US20190022116A1 - N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto - Google Patents

N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto Download PDF

Info

Publication number
US20190022116A1
US20190022116A1 US15/537,087 US201515537087A US2019022116A1 US 20190022116 A1 US20190022116 A1 US 20190022116A1 US 201515537087 A US201515537087 A US 201515537087A US 2019022116 A1 US2019022116 A1 US 2019022116A1
Authority
US
United States
Prior art keywords
alkyl
amino
aryl
alkoxy
hydroxy
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/537,087
Other languages
English (en)
Inventor
George R. Painter
David Guthrie
Gregory R. BLUEMLING
Michael G. Natchus
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Emory University
Original Assignee
Emory University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=56151433&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20190022116(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Emory University filed Critical Emory University
Priority to US15/537,087 priority Critical patent/US20190022116A1/en
Assigned to EMORY UNIVERSITY reassignment EMORY UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GUTHRIE, DAVID B., BLUEMLING, GREGORY R., NATCHUS, MICHAEL G., PAINTER, GEORGE R.
Assigned to EMORY UNIVERSITY reassignment EMORY UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BLUEMLING, GREGORY R., GUTHRIE, DAVID B., NATCHUS, MICHAEL G., PAINTER, GEORGE R.
Publication of US20190022116A1 publication Critical patent/US20190022116A1/en
Assigned to DEFENSE THREAT REDUCTION AGENCY, US DOD reassignment DEFENSE THREAT REDUCTION AGENCY, US DOD CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: EMORY UNIVERSITY
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/067Pyrimidine radicals with ribosyl as the saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/073Pyrimidine radicals with 2-deoxyribosyl as the saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • C07H19/11Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids containing cyclic phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This disclosure relates to N4-hydroxycytidine nucleoside derivatives, compositions, and methods related thereto. In certain embodiments, the disclosure relates to the treatment and prophylaxis of viral infections.
  • the causative agents for Eastern, Western, and Venezuelan Equine Encephalitis (EEE, WEE and VEE, respectively) and Chikungunya fever (CHIK) are vector-borne viruses (family Togaviridae, genus Alphavirus) that can be transmitted to humans through mosquito bites.
  • the equine encephalitis viruses are CDC Category B pathogens, and the CHIK virus is Category C.
  • CHIK virus is Category C.
  • This disclosure relates to N4-hydroxycytidine and derivatives, pharmaceutical compositions, and uses related thereto.
  • the disclosure relates to a compound having formula I,
  • the disclosure contemplates derivatives of compounds disclosed herein such as those containing one or more, the same or different, substituents.
  • the disclosure contemplates pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a compound disclosed herein.
  • the pharmaceutical composition is in the form of a tablet, capsule, pill, or aqueous buffer, such as a saline or phosphate buffer.
  • the pharmaceutical composition comprises a compound disclosed herein and a propellant.
  • the propellant is an aerosolizing propellant is compressed air, ethanol, nitrogen, carbon dioxide, nitrous oxide, hydrofluoroalkanes (HFAs), 1,1,1,2,-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoropropane or combinations thereof.
  • the disclosure contemplates a pressurized or unpressurized container comprising a compound or pharmaceutical composition as described herein.
  • the container is a manual pump spray, inhaler, meter-dosed inhaler, dry powder inhaler, nebulizer, vibrating mesh nebulizer, jet nebulizer, or ultrasonic wave nebulizer.
  • the disclosure relates to methods of treating or preventing a viral infection comprising administering an effective amount of a compound or pharmaceutical composition disclosed herein to a subject in need thereof.
  • the viral infection is an alphavirus or coronaviruses and flavivirus. In certain embodiments, the viral infection is an orthomyxoviridae or paramyxoviridae. In certain embodiments, the viral infection is selected from MERS coronavirus, Eastern equine encephalitis virus, Western equine encephalitis virus, Venezuelan equine encephalitis virus, Ross River virus, Powassan virus, Barmah Forest virus and Chikungunya virus.
  • the compound or pharmaceutical composition is administered orally, intravenously, or through the lungs.
  • the disclosure relates to the use of a compound as described herein in the production of a medicament for the treatment of or prevention of a viral infection.
  • the disclosure relates to method of making compounds disclosed herein by mixing starting materials and reagents disclosed herein under conditions such that the compounds are formed.
  • FIG. 1 illustrates the preparation of ⁇ -D-N-hydroxycytidine.
  • a TBSCl, DMAP, DIPEA, DCM;
  • b (2,4,6-iPr)PhSO 2 Cl, DIPEA, DMAP, DCM;
  • c NH 2 OH—HCl, DIPEA, DCM;
  • F— source e. aq NH 2 OH, AcOH, 50° C.
  • FIG. 2 illustrates certain embodiments of the disclosure.
  • FIG. 3 illustrates certain embodiments of the disclosure.
  • FIG. 4 shows EIDD-01931 mean plasma concentrations and pharmacokinetic parameters from mice dosed with EIDD-01931
  • FIG. 5 shows EIDD-01931 nucleoside accumulation in mouse organs
  • FIG. 6 shows EIDD-01931 triphosphate accumulation in mouse organs
  • FIG. 7 shows reduction in footpad swelling in CHIKV challenged mice treat with EIDD-01931
  • FIG. 8 shows reduction of CHIKV RNA copies by PCR in CHIKV challenged mice treated with EIDD-01931
  • Embodiments of the present disclosure will employ, unless otherwise indicated, techniques of medicine, organic chemistry, biochemistry, molecular biology, pharmacology, and the like, which are within the skill of the art. Such techniques are explained fully in the literature.
  • a pharmaceutical agent which may be in the form of a salt or prodrug, is administered in methods disclosed herein that is specified by a weight. This refers to the weight of the recited compound. If in the form of a salt or prodrug, then the weight is the molar equivalent of the corresponding salt or prodrug.
  • Subject refers any animal, preferably a human patient, livestock, or domestic pet.
  • the terms “prevent” and “preventing” include the prevention of the recurrence, spread or onset. It is not intended that the present disclosure be limited to complete prevention. In some embodiments, the onset is delayed, or the severity of the disease is reduced.
  • the terms “treat” and “treating” are not limited to the case where the subject (e.g. patient) is cured and the disease is eradicated. Rather, embodiments, of the present disclosure also contemplate treatment that merely reduces symptoms, and/or delays disease progression.
  • the term “combination with” when used to describe administration with an additional treatment means that the agent may be administered prior to, together with, or after the additional treatment, or a combination thereof.
  • alkyl means a noncyclic straight chain or branched, unsaturated or saturated hydrocarbon such as those containing from 1 to 10 carbon atoms.
  • a “higher alkyl” refers to unsaturated or saturated hydrocarbon having 6 or more carbon atoms.
  • a “C 6 -C 16 ” refers to an alkyl containing 6 to 16 carbon atoms.
  • a “C 6 -C 22 ” refers to an alkyl containing 6 to 22 carbon atoms.
  • saturated straight chain alkyls include methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-septyl, n-octyl, n-nonyl, and the like; while saturated branched alkyls include isopropyl, sec-butyl, isobutyl, tert-butyl, isopentyl, and the like.
  • Unsaturated alkyls contain at least one double or triple bond between adjacent carbon atoms (referred to as an “alkenyl” or “alkynyl”, respectively).
  • Representative straight chain and branched alkenyls include ethylenyl, propylenyl, 1-butenyl, 2-butenyl, isobutylenyl, 1-pentenyl, 2-pentenyl, 3-methyl-1-butenyl, 2-methyl-2-butenyl, 2,3-dimethyl-2-butenyl, and the like; while representative straight chain and branched alkynyls include acetylenyl, propynyl, 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl, 3-methyl-1-butynyl, and the like.
  • Non-aromatic mono or polycyclic alkyls are referred to herein as “carbocycles” or “carbocyclyl” groups.
  • Representative saturated carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like; while unsaturated carbocycles include cyclopentenyl and cyclohexenyl, and the like.
  • Heterocarbocycles or heterocarbocyclyl groups are carbocycles which contain from 1 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur which may be saturated or unsaturated (but not aromatic), monocyclic or polycyclic, and wherein the nitrogen and sulfur heteroatoms may be optionally oxidized, and the nitrogen heteroatom may be optionally quaternized.
  • Heterocarbocycles include morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyridinyl, tetrahydroprimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, tetrahydropyrimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, and the like.
  • aryl refers to aromatic homocyclic (i.e., hydrocarbon) mono-, bi- or tricyclic ring-containing groups preferably having 6 to 12 members such as phenyl, naphthyl and biphenyl. Phenyl is a preferred aryl group.
  • substituted aryl refers to aryl groups substituted with one or more groups, preferably selected from alkyl, substituted alkyl, alkenyl (optionally substituted), aryl (optionally substituted), heterocyclo (optionally substituted), halo, hydroxy, alkoxy (optionally substituted), aryloxy (optionally substituted), alkanoyl (optionally substituted), aroyl, (optionally substituted), alkylester (optionally substituted), arylester (optionally substituted), cyano, nitro, amino, substituted amino, amido, lactam, urea, urethane, sulfonyl, and, the like, where optionally one or more pair of substituents together with the atoms to which they are bonded form a 3 to 7 member ring.
  • heteroaryl or “heteroaromatic” refers an aromatic heterocarbocycle having 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur, and containing at least 1 carbon atom, including both mono- and polycyclic ring systems.
  • Polycyclic ring systems may, but are not required to, contain one or more non-aromatic rings, as long as one of the rings is aromatic.
  • heteroaryls are furyl, benzofuranyl, thiophenyl, benzothiophenyl, pyrrolyl, indolyl, isoindolyl, azaindolyl, pyridyl, quinolinyl, isoquinolinyl, oxazolyl, isooxazolyl, benzoxazolyl, pyrazolyl, imidazolyl, benzimidazolyl, thiazolyl, benzothiazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, cinnolinyl, phthalazinyl, and quinazolinyl. It is contemplated that the use of the term “heteroaryl” includes N-alkylated derivatives such as a 1-methylimidazol-5-yl substituent.
  • heterocycle or “heterocyclyl” refers to mono- and polycyclic ring systems having 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur, and containing at least 1 carbon atom.
  • the mono- and polycyclic ring systems may be aromatic, non-aromatic or mixtures of aromatic and non-aromatic rings.
  • Heterocycle includes heterocarbocycles, heteroaryls, and the like.
  • Alkylthio refers to an alkyl group as defined above with the indicated number of carbon atoms attached through a sulfur bridge.
  • An example of an alkylthio is methylthio, (i.e., —S—CH 3 ).
  • Alkoxy refers to an alkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge.
  • alkoxy include, but are not limited to, methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, s-butoxy, t-butoxy, n-pentoxy, and s-pentoxy.
  • Preferred alkoxy groups are methoxy, ethoxy, n-propoxy, propoxy, n-butoxy, s-butoxy, t-butoxy.
  • Alkylamino refers an alkyl group as defined above with the indicated number of carbon atoms attached through an amino bridge.
  • An example of an alkylamino is methylamino, (i.e., —NH—CH 3 ).
  • Alkanoyl refers to an alkyl as defined above with the indicated number of carbon atoms attached through a carbonyl bride (i.e., —(C ⁇ O)alkyl).
  • Alkylsulfonyl refers to an alkyl as defined above with the indicated number of carbon atoms attached through a sulfonyl bridge (i.e., —S( ⁇ O) 2 alkyl) such as mesyl and the like, and “Arylsulfonyl” refers to an aryl attached through a sulfonyl bridge (i.e., —S( ⁇ O) 2 aryl).
  • Alkylsulfamoyl refers to an alkyl as defined above with the indicated number of carbon atoms attached through a sulfamoyl bridge (i.e., —NHS( ⁇ O) 2 alkyl), and an “Arylsulfamoyl” refers to an alkyl attached through a sulfamoyl bridge (i.e., —NHS( ⁇ O) 2 aryl).
  • Alkylsulfinyl refers to an alkyl as defined above with the indicated number of carbon atoms attached through a sulfinyl bridge (i.e. —S( ⁇ O)alkyl).
  • cycloalkyl and cycloalkenyl refer to mono-, bi-, or tri homocyclic ring groups of 3 to 15 carbon atoms which are, respectively, fully saturated and partially unsaturated.
  • cycloalkenyl includes bi- and tricyclic ring systems that are not aromatic as a whole, but contain aromatic portions (e.g., fluorene, tetrahydronapthalene, dihydroindene, and the like).
  • the rings of multi-ring cycloalkyl groups may be either fused, bridged and/or joined through one or more spiro unions.
  • substituted cycloalkyl and “substituted cycloalkenyl” refer, respectively, to cycloalkyl and cycloalkenyl groups substituted with one or more groups, preferably selected from aryl, substituted aryl, heterocyclo, substituted heterocyclo, carbocyclo, substituted carbocyclo, halo, hydroxy, alkoxy (optionally substituted), aryloxy (optionally substituted), alkylester (optionally substituted), arylester (optionally substituted), alkanoyl (optionally substituted), aryol (optionally substituted), cyano, nitro, amino, substituted amino, amido, lactam, urea, urethane, sulfonyl, and the like.
  • halogen and “halo” refer to fluorine, chlorine, bromine, and iodine.
  • substituted refers to a molecule wherein at least one hydrogen atom is replaced with a substituent. When substituted, one or more of the groups are “substituents.” The molecule may be multiply substituted. In the case of an oxo substituent (“ ⁇ O”), two hydrogen atoms are replaced.
  • Example substituents within this context may include halogen, hydroxy, alkyl, alkoxy, nitro, cyano, oxo, carbocyclyl, carbocycloalkyl, heterocarbocyclyl, heterocarbocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, —NRaRb, —NRaC( ⁇ O)Rb, —NRaC( ⁇ O)NRaNRb, —NRaC( ⁇ O)ORb, —NRaSO 2 Rb, —C( ⁇ O)Ra, —C( ⁇ O)ORa, —C( ⁇ O)NRaRb, —OC( ⁇ O)NRaRb, —ORa, —SRa, —SORa, —S( ⁇ O) 2 Ra, —OS( ⁇ O) 2 Ra and —S( ⁇ O) 2 ORa.
  • Ra and Rb in this context may be the same or different and independently hydrogen, halogen hydroxyl, alkyl, alkoxy, alkyl, amino, alkylamino, dialkylamino, carbocyclyl, carbocycloalkyl, heterocarbocyclyl, heterocarbocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl.
  • the disclosure relates to a compound of Formula I,
  • Q is O, —O(C ⁇ O)—, —O(C ⁇ O)Lipid, —O(C ⁇ O)V—, NH, or NR 7 ;
  • V is O, NH, NR 7 , S, CH 2 , or CHR 7 ;
  • W is CH 2 , NH, S or O
  • X is CH 2 , CHMe, CMe 2 , CHF, CF 2 , or CD 2 ;
  • Y is N or CR′′
  • Z is N or CR′′
  • each R′′ is independently selected from H, D, F, Cl, Br, I, CH 3 , CD 3 , CF 3 , alkyl, acyl, alkenyl, alkynyl, hydroxyl, formyl or SCH 3 ;
  • R 1 is hydrogen, monophosphate, diphosphate, triphosphate,
  • alkyl halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, carbanoyl, esteryl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, phosphoramidyl, or phosphate wherein R 1 is optionally substituted with one or more, the same or different, R 20 ;
  • Y 1 is O or S
  • Y 2 is OH, OR 12 , OAlkyl, or BH 3 ⁇ M + ;
  • Y 3 is OH or BH 3 ⁇ M + ;
  • R 2 is hydrogen, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, azido, or heterocyclyl, wherein R 2 is optionally substituted with one or more, the same or different, R 20 ;
  • R 3 is hydrogen, hydroxy, alkyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 3 is optionally substituted with one or more, the same or different, R 20 ;
  • R 4 is hydrogen, hydroxy, alkyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 4 is optionally substituted with one or more, the same or different, R 20 ;
  • R 5 is hydrogen, hydroxy, alkoxy, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 5 is optionally substituted with one or more, the same or different, R 20 ;
  • R 6 is hydrogen, hydroxy, alkoxy, alkyl, ethynyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 6 is optionally substituted with one or more, the same or different, R 20 ;
  • each R 7 is independently selected from absent, hydrogen, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)N-dialkyl, —(C ⁇ O)Salkyl, hydroxy, alkoxy, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein each R 7 is optionally substituted with one or more, the same or different, R 20 ;
  • R 8 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, benzyloxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 8 is optionally substituted with one or more, the same or different, R 20 ;
  • R 9 is hydrogen, methyl, ethyl, tert-butyl, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, cycloalkyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 9 is optionally substituted with one or more, the same or different, R 20 ;
  • R 10 is hydrogen, alkyl, branched alkyl, cycloalkyl, lipid methyl, ethyl, isopropyl, cyclopentyl, cyclohexyl, butyl, pentyl, hexyl, neopentyl, benzyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 10 is optionally substituted with one or more, the same or different, R 20 ;
  • R 11 is hydrogen, deuterium, alkyl, methyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 11 is optionally substituted with one or more, the same or different, R 20 ;
  • R 12 is hydrogen, alkyl, aryl, phenyl, 1-naphthyl, 2-naphthyl, aromatic, heteroaromatic, 4-substituted phenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, naphthyl, or heterocyclyl, wherein R 12 is optionally substituted with one or more, the same or different, R 20 ;
  • R 13 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 20 ;
  • R 14 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 14 is optionally substituted with one or more, the same or different, R 20 ;
  • R 20 is deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 21 ; and
  • R 21 is halogen, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, formyl, carboxy, carbamoyl, mercapto, sulfamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, N-methyl-N-ethylamino, acetylamino, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, N-methyl-N-ethylcarbamoyl, methylthio, ethylthio, methylsulfinyl, ethylsulfinyl, mesyl, ethyl sulfon
  • Lipid as used herein, is a C 6-22 alkyl, alkoxy, polyethylene glycol, or aryl substituted with an alkyl group.
  • the lipid is a fatty alcohol, fatty amine, or fatty thiol derived from essential and/or non-essential fatty acids.
  • the lipid is an unsaturated, polyunsaturated, omega unsaturated, or omega polyunsaturated fatty alcohol, fatty amine, or fatty thiol derived from essential and/or non-essential fatty acids.
  • the lipid is a fatty alcohol, fatty amine, or fatty thiol derived from essential and non-essential fatty acids that have one or more of its carbon units substituted with an oxygen, nitrogen, or sulfur.
  • the lipid is an unsaturated, polyunsaturated, omega unsaturated, or omega polyunsaturated fatty alcohol, fatty amine, or fatty thiol derived from essential and/or non-essential fatty acids that have one or more of its carbon units substituted with an oxygen, nitrogen, or sulfur.
  • the lipid is a fatty alcohol, fatty amine, or fatty thiol derived from essential and/or non-essential fatty acids that is optionally substituted.
  • the lipid is an unsaturated, polyunsaturated, omega unsaturated, or omega polyunsaturated fatty alcohol, fatty amine, or fatty thiol derived from essential and/or non-essential fatty acids that is optionally substituted.
  • the lipid is a fatty alcohol, fatty amine, or fatty thiol derived from essential and/or non-essential fatty acids that have one or more of its carbon units substituted with an oxygen, nitrogen, or sulfur that is optionally substituted.
  • the lipid is an unsaturated, polyunsaturated, omega unsaturated, or omega polyunsaturated fatty alcohol, fatty amine, or fatty thiol derived from essential and/or non-essential fatty acids that have one or more of its carbon units substituted with an oxygen, nitrogen, or sulfur that is also optionally substituted.
  • the lipid is hexadecyloxypropyl.
  • the lipid is 2-aminohexadecyloxypropyl.
  • the lipid is 2-aminoarachidyl.
  • the lipid is 2-benzyloxyhexadecyloxypropyl.
  • the lipid is lauryl, myristyl, palmityl, stearyl, arachidyl, behenyl, or lignoceryl.
  • the lipid is a sphingolipid having the formula:
  • R 8 of the sphingolipid is hydrogen, alkyl, C( ⁇ O)R 12 , C( ⁇ O)OR 12 , or C( ⁇ O)NHR 12 ;
  • R 9 of the sphingolipid is hydrogen, fluoro, OR 12 , OC( ⁇ O)R 12 , OC( ⁇ O)OR 12 , or OC( ⁇ O)NHR 12 ;
  • R 10 of the sphingolipid is a saturated or unsaturated alkyl chain of greater than 6 and less than 22 carbons optionally substituted with one or more halogen or hydroxy or a structure of the following formula:
  • n 8 to 14 or less than or equal to 8 to less than or equal to 14
  • o is 9 to 15 or less than or equal to 9 to less than or equal to 15
  • the total or m and n is 8 to 14 or less than or equal to 8 to less than or equal to 14
  • the total of m and o is 9 to 15 or less than or equal to 9 to less than or equal to 15;
  • n is 4 to 10 or less than or equal to 4 to less than or equal to 10
  • o is 5 to 11 or less than or equal to 5 to less than or equal to 11
  • the total of m and n is 4 to 10 or less than or equal to 4 to less than or equal to 10
  • the total of m and o is 5 to 11 or less than or equal to 5 to less than or equal to 11;
  • n 6 to 12 or n is less than or equal to 6 to less than or equal to 12, the total of m and n is 6 to 12 or n is less than or equal to 6 to less than or equal to 12;
  • R 11 of the sphingolipid is OR 12 , OC( ⁇ O)R 12 , OC( ⁇ O)OR 12 , or OC( ⁇ O)NHR 12 ;
  • R 12 of the sphingolipid is hydrogen, a branched or strait chain C 1-12 alkyl, C 13-22 alkyl, cycloalkyl, or aryl selected from benzyl or phenyl, wherein the aryl is optionally substituted with one or more, the same or different R 13 ;
  • R 13 of the sphingolipid is halogen, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, formyl, carboxy, carbamoyl, mercapto, sulfamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, N-methyl-N-ethylamino, acetylamino, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, N-methyl-N-ethylcarbamoyl, methylthio, ethylthio, methylsulfinyl, ethylsulfinyl, mesyl, eth
  • R 12 of the sphingolipid is H, alkyl, methyl, ethyl, propyl, n-butyl, branched alkyl, isopropyl, 2-butyl, 1-ethylpropyl, 1-propylbutyl, cycloalkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, benzyl, phenyl, monosubstituted phenyl, disubstituted phenyl, trisubstituted phenyl, or saturated or unsaturated C12-C19 long chain alkyl.
  • the sphingolipid has the formula:
  • R 8 of the sphingolipid is hydrogen, hydroxy, fluoro, OR 12 , OC( ⁇ O)R 12 , OC( ⁇ O)OR 12 , or OC( ⁇ O)NHR 12 ;
  • R 9 of the sphingolipid is hydrogen, hydroxy, fluoro, OR 12 , OC( ⁇ O)R 12 , OC( ⁇ O)OR 12 , or OC( ⁇ O)NHR 12 ;
  • R 10 of the sphingolipid is a saturated or unsaturated alkyl chain of greater than 6 and less than 22 carbons optionally substituted with one or more halogens or a structure of the following formula:
  • n 8 to 14 or less than or equal to 8 to less than or equal to 14, the total or m and n is 8 to 14 or less than or equal to 8 to less than or equal to 14;
  • R 12 of the sphingolipid is hydrogen, a branched or strait chain C 1-12 alkyl, C 13-22 alkyl, cycloalkyl, or aryl selected from benzyl or phenyl, wherein the aryl is optionally substituted with one or more, the same or different R 13 ;
  • R 13 of the sphingolipid is halogen, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, formyl, carboxy, carbamoyl, mercapto, sulfamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, N-methyl-N-ethylamino, acetylamino, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, N-methyl-N-ethylcarbamoyl, methylthio, ethylthio, methylsulfinyl, ethylsulfinyl, mesyl, eth
  • R 12 of the sphingolipid is H, alkyl, methyl, ethyl, propyl, n-butyl, branched alkyl, isopropyl, 2-butyl, 1-ethylpropyl, 1-propylbutyl, cycloalkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, benzyl, phenyl, monosubstituted phenyl, disubstituted phenyl, trisubstituted phenyl, or saturated or unsaturated C 12 -C 19 long chain alkyl.
  • Suitable sphingolipids include, but are not limited to, sphingosine, ceramide, or sphingomyelin, or 2-aminoalkyl optionally substituted with one or more substituents.
  • Suitable sphingolipids include, but are not limited to, 2-aminooctadecane-3,5-diol; (2S,3S,5S)-2-aminooctadecane-3,5-diol; (2S,3R,5S)-2-aminooctadecane-3,5-diol; 2-(methylamino)octadecane-3,5-diol; (2S,3R,5S)-2-(methylamino)octadecane-3,5-diol; 2-(dimethylamino)octadecane-3,5-diol; (2R,3S,5S)-2-(dimethylamino)octadecane-3,5-diol; 1-(pyrrolidin-2-yl)hexadecane-1,3-diol; (1S,3S)-1-((S)-pyrroli
  • Q is O
  • each R 7 is independently selected from hydrogen, —(C ⁇ O)O(C 6 -C 16 )alkyl or —(C ⁇ O)O(C 6 -C 22 )alkyl.
  • R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • R 8 is hydrogen, hydroxy, or benzyloxy.
  • R 9 is higher alkyl, (C 6 -C 16 )alkyl or (C 6 -C 22 )alkyl.
  • R 9 is tert-butyl or isobutyl.
  • W is O
  • Z is H.
  • R 1 is hydrogen, monophosphate, diphosphate, triphospate,
  • R 8 is hydrogen, hydroxy, or benzyloxy.
  • R 9 is higher alkyl, (C 6 -C 16 )alkyl or (C 6 -C 22 )alkyl.
  • R 10 is isopropyl.
  • R 11 is methyl
  • R 12 is phenyl
  • R 13 is hydrogen
  • R 14 is hydrogen
  • R 2 is hydrogen
  • R 3 is hydroxy
  • R 4 is hydrogen, hydroxy, alkyl, halogen, or fluoro.
  • R 5 is hydrogen, hydroxy, alkoxy, alkyl, methyl, ethynyl, or allenyl.
  • R 6 is hydrogen
  • each R 7 is independently selected from hydrogen, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)Salkyl, —(C ⁇ O)O(C 6 -C 16 )alkyl, —(C ⁇ O)(C 6 -C 16 ) alkyl, —(C ⁇ O)NH(C 6 -C 16 )alkyl, or —(C ⁇ O)S(C 6 -C 16 )alkyl.
  • the compound is selected from:
  • the disclosure relates to a compound of formula I having formula IA,
  • X is CH 2 , CHMe, CMe 2 , CHF, CF 2 , or CD 2 ;
  • Y is H, D, F, Cl, Br, I, CH 3 , CD 3 , CF 3 , alkyl, acyl, alkenyl, alkynyl, hydroxyl, formyl or SCH 3 ;
  • R 1 is hydrogen, monophosphate, diphosphate, triphosphate,
  • alkyl halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, carbanoyl, esteryl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, phosphoramidyl, or phosphate wherein R 1 is optionally substituted with one or more, the same or different, R 20 ;
  • Y 1 is O or S
  • Y 2 is OH, OR 12 , OAlkyl, or BH 3 ⁇ M + ;
  • Y 3 is OH or BH 3 ⁇ M + ;
  • R 4 is hydrogen, hydroxy, alkyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 4 is optionally substituted with one or more, the same or different, R 20 ;
  • R 5 is hydrogen, hydroxy, alkoxy, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 5 is optionally substituted with one or more, the same or different, R 20 ;
  • Each R 7 is independently selected from hydrogen, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)N-dialkyl, —(C ⁇ O)Salkyl, hydroxy, alkoxy, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein each R 7 is optionally substituted with one or more, the same or different, R 20 ;
  • R 8 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, benzyloxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 8 is optionally substituted with one or more, the same or different, R 20 ;
  • R 9 is hydrogen, methyl, ethyl, tert-butyl, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, cycloalkyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 9 is optionally substituted with one or more, the same or different, R 20 ;
  • R 10 is hydrogen, alkyl, branched alkyl, cycloalkyl, lipid methyl, ethyl, isopropyl, cyclopentyl, cyclohexyl, butyl, pentyl, hexyl, neopentyl, benzyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 10 is optionally substituted with one or more, the same or different, R 20 ;
  • R 11 is hydrogen, deuterium, alkyl, methyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 11 is optionally substituted with one or more, the same or different, R 20 ;
  • R 12 is hydrogen, alkyl, aryl, phenyl, 1-naphthyl, 2-naphthyl, aromatic, heteroaromatic, 4-substituted phenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, naphthyl, or heterocyclyl, wherein R 12 is optionally substituted with one or more, the same or different, R 20 ;
  • R 13 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 20 ;
  • R 14 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 14 is optionally substituted with one or more, the same or different, R 20 ;
  • R 20 is deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 21 ; and
  • R 21 is halogen, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, formyl, carboxy, carbamoyl, mercapto, sulfamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, N-methyl-N-ethylamino, acetylamino, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, N-methyl-N-ethylcarbamoyl, methylthio, ethylthio, methylsulfinyl, ethylsulfinyl, mesyl, ethyl sulfon
  • the disclosure relates to a compound of formula I has formula IB,
  • V is absent, O, NH, NR 15 , S, CH 2 , or CHR 15 ;
  • X is CH 2 , CHMe, CMe 2 , CHF, CF 2 , or CD 2 ;
  • Y is H, D, F, Cl, Br, I, CH 3 , CD 3 , CF 3 , alkyl, acyl, alkenyl, alkynyl, hydroxyl, formyl or SCH 3 ;
  • R 1 is hydrogen, monophosphate, diphosphate, triphosphate,
  • alkyl halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, carbanoyl, esteryl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, phosphoramidyl, or phosphate wherein R 1 is optionally substituted with one or more, the same or different, R 20 ;
  • Y 1 is O or S
  • Y 2 is OH, OR 12 , OAlkyl, or BH 3 ⁇ M + ;
  • Y 3 is OH or BH 3 ⁇ M + ;
  • R 4 is hydrogen, hydroxy, alkyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 4 is optionally substituted with one or more, the same or different, R 20 ;
  • R 5 is hydrogen, hydroxy, alkoxy, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 5 is optionally substituted with one or more, the same or different, R 20 ;
  • R 8 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, benzyloxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 8 is optionally substituted with one or more, the same or different, R 20 ;
  • R 9 is hydrogen, methyl, ethyl, tert-butyl, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, cycloalkyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 9 is optionally substituted with one or more, the same or different, R 20 ;
  • R 10 is hydrogen, alkyl, branched alkyl, cycloalkyl, lipid methyl, ethyl, isopropyl, cyclopentyl, cyclohexyl, butyl, pentyl, hexyl, neopentyl, benzyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 10 is optionally substituted with one or more, the same or different, R 20 ;
  • R 11 is hydrogen, deuterium, alkyl, methyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 11 is optionally substituted with one or more, the same or different, R 20 ;
  • R 12 is hydrogen, alkyl, aryl, phenyl, 1-naphthyl, 2-naphthyl, aromatic, heteroaromatic, 4-substituted phenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, naphthyl, or heterocyclyl, wherein R 12 is optionally substituted with one or more, the same or different, R 20 ;
  • R 13 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 20 ;
  • R 14 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 14 is optionally substituted with one or more, the same or different, R 20 ;
  • R 15 is hydrogen, Lipid, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)N-dialkyl, —(C ⁇ O)Salkyl, hydroxy, alkoxy, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 15 is optionally substituted with one or more, the same or different, R 20 ;
  • R 20 is deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 21 ; and
  • R 21 is halogen, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, formyl, carboxy, carbamoyl, mercapto, sulfamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, N-methyl-N-ethylamino, acetylamino, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, N-methyl-N-ethylcarbamoyl, methylthio, ethylthio, methylsulfinyl, ethylsulfinyl, mesyl, ethyl sulfon
  • the disclosure relates to a compound of formula I having formula IC,
  • X is CH 2 , CHMe, CMe 2 , CHF, CF 2 , or CD 2 ;
  • Y is H, D, F, Cl, Br, I, CH 3 , CD 3 , CF 3 , alkyl, acyl, alkenyl, alkynyl, hydroxyl, formyl or SCH 3 ;
  • R 1 is hydrogen, monophosphate, diphosphate, triphosphate,
  • alkyl halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, carbanoyl, esteryl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, phosphoramidyl, or phosphate wherein R 1 is optionally substituted with one or more, the same or different, R 20 ;
  • Y 1 is O or S
  • Y 2 is OH, OR 12 , OAlkyl, or BH 3 ⁇ M + ;
  • Y 3 is OH or BH 3 ⁇ M + ;
  • R 4 is hydrogen, hydroxy, alkyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 4 is optionally substituted with one or more, the same or different, R 20 ;
  • R 5 is hydrogen, hydroxy, alkoxy, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 5 is optionally substituted with one or more, the same or different, R 20 ;
  • R 8 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, benzyloxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 8 is optionally substituted with one or more, the same or different, R 20 ;
  • R 9 is hydrogen, methyl, ethyl, tert-butyl, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, cycloalkyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 9 is optionally substituted with one or more, the same or different, R 20 ;
  • R 10 is hydrogen, alkyl, branched alkyl, cycloalkyl, lipid methyl, ethyl, isopropyl, cyclopentyl, cyclohexyl, butyl, pentyl, hexyl, neopentyl, benzyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 10 is optionally substituted with one or more, the same or different, R 20 ;
  • R 11 is hydrogen, deuterium, alkyl, methyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 11 is optionally substituted with one or more, the same or different, R 20 ;
  • R 12 is hydrogen, alkyl, aryl, phenyl, 1-naphthyl, 2-naphthyl, aromatic, heteroaromatic, 4-substituted phenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, naphthyl, or heterocyclyl, wherein R 12 is optionally substituted with one or more, the same or different, R 20 ;
  • R 13 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 20 ;
  • R 14 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 14 is optionally substituted with one or more, the same or different, R 20 ;
  • R 15 is hydrogen, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)N-dialkyl, —(C ⁇ O)Salkyl, hydroxy, alkoxy, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 15 is optionally substituted with one or more, the same or different, R 20 ;
  • R 20 is deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 21 ; and
  • R 21 is halogen, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, formyl, carboxy, carbamoyl, mercapto, sulfamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, N-methyl-N-ethylamino, acetylamino, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, N-methyl-N-ethylcarbamoyl, methylthio, ethylthio, methylsulfinyl, ethylsulfinyl, mesyl, ethyl sulfon
  • the disclosure relates to a compound of formula I having formula ID,
  • W is CH 2 , NH, S or O
  • X is CH 2 , CHMe, CMe 2 , CHF, CF 2 , or CD 2 ;
  • Y is N or CR′′
  • Z is N or CR′′
  • each R′′ is independently selected from is H, D, F, Cl, Br, I, CH 3 , CD 3 , CF 3 , alkyl, acyl, alkenyl, alkynyl, hydroxyl, formyl or SCH 3 ;
  • R 1 is hydrogen, monophosphate, diphosphate, triphosphate,
  • alkyl halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, carbanoyl, esteryl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, phosphoramidyl, or phosphate wherein R 1 is optionally substituted with one or more, the same or different, R 20 ;
  • Y 1 is O or S
  • Y 2 is OH, OR 12 , OAlkyl, or BH 3 ⁇ M + ;
  • Y 3 is OH or BH 3 ⁇ M + ;
  • R 2 is hydrogen, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, azido, or heterocyclyl, wherein R 2 is optionally substituted with one or more, the same or different, R 20 ;
  • R 3 is hydrogen, hydroxy, alkyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 3 is optionally substituted with one or more, the same or different, R 20 ;
  • R 4 is hydrogen, hydroxy, alkyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 4 is optionally substituted with one or more, the same or different, R 20 ;
  • R 5 is hydrogen, hydroxy, alkoxy, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 5 is optionally substituted with one or more, the same or different, R 20 ;
  • R 6 is hydrogen, hydroxy, alkoxy, alkyl, ethynyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 6 is optionally substituted with one or more, the same or different, R 20 ;
  • R 8 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, benzyloxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 8 is optionally substituted with one or more, the same or different, R 20 ;
  • R 9 is hydrogen, methyl, ethyl, tert-butyl, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, cycloalkyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 9 is optionally substituted with one or more, the same or different, R 20 ;
  • R 10 is hydrogen, alkyl, branched alkyl, cycloalkyl, lipid methyl, ethyl, isopropyl, cyclopentyl, cyclohexyl, butyl, pentyl, hexyl, neopentyl, benzyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 10 is optionally substituted with one or more, the same or different, R 20 ;
  • R 11 is hydrogen, deuterium, alkyl, methyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 11 is optionally substituted with one or more, the same or different, R 20 ;
  • R 12 is hydrogen, alkyl, aryl, phenyl, 1-naphthyl, 2-naphthyl, aromatic, heteroaromatic, 4-substituted phenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, naphthyl, or heterocyclyl, wherein R 12 is optionally substituted with one or more, the same or different, R 20 ;
  • R 13 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 20 ;
  • R 14 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 14 is optionally substituted with one or more, the same or different, R 20 ;
  • R 15 is hydrogen, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)N-dialkyl, —(C ⁇ O)Salkyl, hydroxy, alkoxy, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 15 is optionally substituted with one or more, the same or different, R 20 ;
  • R 15′ is hydrogen, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)N-dialkyl, —(C ⁇ O)Salkyl, hydroxy, alkoxy, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein each R 7 is optionally substituted with one or more, the same or different, R 20 ;
  • R 15 and R 15′ can form a ring that is optionally substituted with one or more, the same or different, R 20 ;
  • R 20 is deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 21 ; and
  • R 21 is halogen, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, formyl, carboxy, carbamoyl, mercapto, sulfamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, N-methyl-N-ethylamino, acetylamino, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, N-methyl-N-ethylcarbamoyl, methylthio, ethylthio, methylsulfinyl, ethylsulfinyl, mesyl, ethyl sulfon
  • the disclosure relates to a compound of formula I having formula IE,
  • Q is O, —O(C ⁇ O)—, —O(C ⁇ O)Lipid, —O(C ⁇ O)V—, NH, or NR 7 ;
  • V is O, NH, NR 7 , S, CH 2 , or CHR 7 ;
  • W is CH 2 , NH, S or O
  • X is CH 2 , CHMe, CMe 2 , CHF, CF 2 , or CD 2 ;
  • Y is N or CR′′
  • Z is N or CR′′
  • each R′′ is independently selected from is H, D, F, Cl, Br, I, CH 3 , CD 3 , CF 3 , alkyl, acyl, alkenyl, alkynyl, hydroxyl, formyl or SCH 3 ;
  • R 2 is hydrogen, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, azido, or heterocyclyl, wherein R 2 is optionally substituted with one or more, the same or different, R 20 ;
  • R 3 is hydrogen, hydroxy, alkyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 3 is optionally substituted with one or more, the same or different, R 20 ;
  • R 4 is hydrogen, hydroxy, alkyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 4 is optionally substituted with one or more, the same or different, R 20 ;
  • R 5 is hydrogen, hydroxy, alkoxy, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 5 is optionally substituted with one or more, the same or different, R 20 ;
  • R 6 is hydrogen, hydroxy, alkoxy, alkyl, ethynyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 6 is optionally substituted with one or more, the same or different, R 20 ;
  • each R 7 is independently selected from absent, hydrogen, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)N-dialkyl, —(C ⁇ O)Salkyl, hydroxy, alkoxy, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein each R 7 is optionally substituted with one or more, the same or different, R 20 ;
  • R 15 is hydrogen, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)N-dialkyl, —(C ⁇ O)Salkyl, hydroxy, alkoxy, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 15 is optionally substituted with one or more, the same or different, R 20 ;
  • R 15′ is hydrogen, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)N-dialkyl, —(C ⁇ O)Salkyl, hydroxy, alkoxy, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein each R 7 is optionally substituted with one or more, the same or different, R 20 ;
  • R 15 and R 15′ can form a ring that is optionally substituted with one or more, the same or different, R 20 ;
  • R 7 s can together form a ring that is optionally substituted with one or more, the same or different, R 20 ;
  • R 20 is deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 21 ; and
  • R 21 is halogen, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, formyl, carboxy, carbamoyl, mercapto, sulfamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, N-methyl-N-ethylamino, acetylamino, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, N-methyl-N-ethylcarbamoyl, methylthio, ethylthio, methylsulfinyl, ethylsulfinyl, mesyl, ethyl sulfon
  • the disclosure relates to a compound of Formula II,
  • Q is O, —O(C ⁇ O)—, —O(C ⁇ O)Lipid, —O(C ⁇ O)V—, NH, or NR 7 ;
  • V is O, NH, NR 7 , S, CH 2 , or CHR 7 ;
  • W is CH 2 , NH, S or O
  • X is CH 2 or O
  • Y is N or CR′′
  • Z is N or CR′′
  • each R′′ is independently selected from is H, D, F, Cl, Br, I, CH 3 , CD 3 , CF 3 , alkyl, acyl, alkenyl, alkynyl, hydroxyl, formyl or SCH 3 ;
  • R 1 is monophosphate, diphosphate, triphosphate
  • Y 1 is O or S
  • Y 2 is OH, OR 12 , OAlkyl, or BH 3 ⁇ M + ;
  • Y 3 is OH or BH 3 ⁇ M + ;
  • R 2 is hydrogen, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, azido, or heterocyclyl, wherein R 2 is optionally substituted with one or more, the same or different, R 20 ;
  • R 3 is hydrogen, hydroxy, alkyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 3 is optionally substituted with one or more, the same or different, R 20 ;
  • R 4 is hydrogen, hydroxy, alkyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 4 is optionally substituted with one or more, the same or different, R 20 ;
  • R 5 is hydrogen, hydroxy, alkoxy, alkyl, alkenyl, alkynyl, ethynyl, fluoromethyl, difluoromethyl, trifluoromethyl, hydroxymethyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 5 is optionally substituted with one or more, the same or different, R 20 ;
  • R 6 is hydrogen, hydroxy, alkoxy, alkyl, ethynyl, allenyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 6 is optionally substituted with one or more, the same or different, R 20 ;
  • each R 7 is independently selected from absent, hydrogen, —(C ⁇ O)Oalkyl, —(C ⁇ O)alkyl, —(C ⁇ O)NHalkyl, —(C ⁇ O)N-dialkyl, —(C ⁇ O)Salkyl, hydroxy, alkoxy, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein each R 7 is optionally substituted with one or more, the same or different, R 20 ;
  • R 8 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, benzyloxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 8 is optionally substituted with one or more, the same or different, R 20 ;
  • R 9 is hydrogen, methyl, ethyl, tert-butyl, alkyl, higher alkyl, (C 6 -C 16 )alkyl, (C 6 -C 22 )alkyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, cycloalkyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 9 is optionally substituted with one or more, the same or different, R 20 ;
  • R 10 is hydrogen, alkyl, branched alkyl, cycloalkyl, lipid methyl, ethyl, isopropyl, cyclopentyl, cyclohexyl, butyl, pentyl, hexyl, neopentyl, benzyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 10 is optionally substituted with one or more, the same or different, R 20 ;
  • R 11 is hydrogen, deuterium, alkyl, methyl, halogen, nitro, cyano, hydroxy, amino, mercapto, formyl, carboxy, carbamoyl, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkylsulfonyl, arylsulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 11 is optionally substituted with one or more, the same or different, R 20 ;
  • R 12 is hydrogen, alkyl, aryl, phenyl, 1-naphthyl, 2-naphthyl, aromatic, heteroaromatic, 4-substituted phenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, naphthyl, or heterocyclyl, wherein R 2 is optionally substituted with one or more, the same or different, R 20 ;
  • R 13 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein IC is optionally substituted with one or more, the same or different, R 20 ;
  • R 14 is hydrogen, deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, lipid, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 4 is optionally substituted with one or more, the same or different, R 20 ;
  • R 7 s can together form a ring that is optionally substituted with one or more, the same or different, R 20 ;
  • R 20 is deuterium, alkyl, alkenyl, alkynyl, halogen, nitro, cyano, hydroxy, amino, amido, mercapto, formyl, carboxy, carbamoyl, azido, alkoxy, alkylthio, alkylamino, (alkyl) 2 amino, alkylsulfinyl, alkyl sulfonyl, aryl sulfonyl, carbocyclyl, aryl, or heterocyclyl, wherein R 13 is optionally substituted with one or more, the same or different, R 21 ; and
  • R 21 is halogen, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, formyl, carboxy, carbamoyl, mercapto, sulfamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, N-methyl-N-ethylamino, acetylamino, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, N-methyl-N-ethylcarbamoyl, methylthio, ethylthio, methylsulfinyl, ethylsulfinyl, mesyl, ethyl sulfon
  • any citation of higher alkyl, (C 6 -C 16 )alkyl may be substituted with a (C 6 -C 22 )alkyl.
  • any citation of higher alkyl, (C 6 -C 16 )alkyl or (C 6 -C 22 )alkyl may be substituted with polyethylene glycol or —CH 2 (CH 2 OCH 2 ) n CH 3 , wherein n is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11-20, or 30-100.
  • the disclosure relates to methods of treating or preventing a viral infection comprising administering in effective amount of a compound disclosed herein to a subject in need thereof.
  • the viral infection is, or is caused by, an alphavirus, flavivirus or coronaviruses orthomyxoviridae or paramyxoviridae, or RSV, influenza, Powassan virus or filoviridae or ebola.
  • the viral infection is, or is caused by, a virus selected from MERS coronavirus, Eastern equine encephalitis virus, Western equine encephalitis virus, Venezuelan equine encephalitis virus, Ross River virus, Barmah Forest virus, Powassan virus and Chikungunya virus.
  • a virus selected from MERS coronavirus, Eastern equine encephalitis virus, Western equine encephalitis virus, Venezuelan equine encephalitis virus, Ross River virus, Barmah Forest virus, Powassan virus and Chikungunya virus.
  • the compound is administered by inhalation through the lungs.
  • the subject is at risk of, exhibiting symptoms of, or diagnosed with influenza A virus including subtype H1N1, H3N2, H7N9, or H5N1, influenza B virus, influenza C virus, rotavirus A, rotavirus B, rotavirus C, rotavirus D, rotavirus E, human coronavirus, SARS coronavirus, MERS coronavirus, human adenovirus types (HAdV-1 to 55), human papillomavirus (HPV) Types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, and 59, parvovirus B19, molluscum contagiosum virus, JC virus (JCV), BK virus, Merkel cell polyomavirus, coxsackie A virus, norovirus, Rubella virus, lymphocytic choriomeningitis virus (LCMV), Dengue virus, chikungunya, Eastern equine encephalitis virus (EEEV), Western equine
  • influenza A virus including subtypes H1N1, H3N2, H7N9, H5N1 (low path), and H5N1 (high path) influenza B virus, influenza C virus, rotavirus A, rotavirus B, rotavirus C, rotavirus D, rotavirus E, SARS coronavirus, MERS-CoV, human adenovirus types (HAdV-1 to 55), human papillomavirus (HPV) Types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, and 59, parvovirus B19, molluscum contagiosum virus, JC virus (JCV), BK virus, Merkel cell polyomavirus, coxsackie A virus, norovirus, Rubella virus, lymphocytic choriomeningitis virus (LCMV), yellow fever virus, measles virus, mumps virus, respiratory syncytial virus, parainfluenza viruses 1 and 3, rinder
  • the subject is diagnosed with gastroenteritis, acute respiratory disease, severe acute respiratory syndrome, post-viral fatigue syndrome, viral hemorrhagic fevers, acquired immunodeficiency syndrome or hepatitis.
  • compounds and pharmaceutical compositions disclosed herein are contemplated to be administered in combination with other the antiviral agent(s) such as abacavir, acyclovir, acyclovir, adefovir, amantadine, amprenavir, ampligen, arbidol, atazanavir, atripla, boceprevir, cidofovir, combivir, daclatasvir, darunavir, dasabuvir, delavirdine, didanosine, docosanol, edoxudine, efavirenz, emtricitabine, enfuvirtide, entecavir, famciclovir, fomivirsen, fosamprenavir, foscarnet, fosfonet, ganciclovir, ibacitabine, imunovir, idoxuridine, imiquimod, indinavir, inosine, interferon type III, interferon type II,
  • compositions disclosed herein may be in the form of pharmaceutically acceptable salts, as generally described below.
  • suitable pharmaceutically acceptable organic and/or inorganic acids are hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, acetic acid and citric acid, as well as other pharmaceutically acceptable acids known per se (for which reference is made to the references referred to below).
  • the compounds of the disclosure may also form internal salts, and such compounds are within the scope of the disclosure.
  • a compound contains a hydrogen-donating heteroatom (e.g. NH)
  • salts are contemplated to covers isomers formed by transfer of said hydrogen atom to a basic group or atom within the molecule.
  • Pharmaceutically acceptable salts of the compounds include the acid addition and base salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydr
  • Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts. Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts.
  • suitable salts see Handbook of Pharmaceutical Salts: Properties, Selection, and Use by Stahl and Wermuth (Wiley-VCH, 2002), incorporated herein by reference.
  • a prodrug can include a covalently bonded carrier which releases the active parent drug when administered to a mammalian subject.
  • Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds.
  • Prodrugs include, for example, compounds wherein a hydroxyl group is bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl group.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol functional groups in the compounds.
  • prodrugs form the active metabolite by transformation of the prodrug by hydrolytic enzymes, the hydrolysis of amide, lactams, peptides, carboxylic acid esters, epoxides or the cleavage of esters of inorganic acids.
  • compositions for use in the present disclosure typically comprise an effective amount of a compound and a suitable pharmaceutical acceptable carrier.
  • the preparations may be prepared in a manner known per se, which usually involves mixing the at least one compound according to the disclosure with the one or more pharmaceutically acceptable carriers, and, if desired, in combination with other pharmaceutical active compounds, when necessary under aseptic conditions.
  • the compounds may be formulated as a pharmaceutical preparation comprising at least one compound and at least one pharmaceutically acceptable carrier, diluent or excipient and/or adjuvant, and optionally one or more further pharmaceutically active compounds.
  • the pharmaceutical preparations of the disclosure are preferably in a unit dosage form, and may be suitably packaged, for example in a box, blister, vial, bottle, sachet, ampoule or in any other suitable single-dose or multi-dose holder or container (which may be properly labeled); optionally with one or more leaflets containing product information and/or instructions for use.
  • unit dosages will contain between 1 and 1000 mg, and usually between 5 and 500 mg, of the at least one compound of the disclosure, e.g. about 10, 25, 50, 100, 200, 300 or 400 mg per unit dosage.
  • the compounds can be administered by a variety of routes including the oral, ocular, rectal, transdermal, subcutaneous, intravenous, intramuscular or intranasal routes, depending mainly on the specific preparation used.
  • the compound will generally be administered in an “effective amount”, by which is meant any amount of a compound that, upon suitable administration, is sufficient to achieve the desired therapeutic or prophylactic effect in the subject to which it is administered.
  • such an effective amount will usually be between 0.01 to 1000 mg per kilogram body weight of the patient per day, more often between 0.1 and 500 mg, such as between 1 and 250 mg, for example about 5, 10, 20, 50, 100, 150, 200 or 250 mg, per kilogram body weight of the patient per day, which may be administered as a single daily dose, divided over one or more daily doses.
  • the amount(s) to be administered, the route of administration and the further treatment regimen may be determined by the treating clinician, depending on factors such as the age, gender and general condition of the patient and the nature and severity of the disease/symptoms to be treated.
  • Formulations containing one or more compounds can be prepared in various pharmaceutical forms, such as granules, tablets, capsules, suppositories, powders, controlled release formulations, suspensions, emulsions, creams, gels, ointments, salves, lotions, or aerosols and the like.
  • these formulations are employed in solid dosage forms suitable for simple, and preferably oral, administration of precise dosages.
  • Solid dosage forms for oral administration include, but are not limited to, tablets, soft or hard gelatin or non-gelatin capsules, and caplets.
  • liquid dosage forms such as solutions, syrups, suspension, shakes, etc. can also be utilized.
  • the formulation is administered topically.
  • suitable topical formulations include, but are not limited to, lotions, ointments, creams, and gels.
  • the topical formulation is a gel.
  • the formulation is administered intranasally.
  • Formulations containing one or more of the compounds described herein may be prepared using a pharmaceutically acceptable carrier composed of materials that are considered safe and effective and may be administered to an individual without causing undesirable biological side effects or unwanted interactions.
  • the carrier is all components present in the pharmaceutical formulation other than the active ingredient or ingredients.
  • carrier includes, but is not limited to, diluents, binders, lubricants, disintegrators, fillers, pH modifying agents, preservatives, antioxidants, solubility enhancers, and coating compositions.
  • Carrier also includes all components of the coating composition which may include plasticizers, pigments, colorants, stabilizing agents, and glidants. Delayed release, extended release, and/or pulsatile release dosage formulations may be prepared as described in standard references such as “Pharmaceutical dosage form tablets”, eds. Liberman et. al. (New York, Marcel Dekker, Inc., 1989), “Remington—The science and practice of pharmacy”, 20th ed., Lippincott Williams & Wilkins, Baltimore, Md., 2000, and “Pharmaceutical dosage forms and drug delivery systems”, 6th Edition, Ansel et al., (Media, Pa.: Williams and Wilkins, 1995). These references provide information on carriers, materials, equipment and process for preparing tablets and capsules and delayed release dosage forms of tablets, capsules, and granules.
  • suitable coating materials include, but are not limited to, cellulose polymers such as cellulose acetate phthalate, hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate and hydroxypropyl methylcellulose acetate succinate; polyvinyl acetate phthalate, acrylic acid polymers and copolymers, and methacrylic resins that are commercially available under the trade name EUDRAGIT® (Roth Pharma, Westerstadt, Germany), zein, shellac, and polysaccharides.
  • cellulose polymers such as cellulose acetate phthalate, hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate and hydroxypropyl methylcellulose acetate succinate
  • polyvinyl acetate phthalate acrylic acid polymers and copolymers
  • methacrylic resins that are commercially available under the trade name EUDRAGIT® (Roth Pharma, Westerstadt, Germany), ze
  • the coating material may contain conventional carriers such as plasticizers, pigments, colorants, glidants, stabilization agents, pore formers and surfactants.
  • Optional pharmaceutically acceptable excipients present in the drug-containing tablets, beads, granules or particles include, but are not limited to, diluents, binders, lubricants, disintegrants, colorants, stabilizers, and surfactants.
  • Diluents also referred to as “fillers,” are typically necessary to increase the bulk of a solid dosage form so that a practical size is provided for compression of tablets or formation of beads and granules.
  • Suitable diluents include, but are not limited to, dicalcium phosphate dihydrate, calcium sulfate, lactose, sucrose, mannitol, sorbitol, cellulose, microcrystalline cellulose, kaolin, sodium chloride, dry starch, hydrolyzed starches, pregelatinized starch, silicone dioxide, titanium oxide, magnesium aluminum silicate and powdered sugar.
  • Binders are used to impart cohesive qualities to a solid dosage formulation, and thus ensure that a tablet or bead or granule remains intact after the formation of the dosage forms.
  • Suitable binder materials include, but are not limited to, starch, pregelatinized starch, gelatin, sugars (including sucrose, glucose, dextrose, lactose and sorbitol), polyethylene glycol, waxes, natural and synthetic gums such as acacia, tragacanth, sodium alginate, cellulose, including hydroxypropylmethylcellulose, hydroxypropylcellulose, ethylcellulose, and veegum, and synthetic polymers such as acrylic acid and methacrylic acid copolymers, methacrylic acid copolymers, methyl methacrylate copolymers, aminoalkyl methacrylate copolymers, polyacrylic acid/polymethacrylic acid and polyvinylpyrrolidone.
  • Lubricants are used to facilitate tablet manufacture.
  • suitable lubricants include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, glycerol behenate, polyethylene glycol, talc, and mineral oil.
  • Disintegrants are used to facilitate dosage form disintegration or “breakup” after administration, and generally include, but are not limited to, starch, sodium starch glycolate, sodium carboxymethyl starch, sodium carboxymethylcellulose, hydroxypropyl cellulose, pregelatinized starch, clays, cellulose, alginine, gums or cross linked polymers, such as cross-linked PVP (Polyplasdone XL from GAF Chemical Corp).
  • starch sodium starch glycolate, sodium carboxymethyl starch, sodium carboxymethylcellulose, hydroxypropyl cellulose, pregelatinized starch, clays, cellulose, alginine, gums or cross linked polymers, such as cross-linked PVP (Polyplasdone XL from GAF Chemical Corp).
  • Stabilizers are used to inhibit or retard drug decomposition reactions which include, by way of example, oxidative reactions.
  • Surfactants may be anionic, cationic, amphoteric or nonionic surface active agents.
  • Suitable anionic surfactants include, but are not limited to, those containing carboxylate, sulfonate and sulfate ions.
  • anionic surfactants include sodium, potassium, ammonium of long chain alkyl sulfonates and alkyl aryl sulfonates such as sodium dodecylbenzene sulfonate; dialkyl sodium sulfosuccinates, such as sodium dodecylbenzene sulfonate; dialkyl sodium sulfosuccinates, such as sodium bis-(2-ethylthioxyl)-sulfosuccinate; and alkyl sulfates such as sodium lauryl sulfate.
  • Cationic surfactants include, but are not limited to, quaternary ammonium compounds such as benzalkonium chloride, benzethonium chloride, cetrimonium bromide, stearyl dimethylbenzyl ammonium chloride, polyoxyethylene and coconut amine.
  • nonionic surfactants include ethylene glycol monostearate, propylene glycol myristate, glyceryl monostearate, glyceryl stearate, polyglyceryl-4-oleate, sorbitan acylate, sucrose acylate, PEG-150 laurate, PEG-400 monolaurate, polyoxyethylene monolaurate, polysorbates, polyoxyethylene octylphenylether, PEG-1000 cetyl ether, polyoxyethylene tridecyl ether, polypropylene glycol butyl ether, Poloxamer® 401, stearoyl monoisopropanolamide, and polyoxyethylene hydrogenated tallow amide.
  • amphoteric surfactants include sodium N-dodecyl-.beta.-alanine, sodium N-lauryl-.beta.-iminodipropionate, myristoamphoacetate, lauryl betaine and lauryl sulfobetaine.
  • the tablets, beads, granules, or particles may also contain minor amount of nontoxic auxiliary substances such as wetting or emulsifying agents, dyes, pH buffering agents, or preservatives.
  • the concentration of the compound to carrier and/or other substances may vary from about 0.5 to about 100 wt. % (weight percent).
  • the pharmaceutical formulation will generally contain from about 5 to about 100% by weight of the active material.
  • the pharmaceutical formulation will generally have from about 0.5 to about 50 wt. % of the active material.
  • compositions described herein can be formulation for modified or controlled release.
  • controlled release dosage forms include extended release dosage forms, delayed release dosage forms, pulsatile release dosage forms, and combinations thereof.
  • the extended release formulations are generally prepared as diffusion or osmotic systems, for example, as described in “Remington—The science and practice of pharmacy” (20th ed., Lippincott Williams & Wilkins, Baltimore, Md., 2000).
  • a diffusion system typically consists of two types of devices, a reservoir and a matrix, and is well known and described in the art.
  • the matrix devices are generally prepared by compressing the drug with a slowly dissolving polymer carrier into a tablet form.
  • the three major types of materials used in the preparation of matrix devices are insoluble plastics, hydrophilic polymers, and fatty compounds.
  • Plastic matrices include, but are not limited to, methyl acrylate-methyl methacrylate, polyvinyl chloride, and polyethylene.
  • Hydrophilic polymers include, but are not limited to, cellulosic polymers such as methyl and ethyl cellulose, hydroxyalkylcelluloses such as hydroxypropyl-cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and Carbopol® 934, polyethylene oxides and mixtures thereof.
  • Fatty compounds include, but are not limited to, various waxes such as carnauba wax and glyceryl tristearate and wax-type substances including hydrogenated castor oil or hydrogenated vegetable oil, or mixtures thereof.
  • the plastic material is a pharmaceutically acceptable acrylic polymer, including but not limited to, acrylic acid and methacrylic acid copolymers, methyl methacrylate, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, aminoalkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamine copolymer poly(methyl methacrylate), poly(methacrylic acid)(anhydride), polymethacrylate, polyacrylamide, poly(methacrylic acid anhydride), and glycidyl methacrylate copolymers.
  • acrylic acid and methacrylic acid copolymers including but not limited to, acrylic acid and methacrylic acid copolymers, methyl methacrylate, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, aminoalkyl me
  • the acrylic polymer is comprised of one or more ammonio methacrylate copolymers.
  • Ammonio methacrylate copolymers are well known in the art, and are described in NF XVII as fully polymerized copolymers of acrylic and methacrylic acid esters with a low content of quaternary ammonium groups.
  • the acrylic polymer is an acrylic resin lacquer such as that which is commercially available from Rohm Pharma under the tradename Eudragit®.
  • the acrylic polymer comprises a mixture of two acrylic resin lacquers commercially available from Rohm Pharma under the tradenames Eudragit® RL30D and Eudragit RS30D, respectively.
  • Eudragit® RL30D and Eudragit® RS30D are copolymers of acrylic and methacrylic esters with a low content of quaternary ammonium groups, the molar ratio of ammonium groups to the remaining neutral (meth)acrylic esters being 1:20 in Eudragit® RL30D and 1:40 in Eudragit® RS30D.
  • the mean molecular weight is about 150,000.
  • Edragit® S-100 and Eudragit® L-100 are also preferred.
  • the code designations RL (high permeability) and RS (low permeability) refer to the permeability properties of these agents.
  • Eudragit® RL/RS mixtures are insoluble in water and in digestive fluids. However, multiparticulate systems formed to include the same are swellable and permeable in aqueous solutions and digestive fluids.
  • the polymers described above such as Eudragit® RL/RS may be mixed together in any desired ratio in order to ultimately obtain a sustained-release formulation having a desirable dissolution profile. Desirable sustained-release multiparticulate systems may be obtained, for instance, from 100% Eudragit® RL, 50% Eudragit® RL and 50% Eudragit® RS, and 10% Eudragit® RL and 90% Eudragit® RS.
  • Desirable sustained-release multiparticulate systems may be obtained, for instance, from 100% Eudragit® RL, 50% Eudragit® RL and 50% Eudragit® RS, and 10% Eudragit® RL and 90% Eudragit® RS.
  • acrylic polymers may also be used, such as, for example, Eudragit® L.
  • extended release formulations can be prepared using osmotic systems or by applying a semi-permeable coating to the dosage form.
  • the desired drug release profile can be achieved by combining low permeable and high permeable coating materials in suitable proportion.
  • the devices with different drug release mechanisms described above can be combined in a final dosage form comprising single or multiple units.
  • multiple units include, but are not limited to, multilayer tablets and capsules containing tablets, beads, or granules.
  • An immediate release portion can be added to the extended release system by means of either applying an immediate release layer on top of the extended release core using a coating or compression process or in a multiple unit system such as a capsule containing extended and immediate release beads.
  • Extended release tablets containing hydrophilic polymers are prepared by techniques commonly known in the art such as direct compression, wet granulation, or dry granulation. Their formulations usually incorporate polymers, diluents, binders, and lubricants as well as the active pharmaceutical ingredient.
  • the usual diluents include inert powdered substances such as starches, powdered cellulose, especially crystalline and microcrystalline cellulose, sugars such as fructose, mannitol and sucrose, grain flours and similar edible powders.
  • Typical diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, inorganic salts such as sodium chloride and powdered sugar.
  • Powdered cellulose derivatives are also useful.
  • Typical tablet binders include substances such as starch, gelatin and sugars such as lactose, fructose, and glucose.
  • Natural and synthetic gums including acacia, alginates, methylcellulose, and polyvinylpyrrolidone can also be used.
  • Polyethylene glycol, hydrophilic polymers, ethylcellulose and waxes can also serve as binders.
  • a lubricant is necessary in a tablet formulation to prevent the tablet and punches from sticking in the die.
  • the lubricant is chosen from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils.
  • Extended release tablets containing wax materials are generally prepared using methods known in the art such as a direct blend method, a congealing method, and an aqueous dispersion method.
  • the congealing method the drug is mixed with a wax material and either spray—congealed or congealed and screened and processed.
  • Delayed release formulations are created by coating a solid dosage form with a polymer film, which is insoluble in the acidic environment of the stomach, and soluble in the neutral environment of the small intestine.
  • the delayed release dosage units can be prepared, for example, by coating a drug or a drug-containing composition with a selected coating material.
  • the drug-containing composition may be, e.g., a tablet for incorporation into a capsule, a tablet for use as an inner core in a “coated core” dosage form, or a plurality of drug-containing beads, particles or granules, for incorporation into either a tablet or capsule.
  • Preferred coating materials include bioerodible, gradually hydrolyzable, gradually water-soluble, and/or enzymatically degradable polymers, and may be conventional “enteric” polymers.
  • Enteric polymers become soluble in the higher pH environment of the lower gastrointestinal tract or slowly erode as the dosage form passes through the gastrointestinal tract, while enzymatically degradable polymers are degraded by bacterial enzymes present in the lower gastrointestinal tract, particularly in the colon.
  • Suitable coating materials for effecting delayed release include, but are not limited to, cellulosic polymers such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxymethyl cellulose, hydroxypropyl methyl cellulose, hydroxypropyl methyl cellulose acetate succinate, hydroxypropylmethyl cellulose phthalate, methylcellulose, ethyl cellulose, cellulose acetate, cellulose acetate phthalate, cellulose acetate trimellitate and carboxymethylcellulose sodium; acrylic acid polymers and copolymers, preferably formed from acrylic acid, methacrylic acid, methyl acrylate, ethyl acrylate, methyl methacrylate and/or ethyl methacrylate, and other methacrylic resins that are commercially available under the tradename Eudragit® (Rohm Pharma; Westerstadt, Germany), including Eudragit® L30D-55 and L100-55 (soluble at pH 5.5 and above), Eudragit® L-100 (soluble at pH
  • the preferred coating weights for particular coating materials may be readily determined by those skilled in the art by evaluating individual release profiles for tablets, beads and granules prepared with different quantities of various coating materials. It is the combination of materials, method and form of application that produce the desired release characteristics, which one can determine only from the clinical studies.
  • the coating composition may include conventional additives, such as plasticizers, pigments, colorants, stabilizing agents, glidants, etc.
  • a plasticizer is normally present to reduce the fragility of the coating, and will generally represent about 10 wt. % to 50 wt. % relative to the dry weight of the polymer.
  • typical plasticizers include polyethylene glycol, propylene glycol, triacetin, dimethyl phthalate, diethyl phthalate, dibutyl phthalate, dibutyl sebacate, triethyl citrate, tributyl citrate, triethyl acetyl citrate, castor oil and acetylated monoglycerides.
  • a stabilizing agent is preferably used to stabilize particles in the dispersion.
  • Typical stabilizing agents are nonionic emulsifiers such as sorbitan esters, polysorbates and polyvinylpyrrolidone. Glidants are recommended to reduce sticking effects during film formation and drying, and will generally represent approximately 25 wt. % to 100 wt. % of the polymer weight in the coating solution.
  • One effective glidant is talc.
  • Other glidants such as magnesium stearate and glycerol monostearates may also be used.
  • Pigments such as titanium dioxide may also be used.
  • Small quantities of an anti-foaming agent such as a silicone (e.g., simethicone), may also be added to the coating composition.
  • the formulation can provide pulsatile delivery of the one or more compounds.
  • pulsatile is meant that a plurality of drug doses are released at spaced apart intervals of time.
  • release of the initial dose is substantially immediate, i.e., the first drug release “pulse” occurs within about one hour of ingestion.
  • This initial pulse is followed by a first time interval (lag time) during which very little or no drug is released from the dosage form, after which a second dose is then released.
  • a second nearly drug release-free interval between the second and third drug release pulses may be designed.
  • the duration of the nearly drug release-free time interval will vary depending upon the dosage form design e.g., a twice daily dosing profile, a three times daily dosing profile, etc.
  • the nearly drug release-free interval has a duration of approximately 3 hours to 14 hours between the first and second dose.
  • the nearly drug release-free interval has a duration of approximately 2 hours to 8 hours between each of the three doses.
  • the pulsatile release profile is achieved with dosage forms that are closed and preferably sealed capsules housing at least two drug-containing “dosage units” wherein each dosage unit within the capsule provides a different drug release profile.
  • Control of the delayed release dosage unit(s) is accomplished by a controlled release polymer coating on the dosage unit, or by incorporation of the active agent in a controlled release polymer matrix.
  • Each dosage unit may comprise a compressed or molded tablet, wherein each tablet within the capsule provides a different drug release profile. For dosage forms mimicking a twice a day dosing profile, a first tablet releases drug substantially immediately following ingestion of the dosage form, while a second tablet releases drug approximately 3 hours to less than 14 hours following ingestion of the dosage form.
  • a first tablet releases drug substantially immediately following ingestion of the dosage form
  • a second tablet releases drug approximately 3 hours to less than 10 hours following ingestion of the dosage form
  • the third tablet releases drug at least 5 hours to approximately 18 hours following ingestion of the dosage form. It is possible that the dosage form includes more than three tablets. While the dosage form will not generally include more than a third tablet, dosage forms housing more than three tablets can be utilized.
  • each dosage unit in the capsule may comprise a plurality of drug-containing beads, granules or particles.
  • drug-containing “beads” refer to beads made with drug and one or more excipients or polymers.
  • Drug-containing beads can be produced by applying drug to an inert support, e.g., inert sugar beads coated with drug or by creating a “core” comprising both drug and one or more excipients.
  • drug-containing “granules” and “particles” comprise drug particles that may or may not include one or more additional excipients or polymers. In contrast to drug-containing beads, granules and particles do not contain an inert support.
  • Granules generally comprise drug particles and require further processing. Generally, particles are smaller than granules, and are not further processed. Although beads, granules and particles may be formulated to provide immediate release, beads and granules are generally employed to provide delayed release.
  • the compound is formulated for topical administration.
  • suitable topical dosage forms include lotions, creams, ointments, and gels.
  • a “gel” is a semisolid system containing a dispersion of the active agent, i.e., compound, in a liquid vehicle that is rendered semisolid by the action of a thickening agent or polymeric material dissolved or suspended in the liquid vehicle.
  • the liquid may include a lipophilic component, an aqueous component or both.
  • Some emulsions may be gels or otherwise include a gel component.
  • Some gels, however, are not emulsions because they do not contain a homogenized blend of immiscible components.
  • the compound described herein can be administered adjunctively with other active compounds.
  • active compounds include but are not limited to analgesics, anti-inflammatory drugs, antipyretics, antidepressants, antiepileptics, antihistamines, antimigraine drugs, antimuscarinics, anxioltyics, sedatives, hypnotics, antipsychotics, bronchodilators, anti-asthma drugs, cardiovascular drugs, corticosteroids, dopaminergics, electrolytes, gastro-intestinal drugs, muscle relaxants, nutritional agents, vitamins, parasympathomimetics, stimulants, anorectics and anti-narcoleptics.
  • Adjunctive administration means the compound can be administered in the same dosage form or in separate dosage forms with one or more other active agents.
  • compounds that can be adjunctively administered with the compounds include, but are not limited to, aceclofenac, acetaminophen, adomexetine, almotriptan, alprazolam, amantadine, amcinonide, aminocyclopropane, amitriptyline, amolodipine, amoxapine, amphetamine, aripiprazole, aspirin, atomoxetine, azasetron, azatadine, beclomethasone, benactyzine, benoxaprofen, bermoprofen, betamethasone, bicifadine, bromocriptine, budesonide, buprenorphine, bupropion, buspirone, butorphanol, butriptyline, caffeine, carbamazepine, carbidopa, carisoprodol, celecoxib, chlordiazepoxide, chlorpromazine, choline salicy
  • the additional active agent(s) can be formulated for immediate release, controlled release, or combinations thereof.
  • Example 1 The synthesis of N4-hydroxycytidine or 1-(3,4-dihydroxy-5-(hydroxymethyl) tetrahydrofuran-2-yl)-4-(hydroxyamino)pyrimidin-2-one (EIDD-01931)
  • the compound can be made in one step from cytidine by heating in a pH-adjusted solution of hydroxylamine. Despite being shorter, this route tends to give lower yields and requires purification by reverse phase flash column chromatography, limiting its use to producing smaller quantities.
  • TLC analysis was performed on silica gel, using illumination with a UV lamp (254 nm) or staining with KMnO 4 and heating.
  • Manual flash column chromatography was performed with 40-60 micron (60 ⁇ particle size) RediSep R f silica gel, purchased from Teledyne Isco, as the stationary phase.
  • Automated gradient flash column chromatography was performed on a Teledyne Isco CombiFlash Companion; normal phase separations were performed with pre-packed RediSep R f silica gel as the stationary phase, and reverse phase separations were performed with pre-packed RediSep R f C 18 High Performance Gold stationary phase.
  • Triphosphate purifications were performed using ion-exchange chromatography, with DEAE (diethylaminoethyl) Sephadex A-25 as the stationary phase, and aqueous TEAB (triethylammonium bicarbonate) as the mobile phase.
  • DEAE diethylaminoethyl
  • TEAB triethylammonium bicarbonate
  • Nominal (low resolution) liquid chromatography/mass spectrometry was performed using an Agilent 1200 series LC (UV absorption detector at 254 nm), using a Zorbax Eclipse XDB C 18 4.6 ⁇ 50 mm, 3.5 micron column, eluting with a MeOH/water mixture (typically 95/5 isocratic) and an Agilent 6120 LCMS quadrupole instrument.
  • High resolution mass spectrometry was performed by the Emory University Mass Spectrometry Center with a Thermo LTQ-FTMS using either APCI or ESI.
  • TEAB triethylammonium bicarbonate
  • the contents of the tube were transferred to a round bottom flask, and concentrated by rotary evaporation.
  • the crude material was taken up in 100 mM TEAB, and chromatography on DEAE followed by lyophilization of the product gave a triethylammonium salt of the desired product.
  • a solution of triethylammonium bis(POM)phosphate was prepared by adding triethylamine (0.362 mL, 2.60 mmol) to a solution of S20 (0.782 g, 2.398 mmol) in THF (8 mL). To a solution of EIDD-1931 (0.518 g, 1.998 mmol) in THF (32 mL) under nitrogen was added the prepared solution of triethylammonium bis(POM)phosphate at rt, then it was cooled to 0° C.
  • a 2 N hydroxylamine (30.0 mL, 60.0 mmol) aqueous solution was made by adjusting a 50% w/w aq. NH 2 OH solution with glacial AcOH and then diluting with water to achieve the desired concentration.
  • a sealable pressure vessel was charged with the above solution, L-cytidine (0.486 g, 2.0 mmol), and a stir bar. The vessel was sealed and the mixture was heated at 50° C. for 40 h. The mixture was cooled to rt and concentrated by rotary evaporation. The crude reside was dissolved in water, and automated reverse phase flash chromatography (100 g column, gradient of 100% water to 100% MeCN) gave 300 mg of semipure material as a yellow flaky solid.
  • a sealable pressure tube was charged with uridine (1.00 g, 4.09 mmol), K 2 CO 3 (0.679 g, 4.91 mmol), and deuterium oxide (8.2 mL). The mixture was purged with nitrogen for 15 minutes, the tubed was sealed, and the contents were heated with stirring at 95° C. for 16 h. The mixture was cooled to rt, the tube was unsealed, and the mixture was transferred to a round-bottom flask and concentrated by rotary evaporation. The resulting crude was coevaporated with MeOH ( ⁇ 3) to remove water. NMR analysis showed >95% deuterium incorporation at the 5-position on the nucleobase.
  • reaction mixture was diluted with dichloromethane (25 mL) and washed with 5% aqueous hydrochloric acid (25 mL) and aqueous sodium bicarbonate (25 mL). The organic layer was dried over Na 2 SO 4 and concentrated by rotary evaporation to give S40. The crude product was taken directly to the next step without further purification.
  • reaction mixture was diluted with dichloromethane (25 mL) and washed with 5% aqueous hydrochloric acid (25 mL) and aqueous sodium bicarbonate (25 mL). The organic layer was dried over Na 2 SO 4 and concentrated by rotary evaporation to give crude S41. The crude product was taken directly to the next step without further purification.
  • reaction mixture was diluted with dichloromethane (70 mL) and washed with 5% aqueous hydrochloric acid (100 mL) followed by aqueous sodium hydrogen carbonate (100 mL) and brine (100 mL). The organic layer was dried over Na 2 SO 4 , filtered, and concentrated by rotary evaporation.
  • CPE Primary Cytopathic Effect
  • test compound is prepared at four log 10 final concentrations, usually 0.1, 1.0, 10, and 100 ⁇ g/ml or ⁇ M.
  • the virus control and cell control wells are on every microplate.
  • a known active drug is tested as a positive control drug using the same method as is applied for test compounds. The positive control is tested with each test run.
  • the assay is set up by first removing growth media from the 96-well plates of cells. Then the test compound is applied in 0.1 ml volume to wells at 2 ⁇ concentration. Virus, normally at ⁇ 100 50% cell culture infectious doses (CCID 50 ) in 0.1 ml volume, is placed in those wells designated for virus infection. Medium devoid of virus is placed in toxicity control wells and cell control wells. Virus control wells are treated similarly with virus. Plates are incubated at 37° C. with 5% CO 2 until maximum CPE is observed in virus control wells. The plates are then stained with 0.011% neutral red for approximately two hours at 37° C. in a 5% CO 2 incubator.
  • CCID 50 cell culture infectious doses
  • the neutral red medium is removed by complete aspiration, and the cells may be rinsed 1 ⁇ with phosphate buffered solution (PBS) to remove residual dye.
  • PBS phosphate buffered solution
  • the PBS is completely removed and the incorporated neutral red is eluted with 50% Sorensen's citrate buffer/50% ethanol (pH 4.2) for at least 30 minutes.
  • Neutral red dye penetrates into living cells, thus, the more intense the red color, the larger the number of viable cells present in the wells.
  • the dye content in each well is quantified using a 96-well spectrophotometer at 540 nm wavelength.
  • the dye content in each set of wells is converted to a percentage of dye present in untreated control wells using a Microsoft Excel computer-based spreadsheet.
  • the 50% effective (EC 50 , virus-inhibitory) concentrations and 50% cytotoxic (CC 50 , cell-inhibitory) concentrations are then calculated by linear regression analysis.
  • the quotient of CC 50 divided by EC 50 gives the selectivity index (SI) value.
  • VYR Virus Yield Reduction
  • This assay involves similar methodology to what is described in the previous paragraphs using 96-well microplates of cells. The differences are noted in this section. Eight half-log 10 concentrations of inhibitor are tested for antiviral activity and cytotoxicity. After sufficient virus replication occurs, a sample of supernatant is taken from each infected well (three replicate wells are pooled) and held for the VYR portion of this test, if needed. Alternately, a separate plate may be prepared and the plate may be frozen for the VYR assay. After maximum CPE is observed, the viable plates are stained with neutral red dye. The incorporated dye content is quantified as described above. The data generated from this portion of the test are neutral red EC 50 , CC 50 , and SI values.
  • VYR assay Compounds observed to be active above are further evaluated by VYR assay.
  • the VYR test is a direct determination of how much the test compound inhibits virus replication. Virus that was replicated in the presence of test compound is titrated and compared to virus from untreated, infected controls. Titration of pooled viral samples (collected as described above) is performed by endpoint dilution. This is accomplished by titrating log 10 dilutions of virus using 3 or 4 microwells per dilution on fresh monolayers of cells by endpoint dilution. Wells are scored for presence or absence of virus after distinct CPE (measured by neutral red uptake) is observed.
  • the assay is set up by first removing growth media from the 12-well plates of cells, and infecting cells with 0.01 MOI of LASV strain Josiah. Cells will be incubated for 90 min: 500 ⁇ l inoculum/M12 well, at 37° C., 5% CO2 with constant gentle rocking. The inoculums will be removed and cells will be washed 2 ⁇ with medium. Then the test compound is applied in 1 ml of total volume of media. Tissue culture supernatant (TCS) will be collected at appropriate time points. TCS will then be used to determine the compounds inhibitory effect on virus replication. Virus that was replicated in the presence of test compound is titrated and compared to virus from untreated, infected controls.
  • serial ten-fold dilutions will be prepared and used to infect fresh monolayers of cells.
  • Cells will be overlaid with 1% agarose mixed 1:1 with 2 ⁇ MEM supplemented with 10% FBS and 1% penecillin, and the number of plaques determined. Plotting the log 10 of the inhibitor concentration versus log 10 of virus produced at each concentration allows calculation of the 90% (one log 10 ) effective concentration by linear regression.
  • the secondary assay involves similar methodology to what is described in the previous paragraphs using 12-well plates of cells. The differences are noted in this section. Cells are being infected as described above but this time overlaid with 1% agarose diluted 1:1 with 2 ⁇ MEM and supplemented with 2% FBS and 1% penicillin/streptomycin and supplemented with the corresponding drug concentration. Cells will be incubated at 37° C. with 5% CO2 for 6 days. The overlay is then removed and plates stained with 0.05% crystal violet in 10% buffered formalin for approximately twenty minutes at room temperature. The plates are then washed, dried and the number of plaques counted. The number of plaques is in each set of compound dilution is converted to a percentage relative to the untreated virus control. The 50% effective (EC50, virus-inhibitory) concentrations are then calculated by linear regression analysis.
  • test compound is prepared at four log 10 final concentrations, usually 0.1, 1.0, 10, and 100 ⁇ g/ml or ⁇ M.
  • the virus control and cell control will be run in parallel with each tested compound. Further, a known active drug is tested as a positive control drug using the same experimental set-up as described for the virus and cell control. The positive control is tested with each test run.
  • the assay is set up by first removing growth media from the 12-well plates of cells. Then the test compound is applied in 0.1 ml volume to wells at 2 ⁇ concentration. Virus, normally at approximately 200 plaque-forming units in 0.1 ml volume, is placed in those wells designated for virus infection. Medium devoid of virus is placed in toxicity control wells and cell control wells. Virus control wells are treated similarly with virus. Plates are incubated at 37° C. with 5% CO 2 for one hour. Virus-compound inoculums will be removed, cells washed and overlaid with 1.6% tragacanth diluted 1:1 with 2 ⁇ MEM and supplemented with 2% FBS and 1% penicillin/streptomycin and supplemented with the corresponding drug concentration.
  • Cells will be incubated at 37° C. with 5% CO 2 for 10 days.
  • the overlay is then removed and plates stained with 0.05% crystal violet in 10% buffered formalin for approximately twenty minutes at room temperature. The plates are then washed, dried and the number of plaques counted. The number of plaques is in each set of compound dilution is converted to a percentage relative to the untreated virus control.
  • the 50% effective (EC 50 , virus-inhibitory) concentrations are then calculated by linear regression analysis.
  • the secondary assay involves similar methodology to what is described in the previous paragraphs using 12-well plates of cells. The differences are noted in this section. Eight half-log 10 concentrations of inhibitor are tested for antiviral activity. One positive control drug is tested per batch of compounds evaluated.
  • cells are infected with virus. Cells are being infected as described above but this time incubated with DMEM supplemented with 2% FBS and 1% penicillin/streptomycin and supplemented with the corresponding drug concentration. Cells will be incubated for 10 days at 37° C. with 5% CO 2 , daily observed under microscope for the number of green fluorescent cells. Aliquots of supernatant from infected cells will be taken daily and the three replicate wells are pooled.
  • the pooled supernatants are then used to determine the compounds inhibitory effect on virus replication.
  • Virus that was replicated in the presence of test compound is titrated and compared to virus from untreated, infected controls.
  • serial ten-fold dilutions will be prepared and used to infect fresh monolayers of cells.
  • Cells are overlaid with tragacanth and the number of plaques determined. Plotting the log 10 of the inhibitor concentration versus log 10 of virus produced at each concentration allows calculation of the 90% (one log 10 ) effective concentration by linear regression.
  • BHK21 cells (Syrian golden hamster kidney cells, ATCC catalog # CCL-I 0), Vero cells (African green monkey kidney cells, ATCC catalog# CCL-81), or Huh-7 cells (human hepatocyte carcinoma) were passaged in DMEM supplemented with 10% FBS, 2 mM L-glutamine, 100 U/mL penicillin, and 100 ⁇ g/mL streptomycin in T-75 flasks prior to use in the antiviral assay. On the day preceding the assay, the cells were split 1:2 to assure they were in an exponential growth phase at the time of infection. Total cell and viability quantification was performed using a hemocytometer and Trypan Blue dye exclusion.
  • Cell viability was greater than 95% for the cells to be utilized in the assay.
  • the cells were resuspended at 3 ⁇ 10 3 (5 ⁇ 10 5 for Vero cells and Huh-7 cells) cells per well in tissue culture medium and added to flat bottom microtiter plates in a volume of 100 ⁇ L. The plates were incubated at 37° C./5% CO 2 overnight to allow for cell adherence. Monolayers were observed to be approximately 70% confluent.
  • Virus Preparation The Dengue virus type 2 New Guinea C strain was obtained from ATCC (catalog# VR-1584) and was grown in LLC-MK2 (Rhesus monkey kidney cells; catalog #CCL-7.1) cells for the production of stock virus pools. An aliquot of virus pretitered in BHK21 cells was removed from the freezer ( ⁇ 80° C.) and allowed to thaw slowly to room temperature in a biological safety cabinet.
  • Virus was resuspended and diluted into assay medium (DMEM supplemented with 2% heat-inactivated FBS, 2 mM L-glutamine, 100 U/mL penicillin, and 100 ⁇ g/mL streptomycin) such that the amount of virus added to each well in a volume of 100 ⁇ L was the amount determined to yield 85 to 95% cell killing at 6 days post-infection.
  • assay medium DMEM supplemented with 2% heat-inactivated FBS, 2 mM L-glutamine, 100 U/mL penicillin, and 100 ⁇ g/mL streptomycin
  • Plate Format Each plate contains cell control wells (cells only), virus control wells (cells plus virus), triplicate drug toxicity wells per compound (cells plus drug only), as well as triplicate experimental wells (drug plus cells plus virus).
  • XTT-tetrazolium was metabolized by the mitochondrial enzymes of metabolically active cells to a soluble formazan product, allowing rapid quantitative analysis of the inhibition of virus-induced cell killing by antiviral test substances.
  • XTT solution was prepared daily as a stock of 1 mg/mL in RPMI 1640.
  • Phenazine methosulfate (PMS) solution was prepared at 0.15 mg/mL in PBS and stored in the dark at ⁇ 20° C.
  • XTT/PMS stock was prepared immediately before use by adding 40 ⁇ L of PMS per ml of XTT solution. Fifty microliters of XTT/PMS was added to each well of the plate and the plate was reincubated for 4 hours at 37° C. Plates were sealed with adhesive plate sealers and shaken gently or inverted several times to mix the soluble formazan product and the plate was read spectrophotometrically at 450/650 nm with a Molecular Devices Vmax plate reader.
  • Cell Preparation-HEp2 cells (human epithelial cells, A TCC catalog# CCL-23) were passaged in DMEM supplemented with 10% FBS, 2 mM L-glutamine, 100 U/mL penicillin, 100 ⁇ g/mL streptomycin 1 mM sodium pyruvate, and 0.1 mM NEAA, T-75 flasks prior to use in the antiviral assay. On the day preceding the assay, the cells were split 1:2 to assure they were in an exponential growth phase at the time of infection. Total cell and viability quantification was performed using a hemocytometer and Trypan Blue dye exclusion. Cell viability was greater than 95% for the cells to be utilized in the assay.
  • the cells were resuspended at 1 ⁇ 10 4 cells per well in tissue culture medium and added to flat bottom microtiter plates in a volume of 100 ⁇ L. The plates were incubated at 37° C./5% CO 2 overnight to allow for cell adherence.
  • Virus Preparation The RSV strain Long and RSV strain 9320 were obtained from ATCC (catalog# VR-26 and catalog #VR-955, respectively) and were grown in HEp2 cells for the production of stock virus pools. A pretitered aliquot of virus was removed from the freezer ( ⁇ 80° C.) and allowed to thaw slowly to room temperature in a biological safety cabinet.
  • Virus was resuspended and diluted into assay medium (DMEMsupplemented with 2% heat-inactivated FBS, 2 mM L-glutamine, 100 U/mL penicillin, 100 ⁇ g/mL streptomycin, 1 mM sodium pyruvate, and 0.1 mM NEAA) such that the amount of virus added to each well in a volume of 100 ⁇ L was the amount determined to yield 85 to 95% cell killing at 6 days post-infection. Efficacy and Toxicity XTT-Plates were stained and analyzed as previously described for the Dengue cytoprotection assay.
  • assay medium DMEMsupplemented with 2% heat-inactivated FBS, 2 mM L-glutamine, 100 U/mL penicillin, 100 ⁇ g/mL streptomycin, 1 mM sodium pyruvate, and 0.1 mM NEAA
  • Cell Preparation-MOCK cells (canine kidney cells, ATCC catalog# CCL-34) were passaged in DMEM supplemented with 10% FBS, 2 mM L-glutamine, 100 U/mL penicillin, 100 ⁇ g/mL streptomycin 1 mM sodium pyruvate, and 0.1 mM NEAA, T-75 flasks prior to use in the antiviral assay. On the day preceding the assay, the cells were split 1:2 to assure they were in an exponential growth phase at the time of infection. Total cell and viability quantification was performed using a hemocytometer and Trypan Blue dye exclusion. Cell viability was greater than 95% for the cells to be utilized in the assay.
  • the cells were resuspended at 1 ⁇ 10 4 cells per well in tissue culture medium and added to flat bottom microtiter plates in a volume of 100 ⁇ L. The plates were incubated at 37° C./5% CO 2 overnight to allow for cell adherence.
  • influenza A/PR/8/34 (A TCC #VR-95), A/CA/201709 (CDC),A/NY/18/09 (CDC) and A/NWS/33 (ATCC #VR-219) strains were obtained from ATCC or from the Center of Disease Control and were grown in MDCK cells for the production of stock virus pools. A pretitered aliquot of virus was removed from the freezer ( ⁇ 80° C.) and allowed to thaw slowly to room temperature in a biological safety cabinet.
  • Virus was resuspended and diluted into assay medium (DMEM supplemented with 0.5% BSA, 2 mM L-glutamine, 100 U/mL penicillin, 100 ⁇ g/mL streptomycin, 1 mM sodium pyruvate, 0.1 mM NEAA, and 1 ⁇ g/ml TPCK-treated trypsin) such that the amount of virus added to each well in a volume of 100 ⁇ L was the amount determined to yield 85 to 95% cell killing at 4 days post-infection. Efficacy and Toxicity XTT-Plates were stained and analyzed as previously described for the Dengue cytoprotection assay.
  • assay medium DMEM supplemented with 0.5% BSA, 2 mM L-glutamine, 100 U/mL penicillin, 100 ⁇ g/mL streptomycin, 1 mM sodium pyruvate, 0.1 mM NEAA, and 1 ⁇ g/ml TPCK-treated trypsin
  • the reporter cell line Huh-luc/neo-ET was obtained from Dr. Ralf Bartenschlager (Department of Molecular Virology, Hygiene Institute, University of Heidelberg, Germany) by ImQuest BioSciences through a specific licensing agreement.
  • This cell line harbors the persistently replicating I 389 luc-ubi-neo/NS3-3′/ET replicon containing the firefly luciferase gene-ubiquitin-neomycin phosphotransferase fusion protein and EMCV IRES driven NS3-5B HCV coding sequences containing the ET tissue culture adaptive mutations (E1202G, T12081, and K1846T).
  • a stock culture of the Huh-luc/neo-ET was expanded by culture in DMEM supplemented with I 0% FCS, 2 mM glutamine, penicillin (100 ⁇ U/mL)/streptomycin (100 ⁇ g/mL) and I X nonessential amino acids plus 1 mg/mL G418.
  • the cells were split 1:4 and cultured for two passages in the same media plus 250 ⁇ g/mL G418.
  • the cells were treated with trypsin and enumerated by staining with trypan blue and seeded into 96-well tissue culture plates at a cell culture density 7.5 ⁇ 10 3 cells per well and incubated at 37° C. 5% CO 2 for 24 hours.
  • the cell culture monolayers from treated cells were stained with the tetrazolium dye XTT to evaluate the cellular viability of the Huh-luc/neo-ET reporter cell line in the presence of the compounds.
  • Virus Replication-HCV replication from the replicon assay system was measured by luciferase activity using the britelite plus luminescence reporter gene kit according to the manufacturer's instructions (Perkin Elmer, Shelton, Conn.). Briefly, one vial of britelite plus lyophilized substrate was solubilized in 10 mL of britelite reconstitution buffer and mixed gently by inversion. After a 5 minute incubation at room temperature, the britelite plus reagent was added to the 96 well plates at 100 ⁇ L per well. The plates were sealed with adhesive film and incubated at room temperature for approximately 10 minutes to lyse the cells.
  • the well contents were transferred to a white 96-well plate and luminescence was measured within 15 minutes using the Wallac 1450Microbeta Trilux liquid scintillation counter.
  • the data were imported into a customized Microsoft Excel 2007 spreadsheet for determination of the 50% virus inhibition concentration (EC 50 ).
  • Cell Preparation-HEp2 cells (human epithelial cells, ATCC catalog# CCL-23) were passaged in DMEM supplemented with 10% FBS, 2 mM L-glutamine, 100 U/mL penicillin, 100 ⁇ g/mL streptomycin 1 mM sodium pyruvate, and 0.1 mM NEAA, T-75 flasks prior to use in the antiviral assay. On the day preceding the assay, the cells were split 1:2 to assure they were in an exponential growth phase at the time of infection. Total cell and viability quantification was performed using a hemocytometer and Trypan Blue dye exclusion. Cell viability was greater than 95% for the cells to be utilized in the assay.
  • the cells were resuspended at 1 ⁇ 10 4 cells per well in tissue culture medium and added to flat bottom microtiter plates in a volume of 100 ⁇ L. The plates were incubated at 37° C./5% CO 2 overnight to allow for cell adherence.
  • Virus Preparation The Parainfluenza virus type 3 SF4 strain was obtained from ATCC (catalog# VR-281) and was grown in HEp2 cells for the production of stock virus pools. A pretitered aliquot of virus was removed from the freezer ( ⁇ 80° C.) and allowed to thaw slowly to room temperature in a biological safety cabinet. Virus was resuspended and diluted into assay medium (DMEM supplemented with 2% heat-inactivated FBS, 2 mM L-glutamine, 100 U/mL penicillin, and 100 ⁇ g/mL streptomycin) such that the amount of virus added to each well in a volume of 100 ⁇ L was the amount determined to yield 85 to 95% cell killing at 6 days post-infection.
  • assay medium DMEM supplemented with 2% heat-inactivated FBS, 2 mM L-glutamine, 100 U/mL penicillin, and 100 ⁇ g/mL streptomycin
  • Each plate contains cell control wells (cells only), virus control wells (cells plus virus), triplicate drug toxicity wells per compound (cells plus drug only), as well a triplicate experimental wells (drug plus cells plus virus).
  • XTT-tetrazolium was metabolized by the mitochondrial enzymes of metabolically active cells to a soluble formazan product, allowing rapid quantitative analysis of the inhibition of virus-induced cell killing by antiviral test substances.
  • XTT solution was prepared daily as a stock of 1 mg/mL in RPMI1640.
  • Phenazine methosulfate (PMS) solution was prepared at 0.15 mg/mL in PBS and stored in the dark at ⁇ 20° C.
  • XTT/PMS stock was prepared immediately before use by adding 40 ⁇ L of PMS per ml of XTT solution. Fifty microliters of XTT/PMS was added to each well of the plate and the plate was reincubated for 4 hours at 3 7° C. Plates were sealed with adhesive plate sealers and shaken gently or inverted several times to mix the soluble fomlazan product and the plate was read spectrophotometrically at 450/650 nm with a Molecular Devices Vmax plate reader.
  • Virus Preparation Purified influenza virus A/PR/8/34 (1 ml) was obtained from Advanced Biotechnologies, Inc. (Columbia, Md.), thawed and dispensed into five aliquots for storage at ⁇ 80° C. until use. On the day of assay set up, 20 ⁇ L of 2.5% Triton N-101 was added to 180 ⁇ L of purified virus. The disrupted virus was diluted 1:2 in a solution containing 0.25% Triton and PBS. Disruption provided the source of influenza ribonucleoprotein (RNP) containing the influenza RNA-dependent RNA polymerase and template RNA. Samples were stored on ice until use in the assay.
  • RNP influenza ribonucleoprotein
  • each 50 ⁇ L polymerase reaction contained the following: 5 ⁇ L of the disrupted RNP, 100 mM Tris-HCl (pH 8.0), 100 mM KCl, 5 mM MgCl 2 . 1 mM dithiothreitol, 0.25% Triton N-101, 5 ⁇ Ci of [ ⁇ - 32 P] GTP, 100 ⁇ M ATP, 50 ⁇ M each (CTP, UTP), 1 ⁇ M GTP, and 200 ⁇ M adenyl (3′-5′) guanosine.
  • the reactions contained the inhibitor and the same was done for reactions containing the positive control (2′-Deoxy-2′-fluoroguanosine-5′-triphosphate).
  • Other controls included RNP+reaction mixture, and RNP+I % DMSO.
  • the reaction mixture without the ApG primer and NTPs was incubated at 30° C. for 20 minutes. Once the ApG and NTPs were added to the reaction mixture, the samples were incubated at 30° C. for 1 hour then immediately followed by the transfer of the reaction onto glass-fiber filter plates and subsequent precipitation with 10% trichloroacetic acid (TCA). The plate was then washed five times with 5% TCA followed by one wash with 95% ethanol. Once the filter had dried, incorporation of [ ⁇ - 32 P] GTP was measured using a liquid scintillation counter (Micro beta).
  • Each test plate contained triplicate samples of the three compounds (6 concentrations) in addition to triplicate samples of RNP+reaction mixture (RNP alone), RNP+1% DMSO, and reaction mixture alone (no RNP).
  • the “percent inhibition values” were obtained by dividing the mean value of each test compound by the RNP+1% DMSO control. The mean obtained at each concentration of 2DFGTP was compared to the RNP+reaction control. The data was then imported into Microsoft Excel spreadsheet to calculate the IC 50 values by linear regression analysis.
  • HCV NS5B polymerase assays were performed in 20 ⁇ L volumes in 96 well reaction plates.
  • Each reaction contained 40 ⁇ g/ ⁇ L purified recombinant NS5B ⁇ 22 genotype-1b polymerase, 20 ⁇ g/ ⁇ L of HCV genotype-1b complimentary IRES template, 1 ⁇ M of each of the four natural ribonucleotides, 1 U/mL Optizyme RNAse inhibitor (Promega, Madison, Wis.), 1 mM MgCl 2 , 0.75 mM MnCl 2 , and 2 mM dithiothreitol (DTT) in 50 mM HEPES buffer (pH 7.5). Reaction mixtures were assembled on ice in two steps.
  • Step 1 consisted of combining all reaction components except the natural nucleotides and labeled UTP in a polymerase reaction mixture.
  • Ten microliters (10 ⁇ L) of the polymerase mixture was dispensed into individual wells of the 96 well reaction plate on ice.
  • Polymerase reaction mixtures without NS5B polymerase were included as no enzyme controls.
  • Serial half-logarithmic dilutions of test and control compounds, 2′-O-Methyl-CTP and 2′-O-Methyl-GTP (Trilink, San Diego, Calif.) were prepared in water and 5 ⁇ L of the serial diluted compounds or water alone (no compound control) were added to the wells containing the polymerase mixture.
  • RNA products were applied to a Hybond-N+ membrane (GE Healthcare, Piscataway, N.J) under vacuum pressure using a dot blot apparatus.
  • the membrane was removed from the dot blot apparatus and washed four times with 4 ⁇ SSC (0.6 M NaCl, and 60 mM sodium citrate), and then rinsed one time with water and once with 100% ethanol.
  • the membrane was air dried and exposed to a phosphoimaging screen and the image captured using a Typhoon 8600 Phospho imager. Following capture of the image, the membrane was placed into a Micro beta cassette along with scintillation fluid and the CPM in each reaction was counted on a Micro beta 1450. CPM data were imported into a custom Excel spreadsheet for determination of compound IC 50 s.
  • the human DNA polymerase alpha (catalog#1075), beta (catalog#1077), and gamma (catalog#1076) were purchased from CHIMERx (Madison, Wis.). Inhibition of beta and gamma DNA polymerase activity was assayed in microtiter plates in a 50 uL reaction mixture containing 50 mM Tris-HCl (pH 8.7), KCl (10 mM for beta and 100 mM for gamma), 10 mM MgCl 2 , 0.4 mg/mL BSA, 1 mM DTT, 15% glycerol, 0.05 mM of dCTP, dTTP, and dATP, 10 uCi [ 32 P]-alpha-dGTP (800 Ci/mmol), 20 ug activated calf thymus DNA and the test compound at indicated concentrations.
  • Tris-HCl pH 8.7
  • KCl 10 mM for beta and 100 mM
  • the alpha DNA polymerase reaction mixture was as follows in a 50 uL volume per sample: 20 mM Tris-HCl (pH 8), 5 mM magnesium acetate, 0.3 mg/mL BSA, 1 mM DTT, 0.1 mM spermine, 0.05 mM of dCTP, dTTP, and dATP, 10 uCi [ 32 P]-alpha-dGTP (800 Ci/mmol), 20 ug activated calf thymus DNA and the test compound at the indicated concentrations.
  • the enzyme reactions were allowed to proceed for 30 minutes at 37° C. followed by the transfer onto glass-fiber filter plates and subsequent precipitation with 10% trichloroacetic acid (TCA). The plate was then washed with 5% TCA followed by one wash with 95% ethanol. Once the filter had dried, incorporation of radioactivity was measured using a liquid scintillation counter (Microbeta).
  • PBMCs peripheral blood mononuclear cells
  • cell number was determined by Trypan Blue dye exclusion and cells were re-suspended at 1 ⁇ 10 1 ⁇ 6 cells/mL in RPMI 1640 with 15% Fetal Bovine Serum (FBS), 2 mmol/L L-glutamine, 2 ug/mL PHA-P, 100 U/mL penicillin and 100 ug/mL streptomycin and allowed to incubate for 48-72 hours at 37° C.
  • FBS Fetal Bovine Serum
  • PBMCs were centrifuged and resuspended in tissue culture medium. The cultures were maintained until use by half-volume culture changes with fresh IL-2 containing tissue culture medium every 3 days. Assays were initiated with PBMCs at 72 hours post PHA-P stimulation.
  • PBMCs employed in the assay were a mixture of cells derived from 3 donors. Immediately prior to use, target cells were resuspended in fresh tissue culture medium at 1 ⁇ 10 1 ⁇ 6 cells/mL and plated in the interior wells of a 96-well round bottom microtiter plate at 50 uL/well. Then, 100 uL of 2 ⁇ concentrations of compound-containing medium was transferred to the 96-well plate containing cells in 50 uL of the medium. AZT was employed as an internal assay standard.
  • PBMCs were exposed in triplicate to virus and cultured in the presence or absence of the test material at varying concentrations as described above in the 96-well microtiter plates. After 7 days in culture, HIV-1 replication was quantified in the tissue culture supernatant by measurement of reverse transcriptase (RT) activity. Wells with cells and virus only served as virus controls. Separate plates were identically prepared without virus for drug cytotoxicity studies.
  • Reverse Transcriptase Activity Assay Reverse transcriptase activity was measured in cell-free supernatants using a standard radioactive incorporation polymerization assay. Tritiated thymidine triphosphate (TTP; New England Nuclear) was purchased at 1 Ci/mL and 1 uL was used per enzyme reaction. A rAdT stock solution was prepared by mixing 0.5 mg/mL poly rAand 1.7 U/mL oligo dT in distilled water and was stored at ⁇ 20° C.
  • the RT reaction buffer was prepared fresh daily and consists of 125 uL of 1 mol/L EGTA, 125 uL of dH 2 O, 125 uL of 20% Triton X-100, 50 uL of 1 mol/L Tris (pH 7.4), 50 uL of 1 mol/L DTT, and 40 uL of 1 mol/L MgCl 2 .
  • 1 uL of TTP, 4 uL of dH 2 O, 2.5 uL of rAdT, and 2.5 uL of reaction buffer were mixed.
  • Ten microliters of this reaction mixture was placed in a round bottom microtiter plate and 15 uL of virus-containing supernatant was added and mixed.
  • the plate was incubated at 37° C. in a humidified incubator for 90 minutes. Following incubation, 10 uL of the reaction volume was spotted onto a DEAE filter mat in the appropriate plate format, washed 5 times (5 minutes each) in a 5% sodium phosphate buffer, 2 times (1 minute each) in distilled water, 2 times (1 minute each) in 70% ethanol, and then air dried. The dried filtermat was placed in a plastic sleeve and 4 mL of Opti-Fluor 0 was added to the sleeve. Incorporated radioactivity was quantified utilizing a Wallac 1450 Microbeta Trilux liquid scintillation counter.
  • HepG2.2.15 cells 100 ⁇ L in RPMI1640 medium with 10% fetal bovine serum was added to all wells of a 96-well plate at a density of 1 ⁇ 10 4 cells per well and the plate was incubated at 37° C. in an environment of 5% CO 2 for 24 hours. Following incubation, six ten-fold serial dilutions of test compound prepared in RPMI1640 medium with 10% fetal bovine serum were added to individual wells of the plate in triplicate. Six wells in the plate received medium alone as a virus only control. The plate was incubated for 6 days at 37° C. in an environment of 5% CO 2 . The culture medium was changed on day 3 with medium containing the indicated concentration of each compound. One hundred microliters of supernatant was collected from each well for analysis of viral DNA by qPCR and cytotoxicity was evaluated by XTT staining of the cell culture monolayer on the sixth day.
  • qPCR dilution buffer 40 ⁇ g/mL sheared salmon sperm DNA
  • SDS 2.4 software Ten microliters of cell culture supernatant collected on the sixth day was diluted in qPCR dilution buffer (40 ⁇ g/mL sheared salmon sperm DNA) and boiled for 15 minutes. Quantitative real time PCR was performed in 386 well plates using an Applied Biosystems 7900HT Sequence Detection System and the supporting SDS 2.4 software.
  • HBV-AD38-qF1 (5′-CCG TCT GTG CCT TCT CAT CTG-3′)
  • HBV-AD38-qR1 5′-AGT CCA AGA GTY CTC TTA TRY AAG ACC TT-3′
  • HBV-AD38-qP1 5′-FAM CCG TGT GCA/ZEN/CTT CGC TTC ACC TCT GC-3′BHQ1) at a final concentration of 0.2 ⁇ M for each primer in a total reaction volume of 15 ⁇ L.
  • the HBV DNA copy number in each sample was interpolated from the standard curve by the SDS.24 software and the data were imported into an Excel spreadsheet for analysis.
  • the 50% cytotoxic concentration for the test materials are derived by measuring the reduction of the tetrazolium dye XTT in the treated tissue culture plates.
  • XTT is metabolized by the mitochondrial enzyme NADPH oxidase to a soluble formazan product in metabolically active cells.
  • XTT solution was prepared daily as a stock of 1 mg/mL in PBS.
  • Phenazine methosulfate (PMS) stock solution was prepared at 0.15 mg/mL in PBS and stored in the dark at ⁇ 20° C.
  • XTT/PMS solution was prepared immediately before use by adding 40 ⁇ L of PMS per 1 mL of XTT solution.
  • XTT/PMS Fifty microliters of XTT/PMS was added to each well of the plate and the plate incubated for 2-4 hours at 37° C. The 2-4 hour incubation has been empirically determined to be within linear response range for XTT dye reduction with the indicated numbers of cells for each assay.
  • Adhesive plate sealers were used in place of the lids, the sealed plate was inverted several times to mix the soluble formazan product and the plate was read at 450 nm (650 nm reference wavelength) with a Molecular Devices SpectraMax Plus 384 spectrophotometer. Data were collected by Softmax 4.6 software and imported into an Excel spreadsheet for analysis.
  • RNA polymerase assay was performed at 30° C. using 100 ⁇ l reaction mix in 1.5 ml tube. Final reaction conditions were 50 mM Hepes (pH 7.0), 2 mM DTT, 1 mM MnCl 2 , 10 mM KCl, 100 nM UTR-Poly A (self-annealing primer), 10 ⁇ M UTP, 26 nM RdRp enzyme. The reaction mix with different compounds (inhibitors) was incubated at 30° C. for 1 hour. To assess amount of pyrophosphate generated during polymerase reaction, 30 ⁇ l of polymerase reaction mix was mixed with a luciferase coupled-enzyme reaction mix (70 ⁇ l).
  • Huh-7 cells were seeded at 0.5 ⁇ 10 ⁇ 6 cells/well in 1 mL of complete media in 12 well tissue culture treated plates. The cells were allowed to adhere overnight at 37°/5% CO 2 .
  • a 40 ⁇ M stock solution of test article was prepared in 100% DMSO. From the 40 ⁇ M stock solution, a 20 ⁇ M solution of test article in 25 ml of complete DMEM media was prepared. For compound treatment, the media was aspirated from the wells and 1 mL of the 20 ⁇ M solution was added in complete DMEM media to the appropriate wells. A separate plate of cells with “no” addition of the compound was also prepared. The plates were incubated at 37°/5% CO 2 for the following time points: 1, 3, 6 and 24 hours.
  • the cells were washed 2 ⁇ with 1 mL of DPBS.
  • the cells were extracted by adding 500 ⁇ l of 70% methanol/30% water spiked with the internal standard to each well treated with test article.
  • the non-treated blank plate was extracted with 500 ul of 70% methanol/30% water per well. Samples were centrifuged at 16,000 rpm for 10 minutes at 4° C. Samples were analyzed by LC-MS/MS using an ABSCIEX 5500 QTRAP LC-MS/MS system with a Hypercarb (PGC) column.
  • PPC Hypercarb
  • mice (6-8 weeks old, female) were acclimated for ⁇ 2 days after receipt. Mice were weighed the day before dosing to calculate dosing volumes. Mice were dosed by oral gavage with drug at 30 mg/kg, 100 mg/kg & 300 mg/kg. The mice were sampled at 8 time points: 0.5, 1, 2, 3, 4, 8 and 24 hrs (3 mice per time point for test drug). The mice were euthanized and their organs were collected (see below). In order to collected blood, mice with euthanized by CO 2 at the appropriate time point listed above. Blood was obtained by cardiac puncture (0.3 ml) at each time point. Following blood collection, the organs were removed from the mice (see below).
  • the blood was processed by inverting Li-Heparin tube with blood gently 2 or 3 times to mix well. The tubes were then placed in a rack in ice water until able to centrifuge ( ⁇ 1 hour). As soon as practical, the blood was centrifuged at ⁇ 2000 ⁇ g for 10 min in a refrigerated centrifuge to obtain plasma. Then, using a 200 ⁇ L pipette, the plasma was transferred to a labeled 1.5 ml Eppendorf tube in ice water. The plasma was then frozen in freezer or on dry ice. The samples were stored at ⁇ 80° C. prior to analysis. Organs were collected from euthanized mice.
  • the organs (lungs, liver, kidney, spleen and heart) were removed, placed in a tube, and immediately frozen in liquid nitrogen. The tubes were then transferred to dry ice. The samples were saved in cryogenic tissue vials. Samples were analyzed by LC-MS/MS using an ABSCIEX 5500 QTRAP LC-MS/MS system with a Hypercarb (PGC) column.
  • PPC Hypercarb
  • C57BL-6J mice were injected with 100 pfus CHIK virus in the footpad.
  • the test groups consisted of an unifected and untreated group, an infected and untreated group, an infected group receiving a high dose of 35 mg/kg i.p. of EIDD-01931, and an infected group receiving a low dose of 25 mg/kg i.p. of EIDD-01931.
  • the two test groups receiving EIDD-01931 received compound 12 hours before challenge and then daily for 7 days. Footpads were evaluated for inflammation (paw thickness) daily for 7 days. CHIK virus induced arthritis (histology) was assessed in ankle joints using PCR after 7 days.
  • EIDD-01931 is orally available and dose-proportional with a calculated bioavailability (% F) of 28%.
  • Organ samples (brain, lung, spleen, kidney and liver) were collected at 2.5 and 24 hours post-dose from the 50 mg/kg dose group.
  • EIDD-01931 was well distributed into all tissues tested; of particular note, it was readily distributed into brain tissue at therapeutic levels of drug, based on estimates from cellular data. Once in the brain, EIDD-01931 was rapidly metabolized to its active 5′-triphosphate form to give brain levels of 526 and 135 ng/g at 2.5 and 24 hours, respectively. Even after 24 hours levels of EIDD-01931 and its 5′-triphosphate in the brain are considerable, suggesting that once-daily oral dosing may be adequate for treatment.
  • EIDD-01931 has an acceptable toxicology profile after 6 day q.d. intraperitoneal (IP) injections in mice, with the NOEL (NO Effect Level) to be 33 mg/kg; weight loss was observed at the highest dose tested (100 mg/kg), which reversed on cessation of dosing.
  • IP intraperitoneal
  • EIDD-01931 Several derivatives of EIDD-01931 have shown antiviral activity in screening against various viruses. Activity data is shown in the tables below.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Epidemiology (AREA)
  • Pulmonology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Otolaryngology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Saccharide Compounds (AREA)
  • Medicinal Preparation (AREA)
US15/537,087 2014-12-26 2015-12-16 N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto Abandoned US20190022116A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/537,087 US20190022116A1 (en) 2014-12-26 2015-12-16 N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201462096915P 2014-12-26 2014-12-26
US201562201140P 2015-08-05 2015-08-05
US15/537,087 US20190022116A1 (en) 2014-12-26 2015-12-16 N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto
PCT/US2015/066144 WO2016106050A1 (fr) 2014-12-26 2015-12-16 N4-hydroxycytidine, ses dérivés et utilisations anti-virales

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/066144 A-371-Of-International WO2016106050A1 (fr) 2014-12-26 2015-12-16 N4-hydroxycytidine, ses dérivés et utilisations anti-virales

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/921,359 Continuation US11628181B2 (en) 2014-12-26 2020-07-06 N4-hydroxycytidine and derivatives and anti-viral uses related thereto

Publications (1)

Publication Number Publication Date
US20190022116A1 true US20190022116A1 (en) 2019-01-24

Family

ID=56151433

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/537,087 Abandoned US20190022116A1 (en) 2014-12-26 2015-12-16 N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto
US16/921,359 Active US11628181B2 (en) 2014-12-26 2020-07-06 N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US18/184,288 Pending US20230293566A1 (en) 2014-12-26 2023-03-15 N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/921,359 Active US11628181B2 (en) 2014-12-26 2020-07-06 N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US18/184,288 Pending US20230293566A1 (en) 2014-12-26 2023-03-15 N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto

Country Status (22)

Country Link
US (3) US20190022116A1 (fr)
EP (2) EP3236972B1 (fr)
JP (3) JP7381190B2 (fr)
KR (2) KR20170123308A (fr)
CN (1) CN107427529A (fr)
AU (3) AU2015370004B2 (fr)
BR (1) BR112017013858A2 (fr)
CA (1) CA2972259A1 (fr)
CY (1) CY1124663T1 (fr)
DK (1) DK3236972T3 (fr)
EA (1) EA201791460A1 (fr)
ES (1) ES2892123T3 (fr)
HR (1) HRP20211456T1 (fr)
HU (1) HUE056470T2 (fr)
IL (3) IL296496A (fr)
LT (1) LT3236972T (fr)
PL (1) PL3236972T3 (fr)
PT (1) PT3236972T (fr)
RS (1) RS62434B1 (fr)
SG (2) SG11201705069YA (fr)
SI (1) SI3236972T1 (fr)
WO (1) WO2016106050A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11331331B2 (en) 2017-12-07 2022-05-17 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2022159787A1 (fr) * 2021-01-22 2022-07-28 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Composés pour le traitement d'une maladie ou d'un trouble, méthodes d'identification desdits composés
WO2023012329A1 (fr) 2021-08-06 2023-02-09 Intervet International B.V. Méthode de traitement de maladies virales vétérinaires
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3033756A1 (fr) 2016-09-02 2018-03-08 Dicerna Pharmaceuticals, Inc. Analogues de 4'-phosphate et oligonucleotides comprenant ceux-ci
JP7016170B2 (ja) * 2016-12-16 2022-02-04 国立大学法人東海国立大学機構 ヌクレオシド誘導体及びその利用
EP3712160A4 (fr) 2017-10-31 2021-09-01 Yamasa Corporation Dérivé de nucléoside et utilisation associée
GB201720279D0 (en) 2017-12-05 2018-01-17 Nucana Biomed Ltd Anticancer compounds
CN108586294A (zh) * 2018-05-29 2018-09-28 王若锴 一种脲类衍生物及其在防治炎症中的应用
KR20200095934A (ko) * 2019-02-01 2020-08-11 재단법인 경기도경제과학진흥원 중동호흡기증후군 치료 또는 예방용 약학 조성물
CN112390838A (zh) * 2019-08-14 2021-02-23 斯微(上海)生物科技有限公司 一种改性核苷及其合成方法
JP2023512666A (ja) * 2020-02-07 2023-03-28 エモリー ユニバーシティー N4-ヒドロキシシチジンおよび誘導体ならびにそれらに関連する抗ウイルス用途
CN111548384B (zh) * 2020-03-29 2021-04-27 常州安蒂卫生物科技有限公司 用于抗病毒治疗的被取代的n4-羟基胞苷衍生物及其前药
US11642372B2 (en) 2020-05-01 2023-05-09 Tygrus, LLC Therapeutic material with low pH and low toxicity active against at least one pathogen for addressing patients with respiratory illnesses
US11826382B2 (en) 2020-05-01 2023-11-28 Tygrus, LLC Therapeutic material with low pH and low toxicity active against at least one pathogen for addressing patients with respiratory illnesses
US11702440B2 (en) * 2020-12-10 2023-07-18 Optimus Drugs Private Limited Pharmaceutical polymorphs of Molnupiravir
CN114621229A (zh) * 2020-12-11 2022-06-14 嘉兴金派特生物科技有限公司 治疗或预防猫传染性腹膜炎的化合物或组合物
CN112279877B (zh) * 2020-12-15 2021-08-06 南京颐媛生物医学研究院有限公司 一种核苷磷酸酯及其合成方法
CN112608357B (zh) * 2020-12-21 2022-12-09 杭州科巢生物科技有限公司 一种抗病毒药物Molnupiravir的制备方法
CN112939981B (zh) * 2021-01-19 2022-08-26 嘉兴金派特生物科技有限公司 核苷化合物、药学上可接受的盐、组合物及其应用
US11407779B1 (en) 2021-04-23 2022-08-09 Divi's Laboratories Ltd. Process for the preparation of molnupiravir
EP4334329A1 (fr) * 2021-05-06 2024-03-13 Council Of Scientific & Industrial Research Procédé de préparation de n4-hydroxycytidine et de ses dérivés
WO2022251663A2 (fr) * 2021-05-27 2022-12-01 Emory University Nouveaux agents viraux anti-arn universels
WO2022262845A1 (fr) * 2021-06-18 2022-12-22 Suzhou Spring-Sea Bio-Pharmaceuticals Co., Ltd. Dérivés d'ester de n4-hydroxycytidine et leur utilisation
CN113321694A (zh) * 2021-06-22 2021-08-31 药康众拓(江苏)医药科技有限公司 N4-羟基胞苷衍生物及其制备方法和用途
CN113735929A (zh) * 2021-07-21 2021-12-03 海化生命(厦门)科技有限公司 一种抗冠状病毒的化合物及其制备方法与应用
CN117886870A (zh) * 2021-08-27 2024-04-16 南京知和医药科技有限公司 一种新型的胞苷衍生物及其药物组合物和用途
CN113980074A (zh) * 2021-10-21 2022-01-28 苏州立新制药有限公司 N4-羟基胞苷的晶型a与制备方法和用途
TW202333706A (zh) 2021-11-01 2023-09-01 德商埃慕尼克股份公司 N—羥基胞磷膽鹼(citicoline)化合物的醫藥用途
WO2023083295A1 (fr) * 2021-11-12 2023-05-19 Suzhou Spring-Sea Bio-Pharmaceuticals Co., Ltd. Dérivés d'ester de n4-hydroxycytidine et leur utilisation
US11541071B1 (en) 2021-12-16 2023-01-03 Ascletis BioScience Co., Ltd Nucleoside derivatives and methods of use thereof
CN116554249A (zh) * 2022-01-28 2023-08-08 北京恩泰伟医药科技有限公司 抗病毒化合物及其用途
TW202348228A (zh) 2022-02-24 2023-12-16 德商艾斯巴赫生物有限公司 病毒組合療法
WO2023167488A1 (fr) * 2022-03-02 2023-09-07 유재혁 Composition vétérinaire pour administration topique de molnupiravir et son utilisation
CN116731087A (zh) * 2022-03-03 2023-09-12 上海科胜药物研发有限公司 一种莫那比拉韦及其中间体的制备方法
KR20230130898A (ko) * 2022-03-04 2023-09-12 동아대학교 산학협력단 N4-이소부티릴옥시시티딘 모방체 합성과 이의 항바이러스 용도를 포함하는 바이러스 감염 치료용 조성물
CN114573651A (zh) * 2022-03-11 2022-06-03 山东大学 一种n4-羟基胞苷脂质前药及其制备方法与应用
CN114805458B (zh) * 2022-03-31 2023-09-08 中国人民解放军军事科学院军事医学研究院 类核苷广谱抗病毒药物的脂肪酸前药及其制备方法和用途

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9809616B2 (en) * 2012-10-29 2017-11-07 Emory University Pyrimidine nucleosides and their monophosphate prodrugs for the treatment of viral infections and cancer
US10100076B2 (en) * 2000-10-18 2018-10-16 Gilead Pharmasset Llc Modified nucleosides for the treatment of viral infections and abnormal cellular proliferation
US20190083520A1 (en) * 2016-03-10 2019-03-21 Emory University N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto

Family Cites Families (273)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1325798A (en) 1970-09-24 1973-08-08 Upjohn Co Derivatives of 2,2-anhydro-ara-cytidine
GB1386334A (en) 1972-09-22 1975-03-05 Kohjin Co 2,2-cyclocytidine derivatives
GB1480120A (en) 1973-11-29 1977-07-20 Kohjin Co N4-substituted-2,2'-cyclocytidine derivatives
US4096324A (en) 1975-07-07 1978-06-20 The Upjohn Company Cytidine nucleoside compound
US5736531A (en) 1987-10-28 1998-04-07 Pro-Neuron, Inc. Compositions of chemotherapeutic agent or antiviral agent with acylated pyrimidine nucleosides
US5470838A (en) 1987-10-28 1995-11-28 Pro-Neuron, Inc. Method of delivering exogenous uridine or cytidine using acylated uridine or cytidine
EP0476071A4 (en) 1989-06-05 1992-11-04 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides and methods for preparing the same
WO1992009705A1 (fr) 1990-11-23 1992-06-11 Gilead Sciences, Inc. Oligomeres formant un triplex et contenant des bases modifiees
FI105556B (fi) * 1991-09-30 2000-09-15 Sankyo Co Menetelmä lääkeaineina käyttökelpoisten pyrimidiiniukleosidijohdannaisten valmistamiseksi, joilla on kasvaimen vastaista vaikutusta
EP1256589A3 (fr) 1991-11-26 2003-09-17 Isis Pharmaceuticals, Inc. Oligomères contenant des pyrimidines modifiées
US6057305A (en) 1992-08-05 2000-05-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Antiretroviral enantiomeric nucleotide analogs
IS2334B (is) 1992-09-08 2008-02-15 Vertex Pharmaceuticals Inc., (A Massachusetts Corporation) Aspartyl próteasi hemjari af nýjum flokki súlfonamíða
AU6632094A (en) 1993-04-19 1994-11-08 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5349947A (en) * 1993-07-15 1994-09-27 Newhouse Michael T Dry powder inhaler and process that explosively discharges a dose of powder and gas from a soft plastic pillow
AU690587B2 (en) 1993-09-17 1998-04-30 Gilead Sciences, Inc. Method for dosing therapeutic compounds
DE69435319D1 (de) 1993-09-17 2010-12-16 Gilead Sciences Inc Nukleotidanaloge
BR9607098A (pt) 1995-02-27 1997-11-04 Gilead Sciences Inc Novos inibidores seletivos de neuraminidas virais ou bacterianas
US6043358A (en) 1995-11-01 2000-03-28 Merck & Co., Inc. Hexahydro-5-imino-1,4-heteroazepine derivatives as inhibitors of nitric oxide synthases
CA2268756A1 (fr) 1996-10-21 1998-04-30 Gilead Sciences, Inc. Composes de piperidine
GB9718913D0 (en) 1997-09-05 1997-11-12 Glaxo Group Ltd Substituted oxindole derivatives
US6086376A (en) * 1998-01-30 2000-07-11 Rtp Pharma Inc. Dry aerosol suspension of phospholipid-stabilized drug microparticles in a hydrofluoroalkane propellant
US6369087B1 (en) 1999-08-26 2002-04-09 Robert R. Whittle Alkoxy substituted benzimidazole compounds, pharmaceutical preparations containing the same, and methods of using the same
YU92202A (sh) 2000-05-26 2006-01-16 Idenix (Cayman) Limited Metode i smeše za lečenje flavi virusa i pesti virusa
US20030008841A1 (en) 2000-08-30 2003-01-09 Rene Devos Anti-HCV nucleoside derivatives
EP2141244A1 (fr) 2000-10-18 2010-01-06 Pharmasset, Inc. Quantification simultanée d'acides nucléiques dans des cellules malades
CA2743451A1 (fr) 2000-10-18 2002-04-25 Pharmasset, Inc. Nucleosides modifies pour le traitement d'infections virales et de proliferations cellulaires anormales
PL209031B1 (pl) 2001-04-30 2011-07-29 Trommsdorff Arzneimittel Estry urydyny, ich zastosowanie i sposób wytwarzania, oraz kompozycje farmaceutyczne je zawierające
GB0114286D0 (en) 2001-06-12 2001-08-01 Hoffmann La Roche Nucleoside Derivatives
US7049303B2 (en) 2001-11-07 2006-05-23 Medical Research Council Inhibition of viruses
EA008775B1 (ru) 2002-04-26 2007-08-31 Джилид Сайэнс, Инк. Ингибиторы протеазы вич для лечения инфекции вич и фармацевтическая композиция
US20050239054A1 (en) 2002-04-26 2005-10-27 Arimilli Murty N Method and compositions for identifying anti-HIV therapeutic compounds
GB0215293D0 (en) 2002-07-03 2002-08-14 Rega Foundation Viral inhibitors
CN100536852C (zh) 2002-07-15 2009-09-09 基利得科学公司 L-fmau在制备联合治疗乙型肝炎病毒感染的药物中的应用
AU2003264406A1 (en) 2002-09-11 2004-04-30 Michio Ishibashi Drug or cosmetic
US20040194155A1 (en) 2002-10-01 2004-09-30 Gilead Sciences, Inc. HBV mutations associated with reduced susceptibility to adefovir
US20040157804A1 (en) 2002-10-16 2004-08-12 Gilead Sciences, Inc. Pre-organized tricyclic integrase inhibitor compounds
AU2003287606A1 (en) * 2002-11-12 2004-06-03 Enzon Pharmaceuticals, Inc. Novel acylating reagents
TWI332507B (en) 2002-11-19 2010-11-01 Hoffmann La Roche Antiviral nucleoside derivatives
WO2004050613A2 (fr) 2002-12-02 2004-06-17 Gilead Sciences, Inc. Derives de 3-propenamide substitue en 2 et procedes d'utilisation de ces derives
SI1583542T1 (sl) 2003-01-14 2008-12-31 Gilead Sciences Inc Sestavki in postopki za kombinacijsko antivirusnoterapijo
CN101410120A (zh) 2003-04-25 2009-04-15 吉里德科学公司 抗炎的膦酸酯化合物
MXPA05011294A (es) 2003-04-25 2006-03-15 Gilead Sciences Inc Compuestos de fosfonato anti-inflamatorios.
WO2004096287A2 (fr) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Composes de phosphonate inhibiteurs de l'inosine monophosphate deshydrogenase
DK1628685T3 (da) 2003-04-25 2011-03-21 Gilead Sciences Inc Antivirale phosphonatanaloge
WO2005002626A2 (fr) 2003-04-25 2005-01-13 Gilead Sciences, Inc. Composes de phosphonate therapeutiques
US20060252729A1 (en) 2003-07-30 2006-11-09 Krawczyk Steven H Nucleobase phosphonate analogs for antiviral treatment
CA2537325A1 (fr) 2003-09-19 2005-03-31 Gilead Sciences, Inc. Composes d'aza-quinolinol phosphonate inhibiteurs de l'integrase
RU2264409C2 (ru) * 2003-10-01 2005-11-20 Государственный научный центр вирусологии и биотехнологии "Вектор" (ГНЦ ВБ "Вектор") 2`-амино-2`-дезоксинуклеозиды - ингибиторы репродукции вирусов кори и марбург
JP2007508843A (ja) 2003-10-24 2007-04-12 ギリアード サイエンシーズ, インコーポレイテッド 治療用化合物の同定のための方法および組成物
EP1680435A1 (fr) 2003-11-03 2006-07-19 Cognis IP Management GmbH Ribonucleosides d'acyle et desoxyribonucleosides d'acyle
EP2161273B1 (fr) 2003-12-22 2015-04-15 K.U.Leuven Research & Development Composés d'Imidazo[4,5-c]pyridine et procédés de traitement anti-viral
MXPA06006899A (es) 2003-12-22 2006-09-04 Gilead Sciences Inc Derivados de carbovir y abacavir 4'-sustituidos asi como compuestos relacionados con actividad antiviral de virus de inmunodeficiencia humana y virus de la hepatitis c.
SI2204374T1 (sl) 2003-12-30 2012-09-28 Gilead Sciences Inc Nukleozidni fosfonati in njihovi analogi za zdravljenje HPV-infekcij
CN1934093A (zh) 2004-01-12 2007-03-21 吉里德科学公司 嘧啶基膦酸酯抗病毒化合物和使用方法
DE602005017131D1 (de) 2004-01-21 2009-11-26 Gilead Sciences Inc Verwendung von adefovir oder tenofovir zur hemmung von mmtv-artigen viren im zusammenhang mit brustkrebs und primärer biliärer zirrhose
DK1742642T3 (da) 2004-04-14 2009-02-16 Gilead Sciences Inc Phosphonatanaloge af HIV-integraseinhibitorforbindelser
UA93354C2 (ru) 2004-07-09 2011-02-10 Гилиад Сайенсиз, Инк. Местный противовирусный препарат
AU2005274700B2 (en) 2004-07-16 2011-11-10 Gilead Sciences, Inc. Antiviral compounds
BRPI0513811A (pt) 2004-07-27 2008-07-15 Gilead Sciences Inc imidazo [4,5-d] pirimidinas, seus usos e processos de preparação
US7871991B2 (en) 2004-07-27 2011-01-18 Gilead Sciences, Inc. Phosphonate analogs of HIV inhibitor compounds
DE102004051804A1 (de) * 2004-10-21 2006-04-27 Max-Delbrück-Centrum Für Molekulare Medizin (Mdc) Beta-L-N4-Hydroxycytosin-Desoxynucleoside und ihre Verwendung als pharmazeutische Mittel zur Prophylaxe oder Therapie von viralen Erkrankungen
CA2585353A1 (fr) 2004-10-26 2006-05-04 Gilead Sciences, Inc. Derives de phosphonate d'acide mycophenolique
DK1841765T5 (da) 2004-12-21 2009-12-21 Gilead Sciences Inc Imidazo[4,5-C]pyridinforbindelse og fremgangsmåde til antiviral behandling
WO2006091905A1 (fr) 2005-02-25 2006-08-31 Gilead Sciences, Inc. Derives de bicyclo(3.1.0) hexane utilises comme composes antiviraux
WO2006125048A2 (fr) 2005-05-16 2006-11-23 Gilead Sciences, Inc. Composes inhibant l'integrase
EP2526942B1 (fr) 2005-06-08 2017-08-09 The University of North Carolina At Chapel Hill Procédés facilitant la survie de cellules neuronales à l'aide de mimétiques de neurotrophine BDNF peptidiques et non peptidiques
TW200738742A (en) 2005-07-14 2007-10-16 Gilead Sciences Inc Antiviral compounds
TWI389908B (zh) 2005-07-14 2013-03-21 Gilead Sciences Inc 抗病毒化合物類
WO2007014174A2 (fr) 2005-07-25 2007-02-01 Gilead Sciences, Inc. Mutants résistant aux médicaments du virus de l'hépatite c
JP2009502964A (ja) 2005-07-27 2009-01-29 ギリアード サイエンシーズ, インコーポレイテッド Hivを阻害するための抗ウイルス性ホスホン酸結合体
CA2619049A1 (fr) 2005-08-16 2007-02-22 Irm, Llc Composes et compositions en tant qu'inhibiteurs de proteine kinase
JP4885963B2 (ja) 2005-09-22 2012-02-29 エフ.ホフマン−ラ ロシュ アーゲー ヌクレオシドの選択的o−アシル化
EP1976517A1 (fr) 2005-12-30 2008-10-08 Gilead Sciences, Inc. Procedes pour l'amelioration de la pharmacocinetique d'inhibiteurs de l'integrase vih
UA97241C2 (uk) 2006-03-29 2012-01-25 Гіліад Сайєнсіз, Інк. Спосіб одержання інгібіторів віл-протеази
US20090318456A1 (en) 2006-07-06 2009-12-24 Gilead Sciences, Inc. Substituted pteridines for the treatment and prevention of viral infections
AU2007269557B2 (en) 2006-07-07 2013-11-07 Gilead Sciences, Inc. Modulators of toll-like receptor 7
AU2007275860C1 (en) 2006-07-07 2014-06-12 Gilead Sciences, Inc. Modulators of pharmacokinetic properties of therapeutics
JP4958974B2 (ja) 2006-07-07 2012-06-20 ギリアード サイエンシーズ, インコーポレイテッド 新規ピリダジン化合物およびそれらの使用
WO2008005542A2 (fr) 2006-07-07 2008-01-10 Gilead Sciences, Inc., Composés antiviraux à base de phosphinate
US8338435B2 (en) 2006-07-20 2012-12-25 Gilead Sciences, Inc. Substituted pyrido(3,2-D) pyrimidines and pharmaceutical compositions for treating viral infections
WO2008009078A2 (fr) 2006-07-20 2008-01-24 Gilead Sciences, Inc. Dérivés de la quinazoline tri-substitués en 4,6-dl et en 2,4,6 utilisables pour traiter les infections virales
US10144736B2 (en) 2006-07-20 2018-12-04 Gilead Sciences, Inc. Substituted pteridines useful for the treatment and prevention of viral infections
US8673929B2 (en) 2006-07-20 2014-03-18 Gilead Sciences, Inc. 4,6-di- and 2,4,6-trisubstituted quinazoline derivatives and pharmaceutical compositions useful for treating viral infections
US7723380B2 (en) 2006-07-21 2010-05-25 Gilead Sciences, Inc. Antiviral protease inhibitors
NZ573888A (en) 2006-07-21 2012-02-24 Gilead Sciences Inc Aza-peptide protease inhibitors
AU2007277253A1 (en) 2006-07-24 2008-01-31 Gilead Sciences, Inc. Bisfuranyl protease inhibitors
CN101495461A (zh) 2006-07-24 2009-07-29 吉里德科学公司 新的hiv逆转录酶抑制剂
MX2009003403A (es) 2006-09-27 2009-04-09 Coley Pharm Group Inc Composiciones de ligandos de tlr y antivirales.
EP2094702B1 (fr) 2006-12-14 2015-09-02 Gilead Sciences, Inc. Inhibiteurs viraux
TW200840584A (en) 2006-12-26 2008-10-16 Gilead Sciences Inc Pyrido(3,2-d)pyrimidines useful for treating viral infections
WO2008077650A1 (fr) 2006-12-26 2008-07-03 Gilead Sciences, Inc. Pyrido(3,2-d)pyrimidines utiles pour le traitement d'infections virales
WO2008077649A1 (fr) 2006-12-26 2008-07-03 Gilead Sciences, Inc. Pyrido(3,2-d)pyrimidines utiles pour traiter des infections virales
US8324179B2 (en) 2007-02-09 2012-12-04 Gilead Sciences, Inc. Nucleoside analogs for antiviral treatment
LT2487166T (lt) 2007-02-23 2016-11-10 Gilead Sciences, Inc. Terapinių agentų farmakokinetinių savybių moduliatoriai
CN101668515B (zh) 2007-03-29 2013-03-20 弗·哈夫曼-拉罗切有限公司 药物组合物和方法
TW200918507A (en) 2007-06-29 2009-05-01 Gilead Sciences Inc Novel HIV reverse transcriptase inhibitors
AU2008270630B2 (en) 2007-06-29 2014-01-16 Gilead Sciences, Inc. Therapeutic compositions and the use thereof
AU2008271116B2 (en) 2007-06-29 2012-09-20 Gilead Sciences, Inc. Antiviral compounds
US8354421B2 (en) 2007-06-29 2013-01-15 Korea Research Insitute Of Chemical Technology HIV reverse transcriptase inhibitors
PT2170888E (pt) 2007-06-29 2015-08-21 Gilead Sciences Inc Purina derivados e sua utilização como moduladores de recetor de tipo toll 7
US20110009411A1 (en) 2007-06-29 2011-01-13 Gilead Sciences ,Inc. Therapeutic compositions and the use thereof
US20090047252A1 (en) 2007-06-29 2009-02-19 Gilead Sciences, Inc. Antiviral compounds
CN101801925A (zh) 2007-06-29 2010-08-11 吉里德科学公司 抗病毒组合物
UA99466C2 (en) 2007-07-06 2012-08-27 Гилиад Сайенсиз, Инк. Crystalline pyridazine compound
WO2009058800A2 (fr) 2007-10-29 2009-05-07 President And Fellows Of Harvard College Synthèse de nucléosides
JP2011508740A (ja) * 2007-12-27 2011-03-17 エピファニー バイオサイエンシズ, インク. 抗ウイルス化合物
WO2009088719A1 (fr) 2008-01-04 2009-07-16 Gilead Sciences, Inc. Inhibiteurs du cytrochrome p450
TWI444384B (zh) 2008-02-20 2014-07-11 Gilead Sciences Inc 核苷酸類似物及其在治療惡性腫瘤上的用途
NZ588400A (en) 2008-04-23 2012-08-31 Gilead Sciences Inc 1'-substituted carba-nucleoside analogs for antiviral treatment
WO2009143011A1 (fr) * 2008-05-20 2009-11-26 Novartis Ag Compositions antivirales, procédés de fabrication et d’utilisation de ces compositions, et système de délivrance pulmonaire de ces compositions
WO2010002998A1 (fr) 2008-07-03 2010-01-07 Gilead Sciences, Inc. Pyrido(3,2-d)pyrimidines trisubstituées en position 2,4,6 utiles pour traiter des infections virales
MX2011000306A (es) 2008-07-08 2011-03-15 Gilead Sciences Inc Sales de compuestos inhibidores de hiv.
TW201016698A (en) 2008-07-25 2010-05-01 Gilead Sciences Inc Antiviral compounds
CN102272134B (zh) 2008-12-09 2013-10-16 吉里德科学公司 Toll样受体调节剂
US20100160403A1 (en) 2008-12-19 2010-06-24 Gilead Sciences, Inc. Hcv ns3 protease inhibitors
TW201036612A (en) 2008-12-22 2010-10-16 Gilead Sciences Inc Antiviral compounds
PE20120257A1 (es) 2009-02-10 2012-03-29 Gilead Sciences Inc Analogos carba-nucleosidicos para tratamiento antiviral
RS56654B8 (sr) 2009-05-13 2021-06-30 Gilead Pharmasset Llc Antiviralna jedinjenja
TWI576352B (zh) 2009-05-20 2017-04-01 基利法瑪席特有限責任公司 核苷磷醯胺
AR077139A1 (es) 2009-06-23 2011-08-03 Gilead Sciences Inc Composiciones farmaceuticas utiles para tratar el vch
AR077138A1 (es) 2009-06-23 2011-08-03 Gilead Sciences Inc Composiciones farmaceuticas utiles para tratar el vhc
WO2010151472A1 (fr) 2009-06-23 2010-12-29 Gilead Sciences, Inc. Combinaison d'un inhibiteur de polymérase ns5b et d'un inhibiteur de protéase ns3 du virus de l'hépatite c (vhc) pour le traitement du vhc
US8999969B2 (en) 2009-07-09 2015-04-07 Gilead Sciences, Inc. Anti-RSV compounds
JP5841531B2 (ja) 2009-07-21 2016-01-13 ギリアード サイエンシーズ, インコーポレイテッド フラビウイルス科ウイルスの阻害剤
EP2507249B1 (fr) 2009-09-09 2015-11-11 Gilead Sciences, Inc. Inhibiteurs de virus de la famille des flaviviridae
AU2010292102B2 (en) 2009-09-14 2015-04-09 Gilead Sciences, Inc. Modulators of Toll-like receptors
US7973013B2 (en) 2009-09-21 2011-07-05 Gilead Sciences, Inc. 2'-fluoro substituted carba-nucleoside analogs for antiviral treatment
RS55699B1 (sr) 2009-09-21 2017-07-31 Gilead Sciences 2' -fluoro supstituisani karba-nukleozidni analozi zaantiviralno lečenje
WO2011049825A1 (fr) 2009-10-22 2011-04-28 Gilead Sciences, Inc. Dérivés de purine ou de désazapurine utiles pour le traitement (entre autres) d'infections virales
US20110150836A1 (en) 2009-12-22 2011-06-23 Gilead Sciences, Inc. Methods of treating hbv and hcv infection
PL2523950T3 (pl) 2010-01-15 2017-09-29 Gilead Sciences, Inc. Inhibitory wirusów flaviviridae
CN102712632A (zh) 2010-01-15 2012-10-03 吉里德科学公司 黄病毒科病毒的抑制剂
TW201141488A (en) 2010-01-28 2011-12-01 Hoffmann La Roche Antiviral nucleosides
US20110223131A1 (en) 2010-02-24 2011-09-15 Gilead Sciences, Inc. Antiviral compounds
AR081377A1 (es) 2010-05-10 2012-08-29 Gilead Sciences Inc Compuestos de pirazolopiridina bifuncionales
EP2569285A1 (fr) 2010-05-10 2013-03-20 Gilead Sciences, Inc. Dérivés quinoliniques bifonctionnels
WO2011146817A1 (fr) 2010-05-21 2011-11-24 Gilead Sciences, Inc. Inhibiteurs des virus flaviviridae hétérocycliques
TW201201815A (en) 2010-05-28 2012-01-16 Gilead Sciences Inc 1'-substituted-carba-nucleoside prodrugs for antiviral treatment
TW201210597A (en) 2010-06-09 2012-03-16 Gilead Sciences Inc Inhibitors of hepatitis C virus
WO2011156757A1 (fr) 2010-06-10 2011-12-15 Gilead Sciences, Inc. Combinaison de composés anti-vhc avec de la ribavirine pour le traitement du vhc
WO2011156416A1 (fr) 2010-06-11 2011-12-15 Gilead Sciences, Inc. Formulations antivirales topiques pour la prévention de la transmission du vhs-2
EP3012258B1 (fr) 2010-06-24 2018-09-19 Gilead Sciences, Inc. Composition pharmaceutique comprenant un dérivé pyrazolo[1,5-a] pyrimidine en tant qu'agent antiviral
CN103140474A (zh) 2010-07-02 2013-06-05 吉里德科学公司 治疗aids的萘-2-基乙酸衍生物
CN103209963A (zh) 2010-07-02 2013-07-17 吉里德科学公司 作为hiv抗病毒化合物的2-喹啉基-乙酸衍生物
US9090642B2 (en) 2010-07-19 2015-07-28 Gilead Sciences, Inc. Methods for the preparation of diasteromerically pure phosphoramidate prodrugs
BR122020020745B8 (pt) 2010-07-22 2023-10-31 Gilead Sciences Inc Composto antiviral para o tratamento de infecções por paramyxoviridae e composição farmacêutica que o compreende
CN102351931B (zh) * 2010-09-07 2014-01-22 河南省科学院高新技术研究中心 嘧啶核苷衍生物、合成方法及其在制备抗肿瘤、抗病毒药物中的应用
TW201305185A (zh) 2010-09-13 2013-02-01 Gilead Sciences Inc 用於抗病毒治療之2’-氟取代之碳-核苷類似物
KR101879887B1 (ko) 2010-09-20 2018-07-18 길리애드 사이언시즈, 인코포레이티드 항바이러스 치료용 2'-플루오로 치환된 카바-누클레오시드 유사체
KR20190012271A (ko) 2010-11-17 2019-02-08 길리애드 파마셋 엘엘씨 항바이러스 화합물
UA114075C2 (xx) 2010-11-19 2017-04-25 БАГАТОШАРОВА ТАБЛЕТКА, ЩО МІСТИТЬ РИЛПІВІРИН HCl, ЕМТРИЦИТАБІН І ТЕНОФОВІРУ ДИЗОПРОКСИЛФУМАРАТ
UY33775A (es) 2010-12-10 2012-07-31 Gilead Sciences Inc Inhibidores macrocíclicos de virus flaviviridae, composiciones farmacéuticas que los comprenden y sus usos
CA2822037A1 (fr) 2010-12-20 2012-06-28 Gilead Sciences, Inc. Combinations pour traiter le vhc
WO2012088153A1 (fr) 2010-12-21 2012-06-28 Gilead Sciences, Inc. Inhibiteurs de cytochrome p450
US20130274254A1 (en) 2010-12-21 2013-10-17 Gilead Sciences, Inc. Inhibitors of cytochrome p450 (cyp3a4)
US8987313B2 (en) 2010-12-21 2015-03-24 Gilead Sciences, Inc. Inhibitors of cytochrome P450
WO2012138669A1 (fr) 2011-04-04 2012-10-11 Gilead Sciences, Inc. Formes à l'état solide d'un inhibiteur du vih
TW201302760A (zh) 2011-04-04 2013-01-16 Gilead Sciences Inc 製備hiv整合酶抑制劑之方法
BR112013026219A2 (pt) 2011-04-13 2016-07-26 Gilead Sciences Inc análogos de n-nucleosídeo 1'-substituída pirimidina para tratamento antiviral
BR112013027096A2 (pt) 2011-04-21 2016-12-27 Gilead Sciences Inc compostos de benzotiazol e seu uso farmacêutico
EP2705027B1 (fr) 2011-05-02 2015-06-03 Gilead Sciences, Inc. Sels solides et amorphes de Cobicistat (GS-9350)
EP2729490A1 (fr) 2011-07-06 2014-05-14 Gilead Sciences, Inc. Réplicons du génotype 4 du vhc
AU2012278960B2 (en) 2011-07-06 2015-08-27 Gilead Sciences, Inc. HCV genotype 3 replicons
JP6205354B2 (ja) 2011-07-06 2017-09-27 ギリアード サイエンシーズ, インコーポレイテッド Hivの処置のための化合物
PT2734515E (pt) 2011-07-13 2016-03-11 Gilead Sciences Inc Derivados de ácido tiofen-2-carboxílico úteis como inibidores de vírus flaviviridae
MX336627B (es) 2011-08-16 2016-01-26 Gilead Sciences Inc Hemifumarato de tenofovir alafenamida.
DE202012013074U1 (de) * 2011-09-16 2014-10-29 Gilead Pharmasset Lcc Zusammensetzungen zur Behandlung von HCV
BR112014010295A2 (pt) 2011-10-31 2017-04-18 Gilead Pharmassett Llc métodos e composições para o tratamento de vírus de hepatite c
US20130164260A1 (en) 2011-11-16 2013-06-27 Gilead Sciences, Inc. Antiviral compounds
CN104039319B (zh) 2011-11-29 2017-12-29 吉利德制药有限责任公司 用于治疗hcv的组合物和方法
WO2013090929A1 (fr) 2011-12-15 2013-06-20 Gilead Sciences, Inc. Dérivés d'amino-quinoléine inhibiteurs de vhc
WO2013090840A1 (fr) 2011-12-15 2013-06-20 Gilead Sciences, Inc. Dérivés de 2-amino-pyrido[3,2-d]pyrimidine en tant qu'inhibiteurs du vhc
AU2012358994A1 (en) 2011-12-20 2014-08-07 Gilead Sciences, Inc. Pharmaceutical compositions and methods for treating gastrointestinal infections and disorders
PT2794611T (pt) 2011-12-22 2017-12-06 Gilead Sciences Inc Pirazolo[1,5-a]pirimidinas como agentes antivirais
UA117095C2 (uk) 2011-12-22 2018-06-25 Аліос Біофарма, Інк. Нуклеозидна сполука або її фармацевтично прийнятна сіль
WO2013103738A1 (fr) 2012-01-04 2013-07-11 Gilead Sciences, Inc. Dérivés d'acide naphtalène acétique contre l'infection par le vih
US9376392B2 (en) 2012-01-04 2016-06-28 Gilead Sciences, Inc. 2-(tert-butoxy)-2-(7-methylquinolin-6-yl) acetic acid derivatives for treating AIDS
CA2860995A1 (fr) 2012-01-12 2013-07-18 Gilead Sciences, Inc. Compositions pharmaceutiques et procedes pour leur preparation
EA026138B1 (ru) 2012-02-03 2017-03-31 Джилид Сайэнс, Инк. Комбинированная терапия, включающая тенофовир алафенамида гемифумарат и кобицистат, для применения для лечения вирусных инфекций
WO2013115916A1 (fr) 2012-02-03 2013-08-08 Gilead Sciences, Inc. Polythérapie comprenant le gs-7340 et le cobicistat destinée à être utilisée dans le traitement d'infections virales
US9481704B2 (en) 2012-03-13 2016-11-01 Gilead Sciences, Inc. 2′-substituted carba-nucleoside analogs for antiviral treatment
EA035159B1 (ru) * 2012-03-21 2020-05-06 Янссен Байофарма, Инк. Замещенные нуклеозиды, нуклеотиды и их аналоги
CA2870024C (fr) 2012-04-17 2020-04-07 Gilead Sciences, Inc. Composes et procedes pour un traitement antiviral
MD20140063A2 (ro) 2012-04-20 2014-12-31 Gilead Sciences, Inc. Derivaţi de acid benzotiazol-6-il acetic şi utilizarea acestora pentru tratarea unei infecţii HIV
US20130309196A1 (en) 2012-05-16 2013-11-21 Gilead Sciences, Inc. Antiviral compounds
US9079887B2 (en) 2012-05-16 2015-07-14 Gilead Sciences, Inc. Antiviral compounds
JP6209601B2 (ja) 2012-06-08 2017-10-04 ギリアード サイエンシーズ, インコーポレイテッド フラビウイルス科ウイルスの大環状阻害剤
SI2859009T1 (sl) 2012-06-08 2017-12-29 Gilead Sciences, Inc. Makrociklični inhibitorji flaviviridae virusov
AR091279A1 (es) 2012-06-08 2015-01-21 Gilead Sciences Inc Inhibidores macrociclicos de virus flaviviridae
UA119315C2 (uk) 2012-07-03 2019-06-10 Гіліад Фармассет Елелсі Інгібітори вірусу гепатиту с
US8841340B2 (en) 2012-08-17 2014-09-23 Gilead Sciences, Inc. Solid forms of an antiviral compound
US20140094485A1 (en) 2012-10-03 2014-04-03 Gilead Sciences, Inc. Solid state forms of hiv inhibitor
KR102159337B1 (ko) 2012-10-26 2020-09-24 코닌클리케 필립스 엔.브이. 투명 동작 모드를 가지는 무안경 입체영상 디스플레이 디바이스
CA2889208A1 (fr) 2012-10-30 2014-05-08 Gilead Sciences, Inc. Methodes therapeutiques et de diagnostic se rapportant a la lysyl oxydase de type 2 (loxl2)
AU2013341227A1 (en) 2012-11-07 2015-05-07 Gilead Sciences, Inc. HCV genotype 6 replicons
US9211300B2 (en) 2012-12-19 2015-12-15 Idenix Pharmaceuticals Llc 4′-fluoro nucleosides for the treatment of HCV
US9233974B2 (en) 2012-12-21 2016-01-12 Gilead Sciences, Inc. Antiviral compounds
LT2935303T (lt) 2012-12-21 2021-03-25 Janssen Biopharma, Inc. 4'-fluor-nukleozidai, 4'-fluor-nukleotidai ir jų analogai, skirti hcv gydymui
CN111303152B (zh) 2012-12-21 2023-04-11 吉利德科学公司 多环-氨基甲酰基吡啶酮化合物及其药物用途
TW201443037A (zh) 2013-01-09 2014-11-16 Gilead Sciences Inc 治療用化合物
JP5860197B1 (ja) 2013-01-09 2016-02-16 ギリアード サイエンシーズ, インコーポレイテッド ウイルス感染症を処置するための治療用化合物
ES2614053T3 (es) 2013-01-09 2017-05-29 Gilead Sciences, Inc. Heteroarilos de 5 miembros y su uso como antivirales
WO2014124430A1 (fr) * 2013-02-11 2014-08-14 Emory University Compositions thérapeutiques renfermant des nucléotides et des nucléosides et utilisations associées
TWI706945B (zh) 2013-03-01 2020-10-11 美商基利科學股份有限公司 供治療反轉錄病毒科病毒感染之治療性化合物
ES2735355T3 (es) 2013-03-15 2019-12-18 Gilead Sciences Inc Inhibidores macrocíclicos y bicíclicos de virus de hepatitis C
US20140273023A1 (en) * 2013-03-15 2014-09-18 Saladax Biomedical Inc. Gemcitabine immunoassay
US9187515B2 (en) 2013-04-01 2015-11-17 Idenix Pharmaceuticals Llc 2′,4′-fluoro nucleosides for the treatment of HCV
CN105705511A (zh) * 2013-04-12 2016-06-22 艾其林医药公司 用于治疗hcv的氘化核苷前药
SG11201509424VA (en) 2013-05-16 2015-12-30 Riboscience Llc 4'-fluor0-2'-methyl substituted nucleoside derivatives
CA2913206C (fr) 2013-06-26 2022-08-02 Alios Biopharma, Inc. Nucleosides, nucleotides substitues et leurs analogues
US20150072418A1 (en) 2013-08-16 2015-03-12 Gilead Sciences, Inc. Hcv genotype 4d replicons
JP6762873B2 (ja) 2013-09-11 2020-09-30 エモリー・ユニバーシテイ ヌクレオチドおよびヌクレオシド組成物ならびにこれらに関連する使用
KR102314960B1 (ko) 2013-10-11 2021-10-19 얀센 바이오파마, 인코퍼레이트. 치환된 뉴클레오사이드, 뉴클레오타이드 및 이의 유사체
UA119050C2 (uk) 2013-11-11 2019-04-25 Ґілеад Саєнсиз, Інк. ПІРОЛО[1.2-f][1.2.4]ТРИАЗИНИ, ЯКІ ВИКОРИСТОВУЮТЬСЯ ДЛЯ ЛІКУВАННЯ РЕСПІРАТОРНО-СИНЦИТІАЛЬНИХ ВІРУСНИХ ІНФЕКЦІЙ
WO2015084741A2 (fr) 2013-12-02 2015-06-11 Gilead Pharmasset Llc Procédés de traitement d'une infection par le virus de l'hépatite c chez des sujets souffrant d'une cirrhose
WO2015100144A1 (fr) 2013-12-23 2015-07-02 Gilead Sciences, Inc. Formes cristallines d'un composé antiviral
TW201609785A (zh) 2013-12-23 2016-03-16 吉李德製藥公司 固體型之抗病毒化合物
TWI660965B (zh) 2014-01-15 2019-06-01 美商基利科學股份有限公司 泰諾福韋之固體形式
US9463194B2 (en) 2014-02-05 2016-10-11 Gilead Sciences, Inc. Methods of treating patients co-infected with HIV and tuberculosis
US10202353B2 (en) 2014-02-28 2019-02-12 Gilead Sciences, Inc. Therapeutic compounds
WO2015130966A1 (fr) 2014-02-28 2015-09-03 Gilead Sciences, Inc. Agents antiviraux
WO2015179448A1 (fr) 2014-05-21 2015-11-26 Gilead Sciences, Inc. Composés thérapeutiques
AU2015274863B2 (en) 2014-06-12 2018-03-22 Gilead Sciences, Inc. Antiviral compounds
AU2015274626C1 (en) 2014-06-13 2018-08-09 Gilead Sciences, Inc. Quinazolinone derivatives as phosphatidylinositol 3-kinase inhibitors
ES2750357T3 (es) 2014-06-13 2020-03-25 Gilead Sciences Inc Inhibidores de fosfatidilinositol 3-quinasa
SG11201610095SA (en) 2014-06-13 2016-12-29 Gilead Sciences Inc Phosphatidylinositol 3-kinase inhibitors
NZ726360A (en) 2014-06-13 2018-04-27 Gilead Sciences Inc Phosphatidylinositol 3-kinase inhibitors
WO2015191745A1 (fr) 2014-06-13 2015-12-17 Gilead Sciences, Inc. Inhibiteur de la phosphatidylinositol 3-kinase
TW201613936A (en) 2014-06-20 2016-04-16 Gilead Sciences Inc Crystalline forms of(2R,5S,13aR)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazepine-10-carboxamide
AP2016009653A0 (en) 2014-06-24 2016-12-31 Alios Biopharma Inc Substituted nucleosides, nucleotides and analogs thereof
JP6728075B2 (ja) 2014-06-24 2020-07-22 ヤンセン バイオファーマ インク. 置換ヌクレオシド、ヌクレオチドおよびその類似体
CN105288635A (zh) 2014-06-26 2016-02-03 昆明积大制药股份有限公司 一种含有阿糖胞苷5’-0-氨基酸酯的药物组合物
AU2015287773B2 (en) 2014-07-11 2018-03-29 Gilead Sciences, Inc. Modulators of toll-like receptors for the treatment of HIV
TWI678369B (zh) 2014-07-28 2019-12-01 美商基利科學股份有限公司 用於治療呼吸道合胞病毒感染之噻吩並[3,2-d]嘧啶、呋喃並[3,2-d]嘧啶及吡咯並[3,2-d]嘧啶化合物類
TWI673283B (zh) 2014-08-21 2019-10-01 美商基利科學股份有限公司 2’-氯胺基嘧啶酮及嘧啶二酮核苷類
WO2016033243A1 (fr) 2014-08-29 2016-03-03 Gilead Sciences, Inc. Agents antirétroviraux
US20160067255A1 (en) 2014-09-04 2016-03-10 Gilead Sciences, Inc. Methods of treating or preventing hiv in patients using a combination of tenofovir alafenamide and dolutegravir
EP3034499A1 (fr) 2014-12-17 2016-06-22 Gilead Sciences, Inc. Nouveaux composés de modulation (FXR NR1H4)
TWI695003B (zh) 2014-12-23 2020-06-01 美商基利科學股份有限公司 多環胺甲醯基吡啶酮化合物及其醫藥用途
TWI699355B (zh) 2014-12-24 2020-07-21 美商基利科學股份有限公司 喹唑啉化合物
CN107428693B (zh) 2014-12-24 2020-05-29 吉利德科学公司 用于hiv治疗的异喹啉化合物
NZ733135A (en) 2014-12-24 2018-06-29 Gilead Sciences Inc Fused pyrimidine compounds for the treatment of hiv
BR112017013858A2 (pt) 2014-12-26 2018-02-27 Univ Emory n4-hidroxicitidina e derivados e usos antivirais relacionados aos mesmos
WO2016134054A1 (fr) 2015-02-18 2016-08-25 Abbvie Inc. Composés anti-viraux
PL3321265T3 (pl) 2015-03-04 2020-11-16 Gilead Sciences, Inc. Związki 4,6-diamino-pirydo[3,2-d]pirymidynowe i ich wykorzystanie jako modulatorów receptorów toll-podobnych
EP3466490B1 (fr) 2015-04-02 2020-10-21 Gilead Sciences, Inc. Dérivés de carbamoylpyridone polycycliques et leur emploi pharmaceutique
US20160303095A1 (en) 2015-04-14 2016-10-20 Gilead Sciences, Inc. Methods of treating hepatitis b virus
NZ736709A (en) 2015-05-15 2019-05-31 Gilead Sciences Inc Benzimidazole and imidazopyridine carboximidamide compounds having activity as inhibitors of indoleamine 2,3-dioxygenase
MA46677A (fr) 2015-06-17 2019-09-11 Gilead Sciences Inc Co-cristaux, sels et formes solides de ténofovir alafénamide
AU2016287500B2 (en) 2015-06-30 2019-05-02 Gilead Sciences, Inc. Pharmaceutical formulations comprising tenofovir and emtricitabine
CA2921336A1 (fr) 2015-06-30 2016-12-30 Gilead Sciences, Inc. Formulations pharmaceutiques
MX2018001073A (es) 2015-08-06 2018-06-12 Chimerix Inc Nucleosidos de pirrolopirimidina y analogos de los mismos utiles como agentes antivirales.
JP2018525412A (ja) 2015-08-26 2018-09-06 ギリアード サイエンシーズ, インコーポレイテッド 重水素化トール様受容体調節因子
WO2017040896A1 (fr) 2015-09-02 2017-03-09 Abbvie Inc. Composés anti-viraux
WO2017040895A1 (fr) 2015-09-02 2017-03-09 Abbvie Inc. Composés anti-viraux
CA2997955A1 (fr) 2015-09-15 2017-03-23 Gilead Sciences, Inc. Modulateurs de recepteurs de type toll pour le traitement du vih
LT3785717T (lt) 2015-09-16 2022-04-11 Gilead Sciences, Inc. Coronaviridae infekcijų gydymo būdai
CA2999516A1 (fr) 2015-09-30 2017-04-06 Gilead Sciences, Inc. Composes et combinaisons pour le traitement du vih
JP2018530550A (ja) 2015-10-01 2018-10-18 ギリアド サイエンシズ, インコーポレイテッド 癌を治療するためのbtk阻害剤とチェックポイント阻害剤との組合せ
EP3346995B1 (fr) 2015-11-09 2019-08-28 Gilead Sciences, Inc. Compositions thérapeutiques pour le traitement du virus de l'immunodéficience humaine
EA202091898A1 (ru) 2015-12-15 2021-06-22 Джилид Сайэнс, Инк. Антитела, нейтрализующие вирус иммунодефицита человека
CN108430993A (zh) 2015-12-17 2018-08-21 吉利德科学公司 Tank-结合激酶抑制剂化合物
US11963972B2 (en) 2016-03-23 2024-04-23 Emory University Antiviral agents and nucleoside analogs for treatment of Zika virus
JP6970086B2 (ja) 2016-04-08 2021-11-24 クリスタル バイオテック,インコーポレイティド 皮膚の創傷、障害、及び疾患を処置するための組成物及び方法
WO2017184668A1 (fr) 2016-04-20 2017-10-26 Gilead Sciences, Inc. Méthodes de traitement d'infections virales à flaviviridae
WO2017184670A2 (fr) 2016-04-22 2017-10-26 Gilead Sciences, Inc. Méthodes de traitement d'infections à virus zika
BR102017010009A2 (pt) 2016-05-27 2017-12-12 Gilead Sciences, Inc. Compounds for the treatment of hepatitis b virus infection
JP6770098B2 (ja) 2016-05-27 2020-10-14 ギリアード サイエンシーズ, インコーポレイテッド Ns5a、ns5bまたはns3阻害剤を使用する、b型肝炎ウイルス感染症を処置するための方法
WO2017223020A1 (fr) 2016-06-20 2017-12-28 Merck Sharp & Dohme Corp. Dérivés de nucléosides cycliques à substitution phosphate et leurs procédés d'utilisation pour le traitement de maladies virales
WO2017223268A1 (fr) 2016-06-22 2017-12-28 Yale University Compositions et procédés de resensibilisation de cellules à des inhibiteurs des protéines de bromodomaine et de domaine extraterminal (beti)
MX2020005392A (es) * 2017-12-07 2020-12-07 Univ Emory N4-hidroxicitidina y derivados y usos antivirales relacionados con los mismos.
US20210308168A1 (en) 2018-03-07 2021-10-07 Emory University 4'-halogen containing nucleotide and nucleoside therapeutic compositions and uses related thereto
JP2023512666A (ja) * 2020-02-07 2023-03-28 エモリー ユニバーシティー N4-ヒドロキシシチジンおよび誘導体ならびにそれらに関連する抗ウイルス用途

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10100076B2 (en) * 2000-10-18 2018-10-16 Gilead Pharmasset Llc Modified nucleosides for the treatment of viral infections and abnormal cellular proliferation
US9809616B2 (en) * 2012-10-29 2017-11-07 Emory University Pyrimidine nucleosides and their monophosphate prodrugs for the treatment of viral infections and cancer
US20190083520A1 (en) * 2016-03-10 2019-03-21 Emory University N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11331331B2 (en) 2017-12-07 2022-05-17 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11903959B2 (en) 2017-12-07 2024-02-20 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2022159787A1 (fr) * 2021-01-22 2022-07-28 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Composés pour le traitement d'une maladie ou d'un trouble, méthodes d'identification desdits composés
WO2023012329A1 (fr) 2021-08-06 2023-02-09 Intervet International B.V. Méthode de traitement de maladies virales vétérinaires

Also Published As

Publication number Publication date
BR112017013858A2 (pt) 2018-02-27
IL279663B (en) 2022-10-01
US20210060050A1 (en) 2021-03-04
US11628181B2 (en) 2023-04-18
AU2021203840B2 (en) 2023-08-24
LT3236972T (lt) 2021-11-10
WO2016106050A1 (fr) 2016-06-30
SG10202105371YA (en) 2021-07-29
AU2023270335A1 (en) 2023-12-14
CA2972259A1 (fr) 2016-06-30
HUE056470T2 (hu) 2022-02-28
SG11201705069YA (en) 2017-07-28
JP2018500354A (ja) 2018-01-11
SI3236972T1 (sl) 2021-12-31
EP3236972A4 (fr) 2019-01-23
DK3236972T3 (en) 2021-10-04
JP2022058363A (ja) 2022-04-12
EP3939985A1 (fr) 2022-01-19
PT3236972T (pt) 2021-09-24
HRP20211456T1 (hr) 2021-12-24
JP2023153865A (ja) 2023-10-18
RS62434B1 (sr) 2021-11-30
CY1124663T1 (el) 2022-07-22
JP7381190B2 (ja) 2023-11-15
KR20170123308A (ko) 2017-11-07
IL296496A (en) 2022-11-01
EA201791460A1 (ru) 2017-12-29
AU2021203840C1 (en) 2024-01-25
IL279663B2 (en) 2023-02-01
EP3939985B1 (fr) 2024-05-08
PL3236972T3 (pl) 2022-03-07
US20230293566A1 (en) 2023-09-21
ES2892123T3 (es) 2022-02-02
EP3236972A1 (fr) 2017-11-01
AU2015370004B2 (en) 2021-03-11
AU2015370004A1 (en) 2017-07-13
IL252997A0 (en) 2017-08-31
KR20230130175A (ko) 2023-09-11
EP3236972B1 (fr) 2021-07-28
AU2021203840A1 (en) 2021-07-08
CN107427529A (zh) 2017-12-01
IL252997B (en) 2021-01-31
IL279663A (en) 2021-03-01

Similar Documents

Publication Publication Date Title
US11628181B2 (en) N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US10874683B2 (en) N4-hydroxycytidine and derivatives and anti-viral uses related thereto
KR102248165B1 (ko) N4-하이드록시사이티딘 및 유도체 및 이와 관련된 항-바이러스 용도
EA045500B1 (ru) Противовирусное применение производных n4-гидроксицитидина

Legal Events

Date Code Title Description
AS Assignment

Owner name: EMORY UNIVERSITY, GEORGIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PAINTER, GEORGE R.;GUTHRIE, DAVID B.;BLUEMLING, GREGORY R.;AND OTHERS;SIGNING DATES FROM 20150805 TO 20150807;REEL/FRAME:043857/0039

Owner name: EMORY UNIVERSITY, GEORGIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PAINTER, GEORGE R.;GUTHRIE, DAVID B.;BLUEMLING, GREGORY R.;AND OTHERS;REEL/FRAME:043857/0150

Effective date: 20150107

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

AS Assignment

Owner name: DEFENSE THREAT REDUCTION AGENCY, US DOD, VIRGINIA

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:EMORY UNIVERSITY;REEL/FRAME:052689/0661

Effective date: 20200518

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION