AU2021254780A1 - Compositions and methods to reduce the infectivity of a virus - Google Patents

Compositions and methods to reduce the infectivity of a virus Download PDF

Info

Publication number
AU2021254780A1
AU2021254780A1 AU2021254780A AU2021254780A AU2021254780A1 AU 2021254780 A1 AU2021254780 A1 AU 2021254780A1 AU 2021254780 A AU2021254780 A AU 2021254780A AU 2021254780 A AU2021254780 A AU 2021254780A AU 2021254780 A1 AU2021254780 A1 AU 2021254780A1
Authority
AU
Australia
Prior art keywords
pharmaceutical composition
composition
aerosol
inhibitor
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
AU2021254780A
Inventor
Steven C. Quay
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.)
Atossa Therapeutics Inc
Original Assignee
Atossa Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Atossa Therapeutics Inc filed Critical Atossa Therapeutics Inc
Publication of AU2021254780A1 publication Critical patent/AU2021254780A1/en
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
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/23Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms
    • A61K31/231Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms having one or two double bonds
    • 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
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Otolaryngology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Emergency Medicine (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Agricultural Chemicals And Associated Chemicals (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Compositions, devices, and methods for treating a viral infection may comprise use of an enzyme inhibitor, for example a protease inhibitor. A composition of the present disclosure may comprise a dual -action enzyme inhibitor that inhibits two or more enzymes and reduces the infectivity of a virus for a host cell. Additional components may include a bioadhesive that disrupts viral envelopes. A method of treating a viral infection may comprise administering an enzyme inhibitor, such as a dual-action enzyme inhibitor, to a subject and reducing the infectivity of the virus for a host cell of the subject. The compositions may be administered via nasal administration to reduce the infectivity of a viral infection, for example a coronavirus infection.

Description

COMPOSITIONS AND METHODS TO REDUCE THE INFECTIVITY OF A VIRUS
CROSS-REFERENCE
[0001] The present application claims the benefit of U.S. Provisional Application No.
63/011,291, entitled “SERINE PROTEASE INHIBITORS FOR THE TREATMENT OF VIRAL INFECTIONS,” filed on April 16, 2020; U.S. Provisional Application No. 63/012,818, entitled “PROTEASE INHIBITORS FOR THE TREATMENT OF VIRAL INFECTIONS,” filed on April 20, 2020; U.S. Provisional Application No. 63/015,370, entitled “PROTEASE INHIBITORS FOR THE TREATMENT OF VIRAL INFECTIONS,” filed on April 24, 2020; U.S. Provisional Application No. 63/032,435, entitled “PROTEASE INHIBITORS FOR THE TREATMENT OF VIRAL INFECTIONS,” filed on May 29, 2020; U.S. Provisional Application No. 63/051,800, entitled “PROTEASE INHIBITORS FOR THE TREATMENT OF VIRAL INFECTIONS,” filed on July 14, 2020; and U.S.
Provisional Application No. 63/112,112, entitled “PROTEASE INHIBITORS FOR THE TREATMENT OF VIRAL INFECTIONS,” filed on November 10, 2020, each of which applications are herein incorporated by reference in their entireties for all purposes.
BACKGROUND
[0002] Viral infections are responsible for hundreds of thousands of deaths each year. However, treatment options are limited for many viruses. Additionally, carriers of a virus may be asymptomatic, leading to high transmission rates from infected but asymptomatic individuals. There is a need for improved drugs to treat viral infections in both symptomatic and asymptomatic individuals. Furthermore, people such as healthcare workers who are in contact with infected individuals are at high-risk of infection. There is a need for drugs to prevent viral infections in at-risk individuals and other members of the population.
SUMMARY
[0003] In various aspects, the present disclosure provides a method of reducing the infectivity of a coronavirus in a subject, the method comprising: administering a pharmaceutical composition comprising an enzyme inhibitor and a bioadhesive to the subject; and reducing the infectivity of the coronavirus for a host cell of the subject.
[0004] In some aspects, the bioadhesive is glyceryl monooleate. In some aspects, wherein the glyceryl monooleate inactivates an envelope of the coronavirus. In some aspects, wherein the enzyme inhibitor is disodium edetate, citrate, argatroban, hexa-D-arginine, or a combination thereof. In some aspects, the disodium edetate inhibits angiotensin converting enzyme 2, furin, or a combination thereof. In some aspects, the citrate inhibits furin. In some aspects, the argatroban inhibits type II transmembrane serine protease. In some aspects, the hexa-D- arginine inhibits furin. In some aspects, the method further comprises inhibiting the enzyme, wherein inhibiting the enzyme inhibits entry of the coronavirus into the host cell of the subject. In some aspects, the enzyme is a type II transmembrane serine protease, angiotensin converting enzyme 2, a cathepsin B, a cathepsin L, a furin, or a combination thereof.
[0005] In some aspects, the pharmaceutical composition further comprises a stabilizer. In some aspects, the stabilizer is polysorbate 80, microcrystalline cellulose, carboxymethyl cellulose, or a combination thereof. In some aspects, the pharmaceutical composition further comprises an antioxidant. In some aspects, the antioxidant is butylated hydroxytoluene. In some aspects, the pharmaceutical composition further comprises a bacteriostatic agent. In some aspects, the bacteriostatic agent is selected from the group consisting of sodium benzoate, benzalkonium chloride, thiomersal, chlorobutanol, chlorbutol, potassium sorbate, and methyl paraben, or a combination thereof. In some aspects, the bacteriostatic agent is benzalkonium chloride.
[0006] In some aspects, the method comprises nasally administering the pharmaceutical composition to the subject. In some aspects, the method comprises: providing an actuator comprising an actuator tip; producing an aerosol comprising droplets of the pharmaceutical composition from the actuator tip; and dispensing the aerosol into a nose of the subject. In some aspects, the method comprises producing a spray pattern ellipticity ratio of no less than 1.0 to not more than 1.4. In some aspects, the method comprises producing a droplet size distribution such that no more than about 5% of the aerosol volume forms droplets that are less than about 10 pm in diameter. In some aspects, the method comprises producing a droplet size distribution such that at least 0.4% of the aerosol volume forms droplets that are less than about 10 pm in diameter.
[0007] In some aspects, the method comprises producing a droplet size distribution such that no more than about 50% of the aerosol volume forms droplets that are less than about 26.9 pm in diameter. In some aspects, the method comprises producing a droplet size distribution such that a diameter of droplet for which 50% of the aerosol volume forms droplets of smaller diameter is no less than 25 pm and not more than 75 pm. In some aspects, the method comprises producing a droplet size distribution such that a diameter of droplet for which 10% of the aerosol volume forms droplets of smaller diameter is no less than 15 pm and not more than 35 pm. In some aspects, the method comprises producing a droplet size distribution such that a diameter of droplet for which 90% of the aerosol volume forms droplets of smaller diameter is no less than 70 mih and not more than 150 mih. In some aspects, the method comprises producing a spray pattern with a major axis of at least 25 mm and not more than 40 mm and a minor axis of at least 25 mm and not more than 40 mm.
[0008] In various aspects, the present disclosure provides a pharmaceutical composition comprising disodium edetate, glyceryl monooleate, polysorbate 80, microcrystalline cellulose or carboxymethyl cellulose, citrate, and benzalkonium chloride, wherein the pharmaceutical composition is formulated for nasal delivery.
[0009] In some aspects, the disodium edetate is present at a concentration of at least 1 mg/mL and not more than 10 mg/mL, at least 1 mg/mL and not more than 6 mg/mL, or at least 2 mg/mL and not more than 6 mg/mL. In some aspects, the disodium edetate is present at a concentration of at least 3 mg/mL and not more than 5 mg/mL. In some aspects, the glyceryl monooleate is present at a concentration at least 0.5 mg/mL and not more than 10 mg/mL, at least 0.5 mg/mL and not more than 8 mg/mL, at least 1 mg/mL at not more than 6 mg/mL, or at least 1 mg/mL and not more than 5 mg/mL. In some aspects, the glyceryl monooleate is present at a concentration of at least 1.5 mg/mL and not more than 3.5 mg/mL.
[0010] In some aspects, the polysorbate 80 is present at a concentration of at least 1 mg/mL and not more than 10 mg/mL, at least 2 mg/mL and not more than 8 mg/mL, or at least 4 mg/mL and not more than 6 mg/mL. In some aspects, the polysorbate 80 is present at a concentration of at least 4 mg/mL and not more than 6 mg/mL. In some aspects, the microcrystalline cellulose and carboxymethyl cellulose are present at a combined concentration of at least 10 mg/mL and not more than 50 mg/mL, at least 10 mg/mL and not more than 40 mg/mL, at least 20 mg/mL and not more than 50 mg/mL, or at least 20 mg/mL and not more than 40 mg/mL. In some aspects, the microcrystalline cellulose and carboxymethyl cellulose are present at a combined concentration of at least 20 mg/mL and not more than 40 mg/mL.
[0011] In some aspects, the citrate is present at a concentration of at least 1 mg/mL and not more than 10 mg/mL, at least 2 mg/mL and not more than 8 mg/mL, or at least 3.0 mg/mL and not more than 5.0 mg/mL. In some aspects, the citrate is present at a concentration of at least 3.5 mg/mL and not more than 5.5 mg/mL. In some aspects, the benzalkonium chloride is present at a concentration of at least 0.05 mg/mL and not more than 2 mg/mL, at least 0.05 mg/mL and not more than 0.1 mg/mL, or at least 0.1 mg/mL and not more than 0.5 mg/mL.
In some aspects, the benzalkonium chloride is present at a concentration of at least 0.1 mg/mL and not more than 1.5 mg/mL. In some aspects, the benzalkonium chloride is present at a concentration of no more than 0.1% (w/w). In some aspects, the benzalkonium chloride is present at a concentration of at least 0.01% and no more than 0.2% (w/w).
[0012] In some aspects, the pharmaceutical composition further comprises butylated hydroxytoluene. In some aspects, the butylated hydroxytoluene is present at a concentration of at least 0.1 mg/mL and not more than 3 mg/mL, at least 0.1 mg/mL at not more than 2 mg/mL, at least 0.2 mg/mL and not more than 1.0 mg/mL, or at least 0.4 mg/mL and not more than 0.6 mg/mL. In some aspects, the butylated hydroxytoluene is present at a concentration of at least 0.4 mg/mL and not more than 0.6 mg/mL. In some aspects, the composition is at a pH of at least 4 and not more than 6, at least 4.2 and not more than 5.8, or at least 4.5 and not more than 5.5.
[0013] In some aspects, the pharmaceutical composition further comprises argatroban. In some aspects, the argatroban is present at a concentration of at least 0.001 mg/mL but not more than 2 mg/mL, at least 0.05 mg/mL but not more than 1 mg/mL, at least 0.05 mg/mL but not more than 0.5 mg/mL, or at least 0.05 mg/mL and not more than 0.15 mg/mL. In some aspects, the argatroban is present at a concentration of at least 0.05 mg/mL and not more than 0.15 mg/mL.
[0014] In some aspects, the pharmaceutical composition further comprises Hexa-D-arginine. In some aspects, the Hexa-D-arginine is present at a concentration of at least 1 mg/mL and not more than 20 mg/mL, at least 1 mg/mL and not more than 10 mg/mL, or at least 2 mg/mL and not more than 8 mg/mL. In some aspects, the Hexa-D-arginine is present at a concentration of at least 3 mg/mL and not more than 7 mg/mL. In some aspects, the pharmaceutical composition is formulated at a pH of at least 4.0 and not more than 6.0.
[0015] In various aspects, the present disclosure provides a pharmaceutical composition comprising an enzyme inhibitor, a bioadhesive, and a stabilizer, wherein the pharmaceutical composition is formulated for nasal delivery.
[0016] In some aspects, the enzyme inhibitor is a dual-action enzyme inhibitor. In some aspects, the enzyme inhibitor chelates calcium, zinc, or a combination thereof. In some aspects, the enzyme inhibitor comprises disodium edetate, EDTA-magnesium, or EGTA. In some aspects, the enzyme inhibitor is present at a concentration of at least 1 mg/mL and not more than 10 mg/mL, at least 1 mg/mL and not more than 6 mg/mL, or at least 2 mg/mL and not more than 6 mg/mL. In some aspects, the enzyme inhibitor is argatroban. In some aspects, the stabilizer comprises microcrystalline cellulose, carboxymethyl cellulose, polysorbate 80, or combinations thereof. In some aspects, the bioadhesive is glyceryl monooleate. [0017] In some aspects, the pharmaceutical composition further comprises an antioxidant. In some aspects, the antioxidant is butylated hydroxy toluene. In some aspects, the pharmaceutical composition further comprises a bacteriostatic agent. In some aspects, the bacteriostatic agent is selected from the group consisting of sodium benzoate, benzalkonium chloride, thiomersal, chlorobutanol, chlorbutol, potassium sorbate, and methyl paraben, or a combination thereof. In some aspects, the bacteriostatic agent is benzalkonium chloride. [0018] In some aspects, the pharmaceutical composition further comprises a buffer. In some aspects, the buffer is citrate. In some aspects, the pharmaceutical composition further comprises a furin peptide inhibitor. In some aspects, the furin peptide inhibitor is present at a concentration of at least 2 mg/mL, at least 4 mg/mL, or at least 5 mg/mL. In some aspects, the furin peptide inhibitor is present at a concentration of at least 1 mg/mL and not more than 20 mg/mL, at least 1 mg/mL and not more than 10 mg/mL, or at least 2 mg/mL and not more than 8 mg/mL. In some aspects, the furin peptide inhibitor is present at a concentration of at least 3 mg/mL and not more than 7 mg/mL. In some aspects, the furin peptide inhibitor is [W5R4C], [WR]5, C12-[R5], W4-[R5], hexa-D-arginine, or nona-D-arginine. In some aspects, the furin peptide inhibitor is Hexa-D-arginine. In some aspects, the pharmaceutical composition is at a pH of at least 4 and not more than 6, at least 4.2 and not more than 5.8, or at least 4.5 and not more than 5.5. In some aspects, the pharmaceutical composition is free of endotoxins. In some aspects, the pharmaceutical composition is aqueous.
[0019] In some aspects, the pharmaceutical composition is formulated for delivery as an aerosol from an actuator tip. In some aspects, the aerosol has a spray pattern ellipticity ratio of no less than 1.0 to not more than 1.4. In some aspects, the aerosol comprises droplets, and wherein no more than about 5% of the aerosol volume forms droplets that are less than about 10 pm in diameter. In some aspects, the aerosol comprises droplets, and wherein at least 0.4% of the aerosol volume forms droplets that are less than about 10 pm in diameter.
[0020] In some aspects, the aerosol comprises droplets, and wherein no more than about 50% of the aerosol volume forms droplets that are less than about 26.9 pm in diameter. In some aspects, the aerosol comprises droplets, and wherein a diameter of droplet for which 50% of the aerosol volume forms droplets of smaller diameter is no less than 25 pm and not more than 75 pm. In some aspects, the aerosol comprises droplets, and wherein a diameter of droplet for which 10% of the aerosol volume forms droplets of smaller diameter is no less than 15 pm and not more than 35 pm. In some aspects, the aerosol comprises droplets, and wherein a diameter of droplet for which 90% of the aerosol volume forms droplets of smaller diameter is no less than 70 pm and not more than 150 pm. In some aspects, the aerosol has a spray pattern with a major axis of at least 25 mm and not more than 40 mm and a minor axis of at least 25 mm and not more than 40 mm. In some aspects, the aerosol has a spray pattern with a plume angle of at least 30° and no more than 70°.
[0021] In various aspects, the present disclosure provides a pharmaceutical device comprising: a pharmaceutical composition described herein, and an actuator comprising an actuator tip; wherein the actuator is configured to produce an aerosol of the pharmaceutical composition.
[0022] In some aspects, the aerosol has a spray pattern ellipticity ratio of no less than 1.0 to not more than 1.4. In some aspects, the aerosol comprises droplets, and wherein no more than about 5% of the aerosol volume forms droplets that are less than about 10 pm in diameter. In some aspects, the aerosol comprises droplets, and wherein at least 0.4% of the aerosol volume forms droplets that are less than about 10 pm in diameter.
[0023] In some aspects, the aerosol comprises droplets, and wherein no more than about 50% of the aerosol volume forms droplets that are less than about 26.9 pm in diameter. In some aspects, the aerosol comprises droplets, and wherein a diameter of droplet for which 50% of the aerosol volume forms droplets of smaller diameter is no less than 25 pm and not more than 75 pm. In some aspects, the aerosol comprises droplets, and wherein a diameter of droplet for which 10% of the aerosol volume forms droplets of smaller diameter is no less than 15 pm and not more than 35 pm. In some aspects, the aerosol comprises droplets, and wherein a diameter of droplet for which 90% of the aerosol volume forms droplets of smaller diameter is no less than 70 pm and not more than 150 pm.
[0024] In some aspects, the aerosol has a spray pattern with a major axis of at least 25 mm and not more than 40 mm and a minor axis of at least 25 mm and not more than 40 mm. In some aspects, the aerosol has a spray pattern with a plume angle of at least 30° and not more than 70°.
[0025] In various aspects, the present disclosure provides a method of reducing the infectivity of a coronavirus in a subject, the method comprising administering a pharmaceutical composition described herein to the subject using a pharmaceutical device described herein, and reducing the infectivity of the coronavirus.
[0026] In some aspects, the subject has a coronavirus infection. In some aspects, the subject is at risk of developing a coronavirus infection. INCORPORATION BY REFERENCE
[0027] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
BRIEF DESCRIPTION OF THE DRAWINGS [0028] The novel features of the disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present disclosure will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the disclosure are utilized, and the accompanying drawings of which:
[0029] FIG. 1 schematically illustrates a SARS-CoV-2 virion and a target cell angiotensin converting enzyme-2 (ACE-2) receptor.
[0030] FIG. 2 schematically illustrates binding of a SARS-CoV-2 virion to a transmembrane ACE-2 receptor, mediating infection of a target cell. Binding of the virion to the ACE-2 receptor may be inhibited by an ACE-2 inhibitor (top). Internalization may be activated by type II transmembrane serine proteases (TMPRSSs) of the host cell interacting with an arginine cluster (R685, R683, and R682) in the SI and S2 regions of a SARS-CoV-2 spike protein (bottom).
[0031] FIG. 3 shows expression levels of TMPRSS2 in different tissue types.
[0032] FIG. 4 shows examples of small molecule ACE-2 inhibitors and methods of synthesis.
[0033] FIG. 5 shows fluorescent images of VERO cells contacted with SARS-CoV-2 virus pre-treated with serial dilutions of a dual-action inhibitor composition formulated for nasal delivery. Fluorescence indicates the presence of the SARS-CoV-2 N-protein and is representative of viral infection of the cells.
[0034] FIG. 6 illustrates a spray pattern (top) and a plume geometry (bottom) of a first device for nasal delivery of a solution to reduce the infectivity of a virus.
[0035] FIG. 7 illustrates a spray pattern (top) and a plume geometry (bottom) of a second device for nasal delivery of a solution to reduce the infectivity of a virus. DETAILED DESCRIPTION
[0036] Disclosed herein are compositions, methods, and treatment plans for treating an individual who is at risk of having a viral infection, has mild symptoms of a viral infection, or has severe symptoms of a viral infection. A composition of the present disclosure may be used to treat, prevent, or reduce the infectivity of a viral infection, such as a respiratory viral infection. A treatment plan may comprise administering a composition (e.g., a composition comprising an enzyme inhibitor, a bioadhesive, or both) to an individual at risk of having a viral infection or who has a viral infection, and reducing the infectivity of the virus, thereby preventing or treating the viral infection. In some embodiments, the infectivity of a virus may be reduced by disrupting an envelope of the virus (e.g., with a bioadhesive). In some embodiments, the infectivity of a virus may be reduced by inhibiting an enzyme associated with viral infection. For example, a composition of the present disclosure may comprise an enzyme inhibitor that inhibits a type II transmembrane serine protease (TMPRSS), an epithelial angiotensin converting enzyme 2 (ACE2), an endosomal cysteine protease, such as cathepsin B and L (CatB/L), a furin enzyme, or combinations thereof. In some embodiments, an enzyme inhibitor may be a dual action protease inhibitor that inhibits two or more proteases (e.g., two or more of TMPRSS, ACE2, furin, cathepsin B, or cathepsin L).
[0037] FIG.l shows infection of a host cell with SARS-CoV-2 mediated by interactions between the SRS-CoV-2 spike (S) proteins and transmembrane ACE-2 receptors of the host cell. In some embodiments, a viral infection may be prevented by disrupting interactions between a viral surface proteins and host cell proteins that activate or enhance insertion of the viral genetic material into the host cell. FIG. 2 shows interactions between a SARS-CoV-2 spike protein, a host cell ACE-2 receptor, and host cell type II transmembrane serine proteases (TMPRSSs). The TMPRSSs may interact with an arginine cluster within the SARS-CoV-2 spike protein, thereby activating or enhancing viral invasion of the host cell. [0038] A composition of the present disclosure may be formulated as a nasal spray and administered as a for treating or preventing a respiratory viral infection (e.g., a coronavirus infection such as COVID-19). Administration of the composition to the nasal cavity of a subject may form or maintain a protective mucosal-like barrier that reduces the infectivity of the respiratory virus. The compositions of the present disclosure (e.g., compositions formulated for nasal delivery) may be administered to a subject who may be at risk of contracting a viral infection (e.g., COVID-19). For example, the compositions of the present disclosure may be administered to individuals in high risk environments (e.g., healthcare workers), individuals who have been or who are suspected to have been exposed to a virus (e.g., SARS-CoV-2), or individuals who have tested positive for a viral infection. A composition of the present disclosure may be administered to an individual who is displaying symptoms of a respiratory infection (e.g., a SARS-CoV-2 infection) or who is asymptomatic at the time of administration. A composition of the present disclosure may be administered to a subject upon diagnosis of the subject with a respiratory virus. In some embodiments, administration of the composition may reduce symptoms associated the virus. In some embodiments, administration of the composition may slow the infection rate of the virus, enabling the immune system of the subject to more effectively fight the viral infection. The compositions of the present disclosure may be self-administered by the individual (e.g., as a nasal spray) and may be administered outside of a medical facility (e.g., at home).
[0039] In some embodiments, the methods and compositions provided herein may prevent or reduce the infectivity of a viral infection by preventing internalization of a virus into a cell of the subject or by preventing internalization of a viral genome into a cell of the subject. In some embodiments, a composition provided herein may disrupt or prevent an interaction between a viral surface protein (e.g., a spike protein or an envelope protein) and a host receptor protein (e.g., an epithelial angiotensin converting enzyme (ACE) or a n epithelial furin enzyme). For example, an inhibitor (e.g., an TMPRSS inhibitor, an ACE2 inhibitor, or a furin inhibitor) may block internalization of a coronavirus into a cell of a subject by blocking or disrupting interactions between a coronavirus spike protein and a host receptor protein, as illustrated in FIG. 1. Administering an inhibitor (e.g., an TMPRSS inhibitor, an ACE2 inhibitor, or a furin inhibitor) to a subject at risk for a viral infection may reduce the risk of coronavirus infection in the subject.
[0040] In some embodiments, a composition of the present disclosure (e.g., a composition comprising glyceryl monooleate or polysorbate 80) may reduce the infectivity of a virus by disrupting the envelope of an envelope virus (e.g., a coronavirus, a bunyavirus, a filovirus, a flavivirus, a herpesvirus, a hepadnavirus, a paramyxovirus, a poxvirus, a rhabdovirus, or a retrovirus). In some embodiments, the methods and compositions provided herein may reduce the infectivity of a virus by disrupting an interaction between a viral surface protein (e.g., a spike protein or an envelope protein) and an activating host cell protein (e.g., a type II transmembrane serine protease). A composition to reduce the infectivity of a virus may comprise an enzyme inhibitor (e.g., an TMPRSS inhibitor, an ACE2 inhibitor, or a furin inhibitor). In some embodiments, the enzyme inhibitor may be an inhibitor of an epithelial angiotensin converting enzyme 2 (ACE2) or an inhibitor of a type II transmembrane serine protease (TMPRSS2). A composition to reduce the infectivity of a virus may comprise a bioadhesive (e.g., glyceryl monooleate) to disrupt the membrane of an envelope virus (e.g., a coronavirus, a bunyavirus, a filovirus, a flavivirus, a herpesvirus, a hepadnavirus, a paramyxovirus, a poxvirus, a rhabdovirus, or a retrovirus).
[0041] The methods and compositions disclosed herein may be used to treat, prevent, or reduce the infectivity of a viral infection (e.g., a respiratory viral infection). In some embodiments, the viral infection may be a coronavirus infection. The coronavirus may be SARS-CoV, SARS-CoV-2, MERS-CoV, HKU1, OC43, or 229E. The coronavirus may be a beta-coronavirus. Pathogens with long incubation periods, such as SARS-CoV-2 which has a median incubation period of about five days, may have high risk of transmission since many infected individuals may be unaware that they are infected. Additionally, carriers of coronavirus may frequently be asymptomatic or have mild symptoms, leading to unknowing contact between a viral host and other members of a population. A subject at risk for a coronavirus infection may come in contact with an asymptomatic carrier of the coronavirus infection, thereby unknowingly contracting the coronavirus infection. Methods and compositions are needed to prevent coronavirus infections in at-risk individuals (e.g., individuals who have come in contact with a carrier of a coronavirus or who may come in contact with a carrier of a coronavirus).
[0042] In some embodiments, the methods and compositions disclosed herein may treat or prevent an infection caused by a virus from one or more of arenaviridae (e.g., Pichinde virus, Lymphocytic Choriomeningitis Virus (LCMV), Lassa virus (causing Lassa fever) and Argentine hemorrhagic fever (AHF)), Paramyxoviridae (e.g., respiratory syncytial virus (RSV), measles virus (causing subacute sclerosing panencephalitis), mumps virus), herpesviridae (e.g., varicella-zoster (VZV), herpes simplex virus (HSV), human herpes virus- 6 (HHV-6), cytomegalovirus (CMV), and Epstein Barr virus (EBV)), orthomyxoviridae (e.g., influenza A and B virus), picornaviridae (enteroviruses (3 polioviruses (PV), 28 echoviruses (ECV), 23 group A and 6 group B coxsackieviruses (CVA and CBV, respectively), or Theiler's virus), or 4 numbered enteroviruses), poxviridae (e.g., smallpox (variola), cowpox virus (CV), camelpox, monkeypox, or vaccinia viruses), reoviridae (e.g., bluetongue virus, rotavirus, simian (SA11) rotavirus or Colorado tick fever virus (CTFV)), polyomaviridae (e.g., JC Virus (JCV, causing PML in immune compromised patients), BK Virus (BKV), or simian virus 40 (SV40)), filoviridae (e.g., Marburg virus, or Ebola virus), bunyavirales (e.g., hantavirus), rhabdoviridae (e.g., rabies), retroviridae (e.g., Human T-lymphotropic virus (HTLV, type I and II), or Human immunodeficiency virus (HIV, type I and II)), coronaviridae (e.g., coronavirus, or torovirus), adenoviridae, or iridoviridae. [0043] In some embodiments, the compositions, methods, or treatment regiments disclosed herein may treat or prevent a SARS-CoV-2 infection (e.g., COVID-19). A SARS-CoV-2 infection may depend on host cell ACE-2 and TMPRSS2 enzymes. In some embodiments, a SARS-CoV-2 infection may be blocked (e.g., prevented, treated, or slowed) by a protease inhibitor (e.g., a serpin). Cell entry of a coronavirus (e.g., SARS-CoV-2) may depend on binding of the viral spike (S) proteins to cellular receptors and on S protein priming by host cell proteases. For example, SARS-CoV-2 may use an ACE-2 receptor for entry and a serine protease TMPRSS2 for S protein priming. A composition of the present disclosure to treat or prevent coronavirus infection may comprise an inhibitor of TMPRSS2, an inhibitor of ACE- 2, or a combination thereof.
Compositions
[0044] A composition of the present disclosure may comprise one or more active agents. In some embodiments, an active agent may be an agent to prevent, treat, or reduce the infectivity of a viral infection. In some embodiments, treating a viral infection may comprise reducing the infectivity of the virus. In some embodiments, preventing a viral infection may comprise reducing the infectivity of the virus. A composition of the present disclosure may comprise an active agent to prevent a viral infection, an active agent to treat a viral infection, an active agent to reduce the infectivity of a viral infection, or a combination thereof. In some embodiments, an active agent may be a protease inhibitor (e.g., a serine protease inhibitor). Serine proteases may also be referred to herein as “serine endopeptidases.” A protease may be an enzyme that cleave peptide bonds in proteins. A serine protease (or a serine endopeptidase) may be an enzyme that cleave peptide bonds in proteins, in which serine serves as the nucleophilic amino acid at the (enzyme's) active site. Proteases, including serine proteases, may be found in both eukaryotes and prokaryotes. In some embodiments, a protease, such as a serine protease, may be categorized based on structure. For example, a serine protease may be a chymotrypsin-like (trypsin-like) protease, or a serine protein may be a subtili sin-like serine protease. A composition comprising a protease inhibitor may treat, prevent, or reduce the infectivity of a viral infection (e.g., a coronavirus infection) by inhibiting a protease (e.g., ACE-2, furin, or TMPRSS2) associated with the viral infection, thereby preventing or treating the viral infection.
[0045] In some embodiments, a serine protease inhibitor may be a serpin (e.g., a protein with serine protease inhibitor activity). Serpins are a superfamily of proteins with similar structures that have protease inhibition activity. Some serpins may act on (e.g., inhibit) chymotrypsin-like serine proteases. In some embodiments, a serpin may irreversibly inhibit a target protease by undergoing a large conformational change to disrupt the active site of the protease. In some embodiments, a protease inhibitor may inhibit a protease through a competitive mechanism comprising binding and blocking access to the protease active site. [0046] A composition of the present disclosure (e.g., a composition comprising an active agent) may inhibit a protease associated with a viral infection. For example, a composition may inhibit a TMPRSS2 serine protease, thereby treating, preventing, or reducing the infectivity of a SARS-CoV-2 infection (e.g., COVID-19). A composition of the present disclosure may inhibit an ACE protease (e.g., ACE-2), thereby preventing or reducing the infectivity of a SARS-CoV-2 infection.
[0047] In some embodiments, a composition of the present disclosure may comprise an active agent that disrupts an envelope of an envelope virus (e.g., a coronavirus, a bunyavirus, a filovirus, a flavivirus, a herpesvirus, a hepadnavirus, a paramyxovirus, a poxvirus, a rhabdovirus, or a retrovirus). For example, a composition may comprise a bioadhesive (e.g., glyceryl monooleate) that disrupts the viral envelope. In some embodiments, a composition may comprise one or more agents to disrupt a viral envelope (e.g., one or more of glyceryl monooleate or polysorbate 80).
TMPRSS2
[0048] The TMPRSS2 gene encodes a protein that belongs to the serine protease family. The encoded protein contains a type II transmembrane domain, a receptor class A domain, a scavenger receptor cysteine-rich domain and a protease domain. TMPRSS2 may be up- regulated by androgenic hormones in prostate cancer cells and down-regulated in androgen- independent prostate cancer tissue. The protease domain of TMPRSS2 may be cleaved and secreted into cell media after autocleavage. Expression of TMPRSS2 is elevated in the prostate and other androgen tissue as compared to other tissue types (FIG. 3). However, the biological function of this gene is unknown.
[0049] The cellular serine protease TMPRSS2 may prime the S-protein of a coronavirus (e.g., a SARS-CoV-2 coronavirus) for entry into a host cell. The compositions disclosed herein (e.g., a composition comprising a serine protease inhibitor) may block a coronavirus infection by preventing priming of the S-protein by TMPRSS2, thereby inhibiting entry of the coronavirus into the host cell. In some embodiments, the composition may prevent coronavirus infection of lung cells. [0050] A coronavirus infection (e.g., a SARS-CoV-2 infection) may be facilitated the endosomal cysteine proteases CatB/L, which may prime a viral S protein for entry into a host cell. In some embodiments, a viral S protein may be primed by TMPRSS2, CatB/L, or a combination thereof. In some embodiments, spread of the viral infection may be facilitated by TMPRSS2 activity. A composition of the present disclosure (e.g., a composition comprising a TMPRSS2 inhibitor, a CatB/L inhibitor, or a combination thereof) may prevent or treat a viral infection by inhibiting priming of the S protein by TMPRSS2, CatB/L, or a combination thereof, thereby inhibiting entry of the coronavirus into the host cell. A composition comprising a protease inhibitor (e.g., a serine protease inhibitor or a dual-action protease inhibitor) may inhibit priming of the S-protein. For example, a composition may comprise camostat mesylate (also referred to herein as “camostat” or “camostat mesylate”). Camostat may inhibit S-protein priming by TMPRSS2. Camostat may effective against viral infections (e.g., coronavirus infections). In some embodiments, furin-mediated pre-cleavage at the S1/S2 site in infected cells might promote subsequent TMPRSS2-dependent entry into target cells. In some embodiments, a composition to treat, prevent, or reduce the infectivity of a coronavirus may comprise a protease inhibitor that inhibits furin, thereby inhibiting furin- mediated pre-cleavage at the S1/S2 site. For example, a composition may comprise edetate disodium (also referred to herein as “disodium edetate,” “disodium EDTA,” “EDTA,” or “ethylenediaminetetraacetic acid”). In some embodiments, EDTA may function as a dual action inhibitor to furin and one or more additional enzymes. A composition comprising a TMPRSS2 inhibitor (e.g., camostat), a furin inhibitor, or a dual-action protease inhibitor may prevent, treat, reduce the infectivity of, or slow a coronavirus infection (e.g., a SARS-CoV infection, a SARS-CoV-2 infection, or a MERS-CoV infection). Camostat may inhibit TMPRSS2. Inhibition of TMPRSS2 may block, prevent, reduce the infectivity of, or treat infection by a coronavirus (e.g., SARS-CoV, Human coronavirus NL63, SARS-CoV-2, or MERS-CoV). For example, camostat may reduce the infection of Calu-3 lung cells by SARS- CoV-2, thereby preventing or treating COVID-19. EDTA may inhibit furin. Inhibition of furin may block, prevent, reduce the infectivity of, or treat infection by a coronavirus (e.g., SARS-CoV, Human coronavirus NL63, SARS-CoV-2, or MERS-CoV).
[0051] Serine proteases (e.g., TMPRSS2) that proteolytically cleave and activate the viral spike glycoproteins may facilitate virus-cell membrane fusions. Spike proteins may be synthesized and maintained in precursor intermediate folding states and proteolysis may permit the refolding and energy release required to create stable virus-cell linkages and membrane coalescence. TMPRSS2may facilitate human SARS coronavirus (SARS-CoV) infection via two independent mechanisms: proteolytic cleavage of ACE-2, which might promote viral uptake, and cleavage of coronavirus spike glycoprotein which activates the glycoprotein for cathepsin L-independent host cell entry. TMPRSS2may proteolytically cleave and activate the spike glycoproteins of human coronavirus 229E (HCoV-229E) and human coronavirus EMC (HCoV-EMC) and the fusion glycoproteins F0 of Sendai virus (SeV), human metapneumovirus (HMPV), human parainfluenza 1, 2, 3, 4a and 4b viruses (HPIV). TMPRSS2may facilitate spread and pathogenesis of influenza A virus (strains H1N1, H3N2 and H7N9); involved in proteolytic cleavage and activation of hemagglutinin (HA) protein which is essential for viral infectivity. Inhibiting serine proteases, such as a type II transmembrane serine protease (TMPRSS2), may reduce the infectivity of a coronavirus for a host cell by preventing activation of the viral spike glycoproteins. Thus, a composition comprising an inhibitor of TMPRSS2may treat or prevent a viral infection, for example an infection resulting from human coronavirus 229E (HCoV-229E), human coronavirus EMC (HCoV-EMC), the fusion glycoproteins F0 of Sendai virus (SeV), human metapneumovirus (HMPV), or human parainfluenza 1, 2, 3, 4a or 4b viruses (HPIV).
[0052] A composition of the present disclosure may comprise a TMPRSS2 inhibitor (e.g., camostat or argatroban). In some embodiments, a composition may comprise from about 1 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 30 mg/mL, from about 30 mg/mL to about 35 mg/mL, from about 35 mg/mL to about 40 mg/mL, from about 40 mg/mL to about 45 mg/mL, from about 45 mg/mL to about 50 mg/mL, from about 50 mg/mL to about 60 mg/mL, from about 60 mg/mL to about 70 mg/mL, from about 70 mg/mL to about 80 mg/mL, from about 80 mg/mL to about 90 mg/mL, from about 90 mg/mL to about 100 mg/mL, from about 100 mg/mL to about 110 mg/mL, from about 110 mg /mL to about 120 mg/mL, from about 120 mg/mL to about 130 mg/mL, from about 130 mg/mL to about 140 mg/mL, from about 140 mg/mL to about 150 mg/mL, from about 150 mg/mL to about 175 mg/mL, from about 175 mg/mL to about 200 mg/mL, from about 200 mg/mL to about 250 mg/mL, from about 250 mg/mL to about 300 mg/mL, from about 300 mg/mL to about 350 mg/mL, from about 350 mg/mL to about 400 mg/mL, from about 400 mg/mL to about 450 mg/mL, from about 450 mg/mL to about 500 mg/mL, or from about 500 mg/mL to about 1000 mg/mL of a TMPRSS2 inhibitor (e.g., camostat or argatroban).
[0053] In some embodiments, a composition comprising a TMPRSS2 inhibitor (e.g., camostat or argatroban) may be administered nasally, orally, by inhalation, or by any other administration method disclosed herein. A composition comprising a TMPRSS2 inhibitor (e.g., camostat or argatroban) may be administered 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times per day. A composition comprising a TMPRSS2 inhibitor (e.g., camostat or argatroban) may be administered every 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 days. In some embodiments, a composition comprising a TMPRSS2 inhibitor (e.g., camostat) may be administered for 1, 2, 3, 4, 5, 6, 7,
8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 25, or 30 days. A composition comprising a TMPRSS2 inhibitor (e.g., camostat or argatroban) may be administered for 2, 3, 4, 5, 6, 7, 8,
9, 10, 11, 12, 13, 14, or 15 weeks. A composition comprising a TMPRSS2 inhibitor (e.g., camostat or argatroban) may be formulated for nasal delivery.
[0054] The compositions described herein (e.g., compositions comprising a TMPRSS2 inhibitor) may be administered to a subject (e.g., a human subject) to treat or prevent a viral infection without substantial negative side effects on the subject.
ACE-2
[0055] ACE-2 (also referred to herein as “ACE2”) is a type I integral membrane protein and is a serine protease. It is a metallocarboxypeptidase. The active site domain of ACE-2 may be exposed to the extracellular surface of endothelial cells and the renal tubular epithelium. ACE-2 contains a 17 amino acid N-terminal signal sequence and a 22 amino acid hydrophobic transmembrane sequence near the C-terminus followed by a 43 amino acid cytoplasmic domain, which contains potential phosphorylation sites. The complete cDNA for human ACE-2 encodes a protein of 805 amino acids that exhibits 40% identity and 61% similarity to human ACE. Human The juxtamembrane, transmembrane and cytoplasmic domains of ACE-2 do not resemble ACE but share similarity with a 220 amino acid transmembrane glycoprotein termed collectrin, which is localized to the renal collecting ducts. Collectrin has no protease domain and its function is unknown. The homology of ACE-2 and ACE is particularly striking around the HEXXH zinc-binding motif which is identical in the two proteins. ACE-2 also contains eight cysteine residues, six of which are conserved in the N- and C-terminal domains of endothelial ACE, and has seven potential Af- linked glycosylation sites. The ACE-2 gene, located on chromosome Xp22, contains 18 exons and many of those resemble the corresponding exons in the ACE gene. ACE-2 may metabolize circulating peptides including angiotensin II, a potent vasoconstrictor and the product of angiotensin I cleavage by ACE. To this end, ACE-2 may counterbalance the effects of ACE within the renin-angiotensin system (RAS). Indeed, ACE2 has been implicated in the regulation of heart and renal function where it is proposed to control the levels of angiotensin II relative to its hypotensive metabolite, angiotensin. ACE-2 may play a unique role in the renin-angiotensin system and mediate cardiovascular and renal function. [0056] ACE-2 may serve as a functional receptor for a coronavirus (e.g., a SARS-CoV or a SARS-CoV-2). For example, ACE-2 may facilitate respiratory tract infection of a coronavirus (e.g., SARS-CoV, SARS-CoV-2, human respiratory coronavirus NL63, or MERS-CoV). ACE-2 may be expressed in airway epithelial cells, contributing to viral infection. In some embodiments, ACE-2, but not ACE, may facilitate the association between host cells and a coronavirus S protein. Tissue distribution of ACE-2 may be consistent with the pathology of SARS-CoV. ACE-2 may be abundantly expressed in the epithelia of the lung and the small intestine, possible entry sites for a coronavirus. Nasal epithelial cells, specifically goblet/secretory cells and ciliated cells, may display the high ACE-2 expression. The skewed expression of viral receptors/entry-associated proteins towards the upper airway may be correlated with enhanced transmissivity of a viral infection. Genes associated with ACE-2 airway epithelial expression may be innate immune-associated, antiviral genes, highly enriched in the nasal epithelial cells, contributing to viral infection of airway epithelial cells. [0057] A composition of the present disclosure may comprise an angiotensin-converting enzyme-related carboxypeptidase (ACE-2) inhibitor. An ACE-2 inhibitor may block an association between a host cell and a coronavirus S protein. Blocking the association between the host cell and the coronavirus S protein may reduce the infectivity of the coronavirus for the host cell, thereby treating or preventing the coronavirus infection. In some embodiments, the composition comprising the ACE-2 inhibitor may be administered nasally.
[0058] In some embodiments, an ACE2 inhibitor may comprise disodium edetate (EDTA). In some embodiments, an ACE-2 inhibitor may comprise a chemical structure of
[0059] In some embodiments, R1 may be Ph, cyclohexyl CFh, cyclohexyl (ϋ¼)2, 4-NO?Ph, 4-ClPh, 4-CF3OPh, 4-MePh, 2-MePh, 3-MePh, 3,4-diMePh, 3,5-diMePh, or 3,5-diClPh, wherein Ph is a phenyl group, and Me is a methyl group. In some embodiments, R2 may be Me, Ph, or CH(CH3)2, wherein Ph is a phenyl group, and Me is a methyl group. For example, an ACE-2 inhibitor may comprise an R1 group of 3,5-diClPh and an R2 group of CH(CH3)2. Examples of ACE-2 inhibitors that may be used in the compositions of this disclosure, and methods of synthesis, are provided in FIG. 4.
[0060] In some embodiments, an ACE-2 inhibitor may comprise a soluble human ACE-2.
The soluble ACE-2 may function as a decoy for binding of a coronavirus (e.g., a SARS-CoV- 2), thereby preventing or treating a coronavirus infection. In some embodiments, a composition of the present disclosure may comprise a clinical grade soluble ACE-2 (e.g., hrsACE2). For example, a soluble ACE-2 (e.g., hrsACE2) may inhibit SARS-CoV-2 infection of human blood vessel organoids and human kidney organoids. In some embodiments, a composition comprising hrsACE2 may prevent or treat a coronavirus infection by blocking an early stage of a coronavirus infection (e.g., a SARS-CoV-2 infection).
[0061] In some embodiments, a composition to treat or prevent a viral infection (e.g., a coronavirus infection) may comprise a peptide inhibitor of ACE-2. In some embodiments, a composition to treat or prevent a viral infection (e.g., a coronavirus infection) may comprise bacteriophage which display a peptide inhibitor of ACE-2. A peptide inhibitor of ACE-2 may have an equilibrium inhibition constant (Ki value) from about 0.38 to about 1.7 M. In some embodiments, a peptide inhibitor of ACE-2 may have a Ki value of less than about 0.14 M. In some embodiments, a peptide inhibitor of ACE-2 may have a Ki of about 2.8 nM. In some embodiments, a peptide inhibitor of ACE-2 may have a Ki of from about 1 nM to about 10 nM, from about 10 nM to about 100 nM, from about 100 nM to about 1 mM, from about 1 mM to about 10 pM, from about 10 pM to about 100 pM, from about 100 pM to about 1 mM, from about 1 mM to about 10 mM, from about 10 mM to about 100 mM, from about 100 mM to about 1 M, or from about 1 M to about 10 M. An ACE-2 inhibitor peptide may be a DX600 peptide.
[0062] An ACE-2 inhibitor peptide may exhibit mixed competitive and non-competitive type of inhibition. An ACE-2 inhibitor peptide may not be hydrolyzed by ACE-2. An ACE-2 inhibitor peptide may not inhibit ACE activity. An ACE-2 inhibitor peptide may be specific for ACE-2. An ACE-2 inhibitor peptide may inhibit ACE-2 activity toward its natural substrate angiotensin I. In some embodiments, library selection by phage display technology may be used to rapidly and efficiently identify specific peptide inhibitors of ACE-2. A peptide library may be constructed and selected against FLAG-tagged ACE2 target. In some embodiments, a composition may comprise a synthetic peptide inhibitor of ACE-2. In some embodiments, a composition may comprise a Ml 3 bacteriophage peptide inhibitor of ACE-2. The ACE-2 inhibitors (e.g., the peptide inhibitors of ACE-2) may be used in a nasal spray comprising a composition disclosed herein.
[0063] A composition of the present disclosure may comprise an ACE-2 inhibitor (e.g., a small molecule inhibitor provided in FIG. 4, a soluble ACE-2, an ACE-2 peptide inhibitor, a dual-action enzyme inhibitor, or a bacteriophage displaying an ACE-2 peptide inhibitor). In some embodiments, a composition may comprise from about 1 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 30 mg/mL, from about 30 mg/mL to about 35 mg/mL, from about 35 mg/mL to about 40 mg/mL, from about 40 mg/mL to about 45 mg/mL, from about 45 mg/mL to about 50 mg/mL, from about 50 mg/mL to about 60 mg/mL, from about 60 mg/mL to about 70 mg/mL, from about 70 mg/mL to about 80 mg/mL, from about 80 mg/mL to about 90 mg/mL, from about 90 mg/mL to about 100 mg/mL, from about 100 mg/mL to about 110 mg/mL, from about 110 mg/mL to about 120 mg/mL, from about 120 mg/mL to about 130 mg/mL, from about 130 mg/mL to about 140 mg/mL, from about 140 mg/mL to about 150 mg/mL, from about 150 mg/mL to about 175 mg/mL, from about 175 mg/mL to about 200 mg/mL, from about 200 mg/mL to about 250 mg/mL, from about 250 mg/mL to about 300 mg/mL, from about 300 mg/mL to about 350 mg/mL, from about 350 mg/mL to about 400 mg/mL, from about 400 mg/mL to about 450 mg/mL, from about 450 mg/mL to about 500 mg/mL, or from about 500 mg/mL to about 1000 mg/mL of an ACE-2 inhibitor. A composition comprising an ACE-2 inhibitor may treat, prevent, or reduce the infectivity of a coronavirus infection (e.g., a SARS-CoV-2 infection).
[0064] In some embodiments, a composition comprising an ACE-2 inhibitor may be administered nasally, orally, by inhalation, or by any other administration method disclosed herein. A composition comprising an ACE-2 inhibitor may be administered 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times per day. A composition comprising an ACE-2 inhibitor may be administered every 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 days. In some embodiments, a composition comprising an ACE-2 inhibitor may be administered for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 25, or 30 days. A composition comprising an ACE-2 inhibitor may be administered for 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 weeks. In some embodiments, a composition comprising an ACE-2 inhibitor may be formulated for nasal delivery.
