WO2021263139A1 - Methods of treating coronavirus infection using hsp90 inhibitors - Google Patents

Methods of treating coronavirus infection using hsp90 inhibitors Download PDF

Info

Publication number
WO2021263139A1
WO2021263139A1 PCT/US2021/039146 US2021039146W WO2021263139A1 WO 2021263139 A1 WO2021263139 A1 WO 2021263139A1 US 2021039146 W US2021039146 W US 2021039146W WO 2021263139 A1 WO2021263139 A1 WO 2021263139A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
alkyl
cyclo
aryl
Prior art date
Application number
PCT/US2021/039146
Other languages
French (fr)
Inventor
Sallie PERMAR
Maria BLASI
Ria GOSWAMI
Timothy Haystead
Original Assignee
Duke University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Duke University filed Critical Duke University
Publication of WO2021263139A1 publication Critical patent/WO2021263139A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses

Abstract

This present disclosure is directed to methods of use related to heat shock protein 90 inhibitors (HSP90i) to treat, prevent and/or reduce the incidence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, or treating or inhibiting coronavirus disease (COVID-19) in a subject.

Description

METHODS OF TREATING CORONAVIRUS INFECTION USING HSP90 INHIBITORS [0001] All patents, patent applications and publications cited herein are hereby incorporated by reference in their entirety. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art as known to those skilled therein as of the date of the disclosure described and claimed herein. [0002] This patent disclosure contains material that is subject to copyright protection. The copyright owner has no objection to the facsimile reproduction by anyone of the patent document or the patent disclosure as it appears in the U.S. Patent and Trademark Office patent file or records, but otherwise reserves any and all copyright rights. FIELD OF THE DISCLOSURE [0003] This disclosure is directed to methods of use related to heat shock protein 90 inhibitors (HSP90i) to treat and/or prevent a coronavirus infection in a subject (e.g., a human patient), for example a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in the subject. BACKGROUND OF THE DISCLOSURE [0004] In the last 20 years, there have been 3 major Coronavirus (CoV) outbreaks in the world, the most recent being the ongoing SARS-CoV-2 pandemic, which has already infected >5,000,000 people and claimed >350,000 lives around the globe. Hence, new treatment options for patients suffering from CoV-related infections are needed. SUMMARY OF THE DISCLOSURE [0005] An aspect of this disclosure is drawn towards a method for treating a coronavirus infection in a subject. In one embodiment, the method comprises administering to a subject in need thereof a therapeutically effective amount of an HSP90 inhibitor (also abbreviated herein as “HSP90i”), wherein the inhibitor comprises a compound of Formula (I) as described herein:
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof. [0006] An aspect of the disclosure is drawn towards a method for preventing a coronavirus infection in a subject. In an embodiment, the method comprises administering to a subject in need thereof a therapeutically effective amount of an HSP90 inhibitor, wherein the inhibitor comprises a compound of Formula (I) as described herein:
Figure imgf000004_0002
or a pharmaceutically acceptable salt thereof. [0007] An aspect of the disclosure is drawn towards a method of treating and/or reducing the effects (e.g., symptoms) of a coronavirus infection in a subject in need of such treatment. In an embodiment, the method comprises administering a therapeutically effective amount of an HSP90 inhibitor, wherein the inhibitor comprises a compound of Formula (I) as described herein:
Figure imgf000004_0003
or a pharmaceutically acceptable salt thereof. [0008] An aspect of the disclosure is drawn towards a method of treating a subject having a respiratory tract infection caused by a coronavirus. In an embodiment, the method comprises administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the inhibitor comprises a compound of Formula (I) as described herein:
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof. [0009] In an embodiment, the coronavirus infection is caused by a human coronavirus or a zoonotic coronavirus. In an embodiment, the coronavirus infection comprises SARS-CoV, SARS-CoV-2, MERS-CoV, or a virus from the Coronaviridae family, and/or Orthocoronavirinea subfamily. In a futher embodiment, the coronavirus infection comprises SARS-CoV-2. [0010] In an embodiment, the HSP90 inhibitor compound of Formula (I) comprises, consists essentially of, or consists of SNX5422 represented by the structure below:
Figure imgf000005_0002
or a pharmaceutically acceptable salt thereof. [0011] In an embodiment, the disclosure futher comprises administering to the subject suffering a CoV infection a therapeutically effective amount of at least one additional active agent. In an embodiment, the at least one additional active agent comprises an antiviral agent, an anti-inflammatory agent, a pain reliever, a steroid, or a combination thereof. [0012] In an embodiment, the therapeutically effective amount of an HSP90 inhibitor comprises a dosage that is administered to a subject from about 0.5 mg to about 7 g, either in a single dose or multiple consecutive doses. In another embodiment, the HSP90 inhibitor is administered orally, parenterally, topically, or intranasally. In various instances, the HSP90 inhibitor is administered orally. [0013] In an embodiment, the HSP90 inhibitor is provided as a pharmaceutical composition comprising a pharmaceutically acceptable carrier, excipient or diluent. [0014] An aspect of the disclosure is drawn towards a treatment regimen for treating a coronavirus infection in a subject. In an embodiment the treatment regimen comprises administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the inhibitor comprises a compound of Formula (I):
Figure imgf000006_0001
or a pharmaceutically acceptable salt thereof, and wherein administration commences within three days of onset of symptoms, within two days of onset of symptoms, or within 24-36 hours of onset of symptoms. In an embodiment, the HSP90 inhibitor compound of Formula (I) comprises, consists essentially of, or consists of SNX5422 represented by the structure below:
Figure imgf000006_0002
or a pharmaceutically acceptable salt thereof. [0015] In various aspects, provided herein is a method for treating a coronavirus infection in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000007_0001
or a pharmaceutically acceptable salt thereof, wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2, 3, or 4. [0016] In various aspects, provided herein is a method for preventing or reducing the incidence of a coronavirus infection in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000008_0001
or a pharmaceutically acceptable salt thereof, wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2, 3, or 4. [0017] In various aspects, provided herein is a method of reducing the effects and/or symptoms of a coronavirus infection in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000009_0001
or a pharmaceutically acceptable salt thereof, wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2, 3, or 4. [0018] In various aspects, provided herein is a method of treating a subject having a respiratory tract infection caused by a coronavirus, the method comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000010_0001
or a pharmaceutically acceptable salt thereof, wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2, 3, or 4. acceptable salt thereof. [0019] In various aspects, provided herein is a treatment regimen for treating a coronavirus infection in a subject, the treatment regimen comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000011_0001
or a pharmaceutically acceptable salt thereof, wherein administration commences within three days of onset of symptoms, within two days of onset of symptoms, or within 24-36 of onset of symptoms, and wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2, 3, or 4. [0020] In some instances, R1, R4, and R5 are each H. In some instances, R2 is NR7R8. In some instances, R3 is substituted or unsubstituted C1-6 carbonyl. In some instances, X is N. In some instances, Y is CR9. In some instances, n is 1. In some instances, R7 is H, and R8 is substituted or unsubstituted cyclo(C3-8) heteroalkyl. In some instances, the HSP90 inhibitor is:
Figure imgf000012_0002
, or a pharmaceutically acceptable salt thereof. [0021] In other embodiments, the HSP90 inhibitor is:
Figure imgf000012_0001
, or a pharmaceutically acceptable salt thereof. [0022] In some aspects, the coronavirus is a human coronavirus or a zoonotic coronavirus. In some aspects, the coronavirus comprises SARS-CoV, SARS-CoV-2, MERS-CoV, or a virus from the Coronaviridae family, Orthocoronavirinea subfamily. In some aspects, the coronavirus comprises SARS-CoV-2. In some aspects, any of these methods further comprises administering a therapeutically effective amount of at least one additional active agent. In some aspects, the at least one additional active agent comprises an anti-viral agent, an anti-inflammatory agent, a pain reliever, a steroid, or a combination thereof. In some aspects, the therapeutically effective amount of the HSP90 inhibitor comprises a dosage amount of between about 0.5 mg to about 7g. In some aspects, the HSP90 inhibitor is administered orally or intranasally. In such cases, the HSP90 inhibitor can be administered orally. In some aspects, the HSP90 inhibitor is administered as a pharmaceutical composition comprising a pharmaceutically acceptable carrier, excipient or diluent. [0023] Other objects and advantages of this disclosure can become readily apparent from the ensuing description. BRIEF DESCRIPTION OF THE FIGURES [0024] The novel features of the disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present disclosure will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the disclosure are utilized, and the accompanying drawings of which: [0025] FIG.1 shows graphs of viral loads. FIG.1A shows that HSP90i SNX5422 attenuates SARS-CoV-2 replication in vitro in a dose-dependent manner with a cell-free SARS-CoV-2 viral load. The dotted line represents median of DMSO treated infected cells. Each dot represents a single PCR technical replicate. FIG.1B is a graph showing cell-free SARS- CoV-2 viral load. Vero-E6 cells were infected with SARS-CoV-2-USA-WA-1 strain at a multiplicity of infection (MOI) of 0.01 for 1 hr. The infected cells were treated with SNX5422 at 1 hr post infection at indicated concentrations and incubated for 48 hrs. Viral RNA (vRNA) was extracted from cell supernatant and viral RNA copies were monitored using RT-qPCR. Each symbol represents a single experimental replicate. FIG.1C shows that Vero cells were immuno-stained for SARS-CoV-2 nucleocapsid protein (NP) (green). Nuclei were counterstained (blue) prior to imaging. FIG.1D shows immuno-fluorescent images that were quantified to evaluate proportions of SARS- CoV-2-NP+ cells. The data incidate that SNX5422 attenuated intracellular NP expression and proportion of NP+ cells. FIG.1E shows cell-free viral RNA that was detected and measured in the supernatant of infected and treated Vero-E6 cells using qRT-PCR of the viral N gene. FIG.1F shows cell-free viral RNA that was detected and measured in the supernatant of infected and treated Calu-3 cells using qRT- PCR of the viral N gene. FIG.1G shows the infectious viral titers of the supernatant of Vero E6 cells determined by plaque assay. FIG.1H shows the infectious viral titers of the supernatant of Calu-3 cells determined by plaque assay. FIG.1I shows percent Vero E6 cell viability following treatment of the cells with either 0.1% DMSO (drug-vehicle) or 0.01-100 µM SNX5422, or 5 µM remdesivir (RDV). FIG.1J shows percent Calu-3 cell viability following treatment of the cells with either 0.1% DMSO (drug-vehicle) or 0.01-100 µM SNX5422, or 5 µM RDV. [0026] FIG.2 shows a graph of the cytotoxicity of HSP90i SNX5422 in Vero-E6 cells as a function of inhibitor concentration. Each dot represents a single experimental replicate. The inhibitor cytotoxic concentrations at 50% (CC50 = 67.17 μM) and 20% (CC20 = 13.97 μM) were calculated for SNX5422. [0027] FIG.3A shows a schematic diagram illustrating treatment of human tracheobronchial epithelial (TBE) cells. TBE cells from 3 independent healthy donors were cultured in air- liquid interface and treated with either 0.1% DMSO (drug-vehicle) or 1 µM SNX-5422 reconstituted in DMSO, for 48 hrs. RNA was then extracted from the cells and bulk RNA sequencing was performed. FIG.3B shows a Volcano plot demonstrating protein coding genes altered upon SNX-5422 treatment of human TBE cells. Red (left, indicated by arrow #1), blue (right, indicated by arrow #2), and grey (middle) regions of the plot indicate downregulated, upregulated and non-differentially expressed genes, respectively. FIG.3C shows downregulated and FIG.3D upregulated Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways with an adjusted p value of < 0.05. Dot size represented gene ratio and color schema represents adjusted p values. [0028] FIG.4A shows that SNX-5422 dampened expression of cellular genes associated with SARS CoV-2 -mediated proinflammatory response and hyper-cytokinemia. FIG.4B shows data indicating that of the 55 genes altered by SNX-5422 treatment in the context of SARS-CoV-2 replication, 27 genes had an imbalanced cellular expression, in virally infected cells. [0029] FIG.5 shows an LC-MS/MS chromatogram of the concentration (in ng/mL) of HSP90i SNX5422 in adult and infant rhesus macaques’ plasma as a function of time following administration. [0030] FIG.6 shows a graph of SARS-CoV-2 infection in control group (Group 1) and a treatment group (Group 2) of rhesus macques. DETAILED DESCRIPTION OF THE DISCLOSURE [0031] There remains a critical gap in the development of effective therapeutics for SARS- CoV-2 (and related diseases) that are orally bioavailable and can reduce overall virus replication, and thereby transmission, as well as improve outcomes for infected individuals early on in their disease course. As many infected individuals remain asymptomatic after infection, a treatment modality targeting host factors that facilitate CoV replication can be an approach that is disclosed herein. Such approach can allow for development of a broad- spectrum treatment modality that can prevent or attenuate the replication of genetically diverse circulating human and/or zoonotic CoV strains, thereby serving as a universal first line therapy for current and future emerging CoV infections. [0032] As SARS-CoV-2 remains a global health concern due to the lack of specific therapies and vaccines, understanding host-CoV interactions and development of orally bioavailable broad-spectrum therapeutics that are effective in suppressing replication of SARS-CoV-2 and other genetically unrelated CoVs is desirable, and can help address the immediate unmet medical need to end the SARS-CoV-2 pandemic, as well as facilitate future pandemic preparedness with a ready-to-employ therapeutic. [0033] Disclosed herein are HSP90 inhibitory compounds and corresponding host targets that facilitate SARS-CoV-2 replication. Further described herein are orally bioavailable broad- spectrum HSP90 inhibitors that can effectively suppress virus replication for the current and furture CoVs outbreaks. [0034] Hence, aspects of the disclosure are drawn to methods of treating or preventing a coronavirus infection in a subject comprising administering to a subject a therapeutically effective amount of an HSP90 inhibitor. [0035] Further aspects of the disclosure are drawn to methods for treating or reducing the effects of a coronavirus infection in a subect comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor. [0036] Still futher, aspects of the disclosure are directed towards methods of treating or preventing a respiratory tract infection in a subject comprising administering to a subject a therapeutically effective amount of an HSP90 inhibitor. [0037] Detailed descriptions of one or more embodiments are provided herein. It is to be understood, however, that the present disclosure can be embodied in various forms. Therefore, specific details disclosed herein are not to be interpreted as limiting, but rather as a basis for the claims and as a representative basis for teaching one skilled in the art to employ the present disclosure in any appropriate manner. [0038] The singular forms “a”, “an” and “the” include plural reference unless the context clearly dictates otherwise. The use of the word “a” or “an” when used in conjunction with the term “comprising” in the claims and/or the specification may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.” [0039] Wherever any of the phrases “for example,” “such as,” “including” and the like are used herein, the phrase “and without limitation” is understood to follow unless explicitly stated otherwise. Similarly, “an example,” “exemplary” and the like are understood to be nonlimiting. [0040] The term “substantially” allows for deviations from the descriptor that do not negatively impact the intended purpose. Descriptive terms are understood to be modified by the term “substantially” even if the word “substantially” is not explicitly recited. [0041] The terms “comprising” and “including” and “having” and “involving” (and similarly “comprises”, “includes,” “has,” and “involves”) and the like are used interchangeably and have the same meaning. Specifically, each of the terms is defined consistent with the common United States patent law definition of “comprising” and is therefore interpreted to be an open term meaning “at least the following,” and is also interpreted not to exclude additional features, limitations, aspects, etc. Thus, for example, “a process involving steps a, b, and c” means that the process includes at least steps a, b and c. Wherever the terms “a” or “an” are used, “one or more” is understood, unless such interpretation is nonsensical in context. [0042] As used herein the term “about” is used herein to mean approximately, roughly, around, or in the region of. When the term “about” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term “about” is used herein to modify a numerical value above and below the stated value by a variance of 20 percent up or down (higher or lower). In embodiments, the methods described herein can treat a subject afflicted with a coronavirus infection. [0043] As used herein, the terms “treatment” and “treating” can refer to the management and care of a subject for the purpose of combating a condition, disease or disorder, such as a coronavirus infection, coronavirus disease, or coronavirus induced inflammatory response, in any manner in which one or more of the symptoms of a disease or disorder are ameliorated or otherwise beneficially altered. The term can include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound for the purpose of: alleviating or relieving symptoms or complications; delaying the progression of the condition, disease or disorder; curing or eliminating the condition, disease or disorder; and/or preventing the condition, disease or disorder, wherein “preventing” or “prevention” can refer to the management and care of a patient for the purpose of hindering the development of the condition, disease or disorder, and includes the administration of the active compounds to prevent or reduce the risk of the onset of symptoms or complications. For example, the terms “treatment” and “treating” can refer to: (i) inhibiting the progression the disease; (ii) prophylactic use, for example, preventing or limiting development of a disease, condition or disorder in an individual who may be predisposed or otherwise at risk to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease; (iii) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder; (iv) ameliorating the referenced disease state, for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing or improving the pathology and/or symptomatology) such as decreasing the severity of disease; and/or (v) eliciting the referenced biological effect. [0044] In some embodiments of this disclosure, the subject in need is a human subject or patient. In some embodiments the subject, e.g., a human, has been previously treated with an antiviral therapy. In some other embodiments the subject has not been previously treated with an antiviral therapy. The term “subject” or “patient” can refer to any organism to which aspects of the disclosure can be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. For example, subjects to which compounds of the disclosure can be administered include animals, such as mammals. Non-limiting examples of mammals include primates, such as humans. For veterinary applications, a wide variety of subjects can be suitable, e.g., livestock, for example cattle, sheep, goats, cows, and swine; poultry, for example chickens, ducks, geese, and turkeys; and domesticated animals, for example pets such as dogs and cats. For diagnostic or research applications, a wide variety of mammals can be suitable subjects, including rodents (e.g., mice, rats, hamsters), rabbits, primates, and swine such as inbred pigs and the like. The term “living subject” can refer to a subject described herein or another organism that is alive. The term “living subject” can refer to the entire subject or organism and not just a part excised (e.g., a liver or other organ) from the living subject. [0045] The phrase “alleviating a symptom of” can refer to ameliorating, reducing, or eliminating any condition or symptom associated with a viral infection, viral disease, or viral induced inflammatory response. Non-limiting examples of symptoms of coronavirus infection, coronavirus disease or coronavirus induced inflammatory response comprise high viral loads, respiratory distress, and pulmonary damage correlated with high cytokine abundance. Cytokines coordinate the body’s response to infection, trigger inflammation, and in COVID-19 (SARS-CoV-2) they can be generated in uncontrolled amounts. Generation of uncontrolled amounts of cytokines can be referred to as a “cytokine storm”. The term “cytokine storm” can refer to a series of events that result in a devastating and sometime fatal immune reaction that comprises a positive feedback loop between cytokines and immune cells that in turn leads to highly elevated levels of various cytokines. Cytokines that are induced during cytokine storm include, e.g., one or more of the following: IL4, IL2, IL1β, IL12, TNF, IFNγ, IL6, IL8, and IL10. Cytokine storm can lead to multi-organ failure (heart, lung, kidneys) and lead to death. Non-limiting examples of symptoms of viral infections, for example SARS-CoV-2, include cough, shortness of breath, difficulty breathing, fever, chills, muscle pain, headache, exhaustion, sore throat, loss of taste or small, nausea, vomiting and/or diarrhea. Symptoms may appear 2, 5, 14, 28, or greater than 28 days after exposure to the virus. [0046] In an embodiment, the coronavirus infection comprises or can be caused by a human coronavirus or a zoonotic coronavirus. [0047] The term “coronavirus” can refer to a virus in the Coronaviridae family, Orthocoronavirinea subfamily. [0048] The term “zoonotic virus” can refer to a virus that can be transmitted between vertebrate animals to people under natural conditions. [0049] In an embodiment, the coronavirus infection comprises severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or middle east respiratory coronavirus (MERS- CoV). SARS-CoV-2 is a recently discovered coronavirus that causes coronavirus disease (COVID-19). COVID-19 can present asymptomatically or symptomatically in subjects infected with SARS-CoV-2. COVID-19 can lead to respiratory illness. Non-limiting examples of symptoms COVID-19 include fever or chills, couch, shortness of breath or difficulty breathing, fatigue, muscle or body aches, headache, sore throat, congestion or runny nose, nausea or vomiting, and diarrhea. MERS-CoV is a coronavirus that causes Middle East respiratory syndrome (MERS). MERS can present asymptomatically or symptomatically in an individual infected with MERS-CoV. Non-limiting examples of symptoms of MERS include fever, cough, shortness of breath, diarrhea, and nausea or vomiting. [0050] In embodiments, the method comprises administering to a subject in need thereof an HSP90 inhibitor or a pharmaceutical composition comprising the same. The term “administration” can refer to introducing a composition described herein into a subject. Non- limiting examples of routes of administration of the composition comprise topical administration, oral administration, or intranasal administration. However, any route of administration, such as intravenous, subcutaneous, peritoneal, intra-arterial, inhalation, vaginal, rectal, introduction into the cerebrospinal fluid, intravascular either veins or arteries, or instillation into body compartments can be used. As used herein, the term “administering” can refer to introducing a substance, such as a HSP90 inhibitor, into a subject. Any route of administration can be utilized including, for example, intranasal, topical, oral, parenteral, intravitreal, intraocular, ocular, subretinal, intrathecal, intravenous, subcutaneous, transcutaneous, intracutaneous, intracranial and the like administration. In embodiments, “administering” can also refer to providing a therapeutically effective amount of a Formulation or pharmaceutical composition to a subject. The Formulation or pharmaceutical compound can be administered alone, but can be administered with other compounds, excipients, fillers, binders, carriers or other vehicles selected based upon the chosen route of administration and standard pharmaceutical practice. Administration can be by way of carriers or vehicles, such as injectable solutions, including sterile aqueous or non-aqueous solutions, or saline solutions; creams; lotions; capsules; tablets; granules; pellets; powders; suspensions, emulsions, or microemulsions; patches; micelles; liposomes; vesicles; implants, including microimplants; eye drops; other proteins and peptides; synthetic polymers; microspheres; nanoparticles; and the like. [0051] The term “pharmaceutical composition” is used in its widest sense, and can encompass all pharmaceutically applicable compositions containing at least one active substance, and optional carriers, adjuvants, diluents, and constituents. The term “pharmaceutical composition” can also encompass a composition comprising the active substance in the form of derivative or pro-drug, such as pharmaceutically acceptable salts or esters. The manufacture of pharmaceutical compositions for different routes of administration falls within the capabilities of a person skilled in medicinal chemistry. [0052] Embodiments of the pharmaceutical composition can be administered to a subject in one or more doses. Those of skill can readily appreciate that dose levels can vary as a function of the specific the pharmaceutical composition administered, the severity of the symptoms and the susceptibility of the subject to side effects. Dosages for a given compound are readily determinable by those of skill in the art by a variety of means. [0053] In an embodiment, multiple doses of the pharmaceutical composition are administered. The frequency of administration of the pharmaceutical composition can vary depending on any of a variety of factors, e.g., severity of the symptoms, and the like. For example, in an embodiment, the pharmaceutical composition can be administered once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (qid), three times a day (tid), or four times a day. As described herein, in an embodiment, the pharmaceutical composition is administered 1 to 4 times a day over a 1 to 10-day time period. [0054] The duration of administration of the composition or pharmaceutical composition analogue, e.g., the period of time over which the composition or pharmaceutical composition is administered, can vary, depending on any of a variety of factors, e.g., patient response, etc. For example, the composition or pharmaceutical composition in combination or separately, can be administered over a period of time of about one day to one week, about one day to two weeks. [0055] The amount of the compositions and pharmaceutical compositions described herein can be effective in treating the condition or disease can be determined by standard clinical techniques. In addition, in vitro or in vivo assays can be employed to help identify optimal dosage ranges. The precise dose to be employed can also depend on the route of administration, and can be decided according to the judgment of the practitioner and each patient's circumstances. [0056] As used herein, the nomenclature alkyl, alkoxy, and carbonyl, for example, is used as is understood by those of skill in the chemical art. [0057] The term “alkoxy” can represent an alkyl group of indicated number of carbon atoms attached to the parent molecular moiety through an oxygen atom. Non-limiting examples of alkoxy groups can include, for example, methoxy, ethoxy, propoxy and isopropoxy. [0058] As used herein, the term “alkyl” can include those alkyl groups of a designated number of carbon atoms. Alkyl groups can be straight or branched. Non-limiting examples of alkyl can include methyl, ethyl, propyl, isopropyl, butyl, iso-, sec- and tert-butyl, pentyl, hexyl, heptyl, and 3-ethylbutyl. [0059] The term “alkenyl” as used herein can refer to a straight or branched chain hydrocarbon containing from 2 to 10 carbons and containing at least one carbon- carbon double bond formed by the removal of two hydrogens. Representative examples of alkenyl include, but are not limited to, ethenyl, 2-propenyl, 2-methyl-2- propenyl, 3-butenyl, 4- pentenyl, 5-hexenyl, 2-heptenyl, 2-methyl-1-heptenyl, and 3-decenyl. [0060] The term “alkenoxy” can refer to an alkenyl group attached to the parent group through an oxygen atom. [0061] The term “alkynyl” as used herein can refer to a straight or branched chain hydrocarbon group containing from 2 to 10 carbon atoms and containing at least one carbon- carbon triple bond. Representative examples of alkynyl include, but are not limited, to acetylenyl, 1 -propynyl, 2-propynyl, 3-butynyl, 2-pentynyl, and 1 -butynyl. [0062] The term “aryl” can refer to an aromatic hydrocarbon ring system containing at least one aromatic ring. The aromatic ring can optionally be fused or otherwise attached to other aromatic hydrocarbon rings or non-aromatic hydrocarbon rings. Examples of aryl groups include, for example, phenyl, naphthyl, anthracenyl, 1, 2, 3, 4- tetrahydronaphthalene and biphenyl. The aryl groups of the disclosure can be substituted with various groups as provided herein. Thus, any carbon atom present within an aryl ring system and available for substitution can be further bonded to a variety of ring substituents, such as, for example, halogen, hydroxy, nitro, cyano, amino, C1-C8alkyl, C1-C8alkoxy, mono- and di(C1- C8alkyl)amino, C3-C10cycloalkyl, (C3-C10cycloalkyl)alkyl, (C3-C10cycloalkyl)alkoxy, C2- C9heterocycloalkyl, C1-C8alkenyl, C1-C8alkynyl, halo(C1-C8)alkyl, halo(C1-C8)alkoxy, oxo, amino(C1-C8)alkyl, mono- and di(C1-C8alkyl)amino(C1-C8)alkyl, C1-C8acyl, C1-C8acyloxy, C1-C8sulfonyl, C1-C8thio, C1-C8sulfonamido, and C1-C8aminosulfonyl. [0063] The term “carboxy” as used herein can refer to a -CO2H group. [0064] The term “cycloalkyl” can refer to a C3-C8 cyclic hydrocarbon. Non-limiting examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. Examples can include C3-C6 cycloalkyl groups. The cycloalkyl groups of the disclosure can be substituted with various groups as provided herein. Thus, any carbon atom present within a cycloalkyl ring system and available for substitution can be further bonded to a variety of ring substituents, such as, for example, halogen, hydroxy, nitro, cyano, amino, C1-C8alkyl, C1-C8alkoxy, mono- and di(C1-C8alkyl)amino, C3-C10cycloalkyl, (C3- C10cycloalkyl)alkyl, (C3-C10cycloalkyl)alkoxy, C2-C9heterocycloalkyl, C1-C8alkenyl, C1- C8alkynyl, halo(C1-C8)alkyl, halo(C1-C8)alkoxy, oxo, amino(C1-C8)alkyl and mono- and di(C1-C8alkyl)amino(C1-C8)alkyl. [0065] The terms “halogen” or “halo” can refer to fluorine, chlorine, bromine, and iodine. [0066] The term “haloalkoxy” can refer to an alkoxy group substituted with one or more halogen atoms, where each halogen is independently F, Cl, Br or I. Haloalkoxy groups can contain 1-6 carbons, such as 1-4 carbons, and such as 1-2 carbons. “Haloalkoxy” can include perhaloalkoxy groups, such as OCF3 or OCF2CF3. An example of a haloalkoxy group is trifluoromethoxy. [0067] The term “haloalkyl” can refer to an alkyl group substituted with one or more halogen atoms, where each halogen is independently F, Cl, Br or I. Haloalkyl groups can contain 1-6 carbons, such as 1-4 carbons, and such as 1-2 carbons. “Haloalkyl” can include perhaloalkyl groups, such as CF3 or CF2CF3. An example of a haloalkyl group is trifluoromethyl. [0068] The term “heterocycloalkyl” can refer to a ring or ring system containing at least one heteroatom selected from nitrogen, oxygen, and sulfur, wherein said heteroatom is in a non- aromatic ring. The heterocycloalkyl ring is optionally fused to or otherwise attached to other heterocycloalkyl rings and/or non-aromatic hydrocarbon rings and/or phenyl rings. Useful heterocycloalkyl groups can have from 3 to 7 members. In embodiments, heterocycloalkyl groups can have 5 or 6 members. Examples of heterocycloalkyl groups can include, for example, 1, 2, 3, 4-tetrahydroisoquinolinyl, piperazinyl, morpholinyl, piperidinyl, tetrahydrofuranyl, pyrrolidinyl, pyridinonyl, dihydropyrrolidinyl, pyrrol id inonyl, and pyrazolidinyl. The heterocycloalkyl groups of the disclosure can be substituted with various groups as provided herein. Thus, any atom present within a heterocycloalkyl ring and available for substitution can be further bonded to a variety of ring substituents, such as, for example, halogen, hydroxy, nitro, cyano, amino, C1-C8alkyl, C1-C8alkoxy, mono- and di(C1- C8alkyl)amino, C3-C10cycloalkyl, (C3-C10cycloalkyl)alkyl, (C3-C10cycloalkyl)alkoxy, C2- C9heterocycloalkyl, C1-C8alkenyl, C1-C8alkynyl, halo(C1-C8)alkyl, halo(C1-C8)alkoxy, oxo, amino(C1-C8)alkyl and mono- and di(C1-C8alkyl)amino(C1-C8)alkyl. [0069] The term “heteroaryl” can refer to an aromatic ring system containing at least one heteroatom selected from nitrogen, oxygen, and sulfur. The heteroaryl ring can be fused or otherwise attached to one or more heteroaryl rings, aromatic or non-aromatic hydrocarbon rings or heterocycloalkyl rings. Examples of heteroaryl groups include, for example, pyridine, furan, thienyl, 5, 6, 7, 8-tetrahydroisoquinoline and pyrimidines. The heteroaryl groups of the disclosure can be substituted with various groups as provided herein. Thus, any carbon atom present within an heteroaryl ring system and available for substitution can be further bonded to a variety of ring substituents, such as, for example, halogen, hydroxy, nitro, cyano, amino, C1-C8alkyl, C1-C8alkoxy, mono- and di(C1-C8alkyl)amino, C3-C10cycloalkyl, (C3-C10cycloalkyl)alkyl, (C3-C10 ocycloalkyl)alkoxy, C2-C9heterocycloalkyl, C1-C8alkenyl, C1-C8alkynyl, halo(C1-C8)alkyl, halo(C1-C8)alkoxy, oxo, amino(C1-C8)alkyl and mono-and di(C1-C8alkyl)amino(C1-C8)alkyl. [0070] Non-limiting examples of heteroaryl groups include thienyl, benzothienyl, pyridyl, quinolyl, pyrazolyl, pyrimidyl, imidazolyl, benzimidazolyl, furanyl, benzofuranyl, dibenzofuranyl, thiazolyl, benzothiazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, benzisothiazolyl, triazolyl, pyrrolyl, indolyl, pyrazolyl, and benzopyrazolyl. [0071] The compounds of this disclosure can contain one or more asymmetric carbon atoms, so that the compounds can exist in different stereoisomeric forms. These compounds can be, for example, racemates, chiral non-racemic or diastereomers. In these situations, the single enantiomers, i.e., optically active forms, can be obtained by asymmetric synthesis or by resolution of the racemates. Resolution of the racemates can be accomplished, for example, by conventional methods such as crystallization in the presence of a resolving agent; chromatography, using, for example a chiral HPLC column; or derivatizing the racemic mixture with a resolving reagent to generate diastereomers, separating the diastereomers via chromatography, and removing the resolving agent to generate the original compound in enantiomerically enriched form. Any of the procedures described herein can be repeated to increase the enantiomeric purity of a compound. [0072] When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless otherwise specified, it is intended that the compounds include the cis, trans, Z- and E- configurations. Likewise, all tautomeric forms are also intended to be included. [0073] As used herein, “pharmaceutically acceptable derivatives” of a compound can include salts, esters, enol ethers, enol esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, hydrates or prodrugs thereof. Such derivatives can be readily prepared by those of skill in this art using known methods for such derivatization. The compounds produced can be administered to animals or humans without substantial toxic effects and either are pharmaceutically active or are prodrugs. [0074] Pharmaceutically acceptable salts can include, but are not limited to, amine salts, such as but not limited to N,N'-dibenzylethylenediamine, chloroprocaine, choline, ammonia, diethanolamine and other hydroxyalkylamines, ethylenediamine, N-methylglucamine, procaine, N-benzylphenethylamine, 1-para-chlorobenzyl-2-pyrrolidin-1’- ylmethylbenzimidazole, diethylamineand other alkylamines, piperazine and tris(hydroxymethyl) aminomethane; alkali metal salts, such as but not limited to lithium, potassium and sodium; alkali earth metal salts, such as but not limited to barium, calcium and magnesium; transition metal salts, such as but not limited to zinc; and other metal salts, such as but not limited to sodium hydrogen phosphate and disodium phosphate; and also including, but not limited to, salts of mineral acids, such as but not limited to hydrochlorides and sulfates; and salts of organic acids, such as but not limited to acetates, lactates, malates, tartrates, citrates, ascorbates, succinates, butyrates, valerates and fumarates. [0075] Pharmaceutically acceptable esters can include, but are not limited to, alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl and heterocyclyl esters of acidic groups, including, but not limited to, carboxylic acids, phosphoric acids, phosphinic acids, sulfonic acids, sulfinic acids and boronic acids. [0076] Pharmaceutically acceptable enol ethers can include, but are not limited to, derivatives of Formula C=C(OR) where R is hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, or heterocyclyl. Pharmaceutically acceptable enol esters include, but are not limited to, derivatives of Formula C=C(OC(O)R) where R is hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl ar heterocyclyl. [0077] Pharmaceutically acceptable solvates and hydrates are complexes of a compound with one or more solvent or water molecules, or 1 to about 100, or 1 to about 10, or one to about 2, 3 or 4, solvent or water molecules. [0078] “Formulation” as used herein can refer to any collection of components of a compound, mixture, or solution selected to provide optimal properties for a specified end use, including product specifications and/or service conditions. The term Formulation can include liquids, semi-liquids, colloidal solutions, dispersions, emulsions, microemulsions, and nanoemulsions, including oil-in-water emulsions and water-in-oil emulsions, pastes, powders, and suspensions. The Formulations can also be included, or packaged, with other non-toxic compounds, such as cosmetic carriers, excipients, binders and fillers, and the like. For example, the acceptable cosmetic carriers, excipients, binders, and fillers for use in the practice of the present disclosure are those which render the compounds amenable to oral delivery and/or provide stability such that the Formulations of the present disclosure exhibit a commercially acceptable storage shelf life. [0079] The compounds described herein can be administered orally, topically, parenterally, by inhalation or spray or rectally in dosage unit Formulations containing conventional non- toxic pharmaceutically acceptable carriers, adjuvants and vehicles. The term parenteral as used herein includes percutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, or intrathecal injection or infusion techniques and the like. The pharmaceutical compositions described herein may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. [0080] Compositions intended for oral use can be prepared according to any method known in the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preservative agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients can be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets can be uncoated or they can be coated by known techniques. In some cases, such coatings can be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be employed. [0081] Formulations for oral use can also be presented as hard gelatin capsules, wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil. [0082] Formulations for oral use can also be presented as lozenges. [0083] Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions can also contain one or more preservatives, for example ethyl, or n-propyl p- hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin. [0084] Oily suspensions can be Formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions can contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents and flavoring agents can be added to provide palatable oral preparations. These compositions can be preserved by the addition of an antioxidant such as ascorbic acid. [0085] Pharmaceutical compositions of the disclosure can also be in the form of oil-in- water emulsions. The oily phase can be a vegetable oil or a mineral oil or mixtures of these. Suitable emulsifying agents can be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions can also contain sweetening and flavoring agents. [0086] Syrups and elixirs can be Formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such Formulations may also contain a demulcent, a preservative and flavoring and coloring agents. The pharmaceutical compositions can be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension can be Formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been described herein. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1 ,3-butanediol. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil can be employed including synthetic mono-or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. The compositions disclosed herein can also be administered in the form of suppositories, e.g., for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and can therefore melt in the rectum to release the drug. Such materials can include cocoa butter and polyethylene glycols. [0087] The compositions disclosed herein can be administered parenterally in a sterile medium. The drug, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle. Adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle. [0088] In embodiments, the active ingredients can be Formulated in a cream with an oil- in- water cream base. If desired, the aqueous phase of the cream base can include, for example at least 30% w/w of a polyhydric alcohol such as propylene glycol, butane-1 ,3-diol, mannitol, sorbitol, glycerol, polyethylene glycol and mixtures thereof. The topical Formulation can include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas, such as a penetration enhancer. Examples of such dermal penetration enhancers can include dimethylsulfoxide and related analogs. The compounds of this disclosure can also be administered by a transdermal device. Topical administration can be accomplished using a patch either of the reservoir and porous membrane type or of a solid matrix variety. In either case, the active agent can be delivered continuously from the reservoir or microcapsules through a membrane into the active agent permeable adhesive, which is in contact with the skin or mucosa of the recipient. If the active agent is absorbed through the skin, a controlled and predetermined flow of the active agent is administered to the recipient. In the case of microcapsules, the encapsulating agent can also function as the membrane. The transdermal patch can include the compound in a suitable solvent system with an adhesive system, such as an acrylic emulsion, and a polyester patch. The oily phase of the emulsions of this disclosure can be constituted from known ingredients in a known manner. While the phase can comprise merely an emulsifier, it can comprise a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. In embodiments, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. Embodiments can also include both an oil and a fat. Together, the emulsifier(s) with or without stabilizer(s) make-up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream Formulations. Emulsifiers and emulsion stabilizers suitable for use in the Formulation of the present disclosure can include Tween 60, Span 80, cetostearyl alcohol, myristyl alcohol, glyceryl monostearate, and sodium lauryl sulfate, among others. The choice of suitable oils or fats for the Formulation is based on achieving the desired cosmetic properties, since the solubility of the active compound in most oils likely to be used in pharmaceutical emulsion Formulations is very low. Thus, the cream can be a non-greasy, non-staining and washable product with suitable consistency to avoid leakage from tubes or other containers. Straight or branched chain, mono- or dibasic alkyl esters such as di- isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or a blend of branched chain esters may be used. These can be used alone or in combination depending on the properties required. In embodiments, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils can be used. [0089] Formulations for parenteral administration can be in the form of aqueous or non- aqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions can be prepared from sterile powders or granules having one or more of the carriers or diluents mentioned for use in the Formulations for oral administration. The compounds can be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, and/or various buffers. Other adjuvants and modes of administration are well and widely known in the pharmaceutical art. The Formulations or pharmaceutical composition can also be included, or packaged, with other non-toxic compounds, such as pharmaceutically acceptable carriers, excipients, binders and fillers including, but not limited to, glucose, lactose, gum acacia, gelatin, mannitol, xanthan gum, locust bean gum, galactose, oligosaccharides and/or polysaccharides, starch paste, magnesium trisilicate, talc, corn starch, starch fragments, keratin, colloidal silica, potato starch, urea, dextrans, dextrins, and the like. For example, the pharmaceutically acceptable carriers, excipients, binders, and fillers for use in the practice of the present disclosure can be those which render the compounds of the disclosure amenable to intranasal delivery, oral delivery, parenteral delivery, intravitreal delivery, intraocular delivery, ocular delivery, subretinal delivery, intrathecal delivery, intravenous delivery, subcutaneous delivery, transcutaneous delivery, intracutaneous delivery, intracranial delivery, topical delivery and the like. Moreover, the packaging material can be biologically inert or lack bioactivity, such as plastic polymers or silicone, and can be processed internally by the subject without affecting the effectiveness of the composition/Formulation packaged and/or delivered therewith. [0090] Different forms of the present inventive Formulation can be calibrated in order to adapt both to different individuals and to the different needs of a single individual. However, the present Formulation need not counter every cause in every individual. Rather, by countering the necessary causes, the present Formulation can restore the body to its normal function. Then the body can correct the remaining deficiencies. [0091] The term “therapeutically effective amount” as used herein can refer to that amount of an embodiment of the composition or pharmaceutical composition being administered that can relieve at least to some extent one or more of the symptoms of the disease or condition being treated, and/or that amount that can prevent, to some extent, one or more of the symptoms of the condition or disease that the subject being treated has or is at risk of developing. As used interchangeably herein, “subject,” “individual,” or “patient,” can refer to a vertebrate, such as a mammal, for example a human. Mammals include, but are not limited to, murines, simians, humans, farm animals, sport animals, and pets. The term “pet” can refer to, for example, a dog, cat, guinea pig, mouse, rat, rabbit, ferret, and the like. The term farm animal can refer to, for example, a horse, sheep, goat, chicken, pig, cow, donkey, llama, alpaca, turkey, and the like. [0092] A “pharmaceutically acceptable excipient,” “pharmaceutically acceptable diluent,” “pharmaceutically acceptable carrier,” or “pharmaceutically acceptable adjuvant” can refer to an excipient, diluent, carrier, and/or adjuvant that are useful in preparing a pharmaceutical composition that are generally safe, non-toxic and neither biologically nor otherwise undesirable, and include an excipient, diluent, carrier, and adjuvant that are acceptable for veterinary use and/or human pharmaceutical use. “A pharmaceutically acceptable excipient, diluent, carrier and/or adjuvant” as used herein can refer to, for example, one and more such excipients, diluents, carriers, and adjuvants. [0093] The phrase “pharmaceutical composition” or a “pharmaceutical Formulation” can refer to a composition or pharmaceutical composition suitable for administration to a subject, such as a mammal, especially a human and that can refer to the combination of an active agent(s), or ingredient with a pharmaceutically acceptable carrier or excipient, making the composition suitable for diagnostic, therapeutic, or preventive use in vitro, in vivo, or ex vivo. A “pharmaceutical composition” can be sterile and can be free of contaminants that can elicit an undesirable response within the subject (e.g., the compound(s) in the pharmaceutical composition is pharmaceutical grade). Pharmaceutical compositions can be designed for administration to subjects or patients in need thereof via a number of different routes of administration including oral, intranasal, topical, intravenous, buccal, rectal, parenteral, intraperitoneal, intradermal, intracheal, intramuscular, subcutaneous, by stent-eluting devices, catheters-eluting devices, intravascular balloons, inhalational and the like. [0094] In embodiments, the pharmaceutical composition can comprise a therapeutically effective amount of a HSP90 inhbitor, and a therapeutically effective amount of one or more additional active agents (such as one or more anti-oxidants, anti-allergenics, anti- inflammatory agents, anti-viral agents, pain relievers, or antipyretics). For example, the one or more anti-oxidants can be synthetic antioxidants, natural antioxidants, or a combination thereof. The term “anti-oxidant” can refer to a substance that can prevent, delay, or otherwise inhibit the oxidation of a compound or biological substance. The term “anti-allergenic” can refer to any substance that can counter an allergic reaction. The term anti-inflammatory agent can refer to an agent or compound that has anti-inflammatory effects. Non-limiting examples of anti-inflammatory agents include acetylsalicylic acid (aspirin), ibuprofen, naproxen sodium, and acetaminophen. The term “anti-viral” can refer to any compound or drug which targets one or more steps in the virus life cycle with direct anti-viral therapeutic effects. The compounds of the present disclosure can be administered to a subject in need thereof together with one or more antiviral agents either simultaneously or sequentially. The term “pain reliever” can refer to any agent administered to alleviate to treat a subject experiencing pain or discomfort. The term “antipyretics” can refer to any compound which treats, alleviates, or reduces a fever. [0095] Dosage levels from about 0.1 mg to about 140 mg per kilogram of body weight per day can be useful in the treatment of the conditions indicated herein (about 0.5 mg to about 7 g per patient per day). For example, dosage levels comprise less than 0.1 mg, about 0.1 mg, about 0.5 mg, about 1.0 mg, about 2.5 mg, about 5 mg, about 7.5 mg, about 10 mg, about 15 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 250 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1.0 g, and greater than 1.0 g per kilogram of body weight per day.The amount of active ingredient that can be combined with the carrier materials to produce a single dosage form can vary depending upon the host treated and the particular mode of administration. [0096] Dosage unit forms can generally contain between from about 1 mg to about 500 mg of an active ingredient. For example, dosage unit forms can contain an active ingredient comprising the range of less than 0.1 mg, about 0.1 mg, about 0.5 mg, about 1.0 mg, about 2.5 mg, about 5 mg, about 7.5 mg, about 10 mg, about 15 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 250 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1.0 g, and greater than 1.0 g. The daily dose can be administered in one dose per day, or more than one dose per day, such as two doses per day, three doses per day, four doses per day, or more than four doses per day. [0097] It will be understood, however, that the specific dose level for any particular subject can depend upon a variety of factors, non-limiting examples of which include the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy. [0098] As used herein, “pharmaceutically acceptable derivatives” of a compound can include salts, esters, enol ethers, enol esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, hydrates or prodrugs thereof. Such derivatives can be readily prepared by those of skill in this art using known methods for such derivatization. The compounds produced can be administered to animals or humans without substantial toxic effects and either are pharmaceutically active or are prodrugs. [0099] For example, an active ingredient can comprise an antivirial. The compounds of the present disclosure can be administered alone or in combination with at least one antiviral agent. I. HSP90 Inhibitors and Methods of Using the Same [00100] Some HSP90 inhibitors are known in the art. For example, HSP90 inhibitors are disclosed in International Publication Nos. WO 2006/091963, WO 2007/101156, WO 2008/130879, and WO 2016/08153, all of which are incorporated herein by reference in their entireties. [00101] In one embodiment of this disclosure, the HSP90 inhibitor useful in the methods and compositions of the disclosure is of Formula (I):
Figure imgf000032_0001
or a pharmaceutically acceptable salt thereof, wherein, R3 and R4 are independently (a) H, (b) halo, or (c) a C1-C15 alkyl group where up to six of the carbon atoms in said alkyl group are optionally replaced independently by R22, carbonyl, ethenyl, ethynyl or a moiety selected from N, O, S, SO2, or SO, with the proviso that two O atoms, two S atoms, or an O and S atom are not immediately adjacent each other, wherein R22 is (i) heteroaryl, (ii) aryl, (iii) saturated or unsaturated C3-C10 cycloalkyl, or (iv) saturated or unsaturated C2-C10 heterocycloalkyl, wherein each aryl, heteroaryl, saturated or unsaturated cycloalkyl, or saturated or unsaturated heterocycloalkyl, independently, is optionally substituted with at least one group, which independently is hydroxy, halo, amino, cyano, carboxy, carboxamido, nitro, oxo, -S-(C1-C6)alkyl, -SO2-(C1- C6)alkyl, -SO2- aryl, -SO-(C1-C6)alkyl, -SO-aryl, -SO2NH2, -SO2NH-(C1-C6)alkyl, -SO2NH- aryl, (C1-C6)alkoxy, or mono- or di-(C1-C10)alkylamino; and each R22 is optionally fused to a C6-C10 aryl group, C5-C8 saturated cyclic group, or a C5-C10 heterocycloalkyl group; wherein each moiety is optionally substituted at any available position with C1-C10 alkyl, C1-C10 haloalkyl, C2-C10 alkenyl, C2-C10 alkynyl, hydroxy, carboxy, carboxamido, oxo, halo, amino, cyano, nitro, -SH, -S-(C1-C6)alkyl, -SO2-(CrC6)alkyl, -SO2NH2, -SO2NH-(C1- C6)alkyl, -SO2NH- aryl, -SO2-aryl, -SO-(C1-C6)alkyl, -SO2-aryl, C1-C6 alkoxy, C2- C10 alkenyloxy, C2-C10 alkynyloxy, mono- or di-(C1-C10)alkylamino, -OC1-C10 alkyl-Z, -O- C(O)C1-C10 alkyl-Z, or R23, wherein Z is OR30 or -N(R30)2, wherein each R30 is independently -H or C1-C6 alkyl, or N(R30)2 represents pyrrolidinyl, piperidinyl, piperazinyl, azepanyl, 1 ,3- or 1 ,4-diazepanyl, or morpholinyl, each of which is optionally substituted with hydroxy, amino, aminoalkyl, C1-C6 alkyl, mono- or di(C1- C6)alkylamino, C1-C6 alkoxy, or halogen; R0 is -H, -C1-C10 alkyl, -C2-C10 alkenyl, -C2-C10 alkynyl, aryl, heteroaryl, or -C1-C6 acyl; R23 is (1) heteroaryl, (2) aryl, (3) saturated or unsaturated C5-C10 cycloalkyl, or (4) saturated or unsaturated C5-C10 heterocycloalkyl, and the R23 groups are optionally substituted with at least one group which independently is hydroxy, oxo, halo, amino, cyano, nitro, -SH, -S-(C1-C6)alkyl, -SO2-(C1-C6)alkyl, -SO2-aryl, -SO- (C1-C6)alkyl, -SO-aryl, -SO2NH2, -SO2NH-(C1-C6)alkyl, -SO2NH-aryl, (C1- C6)alkoxy, or mono- or di-(C1-C10)alkylamino; and wherein one or both of R3 and R4 is optionally substituted with a group R50, wherein R50 is:
Figure imgf000033_0001
wherein d and k are integers independently selected from 1 and 2; R201 is (C1-C6)alkyl where the alkyl is optionally substituted with (C3-C7)cycloalkyl, ( C2-C6) alkenyl, (C2-C6)alkynyl, hydroxy, halogen, nitro, or cyano; and T is O or NR202 where R202 is hydrogen or (C1-C6)alkyl; and R301 and R302 are independently hydrogen or (C1-C6)alkyl, and R303 is absent, hydrogen, or (C1-C6)alkyl; R7 is O, S, NH, N-OH, N-NH2, N-NHR22, N-NH-(C1-C6 alkyl), N-O-(C0- C6)alkyl, -R22, N-(C1-C6 alkenoxy); or N-(C1-C6 alkoxy optionally substituted with carboxy); Y is N or CRc, wherein each Rc independently is hydrogen, halogen, cyano, nitro, -C(O)Rc’, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3- C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl, wherein each alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl group is optionally substituted with from 1-4 groups that are independently C1-C6 alkyl, C1-C6 alkoxy, halogen, hydroxy, amino, mono- or di-(C1-C6) alkylamino, cyano, nitro, halo(C1-C6)alkyl, halo(C1-C6)alkoxy, carboxamide, heterocycloalkyl, aryl, or heteroaryl, whereinthe aryl and heteroaryl groups are optionally substituted with from 1-4 groups that are independently C1-C6 alkyl, C1-C6 alkoxy, halogen, hydroxy, amino, mono- or di-(C1-C6) alkylamino, halo(C1-C6)alkyl, or carboxamide; Rc’ is -C1-C6 alkyl, -ORc”, or -N(RCN)2, wherein Rc” is -H, C1-C10 alkyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; each RCN is independently -H, -C1-C10 alkyl, -C1-C10 - aloalkyl, -C3-C7 cycloalkyl, -heterocycloalkyl, -C1-C6 acyl, -aryl, or -heteroaryl, wherein each alkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl group is optionally substituted with from 1-4 groups that are independently C1-C6 alkyl, C1-C6 alkoxy, halogen, hydroxy, amino, mono- or di- (C1-C6) alkylamino, nitro, halo(C1-C6)alkyl, halo(C1- C6)alkoxy, or carboxamide; X1 is N or CRC; Q1, Q2, and Q3 are independently N or CRQ, wherein one and only one of Q1, Q2, and Q3 is C-R21, and wherein each RQ is independently hydrogen, halogen, -N(RCN)2, C1-C6 alkyl, C1-C6 haloalkyl, C3-C7 cycloalkyl, aryl, or heteroaryl, or R21, wherein each alkyl, cycloalkyl, aryl, and heteroaryl group is optionally substituted with from 1-4 groups that are independently C1-C6 alkyl, C1-C6 alkoxy, halogen, hydroxy, amino, mono- or di-(C1-C6) alkylamino, halo(C1- C6)alkyl, halo(C1-C6)alkoxy, or carboxamide; R21 is cyano, -C(O)OH, -C(O)-O(C1-C6 alkyl), or a group of the Formula
Figure imgf000034_0001
wherein R1 and R2 are independently H, hydroxy, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, heteroaryl, aryl, C3-C8 cycloalkyl, heterocycloalkyl, wherein each alkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl group is optionally substituted with from 1-4 groups that are independently C1-C6 alkyl, C1-C6 alkoxy, halogen, hydroxy, amino, mono- or di-(C1- C6) alkylamino, nitro, halo(C1-C6)alkyl, halo(C1-C6)alkoxy, or carboxamide; or R1 and R2 together with the nitrogen to which they are both attached, form a heterocycloalkyl which optionally contains one or more additional heteroatoms which are, independently, O, N, S, or N(RCN); and X4 is O, S, NH, NOH, N-NH2, N-NHaryl, N-NH-(C1-C6 alkyl), or N-(C1-C6 alkoxy); X2 and X3 are independently C, O, N, or S(O)p wherein p is 0, 1, or 2; and n is 0, 1, 2, 3, or 4; provided that when (i) X2 is C, then R5 and R6 are independently H, C1-C6 alkyl, or aryl, wherein the aryl is optionally substituted with from 1-4 groups that are independently C1- C6 alkyl, C1-C6 alkoxy, halogen, hydroxy, amino, mono- or di-(C1-C6) alkylamino, nitro, halo(C1-C6)alkyl, halo(C1-C6)alkoxy, or carboxamide, wherein any two adjacent substituted aryl positions, together with the carbon atoms to which they are attached, form an unsaturated cycloalkyl or heterocycloalkyl; or R5 and R6 together with the carbon to which they are attached form a 3-8 membered ring; (ii) X2 is N, then R6 is absent and R5 is H or C1-C6 alkyl; (iii) X3 is C, then it is substituted with two groups that are independently H, C1-C6 alkyl, or mono- or di-(C1-C6)alkylamino(C1-C6)alkyl; and (iv) X2 is O or S(O)p, then R6 and R5 are absent. [00102] In Formula I, R3 and R4 are, as noted herein, independently (a) hydrogen, (b) halo, or (c) an alkyl group having from 1 -15 carbon atoms. All, but no more than about six, of the carbon atoms in the alkyl group may be replaced independently by the various groups listed herein in connection with Formula I. [00103] Thus, when the alkyl group is methyl, i.e., a one carbon atom alkyl group, replacement of that carbon atom with, for example, nitrogen or sulfur, the resulting group is not an alkyl group but instead can be an amino or thio group, respectively. Similarly, when the carbon atom being replaced terminates the alkyl group, the terminal group can become another moiety such as pyrimidinyl, amino, phenyl, or hydroxy. [00104] Replacement of a carbon atom with a group such as, for example, oxygen, nitrogen, or sulfur can require appropriate adjustment of the number of hydrogens or other atoms required to satisfy the replacing atom's valency. Thus, when the replacement is N or O, the number of groups attached to the atom being replaced can be reduced by one or two to satisfy the valency of the nitrogen or oxygen respectively. Similar considerations can be readily apparent to those skilled in the art with respect to replacement by ethenyl and ethynyl. [00105] Thus, replacement as permitted herein results in the term “C1-C15 alkyl” as defined in connection with Formula I encompassing groups such as, but not limited to: amino, hydroxy, phenyl, benzyl, propylaminoethoxy, butoxyethylamino, pyrid-2-ylpropyl, diethylaminomethyl, pentylsulfonyl, methylsulfonamidoethyl, 3-[4- (butylpyrimidin-2- yl)ethyl]phenyl, butoxy, dimethylamino, 4-(2-(benzylamino)ethyl)pyridyl, but-2-enylamino, 4-(1 -(methylamino)pent-3-en-2- ylthio)phenyl, 2-(N-methyl-hexanamido)ethoxy)methyl, and 4-(((3-methoxy-4-(4- methyl-1 H-imidazol-2-yl)but-1 -enyl)(methyl)amino)- methyl)phenyl. [00106] Compounds of Formula I include, for example, those where X1 is carbon optionally substituted with C1-C6 alkyl, such as C1-C3 alkyl. Other compounds of Formula I are those where X1 is carbon optionally substituted with C1-C6 alkyl and Y is CRC wherein Rc is -H, C1-C6 alkyl, C1-C3 haloalkyl, C3-C7 cycloalkyl, or C3-C7 cycloalkyl(C1-C6)alkyl. In compounds of Formula I, X1 can be carbon optionally substituted with C1-C2 alkyl and Y is CRC wherein RC is -H, C1-C4 alkyl, C1-C3 haloalkyl, cyclopropyl, or cyclopropyl(C1-C2)alkyl. [00107] In embodiments, compounds of Formula I can be those where X1 is CH. Other compounds of Formula I can be those where X1 is CH and Y is CRC wherein RC is -H, C1-C3 alkyl, C1-C3 haloalkyl, C3-C5 cycloalkyl, or C3-C5 cycloalkyl(C1-C2)alkyl. In embodiments, compounds of Formula I can be those where X1 is CH and Y is CRC wherein RC is -H, methyl, ethyl, trifluoromethyl, cyclopropyl, or cyclopropylmethyl. In embodiments, compounds of Formula I can be those where X1 is CH and Y is CRC wherein RC is methyl, ethyl, or cyclopropyl. Other compounds of Formula I can be those where X1 is CH and Y is CRC wherein RC is trifluoromethyl. Other compounds of Formula I can be those where X1 is CH and Y is CRC wherein RC is methyl. Other compounds of Formula I can be those where X1 is CH and Y is CRC wherein RC is ethyl. Other compounds of Formula I can be those where X1 is CH and Y is CRC wherein RC is cyclopropyl. [00108] In embodiments, compounds of Formula I can be those where X1 is N. Other compounds of Formula I can be those where X1 is N and Y is CRC wherein RC is -H, C1-C3 alkyl, C1-C3 haloalkyl, C3-C5 cycloalkyl, or C3-C5 cycloalkyl(C1-C2)alkyl. In embodiments, compounds of Formula I can be those where X1 is N and Y is CRC wherein RC is -H, methyl, ethyl, trifluoromethyl, cyclopropyl, or cyclopropylmethyl. In embodiments, compounds of Formula I can be those where X1 is N and Y is CRC wherein RC is methyl, ethyl, or cyclopropyl. In embodiments, compounds of Formula I can be those where X1 is N and Y is CRC wherein RC is trifluoromethyl. In embodiments, compounds of Formula I can be those where X1 is N and Y is CRC wherein RC is methyl. In embodiments, compounds of Formula I can be those where X1 is N and Y is CRC wherein RC is ethyl. In embodiments, compounds of Formula I can be those where X1 is N and Y is CRC wherein RC is cyclopropyl. [00109] In embodiments, compounds of Formula I can be those where Q3 is CR21, wherein R21 is a group of the Formula,
Figure imgf000037_0001
R7 is O; and Y is CRC, wherein RC is hydrogen, C1-C3 alkyl, C3-C5 cycloalkyl, trifluoromethyl, or C3-C5 cycloalkyl(C1-C2)alkyl. Such compounds are compounds of Formula II herein. [00110] In embodiments, compounds of Formula I can be those where Q3 is CR21, wherein R21 is a group of the Formula,
Figure imgf000037_0002
X3 is C substituted with R9a and R9b, wherein R9a and R9b are independently H or C1- C6 alkyl. Such compounds are hereinafter compounds of Formula III. [00111] In embodiments, compounds of Formula I can be those where Q3 is CR21, wherein R21 is a group of the Formula
Figure imgf000037_0003
Q1 and Q2 are independently C substituted with R10a and R10b respectively, wherein R10a and R10b are independently H or C1-C6 alkyl. Such compounds are hereinafter compounds of Formula IV. [00112] In embodiments, compounds of Formula I can be those where Q3 is CR21, wherein R21 is a group of the Formula,
Figure imgf000038_0001
X1 is C substituted with R11 where R11 hydrogen, halogen, cyano, nitro, - C(O)Rc’, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cydoalkyl, C3-C7 cycloalkyl- (C1 -C10)alkyl, heterocydoalkyl, aryl, or heteroaryl, wherein Rc’ is -C1-C6 alkyl, -ORC”, or -N(RCN)2, wherein RC” is -H, C1-C10 alkyl, C1-C10 haloalkyl, C3-C7 cydoalkyl, heterocydoalkyl, aryl, or heteroaryl; each RCN is independently -H, -C1-C10 alkyl, -C1-C10 -haloalkyl, -C3-C7 cycloalkyl, - heterocycloalkyl, -C1-C6 acyl, -aryl, or -heteroaryl. Such compounds are hereinafter compounds of Formula V. [00113] In embodiments, compounds of Formula V can be those where R11 is hydrogen, halogen, C1-C10 alkyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, aryl, or heteroaryl. [00114] In embodiments, compounds of Formula V can be those where Rn is H or C1 -C-6 alkyl. Other compounds of Formula I are those where Q3 is CR21, wherein R21 is a group of the Formula,
Figure imgf000038_0002
X1 is N. Such compounds are hereinafter compounds of Formula V. [00115] In embodiments, compounds of Formula I can be those where Q3 is CR21, wherein R21 is a group of the Formula,
Figure imgf000038_0003
X2 is C substituted with R5 and R6, wherein R5 and R6 are independently H or C1-C4 alkyl. Such compounds are hereinafter compounds of Formula VI. [00116] In embodiments, compounds of the disclosure can be those of Formula I wherein Q3 is CR21. [00117] In embodiments, compounds of the disclosure can be those of Formula I wherein R1 and R2 are independently H or C1-C4 alkyl; Q1 and Q2 are both CH; X2 is C substituted with two independently selected C1-C4 alkyl groups; and n is 1. [00118] In embodiments, compounds of the disclosure can be those having the Formula VII
Figure imgf000039_0001
VII wherein X1 and RC are as defined in Formula I; R5 and R6 are independently H or C1-C4 alkyl; R11 is H or C1-C6 alkyl; R10a and R10b are independently H or C1-C6 alkyl; R9a and R9b are independently H or C1-C6 alkyl. [00119] In embodiments, compounds of Formula VII can be those where R1 and R2 are independently H or C1-C4 alkyl; R10a and R10b are both H; and R5 and R6 are independently C1-C4 alkyl. [00120] In embodiments, compounds of Formula VII can be those where Xi is N. In embodiments, compounds of Formula VII can be those where X1 is CRC, wherein RC is hydrogen, methyl, ethyl, cyclopropyl, cyclopropylmethyl, fluoromethyl, difluoromethyl, or trifluoromethyl. In another embodiment of this aspect, the RC group derived from X1 is hydrogen, methyl, or trifluoromethyl, and the RC group derived from Y carries the definition given in connection with Formula I. [00121] In embodiments, compounds of Formula I can be those of Formula VIII,
Figure imgf000040_0001
VIII wherein RC is H, C1-C6 alkyl, trifluoromethyl, or cyclopropyl; and R1-R6, X1, and X4 carry the same definitions as for Formula I. [00122] In embodiments, compounds of Formula VIII can be those where X1 is N. In embodiments, compounds of Formula VIII can be those where Xi is CRC, wherein RC is hydrogen, methyl, ethyl, cyclopropyl, cyclopropylmethyl, fluoromethyl, difluoromethyl, or trifluoromethyl. In a further embodiment of this aspect, the RC group derived from X1 is hydrogen, methyl, or trifluoromethyl, and the RC group derived from Y carries the definition given in connection with Formula I. [00123] In embodiments, compounds of Formula I can be those of Formula IX:
Figure imgf000040_0002
IX where R11 is hydrogen or methyl, (for example, in an embodiment, R11 is hydrogen); RC is H, C1-C2 alkyl, trifluoromethyl, or cyclopropyl; and R3, R4, and X4 carry the same definitions as for Formula I. Compounds of Formula IX can include those where RC is C1-C2 alkyl, trifluoromethyl, or cyclopropyl. [00124] Other compounds of Formula I can be those where R21 is cyano, R7 is O, and Y is CRC, wherein RC is H, methyl, ethyl, trifluoromethyl, or cyclopropyl. Other compounds of Formula I can be those where, R21 is cyano; R7 is O; and Y is CRC, wherein RC is H, methyl, trifluoromethyl, or cyclopropyl. [00125] Yet other compounds of Formula I can be those where R21 is cyano, and X3 is C substituted with two groups that are independently H or C1-C6 alkyl. Compounds of Formula I can be those where R21 is cyano, and Q1 and Q2 are independently C substituted with H or C1-C6 alkyl. Yet other compounds of Formula I can be those where R21 is cyano, and X1 is C substituted with H or C1-C6 alkyl. Still other compounds of Formula I can be those where R21 is cyano, and X2 is C substituted with two groups that are independently H or C1-C4 alkyl. [00126] In embodiments, compounds of Formula I can be those of Formula X,
Figure imgf000041_0001
X wherein X1-X4, Q1, Q2, RC, and R1-R4 are as defined in Formula I. [00127] In embodiments, compounds of Formula X can be those where Q1 and Q2 are each independently hydrogen or C1-C6 alkyl. In embodiments, compounds of Formula X can be those where RC is C1-C6alkyl, C3- C7cycloalkyl, C1-C6 haloalkyl, C3-C7cycloalkyl(C1- C6)alkyl, or heterocycloalkyl. In embodiments, compounds of Formula X can be those where RC is C3-C7cycloalkyl, C1-C6 haloalkyl, heterocycloalkyl, or C3-C7cycloalkyl(C1-C6)alkyl. In embodiments, compounds of Formula X can be those where RC is C1-C3alkyl, C3- C5cycloalkyl, C3-C5cycloalkyl(C1-C3)alkyl, or C1-C2 haloalkyl. [00128] In embodiments, compounds of Formula X can be those where X1 is N. Such compounds are referred to herein as compounds of Formula XI. Additional compounds of any of Formulas l-X can include those wherein R3 is substituted with R50, and R50 is
Figure imgf000041_0002
[00129] Other compounds of any of Formulas I-X can include those wherein Q3 is CR21, wherein R21 is a group of the Formula,
Figure imgf000042_0001
R7 is O; and Y is CRC, wherein RC is -H, -CH3, ethyl, cyclopropyl, or -CF3. Other compounds of any of Formulas I-X can include those wherein Q3 is CR21, wherein R21 is a group of the Formula,
Figure imgf000042_0002
and X2 is C substituted with two groups that are independently H or C1-C6 alkyl. [00130] Other compounds of any of Formulas I-X can include those wherein Q3 is CR21, wherein R21 is a group of the Formula,
Figure imgf000042_0003
and Q1 and Q2 are independently C substituted with H or C1-C6 alkyl. Other compounds of any of Formulas I-X can include those wherein X1 is N. Other compounds of any of Formulas I-X can include those wherein X1 is CRC. [00131] Other compounds of any of Formulas l-X can include those wherein Q3 is CR21, wherein R21 is a group of the Formula,
Figure imgf000042_0004
and X1 is C substituted with H or C1-C6 alkyl. [00132] Other compounds of any of Formulas I-X can include those wherein Q3 is CR21, wherein R21 is a group of the Formula
Figure imgf000042_0005
and X2 is C substituted with two groups that are independently H or C1-C4 alkyl. [00133] Other compounds of any of Formulas I-X can include those wherein R21 is a group of the Formula,
Figure imgf000043_0001
R3 is -Z1Rz1, wherein Z1 is -O- or -NH-; and RZ1 is a saturated or unsaturated C3-C10 cycloalkyl, each of which is optionally substituted at any available position with C1-C10 alkyl, C1-C10 haloalkyl, C2-C10 alkenyl, C2- C10 alkynyl, hydroxy, carboxy, carboxamido, oxo, halo, amino, cyano, nitro, -SH, -S-(C1- C6)alkyl, -SO2-(C1-C6)alkyl, -SO2NH2, -SO2NH-(C1-C6)alkyl, -SO2NH-aryl, -SO2-aryl, -SO- (C1-C6)alkyl, -SO2-aryl, C1-C6 alkoxy, C2-C10 alkenyloxy, C2-C10 alkynyloxy, mono- or di- (C1- C10)alkylamino, -OC1-C10 alkyl-Z, -O-C(O)C1-C10 alkyl-Z, or R23; and R4 is H or halogen. [00134] Other compounds of any of Formulas I-X can include those wherein RZ1 is a saturated C5-C7 cycloalkyl. [00135] Other compounds of any of Formulas I-X can include those wherein wherein RZ1 is a unsaturated C5-C7 cycloalkyl. Other compounds of any of Formulas I-X can include those wherein X1 is N. Other compounds of any of Formulas I-X can include those wherein X1 is CRC. Other compounds of any of Formulas I-X can include those wherein X1 is CH. Other compounds of any of Formulas I-X can include those wherein R1 and R2 are independently H or C1-C4 alkyl; Q1 and Q2 are both CH; X2 is C substituted with two independently selected C1-C4 alkyl groups; and n is 1. [00136] Other compounds of any of Formulas I-X can include those of the Formula,
Figure imgf000043_0002
wherein RQ1 is H or halogen; and RQ2 is H or halogen. Other compounds of any of Formulas I-X can include those wherein R3 is -Z1-cyclohexyl which is optionally substituted at any available position with C1- C10 alkyl, C1-C10 haloalkyl, C2-C10 alkenyl, C2-C10 alkynyl, hydroxy, carboxy, carboxamido, oxo, halo, amino, cyano, nitro, -SH, -S-(C1-C6)alkyl, -SO2-(C1-C6)alkyl, -SO2NH2, -S02NH-(C1-C6)alkyl, -SO2NH-aryl, -SO2-aryl, -SO-(C1-C6)alkyl, -SO2- aryl, C1-C6 alkoxy, C2-C10 alkenyloxy, C2-C10 alkynyloxy, mono- or di-(C1- C10)alkylamino, -OC1-C10 alkyl-Z, -O-C(O)C1-C10 alkyl-Z, or R23; and R4 is H or fluoro. [00137] In some embodiments, the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000044_0001
or a pharmaceutically acceptable salt thereof, wherein administration commences within three days of onset of symptoms, within two days of onset of symptoms, or within 24-36 of onset of symptoms, and wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2, 3, or 4. In some instances, R1, R4, and R5 are each H. In some instances, R2 is NR7R8. In some instances, R3 is substituted or unsubstituted C1-6 carbonyl. In some instances, X is N. In some instances, Y is CR9. In some instances, n is 1. In some instances, R7 is H, and R8 is substituted or unsubstituted cyclo(C3-8) heteroalkyl. [00138] In some embodiments, the HSP90 inhibitor is 4-(6,6-dimethyl-4-oxo-3- trifluoromethyl-4,5,6,7-tetrahydro-indazol-1-yl)-2-(trans-4-hydroxy-cyclohexylamino)- benzamide (Compound 9):
Figure imgf000045_0001
(Compound 9), or trans-4-({2-(aminocarbonyl)-5-[6,6-dimethyl-4-oxo-3-(trifluoromethyl)-4, 5,6,7- tetrahydro-1 H-indazol-1-yl]-phenyl}amino)cyclohexyl glycinate (Compound 10):
Figure imgf000045_0002
(Compound 10), or pharmaceutically acceptable salts thereof. [00139] Synthesis and characterization data for these compounds are described in U.S. Patent No.7,358,370, which is incorporated by reference in its entirety. [00140] Examples of HSP90 inhibitors suitable for use in the methods and compositions of the disclosure include, but are not limited to, compounds listed in TABLE 1. [00141] TABLE 1: Examples of HSP90 Inhibitors useful for the methods described herein.
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
53
Figure imgf000056_0001
Figure imgf000057_0001
- 55 -
Figure imgf000058_0001

Figure imgf000059_0001

Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
- 69 -
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
- 72 -
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000078_0002
Figure imgf000078_0003
Figure imgf000078_0004
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000082_0002
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
[00142] In various embodiments, a compound, e.g., an HSP90i of the present disclosure (e.g., one according to Formula (Ia)), can be used to alter biological pathways following administration to a subject, wherein such pathway alteration can provide treatment and/or prevention of a CoV infection in a subject in need thereof. In some instances, the methods provided herein can comprise administering a compound of this disclosure to a subject, and downregulating in the subject cytokines and chemokines that serve as chemoattractant for lymphocytes and neutrophils (e.g., CCL20, CXCL1), regulators of cellular inflammation (e.g., CSF3, S100A8) and proliferation (e.g., CDC20), antigen presentation pathway- associated genes (e.g., HLA-DOA, HLA-DMB), genes functioning as solute transporters (e.g., SLC13A2, SLC26A4, SLC5A1, SLC5A5), or any combination thereof. [00143] In some embodiments, the methods provided herein can further comprise downregulating one or more cellular inflammatory pathways by administering an HSP90 inhibitor of this disclosure (e.g., one according to Formula (Ia), such pathways including IL- 17 signaling pathways, cytokine-cytokine receptor interactions, TNF-signaling pathways, Th17 cell differentiations, NOD-like receptor signaling and chemokine signaling pathways. In addition, signaling pathways associated with regulation of cell cycle and cellular senescence, and metabolic pathways such as biosynthesis of amino acids and tryptophan metabolism, can also be downregulated by HSP90 treatment as described herein. [00144] Furthermore, administration (e.g., oral administration) of an HSP90 inhibitor can result in upregulation of key cellular pathways involved in physiological functions such as cellular-adhesion, regulation of apoptosis and cellular proliferation. In such instances, such inflammatory genes can include those coding for chemokines (e.g., CXCL1, CXCL5, CXCL6, CXCL3 and CXCL2), chemokine-receptor ligands (e.g., CCL20, CX3CL1), cytokines (e.g., IL36G), interleukins (e.g., IL-19, CXCL8, IL-32), tumor necrosis factor alpha-induced protein (e.g., TNFAIP2), regulators of inflammatory responses (e.g., CSF3, S100A8, S100A9, SCGB3A1, S100A2), interferon-induced proteins (e.g., RSAD2, IFI44L, IFITM1, MX2, IFI27, ISG15), and combinations thereof. [00145] In various instances, the methods disclosed herein can further comprise modulation of gene activity in treated cells (e.g., of cells a subject and receiving treatment), wherein such genes include those regulating innate immunity (e.g., SERPINA3, BPIFA1, and PGLYRP4), cell cycle (e.g., UBE2C, CENPI, BIRC5, CCNB2, BUB1B, BUB1 and CENPF), mucosal structural proteins (e.g., MMP9 and FCGBP), blood coagulation (e.g., SERPINA3, SERPINE1 and SERPINF2), and combinations thereof. Furthermore, cellular genes whose expression can be altered by HSP90i treatment herein can encode for proteins that are associated with SARS-CoV-2 replication phases (e.g., MMP9 and SERPINE1) or those interacting with SARS-CoV-2 structural and non-structural proteins (e.g., BUB1B, BUB1, CENPF and ID1), and combinations thereof. Hence, the herein disclosed methods can comprise modulation of key biochemical pathways that are associated with viral infection, replication, and which pathways may be utilized to drive viral infection of a subject. Hence, the methods described herein can provide new treatment options for patients suffering from viral infections, particularly infections caused by a coronavirus. [00146] The synthesis and characterization data of the compounds listed herein can be conducted using methods known in the art, e.g., as described in U.S. Patent No.7,358,370, which is incorporated by reference herein in its entirety. [00147] In embodiments, the pharmaceutical composition can further comprise one or more “nutritional components.” The term “nutritional component” as used herein can refer to such as protein, a carbohydrate, vitamins, minerals and other beneficial nutrients including functional ingredients of the disclosure, that is, ingredients that can produce specific benefits to a person consuming the food. The carbohydrate can be, but is not limited to, glucose, sucrose, fructose, dextrose, tagatose, lactose, maltose, galactose, xylose, xylitol, dextrose, polydextrose, cyclodextrins, trehalose, raffinose, stachyose, fructooligosaccharide, maltodextrins, starches, pectins, gums, carrageenan, inulin, cellulose based compounds, sugar alcohols, sorbitol, mannitol, maltitol, xylitol, lactitol, isomalt, erythritol, pectins, gums, carrageenan, inulin, hydrogenated indigestible dextrins, hydrogenated starch hydrolysates, highly branched maltodextrins, starch and cellulose. [00148] Commercially available sources of nutritional proteins, carbohydrates, and the like and their specifications are known, or can be ascertained easily, by those of ordinary skill in the art of processed food Formulation. [00149] The pharmaceutical compositions that include nutritional components can be food preparations that can be, but are not limited to, “snack sized,” or “bite sized” compositions that is, smaller than what might normally be considered to be a food bar. For instance, the food bar can be indented or perforated to allow the consumer to break off smaller portions for eating, or the food “bar” can be small pieces, rather than a long, bar-shaped product. The smaller pieces can be individually coated or enrobed. They can be packaged individually or in groups. [00150] The food can include solid material that is not ground to a homogeneous mass, such as, without limitation. The food can be coated or enrobed, such as, and without limitation, with chocolate, including dark, light, milk or white chocolate, carob, yogurt, other confections, nuts or grains. The coating can be a compounded confectionary coating or a non- confectionary (e.g., sugar free) coating. The coating can be smooth or can contain solid particles or pieces. II. Kits comprising HSP90 Inhibitors [00151] Compounds and pharmaceutical compositions described herein can be provided in a kit. In one embodiment, the kit includes (a) a container that contains a composition that includes an HSP90 inhibitor, and optionally (b) informational material. The informational material can be descriptive, instructional, marketing or other material that relates to the methods described herein and/or the use of the agents for therapeutic benefit. In an embodiment, the kit also includes a second agent for treating or alleviating a symptom of a coronavirus infection in a subject. For example, the kit includes a first container that contains a composition that includes the HSP90 inhibitor, and a second container that includes the second agent. [00152] The informational material of the kits is not limited in its form. In one embodiment, the informational material can include information about production of the compound, molecular weight of the compound, concentration, date of expiration, batch or production site information, and so forth. In one embodiment, the informational material relates to methods of administering the HSP90 inhibitor, e.g., in a suitable dose, dosage form, or mode of administration (e.g., a dose, dosage form, or mode of administration described herein), to treat a subject who has a coronavirus infection. The information can be provided in a variety of formats, include printed text, computer readable material, video recording, or audio recording, or as information that provides a link or address to substantive material. [00153] In addition to the HSP90 inhibitor, the composition in the kit can include other ingredients, such as a solvent or buffer, a stabilizer, or a preservative. The antagonist can be provided in any form, e.g., liquid, dried or lyophilized form, for example substantially pure and/or sterile. When the agents are provided in a liquid solution, the liquid solution can be an aqueous solution. When the agents are provided as a dried form, reconstitution generally is by the addition of a suitable solvent. The solvent, e.g., sterile water or buffer, can optionally be provided in the kit. [00154] The kit can include one or more containers for the composition or compositions containing the agents. In some embodiments, the kit contains separate containers, dividers or compartments for the composition and informational material. For example, the composition can be contained in a bottle, vial, or syringe, and the informational material can be contained in a plastic sleeve or packet. In other embodiments, the separate elements of the kit are contained within a single, undivided container. For example, the composition is contained in a bottle, vial or syringe that has attached thereto the informational material in the form of a label. In some embodiments, the kit includes a plurality (e.g., a pack) of individual containers, each containing one or more unit dosage forms (e.g., a dosage form described herein) of the agents. The containers can include a combination unit dosage, e.g., a unit that includes both the HSP90 inhibitor and the second agent, e.g., in a desired ratio. For example, the kit includes a plurality of syringes, ampules, foil packets, blister packs, or medical devices, e.g., each containing a single combination unit dose. The containers of the kits can be air-tight, waterproof (e.g., impermeable to changes in moisture or evaporation), and/or light-tight. The kit optionally includes a device suitable for administration of the composition, e.g., a syringe or other suitable delivery device. The device can be provided pre-loaded with one or both of the agents or can be empty, but suitable for loading. III. Experimental Procedures [00155] Cell culture. Fully differentiated human tracheobronchial epithelial (TBE) cells (EpiAirwayTM) from 3 independent donors with no reported respiratory disease or smoking history can be obtained from MatTek (Ashland, MA). The cells can be cultured at the air- liquid interface in 1 ml of AIR-100-MM culture medium (MatTek) in 6 well plates at 37 °C in 5% CO2. Upon receipt of cells, the cultures can be acclimated for 16-24 hr prior to the start of experiments. Vero E6 cells (ATCC) can be cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 1X Penicillin/Streptomycin (Gibco), and 1X non-essential amino acid (NEAA) mixture (Gibco) and maintained at 37 °C in 5% CO2. Calu-3 cells (ATCC) can be cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 20% fetal bovine serum (FBS), 25 mM HEPES, 1X Penicillin/ Streptomycin (Gibco), and maintained at 37 °C in 5% CO2. [00156] Compounds. The small molecule inhibitors of HSP90 of the present disclosure, e.g., SNX-5422, can be synthesized using methods known in the art and characterized by proton nuclear magnetic resonance (NMR) and liquid chromatography/mass-spectrometry (LC/MS). The compound can then be solubilized in 100% DMSO to a 10 mM stock concentration. Additional compounds herein, such as Remdesivir (MedChemExpress LLC, USA), can be reconstituted in 100% DMSO to a concentration of 10mM. [00157] SARS-CoV-2 propagation and titering. SARS-CoV-2 USA-WA1/2020 (BEI Resources; NR-52281) can be propagated on Vero E6 cells at a multiplicity of infection (MOI) of 0.001 in virus diluent (DMEM supplemented with 2% FBS, 1X Penicillin/ Streptomycin (Gibco), 1 mM sodium pyruvate (Gibco) and 1X NEAA (Gibco)) at 37 °C in 5% CO2. At day 4 post-infection (pi), cell supernatant containing the released virus can be harvested, spun at 1500 rpm for 5 minutes, filtered through a 0.22µM sterile vacuum filtration system, aliquoted and stored at -80°C until further use. Stock viral titer can be determined herein by plaque assay. Essentially, 0.72 X 106 Vero E6 cells can be seeded in 6 well plates. The stock virus can be serially diluted and incubated on cellular monolayer at 37 °C in 5% CO2. After 1 hr, virus can be aspirated, and cells can be overlayed with carboxy- methyl cellulose (CMC) containing media (0.6% CMC, MEM supplemented with 1X Penicillin/Streptomycin (Gibco), 2% FBS, 1mM sodium pyruvate (Gibco), 1X NEAA (Gibco), 0.3% sodium bicarbonate (Gibco), and 1X GlutaMAX (Gibco). After 4 days of incubation at 37 °C in 5% CO2, plaque assays stained with 1% crystal violet in 10% neutral buffered formalin (NBF), and plaque forming unit/mL (Pfu/mL) can be determined. [00158] SARS-CoV-2 infection and treatment. 1.4 X 105 Vero E6 or Calu-3 cells can be seeded in 24 well plates. After 24 hrs, cells can be incubated with the SARS-CoV-2 isolate at an MOI of 0.01-0.02 at 37 °C and 5% CO2, with intermittent plate rocking. After 1 hr, the virus can be aspirated, cells can be rinsed twice with 1X PBS and fresh maintenance medium containing dilutions of HSP90 inhibitor, e.g., SNX-5422, or compounds such as Remdesivir, or 0.1% DMSO as a control can be added. The cells can then be incubated for 48 hrs at 37 °C and 5% CO2. [00159] Estimation of cellular cytotoxicity using flow cytometry. Suspensions of Vero E6 and Calu-3 cells can be stained with LIVE/DEAD Fixable Aqua Dead Cell Stain kit (Thermo Fisher) according to manufacturer’s instructions. The stained cells can be fixed using methanol-free 4% formaldehyde (Thermo Fisher) for 30 minutes and data can be acquired on an LSRII flow cytometer (BD 441 Biosciences) using BD FACS Diva software and analyzed with FlowJo software version 10.1 (Tree Star, Inc). LIVE/DEAD marker negative viable cells can be selected from total cells after sequential selection of forward scatter and side scatter singlets. [00160] Infectious viral titer of supernatant. Cellular supernatant can be collected from infected and drug-treated cells, 48 hrs after infection. The supernatant can be clarified by, e.g., spinning at 1500 rpm for 5 minutes and infectious viral titer can then be measured by plaque assay as described herein. [00161] qRT-PCR for detecting cell-free viral RNA. SARS-CoV-2 RNA from cell supernatant can be extracted using the QIAamp viral RNA mini kit (Qiagen). A two-step qRT-PCR can be used to detect viral RNA released in the cell supernatant. In the first step, viral cDNA for the nucleocapsid (N) gene was generated using SuperScript III Reverse Transcriptase (Invitrogen) and an N-reverse primer, following manufacturer’s instructions. In the second step, 7 μL cDNA from step-1 can be amplified using an N gene forward primer, an N gene reverse primer and a probe using Taqman mastermix (Thermo Fisher) following manufacturer’s instructions. The thermal cycling steps can be: 50 °C for 2 min, 95 °C for 10 min, and 40 cycles of 95 °C for 15 s and 60 °C for 1 min, and qPCR can be performed on a Step-One-Plus real time PCR machine (Applied Biosystems) using the StepOne Software v2.3. Viral RNA copy number/mL supernatant can be assessed using pCDNA3.1(+)-N-eGFP plasmid (GenScript) as standard. [00162] Immunofluorescence assay for detection of SARS-CoV-2 N protein.1.4 X 105 Vero E6 cells can be seeded in 4-well chamber slides (Corning). After 24 hrs, cells can be incubated with SARS CoV-2 USA-WA1/2020 strain at an MOI of 0.01 at 37 °C and 5% CO2, with intermittent rocking. After 1 hr, the virus can be aspirated, cells can then be rinsed twice with 1X PBS and fresh maintenance medium containing dilutions of HSP90i (e.g., SNX- 5422), or Remdesivir, or 0.1% DMSO can be added. After 48 hrs, cells can be fixed by submerging the chamber slides in 10% NBF for 2 hrs. Fixed cells can then be washed three times with DPBS (Sigma) + 0.02% Triton X-100. For immunostaining, cells can be permeabilized with DPBS+ 0.1% Triton-X-100 for 20 minutes and blocked in DPBS supplemented with 5% BSA, 0.02% Tween-20 and 10% donkey sera (Sigma) for 1 hr. The cells can then be incubated with anti-SARS-CoV-2 N protein antibody (Sinobiological; 1:100) for 2 hrs at room temperature. After subsequent washing of the samples, the cells can be treated with Alexa Fluor 488 donkey anti-rabbit secondary antibody (Invitrogen; 1:1000) for 1 hr at room temperature. After further washing, the coverglasses can be mounted on glass slides with ProLong Glass Antifade with NucBlue (Invitrogen) and sealed. Coverglasses can be allowed to cure overnight prior to imaging using Axio Imager fluorescent microscope (Carl Zeiss). Images can be captured with 20X objective, processed using Zeiss Zen Black software and proportion of SARS-CoV-2-infected cells can be counted using Fiji, utilizing the analyze particle function. [00163] CC50 and IC50 calculations. The 50% cytotoxic concentration (CC50) and 50% inhibitory concentration (IC50) values herein can be calculated, e.g., using Graph Pad Prism using curve fitting procedures. [00164] Drug treatment and bulk RNA-sequencing of human TBE cells. Human tracheobronchial epithelial (TBE) cells from 3 independent donors can be treated with 1 µM SNX-5422 or 0.1% DMSO added to the media on the basolateral side of the culture, in three biological replicates (see, e.g., FIG.3A). After 48 hrs, cells can be resuspended in TRIzol reagent (Thermo Fisher) and total RNA from the cells can be extracted by phase separation with chloroform and subsequently using the RNeasy Mini Kit (Qiagen). RNA-Seq libraries can be prepared using TruSeq RNA library Prep Kit v2 (Illumina, Inc. USA). Before pooling and sequencing, fragment length distribution and library quality can be assessed on a TapeStation 2200 (Agilent Technologies), and the libraries can be validated by Qubit Fluorometers (Thermo Fisher). All libraries can then be pooled in a concentration at 4 nM and sequenced on a NextSeq 500 Illumina sequencing platform system using NextSeq 500/550 High Output Kit v2.0 (150 cycles) (Illumina, Inc. USA). [00165] Analysis of the bulk RNA-seq data. RNA-Seq data can be quality checked with FastQC (Andrews, 2010) and preprocessing can be carried out using TrimGalore (Krueger) toolkit to trim low-quality bases and Illumina adapter sequences using default settings. Reads can be aligned to the ENSEMBL Homo_sapiens.GRCh38.dna.primary_assembly genome using the ENSEMBL Homo_sapiens.GRCh38.100 transcript (Kersey et al., 2012) annotation file with STAR splice-aware RNA-seq alignment tool in paired mode allowing maximum multimapping of 3. Gene level counts can be quantified using FeatureCounts tool, counting unique features in non-stranded mode and retaining both gene ID, gene name, and gene biotype mapping from the ENSEMBL annotation file. Prior to differential expression analysis, count data can be collapsed to donor level and genes for which mean raw count with at least 15 can be kept. Normalization and differential expression can be carried out with DESeq2 Bioconductor package, utilizing the ‘apeglm’ Bioconductor package for log fold change shrinkage, in R statistical programming environment. The design Formula can be constructed to test for the effect of treatment while controlling for donor. Principal component analysis (PCA) can be performed with plotPCA in ‘DESeq2’ R package. PCA can be performed on variance stabilizing transformed (vst) count data and batch corrected with ‘limma’ package for genes with an average raw count of at least 15 across samples. Volcano plot with differentially expressed genes can be generated using ‘EnhancedVolcano’ package in R, after filtering for described differential expression cutoffs. Heatmaps for differential expression of genes can be generated using ‘pheatmap’ package in R. A vst can be applied to count data, and batch corrected with ‘limma’ package, followed by Z-score normalization. Dot plots demonstrating upregulated and downregulated KEGG pathways can be generated using ‘ClusterProfiler’ package in R using a “universe” of all human genes. Protein-protein interaction 520 (PPI) network can be constructed using STRING and Cytoscape (vs 3.8.2). EXAMPLES [00166] Examples are provided below to facilitate a more complete understanding of the disclosure. The following examples illustrate the exemplary modes of making and practicing the disclosure. However, the scope of the disclosure is not limited to specific embodiments disclosed in these Examples, which are for purposes of illustration only, since alternative methods can be utilized to obtain similar results. EXAMPLE 1 HSP90 Inhibition Attenuates Dose-dependently SARS-CoV-2 Replication in Vitro [00167] This example describes an in vitro experiment demonstrating that SNX5422 (Compound 10) inhibits SARS-CoV-2 replication. [00168] African green monkey kidney epithelial Vero-E6 cells were infected with 0.3 multiplicity of infection (MOI) of SARS-CoV-2 isolate USA-WA1/2020 (BEI Resources). Infected Vero-E6 cells were then treated with 10 µM, 1 µM and 0.1 µM concentrations of SNX5422 for 48 hrs. Cell supernatant was collected, viral RNA was extracted from the supernatant and qRT-PCR was performed on the viral nucleocapsid (N) gene. [00169] The data indicate that supernatant from cells treated with increasing concentrations of SNX5422 required higher number of PCR cycles to get amplified (FIG.1A), indicating that SNX5422 blocked SAR-CoV-2 replication in a dose-dependent manner. [00170] Furthermore, FIG.1B is a graph showing the cell-free SARS-CoV-2 viral load. Vero-E6 cells were infected with SARS-CoV-2-USA-WA-1 strain at an MOI of 0.01 for 1 hr. The infected cells were treated with SNX5422 at 1 hr post infection at indicated concentrations and incubated for 48 hrs. Viral RNA (vRNA) was extracted from cell supernatant and viral RNA copies were monitored using RT-qPCR. Each symbol represents a single experimental replicate. FIG.1B demontrates that HSP90i, e.g., SNX5422, significantly reduced replication of SARS-CoV-2 viral material. [00171] Additional experiments were conducted in which Vero E6 and Calu-3 cells were infected with SARS-CoV-2 strain USA-WA1/2020 and treated with either 0.1% DMSO (drug-vehicle) or 0.31-2.5 µM SNX5422, or 5 µM remdesivir (RDV). Both SNX5422 and RDV were reconstituted in DMSO. After 48 hr post-infection (pi), FIG.1C shows that Vero cells were immuno-stained for SARS-CoV-2 nucleocapsid protein (NP) (green). Nuclei was counterstained (blue) prior to imaging. The data incidate that SNX5422 attenuated intracellular NP expression and proportion of NP+ cells as shown in FIG.1D, illustrating that immuno-fluorescent images were quantified to evaluate proportions of SARS- CoV-2-NP+ cells. The 50% inhibitory concentration (IC50) was calculated to 2.3 μM for SNX5422. Each symbol and color represent an independent experiment and each data point represents an independently quantified image field. Medians of data points were reported as grey bars. Dotted line represents detection cut off, which is the mean+3 standard deviation (SD) of the percentage of NP+ cells in uninfected controls. Subsequently, cell-free viral RNA in the supernatant of infected and treated Vero-E6 (FIG.1E) and Calu-3 (FIG.1F) cells were measured by qRT-PCR of the viral N gene. The infectious viral titers of the supernatant of Vero E6 (FIG.1G) and Calu-3 (FIG.1H) cells were determined by plaque assay. Furthermore, Vero E6 (FIG.1I) and Calu-3 (FIG.1J) cells were treated with either 0.1% DMSO (drug-vehicle) or 0.01-100 µM SNX5422, or 5 µM RDV. After 48 hours, proportions of viable cells were evaluated by flow cytometry. Each symbol and color represents an independent experiment and medians of the data points are reported as grey bars. [00172] The data demonstrate that SNX-5422 treatment resulted in a reduction of cell-free viral genomic copies with an IC50 of 0.2 μM in both cell types. Additionally, SNX-5422- treatment reduced cell-free infectious viral titers, with an IC50 of 0.38 μM in Vero E6 cells and 0.4 μM in Calu-3 cells, respectively. [00173] Subsequently, uninfected Vero E6 and Calu-3 cells were treated with increasing concentrations of SNX-5422 and the frequency of live cells was monitored by flow cytometry. The 50% cellular cytotoxicity (CC50) of SNX-5422 was calculated to 91.97μM and >100 μM in Vero E6 and Calu-3 cells, respectively, highlighting the minimal cytotoxic effects of the drug on these two cell types. Consequently, the selectivity index (SI=CC50/IC50) of SNX-5422 in suppressing viral replication was evaluated to be high in both cell types as shown in TABLE 2, summarizing the cell-specific potency and selectivity of HSP90 inhibition using SNX5422. TABLE 2 – Cell-specific SNX-5422 Potency and Selectivity
Figure imgf000101_0001
aCC50: 50% cytotoxic concentration bIC50: 50% inhibitory concentration cSI: Selectivity index=CC50/IC50 [00174] Collectively, these data suggest that treatment with an oral inhibitor of HSP90, e.g., SNX5422, upon SARS-CoV-2 exposure, can potently attenuate viral replication, at least in vitro, at a high selectivity index. EXAMPLE 2 Cytotoxicity Assessment of HSP90i Treatment on SARS-CoV-2 infected Vero-E6 cells [00175] This example demonstrates the cytotoxicity of HSP90 inhibiors, e.g., SNX5422 (Compound 10), on SARS-CoV-2 infected Vero-E6 cells. [00176] To assess the cytotoxicity of SNX5422 (Compound 10) on Vero-E6 cells, and compute the 50% cellular cytotoxic dose (CC50), Vero-E6 cells were incubated with increasing concentrations of SNX5422 for 48 hrs and cellular viability was measured using the Cell-Titer Glo Cell Viability Assay (Promega). The Reed and Muench method was subsequently employed and the CC50 of SNX5422 calculated to 67.17µM. [00177] FIG.2 shows a graph of the cytotoxicity of SNX5422 in Vero-E6 cells as a function of inhibitor concentration. Each dot represents a single experimental replicate. The inhibitor concentrations at 50% cytotoxicity (CC50 = 67.17 μM) and 20% cytotoxicity (CC20 = 13.97 μM) were also calculated. Of note, the SARS-CoV-2 inhibitory concentration of 10 µM as identified in FIG.2 was less than the estimated CC20 of the HSP90 inhibitor SNX5422. [00178] These data demonstrate the cytotoxic potency of HSP90 inhibiors, e.g., SNX5422 (Compound 10), on SARS-CoV-2 infected Vero-E6 cells, and their potential utlility in the treatment of CoV infections in a subject. EXAMPLE 3 SARS-CoV-2 Host Protein Expression is Altered by HSP90 Inhibitors [00179] This example demonstrates that HSP90 inhibiors, e.g., SNX5422 (Compound 10), can significantly downregulate host production of viral proteins in infected cells. [00180] To that end, FIG.3A shows a schematic diagram illustrating treatment of human tracheobronchial epithelial (TBE) cells. TBE cells from 3 independent healthy donors were cultured in air-liquid interface and treated with either 0.1% DMSO (drug-vehicle) or 1 µM SNX-5422 reconstituted in DMSO, for 48 hrs. RNA was then extracted from the cells and bulk RNA sequencing was performed. FIG.3B shows a Volcano plot demonstrating protein coding genes altered upon SNX-5422 treatment of human TBE cells. Red (left, indicated by arrow #1), blue (right, indicated by arrow #2), and grey (middle) regions of the plot indicate downregulated, upregulated and non-differentially expressed genes, respectively. Differentially expressed genes (DEGs) were defined as protein coding genes with a Log2 fold change >2 between the drug-treated and control groups and adjusted p value <0.001. Open symbols represent genes with Log2 fold change >6 between the drug-treated and control groups. Cellular pathway overrepresentation analysis was performed with DEGs with a normalized average read count across donors and experimental conditions ≥300. FIG.3C shows downregulated and FIG.3D upregulated Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways with an adjusted p value of < 0.05. Dot size represented gene ratio and color schema represents adjusted p values. [00181] In sum, fully differentiated human TBE cells from three independent donors without known respiratory disease or smoking history were cultured ex vivo at the air-liquid interface. The basolateral side of the cell culture was treated with either 0.1% DMSO (drug-vehicle) or 1μM SNX-5422 for 48 hrs, followed by bulk RNA sequencing of the total cellular RNA (FIG.3A). Principal component analysis of the read counts revealed that treatment with SNX-5422 formed distinct clusters compared to the control group, highlighting differential expression of cellular genes upon drug treatment. Differentially expressed genes (DEGs) between the control group and the SNX-5422-treated group were defined as protein coding genes with a Log2 fold change (FC) ratio > 2 and adjusted p value ≤ 0.001. A volcano plot of the protein coding genes indicated that 1361 genes were differentially expressed between the two groups, with 470 cellular genes being upregulated and 891 cellular genes being downregulated upon SNX-5422 treatment (FIG.3B). To identify cellular genes whose expression was highly regulated upon SNX-5422 treatment of TBE cells, collated DEGs with a Log2 FC >6, for which the average normalized read count across donors and experimental conditions was ≥300 was utilized. Using this stringent threshold, 16 cellular genes that were downregulated and 4 cellular genes that were upregulated upon drug treatment (FIG.3B) were indentified. Further categorization of those genes based on functions revealed that SNX- 5422-treatment of TBE cells downregulated cytokines and chemokines that serve as chemoattractant for lymphocytes and neutrophils (CCL20, CXCL1), regulators of cellular inflammation (CSF3, S100A8) and proliferation (CDC20), antigen presentation pathway- associated genes (HLA-DOA, HLA-DMB) and genes functioning as solute transporters (SLC13A2, SLC26A4, SLC5A1, SLC5A5) (FIG.3B). [00182] Next, the cellular signaling pathways that were significantly altered upon SNX5422 treatment of human TBE cells were mapped, by performing a Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway overrepresentation analysis on the DEGs (FIG.3C and FIG. 3D). Notably, cellular inflammatory pathways such as IL-17 signaling pathway, cytokine- cytokine receptor interaction, TNF-signaling pathway, Th17 cell differentiation, NOD-like receptor signaling and chemokine signaling pathways were among the top 20 identified pathways that were downregulated by SNX5422 treatment. In addition, signaling pathways associated with regulation of cell cycle and cellular senescence, and metabolic pathways such as biosynthesis of amino acids and tryptophan metabolism, were also downregulated by SNX5422 (FIG.3C). Furthermore, treatment with this oral HSP90 inhibitor resulted in upregulation of key cellular pathways involved in physiological functions such as cellular- adhesion, regulation of apoptosis and cellular proliferation (FIG.3D). [00183] Taken together, these data suggest that HSP90 inhibition, e.g., using the compound SNX5422, can alter biological pathways in a therapeutic and/or preventative manner in subjects suffering from a CoV infection. EXAMPLE 4 HSP90 Inhibition Suppresses Inflammatory Pathways associated with SARS-CoV-2 Replication [00184] This example demonstrates that HSP90 inhibition, e.g., using SNX5422, can suppress inflammatory pathways associated with SARS-CoV-2 replication. [00185] To that end, and based on the results presented in EXAMPLE 3, a gene set was selected whose expression was stringently regulated upon SNX-5422 treatment of human TBE cells (Log2 FC >3, total 287 genes). This set included genes and pathways reported in ex vivo SARS-CoV-2-infected primary airway epithelial cells or those identified in blood, bronchioalveolar lavage fluid (BALF) and lung biopsies of SARS-CoV-2 positive patients. A total of 55 genes were identified that were regulated by SNX-5422, and which have been associated with SARS-CoV-2 pathogenesis and disease progression. A protein-protein interaction (PPI) network and functional enrichment analysis mapped several of these 55 genes to cellular inflammatory pathways including IL-17 signaling pathway, TNF signaling pathway, cytokine-cytokine receptor interaction and chemokine signaling pathway. SNX- 5422 dampened expression of cellular genes associated with SARS CoV-2 -mediated proinflammatory response and hyper-cytokinemia (FIG.4A). Specifically, these inflammatory genes could be broadly categorized into chemokines (e.g., CXCL1, CXCL5, CXCL6, CXCL3 and CXCL2), chemokine-receptor ligands (e.g., CCL20, CX3CL1), cytokines (e.g., IL36G), interleukins (e.g., IL-19, CXCL8, IL-32), tumor necrosis factor alpha-induced protein (e.g., TNFAIP2), regulators of inflammatory responses (e.g., CSF3, S100A8, S100A9, SCGB3A1, S100A2) and interferon-induced proteins (e.g., RSAD2, IFI44L, IFITM1, MX2, IFI27, ISG15). [00186] Collectively, these results suggest that early treatment with SNX-5422 may mitigate SARS-CoV-2-mediated hyperinflammation in a subject in need thereof, thereby improving clinical outcomes of COVID-19. [00187] Furthermore, it was investigated whether SNX-5422 treatment can regulate expression of identified SARS-CoV-2 host factors and cellular mechanisms imbalanced upon infection. [00188] The data indicate that of the 55 genes altered by SNX-5422 treatment in the context of SARS-CoV-2 replication, 27 genes had an imbalanced cellular expression, in virally infected cells (FIG.4B). Further categorization of these 55 genes revealed that the majority of these genes fall into 4 functional categories,- namely, regulators of innate immunity (e.g., SERPINA3, BPIFA1, and PGLYRP4), regulators of cell cycle (e.g., UBE2C, CENPI, BIRC5, CCNB2, BUB1B, BUB1 and CENPF), mucosal structural proteins (e.g., MMP9 and FCGBP) and regulators of blood coagulation (e.g., SERPINA3, SERPINE1 and SERPINF2). Furthermore, cellular genes whose expression was impacted by SNX-5422 treatment, encoded for proteins that are associated with SARS-CoV-2 replication phases (e.g., MMP9 and SERPINE1) or those interacting with SARS-CoV-2 structural and non-structural proteins (e.g., BUB1B, BUB1, CENPF and ID1). [00189] Taken together, these data demonstrate that SNX-5422 treatment interrupted cellular pathways and genes associated with replication of SARS-CoV-2, and hence HSP90 inhibition may be a new and effective treatment approach to prevent SARS-CoV-2 infection and/or mitigate COVID-19 symptons in a subject (e.g., a human or a rodent). EXAMPLE 5 Pharmokinetics and Pharmacodynamics of HSP90 Inhibition to Treat SARS-CoV-2 Infections [00190] This example demonstrates pharmacokinetic and pharmacodynamic evaluation of HSP90 inhibition to treat CoV infections in subjects in need thereof. [00191] The data provided in EXAMPLES 1-4 herein demonstrate that the orally bioavailable HSP90 inhibitor SNX5422 can suppress SARS-CoV-2 replication in vitro at non-cytotoxic doses. Based on these results, it can be assumed that the orally bioavailable HSP90 inhibitor SNX5422 can be well tolerated in SARS-CoV-2-infected subjects (e.g., patients or rhesus macaques (RMs)), with no major side effects, and may be associated with lower viral loads, dampened lung inflammatory response and better disease outcomes. [00192] To assess whether a single dose of SNX5422 can provide detectable plasma levels, SNX5422 was orally administered in 5 mg/kg doses to 2 adult and to 2 infant (4 months of age) RMs. The plasma levels of SNX5422 at multiple time points were then measured, using a liquid chromatography-tandem mass spectrometry (LC-MS) (FIG.5). Additionally, serum biochemistry and complete blood count (CBC) of these macaques indicated that a 5mg/kg single oral dose of the drug was well tolerated in both adult and infant RMs. [00193] Next, safety and tolerability of orally administered SNX5422 is investigated. To that end, an HSP90 inhibitor (abbreviated as “HSP90i”) at a dose of 21mg/kg is administered, which is less than the maximum tolerable dose (MTD) reported in humans, three times a week for 2 weeks. To evaluate if a short-term repeated treatment of HSP90i results in any severe adverse side effects in RMs, a serum biochemistry panel, CBC and a flow-based evaluation of CD3+, CD4+, CD8+ and CD20+ cells are performed on a weekly basis. These analyses can indicate the impact of HSP90i on hematologic, immunologic and metabolic functions, as well as liver and kidney functions. Furthermore, at necropsy, tissues from multiple organs such as kidney, lung, liver, gut, lymph nodes, spleen, brain, and eyeball, are harvested, fixed and paraffin embedded. To assess morphometric and histopathological changes of different organs upon HSP90i treatment, tissue sections are stained with hematoxylin and eosin (H&E) stain and Masson Trichrome (MT) stain, respectively. [00194] In a subsequent step, HSP90i treatment of a SARS-CoV-2 infected RM model is investigated. To that end, 12 SIV and STLV negative Indian origin RM from the California National Primate Research Center (CNPRC) breeding colony, between ages 3 and 12 are selected. Animals are brought into BLS3 housing and acclimated prior to inoculation. SARS- CoV-2 isolates USA-WA1/2020 (BEI Resources) are produced and expanded in BSL3 facility. Animals are inoculated mucosally. Approximately 6 x 106 TCID50 of total virus dose are instilled into the conjunctiva, nostrils, and trachea of anesthetized monkeys in 5 ml of 0.9% sterile saline (trachea (4 ml), nares (0.5 ml) and conjunctivae (0.5 ml) to recapitulate relevant transmission routes of COVID-19 using established protocols. After 3 days of infection, an HSP90 inibitor, e.g., SNX5422, at a dosage optimized by our PK study described above are administered in 6 RMs three times a week by oral intubation for 27 days as outlined in FIG.6. [00195] Physiologic parameters (body temperature, weight, and activity) are monitored prior to, and after, animals are infected. CBCs and serum chemistry are obtained on all blood samples to monitor host responses and organ function after infection. The sampling schedule is designed to comprehensively characterize viral shedding and cytokine responses. Intensive sampling during the first week of infection can enable the study of the acute virology and host responses in the lung and systemic compartments. Animals are necropsied 30 days post- infection to perform histologic examination of the tissues. Nasal and tracheal washes are analyzed for SARS CoV-2 RNA levels and infectious virus every other day for the first 2 weeks after infection. Since the primary receptor necessary for entry of SARS-CoV-2 into a cell, ACE2, is expressed in the gastrointestinal and genitourinary tracts of rhesus and humans, and virus shedding in saliva, urine, and stool samples is also analyzed. Tissue samples are homogenized in DMEM, supernatants are removed and immediately used for RNA extraction and cDNA synthesis. Taqman PCR to amplify a segment of SARS-CoV-2 nucleoprotein (N) gene sequence is conducted. In addition, in order to differentiate input virus from replicating virus in respiratory samples, levels of sub-genomic messenger RNA of the E gene that is not packaged into the viral particle are also assessed. Finally, levels of infectious virus via plaque forming unit assays by culturing dilutions of the respiratory and GI samples in Vero E6 monolayers is analyzed, followed by methylcellulose overlay and crystal violet staining. At necropsy, (FIG.6) all relevant tissues are collected including the salivary glands, lungs, mediastinal lymph nodes, kidney, and gut tissue among others to evaluate virus localization and immune responses by RT-PCR, quantitative molecular histology and flow cytometry. Tissues are evaluated for gross pathology, histopathology, and tissue vRNA levels by qRT- PCR. Lung tissue and PBMCs from NHPs are collected at necropsy. qRT-PCR and flow cytometry on epithelial cells and immune cells from lung and PBMC in treated and untreated macaques are conducted to validate downregulation of SARS-CoV-2 host proteins and inflammatory genes. Furthermore, immune analysis in respiratory secretions and plasma are analyzed to assess cytokine production. Serum and respiratory washes are collected and assayed for cytokines by bead array assay on a Luminex platform that is in BSL3 at the CNPRC. A bead array assay is performed to determine cytokine and chemokine levels including IL-12, IL-1β, IL-8, IL-2; MIP1α, MIP1β, MIP3α, MIP3β; MCP-1; CXCR3 ligands; TNFα; IFNs such as IFN-α,-β, or -γ and Th17 responses (IL-17, IL-6, TGF-ß, IL-21, IL-23 and the CCR6 ligand CCL20). [00196] To end the currently ongoing SARS-CoV-2 pandemic and improve global preparedness for future CoV outbreaks, a broad-spectrum easy-to-administer therapeutic modality that can suppress SARS-CoV-2 replication is urgently needed. Blocking host factors that facilitate viral replication and dampening virus-mediated lung inflammation can be crucial for achieving better SARS-CoV-2 disease outcomes. The safety, tolerability, efficacy and mechanism as determined herein of HSP90 inhibitors as potent and orally administered host-directed SARS-CoV-2 therapeutics can meet such immediate unmet need for treating early SARS-CoV-2 infection and to limit virus replication, prevent severe disease, and reduce transmissions.

Claims

CLAIMS What is claimed is: 1. A method for treating a coronavirus infection in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000108_0001
or a pharmaceutically acceptable salt thereof, wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2, 3, or 4.
2. A method for preventing or reducing the incidence of a coronavirus infection in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000109_0001
or a pharmaceutically acceptable salt thereof, wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2, 3, or 4. 3. A method of reducing the effects and/or symptoms of a coronavirus infection in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000110_0001
or a pharmaceutically acceptable salt thereof, wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2,
3, or 4.
4. A method of treating a subject having a respiratory tract infection caused by a coronavirus, the method comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000111_0001
or a pharmaceutically acceptable salt thereof, wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2, 3, or 4.
5. A treatment regimen for treating a coronavirus infection in a subject, the treatment regimen comprising administering to the subject a therapeutically effective amount of an HSP90 inhibitor, wherein the HSP90 inhibitor is a compound according to Formula (Ia):
Figure imgf000112_0001
or a pharmaceutically acceptable salt thereof, wherein administration commences within three days of onset of symptoms, within two days of onset of symptoms, or within 24-36 of onset of symptoms, and wherein, R1, R2, R3, R4, and R5 are each independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C1-6 heteroalkyl, substituted or unsubstituted C1-6 alkoxy, substituted or unsubstituted C1-6 thioalkoxy, substituted or unsubstituted C1-6 carbonyl, substituted or unsubstituted C1-6 carboxyl, -NR7R8, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R6 is O, S, NH, N-OH, N-NH2, N-NHR22, or N-NH-(C1-C6 alkyl); R7 and R8 are independently H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted cyclo(C3-8) alkyl, substituted or unsubstituted cyclo(C3-8) heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; X is N or CH; Y is N or CR9, wherein R9 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C1-C10 haloalkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C10)alkyl, heterocycloalkyl, aryl, or heteroaryl; and n is 0, 1, 2, 3, or 4.
6. The method of any one of claims 1-5, wherein R1, R4, and R5 are each H.
7. The method of any one of claims 1-6, wherein R2 is NR7R8.
8. The method of any one of claims 1-7, wherein R3 is substituted or unsubstituted C1-6 carbonyl.
9. The method of any one of claims 1-8, wherein X is N.
10. The method of any one of claims 1-9, wherein Y is CR9.
11. The method of any one of claims 1-10, wherein n is 1.
12. The method of any one of claims 1-11, wherein R7 is H, and R8 is substituted or unsubstituted cyclo(C3-8) heteroalkyl.
13. The method of any one of claims 1-12, wherein the HSP90 inhibitor is:
Figure imgf000113_0001
or a pharmaceutically acceptable salt thereof.
14. The method of any one of claims 1-11, wherein the HSP90 inhibitor is:
Figure imgf000113_0002
, or a pharmaceutically acceptable salt thereof.
15. The method of any one of claims 1-14, wherein the coronavirus is a human coronavirus or a zoonotic coronavirus.
16. The method of any one of claims 1-15, wherein the coronavirus comprises SARS- CoV, SARS-CoV-2, MERS-CoV, or a virus from the Coronaviridae family, Orthocoronavirinea subfamily.
17. The method of any one of claims 1-16, wherein the coronavirus comprises SARS- CoV-2.
18. The method of any one of claims 1-17, further comprising administering a therapeutically effective amount of at least one additional active agent.
19. The method of claim 18, wherein the at least one additional active agent comprises an anti-viral agent, an anti-inflammatory agent, a pain reliever, a steroid, or a combination thereof.
20. The method of any one of claims 1-19, wherein the therapeutically effective amount of the HSP90 inhibitor comprises a dosage amount of between about 0.5 mg to about 7g.
21. The method of any one of claims 1-20, wherein the HSP90 inhibitor is administered orally or intranasally.
22. The method of any one of claims 1-21, wherein the HSP90 inhibitor is administered as a pharmaceutical composition comprising a pharmaceutically acceptable carrier, excipient or diluent.
PCT/US2021/039146 2020-06-26 2021-06-25 Methods of treating coronavirus infection using hsp90 inhibitors WO2021263139A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063044830P 2020-06-26 2020-06-26
US63/044,830 2020-06-26

Publications (1)

Publication Number Publication Date
WO2021263139A1 true WO2021263139A1 (en) 2021-12-30

Family

ID=79281937

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/039146 WO2021263139A1 (en) 2020-06-26 2021-06-25 Methods of treating coronavirus infection using hsp90 inhibitors

Country Status (1)

Country Link
WO (1) WO2021263139A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090156473A1 (en) * 2006-06-01 2009-06-18 Virologik Gmbh Pharmaceutical composition for the treatment of viral infections and/or tumor diseases by inhibiting protein folding and protein breakdown
US20160143884A1 (en) * 2014-11-26 2016-05-26 Esanex, Inc. Use of tetrahydroindazolylbenzamide and tetrahydroindolylbenzamide derivatives for the treatment of human immunodeficiency virus (hiv) and acquired immune deficiency syndrome (aids)

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090156473A1 (en) * 2006-06-01 2009-06-18 Virologik Gmbh Pharmaceutical composition for the treatment of viral infections and/or tumor diseases by inhibiting protein folding and protein breakdown
US20160143884A1 (en) * 2014-11-26 2016-05-26 Esanex, Inc. Use of tetrahydroindazolylbenzamide and tetrahydroindolylbenzamide derivatives for the treatment of human immunodeficiency virus (hiv) and acquired immune deficiency syndrome (aids)

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LI Y-H,ET AL: "Geldanamycin, a ligand of heat shock protein 90, inhibits the replication of herpes simplex virus type 1 in vitro.", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 48, no. 3, 1 March 2004 (2004-03-01), US , pages 867 - 872, XP002343814, ISSN: 0066-4804, DOI: 10.1128/AAC.48.3.867-872.2004 *
SILVA FLAVIA RODRIGUES DA, GUERREIRO RENATO DE CARVALHO, ANDRADE HENRIQUE DE ARAÚJO, STIELER EDUARDO, SILVA ANDRESSA, DE MELLO MAR: "Does the compromised sleep and circadian disruption of night and shiftworkers make them highly vulnerable to 2019 coronavirus disease (COVID-19)?", CHRONOBIOLOGY INTERNATIONAL, INFORMA HEALTHCARE, GB, vol. 37, no. 5, 3 May 2020 (2020-05-03), GB , pages 607 - 617, XP055896174, ISSN: 0742-0528, DOI: 10.1080/07420528.2020.1756841 *

Similar Documents

Publication Publication Date Title
US11072648B2 (en) Mast cell stabilizers for treatment of fever
US11318135B2 (en) Use of Favipiravir in treatment of coronavirus infection
JP5781606B2 (en) Use of levocetirizine and montelukast in the treatment of influenza, common colds and inflammation
CN102065854A (en) Combination of pilocarpin and methimazol for treating charcot-marietooth disease and related disorders
AU2018206907B2 (en) Prophylactic and therapeutic drug for nonalcoholic fatty liver disease
WO2013044871A1 (en) The use of chloroquine, chlorpromazine, derivatives thereof, or mixtures thereof in preparing medications for treating and/or preventing pulmonary infection and injury
Preston et al. Temporary treatment interruptions with oral selexipag in pulmonary arterial hypertension: Insights from the Prostacyclin (PGI2) Receptor Agonist in Pulmonary Arterial Hypertension (GRIPHON) study
Sullo et al. Nociceptin/orphanin FQ receptor activation decreases the airway hyperresponsiveness induced by allergen in sensitized mice
Pai et al. Lithium toxicity at therapeutic doses as a fallout of COVID-19 infection: a case series and possible mechanisms
US20230117397A1 (en) Pirfenidone for coronavirus treatment
CN1638772A (en) Anti-viral compounds
Feng et al. Pharmacokinetics of florfenicol in the orange‐spotted grouper, Epinephelus coioides, following oral administration in warm seawater
WO2021263139A1 (en) Methods of treating coronavirus infection using hsp90 inhibitors
CA3105834A1 (en) Compounds for treatment of inflammatory bowel disease and methods thereof
JP6178058B2 (en) Anti-inflammatory and / or antihistamine composition
JP2010083871A (en) Medicinal composition containing anti-adenoviral agent
CA3190278A1 (en) Antiviral use of liraglutide and gefitinib
JP6100510B2 (en) Anti-cold medicine
Trummer‐Menzi et al. Evolution of gene expression changes in newborn rats after mechanical ventilation with reversible intubation
JP5364330B2 (en) Pharmaceutical composition containing azelastine and ambroxol
JP2016175866A (en) PREVENTIVE OR IMPROVING AGENT OF BRONCHIAL ASTHMA CAUSED BY A(H1 N1)pdm09 INFLUENZA VIRUS INFECTION
US20230149328A1 (en) Method for Treating SARS and Treating or Preventing ARDS
EP3441066B1 (en) Mapk inhibitors for the treatment of parkinson&#39;s disease
WO2023205635A1 (en) Compositions and methods of treatment for congenital diarrheal disorder
Koyama Animal Models for the Study of Human Disease: Chapter 36. Animal Models of Febrile Seizures

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21828837

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21828837

Country of ref document: EP

Kind code of ref document: A1