Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
THIOL-CONTAINING COMPOUNDS FOR USE IN TREATING CORONAVIRUS
Document Type and Number:
WIPO Patent Application WO/2021/231421
Kind Code:
A1
Abstract:
Provided herein, inter alia, are methods and compositions including thiol-containing compounds for treating coronavirus infection.

Inventors:
FAHY JOHN VINCENT (US)
GITLIN IRINA (US)
RAYMOND WILFRED (US)
Application Number:
PCT/US2021/031772
Publication Date:
November 18, 2021
Filing Date:
May 11, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV CALIFORNIA (US)
International Classes:
A61K31/4409; A61K9/00; A61K9/48
Foreign References:
US20060189542A12006-08-24
US20110218241A12011-09-08
US20180344751A12018-12-06
US20090068142A12009-03-12
US20050267172A12005-12-01
US20170056347A12017-03-02
Attorney, Agent or Firm:
JENKINS, Kenneth et al. (US)
Download PDF:
Claims:
CLAIMS

WHAT IS CLAIMED IS:

1. A method of treating a coronavirus infection in a subject in need thereof, the method comprising administering to said subject an effective amount of a thiol-containing compound in a pharmaceutically acceptable carrier.

2. The method of claim 1, wherein the coronavirus infection is a SARS-CoV-2 virus infection.

3. The method of claim 1, wherein the subject has or is suspected of having COVID-19.

4. The method of claim 1, wherein the effective amount is administered within 48-96 hours of the onset of one or more symptoms of the infection.

5. The method of claim 4, wherein the effective amount is administered within 72 hours of the onset of the one or more symptoms.

6. The method of claim 4, wherein the effective amount is administered within 48 hours of the onset of the one or more symptoms.

7. The method of claim 1, wherein the effective amount is effective to reduce viral load in the subject, optionally wherein the viral load is reduced in the lung.

8. The method of claim 1, wherein the thiol-containing compound is administered by pulmonary delivery.

9. The method of claim 8, wherein the thiol-containing compound is administered as a liquid aerosol or a dry powder.

10. The method of claim 1, wherein the thiol-containing compound is administered orally.

11. The method of claim 10, wherein the thiol-containing compound is administered as a lozenge.

12. The method of claim 1, wherein the thiol-containing compound is administered as a nasal spray.

13. The method of claim 1, wherein the thiol-containing compound is administered intravenously.

14. The method of claim 1, wherein the subject is not hospitalized.

15. The method of claim 1, wherein the subject is hospitalized.

16. The method of claim 15, wherein the subject is in an intensive care unit.

17. The method of claim 1, wherein the thiol-containing compound is a monothiol.

18. The method of claim 1, wherein the thiol-containing compound comprises two or more thiols.

19. The method of claim 1, wherein the thiol-containing compound is a dithiol.

20. The method of claim 1, wherein the thiol-containing compound has a molecular weight of at least 164 g/mol.

21. The method of claim 1, wherein the thiol-containing compound is thiomandelic acid, DL-Captopril, DL-Thiophan, N-acetylcysteine, Meso-2,3,-dimercaptosuccinic acid, 2,3- dimercaprol, D-(-)-Penicillamine, Glutathione, L-Cysteine, Zofenoprilat, Tiopronin, N- acystelyn, carbocysteine, cysteamine, sodium-2-mercaptoethane sulfonate, WR-1065, an analogue or derivative thereof, or a combination thereof.

22. The method of claim 1, wherein the thiol-containing compound is Bucillamine.

23. The method of claim 1, wherein administering the thiol-containing compound comprises administering a prodrug thereof, wherein the prodrug is metabolized by the subject to form the thiol-containing compound.

24. The method of claim 23, wherein the prodrug is amifostine.

25. The method of claim 1, wherein the thiol-containing compound is not N-acetylcysteine, glutathione, cysteamine, bucillamine, or dithiothreitol.

26. The method of claim 1, wherein the thiol-containing compound is coadministered with one or more additional antiviral compound, and/or one or more anti-inflammatory compounds.

27. The method of claim 1, wherein the thiol-containing compound is not coadministered with heparin, an antibiotic, or methylene blue.

28. The method of claim 1, further comprising reducing inflammation, wherein the inflammation is in the lung.

29. A composition comprising a thiol-containing compound for use in a method of claim 1.

Description:
THIOL-CONTAINING COMPOUNDS FOR USE IN TREATING

CORONAVIRUS

CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims priority to U.S. Provisional Application No. 63/024,315, filed May 13, 2020, which is hereby incorporated by reference in its entirety and for all purposes.

REFERENCE TO A “SEQUENCE LISTING,” A TABLE, OR A COMPUTER PROGRAM LISTING APPENDIX SUBMITTED AS AN ASCII FILE [0002] The Sequence Listing written in file 048536-688001WO_Sequence_Listing_ST25.txt, created May 4, 2021, 5,101 bytes, machine format IBM-PC, MS Windows operating system, is hereby incorporated by reference.

STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT [0003] This invention was made with government support under grant nos. P01 HL128191 and R01 HL080414, awarded by the National Institutes of Health. The government has certain rights in the invention.

BACKGROUND

[0004] Coronavimses are enveloped RNA viruses that cause respiratory tract infections. The novel 2019 strain of coronavirus (SARS-CoV-2) causes Coronavirus Disease 2019 (COVID-19), characterized by severe systemic inflammation and pneumonia. COVID-19 has rapidly become a source of profound morbidity and mortality worldwide.

SUMMARY

[0005] Therapeutic agents for CO VID- 19 and related coronavirus infections are needed. The present disclosure addresses this need, and provides additional benefits as well.

[0006] In an aspect is provided a method of treating a coronavirus infection in a subject in need thereof. In embodiments, the method includes administering to the subject an effective amount of a thiol-containing compound in a pharmaceutically acceptable carrier.

[0007] In an aspect is provided a composition including a thiol-containing compound for use in the methods provided herein including embodiments thereof. BRIEF DESCRIPTION OF THE DRAWINGS [0008] FIG. 1 is a graph illustrating that 6,6’-dithiotrehalose (“MUC”) and N-acetyl-L- cysteine (NAC) inhibit binding of SARS-CoV-2 spike protein (SARS-2-S) to ACE2.

[0009] FIG. 2 is a schematic depicting generation of a vesicular stomatitus virus (VSV) pseudotype. The VSV pseudotype (VSV-S) includes the gene encoding luciferase and bears the SARS-CoV-2 spike protein (SARS-2-S) on its viral envelope.

[0010] FIG. 3 is a schematic illustrating VSV-S entering target cells in the absence of an inhibitor compound (top panel) and inhibition of VSV-S cellular entry when incubated with MUC or NAC (bottom panel).

[0011] FIG. 4 is a bar graph showing VSV-S is inhibited from entering ACE2 expressing cells when treated with MUC or NAC.

[0012] FIGS. 5A-5D shows cystine mapping and conservation of cystines in beta coronavirus RBD. FIG. 5A: Cystine map for SARS-2-S domain SI, amino acids 15 - 685, comprising the sequence from the mature N-terminus to the first TMPRSS2 proteolytic site R685 (UniProt Entry: P0DTC2). Ten cystine linkages are denoted by dashed lines with amino acid residue number above. The dark gray region is the receptor binding domain (RBD), and the lighter gray box highlights the ACE2 binding motif, a cluster of amino acids that make contact with ACE2. FIG. 5B: Amino acid alignment of SARS-2-S RBD domain (SEQ ID NO:l, aa 319-541, PDB Entry 6M0J) and SARS-l-S RBD domain (SEQ ID NO:2, aa 306-517, PDB Entry 3 SCI). Residues that are shared are highlighted by black boxes and residues that represent a similar amino acid class replacement are bound by gray boxes. The solid lines link cystine forming cysteines. The solid line and numbers 480-488 and 467-474 highlight the conserved cystine bridge in the RBDs for both viruses. Asterisks denote amino acids that are within 4 angstroms of ACE2 in their respective solved structures. FIG. 5C: A surface rendering of SARS-2-RBD (PDB Entry 6M0J) generated with UCSF Chimera software oriented with the ACE2 binding region facing forward. FIG. 5D: Amino acid sequence of SAR-2-S RBD (SEQ ID NO:l, aa 319-541, PDB Entry 6M0J) highlighting the RBD mutations identified in the circulating SARS CoV2 variants B.l.1.7, B.1.351, P.1, P.2 and B.1.525. Amino acids are noted with single letter code and sequence number. The conserved RBD cystine formed by C480 and C488 is highlighted.

[0013] FIGS. 6A-6J illustrates binding of SARS-CoV-2 RBD to ACE2 is inhibited by thiol- based drugs. FIG. 6A: Schematic representation of a SARS CoV-2 RBD to ACE2 binding assay. RBD was covalently coupled to plates functionalized with primary amine-reactive maleic anhydride. ACE2 binding was then evaluated after RBD exposure to thiol-based drugs for 60 minutes. FIG. 6B shows percent of binding of RBD ongmal to ACE2 in the presence of the drugs (n = 4 - 6). Without drug treatment, the binding was 100%, whereas treatment with the thiol- based drugs showed a decrease in the binding % relative to no drug control. The X axis is scaled to log2. At the highest illustrated concentrations, points in the graph from top to bottom correspond to carbocysteine, amifostine, tiopronin, and NAC, respectively. FIG. 6C shows area under the curve (AUC) analysis for effects of the thiol-based drugs on RBD original to ACE2 binding. Reference AUC was calculated from RBD ongmal to ACE2 binding with no drug control; dashed line represents 50% of reference AUC. FIG. 6D shows binding of RBD ongmal to ACE2 at one and two hours post TM21, WR-1065, cysteamine, Mesna or bucillamine exposure and washout (n = 4 - 5). At two hours, points from top to bottom correspond to MESNA, TS21, bucillamine, cysteamine, and WR-1065, respectively. FIG. 6E shows the fold change in the binding of RBD N501Y to ACE2 with respect to RBD ongmal (n=7). FIG. 6F shows percent of binding of RBD N501Y to ACE2 in the presence of TS21, cysteamine and carbocysteine (n = 4 - 6). The X axis is scaled to log2. Compounds noted in the legend correspond to the plots, from top to bottom, respectively. FIG. 6G shows area under the curve (AUC) analysis for effects of TS21, cysteamine and carbocysteine on RBD N501Y to ACE2 binding. Reference AUC was calculated from RBD N501Y to ACE2 binding with no drug control; dashed line represents 50% of reference AUC. FIG. 6H shows schematic representation of a BODIPY assay. FIG. 61 shows the change in fluorescence with respect to time when thiol-based drugs react with BODIPY FL cystine. Dotted lines indicate SEM for the graph. Beginning with the most verical plot at the left, plots in clockwise order are as follows: WR-1065, cysteamine, TS21, bucillamine, tiopronin, MESNA, NAC, amifostine, carbocysteine. FIG. 6J shows maximum slope (Max V) for the fluorescence vs time graph in FIG. 61 for the thiol-based drugs-BODIPY cystine reaction. Data are mean ± SEM. Statistical significance for FIG. 6C, 6G, and 6J was analyzed by one way ANOVA followed by Dunnett’s post-hoc analysis. Significance indicates differences from reference AUC. Statistical significance for FIG. 6E inset was analyzed by two tailed unpaired t- test. **p < 0.01, *** p < 0.005, ****, p < 0.0001.

[0014] FIGS. 7A-7E shows entry of SARS-CoV-2 pseudoviruses into 293T-ACE2- TMPRSS2 cells is inhibited by thiol-based drugs. Pseudovirus (PV) entry efficiency, quantified by luciferase activity, when the pseudoviruses were exposed to thiol-based drugs prior to cell transduction (n = 3-4). The effects of drugs on 293T-ACE2-TMPRSS2 cell viability was quantified using Cell Titer Glo 2.0 with lower drug dose exposures, reflecting 66-fold dilution of drugs when pseudovirus/drug mixture was incubated with cells (n=3). X-axes are scaled to loglO - the lower X-axis refers to concentration of drugs on the pseudovirus and the upper X- axis refers to equivalent concentration of drugs on the cells. The left Y-axis refers to PV entry efficiency and the right Y -axis refers to cell viability. Percentage changes are with respect to no drug control which is set as 100%. ICso of the drugs was determined using the non-linear regression fitting with a variable slope. Data are mean ± SD. In each graph, the most downward-trending plot corresponds to pseudovirus entry.

[0015] FIGS. 8A-8H illustrates effects of TS21 and cysteamine on a Syrian hamster model of SARS CoV2 infection. FIG. 8A shows the study design for assessing the effect of thiol-based drugs in Syrian hamster model of COVID-19. TS21 (0.5mg/kg lung deposited dose) was given to hamsters via nose-only inhalation exposure for 3 days (Days 0-2). Cysteamine hydrochloride (147mg/kg) was administered to hamsters via intraperitoneal injection for 5 days (Days 0-4). Both drugs were given twice daily, with the first dose given 2 hours prior to the virus inoculation on Day 0. SARS CoV2 virus inoculation was carried out by intranasal administration at lE+05TCID5o/animal. All animals were sacrificed on Day5. FIG. 8B shows viral RNA levels in the lungs of animals treated with TS21 and cysteamine relative to the respective vehicle control groups. FIG. 8C shows the lung weights, normalized to the terminal body weights, of the animals. FIG. 8D shows total leukocyte counts in the BAL fluid of hamsters treated with TS21 and cysteamine with respected to the vehicle controls. FIGS. 8E- 8H: Differential leukocyte counts in the BAL fluid of animals, with FIG. 8E showing neutrophil, FIG. 8F showing macrophage, FIG. 8G showing lymphocyte and FIG. 8H showing eosinophil counts in treated and vehicle control groups. Aero control - aerosol vehicle control group; IP control - intraperitoneal vehicle control group. Each group had N=10 animals (5 Males, 5 females). One animal in the aero control group died during inhalation exposure. 2 BAL samples from the cysteamine group were not analyzed because of a technical error. Data are mean ± SEM. Statistical significance was analyzed by two tailed, unpaired t-test between treated and respective control groups (aero control vs TS21; IP control vs cysteamine). * p < 0.05, ** p < 0.01 *** p < 0.005, **** p < 0.0001.

[0016] FIGS. 9A-9C illustrate that thiol-based drugs inhibit SARS-CoV-2 virus infectivity in VeroE6 cells. Cytopathic effects (CPE) quantified by visual inspection when virus is exposed to drugs prior to infection in Vero E6-TMPRSS2 cells (n = 3). The effects of drugs on Vero E6 cell viability was quantified with exposure of cell to lower drug doses, reflecting the 24-fold dilution of drugs when virus/drug mixture was incubated with cells (n=3). Percentage changes are with respect to no drug control which is set as 100%. IC50 of the drugs was determined using the non-linear regression fitting with a variable slope. Data are mean ± SD. DET AILED DESCRIPTION

I. Definitions

[0017] The practice of the technology described herein will employ, unless indicated specifically to the contrary, conventional methods of chemistry, biochemistry, organic chemistry, molecular biology, microbiology, recombinant DNA techniques, genetics, immunology, and cell biology that are within the skill of the art, many of which are described below for the purpose of illustration. Examples of such techniques are available in the literature. See, e.g., Singleton et al., DICTIONARY OF MICROBIOLOGY AND MOLECULAR BIOLOGY 2nd ed., J. Wiley & Sons (New York, NY 1994); and Sambrook and Green, Molecular Cloning: A Laboratory Manual, 4th Edition (2012). Methods, devices and materials similar or equivalent to those described herein can be used in the practice of this invention.

[0018] All patents, patent applications, articles and publications mentioned herein, both supra and infra, are hereby expressly incorporated herein by reference in their entireties.

[0019] Unless defined otherwise herein, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Various scientific dictionaries that include the terms included herein are well known and available to those in the art. Although any methods and materials similar or equivalent to those described herein find use in the practice or testing of the disclosure, some preferred methods and materials are described. Accordingly, the terms defined immediately below are more fully described by reference to the specification as a whole. It is to be understood that this disclosure is not limited to the particular methodology, protocols, and reagents described, as these may vary, depending upon the context in which they are used by those of skill in the art. The following definitions are provided to facilitate understanding of certain terms used frequently herein and are not meant to limit the scope of the present disclosure.

