-lentiginous melanoma, amelanotic melanoma, benign juvenile melanoma, Cloudman's melanoma, S91 melanoma. Harding-Passey melanoma, juvenile melanoma, lentigo maligna melanoma, malignant melanoma, nodular melanoma, subungal melanoma, or superficial spreading melanoma.
[000120] The term "carcinoma" refers to a malignant new growth made up of epithelial cells tending to infiltrate the surrounding tissues and give rise to metastases. Exemplary carcinomas that may be prevented or treated with a compound or method provided herein include, for example, medullary thyroid carcinoma, familial medullary thyroid carcinoma, acinar carcinoma, acinous carcinoma, adenocystic carcinoma, adenoid cystic carcinoma, carcinoma adenomatosum. carcinoma of adrenal cortex, alveolar carcinoma, alveolar cell carcinoma, basal cell carcinoma, carcinoma basocellulare, basaloid carcinoma, basosquamous cell carcinoma, bronchioalveolar carcinoma, bronchiolar carcinoma, bronchogenic carcinoma, cerebriform carcinoma, cholangiocellular carcinoma, chorionic carcinoma, colloid carcinoma, comedo carcinoma, corpus carcinoma, cribriform carcinoma, carcinoma en cuirasse, carcinoma cutaneum, cylindrical carcinoma, cylindrical cell carcinoma, duct carcinoma, carcinoma durum, embryonal carcinoma, encephaloid carcinoma, epiermoid carcinoma, carcinoma epitheliale adenoides, exophytic carcinoma, carcinoma ex ulcere, carcinoma fibrosum, gelatinifomi carcinoma, gelatinous carcinoma, giant cell carcinoma, carcinoma gigantocellulare, glandular carcinoma, granulosa cell carcinoma, hair-matrix carcinoma, hematoid carcinoma, hepatocellular carcinoma, Hurthle cell carcinoma, hyaline carcinoma, hypemephroid carcinoma, infantile embryonal carcinoma, carcinoma in situ, intraepidermal carcinoma, intraepithelial carcinoma, Krompecher's carcinoma, Kulchitzky- cell carcinoma, large-cell carcinoma, lenticular carcinoma, carcinoma lenticulare, lipomatous carcinoma, lymphoepithelial carcinoma, carcinoma medullare, medullary carcinoma, melanotic carcinoma, carcinoma molle, mucinous carcinoma, carcinoma muciparum. carcinoma mucocellulare, mucoepidermoid carcinoma, carcinoma mucosum, mucous carcinoma, carcinoma myxomatodes, nasopharyngeal carcinoma, oat cell carcinoma, carcinoma ossificans, osteoid carcinoma, papillary carcinoma, periportal carcinoma, preinvasive carcinoma, prickle cell carcinoma, pultaceous carcinoma, renal cell carcinoma of kidney, reserve cell carcinoma, carcinoma sarcomatodes, Schneiderian carcinoma, scirrhous carcinoma, carcinoma scroti, signet-ring cell carcinoma, carcinoma simplex, small-cell carcinoma, solanoid carcinoma, spheroidal cell carcinoma, spindle cell carcinoma, carcinoma spongiosum, squamous carcinoma, squamous cell carcinoma, string carcinoma, carcinoma telangiectaticum, carcinoma telangiectodes, transitional cell carcinoma, carcinoma tuberosum, tuberous carcinoma, verrucous carcinoma, or carcinoma villosum.
[000121] In some embodiments, the pharmaceutical agent further comprises a whole cell cancer vaccine. The advantage of whole tumor cells used as a vaccine rather than a specific protein or peptide tumor antigen is that the cells provide a source of all potential antigens, eliminating the need to identify the most optimal antigen to target in a particular type of cancer. This way, multiple tumor antigens can be targeted at once, generating immune responses to more than one tumor antigen, thereby bypassing issues of tumor antigen loss. Furthermore, immunized lymphocyte and serologic responses can be exploited to identify novel tumor antigens or categorize the importance of a response to a particular tumor antigen through the comparison of immune responses pre- and post- vaccination and by correlating responses with prognosis.
[000122] An autologous approach employs tumor cells from the same patient to be treated and ensures that patients are vaccinated with cells containing the same tumor antigens that their tumor expresses. Vaccines made from allogeneic cells circumvent the issue of individualizing each patient's therapy and by using several cell lines derived from different tumors in the vaccine, there is an increased likelihood that the patient's tumor will share antigens (e g., neoantigens or TAAs) expressed by the vaccine cells, including important tumor antigens overexpressed or mutated in a high percentage of that particular cancer. A concern of using allogeneic cells is that HLA mismatch between vaccinating cell lines and the patient will result in a response directed against foreign HLA molecules rather than tumor antigens. While anti-HLA responses do develop, they have not been shown to inhibit the tumor antigen response and have actually been associated with clinical response to whole cell vaccination.
[000123] In some embodiments, the second composition comprises two or more biological macromolecules.
[000124] In some embodiments, a first macromolecule of the two or more biological macromolecules comprises an antigenic protein or a fragment thereof, and a second macromolecule comprises an immunomodulatory protein. The immunomodulatory protein may include, but is not necessarily limited to, a cytokine, a chemokine, or an interleukin.
[000125] In some embodiments, the pharmaceutical agent further compnses a lipid formulation.
[000126] In some embodiments, the lipid formulation is selected from a lipoplex, a liposome, a lipid nanoparticle, a polymer-based earner, an exosome, a lamellar body, a micelle, and an emulsion. In some aspects, the lipid formulation is a liposome selected from a cationic liposome, a nanoliposome, a proteoliposome, a unilamellar liposome, a multilamellar liposome, a ceramide-containing nanoliposome, and a multivesicular liposome. In some aspects, the lipid formulation is a lipid nanoparticle. In some aspects, the lipid nanoparticle has a size of less than about 200 nm. In some aspects, the lipid nanoparticle has a size of less than about 150 nm. In some aspects, the lipid nanoparticle has a size of less than about 100 nm. In some aspects, the lipid nanoparticle has a size of about 55 nm to about 90 nm. In some embodiments, the lipid formulation comprises polymer-based carriers such as polyethyleneimine (PEI), lipid nanoparticles and liposomes, nanoliposomes, ceramide- containing nanoliposomes, multivesicular liposomes, proteoliposomes, both natural and synthetically-derived exosomes, natural, synthetic and semi-synthetic lamellar bodies, nanoparticulates, micelles, and emulsions. [000127] Conventional liposomes are vesicles that consist of at least one bilayer and an internal aqueous compartment. Bilayer membranes of liposomes are typically formed by amphiphilic molecules, such as lipids of synthetic or natural origin that comprise spatially- separated hydrophilic and hydrophobic domains (Lasic. Trends Biotechnol., 16: 307-321, 1998). Bilayer membranes of the liposomes can also be formed by amphiphilic polymers and surfactants (e.g., polymerosomes, niosomes, etc.). They generally present as spherical vesicles and can range in size from 20 nm to a few microns. Liposomal formulations can be prepared as a colloidal dispersion or they can be lyophilized to reduce stability risks and to improve the shelf-life for liposome-based drugs. Methods of preparing liposomal compositions are known in the art and w ould be within the skill of an ordinary artisan.