[0065] A composition of the present disclosure may comprise a dual-action enzyme inhibitor (e.g., EDTA) that inhibits ACE-2 and one or more additional enzymes (e.g., furin). For example, a dual action enzyme inhibitor may inhibit ACE-2 and furin. In some embodiments, a composition may comprise from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, or from about 9 mg/mL to about 10 mg/mL of a dual-action enzyme inhibitor (e.g., EDTA). A composition comprising a dual-action protease inhibitor may treat, prevent, or reduce the infectivity of a coronavirus infection (e.g., a SARS-CoV-2 infection).
[0066] In some embodiments, a composition comprising a dual-action enzyme inhibitor may be administered nasally, orally, by inhalation, or by any other administration method disclosed herein. A composition comprising a dual-action enzyme inhibitor may be administered 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times per day. A composition comprising a dual action enzyme inhibitor may be administered every 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 days. In some embodiments, a composition comprising a dual-action enzyme inhibitor may be administered for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 25, or 30 days. A composition comprising a dual-action enzyme inhibitor may be administered for 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 weeks. In some embodiments, a composition comprising a dual-action enzyme inhibitor may be formulated for nasal delivery.
Serine Protease Inhibitor, Dual-Action Enzyme Inhibitor, and Cysteine Protease Inhibitor Treatments
[0067] A composition to treat or prevent a viral infection (e.g., a coronavirus infection) may comprise a serine protease inhibitor, a cysteine protease inhibitor, a dual-action enzyme inhibitor, or a combination thereof. For example, a composition may comprise an inhibitor of the type II transmembrane protease TMPRSS2. TMPRSS2 may activate the spike (S) protein of a coronavirus (e.g., SARS-CoV, SARS-CoV-2, or MERS-CoV) on the cell surface following receptor binding during viral entry into cells. In the absence of TMPRSS2 (e.g., in the presence of a TMPRSS2 inhibitor), a coronavirus may enter a cell via an endosomal pathway in which cathepsin L may play an important role (e.g., the activation of spike protein fusogenicity). In some embodiments, a serine protease inhibitor (e.g., camostat) may block an infection by a coronavirus (e.g., SARS-CoV, human coronavirus NL63 (HCoV-NL63), MERS-CoV, or SARS-CoV-2) in cells expressing the receptor angiotensin-converting enzyme 2 (ACE2) and TMPRSS2. In some embodiments, a composition may comprise a serine protease inhibitor (e.g., camostat) and a cathepsin inhibitor (e.g., (23,25)-trans- epoxysuccinyl-L-leucylamindo-3-methylbutane ethyl ester (EST)). A cathepsin inhibitor may comprise (23, 25 )/ra//.s-epoxysuccinyl -1-1 eucyl ami ndo-3 -methyl butane ethyl ester, balicatib, calpeptin, /V-acetyl -L-l eucyl -L-l eucyl - L-m ethi onal , N-[[(2S,3S)-3-[(Propylamino)carbonyl]-
2-oxiranyl]carbonyl]-L-isoleucyl-L-proline; (2S,3S)-3-[[[(lS)-l-[[[4- (Aminoiminomethyl)amino]butyl]amino]carbonyl]-3-methylbutyl]amino]carbonyl]-2- oxiranecarboxylic acid; (2S,3S)-3-[[[(lS)-3-Methyl-l-[[(3- methylbutyl)amino]carbonyl]butyl]amino]carbonyl]-2-oxiranecarboxylic acid ethyl ester; N- [l-[[(Cyanomethyl)amino]carbonyl]cyclohexyl]-4-[2-(4-methyl-l-piperazinyl)-4- thiazolyljbenzamide; N-[N-[(Phenylmethoxy)carbonyl]-L-valyl]-phenylalaninal; or N-[(l, 1- Dimethylethoxy)carbonyl]-L-tryptophan-2-[[[2-[(2-ethylphenyl)amino]-2- oxoethyl]thio]carbonyl]hydrazide. A serine protease inhibitor may comprise camostat, nafamostat, gabexate, sivelestat, rivaroxaban, telaprevir, ulinastatin, or simeprevir. Simultaneous treatment of the cells with a serine protease inhibitor and a cathepsin inhibitor, may prevent both cell entry and the multistep growth of a coronavirus infection.
[0068] A composition of the present disclosure may comprise a serine protease inhibitor (e.g., camostat) and a cathepsin inhibitor (e.g., (23,25)-trans-epoxysuccinyl-L-leucylamindo-
3-methylbutane ethyl ester (EST)). In some embodiments, a composition may comprise from about 1 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 30 mg/mL, from about 30 mg/mL to about 35 mg/mL, from about 35 mg/mL to about 40 mg/mL, from about 40 mg/mL to about 45 mg/mL, from about 45 mg/mL to about 50 mg/mL, from about 50 mg/mL to about 60 mg/mL, from about 60 mg/mL to about 70 mg/mL, from about 70 mg/mL to about 80 mg/mL, from about 80 mg/mL to about 90 mg/mL, from about 90 mg/mL to about 100 mg/mL, from about 100 mg/mL to about 110 mg/mL, from about 110 mg/mL to about 120 mg/mL, from about 120 mg/mL to about 130 mg/mL, from about 130 mg/mL to about 140 mg/mL, from about 140 mg/mL to about 150 mg/mL, from about 150 mg/mL to about 175 mg/mL, from about 175 mg/mL to about 200 mg/mL, from about 200 mg/mL to about 250 mg/mL, from about 250 mg/mL to about 300 mg/mL, from about 300 mg/mL to about 350 mg/mL, from about 350 mg/mL to about 400 mg/mL, from about 400 mg/mL to about 450 mg/mL, from about 450 mg/mL to about 500 mg/mL, or from about 500 mg/mL to about 1000 mg/mL of a serine protease inhibitor (e.g., camostat). In some embodiments, a composition may comprise from about 1 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 30 mg/mL, from about 30 mg/mL to about 35 mg/mL, from about 35 mg/mL to about 40 mg/mL, from about 40 mg/mL to about 45 mg/mL, from about 45 mg/mL to about 50 mg/mL, from about 50 mg/mL to about 60 mg/mL, from about 60 mg/mL to about 70 mg/mL, from about 70 mg/mL to about 80 mg/mL, from about 80 mg/mL to about 90 mg/mL, from about 90 mg/mL to about 100 mg/mL, from about 100 mg/mL to about 110 mg/mL, from about 110 mg/mL to about 120 mg/mL, from about 120 mg/mL to about 130 mg/mL, from about 130 mg/mL to about 140 mg/mL, from about 140 mg/mL to about 150 mg/mL, from about 150 mg/mL to about 175 mg/mL, from about 175 mg/mL to about 200 mg/mL, from about 200 mg/mL to about 250 mg/mL, from about 250 mg/mL to about 300 mg/mL, from about 300 mg/mL to about 350 mg/mL, from about 350 mg/mL to about 400 mg/mL, from about 400 mg/mL to about 450 mg/mL, from about 450 mg/mL to about 500 mg/mL, or from about 500 mg/mL to about 1000 mg/mL of a cathepsin inhibitor (e.g., (23,25)-trans-epoxysuccinyl-L-leucylamindo-3-methylbutane ethyl ester (EST)).
[0069] In some embodiments, a composition comprising a serine protease inhibitor (e.g., camostat) and a cathepsin inhibitor (e.g., (23,25)-trans-epoxysuccinyl-L-leucylamindo-3- methylbutane ethyl ester (EST)) may be administered nasally, orally, by inhalation, or by any other administration method disclosed herein. A composition comprising a serine protease inhibitor (e.g., camostat) and a cathepsin inhibitor (e.g., (23,25)-trans-epoxysuccinyl-L- leucylamindo-3-methylbutane ethyl ester (EST)) may be administered 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times per day. A composition comprising a serine protease inhibitor and a cathepsin inhibitor (e.g., (23,25)-trans-epoxysuccinyl-L-leucylamindo-3-methylbutane ethyl ester (EST)) may be administered every 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 days. In some embodiments, a composition comprising a serine protease inhibitor and a cathepsin inhibitor (e.g., (23,25)- trans-epoxysuccinyl-L-leucylamindo-3-methylbutane ethyl ester (EST)) may be administered for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 25, or 30 days. A composition comprising a serine protease inhibitor and a cathepsin inhibitor (e.g., (23,25)- trans-epoxysuccinyl-L-leucylamindo-3-methylbutane ethyl ester (EST)) may be administered for 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 weeks. A composition comprising a serine protease inhibitor and a cathepsin inhibitor may be formulated for nasal delivery. Fibr oblast Growth Factor (FGF)-23 , a-Klotho and Furin Protease
[0070] A composition of the present disclosure may comprise an inhibitor of one or more of Fibroblast Growth Factor (FGF)-23, a-Klotho, Furin Protease, or a combination thereof. Furin, also called as paired basic amino acid cleavage enzyme (PACE), is a product of the gene that resides upstream of an oncogene, FES, and hence given the name (furin, F- upstream region). It is a subtilisin-like apo -/ pro-protein convertase, endo-peptidase in nature, that requires calcium ion and in which a serine residue participates in catalytic activity. Furin, a serine protease, is enriched in trans-golgi network, where it cleaves other proteins into their mature active forms. Although ubiquitously expressed at very low levels, furin is found in highest concentrations in liver and kidney, whereas brain, spleen and thymus possess much lower levels. Furin family of proteins exist as pro-enzymes in which *10 kDa heterogeneous amino-terminal, called the proregion, is cleaved prior to its activation. This is followed by a highly conserved *55 kDa catalytic domain and a C-terminal tail that has variable length as well as amino acid sequence. Furin is the only pro-convertase that possesses a trans membrane domain; it cleaves the target proteins down-stream of basic amino acid consensus sequence of ‘RX(R/K)R\ While the acidic amino acid sequence 771 CPSDSEEDEG 780 appears to be responsible for its trans-Golgi-network localization, the second domain, 759 YKGL 762, directs the internalization from cell surface. It is the phosphorylation of 775Serine residue that modulates intracellular routing. The activity of the furin protease may regulate the half-life of FGF-23; the latter in turn is responsible, after proper docking on its receptor (FGF-R), made possible by the participation of a-klotho, for the regulation of renal a-1 hydroxylase of vitamin D2 and repression of Na+/Pi-2a and 2c transporters. In some embodiments, a composition to treat, prevent, or reduce the infectivity of a coronavirus may comprise a protease inhibitor that inhibits furin, thereby inhibiting furin-mediated pre cleavage at the S1/S2 site of a viral spike protein and inhibiting entry of the virus into the host cell. Inhibiting entry of the virus into the host cell may reduce the infectivity of the virus, thereby treating or preventing the viral infection. In some embodiments, furin inhibitor may comprise disodium edetate, hexa-D-arginine, citrate, or combinations thereof. In some embodiments, a furin inhibitor may be a dual-action enzyme inhibitor. For example, a composition may comprise edetate disodium (EDTA). In some embodiments, EDTA may function as a dual-action inhibitor to furin and one or more additional enzymes (e.g., ACE2). [0071] A composition to treat or prevent a viral infection may comprise heparin. In some embodiments, heparin may inhibit furin cleavage of fibroblast growth factor. In some embodiments, heparin may inhibit activation of latent transforming growth factor-b 1. Heparin may not interfere with the receptor binding of TGF-bI, but may inhibit furin-like proprotein convertase-mediated activation of platelet LTGF-bI. Heparin may not inhibit the activity of furin-like proprotein convertase. In addition, heparin may inhibit acid activations of recombinant small LTGF-bI, platelet LTGF-bI and LTGF-b I s secreted in the supernatant of cultured cells. A composition of the present disclosure may comprise low-molecular- weight heparins, including dalteparin, enoxaparin and nadroparin, which may inhibit furin- like proprotein convertase-mediated or acid activation of platelet LTGF-bI. Heparin may inhibit LTGF-bI activation, possibly by binding simultaneously to TGF-bI and LAP.
Heparin may treat or prevent a viral infection through inhibition of LTGF-bI activation. [0072] A composition may comprise from 500 IU to 1000 IU, from 1000 IU to 1500 IU, from 1500 IU to 2000 IU, from 2000 IU to 2500 IU, from 2500 IU to 3000 IU, from 3000 IU to 3500 IU, from 3500 IU to 4000 IU, from 4000 IU to 4500 IU, from 4500 IU to 5000 IU, from 5000 IU to 5500 IU, from 5500 IU to 6000 IU, from 6000 IU to 6500 IU, from 6500 IU to 7000 IU, from 7000 IU to 7500 IU, from 7500 IU to 8000 IU, from 8000 IU to 8500 IU, from 8500 IU to 9000 IU, from 9000 IU to 9500 IU, from 9500 IU to 10000 IU, from 10000 IU to 20000 IU, from 20000 IU to 30000 IU, from 30000 IU to 40000 IU, from 40000 IU to 50000 IU, from 50000 IU to 60000 IU, from 60000 IU to 70000 IU, from 70000 IU to 80000 IU, from 80000 IU to 90000 IU, from 90000 IU to 100000 IU, from 100000 IU to 110000 IU, from 110000 IU to 120000 IU, from 120000 IU to 130000 IU, from 130000 IU to 140000 IU, from 140000 IU to 150000 IU, from 150000 IU to 160000 IU, from 160000 IU to 170000 IU, from 170000 IU to 180000 IU, from 180000 IU to 190000 IU, or from 190000 IU to 200000 IU of heparin. In some embodiments, from 500 IU to 1000 IU, from 1000 IU to 1500 IU, from 1500 IU to 2000 IU, from 2000 IU to 2500 IU, from 2500 IU to 3000 IU, from 3000 IU to 3500 IU, from 3500 IU to 4000 IU, from 4000 IU to 4500 IU, from 4500 IU to 5000 IU, from 5000 IU to 5500 IU, from 5500 IU to 6000 IU, from 6000 IU to 6500 IU, from 6500 IU to 7000 IU, from 7000 IU to 7500 IU, from 7500 IU to 8000 IU, from 8000 IU to 8500 IU, from 8500 IU to 9000 IU, from 9000 IU to 9500 IU, from 9500 IU to 10000 IU, from 10000 IU to 20000 IU, from 20000 IU to 30000 IU, from 30000 IU to 40000 IU, from 40000 IU to 50000 IU, from 50000 IU to 60000 IU, from 60000 IU to 70000 IU, from 70000 IU to 80000 IU, from 80000 IU to 90000 IU, from 90000 IU to 100000 IU, from 100000 IU to 110000 IU, from 110000 IU to 120000 IU, from 120000 IU to 130000 IU, from 130000 IU to 140000 IU, from 140000 IU to 150000 IU, from 150000 IU to 160000 IU, from 160000 IU to 170000 IU, from 170000 IU to 180000 IU, from 180000 IU to 190000 IU, or from 190000 IU to 200000 IU of heparin may be administered to a subject per day. The terms “international standard units,” “IU,” and “units” are used interchangeably herein to refer to an amount of an active compound.
[0073] A composition of the present disclosure may comprise a furin inhibitor peptide. For example, a composition to treat or prevent a viral infection may comprise a synthetic cyclic peptide inhibitor of furin. Examples of synthetic cyclic peptide inhibitors of furin are provided in TABLE 1. A composition to treat or prevent a viral infection may comprise a peptide inhibitor provided in TABLE 1.
TABLE 1 - Ki values of synthetic cyclic peptide inhibitors of furin
[0074] A composition may comprise from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, from about 9 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 11 mg/mL, from about 11 mg/mL to about 12 mg/mL, from about 12 mg/mL to about 13 mg/mL, from about 13 mg/mL to about 14 mg/mL, from about 14 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 16 mg/mL, from about 16 mg/mL to about 17 mg/mL, from about 17 mg/mL to about 18 mg/mL, from about 18 mg/mL to about 19 mg/mL, from about 19 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 21 mg/mL, from about 21 mg/mL to about 22 mg/mL, from about 22 mg/mL to about 23 mg/mL, from about 23 mg/mL to about 24 mg/mL, from about 24 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 26 mg/mL, from about 26 mg/mL to about 27 mg/mL, from about 27 mg/mL to about 28 mg/mL, from about 28 mg/mL to about 29 mg/mL, or from about 29 mg/mL to about 30 mg/mL of a synthetic cyclic peptide inhibitor of furin. A composition may comprise from about 1 mg/mL to about 20 mg/mL, from about 1 mg/mL to about 10 mg/mL, or from about 2 mg/mL to about 8 mg/mL of a synthetic cyclic peptide inhibitor of furin. A composition may comprise at least about 2 mg/mL, at least about 4 mg/mL, or at least about 5 mg/mL of a synthetic cyclic peptide inhibitor of furin. In some embodiments, a furin inhibitor may comprise Hexa-D-arginine. Hexa-D-arginine may be a potent and specific a furin inhibitor, for example hexa-L-arginine may have Ki values of 106 nM, 580 nM, and 13.2 mM for furin, PACE4, and PCI, respectively. A composition to treat or prevent a viral infection may comprise Hexa-D-arginine.
Dual-Action Enzyme Inhibitors
[0075] A composition for the treatment or prevention of a coronavirus infection may comprise one or more enzyme inhibitors that function as dual-action enzyme inhibitors. A dual-action enzyme inhibitor may be capable of inhibiting two-or more enzymes associated with a coronavirus infection. For example, a dual-action inhibitor may prevent a coronavirus infection (e.g., a SARS-CoV2 infection) by inhibiting ACE-2 and furin. In some embodiments, a dual-action inhibitor may inhibit one or more calcium-dependent enzymes, one or more zinc-dependent enzymes, or one or more zinc-dependent enzymes and one or more calcium-dependent enzymes. For example, a dual-action enzyme inhibitor may inhibit the zinc-dependent enzyme, ACE-2 and the calcium-dependent enzyme, furin. A dual-action enzyme inhibitor may function by chelating zinc, thereby inhibiting zinc-dependent enzyme (e.g., furin), by chelating calcium, thereby inhibiting calcium-dependent enzymes (e.g., furin), or by chelating both calcium and zinc, thereby inhibiting calcium-dependent enzymes and zinc-dependent enzymes. Examples of dual-action enzyme inhibitors include EDTA, EDTA-magnesium, and EGTA. In some embodiments, EDTA may function as a dual-action inhibitor by inhibiting ACE-2 and furin. In some embodiments, EDTA-magnesium may function as a dual-action inhibitor by inhibiting ACE-2 and furin. In some embodiments, EGTA may function as a dual-action inhibitor by inhibiting ACE-2 and furin.
[0076] A composition of the present disclosure may comprise Disodium Edetate (EDTA). A composition may comprise from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, or from about 9 mg/mL to about 10 mg/mL of Disodium Edetate. A composition may comprise from about 1 mM to about 3 mM, from about 2 mM to about 4 mM, from about 3 mM to about 5 mM, from about 4 mM to about 6 mM, from about 5 mM to about 10 mM, or from about 1 mM to about 15 mM of Disodium Edetate. A composition may comprise from about 0.001% (w/w) to about 0.005% (w/w), from about 0.005% (w/w) to about 0.01% (w/w), from about 0.01% (w/w) to about 0.05% (w/w), from about 0.05% (w/w) to about 0.10% (w/w), from about 0.10% (w/w) to about 0.20% (w/w), from about 0.20% (w/w) to about 0.30% (w/w), from about 0.30% (w/w) to about 0.40% (w/w), from about 0.40% (w/w) to about 0.50% (w/w), from about 0.50% (w/w) to about 0.60% (w/w), from about 0.60% (w/w) to about 0.70% (w/w), from about 0.70% (w/w) to about 0.80% (w/w), from about 0.80% (w/w) to about 0.90% (w/w), from about 0.90% (w/w) to about 1.00% (w/w), from about 1% (w/w) to about 2% (w/w), from about 2% (w/w) to about 3% (w/w), from about 3% (w/w) to about 4% (w/w), from about 4% (w/w) to about 5% (w/w), from about 5% (w/w) to about 6% (w/w), from about 6% (w/w) to about 7% (w/w), from about 7% (w/w) to about 8% (w/w), from about 8% (w/w) to about 9% (w/w), from about 9% (w/w) to about 10% (w/w), from about 10% (w/w) to about 15% (w/w), from about 15% (w/w) to about 20% (w/w), from about 20% (w/w) to about 25% (w/w), or from about 25% (w/w) to about 30% (w/w) of Di sodium Edetate. Disodium Edetate may be added as a chelator of Zn to inhibit ACE-2. Disodium Edetate may be added as a chelator of calcium to inhibit furin. A composition may be administered at a concentration of about 2.0 mg/mL Disodium Edetate. Disodium Edetate may have a molecular weight of about 336.2. A composition comprising 2 mg/mL of Disodium Edetate may be is 6 mM. Furin and ACE-2 may be inhibited by 2 mM EDTA, 5 mM EDTA, or by 10 mM EDTA. In some embodiments, EDTA may be EDTA-magnesium. [0077] A composition of the present disclosure may comprise egtazic acid (EGTA). A composition may comprise from about 0.1 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, or from about 9 mg/mL to about 10 mg/mL of EGTA. A composition may comprise from about 1 mM to about 3 mM, from about 2 mM to about 4 mM, from about 3 mM to about 5 mM, from about 4 mM to about 6 mM, from about 5 mM to about 10 mM, or from about 1 mM to about 15 mM of EGTA. A composition may comprise from about 0.001% (w/w) to about 0.005% (w/w), from about 0.005% (w/w) to about 0.01% (w/w), from about 0.01% (w/w) to about 0.05% (w/w), from about 0.05% (w/w) to about 0.10% (w/w), from about 0.10% (w/w) to about 0.20% (w/w), from about 0.20% (w/w) to about 0.30% (w/w), from about 0.30% (w/w) to about 0.40% (w/w), from about 0.40% (w/w) to about 0.50% (w/w), from about 0.50% (w/w) to about 0.60% (w/w), from about 0.60% (w/w) to about 0.70% (w/w), from about 0.70% (w/w) to about 0.80% (w/w), from about 0.80% (w/w) to about 0.90% (w/w), from about 0.90% (w/w) to about 1.00% (w/w), from about 1% (w/w) to about 2% (w/w), from about 2% (w/w) to about 3% (w/w), from about 3% (w/w) to about 4% (w/w), from about 4% (w/w) to about 5% (w/w), from about 5% (w/w) to about 6% (w/w), from about 6% (w/w) to about 7% (w/w), from about 7% (w/w) to about 8% (w/w), from about 8% (w/w) to about 9% (w/w), from about 9% (w/w) to about 10% (w/w), from about 10% (w/w) to about 15% (w/w), from about 15% (w/w) to about 20% (w/w), from about 20% (w/w) to about 25% (w/w), or from about 25% (w/w) to about 30% (w/w) of EGTA. EGTA may be added as a chelator of Zn to inhibit ACE-2. EGTA may be added as a chelator of calcium to inhibit furin. A composition may be administered at a concentration of about 2.0 mg/mL EGTA. Disodium Edetate may have a molecular weight of about 380.35 g/mol. A composition comprising 2 mg/mL of EGTA may be is 6 mM. Furin and ACE-2 may be inhibited by 2 mM EGTA, 5 mM EGTA, or by 10 mM EGTA.
Bacteriostatic Agents
[0078] In some embodiments, the compositions of the present disclosure may comprise a bacteriostatic agent. For example, a bacteriostatic agent may be an agent provided in TABLE 2
TABLE 2 - Bacteriostatic Agents [0079] A bacteriostatic agent may be Benzoic Acid (sodium benzoate), Benzalkonium chloride, Thiomersal, Chlorobutanol, Chlobutol, Potassium Sorbate, or Methyl Paraben. In some embodiments, a composition comprising a bacteriostatic agent may be formulated for nasal delivery.
[0080] A composition may comprise from about 0.1% (w/w) to about 0.2% (w/w) benzoic acid (sodium benzoate). A composition may comprise up to about 0.1% (w/w) benzalkonium chloride. A composition may comprise from about 0.0003% (w/w) to about 0.01% (w/w) thiomersal. A composition may comprise about 0.5% (w/w) chlorobutanol. A composition may comprise about 0.25% (w/w) chlobutol. A composition may comprise from about 0.1% (w/w) to about 0.2% (w/w) potassium sorbate. A composition may comprise from about 0.1% (w/w) to about 0.25% (w/w) methyl paraben.
[0081] A composition may comprise from about 0.005 % (w/w) to about 0.01 % (w/w), from about 0.01 % (w/w) to about 0.02 % (w/w), from about 0.02 % (w/w) to about 0.05 % (w/w), from about 0.05 % (w/w) to about 0.1 % (w/w), from about 0.1 % (w/w) to about 0.5 % (w/w), from about 0.5 % (w/w) to about 1 % (w/w), from about 1 % (w/w) to about 2 % (w/w), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/w) to about 5 % (w/w), from about 5 % (w/w) to about 6 % (w/w), from about 6 % (w/w) to about 7 % (w/w), from about 7 % (w/w) to about 8 % (w/w), from about 8 % (w/w) to about 9 % (w/w), from about 9 % (w/w) to about 10 % (w/w), from about 10 % (w/w) to about 11 % (w/w), from about 11 % (w/w) to about 12 % (w/w), from about 12 %
(w/w) to about 13 % (w/w), from about 13 % (w/w) to about 14 % (w/w), from about 14 %
(w/w) to about 15 % (w/w), from about 15 % (w/w) to about 16 % (w/w), from about 16 %
(w/w) to about 17 % (w/w), from about 17 % (w/w) to about 18 % (w/w), from about 18 %
(w/w) to about 19 % (w/w), or from about 19 % (w/w) to about 20 % (w/w) benzoic acid (sodium benzoate).
[0082] A composition may comprise from about 0.005 % (w/w) to about 0.01 % (w/w), from about 0.01 % (w/w) to about 0.02 % (w/w), from about 0.02 % (w/w) to about 0.05 % (w/w), from about 0.05 % (w/w) to about 0.1 % (w/w), from about 0.1 % (w/w) to about 0.5 % (w/w), from about 0.5 % (w/w) to about 1 % (w/w), from about 1 % (w/w) to about 2 % (w/w), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/w) to about 5 % (w/w), from about 5 % (w/w) to about 6 % (w/w), from about 6 % (w/w) to about 7 % (w/w), from about 7 % (w/w) to about 8 % (w/w), from about 8 % (w/w) to about 9 % (w/w), from about 9 % (w/w) to about 10 % (w/w), from about 10 % (w/w) to about 11 % (w/w), from about 11 % (w/w) to about 12 % (w/w), from about 12 % (w/w) to about 13 % (w/w), from about 13 % (w/w) to about 14 % (w/w), from about 14 %
(w/w) to about 15 % (w/w), from about 15 % (w/w) to about 16 % (w/w), from about 16 %
(w/w) to about 17 % (w/w), from about 17 % (w/w) to about 18 % (w/w), from about 18 %
(w/w) to about 19 % (w/w), or from about 19 % (w/w) to about 20 % (w/w) benzalkonium chloride.
[0083] A composition may comprise from about 0.005 % (w/w) to about 0.01 % (w/w), from about 0.01 % (w/w) to about 0.02 % (w/w), from about 0.02 % (w/w) to about 0.05 % (w/w), from about 0.05 % (w/w) to about 0.1 % (w/w), from about 0.1 % (w/w) to about 0.5 % (w/w), from about 0.5 % (w/w) to about 1 % (w/w), from about 1 % (w/w) to about 2 % (w/w), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/w) to about 5 % (w/w), from about 5 % (w/w) to about 6 % (w/w), from about 6 % (w/w) to about 7 % (w/w), from about 7 % (w/w) to about 8 % (w/w), from about 8 % (w/w) to about 9 % (w/w), from about 9 % (w/w) to about 10 % (w/w), from about 10 % (w/w) to about 11 % (w/w), from about 11 % (w/w) to about 12 % (w/w), from about 12 %
(w/w) to about 13 % (w/w), from about 13 % (w/w) to about 14 % (w/w), from about 14 %
(w/w) to about 15 % (w/w), from about 15 % (w/w) to about 16 % (w/w), from about 16 %
(w/w) to about 17 % (w/w), from about 17 % (w/w) to about 18 % (w/w), from about 18 %
(w/w) to about 19 % (w/w), or from about 19 % (w/w) to about 20 % (w/w) thiomersal. [0084] A composition may comprise from about 0.005 % (w/w) to about 0.01 % (w/w), from about 0.01 % (w/w) to about 0.02 % (w/w), from about 0.02 % (w/w) to about 0.05 % (w/w), from about 0.05 % (w/w) to about 0.1 % (w/w), from about 0.1 % (w/w) to about 0.5 % (w/w), from about 0.5 % (w/w) to about 1 % (w/w), from about 1 % (w/w) to about 2 % (w/w), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/w) to about 5 % (w/w), from about 5 % (w/w) to about 6 % (w/w), from about 6 % (w/w) to about 7 % (w/w), from about 7 % (w/w) to about 8 % (w/w), from about 8 % (w/w) to about 9 % (w/w), from about 9 % (w/w) to about 10 % (w/w), from about 10 % (w/w) to about 11 % (w/w), from about 11 % (w/w) to about 12 % (w/w), from about 12 %
(w/w) to about 13 % (w/w), from about 13 % (w/w) to about 14 % (w/w), from about 14 %
(w/w) to about 15 % (w/w), from about 15 % (w/w) to about 16 % (w/w), from about 16 %
(w/w) to about 17 % (w/w), from about 17 % (w/w) to about 18 % (w/w), from about 18 %
(w/w) to about 19 % (w/w), or from about 19 % (w/w) to about 20 % (w/w) chlorobutanol. [0085] A composition may comprise from about 0.005 % (w/w) to about 0.01 % (w/w), from about 0.01 % (w/w) to about 0.02 % (w/w), from about 0.02 % (w/w) to about 0.05 % (w/w), from about 0.05 % (w/w) to about 0.1 % (w/w), from about 0.1 % (w/w) to about 0.5 % (w/w), from about 0.5 % (w/w) to about 1 % (w/w), from about 1 % (w/w) to about 2 % (w/w), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/w) to about 5 % (w/w), from about 5 % (w/w) to about 6 % (w/w), from about 6 % (w/w) to about 7 % (w/w), from about 7 % (w/w) to about 8 % (w/w), from about 8 % (w/w) to about 9 % (w/w), from about 9 % (w/w) to about 10 % (w/w), from about 10 % (w/w) to about 11 % (w/w), from about 11 % (w/w) to about 12 % (w/w), from about 12 %
(w/w) to about 13 % (w/w), from about 13 % (w/w) to about 14 % (w/w), from about 14 %
(w/w) to about 15 % (w/w), from about 15 % (w/w) to about 16 % (w/w), from about 16 %
(w/w) to about 17 % (w/w), from about 17 % (w/w) to about 18 % (w/w), from about 18 %
(w/w) to about 19 % (w/w), or from about 19 % (w/w) to about 20 % (w/w) chlobutol.
[0086] A composition may comprise from about 0.005 % (w/w) to about 0.01 % (w/w), from about 0.01 % (w/w) to about 0.02 % (w/w), from about 0.02 % (w/w) to about 0.05 % (w/w), from about 0.05 % (w/w) to about 0.1 % (w/w), from about 0.1 % (w/w) to about 0.5 % (w/w), from about 0.5 % (w/w) to about 1 % (w/w), from about 1 % (w/w) to about 2 % (w/w), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/w) to about 5 % (w/w), from about 5 % (w/w) to about 6 % (w/w), from about 6 % (w/w) to about 7 % (w/w), from about 7 % (w/w) to about 8 % (w/w), from about 8 % (w/w) to about 9 % (w/w), from about 9 % (w/w) to about 10 % (w/w), from about 10 % (w/w) to about 11 % (w/w), from about 11 % (w/w) to about 12 % (w/w), from about 12 %
(w/w) to about 13 % (w/w), from about 13 % (w/w) to about 14 % (w/w), from about 14 %
(w/w) to about 15 % (w/w), from about 15 % (w/w) to about 16 % (w/w), from about 16 %
(w/w) to about 17 % (w/w), from about 17 % (w/w) to about 18 % (w/w), from about 18 %
(w/w) to about 19 % (w/w), or from about 19 % (w/w) to about 20 % (w/w) potassium sorb ate.
[0087] A composition may comprise from about 0.005 % (w/w) to about 0.01 % (w/w), from about 0.01 % (w/w) to about 0.02 % (w/w), from about 0.02 % (w/w) to about 0.05 % (w/w), from about 0.05 % (w/w) to about 0.1 % (w/w), from about 0.1 % (w/w) to about 0.5 % (w/w), from about 0.5 % (w/w) to about 1 % (w/w), from about 1 % (w/w) to about 2 % (w/w), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/w) to about 5 % (w/w), from about 5 % (w/w) to about 6 % (w/w), from about 6 % (w/w) to about 7 % (w/w), from about 7 % (w/w) to about 8 % (w/w), from about 8 % (w/w) to about 9 % (w/w), from about 9 % (w/w) to about 10 % (w/w), from about 10 % (w/w) to about 11 % (w/w), from about 11 % (w/w) to about 12 % (w/w), from about 12 %
(w/w) to about 13 % (w/w), from about 13 % (w/w) to about 14 % (w/w), from about 14 % (w/w) to about 15 % (w/w), from about 15 % (w/w) to about 16 % (w/w), from about 16 % (w/w) to about 17 % (w/w), from about 17 % (w/w) to about 18 % (w/w), from about 18 % (w/w) to about 19 % (w/w), or from about 19 % (w/w) to about 20 % (w/w) methyl paraben.
Additional Compositions
[0088] A composition of the present disclosure to treat or prevent a viral infection (e.g., a coronavirus infection) may comprise Argatroban, Disodium Edetate, Hexa-D-Arginine Amide (Chloride Salt), Glyceryl Monooleate (e.g., 90% monosub stituted or 40% monosub stituted), Polysorbate 80, Benzalkonium chloride, microcrystalline cellulose and carboxymethyl cellulose sodium (e.g., VIVAPURMCG 811 P), Glacial acetic acid (100 mM), Sodium Hydroxide, Purified Water, or a combination thereof. For example, a composition to treat or prevent a viral infection may comprise about 0.1 mg/mL Argatroban, about 4 mg/mL Disodium Edetate, about 5 mg/mL Hexa-D-Arginine Amide, Chloride Salt (CAS No.: 673202-67-0), about 2.5 mg/mL Glyceryl Monooleate (e.g., 90% monosub stituted or 40% monosub stituted), about 5 mg/mL Polysorbate 80, about 1.2 mg/mL Benzalkonium chloride, about 20 mg/mL microcrystalline cellulose and carboxymethyl cellulose sodium (e.g., VIVAPUR MCG 811 P), about 6 mg/mL glacial acetic acid (100 mM), a sufficient quantity of purified water, and sodium hydroxide as required to reach a target pH.
[0089] A composition may be formulated without acetic acid. For example, a composition to treat or prevent a viral infection may comprise about 0.1 mg/mL Argatroban, about 4 mg/mL Disodium Edetate, about 5 mg/mL Hexa-D-Arginine Amide, Chloride Salt (CAS No.: 673202-67-0), about 2.5 mg/mL Glyceryl Monooleate (e.g., 90% monosub stituted or 40% monosub stituted), about 5 mg/mL Polysorbate 80, about 1.2 mg/mL Benzalkonium chloride, about 20 mg/mL microcrystalline cellulose and carboxymethyl cellulose sodium (e.g., VIVAPUR MCG 811 P), a sufficient quantity of purified water, and sodium hydroxide as required to reach a target pH.
[0090] A composition of the present disclosure may be formulated as a preservative-free formulation. For example, a preservative-free composition may comprise about 0.1 mg/mL Argatroban, about 4 mg/mL Disodium Edetate, about 5 mg/mL Hexa-D-Arginine Amide, Chloride Salt (e.g., CAS No.: 673202-67-0), about 2.5 mg/mL Glyceryl Monooleate, about 5 mg/mL Polysorbate 80, about 20 mg/mL microcrystalline cellulose and carboxymethyl cellulose sodium (e.g., VIVAPUR MCG), about 6 mg/mL glacial acetic acid (100 mM), a sufficient quantity of purified water, and sodium hydroxide as required to reach a target pH. For example, a preservative-free composition may comprise about 0.1 mg/mL Argatroban, about 4 mg/mL Disodium Edetate, about 5 mg/mL Hexa-D-Arginine Amide, Chloride Salt (CAS No.: 673202-67-0), about 2.5 mg/mL Glyceryl Monooleate, about 5 mg/mL Polysorbate 80, about 20 mg/mL microcrystalline cellulose and carboxymethyl cellulose sodium (e.g., VIVAPUR MCG), a sufficient quantity of purified water, and sodium hydroxide as required to reach a target pH. In some embodiments, a composition may further comprise ethanol to facilitate solubility of one or more agents in the composition. A composition may be formulated for nasal delivery.
[0091] In some embodiments, a composition of the present disclosure to treat or prevent a viral infection (e.g., a coronavirus infection) may comprise microcrystalline cellulose and carboxymethyl cellulose, polysorbate 80, glyceryl monooleate (40% monosub stituted), butylated hydroxytoluene, disodium edetate (EDTA), benzalkonium chloride, and trisodium citrate, a sufficient quantity of purified water, and hydrochloric acid to reach a target pH. For example, a composition may comprise about 30 mg/mL microcrystalline cellulose and carboxymethyl cellulose, about 5.00 mg/mL polysorbate 80, about 2.50 mg/mL glyceryl monooleate (40% monosub stituted), about 0.5 mg/mL butylated hydroxytoluene, about 4.00 mg/mL disodium edetate (EDTA), about 0.20 mg/mL benzalkonium chloride, about 4.41 mg/mL trisodium citrate, a sufficient quantity of purified water, and hydrochloric acid to reach a pH of about 5.0. The composition may be formulated for nasal delivery.
[0092] A composition of the present disclosure may comprise Argatroban. A composition may comprise from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.15 mg/mL, from about 0.15 mg/mL to about 0.2 mg/mL, from about 0.2 mg/mL to about 0.25 mg/mL, from about 0.25 mg/mL to about 0.3 mg/mL, from about 0.3 mg/mL to about 0.35 mg/mL, from about 0.35 mg/mL to about 0.4 mg/mL, from about 0.4 mg/mL to about 0.45 mg/mL, from about 0.45 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 0.55 mg/mL, from about from about 0.5 mg/mL to about 1.0 mg/mL, from about 1.0 mg/mL to about 2.0 mg/mL, from about 2.0 mg/mL to about 3.0 mg/mL, from about 3.0 mg/mL to about 4.0 mg/mL, or from about 4.0 mg/mL to about 5.0 mg/mL Argatroban. In some embodiments, a composition comprising Argatroban may be administered intravenously. In some embodiments, a dose of a composition comprising Argatroban may be selected such that the median plasma concentration is from about 0.35 pg/mL plasma to about 0.41 pg/mL. Argatroban may have a molecular weight of about 508.6, so a steady state plasma level of 0.4 mg/L may be about 0.8 pM Argatroban. In some embodiments, a dose of a composition comprising Argatroban may be selected such that 50% inhibition is achieved. Argatroban may have an inhibitor constant of 100 nM. A composition may comprise about 10 mM Argatroban.
[0093] A composition of the present disclosure may comprise Disodium Edetate (EDTA). A composition may comprise from about 0.1 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, or from about 9 mg/mL to about 10 mg/mL of Disodium Edetate. A composition may comprise from about 1 mM to about 3 mM, from about 2 mM to about 4 mM, from about 3 mM to about 5 mM, from about 4 mM to about 6 mM, from about 5 mM to about 10 mM, or from about 1 mM to about 15 mM of Disodium Edetate. Disodium Edetate may be added as a chelator of Zn to inhibit ACE-2. A composition may be administered at a concentration of about 2.0 mg/mL Disodium Edetate. Disodium Edetate may have a molecular weight of about 336.2. A composition comprising 2 mg/mL of Disodium Edetate may be is 6 mM. ACE-2 may be inhibited by 2 mM EDTA, 5 mM EDTA, or by 10 mM EDTA. In some embodiments, EDTA may be EDTA-magnesium.
TABLE 3 - Examples of Disodium Edetate Compositions for Nasal Administration
[0094] A composition of the present disclosure may comprise egtazic acid (EGTA). A composition may comprise from about 0.1 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, or from about 9 mg/mL to about 10 mg/mL of EGTA. A composition may comprise from about 1 mM to about 3 mM, from about 2 mM to about 4 mM, from about 3 mM to about 5 mM, from about 4 mM to about 6 mM, from about 5 mM to about 10 mM, or from about 1 mM to about 15 mM of EGTA. EGTA may be added as a chelator of Zn to inhibit ACE-2. A composition may be administered at a concentration of about 2.0 mg/mL EGTA. Disodium Edetate may have a molecular weight of about 380.35 g/mol. A composition comprising 2 mg/mL of EGTA may be is 6 mM. ACE-2 may be inhibited by 2 mM EGTA, 5 mM EGTA, or by 10 mM EGTA.
[0095] A composition of the present disclosure may comprise Hexa-D-Arginine Amide (e.g., a chloride salt of Hexa-D-Arginine Amide). A composition may comprise from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, from about 9 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 11 mg/mL, from about 11 mg/mL to about 12 mg/mL, from about 12 mg/mL to about 13 mg/mL, from about 13 mg/mL to about 14 mg/mL, from about 14 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 16 mg/mL, from about 16 mg/mL to about 17 mg/mL, from about 17 mg/mL to about 18 mg/mL, from about 18 mg/mL to about 19 mg/mL, from about 19 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 21 mg/mL, from about 21 mg/mL to about 22 mg/mL, from about 22 mg/mL to about 23 mg/mL, from about 23 mg/mL to about 24 mg/mL, from about 24 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 26 mg/mL, from about 26 mg/mL to about 27 mg/mL, from about 27 mg/mL to about 28 mg/mL, from about 28 mg/mL to about 29 mg/mL, or from about 29 mg/mL to about 30 mg/mL Hexa-D-Arginine Amide (e.g., a chloride salt of Hexa-D-Arginine Amide).
[0096] A composition of the present disclosure may comprise butylated hydroxytoluene. A composition may comprise from about 0.1 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, or from about 9 mg/mL to about 10 mg/mL of butyl ated hydroxytoluene.
[0097] A composition of the present disclosure may comprise Glyceryl Monooleate (e.g., 90% monosub stituted or 40% monosubstituted). In some embodiments, glyceryl monooleate may be chemically described as 2,3-dihydroxypropyl (Z)-octadec-9-enoate. In some embodiments, Glyceryl Monooleate may function as an antiviral by inactivating envelope viruses. In some embodiments, Glyceryl Monooleate act as a bioadhesive to facilitate retention of the composition at the site of application (e.g., the nasal cavity). In some embodiments, a composition may comprise cubic phase of glyceryl monooleate, which may be a transparent, stiff, gel-like substance built up of three-dimensional network of curved lipid bilayers separated by a network of congruent water channels. The cubic phase may serve as a drug delivery system, or an anti-viral to inactivate envelope viruses, or both. In some embodiments, glyceryl monooleate may reduce the infectivity of an envelope virus (e.g., a coronavirus, a bunyavirus, a filovirus, a flavivirus, a herpesvirus, a hepadnaviruses, a paramyxovirus, a poxvirus, a rhabdovirus, or a retrovirus). A composition may comprise from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, from about 9 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 11 mg/mL, from about 11 mg/mL to about 12 mg/mL, from about 12 mg/mL to about 13 mg/mL, from about 13 mg/mL to about 14 mg/mL, from about 14 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 16 mg/mL, from about 16 mg/mL to about 17 mg/mL, from about 17 mg/mL to about 18 mg/mL, from about 18 mg/mL to about 19 mg/mL, from about 19 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 21 mg/mL, from about 21 mg/mL to about 22 mg/mL, from about 22 mg/mL to about 23 mg/mL, from about 23 mg/mL to about 24 mg/mL, from about 24 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 26 mg/mL, from about 26 mg/mL to about 27 mg/mL, from about 27 mg/mL to about 28 mg/mL, from about 28 mg/mL to about 29 mg/mL, or from about 29 mg/mL to about 30 mg/mL Glyceryl Monooleate (e.g., 90% monosub stituted or 40% monosub stituted).
[0098] A composition of the present disclosure may comprise Polysorbate 80. In some embodiments, Polysorbate 80 may function as a non-ionic surfactant and inactivate viruses by solvating the viral envelope and disrupting the nucleocapsid. Linkages between the hydrophobic and hydrophilic segments of Polysorbate 80 may play a role in the viral disinfectant function of Polysorbate 80. In some embodiments, Polysorbate 80 may function as a stabilizer. For example, Polysorbate may stabilize a composition comprising glyceryl monooleate by emulsifying the glyceryl monooleate. Polysorbate 80 is a nonionic surfactant and emulsifier often used in foods and cosmetics. This synthetic compound is a viscous, water-soluble yellow liquid. Polysorbate 80 may be derived from polyethoxylated sorbitan and oleic acid. The hydrophilic groups in this compound are polyethers also known as polyoxyethylene groups, which are polymers of ethylene oxide. In the nomenclature of polysorbates, the numeric designation following polysorbate refers to the lipophilic group, in this case the oleic acid. Polysorbate 80 may also be referred to as Polyoxyethylene (20) sorbitan monooleate, or (x)-sorbitan mono-9-octadecenoate poly(oxy-l,2-ethanediyl). The critical micelle concentration of polysorbate 80 in pure water may be about as 0.012 mM.
The molecular weight may be about 1310 g/mole. A composition comprising 1 mM of polysorbate 80 may comprise about 1310 mg/L or 1.3 mg/mL. A composition comprising 0.01 mM of polysorbate 80 may comprise about 0.013 mg/mL. A composition may comprise an amount of polysorbate 80 above the critical micelle concentration (CMC) for Polysorbate 80. A composition may comprise from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, from about 9 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 11 mg/mL, from about 11 mg/mL to about 12 mg/mL, from about 12 mg/mL to about 13 mg/mL, from about 13 mg/mL to about 14 mg/mL, from about 14 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 16 mg/mL, from about 16 mg/mL to about 17 mg/mL, from about 17 mg/mL to about 18 mg/mL, from about 18 mg/mL to about 19 mg/mL, from about 19 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 21 mg/mL, from about 21 mg/mL to about 22 mg/mL, from about 22 mg/mL to about 23 mg/mL, from about 23 mg/mL to about 24 mg/mL, from about 24 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 26 mg/mL, from about 26 mg/mL to about 27 mg/mL, from about 27 mg/mL to about 28 mg/mL, from about 28 mg/mL to about 29 mg/mL, or from about 29 mg/mL to about 30 mg/mL Polysorbate 80. In some embodiments, a dosage of a composition may comprise 0.05 mg of polysorbate 80. Polysorbate 80 may function as a non-ionic surfactant for inactivation of viruses (e.g., SARS-CoV-2) by solvating the viral envelope and disrupting the nucleocapsid.
TABLE 4 - Examples of Polysorbate 80 Compositions for Nasal Administration
[0099] A composition of the present disclosure may comprise Benzalkonium chloride. A composition may comprise from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, from about 9 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 11 mg/mL, from about 11 mg/mL to about 12 mg/mL, from about 12 mg/mL to about 13 mg/mL, from about 13 mg/mL to about 14 mg/mL, from about 14 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 16 mg/mL, from about 16 mg/mL to about 17 mg/mL, from about 17 mg/mL to about 18 mg/mL, from about 18 mg/mL to about 19 mg/mL, from about 19 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 21 mg/mL, from about 21 mg/mL to about 22 mg/mL, from about 22 mg/mL to about 23 mg/mL, from about 23 mg/mL to about 24 mg/mL, from about 24 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 26 mg/mL, from about 26 mg/mL to about 27 mg/mL, from about 27 mg/mL to about 28 mg/mL, from about 28 mg/mL to about 29 mg/mL, or from about 29 mg/mL to about 30 mg/mL Benzalkonium chloride. Benzalkonium chloride may be used in a nasal spray, for example as provided in TABLE 5. A dose of a composition may comprise 1.2 mg/mL of Benzalkonium chloride.
TABLE 5 - Examples of Benzalkonium Chloride Compositions for Nasal
Administration
[0100] A composition of the present disclosure may comprise microcrystalline cellulose. In some embodiments, microcrystalline cellulose may increase retention time of a composition within the nasal cavity. A composition may comprise from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, from about 9 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 11 mg/mL, from about 11 mg/mL to about 12 mg/mL, from about 12 mg/mL to about 13 mg/mL, from about 13 mg/mL to about 14 mg/mL, from about 14 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 16 mg/mL, from about 16 mg/mL to about 17 mg/mL, from about 17 mg/mL to about 18 mg/mL, from about 18 mg/mL to about 19 mg/mL, from about 19 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 21 mg/mL, from about 21 mg/mL to about 22 mg/mL, from about 22 mg/mL to about 23 mg/mL, from about 23 mg/mL to about 24 mg/mL, from about 24 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 26 mg/mL, from about 26 mg/mL to about 27 mg/mL, from about 27 mg/mL to about 28 mg/mL, from about 28 mg/mL to about 29 mg/mL, or from about 29 mg/mL to about 30 mg/mL microcrystalline cellulose. [0101] A composition of the present disclosure may comprise carboxymethyl cellulose. In some embodiments, carboxymethyl cellulose may increase retention time of a composition within the nasal cavity. In some embodiments, carboxymethyl cellulose may form electrostatic interactions with a virus, thereby sequestering the virus away from the nasal epithelium and preventing infection as well as further dispersion into the lower airway. Sequestering the virus may treat or prevent a viral infection by reducing the infectivity of the virus. A composition may comprise from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, from about 9 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 11 mg/mL, from about 11 mg/mL to about 12 mg/mL, from about 12 mg/mL to about 13 mg/mL, from about 13 mg/mL to about 14 mg/mL, from about 14 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 16 mg/mL, from about 16 mg/mL to about 17 mg/mL, from about 17 mg/mL to about 18 mg/mL, from about 18 mg/mL to about 19 mg/mL, from about 19 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 21 mg/mL, from about 21 mg/mL to about 22 mg/mL, from about 22 mg/mL to about 23 mg/mL, from about 23 mg/mL to about 24 mg/mL, from about 24 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 26 mg/mL, from about 26 mg/mL to about 27 mg/mL, from about 27 mg/mL to about 28 mg/mL, from about 28 mg/mL to about 29 mg/mL, or from about 29 mg/mL to about 30 mg/mL carboxymethyl cellulose.
[0102] In some embodiments, a composition may comprise a combination of microcrystalline cellulose and carboxymethyl cellulose (e.g., VIVAPUR MCG 811 P) at a combined concentration of from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, from about 9 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 11 mg/mL, from about 11 mg/mL to about 12 mg/mL, from about 12 mg/mL to about 13 mg/mL, from about 13 mg/mL to about 14 mg/mL, from about 14 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 16 mg/mL, from about 16 mg/mL to about 17 mg/mL, from about 17 mg/mL to about 18 mg/mL, from about 18 mg/mL to about 19 mg/mL, from about 19 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 21 mg/mL, from about 21 mg/mL to about 22 mg/mL, from about 22 mg/mL to about 23 mg/mL, from about 23 mg/mL to about 24 mg/mL, from about 24 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 26 mg/mL, from about 26 mg/mL to about 27 mg/mL, from about 27 mg/mL to about 28 mg/mL, from about 28 mg/mL to about 29 mg/mL, or from about 29 mg/mL to about 30 mg/mL.
[0103] A composition of the present disclosure may comprise citrate (e.g., trisodium citrate dehydrate). In some embodiments, citrate may act as a buffer. In some embodiments, citrate may chelate calcium and interfere with the calcium-dependent endoprotease activity of furin. In some embodiments, citrate may inhibit activation of a clotting cascade by chelating calcium ions. In some embodiments, citrate may chelate or sequester calcium ions, thereby inhibiting calcium-dependent endoprotease activity of furin. Inhibition of furin by citrate may prevent cleavage of a viral spike protein (e.g., a SARS-CoV-2 spike protein), thereby reducing the infectivity of the virus. A composition may comprise from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, from about 9 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 11 mg/mL, from about 11 mg/mL to about 12 mg/mL, from about 12 mg/mL to about 13 mg/mL, from about 13 mg/mL to about 14 mg/mL, from about 14 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 16 mg/mL, from about 16 mg/mL to about 17 mg/mL, from about 17 mg/mL to about 18 mg/mL, from about 18 mg/mL to about 19 mg/mL, from about 19 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 21 mg/mL, from about 21 mg/mL to about 22 mg/mL, from about 22 mg/mL to about 23 mg/mL, from about 23 mg/mL to about 24 mg/mL, from about 24 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 26 mg/mL, from about 26 mg/mL to about 27 mg/mL, from about 27 mg/mL to about 28 mg/mL, from about 28 mg/mL to about 29 mg/mL, or from about 29 mg/mL to about 30 mg/mL (e.g., trisodium citrate dehydrate). Citrate (e.g., trisodium citrate) may function to prevent activation of a clotting cascade by chelating calcium ions. Chelating and sequestration of calcium ions by citrate may interfere with the calcium-dependent endoprotease activity of furin, thereby preventing cleavage of the spike protein of SARS-CoV-2.
[0104] A composition of the present disclosure may comprise acetic acid (e.g., 100 mM glacial acetic acid). A composition may comprise from about 0.001 mg/mL to about 0.005 mg/mL, from about 0.005 mg/mL to about 0.01 mg/mL, from about 0.01 mg/mL to about 0.05 mg/mL, from about 0.05 mg/mL to about 0.1 mg/mL, from about 0.1 mg/mL to about 0.5 mg/mL, from about 0.5 mg/mL to about 1 mg/mL, from about 1 mg/mL to about 2 mg/mL, from about 2 mg/mL to about 3 mg/mL, from about 3 mg/mL to about 4 mg/mL, from about 4 mg/mL to about 5 mg/mL, from about 5 mg/mL to about 6 mg/mL, from about 6 mg/mL to about 7 mg/mL, from about 7 mg/mL to about 8 mg/mL, from about 8 mg/mL to about 9 mg/mL, from about 9 mg/mL to about 10 mg/mL, from about 10 mg/mL to about 11 mg/mL, from about 11 mg/mL to about 12 mg/mL, from about 12 mg/mL to about 13 mg/mL, from about 13 mg/mL to about 14 mg/mL, from about 14 mg/mL to about 15 mg/mL, from about 15 mg/mL to about 16 mg/mL, from about 16 mg/mL to about 17 mg/mL, from about 17 mg/mL to about 18 mg/mL, from about 18 mg/mL to about 19 mg/mL, from about 19 mg/mL to about 20 mg/mL, from about 20 mg/mL to about 21 mg/mL, from about 21 mg/mL to about 22 mg/mL, from about 22 mg/mL to about 23 mg/mL, from about 23 mg/mL to about 24 mg/mL, from about 24 mg/mL to about 25 mg/mL, from about 25 mg/mL to about 26 mg/mL, from about 26 mg/mL to about 27 mg/mL, from about 27 mg/mL to about 28 mg/mL, from about 28 mg/mL to about 29 mg/mL, or from about 29 mg/mL to about 30 mg/mL (e.g., 100 mM glacial acetic acid). [0105] A composition of the present disclosure may comprise L-arginine. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 %
(w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 %
(w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 %
(w/v) to about 20 % (w/v) L-arginine.
[0106] A composition of the present disclosure may comprise Poly-L-arginine. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 % (w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 % (w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 % (w/v) to about 20 % (w/v) Poly-L-arginine.
[0107] In some embodiments, a composition may have a pH of about 5.5, A composition may have a pH of from about 4.5 to about 6.5 or from about 4 to about 7. A composition may have an osmolality of about 600 mOsm/kg. A composition may have an osmolality of about 290 mOsm/kg. A composition may be isotonic. A composition may have an osmolality of from about 150 mOsm/kg to about 350 mOsm/kg, from about 200 mOsm/kg to about 500 mOsm/kg, from about 400 mOsm/kg to about 700 mOsm/kg or from about 300 mOsm/kg to about 800 mOsm/kg. A composition may have an osmolarity of about 600 mOsm/L. A composition may have an osmolality of about 290 mOsm/L. A composition may be isotonic. A composition may have an osmolarity of from about 150 mOsm/L to about 350 mOsm/L, from about 200 mOsm/L to about 500 mOsm/L, from about 400 mOsm/L to about 700 mOsm/L or from about 300 mOsm/L to about 800 mOsm/L. A composition may have a viscosity of about 2500 cP. A composition may have a viscosity of from about 1500 cP to about 3000 cP or from about 1000 cP to about 3500 cP.
[0108] A composition of the present disclosure may comprise gamma cyclodextrine. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 % (w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 % (w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 % (w/v) to about 20 % (w/v) gamma cyclodextrine.
[0109] A composition of the present disclosure may comprise a-cyclodextrine. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 % (w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 % (w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 % (w/v) to about 20 % (w/v) a-cyclodextrine.
[0110] A composition of the present disclosure may comprise methyl-B-cyclodextrine. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 % (w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 % (w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 % (w/v) to about 20 % (w/v) methyl -B-cyclodextrine.
[0111] A composition of the present disclosure may comprise n-Capric acid sodium. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 % (w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 % (w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 % (w/v) to about 20 % (w/v) n-Capric acid sodium.
[0112] A composition of the present disclosure may comprise chitosan. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 % (w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 % (w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 % (w/v) to about 20 % (w/v) chitosan. [0113] A composition of the present disclosure may comprise L-a-phosphatidylcholine didecanyl. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 % (w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 % (w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 % (w/v) to about 20 % (w/v) L-a-phosphatidylcholine didecanyl.
[0114] A composition of the present disclosure may comprise S-Notroso-N-acetyl- penicillamine. A composition may comprise from about 0.005 % (w/v) to about 0.01 %
(w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 %
(w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 %
(w/v) to about 15 % (w/v), from about 15 % (w/v) to about 16 % (w/v), from about 16 %
(w/v) to about 17 % (w/v), from about 17 % (w/v) to about 18 % (w/v), from about 18 %
(w/v) to about 19 % (w/v), or from about 19 % (w/v) to about 20 % (w/v) S-Notroso-N- acetyl-penicillamine.
[0115] A composition of the present disclosure may comprise pluronic 127. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 %
(w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 %
(w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 %
(w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 %
(w/v) to about 20 % (w/v) Pluronic 127.
[0116] A composition of the present disclosure may comprise sodium nitroprusside. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 % (w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 % (w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 % (w/v) to about 20 % (w/v) sodium nitroprusside.
[0117] A composition of the present disclosure may comprise sodium glycocholate. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 % (w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 % (w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 % (w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 % (w/v) to about 20 % (w/v) sodium glycocholate.
[0118] A composition of the present disclosure may comprise DDPC. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 %
(w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 %
(w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 %
(w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 %
(w/v) to about 20 % (w/v) DDPC.
[0119] A composition of the present disclosure may comprise MBCD. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 %
(w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 %
(w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 %
(w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 %
(w/v) to about 20 % (w/v) MBCD.
[0120] A composition of the present disclosure may comprise EDTA. A composition may comprise from about 0.005 % (w/v) to about 0.01 % (w/v), from about 0.01 % (w/v) to about 0.02 % (w/v), from about 0.02 % (w/v) to about 0.05 % (w/v), from about 0.05 % (w/v) to about 0.1 % (w/v), from about 0.1 % (w/v) to about 0.5 % (w/v), from about 0.5 % (w/v) to about 1 % (w/v), from about 1 % (w/v) to about 2 % (w/v), from about 2 % (w/v) to about 3 % (w/v), from about 3 % (w/v) to about 4 % (w/v), from about 4 % (w/v) to about 5 % (w/v), from about 5 % (w/v) to about 6 % (w/v), from about 6 % (w/v) to about 7 % (w/v), from about 7 % (w/v) to about 8 % (w/v), from about 8 % (w/v) to about 9 % (w/v), from about 9 % (w/v) to about 10 % (w/v), from about 10 % (w/v) to about 11 % (w/v), from about 11 % (w/v) to about 12 % (w/v), from about 12 % (w/v) to about 13 % (w/v), from about 13 %
(w/v) to about 14 % (w/v), from about 14 % (w/v) to about 15 % (w/v), from about 15 %
(w/v) to about 16 % (w/v), from about 16 % (w/v) to about 17 % (w/v), from about 17 %
(w/v) to about 18 % (w/v), from about 18 % (w/v) to about 19 % (w/v), or from about 19 %
(w/v) to about 20 % (w/v) EDTA.
[0121] In some embodiments, a composition to treat or prevent a viral infection may comprise an additional antiviral agent. For example, a composition may comprise chloroquine, Remdesivir, Tocilizumab, Lopinavir, Sarilumab, interferon-B, or a combination thereof. The additional antiviral agent may kill a virus or suppress viral replication. In some embodiments, a composition of the present disclosure may comprise tenofovir disoproxil fumarate, nevirapine, delavirdine, efavirenz, saquinavir, ritonavir, indinavir, nelfmavir, amprenavir, lopinavir, darunavir and atazanavir, peramivir, zanamivir (Tamiflu), oseltamivir (Relenza), amantadine, rimantadine, adefovir dipivoxil, famciclovir, penciclovir, imiquimod, docosanole, foscarnet (PFA), maribavir, BAY 38-4766, GW275175X, MVE-1, MVE-2, AM- 3, AM-5, mannozym, bropirimine, 3, 6-bis(2-p-peri dinoethoxy) acridine trihydrochloride, phenyleneamine, 2-amino-5-halo-6-aryl-4(3H)-pyrimidinones, 2-amino-5-bromo-6-methyl- 4(3H)-pyrimidinone, 7,8-didehydro-7-methyl-8-thioxoguanosine, 7-deazaguanosine, melatonin, 8-chloro-7-deazaguanosine, CL246,738, glycyrrhizin, pleconaril, bananin, iodobananin, vanillinbananin, ansabananin, eubananin, adeninobananin, cloroquine, valinomycin, idoxuridine, aciclovir (acyclovir or acycloguansoine), valaciclovir (valacyclovir), ganciclovir, valganciclovir, adenosine arabinoside (AraA, Vidarabine), AraA monophosphate, cytosine arabinoside (AraC, cytarabine), cytosine arabinoside monophosphate (Ara-CMP), azidothymidine (AZT), l-B-D-ribofuranosyl-l,2,4-triazole-3- carboxamide (ribavirin or RBV), 5-ethynyl-l-B-D-ribofuranosylimidazole-4-carboxamide (EICAR), EICAR-monophosphate, ribamidine, ribavirin 2’, 3’, 5’ -acetate, ribavirin-5 ’- sulfamate, ribavirin 5 ’-triphosphate, ribavirin 5 ’-monophosphate, ZX-2401, mycophenolic acid, tiazofurin, tiazofurin-5’ -monophosphate, tiazofurin 2 ’,3 ’,5 ’-acetate, 7-thia-8- oxoguanosine, selenazofurin, pyrazofurin, furanonaphthoquinone derivatives, merimepodib (VX497), viramidine, 6-azauridine, 9-(2-phosphonylmethoxyethyl)guanine (PMEG), (S)-9- (3-hydroxy-2-phosphonylmethoxypropyl)adenine (HPMPA), 9-(2- phosphonylmethoxyethyl)adenine (PMEA), 9-(2-phosphonylmethoxyethyl)-2,6- diaminopurine (PMEDAP), didenosine (DDI), dideoxycytosine (DDC), stavudine (d4T), Epivir (3TC), abacavir (ABC), iodo-deozyuridine (DU), and bromovinyl deoxiuridine (BVDU or brivudin), (S)-l-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine (HPMPC, cidofovir, CDV or Vistide®), cyclic HPMPC, hexadecyloxypropyl-cidofovir (HDP-CDV, or CMXOOl), 3-deazaguanine (3-DG), 3-deazauridine, 9-(S)-(2,3-dihydroxypropyl)adenine ((S)-DHPA), zidovudine, didanosine, zalcitabine, stavudine, lamivudine, abacavir, emtricitabine, or a combination thereof.
Phage Therapies
[0122] A composition described herein may comprise a phage therapy (e.g., a bacteriophage therapy). Bacteriophages are bacteria-specific viruses that infect and, in the case of obligately lytic phages, destroy their host bacteria. Phage therapy may be used therapeutically to combat bacterial infections. In some embodiments, a phage therapy may be administered parenterally. A phage therapy may be administered by inhalation. A phage therapy may be administered nasally. In some embodiments, a phage therapy may be administered via oral delivery (e.g., to treat a gastrointestinal viral infection). A phage therapy may be delivered locally (e.g., to skin, lungs, nasal passage, ears, or teeth). A phage therapy may be delivered systemically (e.g., through the circulatory system). In some embodiments, a composition comprising a bacteriophage may prevent biofilm formation on a medical device (e.g., a nasal administration device).
[0123] A bacteriophage (“phage”) may be used as an alternative or supplementary treatment option over a conventional antibiotic. In some embodiments, a phage can kill the bacterial host, including those that are resistant to antibiotics. A composition comprising a bacteriophage may be formulated for inhalation and may be suitable for aerosol delivery. Disclosed herein are formulation for aerosol delivery of compositions (e.g., comprising a phage). Inhaled phage therapy may be used as a complementary treatment option with current antibiotics and as a preventative means. In some embodiments, a phage therapy may be used to treat a bacterial infectious agent that is antibiotic resistant. For example, a phage therapy may be used to treat infections caused by the Burkholderia cepacia complex (BCC) since members of the BCC are antibiotic pan-resistant. Formulations
[0124] A composition of the present disclosure (e.g., a composition comprising a bioadhesive, an envelope disruptor, an inhibitor of ACE-2, TMPRSS2, furin, or cathepsin, a dual-action enzyme inhibitor, or combinations thereof) may be formulated for nasal delivery. In some embodiments, the composition may be formulated to be substantially free of endotoxins. The composition may be dispensed from a container configured to produce a desired spray pattern. In some embodiments, a formulation for nasal delivery may comprise an enzyme inhibitor, a stabilizer, a bioadhesive, or combinations thereof. In some embodiments, the protease inhibitor formulation may further comprise an antioxidant, a bacteriostatic agent, a buffer, or combinations thereof. In some embodiments, nasal delivery of the composition may form or maintain a protective mucosal-like barrier in the nasal cavity of a subject.
[0125] In some embodiments, a formulation to reduce the infectivity of viral infection may comprise disodium edetate, glyceryl monooleate, polysorbate 80, microcrystalline cellulose or carboxymethyl cellulose, citrate (e.g., trisodium citrate), and benzalkonium chloride. For example, a protease inhibiter formulation for treatment or prevention of a respiratory viral infection may comprise from about 3 mg/mL to about 5 mg/mL of disodium edetate, from about 1.5 mg/mL to about 3.5 mg/mL of glyceryl monooleate, from about 4 mg/mL to about 6 mg/mL polysorbate 80, from about 20 mg/mL to about 40 mg/mL microcrystalline cellulose or carboxymethyl cellulose, from about 3.5 mg/mL to about 5.5 mg/mL citrate (e.g., trisodium citrate), and from about 0.1 mg/mL to about 1.5 mg/mL benzalkonium chloride. The protease inhibitor formulation may be formulated for nasal delivery.
[0126] In some embodiments, the disodium edetate may inhibit ACE 2, thereby inhibiting viral entry of a coronavirus. In some embodiments, the glyceryl monooleate may inactivate an envelope virus (e.g., a coronavirus, a bunyavirus, a filovirus, a flavivirus, a herpesvirus, a hepadnaviruses, a paramyxovirus, a poxvirus, a rhabdovirus, or a retrovirus). In some embodiments, the citrate may inhibit endoprotease activity of furin, thereby preventing cleavage of a viral spike protein (e.g., a SARS-CoV-2 spike protein). In some embodiments, the polysorbate 80 may inactivate the virus by solvating the viral envelope and disrupting the nucleocapsid. In some embodiments, the carboxymethyl cellulose may form electrostatic interactions with the virus and sequester the virus away from the nasal epithelium. One or more of disodium edetate, glyceryl monooleate, citrate, polysorbate 80, or carboxymethyl cellulose may reduce the infectivity of a virus (e.g., a coronavirus), thereby treating or preventing the viral infection. [0127] In some embodiments, a formulation for treatment or prevention of a respiratory viral infection may further comprise butylated hydroxytoluene. For example, a formulation for treatment or prevention of a respiratory viral infection may comprise from about 0.4 mg/mL to about 0.6 mg/mL butylated hydroxytoluene.
Formulations for Nasal Delivery
[0128] A formulation comprising a composition for nasal deliver may have a pH corresponding to a physiologically acidic nasal pH. The physiologically acidic nasal pH may depend on intact nasal mucosal function. A composition may comprise a pH of about be 6.5 ± 0.5 (5.9 to 7.3) or about 6.7 ± 0.6 (5.3 to 7.6). A composition may comprise a pH of about 3.8-7.7 (mean ± SD 5.7 ± 0.9). A composition for nasal deliver may be in the slightly acidic range. The average pH may have an acidity of pH 5.7.
[0129] In some embodiments, a composition of the present disclosure (e.g., a composition to treat or prevent a viral infection) may be formulated to minimize a chloride ion concentration. A chloride ion concentration may be less than about 1 M, less than about 100 mM, less than about 10 mM, less than about 1 mM, less than about 0.1 mM, or less than about 0.01 mM. A chloride ion bound to an ACE-2 inhibitor may have catalytic activity.
Endotoxin-Free Formulations
[0130] In some embodiments, an endotoxin-free formulation may be a formulation which contains a Y2-receptor-binding peptide and one or more mucosal delivery enhancing agents. The solution may be substantially free of endotoxins and/or related pyrogenic substances. Endotoxins include toxins that are confined inside a microorganism and are released only when the microorganisms are broken down or die. Pyrogenic substances include fever- inducing, thermostable substances (glycoproteins) from the outer membrane of bacteria and other microorganisms. Both of these substances can cause fever, hypotension and shock if administered to humans. Producing formulations that are endotoxin-free can require special equipment, expert artisans, and can be significantly more expensive than making formulations that are not endotoxin-free.
Enzyme Inhibitor Formulations
[0131] A component of the invention is an inhibitor of one or more enzyme that is used by coronaviruses, including SARS-CoV-2, for cell entry and infectivity. Exemplary mucoadhesive polymer-enzyme inhibitor complexes that are useful within the mucosal delivery formulations and methods of the invention include, but are not limited to: Carboxymethylcellulose-pepstatin (with anti-pepsin activity); Poly(acrylic acid)-Bowman- Birk inhibitor (anti-chymotrypsin); Poly(acrylic acid)-chymostatin (anti-chymotrypsin); Poly(acrylic acid)-elastatinal (anti-elastase); Carboxymethylcellulose-elastatinal (anti- elastase); Polycarbophil— elastatinal (anti-elastase); Chitosan— antipain (anti-trypsin); Poly(acrylic acid)— bacitracin (anti-aminopeptidase N); Chitosan— EDTA (anti- aminopeptidase N, anti-carboxypeptidase A); Chitosan— EDTA— antipain (anti-trypsin, anti- chymotrypsin, anti-elastase). As described in further detail below, certain embodiments of the invention will optionally incorporate a novel chitosan derivative or chemically modified form of chitosan. One such novel derivative for use within the invention is denoted as a B-[l 4]- 2-guanidino-2-deoxy-D-glucose polymer (poly-GuD).
[0132] Any inhibitor that inhibits the activity of an enzyme that prevents virus entry or replication may be usefully employed in the compositions and methods of the invention. Useful enzyme inhibitors include, for example, soybean trypsin inhibitor, pancreatic trypsin inhibitor, chymotrypsin inhibitor and trypsin and chrymotrypsin inhibitor isolated from potato (solanum tuberosum L.) tubers. A combination or mixtures of inhibitors may be employed. Additional inhibitors of proteolytic enzymes for use within the invention include ovomucoid-enzyme, gabaxate mesylate, a 1 -antitrypsin, aprotinin, amastatin, bestatin, puromycin, bacitracin, leupepsin, a2-macroglobulin, pepstatin and egg white or soybean trypsin inhibitor. These and other inhibitors can be used alone or in combination. The inhibitor(s) may be incorporated in or bound to a carrier, e.g., a hydrophilic polymer, coated on the surface of the dosage form which is to contact the nasal mucosa, or incorporated in the superficial phase of the surface, in combination with the biologically active agent or in a separately administered (e.g., pre-administered) formulation.
[0133] The amount of the inhibitor, e.g., of a proteolytic enzyme inhibitor that is optionally incorporated in the compositions of the invention will vary depending on (a) the properties of the specific inhibitor, (b) the number of functional groups present in the molecule (which may be reacted to introduce ethylenic unsaturation necessary for copolymerization with hydrogel forming monomers), and (c) the number of lectin groups, such as glycosides, which are present in the inhibitor molecule. It may also depend on the specific therapeutic agent that is intended to be administered. Generally speaking, a useful amount of an enzyme inhibitor is from about 0.1 mg/ml to about 50 mg/ml, often from about 0.2 mg/ml to about 25 mg/ml, and more commonly from about 0.5 mg/ml to 5 mg/ml of the of the formulation (i.e., a separate formulation or combined formulation with the inhibitor and biologically active agent). [0134] In the case of trypsin inhibition, suitable inhibitors may be selected from, e.g., aprotinin, BBI, soybean trypsin inhibitor, chicken ovomucoid, chicken ovoinhibitor, human pancreatic trypsin inhibitor, camostat mesilate, flavonoid inhibitors, antipain, leupeptin, p- aminobenzamidine, AEBSF, TLCK (tosyllysine chloromethylketone), APMSF, DFP, PMSF, and poly(acrylate) derivatives. In the case of chymotrypsin inhibition, suitable inhibitors may be selected from, e.g., aprotinin, BBI, soybean trypsin inhibitor, chymostatin, benzyloxycarbonyl-Pro-Phe-CHO, FK-448, chicken ovoinhibitor, sugar biphenylboronic acids complexes, DFP, PMSF, B-phenylpropionate, and poly(acrylate) derivatives. In the case of elastase inhibition, suitable inhibitors may be selected from, e.g., elastatinal, methoxysuccinyl-Ala-Ala-Pro-Val-chloromethylketone (MPCMK), BBI, soybean trypsin inhibitor, chicken ovoinhibitor, DFP, and PMSF.
[0135] Additional enzyme inhibitors for use within the invention are selected from a wide range of non-protein inhibitors that vary in their degree of potency and toxicity. As described in further detail below, immobilization of these adjunct agents to matrices or other delivery vehicles, or development of chemically modified analogues, may be readily implemented to reduce or even eliminate toxic effects, when they are encountered. Among this broad group of candidate enzyme inhibitors for use within the invention are organophosphorous inhibitors, such as diisopropylfluorophosphate (DFP) and phenylmethylsulfonyl fluoride (PMSF), which are potent, irreversible inhibitors of serine proteases (e.g., trypsin and chymotrypsin). The additional inhibition of acetylcholinesterase by these compounds makes them highly toxic in uncontrolled delivery settings. Another candidate inhibitor, 4-(2-Aminoethyl)- benzenesulfonyl fluoride (AEBSF), has an inhibitory activity comparable to DFP and PMSF, but it is markedly less toxic. (4-Aminophenyl)-methanesulfonyl fluoride hydrochloride (APMSF) is another potent inhibitor of trypsin, but is toxic in uncontrolled settings. In contrast to these inhibitors, 4-(4-isopropylpiperadinocarbonyl)phenyl 1,2,3,4,-tetrahydro-l- naphthoate methanesulphonate (FK-448) is a low toxic substance, representing a potent and specific inhibitor of chymotrypsin. Further representatives of this non-protein group of inhibitor candidates, and also exhibiting low toxic risk, are camostat mesilate (N,N'- dimethylcarbamoylmethyl-p-(p'-guanidino-benzoyloxy)phenyla- cetate methane-sulphonate). [0136] Yet another type of enzyme inhibitory agent for use within the methods and compositions of the invention are amino acids and modified amino acids that interfere with enzymatic degradation of specific therapeutic compounds. For use in this context, amino acids and modified amino acids are substantially non-toxic and can be produced at a low cost. However, due to their low molecular size and good solubility, they are readily diluted and absorbed in mucosal environments. Nevertheless, under proper conditions, amino acids can act as reversible, competitive inhibitors of protease enzymes. Certain modified amino acids can display a much stronger inhibitory activity. A desired modified amino acid in this context is known as a 'transition-state' inhibitor. The strong inhibitory activity of these compounds is based on their structural similarity to a substrate in its transition-state geometry, while they are generally selected to have a much higher affinity for the active site of an enzyme than the substrate itself. Transition-state inhibitors are reversible, competitive inhibitors.
[0137] Examples of this type of inhibitor are a-aminoboronic acid derivatives, such as boro- leucine, boro-valine and boro-alanine. The boron atom in these derivatives can form a tetrahedral boronate ion that is believed to resemble the transition state of peptides during their hydrolysis by aminopeptidases. These amino acid derivatives are potent and reversible inhibitors of aminopeptidases and it is reported that boro-leucine is more than 100-times more effective in enzyme inhibition than bestatin and more than 1000-times more effective than puromycin. Another modified amino acid for which a strong protease inhibitory activity has been reported is N-acetylcysteine, which inhibits enzymatic activity of aminopeptidase N. This adjunct agent also displays mucolytic properties that can be employed within the methods and compositions of the invention to reduce the effects of the mucus diffusion barrier.
[0138] Still other useful enzyme inhibitors for use within the coordinate administration methods and combinatorial formulations of the invention may be selected from peptides and modified peptide enzyme inhibitors. An important representative of this class of inhibitors is the cyclic dodecapeptide, bacitracin, obtained from Bacillus licheniformis. In addition to these types of peptides, certain dipeptides and tripeptides display weak, non-specific inhibitory activity towards some protease. By analogy with amino acids, their inhibitory activity can be improved by chemical modifications. For example, phosphinic acid dipeptide analogues are also 'transition-state' inhibitors with a strong inhibitory activity towards aminopeptidases. They have reportedly been used to stabilize nasally administered leucine enkephalin. Another example of a transition-state analogue is the modified pentapeptide pepstatin, which is a very potent inhibitor of pepsin. Structural analysis of pepstatin, by testing the inhibitory activity of several synthetic analogues, demonstrated the major structure-function characteristics of the molecule responsible for the inhibitory activity. Another special type of modified peptide includes inhibitors with a terminally located aldehyde function in their structure. For example, the sequence benzyloxycarbonyl-Pro-Phe- CHO, which fulfills the known primary and secondary specificity requirements of chymotrypsin, has been found to be a potent reversible inhibitor of this target proteinase. The chemical structures of further inhibitors with a terminally located aldehyde function, e.g. antipain, leupeptin, chymostatin and elastatinal, are also known in the art, as are the structures of other known, reversible, modified peptide inhibitors, such as phosphoramidon, bestatin, puromycin and amastatin.
[0139] Due to their comparably high molecular mass, polypeptide protease inhibitors are more amenable than smaller compounds to concentrated delivery in a drug-carrier matrix. Additional agents for protease inhibition within the formulations and methods of the invention involve the use of complexing agents. These agents mediate enzyme inhibition by depriving the intranasal environment (or preparative or therapeutic composition) of divalent cations, which are co-factors for many proteases. For instance, the complexing agents EDTA and DTPA as coordinately administered or combinatorially formulated adjunct agents, in suitable concentration, will be sufficient to inhibit selected proteases to thereby enhance intranasal delivery of biologically active agents according to the invention. Further representatives of this class of inhibitory agents are EGTA, 1,10-phenanthroline and hydroxychinoline. In addition, due to their propensity to chelate divalent cations, these and other complexing agents are useful within the invention as direct, absorption and/or adsorption-promoting agents.
[0140] As noted in more detail elsewhere herein, it is also contemplated to use various polymers, particularly mucoadhesive polymers, as enzyme inhibiting agents within the coordinate administration, multi-processing and/or combinatorial formulation methods and compositions of the invention. For example, poly(acrylate) derivatives, such as poly(acrylic acid) and polycarbophil, can affect the activity of various proteases, including trypsin, chymotrypsin. The inhibitory effect of these polymers may also be based on the complexation of divalent cations such as Ca2+ and Zn2+. It is further contemplated that these polymers may serve as conjugate partners or carriers for additional enzyme inhibitory agents, as described above. For example, a chitosan-EDTA conjugate has been developed and is useful within the invention that exhibits a strong inhibitory effect towards the enzymatic activity of zinc- dependent proteases. The mucoadhesive properties of polymers following covalent attachment of other enzyme inhibitors in this context are not expected to be substantially compromised, nor is the general utility of such polymers as a delivery vehicle for biologically active agents within the invention expected to be diminished. On the contrary, the reduced distance between the delivery vehicle and mucosal surface afforded by the mucoadhesive mechanism will minimize presystemic metabolism of the active agent, while the covalently bound enzyme inhibitors remain concentrated at the site of drug delivery, minimizing undesired dilution effects of inhibitors as well as toxic and other side effects caused thereby. In this manner, the effective amount of a coordinately administered enzyme inhibitor can be reduced due to the exclusion of dilution effects.
[0141] Exemplary mucoadhesive polymer-enzyme inhibitor complexes that are useful within the mucosal formulations and methods of the invention include, but are not limited to: heparin, N-acetyl-cysteine, Carboxymethylcellulose-pepstatin (with anti-pepsin activity); Poly(acrylic acid)-Bowman-Birk inhibitor (anti-chymotrypsin); Poly(acrylic acid)- chymostatin (anti-chymotrypsin); Poly(acrylic acid)-elastatinal (anti-elastase); Carboxymethylcellulose-elastatinal (anti-elastase); Polycarbophil— elastatinal (anti-elastase); Chitosan— antipain (anti-trypsin); Poly(acrylic acid)— bacitracin (anti-aminopeptidase N); Chitosan— EDTA (anti-aminopeptidase N, anti-carboxypeptidase A); Chitosan— EDT A— antipain (anti -trypsin, anti-chymotrypsin, anti-elastase).
Mucolytic and Mucus-Clearing Agents and Methods
[0142] Effective delivery of therapeutic agents via intranasal administration must take into account the decreased drug transport rate across the protective mucus lining of the nasal mucosa, in addition to drug loss due to binding to glycoproteins of the mucus layer. Normal mucus is a viscoelastic, gel-like substance consisting of water, electrolytes, mucins, macromolecules, and sloughed epithelial cells. It serves primarily as a cytoprotective and lubricative covering for the underlying mucosal tissues. Mucus is secreted by randomly distributed secretory cells located in the nasal epithelium and in other mucosal epithelia. The structural unit of mucus is mucin. This glycoprotein is mainly responsible for the viscoelastic nature of mucus, although other macromolecules may also contribute to this property. In airway mucus, such macromolecules include locally produced secretory IgA, IgM, IgE, lysozyme, and bronchotransferrin, which also play an important role in host defense mechanisms.
[0143] The coordinate administration methods of the instant invention optionally incorporate effective mucolytic or mucus-clearing agents, which serve to degrade, thin or clear mucus from intranasal mucosal surfaces to facilitate absorption and/or adsorption of intranasally administered biotherapeutic agents. Within these methods, a mucolytic or mucus-clearing agent is coordinately administered as an adjunct compound to enhance intranasal delivery of the biologically active agent. Alternatively, an effective amount of a mucolytic or mucus clearing agent is incorporated as a processing agent within a multi-processing method of the invention, or as an additive within a combinatorial formulation of the invention, to provide an improved formulation that enhances intranasal delivery of biotherapeutic compounds by reducing the barrier effects of intranasal mucus.
[0144] A variety of mucolytic or mucus-clearing agents are available for incorporation within the methods and compositions of the invention. Based on their mechanisms of action, mucolytic and mucus clearing agents can often be classified into the following groups: proteases (e.g., pronase, papain) that cleave the protein core of mucin glycoproteins; sulfhydryl compounds that split mucoprotein disulfide linkages; and detergents (e.g., Triton X-100, Tween 20) that break non-covalent bonds within the mucus. Additional compounds in this context include, but are not limited to, bile salts and surfactants, for example, sodium deoxycholate, sodium taurodeoxycholate, sodium glycocholate, and lysophosphatidylcholine. [0145] The effectiveness of bile salts in causing structural breakdown of mucus is in the order deoxycholate>taurocholate>glycocholate. Other effective agents that reduce mucus viscosity or adhesion to enhance intranasal delivery according to the methods of the invention include, e.g., short-chain fatty acids, and mucolytic agents that work by chelation, such as N- acylcollagen peptides, bile acids, and saponins (the latter function in part by chelating Ca2+ and/or Mg2+ which play an important role in maintaining mucus layer structure).
[0146] Additional mucolytic agents for use within the methods and compositions of the invention include N-acetyl-L-cysteine (ACS), a potent mucolytic agent that reduces both the viscosity and adherence of bronchopulmonary mucus and is reported to modestly increase nasal bioavailability of human growth hormone in anesthetized rats (from 7.5 to 12.2%). These and other mucolytic or mucus-clearing agents are contacted with the nasal mucosa, typically in a concentration range of about 0.2 to 20 mM, coordinately with administration of the biologically active agent, to reduce the polar viscosity and/or elasticity of intranasal mucus.
[0147] Still other mucolytic or mucus-clearing agents may be selected from a range of glycosidase enzymes, which are able to cleave glycosidic bonds within the mucus glycoprotein a-amylase and B-amylase are representative of this class of enzymes, although their mucolytic effect may be limited. In contrast, bacterial glycosidases which allow these microorganisms to permeate mucus layers of their hosts.
[0148] For combinatorial use with most biologically active agents within the invention, including peptide and protein therapeutics, non-ionogenic detergents are generally also useful as mucolytic or mucus-clearing agents. These agents typically will not modify or substantially impair the activity of therapeutic polypeptides. Ciliostatic Agents and Methods
[0149] Because the self-cleaning capacity of certain mucosal tissues (e.g., nasal mucosal tissues) by mucociliary clearance is necessary as a protective function (e.g., to remove dust, allergens, and bacteria), it has been generally considered that this function should not be substantially impaired by mucosal medications. Mucociliary transport in the respiratory tract is a particularly important defense mechanism against infections. To achieve this function, ciliary beating in the nasal and airway passages moves a layer of mucus along the mucosa to removing inhaled particles and microorganisms.
[0150] Ciliostatic agents find use within the methods and compositions of the invention to increase the residence time of mucosally (e.g., intranasally) administered biologically active agents against viruses disclosed herein. In particular, the delivery these agents within the methods and compositions of the invention is significantly enhanced in certain aspects by the coordinate administration or combinatorial formulation of one or more ciliostatic agents that function to reversibly inhibit ciliary activity of mucosal cells, to provide for a temporary, reversible increase in the residence time of the mucosally administered active agent(s). For use within these aspects of the invention, the foregoing ciliostatic factors, either specific or indirect in their activity, are all candidates for successful employment as ciliostatic agents in appropriate amounts (depending on concentration, duration and mode of delivery) such that they yield a transient (i.e., reversible) reduction or cessation of mucociliary clearance at a mucosal site of administration to enhance delivery of biologically active agents disclosed herein, without unacceptable adverse side effects.
[0151] Within more detailed aspects, a specific ciliostatic factor is employed in a combined formulation or coordinate administration protocol with one or more Y2 receptor-binding peptide proteins, analogs and mimetics, and/or other biologically active agents disclosed herein. Various bacterial ciliostatic factors isolated and characterized in the literature may be employed within these embodiments of the invention. Ciliostatic factors from the bacterium Pseudomonas aeruginosa include a phenazine derivative, a pyo compound (2-alkyl-4- hydroxy quinolines), and a rhamnolipid (also known as a hemolysin). The pyo compound produced ciliostasis at concentrations of 50 pg/ml and without obvious ultrastructural lesions. The phenazine derivative also inhibited ciliary motility but caused some membrane disruption, although at substantially greater concentrations of 400 pg/ml. Limited exposure of tracheal explants to the rhamnolipid resulted in ciliostasis, which was associated with altered ciliary membranes. More extensive exposure to rhamnolipid was associated with removal of dynein arms from axonemes. Surface Active Agents and Methods
[0152] Within more detailed aspects of the invention, one or more membrane penetration enhancing agents may be employed within a mucosal delivery method or formulation of the invention to enhance mucosal delivery biologically active agents disclosed herein. Membrane penetration enhancing agents in this context can be selected from: (i) a surfactant, (ii) a bile salt, (iii) a phospholipid additive, mixed micelle, liposome, or carrier, (iv) an alcohol, (v) an enamine, (vi) an NO donor compound, (vii) a long-chain amphipathic molecule (viii) a small hydrophobic penetration enhancer; (ix) sodium or a salicylic acid derivative; (x) a glycerol ester of acetoacetic acid (xi) a clyclodextrin or B-cyclodextrin derivative, (xii) a medium- chain fatty acid, (xiii) a chelating agent, (xiv) an amino acid or salt thereof, (xv) an N- acetylamino acid or salt thereof, (xvi) an enzyme degradative to a selected membrane component, (xvii) an inhibitor of fatty acid synthesis, or (xviii) an inhibitor of cholesterol synthesis; or (xix) any combination of the membrane penetration enhancing agents recited in (i) (xix).
[0153] Certain surface-active agents are readily incorporated within the mucosal delivery formulations and methods of the invention as mucosal absorption and/or adsorption enhancing agents. These agents, which may be coordinately administered or combinatorially formulated with other biologically active agents disclosed herein, may be selected from a broad assemblage of known surfactants. Surfactants, which generally fall into three classes: (1) nonionic polyoxyethylene ethers; (2) bile salts such as sodium glycocholate (SGC) and deoxycholate (DOC); and (3) derivatives of fusidic acid such as sodium taurodihydrofusidate (STDHF). The mechanisms of action of these various classes of surface-active agents typically include solubilization of the biologically active agent. For proteins and peptides which often form aggregates, the surface active properties of these absorption and/or adsorption promoters can allow interactions with proteins such that smaller units such as surfactant coated monomers may be more readily maintained in solution. Examples of other surface-active agents are L-a-Phosphatidylcholine Didecanoyl (DDPC) polysorbate 80 and polysorbate 20. These monomers are presumably more transportable units than aggregates. A second potential mechanism is the protection of the peptide or protein from proteolytic degradation by proteases in the mucosal environment. Both bile salts and some fusidic acid derivatives reportedly inhibit proteolytic degradation of proteins by nasal homogenates at concentrations less than or equivalent to those required to enhance protein absorption and/or adsorption. This protease inhibition may be especially important for peptides with short biological half-lives. Vasodilator Agents and Methods
[0154] While generally it is intended the formulations of this invention remain on the nasal mucosa to perform their virus inhibiting action, yet another class of absorption and/or adsorption-promoting agents that shows beneficial utility within the coordinate administration and combinatorial formulation methods and compositions of the invention are vasoactive compounds, more specifically vasodilators. These compounds function within the invention to modulate the structure and physiology of the submucosal vasculature, increasing the transport rate of biologically active agents into or through the mucosal epithelium and/or to specific target tissues or compartments (e.g., the systemic circulation or central nervous system.).
[0155] Vasodilator agents for use within the invention typically cause submucosal blood vessel relaxation by either a decrease in cytoplasmic calcium, an increase in nitric oxide (NO) or by inhibiting myosin light chain kinase. They are generally divided into 9 classes: calcium antagonists, potassium channel openers, ACE inhibitors, angiotensin-II receptor antagonists, a-adrenergic and imidazole receptor antagonists, Bl-adrenergic agonists, phosphodiesterase inhibitors, eicosanoids and NO donors.
[0156] Despite chemical differences, the pharmacokinetic properties of calcium antagonists are similar. Absorption and/or adsorption into the systemic circulation is high, and these agents therefore undergo considerable first-pass metabolism by the liver, resulting in individual variation in pharmacokinetics. Except for the newer drugs of the dihydropyridine type (amlodipine, felodipine, isradipine, nilvadipine, nisoldipine and nitrendipine), the half- life of calcium antagonists is short. Therefore, to maintain an effective drug concentration for many of these may require delivery by multiple dosing, or controlled release formulations, as described elsewhere herein. Treatment with the potassium channel opener minoxidil may also be limited in manner and level of administration due to potential adverse side effects.
[0157] ACE inhibitors prevent conversion of angiotensin-I to angiotensin-II, and are most effective when renin production is increased. Since ACE is identical to kininase-II, which inactivates the potent endogenous vasodilator bradykinin, ACE inhibition causes a reduction in bradykinin degradation. ACE inhibitors provide the added advantage of cardioprotective and cardioreparative effects, by preventing and reversing cardiac fibrosis and ventricular hypertrophy in animal models. The predominant elimination pathway of most ACE inhibitors is via renal excretion. Therefore, renal impairment is associated with reduced elimination and a dosage reduction of 25 to 50% is recommended in patients with moderate to severe renal impairment. [0158] With regard to NO donors, these compounds are particularly useful within the invention for their additional effects on mucosal permeability. In addition to the above-noted NO donors, complexes of NO with nucleophiles called NO/nucleophiles, orNONOates, spontaneously and nonenzymatically release NO when dissolved in aqueous solution at physiologic pH. In contrast, nitro vasodilators such as nitroglycerin require specific enzyme activity for NO release. NONOates release NO with a defined stoichiometry and at predictable rates ranging from <3 minutes for diethylamine/NO to approximately 20 hours for diethylenetriamine/NO (DETANO).
Polymeric Delivery Vehicles and Methods
[0159] Within certain aspects of the invention, biologically active agents disclosed herein, and delivery-enhancing agents as described above, are, individually or combinatorially, incorporated within a mucosally (e.g., nasally) administered formulation that includes a biocompatible polymer functioning as a carrier or base. Such polymer carriers include polymeric powders, matrices or microparticulate delivery vehicles, among other polymer forms. The polymer can be of plant, animal, or synthetic origin. Often the polymer is crosslinked. Additionally, in these delivery systems the Virus entry and infectivity inhibitor, can be functionalized in a manner where it can be covalently bound to the polymer and rendered inseparable from the polymer by simple washing. In other embodiments, the polymer is chemically modified with an inhibitor of enzymes or other agents which may degrade or inactivate the biologically active agent(s) and/or delivery enhancing agent(s). In certain formulations, the polymer is a partially or completely water insoluble but water swellable polymer, e.g., a hydrogel. Polymers useful in this aspect of the invention are desirably water interactive and/or hydrophilic in nature to absorb significant quantities of water, and they often form hydrogels when placed in contact with water or aqueous media for a period of time sufficient to reach equilibrium with water. In more detailed embodiments, the polymer is a hydrogel which, when placed in contact with excess water, absorbs at least two times its weight of water at equilibrium when exposed to water at room temperature. [0160] Drug delivery systems based on biodegradable polymers are preferred in many biomedical applications because such systems are broken down either by hydrolysis or by enzymatic reaction into non-toxic molecules. The rate of degradation is controlled by manipulating the composition of the biodegradable polymer matrix. These types of systems can therefore be employed in certain settings for long-term release of biologically active agents. Biodegradable polymers such as poly(glycolic acid) (PGA), poly-(lactic acid) (PLA), and poly(D,L-lactic-co-glycolic acid) (PLGA), have received considerable attention as possible drug delivery carriers, since the degradation products of these polymers have been found to have low toxicity. During the normal metabolic function of the body these polymers degrade into carbon dioxide and water. These polymers have also exhibited excellent biocompatibility.
