EP4121026A1 - Compositions antimicrobiennes topiques persistantes et leurs méthodes d'utilisation - Google Patents

Compositions antimicrobiennes topiques persistantes et leurs méthodes d'utilisation

Info

Publication number
EP4121026A1
EP4121026A1 EP21771166.2A EP21771166A EP4121026A1 EP 4121026 A1 EP4121026 A1 EP 4121026A1 EP 21771166 A EP21771166 A EP 21771166A EP 4121026 A1 EP4121026 A1 EP 4121026A1
Authority
EP
European Patent Office
Prior art keywords
nanoemulsion
poloxamer
hours
oil
concentration
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.)
Pending
Application number
EP21771166.2A
Other languages
German (de)
English (en)
Other versions
EP4121026A4 (fr
Inventor
David Peralta
Susan Ciotti
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.)
Bluewillow Biologics Inc
Original Assignee
Bluewillow Biologics 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
Priority claimed from US16/828,542 external-priority patent/US20200237689A1/en
Application filed by Bluewillow Biologics Inc filed Critical Bluewillow Biologics Inc
Publication of EP4121026A1 publication Critical patent/EP4121026A1/fr
Publication of EP4121026A4 publication Critical patent/EP4121026A4/fr
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/075Ethers or acetals
    • A61K31/085Ethers or acetals having an ether linkage to aromatic ring nuclear carbon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/14Quaternary ammonium compounds, e.g. edrophonium, choline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4425Pyridinium derivatives, e.g. pralidoxime, pyridostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/74Synthetic polymeric materials
    • A61K31/765Polymers containing oxygen
    • A61K31/77Polymers containing oxygen of oxiranes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/186Quaternary ammonium compounds, e.g. benzalkonium chloride or cetrimide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/44Oils, fats or waxes according to two or more groups of A61K47/02-A61K47/42; Natural or modified natural oils, fats or waxes, e.g. castor oil, polyethoxylated castor oil, montan wax, lignite, shellac, rosin, beeswax or lanolin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin

Definitions

  • the present application is directed to methods of preventing and/or decreasing the risk of microbial infection by topical administration of a persistent, long-acting antimicrobial nanoemulsion compositions.
  • Nanoemulsions have been used as topical antimicrobial formulations as well as vaccine adjuvants. Prior teachings related to nanoemulsions are described in, for example, U.S. Pat. Nos. 6,015,832; 6,506,803; 6,559,189; 6,635,676; and 7,314,624. Methods of using persistant long- acting antimicrobial nanoemulsion compositions, where the site of application can be flushed with a liquid or water and the site still retains the 99.9% antimicrobial killing activity of the topically administered composition, have not been previously disclosed.
  • the present disclosure is directed to a method for preventing or reducing the risk of microbial infection in a subject.
  • the method comprises administering topically, mucosally, ocularly, or nasally a long-acting antimicrobial nanoemulsion to a target site of a subject in need, wherein following a single application the nanoemulsion kills about 99.9% or more of microorganisms present at the target site at the time of administration, and additionally the nanoemulsion kills 99.9% of microorganisms exposed to the target site for about 4 hours or more after administration of the nanoemulsion, and wherein the method prevents or reduces the risk of microbial infection in the subject caused by contact of the microorganism with the target site of the subject.
  • the nanoemulsion comprises droplets having an average or mean diameter of less than about 1 micron, and the nanoemulsion comprises an aqueous phase, an oil phase comprising at least one pharmaceutically acceptable oil, at least one pharmaceutically acceptable organic solvent, at least one nonionic surfactant, and a quaternary ammonium compound selected from the group consisting of cetylpyridimium chloride, benzalkonium chloride, benzethonium chloride, dioctadecyl dimethyl ammonium chloride, octenidine dihydrochloride and a combination thereof. Further, the concentration (w/w) of the quaternary ammonium compound and nonionic surfactant is in a ratio of about 5:1 to about 1:27.
  • the ratio of the concentration of the quaternary ammonium compound to nonionic surfactant is: (a) selected from the group consisting of about 5:1, about 4:1, about 3:1, about 2:1, about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:11, about 1:12, about 1:13, about 1:14, about 1:15, about 1:16, about 1:17, about 1:18, about 1:19, about 1:20, about 1:21, about 1:22, about 1:23, about 1:24, about 1:25, about 1:26, and about 1:27; and/or (b) about 4:1 to about 1:27; and/or (c) selected from the group consisting of about 1:2, about 1:6, about 1:7, about 1:9, about 1:10, and about 1:12; and/or (d) about 1:5 to about 1:10; and/or (e) about 1 :6 to about 1 :9; and/or
  • the target site is flushed with water or another liquid at least once during a period of 4 hours after administration, and (a) following flushing with water or another liquid, the target area retains the 99.9% microbial killing activity of the nanoemulsion; and/or (b) flushing with water or another liquid comprises sweating, submersion or rinsing in freshwater, submersion or rinsing in salt water, and/or rinsing with soap and water; and/or (c) the flushing is for a time period selected from the group consisting of about 1 min, about 2 mins, about 3 mins, about 4 mins, about 5 mins, about 10 mins, about 15 mins, about 20 mins, about 25 mins, about 30 mins, about 45 mins, about 1 hour, about 1.5 hrs, about 2 hours, about 2.5 hours or about 3 hours; and/or (d) the flushing occurs about 0.5 min to about 1 min, about 1 min to about 5 min, about 5 min to about
  • the target site comprises skin, epidermis, dermis, mucosa, squamous epithelium, or a combination thereof of the subject; and/or
  • the nanoemulsion is bioadhesive with the target site; and/or (c) following application of the nanoemulsion to a target area, the nanoemulsion does not result in any noticeable irritation of the target site; and/or (d) when the nanoemulsion is applied to the target site, the nanoemulsion results in increased hydration as compared to a composition comprising the same quaternary ammonium compound at the same concentration but lacking a nanoemulsion, measured at any suitable time period after administration to the target area, and optionally wherein the increase in target site hydration is about 50%, about 75%, about 100%, about 125%, about 150%, about 175%, about 200%, about 225%, about 250%, about 275%, about 300%, about 325%, about 350%, about 375%, about 400%, about 425%, about 450%, about 475%, about 500%, about 525%, about 550%, about 575%, about 600%, about 625%, about 650%, about 675%, about 700%,
  • microorganism (a) is a bacteria, virus, yeast, fungus, or any combination thereof; and/or (b) is a bacteria or virus; and/or
  • (c) is a virus; and/or (d) is selected from the group consisting of SARS-CoV-2, Human coronavirus 229E, human coronavirus OC43, SARS-CoV, HCoVNL63, HKU1, MERS-CoV, SARS-CoV-2, and any combination thereof; and/or (e) is a coronavirus and the coronavirus comprises (i) a polynucleotide comprising the SARS-CoV-2 genome sequence, (ii) an immunogenic fragment of the SARS-CoV-2 genome sequence, or (iii) a polynucleotide having at least about 80%, at least about 90%, at least about 95%, or at least about 99% sequence identity to the polynucleotide comprising the SARS-CoV-2 genome sequence; and/or (f) is selected from the group consisting of Influenza, B Respiratory Syncytial Virus, Staphylococcus aureus , Enterococcus fae
  • Human coronavirus HKU 1 or Murine coronavirus a Hibecovirus such as Bat Hp- betacoronavirus Zhejiang2013; a Merbecovirus such as Hedgehog coronavirus 1, Middle East respiratory syndrome-related coronavirus (MERS-CoV), Pipistrellus bat coronavirus HKU5 or Tylonycteris bat coronavirus HKU4; a Nobecovirus such as Rousettus bat coronavirus GCCDC1 or Rousettus bat coronavirus HKU9, a Sarbecovirus such as a Severe acute respiratory syndrome-related coronavirus, Severe acute respiratory syndrome coronavirus (SARS-CoV) or Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, COVID-19); a Deltacoronavirus; an Andecovirus such as Wigeon coronavirus HKU20; a Buldecovirus such as Bulbul coronavirus HKU11, Porcine coronavirus HKU
  • the quaternary ammonium compound can be: (a) BZK, which is present in an amount of about 0.05% to about 5.0%; and/or (b) BZK, which is present in an amount of about 0.13%; and/or (c)monographed by the US FDA as an antiseptic for topical use; (d) benzalkonium chloride (BZK); and/or (e) BZK present in a concentration of from about 0.05% to about 0.40%; BZK present in a concentration of from about 0.10% to about 0.20%; or BZK present in a concentration of about 0.13%; and/or (f) cetylpyridimium chloride (CPC); and/or (g) CPC present in a concentration of from about 0.05% to about 0.40%; CPC present in a concentration of from about 0.15% to about 0.30%; or CPC present in a concentration of about 0.20%; and/or (h)
  • the nanoemulsion particles have an average or mean diameter of less than or equal to about 900 nm, less than or equal to about 800 nm, less than or equal to about 700 nm, less than or equal to about 600 nm, less than or equal to about 500 nm, less than or equal to about 400 nm, less than or equal to about 300 nm, less than or equal to about 200 nm, less than or equal to about 150 nm, less than or equal to about 100 nm, or less than or equal to about 50 nm; and/or (b)the nanoemulsion particles have an average or mean diameter of about 350 nm.
  • the nanoemulsion is diluted resulting in a formulation of about 0.5% to about 60% nanoemulsion, and wherein: (i) the concentration ratio of the quaternary ammonium compound to nonionic surfactant is about 5: 1 to about 1 :27, and the nanoemulsion enhances delivery of the quaternary ammonium compound into tissue by at least about 25% as compared to a solution with the same concentration of the same quaternary ammonium compound but lacking a nanoemulsion and as compared to a nanoemulsion with compared to a nanoemulsion with compared to a nanoemulsion with a concentration ratio of the quaternary ammonium compound to nonionic surfactant outside of the range from about 5: 1 to about 1 :27; or (ii) the viscosity of the nanoemulsion is less than about lOOOcp, and the nanoemulsion enhances delivery of the quaternary ammonium compound into tissue by at least about 25% as compared to a solution with the same concentration
  • the nonionic surfactant is: (i) selected from the group consisting of a poloxamer surfactant, polysorbate surfactant, Triton ® X-100, nonoxynol-9, and any combination thereof; and/or (ii) selected from the group consisting of polysorbate 20, polysorbate 21, polysorbate 40, polysorbate 60, polysorbate 61, polysorbate 65, polysorbate 80, polysorbate 81, and polysorbate 85; and/or (iii) selected from the group consisting of poloxamer 407, poloxamer 101, poloxamer 105, poloxamer 108, poloxamer 122, poloxamer 123, poloxamer 124, poloxamer 181, poloxamer 182, poloxamer 183, poloxamer 184, poloxamer 185, poloxamer 188, poloxamer 212, poloxamer 215, poloxamer 217,
  • GRAS Generally Recognized as Safe
  • the organic solvent comprises a C1-C12 alcohol, diol, or triol, a dialkyl phosphate, a trialkyl phosphate or a combination thereof; and/or (ii) comprises an alcohol selected from the group consisting of ethanol, isopropyl alcohol, glycerol or a combination thereof.
  • the oil (i) comprises soybean oil, mineral oil, avocado oil, squalene oil, olive oil, canola oil, com oil, rapeseed oil, safflower oil, sunflower oil, fish oils, flavor oils, cinnamon bark, coconut oil, cottonseed oil, flaxseed oil, pine needle oil, silicon oil, essential oils, water insoluble vitamins, other plant oil, or a combination thereof; and/or (ii) comprises soybean oil.
  • the nanoemulsion further comprises a chelating agent.
  • the chelating agent can be, for example, (a) ethylenediaminetetraacetic acid (EDTA), ethylene glycol -bis(P-ami noethyl ether)-N,N,N',N'-tetraacetic acid (EGTA), or a combination thereof; and/or (b) ethylenediaminetetraacetic acid (EDTA); and/or (c) EDTA and EDTA is present at a concentration of about 0.0005% to about 1% (w/w).
  • EDTA ethylenediaminetetraacetic acid
  • EGTA ethylene glycol -bis(P-ami noethyl ether)-N,N,N',N'-tetraacetic acid
  • EDTA ethylenediaminetetraacetic acid
  • EDTA and EDTA is present at a concentration of about 0.0005% to about 1% (w/w).
  • the nanoemulsion can comprise: (i) about 5 vol.
  • % to about 50 vol. % (w/w) of aqueous phase (ii) about 30 vol. % to about 90 vol. % (w/w) of a pharmaceutically acceptable oil phase; (iii) about 0.1% up to about 20% of a pharmaceutically acceptable organic solvent; (iv) about 3 vol. % to about 15 vol. % (w/w) of a nonionic surfactant; and (v) about 0.05% to about 3 vol. % (w/w) of a quaternary ammonium compound.
  • the nanoemulsion can comprise (i) about 5 vol. % to about 50 vol. % (w/w) of aqueous phase; (ii) about 30 vol. % to about 90 vol. % (w/w) soybean oil; (iii) about 0.1% up to about 20% (w/w) glycerol; (iv) about 3 vol. % to about 15 vol. % (w/w) poloxamer 407; (v) benzalkonium chloride at a concentration of about 0.13% (w/w); and (vi) about 0.0005% to about 1% (w/w) EDTA.
  • the nanoemulsion (a) is non-toxic in humans and animals; and/or (b) is thermostable; and/or (c) is stable for at least 3 months at about 50°C; and/or (d) is stable for at least 3 months at 40°C; and/or (e) is stable for at least 3 months at 25°C; and/or (f) is stable for at least 3 months at 5°C; and/or (g) is stable at 5°C for up to at least 60 months; and/or (h) is stable at 50°C for up to at least 12 months; and/or (i) has been autoclaved, and optionally wherein the nanoemulsion retains its structural and/or chemical integrity following autoclaving; (j) is formulated in soap, hand sanitizer, lotion, cream, or spray dosage form; and/or (k) is formulated liquid dosage form, solid dosage form, or semisolid dosage form; and/or (1) is formulated in a nasal spray.
  • nanoemulsion formulated for topical, mucosal, ocular, or nasal administration
  • the nanoemulsion comprises droplets having an average or mean diameter of less than about 1000 nm
  • the nanoemulsion comprises: (i) an aqueous phase; (ii) an oil phase comprising at least one pharmaceutically acceptable oil: (iii) at least one pharmaceutically acceptable organic solvent; (iv) at least one nonionic surfactant; and (v) a quaternary ammonium compound selected from the group consisting of cetylpyridimium chloride, benzalkonium chloride, benzethonium chloride, dioctadecyl dimethyl ammonium chloride, octenidine dihydrochloride and a combination thereof.
  • the concentration (w/w) of the quaternary ammonium compound and nonionic surfactant can be in a ratio of about 5: 1 to about 1:27.
  • the nanoemulsion kills about 99.9% or more of microorganisms present at an application site at the time of administration, and additionally the nanoemulsion kills 99.9% of microorganisms exposed to the application site for about 4 hours or more after administration of the nanoemulsion.
  • the ratio of the concentration of the quaternary ammonium compound to nonionic surfactant is: (a) selected from the group consisting of about 5:1, about 4:1, about 3:1, about 2:1, about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:11, about 1:12, about 1:13, about 1:14, about 1:15, about 1:16, about 1:17, about 1:18, about 1:19, about 1:20, about 1:21, about 1:22, about 1:23, about 1:24, about 1:25, about 1:26, and about 1:27; and/or (b) about 4:1 to about 1:27; and/or (c) selected from the group consisting of about 1:2, about 1:6, about 1:7, about 1:9, about 1:10, and about 1:12; and/or (d) about 1:5 to about 1:10; and/or (e) about 1 :6 to about 1 :9; and/
  • the nanoemulsion kills about 99.9% or more of microorganisms exposed to the nanoemulsion for at least about 4.5 hours, at least about 5 hours, at least about 5.5 hours, at least about 6 hours, at least about 6.5 hours, at least about 7 hours, at least about 7.5 hours, at least about 8 hours, at least about 8.5 hours, at least about 9 hours, at least about 9.5 hours, at least about 10 hours, at least about 10.5 hours, at least about 11 hours, at least about 11.5 hours, at least about 12 hours, or greater than about 12 hours after application of the nanoemulsion; and/or (b) is water resistant for at least about 4 hours, at least about 4.5 hours, at least about 5 hours, at least about 5.5 hours, at least about 6 hours, at least about 6.5 hours, at least about 7 hours, at least about 7.5 hours, at least about 8 hours, at least about 8.5 hours, at least about 9 hours, at least about 9.5 hours,
  • the microorganism (a) is a bacteria, virus, yeast, fungus, or any combination thereof; and/or (b) is a bacteria or virus; and/or (c) is a virus.
  • the nanoemulsion particles have an average diameter of less than or equal to about 900 nm, less than or equal to about 800 nm, less than or equal to about 700 nm, less than or equal to about 600 nm, less than or equal to about 500 nm, less than or equal to about 400 nm, less than or equal to about 300 nm, less than or equal to about 200 nm, less than or equal to about 150 nm, less than or equal to about 100 nm, or less than or equal to about 50 nm; and/or (b) the nanoemulsion particles have an average diameter of about 350 nm.
  • the quaternary ammonium compound is: (a) monographed by the US FDA as an antiseptic for topical use; (b) benzalkonium chloride (BZK); and/or (c) BZK present in a concentration of from about 0.05% to about 0.40%; BZK present in a concentration of from about 0.10% to about 0.20%; or BZK present in a concentration of about 0.13%; and/or (d) cetylpyridimium chloride (CPC); and/or (e) CPC present in a concentration of from about 0.05% to about 0.40%; CPC present in a concentration of from about 0.15% to about 0.30%; or CPC present in a concentration of about 0.20%; and/or (f) benzethonium chloride (BEC); and/or (g) BEC present in a concentration of from about 0.05% to about 1%; BEC present in a concentration of
  • the nonionic surfactant is: (i) selected from the group consisting of a poloxamer surfactant, polysorbate surfactant, Triton ® X- 100, nonoxynol-9, and any combination thereof; and/or (ii) selected from the group consisting of polysorbate 20, polysorbate 21, polysorbate 40, polysorbate 60, polysorbate 61, polysorbate 65, polysorbate 80, polysorbate 81, and polysorbate 85; and/or (iii) selected from the group consisting of poloxamer 407, poloxamer 101, poloxamer 105, poloxamer 108, poloxamer 122, poloxamer 123, poloxamer 124, poloxamer 181, poloxamer 182, poloxamer 183, poloxamer 184, poloxamer 185, poloxamer 188, poloxamer 212, poloxamer 215, poloxamer
  • the organic solvent comprises a C1-C12 alcohol, diol, or triol, a dialkyl phosphate, a trialkyl phosphate or a combination thereof; and/or (ii) comprises and alcohol selected from the group consisting of ethanol, isopropyl alcohol, glycerol or a combination thereof.
  • the oil comprises soybean oil, mineral oil, avocado oil, squalene oil, olive oil, canola oil, corn oil, rapeseed oil, safflower oil, sunflower oil, fish oils, flavor oils, cinnamon bark, coconut oil, cottonseed oil, flaxseed oil, pine needle oil, silicon oil, essential oils, water insoluble vitamins, other plant oil, or a combination thereof; and/or (ii) comprises soybean oil.
  • the nanoemulsion further comprises a chelating agent.
  • the chelating agent can be, for example, (a) ethylenediaminetetraacetic acid (EDTA), ethylene glycol -bis(P-ami noethyl ether)-N,N,N',N'-tetraacetic acid (EGTA), or a combination thereof; and/or (b) ethylenediaminetetraacetic acid (EDTA); and/or (c) EDTA and EDTA is present at a concentration of about 0.0005% to about 1% (w/w).
  • EDTA ethylenediaminetetraacetic acid
  • EGTA ethylene glycol -bis(P-ami noethyl ether)-N,N,N',N'-tetraacetic acid
  • EDTA ethylenediaminetetraacetic acid
  • EDTA and EDTA is present at a concentration of about 0.0005% to about 1% (w/w).
  • the nanoemulsion comprises: (i) about 5 vol. % to about 50 vol. % (w/w) of aqueous phase; (ii) about 30 vol. % to about 90 vol.
  • % (w/w) of a pharmaceutically acceptable oil phase (iii) about 0.1% up to about 20% of a pharmaceutically acceptable organic solvent; (iv) about 3 vol. % to about 15 vol. % (w/w) of a nonionic surfactant; and (v) about 0.05% to about 3 vol. % (w/w) of a quaternary ammonium compound.
  • the nanoemulsion comprises: (i) about 5 vol. % to about 50 vol. % (w/w) of aqueous phase; (ii) about 30 vol. % to about 90 vol.
  • % (w/w) soybean oil (iii) about 0.1% up to about 20% (w/w) glycerol; (iv) about 3 vol. % to about 15 vol. % (w/w) poloxamer 407; (v) benzalkonium chloride at a concentration of about 0.13% (w/w); and (vi) about 0.0005% to about 1% (w/w) EDTA.
  • the nanoemulsion in a further aspect of the nanoemulsion of the disclosure, the nanoemulsion: (a) is non toxic in humans and animals; and/or (b) is thermostable; and/or (c) is stable for at least 3 months at about 50°C; and/or (d) is stable for at least 3 months at 40°C; and/or (e) is stable for at least 3 months at 25°C; and/or (f) is stable for at least 3 months at 5°C; and/or (g) is stable at 5°C for up to at least 60 months; and/or (h) is stable at 50°C for up to at least 12 months; and/or (i) has been autoclaved, and optionally wherein the nanoemulsion retains its structural and/or chemical integrity following autoclaving; (j) the nanoemulsion is formulated in soap, hand sanitizer, lotion, cream, or spray dosage form; and/or (k) the nanoemulsion is formulated liquid dosage form, solid dosage form, or semisolid dosage
  • Figure 1 shows epidermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of NE-1 formulations (0.13% BZK) with surfactant blend ratios 1:5 and 1 :9 and Purell ® Foam (0.13% BZK).
  • Figure 2 shows dermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of NE-1 formulations (0.13% BZK) with surfactant blend ratios 1:5 and 1 :9 and Purell ® Foam.
  • Figure 3 shows the epidermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of NE-1 formulations (0.13% BZK) with different surfactant blend ratios (5:1, 2:1, 1:1, 1:2, 1:5, 1:9, 1:14, 1:18, and 1 :27) and Purell ® Foam (0.13% BZK).
  • Figure 4 shows the dermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of NE-1 formulations (0.13% BZK) with different surfactant blend ratios (5:1, 2:1, 1:1, 1:2, 1:5, 1:9, 1:14, 1:18, and 1 :27) and Purell ® Foam (0.13% BZK).
  • Figure 5 shows the log killing ofNE-2 (surfactant blend ratio: 1:5; 0.13% BZK) microorganisms and virus following one-minute exposure.
  • Figure 6 shows skin hydration study results of NE-1 (surfactant blend ratio: 1:5; 0.13% BZK) and Purell ® Foam (0.13% BZK).
  • Figure 7 shows the % of BZK dispensed from the wipe (spunlace washcloth) with aqueous BZK (0.13%BZK), NE-1 (surfactant blend ratio: 1:9; 0.13% BZK), and Purell ® Foam (0.13% BZK) at the following time points: initial, 2 hours and 5 days.
  • Figure 8 shows the % of BZK dispensed from the wipe (airlaid washcloth) with aqueous BZK (0.13% BZK), NE-1 (surfactant blend ratio: 1:9; 0.13% BZK), and Purell® Foam (0.13% BZK) at the following time points: initial, 2 hours and 5 days.
  • Figure 9 shows a diagram of the mucin coated Transwell® membrane in a 24 well plate.
  • Figure 10 shows the results of the in vitro mucin permeation studies of Compound A with the commercially available intranasal product of Compound A (0.50% Compound A) and the NE-1 (surfactant blend ratio: 1 :9) with 0.50% and 0.25% of Compound A.
  • Figure 11 shows the % increase in serum levels of Compound A following intranasal administration with the commercially available intranasal product of Compound A (0.50% Compound A) and the NE-2 (surfactant blend ratios: 1 :9, 1:5, and 1 :2) and NE-4 (surfactant blend ratios: 1:5 and 1:2) formulations with 0.50% or 0.25% of Compound A.
  • NE-2 surfactant blend ratios: 1 :9, 1:5, and 1 :2
  • NE-4 surfactant blend ratios: 1:5 and 1:2
  • Figure 12 shows the serum levels of Compound A following one administration with the commercially available intranasal product of Compound A (0.50% Compound A) and the NE-2 and NE-4 formulations (surfactant blend ratios: 1:5 and 1 :2) with 0.50% of Compound A.
  • Figure 13 shows the epidermal levels of terbinafme (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE-1 formulation (surfactant ratio of 1 :9 with 1% terbinafme) with Lamisil AT ® (1% terbinafme).
  • Figure 14 shows the dermal levels of terbinafme (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of NE-1 formulation (surfactant ratio of 1:9 with 1% terbinafme) with Lamisil AT® (1% terbinafme).
  • Figure 15 shows the epidermal levels of miconazole (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of the NE-1 formulation (surfactant ratio of 1 : 12 with 2% miconazole) with Monistat® (2% miconazole).
  • Figure 16 shows the dermal levels of miconazole (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of NE-1 formulation (surfactant ratio of 1 : 12 with 2% miconazole) with Monistat® (2% miconazole).
  • Figure 17 shows the epidermal levels of salicylic acid (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE-1 formulation (surfactant ratio of 1 : 12 with 1% and 2 % salicylic acid) with Dermarest ® (3% salicylic acid).
  • Figure 18 shows the epidermal levels of hydrocortisone (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of the NE- 1 formulation (surfactant ratio of 1:9 with 1% hydrocortisone) with Cortizone-10 ® (1% hydrocortisone).
  • Figure 19 shows the dermal levels of hydrocortisone (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of NE-1 formulation (surfactant ratio of 1:9 with 1% hydrocortisone) with Cortizone-10 ® (1% hydrocortisone).
  • Figure 20 shows the epidermal levels of adapalene (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE-1 formulation (surfactant ratio of 1 :9 with 0.1% adapalene) with Differin ® (0.1% adapalene).
  • Figure 21 shows the dermal levels of adapalene (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of NE-1 formulation (surfactant ratio of 1 :9 with 0.1% adapalene) with Differin ® (0.1% adapalene).
  • Figure 22 shows the epidermal levels of peanut proteins Ara h2, Ara hi, Ara h3, and Ara hX (pg/g tissue) in human abdominal skin following one application (occluded dose of 100 pl/cm 2 , measured at 18 hours) of the NE-1 formulation (surfactant ratio of 1:6 with 0.1% peanut protein) with an aqueous formulation (0.1% peanut protein).
  • Figure 23 shows the dermal levels of peanut proteins Ara h2, Ara hi, Ara h3, and Ara hX (pg/g tissue) in human abdominal skin following one application (occluded dose of 100 pl/cm 2 , measured at 18 hours) of NE-1 formulation (surfactant ratio of 1:6), NE-2 formulation (surfactant ratio of 1 :6), and NE-3 formulation (surfactant ratio of 1 :9) with 0.1% peanut protein with aqueous formulation (0.1% peanut protein).
  • NE-1 formulation surfactant ratio of 1:6
  • NE-2 formulation surfactant ratio of 1 :6
  • NE-3 formulation surfactant ratio of 1 :9
  • Figure 24 shows the epidermal levels of BEC (pg/g tissue) in human abdominal skin following one application (single dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE formulation (surfactant ratio of 1 :6 with 0.2% BEC) with an aqueous formulation (0.2% BEC), New-Skin® spray (0.2% BEC), and CVS Liquid Bandage (0.2% BEC).
  • Figure 25 shows the dermal levels of BEC (pg/g tissue) in human abdominal skin following one application (single dose of 100 pl/cm 2 , measured at 24 hours) of the NE formulation (surfactant ratio of 1 :6 with 0.2% BEC) with an aqueous formulation (0.2% BEC), New-Skin® spray (0.2% BEC), and CVS Liquid Bandage (0.2% BEC).
  • Figure 26 shows the epidermal levels of PCMX (pg/g tissue) in human abdominal skin following one application (single dose of 100 pl/cm 2 , measured at 24 hours) of the NE formulation (surfactant ratio of 1 :6 with 3.0% PCMX) with an 70% ethanol formulation (3% PCMX).
  • Figure 27 shows the dermal levels of PCMX (pg/g tissue) in human abdominal skin following one application (single dose of 100 pl/cm 2 , measured at 24 hours) of the NE formulation (surfactant ratio of 1 :6 with 3.0% PCMX) with an 70% ethanol formulation (3% PCMX).
  • Figure 28 shows the epidermal levels of chlorhexidine (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of the NE-1 formulation (surfactant ratio of 1 :9 with 2% chlorhexidine) with a 70% IPA solution containing 2% chlorhexidine.
  • Figure 29 shows the dermal levels of chlorhexidine (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of NE-1 formulation (surfactant ratio of 1 :9 with 2% chlorhexidine) with a 70% IPA solution containing 2% chlorhexidine.
  • Figure 30 shows epidermal permeability results for nanoemulsion formulations of various nanoemulsion concentrations (0.5%, 1%, 2.5%, 5%, 10%, 20%, 30%, 40%, and 60%) and Purell ® Foam (0.13% BZK).
  • Figure 31 shows dermal permeability results for nanoemulsion formulations of various nanoemulsion concentrations (0.5%, 1%, 2.5%, 5%, 10%, 20%, 30%, 40%, and 60%) and Purell ® Foam (0.13% BZK).
  • Figure 32 shows epidermal permeability results for nanoemulsion formulations relative to their viscosity (1.33 cp, 1.36 cp, 1.37 cp, 1.39 cp, 1.52 cp, 2.06 cp, 3.32 cp, 6.08 cp, and 261 cp) and Purell ® Foam (0.13% BZK).
  • Figure 33 shows dermal permeability results for nanoemulsion formulations relative to their viscosity (1.33 cp, 1.36 cp, 1.37 cp, 1.39 cp, 1.52 cp, 2.06 cp, 3.32 cp, 6.08 cp, and 261 cp) and Purell ® Foam (0.13% BZK).
  • Figure 34 shows epidermal permeability results for nanoemulsion formulations relative to their zeta potential (75.2 mV, 47.6 mV, 34.7 mV, 34.8 mV, 28.3 mV, 27.8 mV, 27.0 mV, 27.4 mV) and Purell ® Foam (0.13% BZK).
  • Figure 35 shows dermal permeability results for nanoemulsion formulations relative to their zeta potential (75.2 mV, 47.6 mV, 34.7 mV, 34.8 mV, 28.3 mV, 27.8 mV, 27.0 mV, 27.4 mV) and Purell ® Foam (0.13% BZK).
  • Figure 36 shows epidermal permeability results for nanoemulsion formulations relative to their entrapment of the quaternary ammonium salt (19.86%, 11.04%, 2.85%, 1.48%, 0.86%, 0.57%, 0.32%, 0.26%, 0.21%) and Purell ® Foam (0.13% BZK).
  • Figure 37 shows dermal permeability results for nanoemulsion formulations relative to their entrapment of the quaternary ammonium salt (19.86%, 11.04%, 2.85%, 1.48%, 0.86%, 0.57%, 0.32%, 0.26%, 0.21%) and Purell ® Foam (0.13% BZK).
  • Figure 38 shows epidermal permeability results for nanoemulsion formulations of the disclosure relative to the formulation’s stability as measured by the percent (%) change in mean droplet size following prolonged centrifugation (0.2%, 2.0%, 0.5%, 1.8%, 2.9%, 2.2%, 5.4%, 0.2%, 0.5%) and Purell ® Foam (0.13% BZK).
  • Figure 39 shows dermal permeability results for nanoemulsion formulations of the disclosure relative to the formulation’s stability as measured by the percent (%) change in mean droplet size following prolonged centrifugation (0.2%, 2.0%, 0.5%, 1.8%, 2.9%, 2.2%, 5.4%, 0.2%, 0.5%) and Purell ® Foam (0.13% BZK).
  • Figure 40 shows epidermal levels of lidocaine delivered by Salonpas patch (left), nanoemulsion (NB liquid, center), and nanoemulsion patch (NB patch, right).
  • Figure 41 shows dermal levels of lidocaine delivered by Salonpas patch (left), nanoemulsion (NB liquid, center), and nanoemulsion patch (NB patch, right).
  • Figure 42 shows levels of transdermal lidocaine delivered to the receptor by Salonpas patch (left), nanoemulsion (NB liquid, center), and nanoemulsion patch (NB patch, right).
  • Figure 43 shows images depicting nanoemulsion sample after centrifugation. Image taken under normal lighting conditions (left) and corresponding negative image (right).
  • Figure 44A shows shows epidermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of NE-1 formulations (NanoBio protect ) (0.13% BZK) with a surfactant blend ratio of 1 :9, Purell ® Foam (0.13% BZK), and aqueous 0.13% BZK.
  • Figure 44B shows dermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of NE-1 formulations (0.13% BZK) with a surfactant blend ratio of 1 :9, Purell ® Foam (0.13% BZK), and aqueous 0.13% BZK.
  • Figure 45 shows cross sections of nasal mouse epithelium 24 hours post application of green fluorescent protein (GFP) in aqueous solution (left) (Fig. 45A) and in nanoemulsion with a surfactant blend ratio of 1 :9 (right) (Fig. 45B).
  • GFP green fluorescent protein
  • Figure 46 shows nasal nanoemulsion antiseptic formulations (NE1, NE2, and NE3, having different surfactant ratios) significantly enhanced survival in mice that were challenged with a lethal dose of influenza virus 90 minutes after application.
  • Pretreatment of mouse nares with three nanoemulsion formulations followed by five minute exposure to aerosolized influenza A virus at a concentration of 5xl0 5 pfu/ml was performed to determine the ability of these compounds to protect mice against inhaled virus particles.
  • Control mice were pretreated with an intranasal application of PBS. 81.25% (13/16) of mice pretreated with PBS died, while 31.91% (15/47) of mice pretreated with nanoemulsion died.
  • Figure 47 shows that in a nanoemulion, the active or quaternary ammonium compound (e.g., BZK) resides at the interface between the oil and water phases of the nanodroplet, s with the hydrophobic tail distributed in the oil core and the polar cationic head group residing at the water phase.
  • the active or quaternary ammonium compound e.g., BZK
  • Figure 48 shows log reduction in viral and bacterial pathogens treated with a topical, persistent antimicrobial nanoemulsion having a preferred ratio of quaternary ammonium compoundmon-ionic surfactant.
  • Figure 49 shows skin substantivity of BZK from a nanoemulsion comprising BZK as compared to aqueous BZK after water and soap-water washing procedure at 4 hours and at 8 hours following application.
  • Figure 50 shows skin hydration levels following one topical application of NE-BZK and two commercially available hand sanitizers.
  • the present invention is directed to the surprising discovery that antibacterial nanoemulsion compositions, applied either topically, mucosally, ocularly or nasally, and comprising a quaternary ammonium compound and a nonionic surfactant, where the quaternary ammonium compound and surfactant are present in a defined ratio, exhibit highly unexpected persistence when applied to a skin or mucosal surface.
  • the highly unexpected substantivity (e.g., “stickiness”) of the compositions to a biological surface means that the compositions can function as long acting sanitizers, killing 99.9% of microorganisms present on the application site (e.g., skin or mucosal surface) at the time of application or for up to about 12 hours after application.
  • the compositions function to kill 99.9% of microorganisms, such as bacteria and viruses, present at the application site at the time of application.
  • the compositions also kill 99.9% of microoganisms exposed to the application site for up to 12 hours after application.
  • This “substantivity” is similar to the function of sunscreens and insect repellant, which function for a period of time after application.
  • the disclosure encompasses methods of treating and/or preventing a microbial infection, e.g., a viral or bacterial infection, comprising topically, mucosally, ocularly, or nasally (e.g., via a spray or swab) administering an antibacterial nanoemulsion composition described herein.
  • a microbial infection e.g., a viral or bacterial infection
  • nasally e.g., via a spray or swab
  • the site of application can be, for example, mucosa, ocular, dermis, epidermis, skin, and/or squamous epithelium (the nasal vestibule is completely lined by squamous epithelium).
  • the antibacterial nanoemulsion comprises an aqueous phase, an oil phase comprising at least one pharmaceutically acceptable oil, at least one pharmaceutically acceptable organic solvent, and a combination of at least one a quaternary ammonium compound and at least one non-ionic surfactant, wherein the ratio of the concentration of the quaternary ammonium compound to nonionic surfactant is about 5:1 to about 1:27. In other embodiments, the ratio of the concentration of the quaternary ammonium compound to nonionic surfactant is about 1:2 to about 1:18.
  • compositions to be used in the methods described herein are compositions to be used in the methods described herein.
  • Nanoemulsions kill viruses at concentrations that are nontoxic in humans.
  • the nanoemulsions function by fusing with lipid bilayers of cell membranes, thereby destabilizing the lipid membrane of the microbial pathogen.
  • the nanoemulsion “dissolves” the membrane or envelope surrounding the microbe (e.g., virus or bacteria), thus inactivating the virus or bacteria and eliminating the ability of the virus or bacteria to infect a host.
  • the antimicrobial activity of nanoemulsions is nonspecific, unlike that of typical small molecule antimicrobial compositions, thus allowing broad-spectrum antimicrobial activity while limiting the capacity for the generation of resistance.
  • a key aspect of the disclosure is that the described nanoemulsions have a high permeation and residence time at the site of mucosal, ocular, or skin application, with significant microbial-killing nanoemulsion residing at the side of application for an extended period of time - e.g., about 4, about 8, or about 12 hours or more.
  • the nanomemulsions can be routinely applied to sites of potential microbial infection, e.g., skin, nasal mucosa, ocular sites, and the mouth area, and the nanoemulsions will function to kill any microbes present at the application site, thereby preventing microbial infection.
  • the nanoemulsion will kill microorganisms which are present at the site of nanoemulsion application, where the microorganism is present either before or after the nanoemulsion is applied. This is because the nanoemulsion will function to kill microorganisms already present prior to application and the nanoemulsion will reside at the site of application in a sufficient amount to kill microorganisms that comes into contact with the application site up to 24 hours after nanoemulsion application.
  • Nanoemulsion compositions may be applied periodically to mucosal, ocular, or skin surfaces of subjects at risk for microbial infection. Periodic applications will allow for constant residence of the microbial-killing nanoemulsion composition at and within the biological surface, and thus defend the subject from microbial particles contacted with these surfaces.
  • compositions that can minimize or prevent microbial infection.
  • barrier methods comprising using medical grade masks that cover the nose and mouth combined with eye goggles or face shields. The present disclosure therefore satisfies an urgent need in the art.
  • An exemplary patient population includes subjects susceptible to exposure to microbial infections, such as healthcare workers, subjects in coronavirus quarantine zones, subjects caring for COVID-19 patients, and subjects with pre-existing conditions including but not limited to diabetes, heart conditions, or respiratory conditions such as asthma or COPD.
  • microbial infections such as healthcare workers, subjects in coronavirus quarantine zones, subjects caring for COVID-19 patients, and subjects with pre-existing conditions including but not limited to diabetes, heart conditions, or respiratory conditions such as asthma or COPD.
  • the nasal cavity is a primary route of entry for viral and bacterial respiratory pathogens.
  • Recently published studies on SARS-CoV-2 highlight the prominent role of nasal epithelium in initial infection, viral replication and transmission (He et al., “Temporal dynamics in viral shedding and transmissibility of COVID-19,” Nat. Med. (2020); Zou et al., “SARS-CoV-2 Viral Load in Upper Respiratory Specimens of Infected Patients,” N. Engl. ./. Med ., 382: 1177-1179 (2020); and Wang et al., “Detection of SARS-CoV-2 in Different Types of Clinical Specimens,” JAMA, El (2020)).
  • Bacterial pathogens such as Staphylococcus aureus
  • Bacterial pathogens can reside in nasal mucosal tissues for long periods serving as a source for infection in other areas of the host or opportunistic respiratory infections (Chmielowiec-Korzeniowska et al., “ Staphylococcus aureus carriage state in healthy adult population and phenotypic and genotypic properties of isolated strains. Postepy Dermatol Alergok, 37(2):184-189 (Apr. 2020)).
  • An exemplary nanoemulsion described herein is a nanoemulsion comprising benzalkonium chloride as the quaternary ammonium compound (NE-BZK).
  • This composition incorporates the FDA-monographed active ingredient benzalkonium chloride (0.13% BZK) in plant oil nanodroplets emulsified in water.
  • the nanodroplets are approximately 350 nm in size and have positively charged surfaces. Given their size, they persist following application for up to 24 hours in hair follicles, sweat glands and sebaceous glands which can harbor pathogens.
  • the nanodroplets repel each other which keeps the active ingredient BZK from crystallizing, which has clinical relevance in that once crystallized on the skin, BZK loses its antimicrobial activity and can cause skin irritation.
  • the surfaces of enveloped viruses e.g. SARS-CoV-2, influenza, RSV
  • cell-walled bacteria e.g. Staphylococcus, Enterococcus, Pseudomonas
  • Zanin et ah “The interaction between respiratory pathogens and mucus,” Cell Host Microbe, 19(2): 159-168 (2016). Therefore, the positively charged nanodroplets in NE-BZK are attracted to these microbes, delivering the antiseptic directly to the surface of the pathogen where killing occurs.
  • Ethyl alcohol products comprise over 80% of skin sanitizers available in the U.S. and have been promoted by the Centers for Disease Control and Prevention as preferred for infection risk reduction (https://wvw cdc.gov/coronavirus/2019-ncov/hcp/itand-hvgiene.html: and Guideline for Hand Hygiene in Health-Care Settings Recommendations of the Healthcare Infection Control Practices Advisory Committee and the HICPAC/SHEA/APIC/IDSA Hand Hygiene Task Force; CDC Morbidity and Mortality Weekly Report October 25, 2002 / Vol. 51 / No. RR-16). Aqueous 0.13% BZK preparations are also widely available.
  • the antispectic, antimicrobial nanoemulsions described herein are a novel approach which can be used as an alternative to conventional topical and mucosal formulations to increase substantivity.
  • This system comprises the active (e.g. a quaternary ammonium compound such as BZK) and upon contact with the skin, leaves behind a layer of excipients along with the active upon evaporation.
  • the formed layer is the nanoemulsion droplet material that act a residual liquid film which is rapidly adhering to the stratum corneum.
  • the co-surfactant in the nanoemulsion is a thermo-responsive hydrogel, which is extremely bioadhesive at the surface temperature of the skin and increases the interaction between two materials, the nanoemulsion droplets and the skin stratum corneum for a given period through interfacial forces.
  • thermo-responsive hydrogel which is extremely bioadhesive at the surface temperature of the skin and increases the interaction between two materials, the nanoemulsion droplets and the skin stratum corneum for a given period through interfacial forces.
  • NE-BZK demonstrated broad-spectrum in-vitro activity against every enveloped virus, gram-positive bacteria and gram-negative bacteria tested including but not limited to SARS- CoV-2, Influenza and MRSA.
  • Coronaviruses are a family of hundreds of viruses that can cause fever, respiratory problems, and sometimes gastrointestinal symptoms. Coronavirus Disease 2019 (COVID-19) is one of seven members of this family known to infect humans, and the third in the past three decades to jump from animals to humans. Since emerging in China in December 2019, this new coronavirus has caused a global health emergency.
  • Patient populations at risk for more serious COVID-19 disease include elderly subjects, those with weakened immune systems, and those with preexisting health conditions, such diabetes, heart disease, and asthma or other respiratory conditions such as COPD.
  • “Elderly subjects” can be defined as subjects aged about 50 or older, aged about 55 or older, aged about 60 or older, aged about 65 or older, aged about 70 or older, aged about 75 or older, or aged about 80 or older.
  • Human coronaviruses most commonly spread from an infected person to others through respiratory droplets produced when an infected person coughs or sneezes, close personal contact (such as caring for or living with an infected person), or touching an object or surface with the virus on it and then touching the mouth or eyes prior to hand washing.
  • Three human coronaviruses SARS-CoV, MERS-CoV, and 2019-nCoV are also thought to spread from infected animals to people through contact.
  • the new 2019-nCoV virus spreads much more readily than the one that caused severe acute respiratory syndrome, or SARS (also a coronavirus).
  • Viruses such as influenza and coronavirus infect a subject by entering the nasal or respiratory tract, where they replicate in epithelial cells. Interruption of the replication process can be a tactic used to prevent infection. Moreover, coronaviruses need a period of time to replicate for successful infection. Thus, this period of time offers a window into an infection prevention strategy, as well as a method for minimizing the risk of infection.
  • coronavirus can be transmitted via air.
  • droplet form the coronavirus is airborne for a few seconds after someone sneezes or coughs.
  • a coronavirus droplet is able to travel only a short distance before gravitational forces pull it down. Someone close enough for the virus particles to reach in that brief period can therefore be infected. So can anyone who comes into contact with virus-containing droplets that fall onto a surface.
  • the new coronavirus can survive on surfaces for several hours; hence the importance of hand-washing after touching a surface in a public place.
  • An aerosol is a wholly different physical state:
  • Fog is an aerosol; water droplets are suspended in air. The suspended particles remain for hours or more, depending on factors such as heat and humidity. If virus particles, probably on droplets of mucus or saliva, can be suspended in air for more than a few seconds, as the measles virus can, then anyone passing through that pathogenic cloud could become infected. Nevertheless, the fact that comonaviruses spread via airborne droplets demonstrates the need for more rigorous methods of preventing or minimizing the risk of infection, particularly for patent populations at greater risk for infection, or for patent populations at greater risk for significant adverse effects following infection.
  • Example 20 details experimental results demonstrating the persistence, or substantivity, of the antibacterial nanoemulsions when applied to a skin or mucosal surface.
  • a nanoemulsion comprising the quaternary ammonium compound BSK at 0.13% concentration.
  • Fig. 48 illustrates the antiviral and antibacterial activity of the nanoemulsion composition tested (NE- BZK) against several common pathogens, including the novel coronavirus SARS-CoV-2, HCoV 229E, Influenza B, RSV, S. aureus , E. faecium , S. epidermidis , C A-MRS A, P. aeruginosa, S. marcescens, A. baumannii, and K. pneumoniae.
  • NE-BZK deactivated >99.99% of all viruses following five minutes of exposure (the earliest time point measured) and >99.99% of all bacterial pathogens following one minute of exposure.
  • Example 21 details the results of an evaluation of the antibacterial activity against human coronavirus and MRSA of an antibacterial nanoemulsion comprising BZK (NE-BZK) as compared to aqueous BZK (AQ-BZK).
  • Antiviral activity was measured against human coronavirus (HCoV229E) in a time-kill study following 5 minutes exposure.
  • HCV229E human coronavirus
  • both the nanoemulsion antiseptic and AQ-BZK formulations achieved >99.99% killing when formulated at full strength or a 1/10 dilution.
  • the aqueous AQ-BZK formulation lost all activity while in dramatic contrast the nanoemulsion antiseptic continued to demonstrate >99.99% killing.
  • FIG. 49 and Table 34 shows that a model nanoemulsion comprising BZK (NE-BZK) exhibited a 5.8 and 3.8 fold increase substantivity on volunteers’ skin surface at 4 hours after two washing protocols (e.g. (1) water rinse and (2) soap rub +water rinse) and 16.5 and 7.3 fold increase, respectively, after 8 hours after a two washing protocols as compared to a 0.13% AQ- BZK solution.
  • two washing protocols e.g. (1) water rinse and (2) soap rub +water rinse
  • Example 21 details measurement of antiviral activity of the model nanoemulsion NE-BZK against human coronavirus (HCoV229E) ex vivo in a time-kill study following 15 minutes exposure of skin pre-treated with the nanoemulsion antiseptic (0.13% BZK) or AQ-BZK for 4 and 8 hours.
  • NE-BZK achieved >4.7% log killing at both the 4- and 8hour time points.
  • aqueous BZK exhibited only 1.5 log killing at 4 hours and below the limit of detection at 8 hours.
  • NE-BZK the antimicrobial activity of the model nanoemulsion NE-BZK was measured against MRS A ex vivo in a time-kill study following 15 minutes exposure of skin pre-treated and compared to AQ-BZK (0.13% BZK) and an alcohol-based nasal sanitizer (0.62% ethyl alcohol).
  • AQ-BZK 0.13% BZK
  • alcohol-based nasal sanitizer 0.62% ethyl alcohol
  • Example 6 shows that in a comparison of a non-nanoemulsion formulation having 0.13% BKC (Purell ® Foam) with nanoemulsion (NE) formulations having 0.13% BZK and surfactant blend ratios of 1:5 and 1:9, the amount of BZK delivered into human abdominal skin epidermal tissue was almost 600% higher for the nanoemulsion formulation having a 1 :9 surfactant blend ratio as compared to the non-nanoemulsion formulation (6642 ng BZK/gram tissue, as compared to 953 ng BZK/gram tissue for the Purell ® Foam).
  • Figs. 2 epidermis
  • 3 dermis
  • graphs of levels of BZK pg/g tissue
  • the nanoemulsion formulation delivered almost 4 to 7 times more BZK into the epidermis as compared to a marketed 0.13% Purell ® Foam (Fig. 1).
  • the nanoemulsion formulation delivered 3 to 4 times more BZK as compared to the marketed product, Purell ® Foam, indicating that the BZK was able to penetrate into the deeper dermal levels of the skin from the nanoemulsion formulations (Fig. 2).
  • nanoemulsions having representative surfactant ratios of 1:5 and 1:9 showed dramatic and significantly greater permeation (amount of BZK (ng)/ tissue weight (g)) as compared to a non-nanoemulsion formulation having the same quantity of BZK.
  • FIG. 3 A clear bell curve of permeation vs. surfactant blend ratio is depicted in Figs. 3 and 4, demonstrating that nanoemulsions having a preferred surfactant blend ratio show dramatic and significant increased permeation in the epidermis (Fig. 3) and dermis (Fig. 4) as compared to non-nanoemulsion formulations of the same quaternary ammonium compound at the same concentration (Purell ® Foam), and as compared to nanoemulsion formulations having surfactant blend ratios outside the claimed range of about 5: 1 up to about 1 :27. Outside the claimed surfactant blend ratio, the amount of drug in the epidermis (Fig. 3) and dermis (Fig. 4) is dramatically less. The impact of the claimed narrow range of surfactant blend ratios on permeation was not known prior to the present invention.
  • This enhanced permeability allows for the nanoemulsion compositions described herein to deliver more of the quaternary ammonium compound to the site of application, as well as any additional therapeutic agent present in the nanoemulsion, and to also have a longer residence time at the site of application as compared to non-nanoemulsion compositions containing the same quaternary ammonium compound present at the same concentration. This property is critical to effective prevention of microbial infections.
  • the nanoemulsion compositions described herein can also comprise a therapeutic agent suitable for topical, mucosal, ocular, or intranasal delivery.
  • the enhanced permeability of the nanoemulsions described herein allows for the nanoemulsion compositions to deliver more of the therapeutic agent to a site of application, and to also have a longer residence time of the therapeutic agent at the site of application, as compared to non-nanoemulsion compositions containing the same therapeutic agent at the same concentration.
  • the site of application can be, for example, mucosa, ocular, dermis, epidermis, skin, and/or squamous epithelium (the nasal vestibule is completely lined by squamous epithelium).
  • the permeation of a representative model therapeutic agent Compound A was significantly greater when present in a nanoemulsion formulation as compared to a non-nanoemulsion formulation, having the same drug concentration.
  • the commercial product of Compound A having a drug concentration of 50% present in non-nanoemulsion formulation, showed a cumulative concentration of Compound A (pg/mL) at 6 hours following application of about 325 pg/mL, in contrast to a concentration of about 730 pg/mL for the nanoemulsion having a surfactant ratio of 1:9 and a drug concentration of 50%, an increase in drug permeation of about 125%.
  • Examples 11 and 12 show in vitro and in vivo data, respectively, for a nanoemulsion having a model Compound A incorporated within the nanoemulsion.
  • In vitro all of the nanoemulsion formulations resulted in significantly greater serum levels of Compound A (pg/mL) - all greater than about 3500 pg/mL - as compared to the conventional, non nanoemulsion formulation having the same compound at the same concentration; e.g ., about 2750 pg/mL - a difference of about 30% (Fig. 12).
  • Example 12 demonstrate that greater mucin penetration of Compound A incorporated in a nanoemulsion measured in vitro directly correlates with Compound A penetration in the nasal epithelium in vivo when animals are intranasally treated with the NE-Compound A formulations, and leads to greater systemic drug delivery as compared to the commercially available product containing the same concentration of Compound A.
  • nanoemulsion formulations having a preferred surfactant blend ratio significantly enhance the permeation of a component therapeutic agent.
  • these results show that nanoemulsion formulations having a preferred surfactant blend ratio significantly enhance the systemic absorption of a representative incorporated therapeutic agent (Compound A) in vivo as compared to a non-nanoemulsion commercial product having the same active at the same concentration.
  • a significantly lower amount of a therapeutic agent can be administered with any one of the nanoemulsion compositions described herein to achieve systemic absorption equivalent or greater than a non-nanoemulsion composition having the same therapeutic agent.
  • a method of preventing or reducing the risk of infection in a subject caused by exposure to a microorganism comprising administering a composition comprising a nanoemulsion to the skin, nasal vestibule or passages, ocular, or the mucosa of the mouth, of the subject, either before or after the microbial exposure.
  • the nanoemulsion composition can be repeatedly replied, such at least once every 24 hours, or periodically during a 24 hr period as described herein.
  • Other exemplary application schedules include about once every hour, once every about 2 hours, once every about 3 hours, once every about 4 hours, once every about 5 hours, once every about 6 hours, once every about 7 hours, once every about 8 hours, once every about 9 hours, once every about 10 hours, once every about 11 hours, once every about 12 hours, once every about 13 hours, once every about 14 hours, once every about 15 hours, once every about 16 hours, once every about 17 hours, once every about 18 hours, once every about 19 hours, once every about 20 hours, once every about 21 hours, once every about 22 hours, once every about 3 hours, once every about 4 hours, once every about 5 hours, once every about 23 hours, or once every about 24 hours.
  • the enhanced permeability also results in increased skin, mucosa, and/or squamous epithelium hydration.
  • the increase in skin, mucosa, and/or squamous epithelium hydration can be about 25%, about 50%, about 75%, about 100%, about 125%, about 150%, about 175%, or about 200%, as compared to the skin, mucosa, and/or squamous epithelium hydration prior to application of the nanoemulsion.
  • the nanoemulsions described herein with a specific surfactant blend ratio exhibit surprising and unexpected long-term stability even at high temperatures.
  • Example 8 details data demonstrating that a nanoemulsion having a surfactant blend ratio of 1 :5 was stable for 1 month even at the most extreme storage condition of 50 °C (122 °F). Additional data (not shown) demonstrates that nanoemulsions according to the invention, including nanoemulsions comprising an incorporated therapeutic agent, are stable for at least 3 months at up to 50 °C, up to 12 months at 50 °C, and up to 60 months at 5 °C. This is highly unexpected.
  • a nanoemulsion is a composition comprising an aqueous phase, at least one oil, and at least one organic solvent.
  • emulsion refers to, without limitation, any oil-in-water dispersions or droplets, including lipid structures that can form as a result of hydrophobic forces that drive apolar residues (e.g., long hydrocarbon chains) away from water and polar head groups toward water, when a water immiscible phase is mixed with an aqueous phase.
  • lipid structures include, but are not limited to, unilamellar, paucilamellar, and multilamellar lipid vesicles, micelles, and lamellar phases.
  • the nanoemulsion can comprise an aqueous phase, at least one pharmaceutically acceptable oil, at least one pharmaceutically acceptable organic solvent, at least one pharmaceutically acceptable quaternary ammonium compound selected from the group consisting of benzalkonium chloride (BZK), cetylpyridimium chloride (CPC), benzethonium chloride (BEC), dioctadecyl dimethyl ammonium chloride (DODAC), and octenidine dihydrochloride (OCT); and at least one pharmaceutically acceptable nonionic surfactant.
  • BZK benzalkonium chloride
  • CPC cetylpyridimium chloride
  • BEC benzethonium chloride
  • DODAC dioctadecyl dimethyl ammonium chloride
  • OCT octenidine dihydrochloride
  • the concentration ratio of the quaternary ammonium compound to nonionic surfactant is from about 5: 1 to about 1: 27.
  • the nanoemulsion comprises droplets having an average or mean particle size diameter of less than about 1 micron or less than about 1000 nm, less than about 950 nm, less than about 900 nm, less than about 850 nm, less than about 800 nm, less than about 750 nm, less than about 700 nm, less than about 650 nm, less than about 600 nm, less than about 550 nm, less than about 500 nm, less than about 450 nm, less than about 400 nm, less than about 350 nm, less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, or less than about 100 nm.
  • the nanoemulsion comprises droplets having an average or mean particle size diameter of less than about 1000 nm.
  • the nanoemulsion comprises droplets having an average or mean particle size diameter of about 250 nm to about 1000 nm.
  • the nanoemulsion composition described herein comprises BZK at a concentration of about 0.13%, poloxamer 407, soybean oil, EDTA, and water.
  • the nanoemulsion composition comprises an aqueous phase.
  • the aqueous phase may be any type of aqueous phase including, but not limited to, water (e.g., EhO, distilled water, tap water), solutions (e.g., phosphate-buffered saline (PBS) solution), or any combination thereof.
  • the aqueous phase comprises water at a pH of about 4 to about 10, preferably about 6 to about 8.
  • the aqueous phase is deionized.
  • the aqueous is purified.
  • the aqueous phase is sterile and/or pyrogen free.
  • the aqueous phase is present in a concentration that is greater than about 50%, greater than about 55%, greater than about 60%, greater than about 65%, greater than about 70%, greater than about 75%, greater than about 80%, greater than about 85%, greater than about 90%, or greater than about 95%. In some embodiments, the aqueous phase is present in a concentration that is from about 50% to about 99%.
  • the nanoemulsion compositions described herein comprise at least one oil.
  • the oil in the nanoemulsion composition described herein may be any cosmetically or pharmaceutically acceptable oil.
  • the oil may be volatile or non-volatile, and may be chosen from animal oil, plant oil, vegetable oil, natural oil, synthetic oil, hydrocarbon oils, silicone oils, semi-synthetic derivatives thereof, and combinations thereof.
  • the oil is an animal oil, plant oil, or a vegetable oil.
  • the oil is present in a concentration that is equal to or less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, or less than about 1%. In some embodiments, the oil is present in a concentration that is from about 1% to about 30%.
  • Suitable oils include, but are not limited to, mineral oil, squalene oil, flavor oils, silicon oil, essential oils, water insoluble vitamins, isopropyl stearate, butyl stearate, octyl palmitate, cetyl palmitate, tridecyl behenate, diisopropyl adipate, dioctyl sebacate, menthyl anthranhilate, cetyl octanoate, octyl salicylate, isopropyl myristate, neopentyl glycol dicarpate cetols, Ceraphyls®, decyl oleate, diisopropyl adipate, C12-15 alkyl lactates, cetyl lactate, lauryl lactate, isostearyl neopentanoate, myristyl lactate, isocetyl stearoyl stearate, octyld
  • the oil comprises soybean oil, avocado oil, squalene oil, olive oil, canola oil, corn oil, rapeseed oil, safflower oil, sunflower oil, fish oils, cinnamon bark, coconut oil, cottonseed oil, flaxseed oil, pine needle oil, silicon oil, mineral oil, essential oil, flavor oils, water insoluble vitamins, and combinations comprising one or more of the foregoing oils.
  • the oil comprises soybean oil.
  • the nanoemulsions described herein can optionally comprise at least one organic solvent.
  • Organic solvents contemplated for use include but are not limited to C1-C12 alcohols, diols, triols, or a combination thereof.
  • Organic phosphate solvents, alcohols and combinations thereof are also contemplated for use as organic solvents.
  • Suitable organic phosphate solvents include, but are not limited to, dialkyl and trialkyl phosphates having one to ten carbon atoms, more preferably two to eight carbon atoms.
  • the alkyl groups of the di- or trialkyl phosphate can all the same or the alkyl groups can be different.
  • the trialkyl phosphate is tri-n- butyl phosphate.
  • the organic solvent comprises a C1-C12 alcohol, diol, or triol, a dialkyl phosphate, a trialkyl phosphate, or a combination thereof. In some embodiments, the organic solvent is present in a concentration that is about 0.1% up to about 20%, less than about 15%, less than about 10%, less than about 5%, less than about 1%, less than about 0.5%, less than about 0.1%. In some embodiments, the organic solvent is present in a concentration that is from about 0.1% to about 5%.
  • Suitable organic solvents for the nanoemulsion include, but are not limited to, ethanol, methanol, isopropyl alcohol, glycerol, medium chain triglycerides, diethyl ether, ethyl acetate, acetone, dimethyl sulfoxide (DMSO), acetic acid, «-butanol, butylene glycol, perfumers alcohols, isopropanol, «-propanol, formic acid, propylene glycols, glycerol, sorbitol, industrial methylated spirit, triacetin, hexane, benzene, toluene, diethyl ether, chloroform, 1,4-dixoane, tetrahydrofuran, dichloromethane, acetone, acetonitrile, dimethylformamide, dimethyl sulfoxide, formic acid, semi-synthetic derivatives thereof, and a combination thereof.
  • DMSO dimethyl s
  • the quaternary ammonium compound may be benzalkonium chloride (BZK), cetylpyridinium chloride (CPC), benzethonium chloride (BEC), dioctadecyl dimethyl ammonium chloride (DODAC) and/or octenidine dihydrochloride (OCT).
  • BZK benzalkonium chloride
  • CPC cetylpyridinium chloride
  • BEC benzethonium chloride
  • DODAC dioctadecyl dimethyl ammonium chloride
  • OCT octenidine dihydrochloride
  • the quaternary ammonium compound is a cationic surfactant or is part of a zwitterionic surfactant.
  • BZK is present as the quaternary ammonium compound, then the BZK is present at a concentration of from about 0.05% to about 5.0%, or any amount in-between these two amounts. In some embodiments, the BZK is present at a concentration of from about 0.05% to about 0.40%. In some embodiments, the BZK is present at a concentration of from about 0.05% to about 0.20%. In some embodiments, the BZK is present at a concentration of from about 0.10% to about 0.20%. In some embodiments, the BZK is present at a concentration of from about 0.10% to about 0.15%.
  • the BZK is present at a concentration of about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.10%, about 0.11%, about 0.12%, about 0.13%, about 0.14%, about 0.15%, about 0.16%, about 0.17%, about 0.18%, about 0.19%, about 0.20%, about 0.21%, about 0.22%, about 0.23%, about 0.24%, about 0.25%, about 0.26%, about 0.27%, about 0.28%, about 0.29%, about 0.30%, about 0.31%, about 0.32%, about 0.33%, about 0.34%, about 0.35%, about 0.36%, about 0.37%, about 0.38%, about 0.39%, or about 0.40%.
  • the BZK is present at a concentration of 0.13%.
  • the quaternary ammonium compound is monographed by the US FDA as an antiseptic for topical use.
  • the monographed quaternary ammonium compound can be BZK.
  • cetylpyridinium chloride is present as the quaternary ammonium compound, then the CPC is present at a concentration of from about 0.05% to about 5.0%, or any amount in- between these two amounts. In some embodiments, the CPC is present at a concentration of from about 0.05% to about 0.40%. In some embodiments, the CPC is present at a concentration of from about 0.05% to about 0.20%. In some embodiments, the CPC is present at a concentration of from about 0.15% to about 0.30%. In some embodiments, the CPC is present at a concentration of from about 0.08% to about 0.15%. In some embodiments, the CPC is present at a concentration of about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about
  • the CPC is present at a concentration of 0.10%. In some embodiments, the CPC is present at a concentration of 0.20%.
  • BEC benzethonium chloride
  • the BEC is present at a concentration of from about 0.05% to about 5.0%, or any amount in- between these two amounts. In some embodiments, the BEC is present in a concentration of: (a) from about 0.05% to about 1%; or (b) from about 0.10% to about 0.30%.
  • the BEC is present at a concentration of about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.10%, about 0.11%, about 0.12%, about 0.13%, about 0.14%, about 0.15%, about 0.16%, about 0.17%, about 0.18%, about 0.19%, about 0.20%, about 0.21%, about 0.22%, about 0.23%, about 0.24%, about 0.25%, about 0.26%, about 0.27%, about 0.28%, about 0.29%, or about 0.30%. In some embodiments, the BEC is present in a concentration of about 0.2%.
  • DODAC dioctadecyl dimethyl ammonium chloride
  • the DODAC is present at a concentration of about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.10%, about 0.11%, about 0.12%, about 0.13%, about 0.14%, about 0.15%, about 0.16%, about 0.17%, about 0.18%, about 0.19%, about 0.20%, about 0.21%, about 0.22%, about 0.23%, about 0.24%, about 0.25%, about 0.26%, about 0.27%, about 0.28%, about 0.29%, about 0.30%, about 0.31%, about 0.32%, about 0.33%, about 0.34%, about 0.35%, about 0.36%, about 0.37%, about 0.38%, about 0.39%, or about 0.40%.
  • the DODAC is present in a concentration of about 0.2%.
  • OCT octenidine dihydrochloride
  • the OCT is present at a concentration of about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.10%, about 0.11%, about 0.12%, about 0.13%, about 0.14%, about 0.15%, about 0.16%, about 0.17%, about 0.18%, about 0.19%, about 0.20%, about 0.21%, about 0.22%, about 0.23%, about 0.24%, about 0.25%, about 0.26%, about 0.27%, about 0.28%, about 0.29%, about 0.30%, about 0.31%, about 0.32%, about 0.33%, about 0.34%, about 0.35%, about 0.36%, about 0.37%, about 0.38%, about 0.39%, or about 0.40%.
  • the OCT is present in a concentration of about 0.2%.
  • nonionic surfactants described herein are Generally Recognized as Safe (GRAS) by the US Food and Drug Administration.
  • GRAS Generally Recognized as Safe
  • Exemplary useful surfactants are described in Applied Surfactants: Principles and Applications, Tharwat F. Tadros (Copyright 2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-30629-3), which is specifically incorporated by reference.
  • Suitable nonionic surfactants include polysorbate surfactants (i.e., polyoxyethylene ethers), poloxamers, or a combination thereof.
  • polysorbate detergents include the following sold under the tradenames: TWEEN ® 20, TWEEN ® 21, TWEEN ® 40, TWEEN ® 60, TWEEN ® 61, TWEEN ® 65, TWEEN ® 80, TWEEN ® 81, and TWEEN ® 85.
  • Poloxamers are polymers made of a block of polyoxyethylene, followed by a block of polyoxypropylene, followed by a block of polyoxyethylene. The average number of units of polyoxyethylene and polyoxypropylene varies based on the number associated with the polymer.
  • Poloxamer 101 consists of a block with an average of 2 units of polyoxyethylene, a block with an average of 16 units of polyoxypropylene, followed by a block with an average of 2 units of polyoxyethylene.
  • poloxamers examples include, but are not limited to, Poloxamer 101, Poloxamer 105, Poloxamer 108, Poloxamer 122, Poloxamer 123, Poloxamer 124, Poloxamer 181, Poloxamer 182, Poloxamer 183, Poloxamer 184, Poloxamer 185, Poloxamer 188, Poloxamer 212, Poloxamer 215, Poloxamer 217, Poloxamer 231, Poloxamer 234, Poloxamer 235, Poloxamer 237, Poloxamer 238, Poloxamer 282, Poloxamer 284, Poloxamer 288, Poloxamer 331, Poloxamer 333, Poloxamer 334, Poloxamer 335, Poloxamer 338, Poloxamer 401, Poloxamer 402, Poloxamer 403, Poloxamer 407, Poloxamer 105 Benzoate, and Poloxamer 182 Dibenzoate.
  • Nonionic surfactants can also include, but are not limited to, an ethoxylated surfactant, an alcohol ethoxylated, an alkyl phenol ethoxylated, a fatty acid ethoxylated, a monoalkaolamide ethoxylated, a sorbitan ester ethoxylated, a fatty amino ethoxylated, an ethylene oxide-propylene oxide copolymer, Bis(polyethylene glycol bis[imidazoyl carbonyl]), nonoxynol-9, Bis(polyethylene glycol bis[imidazoyl carbonyl]), Brij ® 35, Brij ® 56, Brij ® 72, Brij ® 76, Brij ®
  • nanoemulsion compositions with certain concentration ratios of quaternary ammonium compound to nonionic surfactant provide greater delivery of the quaternary ammonium compound (or an additional active agent present in the composition) to the site of application and/or increased skin hydration when the nanoemulsions are applied to the skin as compared to non-nanoemulsion compositions comprising the same quaternary ammonium compound (or additional active agent).
  • the ratio of the concentration of the quaternary ammonium compound to nonionic surfactant is about 5: 1 to about 1 :27.
  • the ratio of the concentration of the quaternary ammonium compound to nonionic surfactant is selected from the group consisting of about 5:1, about 4:1, about 3:1, about 2:1, about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:11, about 1:12, about 1:13, about 1:14, about 1:15, about 1:16, about 1:17, about 1:18, about 1:19, about 1:20, about 1:21, about 1:22, about 1:23, about 1:24, about 1:25, about 1 :26, and about 1 :27.
  • the ratio of the concentration of the quaternary ammonium compound to the nonionic surfactant is from about 4: 1 to about 1 :27.
  • the ratio of the concentration of the quaternary ammonium compound to the nonionic surfactant is selected from the group consisting of about 1:2, about 1:5, about 1:9, about 1:14, and about 1:18. In certain embodiments, the concentration of the quaternary ammonium compound to the nonionic surfactant is about 1 :2 to about 1:18.
  • nanoemulsion compositions described herein may further comprise one or more active or therapeutic agents suitable for topical, transdermal, nasal or mucosal administration.
  • the active agents may include any active agent that kills, or inactivates a microorganism, such as a coronavirus, for example, SARS-CoV-2 (SEQ ID NO: 1).
  • a coronavirus for example, SARS-CoV-2 (SEQ ID NO: 1).
  • antiviral compounds include for example, chloroquine, darunavir, galidesivir, interferon beta, lopinavir, ritonavir, remdesivir, and triazavirin, may be included.
  • the therapeutic agent is present in a concentration of from about 0.01% to about 10%; from about 0.01% to about 1%; from about 0.01% to about 0.75%; and from about 0.1% to about 0.5%. In some embodiments, the therapeutic agent is present in a concentration of from about 0.01%, about 0.02%, about 0.05%, about 0.1%, about 0.15%, about 0.2%, about 0.25%, about 0.3%, about 0.35%, about 0.4%, about 0.45%, about 0.5%, about 0.55%, about 0.6%, about 0.65%, about 0.7%, about 0.75%, about 0.8%, about 0.85%, about 0.9%, about 0.95%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%.
  • the amount present can be from about 1 to about 250 pg/per dose.
  • the composition when the composition further comprises a therapeutic or active agent, after a single application of the composition topically, transdermally, nasally, or mucosally (e.g. intranasal, ocular), the composition delivers a greater amount of therapeutic agent to the dermis, epidermis, mucosa, and/or squamous epithelium as compared to a composition comprising the same therapeutic agent at the same concentration but lacking a nanoemulsion, and applied using the same method, measured at any suitable time point after application.
  • a therapeutic or active agent after a single application of the composition topically, transdermally, nasally, or mucosally (e.g. intranasal, ocular)
  • the composition delivers a greater amount of therapeutic agent to the dermis, epidermis, mucosa, and/or squamous epithelium as compared to a composition comprising the same therapeutic agent at the same concentration but lacking a nanoemulsion, and applied using the same method, measured at any
  • the composition after a single application of the composition to skin, mucosa, or squamous epithelium, the composition delivers at least about 25% more of the therapeutic agent to the epidermis, and/or at least about 25% more of the therapeutic agent to the dermis, and/or about 25% more of the therapeutic agent to the mucosa, and/or about 25% more of the therapeutic agent to the squamous epithelium as compared to a composition comprising the same therapeutic agent at the same concentration but lacking a nanoemulsion, and applied using the same method, measured at any suitable time point after application.
  • the composition when the composition further comprises a therapeutic or active agent, after a single application or administration of the composition topically, transdermally, mucosally, ocularly, or nasally, the composition delivers at least about 25%, at least about 50%, at least about 100%, at least about 125%, at least about 150%, at least about 175%, at least about 200%, at least about 225%, at least about 250%, at least about 275%, at least about 300%, at least about 325%, at least about 350%, at least about 375%, at least about 400%, at least about 425%, at least about 450%, at least about 475%, or at least about 500% more of the therapeutic agent to the dermis, epidermis, mucosa, and/or squamous epithelium as compared to a composition comprising the same therapeutic agent at the same concentration but lacking a nanoemulsion, and applied using the same method, measured at any suitable time point after application or administration.
  • the composition when the composition further comprises a therapeutic or active agent, after a single application or administration of the composition topically, transdermally, ocularly, nasally, or mucosally, the composition has a longer residence time at the site of application or administration as compared to a composition comprising the same therapeutic agent at the same concentration but lacking a nanoemulsion, and applied using the same method, measured at any suitable time point after application.
  • the longer residence time can be determined by comparing the amount of the therapeutic agent present at the site of application or administration for the nanoemulsion composition as compared to the non-nanoemulsion composition, measured at any suitable time point after application.
  • the longer residence time at the site of application can be, for example, an increase of about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 125%, about 150%, about 175%, or about 200%, as compared to the residence time of the same quaternary ammonium compound, present at the same concentration, and applied using the same method, measured at any suitable time point after application or administration.
  • the composition when the composition further comprises a therapeutic or active agent, after a single application or administration of the composition topically, transdermally, ocularly, nasally, or mucosally, the composition delivers at least about 25% more, at least about 50% more, at least about 75% more, at least about 100% more, at least about 125% more, at least about 150% more, at least about 175% more, or at least about 200% more of the quaternary ammonium compound to the epidermis, dermis, nasal tissue, mucosa, and/or squamous epithelium as compared to a composition comprising the same therapeutic agent at the same concentration but lacking a nanoemulsion, and applied using the same method, measured at any suitable time point after application or administration.
  • Additional compounds suitable for use in the disclosed methods or compositions include, but are not limited to, one or more solvents, such as an organic phosphate-based solvent, bulking agents, coloring agents, pharmaceutically acceptable carriers, a preservative, pH adjuster, buffer, chelating agent, an auxiliary surfactant, a suds suppressor, a detergent builder, etc.
  • solvents such as an organic phosphate-based solvent, bulking agents, coloring agents, pharmaceutically acceptable carriers, a preservative, pH adjuster, buffer, chelating agent, an auxiliary surfactant, a suds suppressor, a detergent builder, etc.
  • the additional compounds can be admixed into a previously formulated composition, or the additional compounds can be added to the original mixture to be further formulated.
  • one or more additional compounds are admixed into an existing disclosed composition immediately prior to its use.
  • Suitable preservatives in the disclosed composition include, but are not limited to, cetylpyridinium chloride, benzalkonium chloride, benzyl alcohol, chlorhexidine, imidazolidinyl urea, phenol, potassium sorbate, benzoic acid, bronopol, chlorocresol, paraben esters, phenoxyethanol, sorbic acid, alpha-tocophemol, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, sodium ascorbate, sodium metabisulphite, citric acid, edetic acid, semi-synthetic derivatives thereof, and combinations thereof.
  • Suitable preservatives include, but are not limited to, benzyl alcohol, chlorhexidine (bis (p- chlorophenyldiguanido) hexane), chlorphenesin (3-(-4-chloropheoxy)-propane-l,2-diol), Kathon CG (methyl and methylchloroisothiazolinone), parabens (methyl, ethyl, propyl, butyl hydrobenzoates), phenoxyethanol (2 -phenoxyethanol), sorbic acid (potassium sorbate, sorbic acid), Phenonip (phenoxyethanol, methyl, ethyl, butyl, propyl parabens), Phenoroc (phenoxyethanol 0.73%, methyl paraben 0.2%, propyl paraben 0.07%), Liquipar Oil (isopropyl, isobutyl, butylparabens), Liquipar PE (70% phenoxyethanol, 30% liquipar oil), Ni
  • Suitable pH adjusters include, but are not limited to, diethyanolamine, lactic acid, monoethanolamine, triethylanolamine, sodium hydroxide, sodium phosphate, semi-synthetic derivatives thereof, and combinations thereof.
  • Suitable buffers include pharmaceutically acceptable buffering agents. Examples of buffering agents are disclosed in U.S. Patent Publication No. 2010/0226983
  • the disclosed composition can comprise a chelating agent.
  • the chelating agent is present in an amount of about 0.0005% to about 1%.
  • chelating agents include, but are not limited to, ethylenediamine, ethylenediaminetetraacetic acid (EDTA), ethylene glycol -bis(P-ami noethyl ether)-N,N,N',N'- tetraacetic acid (EGTA), phytic acid, polyphosphoric acid, citric acid, gluconic acid, acetic acid, lactic acid, dimercaprol, or any combination thereof.
  • the chelating agent is ethylenediaminetetraacetic acid.
  • Suitable auxiliary surfactants include compounds that enhance the properties of a nanoemulsion composition.
  • the choice of auxiliary surfactant depends on the desire of the user with regard to the intended purpose of the composition and the commercial availability of the surfactant.
  • the auxiliary surfactant is an organic, water-soluble surfactant.
  • Suitable suds suppressors are low-foaming co- surfactants that prevents excessive sudsing during employment of the compositions on hard surfaces. Suds suppressors are also useful in formulations for no-rinse application of the composition. Concentrations of about 0.5 vol % to about 5 vol % are generally effective. Selection of a suds suppressor depends on its ability to formulate in a nanoemulsion composition and the residue as well as the cleaning profile of the composition. The suds suppressor should be chemically compatible with the components in a nanoemulsion composition and functional at the pH of a given composition. In one embodiment the suds suppressor or composition containing a suds suppressor does not leave a visible residue on surfaces on which a composition is applied.
  • Low-foaming co-surfactants can be used as a suds suppressor to mediate the suds profile in a nanoemulsion composition.
  • suitable suds suppressors include block copolymers, alkylated primary and secondary alcohols, and silicone-based materials.
  • Exemplary block co-polymers include, e.g., Pluronic ® and Tetronic ® (BASF Company).
  • Alkylated alcohols include those which are ethoxylated and propoxylated, such as, tergitol (Union Carbide) or poly- tergent ® (Olin Corp.).
  • Silicone-based materials include DSE (Dow Corning).
  • Suitable detergent builders include compounds that sequester calcium and magnesium ions that might otherwise bind with and render less effective the auxiliary surfactants or co surfactants.
  • Detergent builders are particularly useful when auxiliary surfactants are used, and when the compositions are diluted prior to use with hard tap water, especially water having a hardness of, above about 12 grains/gallon.
  • the disclosed methods and compositions can comprise one or more emulsifying agents to aid in the formation of emulsions.
  • Emulsifying agents include compounds that aggregate at the oil/water interface to form a kind of continuous membrane that prevents direct contact between two adjacent droplets.
  • Certain embodiments of the present disclosure feature nanoemulsion compositions that may readily be diluted with water or another aqueous phase to a desired concentration without impairing their desired properties.
  • a composition for topical, transdermal, mucosal, ocular, or nasal application or administration.
  • the composition comprises an oil-in-water nanoemulsion, the nanoemulsion comprising: (a) an aqueous phase; (b) at least one oil; (c) at least one quaternary ammonium compound; and (d) at least one nonionic surfactant; wherein the droplets of the nanoemulsion have a mean droplet size of less than about 1 micron; and wherein (i) the nanoemulsion is diluted resulting in a formulation of about 0.5% to about 60% nanoemulsion; (ii) the viscosity of the nanoemulsion is less than about 1000 cp; and (iii) the nanoemulsion enhances delivery of the quaternary ammonium compound into tissue by at least about 25% as compared to a solution with the same concentration of quaternary ammonium compound but lacking a nanoemulsion and
  • the nanoemulsion compositions described herein have a viscosity of less than about 1000 cP. In some embodiments, the nanoemulsion compositions described herein have a viscosity of less than about 900, less than about 800, less than about 700, less than about 600, less than about 500, less than about 400, less than about 300, less than about 275, less than about 250, less than about 225, less than about 200, less than about 100, less than about 75, less than about 50, less than about 25, less than about 20, less than about 10, less than about 9, less than about 8, less than about 7, less than about 6, less than about 5, less than about 4, less than about 3, less than about 2, or less than about 1.5 cP.
  • the viscosity is greater than 0
  • the viscosity is from about 1 cP to about 1000 cP; or from about 1.2 cP to about 275 cP.
  • nanoemulsions described herein enhance delivery of the quaternary ammonium compound (and/or additional active/therapeutic agent) into tissue by at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 100%, as compared to a solution with the same concentration of quaternary ammonium compound but lacking a nanoemulsion and as compared to a nanoemulsion with a viscosity greater than the referenced viscosity (e.g., greater than about 1000, greater than about 900, greater than about 800, . . . greater than about 300, greater than about 275 cP . . . , or greater than any other viscosity amount described herein).
  • a composition for topical, transdermal, mucosal, nasal or ocular application or administration, the composition comprising an oil-in-water nanoemulsion, the nanoemulsion comprising: (a) an aqueous phase; (b) at least one oil; (c) at least one quaternary ammonium compound; and (d) at least one nonionic surfactant; wherein (i) the droplets of the nanoemulsion have a mean droplet size of less than about 1 micron; (ii) the nanoemulsion is diluted resulting in a formulation of about 0.5% to about 60% nanoemulsion; (iii) the zeta potential of the nanoemulsion is greater than about 20mV; and (iv) the nanoemulsion enhances delivery of the quaternary ammonium compound (and/or additional active/therapeutic agent) into tissue by at least about 25% as compared to a solution with the same concentration of quaternary am
  • Zeta potential is a scientific term for electrokinetic potential in colloidal dispersions. The usual units are volts (V) or millivolts (mV). From a theoretical viewpoint, the zeta potential is the electric potential in the interfacial double layer (DL) at the location of the slipping plane relative to a point in the bulk fluid away from the interface. In other words, zeta potential is the potential difference between the dispersion medium and the stationary layer of fluid attached to the dispersed particle.
  • the nanoemulsion has a zeta potential from about 20 mV to about 40 mV; from about 40 mV to about 60 mV; from about 60 mV to about 80 mV; or from about 80 mV to about 100 mV.
  • the nanoemulsion has a zeta potential of greater than or equal to about 20m V, about 25m V, about 30mV, about 35mV, about 40m V, about 45m V, about 50mV, about 55mV, about 60mV, about 65m V, about 70mV, about 75m V, about 80m V, about 85mV, about 90m V, about 95mV, or greater than or equal to about lOOmV.
  • nanoemulsions described herein enhance delivery of the quaternary ammonium compound (and/or additional active/therapeutic agent) into tissue by at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 100%, as compared to a solution with the same concentration of quaternary ammonium compound but lacking a nanoemulsion and as compared to a nanoemulsion with a zeta potential less than the referenced zeta potential (e.g., less than 20m V, less than about 30mV, or less than any other zeta potential amount described herein for the described nanoemulsions).
  • a composition for topical, transdermal, mucosal, ocular, or nasal application or administration, the composition comprising an oil-in-water nanoemulsion, the nanoemulsion comprising: (a) an aqueous phase; (b) at least one oil; (c) at least one quaternary ammonium compound; and (d) at least one nonionic surfactant; wherein (i) the droplets of the nanoemulsion have a mean droplet size of less than about 1 micron; (ii) the nanoemulsion is diluted resulting in a formulation of about 0.5% to about 60% nanoemulsion; (iii) at least about 33% of the quaternary ammonium compound is entrapped in the oil phase of the nanoemulsion and at least about 0.2% of the weight of the oil phase of the nanoemulsion is attributed to entrapment of the quaternary ammonium compound; and (iv) the nanoemulmulsion
  • (a) at least about 33% of the quaternary ammonium compound is entrapped in the oil phase of the nanoemulsion; (b) at least about 0.2% of the weight of the oil phase of the nanoemulsion is attributed to the quaternary ammonium compound; or (c) the composition satisfies both (a) and (b).
  • any combination of the percentage of the quaternary ammonium compound entrapped in the oil phase of the nanoemulsion described herein can be combined with any percentage of the weight of the oil phase of the nanoemulsion attributed to entrapment of the quaternary ammonium compound described herein (e.g., at least about 0.2% up to about 25%).
  • (a) at least about 90 % of the quaternary ammonium compound is entrapped in the oil phase of the nanoemulsion; (b) at least about 0.2% of the weight of the oil phase of the nanoemulsion is attributed to entrapment of the quaternary ammonium compound; or (c) the composition satisfies both (a) and (b).
  • (a) at least about 33% of the quaternary ammonium compound is entrapped in the oil phase of the nanoemulsion; (b) at least about 0.4% of the weight of the oil phase of the nanoemulsion is attributed to entrapment of the quaternary ammonium compound; or (c) the composition satisfies both (a) and (b).
  • (a) at least about 33% of the quaternary ammonium compound is entrapped in the oil phase of the nanoemulsion; (b) at least about 0.6% of the weight of the oil phase of the nanoemulsion is attributed to entrapment of the quaternary ammonium compound; or (c) the composition satisfies both (a) and (b).
  • (a) at least about 33% of the quaternary ammonium compound is entrapped in the oil phase of the nanoemulsion; (b) at least about 0.8% of the weight of the oil phase of the nanoemulsion is attributed to entrapment of the quaternary ammonium compound; or (c) the composition satisfies both (a) and (b).
  • (a) at least about 33% of the quaternary ammonium compound is entrapped in the oil phase of the nanoemulsion; (b) at least about 1.0% of the weight of the oil phase of the nanoemulsion is attributed to entrapment of the quaternary ammonium compound; or (c) the composition satisfies both (a) and (b).
  • nanoemulsions described herein enhance delivery of the quaternary ammonium compound (and/or additional active/therapeutic agent) into tissue by at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 100%, as compared to a solution with the same concentration of quaternary ammonium compound but lacking a nanoemulsion and as compared to a nanoemulsion with a less than about 0.20% of the weight of the oil phase of the nanoemulsion attributed to entrapment of the quaternary ammonium compound.
  • the nanoemulsion compositions described herein have droplets having an average or mean particle size diameter of about 250 nm to about 1000 nm. In some embodiments, the droplets have an average or mean particle size diameter of about 250 nm to about 600 nm. In some embodiments, the droplets have an average or mean particle size diameter of about 300 nm to about 600 nm.
  • the droplets have an average or mean particle size diameter of about 150 nm or less, about 200 nm or less, about 250 nm or less, about 260 nm or less, about 270 nm or less, about 280 nm or less, about 290 nm or less, about 300 nm or less, about 310 nm or less, about 320 nm or less, about 330 nm or less, about 340 nm or less, about 350 nm or less, about 360 nm or less, about 370 nm or less, about 380 nm or less, about 390 nm or less, about 400 nm or less, about 410 nm or less, about 420 nm or less, about 430 nm or less, about 440 nm or less, about 450 nm or less, about 460 nm or less, about 470 nm or less, about 480 nm or less, about 490 nm or less, about 500
  • the mean droplet size of the nanoemulsion does not change by more than about 10% after centrifuging the nanoemulsion at a speed of about 200,000 rpm for about one hour. In other embodiments, the mean droplet size of the nanoemulsion does not change by more than about 9%, more than about 8%, more than about 7%, more than about 6%, more than about 5%, more than about 4%, more than about 3%, more than about 2%, more than about 1%, more than about 0.9%, more than about 0.8%, more than about 0.7%, more than about 0.6%, more than about 0.5%, more than about 0.4%, more than about 0.3%, or more than about 0.2%, after centrifuging the nanoemulsion at a speed of about 200,000 rpm for about one hour.
  • nanoemulsions described herein enhance delivery of the quaternary ammonium compound (and/or additional active/therapeutic agent) into tissue by at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 100%, as compared to a solution with the same concentration of quaternary ammonium compound but lacking a nanoemulsion and as compared to a nanoemulsion with a change in mean droplet size, following centrifuging the nanoemulsion at a speed of about 200,000 rpm for about one hour, of greater than about 10%.
  • the nanoemulsion compositions described herein are stable. In certain embodiments, the nanoemulsion compositions herein demonstrate stability even under storage conditions at high temperatures (e.g., about 50 °C). In some embodiments, the nanoemulsion compositions described herein are thermostable. In some embodiments, the compositions are stable for at least about 1 month, at least about 2, at least about 3, at least about 4, at least about 5, at least about 6, at least about 12, at least about 24, at least about 30, at least about 36, at least about 42, at least about 48, at least about 54, or at least about 60 months at about 5 °C, about 25 °C, about 40 °C, and/or about 50 °C.
  • the compositions are stable for at least about 3 months at about 5 °C, about 25 °C, about 40 °C, and/or about 50 °C. In some embodiments, the compositions are stable for at least about 60 months at 5 °C. In other embodiments the compositions are stable for at least about 12 months at 50 °C.
  • the nanoemulsion compositions of the invention are highly thermostable, the nanoemulsion compositions can be autoclaved without losing the structural or chemical integrity of the compositions. This is desirable as sterile formulations may be preferable for some disease indications and/or patient populations.
  • stability of a nanoemulsion according to the invention is measured by a lack of a substantial increase in average particle size over time and/or upon exposure to elevated temperatures.
  • a “lack of a substantial increase in average particle size” of a nanoemulsion can mean a particle size growth of less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, or less than about 3%.
  • the period of time over which stability is measured can be any suitable period of time, such as about 1 month, at least about 2, at least about 3, at least about 4, at least about 5, at least about 6, at least about 12, at least about 24, at least about 30, at least about 36, at least about 42, at least about 48, at least about 54, or at least about 60 months.
  • stability is measured by the ability of the composition upon exposure to elevated temperatures, and/or prolonged storage, to exhibit minimal particle aggregation formation and/or retain at an at least 80% label claim of an active agent and/or of the quaternary ammonium compound present in the nanoemulsion.
  • Time points for measurement can be as described above.
  • Other label claim thresholds can be about 85%, about 90%, or about 95% (see e.g. the methodology of Example 8).
  • the nanoemulsion compositions described herein have antiviral activity.
  • the composition is non-toxic in human and animals.
  • the composition kills at least about 99.9% of microorganisms (i.e., coronaviruses) following a 60 second exposure using the ASTM E2315-16 Standard Guide for Assessment of Antimicrobial Activity Using a Time-Kill Procedure.
  • the microorganism killed by the nanoemulsion can be a virus, bacteria, fungus or yeast.
  • the bacteria can be a gram negative or gram positive bacteria.
  • the microorganism are selected from a coronavirus selected from the group consisting of an Alphacoronavirus; a Colacovirus such as Bat coronavirus CDPHE15; a Decacovirus such as Bat coronavirus HKU10 or Rhinolophus ferrumequinum alphacoronavirus HuB-2013; a Duvinacovirus such as Human coronavirus 229E; a Luchacovirus such as Lucheng Rn rat coronavirus; a Minacovirus such as a Ferret coronavirus or Mink coronavirus 1; a Minunacovirus such as Miniopterus bat coronavirus 1 or Miniopterus bat coronavirus HKU8; a Myotacovirus such as Myotis ricketti alphacoronavirus Sax-2011; a nyctacovirus such as Nyctalus velutinus alphacoronavirus SC-2013; a Pedacovirus such
  • Human coronavirus HKU 1 or Murine coronavirus a Hibecovirus such as Bat Hp- betacoronavirus Zhejiang2013; a Merbecovirus such as Hedgehog coronavirus 1, Middle East respiratory syndrome-related coronavirus (MERS-CoV), Pipistrellus bat coronavirus HKU5 or Tylonycteris bat coronavirus HKU4; a Nobecovirus such as Rousettus bat coronavirus GCCDC1 or Rousettus bat coronavirus HKU9, a Sarbecovirus such as a Severe acute respiratory syndrome-related coronavirus, Severe acute respiratory syndrome coronavirus (SARS-CoV) or Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, COVID-19); a Deltacoronavirus; an Andecovirus such as Wigeon coronavirus HKU20; a Buldecovims such as Bulbul coronavirus HKU11, Porcine coronavirus H
  • the composition delivers at least 25% more of the quaternary ammonium compound to the epidermis, and/or at least 25% more of the quaternary ammonium compound to the dermis, and/or at least 25% more of the quaternary ammonium compound to the mucosa, and/or at least 25% more of the quaternary ammonium compound to the squamous epithelium as compared to a composition comprising the same quaternary ammonium compound at the same concentration but lacking a nanoemulsion, measured at any suitable time point after application, such as 24 hours after application.
  • the composition after a single application of the composition, has a longer residence time at the site of application as compared to a composition comprising the same quaternary ammonium compound at the same concentration but lacking a nanoemulsion, wherein the longer residence time is determined by comparing the amount of the quaternary ammonium compound present at the site of application for the nanoemulsion composition as compared to the non-nanoemulsion composition.
  • the composition delivers at least about 1.25x, at least about 1.5x, at least about 1.75x, at least about 2x, at least about 2.25x, at least about 2.5x, at least about 2.75x, at least about 3x, at least about 3.25x, at least about 3.5x, at least about 3.75x, at least about 4x, at least about 5x, at least about 6x, at least about 7x, at least about 8x, at least about 9x, or at least about lOx more of the quaternary ammonium compound to the epidermis, dermis, mucosa, and/or squamous epithelium as compared to a composition comprising the same quaternary ammonium compound at the same concentration but lacking a nanoemulsion.
  • the composition delivers at least about 25%, at least about 50%, at least about 100%, at least about 125%, at least about 150%, at least about 175%, at least about 200%, at least about 225%, at least about 250%, at least about 275%, at least about 300%, at least about 325%, at least about 350%, at least about 375%, at least about 400%, at least about 425%, at least about 450%, at least about 475%, or at least about 500% more of the quaternary ammonium compound to the epidermis, dermis, mucosa, and/or squamous epithelium as compared to a composition comprising the same quaternary ammonium compound at the same concentration but lacking a nanoemulsion.
  • the composition delivers from about 25% to about 500% more of the quaternary ammonium compound to the epidermis, dermis, mucosa, and/or squamous epithelium as compared to a composition comprising the same quaternary ammonium compound at the same concentration but lacking a nanoemulsion.
  • the composition when the composition is applied to skin, mucosa and/or squamous epithelium, the composition results in increased skin, mucosa and/or squamous epithelium hydration as compared to a composition comprising the same quaternary ammonium compound at the same concentration but lacking a nanoemulsion.
  • the increase in skin, mucosa and/or squamous epithelium hydration is from about 50% to about 1000%. In some embodiments, the increase in skin, mucosa and/or squamous epithelium hydration is about 50%, about 75%, about 100%, about 125%, about 150%, about 175%, about 200%, about 225%, about 250%, about 275%, about
  • nanoemulsions of the present disclosure may be formulated into pharmaceutical compositions that are administered in a therapeutically effective amount to a subject and may further comprise one or more suitable, pharmaceutically-acceptable excipients, additives, or preservatives. Suitable excipients, additives, and/or preservatives are well known in the art.
  • Suitable pharmaceutically acceptable excipients or pharmaceutically acceptable carriers may include solvents, dispersion media, coatings, isotonic and absorption delaying agents and the like, and combinations comprising one or more of the foregoing carriers as described, for instance, in Remington ’s Pharmaceutical Sciences , 15th Ed. Easton: Mack Publishing Co. pp. 1405-1412 and 1461-1487 (1975), and The National Formulary XIV 14th Ed., Washington: American Pharmaceutical Association (1975).
  • Suitable carriers include, but are not limited to, calcium carbonate, carboxymethylcellulose, cellulose, citric acid, dextrate, dextrose, ethyl alcohol, glucose, hydroxymethylcellulose, lactose, magnesium stearate, maltodextrin, mannitol, microcrystalline cellulose, oleate, polyethylene glycols, potassium diphosphate, potassium phosphate, saccharose, sodium diphosphate, sodium phosphate, sorbitol, starch, stearic acid and its salts, sucrose, talc, vegetable oils, water, and combinations comprising one or more of the foregoing carriers. Except insofar as any conventional media or agent is incompatible with the emulsions of the present invention, their use in therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions.
  • pharmaceutically acceptable carriers can take the form of a liquid, cream, foam, lotion, or gel, and may additionally comprise organic solvents, emulsifiers, gelling agents, moisturizers, stabilizers, surfactants, wetting agents, preservatives, time release agents, and minor amounts of humectants, sequestering agents, dyes, perfumes, and other components commonly used in pharmaceutical compositions for topical and mucosal administration.
  • terapéuticaally effective amount it is meant any amount of the composition that is effective in killing or inhibiting the growth of any one of the microorganisms described herein.
  • Topical administration includes administration to the skin, mucosa, and squamous epithelium, including surface of the hair follicle and pilosebaceous unit.
  • the composition enters the epidermis, dermis, mucosa, squamous epithelium, or any combination thereof.
  • the composition permeates into the epidermis and dermis via the follicular route using skin pores and hair follicles.
  • the composition diffuses through the skin, skin pores, nail, scalp, hair follicles, lateral or proximal folds, nail, hyponichium, or a combination thereof.
  • compositions for administration include, but are not limited to, ointments, creams, liquids, emulsions, lotions, gels, bioadhesive gels, aerosols, pastes, foams, or in the form of an article or carrier, such as a bandage, insert, syringe-like applicator, pessary, powder, talc or other solid, cleanser, and agents that favor penetration within the pilosebaceous gland.
  • the composition is administered in the form of a liquid, lotion, cream, ointment, salve, or spray.
  • the pharmaceutical compositions may be formulated for immediate release, sustained release, controlled release, delayed release, or any combinations thereof, into the epidermis or dermis, with no systemic absorption.
  • the formulations may comprise a penetration-enhancing agent for enhancing penetration of the nanoemulsion through the stratum corneum and into the epidermis or dermis.
  • Suitable penetration-enhancing agents include, but are not limited to, alcohols such as ethanol, triglycerides and aloe compositions.
  • the amount of the penetration-enhancing agent may comprise from about 0.5% to about 40% by weight of the formulation.
  • the pharmaceutical compositions may be applied in a single administration or in multiple administrations.
  • the pharmaceutical compositions can be applied for any suitable time period, such as lx or multiples times per day.
  • the compositions can be applied for at least once a week, at least twice a week, at least once a day, at least twice a day, multiple times daily, multiple times weekly, biweekly, at least once a month, or any combination thereof.
  • the pharmaceutical compositions are applied for a period of time of about one month, about two months, about three months, about four months, about five months, about six months, about seven months, about eight months, about nine months, about ten months, about eleven months, about one year, about 1.5 years, about 2 years, about 2.5 years, about 3 years, about 3.5 years, about 4 years, about 4.5 years, and about 5 years. Between applications, the application area may be washed to remove any residual nanoemulsion.
  • compositions described herein are formulated for mucosal delivery, for example by contacting any one of the compositions described herein to a nasal mucosal epithelium, a bronchial or pulmonary mucosal epithelium, oral mucosa, or ocular application.
  • the compositions described herein are formulated for intranasal delivery, (e.g., nasal mucosal delivery or intranasal mucosal delivery).
  • administration comprises contacting the nasal swab or wipe to the subject.
  • a wipe impregnated with a nanoemulsion can be used to sanitize a subject’s hands or any other surface that may come in contact with a microorganism, such as a coroavirus.
  • the nasal swab, or wipe dispenses a greater amount of the quaternary ammonium compound and/or incorporated active or therapeutic agent to an application site, as compared to a nasal swab or wipe impregnated or saturated with or incorporating a composition comprising the same quaternary ammonium compound and/or incorporated active or therapeutic agent at the same concentration but lacking a nanoemulsion.
  • the nasal swab or wipe dispenses about 20% to about 100% more of the quaternary ammonium compound and/or incorporated active or therapeutic agent to an application site, as compared to a nasal swab or wipe impregnated or saturated with or incorporating a composition comprising the same quaternary ammonium compound and/or incorporated active or therapeutic agent at the same concentration but lacking a nanoemulsion.
  • the nasal swab or wipe dispenses about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% more of the quaternary ammonium compound and/or incorporated active or therapeutic agent to an application site, as compared to a nasal swab or wipe impregnated or saturated with or incorporating a composition comprising the same quaternary ammonium compound and/or incorporated active or therapeutic agent at the same concentration but lacking a nanoemulsion.
  • Example 10 As detailed in Example 10, a comparison of a wipe saturated with a non-nanoemulsion formulation and compared to a wipe saturated with a nanoemulsion formulation revealed that the nanoemulsion-saturated wipe released much more of the component active agent (e.g., the cationic active agent). It is theorized that the active agent in the non-nanoemulsion formulation binds to the fibers or compounds in the wipe, preventing a significant portion of the active agent from being deposited on the surface or skin where the wipe is applied. This lack of active agent deposition is undesirable, as the result is a reduced effectiveness - e.g., a reduced effectiveness in antimicrobial activity when the wipe is used for disinfection.
  • the component active agent e.g., the cationic active agent
  • a nasal swab, dropper, or spray for use with any nanoemulsion composition described herein.
  • the nasal swab, dropper, or spray can be impregnated or saturated with or incorporating the any nanoemulsion composition described herein, or the nasal swab, dropper, or spray can be packaged in a kit with a container comprising a nanoemulsion composition described herein, with the swab being exposed to the nanoemulsion prior to use.
  • Such swabs are useful to prevent and/or minimize infections in hospital settings.
  • a nasal spray comprising a nanoemulsion according to the invention can also be used to treat and/or prevent viral infections originating in the nasal cavities.
  • both the nasal swab, dropper, and spray are hydrating, as hydration is a feature of the nanoemulsions described herein.
  • the swab and spray will hydrate the nasal mucosa, as well as be antiviral.
  • the methods of the invention are useful in preventing or reducing the risk of infection in a subject caused by exposure to a coronavirus, the method comprising administering to the nasal vestibule or passages, ocular region, or mouth mucosa of the subject, either before or after the exposure, a composition comprising a nanoemulsion as disclosed herein.
  • the composition, wipe, and/or swab permeates into the epidermis, dermis, mucosa, and/or squamous epithelium via the follicular route using skin pores and hair follicles.
  • the composition, wipe, and/or swab diffuses through the skin, skin pores, nail, scalp, hair follicles, lateral or proximal folds, nail, hyponichium, or a combination thereof.
  • nanoemulsions, wipes and swabs described herein are beneficial for use of the compositions, wipes and/or swabs. This is because the mechanism of action in killing the viruses does not result in drug-resistant viruses.
  • nanoemulsions lyse viral pathogens such as coronaviruses upon contact, thereby overcoming existing resistance mechanisms.
  • DR drug-resistant
  • the nanoemulsions of the invention can be formed using classic emulsion forming techniques. See e.g. , U.S. 2004/0043041.
  • the oil is mixed with the aqueous phase under relatively high shear forces (e.g., using high hydraulic and mechanical forces) to obtain a nanoemulsion comprising oil droplets having an average diameter of less than about 1000 nm.
  • relatively high shear forces e.g., using high hydraulic and mechanical forces
  • Some embodiments of the invention employ a nanoemulsion having an oil phase comprising an alcohol such as ethanol.
  • the oil and aqueous phases can be blended using any apparatus capable of producing shear forces sufficient to form an emulsion, such as French Presses or high shear mixers (e.g., FDA approved high shear mixers are available, for example, from Admix, Inc., Manchester, N.H.). Methods of producing such emulsions are described in U.S. Pat. Nos. 5,103,497 and 4,895,452, herein incorporated by reference in their entireties.
  • the nanoemulsions used in the methods of the invention comprise droplets of an oily discontinuous phase dispersed in an aqueous continuous phase, such as water or PBS.
  • the nanoemulsions of the invention are stable, and do not deteriorate even after long storage periods.
  • Certain nanoemulsions of the invention are non-toxic and safe when swallowed, inhaled, or contacted to the skin of a subject.
  • compositions of the invention can be produced in large quantities and are stable for many months at a broad range of temperatures.
  • the nanoemulsion can have textures ranging from that of a semi-solid cream to that of a thin lotion, to that of a liquid and can be applied topically, transdermally, mucosally, ocularly, or nasally by any pharmaceutically acceptable method as stated above, e.g., by hand, or nasal drops/spray, or via any other pharmaceutically acceptable method.
  • the present invention contemplates that many variations of the described nanoemulsions will be useful in the methods of the present invention.
  • three criteria are analyzed. Using the methods and standards described herein, candidate emulsions can be easily tested to determine if they are suitable. First, the desired ingredients are prepared using the methods described herein, to determine if a nanoemulsion can be formed. If a nanoemulsion cannot be formed, the candidate is rejected. Second, the candidate nanoemulsion should form a stable emulsion. A nanoemulsion is stable if it remains in emulsion form for a sufficient period to allow its intended use.
  • nanoemulsions that are to be stored, shipped, etc.
  • Typical nanoemulsions that are relatively unstable, will lose their form within a day.
  • the candidate nanoemulsion should have efficacy for its intended use.
  • the nanoemulsion of the invention can be provided in many different types of containers and delivery systems.
  • the nanoemulsions can be delivered (e.g., to a subject or customers) in any suitable container. Suitable containers can be used that provide one or more single use or multi-use dosages of the nanoemulsion for the desired application.
  • the nanoemulsions are provided in a suspension or liquid form.
  • Such nanoemulsions can be delivered in any suitable container including spray bottles and any suitable pressurized spray device. Such spray bottles may be suitable for example for delivering the nanoemulsions intranasally or via inhalation.
  • administering can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell being treated, the disease being treated and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents are known in the art.
  • buffer or “buffering agents” refer to materials which when added to a solution, cause the solution to resist changes in pH.
  • ammonium compound refers to a compound containing an ammonium moiety.
  • the ammonium moiety may include four bonds to a positively charged nitrogen atom.
  • compositions and methods include the recited elements, but not excluding others.
  • Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the composition or method.
  • Consisting of shall mean excluding more than trace elements of other ingredients for claimed compositions and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this disclosure. Accordingly, it is intended that the methods and compositions can include additional steps and components (comprising) or alternatively including steps and compositions of no significance (consisting essentially of) or alternatively, intending only the stated method steps or compositions (consisting of).
  • chelator or “chelating agent” refer to any materials having more than one atom with a lone pair of electrons that are available to bond to a metal ion.
  • nasal(ly) refers to application of the compositions of the present disclosure to the surface of the skin and mucosal cells and tissues of the nasal passages, e.g., nasal mucosa, sinus cavity, nasal turbinates, or other tissues and cells which line the nasal passages.
  • microorganism refers to without limitation, bacteria, viruses, bacterial spores, molds, fungi, and the like. Also included are biological microorganisms that are capable of producing an undesirable effect upon a host animal, and includes, for example, without limitation, bacteria, viruses, bacterial spores, molds, fungi, and the like. This includes all such biological microorganisms, regardless of their origin or of their method of production
  • nanoemulsion includes small oil-in-water dispersions or droplets, as well as other lipid structures which can form as a result of hydrophobic forces which drive apolar residues (i.e., long hydrocarbon chains) away from water and drive polar head groups toward water, when a water immiscible oily phase is mixed with an aqueous phase.
  • lipid structures include, but are not limited to, unilamellar, paucilamellar, and multilamellar lipid vesicles, micelles, and lamellar phases.
  • present disclosure contemplates that one skilled in the art will appreciate this distinction when necessary for understanding the specific embodiments herein disclosed.
  • compositions that do not substantially produce adverse allergic or adverse immunological reactions when administered to a host (e.g., an animal or a human).
  • a host e.g., an animal or a human
  • Such formulations include any pharmaceutically acceptable dosage form.
  • pharmaceutically acceptable dosage forms include, but are not limited to, dips, sprays, seed dressings, stem injections, lyophilized dosage forms, sprays, and mists.
  • “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, wetting agents (e.g., sodium lauryl sulfate), isotonic and absorption delaying agents, disintegrants (e.g., potato starch or sodium starch glycolate), and the like.
  • topical(ly) refers to application of the compositions of the present disclosure to the surface of the skin, mucosal, and squamous epithelium cells and tissues (e.g., buccal, lingual, sublingual, masticatory, respiratory or nasal mucosa, nasal turbinates and other tissues and cells which line hollow organs or body cavities).
  • topical(ly) is in reference to application to the surface of the skin.
  • subject refers to any subject, patient, or individual, and the terms are used interchangeably herein.
  • the terms “subject,” “patient,” and “individual” includes mammals, and, in particular humans.
  • the term “subject,” “patient,” or “individual” intends any subject, patient, or individual having or at risk for a specified symptom or disorder.
  • stable when referring to a “stable nanoemulsion” means that the nanoemulsion retains its structure as an emulsion.
  • a desired nanoemulsion structure for example, may be characterized by a desired size range, macroscopic observations of emulsion science (is there one or more layers visible, is there visible precipitate), pH, and a stable concentration of one or more the components.
  • surfactant refers to any molecule having both a polar head group, which energetically prefers solvation by water, and a hydrophobic tail which is not well solvated by water.
  • cationic surfactant refers to a surfactant with a cationic head group.
  • the phrase “therapeutically effective” or “effective” in context of a “dose” or “amount” means a dose or amount that provides the specific pharmacological effect for which the compound or compounds are being administered. It is emphasized that a therapeutically effective amount will not always be effective in achieving the intended effect in a given subject, even though such dose is deemed to be a therapeutically effective amount by those of skill in the art. For convenience only, exemplary dosages are provided herein. Those skilled in the art can adjust such amounts in accordance with the methods disclosed herein to treat a specific subject suffering from a specified symptom or disorder. The therapeutically effective amount may vary based on the route of administration and dosage form.
  • treatment includes reducing, ameliorating, or eliminating (i) one or more specified symptoms and/or (ii) one or more symptoms or effects of a specified disorder.
  • prevention includes reducing, ameliorating, or eliminating the risk of developing (i) one or more specified symptoms and/or (ii) one or more symptoms or effects of a specified disorder.
  • the antiviral activity of the nanoemulsion formulations described were assessed by inoculating the test samples with a suspension of RSV viral particles at a final concentration of 1-3X10 6 PFU/mL. At a predetermined exposure time an aliquot was removed and naturized by diluting into EMEM media containing 2% FBS. Residual concentration of active virus particles in treated sample was determined quantitively using a qualified plaque assay described in ATP- 12-213.01 -Plaque Assay of Respiratory Syncytial Virus. Briefly, serially diluted sample were plated on to Vero cells grown overnight at 80-9-% confluency.
  • test formulation was inoculated with a suspension of a test viral particle or organism.
  • an aliquot was removed, neutralized in BPB+ and plated onto TSA agar to be quantitatively assayed for surviving test viral particle or organisms.
  • the plates were incubated for 24 hours and the survivors were enumerated. Plate counts were converted into log 10 format and compared to an initial starting population to determine log reduction.
  • *- a greater than symbol (>) indicates that 100% of the bacteria sample was killed.
  • BZK benzalkonium chloride
  • Nanoemulsions comprising 0.13% BZK were topically applied to dermatomed cadaver human skin in a Franz diffusion cell chamber and compared against each other and against a marketed non-nanoemulsion product comprising the same concentration of BZK, 0.13% (Purell ® Foam). Permeation was measured by HPLC in the epidermis and dermis 24 hours after a single topical dose.
  • the in vitro human cadaver skin model has proven to be a valuable tool for the study of percutaneous absorption of topically applied compounds.
  • the model uses human cadaver skin mounted in specially designed diffusion chambers that allow the skin to be maintained at a temperature and humidity that match typical in vivo conditions.
  • a finite dose of formulation is applied to the epidermal layer, e.g., the outer surface of the skin, and compound absorption is measured by monitoring the compound’s rate of appearance in the receptor solution bathing the dermal surface of the skin.
  • Data defining total absorption, rate of absorption, as well as skin content can be accurately determined in this model.
  • the method has historic precedent for accurately predicting in vivo percutaneous absorption kinetics.
  • Franz, TJ “Percutaneous absorption: on the relevance of in vitro data,” J. Invest. Dermatol ., 64 190-195 (1975).
  • Cryopreserved, dermatomed human cadaver abdominal skin from a 67-year-old Caucasian female donor was used in permeation studies and obtained from Science Care (Phoenix, A Z) organ donor bank.
  • Cadaver skin was stored in aluminum foil pouches at -70°C until use. At the time of use, the skin was thawed by placing the sealed pouch in 37 °C water for approximately five minutes. Thawed skin was removed from the pouch and cut into circular discs (30 mm diameter) to fit between the donor and receiver sides of the permeation chambers.
  • Percutaneous absorption was measured using the in-vitro cadaver skin finite dose technique.
  • Franz et ak “The finite dose technique as a valid in vitro model for the study of percutaneous absorption in man,” In Skin: Drug Application and Evaluation of Environmental Hazards, Current Problems in Dermatology , vol. 7, G, edited by Simon et ak, pp 58-68 (Basel, Switzerland, S. Karger, 1978).
  • the receptor compartment was filled with 7.0 mL of distilled water, comprising 10% (v/v) ethanol in water, and was placed in the donor compartment and left open to ambient laboratory conditions.
  • the receptor compartment spout was covered with a Teflon screw cap to minimize evaporation of the receptor solution.
  • the skin was equilibrated for a period of 30 minutes before applying a 113 pL dose (over a dosing area of 1.13 cm 2 ) of the test formulations onto the epidermal surface of the donor chamber of the diffusion cells using a positive displacement pipette.
  • the exposed dosing epidermal surface area was 1.13 cm 2 .
  • Twenty-four hours after the application of the first dose the surface of the skin was rinsed with 1 ml of 70% ethanol/water solution and then cleaned with a 70% ethanol-soaked cotton swab, four times. Following alcohol swabbing, the donor cap was removed, and the skin was removed from the apparatus.
  • the epidermis was removed from the dermis via a scraping method and placed in a tarred scintillation vial. A punch biopsy was taken through the dermis and placed in a tarred scintillation vial. Weights of dermis and epidermis were recorded.
  • the epidermal and dermal tissues were extracted with a 200 proof ethanol solution, sonicated for 30 minutes, filtered through a 25 mm, 0.45 pm PTFE membrane syringe filter into HPLC vials and assayed using HPLC. The excess skin portion was placed in scintillation vial with the surface swabs.
  • One mL of the receptor solution was also sampled at 24 hours from the receptor of each cell and filtered through a 0.45 pm PTFE (25 mm) membrane syringe filter. The filtrates were collected in HPLC snap cap vials.
  • the amount of BZK that permeated into the epidermis, dermis, and the receptor compartment (at 24 hours after first dose) was determined by HPLC.
  • the concentration of BZK in the dosing area was determined with respect to a standard preparation.
  • the level of BZK each skin area is represented as the amount per wet tissue weight (ng/grams) ⁇ the standard deviation.
  • the number of replicas used in the calculation was 5 for each formulation.
  • the amount of BZK delivered into the human abdominal skin epidermal tissue was the highest with NE-2 (Surfactant Blend Ratio 1 :9), with 6642 ng BZK/gram tissue, as compared to 953 ng BZK/gram tissue for the Purell ® Foam with the same percentage of 0.13% BZK (0.13%) in each formulation, e.g., equivalent to a 597% increase in permeation with the nanoemulsion formulation having a 1 :9 surfactant blend ratio.
  • the nanoemulsion having a 1 :5 surfactant blend ratio showed an about 300% increase in permeation as compared to the non nanoemulsion formulation (Purell ® Foam).
  • Figure 44A graphically shows the epidermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours), and Figure 44B shows the dermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours).
  • nanoemulsions showed dramatic and significantly greater permeation (amount of BZK (ng)/ tissue weight (g)) as compared to a non nanoemulsion formulation having the same quantity of BZK.
  • amount of BZK (ng)/ tissue weight (g) As clearly depicted in Figs. 45A and 45B, nanoemulsions showed dramatic and significantly greater permeation (amount of BZK (ng)/ tissue weight (g)) as compared to a non nanoemulsion formulation having the same quantity of BZK.
  • Receptor is total amount of BZK (pg): mean of replicates ⁇ SD).
  • FIG. 45 A shows that when an aqueous solution is applied topically, no GFP is delivered into the skin (left panel).
  • Fig. 45B right panel is the NE+GFP and shows the distribution of GFP in the epidermis and dermis.
  • Figure 46 shows nasal nanoemulsion antiseptic formulations (NE1, NE2, and NE3, having different surfactant ratios) significantly enhanced survival in mice that were challenged with a lethal dose of influenza virus 90 minutes after application.
  • Pretreatment of mouse nares with three nanoemulsion formulations followed by five minute exposure to aerosolized influenza A virus at a concentration of 5xl0 5 pfu/ml was performed to determine the ability of these compounds to protect mice against inhaled virus particles.
  • Control mice were pretreated with an intranasal application of PBS. 81.25% (13/16) of mice pretreated with PBS died, while 31.91% (15/47) of mice pretreated with nanoemulsion died.
  • the nanoemulsion test formulations comprised 0.13% BZK or 0.10% CPC, and were made using conventional homogenization techniques.
  • the compositions of the BZK or CPC formulations are listed in Tables 5, 6, and 7 as NE-1, NE-2, and NE-3 formulations, respectively.
  • the water soluble ingredients are first dissolved in water.
  • the oil is then added and the mixture is mixed using high shear homogenization and/ or microfluidization until a viscous white emulsion is formed.
  • the emulsion may be further diluted with water to yield the desired concentration of emulsion or quaternary ammonium compound.
  • Nanoemulsions used in this study are oil-in-water (o/w) emulsions with mean droplet diameters of 300-600 nm.
  • BZK or CPC resides at the interface between the oil and water phases.
  • the hydrophobic tail of the surfactant distributes in the oil core and its polar head group resides in the water phase.
  • the nanoemulsions described herein are made from surfactants approved for human consumption and common food substances and are 'Generally Recognized as Safe' (GRAS) by the FDA. These emulsions are produced by mixing a water-immiscible oil phase into an aqueous phase. The two phases (aqueous phase and oil phase) are combined and processed to yield an emulsion. The emulsion is further processed to achieve the desired particle size.
  • GRAS 'Generally Recognized as Safe'
  • BZK benzalkonium chloride
  • Nanoemulsions comprising 0.13% BZK were topically applied to dermatomed cadaver human skin in a Franz diffusion cell chamber and compared against each other and against a marketed non-nanoemulsion product comprising the same concentration of BZK, 0.13% (Purell ® Foam). Permeation was measured by HPLC in the epidermis and dermis 24 hours after a single topical dose.
  • the in vitro human cadaver skin model has proven to be a valuable tool for the study of percutaneous absorption of topically applied compounds.
  • the model uses human cadaver skin mounted in specially designed diffusion chambers that allow the skin to be maintained at a temperature and humidity that match typical in vivo conditions.
  • a finite dose of formulation is applied to the epidermal layer, e.g., the outer surface of the skin, and compound absorption is measured by monitoring the compound’s rate of appearance in the receptor solution bathing the dermal surface of the skin.
  • Data defining total absorption, rate of absorption, as well as skin content can be accurately determined in this model.
  • the method has historic precedent for accurately predicting in vivo percutaneous absorption kinetics.
  • Franz, TJ “Percutaneous absorption: on the relevance of in vitro data,” J. Invest. Dermatol ., 64 190-195 (1975).
  • Cryopreserved, dermatomed human cadaver abdominal skin from a 67-year-old Caucasian female donor was used in permeation studies and obtained from Science Care (Phoenix, AZ) organ donor bank.
  • Cadaver skin was stored in aluminum foil pouches at -70°C until use. At the time of use, the skin was thawed by placing the sealed pouch in 37 °C water for approximately five minutes. Thawed skin was removed from the pouch and cut into circular discs (30 mm diameter) to fit between the donor and receiver sides of the permeation chambers.
  • Percutaneous absorption was measured using the in-vitro cadaver skin finite dose technique.
  • Franz et ak “The finite dose technique as a valid in vitro model for the study of percutaneous absorption in man,” In Skin: Drug Application and Evaluation of Environmental Hazards, Current Problems in Dermatology , vol. 7, G, edited by Simon et ak, pp 58-68 (Basel, Switzerland, S. Karger, 1978).
  • the receptor compartment was filled with 7.0 mL of distilled water, comprising 10% (v/v) ethanol in water, and was placed in the donor compartment and left open to ambient laboratory conditions.
  • the receptor compartment spout was covered with a Teflon screw cap to minimize evaporation of the receptor solution.
  • the skin was equilibrated for a period of 30 minutes before applying a 113 pL dose (over a dosing area of 1.13 cm 2 ) of the test formulations onto the epidermal surface of the donor chamber of the diffusion cells using a positive displacement pipette.
  • the exposed dosing epidermal surface area was 1.13 cm 2 .
  • Twenty-four hours after the application of the first dose the surface of the skin was rinsed with 1 ml of 70% ethanol/water solution and then cleaned with a 70% ethanol-soaked cotton swab, four times. Following alcohol swabbing, the donor cap was removed, and the skin was removed from the apparatus.
  • the epidermis was removed from the dermis via a scraping method and placed in a tarred scintillation vial. A punch biopsy was taken through the dermis and placed in a tarred scintillation vial. Weights of dermis and epidermis were recorded.
  • the epidermal and dermal tissues were extracted with a 200 proof ethanol solution, sonicated for 30 minutes, filtered through a 25 mm, 0.45 pm PTFE membrane syringe filter into HPLC vials and assayed using HPLC. The excess skin portion was placed in scintillation vial with the surface swabs.
  • One mL of the receptor solution was also sampled at 24 hours from the receptor of each cell and filtered through a 0.45 pm PTFE (25 mm) membrane syringe filter. The filtrates were collected in HPLC snap cap vials.
  • the mobile phase composition was acetate buffer and acetonitrile (ACN) in the ratio of 48:52 in isocratic mode.
  • the method was qualified for linearity and for specificity. Experimental conditions are tabulated below in Table 9.
  • the amount of BZK that permeated into the epidermis, dermis, and the receptor compartment (at 24 hours after first dose) was determined by HPLC.
  • the concentration of BZK in the dosing area was determined with respect to a standard preparation.
  • the level of BZK each skin area is represented as the amount per wet tissue weight (ng/grams) ⁇ the standard deviation.
  • the number of replicas used in the calculation was 5 for each formulation.
  • the amount of BZK delivered into the human abdominal skin epidermal tissue was the highest with NE-2 (Surfactant Blend Ratio 1 :9), with 6642 ng BZK/gram tissue, as compared to 953 ng BZK/gram tissue for the Purell ® Foam with the same percentage of 0.13% BZK (0.13%) in each formulation, e.g., equivalent to a 597% increase in permeation with the nanoemulsion formulation having a 1 :9 surfactant blend ratio.
  • the nanoemulsion having a 1 :5 surfactant blend ratio showed an about 300% increase in permeation as compared to the non nanoemulsion formulation (Purell ® Foam).
  • Figure 1 graphically shows the epidermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours), and Figure 2 shows the dermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours).
  • nanoemulsions having surfactant ratios of 1:5 and 1:9 showed dramatic and significantly greater permeation (amount of BZK (ng)/ tissue weight (g)) as compared to a non-nanoemulsion formulation having the same quantity of BZK.
  • Example 6 Following the ex vivo skin permeation study outlined in Example 6, the following 0.13% BZK NE-1 formulations were evaluated against the Purell ® Foam using the same methodology of Example 6:
  • Figure 3 graphically shows the epidermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of the different NE-1 formulations with different surfactant blend ratios and Purell ® Foam.
  • Figure 4 shows the dermal levels of BZK (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of different NE-1 formulations with different surfactant blend ratios and Purell ® Foam.
  • Stability at extremely high temperatures e.g. 50°C; 122°F
  • robust packaging components e.g. PET plastic bottles with sprayers, not glass vials
  • NE-2 (Surfactant Blend Ratio: 1:5; 0.13% BZK) was produced at a 4 kg scale and placed on stability at 5°C, 25°C, 40°C, and 50°C (122°F).
  • Table 12 shows that NE-2 (Surfactant Blend Ratio: 1:5; 0.13% BZK) is stable for 1 month even at the most extreme storage condition of 50 °C (122 °F). This is highly unexpected. At severely high temperatures, emulsions are prone to rapid destabilization within a few hours to a couple of days. This data demonstrates that the nanoemulsion formulations having the claimed surfactant blend ratio will offer key advantages for use in extremely high temperature climates.
  • the BZK Potency was determined with RP-HPLC, as described previously (e.g. permeation section). The appearance was determined via a visual assessment of color, creaming, settling and phase separation with predetermined acceptance criteria.
  • the particle size and polydispersity index (Pdl) of the sample were measured by dynamic light scattering using photon correlation spectroscopy with a Malvern Zetasizer Nano ZS90 (Malvern Instruments, Worcestershire, UK), according to SOP#208.01 version 1: Particle Sizing (Malvern). All measurements were carried out at 25 °C after appropriate dilution with double distilled 0.22 pm filtered water.
  • Figure 6 shows skin hydration study results of NE-1 (surfactant blend ratio: 1:5; 0.13% BZK) and Purell ® Foam (0.13% BZK), with the figure clearly and unequivocally showing significant and dramatically improved hydration with nanoemulsion formulations according to the invention as compared to a non-nanoemulsion formulation comprising the same quaternary ammonium compound at the same concentration.
  • the objective of this study was to compare the NE formulations comprising BZK described herein to other products comprising the same amount of BZK but lacking a nanoemulsion.
  • Two different wipe materials were tested: spunlace washcloth and airlaid washcloth.
  • Three test formulations comprising the same amount of BZK were tested: (i) an aqueous solution of 0.13% BZK; (ii) NE-1 (surfactant blend ratio: 1 :9; 0.13% BZK); and (iii) Purell ® Foam (0.13% BZK).
  • the wipes were saturated with consistent volumes of each tested formulation and the amount of BZK dispensed was measured at the following three time points - initial, 2 hours and 5 days.
  • Figure 7 shows the percent (%) of BZK dispensed from the wipe (spunlace washcloth) with aqueous BZK (0.13%BZK), NE-1 (surfactant blend ratio: 1:9; 0.13% BZK), and Purell ® Foam (0.13% BZK) at the following time points: initial, 2 hours and 5 days.
  • the results graphically depicted in the figure show that the aqueous BZK and Purell ® Foam formulations had significantly less compound (BSK) dispensed from the wipe as compared to the nanoemulsion formulation. This result is significant, as the goal of a wipe-dispensed product is to dispense as much drug as possible. Retention of drug in a wipe is contrary to the goal of drug dispension.
  • Figure 7 shows that the nanoemulsion formulation dispensed over 95% of the BZK label claim from the wipe at each of the tested time points, with over a 110% measurement at 5 days.
  • the aqueous BZK formulation had a high BZK% label claim of 60% at the initial time point
  • the Purell ® Foam formulation had a high of an initial %BZK label claim of about 73%, also at the initial time point.
  • Figure 8 shows the % of BZK dispensed from the wipe with aqueous BZK (0.13% BZK), NE-1 (surfactant blend ratio: 1:9; 0.13% BZK), and Purell ® Foam (0.13% BZK) at the following time points: initial, 2 hours and 5 days.
  • the data shown in the figure demonstrates that the nanoemulsion formulation dispensed about 85% of the %BZK label claim at the initial and 2 hour test points, and about 95% of the %BZK label claim at 5 days.
  • the aqueous BZK formulation had a high of about a 35% of the %BZK label claim at the initial test point, with the percentage decreasing at the 2 hour and 5 day test points.
  • the Purell ® Foam formulation had a high of just over 40% of the %BZK label claim at the initial time point, with decreasing amounts at the 2 hour (35%) and 5 day (20%) time points.
  • the objective of this study was to compare the in vitro permeation of Compound A, a therapeutic compound, across a mucin layer (as a surrogate for the nasal mucous) using a commercially available intranasal product and the nanoemulsion emulsion formulations described herein.
  • Porcine stomach mucin type III (a mixture of different mucins) and HEPES (4-(2- Hydroxyethyl)piperazine-l-ethanesulfonic acid, N-(2 -Hydroxy ethyl)piperazine-N'-(2- ethanesulfonic acid) were purchased from Sigma-Aldrich (St. Louis, MO).
  • Transwell® membranes (6.5 mm diameter inserts, 3.0 pm pore size in polycarbonate membrane) were purchase from Corning Incorporated (Kennebunk ME). 24 well plates were purchased from VWR (Radnor, PA).
  • Porcine gastric mucin type III was rehydrated at 10 mg/mL in 1 mM HEPES, pH 7 at 25°C for 30 minutes.
  • Transwell® membranes were coated with 10 mg/mL mucin in 1 mM HEPES, pH 7 overnight at 37°C hanging in a lower buffer reservoir (1 mM HEPES, pH 7). Mucin coated Transwell® membranes were moved to a fresh reservoir containing 600 pL of fresh 1 mM HEPES buffer, pH 7, at 37°C.
  • Buffer contains 0.4% sodium citrate and 0.15% citric acid in purified water.
  • Figure 10 shows the results of the in vitro mucin permeation studies of Compound A with the commercially available intranasal product of Compound A (0.50% Compound A) and the NE-1 (surfactant blend ratio: 1 :9) with 0.50% and 0.25% of Compound A.
  • the permeation of Compound A was greater when present in a nanoemulsion formulation as compared to a non-nanoemulsion formulation.
  • the commercial product of Compound A having a drug concentration of 50%, showed a cumulative concentration of compound A (pg/mL) at 6 hours following application of about 325 pg/mL, in contrast to a concentration of about 730 pg/mL for the nanoemulsion having a surfactant ratio of 1 :9 and a drug concentration of 50%, an increase in drug permeation of 125%.
  • Sprague-Dawley rats were purchased from Charles River Laboratories (Wilmington, MA; Source; Stock #400) and were 6 weeks old upon arrival. Rats were housed in specific pathogen - free conditions. All procedures were approved by the University Committee on the Use and Care of Animals (UCUCA) at the University of Michigan (ULAM IV AC#: IV1060). Animals were housed in ventilated racks, 3 rats per cage. The in-life duration of the study included 50 pL intranasal administration (25 pL per nare) of each test formulation to three separate rats, timed bleeds, and euthanasia of the animals. The intranasal administration was performed under brief anesthesia.
  • test formulations included: (1) a commercial product with 0.5% Compound A (a representative therapeutic agent) or (2) nanoemulsion formulated with either 0.25% or 0.5% Compound A (NE-2 with surfactant blend ratio of 1 :2, 1 :5, 1 :9, and NE-4 with surfactant blend ratio of 1:2 and 1:5).
  • the unlabeled Compound A competes with its labeled species for the limited number of available binding sites on its specific binder, thus reducing the amount of labeled Compound A bound. Therefore, the level of radioactivity bound is inversely related to the concentration in the rat serum sample or standard.
  • Figure 11 shows the % increase in serum levels of Compound A following intranasal administration with the commercially available intranasal product of Compound A (0.50% Compound A) and the NE-2 (surfactant blend ratios: 1 :9, 1:5, and 1 :2) and NE-4 formulations (surfactant blend ratios: 1:5 and 1 :2) with 0.50% or 0.25% of Compound A.
  • the non-nanoemulsion product of Compound A had a 50 percent (%) increase in serum levels of Compound A at 24 hours vs baseline. This is in contrast to increases of up to 150% for a nanoemulsion having the same drug concentration and a surfactant ratio of 1 :5.
  • a nanoemulsion having a surfactant ratio of 1 :5 and half the quantity of drug e.g., 0.25% concentration
  • Figure 12 shows the serum levels of Compound A following one intranasal administration with the commercially available intranasal product of Compound A (0.50% Compound A) and the NE-2 and NE-4 formulations (surfactant blend ratios: 1:5 and 1 :2) with 0.50% of Compound A. All of the nanoemulsion formulations resulted in significantly greater serum levels of Compound A (pg/mL) - all greater than about 3500 pg/mL - as compared to the conventional, non-nanoemulsion formulation - about 2750 pg/mL - a difference of about 30%.
  • the nanoemulsion tested had a surfactant ratio of 1 :9 and a BZK amount of 0.13% (NE-1 from Table 5, supra).
  • the positive control was 3M Skin and Nasal Antiseptic Povidone -Iodine Solution 5% (w/w) USP REF 192401 Lot 0006461182 (Exp 2020-06-21) (St Paul, MN).
  • the negative control was PBS (IX).
  • tissue explants were placed in a 50 mL sterile conical tube and washed with 15 mL of RPMI 1640 (antibiotics-free) medium for 1 minute with gentle swirling. The skin explants were then placed stratum corneum side up on a 0.4 pm cell culture insert in a 6-well plate with 1 mL of RPMI 1640 (antibiotics-free) medium. 12 tissue explants were placed in a 50 mL sterile conical tube and washed with 15 mL RPMI 1640 (antibiotics-free) medium plus 2% human serum for 1 minute with gentle swirling.
  • the skin explants were placed stratum corneum side up on a 0.4 pm cell culture insert in a 6-well plate with 1 mL RPMI1640 (antibiotics-free) medium.
  • RPMI1640 antibiotics-free
  • 1.2 mL/well of the appropriate medium e.g. RPMI 1640 (antibiotics-free) medium +/- 2% (v/v) human serum was plaed into 6-well plate and placed in an incubator at 37°C and 7% CO2.
  • S. aureus Bacteria S. aureus was inoculated into a TSA plate and incubated overnight at 37°C and 7% CO2. A single colony of S. aureus was chosen from the TSA plate and resuspended in RPMI 1640 (antibiotics-free) medium to a concentration of approximately 5xl0 8 CFU/mL to be used as the inoculum.
  • RPMI 1640 antibiotics-free
  • Topical Application of Test Formulations to Skin Explant After S. aureus infection, 50 pL of each test formulation was applied on top of skin surface of three skin explants with a pipette. After 30 seconds, another 50 pL of the test formulation was applied for a total dosing volume of 100 pL. Incubated for 1 hour at 37°C and 7% CO2. Wash Skin Explants: 1 mL of PBS (IX) was applied in each insert to wash the tissue for 10 seconds, while swirling the plate gently to wash the tissues. 1 mL wash was removed from each insert and discarded. Incubate Skin Extracts: incubation was continued for 1 hour at 37°C and 7% CO2.
  • Figure 13 shows the epidermal levels of terbinafme (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE-1 formulation (surfactant ratio of 1 :9 with 1% terbinafme) with Lamisil AT ® (1% terbinafme).
  • Figure 14 shows the dermal levels of terbinafme (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of NE-1 formulation (surfactant ratio of 1 :9 with 1% terbinafme) with Lamisil AT ® (1% terbinafme).
  • the nanoemulsions having surfactant ratios of 1:9 showed dramatic and significantly greater permeation (amount of terbinafme (pg)/ tissue weight (g)) as compared to a non-nanoemulsion formulation having the same quantity of terbinafme.
  • mice [0331] Miconazole Delivery: The nanoemulsion tested had a surfactant ratio of 1 : 12 and a miconazole amount of 2.0% as shown in the below table. This nanoemulsion was evaluated against the Monistat® (2% miconazole) using the same methodology of Example 6:
  • Figure 15 graphically shows the epidermal levels of miconazole (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE- 1 formulation (surfactant ratio of 1 : 12 with 2% miconazole) with Monistat ® (2% miconazole).
  • Figure 16 shows the dermal levels of miconazole (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of NE-1 formulation (surfactant ratio of 1 : 12 with 2% miconazole) with Monistat ® (2% miconazole).
  • the nanoemulsion having surfactant ratio of 1:12 showed dramatic and significantly greater permeation (amount of miconazole (pg)/ tissue weight (g)) as compared to a non-nanoemulsion formulation having the same quantity of miconazole.
  • Figure 17 graphically shows the epidermal levels of salicylic acid (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of the NE- 1 formulation (surfactant ratio of 1 : 12 with 1% and 2 % salicylic acid) with Dermarest ® (3% salicylic acid).
  • the nanoemulsions having surfactant ratio of 1:12 showed dramatic and significantly greater permeation (amount of salicylic acid (pg)/ tissue weight (g)) as compared to a non-nanoemulsion formulation having the greater quantity of salicylic acid.
  • Figure 18 graphically shows the epidermal levels of hydrocortisone (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE-1 formulation (surfactant ratio of 1:9 with 1% hydrocortisone) with Cortizone-10 ® (1% hydrocortisone).
  • Figure 19 shows the dermal levels of hydrocortisone (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of NE-1 formulation (surfactant ratio of 1:9 with 1% hydrocortisone) with Cortizone-10 ® (1% hydrocortisone).
  • the nanoemulsion having a surfactant ratio of 1:9 showed dramatic and significantly greater permeation (amount of hydrocortisone (pg)/ tissue weight (g)) as compared to a non-nanoemulsion formulation having the same quantity of hydrocortisone.
  • Figure 20 graphically shows the epidermal levels of adapalene (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE- 1 formulation (surfactant ratio of 1 :9 with 0.1% adapalene) with Differin ® (0.1% adapalene).
  • Figure 21 shows the dermal levels of adapalene (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of NE-1 formulation (surfactant ratio of 1:9 with 0.1% adapalene) with Differin ® (0.1% adapalene).
  • the nanoemulsion having surfactant ratio of 1:9 showed dramatic and significantly greater permeation (amount of adapalene (pg)/ tissue weight (g)) as compared to a non-nanoemulsion formulation having the same quantity of adapalene.
  • Topical Protein Delivery The nanoemulsions tested had a surfactant ratio of 1 :6 and 1 :9 and a peanut extract protein amount of 0.1% as shown in the below table, where each of the following peanut proteins were used: Ara h2, Ara hi, Ara h3 and Ara hX. This nanoemulsion was evaluated against an aqueous formulation (0.1% peanut protein) using the same methodology of Example 6:
  • Peanut Extract Protein one of the following: Ara h2, Ara hi, Ara h3, and Ara hX
  • Figure 22 graphically shows the epidermal levels of peanut proteins Ara h2, Ara hi, Ara h3, and Ara hX (pg/g tissue) in human abdominal skin following one application (occluded dose of 100 m ⁇ /cm 2 , measured at 18 hours) of the NE-1 formulation (surfactant ratio of 1 :6 with 0.1% peanut protein) with an aqueous formulation (0.1% peanut protein).
  • Figure 23 shows the dermal levels of peanut proteins Ara h2, Ara hi, Ara h3, and Ara hX (pg/g tissue) in human abdominal skin following one application (occluded dose of 100 pl/cm 2 , measured at 18 hours) of NE-1 formulation (surfactant ratio of 1 :6), NE-2 formulation (surfactant ratio of 1 :6), and NE-3 formulation (surfactant ratio of 1 :9) using three different quaternary ammonium compounds and two different nonionic surfactants combined with 0.1% peanut protein) with aqueous formulation (0.1% peanut protein).
  • NE-1 formulation surfactant ratio of 1 :6
  • NE-2 formulation surfactant ratio of 1 :6
  • NE-3 formulation surfactant ratio of 1 :9
  • each of the nanoemulsions having surfactant ratios of 1 :6 and 1 :9 showed dramatic and significantly greater permeation (amount of peanut protein (pg)/ tissue weight (g)) as compared to a non-nanoemulsion formulation having the same quantity of peanut protein. Furthermore, by interchanging the three quaternary ammonium compounds and two nonionic surfactants in the nanoemulsion formulations tested in Figure 23 and still achieving significantly greater permeation in each case as compared to a non nanoemulsion formulation, the importance of the concentration ratio of the quaternary ammonium compound to the nonionic surfactant as opposed to the specific surfactants used in each formulation is demonstrated.
  • Topical BEC Delivery The nanoemulsion tested had a surfactant ratio of 1 :6 and a BEC amount of 0.2% as shown in the below table. This nanoemulsion was evaluated against an aqueous formulation (0.2% BEC), New-Skin® spray (0.2% BEC), and CVS Liquid Bandage (0.2% BEC) using the same methodology of Example 6:
  • Figure 24 graphically shows the epidermal levels of BEC (pg/g tissue) in human abdominal skin following one application (single dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE formulation (surfactant ratio of 1 :6 with 0.2% BEC) with an aqueous formulation (0.2% BEC), New-Skin® spray (0.2% BEC), and CVS Liquid Bandage (0.2% BEC).
  • Figure 25 graphically shows the dermal levels of BEC (pg/g tissue) in human abdominal skin following one application (single dose of 100 pl/cm 2 , measured at 24 hours) of the NE formulation (surfactant ratio of 1 :6 with 0.2% BEC) with an aqueous formulation (0.2% BEC), New-Skin® spray (0.2% BEC), and CVS Liquid Bandage (0.2% BEC).
  • Figure 26 graphically shows the epidermal levels of PCMX (pg/g tissue) in human abdominal skin following one application (single dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE formulation (surfactant ratio of 1 :6 with 3.0% PCMX) with an 70% ethanol formulation (3% PCMX).
  • Figure 27 graphically shows the dermal levels of PCMX (pg/g tissue) in human abdominal skin following one application (single dose of 100 pl/cm 2 , measured at 24 hours) of the NE formulation (surfactant ratio of 1 :6 with 3.0% PCMX) with an 70% ethanol formulation (3% PCMX).
  • the nanoemulsion having surfactant ratio of 1:6 showed dramatic and significantly greater permeation (amount of PCMX (pg)/ tissue weight (g)) as compared to a non-nanoemulsion formulation having the same quantity of PCMX.
  • Figure 28 shows the epidermal levels of chlorhexidine (pg/g tissue) in human abdominal skin following one application (dose of 100 m ⁇ /cm 2 , measured at 24 hours) of the NE-1 formulation (surfactant ratio of 1 :9 with 2% chlorhexidine) with a 70% IPA solution containing 2% chlorhexidine.
  • Figure 29 shows the dermal levels of chlorhexidine (pg/g tissue) in human abdominal skin following one application (dose of 100 pl/cm 2 , measured at 24 hours) of NE-1 formulation (surfactant ratio of 1 :9 with 2% chlorhexidine) with a 70% IPA solution containing 2% chlorhexidine.
  • the nanoemulsions having surfactant ratios of 1:9 showed dramatic and significantly greater permeation (amount of chlorhexidine (pg)/ tissue weight (g)) as compared to a non-nanoemulsion formulation having the same quantity of chlorhexidine.
  • NE samples ranging from 0.5% NE to 100% NE
  • Brookfield Viscometers Models LV and RV Brookfield Engineering Laboratories, Inc., USA
  • the viscometers and NE samples were allowed come to 22.0 ⁇ 1°C.
  • Each NE sample was placed in a BD FalconTM 50 mL Conical Centrifuge Tube wide enough to properly cover the specified spindle. The tube containing the NE sample was placed under the spindle and centered to the immersion line.
  • a LV viscometer using an UL adaptor was used for NE samples 0.5% NE to 60% NE.
  • the viscosity of each NE sample was measured at a property speed of either 100, 50 or 1 rpm.
  • the viscosity (cP) readings were recorded.
  • the 80% NE sample was measured using a LV viscometer using a LV2 spindle at a speed of 3 rpm. Due to tremendous increase in viscosity of the 100% NE sample, an RV viscosity with a F spindle at 100 rpm was used to determine the viscosity.
  • Figure 30 shows epidermal permeability results
  • Figure 31 shows dermal permeability results, for nanoemulsion formulations of various nanoemulsion concentrations
  • Figure 32 shows epidermal permeability
  • Figure 33 shows dermal permeability for nanoemulsions having various viscosities.
  • Nanoemulsions falling within the preferred viscosity range of the present disclosure shown in the shaded box have significant and dramatic increased permeability as compared to the nanoemulsion formulations outside the viscosity range of the disclosure.
  • the mean particle size (Z-AVE), polydispersity index (Pdl) and zeta potential were determined for samples by dynamic light scattering using photon correlation spectroscopy in a Malvern Zetasizer Nano ZS90 (Malvern Instruments, Worcestershire, UK).
  • the test sample of nanoemulsion diluted was to 1% final nanoemulsion concentration.
  • the test sample of nanoemulsion diluted was to 0.1% final nanoemulsion concentration. All measurements were carried out at 25°C after appropriate dilution with double distilled 0.2 pm filtered water.
  • Figure 34 shows epidermal permeability results
  • Figure 35 shows dermal permeability results, for nanoemulsion formulations within preferred zeta potential range and outside the scope of the disclosure relative to the formulation’s zeta potential.
  • Nanoemulsions of the disclosure shown in the shaded box show significant and dramatic increased permeability as compared to the nanoemulsion formulations outside the claimed zeta potential range.
  • Example 17 Centrifugation Study to Determine Quaternary Ammonium Compound Entrapment
  • the amount of BZK in the external (aqueous phase) the nanoemulsion was determined.
  • the experiment required separation of the nanoemulsion droplets from the external aqueous phase of the formulation using centrifugation while maintaining emulsion droplet structure (i.e. intact droplets in close proximity to each other) and not to cause coalescence (fusing of the droplets and then measuring the concentration of quaternary ammonium compound.
  • the nanoemulsion droplets concentrate at the top of the tube and the clear aqueous phase below the emulsion droplets. Images depicting nanoemulsion sample after centrifugation are shown in Figure 43. Image taken under normal lighting conditons (left) and corresponding negative image (right). The negative image illustrates the clarity of the aqueous phase. A portion of the aqueous phase was removed from the tube with a 26-gauge needle and syringe without distributing the top layer of the nanoemulsion droplets. The sample of the aqueous phase was than assayed for BZK using RP-HPLC.
  • the concentration of BZK in the extracted aqueous phase, the weight of the nanoemulsion in each tube and the percentage of the aqueous phase verses the oil phase was used to determine the entrapment of the quaternary ammonium compound in the oil phase of the nanoemulsion.
  • Figure 38 shows epidermal permeability results
  • Figure 39 shows dermal permeability results, for nanoemulsion formulations falling within the disclosure and outside the scope of the disclosure relative to the formulation’s entrapment of the quaternary ammonium compound in the oil phase of the nanoemulsion.
  • Nanoemulsions falling within the disclosure are shown in the shaded box and show significant and dramatic increased permeability as compared to the nanoemulsion formulations outside the range of the disclosure (e.g., 80% and 100% nanoemulsion (NE)) and a current commercial formulation (Purell ® ).
  • Nanoemulsion samples were placed under a very high centrifugal force and long duration to force the nanoemulsion droplets from the external aqueous phase to come near each other. If the interface of the nanoemulsion droplets is not strong, the droplets will coalescence (fusing of the droplets) and the mean particle size will be effected.
  • Figure 38 shows epidermal permeability results
  • Figure 39 shows dermal permeability results, for nanoemulsion formulations falling within the disclosure and formulations outside the scope of the disclosure, relative to the formulation’s stability (as measured by change in mean droplet size) following prolonged centrifugation.
  • Nanoemulsions of the present disclosure are shown in the shaded box show significant and dramatic increased permeability as compared to the nanoemulsion formulations outside the claimed range (e.g., 80% and 100% nanoemulsion (NE)) and a current commercial formulation (Purell ® ).
  • Cryopreserved, dermatomed human cadaver male thigh skin from a donor was used in permeation studies and obtained from Science Care (Tucson, A Z) tissue organ donor bank.
  • Cadaver skin was stored in aluminum foil pouches at -70° C until use. At the time of use, the skin was thawed by placing the sealed pouch in 37°C water for approximately five minutes. Thawed skin was removed from the pouch and cut into circular discs (30 mm diameter) to fit between the donor and receiver sides of the permeation chambers.
  • the receptor compartment was filled with 7.0 mL of distilled water, and was placed in the donor compartment.
  • the receptor compartment spout was covered with a Teflon screw cap to minimize evaporation of the receptor solution.
  • Correctly-sized human cadaver skin was placed onto the opening on the permeation cell. All cells were individually clamped with a clamp-support and placed in a heating bath which was maintained at 37°C by a circulating water bath on the outside of the cells.
  • the receptor compartment was maintained at 37°C with the water bath and magnetic stirring.
  • the surface temperature of the skin was appropriately 32°C as determined by an IR surface temperature probe.
  • test articles included the following: Salonpas Gel Patch with 4% Lidocaine, NDC#46581-830-06 (Hisamitsu, Japan), Salonpas Roll on Liquid with 4% Lidocaine, 10% Benzyl Alcohol, NDC#55328-901-03, (Hisamitsu, Japan), 20% NE with 0.13% BZK and 4% Lidocaine (non-occluded), 20% NE with 0.13% BZK and 4% Lidocaine (occluded).
  • the composition of the NE is shown in Table 29.
  • the skin was equilibrated for a period of 30 minutes before dosing.
  • a 113 pL (over a dosing area of 1.13 cm 2 ) dose of the liquid test formulations were topically applied onto the epidermal surface of the cadaver skin mounted on the donor chamber of the diffusion cells using a positive displacement pipette.
  • Half of the cells with the NE formulation was left non-occluded and half were occluded with a parafilm film placed over the donor cap to stop any evaporation of the NE from the skin surface.
  • Salonpas Gel Patch a piece of the patch was cut to fit a surface area of 1.13 cm 2 area and the donor cap was clamped into the cell.
  • the amount of active agent (Lidocaine) that permeated into the epidermis (at 1 and 8 hours, see Figure 40), dermis (at 1 and 8 hours, see Figure 41) and the receptor compartment (at 8 hours, see Figure 42) was determined by HPLC.
  • the levels of the active agent (Lidocaine) in each skin area are represented as the amount per wet tissue weight (pg/grams) ⁇ the standard deviation.
  • the number of replicas used in the calculation was 4 or 5 for each formulation.
  • NanoBio® Protect Nasal (BlueWillow Biologies, Ann Arbor, MI) is a nanoemulsion with a mean droplet diameter of approximately 350 nm.
  • the nanemulsion components are depicted above in Table 5 and shown again below.
  • the composition has a surfactant blend ratio of 1 :5.
  • the active agent in the nanoemulsion is benzalkonium chloride (BZK), a quaternary ammonium compound, which has antimicrobial activity and is a skin antiseptic under the FDA skin antiseptic monograph (Department of Health and Huma Services, Food and Drug Administration Safety and Effectiveness for Health Care Antiseptics; Topical Antimicrobial Drug Products for Over-the-Counter Human Use; Proposed Amendment of the Tentative Final Monograph; Reopening of Administrative Record Docket No. FDA-2015-N-0101.)
  • BZK benzalkonium chloride
  • BZK resides at the interface between the oil and water phases of the nanodroplets with the hydrophobic tail distributed in the oil core and the polar cationic head group residing at the water phase as shown in Figure 1.
  • Virus Strains SARS-CoV-2 Victoria/1/2020 strain (Public Health England (PHE), Porton Down, Salisbury, UK); Human coronavirus 229E (ATCC: VR-740); Influenza B (VR-1931); Respiratory Syncytial Virus (BlueWillow Biologies in-house strain: NBL-14-001- 2UC).
  • the virus growth media was either Minimum Essential Medium - Eagle with Earle's BSS (MEM Eagle EBSS) from Lonza (Rochester, NY) or Dulbecco’s Modified Eagle’s Medium (DMEM) from Corning Inc (Coming, NY).
  • ATTC Manassas, VA
  • vero E6 cells ATCC# CRL 1586
  • MRC-5 cells for HCoV229E
  • MDCK cells for Influenza B
  • vero cells for RSV (CCL-81 ATCC).
  • Bacteria Staphylococcus aureus (ATCC: 6538), Enterococcus faecium (ATCC: 51559), Staphylococcus epidermidis (ATCC: 12228). Methicillin Resistant Staphylococcus aureus (MRS A, USA 300), Pseudomonas aeruginosa (ATCC: 9027), Serratia marcescens (ATCC: 14756), Acenitobacter baumannii (ATCC: 19606), Klebsiella pneumoniae (ATCC: 13883).
  • TSA Tryptic Soy Agar
  • FBS Fetal Bovine Serum
  • the neutralizing buffer used was Butterfield’s Buffer and was obtained from Hardy Diagnostics (Santa Maria, CA).
  • Incubators (C02 , oxygen controls and temperate monitoring) for bacterial and viral studies were obtained from ThermoFisher Scientific (Waltham, MA).
  • An infrared surface temperature thermometer was purchased from VRW Scientific (Radnor, PA) to measure skin surface temperatures.
  • nanoemulsion antiseptic was inoculated with test organisms at 10% or 50% (v/v), and incubated at RT for 1, 5, 15 and 30 minutes.
  • Bacterial and viral loads of treated samples were determined by plating the serial dilutions of samples onto TSA plates for bacteria or appropriate host cells for viruses.
  • HCoV 229E was also evaluated at no dilution, as well as a 1/10, 1/20 dilution and 1/40 dilution, and compared against an aqueous BZK (0.13%) test sample. Concentration of active virus particles was determined quantitively by plaque or TCID50 assay. Briefly, serially diluted samples were plated onto 80- 90% confluent vero E6 cells for SARS-CoV-2, MRC-5 cells for HCoV229E, MDCK for Influenza B and vero cells for RSV. Plates were incubated for 5-7 days at 35°C for human corona viruses and influenza B, and at 37°C for RSV, under 5% C02.
  • TCID50 was calculated by the Karber method as referenced by Lambert (Titration of Human Coronaviruses, HCoV-229E and HCoV-OC43, by an Indirect Immunoperoxidase Assay, Lambert, F. , Helene Jacomy, H., Marceau, G. and Pierre J. Talbot. Methods in Molecular Biology, vol. 454: SARS- and Other Coronaviruses, 93, Edited by: D. Cavanagh, DOI: 10.1007/978-1-59745-181-9 C _ 8, Humana Press, New York, NY), based on the presence of cytopathic effect in host cells.
  • NCLS Methods for Determining Bacterial Activity of Antimicrobial Agents: Approved Guideline Document MS26-1, 1999).
  • a nanoemulsion sample was inoculated with a suspension of bacteria at a final concentration 1x107 to 1x109 CFU/ml (ASTM E2315-16, Standard Guide for Assessment of Antimicrobial Activity Using a Time-Kill Procedure, ASTM International, West Conshohocken, PA, 2016, www.astm.org.)
  • ASTM E2315-16 Standard Guide for Assessment of Antimicrobial Activity Using a Time-Kill Procedure, ASTM International, West Conshohocken, PA, 2016, www.astm.org.
  • an aliquot was removed, serially diluted and plated on to TSA agar to determine the surviving test microorganisms.
  • the nanoemulsion was diluted 1 : 100 in a neutralizing buffer to remove residual activity of the product.
  • the plates were incubated at 35°C for 24 to 48 hours and the numbers of CFU per plate were counted, converted into log 10 and compared to an initial starting population to determine log reduction.
  • the active agent is considered bactericidal at the concentration and contact time that demonstrates a 34ogl0 (99.9%) or greater reduction in bacterial viability for the strains tested.
  • D- Squame® adhesive tape CuDerm Corp, Dallas, TX
  • D-Squame® adhesive tape CuDerm Corp, Dallas, TX
  • D-Squame tape uniformly removes a sample from a fixed surface area on the stratum corneum on the skin surface (Serup, L, Winter, C., Blichmann, A. simple method for the study of scale pattern and effects of a moisture-qualitative and quantitative evaluation by D-Squame® tape compared with parameters of epidermal hydration. Clinical and Experimental Dermatology, 1365-2230 (1989).)
  • the BZK in the stripes in each vial were extracted with a 70% ethanol solution, sonication for 30 minutes and then filtered through a 0.45 pm PTFE membrane syringe filter into HPLC vials.
  • the amount of BZK (pg/cm 2 surface area) was measured by HPLC, 4 or 8 hours after each washing procedure by reverse phase-high performance liquid chromatography (RP- HPLC).
  • the HPLC was equipped with UV detector set at 254 nm, column Luna CN, 250 x 4 mm, 5 pm at 55°C, mobile phase; acetate buffer and acetonitrile (ACN) in the ratio of 40:60 (v/v) in isocratic mode.
  • the washes were pooled and neutralized to remove residual effect of the test formulation by diluting at 1:100 dilution in virus growth media containing 1-2% FBS. Concentration of active virus particles was determined quantitively by plaque or TCID50 assay. Briefly, serially diluted samples were plated onto 80-90% confluent MRC-5 cells. Plates were incubated for 4-6 days at 35°C under 5% CO2.
  • TCID50 was calculated by the Karber method as reference by Lambert (Titration of Human Coronaviruses, HCoV-229E and HCoV-OC43, by an Indirect Immunoperoxidase Assay, Lambert, F., Helene Jacomy, H., Marceau, G. and Pierre J. Talbot. Methods in Molecular Biology, vol. 454: SARS- and Other Coronaviruses, 93, Edited by: D. Cavanagh, DOI: 10.1007/978-1-59745-181-9 C _ 8, Humana Press, New York, NY., based on the presence of cytopathic effect in host cells.
  • NCLS Methods for Determining Bacterial Activity of Antimicrobial Agents: Approved Guideline Document MS26-1, 1999.
  • NE-BZK nanoemulsion antiseptic
  • AQ-BZK aqueous BZK
  • alcohol-based nasal sanitizer Nozin Nasal Sanitizer ® , Global Life Technologies Corp, Bethesda, MD
  • WFI Sterile water for injection
  • MRSA inoculum in saline final concentration of lxlO 7 to lxlO 9 CFU/ml
  • the skin surface was washed, the wash was removed and neutralized to remove residual effect of the products by diluting 1:100 in TAT neutralizing buffer.
  • the samples were serially diluted and plated on to TSA agar to determine the surviving test microorganisms. The plates were incubated at 35°C for 24 to 48 hours and the numbers of CFU per plate were counted, converted into log 10 and compared to an initial starting population to determine log reduction.
  • the hydration effect of applying NE-BZK on human skin was evaluated by measuring the moisture level of tested skin area on human forearm and back arm of 5 human volunteers (3 females and 2 males) ranging in age from 30 to 62 years.
  • the Delfin Meter SC (Miami, FL) uses a precise (1.25 MHz) electromagnetic field to measure the skin’s dielectric constant giving more accurate and reproducible results (Alanen et ak, “Measurement of hydration in the stratum corneum with the MoistureMeter and comparison with the Comeometer,” Skin Res Technol , 10:32-37 (2004)).
  • FIG. 48 illustrates the antiviral and antibacterial activity of NE-BZK against several common pathogens, including the novel coronavirus SARS-CoV-2.
  • the purpose of this example was to evaluate the antibacterial activity against human coronavirus and MRSA of an antibacterial nanoemulsion comprising BZK (NE-BZK) as compared to aqueous BZK (AQ-BZK).
  • NE-BZK antibacterial nanoemulsion comprising BZK
  • AQ-BZK aqueous BZK
  • AQ-BZK formulations are the most commonly marketed BZK skin antiseptics. Three different concentrations of NE-BZK, including full-strength (0.13% BZK), 1/10 dilution (0.013% BZK) and 1/20 dilution (0.0065% BZK), were tested for in vitro antiviral activity as compared to the same concentrations of AQ-BZK.
  • Figure 49 shows that the NE-BZK exhibited a 5.8 and 3.8 fold increase substantivity on volunteers’ skin surface at 4 hours after two washing protocols (e.g. 1) water rinse and 2) soap rub +water rinse) and 16.5 and 7.3 fold increase, respectively, after 8 hours after a two washing protocols as compared to a 0.13% AQ-BZK solution.
  • the data from Fig. 49 is shown below in Table 34.
  • NE-BZK Antiviral activity of NE-BZK was measured against human coronavirus (HCoV229E) ex vivo in a time-kill study following 15 minutes exposure of skin pre-treated with the nanoemulsion antiseptic (0.13% BZK) or AQ-BZK for 4 and 8 hours. As presented in Table 35, NE-BZK achieved >4.7% log killing at both the 4- and 8hour time points. AQ-BZK exhibited only 1.5 log killing at 4 hours and below the limit of detection at 8 hours.
  • Virus killing is calculated based on recovery from WFI control samples (5.70 Logs per skin sample)
  • NE- BZK achieved >5.2% log killing at 4, 8 and 12 hours.
  • AQ-BZK formulation exhibited only 0.5 log killing at 4 hours and 0.21 log killing at 8 hours.
  • the alcohol -based nasal sanitizer demonstrated no antimicrobial activity at either 8 or 12 hours after application.
  • NE-BZK significantly enhanced skin hydration for up to three hours after a single application compared to the other formulations tested.

Abstract

La présente divulgation concerne des compositions de nanoémulsion présentant certains rapports de mélange de tensioactifs qui confèrent une perméabilité améliorée. De telles compositions sont utiles pour des applications topiques, mucosales et intranasales et permettent l'administration plus importante d'un ou de plusieurs principes actifs au niveau du site d'application pour prévenir une infection par un micro-organisme.
EP21771166.2A 2020-03-17 2021-03-16 Compositions antimicrobiennes topiques persistantes et leurs méthodes d'utilisation Pending EP4121026A4 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US202062990534P 2020-03-17 2020-03-17
US16/828,542 US20200237689A1 (en) 2018-11-15 2020-03-24 Prevention and treatment of coronavirus and other respiratory infections using nanoemulsion compositions
PCT/US2021/022534 WO2021188518A1 (fr) 2020-03-17 2021-03-16 Compositions antimicrobiennes topiques persistantes et leurs méthodes d'utilisation

Publications (2)

Publication Number Publication Date
EP4121026A1 true EP4121026A1 (fr) 2023-01-25
EP4121026A4 EP4121026A4 (fr) 2024-04-10

Family

ID=77771573

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21771166.2A Pending EP4121026A4 (fr) 2020-03-17 2021-03-16 Compositions antimicrobiennes topiques persistantes et leurs méthodes d'utilisation

Country Status (2)

Country Link
EP (1) EP4121026A4 (fr)
WO (1) WO2021188518A1 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023089433A2 (fr) * 2021-11-17 2023-05-25 Council For Scientific And Industrial Research Désinfectant pour les mains antimicrobien sans alcool

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2180636T3 (es) * 1994-05-20 2003-02-16 Novavax Inc Emulsiones antimicrobianas de aceite en agua.
US8232320B2 (en) * 1999-04-28 2012-07-31 The Regents Of The University Of Michigan Antimicrobial nanoemulsion compositions and methods
US20050100601A1 (en) * 2003-11-07 2005-05-12 Viratox, L.L.C. Virucidal activities of cetylpyridinium chloride
AU2009240389A1 (en) * 2008-04-25 2009-10-29 Nanobio Corporation Nanoemulsions for treating fungal, yeast and mold infections
EP3329936A1 (fr) * 2011-08-22 2018-06-06 Nanobio Corporation Vaccin à base de nanoémulsion du virus de l'herpès simplex
US20210393554A1 (en) * 2018-11-15 2021-12-23 Bluewillow Biologics, Inc. Nanoemulsion compositions having enhanced permeability

Also Published As

Publication number Publication date
EP4121026A4 (fr) 2024-04-10
WO2021188518A1 (fr) 2021-09-23

Similar Documents

Publication Publication Date Title
US20200237689A1 (en) Prevention and treatment of coronavirus and other respiratory infections using nanoemulsion compositions
US11369578B2 (en) Persistent topical antimicrobial compositions and methods of using the same
US20100226983A1 (en) Compositions for treatment and prevention of acne, methods of making the compositions, and methods of use thereof
US9782426B2 (en) Anti-viral therapeutic composition
US20210077544A1 (en) Sanitizer composition with probiotic/prebiotic active ingredient
US20100075914A1 (en) Methods for treating herpes virus infections
JP2005514325A (ja) 抗微生物ナノエマルジョンの組成物および方法
WO2010059366A2 (fr) Savons antimicrobiens moussants
US6413921B1 (en) Antimicrobial composition containing parachlorometaxylenol (PCMX)
WO2009132343A1 (fr) Nanoemulsions destinées à traiter l'onychomycose
AU2017240654A1 (en) Topical cleansing composition with prebiotic/probiotic additive
US20200352164A1 (en) Antimicrobial preservative compositions
AU2017365025A1 (en) Topical cleansing composition with prebiotic/probiotic additive
WO2019171385A1 (fr) Conservation de compositions de soins personnels
EP4121026A1 (fr) Compositions antimicrobiennes topiques persistantes et leurs méthodes d'utilisation
JP2007217394A (ja) 医薬組成物
WO2009054992A1 (fr) Compositions antimicrobiennes comprenant du docusate
KR101326164B1 (ko) 항균성 피부 외용제 조성물
US20170304411A1 (en) Topical formulation for treating skin or mucosal infections, preparation method and uses thereof
EP1638516A2 (fr) Perfectionnements apportes a une matiere organique ou concernant celle-ci
JP2010209109A (ja) 抗微生物ナノエマルジョンの組成物および方法

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20221014

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Free format text: PREVIOUS MAIN CLASS: A61K0031140000

Ipc: A61K0009000000

A4 Supplementary search report drawn up and despatched

Effective date: 20240314

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 47/18 20170101ALI20240307BHEP

Ipc: A61K 47/10 20170101ALI20240307BHEP

Ipc: A61K 31/77 20060101ALI20240307BHEP

Ipc: A61K 31/444 20060101ALI20240307BHEP

Ipc: A61K 31/4425 20060101ALI20240307BHEP

Ipc: A61K 31/085 20060101ALI20240307BHEP

Ipc: A61K 38/21 20060101ALI20240307BHEP

Ipc: A61K 31/4706 20060101ALI20240307BHEP

Ipc: A61P 31/14 20060101ALI20240307BHEP

Ipc: A61K 31/14 20060101ALI20240307BHEP

Ipc: A61K 45/06 20060101ALI20240307BHEP

Ipc: A61K 47/34 20170101ALI20240307BHEP

Ipc: A61K 47/26 20060101ALI20240307BHEP

Ipc: A61K 47/44 20170101ALI20240307BHEP

Ipc: A61K 9/107 20060101ALI20240307BHEP

Ipc: A61K 9/00 20060101AFI20240307BHEP