WO2022081984A1 - Methods and compounds to treat sars infections - Google Patents

Methods and compounds to treat sars infections Download PDF

Info

Publication number
WO2022081984A1
WO2022081984A1 PCT/US2021/055199 US2021055199W WO2022081984A1 WO 2022081984 A1 WO2022081984 A1 WO 2022081984A1 US 2021055199 W US2021055199 W US 2021055199W WO 2022081984 A1 WO2022081984 A1 WO 2022081984A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
group
independently selected
optionally substituted
Prior art date
Application number
PCT/US2021/055199
Other languages
French (fr)
Inventor
Tariq M. Rana
Original Assignee
The Regents Of The University Of California
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 The Regents Of The University Of California filed Critical The Regents Of The University Of California
Publication of WO2022081984A1 publication Critical patent/WO2022081984A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C225/00Compounds containing amino groups and doubly—bound oxygen atoms bound to the same carbon skeleton, at least one of the doubly—bound oxygen atoms not being part of a —CHO group, e.g. amino ketones
    • C07C225/24Compounds containing amino groups and doubly—bound oxygen atoms bound to the same carbon skeleton, at least one of the doubly—bound oxygen atoms not being part of a —CHO group, e.g. amino ketones the carbon skeleton containing carbon atoms of quinone rings
    • C07C225/26Compounds containing amino groups and doubly—bound oxygen atoms bound to the same carbon skeleton, at least one of the doubly—bound oxygen atoms not being part of a —CHO group, e.g. amino ketones the carbon skeleton containing carbon atoms of quinone rings having amino groups bound to carbon atoms of quinone rings or of condensed ring systems containing quinone rings
    • C07C225/30Compounds containing amino groups and doubly—bound oxygen atoms bound to the same carbon skeleton, at least one of the doubly—bound oxygen atoms not being part of a —CHO group, e.g. amino ketones the carbon skeleton containing carbon atoms of quinone rings having amino groups bound to carbon atoms of quinone rings or of condensed ring systems containing quinone rings of condensed quinone ring systems formed by two rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/16Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by singly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/30Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/37Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • C07C311/38Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring having sulfur atoms of sulfonamide groups and amino groups bound to carbon atoms of six-membered rings of the same carbon skeleton
    • C07C311/44Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring having sulfur atoms of sulfonamide groups and amino groups bound to carbon atoms of six-membered rings of the same carbon skeleton having the nitrogen atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/26Sulfones; Sulfoxides having sulfone or sulfoxide groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton
    • C07C317/28Sulfones; Sulfoxides having sulfone or sulfoxide groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton with sulfone or sulfoxide groups bound to acyclic carbon atoms of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/46Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with hetero atoms directly attached to the ring nitrogen atom
    • C07D207/50Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/56Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/56Nitrogen atoms
    • C07D211/58Nitrogen atoms attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • C07D249/101,2,4-Triazoles; Hydrogenated 1,2,4-triazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D249/12Oxygen or sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D279/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D279/101,4-Thiazines; Hydrogenated 1,4-thiazines
    • C07D279/121,4-Thiazines; Hydrogenated 1,4-thiazines not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D293/00Heterocyclic compounds containing rings having nitrogen and selenium or nitrogen and tellurium, with or without oxygen or sulfur atoms, as the ring hetero atoms
    • C07D293/10Heterocyclic compounds containing rings having nitrogen and selenium or nitrogen and tellurium, with or without oxygen or sulfur atoms, as the ring hetero atoms condensed with carbocyclic rings or ring systems
    • C07D293/12Selenazoles; Hydrogenated selenazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D309/08Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D309/14Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06078Dipeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0808Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0827Tripeptides containing heteroatoms different from O, S, or N
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/101Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/02Systems containing only non-condensed rings with a three-membered ring