[000128] Liposomes that have only one bilayer are referred to as being unilamellar, and those having more than one bilayer are referred to as multilamellar. The most common types of liposomes are small unilamellar vesicles (SUV), large unilamellar vesicle (LUV), and multilamellar vesicles (MLV). In contrast to liposomes, lysosomes, micelles, and reversed micelles are composed of monolay ers of lipids. Generally, a liposome is thought of as having a single interior compartment, however some formulations can be multivesicular liposomes (MVL), which consist of numerous discontinuous internal aqueous compartments separated by several nonconcentric lipid bilayers.
[000129] Liposomes have long been perceived as drug delivery vehicles because of their superior biocompatibility, given that liposomes are basically analogs of biological membranes, and can be prepared from both natural and synthetic phospholipids (Int J Nanomedicine. 2014; 9: 1833-1843). In their use as drug delivery vehicles, because a liposome has an aqueous solution core surrounded by a hydrophobic membrane, hydrophilic solutes dissolved in the core cannot readily pass through the bilayer, and hydrophobic compounds will associate with the bilayer. Thus, a liposome can be loaded with hydrophobic and/or hydrophilic molecules. When a liposome is used to cany- a nucleic acid such as RNA, the nucleic acid will be contained within the liposomal compartment in an aqueous phase.
[000130] In contrast to liposomes and cationic liposomes, lipid nanoparticles (LNP) have a structure that includes a single monolayer or bilayer of lipids that encapsulates a compound in a solid phase. Thus, unlike liposomes, lipid nanoparticles do not have an aqueous phase or other liquid phase in its interior, but rather the lipids from the bilayer or monolayer shell are directly complexed to the internal compound thereby encapsulating it in a solid core. Lipid nanoparticles are typically spherical vesicles having a relatively uniform dispersion of shape and size. While sources vary' on what size qualifies a lipid particle as being a nanoparticle, there is some overlap in agreement that a lipid nanoparticle can have a diameter in the range of from 10 nm to 1000 nm. However, more commonly they are considered to be smaller than 120 nm or even 100 nm.
[000131] For lipid nanoparticle nucleic acid delivery' systems, the lipid shell is formulated to include an ionizable cationic lipid which can complex to and associate with the negatively charged backbone of the nucleic acid core. Ionizable cationic lipids with apparent pKa values below about 7 have the benefit of providing a cationic lipid for complexing with the nucleic acid’s negatively charged backbone and loading into the lipid nanoparticle at pH values below the pKa of the ionizable lipid where it is positively charged. Then, at physiological pH values, the lipid nanoparticle can adopt a relatively neutral exterior allowing for a significant increase in the circulation half-lives of the particles following i.v. administration. In the context of nucleic acid delivery, lipid nanoparticles offer many advantages over other lipid- based nucleic acid delivery systems including high nucleic acid encapsulation efficiency, potent transfection, improved penetration into tissues to deliver therapeutics, and low levels of cytotoxicity and immunogenicity.
[000132] In some embodiments, the lipid formulation is selected from a lipoplex, a liposome, a lipid nanoparticle, a polymer-based earner, an exosome, a lamellar body, a micelle, and an emulsion.
[000133] In some embodiments, the lipid formulation further comprises a polyethylene glycol (PEG)-lipid conjugate. The inclusion of polyethylene glycol (PEG) in a lipid formulation as a coating or surface ligand, a technique referred to as PEGylation, helps to protect nanoparticles from the immune system and their escape from RES uptake (Nanomedicine (Lond). 2011 Jun; 6(4): 715-28). PEGylation has been used to stabilize lipid formulations and their payloads through physical, chemical, and biological mechanisms. Detergent-like PEG lipids (e.g., PEG-DSPE) can enter the lipid formulation to form a hydrated layer and steric barrier on the surface. Based on the degree of PEGylation, the surface layer can be generally divided into two types, brush-like and mushroom-like layers. For PEG-DSPE-stabilized formulations, PEG will take on the mushroom conformation at a low degree of PEGylation (usually less than 5 mol%) and will shift to brush conformation as the content of PEG-DSPE is increased past a certain level (Journal of Nanomaterials. 2011 ;2011 : 12). PEGylation leads to a significant increase in the circulation half-life of lipid formulations (Annu. Rev. Biomed. Eng. 2011 Aug 15; 13():507-30; J. Control Release. 2010 Aug 3; 145(3): 178-81).
[000134] Examples of PEG-lipids include, but are not limited to, PEG coupled to dialkyloxypropyls (PEG-DAA), PEG coupled to diacylglycerol (PEG-DAG), PEG coupled to phospholipids such as phosphatidylethanolamine (PEG-PE), PEG conjugated to ceramides, PEG conjugated to cholesterol or a derivative thereof, and mixtures thereof.
[000135] PEG is a linear, water-soluble polymer of ethylene PEG repeating units with two terminal hydroxyl groups. PEGs are classified by their molecular weights and include the following: monomethoxypolyethylene glycol (MePEG-OH), monomethoxypolyethylene glycol- succinate (MePEG-S), monomethoxypolyethylene glycol-succinimidyl succinate (MePEG-S-NHS), monomethoxypolyethylene glycol-amine (MePEG-NH2), monomethoxypolyethylene glycol-tresylate (MePEG-TRES), monomethoxypolyethylene glycol-imidazolyl-carbonyl (MePEG-IM), as well as such compounds containing a terminal hydroxyl group instead of a terminal methoxy group (e.g., HO-PEG-S, HO-PEG-S-NHS, HO-PEG-NH2).
[000136] In some embodiments, the pharmaceutical agent further comprises one or more cryoprotectants. In some embodiments, the cryoprotectant comprises glycerol. In some embodiments, the composition or compositions comprise between about 5% and about 50% glycerol. In some embodiments, the storage medium comprises about 20% glycerol. In some embodiments, the cryoprotectant comprises sucrose. In some embodiments, the composition comprises between about 5% and about 30% sucrose. In some embodiments, the composition comprises about 10% sucrose. In some embodiments, the cryoprotectant comprises dimethylsulfoxide (DMSO). In some embodiments, the DMSO is at a concentration of between 2% and 10%. In specific embodiments, the one or more cryoprotectants are selected from sucrose, glycerol, or a combination of sucrose and glycerol.
[000137] In some embodiments, the second composition is a liquid, semi-liquid, solid, frozen, lyophilized, or vacuum-dried formulation. In embodiments, the second composition is in a liquid formulation. In embodiments, the second composition is in a semi-liquid formulation. In embodiments, the second composition is in a solid formulation. In embodiments, the second composition is in a frozen formulation. In embodiments, the second composition is in a lyophilized formulation. In some embodiments, the second composition is a vacuum-dried composition.
[000138] In some embodiments, the first composition and/or the second composition may be used in combination with a gel or cream. In embodiments, the gel or cream may or may not further comprise hydrogen peroxide. In some embodiments, the gel or cream does not further comprise hydrogen peroxide. In some embodiments, the gel or cream further comprises hydrogen peroxide. Such gels or creams are generally commercially available any may contain from about 0.5% w/w to about 50% w/w hydrogen peroxide. For example, a gel containing about 1% w/w, or about 2% w/w, or about 3% w/w, or about 4% w/w, or about 6% w/w, or about 7% w/w, or about 8% w/w, or about 9% w/w, or about 10% w/w, or about 15% w/w, or about 20% w/w, or about 25% w/w, or about 30% w/w, or about 40% w/w, or about 45% w/w, or about 50% w/w hydrogen peroxide may be used in any of the methods disclosed herein in combination with the first composition and the second composition.
[000139] In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongillcr, and a second composition comprising a vaccine against mycobacteria.
[000140] In some embodiments, the vaccine against mycobacteria is BCG.
[000141] In some embodiments, the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongillcr, and a second composition comprising a vaccine against Mpox virus (formerly known as monkeypox virus).
[000142] In some embodiments, the present disclosure provides a pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongillc, and a second composition comprising a vaccine against rabies virus.
[000143] In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the agent further comprises a T-cell co-stimulation modulator, an antagonist of one or more interleukin receptors, an antagonist of one or more interleukins, an interferon-gamma antagonist, a tissue necrosis factor-alpha antagonist, or a transforming growth factor-beta agonist. [000144] In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the agent further comprises an antagonist of one or more interleukin receptors. In some embodiments, said interleukin receptor is interleukin 1 receptor (IL-1R). IL-IRA, IL-1RB, IL-2R, IL-4R. IL-5R, IL-6R, IL-10R. IL-12R, IL-13R. IL-17R, IL-17RA, IL-21R, IL-22R, IL-23R, IL-31R, IL-35R, or a combination of these.
[000145] In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the agent further comprises an antagonist of one or more interleukins. In some embodiments, said interleukin is IL-1, IL-1A, IL-1B, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, IL-13, IL-17, IL-17A, IL-21, IL-22, IL-23, IL-31, IL-35, or a combination of these.
[000146] In some embodiments, the present disclosure provides a second composition comprising a live-attenuated virus or other pathogen, an inactivated virus or other pathogen, virus-like particles, a subunit vaccine, a conjugate vaccine, and/or a dendritic cell vaccine.
[000147] Live-attenuated vaccines comprise attenuated forms of a pathogen that is still infectious but exhibits reduced virulence while still stimulating a strong and effective immune response. Inactivated vaccines comprise pathogenic entities or organisms that have either been killed or inactivated so that they are no longer infectious but are still able to induce an immune response in a recipient. Virus-like particles are molecules that closely resemble viruses, but are non-infectious because they contain no viral genetic material. The components of virus-like particles are able to induce an effective immune response. Unlike live-attenuated and inactivated virus vaccines, subunit vaccines do not contain the whole pathogen, but contain only the antigenic parts such as proteins, polysaccharides, or peptides. One advantage of such a vaccine strategy is that the vaccine cannot cause an infection and it is therefore safer and more stable than vaccines containing whole pathogens, particularly for immunocompromised individuals. Conjugate vaccines are a ty pe of subunit vaccine which combines a weak antigen with a strong antigen as a carrier so that the immune system has a stronger response to the weak antigen. Most vaccines contain a single antigen; however, some antigens do not elicit a strong immune response. Conjugate vaccines are great for invoking immune responses against weak antigens because the weak antigen is covalently attached to a strong antigen, thereby eliciting a stronger immunological response to the weak antigen. Dendritic cells are professional antigen-presenting cells that process antigen material and present it on their surface to T cells, which then carry out effector functions of the adaptive immune system. Dendritic cells can be isolated from a patient, loaded with an antigen of choice, and then returned to the patient to induce the desired immune response. Methods for generating any or all of these types of vaccines are well known in the art.
[000148] In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 25 x 104 colony-forming units per gram (CFU/g). In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 10 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 5 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 1 x 104 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 5 x 103 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 1 x 104 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 10,000 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 7,500 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 5,000 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 2.500 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 2,000 CFU/g. In another aspect the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 1,500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 1,000 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 750 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 500 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 250 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 200 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 150 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 100 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 75 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 50 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 25 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 20 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 10 CFU/g. In another aspect, the present disclosure provides a pharmaceutical agent for inducing an immune response, wherein the first composition has a combined aerobic and anaerobic microbial content of not more than about 1 CFU/g. The combined aerobic and anaerobic microbial content of the Spongilla materials may be reduced by physical or chemical methods known to those of ordinary skill in the art, such as physical treatment of the materials with heat in the form of steam or dry heat, or chemical treatment in the form of exposure to ethylene oxide gas or treatment by ionizing radiation for a sufficient amount of time to reduce the microbial content to the desired levels. The combined aerobic and anaerobic microbial content of the Spongilla materials may be measured by methods known to those of ordinary skill in the art, such as those described in the United States Pharmacopeia method USP <61> (Microbial Enumeration Tests).