[0161] For prolonging the biological activity of virus entry and infectivity inhibitor and other biologically active agents disclosed herein, as well as optional delivery-enhancing agents, these agents may be incorporated into polymeric matrices, e.g., polyorthoesters, polyanhydrides, or polyesters. This yields sustained activity and release of the active agent(s), e.g., as determined by the degradation of the polymer matrix. Although the encapsulation of biotherapeutic molecules inside synthetic polymers may stabilize them during storage and delivery, the largest obstacle of polymer-based release technology is the activity loss of the therapeutic molecules during the formulation processes that often involve heat, sonication or organic solvents.
[0162] Absorption and/or adsorption-promoting polymers contemplated for use within the invention may include derivatives and chemically or physically modified versions of the foregoing types of polymers, in addition to other naturally occurring or synthetic polymers, gums, resins, and other agents, as well as blends of these materials with each other or other polymers, so long as the alterations, modifications or blending do not adversely affect the desired properties, such as water absorption and/or adsorption, hydrogel formation, and/or chemical stability for useful application. In more detailed aspects of the invention, polymers such as nylon, acrylan and other normally hydrophobic synthetic polymers may be sufficiently modified by reaction to become water swellable and/or form stable gels in aqueous media.
[0163] Absorption and/or adsorption-promoting polymers of the invention may include polymers from the group of homo- and copolymers based on various combinations of the following vinyl monomers: acrylic and methacrylic acids, acrylamide, methacrylamide, hydroxyethyl acrylate or methacrylate, vinylpyrrolidones, as well as polyvinylalcohol and its co- and terpolymers, polyvinylacetate, its co- and terpolymers with the above listed monomers and 2-acrylamido-2-methyl-propanesulfonic acid (AMPS.RTM.). Very useful are copolymers of the above listed monomers with copolymerizable functional monomers such as acryl or methacryl amide acrylate or methacrylate esters where the ester groups are derived from straight or branched chain alkyl, aryl having up to four aromatic rings which may contain alkyl substituents of 1 to 6 carbons; steroidal, sulfates, phosphates or cationic monomers such as N,N-dimethylaminoalkyl(meth)acrylamide, dimethylaminoalkyl(meth)acrylate, (meth)acryloxyalkyltrimethylammonium chloride, (meth)acryloxyalkyldimethylbenzyl ammonium chloride.
[0164] Additional absorption and/or adsorption-promoting polymers for use within the invention are those classified as dextrans, dextrins, and from the class of materials classified as natural gums and resins, or from the class of natural polymers such as processed collagen, chitin, chitosan, pullalan, zooglan, alginates and modified alginates such as “Kelcoloid” (a polypropylene glycol modified alginate) gellan gums such as “Kelocogel”, Xanathan gums such as “Keltrol”, estastin, a-hydroxy butyrate and its copolymers, hyaluronic acid and its derivatives, polylactic and glycolic acids.
[0165] A very useful class of polymers applicable within the instant invention are olefmically-unsaturated carboxylic acids containing at least one activated carbon-to-carbon olefmic double bond, and at least one carboxyl group; that is, an acid or functional group readily converted to an acid containing an olefmic double bond which readily functions in polymerization because of its presence in the monomer molecule, either in the a-b position with respect to a carboxyl group, or as part of a terminal methylene grouping. Olefmically- unsaturated acids of this class include such materials as the acrylic acids typified by the acrylic acid itself, a-cyano acrylic acid, B-methylacrylic acid (crotonic acid), a-phenyl acrylic acid, B-acryloxy propionic acid, cinnamic acid, p-chloro cinnamic acid, l-carboxy-4-phenyl butadiene- 1,3, itaconic acid, citraconic acid, mesaconic acid, glutaconic acid, aconitic acid, maleic acid, fumaric acid, and tricarboxy ethylene. As used herein, the term “carboxylic acid” includes the polycarboxylic acids and those acid anhydrides, such as maleic anhydride, wherein the anhydride group is formed by the elimination of one molecule of water from two carboxyl groups located on the same carboxylic acid molecule.
[0166] Representative acrylates useful as absorption and/or adsorption-promoting agents within the invention include methyl acrylate, ethyl acrylate, propyl acrylate, isopropyl acrylate, butyl acrylate, isobutyl acrylate, methyl methacrylate, methyl ethacrylate, ethyl methacrylate, octyl acrylate, heptyl acrylate, octyl methacrylate, isopropyl methacrylate, 2- ethylhexyl methacrylate, nonyl acrylate, hexyl acrylate, n-hexyl methacrylate, and the like. Higher alkyl acrylic esters are decyl acrylate, isodecyl methacrylate, lauryl acrylate, stearyl acrylate, behenyl acrylate and melissyl acrylate and methacrylate versions thereof. Mixtures of two or three or more long chain acrylic esters may be successfully polymerized with one of the carboxylic monomers. Other comonomers include olefins, including alpha olefins, vinyl ethers, vinyl esters, and mixtures thereof. [0167] Other vinylidene monomers, including the acrylic nitriles, may also be used as absorption and/or adsorption-promoting agents within the methods and compositions of the invention to enhance delivery and adsorption of one or more Y2 receptor-binding peptide proteins, analogs and mimetics, and other biologically active agent(s), including to enhance delivery of the active agent(s) to a target tissue or compartment in the subject (e.g., the liver, hepatic portal vein, CNS tissue or fluid, or blood plasma). Useful alpha, beta-olefmically unsaturated nitriles are preferably monoolefmically unsaturated nitriles having from 3 to 10 carbon atoms such as acrylonitrile, methacrylonitrile, and the like. Most preferred are acrylonitrile and methacrylonitrile. Acrylic amides containing from 3 to 35 carbon atoms including monoolefmically unsaturated amides also may be used. Representative amides include acrylamide, methacrylamide, N-t-butyl acrylamide, N-cyclohexyl acrylamide, higher alkyl amides, where the alkyl group on the nitrogen contains from 8 to 32 carbon atoms, acrylic amides including N-alkylol amides of alpha, beta-olefmically unsaturated carboxylic acids including those having from 4 to 10 carbon atoms such as N-methylol acrylamide, N- propanol acrylamide, N-methylol methacrylamide, N-methylol maleimide, N-methylol maleamic acid esters, N-methylol-p-vinyl benzamide, and the like.
[0168] Yet additional useful absorption and/or adsorption promoting materials are alpha- olefins containing from 2 to 18 carbon atoms, more preferably from 2 to 8 carbon atoms; dienes containing from 4 to 10 carbon atoms; vinyl esters and allyl esters such as vinyl acetate; vinyl aromatics such as styrene, methyl styrene and chloro-styrene; vinyl and allyl ethers and ketones such as vinyl methyl ether and methyl vinyl ketone; chloroacrylates; cyanoalkyl acrylates such as alpha-cyanomethyl acrylate, and the alpha-, beta-, and gamma- cyanopropyl acrylates; alkoxyacrylates such as methoxy ethyl acrylate; haloacrylates as chloroethyl acrylate; vinyl halides and vinyl chloride, vinylidene chloride and the like; divinyls, diacrylates and other polyfunctional monomers such as divinyl ether, diethylene glycol diacrylate, ethylene glycol dimethacrylate, methylene-bis-acrylamide, allylpentaerythritol, and the like; and bis(beta-haloalkyl) alkenyl phosphonates such as bis(B- chloroethyl)vinyl phosphonate and the like as are known to those skilled in the art. Copolymers wherein the carboxy containing monomer is a minor constituent, and the other vinylidene monomers present as major components are readily prepared in accordance with the methods disclosed herein.
[0169] When hydrogels are employed as absorption and/or adsorption promoting agents within the invention, these may be composed of synthetic copolymers from the group of acrylic and methacrylic acids, acrylamide, methacrylamide, hydroxyethyl acrylate (HEA) or methacrylate (HEMA), and vinylpyrrolidones which are water interactive and swellable. Specific illustrative examples of useful polymers, especially for the delivery of peptides or proteins, are the following types of polymers: (meth)acrylamide and 0.1 to 99 wt. % (meth)acrylic acid; (meth)acrylamides and 0.1 75 wt % (meth)acryloxyethyl trimethyammonium chloride; (meth)acrylamide and 0.1 75 wt % (meth)acrylamide; acrylic acid and 0.1 75 wt % alkyl(meth)acrylates; (meth)acrylamide and 0.1 75 wt % AMPS.RTM. (trademark of Lubrizol Corp.); (meth)acrylamide and 0 to 30 wt % alkyl(meth)acrylamides and 0.1 75 wt % AMPS.RTM.; (meth)acrylamide and 0.1 99 wt. % HEMA; (metb)acrylamide and 0.1 to 75 wt % HEMA and 0.1 to 99% (meth)acrylic acid; (meth)acrylic acid and 0.1 99 wt % HEMA; 50 mole % vinyl ether and 50 mole % maleic anhydride; (meth)acrylamide and 0.1 to 75 wt % (meth)acryloxyalky dimethyl benzylammonium chloride; (meth)acrylamide and 0.1 to 99 wt % vinyl pyrrolidone; (meth)acrylamide and 50 wt % vinyl pyrrolidone and 0.1 99.9 wt % (meth)acrylic acid; (meth)acrylic acid and 0.1 to 75 wt % AMPS.RTM. and 0.1 75 wt % alkyl(meth)acrylamide. In the above examples, alkyl means Ci to C30, preferably Ci to C22, linear and branched and C4 to Ci6 cyclic; where (meth) is used, it means that the monomers with and without the methyl group are included. Other very useful hydrogel polymers are swellable, but insoluble versions of poly(vinyl pyrrolidone) starch, carboxym ethyl cellulose and polyvinyl alcohol. [0170] Additional polymeric hydrogel materials useful within the invention include (poly) hydroxyalkyl (meth)acrylate: anionic and cationic hydrogels: poly(electrolyte) complexes; poly(vinyl alcohols) having a low acetate residual: a swellable mixture of crosslinked agar and crosslinked carboxymethyl cellulose: a swellable composition comprising methyl cellulose mixed with a sparingly crosslinked agar; a water swellable copolymer produced by a dispersion of finely divided copolymer of maleic anhydride with styrene, ethylene, propylene, or isobutylene; a water swellable polymer of N-vinyl lactams; swellable sodium salts of carboxymethyl cellulose; and the like.
[0171] Other gelable, fluid imbibing and retaining polymers useful for forming the hydrophilic hydrogel for mucosal delivery of biologically active agents within the invention include pectin; polysaccharides such as agar, acacia, karaya, tragacenth, algins and guar and their crosslinked versions; acrylic acid polymers, copolymers and salt derivatives, polyacrylamides; water swellable indene maleic anhydride polymers; starch graft copolymers; acrylate type polymers and copolymers with water absorbability of about 2 to 400 times its original weight; diesters of polyglucan; a mixture of crosslinked poly(vinyl alcohol) and poly(N-vinyl-2-pyrrolidone); polyoxybutylene-polyethylene block copolymer gels; carob gum; polyester gels; poly urea gels; polyether gels; polyamide gels; polyimide gels; polypeptide gels; polyamino acid gels; poly cellulosic gels; crosslinked indene-maleic anhydride acrylate polymers; and polysaccharides.
[0172] Synthetic hydrogel polymers for use within the invention may be made by an infinite combination of several monomers in several ratios. The hydrogel can be crosslinked and generally possesses the ability to imbibe and absorb fluid and swell or expand to an enlarged equilibrium state. The hydrogel typically swells or expands upon delivery to the nasal mucosal surface, absorbing about 2 5, 5 10, 10 50, up to 50 100 or more times fold its weight of water. The optimum degree of swellability for a given hydrogel will be determined for different biologically active agents depending upon such factors as molecular weight, size, solubility and diffusion characteristics of the active agent carried by or entrapped or encapsulated within the polymer, and the specific spacing and cooperative chain motion associated with each individual polymer.
[0173] Hydrophilic polymers useful within the invention are water insoluble but water swellable. Such water-swollen polymers as typically referred to as hydrogels or gels. Such gels may be conveniently produced from water-soluble polymer by the process of crosslinking the polymers by a suitable crosslinking agent. However, stable hydrogels may also be formed from specific polymers under defined conditions of pH, temperature and/or ionic concentration, according to know methods in the art. Typically the polymers are cross linked, that is, cross-linked to the extent that the polymers possess good hydrophilic properties, have improved physical integrity (as compared to non cross-linked polymers of the same or similar type) and exhibit improved ability to retain within the gel network both the biologically active agent of interest and additional compounds for coadministration therewith such as a cytokine or enzyme inhibitor, while retaining the ability to release the active agent(s) at the appropriate location and time.
[0174] Generally, hydrogel polymers for use within the invention are crosslinked with a difunctional cross-linking in the amount of from 0.01 to 25 weight percent, based on the weight of the monomers forming the copolymer, and more preferably from 0.1 to 20 weight percent and more often from 0.1 to 15 weight percent of the crosslinking agent. Another useful amount of a crosslinking agent is 0.1 to 10 weight percent. Tri, tetra or higher multifunctional crosslinking agents may also be employed. When such reagents are utilized, lower amounts may be required to attain equivalent crosslinking density, i.e., the degree of crosslinking, or network properties that are sufficient to contain effectively the biologically active agent(s). [0175] The crosslinks can be covalent, ionic or hydrogen bonds with the polymer possessing the ability to swell in the presence of water containing fluids. Such crosslinkers and crosslinking reactions are known to those skilled in the art and in many cases are dependent upon the polymer system. Thus a crosslinked network may be formed by free radical copolymerization of unsaturated monomers. Polymeric hydrogels may also be formed by crosslinking preformed polymers by reacting functional groups found on the polymers such as alcohols, acids, amines with such groups as glyoxal, formaldehyde or glutaraldehyde, bis anhydrides and the like.
[0176] The polymers also may be cross-linked with any polyene, e.g. decadiene or trivinyl cyclohexane; acrylamides, such as N,N-methylene-bis(acrylamide); polyfunctional acrylates, such as trimethylol propane triacrylate; or polyfunctional vinylidene monomer containing at least 2 terminal CH2<groups, including, for example, divinyl benzene, divinyl naphthlene, allyl acrylates and the like. In certain embodiments, cross-linking monomers for use in preparing the copolymers are polyalkenyl polyethers having more than one alkenyl ether grouping per molecule, which may optionally possess alkenyl groups in which an olefmic double bond is present attached to a terminal methylene grouping (e.g., made by the etherification of a polyhydric alcohol containing at least 2 carbon atoms and at least 2 hydroxyl groups). Compounds of this class may be produced by reacting an alkenyl halide, such as allyl chloride or allyl bromide, with a strongly alkaline aqueous solution of one or more polyhydric alcohols. The product may be a complex mixture of polyethers with varying numbers of ether groups. Efficiency of the polyether cross-linking agent increases with the number of potentially polymerizable groups on the molecule. Typically, polyethers containing an average of two or more alkenyl ether groupings per molecule are used. Other cross-linking monomers include for example, diallyl esters, dimethallyl ethers, allyl or methallyl acrylates and acrylamides, tetravinyl silane, polyalkenyl methanes, diacrylates, and dimethacrylates, divinyl compounds such as divinyl benzene, polyallyl phosphate, diallyloxy compounds and phosphite esters and the like. Typical agents are allyl pentaerythritol, allyl sucrose, trimethyl olpropane triacrylate, 1,6-hexanediol diacrylate, trimethyl olpropane diallyl ether, pentaerythritol triacrylate, tetramethylene dimethacrylate, ethylene diacrylate, ethylene dimethacrylate, triethylene glycol dimethacrylate, and the like. Allyl pentaerythritol, trimethylolpropane diallylether and allyl sucrose provide suitable polymers. When the cross- linking agent is present, the polymeric mixtures usually contain between about 0.01 to 20 weight percent, e.g., 1%, 5%, or 10% or more by weight of cross-linking monomer based on the total of carboxylic acid monomer, plus other monomers. [0177] In more detailed aspects of the invention, mucosal delivery of biologically active agents disclosed herein, is enhanced by retaining the active agent(s) in a slow-release or enzymatically or physiologically protective carrier or vehicle, for example a hydrogel that shields the active agent from the action of the degradative enzymes. In certain embodiments, the active agent is bound by chemical means to the carrier or vehicle, to which may also be admixed or bound additional agents such as enzyme inhibitors, cytokines, etc. The active agent may alternately be immobilized through sufficient physical entrapment within the carrier or vehicle, e.g., a polymer matrix.
[0178] Polymers such as hydrogels useful within the invention may incorporate functional linked agents such as glycosides chemically incorporated into the polymer for enhancing intranasal bioavailability of active agents formulated therewith. Examples of such glycosides are glucosides, fructosides, galactosides, arabinosides, mannosides and their alkyl substituted derivatives and natural glycosides such as arbutin, phlorizin, amygdalin, digitonin, saponin, and indican. There are several ways in which a typical glycoside may be bound to a polymer. For example, the hydrogen of the hydroxyl groups of a glycoside or other similar carbohydrate may be replaced by the alkyl group from a hydrogel polymer to form an ether. Also, the hydroxyl groups of the glycosides may be reacted to esterify the carboxyl groups of a polymeric hydrogel to form polymeric esters in situ. Another approach is to employ condensation of acetobromoglucose with cholest-5-en-3B-ol on a copolymer of maleic acid. N-substituted polyacrylamides can be synthesized by the reaction of activated polymers with omega-aminoalkylglycosides: (1) (carbohydrate-spacer)(n)-polyacrylamide, 'pseudopolysaccharides'; (2) (carbohydrate spacer)(n)-phosphatidylethanolamine(m)- polyacrylamide, neoglycolipids, derivatives of phosphatidylethanolamine; (3) (carbohydrate- spacer)(n)-biotin(m)-polyacrylamide. These biotinylated derivatives may attach to lectins on the mucosal surface to facilitate absorption and/or adsorption of the biologically active agent(s), e.g., a polymer-encapsulated Y2 receptor-binding peptide.
[0179] Within more detailed aspects of the invention, one or more biologically active agents, disclosed herein, optionally including secondary active agents such as protease inhibitor(s), cytokine(s), additional modulator(s) of intercellular junctional physiology, etc., are modified and bound to a polymeric carrier or matrix. For example, this may be accomplished by chemically binding a peptide or protein active agent and other optional agent(s) within a crosslinked polymer network. It is also possible to chemically modify the polymer separately with an interactive agent such as a glycosidal containing molecule. In certain aspects, the biologically active agent(s), and optional secondary active agent(s), may be functionalized, i.e., wherein an appropriate reactive group is identified or is chemically added to the active agent(s). Most often an ethylenic polymerizable group is added, and the functionalized active agent is then copolymerized with monomers and a crosslinking agent using a standard polymerization method such as solution polymerization (usually in water), emulsion, suspension or dispersion polymerization. Often, the functionalizing agent is provided with a high enough concentration of functional or polymerizable groups to insure that several sites on the active agent(s) are functionalized. For example, in a polypeptide comprising 16 amine sites, it is generally desired to functionalize at least 2, 4, 5, 7, and up to 8 or more of the sites. [0180] After functionalization, the functionalized active agent(s) is/are mixed with monomers and a crosslinking agent that comprise the reagents from which the polymer of interest is formed. Polymerization is then induced in this medium to create a polymer containing the bound active agent(s). The polymer is then washed with water or other appropriate solvents and otherwise purified to remove trace unreacted impurities and, if necessary, ground or broken up by physical means such as by stirring, forcing it through a mesh, ultrasoni cation or other suitable means to a desired particle size. The solvent, usually water, is then removed in such a manner as to not denature or otherwise degrade the active agent(s). One desired method is lyophilization (freeze drying) but other methods are available and may be used (e.g., vacuum drying, air drying, spray drying, etc.).
[0181] To introduce polymerizable groups in peptides, proteins and other active agents within the invention, it is possible to react available amino, hydroxyl, thiol and other reactive groups with electrophiles containing unsaturated groups. For example, unsaturated monomers containing N-hydroxy succinimidyl groups, active carbonates such as p-nitrophenyl carbonate, trichlorophenyl carbonates, tresylate, oxycarbonylimidazoles, epoxide, isocyanates and aldehyde, and unsaturated carboxymethyl azides and unsaturated orthopyridyl-disulfide belong to this category of reagents. Illustrative examples of unsaturated reagents are allyl glycidyl ether, allyl chloride, allylbromide, allyl iodide, acryloyl chloride, allyl isocyanate, allylsulfonyl chloride, maleic anhydride, copolymers of maleic anhydride and allyl ether, and the like.
[0182] All of the lysine active derivatives, except aldehyde, can generally react with other amino acids such as imidazole groups of histidine and hydroxyl groups of tyrosine and the thiol groups of cystine if the local environment enhances nucleophilicity of these groups. Aldehyde containing functionalizing reagents are specific to lysine. These types of reactions with available groups from lysines, cysteines, tyrosine have been extensively documented in the literature and are known to those skilled in the art. [0183] In the case of biologically active agents that contain amine groups, it is convenient to react such groups with an acyloyl chloride, such as acryloyl chloride, and introduce the polymerizable acrylic group onto the reacted agent. Then during preparation of the polymer, such as during the crosslinking of the copolymer of acrylamide and acrylic acid, the functionalized active agent, through the acrylic groups, is attached to the polymer and becomes bound thereto.
[0184] In additional aspects of the invention, biologically active agents, including peptides, proteins, nucleosides, and other molecules which are bioactive in vivo, are conjugation- stabilized by covalently bonding one or more active agent(s) to a polymer incorporating as an integral part thereof both a hydrophilic moiety, e.g., a linear polyalkyl ene glycol, a lipophilic moiety. In one aspect, a biologically active agent is covalently coupled with a polymer comprising (i) a linear polyalkylene glycol moiety and (ii) a lipophilic moiety, wherein the active agent, linear polyalkylene glycol moiety, and the lipophilic moiety are conformationally arranged in relation to one another such that the active therapeutic agent has an enhanced in vivo resistance to enzymatic degradation (i.e., relative to its stability under similar conditions in an unconjugated form devoid of the polymer coupled thereto). In another aspect, the conjugation-stabilized formulation has a three-dimensional conformation comprising the biologically active agent covalently coupled with a polysorbate complex comprising (i) a linear polyalkylene glycol moiety and (ii) a lipophilic moiety, wherein the active agent, the linear polyalkylene glycol moiety and the lipophilic moiety are conformationally arranged in relation to one another such that (a) the lipophilic moiety is exteriorly available in the three-dimensional conformation, and (b) the active agent in the composition has an enhanced in vivo resistance to enzymatic degradation.
[0185] In a further related aspect, a multiligand conjugated complex is provided which comprises a biologically active agent covalently coupled with a triglyceride backbone moiety through a polyalkylene glycol spacer group bonded at a carbon atom of the triglyceride backbone moiety, and at least one fatty acid moiety covalently attached either directly to a carbon atom of the triglyceride backbone moiety or covalently joined through a polyalkylene glycol spacer moiety. In such a multiligand conjugated therapeutic agent complex, the alpha and beta carbon atoms of the triglyceride bioactive moiety may have fatty acid moieties attached by covalently bonding either directly thereto, or indirectly covalently bonded thereto through polyalkylene glycol spacer moieties. Alternatively, a fatty acid moiety may be covalently attached either directly or through a polyalkylene glycol spacer moiety to the alpha and alpha carbons of the triglyceride backbone moiety, with the bioactive therapeutic agent being covalently coupled with the gamma-carbon of the triglyceride backbone moiety, either being directly covalently bonded thereto or indirectly bonded thereto through a polyalkylene spacer moiety. It will be recognized that a wide variety of structural, compositional, and conformational forms are possible for the multiligand conjugated therapeutic agent complex comprising the triglyceride backbone moiety, within the scope of the invention. It is further noted that in such a multiligand conjugated therapeutic agent complex, the biologically active agent(s) may advantageously be covalently coupled with the triglyceride modified backbone moiety through alkyl spacer groups, or alternatively other acceptable spacer groups, within the scope of the invention. As used in such context, acceptability of the spacer group refers to steric, compositional, and end use application specific acceptability characteristics.
[0186] In yet additional aspects of the invention, a conjugation-stabilized complex is provided which comprises a polysorbate complex comprising a polysorbate moiety including a triglyceride backbone having covalently coupled to alpha, alpha and beta carbon atoms thereof functionalizing groups including (i) a fatty acid group; and (ii) a polyethylene glycol group having a biologically active agent or moiety covalently bonded thereto, e.g., bonded to an appropriate functionality of the polyethylene glycol group. Such covalent bonding may be either direct, e.g., to a hydroxy terminal functionality of the polyethylene glycol group, or alternatively, the covalent bonding may be indirect, e.g., by reactively capping the hydroxy terminus of the polyethylene glycol group with a terminal carboxy functionality spacer group, so that the resulting capped polyethylene glycol group has a terminal carboxy functionality to which the biologically active agent or moiety may be covalently bonded.
[0187] In yet additional aspects of the invention, a stable, aqueously soluble, conjugation- stabilized complex is provided which comprises one or more biologically active agent(s)+disclosed herein covalently coupled to a physiologically compatible polyethylene glycol (PEG) modified glycolipid moiety. In such complex, the biologically active agent(s) may be covalently coupled to the physiologically compatible PEG modified glycolipid moiety by a labile covalent bond at a free amino acid group of the active agent, wherein the labile covalent bond is scissionable in vivo by biochemical hydrolysis and/or proteolysis. The physiologically compatible PEG modified glycolipid moiety may advantageously comprise a polysorbate polymer, e.g., a polysorbate polymer comprising fatty acid ester groups selected from the group consisting of monopalmitate, dipalmitate, monolaurate, dilaurate, trilaurate, monoleate, dioleate, trioleate, monostearate, distearate, and tristearate. In such complex, the physiologically compatible PEG modified glycolipid moiety may suitably comprise a polymer selected from the group consisting of polyethylene glycol ethers of fatty acids, and polyethylene glycol esters of fatty acids, wherein the fatty acids for example comprise a fatty acid selected from the group consisting of lauric, palmitic, oleic, and stearic acids.
[0188] Compositions according to the present invention are often administered in an aqueous solution as a nasal or pulmonary spray and may be dispensed in spray form by a variety of methods known to those skilled in the art. Additional aerosol delivery forms may include, e.g., compressed air-, jet-, ultrasonic-, and piezoelectric nebulizers, which deliver the biologically active agent dissolved or suspended in a pharmaceutical solvent, e.g., water, ethanol, or a mixture thereof.
[0189] Nasal and pulmonary spray solutions of the present invention typically comprise the drug or drug to be delivered, optionally formulated with a surface-active agent, such as a nonionic surfactant (e.g., polysorbate-80), and one or more buffers. In some embodiments of the present invention, the nasal spray solution further comprises a propellant. The pH of the nasal spray solution is optionally between about pH 3.0 and 6.0, preferably 5.0±0.3. Suitable buffers for use within these compositions are as described above or as otherwise known in the art. Other components may be added to enhance or maintain chemical stability, including preservatives, surfactants, dispersants, or gases. Suitable preservatives include, but are not limited to, phenol, methyl paraben, paraben, m-cresol, thiomersal, chlorobutanol, benzylalkonimum chloride, and the like. Suitable surfactants include, but are not limited to, oleic acid, sorbitan trioleate, polysorbates, lecithin, phosphotidyl cholines, and various long chain diglycerides and phospholipids. Suitable dispersants include, but are not limited to, ethylenediaminetetraacetic acid, and the like. Suitable gases include, but are not limited to, nitrogen, helium, chlorofluorocarbons (CFCs), hydrofluorocarbons (HFCs), carbon dioxide, air, and the like.
[0190] Within alternate embodiments, mucosal formulations are administered as dry powder formulations comprising the biologically active agent in a dry, usually lyophilized, form of an appropriate particle size, or within an appropriate particle size range, for intranasal delivery. Minimum particle size appropriate for deposition within the nasal or pulmonary passages is often about 0.5m mass median equivalent aerodynamic diameter (MMEAD), commonly about 1m MMEAD, and more typically about 2m MMEAD. Maximum particle size appropriate for deposition within the nasal passages is often about 10m MMEAD, commonly about 8m MMEAD, and more typically about 4m MMEAD. Intranasally respirable powders within these size ranges can be produced by a variety of conventional techniques, such as jet milling, spray drying, solvent precipitation, supercritical fluid condensation, and the like. These dry powders of appropriate MMEAD can be administered to a patient via a conventional dry powder inhaler (DPI), which rely on the patient's breath, upon pulmonary or nasal inhalation, to disperse the power into an aerosolized amount. Alternatively, the dry powder may be administered via air-assisted devices that use an external power source to disperse the powder into an aerosolized amount, e.g., a piston pump.
[0191] Dry powder devices typically require a powder mass in the range from about 1 mg to 20 mg to produce a single aerosolized dose (“puff’). If the required or desired dose of the biologically active agent is lower than this amount, the powdered active agent will typically be combined with a pharmaceutical dry bulking powder to provide the required total powder mass. Preferred dry bulking powders include sucrose, lactose, dextrose, mannitol, glycine, trehalose, human serum albumin (HSA), and starch. Other suitable dry bulking powders include cellobiose, dextrans, maltotriose, pectin, sodium citrate, sodium ascorbate, and the like.
[0192] To formulate compositions for mucosal delivery within the present invention, the biologically active agent can be combined with various pharmaceutically acceptable additives, as well as a base or carrier for dispersion of the active agent(s). Desired additives include, but are not limited to, pH control agents, such as arginine, sodium hydroxide, glycine, hydrochloric acid, citric acid, etc. In addition, local anesthetics (e.g., benzyl alcohol), isotonizing agents (e.g., sodium chloride, mannitol, sorbitol), adsorption inhibitors (e.g., Tween 80), solubility enhancing agents (e.g., cyclodextrins and derivatives thereof), stabilizers (e.g., serum albumin), humectants, and reducing agents (e.g., glutathione) can be included. When the composition for mucosal delivery is a liquid, the tonicity of the formulation, as measured with reference to the tonicity of 0.9% (w/v) physiological saline solution taken as unity, is typically adjusted to a value at which no substantial, irreversible tissue damage will be induced in the nasal mucosa at the site of administration. Generally, the tonicity of the solution is adjusted to a value of about 1/3 to 3, more typically 1/2 to 2, and most often 3/4 to 1.7.
Administration
[0193] Diseases from coronaviruses, including SARS-CoV-2, can be prevented or treated by administering pharmaceuticals intranasally using a nasal spray or aerosol. This is surprising because coronaviruses are thought to infect the lungs initially and that many inhibitors, proteins and peptides have been shown to be sheared or denatured due to the mechanical forces generated by the actuator in producing the spray or aerosol. Nasal administration of a composition may reduce spread of the virus to the lungs from the nasal cavity, thereby reducing respiratory effects of the virus. In some embodiments, a method of treating or preventing a coronavirus infection may comprise administering a composition described here. In some embodiments, a composition of the present disclosure may be administered as a nasal spray to treat or prevent a coronavirus infection. In some embodiments, treating a coronavirus infection may comprise reducing the infectivity of the coronavirus. In some embodiments, preventing a coronavirus infection may comprise reducing the infectivity of the coronavirus.
[0194] A composition of the present disclosure (e.g., a composition for the treatment or prevention of a SARS-CoV-2 infection) may be formulated for nasal delivery and administered via a nasal spray. A nasal spray formulation may be contained within a nasal spray bottle comprising a spray nozzle and a cap. In some embodiments, the bottle may comprise an opaque or semi-opaque material (e.g., opaque plastic or amber glass). In some embodiments, a subject (e.g., a human subject who may have been exposed to SARS-CoV-2) may administer a nasal spray formulation of the present disclosure by gently blowing nose to clear nostrils, shaking the bottle, holding the bottle with thumb under the bottle and the spray nozzle between fingers, close off one nostril by pressing fingers against the outside of the nose, insert the spray nozzle into the other nostril until fingers contact the nose, aim the nozzle toward the back of the nose, and spray into the nostril while sniffing gently. These steps may be repeated for each nostril.
[0195] In this area the following definitions are useful. Aerosol is a product that is packaged under pressure and contains therapeutically active ingredients that are released upon activation of an appropriate valve system. Metered aerosol is a pressurized dosage form comprised of metered dose valves, which allow for the delivery of a uniform quantity of spray upon each activation. Powder aerosol is a product that is packaged under pressure and contains therapeutically active ingredients in the form of a powder, which are released upon activation of an appropriate valve system. Spray aerosol is an aerosol product that utilizes a compressed gas as the propellant to provide the force necessary to expel the product as a wet spray; it generally applicable to solutions of medicinal agents in aqueous solvents. Spray is a liquid minutely divided as by a jet of air or steam. Nasal spray drug products contain therapeutically active ingredients dissolved or suspended in solutions or mixtures of excipients in non-pressurized dispensers. Metered spray is a non-pressurized dosage form consisting of valves that allow the dispensing of a specified quantity of spray upon each activation. Suspension spray is a liquid preparation containing solid particles dispersed in a liquid vehicle and in the form of course droplets or as finely divided solids.
[0196] The fluid dynamic characterization of the aerosol spray emitted by metered nasal spray pumps as a drug delivery device (“DDD”). Spray characterization is an integral part of the regulatory submissions necessary for Food and Drug Administration (“FDA”) approval of research and development, quality assurance and stability testing procedures for new and existing nasal spray pumps.
Spray Patterns and Plume Geometries for Nasal Delivery
[0197] Thorough characterization of the spray's geometry has been found to be a good indicator of the overall performance of nasal spray pumps. In particular, measurements of the spray's divergence angle (plume geometry) as it exits the device; the spray's cross-sectional ellipticity, uniformity and particle/droplet distribution (spray pattern); and the time evolution of the developing spray have been found to be the most representative performance quantities in the characterization of a nasal spray pump. During quality assurance and stability testing, plume geometry and spray pattern measurements are key identifiers for verifying consistency and conformity with the approved data criteria for the nasal spray pumps.
[0198] Plume Height is the measurement from the actuator tip to the point at which the plume angle becomes non-linear because of the breakdown of linear flow. Based on a visual examination of digital images, and to establish a measurement point for width that is consistent with the farthest measurement point of spray pattern. In some embodiments, a height of 30 mm may be used for assessing plume geometry. Major Axis, or Dmax, is the largest chord that can be drawn within the fitted spray pattern that crosses the center of mass of the spray pattern (COMw) in base units (mm). Minor Axis is the smallest chord that can be drawn within the fitted spray pattern that crosses the COMw in base units (mm). Ellipticity Ratio, or ovality, is the ratio of the major axis to the minor axis. Dio is the diameter of droplet for which 10% of the total liquid volume of sample consists of droplets of a smaller diameter (pm). D50 is the diameter of droplet for which 50% of the total liquid volume of sample consists of droplets of a smaller diameter (pm), also known as the mass median diameter. D90 is the diameter of droplet for which 90% of the total liquid volume of sample consists of droplets of a smaller diameter (pm). Span is ameasurement of the width of the distribution. The smaller the value, the narrower the distribution. Span is calculated as: (D90 - DIO)D5O. Percent relative standard deviation (% RSD) is the standard deviation divided by the mean of the series and multiplied by 100, also known as % CV. [0199] In some embodiments, a nasal spray pump may be used to nasally administer a formulation to treat or prevent a respiratory viral infection (e.g., a coronavirus infection). A nasal spray pump may produce a spray pattern with a maximum diameter (Dmax) of from about 10 mm to about 50 mm, from about 20 mm to about 50 mm, from about 30 mm to about 50 mm, from about 40 mm to about 50 mm, from about 10 mm to about 40 mm, from about 20 mm to about 40 mm, from about 30 mm to about 40 mm, from about 10 mm to about 30 mm, from about 20 mm to about 30 mm, or from about 10 mm to about 20 mm. A nasal spray pump may produce a spray pattern with an ovality of from about 1 to about 2, from about 1.2 to about 2, from about 1.3 to about 2, from about 1.4 to about 2, from about 1.5 to about 2, from about 1 to about 1.5, from about 1.2 to about 1.5, from about 1.3 to about 1.5, from about 1.4 to about 1.5, from about 1 to about 1.4, from about 1.2 to about 1.4, from about 1.3 to about 1.4, from about 1 to about 1.3, from about 1.2 to about 1.3, from about 1 to about 1.25, from about 1.15 to about 1.25, from about 1.2 to about 1.25, from about 1 to about 1.2, from about 1.15 to about 1.2, or from about 1 to about 1.15. A nasal spray pump may produce a spray pattern with an area of from about 200 mm2 to about 800 mm2, from about 300 mm2 to about 800 mm2, from about 400 mm2 to about 800 mm2, from about 500 mm2 to about 800 mm2, from about 600 mm2 to about 800 mm2, from about 700 mm2 to about 800 mm2, from about 200 mm2 to about 700 mm2, from about 300 mm2 to about 700 mm2, from about 400 mm2 to about 700 mm2, from about 500 mm2 to about 700 mm2, from about 600 mm2 to about 700 mm2, from about 200 mm2 to about 600 mm2, from about 300 mm2 to about 600 mm2, from about 400 mm2 to about 600 mm2, from about 500 mm2 to about 600 mm2, from about 200 mm2 to about 500 mm2, from about 300 mm2 to about 500 mm2, from about 400 mm2 to about 500 mm2, from about 200 mm2 to about 400 mm2, from about 300 mm2 to about 400 mm2, or from about 200 mm2 to about 300 mm2.
[0200] In some embodiments, a nasal spray pump may produce a plume geometry with a plume angle of from about 30° to about 70°, from about 40° to about 70°, from about 50° to about 70°, from about 60° to about 70°, from about 30° to about 60°, from about 40° to about 60°, from about 50° to about 60°, from about 30° to about 50°, from about 40° to about 50°, or from about 30° to about 40°. A nasal spray pump may produce a plume geometry with a plume width of from about 15 mm to about 40 mm, from about 20 mm to about 40 mm, from about 25 mm to about 40 mm, from about 30 mm to about 40 mm, from about 35 mm to about 40 mm, from about 15 mm to about 35 mm, from about 20 mm to about 35 mm, from about 25 mm to about 35 mm, from about 30 mm to about 35 mm, from about 15 mm to about 30 mm, from about 20 mm to about 30 mm, from about 25 mm to about 30 mm, from about 15 mm to about 25 mm, from about 20 mm to about 25 mm, or from about 15 mm to about 20 mm.
[0201] In some embodiments, a nasal spray pump may produce a droplet size distribution of which the Dio is from about 10 pm to about 30 pm, from about 12 pm to about 30 pm, from about 15 pm to about 30 pm, from about 17 pm to about 30 pm, from about 20 pm to about 30 pm, from about 25 pm to about 30 pm, from about 10 pm to about 25 pm, from about 12 pm to about 25 pm, from about 15 pm to about 25 pm, from about 17 pm to about 25 pm, from about 20 pm to about 25 pm, from about 10 pm to about 20 pm, from about 12 pm to about 20 pm, from about 15 pm to about 20 pm, from about 17 pm to about 20 pm, from about 10 pm to about 17 pm, from about 12 pm to about 17 pm, from about 15 pm to about 17 pm, from about 10 pm to about 15 pm, from about 12 pm to about 15 pm, or from about 10 pm to about 12 pm.
[0202] A nasal spray pump may produce a droplet size distribution of which the D50 is from about 25 pm to about 75 pm, from about 30 pm to about 75 pm, from about 35 pm to about 75 pm, from about 40 pm to about 75 pm, from about 45 pm to about 75 pm, from about 50 pm to about 75 pm, from about 60 pm to about 75 pm, from about 70 pm to about 75 pm, from about 25 pm to about 70 pm, from about 30 pm to about 70 pm, from about 35 pm to about 70 pm, from about 40 pm to about 70 pm, from about 45 pm to about 70 pm, from about 50 pm to about 70 pm, from about 60 pm to about 70 pm, from about 25 pm to about 60 pm, from about 30 pm to about 60 pm, from about 35 pm to about 60 pm, from about 40 pm to about 60 pm, from about 45 pm to about 60 pm, from about 50 pm to about 60 pm, from about 25 pm to about 50 pm, from about 30 pm to about 50 pm, from about 35 pm to about 50 pm, from about 40 pm to about 50 pm, from about 45 pm to about 50 pm, from about 25 pm to about 45 pm, from about 30 pm to about 45 pm, from about 35 pm to about 45 pm, from about 40 pm to about 45 pm, from about 25 pm to about 40 pm, from about 30 pm to about 40 pm, from about 35 pm to about 40 pm, from about 25 pm to about 35 pm, from about 30 pm to about 35 pm, or from about 25 pm to about 30 pm.
[0203] A nasal spray pump may produce a droplet size distribution of which the D90 is from about 70 pm to about 150 pm, from about 80 pm to about 150 pm, from about 90 pm to about 150 pm, from about 100 pm to about 150 pm, from about 110 pm to about 150 pm, from about 130 pm to about 150 pm, from about 70 pm to about 130 pm, from about 80 pm to about 130 pm, from about 90 pm to about 130 pm, from about 100 pm to about 130 pm, from about 110 pm to about 130 pm, from about 70 pm to about 110 pm, from about 80 pm to about 110 pm, from about 90 pm to about 110 pm, from about 100 pm to about 110 pm, from about 70 mih to about 100 mih, from about 80 mih to about 100 mm, from about 90 mih to about 100 mih, from about 70 mih to about 90 mih, from about 80 mm to about 90 mih, or from about 70 mih to about 80 mih.
[0204] A nasal spray pump may produce a droplet size distribution of which the span is from about 1.5 to about 1.9, from about 1.6 to about 1.9, from about 1.65 to about 1.9, from about 1.7 to about 1.9, from about 1.75 to about 1.9, from about 1.8 to about 1.9, from about 1.5 to about 1.8, from about 1.6 to about 1.8, from about 1.65 to about 1.8, from about 1.7 to about 1.8, from about 1.75 to about 1.8, from about 1.5 to about 1.75, from about 1.6 to about 1.75, from about 1.65 to about 1.75, from about 1.7 to about 1.75, from about 1.5 to about 1.7, from about 1.6 to about 1.7, from about 1.65 to about 1.7, from about 1.5 to about 1.65, from about 1.6 to about 1.65, or from about 1.5 to about 1.6.
[0205] A nasal spray pump may produce a droplet size distribution of which the percent of volume below 10 pm is from about 0.40% to about 6%, from about 1% to about 6%, from about 2% to about 6%, from about 3% to about 6%, from about 4% to about 6%, from about 5% to about 6%, from about 0.40% to about 5%, from about 1% to about 5%, from about 2% to about 5%, from about 3% to about 5%, from about 4% to about 5%, from about 0.40% to about 4%, from about 1% to about 4%, from about 2% to about 4%, from about 3% to about 4%, from about 0.40% to about 3%, from about 1% to about 3%, from about 2% to about 3%, from about 0.40% to about 2%, from about 1% to about 2%, or from about 0.40% to about 1%. A nasal spray pump may produce a droplet size distribution of which the percent of volume below 10 pm is at least about 0.4%, at least about 1%, at least about 2%, at least about 3%, at least about 4%, or at least about 5%.
Devices for Nasal Delivery
[0206] A number of devices, such as nasal spray pumps, may be used to nasally administer the compositions of the present disclosure to treat or prevent a respiratory viral infection (e.g., a coronavirus infection such as COVID-19). In some embodiments, a device may be selected for a desired spray pattern, plume geometry, or droplet size. In some embodiments, a device may be selected based on formulation viscosity. For example, a device may be selected to accurately dispense the desired dosage and produce a wide spray pattern when administering a high viscosity formulation. In some embodiments, a device may be selected to reproducibly produce a uniform plume with a desired plume width (e.g., a narrow plume or a wide plume). [0207] In some embodiments, a device for nasal delivery may delivery a composition of the present disclosure at a spray velocity of from about 90 mm/s to about 110 mm/s. The device may produce a spray pattern with a maximum diameter (Dmax) of from about 20 mm to about 32.5 mm, an ovality of from about 1.17 to about 1.38, and an area of about 277 mm2 to about 685 mm2. The device may produce a plume geometry with a plume angle of from about 37° to about 58.2° and a plume width of from about 20 mm to about 33.5 mm. Examples of spray patterns and plume geometries produced by devices for nasal delivery of formulations for treatment or prevention of a respiratory viral infection are shown in FIG. 6 and FIG. 7. In some embodiments, a device for nasal delivery of a formulation may produce a droplet size distribution of which the Dio is from about 13.1 pm to about 27.2 pm, the D50 is from about 33.9 pm to about 70.9 pm, the D90 is from about 81.1 pm to about 146.5 pm, the span is from about 1.674 pm to about 1.8 pm, and the percent of volume below 10 pm is from about 0.41% to about 4.5%.
[0208] FIG. 6 shows the spray pattern and plume geometry of a device that produces a spray pattern with a Dmax of about 28.2 mm, an ovality of about 1.219, an area of about 508 mm2. The device produces a plume geometry with a plume angle of about 41.9° and a plume width of about 24.5 mm. The device produces droplets with a size distribution of which the Dio is about 19.2 pm, the D50 is about 49.2 pm, the D90 is about 106.6 pm, the span is about 1.8 pm, and the percent of volume below 10 pm is about 1.0%.
[0209] FIG. 7 shows the spray pattern and plume geometry of a device that produces a spray pattern with a Dmax of about 29 mm, an ovality of about 1.20, an area of about 542 mm2. The device produces a plume geometry with a plume angle of about 44° and a plume width of about 24 mm. The device produces droplets with a size distribution of which the Dio is about 18.7 pm, the D50 is about 49.0 pm, the D90 is about 106.5 pm, the span is about 1.792 pm, and the percent of volume below 10 pm is about 1.35%.
Nasal Delivery Administration Schedule
[0210] A composition of the present disclosure may be administered nasally in any of the spray patterns described herein. A composition may be administered nasally in from about 0.01 mL to about 0.1 mL, from about 0.05 mL to about 0.15 mL, from about 0.1 mL to about 0.2 mL, from about 0.01 mL to about 0.3 mL, or from about 0.05 mL to about 0.5 mL per spray. A composition of the present disclosure may be administered nasally about 1, 2, 3, 4,
5, 6, 7, 8, 9, or 10 times per day. A composition may be administered nasally for about 1, 2,
3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 days. The total amount of the composition administered during a course of treatment may be from about 1 mL to about 5 mL, from about 2 mL to about 6 mL, from about 3 mL to about 7 mL, from about 4 mL to about 8 mL, from about 5 mL to about 9 mL, from about 6 mL to about 10 mL, from about 7 mL to about 11 mL, from about 8 mL to about 12 mL, from about 9 mL to about 13 mL, or from about 10 mL to about 14 mL.
Pharmaceutical Formulations
[0211] A composition of the present disclosure (comprising one or more active agents) may be formulated as a pharmaceutical composition. A pharmaceutical composition may comprise a pharmaceutically acceptable carrier or excipient. As used herein “pharmaceutically acceptable” or “pharmacologically acceptable” includes molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a subject, as appropriate. “Pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients are often also incorporated into the compositions.
[0212] A pharmaceutical composition comprising an active agent of the present disclosure is formulated according to known methods to prepare pharmaceutically useful compositions, for example, as found in “Excipient Selection in Parenteral Formulation Development” Pramanick et. ah, Pharma Times, Vol. 45, No. 3, March 2013, incorporated in its entirety herein by reference. In some aspects, the active agent is combined with a pharmaceutically acceptable carrier. A composition is said to be a pharmaceutically acceptable carrier if its administration is tolerated by a recipient patient. Sterile phosphate-buffered saline is one example of a pharmaceutically acceptable carrier. Other suitable carriers are well-known to those in the art. See, for example, Gennaro (ed.), Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company 1995).
[0213] Formulations for administration of the active agents of the present disclosure are typically provided but are not limited to as liquid, solid or semi-solid products or dosage forms, exemplified by tablets, capsules, pellets, a powder or a lyophilized product. In some aspects, the active agent is formulated to comprise no additional materials except for a pharmaceutical carrier. In some other aspects, the active agent is formulated such that it comprises a core “matrix material” which encapsulates, binds to, coats or is adjacent to the active agent. In some other aspects, the active agent and matrix material further comprises a protective coating. Various formulations are well-known to those in the art. See, for example, Gennaro (ed.), Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company 1995).
[0214] Suitable excipients for use with the active agents of the present disclosure are often included in formulations for inhalation or for oral delivery. More specifically, formulations which include active agents and one or more but not limited to suitable excipients, exemplified by matrix materials, binders, lubricants, glidants or disintegrates which aid in modulating the pharmacokinetic (PK) profile of administered active agents are preferred. In some aspects, compositions comprising active agents in combination with one or more suitable excipients and one or more specific product characteristics (such as dissolution or water content) which result in improved pharmacokinetic profiles of active agents in vivo. Thus, the in vivo performance of active agent’s dosage forms/products included herein is based upon the composition of the excipients added during manufacturing and/or the final product characteristics generated through specific processing parameters and methods. Other excipients are well-known to those in the art. See, for example, Gennaro (ed.), Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company 1995).
[0215] Suitable carriers for intravenous administration include for example but are not limited to physiological saline or phosphate buffered saline (PBS), Tris, and solutions containing solubilizing agents, such as glucose, polyethylene glycol, polypropylene glycol, additional agents such as histidine, dextrose, mannitol and mixtures thereof. In some aspects, carriers for intravenous administration include a mixture of histidine and dextrose, Tris and dextrose or Tris and mannitol. Other carriers are well-known to those in the art. See, for example, Gennaro (ed.), Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company 1995).
[0216] The formulation often includes an aqueous vehicle. Aqueous vehicles include, by way of example and without limitation, sodium chloride solution, Ringers solution, isotonic dextrose solution, sterile water solution, dextrose and lactated Ringers solution. Nonaqueous vehicles include, by way of example and without limitation, fixed oils of vegetable origin, cottonseed oil, com oil, sesame oil and peanut oil, benzyl benzoate, castor oil, N,N- dimethylacetamide, ethanol, dehydrated ethanol, glycerin, glycerol, N-methyl-2-pyrrolidone, polyethylene glycol and any derivative thereof, propylene glycol, safflower oil and soybean oil. Other vehicles are well-known to those in the art. See, for example, Gennaro (ed.), Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company 1995).
[0217] In some aspects, the composition the pharmaceutically acceptable carrier comprises an osmolyte. In some aspects, the osmolyte comprises a sugar, a sugar alcohol, or a combination thereof.
[0218] In certain aspects, the composition comprises a sugar alcohol selected from sorbitol, inositol, mannitol, xylitol and glycerol, or a combination thereof. In further aspects, the sugar alcohol comprises mannitol. In certain aspects, the composition comprises from 2% to 20% (wt/vol %) mannitol. In some aspects, the composition comprises from 2% to 10% (wt/vol %) mannitol. In further aspects, the composition comprises essentially 5% (wt/vol %) mannitol. [0219] In other aspects, the composition comprises a sugar. In certain aspects, the sugar is selected from trehalose, lactose, sucrose, glucose, galactose, maltose, mannose, fructose, dextrose, or a combination thereof. In additional aspects, the sugar is selected from trehalose, sucrose, or a combination thereof. In some aspects, the composition comprises from 1% to 40% (wt/vol %) of trehalose, sucrose, or a combination of trehalose and sucrose. In other aspects, the composition comprises from 1% to 20% (wt/vol %) of trehalose, sucrose, or a combination of trehalose and sucrose. In additional aspects, the composition comprises 2% (wt/vol %) of trehalose, sucrose, or a combination of trehalose and sucrose.
[0220] In certain aspects, the composition further comprises an osmolyte selected from glycine, carnitine, ethanolamine, their phosphates, mono sugars, or a combination thereof. [0221] In some aspects, the present compositions are isotonic. In other aspects, the compositions are essentially isotonic.
[0222] In certain aspects, the ionic strength of the composition is less than 50 mM. In other aspects, the ionic strength of the composition is less than 10 mM.
[0223] Antimicrobial agents in bacteriostatic or fungistatic concentrations are typically added to preparations packaged in multiple dose containers which include by way of example and without limitation, phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride. Other antimicrobial agents are well-known to those in the art. See, for example, Gennaro (ed.), Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company 1995).
[0224] Buffers include by way of example and without limitation, acetate, ammonium sulfate, ammonium hydroxide, arginine, aspartic acid, benzene sulfonic acid, benzoate sodium, benzoate acid, carbonate, sodium carbonate, carbon dioxide, citrate, diethanolamine, glucono delta lactone, glycine, glycine HC1, histidine, histidine HC1, hydrochloric acid, hydrobromic acid, lysine maleic acid, meglumine, methanesulfonic acid, monoethanolamine, phosphate, sodium phosphate, citrate, succinate sodium, sulfuric acid, tartarate sodium, tromethamine, sodium citrate, hydroxide, sodium hydroxide, Tris base, Tris base -65, Tris acetate, Tris HC1, and Tris HCl-65.
[0225] In various aspects, the pharmaceutically acceptable carrier comprises a buffer. In some aspects, the buffer is selected from tris, HEPES, histidine, ethylene diamine, or a combination thereof. In other aspects, the buffer is selected from tris, histidine, or a combination thereof. In further aspects, the buffer comprises histidine, which is optionally L- histidine. In additional aspects, the composition comprises at least 100 mM histidine. In further aspects, the composition comprises at least 50 mM histidine. In some aspects, the composition comprises at least 20 mM histidine. In additional aspects, the composition comprises 10 to 100 mM histidine. In other aspects, the composition comprises 10 to 20 mM histidine.
[0226] Antioxidants include by way of example and without limitation, sodium bisulfate, acetone sodium bisulfate, argon, ascorbyl palmitate, ascorbate sodium, ascorbate acid, butylated hydroxy anisole, butylated hydroxy toluene, cysteine, cystenate HC1, dithionite sodium, gentistic acid, gentistic acid ethanoloamine, glutamate monosodium, glutathione, formaldehyde solfoxylate sodium, metabisulfite potassium, metabisulfite sodium, methionine, monothioglycerol, nitrogen, propyl gallate, sulfite sodium, tocopherol alpha, alpha tocopherol hydrogen succinate and thioglycolyate sodium.
[0227] In some aspects, the compositions comprise an antioxidant, a free radical scavenger, a quencher, an antioxidant synergist or a combination thereof.
[0228] In some aspects, the antioxidant is selected from methionine, butylated hydroxytoluene, butylated hydroxy anisole, propyl gallate, or a combination thereof. In other aspects, the antioxidant comprises methionine. In further aspects, the antioxidant is L- methionine. In certain aspects, the compositions comprise at least 20 mM methionine. In other aspects, aspects, the compositions comprise at least 10 mM methionine.
[0229] Suspending, emulsifying and/or dispersing agents include by way of example and without limitation, sodium carboxymethylcelluose, hydroxypropyl methylcellulose, Polysorbate 80 (TWEEN® 80) and polyvinylpyrrolidone.
[0230] In various aspects, the compositions comprise a surfactant. In certain aspects, the surfactant is selected from polysorbate 20, polysorbate 80, a pluronic, polyoxyethylene sorbitan mono-oleate, polyethylene mono-laureate, N-actylglucoside, or a combination thereof. In certain aspects, the surfactant is polysorbate 20. In further aspects, the compositions comprise from 0.0001% to 0.1% (wt/vol %) polysorbate 20. In additional aspects, the compositions comprise cyclodextrin. In further aspects, the cyclodextrin comprises (2-hydroxypropyl)-P-cyclodextrin.
[0231] A sequestering or chelating agent of metal ions include by way of example and without limitation, calcium disodium EDTA, disodium EDTA, sodium EDTA, calcium versetaminde sodium, calteridol and DPTA. In some aspects, the present compositions comprise a metal chelator. In certain aspects, the metal chelator is selected from EDTA, deferoxamine mesylate, EGTA, fumaric acid, and malic acid, salts thereof, or combinations thereof. In further aspects, the metal chelator comprises EDTA or salts thereof. In certain aspects, the compositions have an EDTA concentration of about 0.1 mg/ml to about 1.0 mg/ml.
[0232] Other isotonic agents, buffers, antioxidants, anesthetics, suspending and dispersing agents, emulsifying agents and chelating agents are well-known to those in the art. See, for example, Gennaro (ed.), Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company 1995).
[0233] Pharmaceutical carriers also include, by way of example and without limitation, ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles and sodium hydroxide, hydrochloric acid, citric acid or lactic acid. Other pharmaceutical carriers are well-known to those in the art. See, for example, Gennaro (ed.), Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company 1995).
[0234] The active agents described herein are often formulated using a variety of parameters including by way of example and without limitation, pH, molarity, % weight/volume, % volume/volume and the like. Other factors considered in the formulation of, stability of, storage of, shipping of active agents include by way of example and without limitation, the gas environment, container material, container color, cap material, cap color, presence of additional aspects, such as antioxidants, stabilizers, photoprotective compounds, protectants, sugars, ion chelators, ion donors or the like. Any factor which serves as any one of the above factors known to one of ordinary skill in the art is often used with the active agents described herein but not limited as such.
[0235] The preparation of pharmaceutical or pharmacological compositions are known to those of skill in the art in light of the present disclosure. General techniques for formulation and administration are found in “Remington: The Science and Practice of Pharmacy, Twentieth Edition,” Lippincott Williams & Wilkins, Philadelphia, Pa. Tablets, capsules, pills, powders, granules, dragees, gels, slurries, ointments, solutions suppositories, injections, inhalants and aerosols are examples of such formulations.
[0236] The active agents are often stored at various temperatures, including by way of example and without limitation, freezing, for example at about -20° C., about -70° C., about -100° C., about -120° C., about -150° C., about -200° C. or more than about -200° C., cold storage, for example at about 10° C., about 5° C., about 4° C., about 2° C., about 0° C., about -2° C. or more than about -5° C., or any other suitable temperature such that the composition remains stable.
[0237] In some aspects, compositions comprising the compounds described herein are stored as lyophilized solids. In some aspects, the present disclosure provides methods for producing the lyophilized composition, the method comprising providing the composition; and lyophilizing the composition, thereby producing the lyophilized composition.
[0238] Using lyophilization, it is possible to store the compounds in a manner that maintains physiological or otherwise optimal pH, isotonicity and stability. Such materials include pH buffers, preservatives, tonicity adjusting agents, anti -oxidants, other polymers (e.g., viscosity adjusting agents or extenders) and excipients to stabilize the labile protein against the stresses of drying and storage of the dried product. Specific illustrative examples of such additives include phosphate, citrate, or borate buffers; thimerosal; sorbic acid; methyl or propyl paraben, and chlorobutanol preservatives; sodium chloride: polyvinyl alcohol, polyvinyl pyrrolidone; mannitol, dextrose, dextran, lactose, sucrose, ethylene diamine tetra-acetic acid, and the like. Suitable formulations, known in the art, (Remington's Pharmaceutical Sciences (latest edition), Mack Publishing Company, Easton, Pa.; Arakawa et al. (1990), supra; Carpenter et al. (1991), supra; and Pikal (1990), supra).
[0239] In certain aspects, the pharmaceutically acceptable carrier comprises a reconstitution stabilizer. In other aspects, the reconstitution stabilizer comprises a water-soluble polymer. In additional aspects, the water-soluble polymer is selected from a polaxamer, a polyol, a polyethylene glycol, a polyvinylalcohol, a hydroxyethyl starch, dextran, polyvinylpyrrolidene poly(acrylic acid), or a combination thereof.
[0240] The term “reconstitution stabilizer” means any excipient which is capable of preventing aggregation of a reconstituted protein in an aqueous medium. Excipients possessing the necessary characteristics for the present invention are well-known in the art and generally function by the mechanisms of charge repulsion, steric hindrance, hydrophobic binding or specific high-affinity binding to the dried protein. Exemplary excipients include various osmolytes, various salts, water soluble synthetic and natural polymers, surfactants, sulfated polysaccharides, carrier proteins, buffers and the like (Manning et al. (1989), Pharmaceutical Research, 6:903-918; and Paboiji, et al. (1994), Pharmaceutical Research, 11:764-771).
[0241] The present compounds and an effective amount of the reconstitution stabilizer are admixed under conditions effective to reduce aggregation of present compounds upon reconstitution with the reconstitution medium (e.g., a solvent and optionally other components such as antibacterials). The reconstitution stabilizer may be admixed with the compounds at a suitable time before, during or after reconstitution; preferably the reconstitution stabilizer will be pre-dissolved in the reconstitution medium. The compound is reconstituted at a temperature which is above the freezing point of the reconstitution medium, but which will not degrade the compound and which will not be deleterious to the reconstitution stabilizer; preferably the temperature will be between about 2° C. to 50° C. The time taken to mix the reconstitution stabilizer and the dried compound should be for a sufficient period to prepare a suitable admixture; preferably mixing will be for between about 1 to 30 minutes. Generally, the reconstituted formulation is used soon after reconstitution. [0242] In certain aspects, the present compositions are reconstituted from a lyophilized form. In other aspects, the present disclosure provides methods for producing the reconstituted composition, the method comprising providing a lyophilized composition; and reconstituting the composition with a solution to produce a reconstituted composition. In various aspects, the reconstituting solution comprises water. In some aspects, the reconstituting solution is selected from sterile water, physiological saline solution, glucose solution or other aqueous solvents (e.g., alcohols such as ethyl, n-propyl or isopropyl, butyl alcohol), or a combination thereof, which are capable of dissolving the dried composition and compatible with the selected administration route and which does not negatively interfere with the compound and the reconstitution stabilizers employed.
Therapeutic Applications
[0243] An active agent of the present disclosure may be used for various therapeutic applications. An active agent may be administered as a pharmaceutical composition. A pharmaceutical composition of the disclosure can be a combination of any active agent described herein with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients. The pharmaceutical composition facilitates administration of an active agent described herein to an organism. Pharmaceutical compositions can be administered in therapeutically-effective amounts as pharmaceutical compositions by various forms and routes including, for example, intravenous, subcutaneous, intramuscular, rectal, aerosol, parenteral, ophthalmic, pulmonary, transdermal, vaginal, optic, nasal, oral, inhalation, dermal, intra-articular, intrathecal, intranasal, and topical administration. A pharmaceutical composition can be administered in a local or systemic manner, for example, via injection of the active agent described herein directly into an organ, optionally in a depot.
[0244] Parenteral injections can be formulated for bolus injection or continuous infusion. The pharmaceutical compositions can be in a form suitable for parenteral injection as a sterile suspension, solution or emulsion in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of an active agent described herein in water-soluble form. Suspensions of active agents described herein can be prepared as oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions can contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. The suspension can also contain suitable stabilizers or agents which increase the solubility and/or reduces the aggregation of such active agents described herein to allow for the preparation of highly concentrated solutions. Alternatively, the active agents described herein can be lyophilized or in powder form for re-constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. In some embodiments, a purified active agent is administered intravenously.
[0245] An active agent of the disclosure can be applied directly to an organ, or an organ tissue or cells, during a surgical procedure. In some embodiments, an active agent may be applied directly to a cancerous tissue (e.g., a tumor). The active agents described herein can be administered topically and can be formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams, and ointments. Such pharmaceutical compositions can contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
[0246] In practicing the methods of treatment or use provided herein, therapeutically- effective amounts of the active agent described herein described herein are administered in pharmaceutical compositions to a subject suffering from a condition. In some instances, the pharmaceutical composition will affect the physiology of the animal, such as the immune system, inflammatory response, or other physiologic affect. In some embodiments, the subject is a mammal such as a human. A therapeutically-effective amount can vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compounds used, and other factors.
[0247] Pharmaceutical compositions can be formulated using one or more physiologically- acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations that can be used pharmaceutically. Formulation can be modified depending upon the route of administration chosen. Pharmaceutical compositions comprising an active agent described herein can be manufactured, for example, by expressing the active agent in a recombinant system, purifying the active agent, lyophilizing the active agent, mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or compression processes. The pharmaceutical compositions can include at least one pharmaceutically acceptable carrier, diluent, or excipient and compounds described herein as free-base or pharmaceutically-acceptable salt form.
[0248] Methods for the preparation of active agents described herein include formulating the active agent described herein with one or more inert, pharmaceutically-acceptable excipients or carriers to form a solid, semi-solid, or liquid composition. Solid compositions include, for example, powders, tablets, dispersible granules, capsules, cachets, and suppositories. These compositions can also contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and other pharmaceutically-acceptable additives.
[0249] “Product” or “dosage form” as used herein refers to any solid, semi-solid, lyophilized, aqueous, liquid or frozen formulation or preparation used for administration. Upon administration, the rate of release of an active moiety from a product is often greatly influenced by the excipients and/or product characteristics which make up the product itself. For example, an enteric coat on a tablet is designed to separate that tablet's contents from the stomach contents to prevent, for example, degradation of the stomach which often induces gastrointestinal discomfort or injury. According to the currently accepted conventional understanding, systemic exposure of the active moiety will be relatively insensitive to the small formulation changes.
[0250] Non-limiting examples of pharmaceutically-acceptable excipients can be found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams &
Wilkins 1999), each of which is incorporated by reference in its entirety.
[0251] An active agent of the present disclosure may be administered to a patient in an effective amount. The term “effective amount,” as used herein, can refer to a sufficient amount of an agent or a compound being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. Compositions containing such agents or compounds can be administered for prophylactic, enhancing, and/or therapeutic treatments. An appropriate “effective” amount in any individual case can be determined using techniques, such as a dose escalation study.
[0252] The methods, compositions, and kits of this disclosure can comprise a method to prevent, treat, arrest, reverse, or ameliorate the symptoms of a condition. The treatment can comprise treating a subject (e.g., an individual, a domestic animal, a wild animal or a lab animal afflicted with a disease or condition) with an active agent of the disclosure. Active agents of the present disclosure may be administered to treat a disease in a subject. The subject can be a human. A subject can be a human; a non-human primate such as a chimpanzee, or other ape or monkey species; a farm animal such as cattle, horse, sheep, goat, swine; a domestic animal such as a rabbit, dog, and cat; a laboratory animal including a rodent, such as a rat, mouse and guinea pig, or the like. A subject can be of any age. A subject can be, for example, an elderly adult, adult, adolescent, pre-adolescent, child, toddler, infant, or fetus in utero.
[0253] Treatment can be provided to the subject before clinical onset of disease. Treatment can be provided to the subject after clinical onset of disease. Treatment can be provided to the subject after 1 day, 1 week, 6 months, 12 months, or 2 years or more after clinical onset of the disease. Treatment may be provided to the subject for more than 1 day, 1 week, 1 month,
6 months, 12 months, 2 years or more after clinical onset of disease. Treatment may be provided to the subject for less than 1 day, 1 week, 1 month, 6 months, 12 months, or 2 years after clinical onset of the disease. Treatment can also include treating a human in a clinical trial. A treatment can comprise administering to a subject a pharmaceutical composition, such as one or more of the pharmaceutical compositions described throughout the disclosure. A treatment can comprise a once daily dosing. A treatment can comprise delivering an active agent of the disclosure to a subject, either intravenously, subcutaneously, intramuscularly, by inhalation, dermally, intra-articular injection, orally, intrathecally, transdermally, intranasally, via a peritoneal route, or directly onto or into a diseased tissue, e.g., via topical, intra-articular injection route or injection route of application.
[0254] In some embodiments, the present disclosure provides a method for treating a cancer, the method comprising administering to a subject in need thereof an effective amount of an active agent of the present disclosure.
[0255] In some embodiments, the present disclosure provides a method for treating a cancer, the method comprising administering to a patient in need thereof an effective amount of a pharmaceutical composition comprising an active agent of the present disclosure and a pharmaceutically acceptable carrier.
Kits
[0256] The instant invention also includes kits, packages and multi-container units containing the above described pharmaceutical compositions, active ingredients, or means for administering the same for use in the prevention and treatment of diseases and other conditions in mammalian subjects. An active agent of the present disclosure may be provided in various kits. In some embodiments, pharmaceutical compositions comprising an active agent of the present disclosure may be supplied as a kit. A kit may comprise a container that comprises an active agent. Therapeutic active agents can be provided in the form of an injectable solution for single or multiple doses, or as a sterile powder that will be reconstituted before injection. Alternatively, such a kit can include a dry-powder disperser, liquid aerosol generator, or nebulizer for administration of a therapeutic active agents. Such a kit may further comprise written information on indications and usage of the pharmaceutical composition.
[0257] Unless otherwise defined, all technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. Any reference to “or” herein is intended to encompass “and/or” unless otherwise stated.
[0258] Whenever the term “at least,” “greater than,” or “greater than or equal to” precedes the first numerical value in a series of two or more numerical values, the term “at least,” “greater than” or “greater than or equal to” applies to each of the numerical values in that series of numerical values. For example, greater than or equal to 1, 2, or 3 is equivalent to greater than or equal to 1, greater than or equal to 2, or greater than or equal to 3. [0259] Whenever the term “no more than,” “less than,” “less than or equal to,” or “at most” precedes the first numerical value in a series of two or more numerical values, the term “no more than,” “less than” or “less than or equal to,” or “at most” applies to each of the numerical values in that series of numerical values. For example, less than or equal to 3, 2, or 1 is equivalent to less than or equal to 3, less than or equal to 2, or less than or equal to 1. [0260] Where values are described as ranges, it will be understood that such disclosure includes the disclosure of all possible sub-ranges within such ranges, as well as specific numerical values that fall within such ranges irrespective of whether a specific numerical value or specific sub-range is expressly stated.
EXAMPLES
[0261] The following examples are illustrative and non-limiting to the scope of the devices, methods, systems, and kits described herein.
EXAMPLE 1
Formulation of Protease Inhibitors for Treatment of COVID-19 [0262] This example describes a formulation of protease inhibitors for treatment of COVID- 19. A protease inhibitor composition comprising Argatroban, disodium Edetate, and hexa-D- arginine amide protease inhibitors is formulated as shown in TABLE 6.
TABLE 6 - Protease Inhibitor Composition for Treatment of COVID-19
[0263] A preservative-free protease inhibitor composition comprising Argatroban, disodium Edetate, and hexa-D-arginine amide protease inhibitors is formulated as with 1 mg/mL Argatroban, 4 mg/mL Di sodium Edetate, 5 mg/mL Hexa-D-Arginine Amide, Chloride Salt (CAS No.: 673202-67-0), 2.5 mg/mL Glyceryl Monooleate (90% monosub stituted), 5 mg/mL Polysorbate 80, 20 mg/mL microcrystalline cellulose and carboxymethyl cellulose sodium (VIVAPUR MCG 811 P), 6 mg/mL glacial acetic acid (100 mM), a sufficient quantity of purified water, and sodium hydroxide as required to reach a target pH.
[0264] The protease inhibitor composition or the preservative-free protease inhibitor composition is formulated to the specifications provided in TABLE 7. Alternatively, the protease inhibitor composition or the preservative-free protease inhibitor composition may be formulated as an isotonic solution (e.g., about 290 mOsm/L).
TABLE 7 - Parameters for a Protease Inhibitor Composition for Treatment of COVID-
19
[0265] The protease inhibitor composition, prepared as described in TABLE 6 and TABLE 7, or the preservative-free protease inhibitor composition, is administered to an individual at risk of contracting COVID-19, or to an individual who has or is suspected to have COVID- 19. The composition is administered via a nasal spray to the individual. The individual is administered one spray per nostril, 0.1 mL per spray, three times per day (TID), for 14 days. The total amount administered to the individual during the treatment course is approximately 8.4 mL. [0266] The composition may be contained in an amber glass container filled with 9.5 mL of the composition and configured for nasal delivery.
EXAMPLE 2
Composition for Treating or Preventing a Coronavirus Infection [0267] This example describes a composition for treating or preventing a coronavirus infection.
[0268] An example of a formulation for treating or preventing a coronavirus infection following the teachings of the instant specification is prepared and evaluated as follows. [0269] The composition may be formulated as described in TABLE 8.
TABLE 8 - Delivery Agents for Treating or Preventing a Coronavirus Infection EXAMPLE 3
Formulation of Protease Inhibitors for Treatment of COVID-19 [0270] This example describes a formulation of protease inhibitors for treatment of COVID- 19. A protease inhibitor composition comprising Argatroban, disodium Edetate, and hexa-D- arginine amide protease inhibitors is formulated as shown in TABLE 9.
TABLE 9 - Protease Inhibitor Composition for Treatment of COVID-19
[0271] A preservative-free protease inhibitor composition comprising Argatroban, disodium Edetate, and hexa-D-arginine amide protease inhibitors is formulated as with 1 mg/mL Argatroban, 4 mg/mL Di sodium Edetate, 5 mg/mL Hexa-D-Arginine Amide, Chloride Salt (CAS No.: 673202-67-0), 2.5 mg/mL Glyceryl Monooleate (90% monosub stituted), 5 mg/mL Polysorbate 80, 20 mg/mL microcrystalline cellulose and carboxymethyl cellulose sodium (VIVAPUR MCG 811 P), a sufficient quantity of purified water, and sodium hydroxide as required to reach a target pH.
[0272] The protease inhibitor composition or the preservative-free protease inhibitor composition is formulated to the specifications provided in TABLE 7. Alternatively, the protease inhibitor composition or the preservative-free protease inhibitor composition may be formulated as an isotonic solution (e.g., about 290 mOsm/L). [0273] The protease inhibitor composition, prepared as described in TABLE 9 and TABLE 7, or the preservative-free protease inhibitor composition, is administered to an individual at risk of contracting COVID-19, or to an individual who has or is suspected to have COVID- 19. The composition is administered via a nasal spray to the individual. The individual is administered one spray per nostril, 0.1 mL per spray, three times per day (TID), for 14 days. The total amount administered to the individual during the treatment course is approximately 8.4 mL.
[0274] The composition may be contained in an amber glass container filled with 9.5 mL of the composition and configured for nasal delivery.
EXAMPLE 4
Composition Comprising Dual-Action Enzyme Inhibitors for Treating or Preventing a
Coronavirus Infection
[0275] This example describes compositions comprising dual-action protease inhibitors for treating or preventing a coronavirus infection. A dual-action protease inhibitor composition comprising disodium Edetate dual-action enzyme inhibitor is formulated as shown in TABLE 10. Disodium edetate functions as a dual -action enzyme inhibitor by inhibiting both ACE-2 and Furin enzymes.
TABLE 10 - Composition for Treatment of COVID-19 [0276] A composition comprising disodium edetate dual-action enzyme inhibitor is formulated with 4 mg/mL Disodium Edetate, 2.5 mg/mL Glyceryl Monooleate (e90% monosub stituted), 5 mg/mL Polysorbate 80, 20 mg/mL microcrystalline cellulose and carboxymethyl cellulose sodium (VIVAPUR MCG 811 P), 6.0 mg glacial acetic acid (100 mM), a sufficient quantity of purified water, and sodium hydroxide as required to reach a target pH.
[0277] The dual-action enzyme inhibitor composition is formulated as described in TABLE 10 and TABLE 7. Alternatively, the dual-action enzyme inhibitor composition may be formulated as an isotonic solution (e.g., about 290 mOsm/L).
[0278] The dual-action enzyme inhibitor composition is administered to an individual at risk of contracting COVID-19, or to an individual who has or is suspected to have COVID-19. The composition is administered via a nasal spray to the individual. The individual is administered one spray per nostril, 0.1 mL per spray, three times per day (TID), for 14 days. The total amount administered to the individual during the treatment course is approximately 8.4 mL.
[0279] The composition may be contained in an amber glass container filled with 9.5 mL of the composition and configured for nasal delivery.
EXAMPLE 5
Inhibition of a SARS-CoV-2 Infection in VERO Cells using a Dual-Action Inhibitor
Formulated for Nasal Delivery
[0280] This example describes inhiation of a SARS-CoV-2 Infection in VERO Cells using a dual-action inhibitor formulated for nasal delivery. The ability of a composition containing edetate disodium (EDTA) and formulated for nasal delivery to reduce the infectivity of a SARS-CoV-2 virus was tested in VERO cells. The nasal spray formulation contained ingredients, including EDTA, which can block entry of SARS-CoV-2 into nasal epithelial cells by interfering with Spike Protein activation by host proteases and/or by masking receptor binding domains (RBD) via electrostatic mechanisms. The formulation composition is provided in TABLE 11. TABLE 11 - Composition for Nasal Delivery to Treat or Prevent a Coronavirus
Infection
[0281] The purpose of these experiments was to mimic the entry process in vitro by examining the infectivity of VERO monkey kidney cells by authentic SARS-CoV-2 virus particles. Although VERO cells are non-mucosal epithelial cells and therefore are more sensitive to cytotoxicity than would be expected of mucosal cells, they provided an established model system to judge in vitro efficacy.
[0282] Serial dilutions of the dual-action inhibitor formulation provided in TABLE 11 were prepared from 1 to 4 dilution to a 1 to 4096 dilution. SARS-CoV-2 (MOI = 0.05) was incubated in these serial dilutions for 1 hour at 37° C and then the mixture was added to the VERO cells for one hour at 37° C. The mixture was then removed and fresh media was added at 37° C and incubated overnight. The cells were then fixed with 10% formalin overnight and then the cells were permeabilized with 0.5% Triton X-100. H anti-SARS-2 N protein antibody, 1 pg/mL, was added at room temperature for 1 hour. Anti-human-IgG-488 (green) tagged antibody was added at a 1 : 1000 dilution at room temperature for 1 hour. The fixed and labeled cells were imaged, as shown in FIG. 5.
[0283] FIG. 5 shows fluorescent images of VERO cells contacted with SARS-CoV-2 virus pre-treated with serial dilutions of a dual-action inhibitor composition formulated for nasal delivery. Fluorescence indicates the presence of the SARS-CoV-2 N-protein and is representative of viral infection of the cells. Serial dilutions ranged from a 1 to 4 dilution (left-most panel) and going to a 1 to 4096 dilution (second panel from right) were tested. Numbers above each panel indicate the fold-dilution tested. The last panel on the right is a negative control without the dual-action inhibitor composition. The fluorescent yellow specks are the N protein that is formed by SARS-CoV-2 infection of the VERO cells. Beginning with the 1 to 4 dilution up until the 1 to 32 dilution (indicated by an arrow), there was no N protein being formed, suggesting that viral infection of the VERO cells was blocked by the composition. From there, further dilution of the dual-action inhibitor composition provided no further protection of the cells from infection. There was minimal toxicity noted at any concentration of the composition tested.
[0284] The results of this experiment showed that the dual-action inhibitor formulation can prevent infection of VERO cells by SARS-CoV-2 when diluted to at least 1 to 16 with minimal toxicity noted. These data showing efficacy in preventing SARS-CoV-2 infection coupled low toxicity of the formulation provides a strong rationale to begin human testing.
EXAMPLE 6
Clinical Trial of an Anti-Viral Nasal Spray Formulation for Prevention of a SARS-
CoV-2 Infection
[0285] This example describes a clinical trial of an anti-viral nasal spray formulation for prevention of a SARS-CoV-2 infection. The anti-viral nasal spray formulation was developed to act as a frontline barrier to the establishment of SARS-CoV-2 infection in the nasal epithelia. A Phase la clinical trial was performed as a randomized, double-blind, placebo- controlled, ascending dose, multi-cohort study on healthy adult participants. The study was performed in two parts: a single ascending dose part, followed by a multiple ascending dose part. Formulations were administered in either a single installation (one 0.1 mL spray per nostril, for a 0.2 mL dose) or a double installation (two 0.1 mL sprays per nostril, for a 0.4 mL dose). In the first part of the study, a single 0.2 mL or 0.4 mL dose of the anti-viral nasal spray formulation provided in TABLE 12 was nasally administered to each subject. Control subjects received a single 0.2 mL or 0.4 mL dose of a placebo formulation (0.20 mg/mL benzalkonium chloride, 9.00 mg/mL sodium chloride, pH 5.0). In the second part of the study, each subject was nasally administered either 0.2 mL or 0.4 mL of the anti -viral nasal spray formulation provided in TABLE 12 three times per day for 14 days. Control subjects received 0.2 mL or 0.4 mL of the placebo formulation three times per day for 14 days. TABLE 12 - Anti-Viral Nasal Spray Formulation to Treat or Prevent a Coronavirus
Infection
1 Dose is equivalent to 0.2 mL - two sprays, one per nostril
2 Dose is equivalent to 0.4 mL - four sprays, two per nostril
[0286] The objectives of the study were to evaluate the tolerability and safety, including incidence and severity of bronchospasm, of single and multiple doses of the anti -viral nasal spray formulation. During the trial there were no deaths, serious adverse events, or discontinuations in the study. Subjects were monitored for cardiac, pulmonary, and hemodynamic parameters using 12 lead ECG, auscultation, and determination of oxygen saturation levels. Additional evaluations included physical examinations, vital sign measurements (heart rate, blood pressure, temperature and respiratory rate), monitoring of adverse events, pregnancy test (females), recording of concomitant medication use and blood sample collection for hematology, coagulation and chemistry, as well as urine collection for urinalysis.
[0287] The formulation was nasally administered using a nasal spray pump that produced a spray pattern with a Dmax of 28.2 mm, an ovality of 1.219, an area of 508 mm2. The device produced a plume geometry with a plume angle of 41.9° and a plume width of 24.5 mm. The device produced droplets with a size distribution of which the Dio was 19.2 pm, the D50 was 49.2 pm, the D90 was 106.6 pm, the span was 1.8 pm, and the percent of volume below 10 pm was 1.0%. The spray pattern and plume geometry are illustrated in FIG. 6.
[0288] Administration of the formulation inhibits entry of the virus into a host cell of the subject by inhibiting enzymes, such as type II transmembrane serine protease, angiotensin converting enzyme, cathepsin B, cathepsin L, and furin. Additionally, the formulation destabilizes the vims by inactivating the viral envelope. Inhibition of viral entry and/or destabilization of the vims reduces the infectivity of the vims, thereby treating or preventing the viral infection.
[0289] The formulations supplied as a nasal spray in amber glass bottles. The bottle labels were coded for blinding as appropriate to the study design and in compliance with the protocol. The storage conditions and expiry date were indicated on the bottle label. The anti viral formulation and the placebo formulation were stored at controlled room temperature 20°C to 25°C, excursions allowed 15 to 30°C, stored dry and protected from light.
EXAMPLE 7
Second Clinical Trial of an Anti- Viral Nasal Spray Formulation for Prevention of a
SARS-CoV-2 Infection
[0290] This example describes a second clinical trial of an anti-viral nasal spray formulation for prevention of a SARS-CoV-2 infection. Formulations are administered to patients with COVID-19 in a double installation (two 0.1 mL sprays per nostril, for a 0.4 mL dose). A single 0.4 mL dose of the anti-viral nasal spray formulation provided in any one of TABLE 6 or TABLE 9 - TABLE 12 is nasally administered to each subject three times per day for 14 days. Control subjects receive a single 0.4 mL dose of a placebo formulation (0.20 mg/mL benzalkonium chloride, 9.00 mg/mL sodium chloride, pH 5.0) three times per day for 14 days.
[0291] The formulation is nasally administered using a nasal spray pump that produces a spray pattern with a Dmax of 29 mm, an ovality of 1.20, an area of 542 mm2. The device produces a plume geometry with a plume angle of 44° and a plume width of 24 mm. The device produces droplets with a size distribution of which the Dio is 18.7 pm, the D50 is 49.0 pm, the D90 is 106.5 pm, the span is 1.792 pm, and the percent of volume below 10 pm is 1.35%. The spray pattern and plume geometry are illustrated in FIG. 7.
[0292] Administration of the formulation inhibits entry of the virus into a host cell of the subject by inhibiting enzymes, such as type II transmembrane serine protease, angiotensin converting enzyme, cathepsin B, cathepsin L, and furin. Additionally, the formulation destabilizes the virus by inactivating the viral envelope. Inhibition of viral entry and/or destabilization of the virus reduces the infectivity of the virus, thereby treating or preventing the viral infection.
[0293] While preferred embodiments of the present disclosure have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the disclosure. It should be understood that various alternatives to the embodiments of the disclosure described herein may be employed in practicing the disclosure. It is intended that the following claims define the scope of the disclosure and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims (100)

CLAIMS WHAT IS CLAIMED IS:
1. A method of reducing the infectivity of a coronavirus in a subject, the method comprising: administering a pharmaceutical composition comprising an enzyme inhibitor and a bioadhesive to the subject; and reducing the infectivity of the coronavirus for a host cell of the subject.
2. The method of claim 1, wherein the bioadhesive is glyceryl monooleate.
3. The method of claim 2, wherein the glyceryl monooleate inactivates an envelope of the coronavirus.
4. The method of any one of claims 1-3, wherein the enzyme inhibitor is disodium edetate, citrate, argatroban, hexa-D-arginine, or a combination thereof.
5. The method of claim 4, wherein the disodium edetate inhibits angiotensin converting enzyme 2, furin, or a combination thereof.
6. The method of claim 4 or claim 5, wherein the citrate inhibits furin.
7. The method of any one of claims 4-6, wherein the argatroban inhibits type II transmembrane serine protease.
8. The method of any one of claims 4-7, wherein the hexa-D-arginine inhibits furin.
9. The method of any one of claims 4-8, further comprising inhibiting the enzyme, wherein inhibiting the enzyme inhibits entry of the coronavirus into the host cell of the subject.
10. The method of any one of claims 1-9, wherein the enzyme is a type II transmembrane serine protease, angiotensin converting enzyme 2, a cathepsin B, a cathepsin L, a furin, or a combination thereof.
11. The method of any one of claims 1-10, wherein the pharmaceutical composition further comprises a stabilizer.
12. The method of claim 11, wherein the stabilizer is polysorbate 80, microcrystalline cellulose, carboxymethyl cellulose, or a combination thereof.
13. The method of any one of claims 1-12, wherein the pharmaceutical composition further comprises an antioxidant.
14. The method of claim 13, wherein the antioxidant is butylated hydroxytoluene.
15. The method of any one of claims 1-14, wherein the pharmaceutical composition further comprises a bacteriostatic agent.
16. The method of claim 15, wherein the bacteriostatic agent is selected from the group consisting of sodium benzoate, benzalkonium chloride, thiomersal, chlorobutanol, chlorbutol, potassium sorbate, and methyl paraben, or a combination thereof.
17. The method of claim 15 or claim 16, wherein the bacteriostatic agent is benzalkonium chloride.
18. The method of any one of claims 1-17, comprising nasally administering the pharmaceutical composition to the subject.
19. The method of any one of claims 1-18, comprising: providing an actuator comprising an actuator tip; producing an aerosol comprising droplets of the pharmaceutical composition from the actuator tip; and dispensing the aerosol into a nose of the subject.
20. The method of claim 19, comprising producing a spray pattern ellipticity ratio of no less than 1.0 to not more than 1.4.
21. The method of claim 19 or claim 20, comprising producing a droplet size distribution such that no more than about 5% of the aerosol volume forms droplets that are less than about 10 pm in diameter.
22. The method of any one of claims 19-21, comprising producing a droplet size distribution such that at least 0.4% of the aerosol volume forms droplets that are less than about 10 pm in diameter.
23. The method of any one of claims 19-22, comprising producing a droplet size distribution such that no more than about 50% of the aerosol volume forms droplets that are less than about 26.9 pm in diameter.
24. The method of any one of claims 19-23, comprising producing a droplet size distribution such that a diameter of droplet for which 50% of the aerosol volume forms droplets of smaller diameter is no less than 25 pm and not more than 75 pm.
25. The method of any one of claims 19-24, comprising producing a droplet size distribution such that a diameter of droplet for which 10% of the aerosol volume forms droplets of smaller diameter is no less than 15 pm and not more than 35 pm.
26. The method of any one of claims 19-25, comprising producing a droplet size distribution such that a diameter of droplet for which 90% of the aerosol volume forms droplets of smaller diameter is no less than 70 pm and not more than 150 pm.
27. The method of any one of claims 19-26, comprising producing a spray pattern with a major axis of at least 25 mm and not more than 40 mm and a minor axis of at least 25 mm and not more than 40 mm.
28. A pharmaceutical composition comprising disodium edetate, glyceryl monooleate, polysorbate 80, microcrystalline cellulose or carboxymethyl cellulose, citrate, and benzalkonium chloride, wherein the pharmaceutical composition is formulated for nasal delivery.
29. The pharmaceutical composition of claim 28, wherein the disodium edetate is present at a concentration of at least 1 mg/mL and not more than 10 mg/mL, at least 1 mg/mL and not more than 6 mg/mL, or at least 2 mg/mL and not more than 6 mg/mL.
30. The pharmaceutical composition of claim 28 or claim 29, wherein the disodium edetate is present at a concentration of at least 3 mg/mL and not more than 5 mg/mL.
31. The pharmaceutical composition of any one of claims 28-30, wherein the glyceryl monooleate is present at a concentration at least 0.5 mg/mL and not more than 10 mg/mL, at least 0.5 mg/mL and not more than 8 mg/mL, at least 1 mg/mL at not more than 6 mg/mL, or at least 1 mg/mL and not more than 5 mg/mL.
32. The pharmaceutical composition of any one of claims 28-31, wherein the glyceryl monooleate is present at a concentration of at least 1.5 mg/mL and not more than 3.5 mg/mL.
33. The pharmaceutical composition of any one of claims 28-32, wherein the polysorbate 80 is present at a concentration of at least 1 mg/mL and not more than 10 mg/mL, at least 2 mg/mL and not more than 8 mg/mL, or at least 4 mg/mL and not more than 6 mg/mL.
34. The pharmaceutical composition of any one of claims 28-33, wherein the polysorbate 80 is present at a concentration of at least 4 mg/mL and not more than 6 mg/mL.
35. The pharmaceutical composition of any one of claims 28-34, wherein the microcrystalline cellulose and carboxymethyl cellulose are present at a combined concentration of at least 10 mg/mL and not more than 50 mg/mL, at least 10 mg/mL and not more than 40 mg/mL, at least 20 mg/mL and not more than 50 mg/mL, or at least 20 mg/mL and not more than 40 mg/mL.
36. The pharmaceutical composition of any one of claims 28-35, wherein the microcrystalline cellulose and carboxymethyl cellulose are present at a combined concentration of at least 20 mg/mL and not more than 40 mg/mL.
37. The pharmaceutical composition of any one of claims 28-36, wherein the citrate is present at a concentration of at least 1 mg/mL and not more than 10 mg/mL, at least 2 mg/mL and not more than 8 mg/mL, or at least 3.0 mg/mL and not more than 5.0 mg/mL.
38. The pharmaceutical composition of any one of claims 28-37, wherein the citrate is present at a concentration of at least 3.5 mg/mL and not more than 5.5 mg/mL.
39. The pharmaceutical composition of any one of claims 28-38, wherein the benzalkonium chloride is present at a concentration of at least 0.05 mg/mL and not more than 2 mg/mL, at least 0.05 mg/mL and not more than 0.1 mg/mL, or at least 0.1 mg/mL and not more than 0.5 mg/mL.
40. The pharmaceutical composition of any one of claims 28-39, wherein the benzalkonium chloride is present at a concentration of at least 0.1 mg/mL and not more than 1.5 mg/mL.
41. The pharmaceutical composition of any one of claims 28-38, wherein the benzalkonium chloride is present at a concentration of no more than 0.1% (w/w).
42. The pharmaceutical composition of any one of claims 28-40, wherein the benzalkonium chloride is present at a concentration of at least 0.01% and no more than 0.2% (w/w).
43. The pharmaceutical composition of any one of claims 28-42, further comprising butylated hydroxytoluene.
44. The pharmaceutical composition of claim 43, wherein the butylated hydroxytoluene is present at a concentration of at least 0.1 mg/mL and not more than 3 mg/mL, at least 0.1 mg/mL at not more than 2 mg/mL, at least 0.2 mg/mL and not more than 1.0 mg/mL, or at least 0.4 mg/mL and not more than 0.6 mg/mL.
45. The pharmaceutical composition of claim 43 or claim 44, wherein the butylated hydroxytoluene is present at a concentration of at least 0.4 mg/mL and not more than 0.6 mg/mL.
46. The pharmaceutical composition of any one of claims 28-45, wherein the composition is at a pH of at least 4 and not more than 6, at least 4.2 and not more than 5.8, or at least 4.5 and not more than 5.5.
47. The pharmaceutical composition of any one of claims 28-46, further comprising argatroban.
48. The pharmaceutical composition of claim 47, wherein the argatroban is present at a concentration of at least 0.001 mg/mL but not more than 2 mg/mL, at least 0.05 mg/mL but not more than 1 mg/mL, at least 0.05 mg/mL but not more than 0.5 mg/mL, or at least 0.05 mg/mL and not more than 0.15 mg/mL.
49. The pharmaceutical composition of claim 47 or claim 48, wherein the argatroban is present at a concentration of at least 0.05 mg/mL and not more than 0.15 mg/mL.
50. The pharmaceutical composition of any one of claims 28-49, further comprising Hexa-D-arginine.
51. The pharmaceutical composition of claim 50, wherein the Hexa-D-arginine is present at a concentration of at least 1 mg/mL and not more than 20 mg/mL, at least 1 mg/mL and not more than 10 mg/mL, or at least 2 mg/mL and not more than 8 mg/mL.
52. The pharmaceutical composition of claim 50 or claim 51, wherein the Hexa-D- arginine is present at a concentration of at least 3 mg/mL and not more than 7 mg/mL.
53. The pharmaceutical composition of any one of claims 28-52, wherein the pharmaceutical composition is formulated at a pH of at least 4.0 and not more than 6.0.
54. A pharmaceutical composition comprising an enzyme inhibitor, a bioadhesive, and a stabilizer, wherein the pharmaceutical composition is formulated for nasal delivery.
55. The pharmaceutical composition of claim 54, wherein the enzyme inhibitor is a dual action enzyme inhibitor.
56. The pharmaceutical composition of claim 54 or claim 55, wherein the enzyme inhibitor chelates calcium, zinc, or a combination thereof.
57. The pharmaceutical composition of claim 56, wherein the enzyme inhibitor comprises disodium edetate, EDTA-magnesium, or EGTA.
58. The pharmaceutical composition of any one of claims 54-57, wherein the enzyme inhibitor is present at a concentration of at least 1 mg/mL and not more than 10 mg/mL, at least 1 mg/mL and not more than 6 mg/mL, or at least 2 mg/mL and not more than 6 mg/mL.
59. The pharmaceutical composition of claim 54, wherein the enzyme inhibitor is argatroban.
60. The pharmaceutical composition of any one of claims 54-59, wherein the stabilizer comprises microcrystalline cellulose, carboxymethyl cellulose, polysorbate 80, or combinations thereof.
61. The pharmaceutical composition of any one of claims 54-60, wherein the bioadhesive is glyceryl monooleate.
62. The pharmaceutical composition of any one of claims 54-61, further comprising an antioxidant.
63. The pharmaceutical composition of claim 62, wherein the antioxidant is butylated hydroxytoluene.
64. The pharmaceutical composition of any one of claims 54-63, further comprising a bacteriostatic agent.
65. The pharmaceutical composition of claim 64, wherein the bacteriostatic agent is selected from the group consisting of sodium benzoate, benzalkonium chloride, thiomersal, chlorobutanol, chlorbutol, potassium sorbate, and methyl paraben, or a combination thereof.
66. The pharmaceutical composition of claim 64 or claim 65, wherein the bacteriostatic agent is benzalkonium chloride.
67. The pharmaceutical composition of any one of claims 54-66, further comprising a buffer.
68. The pharmaceutical composition of claim 67, wherein the buffer is citrate.
69. The pharmaceutical composition of any one of claims 54-68, further comprising a furin peptide inhibitor.
70. The pharmaceutical composition of claim 69, wherein the furin peptide inhibitor is present at a concentration of at least 2 mg/mL, at least 4 mg/mL, or at least 5 mg/mL.
71. The pharmaceutical composition of claim 69 or claim 70, wherein the furin peptide inhibitor is present at a concentration of at least 1 mg/mL and not more than 20 mg/mL, at least 1 mg/mL and not more than 10 mg/mL, or at least 2 mg/mL and not more than 8 mg/mL.
72. The pharmaceutical composition of any one of claims 69-71, wherein the furin peptide inhibitor is present at a concentration of at least 3 mg/mL and not more than 7 mg/mL.
73. The pharmaceutical composition of any one of claims 69-72, wherein the furin peptide inhibitor is [W5R4C], [WR]s, Ci2-[Rs], W4-[Rs], hexa-D-arginine, or nona-D- arginine.
74. The pharmaceutical composition of any one of claims 69-73, wherein the furin peptide inhibitor is Hexa-D-arginine.
75. The pharmaceutical composition of any one of claims 54-74, wherein the pharmaceutical composition is at a pH of at least 4 and not more than 6, at least 4.2 and not more than 5.8, or at least 4.5 and not more than 5.5.
76. The pharmaceutical composition of any one of claims 28-75, wherein the pharmaceutical composition is free of endotoxins.
77. The pharmaceutical composition of any one of claims 28-76, wherein the pharmaceutical composition is aqueous.
78. The pharmaceutical composition of any one of claims 28-77, wherein the pharmaceutical composition is formulated for delivery as an aerosol from an actuator tip.
79. The pharmaceutical composition of claim 78, wherein the aerosol has a spray pattern ellipticity ratio of no less than 1.0 to not more than 1.4.
80. The pharmaceutical composition of claim 78 or claim 79, wherein the aerosol comprises droplets, and wherein no more than about 5% of the aerosol volume forms droplets that are less than about 10 pm in diameter.
81. The pharmaceutical composition of any one of claims 78-80, wherein the aerosol comprises droplets, and wherein at least 0.4% of the aerosol volume forms droplets that are less than about 10 pm in diameter.
82. The pharmaceutical composition of any one of claims 78-81, wherein the aerosol comprises droplets, and wherein no more than about 50% of the aerosol volume forms droplets that are less than about 26.9 pm in diameter.
83. The pharmaceutical composition of any one of claims 78-82, wherein the aerosol comprises droplets, and wherein a diameter of droplet for which 50% of the aerosol volume forms droplets of smaller diameter is no less than 25 pm and not more than 75 pm.
84. The pharmaceutical composition of any one of claims 78-83, wherein the aerosol comprises droplets, and wherein a diameter of droplet for which 10% of the aerosol volume forms droplets of smaller diameter is no less than 15 pm and not more than 35 pm.
85. The pharmaceutical composition of any one of claims 78-84, wherein the aerosol comprises droplets, and wherein a diameter of droplet for which 90% of the aerosol volume forms droplets of smaller diameter is no less than 70 pm and not more than 150 pm.
86. The pharmaceutical composition of any one of claims 78-85, wherein the aerosol has a spray pattern with a major axis of at least 25 mm and not more than 40 mm and a minor axis of at least 25 mm and not more than 40 mm.
87. The pharmaceutical composition of any one of claims 78-86, wherein the aerosol has a spray pattern with a plume angle of at least 30° and no more than 70°.
88. A pharmaceutical device comprising: a pharmaceutical composition of any one of claims 28-87, and an actuator comprising an actuator tip; wherein the actuator is configured to produce an aerosol of the pharmaceutical composition.
89. The pharmaceutical device of claim 88, wherein the aerosol has a spray pattern ellipticity ratio of no less than 1.0 to not more than 1.4.
90. The pharmaceutical device of claim 88 or claim 89, wherein the aerosol comprises droplets, and wherein no more than about 5% of the aerosol volume forms droplets that are less than about 10 pm in diameter.
91. The pharmaceutical device of any one of claims 88-90, wherein the aerosol comprises droplets, and wherein at least 0.4% of the aerosol volume forms droplets that are less than about 10 pm in diameter.
-HO-
92. The pharmaceutical device of any one of claims 88-91, wherein the aerosol comprises droplets, and wherein no more than about 50% of the aerosol volume forms droplets that are less than about 26.9 pm in diameter.
93. The pharmaceutical device of any one of claims 88-92, wherein the aerosol comprises droplets, and wherein a diameter of droplet for which 50% of the aerosol volume forms droplets of smaller diameter is no less than 25 pm and not more than 75 pm.
94. The pharmaceutical device of any one of claims 88-93, wherein the aerosol comprises droplets, and wherein a diameter of droplet for which 10% of the aerosol volume forms droplets of smaller diameter is no less than 15 pm and not more than 35 pm.
95. The pharmaceutical device of any one of claims 88-94, wherein the aerosol comprises droplets, and wherein a diameter of droplet for which 90% of the aerosol volume forms droplets of smaller diameter is no less than 70 pm and not more than 150 pm.
96. The pharmaceutical device of any one of claims 88-95, wherein the aerosol has a spray pattern with a major axis of at least 25 mm and not more than 40 mm and a minor axis of at least 25 mm and not more than 40 mm.
97. The pharmaceutical device of any one of claims 88-96, wherein the aerosol has a spray pattern with a plume angle of at least 30° and not more than 70°.
98. A method of reducing the infectivity of a coronavirus in a subject, the method comprising administering the pharmaceutical composition of any one of claims 28-87 to the subject using the pharmaceutical device of any one of claims 88-97, and reducing the infectivity of the coronavirus.
99. The method of claim 98, wherein the subject has a coronavirus infection.
100. The method of claim 98, wherein the subject is at risk of developing a coronavirus infection.
AU2021254780A 2020-04-16 2021-04-15 Compositions and methods to reduce the infectivity of a virus Pending AU2021254780A1 (en)

Applications Claiming Priority (13)

Application Number Priority Date Filing Date Title
US202063011291P 2020-04-16 2020-04-16
US63/011,291 2020-04-16
US202063012818P 2020-04-20 2020-04-20
US63/012,818 2020-04-20
US202063015370P 2020-04-24 2020-04-24
US63/015,370 2020-04-24
US202063032435P 2020-05-29 2020-05-29
US63/032,435 2020-05-29
US202063051800P 2020-07-14 2020-07-14
US63/051,800 2020-07-14
US202063112112P 2020-11-10 2020-11-10
US63/112,112 2020-11-10
PCT/US2021/027438 WO2021211808A1 (en) 2020-04-16 2021-04-15 Compositions and methods to reduce the infectivity of a virus

Publications (1)

Publication Number Publication Date
AU2021254780A1 true AU2021254780A1 (en) 2022-10-20

Family

ID=78083678

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2021254780A Pending AU2021254780A1 (en) 2020-04-16 2021-04-15 Compositions and methods to reduce the infectivity of a virus

Country Status (4)

Country Link
EP (1) EP4135660A1 (en)
AU (1) AU2021254780A1 (en)
CA (1) CA3173515A1 (en)
WO (1) WO2021211808A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3198674A1 (en) * 2020-10-15 2022-04-21 Bausch & Lomb Ireland Limited Benzalkonium chloride for use in treating conjunctivitis and/or covid-19
WO2023137404A1 (en) * 2022-01-12 2023-07-20 University Of Virginia Patent Foundation Spike furin cleavage is a sars-cov-2 targeting strategy to break the chain of infection cycle
WO2023220712A1 (en) * 2022-05-13 2023-11-16 The Trustees Of Columbia University In The City Of New York Host proteases essential for parainfluenza spread in the human lung: potential targets for antiviral interventions

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0760650B1 (en) * 1994-05-20 2002-07-31 Novavax, Inc. Antimicrobial oil-in-water emulsions
IL122892A0 (en) * 1998-01-11 1998-08-16 Yeda Res & Dev Pharmaceutical compositions comprising a thiocarbamate
AU4841799A (en) * 1998-06-29 2000-01-17 Nastech Pharmaceutical Company, Inc Methods and pharmaceutical formulations for preventing and treating motion sickness
US7264932B2 (en) * 1999-09-24 2007-09-04 Applera Corporation Nuclease inhibitor cocktail
WO2001028515A1 (en) * 1999-10-22 2001-04-26 Líf-Hlaup ehf. Bio-Gels Pharmaceuticals Inc. Pharmaceutical composition for treatment of mucosal epithelial ulceration and/or erosion
JP5651186B2 (en) * 2009-10-30 2015-01-07 バイオジェニック イノベーションズ, リミテッド ライアビリティ カンパニー Methylsulfonylmethane (MSM) for treating drug-resistant microorganisms
IN2014MN02245A (en) * 2012-04-10 2015-10-09 Rubicon Res Private Ltd
EP2666463A1 (en) * 2012-05-21 2013-11-27 Synthon BV Stabilized liquid composition comprising pemetrexed

Also Published As

Publication number Publication date
CA3173515A1 (en) 2021-10-21
EP4135660A1 (en) 2023-02-22
WO2021211808A1 (en) 2021-10-21

Similar Documents

Publication Publication Date Title
EP4135660A1 (en) Compositions and methods to reduce the infectivity of a virus
US20230110614A1 (en) Heparin and n-acetylcysteine for the treatment of a respiratory virus
US20060069021A1 (en) Compositions and methods for intranasal administration of inactive analogs of PTH or inactivated preparations of PTH or PTH analogs
AU2004312043B2 (en) Intranasal administration of glucose-regulating peptides
EP1951198B1 (en) Intranasal administration of rapid acting insulin
US20060074025A1 (en) Therapeutic formulations for transmucosal administration that increase glucagon-like peptide-1 bioavailability
US20080318861A1 (en) Mucosal Delivery of Stabilized Formulations of Exendin
US20060210614A1 (en) Method of treatment of a metabolic disease using intranasal administration of exendin peptide
BRPI0620586A2 (en) aqueous pharmaceutical formulation for intranasal administration and use of an aqueous pharmaceutical formulation to make a medicament
US20070161563A1 (en) A device for enhanced epithelial permeation of y2 receptor-binding peptides
US20070077283A1 (en) Method of enhancing transmucosal delivery of therapeutic compounds
WO2004103396A1 (en) Compositions for enhanced mucosal delivery of interferon alpha
WO2007061434A2 (en) A pharmaceutical formulation of glp-1 and its use for treating a metabolic syndrome
WO2007146448A1 (en) Pharmaceutical formulations of glp-1 derivatives
WO2022009081A1 (en) Drug for use against the novel coronavirus disease, covid-19
KR20070030264A (en) Intranasal formulations of interferon beta free of stabilizers that are proteins or polypeptides
US20070185035A1 (en) Enhanced mucosal administration of neuroprotective peptides
WO2024026042A1 (en) Heparin compositions for treatment of lung damage and methods of use thereof
KR102664833B1 (en) Formulation for nasal or oral inhalation product containing nafamostat or camostat
MX2008004980A (en) Intranasal administration of rapid acting insulin
MXPA06009331A (en) Compositions and methods for enhanced mucosal delivery of y2 receptor-binding peptides and methods for treating and preventing obesity