Definitions

  • SARS-CoV-2 produces a spike protein that binds to host cell receptor ACE2 for entry.
  • ACE2 host cell receptor
  • the positive genomic RNA of SARS-CoV-2 will attach directly to the host ribosome and translate two large polyproteins, which are then processed by proteolysis into components for packaging new virions.
  • This proteolysis is controlled by two protease enzymes, the coronavirus main protease (M pro ) and the papain-like protease (PL pro ).
  • M pro coronavirus main protease
  • PL pro papain-like protease
  • RdRp RNA-dependent RNA polymerase
  • This disclosure features chemical entities (e.g., a compound (e.g., a compound of Formula (I), (II), (III), or (IV) as defined herein) or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the main protease (M Pro ) of a coronavirus (e.g., SARS-CoV-2).
  • a compound e.g., a compound of Formula (I), (II), (III), or (IV) as defined herein
  • M Pro main protease
  • Said chemical entities are useful, e.g., for treating a coronavirus infection (e.g., SARS-CoV-2 infection (e.g., COVID-19)) in a subject (e.g., a human).
  • a coronavirus infection e.g., SARS-CoV-2 infection (e.g., COVID-19)
  • a subject e.g., a human
  • This disclosure also features compositions containing the same as well as methods of using and making the same.
  • halo refers to fluoro (F), chloro (Cl), bromo (Br), or iodo (I).
  • alkyl refers to a saturated acyclic hydrocarbon radical that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, C 1-10 indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it.
  • Alkyl groups can either be unsubstituted or substituted with one or more substituents. Non-limiting examples include methyl, ethyl, iso-propyl, tert-butyl, n-hexyl.
  • saturated as used in this context means only single bonds present between constituent carbon atoms and other available valences occupied by hydrogen and/or other substituents as defined herein.
  • haloalkyl refers to an alkyl, in which one or more hydrogen atoms is/are replaced with an independently selected halo (e.g., -CF 3 ).
  • alkoxy refers to an -O-alkyl radical (e.g., -OCH 3 ).
  • haloalkoxy refers to an –O-haloalkyl radical (e.g., -OCF 3 ).
  • thioalkoxy refers to an –S-alkyl radical (e.g., -SCH 3 ).
  • aryl refers to a 6-20 carbon mono-, bi-, tri- or polycyclic group wherein at least one ring in the system is aromatic (e.g., 6-carbon monocyclic, 10-carbon bicyclic, or 14- carbon tricyclic aromatic ring system); and wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent.
  • aryl groups include phenyl, naphthyl, tetrahydronaphthyl, and the like.
  • cycloalkyl refers to saturated or partially unsaturated (but not aromatic) cyclic hydrocarbon groups having 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkyl group may be optionally substituted.
  • Examples of cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Cycloalkyl may include multiple fused and/or bridged rings.
  • fused/bridged cycloalkyl includes: bicyclo[1.1.0]butane, bicyclo[2.1.0]pentane, bicyclo[1.1.1]pentane, bicyclo[3.1.0]hexane, bicyclo[2.1.1]hexane, bicyclo[3.2.0]heptane, bicyclo[4.1.0]heptane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[4.2.0]octane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane, and the like.
  • Cycloalkyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom).
  • spirocyclic cycloalkyls include spiro[2.2]pentane, spiro[2.5]octane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[4.4]nonane, spiro[2.6]nonane, spiro[4.5]decane, spiro[3.6]decane, spiro[5.5]undecane, and the like.
  • heteroaryl means a mono-, bi-, tri- or polycyclic group having 5 to 20 ring atoms, alternatively 5, 6, 9, 10, or 14 ring atoms; and having 6, 10, or 14 pi electrons shared in a cyclic array; wherein at least one ring in the system is aromatic (but does not have to be a ring which contains a heteroatom, e.g. tetrahydroisoquinolinyl, e.g., tetrahydroquinolinyl), and at least one ring in the system contains one or more heteroatoms independently selected from the group consisting of N, O, and S.
  • heteroatoms independently selected from the group consisting of N, O, and S.
  • Heteroaryl groups can either be unsubstituted or substituted with one or more substituents.
  • heteroaryl include thienyl, pyridinyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl benzothienyl, benzoxadiazolyl, benzofuranyl, benzimidazolyl, benzotriazolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3-d]pyrimi
  • the heteroaryl is selected from thienyl, pyridinyl, furyl, pyrazolyl, imidazolyl, isoindolinyl, pyranyl, pyrazinyl, and pyrimidinyl.
  • heterocyclyl refers to a mon-, bi-, tri-, or polycyclic saturated or partially unsaturated (but not aromatic) ring system with 3-16 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent.
  • ring atoms e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system
  • heteroatoms selected from O
  • heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like.
  • Heterocyclyl may include multiple fused and bridged rings.
  • Non-limiting examples of fused/bridged heteorocyclyl includes: 2-azabicyclo[1.1.0]butane, 2-azabicyclo[2.1.0]pentane, 2-azabicyclo[1.1.1]pentane, 3-azabicyclo[3.1.0]hexane, 5-azabicyclo[2.1.1]hexane, 3- azabicyclo[3.2.0]heptane, octahydrocyclopenta[c]pyrrole, 3-azabicyclo[4.1.0]heptane, 7- azabicyclo[2.2.1]heptane, 6-azabicyclo[3.1.1]heptane, 7-azabicyclo[4.2.0]octane, 2- azabicyclo[2.2.2]octane, 3-azabicyclo[3.2.1]octane, 2-oxabicyclo[1.1.0]butane, 2- oxabicyclo[2.1.0]pentane, 2-oxabicyclo[1.1.1]
  • Heterocyclyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom).
  • spirocyclic heterocyclyls include 2-azaspiro[2.2]pentane, 4- azaspiro[2.5]octane, 1-azaspiro[3.5]nonane, 2-azaspiro[3.5]nonane, 7-azaspiro[3.5]nonane, 2- azaspiro[4.4]nonane, 6-azaspiro[2.6]nonane, 1,7-diazaspiro[4.5]decane, 7- azaspiro[4.5]decane 2,5-diazaspiro[3.6]decane, 3-azaspiro[5.5]undecane, 2- oxaspiro[2.2]pentane, 4-oxaspiro[2.5]octane, 1-oxaspiro[3.5]nonan
  • inhibiting refers to ceasing biological and/or chemical activity, slowing biological and/or chemical activity, and/or reducing biological and/or chemical activity.
  • the inhibition of stress granule formation can include reducing the formation of stress granules, ceasing the formation of stress granules, and/or slowing the progression of stress granule formation.
  • pharmaceutically acceptable salts is meant to include salts of the compounds that are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
  • base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
  • pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, oxalic, methanesulfonic, and the like.
  • inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic,
  • salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al., “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1-19).
  • Certain specific compounds of the present disclosure contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
  • Certain compounds of the present disclosure can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present disclosure.
  • Certain compounds of the present disclosure may exist in multiple crystalline or amorphous forms.
  • “Pharmaceutically acceptable excipient” and “pharmaceutically acceptable carrier” refer to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present disclosure without causing a significant adverse toxicological effect on the patient.
  • Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, lactated Ringer’s, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such as Ringer's solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethycellulose, polyvinyl pyrrolidine, and colors, and the like.
  • Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure.
  • auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure.
  • auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure.
  • auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents
  • FIG.2. Illustrates docking poses of Formula (I) compounds.
  • a hydrogen bond is observed between the carbonyl oxygen atom of compound 1 and Glu 166.
  • the benzoisoselenazole ring forms ⁇ - ⁇ stacking interactions with catalytic His 41.
  • a hydrogen bond is observed between the carbonyl oxygen atom of compound 19 and Glu 166.
  • the benzoisoselenazole ring forms ⁇ - ⁇ stacking interactions with catalytic His 41.
  • the chlorine atom of compound 19 forms a halogen bond interaction with Gly 143.
  • ⁇ - ⁇ stacking interactions are observed between the benzoisoselenazole ring of compound 20 and catalytic His 41.
  • a hydrogen bond is observed between the carbonyl oxygen atom of compound 20 and Glu 166.
  • FIG.3 Illustrates that ebselen and certain compounds described herein can covalently modify SARS-CoV-2 M pro in the absence of reducing agents.
  • compound 1 ebselen
  • compound 1 is a reversible covalent inhibitor of SARS-CoV-2 M pro .
  • compound 19 is a rapidly reversible inhibitor of SARS-CoV-2 M pro .
  • E. compound 20 is a reversible covalent inhibitor of SARS-CoV-2 M pro .
  • F. compound 25 is a reversible covalent inhibitor of SARS-CoV-2 M pro .
  • FIG. 4 Illustrates that compound 1 (ebselen) and compound 4 are competitive inhibitors of SARS-CoV-2 main protease.
  • C. M pro exposed to varying concentrations of compound 4 reaches a common V max at 30 ⁇ M substrate D.
  • LORGs iPSC-derived human lung organoids
  • A Representative phase contrast image of LORG at 60 Days (60D); scale bar represents 200 ⁇ m.
  • C Confocal images showing labelling for cells surrounding epithelial structures (ECAD) co- labelled with mesenchymal-cell-type marker vimentin (VIM).
  • AT1 lung alveolar type 1 cell
  • G. LORGs express ACE2, TMPRSS2, DPP4, and Furin.
  • Lung organoids are permissive to SARS-CoV-2 pseudovirus infection that is inhibited by Camostat, nafamostat, and EK1 peptide.
  • SARS-CoV-2 isolate USA-WA1/2020 robustly replicates in human lung organoids. Lung organoids were infected with SARS-CoV-2 USA-WA1/2020 virus at MOI of 2 and viral RNAs from J. supernatant and K.
  • M. Organoids pre-treated with 25HC (5 ⁇ M) for 16h and infected with luciferase expressing SARS-CoV-2 pseudovirus at MOI 0.1 for 2 h and treated with fresh medium containing 5 ⁇ M 25HC.
  • this disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the main protease (M Pro ) of a coronavirus (e.g., SARS-CoV-2).
  • Said chemical entities are useful, e.g., for treating a coronavirus infection (e.g., SARS-CoV-2 infection (e.g., COVID-19)) in a subject (e.g., a human).
  • This disclosure also features compositions containing the same as well as methods of using and making the same.
  • R 1 is selected from the group consisting of: H, halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, and C 1-6 haloalkoxy
  • R 2 is phenyl, –CH 2 -phenyl, or –C 4-10 alkyl, each optionally substituted with from 1-4 independently selected R a
  • each occurrence of R a is independently selected from the group consisting of: halo; C 1- 10 alkyl; C 1-6 haloalkyl; cyano; C 1-6 alkoxy; C 1-6 haloalkoxy; C 1-6 thioalkoxy; and –OH.
  • R 1 is selected from the group consisting of: halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, and C 1-6 haloalkoxy; and/or (bb) R 2 is selected from the group consisting of: phenyl substituted with from 1-4 R a ; –CH 2 -phenyl optionally substituted with from 1-4 independently selected R a ; and –C 4-10 alkyl optionally substituted with from 1-4 independently selected R a .
  • R 1 when R 1 is H, R 2 is –CH 2 -phenyl or –C 4-10 alkyl, each optionally substituted with from 1-4 independently selected R a .
  • R 1 when R 2 is phenyl, R 1 is selected from the group consisting of: halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, and C 1-6 haloalkoxy. In some embodiments of Formula (I), R 1 is H.
  • R 1 is selected from the group consisting of: halo, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, and C 1-3 haloalkoxy. In some embodiments of Formula (I), R 1 is selected from the group consisting of: -F, - Cl, -CH 3 , -CF 3 , and –OCH 3 . In some embodiments of Formula (I), R 2 is phenyl which is optionally substituted with from 1-4 independently selected R a . In some embodiments of Formula (I), R 2 is phenyl substituted with from 1-4 independently selected R a .
  • R 2 is phenyl substituted with from 1-2 independently selected R a .
  • R 2 is a1 , wherein: R is selected from the group consisting of: H, F, Cl, CH 3 , CF 3 , OH, OCH 3 , OCF 3 , SCH 3 , CN, C 2 H 5 , C 5 H 11 , C 7 H 15 , and C 8 H 17 ; and R a2 is selected from the group consisting of: H, F, Cl, CH 3 , CF 3 , OH, OCH 3 , OCF 3 , and SCH 3 .
  • R 2 is , wherein R a1 is an independently selected R a ; and R a2 is H or R a .
  • R 2 is , wherein R a1 is an cted R a independently sele ; and R a2 is H or R a .
  • R a1 is selected from the group consisting of: -Br, -Cl, -F, cyano, C 1-3 alkyl, C 1-3 haloalkyl, and C 1-3 alkoxy.
  • R a1 is selected from the group consisting of: -Br, -Cl, C 1-3 alkyl, and C 1-3 haloalkyl. In some embodiments of Formula (I), R a1 is selected from the group consisting of –Br, -Cl, -CH 3 , CH 2 CH 3 , and CF 3 . In some embodiments of Formula (I), R a2 is H or halo. In some embodiments of Formula (I), R 2 is C 4-10 alkyl optionally substituted with from 1-4 independently selected R a . In some embodiments of Formula (I), R 2 is C 4-10 alkyl, such as linear C 4-10 alkyl.
  • R 1 is H
  • R 2 is , wherein: R a1 is selected from the group consisting of: H, F, Cl, CH 3 , CF 3 , OH, OCH 3 , OCF 3 , SCH 3 , CN, C 2 H 5 , C 5 H 11 , C 7 H 15 , and C8H17; and R a2 is selected from the group consisting of: H, F, Cl, CH 3 , CF 3 , OH, OCH 3 , OCF 3 , and SCH 3 .
  • R 1 is H
  • R 2 is , wherein R a1 is an independently selected R a ; and R a2 is H or R a .
  • R 1 is H, and R 2 is , wherein R a1 is an independently selected R a ; and R a2 is H or R a .
  • R 1 is H, and R 2 is C 4-10 alkyl optionally substituted with from 1-4 independently selected R a .
  • R 1 is H, and R 2 is C 4-10 alkyl, such as linear C 4-10 alkyl.
  • the compound is selected from the group consisting of the following:
  • the compound is selected from the group consisting of compounds 2-30, or a pharmaceutically acceptable salt thereof. In some embodiments of Formula (I), the compound is not compound 1, or a pharmaceutically acceptable salt thereof. In some embodiments of Formula (I), the compound is selected from the group consisting of the following: or a pharmaceutically acceptable salt thereof.
  • each R 1 is independently selected from the group consisting of: halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 1-6 thioalkoxy, -OH, cyano, and C(O)O(C 1-6 alkyl); and each R 2 is selected from the group consisting of: halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 1-6 thioalkoxy, -OH, and C(O)O(C 1-6 alkyl).
  • Ar is In some embodiments of Formula (II), Ar is In some embodiments of Formula (II), q is 1. In some embodiments of Formula (II), q is 2. In some embodiments of Formula (II), each R 2 is independently selected from the group consisting of: F, Cl, CN, CH 3 , t Bu, CF 3 , OH, OCH 3 , OCF 3 , SCH 3 , and C(O)OMe. In some embodiments of Formula (II), p is 0. In some embodiments of Formula (II), p is 1, 2, or 3.
  • R 1 is selected from the group consisting of: F, Cl, CH 3 , CF 3 , OH, OCH 3 , OCF 3 , SCH 3 , CN, and C(O)OMe.
  • the compound is selected from the group consisting of:
  • R 1 is selected from the group consisting of: o C1-15 alkyl optionally substituted with from 1-6 independently selected R a ; and o -L 3 -R 3 , wherein: o L 3 is a bond or C 1-6 alkylene optionally substituted with from 1-6 independently selected R a ; and o R 3 is selected from the group consisting of C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with from 1-4 independently selected R b ; and o -(Z 1 -Z 2 ) m -Z 3 , wherein: o each Z 1 is independently C 1-6 alkylene optionally substituted with from 1-6 independently selected R a ; o each Z 2 is independently selected from the group consisting
  • R 2a is nitro. In some embodiments, R 2b , R 2c , and R 2d are each H. In some embodiments of Formula (III), R 2d is nitro. In some embodiments, R 2a , R 2b , and R 2c are each H. In some embodiments of Formula (III), R 2a and R 2d are —OH. In some embodiments, R 2b and R 2c are H. In some embodiments of Formula (III), R 1a is H. In some embodiments of Formula (III), R 1 is C 1-15 alkyl, such as C 4-15 alkyl (e.g., C 4-15 linear alkyl), optionally substituted with from 1-6 independently selected R a .
  • R 1 can be selected from the group consisting of: , , In some embodiments of Formula (III), R 1 is –R 3 or –CH 2 -R 3 . In some embodiments of Formula (III), R 3 is selected from the group consisting of: C 3- 10 cycloalkyl and 4-10 membered heterocyclyl, each optionally substituted with from 1-4 independently selected R b , optionally wherein one occurrence of R b is –(L c ) p -R c . In some embodiments of Formula (III), R 1 is selected from the group consisting of: and .
  • R 3 is selected from the group consisting of: C 6- 10 aryl and 5-10 membered heteroaryl, each optionally substituted with from 1-4 independently selected R b , optionally wherein one occurrence of R b is –(L c ) p -R c .
  • R 1 is selected from the group consisting of: and
  • R 1a and R 1 taken together with the nitrogen atom to which each is attached forms a 4-8 membered heterocyclyl, wherein the heterocyclyl is optionally substituted with from 1-6 independently selected R b , such as wherein R 1a and R 1 taken together with the nitrogen atom to which each is attached forms .
  • R 1 is -(Z 1 -Z 2 ) m -Z 3 , optionally wherein Z 1 is CH 2 CH 2 ; and Z 2 is –O-, such as wherein R 1 is In some embodiments of Formula (III), the compound of selected from the group consisting of the following:
  • R 1 is selected from the group consisting of: H, -OC 1-6 alkyl, -O(C 3-6 cycloalkyl), and NR’R”
  • R 2 is selected from the group consisting of: -C 1-6 alkyl, -C 1-6 hal
  • R 1 is H. In some embodiments of Formula (IV), R 1 is OC 1-6 alkyl, such as OMe. In some embodiments of Formula (IV), R 1 is NR’R” such as NHMe, or NH(C 3-6 cycloalkyl). In some embodiments of Formula (IV), R 2 is In some embodiments of Formula (IV), R 2 is C 1-6 alkyl, such as isopropyl; or R 2 is C1- 6 haloalkyl, such as CF 3 In some embodiments of Formula (IV), R 2 is -(C 1-6 alkylene)-NR’R”, such as – CH 2 NHMe.
  • R 1 is OMe, H, or NHMe;
  • R 1 is OMe, H, or NHMe;
  • R 2 is ; and
  • the compound has the following formula:
  • the compound is selected from the group consisting of:
  • R is C 1-6 alkyl or C 1-6 haloalkyl, such as wherein R” is C 1-6 alkyl, such as isopropyl. Also provided herein is a compound as shown in Table I. Table I. Compounds of Formula (I), (II), and (III).
  • the method comprises administering to the subject one or more chemical entities generically or specifically described herein.
  • the method comprises administering to the subject a compound of Formula (I), Formula (II), Formula (III), Formula (IV), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as described herein.
  • the coronavirus infection is a betacoronavirus infection.
  • the coronavirus infection is selected from the group consisting of SARS-CoV-2, SARS-CoV, and MERS-CoV infection.
  • the coronavirus infection is a SARS-CoV-2 infection.
  • the coronavirus infections can be COVID-19.
  • the subject is human. Also provided herein are methods of treating a coronavirus infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I), Formula (II), Formula (III), Formula (IV) as described anywhere herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
  • the coronavirus infection is a betacoronavirus infection.
  • the coronavirus infection is selected from the group consisting of SARS-CoV- 2, SARS-CoV, and MERS-CoV infection.
  • the coronavirus infection is a SARS-CoV-2 infection.
  • the coronavirus infections can be COVID-19.
  • the subject is human.
  • Solvents methylene chloride, ether, tetrahydrofuran, benzene, and toluene
  • solvents were deoxygenated by sparging with nitrogen for at least 1 hour unless otherwise noted. All other reagents were used directly from the supplier without further purification unless otherwise noted.
  • Organic solutions were concentrated by rotary evaporation at ⁇ 25 mbar in a water bath heated to 40 °C unless otherwise noted.
  • Analytical thin-layer chromatography (TLC) was carried out using 0.2 mm commercial glass- coated silica gel plates (silica gel 60, F254, EMD chemical).
  • Thin layer chromatography plates were visualized by exposure to ultraviolet light and/or exposure to iodine, or to an acidic solution of ceric ammonium molybdate, or a basic solution of potassium permanganate followed by heating on a hot plate.
  • Gas chromatographs were measured using an Agilent 7820 GC. Mass spectra (MS) were obtained on a Karatos MS9, Autospec, or an Agilent 6150 and reported as m/z (relative intensity). Accurate masses are reported for the molecular ion [M+D] + .
  • Step 1 Preparation of intermediates (I)-i1-30.
  • a stirred solution of R 2 NH 2 (2 mmol) in DCM at 0 °C was treated with TEA (2 mmol), followed by the dropwise addition of corresponding acid chloride (I)-SM 1-30 (2 mmol) and the solution was stirred for 3 hours at room temperature.
  • the reaction mixture was diluted in excess DCM, transferred to a separatory funnel, and extracted with water and brine solution. The organic layer was separated and concentrated under reduced pressure.
  • Step 2 Preparation of compounds 1-30 A one-necked round-bottomed flask was fitted with a reflux condenser and with a magnetic stirrer. The flask was charged with corresponding intermediate (I)-i 1-30 (1 mmol), followed by the addition of Se powder (5 mmol) and tBuONa (2 mmol). The reaction mixture was left to stir at 130 °C for 12 hours. After completion of reaction, the reaction mixture was diluted with excess water and extracted with ethyl acetate. The organic layer was separate and concentrated under reduced pressure.
  • reaction mixture changed the color immediately (for aliphatic amines) or slowly (for aromatic amines). Then another 3 mL of EtOH was added into the vigorously stirring system which was heated to 78 ⁇ afterwards to reflux. The consumption of starting materials was followed and monitored by TLC. And after 30 min to 5 hours (the aromatic amine reaction time is longer than the aliphatic ones), the mixture was concentrated by rotary evaporator under reduced pressure. The final compounds were recrystallized in EtOH or purified by chromatographic silica gel column at yield 30-70%. Most of the final compounds are dark red solids.
  • Example 2 In Silico Modeling of Selected Compounds with Schrödinger In silico modeling of selected compounds was performed using the Glide docking module of the Schrödinger 11.5 modeling software suite. A crystal structure of the SARS-CoV- 2 M pro bound to N3 (PDB ID: 6UL7) was first refined using Prime (i. Jin, Z. et al, Nature 2020, 582, 289-293; ii. Jacobson, M. P. et al, J Mol Biol 2002, 320 (3), 597-608; iii. Jacobson, M. P. et al, Proteins 2004, 55 (2), 351-367).
  • Prime i. Jin, Z. et al, Nature 2020, 582, 289-293; ii. Jacobson, M. P. et al, J Mol Biol 2002, 320 (3), 597-608; iii. Jacobson, M. P. et al, Proteins 2004, 55 (2), 351-367).
  • the Optimized Potentials for Liquid Simulations All- Atom (OPLS) force field and the Surface generalized Born (SGB) continuum solution model were used to optimize and minimize the crystal structure.
  • Ligprep was used to generate a minimized 3D structure for all antiviral compounds using the OPLS 2001 force field. Docking was performed with Glide XP (i. Friesner, R. A. et al, J Med Chem 2004, 47 (7), 1739-1749; ii. Halgren, T. A. et al, J Med Chem 2004, 47 (7), 1750-1759; iii.
  • SARS-CoV-2 Main Protease Protein Expression and Purification The GST-tagged SARS-CoV-2 main protease was expressed in E. Coli BL21 competent cells (New England Biolabs) transformed with pET41b plasmid by heat shock and spread on a LB Kanamycin agar plate, then incubated overnight at 37 °C.
  • the pellets were resuspended in lysis buffer (50 mM Tris-HCl pH 7.5, 500 mM NaCl, 1.0% Triton x-100, 0.5 mM EDTA, 1x Complete protease inhibitor cocktail, 1 mM ⁇ -mercaptoethanol in RNase-free water; 6 mL per gram) with DNase 1 (5U per mL, RNase-free) and incubated at 4 °C for 1 hour and sonicated. The suspension was centrifuged at 10,000 g for 20 min and the supernatant was transferred to a glutathione sepharose affinity resin column that had been pre-equilibrized with lysis buffer.
  • lysis buffer 50 mM Tris-HCl pH 7.5, 500 mM NaCl, 1.0% Triton x-100, 0.5 mM EDTA, 1x Complete protease inhibitor cocktail, 1 mM ⁇ -mercaptoethanol in RNase-free water; 6 mL per
  • the supernatant was incubated with the affinity resin column at 4 °C for 2 hours with end-over-end rotation. After incubation, the column was washed with 5 bed volumes of lysis buffer, 3 x 5 bed volumes of wash buffer (50 mM Tris-HCl pH 7.5, 500 mM NaCl, 0.1% Triton x-100, 0.5 mM EDTA, 1x Complete protease inhibitor cocktail, 1 mM ⁇ - mercaptoethanol in RNase-free water), and 5 bed volumes of pre-elution buffer (50 mM Tris- HCl pH 7.5, 100 mM NaCl in RNase-free water).
  • wash buffer 50 mM Tris-HCl pH 7.5, 500 mM NaCl, 0.1% Triton x-100, 0.5 mM EDTA, 1x Complete protease inhibitor cocktail, 1 mM ⁇ - mercaptoethanol in RNase-free water
  • pre-elution buffer 50 mM Tri
  • protease was eluted with elution buffer (50 mM Tris-HCl pH 7.5, 100 mM NaCl, 40 mM glutathione in RNase-free water) and collected in 1 mL fractions until no further protein was collected (3-5 bed volumes total). The purity of the fractions was determined by SDS-page and fractions of purified protein were collected and concentrated to 2 mg/mL.
  • the GST-tagged purified protein was incubated with TEV protease at 37 °C for 4 hours to obtain the untagged SARS-CoV-2 protease. Excess GST and GST-TEV protease were removed by glutathione sepharose affinity column purification.
  • the untagged proteases were eluted in 50 mM Tris-HCl pH 7.5, 100 mM NaCl in RNase-free water. The purity of the fractions was determined by SDS-page and fractions of purified protein were collected and concentrated to 5 ⁇ M. The protein solutions were transferred to a Slyde-A- Lyzer Dialysis Cassette (20,000 MWCO, Thermo Scientific) and dialyzed overnight at 4 °C against dialysis buffer (50 mM Tris-HCl pH 7.4, 200 mM NaCl, 1 mM EDTA, 1 mM DTT in RNase-free water).
  • dialysis buffer 50 mM Tris-HCl pH 7.4, 200 mM NaCl, 1 mM EDTA, 1 mM DTT in RNase-free water.
  • FRET Protease Activity Assay Activity of the SARS-CoV-2 main proteases was determined using fluorescence resonance energy transfer (FRET) assay adapted from Muramatsu et al (Proc Natl Acad Sci U S A 2016, 113 (46), 12997-13002).
  • FRET fluorescence resonance energy transfer
  • the fluorogenic peptide Dabcyl-Val-Asn-Ser-Thr-Leu-Gln- Ser-Gly-Leu-Arg-Lys-EDANS was used as a substrate (AnaSpec Inc).
  • Calu-3 human lung cells approximately 1.5x10 5 Calu-3 cells (ATCC) were plated in 500 ⁇ L of EMEM (Gibco) supplemented with 10% FBS, 2 mM L- glutamine, and 1% penicillin/streptomycin in each well of 24-well flat bottom plates and allowed to adhere overnight. Cells were pre-treated with 0.10-20 ⁇ M test compound in DMSO (0.2 % final v/v) of the same media for 1 hr, washed, and infected with SARS-CoV-2 (USA_WA01/2020) in reduced serum media (2-3%) at 0.1 MOI for 1.5 hrs.
  • Example 8 Lung organoid infection and treatment with compound 24 Human iPSC derived lung organoids were generated using previously published methods and characterized by expression of ACE2, TMPRSS2, and alveolar cell epithelial markers (SFTPC, SFTPB, HOPX). The 60D differentiated lung organoids were pretreated with 5 ⁇ M E24 for 2 h and infected with SARS-CoV-2 as described previously (Tiwari, S. K. et al, Stem Cell Reports 2021, 16 (3), 437-445). SARS-CoV-2 isolate USA-WA1/2020 was obtained from BEI Resources. SARS-CoV-2 was propagated, and infectious units quantified by plaque assay using Vero E6 cells.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This disclosure features chemical entities (e.g., a compound (e.g., a compound of Formula (I), (II), (III), or (IV)), or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the main protease (MPro) of a coronavirus (e.g., SARS-CoV-2). Said chemical entities are useful, e.g., for treating a coronavirus infection (e.g., SARS-CoV-2 infection (e.g., COVID-19)) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same.

Description

Methods and Compounds to Treat SARS Infections I. Federally Sponsored Research and Development This invention was made with Government support under grant No. DA039562 awarded by the National Institutes of Health. The Government has certain rights in the invention. II. BACKGROUND Coronavirus disease 2019 (COVID-19) is a transmissible respiratory disease caused by a novel severe acute respiratory syndrome coronavirus SARS-CoV-2. Since its emergence in December 2019, COVID-19 has rapidly spread worldwide spanning 216 countries. As of October 2020, there are more than 219 million confirmed cases and 4.55 million deaths (WHO COVID Pandemic). Reports indicate that COVID-19 patients present with a cluster of severe respiratory illness symptoms similar to SARS, with which the genome of SARS-CoV-2 shares approximately 80% identity. SARS-CoV-2 produces a spike protein that binds to host cell receptor ACE2 for entry. Upon entry, the positive genomic RNA of SARS-CoV-2 will attach directly to the host ribosome and translate two large polyproteins, which are then processed by proteolysis into components for packaging new virions. This proteolysis is controlled by two protease enzymes, the coronavirus main protease (Mpro) and the papain-like protease (PLpro). An RNA-dependent RNA polymerase (RdRp) is also required to replicate the RNA genome. As such, there is a need for methods and compositions useful in the treatment of coronavirus infections, such as SARS-CoV-2 infection. III. SUMMARY This disclosure features chemical entities (e.g., a compound (e.g., a compound of Formula (I), (II), (III), or (IV) as defined herein) or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the main protease (MPro) of a coronavirus (e.g., SARS-CoV-2). Said chemical entities are useful, e.g., for treating a coronavirus infection (e.g., SARS-CoV-2 infection (e.g., COVID-19)) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same. The term "halo" refers to fluoro (F), chloro (Cl), bromo (Br), or iodo (I). The term "alkyl" refers to a saturated acyclic hydrocarbon radical that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, C1-10 indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it. Alkyl groups can either be unsubstituted or substituted with one or more substituents. Non-limiting examples include methyl, ethyl, iso-propyl, tert-butyl, n-hexyl. The term “saturated” as used in this context means only single bonds present between constituent carbon atoms and other available valences occupied by hydrogen and/or other substituents as defined herein. The term "haloalkyl" refers to an alkyl, in which one or more hydrogen atoms is/are replaced with an independently selected halo (e.g., -CF3). The term "alkoxy" refers to an -O-alkyl radical (e.g., -OCH3). The term “haloalkoxy” refers to an –O-haloalkyl radical (e.g., -OCF3). The term “thioalkoxy” refers to an –S-alkyl radical (e.g., -SCH3). The term "aryl" refers to a 6-20 carbon mono-, bi-, tri- or polycyclic group wherein at least one ring in the system is aromatic (e.g., 6-carbon monocyclic, 10-carbon bicyclic, or 14- carbon tricyclic aromatic ring system); and wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent. Examples of aryl groups include phenyl, naphthyl, tetrahydronaphthyl, and the like. The term "cycloalkyl" as used herein refers to saturated or partially unsaturated (but not aromatic) cyclic hydrocarbon groups having 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkyl group may be optionally substituted. Examples of cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Cycloalkyl may include multiple fused and/or bridged rings. Non-limiting examples of fused/bridged cycloalkyl includes: bicyclo[1.1.0]butane, bicyclo[2.1.0]pentane, bicyclo[1.1.1]pentane, bicyclo[3.1.0]hexane, bicyclo[2.1.1]hexane, bicyclo[3.2.0]heptane, bicyclo[4.1.0]heptane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[4.2.0]octane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane, and the like. Cycloalkyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom). Non- limiting examples of spirocyclic cycloalkyls include spiro[2.2]pentane, spiro[2.5]octane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[4.4]nonane, spiro[2.6]nonane, spiro[4.5]decane, spiro[3.6]decane, spiro[5.5]undecane, and the like. The term “heteroaryl”, as used herein, means a mono-, bi-, tri- or polycyclic group having 5 to 20 ring atoms, alternatively 5, 6, 9, 10, or 14 ring atoms; and having 6, 10, or 14 pi electrons shared in a cyclic array; wherein at least one ring in the system is aromatic (but does not have to be a ring which contains a heteroatom, e.g. tetrahydroisoquinolinyl, e.g., tetrahydroquinolinyl), and at least one ring in the system contains one or more heteroatoms independently selected from the group consisting of N, O, and S. Heteroaryl groups can either be unsubstituted or substituted with one or more substituents. Examples of heteroaryl include thienyl, pyridinyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl benzothienyl, benzoxadiazolyl, benzofuranyl, benzimidazolyl, benzotriazolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3-d]pyrimidinyl, pyrrolo[2,3-b]pyridinyl, quinazolinyl, quinolinyl, thieno[2,3-c]pyridinyl, pyrazolo[3,4-b]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[4,3- c]pyridine, pyrazolo[4,3-b]pyridinyl, tetrazolyl, chromane, 2,3-dihydrobenzo[b][1,4]dioxine, benzo[d][1,3]dioxole, 2,3-dihydrobenzofuran, tetrahydroquinoline, 2,3- dihydrobenzo[b][1,4]oxathiine, isoindoline, and others. In some embodiments, the heteroaryl is selected from thienyl, pyridinyl, furyl, pyrazolyl, imidazolyl, isoindolinyl, pyranyl, pyrazinyl, and pyrimidinyl. The term "heterocyclyl" refers to a mon-, bi-, tri-, or polycyclic saturated or partially unsaturated (but not aromatic) ring system with 3-16 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent. Examples of heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like. Heterocyclyl may include multiple fused and bridged rings. Non-limiting examples of fused/bridged heteorocyclyl includes: 2-azabicyclo[1.1.0]butane, 2-azabicyclo[2.1.0]pentane, 2-azabicyclo[1.1.1]pentane, 3-azabicyclo[3.1.0]hexane, 5-azabicyclo[2.1.1]hexane, 3- azabicyclo[3.2.0]heptane, octahydrocyclopenta[c]pyrrole, 3-azabicyclo[4.1.0]heptane, 7- azabicyclo[2.2.1]heptane, 6-azabicyclo[3.1.1]heptane, 7-azabicyclo[4.2.0]octane, 2- azabicyclo[2.2.2]octane, 3-azabicyclo[3.2.1]octane, 2-oxabicyclo[1.1.0]butane, 2- oxabicyclo[2.1.0]pentane, 2-oxabicyclo[1.1.1]pentane, 3-oxabicyclo[3.1.0]hexane, 5- oxabicyclo[2.1.1]hexane, 3-oxabicyclo[3.2.0]heptane, 3-oxabicyclo[4.1.0]heptane, 7- oxabicyclo[2.2.1]heptane, 6-oxabicyclo[3.1.1]heptane, 7-oxabicyclo[4.2.0]octane, 2- oxabicyclo[2.2.2]octane, 3-oxabicyclo[3.2.1]octane, and the like. Heterocyclyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom). Non-limiting examples of spirocyclic heterocyclyls include 2-azaspiro[2.2]pentane, 4- azaspiro[2.5]octane, 1-azaspiro[3.5]nonane, 2-azaspiro[3.5]nonane, 7-azaspiro[3.5]nonane, 2- azaspiro[4.4]nonane, 6-azaspiro[2.6]nonane, 1,7-diazaspiro[4.5]decane, 7- azaspiro[4.5]decane 2,5-diazaspiro[3.6]decane, 3-azaspiro[5.5]undecane, 2- oxaspiro[2.2]pentane, 4-oxaspiro[2.5]octane, 1-oxaspiro[3.5]nonane, 2-oxaspiro[3.5]nonane, 7-oxaspiro[3.5]nonane, 2-oxaspiro[4.4]nonane, 6-oxaspiro[2.6]nonane, 1,7- dioxaspiro[4.5]decane, 2,5-dioxaspiro[3.6]decane, 1-oxaspiro[5.5]undecane, 3- oxaspiro[5.5]undecane, 3-oxa-9-azaspiro[5.5]undecane and the like. The term “inhibiting”, “inhibitor”, and/or “inhibits”, as used herein refers to ceasing biological and/or chemical activity, slowing biological and/or chemical activity, and/or reducing biological and/or chemical activity. For example, the inhibition of stress granule formation can include reducing the formation of stress granules, ceasing the formation of stress granules, and/or slowing the progression of stress granule formation. The term “pharmaceutically acceptable salts” is meant to include salts of the compounds that are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When compounds of the present disclosure contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt. When compounds of the present disclosure contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, oxalic, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al., “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1-19). Certain specific compounds of the present disclosure contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts. Certain compounds of the present disclosure can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present disclosure. Certain compounds of the present disclosure may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present disclosure and are intended to be within the scope of the present disclosure. “Pharmaceutically acceptable excipient” and “pharmaceutically acceptable carrier” refer to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present disclosure without causing a significant adverse toxicological effect on the patient. Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, lactated Ringer’s, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such as Ringer's solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethycellulose, polyvinyl pyrrolidine, and colors, and the like. Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure. One of skill in the art will recognize that other pharmaceutical excipients are useful in the present disclosure. IV. BRIEF DESCRIPTION OF THE DRAWINGS FIG.1. Illustrates the structure of SARS-CoV-2 Main Protease (highly conserved with SARS Mpro). Catalysis is controlled by a dyad of reactive active site residues including His 41 and Cys 145, which follow covalent mechanism consistent with other Ser, Cys, and Thr proteases. FIG.2. Illustrates docking poses of Formula (I) compounds. A. Docking pose of compound 1 (ebselen) bound to the catalytic site of SARS-CoV-2 Mpro. A hydrogen bond is observed between the carbonyl oxygen atom of compound 1 and Glu 166. The benzoisoselenazole ring forms π-π stacking interactions with catalytic His 41. B. Docking pose of compound 19 bound to the catalytic site of SARS-CoV-2 Mpro. A hydrogen bond is observed between the carbonyl oxygen atom of compound 19 and Glu 166. The benzoisoselenazole ring forms π-π stacking interactions with catalytic His 41. The chlorine atom of compound 19 forms a halogen bond interaction with Gly 143. C. Docking pose of compound 20 bound to the catalytic site of SARS-CoV-2 Mpro. π-π stacking interactions are observed between the benzoisoselenazole ring of compound 20 and catalytic His 41. A hydrogen bond is observed between the carbonyl oxygen atom of compound 20 and Glu 166. D. Docking pose of compound 25 bound to the catalytic site of SARS-CoV-2 Mpro. A hydrogen bond is observed between the carbonyl oxygen atom of compound 25 and Glu 166. π-π stacking interactions are observed between the benzoisoselenazole ring of compound 25 and catalytic His 41. The nitrile group is observed to accept a hydrogen bond from Cys 44. FIG.3. Illustrates that ebselen and certain compounds described herein can covalently modify SARS-CoV-2 Mpro in the absence of reducing agents. A. compound 1 (ebselen) is a reversible covalent inhibitor of SARS-CoV-2 Mpro. The product formation over time was reduced approximately 65% relative to DMSO control (squares) by 1 µM of ebselen (dots) and by 89% by 10 µM of ebselen (inverted triangles) when added simultaneously with fluorogenic substrate. Upon 10x dilution in assay buffer without DTT, product formation of SARS-CoV-2 Mpro pre-incubated with 10 µM of ebselen (diamonds) is observed to reduce product formation by 91%, comparable to an undiluted concentration of 10 µM. When diluted in the presence of DTT, SARS-CoV-2 Mpro pre-incubated with 10 µM of ebselen (triangles) is observed to reduce product formation by 53% relative to DMSO control (squares), similar to that observed for a final concentration of 1 µM. These data indicate that ebselen can covalently modify SARS- CoV-2 Mpro, although this modification is reversible in the presence of strong reducing agents. B. compound 4 is a rapidly reversible inhibitor of SARS-CoV-2 Mpro. Upon 10x dilution, product formation of SARS-CoV-2 Mpro pre-incubated with 10 µM of compound 4 in the absence of DTT (diamonds) is observed to reduce product formation by 48% relative to DMSO control (squares), similar to that observed for a final concentration of 1 µM compound 4 (dots, 44% inhibition), while simultaneous addition of substrate and 10 µM compound 4 (inverted triangles) is observed to reduce product formation by 86%. Similar results are observed for SARS-CoV-2 Mpro pre-incubated with 10 µM of compound 4 in the presence of DTT (triangles,43% inhibition). C. compound 7 is a reversible covalent inhibitor of SARS-CoV-2 Mpro. D. compound 19 is a rapidly reversible inhibitor of SARS-CoV-2 Mpro. E. compound 20 is a reversible covalent inhibitor of SARS-CoV-2 Mpro. F. compound 25 is a reversible covalent inhibitor of SARS-CoV-2 Mpro. Upon 10x dilution, product formation of SARS-CoV- 2 Mpro pre-incubated with 10 µM of compound 25 in the absence of DTT (diamonds) is observed to reduce product formation by 64% relative to DMSO control (squares). Simultaneous addition of substrate and 10 µM 25 reduced product formation by 94%, while simultaneous addition of substrate and 1 µM compound 25 is observed to reduce product formation by 29%. These data suggest that while compound 25 may covalently modify SARS- CoV-2 Mpro, formation of the covalent adduct was incomplete after 30 minutes of incubation. FIG. 4. Illustrates that compound 1 (ebselen) and compound 4 are competitive inhibitors of SARS-CoV-2 main protease. A. Mpro exposed to varying concentrations of ebselen reaches a common Vmax at 30 µM substrate B. All ebselen conditions converge on a common y-intercept, indicating Vmax is independent of inhibitor concentration, consistent with a competitive mechanism of inhibition. C. Mpro exposed to varying concentrations of compound 4 reaches a common Vmax at 30 µM substrate D. All compound 4 conditions converge on a common y-intercept, indicating Vmax is independent of inhibitor concentration, consistent with a competitive mechanism of inhibition. FIG. 5. Illustrates antiviral activity of select Formula (I) compounds. Vero E6 cells were pre-treated with varying doses of compound then infected with SARS-CoV-2 to a multiplicity of infection (MOI) of 0.01. After 72 hours, cell proliferation was determined. Remdesivir is included as a positive control (EC50 = 1.8 ± 1.2 µM). A. Dose-response curve for compound 1 (EC50 = >20 µM). B. Dose-response curve for compound 4 (EC50 = 11.2 ± 1.3 µM). C. Dose-response curve for compound 7 (EC50 = 26.5 ± 1.2 µM). D. Dose-response curve for compound 18 (EC50 = 6.5 ± 2.0 µM). E. Dose-response curve for compound 19 (EC50 = 17.4 ± 3.5 µM). F. Dose-response curve for compound 20 (EC50 = 18.2 ± 3.6 µM). G. Dose-response curve for compound 21 (EC50 = 5.2 ± 1.2 µM). H. Dose-response curve for compound 24 (EC50 = 0.8 ± 0.3 µM). I. Dose-response curve for compound 25 (EC50 = 2.0 ± 1.1 µM). FIG. 6. Illustrates that compound 24 inhibits SARS-CoV-2 replication in infected human iPSC-deriv6 lung organoids. Sixty day old lung organoids were infected with SARS- CoV-2 USA-WA1/2020 virus at MOI of 2 and viral RNAs from A. supernatant, and B. cellular fractions quantified after 72 h of infection; mean ± SEM of n = 3 organoids cultured and infected in different wells; *p <0.05, by Student’s t test. FIG. 7. Illustrates that SARS-CoV-2 robustly replicates in human lung organoids and induces expression of genes involved in innate immunity and inflammation. Generation and characterization of iPSC-derived human lung organoids (LORGs). A. Representative phase contrast image of LORG at 60 Days (60D); scale bar represents 200 µm. B. Hematoxylin and Eosin staining of 60D LORG showing the alveolar like morphology; scale bars = 100 μm. C. Confocal images showing labelling for cells surrounding epithelial structures (ECAD) co- labelled with mesenchymal-cell-type marker vimentin (VIM). LORG is also co-labelled with SOX9 and FOXJ1 markers as well as with goblet cell marker MUC5AC and SOX9; scale bars = 100 μm and for enlarged image scale bar = 50 μm. D. Western blot analysis of 60D old organoid showing protein levels for ECAD, SOX9, FOXJ1, Mucin5AC. N=2 organoids. E. qRT-PCR analysis for 60D LORG shows expression of lung alveolar type 1 cell (AT1) genes AGER and HOPX; mean ± SEM of n = 3; *p < 0.05 and ***p < 0.001. F. qRT-PCR analysis for 60D LORG shows expression of lung alveolar type 2 cell (AT2) genes SFTPB, SFTPC, ABCA3 and SLC34A2; mean ± SEM of n = 3; *p < 0.05, **p < 0.01, and ***p < 0.001. G. LORGs express ACE2, TMPRSS2, DPP4, and Furin. qRT-PCR analysis for 60D LORG shows expression of ACE2, TMPRSS2, DPP4 and Furin; Data is presented as mean ± SEM of n=3; *p < 0.05 and **p < 0.01. Lung organoids are permissive to SARS-CoV-2 pseudovirus infection that is inhibited by Camostat, nafamostat, and EK1 peptide. H. Phase contrast and fluorescence imaging shows infection of SARS-CoV-2-GFP pseudovirus at 24h post infection at MOI=2; scale bars= 200 μm. I. SARS-CoV-2-Luciferase pseudovirus infection is blocked by viral entry inhibitors. Bar graph shows the luciferase activity in the presence and absence of TMPRSS2 inhibitor (Camostat mesylate), nafamostat, and a fusion inhibitor EK1 peptide to lung organoid infected with SARS-CoV-2 pseudovirus; SEM of n = 3; ***p < 0.001, by student t test. SARS-CoV-2 isolate USA-WA1/2020 robustly replicates in human lung organoids. Lung organoids were infected with SARS-CoV-2 USA-WA1/2020 virus at MOI of 2 and viral RNAs from J. supernatant and K. cellular; fractions were quantified at indicated times of infection; mean ± SEM of n = 2; **p < 0.01, and ***p < 0.001, by Student’s t test. L. SARS- CoV-2 isolate USA-WA1/2020 infection induces expression of immunity and inflammation genes. J. LORGs infected as above and gene expression of indicated genes was quantified by qRT-PCR at 72 hours post infection. Bar graph shows expression of immune response genes and cytokines. M. Organoids pre-treated with 25HC (5 µM) for 16h and infected with luciferase expressing SARS-CoV-2 pseudovirus at MOI =0.1 for 2 h and treated with fresh medium containing 5 µM 25HC. Luciferase activity was measured at 24h post-infection; mean ± SD of n = 3; ****p < 0.0001, by Student’s t test. V. DETAILED DESCRIPTION The disclosure is based, in part, on the finding that by targeting the main protease (Mpro) of a coronavirus (e.g., SARS-CoV-2), the replication of SARS-CoV-2 can be impeded. Accordingly, this disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the main protease (MPro) of a coronavirus (e.g., SARS-CoV-2). Said chemical entities are useful, e.g., for treating a coronavirus infection (e.g., SARS-CoV-2 infection (e.g., COVID-19)) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same. Provided herein are compounds of Formula (I):
Figure imgf000011_0001
Formula (I) or a pharmaceutically acceptable salt thereof, wherein: R1 is selected from the group consisting of: H, halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, and C1-6 haloalkoxy; R2 is phenyl, –CH2-phenyl, or –C4-10 alkyl, each optionally substituted with from 1-4 independently selected Ra; and each occurrence of Ra is independently selected from the group consisting of: halo; C1- 10 alkyl; C1-6 haloalkyl; cyano; C1-6 alkoxy; C1-6 haloalkoxy; C1-6 thioalkoxy; and –OH. In some embodiments of Formula (I), one or both of (aa) and (bb) apply: (aa) R1 is selected from the group consisting of: halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, and C1-6 haloalkoxy; and/or (bb) R2 is selected from the group consisting of: phenyl substituted with from 1-4 Ra; –CH2-phenyl optionally substituted with from 1-4 independently selected Ra; and –C4-10 alkyl optionally substituted with from 1-4 independently selected Ra. In some embodiments of Formula (I), when R1 is H, R2 is –CH2-phenyl or –C4-10 alkyl, each optionally substituted with from 1-4 independently selected Ra. In some embodiments of Formula (I), when R2 is phenyl, R1 is selected from the group consisting of: halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, and C1-6 haloalkoxy. In some embodiments of Formula (I), R1 is H. In some embodiments of Formula (I), R1 is selected from the group consisting of: halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, and C1-3 haloalkoxy. In some embodiments of Formula (I), R1 is selected from the group consisting of: -F, - Cl, -CH3, -CF3, and –OCH3. In some embodiments of Formula (I), R2 is phenyl which is optionally substituted with from 1-4 independently selected Ra. In some embodiments of Formula (I), R2 is phenyl substituted with from 1-4 independently selected Ra. In some embodiments of Formula (I), R2 is phenyl substituted with from 1-2 independently selected Ra. In some embodiments of Formula (I), R2 is a1
Figure imgf000012_0001
, wherein: R is selected from the group consisting of: H, F, Cl, CH3, CF3, OH, OCH3, OCF3, SCH3, CN, C2H5, C5H11, C7H15, and C8H17; and Ra2 is selected from the group consisting of: H, F, Cl, CH3, CF3, OH, OCH3, OCF3, and SCH3. In some embodiments of Formula (I), R2 is , wherein Ra1 is an
Figure imgf000012_0002
independently selected Ra; and Ra2 is H or Ra. In some embodiments of Formula (I), R2 is , wherein Ra1 is an cted Ra
Figure imgf000012_0003
independently sele ; and Ra2 is H or Ra. In some embodiments of Formula (I), Ra1 is selected from the group consisting of: -Br, -Cl, -F, cyano, C1-3 alkyl, C1-3 haloalkyl, and C1-3 alkoxy. In some embodiments of Formula (I), Ra1 is selected from the group consisting of: -Br, -Cl, C1-3 alkyl, and C1-3 haloalkyl. In some embodiments of Formula (I), Ra1 is selected from the group consisting of –Br, -Cl, -CH3, CH2CH3, and CF3. In some embodiments of Formula (I), Ra2 is H or halo. In some embodiments of Formula (I), R2 is C4-10 alkyl optionally substituted with from 1-4 independently selected Ra. In some embodiments of Formula (I), R2 is C4-10 alkyl, such as linear C4-10 alkyl. In some embodiments of Formula (I), R1 is H, and R2 is
Figure imgf000012_0004
, wherein: Ra1 is selected from the group consisting of: H, F, Cl, CH3, CF3, OH, OCH3, OCF3, SCH3, CN, C2H5, C5H11, C7H15, and C8H17; and Ra2 is selected from the group consisting of: H, F, Cl, CH3, CF3, OH, OCH3, OCF3, and SCH3. In some embodiments of Formula (I), R1 is H, and R2 is
Figure imgf000013_0001
, wherein Ra1 is an independently selected Ra; and Ra2 is H or Ra. In some embodiments of Formula (I), R1 is H, and R2 is
Figure imgf000013_0005
, wherein Ra1 is an independently selected Ra; and Ra2 is H or Ra. In some embodiments of Formula (I), R1 is H, and R2 is C4-10 alkyl optionally substituted with from 1-4 independently selected Ra. In some embodiments of Formula (I), R1 is H, and R2 is C4-10 alkyl, such as linear C4-10 alkyl. In some embodiments of Formula (I), the compound is selected from the group consisting of the following:
Figure imgf000013_0002
Figure imgf000013_0003
Figure imgf000013_0004
Figure imgf000014_0001
or a pharmaceutically acceptable salt thereof. In some embodiments of Formula (I), the compound is selected from the group consisting of compounds 2-30, or a pharmaceutically acceptable salt thereof. In some embodiments of Formula (I), the compound is not compound 1, or a pharmaceutically acceptable salt thereof. In some embodiments of Formula (I), the compound is selected from the group consisting of the following:
Figure imgf000014_0002
Figure imgf000014_0003
Figure imgf000015_0001
or a pharmaceutically acceptable salt thereof. Also provided herein are compounds of Formula (II):
Figure imgf000016_0001
Formula (II) or a pharmaceutically acceptable salt thereof, wherein: Ar is:
Figure imgf000016_0004
or
Figure imgf000016_0005
; p, q, and r are independently 0, 1, 2, or 3; each R1 is independently selected from the group consisting of: halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 haloalkoxy, C1-6 thioalkoxy, -OH, cyano, and C(O)O(C1-6 alkyl); and each R2 is selected from the group consisting of: halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 haloalkoxy, C1-6 thioalkoxy, -OH, and C(O)O(C1-6 alkyl). In some embodiments of Formula (II), Ar is
Figure imgf000016_0002
In some embodiments of Formula (II), Ar is
Figure imgf000016_0003
In some embodiments of Formula (II), q is 1. In some embodiments of Formula (II), q is 2. In some embodiments of Formula (II), each R2 is independently selected from the group consisting of: F, Cl, CN, CH3, tBu, CF3, OH, OCH3, OCF3, SCH3, and C(O)OMe. In some embodiments of Formula (II), p is 0. In some embodiments of Formula (II), p is 1, 2, or 3. In some embodiments of Formula (II), R1 is selected from the group consisting of: F, Cl, CH3, CF3, OH, OCH3, OCF3, SCH3, CN, and C(O)OMe. In some embodiments of Formula (II), the compound is selected from the group consisting of:
Figure imgf000017_0001
Figure imgf000017_0002
Figure imgf000017_0003
Figure imgf000017_0004
Figure imgf000017_0005
Figure imgf000017_0006
Figure imgf000018_0001
Figure imgf000018_0002
Figure imgf000018_0003
Figure imgf000018_0004
Figure imgf000018_0005
or a pharmaceutically acceptable salt thereof. Also provided herein are compounds of Formula (III):
Figure imgf000018_0006
or a pharmaceutically acceptable salt thereof, wherein: X is halo; R1 is selected from the group consisting of: º C1-15 alkyl optionally substituted with from 1-6 independently selected Ra; and º -L3-R3, wherein: o L3 is a bond or C1-6 alkylene optionally substituted with from 1-6 independently selected Ra; and o R3 is selected from the group consisting of C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with from 1-4 independently selected Rb; and º -(Z1-Z2)m-Z3, wherein: o each Z1 is independently C1-6 alkylene optionally substituted with from 1-6 independently selected Ra; o each Z2 is independently selected from the group consisting of: NH, N(C1-3 alkyl), O, and S; and o Z3 is selected from the group consisting of: (i) C1-6 alkyl optionally substituted with from 1-6 independently selected Ra; and (ii) –L3-R3; and o m is an integer from 1 to 10; R1a is H or C1-3 alkyl; or R1a and R1, taken together with the nitrogen atom to which each is attached forms a 4-8 membered heterocyclyl, wherein the heterocyclyl is optionally substituted with from 1-6 independently selected Rb; R2a, R2b, R2c, and R2d are independently selected from the group consisting of: halo, -OH, C1-6 alkoxy, C1-6 haloalkoxy, and nitro; each occurrence of Ra is independently selected from the group consisting of: halo, C1-6 alkoxy, C1-6 haloalkoxy, C1-6 thioalkoxy, -OH, cyano, C(O)O(C1-6 alkyl), C(O)(C1-6 alkyl), S(O)1-2C1-6 alkyl, S(O)2NR’R”, and C(O)NR’R”; each occurrence of Rb is independently selected from the group consisting of: halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 haloalkoxy, C1-6 thioalkoxy, -OH, cyano, C(O)O(C1-6 alkyl), C(O)(C1-6 alkyl), S(O)1-2C1-6 alkyl, S(O)2NR’R”, C(O)NR’R”, and –(Lc)p-Rc; each p is independently 0, 1, 2, or 3; each Lc is independently selected from the group consisting of CH2, C(=O), O, NH, N(C1-3 alkyl), S, and S(O)1-2; each Rc is independently selected from the group consisting of: C3-10 cycloalkyl, C6-10 aryl, 4-10 membered heterocyclyl, and 5-10 membered heteroaryl, each optionally substituted from 1-4 independently selected Rd; each occurrence of Rd is independently selected from the group consisting of: halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 haloalkoxy, C1-6 thioalkoxy, -OH, cyano, and C(O)O(C1-6 alkyl); and each occurrence of R’ and R” is independently H, C1-6 alkyl, or C3-6 cycloalkyl. In some embodiments of Formula (III), R2a is nitro. In some embodiments, R2b, R2c, and R2d are each H. In some embodiments of Formula (III), R2d is nitro. In some embodiments, R2a, R2b, and R2c are each H. In some embodiments of Formula (III), R2a and R2d are –OH. In some embodiments, R2b and R2c are H. In some embodiments of Formula (III), R1a is H. In some embodiments of Formula (III), R1 is C1-15 alkyl, such as C4-15 alkyl (e.g., C4-15 linear alkyl), optionally substituted with from 1-6 independently selected Ra. For example, R1 can be selected from the group consisting of:
Figure imgf000020_0001
,  , 
Figure imgf000020_0002
In some embodiments of Formula (III), R1 is –R3 or –CH2-R3. In some embodiments of Formula (III), R3 is selected from the group consisting of: C3- 10 cycloalkyl and 4-10 membered heterocyclyl, each optionally substituted with from 1-4 independently selected Rb, optionally wherein one occurrence of Rb is –(Lc)p-Rc. In some embodiments of Formula (III), R1 is selected from the group consisting of: and
Figure imgf000020_0003
. In some embodiments of Formula (III), R3 is selected from the group consisting of: C6- 10 aryl and 5-10 membered heteroaryl, each optionally substituted with from 1-4 independently selected Rb, optionally wherein one occurrence of Rb is –(Lc)p-Rc. In some embodiments of Formula (III), R1 is selected from the group consisting of:
Figure imgf000021_0003
and
Figure imgf000021_0005
Figure imgf000021_0004
In some embodiments of Formula (III), R1a and R1, taken together with the nitrogen atom to which each is attached forms a 4-8 membered heterocyclyl, wherein the heterocyclyl is optionally substituted with from 1-6 independently selected Rb, such as wherein R1a and R1 taken together with the nitrogen atom to which each is attached forms
Figure imgf000021_0001
. In some embodiments of Formula (III), R1 is -(Z1-Z2)m-Z3, optionally wherein Z1 is CH2CH2; and Z2 is –O-, such as wherein R1 is
Figure imgf000021_0006
In some embodiments of Formula (III), the compound of selected from the group consisting of the following:
Figure imgf000021_0002
Figure imgf000021_0007
Figure imgf000022_0001
Figure imgf000023_0001
or a pharmaceutically acceptable sale thereof. In some embodiments of Formula (III), the compound is selected from the group consisting of:
Figure imgf000023_0002
Figure imgf000024_0001
Figure imgf000025_0003
or a pharmaceutically acceptable salt thereof. Also provided herein are compounds of Formula (IV):
Figure imgf000025_0002
Formula (IV) or a pharmaceutically acceptable salt thereof, wherein: R1 is selected from the group consisting of: H, -OC1-6 alkyl, -O(C3-6 cycloalkyl), and NR’R”; R2 is selected from the group consisting of: -C1-6 alkyl, -C1-6 haloalkyl, -NR’R”, -(C1-6 alkylene)-NR’R”, -(C1-6 haloalkylene)-NR’R”, -(C1-6 alkylene)-NR’C(=W)R’, -(C1-6 alkylene)-NR’C(=W)OR’, -(C1-6 alkylene)-NR’C(=W)NR’R”, and
Figure imgf000025_0001
; R3 is selected from the group consisting of: -NHC(=O)R4 and -R5; R4 is selected from the group consisting of: C1-6 alkyl, -(C1-6 alkylene)-NR’R”, -(C1-6 haloalkylene)-NR’R”, and –L6-R6; R5 is selected from the group consisting of: H, -NR’R”, -(C1-6 alkylene)-NR’R”, -(C1- 6 alkylene)-C(O)OH, -(C1-6 alkylene)-C(O)O(C1-6 alkyl), C1-6 alkyl, C1-6 haloalkyl, and -(C1-6 alkylene)-OH; W is O, S, or NR’; -L6 is a bond or C1-6 alkylene; -R6 is C6-10 or 5-10 membered heteroaryl, each optionally substituted with from 1-4 substituents independently selected from the group consisting of: halo, cyano, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, and C1-6 haloalkoxy; and each occurrence of R’ and R” is independently H, C1-6 alkyl, or C3-6 cycloalkyl. In some embodiments of Formula (IV), R1 is H. In some embodiments of Formula (IV), R1 is OC1-6 alkyl, such as OMe. In some embodiments of Formula (IV), R1 is NR’R” such as NHMe, or NH(C3-6 cycloalkyl). In some embodiments of Formula (IV), R2 is
Figure imgf000026_0001
In some embodiments of Formula (IV), R2 is C1-6 alkyl, such as isopropyl; or R2 is C1- 6 haloalkyl, such as CF3 In some embodiments of Formula (IV), R2 is -(C1-6 alkylene)-NR’R”, such as – CH2NHMe. In some embodiments of Formula (IV), R2 is -(C1-6 alkylene)NR’C(=W)NR’R”, such as –CH2-NHC(=S)NH2. In some embodiments of Formula (IV), R3 is -NHC(=O)R4. In some embodiments of Formula (IV), R3 is -NHC(=O)–L6-R6, such as wherein R3 is
Figure imgf000026_0002
. In some embodiments of Formula (IV), R3 is NHC(=O)R4; and R4 is C1-6 alkyl such as methyl. In some embodiments of Formula (IV), R3 is NHC(=O)R4; and R4 is -(C1-6 alkylene)- NR’R”, such as
Figure imgf000026_0003
In some embodiments of Formula (IV), R3 is NHC(=O)R4; and R4 is -(C1-6 haloalkylene)-NR’R”, such as
Figure imgf000026_0004
In some embodiments of Formula (IV), R3 is –R5. In some embodiments of Formula (IV), R3 is NR’R”, -CH2NR’R”, -C1-3 alkyl, or – CH2C(O)OH. In some embodiments of Formula (IV), R1 is OMe, H, or NHMe; R2 is isopropyl, CF3, -NH2, -CH2NHMe, or –CH2NH(C=S)NH2; and R3 is NHC(=O)Me,
Figure imgf000026_0005
In some embodiments of Formula (IV), R1 is OMe, H, or NHMe; R2 is
Figure imgf000027_0001
; and R3 is NHC(=O)Me, methyl, NH2, CH2NH2, or CH2C(=O)OH. In some embodiments of Formula (IV), the compound has the following formula:
Figure imgf000027_0002
In some embodiments of Formula (IV), the compound is selected from the group consisting of:
 
Figure imgf000028_0001
wherein R” is C1-6 alkyl or C1-6 haloalkyl, such as wherein R” is C1-6 alkyl, such as isopropyl. Also provided herein is a compound as shown in Table I. Table I. Compounds of Formula (I), (II), and (III).
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Also provided herein are methods of treating a coronavirus infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an inhibitor for the main protease of the coronavirus. In some embodiments, the method comprises administering to the subject one or more chemical entities generically or specifically described herein. In some embodiments, the method comprises administering to the subject a compound of Formula (I), Formula (II), Formula (III), Formula (IV), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as described herein. In some embodiments, the coronavirus infection is a betacoronavirus infection. In some embodiments, the coronavirus infection is selected from the group consisting of SARS-CoV-2, SARS-CoV, and MERS-CoV infection. In some embodiments, the coronavirus infection is a SARS-CoV-2 infection. For example, the coronavirus infections can be COVID-19. In some embodiments, the subject is human. Also provided herein are methods of treating a coronavirus infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I), Formula (II), Formula (III), Formula (IV) as described anywhere herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In some embodiments, the coronavirus infection is a betacoronavirus infection. In some embodiments, the coronavirus infection is selected from the group consisting of SARS-CoV- 2, SARS-CoV, and MERS-CoV infection. In some embodiments, the coronavirus infection is a SARS-CoV-2 infection. For example, the coronavirus infections can be COVID-19. In some embodiments, the subject is human. VI. OTHER EMBODIMENTS It is to be understood that while the present disclosure has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the disclosure, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims. VII. EXAMPLES It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes. Example 1. Compounds General Procedures. All reactions were performed in flame-dried round-bottomed or modified Schlenk flasks fitted with rubber septa under a positive pressure of argon, unless otherwise noted. Air- and moisture-sensitive liquids and solutions were transferred via syringe or stainless steel cannula. Solvents (methylene chloride, ether, tetrahydrofuran, benzene, and toluene) were purified using a Pure-Solv MD-5 Solvent Purification System (Innovative Technology). Where necessary, solvents were deoxygenated by sparging with nitrogen for at least 1 hour unless otherwise noted. All other reagents were used directly from the supplier without further purification unless otherwise noted. Organic solutions were concentrated by rotary evaporation at ~25 mbar in a water bath heated to 40 °C unless otherwise noted. Analytical thin-layer chromatography (TLC) was carried out using 0.2 mm commercial glass- coated silica gel plates (silica gel 60, F254, EMD chemical). Thin layer chromatography plates were visualized by exposure to ultraviolet light and/or exposure to iodine, or to an acidic solution of ceric ammonium molybdate, or a basic solution of potassium permanganate followed by heating on a hot plate. Gas chromatographs were measured using an Agilent 7820 GC. Mass spectra (MS) were obtained on a Karatos MS9, Autospec, or an Agilent 6150 and reported as m/z (relative intensity). Accurate masses are reported for the molecular ion [M+D]+. Nuclear magnetic resonance spectra (1H NMR and 13C NMR) were recorded with a Varian Gemini (500 MHz, 1H at 500 MHz, 13C at 125 MHz; or 600 MHz, 1H at 600 MHz, 13C at 150 MHz). For CDCl3, (CD3)2SO, and CD3OD solutions, chemical shifts are reported as parts per million (ppm) referenced to residual protium or carbon of the solvent; CDCl3 δ 7.26 (1H) and 77.0 (13C) ppm, (CD3)2SO at δ 2.50 (1H) and 39.5 (13C) ppm, CD3OD δ at 3.31 (1H) and 49.1 (13C)ppm. Coupling constants are reported in Hertz (Hz). Data for 1H-NMR spectra are reported as follows: chemical shift (ppm, referenced to protium; (bs = broad singlet, s = singlet, br d = broad doublet, d = doublet, t = triplet, q = quartet, dd = doublet of doublets, td = triplet of doublets, ddd = doublet of doublet of doublets, m = multiplet, integration, and coupling constants (Hz)). HPLC was performed on an Agilent 1200 series HPLC with a Supelco Analytical Discovery® C18 (25 cm X 10 mm, 5µm) RP-HPLC column to quantify purity. Formula (I) Compounds Method A
Figure imgf000042_0001
The compounds described below were synthesized from commercially available acid chlorides and anilines in two steps. Step 1: Preparation of intermediates (I)-i1-30. A stirred solution of R2NH2 (2 mmol) in DCM at 0 °C was treated with TEA (2 mmol), followed by the dropwise addition of corresponding acid chloride (I)-SM1-30 (2 mmol) and the solution was stirred for 3 hours at room temperature. The reaction mixture was diluted in excess DCM, transferred to a separatory funnel, and extracted with water and brine solution. The organic layer was separated and concentrated under reduced pressure. The crude material was purified by column chromatography (hexane:ethyl acetate, 70:30) yield the intermediates (I)- i1-30 as solids. Step 2: Preparation of compounds 1-30 A one-necked round-bottomed flask was fitted with a reflux condenser and with a magnetic stirrer. The flask was charged with corresponding intermediate (I)-i1-30 (1 mmol), followed by the addition of Se powder (5 mmol) and tBuONa (2 mmol). The reaction mixture was left to stir at 130 °C for 12 hours. After completion of reaction, the reaction mixture was diluted with excess water and extracted with ethyl acetate. The organic layer was separate and concentrated under reduced pressure. The crude material was purified by column chromatography (hexane:ethyl acetate, 70:30) yielded compounds 1-30 as solids. Table A. Formula (I) compounds prepared by Method A.
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0002
Formula (II) Compounds Method B
Figure imgf000051_0001
Table B. Formula (II) compounds prepared by Method B.
Figure imgf000051_0003
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0002
Formula (III) Compounds Method C
Figure imgf000055_0001
To a stirred solution of 2,3-dichloro-5,8-didroxy-1,4-naphthoquinone[(III)-SM1, 50 mg, 193 μmol) or 2,3-dichloro-5-nitro-1,4-naphthoquinone([(III)-SM2, 50 mg, 184 μmol) in DCM (1 mL), appropriate amine (R1NHR1a) (2 equiv.) in EtOH (1 mL) was added, and if the amine was hydrochloride 2 equiv. TEA was added to neutralize this hydrochloride. The reaction mixture changed the color immediately (for aliphatic amines) or slowly (for aromatic amines). Then another 3 mL of EtOH was added into the vigorously stirring system which was heated to 78˚С afterwards to reflux. The consumption of starting materials was followed and monitored by TLC. And after 30 min to 5 hours (the aromatic amine reaction time is longer than the aliphatic ones), the mixture was concentrated by rotary evaporator under reduced pressure. The final compounds were recrystallized in EtOH or purified by chromatographic silica gel column at yield 30-70%. Most of the final compounds are dark red solids. For the 5-nitro-1,4-naphthoquinone reactions, there are two new points in TLC which correspond to the 2-chloro and 3-chloro regioisomers. Compounds corresponding to the faster moving spot in TLC (the upper point) were isolated (only compound 97 is the regioisomer mixture) and determined to be the 3- chloro-position substituted products by NMR via 2D-HMBC. Table C. Formula (III) compounds prepared by Method C.
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Example 2. In Silico Modeling of Selected Compounds with Schrödinger In silico modeling of selected compounds was performed using the Glide docking module of the Schrödinger 11.5 modeling software suite. A crystal structure of the SARS-CoV- 2 Mpro bound to N3 (PDB ID: 6UL7) was first refined using Prime (i. Jin, Z. et al, Nature 2020, 582, 289-293; ii. Jacobson, M. P. et al, J Mol Biol 2002, 320 (3), 597-608; iii. Jacobson, M. P. et al, Proteins 2004, 55 (2), 351-367). The Optimized Potentials for Liquid Simulations All- Atom (OPLS) force field and the Surface generalized Born (SGB) continuum solution model were used to optimize and minimize the crystal structure. The docking grid was generated as a 15 Å cube centered on N3 (x = -14, y = 17.7, z = 67.1). Ligprep was used to generate a minimized 3D structure for all antiviral compounds using the OPLS 2001 force field. Docking was performed with Glide XP (i. Friesner, R. A. et al, J Med Chem 2004, 47 (7), 1739-1749; ii. Halgren, T. A. et al, J Med Chem 2004, 47 (7), 1750-1759; iii. Friesner, R. A. et al, J Med Chem 2006, 49 (21), 6177-6196). Compounds were ranked by docking score and the docking poses were evaluated for interactions with key residues, such as the catalytic dyad His 41 and Cys 145. See, for example FIG.2.   Example 3. SARS-CoV-2 Main Protease Protein Expression and Purification The GST-tagged SARS-CoV-2 main protease was expressed in E. Coli BL21 competent cells (New England Biolabs) transformed with pET41b plasmid by heat shock and spread on a LB Kanamycin agar plate, then incubated overnight at 37 °C. 2-3 colonies were picked and transferred to 5 mL of LB media treated with kanamycin (0.5 mg/mL final concentration), then grown overnight shaking at 37 °C. The overnight culture was then transferred to 2 L of LB kanamycin media and incubated at 37 °C until OD 0.8. The culture was cooled at 4 °C for 20 mins and induced with 0.5 mM isopropyl β-D-1- thiogalactopyranoside (IPTG), then grown shaken at 16 °C. Cell pellets were collected by centrifugation (5,000 g for 10 min at 4 °C) and the supernatant was discarded. The pellets were resuspended in lysis buffer (50 mM Tris-HCl pH 7.5, 500 mM NaCl, 1.0% Triton x-100, 0.5 mM EDTA, 1x Complete protease inhibitor cocktail, 1 mM β-mercaptoethanol in RNase-free water; 6 mL per gram) with DNase 1 (5U per mL, RNase-free) and incubated at 4 °C for 1 hour and sonicated. The suspension was centrifuged at 10,000 g for 20 min and the supernatant was transferred to a glutathione sepharose affinity resin column that had been pre-equilibrized with lysis buffer. The supernatant was incubated with the affinity resin column at 4 °C for 2 hours with end-over-end rotation. After incubation, the column was washed with 5 bed volumes of lysis buffer, 3 x 5 bed volumes of wash buffer (50 mM Tris-HCl pH 7.5, 500 mM NaCl, 0.1% Triton x-100, 0.5 mM EDTA, 1x Complete protease inhibitor cocktail, 1 mM β- mercaptoethanol in RNase-free water), and 5 bed volumes of pre-elution buffer (50 mM Tris- HCl pH 7.5, 100 mM NaCl in RNase-free water). The protease was eluted with elution buffer (50 mM Tris-HCl pH 7.5, 100 mM NaCl, 40 mM glutathione in RNase-free water) and collected in 1 mL fractions until no further protein was collected (3-5 bed volumes total). The purity of the fractions was determined by SDS-page and fractions of purified protein were collected and concentrated to 2 mg/mL. The GST-tagged purified protein was incubated with TEV protease at 37 °C for 4 hours to obtain the untagged SARS-CoV-2 protease. Excess GST and GST-TEV protease were removed by glutathione sepharose affinity column purification. The untagged proteases were eluted in 50 mM Tris-HCl pH 7.5, 100 mM NaCl in RNase-free water. The purity of the fractions was determined by SDS-page and fractions of purified protein were collected and concentrated to 5 µM. The protein solutions were transferred to a Slyde-A- Lyzer Dialysis Cassette (20,000 MWCO, Thermo Scientific) and dialyzed overnight at 4 °C against dialysis buffer (50 mM Tris-HCl pH 7.4, 200 mM NaCl, 1 mM EDTA, 1 mM DTT in RNase-free water). Example 4. FRET Protease Activity Assay Activity of the SARS-CoV-2 main proteases was determined using fluorescence resonance energy transfer (FRET) assay adapted from Muramatsu et al (Proc Natl Acad Sci U S A 2016, 113 (46), 12997-13002). The fluorogenic peptide Dabcyl-Val-Asn-Ser-Thr-Leu-Gln- Ser-Gly-Leu-Arg-Lys-EDANS was used as a substrate (AnaSpec Inc). All reactions were performed in a black 96-well plate with 200 µL assay buffer (50 mM Tris-HCl pH 7.4, 200 mM NaCl, 1 mM EDTA, 1 mM DTT in RNase-free water) with 25 nM SARS-CoV-2 protease and 3 µM substrate. Compounds were screened at a concentration of 10 µM. All inhibitors were dissolved in DMSO and added to a final concentration of 0.2% DMSO. Fluorescent readings were normalized to 200 µL assay buffer with 3 µM substrate and 10 µM of inhibitor with a final concentration of 0.2% DMSO. Cleavage between the Gln and Ser residues was monitored by a BioTek Synergy plate reader, with excitation at 380 nm and emission at 485 nm every 10 minutes for 1 hour. The relative rate for wild type protease and each inhibitor concentration was averaged from three assay results. The relative reaction rates were normalized to wild type protease, and the relative activity was reported as a percentage of the wild type protease activity in Table 1. Additionally, dose-response curves were generated for compounds that showed significant inhibition at 10 µM using the assay conditions described above. The dose-response curve for ebselen (compound 1) was also generated as a positive control. Inhibitors were screened at six concentrations ranging from 0.100-10 µM. As before, cleavage between the Gln and Ser residues was monitored by a BioTek Synergy plate reader, with excitation at 380 nm and emission at 485 nm every 10 minutes for 1 hour. The relative rate for wild type protease and each inhibitor concentration was averaged from three assay results. The relative reaction rates were normalized to wild type protease and dose-response curves were plotted in GraphPad Prism 6. Example curves for Formula (I) compounds are shown in FIG. 5. To determine if the compounds were covalent or rapid, reversible inhibitors of SARS- CoV-2 Mpro, pre-incubation dilution experiments were performed. In assay buffer, 250 nM protease was incubated on ice for 30 mins with 10 µM of either compound ebselen (compound 1), compounds 4, 7, 19, 20 or 25. The reaction mixture was then diluted 10-fold with assay buffer and the fluorogenic peptide substrate was added to a final concentration of 3 µM. The experiment was also repeated with assay buffer free of DTT to determine the effects of the reducing agent on the mechanism of inhibition. For each compound, the rate of product formation after preincubation-dilution was compared to the rate of product formation for Mpro with 0.2% DMSO, 1 µM, and 10 µM compound without dilution. Results are shown in FIG. 3. Example 5. logD Measurements logD values were measured at pH = 7.4 by the shake-flask method, according to the literature (Donovan, S. F. et al, J Chromatogr A 2002, 952 (1-2), 47-61; Box, K. et al, Anal Chem 2003, 75 (4), 883-892). Results are shown in Table 1. Example 6. Inhibition Mechanism The reaction velocities observed over a range of substrate (1.5, 3, 4.5, 6, 9, 15, 22.5, and 30 µM) and inhibitor concentrations (0, 0.5, 1, 5, and 10 µM) for either ebselen (compound 1) or compound 4 were globally fit to a generalized mixed-model of inhibition (See FIG. 4) represented by the following equilibrium reaction scheme: This mechanism allows for inhibitor binding to both the free enzyme and the enzyme-substrate complex with varying affinity described by the following rate equation:
Figure imgf000073_0001
The term Ki describes the affinity of the inhibitor for free enzyme. The mechanism can be evaluated by the term α. When α approaches 1, the inhibitor is considered non-competitive and when α approached infinity, the inhibitor is considered competitive.
Figure imgf000073_0002
Example 7. Antiviral Assays Approximately 2x104 Vero E6 cells (ATCC) were plated in 100 μL of DMEM (Gibco) supplemented with 10% serum in each well of a 96-well flat bottom plate and allowed to adhere overnight. Cells were pre-treated with 0.10-20 μM test compound in 100 μL of the same media for 1 hr, washed, and infected with SARS-CoV-2 (USA_WA01/2020 [World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch]) in 50 μL of media at 0.01 MOI for 1.5 hrs. Cells were then washed, and test compounds reapplied at 0.10- 20 μM in 100 μL media and incubated at 37 °C in 5% CO2 for 72 hrs. Following incubation, 10 μL room temperature WST8 reagent was added to each well, mixed and incubated for an additional 3 hrs at 37 °C and 5% CO2. O.D. at 460 nm was read on a BioTek plate reader. For antiviral assays in Calu-3 human lung cells, approximately 1.5x105 Calu-3 cells (ATCC) were plated in 500 μL of EMEM (Gibco) supplemented with 10% FBS, 2 mM L- glutamine, and 1% penicillin/streptomycin in each well of 24-well flat bottom plates and allowed to adhere overnight. Cells were pre-treated with 0.10-20 μM test compound in DMSO (0.2 % final v/v) of the same media for 1 hr, washed, and infected with SARS-CoV-2 (USA_WA01/2020) in reduced serum media (2-3%) at 0.1 MOI for 1.5 hrs. Cells were then washed with PBS, and test compounds reapplied at 0.10-20 μM in 500 μl media and incubated at 37 °C in 5% CO2 for 24 hrs. At 24 hours post-infection, supernatant and infected cells were collected and lysed using TRIzol and RNA was extracted using a Direct-zol RNA Kit (Zymo) and quantified by RT-qPCR using SARS-CoV-2 N primers, as previously described (Tiwari, S. K. et al, Stem Cell Reports 2021, 16 (3), 437-445; Li, N. et al, Cell Rep 2021, 35 (6), 109091; Wang, S. et al, EMBO J 2020, 39 (21), e106057). The primers are as follows: Forward: CACATTGGCACCCGCAATC; Reverse: GAGGAACGAGAAGAGGCTTG. Results are shown in Table 1. Table 1. Enzymatic and cellular anti-viral activity of Formula (I) compounds
Figure imgf000074_0001
Figure imgf000075_0001
Example 8. Lung organoid infection and treatment with compound 24 Human iPSC derived lung organoids were generated using previously published methods and characterized by expression of ACE2, TMPRSS2, and alveolar cell epithelial markers (SFTPC, SFTPB, HOPX). The 60D differentiated lung organoids were pretreated with 5 µM E24 for 2 h and infected with SARS-CoV-2 as described previously (Tiwari, S. K. et al, Stem Cell Reports 2021, 16 (3), 437-445). SARS-CoV-2 isolate USA-WA1/2020 was obtained from BEI Resources. SARS-CoV-2 was propagated, and infectious units quantified by plaque assay using Vero E6 cells. Human iPSC derived lung organoids, were infected with SARS- CoV-2 at MOI=2 for 2 h at 37 °C. Then cells were washed, and fresh medium was added. At 72 h post infection supernatant and infected organoids were collected and lysed using TRIzol and RNA was extracted using a Direct-zol RNA Kit (Zymo) and quantified by RT-qPCR using SARS-CoV-2 N primers. Results are shown in FIG. 6.

Claims

What is claimed is: 1. A compound of Formula (I):
Figure imgf000076_0001
Formula (I) or a pharmaceutically acceptable salt thereof, wherein: R1 is selected from the group consisting of: H, halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, and C1-6 haloalkoxy; R2 is phenyl, –CH2-phenyl, or –C4-10 alkyl, each optionally substituted with from 1-4 independently selected Ra; and each occurrence of Ra is independently selected from the group consisting of: halo; C1- 10 alkyl; C1-6 haloalkyl; cyano; C1-6 alkoxy; C1-6 haloalkoxy; C1-6 thioalkoxy; –OH; C(O)OC1-6 alkyl; and C(O)C1-6 alkyl.
2. The compound of claim 1, wherein one or both of (aa) and (bb) apply: (aa) R1 is selected from the group consisting of: halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, and C1-6 haloalkoxy; or (bb) R2 is selected from the group consisting of: phenyl substituted with from 1-4 Ra; –CH2-phenyl optionally substituted with from 1-4 independently selected Ra; and –C4-10 alkyl optionally substituted with from 1-4 independently selected Ra.
3. The compound of claims 1 or 2, wherein R1 is H.
4. The compound of claims 1 or 2, wherein R1 is selected from the group consisting of: halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, and C1-3 haloalkoxy.
5. The compound of any one of claims 1-4, wherein R1 is selected from the group consisting of: -F, -Cl, -CH3, -CF3, and –OCH3.
6. The compound of any one of claims 1-5, wherein R2 is phenyl which is optionally substituted with from 1-4 independently selected Ra.
7. The compound of any one of claims 1-6, wherein R2 is phenyl substituted with from 1-4 independently selected Ra.
8. The compound of any one of claims 1-7, wherein R2 is phenyl substituted with from 1-2 independently selected Ra.
9. The compound of any one of claims 1-8, wherein R2 is
Figure imgf000077_0002
, wherein: Ra1 is selected from the group consisting of: H, F, Cl, CH3, CF3, OH, OCH3, OCF3, SCH3, CN, C2H5, C5H11, C7H15, and C8H17; and Ra2 is selected from the group consisting of: H, F, Cl, CH3, CF3, OH, OCH3, OCF3, and SCH3.
10. The compound of any one of claims 1-8, wherein R2 is
Figure imgf000077_0001
, wherein Ra1 is an independently selected Ra; and Ra2 is H or Ra.
11. The compound of any one of claims 1-8, wherein R2 is
Figure imgf000077_0003
, wherein Ra1 is an independently selected Ra; and Ra2 is H or Ra.
12. The compound of claims 10 or 11, wherein Ra1 is selected from the group consisting of: -Br, -Cl, -F, cyano, C1-3 alkyl, C1-3 haloalkyl, and C1-3 alkoxy.
13. The compound of any one of claims 10-12, wherein Ra1 is selected from the group consisting of: -Br, -Cl, C1-3 alkyl, and C1-3 haloalkyl.
14. The compound of any one of claims 10-13, wherein Ra1 is selected from the group consisting of –Br, -Cl, -CH3, CH2CH3, and CF3.
15. The compound of any one of claims 10-14, wherein Ra2 is H or halo.
16. The compound of any one of claims 1-5, wherein R2 is C4-10 alkyl optionally substituted with from 1-4 independently selected Ra.
17. The compound of claim 16, wherein R2 is C4-10 alkyl, such as linear C4-10 alkyl.
18. The compound of any one of claims 1-17, wherein the compound is selected from the group consisting of the following:
Figure imgf000078_0001
Figure imgf000078_0002
Figure imgf000079_0001
or a pharmaceutically acceptable salt thereof.
19. The compound of any one of claims 1-17, wherein the compound is selected from the group consisting of the following:
Figure imgf000079_0002
Figure imgf000079_0003
Figure imgf000080_0001
or a pharmaceutically acceptable salt thereof.
20. A compound of Formula (II):
Figure imgf000081_0001
Formula (II) or a pharmaceutically acceptable salt thereof, wherein: Ar is:
Figure imgf000081_0002
or
Figure imgf000081_0003
; p, q, and r are independently 0, 1, 2, or 3; each R1 is independently selected from the group consisting of: halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 haloalkoxy, C1-6 thioalkoxy, -OH, cyano, and C(O)O(C1-6 alkyl); and each R2 is selected from the group consisting of: halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 haloalkoxy, C1-6 thioalkoxy, -OH, cyano, and C(O)O(C1-6 alkyl).
21. The compound of claim 20, wherein Ar is
Figure imgf000081_0004
22. The compound of claim 20, wherein Ar is
Figure imgf000081_0005
23. The compound of any one of claims 20-22, wherein q is 1.
24. The compound of any one of claims 20-22, wherein q is 2.
25. The compound of any one of claims 20-24, wherein each R2 is independently selected from the group consisting of: F, Cl, CN, CH3, tBu, CF3, OH, OCH3, OCF3, SCH3, and C(O)OMe.
26. The compound of any one of claims 20-25, wherein p is 0.
27. The compound of any one of claims 20-25, wherein p is 1, 2, or 3.
28. The compound of any one of claims 20-25 or 27, wherein R1 is selected from the group consisting of: F, Cl, CH3, CF3, OH, OCH3, OCF3, SCH3, CN, and C(O)OMe.
29. The compound of any one of claims 20-28, wherein the compound is selected from the group consisting of:
Figure imgf000082_0001
Figure imgf000082_0002
Figure imgf000082_0003
Figure imgf000083_0001
Figure imgf000083_0002
Figure imgf000083_0003
Figure imgf000083_0004
Figure imgf000083_0005
Figure imgf000083_0006
Figure imgf000083_0007
or a pharmaceutically acceptable salt thereof.
30. A compound of Formula (III):
Figure imgf000084_0001
or a pharmaceutically acceptable salt thereof, wherein: X is halo (e.g., -Cl); R1 is selected from the group consisting of: º -C1-15 alkyl optionally substituted with from 1-6 independently selected Ra; and º -L3-R3, wherein: o -L3 is a bond or C1-6 alkylene optionally substituted with from 1-6 independently selected Ra; and o -R3 is selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with from 1-4 independently selected Rb; and º -(Z1-Z2)m-Z3, wherein: o each Z1 is independently C1-6 alkylene optionally substituted with from 1-6 independently selected Ra; o each Z2 is independently selected from the group consisting of: -NH-, - N(C1-3 alkyl)-, -O-, and –S-; and o Z3 is selected from the group consisting of: (i) C1-6 alkyl optionally substituted with from 1-6 independently selected Ra; and (ii) –L3-R3; and o m is an integer from 1 to 10; R1a is H or C1-3 alkyl; or R1a and R1, taken together with the nitrogen atom to which each is attached forms a 4- 8 membered heterocyclyl ring, wherein the heterocyclyl ring is optionally substituted with from 1-6 independently selected Rb; R2a, R2b, R2c, and R2d are independently selected from the group consisting of: H, halo, -OH, C1-6 alkoxy, C1-6 haloalkoxy, and nitro; each occurrence of Ra is independently selected from the group consisting of: halo, C1- 6 alkoxy, C1-6 haloalkoxy, C1-6 thioalkoxy, -OH, cyano, C(O)O(C1-6 alkyl), C(O)(C1-6 alkyl), S(O)1-2C1-6 alkyl, S(O)2NR’R”, -NR’R”, and C(O)NR’R”; each occurrence of Rb is independently selected from the group consisting of: halo, C1- 6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 haloalkoxy, C1-6 thioalkoxy, -OH, cyano, C(O)O(C1-6 alkyl), C(O)(C1-6 alkyl), S(O)1-2C1-6 alkyl, S(O)2NR’R”, -NR’R”, C(O)NR’R”, and –(Lc)p-Rc; each p is independently 0, 1, 2, or 3; each Lc is independently selected from the group consisting of CH2, C(=O), O, NH, N(C1-3 alkyl), S, and S(O)1-2; each Rc is independently selected from the group consisting of: C3-10 cycloalkyl, C6-10 aryl, 4-10 membered heterocyclyl, and 5-10 membered heteroaryl, each optionally substituted from 1-4 independently selected Rd; each occurrence of Rd is independently selected from the group consisting of: halo, C1- 6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 haloalkoxy, C1-6 thioalkoxy, -OH, cyano, and C(O)O(C1-6 alkyl); and each occurrence of R’ and R” is independently H, C1-6 alkyl, C3-6 cycloalkyl, C(O)C1-6 alkyl, C(O)OC1-6 alkyl, or S(O)1-2C1-6 alkyl.
31. The compound of claim 30, wherein R2a is nitro.
32. The compound of claims 30 or 31, wherein R2b, R2c, and R2d are each H.
33. The compound of claim 30, wherein R2d is nitro.
34. The compound of claim 33, wherein R2a, R2b, and R2c are each H.
35. The compound of claim 30, wherein R2a and R2d are –OH.
36. The compound of claim 35, wherein R2b and R2c are H.
37. The compound of any one of claims 30-36, wherein R1a is H.
38. The compound of any one of claims 30-37, wherein R1 is C1-15 alkyl, such as C4-15 alkyl (e.g., C4-15 linear alkyl), optionally substituted with from 1-6 independently selected Ra.
39. The compound of any one of claims 30-38, wherein R1 is selected from the group consisting of:
Figure imgf000086_0001
.
40. The compound of any one of claims 30-37, wherein R1 is –R3 or –CH2-R3.
41. The compound of any one of claims 30-37 or 40, wherein R3 is selected from the group consisting of: C3-10 cycloalkyl and 4-10 membered heterocyclyl, each optionally substituted with from 1-4 independently selected Rb, optionally wherein one occurrence of Rb is –(Lc)p-Rc.
42. The compound of any one of claims 30-37 or 41, wherein R1 is selected from the group consisting of:
Figure imgf000086_0002
; ; ; ; and
Figure imgf000086_0003
Figure imgf000086_0004
43. The compound of any one of claims 30-37 or 40, wherein R3 is selected from the group consisting of: C6-10 aryl and 5-10 membered heteroaryl, each optionally substituted with from 1-4 independently selected Rb, optionally wherein one occurrence of Rb is –(Lc)p- Rc.
44. The compound of any one of claims 30-37, 40, or 43, wherein R1 is selected from the group consisting of:
Figure imgf000086_0005
Figure imgf000086_0006
and
Figure imgf000087_0002
45. The compound of any one of claims 30-36, wherein R1a and R1, taken together with the nitrogen atom to which each is attached forms a 4-8 membered heterocyclyl, wherein the heterocyclyl is optionally substituted with from 1-6 independently selected Rb, such as wherein R1a and R1 taken together with the nitrogen atom to which each is attached forms
Figure imgf000087_0003
46. The compound of any one of claims 30-36, wherein R1 is -(Z1-Z2)m-Z3, optionally wherein Z1 is CH2CH2; and Z2 is –O-, such as wherein R1 is
Figure imgf000087_0004
47. The compound of any one of claims 30-46, wherein the compound of selected from the group consisting of the following:
Figure imgf000087_0001
Figure imgf000087_0005
Figure imgf000088_0001
Figure imgf000089_0001
or a pharmaceutically acceptable sale thereof.
48. The compound of any one of claims 30-47, wherein the compound is selected from the group consisting of:
Figure imgf000089_0002
Figure imgf000090_0001
Figure imgf000091_0003
or a pharmaceutically acceptable salt thereof.
49. A compound of Formula (IV):
Figure imgf000091_0001
Formula (IV) or a pharmaceutically acceptable salt thereof, wherein: R1 is selected from the group consisting of: H, -OC1-6 alkyl, -O(C3-6 cycloalkyl), and NR’R”; R2 is selected from the group consisting of: -C1-6 alkyl, -C1-6 haloalkyl, -NR’R”, -(C1-6 alkylene)-NR’R”, -(C1-6 haloalkylene)-NR’R”, -(C1-6 alkylene)-NR’C(=W)R’, -(C1-6 alkylene)-NR’C(=W)OR’, -(C1-6 alkylene)-NR’C(=W)NR’R”, and
Figure imgf000091_0002
R3 is selected from the group consisting of: -NHC(=O)R4 and -R5; R4 is selected from the group consisting of: C1-6 alkyl, -(C1-6 alkylene)-NR’R”, -(C1-6 haloalkylene)-NR’R”, and –L6-R6; R5 is selected from the group consisting of: H, -NR’R”, -(C1-6 alkylene)-NR’R”, -(C1- 6 alkylene)-C(O)OH, -(C1-6 alkylene)-C(O)O(C1-6 alkyl), C1-6 alkyl, C1-6 haloalkyl, and -(C1-6 alkylene)-OH; W is O, S, or NR’; -L6 is a bond or C1-6 alkylene; -R6 is C6-10 or 5-10 membered heteroaryl, each optionally substituted with from 1-4 substituents independently selected from the group consisting of: halo, cyano, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, and C1-6 haloalkoxy; and each occurrence of R’ and R” is independently H, C1-6 alkyl, or C3-6 cycloalkyl.
50. The compound of claim 49, wherein R1 is H.
51. The compound of claim 49, wherein R1 is OC1-6 alkyl, such as OMe.
52. The compound of claim 49, wherein R1 is NR’R” such as NHMe, or NH(C3-6 cycloalkyl).
53. The compound of any one of claims 49-52, wherein R2 is .
54. The compound of any one of claims 49-52, wherein R2 is C1-6 alkyl, such as isopropyl; or R2 is C1-6 haloalkyl, such as CF3
55. The compound of any one of claims 49-52, wherein R2 is -(C1-6 alkylene)- NR’R”, such as –CH2NHMe.
56. The compound of any one of claims 49-52, wherein R2 is -(C1-6 alkylene)NR’C(=W)NR’R”, such as –CH2-NHC(=S)NH2.
57. The compound of any one of claims 49-56, wherein R3 is -NHC(=O)R4.
58. The compound of any one of claims 49-57, wherein R3 is -NHC(=O)–L6-R6, such as wherein R3 is
Figure imgf000092_0002
59. The compound of any one of claims 49-57, wherein R3 is NHC(=O)R4; and R4 is C1-6 alkyl such as methyl.
60. The compound of any one of claims 49-57, wherein R3 is NHC(=O)R4; and R4 is -(C1-6 alkylene)-NR’R”, such a
Figure imgf000092_0001
61. The compound of any one of claims 49-57, wherein R3 is NHC(=O)R4; and R4 is -(C1-6 haloalkylene)-NR’R”, such as
Figure imgf000093_0001
.
62. The compound of any one of claims 49-56, wherein R3 is –R5.
63. The compound of any one of claims 49-56 or 62, wherein R3 is NR’R”, - CH2NR’R”, -C1-3 alkyl, or –CH2C(O)OH.
64. The compound of claim 49, wherein: R1 is OMe, H, or NHMe; R2 is isopropyl, CF3, -NH2, -CH2NHMe, or –CH2NH(C=S)NH2; and R3 is NHC(=O)Me,
Figure imgf000093_0003
65. The compound of claim 49, wherein R1 is OMe, H, or NHMe; R2 is
Figure imgf000093_0004
; and R3 is NHC(=O)Me, methyl, NH2, CH2NH2, or CH2C(=O)OH.
66. The compound of any one of claims 49-63, wherein the compound has the following formula:
Figure imgf000093_0002
.
67. The compound of any one of claims 49-64, wherein the compound is selected from the group consisting of:
Figure imgf000094_0001
 
Figure imgf000095_0001
wherein R” is C1-6 alkyl or C1-6 haloalkyl, such as wherein R” is C1-6 alkyl, such as isopropyl.
68. A compound selected from any one of compounds 1-103 as described herein, or a pharmaceutically acceptable salt thereof.
69. A pharmaceutical composition comprising a compound of any one of claims 1- 68 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients.
70. A method of treating a coronavirus infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an inhibitor for the main protease of the coronavirus.
71. The method of claim 70, comprising administering one or more chemical entities generically or specifically described herein.
72. A method of treating a coronavirus infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 69.
73. The method of any one of claims 70-72, wherein the coronavirus infection is a betacoronavirus infection.
74. The method of any one of claims 70-73, wherein the coronavirus infection is selected from the group consisting of SARS-CoV-2, SARS-CoV, and MERS-CoV infection.
75. The method of any one of claims 70-74, wherein the coronavirus infection is a SARS-CoV-2 infection.
76. The method of any one of claims 70-75, wherein the coronavirus infections is COVID-19.
77. The method of any one of claims 70-76, wherein the subject is human.
PCT/US2021/055199 2020-10-16 2021-10-15 Methods and compounds to treat sars infections WO2022081984A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063093044P 2020-10-16 2020-10-16
US63/093,044 2020-10-16
US202163235977P 2021-08-23 2021-08-23
US63/235,977 2021-08-23

Publications (1)

Publication Number Publication Date
WO2022081984A1 true WO2022081984A1 (en) 2022-04-21

Family

ID=81208683

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/055199 WO2022081984A1 (en) 2020-10-16 2021-10-15 Methods and compounds to treat sars infections

Country Status (1)

Country Link
WO (1) WO2022081984A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4618669A (en) * 1984-06-22 1986-10-21 A. Nattermann & Cie Gmbh S-(carbamoyl-phenylselenyl) derivatives of glutathione and of aminomercaptocarboxylic acids
US4711961A (en) * 1984-03-01 1987-12-08 A. Nattermann & Cie Gmbh Benzisoselenazolethiones and process for the treatment of various diseases in humans
US8592468B2 (en) * 2006-05-22 2013-11-26 Thioredoxin Systems Ab Bacterial thioredoxin reductase inhibitors and methods for use thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4711961A (en) * 1984-03-01 1987-12-08 A. Nattermann & Cie Gmbh Benzisoselenazolethiones and process for the treatment of various diseases in humans
US4618669A (en) * 1984-06-22 1986-10-21 A. Nattermann & Cie Gmbh S-(carbamoyl-phenylselenyl) derivatives of glutathione and of aminomercaptocarboxylic acids
US8592468B2 (en) * 2006-05-22 2013-11-26 Thioredoxin Systems Ab Bacterial thioredoxin reductase inhibitors and methods for use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE PubChem SUBSTANCE 29 November 2016 (2016-11-29), ANONYMOUS : "SUBSTANCE RECORD 2-(4-Chlorophenyl)-1,2-benzisoselenazol-3(2H)-one", XP055934811, Database accession no. SID 319220000 *
DATABASE PubChem SUBSTANCE 5 November 2018 (2018-11-05), ANONYMOUS : "SUBSTANCE RECORD SID 376178501", XP055934807, Database accession no. SID 376178501 *

Similar Documents

Publication Publication Date Title
US10829473B2 (en) Modulators of the cystic fibrosis transmembrane conductance regulator protein and methods of use
CN103097384B (en) Substituted imidazo[1,2-a]pyrimidines and pyridines
CN103384668B (en) Compound and composition for suppressing NAMPT
KR20220119088A (en) KRAS mutant protein inhibitor
Pillaiyar et al. Small-molecule thioesters as SARS-CoV-2 main protease inhibitors: enzyme inhibition, structure–activity relationships, antiviral activity, and X-ray structure determination
CN110248939A (en) Tyrosine amide derivatives as RHO- kinase inhibitor
Malpani et al. Efficient synthesis of 3H, 3′ H-spiro [benzofuran-2, 1′-isobenzofuran]-3, 3′-dione as novel skeletons specifically for influenza virus type B inhibition
EP2788000A1 (en) Pyrrolopyrimidines as janus kinase inhibitors
CN105153142B (en) The Furazan Derivatives and antitumor activity of cumarin parent nucleus
TW201702245A (en) Substituted polycyclic pyridone derivatives and prodrug thereof
JP2018526336A (en) Novel tetracyclic 4-oxo-pyridine-3-carboxylic acid derivatives for the treatment and prevention of hepatitis B virus infection
CN105658640A (en) Alkynyl alcohols and methods of use
AU2007263017A1 (en) Pyridine and pyrazine derivatives as MNK kinase inhibitors
ES2250883T3 (en) DERIVATIVES OF 2-PIRIDINIL AND 2-PIRIMIDINIL-6,7,8,9-TETRAHYDROPIRIMIDO (1,2-A) PIRIMIDIN-4-ONA SUBSTITUTED WITH HETEROARILO.
KR20120106869A (en) Novel antiviral compounds
CN101432284A (en) Dihydropyrazolopyrimidinone derivative
TW201014855A (en) Compounds for the treatment of hepatitis C
WO2021155317A1 (en) Asgpr-binding compounds for the degradation of extracellular proteins
KR20120034607A (en) Imidazo[2,1-b][1,3,4]thiadiazole derivatives
CN104592211A (en) Biphenyl compound and use thereof
JP6805172B2 (en) Histone deacetylase inhibitors and compositions and methods of their use
CN105399756B (en) BTK inhibitor and application thereof
ES2326337T3 (en) NEW PIRROLODIHYDROISOQUINOLINS AS PDE INHIBITORS10.
EP3110815B1 (en) Indoloquinolone compounds as anaplastic lymphoma kinase (alk) inhibitors
WO2021205290A1 (en) Compounds and method of treating covid-19

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: 21881179

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: 21881179

Country of ref document: EP

Kind code of ref document: A1