[000149] The pharmaceutical compositions disclosed herein may be in unit dosage forms suitable for single administration of precise dosages. In another aspect is provided any of the methods or kits disclosed herein, wherein the unit dosage forms of the first composition and/or the second composition are suitable for two administrations, three administrations, four administrations, five administrations, six administrations, seven administrations, eight administrations, 9 administrations, 10 administrations, 11 administrations, 12 administrations, 13 administrations, 14 administrations, 15 administrations, 16 administrations, 17 administrations, 18 administrations, 19 administrations, 20 administrations, 21 administrations, 22 administrations, 23 administrations, 24 administrations, 25 administrations, 26 administrations, 27 administrations. 28 administrations. 29 administrations, 30 administrations, administrations for two months, administrations for three months, administrations for four months, administrations for five months, administrations for six months, administrations for seven months, administrations for eight months, administrations for nine months, administrations for ten months, administrations for eleven months, or administrations for 12 months.
[000150] It will be appreciated that the actual dosages of the compositions disclosed herein, may vary according to the composition being used, the mode of administration, and the particular site of administration. Those skilled in the art using conventional dosagedetermination tests in view of the experimental data for a given composition may ascertain optimal dosages for a given set of conditions. This amount will vary depending upon a variety of factors, including but not limited to the characteristics of the compositions and formulations disclosed herein (including activity, pharmacokinetics, pharmacodynamics, and bioavailability thereof), the physiological condition of the subject (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication) or cells, the nature of the pharmaceutically acceptable carrier mg/kg or carriers in the formulation, and the route of administration. Further, an effective or therapeutically effective amount may vary depending on whether the one or more compositions and formulations disclosed herein is administered alone or in combination with other drug(s), other therapy/therapies or other therapeutic method(s) or modality/ modalities. One skilled in the clinical and pharmacological arts will be able to determine an effective amount or therapeutically effective amount through routine experimentation, namely by monitoring a cell's or subject's response to administration of the one or more compositions and formulations disclosed herein and adjusting the dosage accordingly.
[000151] Dosage regimens using the first composition and the second composition may be adjusted to provide the optimum desired response. Dosage unit form, as used herein, refers to physically discrete units suited as unitary dosages for the subjects; each unit containing a predetermined quantity of the compositions disclosed herein, calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the compositions disclosed herein are dictated by and directly dependent on (a) the characteristics of the composition and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such a composition for the prophylactic or therapeutic treatment a particular condition in a subject.
[000152] Thus, the skilled artisan would appreciate, based upon the disclosure provided herein, that the dose and dosing regimen using the compositions disclosed herein may be adjusted in accordance with methods well-known in the prophylactic and therapeutic arts. That is, the maximum tolerable dose can be readily established, and the effective amount providing a detectable therapeutic benefit to a subject may also be determined, as can the temporal requirements for administering each agent to provide a detectable prophylactic and/or therapeutic benefit to the subject. Accordingly, while certain dose and administration regimens are exemplified herein, these examples in no way limit the dose and administration regimen that may be provided to a subject in practicing the presently disclosed methods. In addition, intradermal vaccination offers a dose-sparing benefit, which reduces the volume of vaccine by 60-80% compared to intramuscular vaccination (Blumberg, L.H.(2018) Rabies vaccines: WHO position paper - April 2018 Vaccine 36:5500-5503). Dose-sparing is beneficial to immunization programs because it reduces the costs of purchase, distribution, and storage of vaccines, thereby leading to increased vaccine availability and effectiveness.
[000153] It is to be noted that dosage values may vary7 with the type and severity of the condition to be alleviated or prevented and may include single or multiple doses. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary7 only and are not intended to limit the scope or practice of the claimed composition. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamic parameters, which may include clinical effects such as toxic effects and/or laboratory values. The embodiments disclosed herein are intended to encompass intrasubject dose-escalation as determined by the skilled artisan. Determining appropriate dosages and regimens for administration of the prophylactic or therapeutic agent are well-known in the relevant art and would be understood to be encompassed by the skilled artisan once provided the teachings disclosed herein.
METHODS
[000154] In another aspect, the present disclosure provides a method of delivering a vaccine to a subj ect in need thereof, comprising administering an effective amount of any of the pharmaceutical agents described herein to the skin or the dermis of the subject.
[000155] The term '‘in need of treatment’’ as used herein refers to a judgment made by a caregiver (e.g., physician, nurse, nurse practitioner, or individual in the case of humans; veterinarian in the case of animals, including non-human mammals) that a subject requires or will benefit from a therapeutic or prophylactic treatment. This judgment is made based on a variety of factors that are in the realm of a caregiver’s expertise, but that include the knowledge that the subject is ill, or will be ill, as the result of a condition that is preventable or treatable by the compositions of the invention.
