WO2022056092A1 - Methods of treating viral infection - Google Patents

Methods of treating viral infection Download PDF

Info

Publication number
WO2022056092A1
WO2022056092A1 PCT/US2021/049609 US2021049609W WO2022056092A1 WO 2022056092 A1 WO2022056092 A1 WO 2022056092A1 US 2021049609 W US2021049609 W US 2021049609W WO 2022056092 A1 WO2022056092 A1 WO 2022056092A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound
subject
pharmaceutically acceptable
group
Prior art date
Application number
PCT/US2021/049609
Other languages
French (fr)
Inventor
Shanthakumar Tyavanagimatt
Sarah BUCKLEY
Jim FONG
Adam CRAIG
Original Assignee
Cti Biopharma Corp.
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 Cti Biopharma Corp. filed Critical Cti Biopharma Corp.
Publication of WO2022056092A1 publication Critical patent/WO2022056092A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • C12Y207/11001Non-specific serine/threonine protein kinase (2.7.11.1), i.e. casein kinase or checkpoint kinase
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • SARS-CoV-2 can cause not only severe respiratory system distress and lung damage, but can also cause dangerous hyperinflammation, such as in the form of a cytokine storm.
  • hyperinflammation is a common complication not only of COVID-19, the disease caused by SARS-CoV-2, but also other viral diseases such as influenza and diseases caused by other coronaviruses such as SARS-CoV-1 and MERS- CoV.
  • Coronavirus disease 2019 (COVID-19) is a contagious respiratory illness with symptoms such as fever, cough, and shortness of breath along with non-specific symptoms including myalgia and fatigue.
  • Acute respiratory distress syndrome is the leading cause of mortality for COVID-19. Cytokines release by cells in the body during viral infection, including those of the immune system, can become overactive and unregulated, resulting in hyperinflammation. ARDS is characterized by an acute inflammatory response in the lung parenchyma. [0004] There is an urgent need for therapies that can address virally induced hyperinflammation, such as COVID-19-related hyperinflammation. BRIEF SUMMARY [0005] Embodiments of the present disclosure are generally directed to methods of treating a subject for a viral infection.
  • Some embodiments of the present disclosure provide a method of treating a subject infected with a virus, comprising administering to the subject effective amount of a compound of Formula I: Formula (I) wherein: R 1 and R 2 are H; Z 2 is–N(H)-; Ar 1 is selected from the group consisting of: wherein R 10 is methoxy or fluorine; k is an integer selected from 0 and 1; Ar 2 is a group of the formula wherein R 11 is H or selected from the group consisting of:
  • the compound of Formula (I) has the following formula: or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound of Formula (I) has the following formula: or a pharmaceutically acceptable salt thereof.
  • the virus is selected from a coronavirus, ebola virus, hepatitis C virus, and Dengue virus. In certain embodiments, the virus is SARS- CoV-2. DETAILED DESCRIPTION [0009] The present disclosure provides methods of treating viral infections (e.g., coronavirus infections) using a compound of Formula (I), such as pacritinib.
  • Treatment refers to medical management of a disease, disorder, or condition of a subject (e.g., a human or non-human mammal, such as a primate, horse, cat, dog, goat, mouse, or rat).
  • a subject e.g., a human or non-human mammal, such as a primate, horse, cat, dog, goat, mouse, or rat.
  • an appropriate dose or treatment regimen comprising a compound of Formula (I) is administered in an amount sufficient to elicit a therapeutic effect or therapeutic benefit.
  • Therapeutic effect or therapeutic benefit includes improved clinical outcome; modulation of immune response to lessen, reduce, or dampen counterproductive inflammatory cytokine activity; modulation of immune response to normalize counterproductive inflammatory cytokine activity; lessening or alleviation of symptoms associated with a disease; decreased occurrence of symptoms; improved quality of life; longer disease-free status; diminishment of extent of disease, stabilization of disease state; delay of disease progression; remission; survival; prolonged survival; or any combination thereof.
  • a prophylactic treatment meant to “prevent” a disease or condition is a treatment administered to a subject who does not exhibit signs of a disease or exhibits only early signs, for the purpose of decreasing the risk of developing pathology or further advancement of the early disease. For example, if an individual at risk of developing a coronavirus induced respiratory illness is treated with the methods of the present disclosure and does not later develop coronavirus induced respiratory illness, then the disease has been prevented, at least over a period of time, in that individual.
  • a prophylactic treatment can mean preventing recurrence of a disease or condition in a patient that has previously been treated for the disease or condition, e.g., by preventing relapse or recurrence of coronavirus induced respiratory illness.
  • a “therapeutically effective amount” or “effective amount” of a compound of Formula (I), such as pacritinib, is an amount of the compound that is sufficient to result in a therapeutic effect, including improved clinical outcome; lessening or alleviation of symptoms associated with a disease; decreased occurrence of symptoms; improved quality of life; longer disease-free status; diminishment of extent of disease, stabilization of disease state; delay of disease progression; remission; survival; or prolonged survival in a statistically significant manner.
  • a “pharmaceutical composition” refers to a formulation of a compound of the disclosure and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes all pharmaceutically acceptable carriers, diluents or excipients thereof.
  • “Pharmaceutically acceptable salt” includes both acid and base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4- acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesul
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • basic ion exchange resins such as ammonia, isoprop
  • organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.
  • pharmaceutically acceptable salts include quaternary ammonium salts such as quaternary amine alkyl halide salts (e.g., methyl bromide).
  • quaternary ammonium salts such as quaternary amine alkyl halide salts (e.g., methyl bromide).
  • N-oxide refers to R3N + ⁇ O ⁇ , where each R is independently an organic group bound to N.
  • “Pharmaceutically acceptable carrier, diluent or excipient” includes any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
  • “Subject” refers to an animal, such as a mammal, for example a human. The methods described herein can be useful in both human therapeutics and veterinary applications. In some embodiments, the subject is a mammal, and in some embodiments, the subject is human.
  • “Mammal” includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.
  • the term “in vivo” refers to an event that takes place in a subject’s body.
  • the term “in vitro” refers to an event that takes place outside a subject’s body.
  • the term “gene” can include not only coding sequences but also regulatory regions such as promoters, enhancers, and termination regions.
  • the term further can include all introns and other DNA sequences spliced from the mRNA transcript, along with variants resulting from alternative splice sites.
  • Gene sequences encoding the particular protein can be DNA or RNA that directs the expression of the particular protein. These nucleic acid sequences may be a DNA strand sequence that is transcribed into RNA or an RNA sequence that is translated into the particular protein.
  • the nucleic acid sequences include both the full-length nucleic acid sequences as well as non-full-length sequences derived from the full-length protein.
  • a therapeutic agent for treating a subject for an infection with a virus (e.g., coronavirus such as SARS-CoV-2).
  • the therapeutic agent is a compound of Formula (I) having the structure: wherein: R 1 and R 2 are H; Z 2 is–N(H)-; Ar 1 is selected from the group consisting of:
  • R 10 is methoxy or fluorine
  • k is an integer selected from 0 and 1
  • Ar 2 is a group of the formula wherein R 11 is H or selected from the group consisting of:
  • the compound of Formula (I) is 11-(2-Pyrrolidin1-yl-ethoxy)- 14,19-dioxa-5,7,26-triazatetracyclo[19.3.1.1 2,6 .1 8,12 ] heptacosa- 1(25),2(26),3,5,8,10,12(27),16,21,23-decaene (pacritinib) or a pharmaceutically acceptable salt or N-oxide thereof, such as its citrate or maleate salts.
  • the compound of Formula (I) is 9E-15-(2-pyrrolidin-1-yl-ethoxy)- 7,12,25-trioxa-19,21,24-triaza-tetracyclo[18.3.1.1(2,5).1(14,18)]hexacosa- 1(24),2,4,9,14,16,18(26),20,22-nonaene, or a pharmaceutically acceptable salt or N- oxide thereof, such as its citrate or maleate salts.
  • X 1 is -CH2OCH2-.
  • X 2 is -CH2OCH2-.
  • Y is selected from the group consisting of: .
  • the compound of Formula (I) is selected from the group consisting of: ;
  • the compound of Formula (I) has the following formula: or a pharmaceutically acceptable salt thereof.
  • the compound of Formula (Iww) is also known as pacritinib.
  • the compound of Formula (I) has the following formula: or a pharmaceutically acceptable salt thereof.
  • Pharmaceutical Compositions [0035] Some embodiments of the present disclosure include administering a pharmaceutical composition to a subject for treating an infection with a virus (e.g., coronavirus such as SARS-CoV-2).
  • the pharmaceutical composition comprises a compound of Formula (I) and a pharmaceutically acceptable carrier, diluent or excipient.
  • the pharmaceutical composition comprises a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is formulated for oral administration.
  • the pharmaceutical composition comprises an oral capsule.
  • the pharmaceutical composition is formulated for injection.
  • the carrier or excipient is selected from the group consisting of cellulose, lactose, carboxymethylcellulose and magnesium stearate.
  • the pharmaceutical compositions comprise a compound of Formula (I) or a pharmaceutically acceptable salt thereof and an additional therapeutic agent (e.g., chemotherapeutic agent). Non-limiting examples of such additional therapeutic agents are described above.
  • Suitable routes of administration include oral, intravenous, rectal, aerosol, parenteral, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration.
  • parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
  • the compound of Formula (I) is administered orally.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof according to certain embodiments is effective over a wide dosage range.
  • dosages from 0.01 to 2000 mg, from 1 to 1000 mg per day, from 50 to 500 mg per day, and from 200 to 400 mg per day are examples of dosages that are used in some embodiments.
  • An exemplary dosage is between about 50 and about 500 mg per day, or is about 200mg per day.
  • the dosage is 50, 100, 150, 200, 250, 300, 350, 400, 450, or 500 mg per day.
  • the exact dosage will depend upon the route of administration, the form in which the compound of Formula (I) or pharmaceutically acceptable salt thereof is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof is administered in a single dose. Such administration may be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes are used as appropriate. A single dose of the compound of Formula (I) or pharmaceutically acceptable salt thereof may also be used for treatment of an acute condition.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof is administered in multiple doses. In some embodiments, dosing is about once, twice, three times, four times, five times, six times, or more than six times per day. In other embodiments, dosing is about once a month, once every two weeks, once a week, or once every other day.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof is administered about once per day to about four times per day. In certain embodiments, the compound of Formula (I) and the additional therapeutic agent are administered separately. In another embodiment the administration of the compound of Formula (I) or pharmaceutically acceptable salt thereof continues for less than about one month, less than about three weeks, or less than about two weeks. In certain embodiments the additional therapeutic agent is administered for less than about a month, less than about three weeks, less than about two weeks, or less than about one week. In yet another embodiment the administration of the compound of Formula (I) continues for more than about 6, 10, 14, or 28 days. In some cases, continuous dosing is achieved and maintained as long as necessary.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof is administered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for the compound of Formula (I) or pharmaceutically acceptable salt thereof may be found by routine experimentation in light of the instant disclosure.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof is formulated into pharmaceutical compositions.
  • pharmaceutical compositions are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and/or auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof described are administered as pharmaceutical compositions in which the compound of Formula (I) or pharmaceutically acceptable salt thereof is mixed with other active ingredients (e.g., additional therapeutic agents), as in combination therapy.
  • active ingredients e.g., additional therapeutic agents
  • a pharmaceutical composition refers to a mixture of the compound of Formula (I) or pharmaceutically acceptable salt thereof with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
  • the pharmaceutical composition facilitates administration of the compound of Formula (I) or pharmaceutically acceptable salt thereof to a subject.
  • therapeutically effective amounts of the compound of Formula (I) or pharmaceutically acceptable salt thereof provided herein are administered in a pharmaceutical composition to a mammal having a disease, disorder or medical condition to be treated.
  • the mammal is a human.
  • therapeutically effective amounts vary depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein are used singly or in combination with one or more therapeutic agents as components of mixtures.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein is formulated for oral administration.
  • Compounds described herein are formulated by combining the active compounds (i.e., a compound of Formula (I) or a pharmaceutically acceptable salt thereof and, optionally, additional therapeutic agents) with, e.g., pharmaceutically acceptable carriers or excipients.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein are formulated in oral dosage forms that include, by way of example, tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like.
  • pharmaceutical preparations for oral use are obtained by mixing one or more solid excipient, with the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate.
  • disintegrating agents are optionally added.
  • Disintegrating agents include, by way of example, cross-linked croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • dosage forms such as dragee cores and tablets, are provided with one or more suitable coating.
  • concentrated sugar solutions are used for coating the dosage form.
  • the sugar solutions optionally contain additional components, such as by way of example, gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs and/or pigments are also optionally added to the coatings for identification purposes.
  • the dyestuffs and/or pigments are optionally utilized to characterize different combinations of active compound doses (i.e., a compound of Formula (I) or a pharmaceutically acceptable salt thereof and, optionally, additional therapeutic agents).
  • active compound doses i.e., a compound of Formula (I) or a pharmaceutically acceptable salt thereof and, optionally, additional therapeutic agents.
  • therapeutically effective amounts of the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein are formulated into other oral dosage forms.
  • Oral dosage forms include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • push-fit capsules contain the active ingredients in admixture with one or more filler.
  • Fillers include, by way of example, lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • soft capsules contain the compound of Formula (I) or pharmaceutically acceptable salt thereof that is dissolved or suspended in a suitable liquid. Suitable liquids include, by way of example, one or more fatty oil, liquid paraffin, or liquid polyethylene glycol. In addition, stabilizers are optionally added. [0050]
  • therapeutically effective amounts of the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein are formulated for buccal or sublingual administration. Formulations suitable for buccal or sublingual administration include, by way of example, tablets, lozenges, or gels.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein are formulated for parental injection, including formulations suitable for bolus injection or continuous infusion.
  • formulations for injection are presented in unit dosage form (e.g., in ampoules) or in multi-dose containers. Preservatives are, optionally, added to the injection formulations.
  • the pharmaceutical compositions are formulated in a form suitable for parenteral injection as sterile suspensions, solutions or emulsions in oily or aqueous vehicles.
  • Parenteral injection formulations optionally contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form.
  • suspensions of the compound of Formula (I) or pharmaceutically acceptable salt thereof are prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles for use in the pharmaceutical compositions described herein include, by way of example, fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • aqueous injection suspensions contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension contains suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient(s) i.e., a compound of Formula (I) or a pharmaceutically acceptable salt thereof and, optionally, additional therapeutic agents
  • a suitable vehicle e.g., sterile pyrogen-free water
  • pharmaceutical compositions are formulated in any conventional manner using one or more physiologically acceptable carriers, diluents or excipients which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • compositions comprising the compound of Formula (I) or pharmaceutically acceptable salt thereof are manufactured in a conventional manner, such as, by way of example, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein encompasses unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms of the compound of Formula (I) or pharmaceutically acceptable salt thereof presented herein are also considered to be disclosed herein.
  • compositions optionally include other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, buffers, and/or other therapeutically valuable substances.
  • Methods for the preparation of compositions comprising the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein include formulating the compound of Formula (I) or pharmaceutically acceptable salt thereof with one or more inert, pharmaceutically acceptable carriers, diluents or excipients to form a solid, semi-solid or liquid.
  • Solid compositions include powders, tablets, dispersible granules, capsules, cachets, and suppositories.
  • Liquid compositions include solutions in which the compound of Formula (I) or pharmaceutically acceptable salt thereof is dissolved, emulsions comprising a compound, or a solution containing liposomes, micelles, or nanoparticles comprising the compound of Formula (I) or pharmaceutically acceptable salt thereof as disclosed herein.
  • Semi-solid compositions include gels, suspensions and creams.
  • the form of the pharmaceutical compositions described herein include liquid solutions or suspensions, solid forms suitable for solution or suspension in a liquid prior to use, or as emulsions. These compositions also optionally contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and so forth.
  • the pharmaceutical composition comprising the compound of Formula (I) or pharmaceutically acceptable salt thereof illustratively takes the form of a liquid where the agents are present in solution, in suspension or both.
  • a first portion of the therapeutic agent e.g., a compound of Formula (I)
  • a second portion of the therapeutic agent e.g., a compound of Formula (I)
  • a liquid composition includes a gel formulation.
  • the liquid composition is aqueous.
  • aqueous suspensions contain one or more polymers as suspending agents.
  • Useful polymers include water-soluble polymers such as cellulosic polymers, e.g., hydroxypropyl methylcellulose, and water-insoluble polymers such as cross-linked carboxyl-containing polymers.
  • Certain pharmaceutical compositions described herein comprise a mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer), poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran.
  • Useful pharmaceutical compositions also, optionally, include solubilizing agents to aid in the solubility of the compound of Formula (I) or pharmaceutically acceptable salt thereof.
  • solubilizing agent generally includes agents that result in formation of a micellar solution or a true solution of the agent.
  • Certain acceptable nonionic surfactants for example polysorbate 80, are useful as solubilizing agents, as can ophthalmically acceptable glycols, polyglycols, e.g., polyethylene glycol 400, and glycol ethers.
  • compositions optionally include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids
  • bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane
  • buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • compositions also, optionally, include one or more salts in an amount required to bring osmolality of the composition into an acceptable range.
  • Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
  • Other pharmaceutical compositions optionally include one or more preservatives to inhibit microbial activity. Suitable preservatives include mercury- containing substances such as merfen and thiomersal; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.
  • compositions include one or more surfactants to enhance physical stability or for other purposes.
  • Suitable nonionic surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40.
  • Still other compositions include one or more antioxidants to enhance chemical stability where required. Suitable antioxidants include, by way of example, ascorbic acid and sodium metabisulfite.
  • aqueous suspension compositions are packaged in single-dose non-reclosable containers.
  • multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition.
  • other delivery systems for hydrophobic pharmaceutical compounds are employed. Liposomes and emulsions are examples of delivery vehicles or carriers useful herein. In certain embodiments, organic solvents such as N-methylpyrrolidone are also employed.
  • the compounds described herein are delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials are useful herein. In some embodiments, sustained-release capsules release the compounds for a few weeks up to over 100 days.
  • the formulations described herein comprise one or more antioxidants, metal chelating agents, thiol containing compounds and/or other general stabilizing agents.
  • stabilizing agents include: (a) about 0.5% to about 2% w/v glycerol, (b) about 0.1% to about 1% w/v methionine, (c) about 0.1% to about 2% w/v monothioglycerol, (d) about 1 mM to about 10 mM EDTA, (e) about 0.01% to about 2% w/v ascorbic acid, (f) about 0.003% to about 0.02% w/v polysorbate 80, (g) 0.001% to about 0.05% w/v.
  • polysorbate 20 (h) arginine, (i) heparin, (j) dextran sulfate, (k) cyclodextrins, (l) pentosan polysulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n) combinations thereof.
  • the concentration of the compound of Formula (I) or pharmaceutically acceptable salt thereof provided in the pharmaceutical compositions is less than about 100%, about 90%, about 80%, about 70%, about 60%, about 50%, about 40%, about 30%, about 20%, about 19%, about 18%, about 17%, about 16%, about 15%, about 14%, about 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1%, about 0.5%, about 0.4%, about 0.3%, about 0.2%, about 0.1%, about 0.09%, about 0.08%, about 0.07%, about 0.06%, about 0.05%, about 0.04%, about 0.03%, about 0.02%, about 0.01%, about 0.009%, about 0.008%, about 0.007%, about 0.006%, about 0.005%, about 0.004%, about 0.003%, about 0.002%, about 0.001%, about 0.0009%, about 0.0008%
  • the concentration of the compound of Formula (I) or pharmaceutically acceptable salt thereof is in the range from approximately 0.0001% to approximately 50%, approximately 0.001% to approximately 40 %, approximately 0.01% to approximately 30%, approximately 0.02% to approximately 29%, approximately 0.03% to approximately 28%, approximately 0.04% to approximately 27%, approximately 0.05% to approximately 26%, approximately 0.06% to approximately 25%, approximately 0.07% to approximately 24%, approximately 0.08% to approximately 23%, approximately 0.09% to approximately 22%, approximately 0.1% to approximately 21%, approximately 0.2% to approximately 20%, approximately 0.3% to approximately 19%, approximately 0.4% to approximately 18%, approximately 0.5% to approximately 17%, approximately 0.6% to approximately 16%, approximately 0.7% to approximately 15%, approximately 0.8% to approximately 14%, approximately 0.9% to approximately 12%, approximately 1% to approximately 10% w/w, w/v or v/v.
  • the concentration of the compound of Formula (I) or pharmaceutically acceptable salt thereof is in the range from approximately 0.001% to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%, approximately 0.03% to approximately 4%, approximately 0.04% to approximately 3.5%, approximately 0.05% to approximately 3%, approximately 0.06% to approximately 2.5%, approximately 0.07% to approximately 2%, approximately 0.08% to approximately 1.5%, approximately 0.09% to approximately 1%, approximately 0.1% to approximately 0.9% w/w, w/v or v/v.
  • the amount of the compound of Formula (I) or pharmaceutically acceptable salt thereof is equal to or less than about 10 g, about 9.5 g, about 9.0 g, about 8.5 g, about 8.0 g, about 7.5 g, about 7.0 g, about 6.5 g, about 6.0 g, about 5.5 g, about 5.0 g, about 4.5 g, about 4.0 g, about 3.5 g, about 3.0 g, about 2.5 g, about 2.0 g, about 1.5 g, about 1.0 g, about 0.95 g, about 0.9 g, about 0.85 g, about 0.8 g, about 0.75 g, about 0.7 g, about 0.65 g, about 0.6 g, about 0.55 g, about 0.5 g, about 0.45 g, about 0.4 g, about 0.35 g, about 0.3 g, about 0.25 g, about 0.2 g, about 0.15 g, about 0.1 g, about 0.09 g,
  • Some embodiments of the present disclosure are directed to methods of treating a subject for a viral infection.
  • the present disclosure provides methods of treating a subject for an infection with a virus (e.g., coronavirus such as SARS-CoV-2) comprising administering to the subject an effective amount of a compound of Formula (I), or a pharmaceutical composition comprising a compound of Formula (I), such a pacritinib.
  • a virus e.g., coronavirus such as SARS-CoV-2
  • Patients or subjects that can be treated by the methods disclosed herein include, but are not limited to, a mammal, such as human or non-human primates (e.g., monkeys and apes). In some embodiments, the subject is human.
  • the subject can be male or female and can be any suitable age, including infant, juvenile, adolescent, adult, and geriatric.
  • the subject is infected with a virus that utilizes the clathrin mediated endocytosis entry pathway.
  • Clathrin mediated endocytosis is a process by which cells absorb metabolites, hormones, proteins, and in some cases viruses, by the inward budding of the plasma membrane (invagination).
  • Viruses that utilize the clathrin mediated endocytosis pathway for entry include coronaviruses (e.g., SARS-CoV1 and SARS-CoV-2), ebola virus, influenza A virus, vesicular stomatitis virus, and hepatitis C virus.
  • coronaviruses e.g., SARS-CoV1 and SARS-CoV-2
  • ebola virus e.g., influenza A virus, vesicular stomatitis virus, and hepatitis C virus.
  • Proteins involved in clathrin mediated endocytosis include “AP2- associated protein kinase 1” or “AAK1,” and “cyclin G-associated kinase” or “GAK.”
  • AAK1 and GAK are known to block viral infectivity of viruses that utilize the clathrin mediated endocytosis pathway, such as ebola virus, Marburg virus, hepatitis C virus, and Dengue virus.
  • SARS-CoV-1, SARS-CoV2, and MERS-CoV are also known to utilize clathrin mediated endocytosis.
  • a compound of Formula (I), pacritinib surprisingly interacts with both AAK1 and GAK and thus the compound may have antiviral effects by inhibiting clathrin mediated endocytosis.
  • the subject has viral-induced hyperinflammation. Following viral infection and proliferation, macrophages migrate to the infection site to kill infected cells, leading to the release of inflammatory cytokines and chemokines. The response to this stimuli is local inflammation and increased blood flow, enabling plasma and leukocytes to travel to the infection site.
  • hyperinflammation is characterized by severe, systemic, and uncontrolled immune cell activation and inflammatory cytokine/chemokine production in response to a persistent trigger such as viremia. Symptoms of hyperinflammation may include unremitting fever, splenomegaly, coagulopathy, hepatitis, cytopenia and, if unrestrained, the hyperinflammation may cause multi-organ failure and death. In some embodiments, hyper-inflammation is associated with a viral infection and is a cytokine storm or Acute respiratory distress syndrome (ARDS).
  • ARDS Acute respiratory distress syndrome
  • Cytokine storm also known as “cytokine release syndrome,” refers to an acute systemic inflammatory response that can be induced by a variety of factors such as infection by a virus, therapeutic antibodies, or cellular immunotherapy. Symptoms include fever, nausea, fatigue and headache. Potentially life-threatening complications of cytokine storm include, renal failure, hepatic failure, respiratory distress, and cardiac dysfunction. Soluble mediators of inflammation, including cytokines and chemokines, contribute to the pathophysiology of cytokine storm. These inflammatory cytokines/chemokines include IL-6, TNF ⁇ , IFN ⁇ , IL-1 ⁇ , IL-2, IL-6, IL-8, and IL-10.
  • Elevated levels of inflammatory cytokines have been identified in patients with viral infections, including Orthomyxoviridae influenza A infections. Cytokine storms are associated with a number of other viral infections, including Epstein-Barr virus of the Herpesviridae family, group A Streptococcus pyogenes, and Orthopoxvirus variola. Betacoronavirus infection, including infection by the SARS-CoV-1, which causes severe acute respiratory syndrome (SARS) is also associated with a cytokine storm characterized by elevated levels of inflammatory cytokines and immunopathological damage. [0075] In some embodiments, a virus associated with hyperinflammation is “SARS-CoV-1”.
  • SARS-CoV-1 refers to the strain of Betacoronavirus that emerged in 2003 and causes severe acute respiratory syndrome (SARS).
  • SARS-CoV-2 a virus associated with hyperinflammation
  • SARS-CoV-2 refers to the strain of Betacoronavirus that emerged in 2019 and causes the respiratory illness Coronavirus disease 2019 (COVID-19).
  • subject infected with a coronavirus has COVID- 19.
  • COVID-19 refers to the infectious disease caused by SARS- CoV-2 and characterized by, for example, fever, cough, respiratory symptoms, rhinorrhea, sore throat, malaise, headache, chills, repeated shaking with chills, diarrhea, new loss of smell or taste, muscle pain, or a combination thereof.
  • the subject exhibits one or more symptoms associated with COVID-19 or exhibits no symptoms associated with COVID-19 (asymptomatic).
  • asymptomatic infection refers to patients diagnosed with COVID-19 by a standardized assay that do not present with COVID-19 symptoms (e.g., fever, cough, respiratory symptoms, rhinorrhea, sore throat, malaise, headache, or muscle pain).
  • COVID-19 symptoms may include mild symptoms such as fever, rhinorrhea, mild cough, sore throat, malaise, headache, muscle pain, or any combination thereof, but with no shortness of breath.
  • Patients with “mild” COVID-19 infection present no signs of lower airway disease and have a normal respiratory rate and heart rate, and oxygen saturation (pulse oximetry) greater than 93% on room air.
  • COVID-19 symptoms may include moderate symptoms such as lower respiratory symptoms, including shortness of breath (at rest or with exertion) or signs of pneumonia, such as reduced oxygen saturation (pulse oximetry).
  • the subject has severe COVID-19. “Severe COVID-19” is defined as confirmed disease in patients who are hospitalized with hypoxia [SpO2 ⁇ 93% on room air], respiratory rate >30, PaO2/FiO2 ⁇ 300, but do not require intermittent mandatory ventilation (IMV).
  • IMV intermittent mandatory ventilation
  • the subject exhibits no symptoms associated with COVID-19 but has been exposed to another subject known or suspected of having COVID-19.
  • the subject with a coronavirus exhibits one or more symptoms selected from dry cough, shortness of breath, and fever.
  • the subject has been hospitalized prior to treatment.
  • the subject has had severe COVID-19 with SARS- CoV-2 infection confirmed by either a) a positive reverse transcriptase polymerase chain reaction (RT PCR) or b) an antigen-based test from any respiratory, nasopharyngeal, saliva, blood, or stool specimen, documented within 1 week prior to the start of treatment.
  • the subject has cancer.
  • the subject does not have cancer.
  • the subject has viral-induced hyperinflammation.
  • the viral induced hyperinflammation may be a cytokine storm or acute respiratory distress syndrome (ARDS).
  • ARDS refers to a respiratory condition characterized by severe hypoxemia and may be induced by viral infection.
  • ARDS is characterized by severe impairment in lung gas exchange and lung mechanics.
  • the innate immune response is overactive in ARDS.
  • Virally-induced inflammation promotes pulmonary epithelial and endothelial cellular damage leading to increased capillary permeability.
  • Patients with ARDS may exhibit buildup of fluid in the lung and have reduced oxygen levels in the blood.
  • the present disclosure also provides prophylactic treatment of a subject prior to receipt of a test result for viral infection (e.g., SARS-CoV-2).
  • the subject may be at risk of contracting the virus.
  • Prophylactic treatment would be administered to individuals at risk of exposure to infected patients, for example, immunocompromised individuals, medical personnel, people with known exposure, and essential workers.
  • treatment of a subject that does not have respiratory symptoms associated with either COVID-19 or ARDS reduces the likelihood of the subject developing one or more of mild, moderate, or severe COVID- 19 or ARDS symptoms.
  • treatment of a subject according to the methods described herein results in reduction of the severity or duration of the severe viral infection symptoms. Severe viral infection symptoms may include severe respiratory symptoms such as, for example, shortness of breath, difficulty breathing, reduced respiratory rate, and wheezing.
  • the methods of treatment described herein result in reduction of one or more inflammatory cytokines and/or chemokines.
  • Inflammatory cytokines or chemokines include, for example, CD40L, EGF, Eotaxin (CCL11), FGF-2, Flt-3 ligand, Fractalkine, G-CSF, GM-CSF, GRO ⁇ (CXCL1), IFN- ⁇ 2, IFN- ⁇ , IL-1 ⁇ , IL-1 ⁇ , IL-1RA, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (CXCL8), IL-9, IL-10, IL-12 (p40), IL-12 (p70), IL-13, IL-15, IL-17A, IL-17E/IL-25, IL-17F, IL-18, IL-22, IL-27, IP-10 (CXCL10), MCP-1 (CCL2), MCP-3, M-CSF, MDC (CCL)
  • the level of the inflammatory cytokine and/or chemokines may be measured in blood plasma, e.g., by enzyme-linked immunosorbent assays (ELISA), bead-based immunoassays and other immunoassays, and/or the transcriptional level of inflammatory cytokines and/or chemokines may be measured by RNA sequencing.
  • ELISA enzyme-linked immunosorbent assays
  • bead-based immunoassays and other immunoassays e.g., bead-based immunoassays and other immunoassays
  • the transcriptional level of inflammatory cytokines and/or chemokines may be measured by RNA sequencing.
  • treatment of a subject according to the methods described herein results in reduction of a symptom associated hyperinflammation.
  • hyperinflammation is cytokine release syndrome (or cytokine storm) or acute respiratory distress syndrome (ARDS).
  • ARDS acute respiratory distress syndrome
  • treatment of a subject according to the methods described herein results in reduction of SARS-CoV-2 viral load.
  • the reduction in viral load may be a reduction in plasma viral load.
  • the viral load is measured by PCR.
  • the virus is SARS-CoV-2 and the plasma SARS-CoV-2 viral load is measured by detecting the nucleocapsid (N) gene.
  • N nucleocapsid
  • use of a compound of Formula (I), pacritinib surprisingly results in decreased viral infectivity of SARS-CoV-2.
  • treatment of a subject according to the methods described herein results in reduced duration or occurrence of hospitalization, ventilation or dialysis of the subject.
  • the treating results a reduced likelihood of the subject requiring intermittent mandatory ventilation (IMV) or extracorporeal membrane oxygenation (ECMO), as compared to a subject with similar symptoms who has not been treated with the compound of Formula (I).
  • IMV intermittent mandatory ventilation
  • ECMO extracorporeal membrane oxygenation
  • “Extracorporeal membrane oxygenation” or “ECMO” refers to a treatment that uses a pump to circulate blood through an artificial lung back into the bloodstream.
  • Intermittent mandatory ventilation” or “IMV” refers to a ventilation mode that provides partial mechanical assistance, whereby mandatory breaths are delivered at a set frequency, tidal volume, and inspiratory flow rate.
  • treatment of a subject according to the methods described herein results in the subject having a faster and/or more extensive recovery than a subject with similar symptoms who has not been treatment with the compound of Formula (I).
  • An appropriate dose, suitable duration, and frequency of administration of the compound of Formula (I) will be determined by such factors as the condition of the patient, size, weight, body surface area, age, sex, type and severity of the disease, particular therapy to be administered, particular form of the active ingredient, time and the method of administration, and other drugs being administered concurrently, which can readily be determined by a person skilled in the art.
  • Certain aspects of the first embodiment include administering an effective amount of pacritinib. Pacritinib may be effective over a wide dosage range.
  • the effective amount ranges from 0.01 to 2000 mg, from 1 to 1000 mg per day, from 50 to 500 mg per day, or from 200 to 400 mg per day. In particular embodiments, the effective amount ranges from between about 50 mg per day to about 500 mg per day, or is about 200mg per day. In various embodiments, the effective amount is 50, 100, 150, 200, 250, 300, 350, 400, 450, or 500 mg per day.
  • Some aspects of the first embodiment include administering one or more further therapeutic agents, such as an antiviral agent or an agent to reduce inflammation, to provide a synergistic or additive therapeutic effect.
  • Agents to reduce inflammation include checkpoint inhibitors.
  • checkpoint molecule refers to one or more proteins, molecules, compounds or complexes providing inhibitory signals to assist in controlling or suppressing an immune response.
  • immune checkpoint molecules include those molecules that partially or totally block immune stimulation; decrease, prevent or delay immune activation; or increase, activate, or up regulate immune suppression.
  • checkpoint molecules include PD-1, PD-L1, PD-L2, CD80, CD86, B7-H3, B7-H4, HVEM, adenosine, GAL9, VISTA, CEACAM-1, PVRL2, CTLA-4, BTLA, KIR, LAG3, TIM3, A2aR, CD244/2B4, CD160, TIGIT, LAIR-1, PVRIG/CD112R, and certain metabolic enzymes, such as arginase, indoleamine 2,3-dioxygenase (IDO).
  • antiviral agents refers to a class of drugs for treating viral infections.
  • An antiviral agent may be, for example, a small molecule, peptide, protein, antibody, nucleic acid, or aptamer that targets one or more components in the viral life cycle: attachment to host cell; release of viral genes and possibly enzymes into the host cell; replication of viral components using host cell machinery; assembly of viral components into viral particles; and release of viral particles to infect new host cells.
  • Targets of antiviral agents include critical viral proteins such as neuraminidase, M2 ion channel protein, hemagglutinin, viral RNA polymerase, NTPase/helicase, spike (S) glycoprotein (S1 domain), and 3C-like cysteine protease.
  • anti-viral agents include seltamivir, zanamivir, laninamivir, laninamivir, peramivir, and remdesivir.
  • the present disclosure also provides a therapeutic composition comprising a compound of Formula (I) and at least one of: an anti-viral therapeutic agent, and an agent to reduce hyperinflammation.
  • EXAMPLES EXAMPLE 1 This Example demonstrates that pacritinib, which is a compound of Formula (I), reduced the cytopathic effects (CPE) of SARS-CoV-2. Confluent or near- confluent cell culture monolayers of Vero 76 cells were prepared in 96-well disposable microplates one day prior to testing.
  • a known protease inhibitor was tested in parallel as a positive control drug using the same method as was applied for pacritinib. The positive control was tested with every test run.
  • Growth media was removed from the cells and the test compound was applied in 0.1 ml volume to wells at 2X concentration.
  • Virus, at ⁇ 60 CCID50 (50% cell culture infectious dose) in 0.1 ml volume was added to the wells designated for virus infection.
  • Medium devoid of virus was placed in toxicity control wells and cell control wells. Plates were incubated at 37 o C with 5% CO2 until marked CPE (>80% CPE) was observed in virus control wells. The plates were then stained with 0.011% neutral red for approximately two hours at 37 o C in a 5% CO2 incubator.
  • the neutral red medium was removed by complete aspiration, and the cells were rinsed 1X with phosphate buffered solution (PBS) to remove residual dye.
  • PBS phosphate buffered solution
  • the PBS was completely removed, and the incorporated neutral red was eluted with 50% Sorensen’s citrate buffer/50% ethanol for 30 minutes.
  • Neutral red dye penetrates into living cells, thus, the presence of intense red color, indicates that viable cells are present in the wells.
  • the dye content in each well was quantified using a spectrophotometer at 540 nm wavelength.
  • the dye content in each set of wells was converted to a percentage of dye present in untreated control wells using a Microsoft Excel computer-based spreadsheet and normalized based on the virus control.
  • VYR Viral Yield Reduction
  • the 90% (one log10) effective concentration (EC90) was calculated by regression analysis by plotting the log10 of the inhibitor concentration versus log10 of virus produced at each concentration. Dividing EC90 by the CC50 gives the SI value for this test.
  • the results of the Viral Yield Reduction (VYR) assay are shown in Table 2. Table 2. [0107] After sufficient virus replication of SARS-CoV-2 (3 days), a sample of supernatant was taken from each infected well (three replicate wells were pooled) and tested for titer determination. Titration of the viral samples was performed by serial dilution and plating into Vero 76 cells. After incubation as described in Example 1, the cells were scored.
  • HEK293 cells transiently expressing NanoLuc®- AAK1 or GAK Fusion Vector were seeded into the wells of 384-well plates.
  • the cells were pre-treated with the NanoBRETTM Tracer K-5 and then treated with reference compound CTx-0294885 for 1 hour.
  • the BRET signal was measured on an Envision 2104 Multilabel Reader.
  • IC50 value was calculated and IC50 curve was plotted using the GraphPad Prism 4 program based on a sigmoidal dose response equation. The results are shown in Table 4. Table 4.
  • Patients will be randomized 1:1 to receive pacritinib (400 mg once daily [QD] on Day 1, then 200 mg twice daily [BID] from Day 2 to Day 14) + SOC or placebo + SOC.
  • the inclusion criteria will include: ⁇ hospitalization prior to randomization for the treatment of severe COVID-19 with SARS-CoV-2 infection confirmed by either a) a positive reverse transcriptase polymerase chain reaction (RT PCR) or b) an antigen-based test from any respiratory, nasopharyngeal, saliva, blood, or stool specimen at Screening or documented within 1 week prior to the start of Screening.
  • RT PCR positive reverse transcriptase polymerase chain reaction
  • the exclusion criteria will include: ⁇ In the opinion of the investigator, progression to death is imminent and inevitable within the next 24 hours, irrespective of the provision of treatments ⁇ ⁇ Currently intubated or intubated between screening and randomization ⁇ ⁇ Suspected active uncontrolled bacterial, fungal, viral, or other infection (besides COVID 19) ⁇ o Prior allogenic hematopoietic stem cell transplantation ⁇ ⁇ Active lung cancer or history of lung cancer within the past 12 months o ⁇ Any active grade 2 or higher hemorrhage ⁇ ⁇ Any active gastrointestinal or metabolic condition that could interfere with absorption of oral medication ⁇ o ⁇ Uncontrolled intercurrent illness that, in the judgment of the treating physician, would limit compliance with study requirements ⁇ ⁇ Known seropositivity for human immunodeficiency virus with cluster of differentiation 4
  • Prophylactic anticoagulation therapy or aspirin ( ⁇ 100mg) are permitted. ⁇ ⁇ Unable to ingest capsules or tablets at randomization [0113] Assigned treatment will continue for up to Day 14 or until the patient experiences intolerable adverse events (AEs), withdraws consent, or initiates another investigational therapy or until the study is terminated. Assigned therapy may be given for an additional 7 days (for a total of 21 days) if, in the opinion of the investigator, the patient's clinical signs and symptoms are improving and the potential benefit outweighs the potential risk. In the event of hospital discharge, patients will complete treatment with the assigned therapy as an outpatient.
  • pacritinib 400 mg will be administered once daily [QD] on Day 1, then 200 mg will be administered twice daily [BID] from Day 2 to Day 14, along with standard of care (SOC) treatment.
  • SOC standard of care
  • 4 placebo capsules will be administered once daily [QD] on Day 1, then 2 capsules will be administered twice daily [BID] from Day 2 to Day 14) + SOC.
  • Both pacritinib and the placebo will be provided in 100 mg capsules.
  • the primary outcome measured will be the proportion of patients who progress to intermittent mandatory ventilation (IMV) and/or extracorporeal membrane oxygenation (ECMO) or death during the 28 days following randomization. The proportion will be calculated as the number of patients who progress divided by the total number of patients in the study population.
  • IMV intermittent mandatory ventilation
  • ECMO extracorporeal membrane oxygenation

Abstract

Provided herein are methods of preventing and treating viral infections (e.g., coronavirus infection) using a compound of Formula (I).

Description

METHODS OF TREATING VIRAL INFECTION BACKGROUND [0001] SARS-CoV-2 can cause not only severe respiratory system distress and lung damage, but can also cause dangerous hyperinflammation, such as in the form of a cytokine storm. Such hyperinflammation is a common complication not only of COVID-19, the disease caused by SARS-CoV-2, but also other viral diseases such as influenza and diseases caused by other coronaviruses such as SARS-CoV-1 and MERS- CoV. [0002] Coronavirus disease 2019 (COVID-19) is a contagious respiratory illness with symptoms such as fever, cough, and shortness of breath along with non-specific symptoms including myalgia and fatigue. [0003] Acute respiratory distress syndrome (ARDS) is the leading cause of mortality for COVID-19. Cytokines release by cells in the body during viral infection, including those of the immune system, can become overactive and unregulated, resulting in hyperinflammation. ARDS is characterized by an acute inflammatory response in the lung parenchyma. [0004] There is an urgent need for therapies that can address virally induced hyperinflammation, such as COVID-19-related hyperinflammation. BRIEF SUMMARY [0005] Embodiments of the present disclosure are generally directed to methods of treating a subject for a viral infection. [0006] Some embodiments of the present disclosure provide a method of treating a subject infected with a virus, comprising administering to the subject effective amount of a compound of Formula I:
Figure imgf000003_0001
Formula (I) wherein: R1 and R2 are H; Z2 is–N(H)-; Ar1 is selected from the group consisting of:
Figure imgf000003_0002
wherein R10 is methoxy or fluorine; k is an integer selected from 0 and 1; Ar2 is a group of the formula
Figure imgf000003_0003
wherein R11 is H or selected from the group consisting of:
Figure imgf000004_0001
L is a group of formula:
Figure imgf000004_0002
wherein X1 is attached to Ar1 and X2 is attached to Ar2, and wherein X1, X2 and Y are selected such that the group L has from 5 to 15 atoms in normal linear chain connecting Ar1 to Ar2, wherein X1 is selected from the group consisting of: (a)-OCH2- (b) –OCH2CH2-, and (c)–CH2OCH2-; wherein X2 is selected from the group consisting of: (a) –CH2O-, (b)–CH2CH2O-, and (c)–CH2OCH2-; Y is a group of formula –CRa=CRb-, wherein Ra and Rb are H, or a pharmaceutically acceptable salt or N-oxide thereof. [0007] In certain embodiments, the compound of Formula (I) has the following formula:
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound of Formula (I) has the following formula:
Figure imgf000005_0002
or a pharmaceutically acceptable salt thereof. [0008] In some embodiments, the virus is selected from a coronavirus, ebola virus, hepatitis C virus, and Dengue virus. In certain embodiments, the virus is SARS- CoV-2. DETAILED DESCRIPTION [0009] The present disclosure provides methods of treating viral infections (e.g., coronavirus infections) using a compound of Formula (I), such as pacritinib. [0010] Prior to setting forth this disclosure in more detail, it may be helpful to an understanding thereof to provide definitions of certain terms to be used herein. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs. Additional definitions are set forth throughout this disclosure. [0011] In the present description, the term “about” means + 20% of the indicated range, value, or structure, unless otherwise indicated. The term “consisting essentially of” limits the scope of a claim to the specified materials or steps and those that do not materially affect the basic and novel characteristics of the claimed invention. It should be understood that the terms “a” and “an” as used herein refer to “one or more” of the enumerated components. The use of the alternative (e.g., “or”) should be understood to mean either one, both, or any combination thereof of the alternatives. As used herein, the terms “include” and “have” are used synonymously, which terms and variants thereof are intended to be construed as non-limiting. The term “comprise” means the presence of the stated features, integers, steps, or components as referred to in the claims, but that it does not preclude the presence or addition of one or more other features, integers, steps, components, or groups thereof. Any ranges provided herein include all the values and narrower ranges in the ranges. [0012] “Treat” or “treatment” or “ameliorate” refers to medical management of a disease, disorder, or condition of a subject (e.g., a human or non-human mammal, such as a primate, horse, cat, dog, goat, mouse, or rat). In general, an appropriate dose or treatment regimen comprising a compound of Formula (I) is administered in an amount sufficient to elicit a therapeutic effect or therapeutic benefit. Therapeutic effect or therapeutic benefit includes improved clinical outcome; modulation of immune response to lessen, reduce, or dampen counterproductive inflammatory cytokine activity; modulation of immune response to normalize counterproductive inflammatory cytokine activity; lessening or alleviation of symptoms associated with a disease; decreased occurrence of symptoms; improved quality of life; longer disease-free status; diminishment of extent of disease, stabilization of disease state; delay of disease progression; remission; survival; prolonged survival; or any combination thereof. [0013] A prophylactic treatment meant to “prevent” a disease or condition (e.g., coronavirus induced respiratory illness in a subject or patient) is a treatment administered to a subject who does not exhibit signs of a disease or exhibits only early signs, for the purpose of decreasing the risk of developing pathology or further advancement of the early disease. For example, if an individual at risk of developing a coronavirus induced respiratory illness is treated with the methods of the present disclosure and does not later develop coronavirus induced respiratory illness, then the disease has been prevented, at least over a period of time, in that individual. A prophylactic treatment can mean preventing recurrence of a disease or condition in a patient that has previously been treated for the disease or condition, e.g., by preventing relapse or recurrence of coronavirus induced respiratory illness. [0014] A “therapeutically effective amount” or “effective amount” of a compound of Formula (I), such as pacritinib, is an amount of the compound that is sufficient to result in a therapeutic effect, including improved clinical outcome; lessening or alleviation of symptoms associated with a disease; decreased occurrence of symptoms; improved quality of life; longer disease-free status; diminishment of extent of disease, stabilization of disease state; delay of disease progression; remission; survival; or prolonged survival in a statistically significant manner. When referring to an individual active ingredient or a cell expressing a single active ingredient, administered alone, a therapeutically effective amount refers to the effects of that ingredient or cell expressing that ingredient alone. When referring to a combination, a therapeutically effective amount refers to the combined amounts of active ingredients or combined adjunctive active ingredient with a cell expressing an active ingredient that results in a therapeutic effect, whether administered serially (i.e., sequentially) or simultaneously. [0015] A "pharmaceutical composition" refers to a formulation of a compound of the disclosure and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes all pharmaceutically acceptable carriers, diluents or excipients thereof. [0016] "Pharmaceutically acceptable salt" includes both acid and base addition salts. [0017] "Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4- acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2- hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo- glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-1,5-disulfonic acid, naphthalene-2- sulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid (e.g., L-(+)-tartaric acid), thiocyanic acid, p-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like. [0018] "Pharmaceutically acceptable base addition salt" refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine. [0019] In some embodiments, pharmaceutically acceptable salts include quaternary ammonium salts such as quaternary amine alkyl halide salts (e.g., methyl bromide). [0020] “N-oxide” refers to R3N+−O, where each R is independently an organic group bound to N. [0021] "Pharmaceutically acceptable carrier, diluent or excipient" includes any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals. [0022] "Subject" refers to an animal, such as a mammal, for example a human. The methods described herein can be useful in both human therapeutics and veterinary applications. In some embodiments, the subject is a mammal, and in some embodiments, the subject is human. [0023] "Mammal" includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like. [0024] The term "in vivo" refers to an event that takes place in a subject’s body. [0025] The term "in vitro" refers to an event that takes place outside a subject’s body. [0026] The term “gene” can include not only coding sequences but also regulatory regions such as promoters, enhancers, and termination regions. The term further can include all introns and other DNA sequences spliced from the mRNA transcript, along with variants resulting from alternative splice sites. Gene sequences encoding the particular protein can be DNA or RNA that directs the expression of the particular protein. These nucleic acid sequences may be a DNA strand sequence that is transcribed into RNA or an RNA sequence that is translated into the particular protein. The nucleic acid sequences include both the full-length nucleic acid sequences as well as non-full-length sequences derived from the full-length protein. [0027] Additional definitions are provided in the sections below. Therapeutic Agent [0028] Various embodiments provide methods including administering an effective amount of a therapeutic agent for treating a subject for an infection with a virus (e.g., coronavirus such as SARS-CoV-2). In some embodiments, the therapeutic agent is a compound of Formula (I) having the structure:
Figure imgf000010_0001
wherein: R1 and R2 are H; Z2 is–N(H)-; Ar1 is selected from the group consisting of:
Figure imgf000011_0001
wherein R10 is methoxy or fluorine; k is an integer selected from 0 and 1; Ar2 is a group of the formula
Figure imgf000011_0002
wherein R11 is H or selected from the group consisting of:
Figure imgf000011_0003
Figure imgf000012_0001
L is a group of formula: –X1-Y-X2- wherein X1 is attached to Ar1 and X2 is attached to Ar2, and wherein X1, X2 and Y are selected such that the group L has from 5 to 15 atoms in normal linear chain connecting Ar1 to Ar2, wherein X1 is selected from the group consisting of: (a)-OCH2- (b) –OCH2CH2-, and (c)–CH2OCH2-; wherein X2 is selected from the group consisting of: (a) –CH2O-, (b)–CH2CH2O-, and (c)–CH2OCH2-; Y is a group of formula –CRa=CRb-, wherein Ra and Rb are H, or a pharmaceutically acceptable salt or N-oxide thereof. In some embodiments the compound of Formula (I) is 11-(2-Pyrrolidin1-yl-ethoxy)- 14,19-dioxa-5,7,26-triazatetracyclo[19.3.1.12,6.18,12] heptacosa- 1(25),2(26),3,5,8,10,12(27),16,21,23-decaene (pacritinib) or a pharmaceutically acceptable salt or N-oxide thereof, such as its citrate or maleate salts. In certain embodiments, the compound of Formula (I) is 9E-15-(2-pyrrolidin-1-yl-ethoxy)- 7,12,25-trioxa-19,21,24-triaza-tetracyclo[18.3.1.1(2,5).1(14,18)]hexacosa- 1(24),2,4,9,14,16,18(26),20,22-nonaene, or a pharmaceutically acceptable salt or N- oxide thereof, such as its citrate or maleate salts. [0029] In certain embodiments, X1 is -CH2OCH2-. [0030] In certain embodiments, X2 is -CH2OCH2-. [0031] In certain embodiments, Y is selected from the group consisting of:
Figure imgf000013_0001
. [0032] In certain embodiments, the compound of Formula (I) is selected from the group consisting of: ;
Figure imgf000013_0002
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
and pharmaceutically acceptable salts thereof. [0033] In certain embodiments, the compound of Formula (I) has the following formula:
Figure imgf000020_0002
or a pharmaceutically acceptable salt thereof. The compound of Formula (Iww) is also known as pacritinib. [0034] In certain embodiments, the compound of Formula (I) has the following formula:
Figure imgf000021_0001
or a pharmaceutically acceptable salt thereof. Pharmaceutical Compositions [0035] Some embodiments of the present disclosure include administering a pharmaceutical composition to a subject for treating an infection with a virus (e.g., coronavirus such as SARS-CoV-2). The pharmaceutical composition comprises a compound of Formula (I) and a pharmaceutically acceptable carrier, diluent or excipient. In some embodiments the pharmaceutical composition comprises a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier or excipient. [0036] In some embodiments, the pharmaceutical composition is formulated for oral administration. For example, in some embodiments, the pharmaceutical composition comprises an oral capsule. In other embodiments, the pharmaceutical composition is formulated for injection. In some more specific embodiments, the carrier or excipient is selected from the group consisting of cellulose, lactose, carboxymethylcellulose and magnesium stearate. [0037] In still more embodiments, the pharmaceutical compositions comprise a compound of Formula (I) or a pharmaceutically acceptable salt thereof and an additional therapeutic agent (e.g., chemotherapeutic agent). Non-limiting examples of such additional therapeutic agents are described above. [0038] Suitable routes of administration include oral, intravenous, rectal, aerosol, parenteral, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration. In addition, by way of example, parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections. In particular embodiments, the compound of Formula (I) is administered orally. [0039] The compound of Formula (I) or pharmaceutically acceptable salt thereof according to certain embodiments is effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 2000 mg, from 1 to 1000 mg per day, from 50 to 500 mg per day, and from 200 to 400 mg per day are examples of dosages that are used in some embodiments. An exemplary dosage is between about 50 and about 500 mg per day, or is about 200mg per day. In various embodiments, the dosage is 50, 100, 150, 200, 250, 300, 350, 400, 450, or 500 mg per day. The exact dosage will depend upon the route of administration, the form in which the compound of Formula (I) or pharmaceutically acceptable salt thereof is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician. [0040] In some embodiments, the compound of Formula (I) or pharmaceutically acceptable salt thereof is administered in a single dose. Such administration may be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes are used as appropriate. A single dose of the compound of Formula (I) or pharmaceutically acceptable salt thereof may also be used for treatment of an acute condition. [0041] In some embodiments, the compound of Formula (I) or pharmaceutically acceptable salt thereof is administered in multiple doses. In some embodiments, dosing is about once, twice, three times, four times, five times, six times, or more than six times per day. In other embodiments, dosing is about once a month, once every two weeks, once a week, or once every other day. In another embodiment the compound of Formula (I) or pharmaceutically acceptable salt thereof is administered about once per day to about four times per day. In certain embodiments, the compound of Formula (I) and the additional therapeutic agent are administered separately. In another embodiment the administration of the compound of Formula (I) or pharmaceutically acceptable salt thereof continues for less than about one month, less than about three weeks, or less than about two weeks. In certain embodiments the additional therapeutic agent is administered for less than about a month, less than about three weeks, less than about two weeks, or less than about one week. In yet another embodiment the administration of the compound of Formula (I) continues for more than about 6, 10, 14, or 28 days. In some cases, continuous dosing is achieved and maintained as long as necessary. [0042] In some embodiments, the compound of Formula (I) or pharmaceutically acceptable salt thereof is administered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for the compound of Formula (I) or pharmaceutically acceptable salt thereof may be found by routine experimentation in light of the instant disclosure. [0043] In some embodiments, the compound of Formula (I) or pharmaceutically acceptable salt thereof is formulated into pharmaceutical compositions. In specific embodiments, pharmaceutical compositions are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and/or auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any pharmaceutically acceptable techniques, carriers, diluents and excipients are used as suitable to formulate the pharmaceutical compositions described herein: Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins1999). [0044] In certain embodiments, the compound of Formula (I) or pharmaceutically acceptable salt thereof described are administered as pharmaceutical compositions in which the compound of Formula (I) or pharmaceutically acceptable salt thereof is mixed with other active ingredients (e.g., additional therapeutic agents), as in combination therapy. Encompassed herein are all combinations of actives set forth in the combination therapies section below and throughout this disclosure. [0045] A pharmaceutical composition, as used herein, refers to a mixture of the compound of Formula (I) or pharmaceutically acceptable salt thereof with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients. In certain embodiments, the pharmaceutical composition facilitates administration of the compound of Formula (I) or pharmaceutically acceptable salt thereof to a subject. In some embodiments, practicing the methods of treatment or use provided herein, therapeutically effective amounts of the compound of Formula (I) or pharmaceutically acceptable salt thereof provided herein are administered in a pharmaceutical composition to a mammal having a disease, disorder or medical condition to be treated. In specific embodiments, the mammal is a human. In certain embodiments, therapeutically effective amounts vary depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. The compound of Formula (I) or pharmaceutically acceptable salt thereof described herein are used singly or in combination with one or more therapeutic agents as components of mixtures. [0046] In some embodiments, the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein is formulated for oral administration. Compounds described herein are formulated by combining the active compounds (i.e., a compound of Formula (I) or a pharmaceutically acceptable salt thereof and, optionally, additional therapeutic agents) with, e.g., pharmaceutically acceptable carriers or excipients. In various embodiments, the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein are formulated in oral dosage forms that include, by way of example, tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like. [0047] In certain embodiments, pharmaceutical preparations for oral use are obtained by mixing one or more solid excipient, with the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. In specific embodiments, disintegrating agents are optionally added. Disintegrating agents include, by way of example, cross-linked croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. [0048] In one embodiment, dosage forms, such as dragee cores and tablets, are provided with one or more suitable coating. In specific embodiments, concentrated sugar solutions are used for coating the dosage form. The sugar solutions, optionally contain additional components, such as by way of example, gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs and/or pigments are also optionally added to the coatings for identification purposes. Additionally, the dyestuffs and/or pigments are optionally utilized to characterize different combinations of active compound doses (i.e., a compound of Formula (I) or a pharmaceutically acceptable salt thereof and, optionally, additional therapeutic agents). [0049] In certain embodiments, therapeutically effective amounts of the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein are formulated into other oral dosage forms. Oral dosage forms include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. In specific embodiments, push-fit capsules contain the active ingredients in admixture with one or more filler. Fillers include, by way of example, lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In other embodiments, soft capsules contain the compound of Formula (I) or pharmaceutically acceptable salt thereof that is dissolved or suspended in a suitable liquid. Suitable liquids include, by way of example, one or more fatty oil, liquid paraffin, or liquid polyethylene glycol. In addition, stabilizers are optionally added. [0050] In other embodiments, therapeutically effective amounts of the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein are formulated for buccal or sublingual administration. Formulations suitable for buccal or sublingual administration include, by way of example, tablets, lozenges, or gels. In still other embodiments, the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein are formulated for parental injection, including formulations suitable for bolus injection or continuous infusion. In specific embodiments, formulations for injection are presented in unit dosage form (e.g., in ampoules) or in multi-dose containers. Preservatives are, optionally, added to the injection formulations. In still other embodiments, the pharmaceutical compositions are formulated in a form suitable for parenteral injection as sterile suspensions, solutions or emulsions in oily or aqueous vehicles. Parenteral injection formulations optionally contain formulatory agents such as suspending, stabilizing and/or dispersing agents. In specific embodiments, pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. In additional embodiments, suspensions of the compound of Formula (I) or pharmaceutically acceptable salt thereof are prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles for use in the pharmaceutical compositions described herein include, by way of example, fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. In certain specific embodiments, aqueous injection suspensions contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension contains suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, in other embodiments, the active ingredient(s) (i.e., a compound of Formula (I) or a pharmaceutically acceptable salt thereof and, optionally, additional therapeutic agents) is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. [0051] In certain embodiments, pharmaceutical compositions are formulated in any conventional manner using one or more physiologically acceptable carriers, diluents or excipients which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any pharmaceutically acceptable techniques, carriers, and excipients are optionally used as suitable. Pharmaceutical compositions comprising the compound of Formula (I) or pharmaceutically acceptable salt thereof are manufactured in a conventional manner, such as, by way of example, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes. [0052] Additionally, the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein encompasses unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compound of Formula (I) or pharmaceutically acceptable salt thereof presented herein are also considered to be disclosed herein. In addition, the pharmaceutical compositions optionally include other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, buffers, and/or other therapeutically valuable substances. [0053] Methods for the preparation of compositions comprising the compound of Formula (I) or pharmaceutically acceptable salt thereof described herein include formulating the compound of Formula (I) or pharmaceutically acceptable salt thereof with one or more inert, pharmaceutically acceptable carriers, diluents or excipients to form a solid, semi-solid or liquid. Solid compositions include powders, tablets, dispersible granules, capsules, cachets, and suppositories. Liquid compositions include solutions in which the compound of Formula (I) or pharmaceutically acceptable salt thereof is dissolved, emulsions comprising a compound, or a solution containing liposomes, micelles, or nanoparticles comprising the compound of Formula (I) or pharmaceutically acceptable salt thereof as disclosed herein. Semi-solid compositions include gels, suspensions and creams. The form of the pharmaceutical compositions described herein include liquid solutions or suspensions, solid forms suitable for solution or suspension in a liquid prior to use, or as emulsions. These compositions also optionally contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and so forth. [0054] In some embodiments, the pharmaceutical composition comprising the compound of Formula (I) or pharmaceutically acceptable salt thereof illustratively takes the form of a liquid where the agents are present in solution, in suspension or both. Typically, when the composition is administered as a solution or suspension a first portion of the therapeutic agent (e.g., a compound of Formula (I)) is present in solution and a second portion of the therapeutic agent (e.g., a compound of Formula (I)) is present in particulate form, in suspension in a liquid matrix. In some embodiments, a liquid composition includes a gel formulation. In other embodiments, the liquid composition is aqueous. [0055] In certain embodiments, aqueous suspensions contain one or more polymers as suspending agents. Useful polymers include water-soluble polymers such as cellulosic polymers, e.g., hydroxypropyl methylcellulose, and water-insoluble polymers such as cross-linked carboxyl-containing polymers. Certain pharmaceutical compositions described herein comprise a mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer), poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran. [0056] Useful pharmaceutical compositions also, optionally, include solubilizing agents to aid in the solubility of the compound of Formula (I) or pharmaceutically acceptable salt thereof. The term "solubilizing agent" generally includes agents that result in formation of a micellar solution or a true solution of the agent. Certain acceptable nonionic surfactants, for example polysorbate 80, are useful as solubilizing agents, as can ophthalmically acceptable glycols, polyglycols, e.g., polyethylene glycol 400, and glycol ethers. [0057] Furthermore, pharmaceutical compositions optionally include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride. Such acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range. [0058] Additionally, compositions also, optionally, include one or more salts in an amount required to bring osmolality of the composition into an acceptable range. Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate. [0059] Other pharmaceutical compositions optionally include one or more preservatives to inhibit microbial activity. Suitable preservatives include mercury- containing substances such as merfen and thiomersal; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride. [0060] Still other compositions include one or more surfactants to enhance physical stability or for other purposes. Suitable nonionic surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40. [0061] Still other compositions include one or more antioxidants to enhance chemical stability where required. Suitable antioxidants include, by way of example, ascorbic acid and sodium metabisulfite. [0062] In certain embodiments, aqueous suspension compositions are packaged in single-dose non-reclosable containers. Alternatively, multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition. [0063] In alternative embodiments, other delivery systems for hydrophobic pharmaceutical compounds are employed. Liposomes and emulsions are examples of delivery vehicles or carriers useful herein. In certain embodiments, organic solvents such as N-methylpyrrolidone are also employed. In additional embodiments, the compounds described herein are delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials are useful herein. In some embodiments, sustained-release capsules release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization are employed. [0064] In certain embodiments, the formulations described herein comprise one or more antioxidants, metal chelating agents, thiol containing compounds and/or other general stabilizing agents. Examples of such stabilizing agents, include: (a) about 0.5% to about 2% w/v glycerol, (b) about 0.1% to about 1% w/v methionine, (c) about 0.1% to about 2% w/v monothioglycerol, (d) about 1 mM to about 10 mM EDTA, (e) about 0.01% to about 2% w/v ascorbic acid, (f) about 0.003% to about 0.02% w/v polysorbate 80, (g) 0.001% to about 0.05% w/v. polysorbate 20, (h) arginine, (i) heparin, (j) dextran sulfate, (k) cyclodextrins, (l) pentosan polysulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n) combinations thereof. [0065] In some embodiments, the concentration of the compound of Formula (I) or pharmaceutically acceptable salt thereof provided in the pharmaceutical compositions is less than about 100%, about 90%, about 80%, about 70%, about 60%, about 50%, about 40%, about 30%, about 20%, about 19%, about 18%, about 17%, about 16%, about 15%, about 14%, about 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1%, about 0.5%, about 0.4%, about 0.3%, about 0.2%, about 0.1%, about 0.09%, about 0.08%, about 0.07%, about 0.06%, about 0.05%, about 0.04%, about 0.03%, about 0.02%, about 0.01%, about 0.009%, about 0.008%, about 0.007%, about 0.006%, about 0.005%, about 0.004%, about 0.003%, about 0.002%, about 0.001%, about 0.0009%, about 0.0008%, about 0.0007%, about 0.0006%, about 0.0005%, about 0.0004%, about 0.0003%, about 0.0002%, or about 0.0001% w/w, w/v or v/v. [0066] In some embodiments, the concentration of the compound of Formula (I) or pharmaceutically acceptable salt thereof is in the range from approximately 0.0001% to approximately 50%, approximately 0.001% to approximately 40 %, approximately 0.01% to approximately 30%, approximately 0.02% to approximately 29%, approximately 0.03% to approximately 28%, approximately 0.04% to approximately 27%, approximately 0.05% to approximately 26%, approximately 0.06% to approximately 25%, approximately 0.07% to approximately 24%, approximately 0.08% to approximately 23%, approximately 0.09% to approximately 22%, approximately 0.1% to approximately 21%, approximately 0.2% to approximately 20%, approximately 0.3% to approximately 19%, approximately 0.4% to approximately 18%, approximately 0.5% to approximately 17%, approximately 0.6% to approximately 16%, approximately 0.7% to approximately 15%, approximately 0.8% to approximately 14%, approximately 0.9% to approximately 12%, approximately 1% to approximately 10% w/w, w/v or v/v. [0067] In some embodiments, the concentration of the compound of Formula (I) or pharmaceutically acceptable salt thereof is in the range from approximately 0.001% to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%, approximately 0.03% to approximately 4%, approximately 0.04% to approximately 3.5%, approximately 0.05% to approximately 3%, approximately 0.06% to approximately 2.5%, approximately 0.07% to approximately 2%, approximately 0.08% to approximately 1.5%, approximately 0.09% to approximately 1%, approximately 0.1% to approximately 0.9% w/w, w/v or v/v. [0068] In some embodiments, the amount of the compound of Formula (I) or pharmaceutically acceptable salt thereof is equal to or less than about 10 g, about 9.5 g, about 9.0 g, about 8.5 g, about 8.0 g, about 7.5 g, about 7.0 g, about 6.5 g, about 6.0 g, about 5.5 g, about 5.0 g, about 4.5 g, about 4.0 g, about 3.5 g, about 3.0 g, about 2.5 g, about 2.0 g, about 1.5 g, about 1.0 g, about 0.95 g, about 0.9 g, about 0.85 g, about 0.8 g, about 0.75 g, about 0.7 g, about 0.65 g, about 0.6 g, about 0.55 g, about 0.5 g, about 0.45 g, about 0.4 g, about 0.35 g, about 0.3 g, about 0.25 g, about 0.2 g, about 0.15 g, about 0.1 g, about 0.09 g, about 0.08 g, about 0.07 g, about 0.06 g, about 0.05 g, about 0.04 g, about 0.03 g, about 0.02 g, about 0.01 g, about 0.009 g, about 0.008 g, about 0.007 g, about 0.006 g, about 0.005 g, about 0.004 g, about 0.003 g, about 0.002 g, about 0.001 g, about 0.0009 g, about 0.0008 g, about 0.0007 g, about 0.0006 g, about 0.0005 g, about 0.0004 g, about 0.0003 g, about 0.0002 g, or about 0.0001 g. Methods of Use [0069] Some embodiments of the present disclosure are directed to methods of treating a subject for a viral infection. In some embodiments, the present disclosure provides methods of treating a subject for an infection with a virus (e.g., coronavirus such as SARS-CoV-2) comprising administering to the subject an effective amount of a compound of Formula (I), or a pharmaceutical composition comprising a compound of Formula (I), such a pacritinib. [0070] Patients or subjects that can be treated by the methods disclosed herein include, but are not limited to, a mammal, such as human or non-human primates (e.g., monkeys and apes). In some embodiments, the subject is human. The subject can be male or female and can be any suitable age, including infant, juvenile, adolescent, adult, and geriatric. [0071] In some embodiments, the subject is infected with a virus that utilizes the clathrin mediated endocytosis entry pathway. Clathrin mediated endocytosis is a process by which cells absorb metabolites, hormones, proteins, and in some cases viruses, by the inward budding of the plasma membrane (invagination). Viruses that utilize the clathrin mediated endocytosis pathway for entry include coronaviruses (e.g., SARS-CoV1 and SARS-CoV-2), ebola virus, influenza A virus, vesicular stomatitis virus, and hepatitis C virus. [0072] Proteins involved in clathrin mediated endocytosis include “AP2- associated protein kinase 1” or “AAK1,” and “cyclin G-associated kinase” or “GAK.” Inhibitors of AAK1 and GAK are known to block viral infectivity of viruses that utilize the clathrin mediated endocytosis pathway, such as ebola virus, Marburg virus, hepatitis C virus, and Dengue virus. SARS-CoV-1, SARS-CoV2, and MERS-CoV are also known to utilize clathrin mediated endocytosis. Without being bound by theory, as demonstrated in Example 3, a compound of Formula (I), pacritinib, surprisingly interacts with both AAK1 and GAK and thus the compound may have antiviral effects by inhibiting clathrin mediated endocytosis. [0073] In one aspect, the subject has viral-induced hyperinflammation. Following viral infection and proliferation, macrophages migrate to the infection site to kill infected cells, leading to the release of inflammatory cytokines and chemokines. The response to this stimuli is local inflammation and increased blood flow, enabling plasma and leukocytes to travel to the infection site. However, severe viral infections are characterized by over-induction of this immune response, termed “hyperinflammation,” Hyperinflammation is characterized by severe, systemic, and uncontrolled immune cell activation and inflammatory cytokine/chemokine production in response to a persistent trigger such as viremia. Symptoms of hyperinflammation may include unremitting fever, splenomegaly, coagulopathy, hepatitis, cytopenia and, if unrestrained, the hyperinflammation may cause multi-organ failure and death. In some embodiments, hyper-inflammation is associated with a viral infection and is a cytokine storm or Acute respiratory distress syndrome (ARDS). [0074] “Cytokine storm,” also known as “cytokine release syndrome,” refers to an acute systemic inflammatory response that can be induced by a variety of factors such as infection by a virus, therapeutic antibodies, or cellular immunotherapy. Symptoms include fever, nausea, fatigue and headache. Potentially life-threatening complications of cytokine storm include, renal failure, hepatic failure, respiratory distress, and cardiac dysfunction. Soluble mediators of inflammation, including cytokines and chemokines, contribute to the pathophysiology of cytokine storm. These inflammatory cytokines/chemokines include IL-6, TNFα, IFNγ, IL-1β, IL-2, IL-6, IL-8, and IL-10. Elevated levels of inflammatory cytokines have been identified in patients with viral infections, including Orthomyxoviridae influenza A infections. Cytokine storms are associated with a number of other viral infections, including Epstein-Barr virus of the Herpesviridae family, group A Streptococcus pyogenes, and Orthopoxvirus variola. Betacoronavirus infection, including infection by the SARS-CoV-1, which causes severe acute respiratory syndrome (SARS) is also associated with a cytokine storm characterized by elevated levels of inflammatory cytokines and immunopathological damage. [0075] In some embodiments, a virus associated with hyperinflammation is “SARS-CoV-1”. Used herein, the term “SARS-CoV-1” refers to the strain of Betacoronavirus that emerged in 2003 and causes severe acute respiratory syndrome (SARS). [0076] In some embodiments, a virus associated with hyperinflammation is “SARS-CoV-2”. Used herein, the term “SARS-CoV-2” refers to the strain of Betacoronavirus that emerged in 2019 and causes the respiratory illness Coronavirus disease 2019 (COVID-19). [0077] In some embodiments, subject infected with a coronavirus has COVID- 19. As used herein, “COVID-19” refers to the infectious disease caused by SARS- CoV-2 and characterized by, for example, fever, cough, respiratory symptoms, rhinorrhea, sore throat, malaise, headache, chills, repeated shaking with chills, diarrhea, new loss of smell or taste, muscle pain, or a combination thereof. [0078] In some embodiments, the subject exhibits one or more symptoms associated with COVID-19 or exhibits no symptoms associated with COVID-19 (asymptomatic). In reference to treatment of patients with different COVID-19 disease severity, “asymptomatic” infection refers to patients diagnosed with COVID-19 by a standardized assay that do not present with COVID-19 symptoms (e.g., fever, cough, respiratory symptoms, rhinorrhea, sore throat, malaise, headache, or muscle pain). [0079] COVID-19 symptoms may include mild symptoms such as fever, rhinorrhea, mild cough, sore throat, malaise, headache, muscle pain, or any combination thereof, but with no shortness of breath. Patients with “mild” COVID-19 infection present no signs of lower airway disease and have a normal respiratory rate and heart rate, and oxygen saturation (pulse oximetry) greater than 93% on room air. [0080] COVID-19 symptoms may include moderate symptoms such as lower respiratory symptoms, including shortness of breath (at rest or with exertion) or signs of pneumonia, such as reduced oxygen saturation (pulse oximetry). [0081] In some embodiments, the subject has severe COVID-19. “Severe COVID-19” is defined as confirmed disease in patients who are hospitalized with hypoxia [SpO2 ≤93% on room air], respiratory rate >30, PaO2/FiO2 <300, but do not require intermittent mandatory ventilation (IMV). [0082] In some embodiments, the subject exhibits no symptoms associated with COVID-19 but has been exposed to another subject known or suspected of having COVID-19. [0083] In some embodiments, the subject with a coronavirus exhibits one or more symptoms selected from dry cough, shortness of breath, and fever. [0084] In some embodiments, the subject has been hospitalized prior to treatment. In some embodiments, the subject has had severe COVID-19 with SARS- CoV-2 infection confirmed by either a) a positive reverse transcriptase polymerase chain reaction (RT PCR) or b) an antigen-based test from any respiratory, nasopharyngeal, saliva, blood, or stool specimen, documented within 1 week prior to the start of treatment. In some embodiments, the subject has cancer. In some embodiments, the subject does not have cancer. [0085] In some embodiments, the subject has viral-induced hyperinflammation. The viral induced hyperinflammation may be a cytokine storm or acute respiratory distress syndrome (ARDS). ARDS refers to a respiratory condition characterized by severe hypoxemia and may be induced by viral infection. ARDS is characterized by severe impairment in lung gas exchange and lung mechanics. The innate immune response is overactive in ARDS. Virally-induced inflammation promotes pulmonary epithelial and endothelial cellular damage leading to increased capillary permeability. Patients with ARDS may exhibit buildup of fluid in the lung and have reduced oxygen levels in the blood. [0086] Additionally, the present disclosure also provides prophylactic treatment of a subject prior to receipt of a test result for viral infection (e.g., SARS-CoV-2). The subject may be at risk of contracting the virus. Prophylactic treatment would be administered to individuals at risk of exposure to infected patients, for example, immunocompromised individuals, medical personnel, people with known exposure, and essential workers. [0087] In some embodiments, treatment of a subject that does not have respiratory symptoms associated with either COVID-19 or ARDS reduces the likelihood of the subject developing one or more of mild, moderate, or severe COVID- 19 or ARDS symptoms. [0088] In some embodiments, treatment of a subject according to the methods described herein results in reduction of the severity or duration of the severe viral infection symptoms. Severe viral infection symptoms may include severe respiratory symptoms such as, for example, shortness of breath, difficulty breathing, reduced respiratory rate, and wheezing. [0089] In some embodiments, the methods of treatment described herein result in reduction of one or more inflammatory cytokines and/or chemokines. Inflammatory cytokines or chemokines include, for example, CD40L, EGF, Eotaxin (CCL11), FGF-2, Flt-3 ligand, Fractalkine, G-CSF, GM-CSF, GROα (CXCL1), IFN-α2, IFN-γ, IL-1α, IL-1β, IL-1RA, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (CXCL8), IL-9, IL-10, IL-12 (p40), IL-12 (p70), IL-13, IL-15, IL-17A, IL-17E/IL-25, IL-17F, IL-18, IL-22, IL-27, IP-10 (CXCL10), MCP-1 (CCL2), MCP-3, M-CSF, MDC (CCL22), MIG (CXCL9), MIP-1α (CCL3), MIP-1β (CCL4), PDGF-AA, PDGF-AB/BB, RANTES (CCL5), TGF- α, TNF-α, TNF-β, TNF-r1, and VEGF-A). The level of the inflammatory cytokine and/or chemokines may be measured in blood plasma, e.g., by enzyme-linked immunosorbent assays (ELISA), bead-based immunoassays and other immunoassays, and/or the transcriptional level of inflammatory cytokines and/or chemokines may be measured by RNA sequencing. [0090] In some embodiments, treatment of a subject according to the methods described herein results in reduction of a symptom associated hyperinflammation. In some embodiments, hyperinflammation is cytokine release syndrome (or cytokine storm) or acute respiratory distress syndrome (ARDS). [0091] In some embodiments, treatment of a subject according to the methods described herein results in reduction of viral load in the subject. In some embodiments, treatment of a subject according to the methods described herein results in reduction of SARS-CoV-2 viral load. The reduction in viral load may be a reduction in plasma viral load. In some embodiments, the viral load is measured by PCR. In some embodiments, the virus is SARS-CoV-2 and the plasma SARS-CoV-2 viral load is measured by detecting the nucleocapsid (N) gene. As shown in Examples 1 and 2, use of a compound of Formula (I), pacritinib, surprisingly results in decreased viral infectivity of SARS-CoV-2. [0092] In some embodiments, treatment of a subject according to the methods described herein results in reduced duration or occurrence of hospitalization, ventilation or dialysis of the subject. [0093] In some embodiments, the treating results a reduced likelihood of the subject requiring intermittent mandatory ventilation (IMV) or extracorporeal membrane oxygenation (ECMO), as compared to a subject with similar symptoms who has not been treated with the compound of Formula (I). [0094] “Extracorporeal membrane oxygenation” or “ECMO” refers to a treatment that uses a pump to circulate blood through an artificial lung back into the bloodstream. [0095] “Intermittent mandatory ventilation” or “IMV” refers to a ventilation mode that provides partial mechanical assistance, whereby mandatory breaths are delivered at a set frequency, tidal volume, and inspiratory flow rate. [0096] In some embodiments, treatment of a subject according to the methods described herein results in the subject having a faster and/or more extensive recovery than a subject with similar symptoms who has not been treatment with the compound of Formula (I). [0097] An appropriate dose, suitable duration, and frequency of administration of the compound of Formula (I) will be determined by such factors as the condition of the patient, size, weight, body surface area, age, sex, type and severity of the disease, particular therapy to be administered, particular form of the active ingredient, time and the method of administration, and other drugs being administered concurrently, which can readily be determined by a person skilled in the art. [0098] Certain aspects of the first embodiment include administering an effective amount of pacritinib. Pacritinib may be effective over a wide dosage range. In certain embodiments, the effective amount ranges from 0.01 to 2000 mg, from 1 to 1000 mg per day, from 50 to 500 mg per day, or from 200 to 400 mg per day. In particular embodiments, the effective amount ranges from between about 50 mg per day to about 500 mg per day, or is about 200mg per day. In various embodiments, the effective amount is 50, 100, 150, 200, 250, 300, 350, 400, 450, or 500 mg per day. [0099] Some aspects of the first embodiment include administering one or more further therapeutic agents, such as an antiviral agent or an agent to reduce inflammation, to provide a synergistic or additive therapeutic effect. [0100] Agents to reduce inflammation include checkpoint inhibitors. As used herein, the term “checkpoint molecule” refers to one or more proteins, molecules, compounds or complexes providing inhibitory signals to assist in controlling or suppressing an immune response. For example, immune checkpoint molecules include those molecules that partially or totally block immune stimulation; decrease, prevent or delay immune activation; or increase, activate, or up regulate immune suppression. Exemplary checkpoint molecules are described in further detail herein and include PD-1, PD-L1, PD-L2, CD80, CD86, B7-H3, B7-H4, HVEM, adenosine, GAL9, VISTA, CEACAM-1, PVRL2, CTLA-4, BTLA, KIR, LAG3, TIM3, A2aR, CD244/2B4, CD160, TIGIT, LAIR-1, PVRIG/CD112R, and certain metabolic enzymes, such as arginase, indoleamine 2,3-dioxygenase (IDO). [0101] As used herein, the term “antiviral agents” refers to a class of drugs for treating viral infections. An antiviral agent may be, for example, a small molecule, peptide, protein, antibody, nucleic acid, or aptamer that targets one or more components in the viral life cycle: attachment to host cell; release of viral genes and possibly enzymes into the host cell; replication of viral components using host cell machinery; assembly of viral components into viral particles; and release of viral particles to infect new host cells. Targets of antiviral agents include critical viral proteins such as neuraminidase, M2 ion channel protein, hemagglutinin, viral RNA polymerase, NTPase/helicase, spike (S) glycoprotein (S1 domain), and 3C-like cysteine protease. Examples of anti-viral agents include seltamivir, zanamivir, laninamivir, laninamivir, peramivir, and remdesivir. [0102] The present disclosure also provides a therapeutic composition comprising a compound of Formula (I) and at least one of: an anti-viral therapeutic agent, and an agent to reduce hyperinflammation. EXAMPLES EXAMPLE 1
Figure imgf000039_0001
[0103] This Example demonstrates that pacritinib, which is a compound of Formula (I), reduced the cytopathic effects (CPE) of SARS-CoV-2. Confluent or near- confluent cell culture monolayers of Vero 76 cells were prepared in 96-well disposable microplates one day prior to testing. Cells were maintained in MEM media supplemented with 2% fetal bovine serum (FBS). For antiviral assays the same medium was used but supplemented with 50-µg/ml gentamicin. Pacritinib was dissolved in DMSO, and was prepared at four serial log10 concentrations: 0.1, 1.0, 10, and 100 µM equivalent to 0.06, 0.6, 6, and 60 µM of pacritinib free base. Five microwells are used per dilution: three for infected cultures and two for uninfected toxicity cultures. Controls for the experiment consist of six microwells that were infected and not treated (virus controls) and six that were untreated and uninfected (cell controls) on every plate. A known protease inhibitor was tested in parallel as a positive control drug using the same method as was applied for pacritinib. The positive control was tested with every test run. [0104] Growth media was removed from the cells and the test compound was applied in 0.1 ml volume to wells at 2X concentration. Virus, at ~60 CCID50 (50% cell culture infectious dose) in 0.1 ml volume was added to the wells designated for virus infection. Medium devoid of virus was placed in toxicity control wells and cell control wells. Plates were incubated at 37 oC with 5% CO2 until marked CPE (>80% CPE) was observed in virus control wells. The plates were then stained with 0.011% neutral red for approximately two hours at 37oC in a 5% CO2 incubator. The neutral red medium was removed by complete aspiration, and the cells were rinsed 1X with phosphate buffered solution (PBS) to remove residual dye. The PBS was completely removed, and the incorporated neutral red was eluted with 50% Sorensen’s citrate buffer/50% ethanol for 30 minutes. Neutral red dye penetrates into living cells, thus, the presence of intense red color, indicates that viable cells are present in the wells. The dye content in each well was quantified using a spectrophotometer at 540 nm wavelength. The dye content in each set of wells was converted to a percentage of dye present in untreated control wells using a Microsoft Excel computer-based spreadsheet and normalized based on the virus control. The 50% effective (EC50, virus-inhibitory) concentrations and 50% cytotoxic (CC50, cell-inhibitory) concentrations were then calculated by regression analysis. The quotient of CC50 divided by EC50 gives the selectivity index (SI) value. Compounds showing SI values of approximately 10 or higher are considered active. The results are shown in Table 1. Table 1.
Figure imgf000040_0001
EXAMPLE 2 A COMPOUND OF FORMULA (I) DEMONSTRATES ANTIVIRAL ACTIVITY AGAINST SARS- COV-2 IN A SECONDARY ASSAY [0105] This Example demonstrates that pacritinib, which is a compound of formula (I), reduces the viral replication of SARS-CoV-2 in cell culture. [0106] First, a Viral Yield Reduction (VYR) assay was performed. Virus yielded in the presence of pacritinib was titrated and compared to virus titers from the untreated virus controls. Titration of the viral samples (collected as described in the paragraph above) was performed by endpoint dilution (Reed and Muench). Serial 1/10 dilutions of virus were made and plated into 4 replicate wells containing fresh cell monolayers of Vero 76 cells. Plates were then incubated, and cells are scored for presence or absence of virus after distinct CPE is observed, and the CCID50 calculated using the Reed-Muench method (24). The 90% (one log10) effective concentration (EC90) was calculated by regression analysis by plotting the log10 of the inhibitor concentration versus log10 of virus produced at each concentration. Dividing EC90 by the CC50 gives the SI value for this test. The results of the Viral Yield Reduction (VYR) assay are shown in Table 2. Table 2.
Figure imgf000041_0001
[0107] After sufficient virus replication of SARS-CoV-2 (3 days), a sample of supernatant was taken from each infected well (three replicate wells were pooled) and tested for titer determination. Titration of the viral samples was performed by serial dilution and plating into Vero 76 cells. After incubation as described in Example 1, the cells were scored. After maximum CPE was observed (at least 80% CPE in virus control wells), the viable plates were stained with neutral red dye. The incorporated dye content was quantified as described above to generate the EC50 and CC50 values. SI = CC50 / EC50. The results are shown in Table 3. Table 3.
Figure imgf000042_0002
EXAMPLE 3 INHIBITION OF VIRAL ENTRY REGULATORS AAK1 AND GAK BY A COMPOUND OF FORMULA (I) [0108] This Example demonstrates the ability of pacritinib, which is a compound of Formula (I), to engage with AAK1 and GAK, which are both important kinases involved in clathrin mediated endocytosis. HEK293 cells transiently expressing NanoLuc®- AAK1 or GAK Fusion Vector were seeded into the wells of 384-well plates. The cells were pre-treated with the NanoBRETTM Tracer K-5 and then treated with reference compound CTx-0294885 for 1 hour. The BRET signal was measured on an Envision 2104 Multilabel Reader. IC50 value was calculated and IC50 curve was plotted using the GraphPad Prism 4 program based on a sigmoidal dose response equation. The results are shown in Table 4. Table 4.
Figure imgf000042_0001
EXAMPLE 4 TREATMENT OF SEVERE COVID-19 WITH OR WITHOUT CANCER USING A COMPOUND OF FORMULA (I) [0109] This is a Phase 3 randomized, double-blind, placebo-controlled, multicenter study to evaluate the efficacy and safety of pacritinib in hospitalized patients with severe COVID-19 with or without cancer. Severe COVID-19 is defined as confirmed disease in patients who are hospitalized with hypoxia (blood oxygen saturation [SpO2] ≤93% on room air at sea level), respiratory rate >30, arterial oxygen partial pressure [PaO2]/ fraction of inspired oxygen [FiO2] <300, or lung infiltrates >50% but do not require intermittent mandatory ventilation (IMV). [0110] Patients will be randomized 1:1 to receive pacritinib (400 mg once daily [QD] on Day 1, then 200 mg twice daily [BID] from Day 2 to Day 14) + SOC or placebo + SOC. [0111] The inclusion criteria will include: ● hospitalization prior to randomization for the treatment of severe COVID-19 with SARS-CoV-2 infection confirmed by either a) a positive reverse transcriptase polymerase chain reaction (RT PCR) or b) an antigen-based test from any respiratory, nasopharyngeal, saliva, blood, or stool specimen at Screening or documented within 1 week prior to the start of Screening. ● Age ≥ 18 years ● Platelet count ≥ 50,000/µL ● If fertile, willing to use effective birth control methods during the study ● Provision of informed consent within 96 hours after hospitalization [0112] The exclusion criteria will include: ● In the opinion of the investigator, progression to death is imminent and inevitable within the next 24 hours, irrespective of the provision of treatments ^ ^ Currently intubated or intubated between screening and randomization ^ ^ Suspected active uncontrolled bacterial, fungal, viral, or other infection (besides COVID 19) ^ º Prior allogenic hematopoietic stem cell transplantation ^ ^ Active lung cancer or history of lung cancer within the past 12 months º ^ Any active grade 2 or higher hemorrhage ^ ^ Any active gastrointestinal or metabolic condition that could interfere with absorption of oral medication ^ º ^ Uncontrolled intercurrent illness that, in the judgment of the treating physician, would limit compliance with study requirements ^ ^ Known seropositivity for human immunodeficiency virus with cluster of differentiation 4 (CD4) count < 200/mm3 within 3 months prior to randomization ^ ^ Pregnant or breastfeeding, or positive pregnancy test in a pre- dose examination ^ ^ Concurrent enrollment in another interventional trial (investigational COVID-19 antiviral studies are permitted) ^ ^ Serum creatinine > 2.5 mg/dL ^ ^ Total bilirubin > 4× the upper limit of normal ^ ^ QT corrected by the Fridericia method (QTcF) prolongation > 480 msec ^ ^ Known history of New York Heart Association Class II, III, or IV congestive heart failure prior to hospital admission ^ ^ Known allergic reaction to any Janus kinase 2 (JAK2) inhibitor ^ ^ Exposure to any JAK2 inhibitor within 28 days ^ ^ Currently receiving a strong CYP3A4 inhibitor or strong P450 inducer (and unable to stop the medication prior to the first dose of study drug and throughout the duration of study drug administration ^ ^ Treatment with cytoreductive chemotherapy administered within 14 days prior to randomization ^ ^ Administration of an IL-1 or IL-6 blocking immunomodulatory agent (such as tocilizumab, canakinumab, sarilumab, anakinra) within 48 hours prior to randomization ^ ^ Currently receiving therapeutic anticoagulation or anti platelet medication and unable to stop the medication prior to randomization. Prophylactic anticoagulation therapy or aspirin (≤ 100mg) are permitted. ^ ^ Unable to ingest capsules or tablets at randomization [0113] Assigned treatment will continue for up to Day 14 or until the patient experiences intolerable adverse events (AEs), withdraws consent, or initiates another investigational therapy or until the study is terminated. Assigned therapy may be given for an additional 7 days (for a total of 21 days) if, in the opinion of the investigator, the patient's clinical signs and symptoms are improving and the potential benefit outweighs the potential risk. In the event of hospital discharge, patients will complete treatment with the assigned therapy as an outpatient. [0114] For the experimental arm, pacritinib 400 mg will be administered once daily [QD] on Day 1, then 200 mg will be administered twice daily [BID] from Day 2 to Day 14, along with standard of care (SOC) treatment. For the placebo arm, 4 placebo capsules will be administered once daily [QD] on Day 1, then 2 capsules will be administered twice daily [BID] from Day 2 to Day 14) + SOC. Both pacritinib and the placebo will be provided in 100 mg capsules. [0115] The primary outcome measured will be the proportion of patients who progress to intermittent mandatory ventilation (IMV) and/or extracorporeal membrane oxygenation (ECMO) or death during the 28 days following randomization. The proportion will be calculated as the number of patients who progress divided by the total number of patients in the study population. [0116] U.S. Provisional Patent Application No.63/077,461, filed September 11, 2020, to which the present application claims priority, is hereby incorporated herein by reference in its entirety.

Claims

CLAIMS 1. A method of treating a subject infected with a virus, comprising administering to the subject an effective amount of a compound of Formula I:
Figure imgf000046_0002
wherein: R1 and R2 are H; Z2 is–N(H)-; Ar1 is selected from the group consisting of:
Figure imgf000046_0001
wherein R10 is methoxy or fluorine; k is an integer selected from 0 and 1; Ar2 is a group of the formula
Figure imgf000047_0001
wherein R11 is H or selected from the group consisting of:
Figure imgf000047_0002
L is a group of formula:
Figure imgf000047_0003
wherein X1 is attached to Ar1 and X2 is attached to Ar2, and wherein X1, X2 and Y are selected such that the group L has from 5 to 15 atoms in normal linear chain connecting Ar1 to Ar2, wherein X1 is selected from the group consisting of: (a)-OCH2- (b) –OCH2CH2-, and (c)–CH2OCH2-; wherein X2 is selected from the group consisting of: (a) –CH2O-, (b)–CH2CH2O-, and (c)–CH2OCH2-; Y is a group of formula –CRa=CRb-, wherein Ra and Rb are H, or a pharmaceutically acceptable salt or N-oxide thereof.
2. The method according to claim 1, wherein X1 is -CH2OCH2-.
3. The method of claim 1 or 2, wherein X2 is -CH2OCH2-.
4. The method of any one of claims 1-3, wherein Y is selected from the group consisting of:
Figure imgf000048_0001
.
5. The method of claim any one of claims 1-4, wherein the compound of Formula (I) is selected from the group consisting of:
Figure imgf000048_0002
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
and pharmaceutically acceptable salts thereof.
6. The method of any of claims 1-5, wherein the compound of Formula (I) has the following formula:
Figure imgf000056_0002
or a pharmaceutically acceptable salt thereof.
7. The method of any of claims 1-5, wherein the compound of Formula (I) has the following formula:
Figure imgf000056_0003
or a pharmaceutically acceptable salt thereof.
8. The method of any of claims 1-7, wherein the virus is selected from a coronavirus, ebola virus, hepatitis C virus, and Dengue virus.
9. The method of claim 8, wherein the virus is a coronavirus.
10. The method of claim 9, wherein the coronavirus is selected from SARS- CoV-1, SARS-CoV-2, and MERS-CoV.
11. The method of claim 9, wherein the coronavirus is SARS-CoV-2.
12. The method of claim 11, wherein the subject exhibits one or more symptoms associated with mild-to-moderate COVID-19.
13. The method of claim 12, wherein the one or more symptoms are selected from sore throat, loss of taste or smell, dry cough, shortness of breath, and fever.
14. The method of claim 11, wherein the subject exhibits one or more symptoms associated with severe COVID-19.
15. The method of claim 14, wherein the subject has acute respiratory distress syndrome (ARDS).
16. The method of one of claim 11, wherein the subject exhibits no symptoms associated with COVID-19.
17. The method of one of claim 11, wherein the subject exhibits no symptoms associated with COVID-19 but has been exposed to another subject known or suspected of having COVID-19.
18. The method of any one of claims 1-17, wherein the treating results in reduced duration or occurrence of hospitalization, ventilation or dialysis of the subject.
19. The method of any one of claims 1-18, wherein the treating results a reduced likelihood of the subject requiring intermittent mandatory ventilation (IMV) or extracorporeal membrane oxygenation (ECMO), as compared to a subject with similar symptoms who has not been treated with the compound of Formula (I).
20. The method of any one of claims 1-19, wherein the subject has a faster and/or more extensive recovery than a subject with similar symptoms who has not been treated with the compound of Formula (I).
21. The method of any one of claims 1-20, wherein the compound is administered together with at least one additional active agent.
22. The method of claim 21, wherein the at least one additional active agent is selected from an immunomodulatory agent or antiviral agent
23. The method of claim 21 or 22, wherein said administration is simultaneous or sequential.
24. The method of any one of claims 1-23, wherein the compound of Formula (I) is administered in a dose of 400mg per day.
25. The method of any one of claims 1-24, wherein the compound of Formula (I) is administered once daily or twice daily.
26. The method of any one of claims 1-25, wherein the compound of Formula (I) is administered orally.
27. The method of any one of claims 1-26, wherein the treating results in a reduction in viral load of the subject.
28. The method of claim 27, the reduction in viral load is a reduction in plasma viral load.
PCT/US2021/049609 2020-09-11 2021-09-09 Methods of treating viral infection WO2022056092A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063077461P 2020-09-11 2020-09-11
US63/077,461 2020-09-11

Publications (1)

Publication Number Publication Date
WO2022056092A1 true WO2022056092A1 (en) 2022-03-17

Family

ID=80629990

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/049609 WO2022056092A1 (en) 2020-09-11 2021-09-09 Methods of treating viral infection

Country Status (1)

Country Link
WO (1) WO2022056092A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090075999A1 (en) * 2005-11-16 2009-03-19 S*Bio Pte Ltd. Oxygen linked pyrimidine derivatives
US20170231986A1 (en) * 2014-08-11 2017-08-17 Acerta Pharma B.V. Therapeutic Combinations of a BTK Inhibitor, a PI3K Inhibitor, a JAK-2 Inhibitor, and/or a BCL-2 Inhibitor
US20170312283A1 (en) * 2011-11-30 2017-11-02 Emory University Antiviral jak inhibitors useful in treating or preventing retroviral and other viral infections

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090075999A1 (en) * 2005-11-16 2009-03-19 S*Bio Pte Ltd. Oxygen linked pyrimidine derivatives
US20170312283A1 (en) * 2011-11-30 2017-11-02 Emory University Antiviral jak inhibitors useful in treating or preventing retroviral and other viral infections
US20170231986A1 (en) * 2014-08-11 2017-08-17 Acerta Pharma B.V. Therapeutic Combinations of a BTK Inhibitor, a PI3K Inhibitor, a JAK-2 Inhibitor, and/or a BCL-2 Inhibitor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BRODERICK JASON M.: "Pacritinib COVID-19 Trial Launched", ONCLIVE, 28 April 2020 (2020-04-28), XP055917311, Retrieved from the Internet <URL:https://www.onclive.com/view/pacritinib-covid19-trial-launched> [retrieved on 20220503] *

Similar Documents

Publication Publication Date Title
Tufan et al. COVID-19, immune system response, hyperinflammation and repurposingantirheumatic drugs
Sahebnasagh et al. Pharmacological treatments of COVID-19
Channappanavar et al. Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice
Govorkova et al. Neuraminidase inhibitor-rimantadine combinations exert additive and synergistic anti-influenza virus effects in MDCK cells
Qin et al. Type 1 responses of human Vγ9Vδ2 T cells to influenza A viruses
KR101755058B1 (en) Combinations of a specific hcv ns5a inhibitor and an hcv ns3 protease inhibitor
Shetty et al. Peramivir for the treatment of influenza
US10801076B2 (en) Influenza A virus variants
McFee COVID-19: Therapeutics and interventions currently under consideration
Fadel et al. CXCR3‐deficiency protects influenza‐infected CCR5‐deficient mice from mortality
US20230157994A1 (en) New compositions and methods of treating covid-19 disease
Mok et al. Anti-inflammatory and antiviral effects of indirubin derivatives in influenza A (H5N1) virus infected primary human peripheral blood-derived macrophages and alveolar epithelial cells
US20230079150A1 (en) Methods for prevention or treatment of virus-induced organ injury or failure with il-22 dimer
CN115867268A (en) Vidofladimor for the treatment or prevention of viral diseases
US20220288155A1 (en) Cxcr4 inhibitor for the treatment of acute respiratory distress syndrome and viral infections
Pascua et al. Pathogenicity and peramivir efficacy in immunocompromised murine models of influenza B virus infection
US20230150984A1 (en) Nicotinamide mononucleotide and nicotinamide riboside derivatives and use thereof in the treatment of viral infections and respiratory complications, in particular caused by influenzavirus or coronavirus
WO2022056092A1 (en) Methods of treating viral infection
US8957108B2 (en) Use of 5,6-dimethylxanthenone-4-acetic acid as an antiviral agent
JP7293225B2 (en) Treatment of RSV with combination products
CN116096364A (en) Use of diffusion enhancing compounds for the treatment of viral and bacterial induced respiratory diseases
Kumova et al. Severity of neonatal influenza infection is driven by type I interferon and oxidative stress
TWI815341B (en) Method against severe acute respiratory syndrome coronavirus 2 infection
TWI764284B (en) Method against coronavirus infection with quinoline compound
JP2023525981A (en) Religlitazone for treating pulmonary inflammation and interstitial lung disease

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: 21867576

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: 21867576

Country of ref document: EP

Kind code of ref document: A1