[0020] As used herein, the singular terms "a", "an", and "the" include the plural reference unless the context clearly indicates otherwise.

[0021] Reference throughout this specification to, for example, "one embodiment", "an embodiment", "another embodiment", "a particular embodiment", "a related embodiment", "a certain embodiment", "an additional embodiment", or "a further embodiment" or combinations thereof means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present disclosure. Thus, the appearances of the foregoing phrases in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments.

[0022] As used herein, the term “about” means a range of values including the specified value, which a person of ordinary skill in the art would consider reasonably similar to the specified value. In embodiments, the term “about” means within a standard deviation using measurements generally acceptable in the art. In embodiments, about means a range extending to +/- 10% of the specified value. In embodiments, about means the specified value.

[0023] Throughout this specification, unless the context requires otherwise, the words "comprise", "comprises" and "comprising" will be understood to imply the inclusion of a stated step or element or group of steps or elements but not the exclusion of any other step or element or group of steps or elements. By "consisting of is meant including, and limited to, whatever follows the phrase "consisting of." Thus, the phrase "consisting of' indicates that the listed elements are required or mandatory, and that no other elements may be present. By "consisting essentially of' is meant including any elements listed after the phrase, and limited to other elements that do not interfere with or contribute to the activity or action specified in the disclosure for the listed elements. Thus, the phrase "consisting essentially of' indicates that the listed elements are required or mandatory, but that no other elements are optional and may or may not be present depending upon whether or not they affect the activity or action of the listed elements.

[0024] The term “pharmaceutically acceptable salts” is meant to include salts of the active compounds that are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When compounds of the present disclosure contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt. When compounds of the present disclosure contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, oxalic, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al, “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1- 19). Certain specific compounds of the present disclosure contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.

[0025] Thus, the compounds of the present disclosure may exist as salts, such as with pharmaceutically acceptable acids. The present disclosure includes such salts. Non-limiting examples of such salts include hydrochlorides, hydrobromides, phosphates, sulfates, methanesulfonates, nitrates, maleates, acetates, citrates, fumarates, proprionates, tartrates (e.g., (+)-tartrates, (-)-tartrates, or mixtures thereof including racemic mixtures), succinates, benzoates, and salts with amino acids such as glutamic acid, and quaternary ammonium salts (e.g., methyl iodide, ethyl iodide, and the like). These salts may be prepared by methods known to those skilled in the art.

[0026] The neutral forms of the compounds are preferably regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound may differ from the various salt forms in certain physical properties, such as solubility in polar solvents.

[0027] In addition to salt forms, the present disclosure provides compounds, which are in a prodrug form. Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present disclosure. Prodrugs of the compounds described herein may be converted in vivo after administration. Additionally, prodrugs can be converted to the compounds of the present disclosure by chemical or biochemical methods in an ex vivo environment, such as, for example, when contacted with a suitable enzyme or chemical reagent. Non-limiting examples of prodrugs of thiol-containing compounds are described in US10526283B2, which is incorporated herein by reference in its entirety.

[0028] Certain compounds of the present disclosure can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present disclosure. Certain compounds of the present disclosure may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present disclosure and are intended to be within the scope of the present disclosure.

[0029] “Pharmaceutically acceptable excipient” and “pharmaceutically acceptable carrier” refer to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present disclosure without causing a significant adverse toxicological effect on the patient. Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, lactated Ringer’s, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such as Ringer’s solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethycellulose, polyvinyl pyrrolidine, and colors. Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure. One of skill in the art will recognize that other pharmaceutical excipients are useful in the present disclosure. In embodiments, the pharmaceutically acceptable excipient is sodium citrate. In embodiments, the pharmaceutically acceptable excipient is sodium chloride. In embodiments, the pharmaceutically acceptable excipient is sodium hydroxide.

[0030] The term “preparation” is intended to include the formulation of the active compound with encapsulating material as a carrier providing a capsule in which the active component with or without other carriers, is surrounded by a carrier, which is thus in association with it. Similarly, cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid dosage forms suitable for oral administration.

[0031] “Contacting” is used in accordance with its plain ordinary meaning and refers to the process of allowing at least two distinct species (e.g., chemical compounds including biomolecules or cells) to become sufficiently proximal to react, interact or physically touch. It should be appreciated; however, the resulting reaction product can be produced directly from a reaction between the added reagents or from an intermediate from one or more of the added reagents that can be produced in the reaction mixture.

[0032] The term “contacting” may include allowing two species to react, interact, or physically touch, wherein the two species may be a compound as described herein and a protein or enzyme. In some embodiments, contacting includes allowing a compound described herein to interact with a protein or enzyme that is involved in a signaling pathway. [0033] As defined herein, the term “activation”, “activate”, “activating”, “activator” and the like in reference to a protein-inhibitor interaction means positively affecting (e.g., increasing) the activity or function of the protein relative to the activity or function of the protein in the absence of the activator. In embodiments activation means positively affecting (e.g., increasing) the concentration or levels of the protein relative to the concentration or level of the protein in the absence of the activator. The terms may reference activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein decreased in a disease. Thus, activation may include, at least in part, partially or totally increasing stimulation, increasing or enabling activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein associated with a disease (e.g., a protein which is decreased in a disease relative to a non-diseased control). Activation may include, at least in part, partially or totally increasing stimulation, increasing or enabling activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein.

[0034] The terms “agonist”, “activator”, “upregulator”, etc. refer to a substance capable of detectably increasing the expression or activity of a given gene or protein. The agonist can increase expression or activity 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a control in the absence of the agonist. In certain instances, expression or activity is 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold or higher than the expression or activity in the absence of the agonist.

[0035] As defined herein, the term “inhibition”, “inhibit”, “inhibiting” and the like in reference to a protein-inhibitor interaction means negatively affecting (e.g., decreasing) the activity or function of the protein relative to the activity or function of the protein in the absence of the inhibitor. In embodiments inhibition means negatively affecting (e.g., decreasing) the concentration or levels of the protein relative to the concentration or level of the protein in the absence of the inhibitor. In embodiments, inhibition refers to reduction of a disease or symptoms of disease. In embodiments, inhibition refers to a reduction in the activity of a particular protein target. Thus, inhibition includes, at least in part, partially or totally blocking stimulation, decreasing, preventing, or delaying activation, or inactivating, desensitizing, or down-regulating signal transduction or enzymatic activity or the amount of a protein. In embodiments, inhibition refers to a reduction of activity of a target protein resulting from a direct interaction (e.g., an inhibitor binds to the target protein). In embodiments, inhibition refers to a reduction of activity of a target protein from an indirect interaction (e.g., an inhibitor binds to a protein that activates the target protein, thereby preventing target protein activation). [0036] The terms “inhibitor”, “repressor”, “antagonist”, or “downregulator” interchangeably refer to a substance capable of detectably decreasing the expression or activity of a given gene or protein. The antagonist can decrease expression or activity 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a control in the absence of the antagonist. In certain instances, expression or activity is 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold or lower than the expression or activity in the absence of the antagonist.

[0037] The term “expression” includes any step involved in the production of the polypeptide including, but not limited to, transcription, post-transcriptional modification, translation, post- translational modification, and secretion. Expression can be detected using conventional techniques for detecting protein (e.g., ELISA, Western blotting, flow cytometry, immunofluorescence, immunohistochemistry, etc.).

[0038] The term “associated” or “associated with” in the context of a substance or substance activity or function associated with a disease means that the disease is caused by (in whole or in part), or a symptom of the disease is caused by (in whole or in part) the substance or substance activity or function.

[0039] The term “aberrant” as used herein refers to different from normal. When used to describe enzymatic activity or protein function, aberrant refers to activity or function that is greater or less than a normal control or the average of normal non-diseased control samples. Aberrant activity may refer to an amount of activity that results in a disease, wherein returning the aberrant activity to a normal or non-disease-associated amount (e.g., by administering a compound or using a method as described herein), results in reduction of the disease or one or more disease symptoms.

[0040] The term “signaling pathway” as used herein refers to a series of interactions between cellular and optionally extra-cellular components (e.g., proteins, nucleic acids, small molecules, ions, lipids) that conveys a change in one component to one or more other components, which in turn may convey a change to additional components, which is optionally propagated to other signaling pathway components. For example, binding of a thioredoxin protein with a compound as described herein may reduce the interactions between the thioredoxin protein and downstream effectors or signaling pathway components, resulting in changes in cell growth, proliferation, or survival.

[0041] The terms “disease” or “condition” refer to a state of being or health status of a patient or subject capable of being treated with a compound, pharmaceutical composition, or method provided herein. In embodiments, the disease is a coronavirus infection. In embodiments, the disease is coronavirus disease 2019 (COVID-19).

[0042] The term “coronavirus” is used in accordance with its plain ordinary meaning and refers to an RNA virus that in humans causes respiratory tract infections. Coronaviruses constitute the subfamily Orthocoronavirinae, in the family Coronaviridae, order Nidovirales, and realm Riboviria. In embodiments, the coronavirus is an enveloped viruses with a positive- sense single-stranded RNA genome.

[0043] The term “severe acute respiratory syndrome coronavirus” or “SARS-CoV-1” refers to the strain of coronavirus that causes severe acute respiratory syndrome (SARS). In embodiments, SARS-CoV-1 is an enveloped, positive-sense, single-stranded RNA virus that infects the epithelial cells within the lungs. In embodiments, the virus enters the host cell by binding to the angiotensin-converting enzyme 2 (ACE2) receptor.

[0044] The term “severe acute respiratory syndrome coronavirus 2” or “SARS-CoV-2” refers to the strain of coronavirus that causes coronavirus disease 2019 (COVID-19). In embodiments, SARS-CoV-2 is a positive-sense single-stranded RNA virus.

[0045] The terms “treating” or “treatment” refer to any indicia of success in the therapy or amelioration of an injury, disease, pathology or condition, including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the injury, pathology or condition more tolerable to the patient; slowing in the rate of degeneration or decline; making the final point of degeneration less debilitating; improving a patient’s physical or mental well-being. The treatment or amelioration of symptoms can be based on objective or subjective parameters; including the results of a physical examination, neuropsychiatric exams, and/or a psychiatric evaluation. The term “treating” and conjugations thereof, may include prevention of an injury, pathology, condition, or disease. In embodiments, treating is preventing. In embodiments, treating does not include preventing. In embodiments, treating refers to treating a subject having a disease.

[0046] “Treating” or “treatment” as used herein (and as well-understood in the art) also broadly includes any approach for obtaining beneficial or desired results in a subject’s condition, including clinical results. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of the extent of a disease, stabilizing (i.e., not worsening) the state of disease, prevention of a disease’s transmission or spread, delay or slowing of disease progression, amelioration or palliation of the disease state, diminishment of the reoccurrence of disease, and remission, whether partial or total and whether detectable or undetectable. In other words, “treatment” as used herein includes any cure, amelioration, or prevention of a disease. Treatment may prevent the disease from occurring; inhibit the disease’s spread; relieve the disease’s symptoms, fully or partially remove the disease’s underlying cause, shorten a disease’s duration, or do a combination of these things.

[0047] “Treating” and “treatment” as used herein include prophylactic treatment. Treatment methods include administering to a subject a therapeutically effective amount of an active agent. The administering step may consist of a single administration or may include a series of administrations. The length of the treatment period depends on a variety of factors, such as the severity of the condition, the age of the patient, the concentration of active agent, the activity of the compositions used in the treatment, or a combination thereof. It will also be appreciated that the effective dosage of an agent used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required. For example, the compositions are administered to the subject in an amount and for a duration sufficient to treat the patient. In embodiments, the treating or treatment is not prophylactic treatment.

[0048] The term “prevent” refers to a decrease in the occurrence of a disease or disease symptoms in a patient. As indicated above, the prevention may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment.

[0049] As used herein, a “symptom” of a disease includes any clinical or laboratory manifestation associated with the disease, and is not limited to what a subject can feel or observe.

[0050] “Patient” or “subject in need thereof’ refers to a living organism suffering from or prone to a disease or condition that can be treated by administration of a pharmaceutical composition as provided herein. Non-limiting examples include humans, other mammals, bovines, rats, mice, dogs, monkeys, goat, sheep, cows, deer, and other non-mammalian animals. In some embodiments, a patient is human.

[0051] An “effective amount” is an amount sufficient for a compound to accomplish a stated purpose relative to the absence of the compound (e.g., achieve the effect for which it is administered, treat a disease, reduce enzyme activity, increase enzyme activity, reduce a signaling pathway, or reduce one or more symptoms of a disease or condition). An example of an “effective amount” is an amount sufficient to contribute to the treatment, prevention, or reduction of a symptom or symptoms of a disease, which could also be referred to as a “therapeutically effective amount.” A “reduction” of a symptom or symptoms (and grammatical equivalents of this phrase) means decreasing of the severity or frequency of the symptom(s), or elimination of the symptom(s). A “prophylactically effective amount” of a drug is an amount of a drug that, when administered to a subject, will have the intended prophylactic effect, e.g., preventing or delaying the onset (or reoccurrence) of an injury, disease, pathology or condition, or reducing the likelihood of the onset (or reoccurrence) of an injury, disease, pathology, or condition, or their symptoms. The full prophylactic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses. Thus, a prophylactically effective amount may be administered in one or more administrations. An “activity decreasing amount,” as used herein, refers to an amount of antagonist required to decrease the activity of an enzyme relative to the absence of the antagonist. A “function disrupting amount,” as used herein, refers to the amount of antagonist required to disrupt the function of an enzyme or protein relative to the absence of the antagonist. The exact amounts will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols. 1-3, 1992); Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); Pickar, Dosage Calculations (1999); and Remington: The Science and Practice of Pharmacy, 20th Edition, 2003, Gennaro, Ed., Lippincott, Williams & Wilkins).

[0052] For any compound described herein, the therapeutically effective amount can be initially determined from binding assays or cell culture assays. Target concentrations will be those concentrations of active compound(s) that are capable of achieving the methods described herein, as measured using the methods described herein or known in the art.

[0053] As is well known in the art, therapeutically effective amounts for use in humans can also be determined from animal models. For example, a dose for humans can be formulated to achieve a concentration that has been found to be effective in animals. The dosage in humans can be adjusted by monitoring compounds effectiveness and adjusting the dosage upwards or downwards, as described above. Adjusting the dose to achieve maximal efficacy in humans based on the methods described above and other methods is well within the capabilities of the ordinarily skilled artisan.

[0054] The term “therapeutically effective amount,” as used herein, refers to that amount of the therapeutic agent sufficient to ameliorate the disorder, as described above. For example, for the given parameter, a therapeutically effective amount will show an increase or decrease of at least 5%, 10%, 15%, 20%, 25%, 40%, 50%, 60%, 75%, 80%, 90%, or at least 100%. Therapeutic efficacy can also be expressed as “-fold” increase or decrease. For example, a therapeutically effective amount can have at least a 1.2-fold, 1.5-fold, 2-fold, 5-fold, or more effect over a control.

[0055] Dosages may be varied depending upon the requirements of the patient and the compound being employed. The dose administered to a patient, in the context of the present disclosure, should be sufficient to effect a beneficial therapeutic response in the patient over time. The size of the dose also will be determined by the existence, nature, and extent of any adverse side-effects. Determination of the proper dosage for a particular situation is within the skill of the practitioner. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under circumstances is reached. Dosage amounts and intervals can be adjusted individually to provide levels of the administered compound effective for the particular clinical indication being treated. This will provide a therapeutic regimen that is commensurate with the severity of the individual’s disease state.

[0056] As used herein, the term “administering” means oral administration, administration as an aerosol, dry powder, nasal spray, suppository, topical contact, intravenous, parenteral, intraperitoneal, intramuscular, intralesional, intrathecal, intranasal or subcutaneous administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject. Administration is by any route, including parenteral and transmucosal (e.g., buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal). Parenteral administration includes, e.g., intravenous, intramuscular, intra-arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial. Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc. In embodiments, the administering does not include administration of any active agent other than the recited active agent.

[0057] “Co-administer” it is meant that a composition described herein is administered at the same time, just prior to, or just after the administration of one or more additional therapies. The compounds provided herein can be administered alone or can be coadministered to the patient. Coadministration is meant to include simultaneous or sequential administration of the compounds individually or in combination (more than one compound). Thus, the preparations can also be combined, when desired, with other active substances (e.g., to reduce metabolic degradation). The compositions of the present disclosure can be delivered transdermally, by a topical route, or formulated as applicator sticks, solutions, suspensions, emulsions, gels, creams, ointments, pastes, jellies, paints, powders, and aerosols. The preparations may also be combined with inhaled mucolytics (e.g., rhDNase, as known in the art) or with inhaled bronchodilators (short or long acting beta agonists, short or long acting anticholinergics), inhaled corticosteroids, or inhaled antibiotics to improve the efficacy of these drugs by providing additive or synergistic effects. The compositions of the present invention can be delivered transdermally, by a topical route, formulated as applicator sticks, solutions, suspensions, emulsions, gels, creams, ointments, nanoparticles, pastes, jellies, paints, powders, and aerosols. Oral preparations include tablets, pills, powder, dragees, capsules, liquids, lozenges, cachets, gels, syrups, slurries, suspensions, etc., suitable for ingestion by the patient. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules. Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water/propylene glycol solutions. The compositions of the present invention may additionally include components to provide sustained release and/or comfort. Such components include high molecular weight, anionic mucomimetic polymers, gelling polysaccharides and finely-divided drug carrier substrates. These components are discussed in greater detail in U.S. Pat. Nos.

4,911,920; 5,403,841; 5,212,162; and 4,861,760. The entire contents of these patents are incorporated herein by reference in their entirety for all purposes. The compositions of the present invention can also be delivered as microspheres for slow release in the body. For example, microspheres can be administered via intradermal injection of drug-containing microspheres, which slowly release subcutaneously (see Rao, J. Biomater Sci. Polym. Ed. 7:623- 645, 1995; as biodegradable and injectable gel formulations (see, e.g., Gao Pharm. Res. 12:857- 863, 1995); or, as microspheres for oral administration (see, e.g., Eyles, J. Pharm. Pharmacol. 49:669-674, 1997). In another embodiment, the formulations of the compositions of the present invention can be delivered by the use of liposomes which fuse with the cellular membrane or are endocytosed, i.e., by employing receptor ligands attached to the liposome, that bind to surface membrane protein receptors of the cell resulting in endocytosis. By using liposomes, particularly where the liposome surface carries receptor ligands specific for target cells, or are otherwise preferentially directed to a specific organ, one can focus the delivery of the compositions of the present invention into the target cells in vivo. (See, e.g., Al-Muhammed, J. Microencapsul. 13:293-306, 1996; Chonn, Curr. Opin. Biotechnol. 6:698-708, 1995; Ostro, Am. J. Hosp. Pharm. 46:1576-1587, 1989). [0058] A “cell” as used herein, refers to a cell carrying out metabolic or other function sufficient to preserve or replicate its genomic DNA. A cell can be identified by well-known methods in the art including, for example, presence of an intact membrane, staining by a particular dye, ability to produce progeny or, in the case of a gamete, ability to combine with a second gamete to produce a viable offspring. Cells may include prokaryotic and eukaryotic cells. Prokaryotic cells include but are not limited to bacteria. Eukaryotic cells include but are not limited to yeast cells and cells derived from plants and animals, for example mammalian, insect (e.g., spodoptera) and human cells. Cells may be useful when they are naturally nonadherent or have been treated not to adhere to surfaces, for example by trypsinization.

[0059] “Control” or “control experiment” is used in accordance with its plain ordinary meaning and refers to an experiment in which the subjects or reagents of the experiment are treated as in a parallel experiment except for omission of a procedure, reagent, or variable of the experiment. In some instances, the control is used as a standard of comparison in evaluating experimental effects. In some embodiments, a control is the measurement of the activity of a protein in the absence of a compound as described herein (including embodiments and examples).

[0060] The terms “bind” and “bound” as used herein is used in accordance with its plain and ordinary meaning and refers to the association between atoms or molecules. The association can be covalent (e.g., by a covalent bond or linker) or non-covalent (e.g., electrostatic interactions (e.g., ionic bond, hydrogen bond, or halogen bond), van der Waals interactions (e.g., dipole- dipole, dipole-induced dipole, or London dispersion), ring stacking (pi effects), hydrophobic interactions, and the like).

[0061] As used herein, the term “conjugated” when referring to two moieties means the two moieties are bonded, wherein the bond or bonds connecting the two moieties may be covalent or non-covalent. In embodiments, the two moieties are covalently bonded to each other (e.g., directly or through a covalently bonded intermediary). In embodiments, the two moieties are non-covalently bonded (e.g., through ionic bond(s), van der Waals bond(s)/interactions, hydrogen bond(s), polar bond(s), or combinations or mixtures thereof).

[0062] The term “thiol” is used in accordance with its ordinary meaning in the art and refers A “thiol-containing compound” is a compound with at least one thiol

(-SH) moiety. A thiol-containing compound having only one thiol moiety is referred to herein as a “monothiol.” A thiol-containing compound having only two thiol moieties is referred to herein as a “dithiol.” Non-limiting examples of thiol-containing comounds, including monothiols and dithiols, are provided herein. Additional examples of thiol-containing compounds are known in the art.

II. Methods

[0063] In an aspect is provided a method of treating a coronavirus infection in a subject in need thereof. In embodiments, the method includes administering to the subject an effective amount of a thiol-containing compound in a pharmaceutically acceptable carrier.

[0064] In an aspect is provided a method of treating inflammation in a subject in need thereof, the method including administering to the subject in need thereof an effective amount of a thiol-containing compound in a pharmaceutically acceptable carrier. In embodiments, the inflammation is in the lung. In embodiments, the inflammation is a symptom of infection, such as coronavirus infection. In embodiments, the inflammation is a consequence of infection, such as coronavirus infection.

[0065] In embodiments, the coronavirus infection is a SARS-CoV-1 infection. In embodiments, the coronavirus infection is Severe Acute Respiratory Disease (SARS).

[0066] In embodiments, the coronavirus infection is a SARS-CoV-2 virus infection. In embodiments, the subject has or is suspected of having COVID-19.

[0067] In embodiments, the subject in need thereof has post acute COVID syndrome (e.g., COVID long haul syndrome) and treatment is administered because the syndrome may be caused by persistent viral infection in the airways and lungs. In embodiments, the coronavirus infection is an HCoV-NL63 coronavirus infection. In embodiments, HCoV-NL63 coronavirus infection is a cause of the common cold. In embodiments, HCoV-NL63 coronavirus infection is a cause of pneumonia. In embodiments, the coronavirus infection is an HCoV-229E coronavirus infection. In embodiments, HCoV-229E coronavirus infection is a cause of the common cold.

In embodiments, HCoV-229E coronavirus infection is a cause of pneumonia. In embodiments, the coronavirus infection is an HCoV-OC43 coronavirus infection. In embodiments, HCoV- OC43 coronavirus infection is a cause of the common cold. In embodiments, HCoV-OC43 coronavirus infection is a cause of pneumonia. In embodiments, the coronavirus infection is an HCoV-HKUl coronavirus infection. In embodiments, HCoV-HKUl coronavirus infection is a cause of the common cold. In embodiments, HCoV-HKUl coronavirus infection is a cause of pneumonia. [0068] In embodiments, the effective amount is administered within 48-96 hours of the onset of one or more symptoms of the infection. In embodiments, the effective amount is administered within 48 hours of the onset of the one or more symptoms. In embodiments, the effective amount is administered within 72 hours of the onset of the one or more symptoms. In embodiments, the symptom is any symptom associated with coronavirus infection. Examples of symptoms include, but are not limited to, cough, shortness of breath or difficulty breathing, fever, chills, repeated shaking with chills, muscle pain, headache, sore throat, and new loss of taste or smell.

[0069] In embodiments, the effective amount is effective to reduce viral load in the subject.

In embodiments, the viral load is reduced in the lung.

[0070] In embodiments, the thiol-containing compound is administered by pulmonary delivery. In embodiments, the thiol-containing compound is administered as a liquid aerosol or a dry powder. In embodiments, the thiol-containing compound is administered orally. In embodiments, the thiol-containing compound is administered as a lozenge. In embodiments, the thiol-containing compound is administered as a nasal spray. In embodiments, the thiol- containing compound is administered intravenously.

[0071] In embodiments, the subject is not hospitalized. In embodiments, the subject is hospitalized. In embodiments, is in an intensive care unit.

[0072] In embodiments, the thiol-containing compound is a monothiol. In embodiments, the thiol-containing compound includes two or more thiols (e.g., 3, 4, or 5 thiols). In embodiments, the thiol-containing compound is a dithiol.

[0073] In embodiments, the thiol-containing compound has a molecular weight of at least 164 g/mol.

[0074] In embodiments, the thiol-containing compound is thiomandelic acid, DL-Captopril, DL-Thiophan, N-acetylcysteine, Meso-2,3,-dimercaptosuccinic acid, 2,3-dimercaprol, D-(-)- Penicillamine, Glutathione, L-Cysteine, Zofenoprilat, Tiopronin, N-acystelyn, carbocysteine, cysteamine, sodium-2-mercaptoethane sulfonate, WR-1065, an analogue or derivative thereof, or a combination thereof. In embodiments, the thiol-containing compound is thiomandelic acid. In embodiments, the thiol-containing compound is DL-Captopril. In embodiments, the thiol- containing compound is DL-Thiophan. In embodiments, the thiol-containing compound is N- acetylcysteine. In embodiments, the thiol-containing compound is Meso-2,3,- dimercaptosuccinic acid. In embodiments, the thiol-containing compound is 2,3-dimercaprol. In embodiments, the thiol-containing compound is D-(-)-Penicillamine. In embodiments, the thiol- containing compound is Glutathione. In embodiments, the thiol-containing compound is L- Cysteine. In embodiments, the thiol-containing compound is Zofenoprilat. In embodiments, the thiol-containing compound is Tiopronin. In embodiments, the thiol-containing compound is N- acystelyn. In embodiments, the thiol-containing compound is carbocysteine. In embodiments, the thiol-containing compound is cysteamine. In embodiments, the thiol-containing compound is sodium-2 -mercaptoethane sulfonate. In embodiments, the thiol-containing compound is WR- 1065. In embodiments, the thiol-containing compound is Bucillamine. In embodiments, the thiol-containing compound is Amifostine.

[0075] In embodiments, the thiol-containing compound is N-acetylcysteine, sodium-2 - mercaptoethane sulfonate, Tiopronin, cysteamine, Amifostine, WR-1065, Erdosteine, Met I, Penicillamine, Glutathione, Bucillamine, Dimercaptosuccinic acid, 2,3-Dimercaprol, or carbocysteine. In embodiments, the thiol-containing compound is N-acetylcysteine. In embodiments, the thiol-containing compound is sodium-2 -mercaptoethane sulfonate. In embodiments, the thiol-containing compound is Tiopronin. In embodiments, the thiol-containing compound is cysteamine. In embodiments, the thiol-containing compound is Amifostine. In embodiments, the thiol-containing compound is WR-1065. In embodiments, the thiol-containing compound is Erdosteine. In embodiments, the thiol-containing compound is Met I. In embodiments, the thiol-containing compound is Penicillamine. In embodiments, the thiol- containing compound is Glutathione. In embodiments, the thiol-containing compound is Bucillamine. In embodiments, the thiol-containing compound is Dimercaptosuccinic acid. In embodiments, the thiol-containing compound is 2,3-Dimercaprol. In embodiments, the thiol- containing compound is carbocysteine.

[0076] Additional non-limiting examples of thiol-containing compounds are disclosed in US9963427B2 and US10106551B2, which are incorporated herein by reference. In embodiments, the thiol-containing compound is selected from Table 1.

[0077] Table 1.

[0078] In embodiments, the administering the thiol-containing compound includes administering a prodrug thereof, wherein the prodrug is metabolized by the subject to form the thiol-containing compound. In embodiments, the prodrug is amifostine.

[0079] In embodiments, the thiol-containing compound is not N-acetylcysteine, glutathione, cysteamine, or dithiothreitol. In embodiments, the thiol-containing compound is not N- acetylcysteine. In embodiments, the thiol-containing compound is not glutathione. In embodiments, the thiol-containing compound is not dithiothreitol. In embodiments, the thiol- containing compound is not cysteamine. In embodiments, the thiol-containing compound is not any of N-acetylcysteine, glutathione, cysteamine, or dithiothreitol. In embodiments, the thiol- containing compound is not bucillamine.

[0080] In embodiments, the thiol-containing compound is coadministered with one or more additional thiol-containing compounds. In embodiments, the thiol-containing compound is coadministered with one or more additional antiviral compounds, and/or one or more anti inflammatory compounds. In general, an antiviral compound is a compound known in the art for use in inhibiting a virus, such as inhibiting viral entry or replication. Several antiviral compounds are known in the art (e.g., remdesivir). In general, an anti-inflammatory compound is a compound known in the art for use in treating inflammation. In embodiments, the coadministered compound is administered by the same route as the thiol-containing compound. In embodiments, the coadministered compound is administered by a different route from that of the thiol-containing compound. In embodiments, a therapeutic composition comprises two or more thiol-containing compounds. In embodiments, a first thiol-containing compound is administered at a first time via a first administration route (e.g., intravenous), and a second thiol- containing compound is administered at a second time via a second administration route (e.g., oral). The first thiol-containing compound administered at the first time and the second thiol- containing compound administered at the second time may be the same or different.

[0081] In embodiments, the thiol-containing compound is not coadministered with heparin, an antibiotic, or methylene blue. In embodiments, the thiol-containing compound is not coadministered with heparin. In embodiments, the thiol-containing compound is not coadministered with an antibiotic. In embodiments, the thiol-containing compound is not coadministered with methylene blue. In embodiments, the thiol-containing compound is not coadministered with any one of heparin, an antibiotic, or methylene blue.

[0082] In an aspect is provided a thiol-containing compound for use in the methods provided herein including embodiments thereof.

III. Formulations

[0083] The thiol-containing compounds disclosed herein can be prepared and administered in a wide variety of inhalation, oral, parenteral, and topical dosage forms. In embodiments, the thiol-containing compound (e.g., as described herein) is administered by injection (e.g., intravenously, intramuscularly, intracutaneously, subcutaneously, intraduodenally, or intraperitoneally). In embodiments, the thiol-containing compound (e.g., as described herein) is administered intravenously. In embodiments, the thiol-containing compound (e.g., as described herein) is administered intramuscularly. In embodiments, the thiol-containing compound (e.g., as described herein) is administered intracutaneously. In embodiments, the thiol-containing compound (e.g., as described herein) is administered subcutaneously. In embodiments, the thiol-containing compound (e.g., as described herein) is administered intraduodenally. In embodiments, the thiol-containing compound (e.g., as described herein) is administered intraperitoneally. In embodiments, the thiol-containing compound (e.g., as described herein) is administered by pulmonary delivery. In embodiments, the thiol-containing compound (e.g., as described herein) is administered as a liquid aerosol or a dry powder. In embodiments, the thiol- containing compound (e.g., as described herein) is administered by inhalation. In embodiments, the thiol-containing compound (e.g., as described herein) is administered orally. In embodiments, the thiol-containing compound (e.g., as described herein) is administered by tablet, pill, or capsule. In embodiments, the thiol-containing compound (e.g., as described herein) is administered as a lozenge. In embodiments, the thiol-containing compound (e.g., as described herein) is administered by the intranasal route. In embodiments, the thiol-containing compound (e.g., as described herein) is administered as a nasal spray. In embodiments, the thiol-containing compound (e.g., as described herein) is administered transdermally. It is also envisioned that multiple routes of administration (e.g., intramuscular, oral, transdermal) can be used to administer the thiol-containing compounds. Accordingly, the present disclosure also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier or excipient and one or more thiol-containing compounds. In embodiments, the thiol-containing compound (e.g., as described herein) is administered topically to an eye. In embodiments, the thiol-containing compound (e.g., as described herein) is administered in an eye drop formulation.

[0084] For preparing pharmaceutical compositions from the thiol-containing compounds described herein, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, pills, capsules, lozenges, wafers, cachets, suppositories, and dispersible granules. A solid carrier can be one or more substance that may also act as diluents, flavoring agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.

[0085] In embodiments, a powder is provided in which the carrier is a finely divided solid in a mixture with the finely divided active component. In embodiments, a tablet is provided in which the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.

[0086] The powders and tablets preferably contain from 5% to 100% of the active compound. Suitable carriers include, but are not limited to, magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like.

[0087] Liquid form preparations include solutions (e.g., for oral ingestion), including, suspensions, elixirs, syrups, solutions, emulsions, for example, water or water/propylene glycol solutions, and effervescent solutions. For parenteral injection, liquid preparations can be formulated in solution in aqueous polyethylene glycol solution.

[0088] When parenteral application is needed or desired, particularly suitable admixtures for the compounds of the invention are injectable, sterile solutions, preferably oily or aqueous solutions, as well as suspensions, emulsions, or implants, including suppositories. In particular, carriers for parenteral administration include aqueous solutions of dextrose, saline, pure water, ethanol, glycerol, propylene glycol, peanut oil, sesame oil, polyoxyethylene-block polymers, and the like. Ampoules are convenient unit dosages. In embodiments, the thiol-containing compound (e.g., as described herein) is incorporated into liposomes or administered via transdermal pumps or patches. Pharmaceutical admixtures suitable for use in the present invention include those described, for example, in PHARMACEUTICAL SCIENCES (17th Ed., Mack Pub. Co., Easton, PA) and WO 96/05309, the teachings of both of which are hereby incorporated by reference.

[0089] Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizers, and thickening agents as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well-known suspending agents.

[0090] Also included are solid form preparations that are intended to be converted, shortly before use, to liquid form preparations for oral or inhaled administration. Such liquid forms include solutions, suspensions, and emulsions. These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.

[0091] The pharmaceutical preparation is preferably in unit dosage form. In such form the preparation is subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.

[0092] The quantity of active component in a unit dose preparation may be varied or adjusted, such as from 0.1 mg to 10,000 mg, 1.0 mg to 1000 mg, or 10 mg to 500 mg, according to the particular application and the potency of the active component. The composition can, if desired, also contain other compatible therapeutic agents.

[0093] Some compounds may have limited solubility in water and therefore may require a surfactant or other appropriate co-solvent in the composition. Such co-solvents include: Polysorbate 20, 60, and 80; Pluronic F-68, F-84, and P-103; cyclodextrin; and polyoxyl 35 castor oil. Such co-solvents are typically employed at a level between about 0.01 % and about 2% by weight.

[0094] Viscosity greater than that of simple aqueous solutions may be desirable to decrease variability in dispensing the formulations, to decrease physical separation of components of a suspension or emulsion of formulation, and/or otherwise to improve the formulation. Such viscosity building agents include, for example, polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose, hydroxy propyl methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose, hydroxy propyl cellulose, chondroitin sulfate and salts thereof, hyaluronic acid and salts thereof, and combinations of the foregoing. Such agents are typically employed at a level between about 0.01% and about 2% by weight.

[0095] The compositions of the present invention may additionally include components to provide sustained release and/or comfort. Such components include high molecular weight, anionic mucomimetic polymers, gelling polysaccharides, and finely-divided drug carrier substrates. These components are discussed in greater detail in U.S. Pat. Nos. 4,911,920; 5,403,841; 5,212,162; and 4,861,760. The entire contents of these patents are incorporated herein by reference in their entirety for all purposes.

[0096] In an aspect is provided a pulmonary pharmaceutical composition comprising a pulmonary pharmaceutical carrier and a thiol-containing compound. The terms “pulmonary pharmaceutical composition” and the like refer to pharmaceutical compositions intended for pulmonary administration. The terms “pulmonary administration” and the like refer, in the usual and customary sense, to administration to achieve inhalation therapy. The term “inhalation therapy” and the like refer to direct delivery of medications to the lungs by inhalation. In embodiments, the thiol-containing compounds disclosed herein are effective when delivered directly to the lung by an inhaled drug delivery system. The term “pulmonary pharmaceutical liquid” refers to a pulmonary pharmaceutical composition which is a liquid. The terms “pulmonary pharmaceutical solid,” “pulmonary pharmaceutical solid” and the like refer to a pulmonary pharmaceutical composition which is a solid (e.g., a powder).

[0097] There are three categories of inhaled drug delivery systems: (i) nebulizers; (ii) pressurized metered-dose inhalers (pMDIs); (iii) dry powder inhalers (DPIs). Nebulizers are distinctly different from both pMDIs and DPIs, in that the active agent is dissolved or suspended in a polar liquid, e.g., water. In contrast, pMDIs and DPIs are bolus drug delivery devices that contain active agent (e.g., solid thiol-containing agent), suspended or dissolved in a nonpolar volatile propellant or in a dry powder mix that is fluidized when the patient inhales. pMDIs and DPIs have considerably reduced treatment time compared with nebulizers. The term “pulmonary pharmaceutical delivery device” and the like refer to an inhaled drug delivery system suitable for delivery (e.g., pulmonary delivery) of a pharmaceutical composition.

[0098] Without wishing to be bound by any theory, it is believed that the lung deposition characteristics and efficacy of an aerosol depend largely on the particle or droplet size. For example, particles of more than 10 pm in diameter are most likely to deposit in the mouth and throat, for those of 5-10 mhi diameter a transition from mouth to airway deposition occurs, and particles smaller than 5 pm in diameter deposit more frequently in the lower airways and are appropriate for pharmaceutical aerosols (e.g., pulmonary pharmaceutical compositions). Aerodynamic particle size distribution is measured by methods known in the art, e.g., cascade impaction method. Micronization is a conventional approach for size reduction. Additional drug particle engineering technologies includes spray drying, sonocrystalization, or super critical fluids, and the like as known in the art. In embodiments, the particle is a nanoparticle, as known in the art. In all of these technologies, the particles can be delivered alone or co-formulated with carriers.

[0099] In embodiments, ideal inhaled particles are characterized as having uniform particle size with mono-dispersion, uniform density, non-cohesiveness, no agglomeration, no compaction, excellent flowability, and ready dispersal when delivered as an aerosol.

[0100] In embodiments, the attributes of an optimized inhaled delivery system include stability (i.e., consistent delivered dose through inhaler life), consistent aerodynamic particle size distribution (i.e., fine particle dose/fraction), and chemical and performance stability, as known in the art.

[0101] In embodiments, formulation considerations for the pulmonary pharmaceutical composition disclosed herein include consistent product performance on stability and through the labeled number of doses, uniform formulation upon shaking to ensure metering and delivery of accurate and consistent doses, drug suspension stabilized by forming loose agglomerates and readily re-dispersed upon shaking after storage, no particle growth due to aggregation or crystal growth to ensure aerosolization performance, no drug loss due to deposition on dispenser to ensure consistent doses through inhaler life, and protection from moisture ingression to ensure long term stability.

[0102] Regarding nebulizers, as known in the art, nebulizers (“atomizers”) may, for example, employ compressor to force a gas air (or a blended mixture of air and oxygen through a solution) or electrical means (e.g., piezoelectric power to break up pharmaceutical compositions (e.g., solutions and suspensions)) into small aerosol droplets that can be directly inhaled from the nebulizer. The term “aerosol” and the like refer, in the usual and customary sense to a mixture of gas and liquid particles. The term “jet nebulizer” and the like refer, in the usual and customary sense, to any of a variety of devices connected by tubing to a compressor that causes compressed air or oxygen to flow at high velocity through a liquid medicine to turn it into an aerosol, which is then inhaled by the patient. Jet nebulizers are commonly used for patients in hospitals who have difficulty using inhalers or who require higher doses of drug than can be delivered with hand held devices such pressurized metered dose inhalers (pMDIs) or dry powder inhalers (DPIs). Jet nebulizers are also common in pediatric practice. The term “vibrating mesh nebulizer” refers to a nebulizer that generates aerosols as liquid passes through a mesh that is oscilated (e.g., by a piezo-element) to generate ultrasonic frequencies, and are becoming preferred devices for home use.

[0103] In embodiments, a thiol-containing compound disclosed herein is administered in an aerosol as described herein. In embodiments, a thiol-containing compound disclosed herein is formulated in a vehicle solution. In embodiments, the vehicle solution includes sodium citrate. In embodiments, the vehicle solution includes sodium chloride. In embodiments, the vehicle solution includes sodium citrate and sodium chloride. In embodiments, the vehicle solution is 20 mM sodium citrate, pH 4.5 with 38.5 mM NaCl. In embodiments, the vehicle solution is about 5 mM to about 50 mM sodium citrate, with a pH from about 3.5 to about 7, with about 10 mM to about 308 mM NaCl.

[0104] In embodiments, the vehicle solution includes about 5 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 7.5 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 10 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 12.5 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 15 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 17.5 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 20 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 22.5 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 25 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 27.5 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 30 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 32.5 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 35 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 37.5 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 40 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 42.5 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 45 mM to about 50 mM sodium citrate. In embodiments, the vehicle solution includes about 47.5 mM to about 50 mM sodium citrate. [0105] In embodiments, the vehicle solution includes about 5 mM to about 47.5 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 45 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 42.5 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 40 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 37.5 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 35 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 32.5 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 30 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 27.5 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 25 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 22.5 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 20 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 17.5 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 15 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 12.5 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 10 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM to about 7.5 mM sodium citrate. In embodiments, the vehicle solution includes about 5 mM, 7.5 mM, 10 mM, 12.5 mM, 15 mM, 17.5 mM, 20 mM, 22.5 mM, 25 mM, 27.5 mM, 30 mM, 32.5 mM, 35 mM, 37.5 mM, 40 mM, 42.5 mM, 45 mM, 47.5 mM or 50 mM sodium citrate. In embodiments, the vehicle solution includes about 20 mM sodium citrate. In embodiments, the vehicle solution includes 20 mM sodium citrate.

[0106] In embodiments, the vehicle solution has a pH from about 3.5 to about 7. In embodiments, the vehicle solution has a pH from about 3.6 to about 7. In embodiments, the vehicle solution has a pH from about 3.7 to about 7. In embodiments, the vehicle solution has a pH from about 3.8 to about 7. In embodiments, the vehicle solution has a pH from about 3.9 to about 7. In embodiments, the vehicle solution has a pH from about 4 to about 7. In embodiments, the vehicle solution has a pH from about 4.1 to about 7. In embodiments, the vehicle solution has a pH from about 4.2 to about 7. In embodiments, the vehicle solution has a pH from about 4.3 to about 7. In embodiments, the vehicle solution has a pH from about 4.4 to about 7. In embodiments, the vehicle solution has a pH from about 4.5 to about 7. In embodiments, the vehicle solution has a pH from about 4.6 to about 7. In embodiments, the vehicle solution has a pH from about 4.7 to about 7. In embodiments, the vehicle solution has a pH from about 4.8 to about 7. In embodiments, the vehicle solution has a pH from about 4.9 to about 7. In embodiments, the vehicle solution has a pH from about 5 to about 7. In embodiments, the vehicle solution has a pH from about 5.1 to about 7. In embodiments, the vehicle solution has a pH from about 5.2 to about 7. In embodiments, the vehicle solution has a pH from about 5.3 to about 7. In embodiments, the vehicle solution has a pH from about 5.4 to about 7. In embodiments, the vehicle solution has a pH from about 5.5 to about 7. In embodiments, the vehicle solution has a pH from about 5.6 to about 7. In embodiments, the vehicle solution has a pH from about 5.7 to about 7. In embodiments, the vehicle solution has a pH from about 5.8 to about 7. In embodiments, the vehicle solution has a pH from about 5.9 to about 7. In embodiments, the vehicle solution has a pH from about 6 to about 7. In embodiments, the vehicle solution has a pH from about 6.1 to about 7. In embodiments, the vehicle solution has a pH from about 6.2 to about 7. In embodiments, the vehicle solution has a pH from about 6.3 to about 7. In embodiments, the vehicle solution has a pH from about 6.4 to about 7. In embodiments, the vehicle solution has a pH from about 6.5 to about 7. In embodiments, the vehicle solution has a pH from about 6.6 to about 7. In embodiments, the vehicle solution has a pH from about 6.7 to about 7. In embodiments, the vehicle solution has a pH from about 6.8 to about 7. In embodiments, the vehicle solution has a pH from about 6.9 to about 7.

[0107] In embodiments, the vehicle solution has a pH from about 3.5 to about 6.9. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.8. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.7. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.6. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.5. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.4. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.3. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.2. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.1. In embodiments, the vehicle solution has a pH from about 3.5 to about 6. In embodiments, the vehicle solution has a pH from about 3.5 to about 5.9. In embodiments, the vehicle solution has a pH from about 3.5 to about 5.8. In embodiments, the vehicle solution has a pH from about 3.5 to about 5.7. In embodiments, the vehicle solution has a pH from about 3.5 to about 5.6. In embodiments, the vehicle solution has a pH from about 3.5 to about 5.5. In embodiments, the vehicle solution has a pH from about 3.5 to about 5.4. In embodiments, the vehicle solution has a pH from about 3.5 to about 5.3. In embodiments, the vehicle solution has a pH from about 3.5 to about 5.2. In embodiments, the vehicle solution has a pH from about 3.5 to about 5.1. In embodiments, the vehicle solution has a pH from about 3.5 to about 5. In embodiments, the vehicle solution has a pH from about 3.5 to about 4.9. In embodiments, the vehicle solution has a pH from about 3.5 to about 4.8. In embodiments, the vehicle solution has a pH from about 3.5 to about 4.7. In embodiments, the vehicle solution has a pH from about 3.5 to about 4.6. In embodiments, the vehicle solution has a pH from about 3.5 to about 4.5. In embodiments, the vehicle solution has a pH from about 3.5 to about 4.4. In embodiments, the vehicle solution has a pH from about 3.5 to about 4.3. In embodiments, the vehicle solution has a pH from about 3.5 to about 4.2. In embodiments, the vehicle solution has a pH from about 3.5 to about 4.1. In embodiments, the vehicle solution has a pH from about 3.5 to about 4. In embodiments, the vehicle solution has a pH from about 3.5 to about 3.9. In embodiments, the vehicle solution has a pH from about 3.5 to about 3.8. In embodiments, the vehicle solution has a pH from about 3.5 to about 3.7. In embodiments, the vehicle solution has a pH from about 3.5 to about 3.6. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.9. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.8. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.7. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.6. In embodiments, the vehicle solution has a pH from about 3.5 to about 6.5. In embodiments, the vehicle solution has a pH of about 3.5, 3.6, 3.7, 3.8, 3.9, 4, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, or 7. In embodiments, the vehicle solution has a pH of about 4.5. In embodiments, the vehicle solution has a pH of 4.5.

[0108] In embodiments, the vehicle solution includes about 10 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 20 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 30 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 40 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 50 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 60 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 70 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 80 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 90 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 100 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 110 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 120 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 130 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 140 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 150 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 160 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 170 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 180 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 190 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 200 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 210 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 220 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 230 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 240 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 250 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 260 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 270 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 280 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 290 mM to about 308 mM NaCl. In embodiments, the vehicle solution includes about 300 mM to about 308 mM NaCl.

[0109] In embodiments, the vehicle solution includes about 10 mM to about 300 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 290 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 280 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 270 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 260 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 250 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 240 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 230 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 220 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 210 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 200 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 190 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 180 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 170 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 160 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 150 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 140 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 130 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 120 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 110 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 100 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 90 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 80 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 70 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 60 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 50 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 40 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 30 mM NaCl. In embodiments, the vehicle solution includes about 10 mM to about 20 mM NaCl. In embodiments, the vehicle solution includes about 10 mM, 20 mM, 30 mM, 40 mM, 50 mM, 60 mM, 70 mM, 80 mM, 90 mM, 100 mM, 110 mM, 120 mM, 130 mM, 140 mM, 150 mM, 160 mM, 170 mM, 180 mM, 190 mM, 200 mM, 210 mM, 220 mM, 230 mM, 240 mM, 250 mM,

260 mM, 270 mM, 280 mM, 290 mM, 300 mM, or 308 mM NaCl. In embodiments, the vehicle solution includes about 38.5 mM NaCl. In embodiments, the vehicle solution includes 38.5 mM NaCl.

[0110] A dry powder inhaler (DPI) is a device that delivers medication to the lungs in the form of a dry powder. When a DPI is actuated, the formulation is fluidized and enters the patient’s airways.

[0111] In embodiments, a thiol-containing compound disclosed herein is administered in an amorphous powder. Non-limiting descriptions relating to amorphous powders are provided in Chen et al. 2016 Amorphous powders for inhalation drug delivery Advanced Drug Delivery Reviews 100: 102-115, the entire content of which is incorporated by reference. In embodiments, the amorphous powder is manufactured by spray-drying. In embodiments, a thiol-containing compound is spray-dried with an excipient suitable for inhalation.

[0112] In embodiments, a thiol-containing compound disclosed herein is administered as a micronized powder.

[0113] In embodiments, a powder composition for use in a DPI is packaged in single dose quantities in blisters or gel capsules containing the powdered medication to be drawn into the lungs by the user’s own breath.

[0114] In embodiments, a DPI formulation must undergo flow, fluidization, and de aggregation. In embodiments, an excipient is added to enhance the physical or chemical stability of the active pharmaceutical ingredient mechanical properties, and/or its pharmaceutical properties, such as dissolution and permeation. [0115] In embodiments, a DPI formulation includes loose agglomerates. In embodiments, the agglomerates consist of particles of disparate sizes, as is the case when agent is prepared with large carrier particles, or particles of similar sizes prepared by unique methods of formation that result in ease of dispersion. In embodiments, a DPI formulation of particles of similar sizes is prepared by spray-drying.

[0116] In embodiments, after the formulation has been produced, it is filled into capsules, multi-dose blisters, or reservoirs for use with the inhaler device.

[0117] Regarding pressurized metered-dose inhalers (pMDIs), a formulation can be made up of the agent (e.g., a thiol-containing compound as described herein), a liquefied gas propellant and, in many cases, stabilizing excipients. The actuator contains the mating discharge nozzle and generally includes a dust cap to prevent contamination. Actuation of the device releases a single metered dose of the formulation which contains the medication either dissolved or suspended in the propellant. Breakup of the volatile propellant into droplets, followed by rapid evaporation of these droplets, results in the generation of an aerosol consisting of micrometer sized medication particles that are then inhaled. One of the most crucial components of a MDI is its propellant. The propellant provides the force to generate the aerosol cloud and is also the medium in which the active component must be suspended or dissolved. Propellants in MDIs typically make up more than 99% of the delivered dose, so it is the properties of the propellant that dominate more than any other individual factor. Suitable propellants must pass a stringent set of criteria, they must: have a boiling point in the range -100 to +30°C have a density of approximately 1.2 to 1.5 g cm 3 (approximately that of the drug to be suspended or dissolved) have a vapor pressure of 40 to 80 psig have no toxicity to the patient, be non-flammable and be able to dissolve common additives. Active ingredients can be either fully soluble or fully insoluble. In the early days of MDIs the most commonly used propellants were the chlorofluorocarbons, but hydrofluoroalkane propellants are now preferred because they have fewer environmental toxicities. General considerations for metered dose inhalers include consideration of the following: agent is dissolved in the liquefied propellant, compliance with applicable rules (e.g., formulation agent (e.g., HFA propellant, surfactant, so-solvent and/or excipient)), container closure system (e.g., can, metering valve), actuator, and dose compliance device, as known in the art. Suspension formulation issues can include micronized drug particles suspended in the liquefied propellant (e.g., air, CO2, HFA134a, 227, and the like). The suspension formulation may contain surfactant and co-solvent to aid suspension, particularly with respect irregular particles, polydispersed (e.g., 0.5-10pm) particles, or amorphous/crystalline particles. [0118] In embodiments of the pulmonary pharmaceutical composition, the pulmonary pharmaceutical carrier is a pulmonary pharmaceutical liquid or pulmonary pharmaceutical powder. In embodiments, the pulmonary pharmaceutical carrier is a pulmonary pharmaceutical liquid. In embodiments, the pulmonary pharmaceutical carrier is a pulmonary pharmaceutical powder.

[0119] In embodiments of the pulmonary pharmaceutical composition, the pulmonary pharmaceutical liquid includes a polar liquid and the thiol-containing compound is dissolved or suspended in the polar liquid. In embodiments, the polar liquid is water.

[0120] In embodiments of the pulmonary pharmaceutical composition, the pulmonary pharmaceutical carrier is lactose, mannitol, a phospholipid or cholesterol. In embodiments, the phospholipid is phosphatidyl choline.

[0121] In embodiments of the pulmonary pharmaceutical composition, the pulmonary pharmaceutical composition is within a pulmonary pharmaceutical delivery device. In embodiments, the pulmonary pharmaceutical delivery device is a pulmonary pharmaceutical nebulizer, a pulmonary pharmaceutical dry powder inhaler, or a pulmonary pharmaceutical pressurized metered dose inhaler.

[0122] In embodiments, the pharmaceutical composition further includes one or more additional therapeutic agents. In embodiments, the pharmaceutical composition further includes one additional therapeutic agent. In embodiments, the pharmaceutical composition further includes a plurality of additional therapeutic agents. In embodiments, the pharmaceutical composition further includes two additional therapeutic agents. In embodiments, the pharmaceutical composition further includes three additional therapeutic agents. In embodiments, the pharmaceutical composition further includes four additional therapeutic agents.

[0123] In embodiments, the additional therapeutic agent is a beta agonist, as known in the art. In embodiments, the additional therapeutic agent is a short-acting beta agonist, as known in the art. In embodiments, the additional therapeutic agent is a long-acting beta agonist, as known in the art. The term “short-acting” in the context of therapeutic agents refers, in the usual and customary sense, a therapeutic agent that elicits a transient effect, e.g., 1-60 seconds, 1-60 minutes, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or even 12 hours, as known in the art. The term “long- acting” in the context of therapeutic agents refers, in the usual and customary sense, a therapeutic agent that elicits a sustained effect, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or even 24 hours, 1, 2, 3, 4, 5, 6, or even 7 days, 1, 2, 3, 4 weeks or longer, as known in the art.

[0124] In embodiments, the additional therapeutic agent is an anticholinergic, as known in the art. In embodiments, the additional therapeutic agent is a short-acting anticholinergic, as known in the art. In embodiments, the additional therapeutic agent is a long-acting anticholinergic, as known in the art.

[0125] In embodiments, the additional therapeutic agent is a steroid as disclosed herein or as known in the art, e.g., fluticasone, budesonide, beclomethasone, momethasone, dexamethasone. In embodiments, the additional therapeutic agent is a corticosteroid as disclosed herein or as known in the art.

[0126] In embodiments, the additional therapeutic agent is an antibiotic, as known in the art.

[0127] In embodiments, the additional therapeutic agent is an anti-viral agent (e.g., remdesivir).

[0128] In embodiments, the additional therapeutic agent is rhDNAse, as known in the art.

EXAMPLES

[0129] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.

Example 1: Inhibiting SARS-CoV-2 entry by targeting the Spike protein

[0130] Betacoronaviruses are enveloped positive-sense, single-stranded RNA viruses that infect humans and generally cause illnesses that induce fever and respiratory symptoms. For example, the SARS-CoV-1 coronavirus strain causes Severe Acute Respiratory Syndrome (SARS), and the SARS-CoV-2 coronavirus strain causes Coronavirus Disease 2019 (COVID- 19), a respiratory illness with symptoms ranging from fever and cough to potentially fatal complications including acute respiratory distress and kidney failure.

[0131] The SARS-CoV-2 spike (S) protein (SARS-2-S) is an envelope glycoprotein trimer that binds angiotensin converting enzyme 2 (ACE2) as an entry receptor. Recently it has been shown that IFNa drives ACE2 expression in primary human nasal epithelial cells. In this manner, SARS-CoV-2 exploits interferon-driven upregulation of ACE2, normally a tissue- protective mediator during lung injury, to enhance infection. A strategy to preventing such infection is to block binding of SARS-CoV-2 to ACE2.

[0132] The SI domain of SARS-2-S includes a receptor binding domain (RBD) that mediates the initial high affinity interaction of SARS-CoV-2 with the ACE2 receptor. In the homologous SARS-CoV-1 Spike protein (SARS-l-S), the SI RBD was previously shown to block S protein- mediated infection more efficiently than the full-length S 1 domain. Further, abolishing disulfide bonds formed by cysteine residues in the SARS-l-S RBD was shown to disrupt viral binding to ACE2 (Wong, S. K. et al. A 193-Amino Acid Fragment of the SARS Coronavirus S Protein Efficiently Binds Angiotensin-converting Enzyme 2. J Biol. Chem. 2004, 279:3197). Thus, conserved cysteine residues in the SARS-2-S RBD may similarly be essential for SARS-2-S associating with ACE2. Without wishing to be bound by theory, cleavage of disulfide bonds in the SARS-2-S protein could alter the native conformation of the RBD and disable SARS-2-S binding to ACE2.

[0133] 6,6’-dithiotrehalose (also referred to herein as “MUC,” “MUC-31,” and “TS21”) and compound 12 in Table 1), is a dithiol substituted trehalose that potently cleaves disulfides and also has multiple other attractive drug attributes, including solubility, stability, and favorable aerosol characteristics. Another compound, N-acetyl-L-cysteine (NAC; compound 1 in Table 1), has previously been shown to reduce disulfide bonds in mucoproteins. Thus, MUC and NAC were screened for inhibiting SARS-2-S binding to ACE2 using a plate-based binding assay. Recombinantly expressed RBD of SARS-2-S was first immobilized to a surface. Immobilized protein was incubated in either MUC or NAC; drug was then washed off. Biotinylated ACE2 was added and allowed to bind to surface-immobilized SARS-2-s. Detection of ACE2 binding was achieved by addition of streptavidin-modified horseradish peroxidase (HRP), followed by a colorimetric substrate for the HRP enzyme. As illustrated in FIG. 1, both compounds MUC and NAC inhibited binding of ACE2 to SARS-2-S, though MUC showed stronger potency in inhibiting ACE2 binding compared to NAC. Notably, the inhibition of binding to ACE2 occurs even when MUC is washed out of the reaction mixture, demonstrating that binding inhibition persists following drug contact.

[0134] MUC and NAC were then tested for their ability to inhibit entry of a SARS-CoV-2 pseudo virus using a cell culture-based assay. The vesicular stomatitis virus (VSV) is promiscuous as to the membrane protein that may be incorporated into its envelope, and thus, VSV may readily form pseudotypes with heterologous membrane proteins. (Whitt, M. A. Generation of VSV Pseudotypes Using Recombinant AG-VSV for Studies on Virus Entry, Identification of Entry Inhibitors, and Immune Responses to Vaccines. J Virol Methods. 2010, 169(2):365). A VSV in which the glycoprotein (G) gene was deleted (VSV-AG) was used to produce the VSV pseudo type. The VSV glycoprotein gene was replaced with the gene encoding luciferase (VSV-AG-Luciferase), enabling detection. As illustrated in FIG. 2, VSV-AG- Luciferase budding from a host cell expressing SARS-2-S resulted in the acquisition of the envelope including the lipid bilayer derived from the host cell plasma membrane, generating a VSV pseudotype bearing the SARS-2-S protein (VSV-S).

[0135] Calu-3 cells were used to determine if MUC inhibits SARS-CoV-2 infection of lung epithelial cells in a cell culture. The cells were incubated with VSV-S for 30 minutes in the presence of either 1 mM MUC, 5 mM MUC, 25 mM MUC, 1 mM NAC, 5 mM NAC, 25 mM NAC, or no compound (control). As shown in the schematic of FIG. 3, VSV-S readily entered Calu-3 cells in the control group, while both MUC and NAC inhibited VSV-S cellular entry. Experiments were also conducted with a 2-hour incubation time. All three concentrations of MUC resulted in potent inhibition of VSV-S entry, while only 5 and 25 mM of NAC resulted in significant cell entry inhibition, as illustrated in FIG. 4. These results demonstrate MUC is more effective than NAC for inhibiting viral entry.

[0136] In additional cell culture experiments using the VSV-S pseudovirus, epithelial cells are pre-incubated with interferons to upregulate ACE2. Additional cell types to test include airway epithelial cells and type 2 alveolar cells.

Example 2: Treatment of COVID-19 with MUC using animal models

[0137] An animal model of COVID-19 is used to determine efficacy of aerosolized MUC in treating COVID-19. It has been shown that Syrian hamsters are susceptible to lung infection with SARS-CoV-2, and infected animals develop high lung viral load, marked cytokine activation and diffuse pneumonia within the first week of virus challenge. Challenged index hamsters consistently infect naive contact hamsters housed within the same cage, resulting in similar pathology.

[0138] Syrian hamsters are administered aerosolized MUC for up to 7 days following infection with SARS-CoV-2. Outcomes will include viral load (copy number/mL) and infectious titer (plaque/TCID50) for characterization of virus, and clinical signs (weight and respiratory rate) and lung histopathology for characterization of COVID-19 outcomes.

Example 3: Treatment of COVID-19 [0139] A subject having COVID-19 and presenting either mild, moderate, or severe disease conditions is treated with a compound of the present disclosure as shown in Table 2. Mild severity may be characterized by the subject’s confirmed COVID-19 diagnosis, and symptoms that do not require hospitalization. Moderate severity may be characterized by hospitalization of the subject, medical care required for the subject’s COVID-19 symptoms, or when the subject has an Sp02 of at least 94%. Severe conditions may be characterized by hospitalization of the subject, or when the subject either has an Sp02 of less than 94% or requires supplemental oxygen, either with or without mechanical ventilation.

[0140] Table 2. Use of different formulation types of thiol-containing compounds for treatment of COVID-19

References: 1. Tran et ah, PLOS One (2012), doi.org/10.1371/joumal.pone.0035797. 2. Wolfel et al, Nature 2020 DOI: 10.1038/s41586-020-2196-x: Reference demonstrates high viral load and replication in upper respiratory tract and throat.

Example 4: Antiviral and anti-inflammatory effects of thiol drugs in COVID-19

[0141] Coronavims disease 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavims 2 (SARS-CoV-2). The SARS-CoV-2 spike protein is an envelope glycoprotein that binds angiotensin converting enzyme 2 (ACE2) as an entry receptor. Treatments that prevent viral entry are needed because entry inhibitors prevent cell infection and interrupt active infection. We considered the possibility that thiol-based dmgs could cleave cystine bridges in the SARS-CoV-2 spike protein (SARS-2-S) and dismpt the native binding interface required for interaction with ACE2. We show that thiol dmgs decrease binding of SARS-CoV-S to ACE2 to decrease the entry efficiency of SARS-CoV-2 and decrease viral titers in the lung. We also show that thiol dmgs can limit SARS-CoV-2 lung inflammation without decreasing SARS-CoV-2 viral infection. Our findings reveal that thiol dmgs can improve COVID-19-related lung disease through either antiviral or anti-inflammatory mechanisms and provide strong rationale for further testing the efficacy of these dmgs in clinical trials.

[0142] The capacity of enveloped vimses such as SARS-CoV-2 to infect host cells often depends on a precise thiol/disulfide balance in their surface glycoprotein complexes. All identified SARS-CoV-2 variants have conserved cystines in their receptor binding domains (RBDs) of the spike protein, but it was unknown if these cystines were critical to maintaining a native binding interface to ACE2 and if dismption of these cystines by thiol-based dmgs is a viable therapeutic strategy against the vims. Using a receptor binding assay, and pseudovims and live vims cell infection assays, we showed that multiple thiol-based dmgs, including cysteamine (approved) and a novel thiol-saccharide dmg (“MUC-31”, in preclinical development), decreased binding of SARS-CoV-2 spike protein to its receptor to decrease the entry efficiency of SARS-CoV-2 spike pseudo typed virus and inhibit live virus infection. Moreover, in SARS-CoV-2-infected hamsters, aerosol delivery of MUC-31 directly to the lung, at a dose which achieves millimolar local concentrations decreased SARS-CoV-2 virus copy number and lung inflammation, whereas intraperitoneal delivery of cysteamine, at a dose which achieves micromolar concentrations or less in the lung did not decrease virus copy number but did decrease lung inflammation. Our findings show that thiol-based drugs have both antiviral and anti-inflammatory effects in CO VID- 19-related lung disease and that direct anti-viral effects can be achieved by aerosol delivery to the lung.

[0143] SARS-CoV-2 causes COVID-19, a multidimensional disease characterized predominantly by pneumonia that can progress to respiratory failure and death 1 2 . The envelope glycoproteins of SARS-CoV-2 include a spike glycoprotein trimer (SARS-2-S) that binds a cell surface protease (angiotensin converting enzyme 2 [ACE2]) as an entry receptor 3 . The viral envelopes of so-called type I enveloped viruses, including coronaviruses, retroviruses, and filoviruses, exhibit similar structural and mechanistic strategies for viral entry 4 . Among these viruses, the capacity of their envelope glycoproteins to mediate fusion of virus to host cell membranes often depends on a precise thiol/disulfide balance in the viral surface glycoprotein complex 5-8 . Natural and specific thiol/disulfide rearrangements in this complex can trigger conformational changes that promote virus entry 9-11 , but removal of disulfide bridges by chemical reduction or by replacement of cysteines by mutagenesis can also disrupt viral binding to prevent infection. For example, chemical reduction of the SI domain of SARS-CoV decreases its binding to ACE2 and inhibits transduction of Vero E6 cells by SARS-CoV pseudovirions, and site-directed mutagenesis replacing cystine forming cysteines with alanines in the SARS- COV spike (hereafter SARS-l-S) RBD prevents binding of ACE2 6 . In addition, molecular dynamics simulations reveal that the binding affinity of SARS-2-S RBD for ACE2 is significantly impaired when all of the disulfide bonds of both ACE2 and SARS-2-S are reduced to thiol groups 12 . Multiple drugs have at least one functional thiol group and can reduce cystines to cysteines, but it is not known if thiol drugs can disrupt cystines in SARS-2-S to inhibit binding to ACE2 or virus entry into cells, and if they can do so at doses that are deliverable safely in vivo.

[0144] To address this question we used published data 17,18 to build a cystine bridge map of the SI domain of SARS-2-S, and we compared the amino acid alignment of the receptor binding domains (RBDs) in SARS-2-S and SARS-l-S. We noted 10 cystine bridges in the SARS-2-S1 domain (FIG. 5 A) and 4 conserved cystines between SARS-l-S and SARS-2-S RBD (FIG. 5B). The conserved Cys467-Cys474 in SARS-l-S and Cys480-Cys488 in SARS-2-S constrain the ACE2 binding domains, and previous studies with SARS-l-S RBD have shown that mutagenesis of either homologous cysteine leads to loss of ACE-2 binding 6 . To further explore if Cys480-Cys488 in SARS-2-S might be vulnerable to chemical cleavage, we used protein modeling software to render the SARS-2-S RBD based on PDB entry 6M0J (FIG. 5C). This rendering shows that Cys480-Cys488 is very near the RBD surface (FIG. 5C) and could be accessible to cleavage by thiol-based drugs. Apart from Cys480-Cys488 cystine, cleavage of the other six cystines in the RBD could also allosterically modify the binding interface in ways that decrease binding to ACE2. Studies in SARS-CoV have shown that cysteine residues flanking the S2 domain - including Cys822 and Cys833 - mediate membrane fusion of SARS CoV (Madu et al., Virology, 2009 Oct 25;393(2):265-71). Amino acid alignment of the spike protein of SARS-CoV and SARS-CoV2 shows that these cysteine residues are conserved in the spike protein of SARS-CoV2 (Cys 840 and Cys 851), which raises the possibility that thiol-based drugs could inhibit membrane fusion in addition to inhibitory effects on receptor binding. Recognizing that multiple variants of SARS CoV2 show sequence variations, including in the RBD, we generated a consensus RBD sequence to highlight and annotate differences among the variants’ amino acid sequences (FIG. 5D). As shown in FIG. 5D, the RBD of the UK variant (Bl.1.7) has an N501Y mutation (RBD N501Y ), the RBD of the Brazilian variant (PI) has an E484K mutation (RBD E484K ), the RBD of the California variant has an E484K mutation (RBD l452r ), and the RBD of the South African variant (B1.351) has K417N, E484K, and N501Y mutations (RBD K417N E484K N501Y ). All of these variants have the four cystine bridges present in the original SARS-CoV2 RBD, and all have the Cys480-Cys488 that is conserved between SARS-CoV and SARS-CoV2 (FIG. 5D).

[0145] To test if thiol drugs can cleave cystines in the RBD of SARS-2-S to disrupt binding to ACE2, we exposed the RBD of the original SARS-CoV isolates (RBD original ) to eight existing thiol drugs and quantified ACE2 binding affinity in a plate-based binding assay. We also tested an investigational thiol saccharide drug (MUC-31) in this system, because of the potential advantages of thiol-saccharides as drugs for inhaled delivery 16 . Carbocysteine and amifostine were included as negative controls because carbocysteine is a sulfur containing drug lacking a free thiol warhead and amifostine is a phosphorothiolate prodrug whose dephosphorylated metabolite (WR-1065) is the active drug metabolite. The ACE2-SARS-2-S RBD binding assay was optimized by modifying a commercially available kit. RBD ongmal was covalently coupled to plates functionalized with primary amine -reactive maleic anhydride, and ACE2 binding was then quantified after exposure of RBD onginal to drugs for 60 minutes (FIG. 6A). We found that all of the thiol drugs inhibited binding of RBD origmal to ACE2 in a dose dependent manner. 2- Mercaptoethane sulfonate, sodium salt (MESNA), bucillamine, cysteamine, WR-1065, and MUC-31 had strong inhibitory effects (FIGS. 6B-6C). We also measured binding ofRBD origmal to ACE2 at one and two hours post exposure and wash to MESNA, bucillamine, cysteamine, MUC-31 and WR-1065 and found that binding inhibition was retained for two hours after drug removal (FIG. 6D). Next, we tested if cysteamine and MUC-31 inhibit binding of RBD N501Y to ACE2. RBD N501Y bound more strongly than RBD origmal to ACE2 (FIG. 6E) confirming recent reports l9 · 20 · 21 , and cysteamine and MUC-31 potently inhibited this binding of RBD N501Y (FIG. 6F). To determine if the variable effects of thiol drugs on RBD binding relates to their cystine cleaving potency, we leveraged the BODIPY FL L-cystine reagent which fluoresces when thiol- specific exchange leads to mixed disulfide formation (FIG. 6H). The potency of the thiol drugs in the BODIPY assay mirrored the potency order seen in the RBD-ACE2 binding assay (FIGS. 6I-6J) suggesting that cystine cleaving potency could explain the drug potency differences in the binding assay.

[0146] To test if thiol drugs can inhibit SARS-COV2 entry into cells, we first tested drug efficacy in pseudovirus and live virus assays. The pseudovirus particles carry SARS-2-S on the surface and enclose a viral genome of recombinant vesicular stomatitis virus (VSV) with a deleted glycoprotein (rVSV-AG) and an insertion of the firefly luciferase gene. In these experiments, we first exposed pseudovirus particles to thiol-based drugs and then quantified cell entry efficiency in human embryonic kidney cells (HEK293T) stably transfected to express huACE2 and transmembrane protease, serine 2 (TMPRSS2, a priming serine protease for SARS-CoV-2 3 ) (293T-ACE2-TMRPRSS2 cells). None of the drugs significantly affected cell viability, and pretreatment of SARS-2-S pseudovirus with carbocysteine and amifostine did not inhibit viral cell entry (FIG. 7 A). In contrast, pretreatment of SARS-2-S pseudovirus with all of the thiol-based drugs significantly decreased viral entry in a dose dependent manner (FIGS. 7B- 7D). The thiol drugs had only small and inconsistent effects on pseudovirus cell entry when the 293T-ACE2-TMPRSS2 cells were first pretreated with thiol drugs and then infected with untreated SARS-2-S pseudovirus. To confirm that these data with pseudovirus particles extend to live virus, we tested the effects of a subset of the drugs (cysteamine, MUC-31, and carbocysteine as negative control) on SARS-CoV-2 infection of Vero E6 cells (FIGS. 9A-9C). We found that cytopathic effects in virus-infected cells was significantly inhibited by all of these drugs, and that inhibition was minimal when the VERO E6 cells were first pretreated with thiol drugs and then infected with untreated SARS-CoV2. Taken together, these data demonstrate that thiol-based drugs inhibit SARS-COV2 entry into cells.

[0147] To determine if thiol-based drugs inhibit SARS-COV2 infection in vivo, we tested two drugs in the Syrian hamster model of COVID- 19 22 25 . Since millimolar drug concentrations of the thiol drugs were most effective in the pseudovirus and live virus assays, we hypothesized that pulmonary delivery of the drug by aerosol will most reliably result in these concentrations in the airways. MUC-31 was selected for in vivo testing by aerosol administration because it is formulated for aerosol delivery. The lung deposited dose of 0.5 mg/kg was calculated to result in millimolar concentrations in the airways and lungs. Cysteamine was selected for in vivo testing because it was the most potent thiol drug in the in vitro assays, but it is not formulated for aerosol delivery and so was given by intraperitoneal (IP) injection. A high dose of 100 mg/Kg of cysteamine free base was selected because cysteamine is used in high doses clinically (2g per day) 26,27 but even this high IP dose is not likely to achieve millimolar concentration in the airways and lungs. Our expectation therefore was that aerosolized MUC-31 would be more effective than IP cysteamine in these hamster experiments. The in vivo protocol is schematized in FIG. 8 A and shows that the first dose of both drugs was given 2 hours prior to intranasal viral inoculation of SARS CoV2 (1E+05 TCIDso/animal). Cysteamine-treated animals were dosed for 5 days and MUC-31 -treated animals were dosed for three days (the initial plan to dose MUC-31 for five days was modified when initial experiments indicated that SARS-CoV2 infected animals would have difficulty tolerating twice daily 20 minute tube confinements for 5 days). MUC-31 -treated animals had significantly lower SARS-CoV2 viral titers in their lungs, but the cysteamine-treated animals did not (FIG. 8A-8B). Despite this difference in viral titer results, both MUC-31 and cysteamine decreased the severity of SARS-CoV2 lung inflammation. Specifically, the lung weights in MUC-31- and cysteamine-treated hamsters were lower than in control groups with cysteamine ’s effects being larger, possibly because of the extra 2 days of treatment (FIG. 8C-8D). In addition, the total leukocyte counts in the bronchoalveolar lavage (BAL) fluid were lower than control in both MUC-31- and cysteamine-treated animals (FIG.

8E), effects driven by decreases in macrophages in MUC-31 -treated hamsters and in neutrophils and lymphocytes in cysteamine-treated hamsters (FIG. 8F). Furthermore, interleukin-6 levels in lung lavage were lower than control in both MUC-31 - and cysteamine-treated animals (FIG.

8F).

[0148] Our study shows that thiol drugs decrease binding of SARS-CoV-2 spike protein to its receptor to decrease the entry efficiency of SARS-CoV-2 and that an aerosolized thiol drug (MUC-31) decreases SARS-CoV-2 viral titers in the lung to limit SARS-COV-2-associated lung inflammation. The antiviral efficacy of MUC-31 in vivo can be attributed to aerosol administration which allows millimolar drug concentrations in the airways and lungs. MUC-31 is an investigational drug not yet approved for human use, but N-acetyl cysteine (NAC) and MESNA are approved thiol drugs that are administered by aerosol. Our data indicate that MESNA is more attractive than NAC as a candidate COVID-19 drug, because it has superior antiviral efficacy in vitro. Surprisingly, cysteamine - delivered IP - has anti-inflammatory effects in the lungs of SARS-CoV-2 infected hamsters, even though it does not decrease SARS-COV-2 viral titers in the lung. The anti-inflammatory effect of cysteamine is plausibly explained by the general anti-oxidant properties of thiol drugs to scavenge reactive oxygen species (ROS) and limit ROS-mediated inflammatory cascades. Some thiol drugs in their disulfide linked dimer form, including cysteamine, also inhibit transglutaminase. Thus, the beneficial effects of thiol drugs on CO VID- 19-related lung disease result from both antiviral and anti-inflammatory mechanisms, but the anti-viral effects were most pronounced at millimolar concentrations in the airways and lungs whereas the anti-inflammatory effect can be achieved at micromol r concentrations or less. Multiple thiol drugs can be administered by oral and intravenous routes, but amifostine (a prodrug whose dephosphorylated metabolite is WR-1065) and cysteamine have potency profiles that make them particularly attractive for repurposing as treatments for COVID-19. Finally, because the anti-viral effect of thiol drugs relates to cystines in the spike protein that are conserved among known SARS-CoV-2 variants, it is unlikely that these variants will escape the thiol drug effect. The anti-inflammatory effects of thiol drugs should operate in lung inflammation caused by all SARS-CoV-2 variants as well. Our work provides rationale for clinical studies of thiol drugs administered by inhaled or systemic routes to combat COVID-19.

[0149] Materials and Methods

[0150] Cells, plasmids and virus

[0151] HEK293T/clonel7 (CRL-11268) and Vero E6 (CRL-1586) cells were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin (Thermo Fischer Scientific). The cells were obtained from ATCC and incubated at 37 °C and 5% CO2. MEXi 293E cells (IBA Lifesciences) were cultured in MEXi culture medium (IBA Lifesciences) at 37 °C, 5% CO2 and 125 RPM as described by the manufacturer. The codon-optimized SARS-CoV-2 spike gene was subcloned from pCG SARS- CoV-2 Spike (provided courtesy of Stefan Polhmann 3 ) into the EBNA-1 dependent expression vector pTT5 for high-level expression in MEXi 293E cells. To boost cell surface expression of SARS-CoV-2 spike for efficient pseudotyping VSV, the C-terminal 21 amino acid containing the ER-retrieval signal (KxHxx) of spike was deleted. Plasmids for engineering lentiviral ACE2 and TMPRSS2 expression constructs: pLK05d.SFFV.dCas9-KRAB.P2A.BSD (a gift from Dirk Heckl, Addgene plasmid) and pDUAL CLDN (GFP) (a gift from Joe Grove, Addgene plasmid). SARS-CoV-2, isolate USA-WA1/2020 (NR-52281) was obtained from BEI resources and passaged in Vero E6 cells. Confluent Vero E6 cells grown in T175 flasks were infected with SARS-CoV-2 and the culture supernatant was collected when widespread cytopathic effect (CPE) was observed. After filtration through 0.45 pm filters, the virus containing culture supernatant was stored at -80 °C in small aliquots.

[0152] Thiol-based drugs and thiol content determination

[0153] N-acetylcysteine (NAC) and MESNA were the pharmaceutical formulations, with NAC manufactured by American Reagent INC at 200mg/ml and MESNA by Baxter at lOOmg/ml USP. Cysteamine (MilliporeSigma), amifostine (MilliporeSigma), WR-1065 (MilliporeSigma) and penicillamine (MP Biomedicals) were lyophilized powders that were solubilized as 500mM concentrated stocks in water. Cysteamine and WR-2065 were at pH 5. Amifostine was at pH 7 which was adjusted to pH 5. To ensure that amifostine does not auto- dephosphorylate to WR-1065, it was made fresh before the experiment each time. Bucillamine (MilliporeSigma) and tiopronin (Spectrum Chemicals) were lyophilized powders that were solubilized as 500mM concentrated stocks in equimolar NaOH to increase the solubility, and the pH was adjusted to pH 5. Carbocysteine (MilliporeSigma) and succimer (MilliporeSigma) were solubilized as 250 mM concentrated stocks in 500 mM NaOH to increase solubility with pH adjusted to pH 5. Free thiol content, and thus concentration of an active drug, was measured before every experiment using Ellman’s Reagent, 5,5'-ditliio-/vv-(2-nitrobcnzoic acid) (DTNB) (Abeam), with the molar extinction coefficient of 14,150 M 'cm ' at 412 nm 30 . Active drug concentration measured by DTNB was within 85 to 99% of nominal drug concentration. The stocks were stored at -20 °C and discarded if the thiol content went below 85%. Drug concentrations reported in plate-binding and viral entry assays are based on active drug concentration in stock.

[0154] Structure Rendering and Analysis

[0155] Space filling images and receptor distance calculations were performed using indicated PDB entries with UCSF Chimera, developed by the Resource for Biocomputing, Visualization, and Informatics at the University of California, San Francisco, with support from NIH P41- GM103311 31 .

[0156] RBD to ACE2 plate based binding assay

[0157] Wells of amine-reactive maleic anhydride-derivatized plates (Thermo Scientific) were washed with PBS + 0.05% Tween-20, and coated overnight with 1 pg/ml recombinant SARS- CoV-2 Receptor binding domain (aa 319-537; ACRO Biosystems). The following day, the plates were washed and blocked with 2% BSA for 1 hour at 37 °C. Wells were then incubated with drugs at concentrations ranging from 0 to 20mM diluted in PBS for an hour at 37 °C. Negative controls included wells with no RBD or no ACE2. After washing, biotinylated soluble recombinant ACE2 (ACRO Biosystems) was added at 0.06 mg/ml and incubated at 37 °C for 60 minutes. After washing, streptavidin-HRP at 0.1 pg/ml (ACRO Biosystems) was added to wells for an hour at 37 °C. The plates were washed and incubated with TMB (Sera Care). The reaction was stopped with 0.5 M hydrochloric acid stop solution and absorbance was read at 450 nm on a Biotek plate reader. Absorbance readings, after subtracting from negative control wells, were transformed to percent binding, with the wells containing no drug set as 100 percent binding.

[0158] To measure the stability of binding of cysteamine, WR-1065, Mesna and bucillamine, wells were incubated with either drugs at 5 mM for 1 hour, followed by three washes. ACE2 was then added to the wells either immediately, after 60 minutes, or after 120 minutes. Wells waiting for ACE2 were filled with dilution buffer. This was followed by the same steps to assess ACE2 binding as described above. For all binding assays, 4-6 independent experiments were carried out for all drugs, with 2 replicates in each.

[0159] To measure the difference in binding of the SARS CoV2 RBD variants, the plate was coated overnight with 1 ug/ml of the respective RBDs. Then the plate was blocked, followed by incubation with varying concentrations of biotinylated ACE2 ranging from 60 ng/ml to 0.7 ng/ml for 1 hour. Finally, the plates were incubated with streptavidin-HRP and developed using TMB. The fold change in the absorbance was assessed for each variant, relative to the original RBD.

[0160] BODIPY FL L-cystine cleaving assay

[0161] BODIPY FL L-cystine (Thermo Fischer scientific) was reconstituted to a final stock concentration of ImM with methanol. In a black Maxi Sorp 96-well flat-bottomed plate (Nunc), 10 uM of BODIPY reagent was added onto 25 uM of the thiol-based drugs and the change in fluorescence was kinetically measured, at 1 minute intervals, for an hour at 37 °C. Fluorescence reads, after subtracting the no drug control reads, were plotted against time. The maximum slope (Max V) for all the thiol-based drugs from this plot was measured and represented as relative fluorescence units/min (RFU/min) to assess the cystine cleaving ability of the drugs. The experiment was repeated three times.

[0162] Production of pseudoviruses

[0163] Pseudoviruses bearing SARS-2-S were generated using recombinant VSVAG- luciferase-based viruses, which lack glycoprotein (G) gene and instead code for reporter gene firefly luciferase. Briefly, MEXi cells were transfected with SARS-CoV-2 Spike expression plasmid (pTT5 SARS-CoV-2 8D21 ), using PEI as described by the manufacturer. Mock transfection served as the ‘no glycoprotein’ control. At 2-3 days post-transfection, the cells were inoculated with VSVG/VSVAG-luc at a multiplicity of infection (MOI) of 0.3. After 6 hours of incubation, the cells were washed twice with PBS by centrifugation and resuspended in culture medium containing 1% II anti-VSV-G hybridoma supernatant (ATCC CRL-2700). At 24 hours post-infection, the culture supernatant was collected by centrifugation and filtered through a 0.45 -pm syringe filter to clear off cellular debris. The supernatant containing viral particles was aliquoted and stored at -80 °C until further use.

[0164] Establishment of HEK293T cells stably expressing ACE2 and TMPRSS2 (293T- ACE2-TMPRSS2)

[0165] Engineering of lentiviral ACE2 and TMPRSS2 expression constructs

[0166] ACE2 and TMPRSS2 were cloned into separate lentiviral expression constructs. ACE2 was cloned into pLK05d.SFFV.dCas9-KRAB.P2A.BSD (a gift from Dirk Heckl, Addgene plasmid) by replacing dCAS9-KRAB with new unique enzyme restriction sites (Spel and Nhel) and subsequently inserting the ACE2 gene sequence into the expression construct downstream of the SFFV promoter based on restriction enzyme cloning. TMPRSS2 was cloned into pDUAL CLDN (GFP) (a gift from Joe Grove, Addgene plasmid). GFP was exchanged with a puromycin cassette using enzyme restriction sites Mlul and Xhol to enable antibiotic selection in cell culture. TMPRSS2 was inserted into the expression construct immediately downstream of the SFFV promoter following the addition of unique enzyme restriction sites (Srfl and Sail). All cloning steps were confirmed by Sanger sequencing.

[0167] Production of lentiviral particles

[0168] Lentiviral particles for delivery of lentiviral ACE2 and TMPRSS2 vectors were produced using a polyethylenimine (PEI; Polysciences, Inc) transfection protocol. Briefly, HEK293T cells were transfected with three plasmids: lentiviral ACE2 or TMPRSS2 constructs, psPAX2, and VSVg, at a ratio of 4:3:1 and a final DNA amount of 1.5 pg prepared in Opti- MEM (ThermoFisher). PEI was added at a ratio of 3:1 PEEDNA (4.5 pg PEI). The transfection mix was vortexed and incubated for 15 min at RT and added to the cells. 16 h post transfection, transfection medium was replaced with standard culture medium, and cells were cultured for another 24h. Cell supernatants containing the newly produced viral particles were then collected 48h post transfection. Supernatants were centrifuged at 4 °C and subsequently filtered using 0.22 pm vacuum filter units (MilliporeSigma). The supernatants were then aliquoted and stored at -80 °C.

[0169] Establishment of cells stably expressing ACE2 and TMPRSS2 (293T-ACE2- TMPRSS2) [0170] To establish HEK293T cells stably expressing ACE2 and TMPRSS2 (293T-ACE2- TMPRSS2 cells), 0.4 xlO 6 cells were seeded in 12-well plates. The following day, cells were transduced with lentiviral particles containing the ACE2 vector by adding 500 mΐ of lentiviral particles and 500 mΐ culture medium per well. 48h post transduction, medium was replaced with blastidicin (BSD; InvivoGen) selection medium at a final concentration of 10 mg/ml BSD. After 5 days of selection, cells were transferred to 75 cm 2 cell culture flasks for further expansion of cells stably expressing ACE2. The process was then repeated to further transduce cells with TMPRSS2 lentiviral particles and cells were cultured in antibiotic selection medium containing 10 pg/ml BSD and 1 pg/ml Puromycin 48 h post transduction. The expression of ACE2 and TMPRSS2 was confirmed by Western Blot and compared to nontransduced cells.

[0171] Pseudovirus transduction experiments

[0172] 293T-ACE2-TMPRSS2 cells were plated in black 96-well tissue culture treated plates (Greiner Bio-one) 18 hours before the experiment. Two experimental strategies of pseudovirus pre-treatment and cell pre -treatment were followed. For pseudo virus pre-treatment, the pseudo viruses were pre-incubated with different concentrations (1.56-100 mM) of the thiol- based drugs for 2 hours at 37 °C, followed by 66-fold dilution with standard culture media. The cells were then transduced with these pre -treated virions for 2 hours at 37 °C. After the incubation, the virions were removed and cells were cultured in standard culture medium. For cell re -treatment, the 293T-ACE2-TMPRSS2 cells were incubated with the different drug concentration (0.02-1.5 mM) for 2 hours at 37 °C, 5% CO2. These concentrations reflect the 66- fold dilution of drugs when virus/drug mix was incubated with the cells in the pseudo virus pre treatment experiment. After incubation, the media was removed and the cells were transduced with untreated pseudoviruses for 2 hours at 37 °C. After the incubation, the virions were removed and the cells were cultured in standard culture medium.

[0173] For both experimental conditions, at 18 hours post-transduction, the cells were lysed and luciferase activity was measured using Promega luciferase assay system and Biotek Synergy HI plate reader. Data was normalized to the viral particles without any viral envelope protein. For each experiment, luciferase reads of no drug control group was set as 100% and the relative transduction efficiencies in the presence of thiol-based drugs were calculated. Three-four independent experiments were carried out for each PV pretreatment and cell pretreatment strategies, with 12 replicates in each for all the drug doses.

[0174] SARS-CoV-2 quantification [0175] Titers of SARS-CoV-2 was measured by TCID50 using Vero E6 cells. Viruses were 10-fold serially diluted in DMEM with 1% FBS prior to addition to cell monolayer in 96-well- plate. For each dilution, viruses were added to 10 replicate wells at 100 pi per well. After two hours of infection, cells were washed and cultured with fresh DMEM medium containing 1% FBS at 37 °C with 5% CO2. Clear CPE was observed two days later. 50% endpoints were calculated with Reed and Muench method 32 .

[0176] Inhibition of SARS-CoV-2 infection

[0177] SARS-CoV-2 of 1.2 x 10 4 TCIDso/ml was incubated with 2-fold serially diluted thiol- based drugs at 37 °C for 2 hrs. Virus-drug mixtures were diluted 12-fold before addition to Vero E6 cell monolayer in 96-well-plate. For each drug concentration, virus-drug mixtures were added to 10 replicate wells at 100 mΐ per well. The final titer of virus added to cells was I x IQ 3 TCIDso/ml (100 TCIDsoper 100 ul per well in 96- well-plate). After two hours of infection, virus-drug inoculum was replaced with fresh DMEM medium containing 1% FBS. Clear CPE developed after two days of incubation at 37 °C with 5% CO2. The experiment was repeated thrice. Wells with clear CPE were counted positive and percentage of positive wells for each concentration of tested drugs were plotted. The effect of thiol-based drugs on Vero E6 cells during the two hours of SARS-CoV-2 infection was evaluated by addition of 8.33 mM or 0.52 mM of each drug and 100 TCID50 SARS-CoV-2 simultaneously to Vero E6 cell monolayer in 96-well-plate. After two hours of infection, cells were washed and then cultured with fresh DMEM medium containing 1% FBS at 37 °C with 5% CO2. Clear CPE developed two days post infection.

[0178] Quantification of cell viability

[0179] The cell viability was quantified using CellTiter-Glo2.0 assay (Promega) which measures cellular ATP content, indicating the metabolically active cells. For all cell viability experiments, the experimental protocol was the same as the main experiment except for the step of pseudovirus/live virus infection. For cell viability measurement corresponding to pseudovirus experiment, 293T-ACE-TMPRSS2 cells were seeded in 96 well black plates 18 hours prior to the experiment. The cells were then incubated with different concentrations (0.02-1.5 mM) of the thiol-based drugs for 2 hours at 37 °C, followed by removal of the drugs and incubation of cells with standard culture medium for 18 hours. The experiment was carried out thrice with 5-6 replicates for each drug. These concentrations reflect the 66-fold dilution of drugs when pseudo virus/drug mix was incubated with the cells in the pseudo virus pretreatment setting. For the cell viability measurement corresponding to the live virus experiment, Vero E6 cells were incubated with different concentrations of the drugs (0.03-8.33 mM) in 1% FBS for 2 days. These concentrations reflect the 12-fold dilution of drugs when virus/drug mix was incubated with the cells in the live virus infection setting. The cell viability experiment on Vero E6 cells was carried out thrice with 6 replicates for each drug. For both cell viability experiments, post the respective incubations, the plates and their contents were equilibrated at room temperature for 30 minutes before addition of equal volumes of CellTiter Glo2.0 reagent. Afterwards, the contents were mixed on a plate shaker to induce cell lysis. The plates were then incubated at room temperature for 10 minutes followed by measurement of luminescence using Biotek plate reader. Luciferase reads of control-treated cells was set as 100% and the relative viability of cells incubated in the presence of thiol-based drugs was calculated.

[0180] Syrian Hamster Model of COVID- 19

[0181] The efficacy of two thiol-based drugs, MUC-31 and cysteamine, was tested in a Syrian hamster model of SARS CoV2 infection. All the antiviral studies were performed in animal biosafety level 3 (ABSL3) facility at the Lovelace Respiratory Research Institute, Albuquerque, New Mexico. All work was conducted under protocols approved by the Institutional Animal Care and Use Committee (IACUC). A total of 40 Syrian hamsters ( Mesocricetus auratus), with a target age of 6-10 weeks old and a target weight of 130-160 g, were on the study. The animals were divided into 4 groups. Groups 1 and 2 included animals receiving aerosol delivery / to be exposed to nose-only inhalation of the vehicle (20 mM citrate, pH 4.5 ± 0.2, and 38.5 mM NaCl), and MUC-31 (0.48 mg/Kg lung deposition dose) respectively. Groups 3 and 4 included animals receiving intraperitoneal dosing of the vehicle (water) and cysteamine hydrochloride (147 mg/kg; MilliporeSigma). The animals were dosed twice daily starting on Day 0, as showing in FIG. 8A. The first dose was administered 2 hours prior to the viral inoculation. All animals were inoculated intranasally with SARS CoV2 (isolate USA-WA1/2020) at 1 xlO 5 TCID5o/animal. Animals in group 3 and 4 received 2X daily dosing on Days 0-4. Animals in groups 1 and 2 received 2X daily dosing on Days 0-2. Animals in all groups were sacrificed on Day 5. Efficacy of the drugs was determined by measuring viral load by RT-qPCR and lung inflammation, as measured by lung weight gain and total and differential cell counts in the bronchoalveolar lavage fluid.

[0182] Aerosol Exposure for MUC-031

[0183] Vehicle solution used as control in the aerosol study was 20 mM sodium citrate, pH 4.5 with 38.5 mM NaCl. MUC-031 was formulated at 11 mg/mL in vehicle. Two separate chambers were used for vehicle and MUC-031 exposures. Aerosols were generated using one Aerogen Solo vibrating mesh nebulizer with nominal forced air dilution of 8.5 ± 0.5 L/min. Animals were exposed for 20 minutes twice a day (to both active and vehicle control formulations). Prior to animal exposures, aerosol trials were conducted to establish the relationship between nebulization times and deposited dose. Aerosol concentration was measured at the breathing zone of the exposure system by collection of the aerosol onto 47-mm glass fiber filters (GE Whatman GF/A membrane filters). Filter samples were collected throughout all exposures at a nominal flowrate of 0.3 F/min. After drying and weighing the filters, total aerosol concentration (AC, mg/F) was measured gravimetrically: AC (mg/F) = A(Filter Weight) / (Exposure time (min) x Flowrate (F/min)). During trials, the deposited material from filters was then extracted into 0.1% TFA/H2O. MUC-031 in extract solution was quantified chromatographically, using a previously established HPLC method at Lovelace. Thus, a relationship could be established between total aerosol concentration and MUC-031 aerosol concentration. That relationship was used to monitor exposure inside BSL3 facility and to subsequently determine pulmonary deposited doses during in vivo exposures (described in Alexander et al). The average mass median aerodynamic diameter (MMAD) of the MUC-031 exposure atmosphere was 2.60 nm (measured with with Mercer-style cascade impactor) and is considered to be within respirable range for rodents (Kuehl et al). Aerosol exposures resulted in average pulmonary deposited dose of MUC-031 of 0.48 mg/kg per exposure, close to the target dose of 0.5 mg/kg.

[0184] Viral titers using RT-qPCR

[0185] Lung samples were homogenized in Trizol using a TissueLyser and centrifuged at 4000 x g for 5 minutes. From the supernatants, RNA was isolated using the QIAGEN RNeasy Kit, according to the manufacturer’s instructions. SARS-CoV-2 viral RNA was quantified by a qPCR assay targeting the SARS CoV-2 nucleocapsid phosphoprotein gene (N gene). Genome copies per g equivalents were calculated from a standard curve generated from RNA standards of known copy concentration. All samples were run in triplicate. The SARS CoV-2 N gene primers and probe sequences are as follows:

[0186] SARS CoV-2 Forward: 5’ TT AC AAAC ATT GGCCGC AAA 3’ (SEQ ID NOG)

[0187] SARS CoV-2 Reverse: 5’ GCGCGACATTCCGAAGAA 3’ (SEQ ID NO:4)

[0188] SARS CoV-2 Probe: 6FAM-AC AATTTGCCCCCAGCGCTT C AG-BHQ- 1 (SEQ ID NOG)

[0189] Amplification and detection was performed using a suitable real-time thermal cycler under the following cycling conditions: 50 °C for 5 minutes, 95 °C for 20 seconds and 40 cycles of 95 °C for 3 seconds, and 60 °C for 30 seconds.

[0190] Bronchoalveolar Lavage (BAL) collection and processing [0191] BAL was performed after the collection of whole lung weight. The left lobe was clamped off and the right lung lobes were lavaged with sterile saline. Half of the BAL collected was UV irradiated for sterilization out of the ABSL3 and used for differential analysis. This aliquot was centrifuged at 1000 g, 2-8 °C, >10 minutes. The supernatant will be collected and frozen at -80 °C for subsequent measurement of inflammatory mediators using ELISA. The cell pellet was resuspended in the appropriate amount of resuspension buffer, and red blood cell lysis buffer was used on samples as necessary. The total cell count was counted using a Nexcelom automated cell counter. A total of 50,000 cells per slide were used to prepare microscope slides by cytocentrifugation. The cells on slides were fixed and stained using Modified Wright’s or Wright-Giemsa Stain. Differential counts on at least 200 nucleated cells per slide were conducted using morphological criteria to classify cells into neutrophils, macrophages, lymphocytes and eosinophils.

[0192] The remaining lavage return volume from each animal was centrifuged at 1000 g, 2-8 °C, >10 minutes, the pellet was treated with 1000 uL of TRI-reagent and stored at -80 °C for RNA isolation and RT-PCR analysis.

[0193] Statistical analysis

[0194] For analyzing the statistical significance of difference in loss of binding for each drug area under the curve (AUC) was plotted and ordinary one -way ANOVA followed by Dunnetf s post hoc analysis was performed. To assess the difference in binding of RBD N501Y and RBD or gmai ? fold change of absorbance was plotted and analyzed using two tailed, unpaired t-test. Data are presented as mean ± SEM [* p < 0.05, ** p < 0.01, *** p < 0.005, **** p < 0.0001] IC50 of the drugs in pseudovirus transduction and live virus experiments was determined using the non-linear regression fitting with a variable slope. Data for pseudovirus and live virus experiments are plotted as mean ± SD. All statistical analyses were performed using GraphPad Prism software (version 8.4.2).

[0195] Table 3. List of currently approved thiol-based drugs or drugs that generate a thiol- containing metabolite*

REFERENCES FOR EXAMPLE 4

[0196] 1. Wiersinga, W. J., Rhodes, A., Cheng, A. C., Peacock, S. J. & Prescott, H. C. JAMA -

Journal of the American Medical Association (2020) doi: 10.1001/jama.2020.12839.

[0197] 2. Zhu, N. et al. N. Engl. J. Med. 382, 727-733 (2020).

[0198] 3. Hoffmann, M. et al. Cell 181, 271-280. e8 (2020).

[0199] 4. Weissenhorn, W. et al. Mol. Membr. Biol. 16, 3-9 (1999).

[0200] 5. Gallagher, T. M. J. Virol. 70, 4683-4690 (1996).

[0201] 6. Wong, S. K., Li, W., Moore, M. J., Choe, H. & Farzan, M. J. Biol. Chem. 279,

3197-3201 (2004).

[0202] 7. Lavillette, D. et al. J. Biol. Chem. 281, 9200-9204 (2006).

[0203] 8. Ryser, H. J. P., Levy, E. M., Mandel, R. & DiSciullo, G. J. Proc. Natl. Acad. Sci. U.

S. A. 91, 4559-4563 (1994).

[0204] 9. Wallin, M., Ekstrom, M. & Garoff, H. EMBO J. 23, 54-65 (2004).

[0205] 10. Abell, B. A. & Brown, D. T. J. Virol. 67, (1993).

[0206] 11. Gallina, A. et al. J. Biol. Chem. 277, 50579-50588 (2002). [0207] 12. Hati, S. & Bhattacharyya, S. ACS Omega 5, 16292-16298 (2020).

[0208] 13. Horowitz, R. L, Freeman, P. R. & Bruzzese, J. Respir. Med. Case Reports 30,

(2020).

[0209] 14. Akerlund, B. et al. Eur. J. Clin. Pharmacol. 50, 457-61 (1996).

[0210] 15. Lobo-Galo, N., Terrazas-Lopez, M., Martinez-Martinez, A. & Diaz-Sanchez, A. G.

J. Biomol. Struct. Dyn. (2020) doi:10.1080/07391102.2020.1764393.

[0211] 16. Yuan, S. et al. Sci. Transl. Med. 7, 276ra27-276ra27 (2015).

[0212] 17. Lan, J. et al. Nature 581, 215-220 (2020).

[0213] 18. Wang, Q. et al. Cell 181, 894-904.e9 (2020).

[0214] 19. Luan, B., Wang, H. & Huynh, T. FEBS Lett. 1873-3468.14076 (2021) doi: 10.1002/1873-3468.14076.

[0215] 20. Tian, F. et al. bioRxiv 2021.02.14.431117 (2021) doklO.l 101/2021.02.14.431117.

[0216] 21. Ali, F., Kasry, A. & Amin, M. Med. Drug Discov. 10, 100086 (2021).

[0217] 22. Sia, S. F. et al. Nature (2020) doi:10.1038/s41586-020-2342-5.

[0218] 23. Roberts, A. et al. J. Virol. 79, 503-511 (2005).

[0219] 24. Chan, J. F. W. et al. Clin. Infect. Dis. (2020) doi:10.1093/cid/ciaa325.

[0220] 25. Imai, M. et al. Proc. Natl. Acad. Sci. U. S. A. 117, 16587-16595 (2020).

[0221] 26. Mylan Pharmaceuticals Inc. Cystagon (cysteamine bitartrate). U.S. Food and Drug

Administration website www. accessdata.fda.gov/drugsatfda_docs/label/2007/020392s010lbl. pdf Revised June 2007.

[0222] 27. Bouazza, N. et al. Orphanet J. Rare Dis. 6, 86 (2011).

[0223] 28. Houk, J., Singh, R. & Whitesides, G. M. Methods Enzymol. 143, 129-140 (1987).

[0224] 29. Nagy, P. Antioxidants and Redox Signaling ol. 18 1623-1641 (2013).

[0225] 30. Riddles, P. W., Blakeley, R. L. & Zemer, B. Anal. Biochem. 94, 75-81 (1979).

[0226] 31. Pettersen, E. F. et al. J. Comput. Chem. 25, 1605-1612 (2004).

[0227] 32. Reed, L. J. & Muench, H. Am. J. Epidemiol. 27, 493^497 (1938).

[0228] 33. Huang, S. & Pang, L. Assay Drug Dev. Technol. 10, 88-96 (2012).

[0229] From the disclosure it will be appreciated that, although specific embodiments of the invention have been described herein for the purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and compositions within the scope of these claims and their equivalents be covered thereby.

P EMBODIMENTS

[0230] P Embodiment 1. A method of treating a coronavirus infection in a subject in need thereof, the method comprising administering to said subject an effective amount of a thiol- containing compound in a pharmaceutically acceptable carrier.

[0231] P Embodiment 2. The method of P Embodiment 1, wherein the coronavirus infection is a SARS-CoV-2 virus infection.

[0232] P Embodiment 3. The method of P Embodiment 1 or 2, wherein the subject has or is suspected of having COVID-19.

[0233] P Embodiment 4. The method of any one of P Embodiments 1-3, wherein the effective amount is administered within 48-96 hours of the onset of one or more symptoms of the infection.

[0234] P Embodiment 5. The method of P Embodiment 4, wherein the effective amount is administered within 72 hours of the onset of the one or more symptoms.

[0235] P Embodiment 6. The method of P Embodiment 4, wherein the effective amount is administered within 48 hours of the onset of the one or more symptoms.

[0236] P Embodiment 7. The method of any one of P Embodiments 1-6, wherein the effective amount is effective to reduce viral load in the subject.

[0237] P Embodiment 8. The method of any one of P Embodiments 1-7, wherein the thiol- containing compound is administered by pulmonary delivery.

[0238] P Embodiment 9. The method of P Embodiment 8, wherein the thiol-containing compound is administered as a liquid aerosol or a dry powder.

[0239] P Embodiment 10. The method of any one of P Embodiments 1-7, wherein the thiol- containing compound is administered orally.

[0240] P Embodiment 11. The method of P Embodiment 10, wherein the thiol-containing compound is administered as a lozenge.

[0241] P Embodiment 12. The method of any one of P Embodiments 1-7, wherein the thiol- containing compound is administered as a nasal spray.

[0242] P Embodiment 13. The method of any one of P Embodiments 1-7, wherein the thiol- containing compound is administered intravenously.

[0243] P Embodiment 14. The method of any one of P Embodiments 1-12, wherein the subject is not hospitalized. [0244] P Embodiment 15. The method of any one of P Embodiments 1-10 or 13, wherein the subject is hospitalized.

[0245] P Embodiment 16. The method of P Embodiment 15, wherein the subject is in an intensive care unit.

[0246] P Embodiment 17. The method of any one of P Embodiments 1-16, wherein the thiol- containing compound is a monothiol.

[0247] P Embodiment 18. The method of any one of P Embodiments 1-16, wherein the thiol- containing compound comprises two or more thiols.

[0248] P Embodiment 19. The method of any one of P Embodiments 1-16, wherein the thiol- containing compound is a dithiol.

[0249] P Embodiment 20. The method of any one of P Embodiments 1-19, wherein the thiol- containing compound has a molecular weight of at least 164 g/mol.

[0250] P Embodiment 21. The method of any one of P Embodiments 1-16, wherein the thiol- containing compound is thiomandelic acid, DL-Captopril, DL-Thiophan, N-acetylcysteine, Meso-2,3,-dimercaptosuccinic acid, 2,3-dimercaprol, D-(-)-Penicillamine, Glutathione, L- Cysteine, Zofenoprilat, Tiopronin, N-acystelyn, carbocysteine, cysteamine, sodium-2- mercaptoethane sulfonate, WR-1065, an analogue or derivative thereof, or a combination thereof.

[0251] P Embodiment 22. The method of any one of P Embodiments 1-16, wherein administering the thiol-containing compound comprises administering a prodrug thereof, wherein the prodrug is metabolized by the subject to form the thiol-containing compound.

[0252] P Embodiment 23. The method of P Embodiment 22, wherein the prodrug is amifostine.

[0253] P Embodiment 24. The method of any one of P Embodiments 1-16, wherein the thiol- containing compound is not N-acetylcysteine, glutathione, cysteamine, or dithiothreitol.

[0254] P Embodiment 25. The method of any one of P Embodiments 1-24, wherein the thiol- containing compound is coadministered with one or more additional antiviral compound, and/or one or more anti-inflammatory compounds.

[0255] P Embodiment 26. The method of any one of P Embodiments 1-24, wherein the thiol- containing compound is not coadministered with heparin, an antibiotic, or methylene blue. [0256] P Embodiment 27. A composition comprising a thiol-containing compound for use in a method of any one of P Embodiments 1-26.

ADDITIONAL EMBODIMENTS

[0257] Embodiment 1. A method of treating a coronavirus infection in a subject in need thereof, the method comprising administering to said subject an effective amount of a thiol- containing compound in a pharmaceutically acceptable carrier.

[0258] Embodiment 2. The method of Embodiment 1, wherein the coronavirus infection is a SARS-CoV-2 virus infection.

[0259] Embodiment 3. The method of Embodiment 1 or 2, wherein the subject has or is suspected of having COVID-19.

[0260] Embodiment 4. The method of any one of Embodiments 1-3, wherein the effective amount is administered within 48-96 hours of the onset of one or more symptoms of the infection.

[0261] Embodiment 5. The method of Embodiment 4, wherein the effective amount is administered within 72 hours of the onset of the one or more symptoms.

[0262] Embodiment 6. The method of Embodiment 4, wherein the effective amount is administered within 48 hours of the onset of the one or more symptoms.

[0263] Embodiment 7. The method of any one of Embodiments 1-6, wherein the effective amount is effective to reduce viral load in the subject, optionally wherein the viral load is reduced in the lung.

[0264] Embodiment 8. The method of any one of Embodiments 1-7, wherein the thiol- containing compound is administered by pulmonary delivery.

[0265] Embodiment 9. The method of Embodiment 8, wherein the thiol-containing compound is administered as a liquid aerosol or a dry powder.

[0266] Embodiment 10. The method of any one of Embodiments 1-7, wherein the thiol- containing compound is administered orally.

[0267] Embodiment 11. The method of Embodiment 10, wherein the thiol-containing compound is administered as a lozenge.

[0268] Embodiment 12. The method of any one of Embodiments 1-7, wherein the thiol- containing compound is administered as a nasal spray. [0269] Embodiment 13. The method of any one of Embodiments 1-7, wherein the thiol- containing compound is administered intravenously.

[0270] Embodiment 14. The method of any one of Embodiments 1-12, wherein the subject is not hospitalized.

[0271] Embodiment 15. The method of any one of Embodiments 1-10 or 13, wherein the subject is hospitalized.

[0272] Embodiment 16. The method of Embodiment 15, wherein the subject is in an intensive care unit.

[0273] Embodiment 17. The method of any one of Embodiments 1-16, wherein the thiol- containing compound is a monothiol.

[0274] Embodiment 18. The method of any one of Embodiments 1-16, wherein the thiol- containing compound comprises two or more thiols.

[0275] Embodiment 19. The method of any one of Embodiments 1-16, wherein the thiol- containing compound is a dithiol.

[0276] Embodiment 20. The method of any one of Embodiments 1-19, wherein the thiol- containing compound has a molecular weight of at least 164 g/mol.

[0277] Embodiment 21. The method of any one of Embodiments 1-16, wherein the thiol- containing compound is thiomandelic acid, DL-Captopril, DL-Thiophan, N-acetylcysteine, Meso-2,3,-dimercaptosuccinic acid, 2,3-dimercaprol, D-(-)-Penicillamine, Glutathione, L- Cysteine, Zofenoprilat, Tiopronin, N-acystelyn, carbocysteine, cysteamine, sodium-2- mercaptoethane sulfonate, WR-1065, an analogue or derivative thereof, or a combination thereof.

[0278] Embodiment 22. The method of any one of Embodiments 1-16, wherein the thiol- containing compound is Bucillamine.

[0279] Embodiment 23. The method of any one of Embodiments 1-16, wherein administering the thiol-containing compound comprises administering a prodrug thereof, wherein the prodrug is metabolized by the subject to form the thiol-containing compound.

[0280] Embodiment 24. The method of Embodiment 23, wherein the prodrug is amifostine.

[0281] Embodiment 25. The method of any one of Embodiments 1-16, wherein the thiol- containing compound is not N-acetylcysteine, glutathione, cysteamine, bucillamine, or dithiothreitol. [0282] Embodiment 26. The method of any one of Embodiments 1-25, wherein the thiol- containing compound is coadministered with one or more additional antiviral compound, and/or one or more anti-inflammatory compounds.

[0283] Embodiment 27. The method of any one of Embodiments 1-25, wherein the thiol- containing compound is not coadministered with heparin, an antibiotic, or methylene blue.

[0284] Embodiments 28. The method of any one of Embodiments 1-25, further comprising reducing inflammation, wherein the inflammation is in the lung.

[0285] Embodiment 29. A composition comprising a thiol-containing compound for use in a method of any one of Embodiments 1-28.