[000156] As used herein, the term “subject” or “patient” refers to a human or non-human animal. Preferably, the subject is a human. Preferably, the subject or patient is in need of prevention or treatment with the composition as described herein, e g., is at risk of becoming infected with an infectious agent or may develop cancer. In some embodiments, the subject is a mammal, including but not limited to humans, dogs, cats, cattle, sheep, and goats. In some embodiments, the subject is a dog. In some embodiments, the subject is a cat. In some embodiments, the subject is a cow. In some embodiments, the subject is a sheep. In some embodiments, the subject is a goat.
[000157] As used herein, the term "‘effective amount” or “therapeutically effective amount” refers to that amount of a Spongilla particle, protein, nucleic acid molecule, composition, or pharmaceutical composition described herein that is sufficient to effect the intended application, including but not limited to inducing an immune response and/or disease treatment, as defined herein. The therapeutically effective amount may vary’ depending upon the intended application (e.g.. inducing an immune response, treatment, application in vivo), or the subject or patient and disease condition being prevented or treated, e g., the weight and age of the subject, the species, the severity' of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary' skill in the art. The term also applies to a dose that will induce a particular response in a target cell. The specific dose will vary depending on the particular Spongilla particle, protein, nucleic acid molecule, composition, or pharmaceutical composition chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
[000158] In some embodiments, the pharmaceutical agent is delivered intradermally.
[000159] In some embodiments, the amount of the first composition comprising Spongilla that is administered to the subject is from about 0.5 grams to about 50 grams. In some embodiments, the amount of the first composition is measured as a dry' weight. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 40 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 35 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 30 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 25 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 20 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 15 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.5 grams to about 10 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.75 grams to about 20 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.75 grams to about 15 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 0.75 grams to about 10 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 20 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 15 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 10 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 9 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 8 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subj ect is from about 1 gram to about 7 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 6 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 5 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 4 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to about 3 grams. In some embodiments, the amount of the first composition including Spongilla administered to the subject is from about 1 gram to 2 grams. In some embodiments, the amount of Spongilla administered, such as those disclosed above, are each measured as a dry weight.
[000160] In some embodiments, the amount of the first composition including Spongilla applied to the skin of the subject is about 0.5 grams, or about 0.75 grams, or about 1 gram, or about 1.25 grams, or about 1.5 grams, or about 1.75 grams, or about 2 grams, or about 2.25 grams, or about 2.5 grams, or about 2.75 grams, or about 3 grams, or about 3.25 grams, or about 3.5 grams, or about 3.75 grams, or about 4 grams, or about 4.25 grams, or about 4.5 grams, or about 4.75 grams, or about 5 grams, or about 5.25 grams, or about 5.5 grams, or about 5.75 grams, or about 6 grams, or about 6.25 grams, or about 6.5 grams, or about 7 grams, or about 7.25 grams, or about 7.5 grams, or about 7.75 grams, or about 8 grams, or about 8.25 grams, or about 8.5 grams, or about 8.75 grams, or about 9 grams, or about 9.25 grams, or about 9.5 grams, or about 9.75 grams, or about 10 grams, or about 11 grams, or about 12 grams, or about 13 grams, or about 14 grams, or about 15 grams, or about 16 grams, or about 17 grams, or about 18 grams, or about 19 grams, or about 20 grams, or about 25 grams, or about 35 grams, or about 40 grams, or about 45 grams, or about 50 grams, or about 75 grams, or about 100 grams, or about 250 grams, or about 500 grams, or about 750 grams, or about 1000 grams, in each case measured as a dry weight.
[000161] In some embodiments, the first composition is administered first, and the second composition is administered second. In some embodiments, the second composition is administered first, and the first composition is administered second. In some embodiments, the first and second composition are each provided separately in separate containers. In some embodiments, the first and second composition are each provided in lyophilized, vacuum- dried, or liquid form in their separate containers. In some embodiments, the first composition is lyophilized and the second composition is vacuum-dried or vice versa. In some embodiments, the first composition is lyophilized and the second composition is in liquid form or vice versa. In some embodiments, the first composition is vacuum-dried and the second composition is in liquid form or vice versa. Any other combination of the above is also envisioned.
[000162] In some embodiments, the first composition and the second composition are administered together. In some embodiments, the first composition and the second composition are administered at the same time. In some embodiments, the first composition and the second composition are administered as a single composition. For example, the first composition and the second composition may be combined prior to administration, by the healthcare professional doing the administering.
[000163] In embodiments, the first composition and the second composition may be a single composition. In some embodiments, the single composition comprising the first and second composition together is provided in one container. In some embodiments, the single composition is provided in lyophilized, vacuum-dried, or liquid form. [000164] In some embodiments, the second composition is lyophilized or vacuum-dried and is reconstituted prior to administration. In some embodiments, the reconstitution reagent may comprise saline, buffered saline, or a hydrogel.
[000165] In yet another aspect, the present disclosure provides a method of preventing an infection in a subject in need thereof, comprising administering any of the pharmaceutical agents described herein by applying an effective amount of the agent to the skin of the subject.
[000166] As used herein, the term “prevent” refers to a decrease in the occurrence of disease symptoms in a patient. The prevention may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment. In some embodiments, the treatment may prevent severe disease, hospitalization and/or death.
[000167] In yet another aspect, the present disclosure provides a method of preventing or treating cancer in a subject in need thereof, comprising administering any of the pharmaceutical agents described herein by applying an effective amount of the agent to the skin of the subject.
[000168] In yet another aspect, the present disclosure provides a method of inducing an immune response in a subject in need thereof comprising administering an effective amount of any of the pharmaceutical agents described herein to the skin of the subject.
[000169] Any type of immune response can be induced using the methods provided herein, including adaptive and innate immune responses. In one aspect, immune responses induced using the methods provided herein include an antibody response, a cellular immune response, or both an antibody response and a cellular immune response.
[000170] In some embodiments, the pharmaceutical agent is administered one time. In some embodiments, the pharmaceutical agent is administered two or more times. In some embodiments, the pharmaceutical agents and/or compositions described herein are administered two or more times at three week intervals, four week intervals, five week intervals, six week intervals, seven week intervals, eight week intervals, nine week interv als, ten week intervals, eleven week intervals, or twelve week intervals. In some embodiments, the pharmaceutical agents and/or compositions described herein are administered two or more times at one month intervals, two month interv als, three month intervals, four month intervals, five month intervals, six month intervals, seven month intervals, eight month intervals, nine month intervals, ten month intervals, eleven month intervals, or twelve month intervals.
[000171] In some embodiments, the pharmaceutical agent is applied to mucosal surfaces.
[000172] In one embodiment, the skin of the subject is cleaned using a non-comedogenic cleanser, water, or a combination of a non-comedogenic cleanser and water following application of the first composition comprising Spongilla. In another embodiment, the skin of the subject is cleaned using a non-comedogenic cleanser, water, or a combination of a non- comedogenic cleanser and water following application of the second composition comprising one or more drugs to the skin of the subject. Non-comedogenic cleansers are those formulated not to cause blocked pores in the skin of subjects to which such cleansers are applied.
[000173] In some embodiments, the compositions may be used in combination with one or more additional compositions. When a combination therapy is used, the one or more additional compositions may be administered sequentially or simultaneously with the first composition and/or the second composition disclosed herein. In some embodiments, the additional compositions is administered to a subject prior to, at the same time as, or following administration of the first composition and/or the second composition disclosed herein. In some embodiments, the additional composition is administered to the subject prior to the administration of the first composition and/or the second composition disclosed herein. In some embodiments, the additional composition is administered to the subject at the same time the first composition and/or the second composition disclosed herein are administered to the subject. In some embodiments, the additional composition is administered to the subject following to the administration of the first composition and/or the second composition disclosed herein.
[000174] As will be understood by one skilled in the art, for all purposes, such as in terms of providing a written description, all ranges disclosed herein also encompass any and all possible values and sub-ranges and combinations of sub-ranges thereof, including endpoints. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like include the number recited and refer to ranges which can be subsequently broken down into sub-ranges as discussed above. Finally, as will be understood by one skilled in the art. a range includes each individual member. Thus, for example, a group having 1-3 articles refers to groups having 1, 2, or 3 articles. Similarly, a group having 1-5 articles refers to groups having 1, 2, 3, 4, or 5 articles, and so forth.
[000175] Headings, e.g., (a), (b), (i) etc., are presented merely for ease of reading the specification and claims. The use of headings in the specification or claims does not require the steps or elements be performed in alphabetical or numerical order or the order in which they are presented.
KITS
[000176] In yet another aspect, the present disclosure provides a kit for inducing an immune response in a subject in need thereof, wherein the kit comprises a first composition comprising Spongilla and a second composition comprising one or more biological macromolecules.
[000177] In some embodiments, the kit or compositions described herein may include a second composition comprising one or more drugs selected from a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials. In another embodiment, said one or more drugs is a biological macromolecule. In another embodiment, said one or more drugs is a chemical compound. In another embodiment, the one or more drugs is a mixture of chemical compounds. In another embodiment, the one or more drugs is a biological macromolecule. In another embodiment, the one or more drugs is an extract made from biological materials.
[000178] In some embodiments, the one or more biological macromolecules comprise a nucleic acid molecule, a protein molecule, or a combination thereof, as described in detail above.
[000179] In some embodiments, the kit further comprises a reconstitution agent. As described above, the reconstitution agent may include, but is not necessarily limited to, saline, buffered saline, or a hydrogel. [000180] In some embodiments, the kit further comprises one or more antiseptic reagents. In some embodiments, the one or more antiseptic reagents is a hydrogen peroxide solution, a hydrogen peroxide-containing hydrogel, a non-comedogenic cleanser, a solution of isopropyl alcohol. In some embodiments, the one or more antiseptic reagents is any acceptable skin antiseptic agent commonly used in surgery, including, but not limited to, chlorhexidine, chlorhexidine-gluconate, iodine, hexachlorophine, benzalkonium chloride, and any reconstitutions thereof.
[000181] In some embodiments, the kit further comprises instructions for use.
[000182] In some embodiments, the compositions and kits described herein do not require cold chain storage.

Claims

Claims
1. A pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongillcr, and a second composition comprising one or more biological macromolecules.
2. The pharmaceutical agent of claim 1, wherein the one or more biological macromolecules comprise a nucleic acid molecule, a protein molecule, or a combination thereof.
3. The pharmaceutical agent of claim 2, wherein the nucleic acid molecule comprises an RNA or a DNA molecule encoding an antigenic protein or a fragment thereof.
4. The pharmaceutical agent of claim 3, wherein the DNA molecule further comprises a promoter.
5. The pharmaceutical agent of claim 4, herein the promoter is a T7 promoter, a T3 promoter, or an SP6 promoter.
6. The pharmaceutical agent of claim 3, wherein the RNA molecule is a self-replicating molecule.
7. The pharmaceutical agent of claim 2, wherein the nucleic acid molecule further comprises a 5’ cap.
8. The pharmaceutical agent of claim 2, wherein the nucleic acid molecule further comprises a 5’ UTR and/or a 3’ UTR.
9. The pharmaceutical agent of claim 8, wherein the 3’ UTR comprises a poly(A) sequence.
10. The pharmaceutical agent of claim 2, wherein the protein molecule is an antigenic protein or a fragment thereof.
11. The pharmaceutical agent of any of claims 3 to 10, wherein the antigenic protein is a viral protein.
12. The pharmaceutical agent of claim 1 1 , wherein the viral protein originates from a human virus.
13. The pharmaceutical agent of any of claims 3 to 12, wherein the antigenic protein is a bacterial protein.
14. The pharmaceutical agent of any of claims 1 to 13, wherein the second composition comprises a vaccine against Mycobacterium tuberculosis .
15. The pharmaceutical agent of claim 14. wherein the vaccine is the Bacillus Calmette- Guerin (BCG) vaccine.
16. The pharmaceutical agent of any of claims 1 to 13, further comprising a vaccine vector of murine origin.
17. The pharmaceutical agent of any of claims 3 to 16, wherein the antigenic protein is a fungal protein.
18. The pharmaceutical agent of any of claims 3 to 16, wherein the antigenic protein is a helminthic protein.
19. The pharmaceutical agent of any of claims 3 to 16, wherein the antigenic protein is a protozoan protein.
20. The pharmaceutical agent of any of claims 3 to 16, wherein the antigenic protein is a prion protein.
21. The pharmaceutical agent of any of claims 3 to 16, wherein the antigenic protein is a cancer protein.
22. The pharmaceutical agent of any of claims 1 to 21, further comprising a whole cell cancer vaccine.
23. The pharmaceutical agent of claim 2, wherein the protein molecule is an immunomodulatory protein.
24. The pharmaceutical agent of claim 1 , wherein the second composition comprises two or more biological macromolecules.
25. The pharmaceutical agent of claim 24, wherein a first macromolecule of the two or more biological macromolecules comprises an antigenic protein or a fragment thereof, and a second macromolecule comprises an immunomodulatory protein.
26. The pharmaceutical agent of any of claims 1 to 25, further comprising a lipid formulation.
27. The pharmaceutical agent of claim 22. wherein the lipid formulation is selected from a lipoplex, a liposome, a lipid nanoparticle, a polymer-based carrier, an exosome. a lamellar body, a micelle, and an emulsion.
28. The pharmaceutical agent of claim 26 or 27. wherein the lipid formulation further comprises a polyethylene glycol (PEG)-lipid conjugate.
29. The pharmaceutical agent of any of claims 1 to 28, wherein the agent further comprises one or more cryoprotectants.
30. The pharmaceutical agent of claim 29, wherein the one or more cryoprotectants are selected from sucrose, glycerol, or a combination of sucrose and glycerol.
31. The pharmaceutical agent of any of claims 1 to 30, wherein the Spongilla is in the form of a powder.
32. The pharmaceutical agent of claim 31, wherein the powder comprises particles that are substantially uniform in size.
33. The pharmaceutical agent of claim 31 or 32, wherein not less than 90% of the particles comprising the Spongilla powder pass through a US 70-mesh screen.
34. The pharmaceutical agent of any one of claims 31 to 33, wherein the particles comprising the Spongilla powder have an average length of from about 50 pm to about 500 pm.
35. The pharmaceutical agent of any one of claims 31 to 34, wherein the particles comprising the Spongilla powder have an average diameter of from about 5 pm to about 50 pm.
36. The pharmaceutical agent of any one of claims 31 to 35, wherein the particles comprising the Spongilla powder have an aspect ratio of from about 1 to 100.
37. The pharmaceutical agent of any one of claims 31 to 36, wherein the particles comprising the Spongilla powder comprise only inorganic Spongilla spicules and any materials that are naturally associated with the spicules.
38. The pharmaceutical agent of any of claims 1 to 37, wherein the second composition is a lyophilized or vacuum-dried composition.
39. The pharmaceutical composition of any of claims 1 to 37, wherein the second composition is a liquid composition.
40. The pharmaceutical agent of any of claims 1 to 39, wherein the first composition is in the form of a paste, gel, or topical cream.
41. The pharmaceutical agent of claim 40, wherein the paste further comprises water or saline or hydrogen peroxide.
42. The pharmaceutical agent of claim 40 or 41, wherein the paste is prepared by mixing a powder comprising Spongilla and water or saline or hydrogen peroxide.
43. The pharmaceutical agent of any of claims 1 to 42, wherein the first composition is packaged prior to use.
44. The pharmaceutical agent according to any of claims 1 to 43, wherein the first composition is prepared by treating with gamma radiation.
45. The pharmaceutical agent of any of claims 1 to 44, wherein the Spongilla is Spongilla lacustris.
46. The pharmaceutical agent of any one of claims 1 to 45, wherein the first composition has a residual moisture content of not more than about 5%.
47. The pharmaceutical agent of any one of claims 1 to 46, wherein the amount of the first composition comprising Spongilla is from about 0.5 grams to about 50 grams.
48. A pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongilla and a second composition comprising a vaccine against mycobacteria.
49. The pharmaceutical agent of claim 48. wherein the vaccine against mycobacteria comprises BCG.
50. A method of delivering a vaccine to a subject in need thereof, comprising administering an effective amount of the pharmaceutical agent of any of claims 1 to 49 to the subject.
51. The method of claim 50, wherein the pharmaceutical agent is delivered intradermally.
52. The method of claim 50 or 51, wherein the first composition is administered first, and the second composition is administered second.
53. The method of claim 50 or 51, wherein the first composition and the second composition are administered together.
54. The method of any of claims 50 to 53, wherein the second composition is lyophilized or vacuum-dried and is reconstituted prior to administration.
55. A method of preventing an infection in a subject in need thereof, comprising administering the pharmaceutical agent of any of claims 1 to 49 to the subj ect.
56. A method of preventing cancer in a subject in need thereof, comprising administering the pharmaceutical agent of any of claims 1 to 49 to the subject.
57. A method of inducing an immune response in a subject in need thereof comprising administering an effective amount of the pharmaceutical agent of any of claims 1 to
49 to the subject.
58. The method of any of claims 50 to 57, wherein the pharmaceutical agent is administered one time.
59. The method of any of claims 50 to 57, wherein the pharmaceutical agent is administered two or more times.
60. The method of any of claims 50 to 59. wherein the pharmaceutical agent is applied to mucosal surfaces.
61. The method of any of claims 50 to 59, wherein the pharmaceutical agent is administered intradermally.
62. A kit for inducing an immune response in a subject in need thereof, wherein the kit comprises a first composition comprising Spongilla and a second composition comprising one or more biological macromolecules.
63. The kit of claim 62, wherein the one or more biological macromolecules comprise a nucleic acid molecule, a protein molecule, or a combination thereof.
64. The kit of claim 62 or 63, further comprising a reconstitution agent.
65. The kit of any of claims 62 to 64, further comprising an antiseptic reagent.
66. The kit of any of claims 62 to 65, further comprising instructions for use.
67. A pharmaceutical agent for inducing an immune response in a subject, wherein the agent comprises a first composition comprising Spongilla, and a second composition comprising a whole cell cancer vaccine.
PCT/US2024/010848 2023-01-13 2024-01-09 Compositions for inducing immune responses Ceased WO2024151612A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363479897P 2023-01-13 2023-01-13
US63/479,897 2023-01-13

Publications (1)

Publication Number Publication Date
WO2024151612A1 true WO2024151612A1 (en) 2024-07-18

Family

ID=89977280

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/010848 Ceased WO2024151612A1 (en) 2023-01-13 2024-01-09 Compositions for inducing immune responses

Country Status (1)

Country Link
WO (1) WO2024151612A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008039390A2 (en) * 2006-09-22 2008-04-03 Government Of The Usa., Dept. Of Health & Human Services Compositions and methods for chitosan enhanced immune response
US7604821B2 (en) 2002-07-01 2009-10-20 Maria Villani Porifera-based therapeutic compositions for treating and preventing skin diseases
CN102617526A (en) * 2011-05-31 2012-08-01 上海长征医院 Dimeric sesquiterpene compound Hippolide J, its preparation method and its purpose
US20150079624A1 (en) * 2002-07-01 2015-03-19 Maria Villani Porifera-based therapeutic compositions for treating and preventing skin diseases
WO2020117698A1 (en) * 2018-12-03 2020-06-11 Dermata Therapeutics, Llc Compositions for the treatment of conditions
WO2022125678A1 (en) * 2020-12-09 2022-06-16 Dermata Therapeutics, Inc. Compositions for the treatment of skin conditions

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7604821B2 (en) 2002-07-01 2009-10-20 Maria Villani Porifera-based therapeutic compositions for treating and preventing skin diseases
US20150079624A1 (en) * 2002-07-01 2015-03-19 Maria Villani Porifera-based therapeutic compositions for treating and preventing skin diseases
WO2008039390A2 (en) * 2006-09-22 2008-04-03 Government Of The Usa., Dept. Of Health & Human Services Compositions and methods for chitosan enhanced immune response
CN102617526A (en) * 2011-05-31 2012-08-01 上海长征医院 Dimeric sesquiterpene compound Hippolide J, its preparation method and its purpose
WO2020117698A1 (en) * 2018-12-03 2020-06-11 Dermata Therapeutics, Llc Compositions for the treatment of conditions
WO2022125678A1 (en) * 2020-12-09 2022-06-16 Dermata Therapeutics, Inc. Compositions for the treatment of skin conditions

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
ANNU. REV. BIOMED. ENG., vol. 13, 15 August 2011 (2011-08-15), pages 507 - 30
BLUMBERG, L.H: "Rabies vaccines: WHO position paper", VACCINE, vol. 36, April 2018 (2018-04-01), pages 5500 - 5503
INT J NANOMEDICINE, vol. 9, 2014, pages 1833 - 1843
J. CONTROL RELEASE, vol. 145, no. 3, 3 August 2010 (2010-08-03), pages 178 - 81
JOURNAL OF NANOMATERIALS, vol. 2011, 2011, pages 12
LASIC, TRENDS BIOTECHNOL., vol. 16, 1998, pages 307 - 321
NANOMEDICINE (LOND, vol. 6, no. 4, June 2011 (2011-06-01), pages 715 - 28

Similar Documents

Publication Publication Date Title
Makowski et al. Advances in lipid and metal nanoparticles for antimicrobial peptide delivery
Kubiatowicz et al. mRNA nanomedicine: Design and recent applications
JP7328187B2 (en) Treatment method and therapeutic composition for cancer and infectious disease
Kischkel et al. Therapies and vaccines based on nanoparticles for the treatment of systemic fungal infections
Ruseska et al. Use of protamine in nanopharmaceuticals—a review
US5994318A (en) Cochleate delivery vehicles
TWI668019B (en) Membrane encapsulated nanoparticles and method of use
Xu et al. Intranasal DNA vaccine for protection against respiratory infectious diseases: the delivery perspectives
Johnson et al. Nanotechnology-based vaccines for allergen-specific immunotherapy: Potentials and challenges of conventional and novel adjuvants under research
US20110305765A1 (en) Preparation and methodology of silk fibroin nanoparticles
Muhammad et al. Inflammation‐modulating nanoparticles for pneumonia therapy
Chelliah et al. Challenges and perspectives of standard therapy and drug development in high-grade gliomas
KR20100123674A (en) Nanotherapeutic colloidal metal compositions and methods
Wu et al. Enhancing antibacterial immunotherapy for bacterial pneumonia via nanovaccines coated with outer membrane vesicles
Zhao et al. Remodeling the hepatic fibrotic microenvironment with emerging nanotherapeutics: a comprehensive review
US20120045396A1 (en) Porous structures with modified biodegradation kinetics
Wu et al. Progress of polymer-based strategies in fungal disease management: Designed for different roles
Zheng et al. COVID‐19 cooling: Nanostrategies targeting cytokine storm for controlling severe and critical symptoms
Manju et al. Nanovaccines to combat drug resistance: The next-generation immunisation
US11252957B2 (en) Nucleic acid-peptide capsule complexes
Chen et al. Exploring new avenues of health protection: plant-derived nanovesicles reshape microbial communities
CA2246754C (en) Cochleate delivery vehicles
Brauer et al. Extracellular vesicles from Aspergillus flavus induce M1 polarization in vitro. mSphere 5: e00190-20
Tampucci et al. Formulations based on natural ingredients for the treatment of nail diseases
WO2024151612A1 (en) Compositions for inducing immune responses

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: 24705824

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE