WO2023144830A1 - Inhibiteurs de la protéine zika m utilisés en tant qu'agents anti-virus zika - Google Patents

Inhibiteurs de la protéine zika m utilisés en tant qu'agents anti-virus zika Download PDF

Info

Publication number
WO2023144830A1
WO2023144830A1 PCT/IL2023/050105 IL2023050105W WO2023144830A1 WO 2023144830 A1 WO2023144830 A1 WO 2023144830A1 IL 2023050105 W IL2023050105 W IL 2023050105W WO 2023144830 A1 WO2023144830 A1 WO 2023144830A1
Authority
WO
WIPO (PCT)
Prior art keywords
zika
protein
day
channel blocker
cell
Prior art date
Application number
PCT/IL2023/050105
Other languages
English (en)
Inventor
Isaiah Arkin
Miriam KRUGLIAK
Original Assignee
Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. filed Critical Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd.
Publication of WO2023144830A1 publication Critical patent/WO2023144830A1/fr

Links

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
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/7036Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin having at least one amino group directly attached to the carbocyclic ring, e.g. streptomycin, gentamycin, amikacin, validamycin, fortimicins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/155Amidines (), e.g. guanidine (H2N—C(=NH)—NH2), isourea (N=C(OH)—NH2), isothiourea (—N=C(SH)—NH2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • A61K31/567Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in position 17 alpha, e.g. mestranol, norethandrolone
    • 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
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • 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
    • A61K31/7072Compounds 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 having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • 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
    • 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

  • the present invention is in the field of anti-viral therapy.
  • Flaviviruses are a positive-sense, single-stranded RNA virus family that contains several important human pathogenic viruses. It includes dengue virus (DenV), Japanese encephalitis virus (JEV), tick-bome encephalitis virus (TBEV), West Nile virus (WNV), yellow fever virus (YFV), and Zika virus (ZikV). Such viruses pose a serious threat to human health and have the potential to cause future epidemics and pandemics. Reports that have surfaced recently about potential outbreaks include DenV, the recent ZikV outbreak in South America, YFV outbreaks in Africa and Brazil, and the spread of WNV across North America.
  • Zika Virus is an enveloped flavivirus with icosahedral symmetry.
  • the virus’ genome is made up of ca. 11 kb long positive-sense single-stranded RNA. It encodes three structural proteins, namely: capsid (C), precursor membrane (prM), envelope (E), and seven non- structural proteins (NS).
  • C capsid
  • prM precursor membrane
  • E envelope
  • NS non- structural proteins
  • prM Precursor membrane, prM, one of the most extensively studied structural proteins from flaviviruses, plays an important role in the viral infectivity cycle and its release. After synthesis, prM protein protects premature fusion of the E protein with transport vesicles at low pH. Before the virus’s release, a furin-like protease from the host cleaves prM into matured M protein in the trans-Golgi network. The cleavage of prM into M is required for membrane fusion between the viral and host endosomal membranes as it triggers rearrangement of the E protein and maturation of the virus. [006] Phylogenetic analyses of ZikV identified African and Asian lineages, with the latter responsible for the epidemics detected so far.
  • the African strain MR766 is known for its higher virulence. It is reported that when the prM gene of MR766 is replaced with the Asian strain PRVABC59, its lethality is reduced in IFNa/p receptor knockout (IFNAR-/-) mice. This finding is associated with reduced neuro-invasiveness of the virus after subcutaneous infection.
  • IFNAR-/- mice IFNa/p receptor knockout mice. This finding is associated with reduced neuro-invasiveness of the virus after subcutaneous infection.
  • the higher neuro-invasiveness of the African variant has been linked to changes in highly conserved amino acids that affect the positive charge and hydrophobicity of the exposed surface of the protein. These conserved amino acids are related to the higher capability of the virus to cross the blood-brain barrier.
  • Viroporins are a family of small, hydrophobic integral membrane proteins that possess channel activity.
  • the M2 channel from the Influenza A virus is one of the most well- characterized viroporins. Inhibition of the M2 channel by amantadine and rimantadine curb the viral infectivity, thereby demonstrating the potential of viroporins to serve as drug targets.
  • the functions of flavivirus M proteins as an ion channel were reported in earlier studies of DenV and WNV.
  • the present invention in some embodiments, is based, at least in part on the surprising findings that drug repurposing screening efforts have yielded several blockers, demonstrating that ZikV-M could be an attractive target to curb the infectivity of the virus. Further disclosed herein, is characterization of matured Zika virus M protein as a potential viroporin utilizing three different bacteria-based assays. The mutational study of the transmembrane region of matured membrane glycoprotein M protein sheds light on the effect of conserved amino acids residues among ZikV, WNV, and DenV.
  • a method of treating or preventing Zika virus virulence in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Zika M protein channel blocker, thereby treating or preventing Zika virus virulence in the subject.
  • a method of preventing Zika virus entry, uncoating and/or release from a cell comprising contacting the cell with a Zika M protein channel blocker, thereby preventing Zika virus cell entry, uncoating and/or release from the cell.
  • a pharmaceutical composition comprising a Zika M protein channel blocker for use in treating or preventing Zika virus virulence in a subject in need thereof.
  • a pharmaceutical composition comprising a Zika M protein channel blocker for use in preventing Zika virus cell entry, uncoating and/or release from a cell.
  • the cell is a cell of a subject, and the contacting is administering to the subject.
  • the subject is a subject infected or suspected as being infected by Zika virus.
  • the Zika M protein channel blocker is at least one molecule selected from the group consisting of: Streptomycin, Spiramycin, AZD-5423, Dabigatran etexillate, Zosuquidar Desogestrel, Floxuridine, Dequalinium chloride, Pentamidine, and Capreomycin, and a salt thereof.
  • the Zika M protein channel blocker is for use at a daily dose of 0.01 to 500 mg/kg.
  • Figs. 1A-1B include graphs showing standardization of ZikV-M as a viroporin in the negative genetic assay. Growth curves (1A) and maximal growth rates (IB) of DH10B bacteria containing MBP-ZikV-M chimera were grown in LB media with an increasing dose of IPTG, as noted. [021] Figs. 2A-2B include graphs showing positive genetic assay utilizing K+-uptake deficient bacteria, which are unable to grow on LB media without potassium supplement. Growth curves (2A) and maximal growth rates (2B) of bacteria harboring the ZikV-M plasmid grown with different concentrations of IPTG, as noted.
  • Figs. 3A-3B include graphs showing proton conductivity of flavivirus viroporins and mutants thereof. Wild-type ZikV-M, WNV MgM, DenV MgM, and mutants were transformed into bacteria that harbor a pH-sensitive green fluorescence protein. Proton flow for uninduced wild type, induced wild type, and mutants (100 pM IPTG) were recorded after two hours of induction. Panel (3 A) depicts the H+ concentration change as a function of time, while the slopes of the individual curves are listed in panel (3B).
  • Figs. 4A-4C include a graph showing the effect of mutants of flavivirus membrane proteins in the positive genetic assay. Maximal growth rates of K + -uptake deficient bacteria that express mutants of ZikV-M (4A), WNV MgM (4B), and DenV MgM (4C) grown with 10 mM IPTG.
  • Fig. 5 include a graph showing the effect of mutants of flavivirus membrane proteins in the positive genetic assay. Maximal growth rates of K+-uptake deficient bacteria that express mutants of ZikV-M, WNV MgM, and DenV MgM grown with 10 pM IPTG.
  • Figs. 6A-6C include graphs showing drug screening on wild-type ZikV-M. Maximal growth rate as a function of the different drugs (100 mM) in the negative genetic assay (6A) or positive (6B) assays. (6C) Effect of drugs on proton flow examined on bacteria that express a pH-sensitive GFP. Note, that effective compounds are expected to increase the growth rate in the negative assay (6A), decrease growth in the positive assay (6B), and decrease H + flow in the pH-assay (6C).
  • Fig. 7 includes a graph showing cell viability under viral; infection in the presence of different concentrations of the indicated drugs. Cell viability was monitored by MTS analyses, and the results are normalized relative to uninfected cells as " 100%” and untreated cells as "0%”.
  • Figs. 8A-8C include graphs showing negative genetic assay of flavivirus viroporins. Negative genetic assay of ZikV-M wild-type and mutants (8A), WNV MgM wild-type and mutants (SB) and DenV MgM wildtype and mutants (8C). All figures depict growth curve as a function of the IPTG inducer. The different mutations are indicated.
  • Figs. 9A-9C include graphs showing positive genetic assay of flavivirus viroporins. Positive genetic assay of ZikV-M wild-type and mutants (9A), WNV MgM wild-type and mutants (9B) and DenV MgM wild-type and mutants (9C). The different mutations are indicated.
  • Figs. 10A-10B include graphs showing proton flow assay of flavivirus viroporins. Proton flow assay of WNV MgM wild-type and mutants (10A) and DenV MgM wild-type and mutants (10B).
  • the present invention in some embodiments, provides compositions comprising a Zika M protein channel blocker for treating or preventing Zika virus virulence in a subject.
  • the present invention in some embodiments, provides compositions comprising a Zika virus M protein ion channel blocker for preventing Zika virus cell entry, uncoating and/or release from a cell.
  • the invention is based, at least in part, on the finding using three bacteria-based assays, that Zika M protein is an ion channel.
  • the invention is further based, at least in part, on a finding that any one of Streptomycin, Spiramycin, AZD-5423, Dabigatran etexillate, Zosuquidar Desogestrel, Floxuridine, Dequalinium chloride, Pentamidine, and Capreomycin, inhibits Zika M protein and therefore can be used to treat and prevent Zika virus virulence.
  • Zika M protein is known to one skilled in the art and has a GenBank Accession no: Zika M (YP_009227197.1). According to some embodiments, the Zika M protein comprises the amino acid sequence:
  • the Zika M protein comprises an analog of SEQ ID NO: 1, such as an analog having at least 85%, at least 90%, at least 95% identity to SEQ ID NO: 1.
  • the invention provides a method of treating or preventing Zika virus virulence in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a Zika M protein channel blocker, thereby treating or preventing Zika virus virulence in said subject.
  • the invention provides a method of treating or preventing Zika fever in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a Zika M protein channel blocker, thereby treating or preventing Zika fever.
  • the invention provides a method of preventing Zika virus release from a cell, the method comprising contacting the cell with a Zika M protein channel blocker, thereby preventing Zika virus release from said cell.
  • the invention provides a method of preventing Zika virus cell entry, the method comprising contacting the cell with a Zika M protein channel blocker, thereby preventing Zika virus cell entry.
  • the invention provides a method of preventing Zika virus uncoating, the method comprising contacting the cell with a Zika M protein channel blocker, thereby preventing Zika virus uncoating.
  • the cell is a cell of a subject. According to some embodiments, contacting is administering to the subject. According to some embodiments, said subject is a subject infected or suspected as being infected by Zika virus.
  • the Zika M protein channel blocker is Spiramycin or any salt thereof.
  • the Zika M protein channel blocker is Dabigatran etexillate or any salt thereof.
  • the Zika M protein channel blocker is Desogestrel or any salt thereof.
  • the Zika M protein channel blocker is Floxuridine or any salt thereof.
  • the Zika M protein channel blocker is Nafamostat, or any salt thereof.
  • the Zika M protein channel blocker Kasugamycin or any salt thereof.
  • the Zika M protein channel blocker is 5- Azacytidine or any salt thereof.
  • the Zika M protein channel blocker is AZD-5423 or any salt thereof.
  • the Zika M protein channel blocker is Streptomycin or any salt thereof. [049] According to some embodiments, the Zika M protein channel blocker is Zosuquidar or any salt thereof.
  • the Zika M protein channel blocker is Dequalinium chloride or any salt thereof.
  • the Zika M protein channel blocker is Capreomycin or any salt thereof.
  • the Zika M protein channel blocker is Pentamidine or any salt thereof.
  • the invention provides a Zika M protein channel blocker for use in treating or preventing Zika virus virulence in a subject in need thereof.
  • the invention provides a Zika M protein channel blocker for use in preventing Zika virus release from a cell.
  • the Zika M protein channel blocker is within a pharmaceutical composition, the pharmaceutical composition further comprising a pharmaceutically acceptable carrier.
  • the invention provides a pharmaceutical composition comprising Spiramycin, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Spiramycin or any salt thereof.
  • Spiramycin includes: 2-[(4R,5S,6S,7R,9R,10R,l lE,13E,16R)-6- ⁇ [(2S,3R,4R,5S,6R)-5- ⁇ [(2S,5S,6S)-4,5-dihydroxy-4,6-dimethyloxan-2-yl]oxy ⁇ -4- (dimethylamino)-3-hydroxy-6-methyloxan-2-yl]oxy]-10- ⁇ [(2R,5S,6R)-5-(dimethylamino)- 6-methyloxan-2-yl]oxy]-4-hydroxy-5-methoxy-9,16-dimethyl-2-oxo-l-oxacyclohexadeca- 11,13 -dien-7-yl] acetaldehyde (CAS: 8025-81-8), as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof.
  • Spira 2-[(4R,5
  • the invention provides a pharmaceutical composition comprising Dabigatran etexillate, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Dabigatran etexillate or any salt thereof.
  • Dabigatran etexillate includes Pradaxa, Pradax, Prazaxa (CAS: 211915-06-9; Ethyl N-[(2- ⁇ [(4- ⁇ N'-)
  • the invention provides a pharmaceutical composition comprising Desogestrel, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Desogestrel or any salt thereof.
  • Desogestrel includes Cerazette, Lovima, Hana, DSG; ORG-2969 (CAS: 54024-22-5; 3-Deketo-l l-methylene-17a-ethynyl-18-methyl-19-nortestosterone; 11 -Methylene- 17a-ethynyl-18-methylestr-4-en-17P-ol, (8S,9S,10R,13S,14S,17R)-13- ethyl-17-ethynyl-l l-methylidene-l,2,3,6,7,8,9,10,12,14,15,16- dodecahydrocyclopenta[a]phenanthren-17-ol), as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof. Desogestrel is described, for example, in Chinese Patent Application No. CN102532232A.
  • the invention provides a pharmaceutical composition comprising Floxuridine, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Floxuridine, an analog or a salt thereof.
  • Floxuridine includes 5-fluorodeoxy uridine (CAS: 50-91-9; 5-Fluoro- l-[4-hydroxy-5-(hydroxymethyl)tetrahydrofuran-2-yl]-lH-pyrimidine-2, 4-dione), as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof.
  • Floxuridine is described, for example, in U.S. Patent(s) 7,842,676.
  • the invention provides a pharmaceutical composition comprising Nafamostat, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Nafamostat an analog or a salt thereof.
  • Nafamostat includes Nafamostat (CAS: 81525-10-2; 6- [amino(imino)methyl]-2-naphthyl 4- ⁇ [amino(imino)methyl]amino]benzoate) as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof. Nafamostat is described, for example, in International Patent Application Publication No. W02014007326.
  • the invention provides a pharmaceutical composition comprising Kasugamycin, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Kasugamycin an analog or a salt thereof.
  • Kasugamycin includes Kasugamycin (Ksg) (CAS: 6980-18-3; 2- amino-2-[(2R,3S,5S,6R)-5-amino-2-methyl-6-[(2R,3S,5S,6S)-2,3,4,5,6- pentahydroxycyclohexyl]oxyoxan-3-yl]iminoacetic acid) as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof. Kasugamycin is described, for example, in U.S. Patent(s) 3,358,001.
  • the invention provides a pharmaceutical composition comprising 5- Azacytidine, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is 5- Azacytidine an analog or a salt thereof.
  • 5-Azacytidine as used herein, includes Ladakamycin (CAS: 320-67-2; 5- Azacitidine, 4-Amino-l-(P-D-ribofuranosyl)-l,3,5-triazin-2(lH)-one) as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof.
  • 5-Azacytidine is described, for example, in U.S. Patent(s) 9,951,098.
  • the invention provides a pharmaceutical composition comprising AZD-5423, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is AZD-5423 an analog or a salt thereof.
  • AZD-5423 includes AZD-5432 (CAS: 1034148-04-3; 2,2,2-Trifluoro-N-[(lR,2S)- 1 - ⁇ [ 1 -(4-fluorophenyl)- 1 H-indazol-5-yl]oxy ⁇ - 1 - (3 -methoxyphenyl)-2-propanyl] acetamide) as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof.
  • the invention provides a pharmaceutical composition comprising Streptomycin, an analog, or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Streptomycin an analog or a salt thereof.
  • Streptomycin includes Streptomycin (CAS: 57-92-1; 5-(2,4- diguanidino-3,5,6-trihydroxy-cyclohexoxy)-4-[4,5-dihydroxy-6-(hydroxymethyl)-3- methylamino-tetrahydropyran-2-yl] oxy-3-hydroxy-2-methyl-tetrahydrofuran-3- carbaldehyde) as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof.
  • the invention provides a pharmaceutical composition comprising Zosuquidar, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Zosuquidar an analog or a salt thereof.
  • Zosuquidar includes LY-335979 (CAS:167354-41-8; (2R)-l- ⁇ 4- [( 1 aR, 1 ObS )- 1 , 1 -difluoro- 1 , 1 a, 6, 1 Ob-tetrahydrodibenzo [a,e] cyclopropa[c] [7] annulen-6- yl]-3-(quinolin-5-yloxy)propan-2-ol) as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof.
  • Zosuquidar is described, for example, in International Patent Application Publication No. W02007008490.
  • the invention provides a pharmaceutical composition comprising Dequalinium chloride, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Dequalinium chloride an analog or a salt thereof.
  • Dequalinium includes Dequalinium chloride (CAS: 6707-58-0; 1,1'- decane- 1 , 10-diylbis(4-amino-2-methylquinolinium) decyl] -2-methyl-4-quinolin- 1 - iumamine dichloride) as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof.
  • Dequalinium chloride is described, for example, in Chinese Patent(s) 106854179.
  • the invention provides a pharmaceutical composition comprising Capreomycin, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Capreomycin an analog or a salt thereof.
  • Capreomycin includes Capreomycin (CAS: 11003-38-6; (3S)-3,6 diamino-N-[[(2S, 5S,8E,1 IS, 15S)-15-amino-l l-[(4R)-2-amino-3, 4,5,6- tetrahydropyrimidin-4-yl]-8-[(carbamoylamino)methylidene]-2-(hydroxymethyl)- 3,6,9,12,16-pentaoxo-l,4,7,10,13-pentazacyclohexadec-5-yl]methyl]hexanamide; (3S)-3,6- diamino-N-[[(2S,5S,8E,HS,15S)-15-amino-l l-[(4R)-2-amino-3,4,5,6- tetrahydropyrimidin-4-yl]-8-[(carbamoylamino)
  • the invention provides a pharmaceutical composition comprising Pentamidine, an analog or a salt thereof, for treating a viral infection.
  • the viral infection is a Zika virus infection.
  • the viral infection is an infection by virus having an M protein being an ion channel.
  • the Zika M protein channel blocker is Pentamidine an analog or a salt thereof.
  • Pentamidine includes Nebupent, and/or Pentam (CAS: 100-33-4; 4,4'-[pentane-l,5-diylbis(oxy)]dibenzenecarboximidamide) as well as pharmaceutically acceptable salts, solvates, hydrates, or mixtures thereof. Pentamidine is described, for example, in International Patent Application Publication No. W02003092616.
  • treatment encompasses alleviation of at least one symptom thereof, a reduction in the severity thereof, or inhibition of the progression thereof. Treatment need not mean that the disease, disorder, or condition is totally cured.
  • a useful composition herein needs only to reduce the severity of a disease, disorder, or condition, reduce the severity of symptoms associated therewith, or provide improvement to a patient or subject’s quality of life.
  • administering refers to any method which, in sound medical practice, delivers a composition containing an active agent to a subject in such a manner as to provide a therapeutic effect.
  • the terms “subject” or “individual” or “animal” or “patient” or “mammal” refers to any subject, particularly a mammalian subject, for whom therapy is desired, for example, a human.
  • a therapeutically effective dose of the composition of the invention is administered.
  • therapeutically effective amount refers to an amount of a drug effective to treat a disease or disorder in a mammal.
  • a therapeutically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. The exact dosage form and regimen would be determined by the physician according to the patient's condition.
  • the dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
  • the route of administration of the pharmaceutical compositions will depend on the disease or condition to be treated. Suitable routes of administration include, but are not limited to, parenteral injections, e.g., intradermal, intravenous, intramuscular, intralesional, subcutaneous, intrathecal, and any other mode of injection as known in the art.
  • compositions of the invention can be lower than when administered via parenteral injection, by using appropriate compositions it is envisaged that it will be possible to administer the compositions of the invention via transdermal, oral, rectal, vaginal, topical, nasal, inhalation, and ocular modes of treatment.
  • the composition of the invention is delivered orally.
  • the composition of the invention is an oral composition.
  • the composition of the invention further comprises orally acceptable carrier, excipient, or a diluent.
  • the Zika M protein channel blocker is for use at a daily dose of 0.01 to 500 mg/kg.
  • the Zika M protein channel blocker is Spiramycin or a salt thereof and is for use at a daily dose of between about 100 mg/day and about 5,000 mg/day, about 500 mg/day and 4,000 mg/day, and 250 mg/day and 7,500 mg/day.
  • the Zika M protein channel blocker is Desogestrel or a salt thereof and is for use at a daily dose of between about 0.1 mg/day and about 1 mg/day, about 0.05 mg/day and 1 mg/day, and 0.1 mg/day and 10 mg/day.
  • the Zika M protein channel blocker is Dabigatran etexillate or a salt thereof and is for use at a daily dose of between about 50 mg/day and about 500 mg/day, about 15 mg/day and 200 mg/day, and 30 mg/day and 250 mg/day.
  • the Zika M protein channel blocker is Floxuridine or a salt thereof and is for use at a daily dose of between about 0.01 mg/day and about 100 mg/day, about 0.01 mg/day and 0.1 mg/day, and 0.01 mg/day and 10 mg/day.
  • the Zika M protein channel blocker is Nafamostat or a salt thereof and is for use at a daily dose of between about 0.1 mg/day and about 10 mg/day, about 0.5 mg/day and 15 mg/day, and 1 mg/day and 20 mg/day.
  • the Zika M protein channel blocker is Kasugamycin or a salt thereof and is for use at a daily dose of between about 1 mg/day and about 100 mg/day, about 5 mg/day and 2,000 mg/day, and 2 mg/day and 400 mg/day.
  • the Zika M protein channel blocker is 5- Azacytidine or a salt thereof and is for use at a daily dose of between about 1 mg/day and about 100 mg/day, about 0.5 mg/day and 50 mg/day, and 0.1 mg/day and 200 mg/day.
  • the Zika M protein channel blocker is AZD-5423 or a salt thereof and is for use at a daily dose of between about 0.1 mg/day and about 10 mg/day, about 0.2 mg/day and 100 mg/day, and 0.5 mg/day and 150 mg/day.
  • the Zika M protein channel blocker is Streptomycin or a salt thereof and is for use at a daily dose of between about 5 mg/day and about 200 mg/day, about 1 mg/day and 250 mg/day, and 10 mg/day and 500 mg/day.
  • the Zika M protein channel blocker is Zosuquidar or a salt thereof and is for use at a daily dose of between about 10 mg/day and about 500 mg/day, about 1 mg/day and 50 mg/day, and 20 mg/day and 1,000 mg/day.
  • the Zika M protein channel blocker is Dequalinium chloride or a salt thereof and is for use at a daily dose of between about 1 mg/day and about 250 mg/day, about 0.1 mg/day and 50 mg/day, and 2 mg/day and 200 mg/day.
  • the Zika M protein channel blocker is Capreomycin or a salt thereof and is for use at a daily dose of between about 0.1 mg/day and about 20 mg/day, about 1 mg/day and 15 mg/day, and 0.01 mg/day and 10 mg/day.
  • the Zika M protein channel blocker is Pentamidine or a salt thereof and is for use at a daily dose of between about 0.01 mg/day and about 1 mg/day, about 0.1 mg/day and 10 mg/day, and 0.5 mg/day and 20 mg/day.
  • the pharmaceutical composition comprises a pharmaceutically acceptable carrier, adjuvant, or excipient.
  • carrier refers to any component of a pharmaceutical composition that is not the active agent.
  • pharmaceutically acceptable carrier refers to non-toxic, inert solid, semi-solid liquid filler, diluent, encapsulating material, formulation auxiliary of any type, or simply a sterile aqueous medium, such as saline.
  • sugars such as lactose, glucose and sucrose, starches such as corn starch and potato starch, cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt, gelatin, talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil and soybean oil; glycols, such as propylene glycol, polyols such as glycerin, sorbitol, mannitol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate, agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline, Ringer's solution; ethy
  • substances which can serve as a carrier herein include sugar, starch, cellulose and its derivatives, powered tragacanth, malt, gelatin, talc, stearic acid, magnesium stearate, calcium sulfate, vegetable oils, polyols, alginic acid, pyrogen-free water, isotonic saline, phosphate buffer solutions, cocoa butter (suppository base), emulsifier as well as other non-toxic pharmaceutically compatible substances used in other pharmaceutical formulations.
  • Wetting agents and lubricants such as sodium lauryl sulfate, as well as coloring agents, flavoring agents, excipients, stabilizers, antioxidants, and preservatives may also be present.
  • any nontoxic, inert, and effective carrier may be used to formulate the compositions contemplated herein.
  • Suitable pharmaceutically acceptable carriers, excipients, and diluents in this regard are well known to those of skill in the art, such as those described in The Merck Index, Thirteenth Edition, Budavari et al., Eds., Merck & Co., Inc., Rahway, N.J. (2001); the CTFA (Cosmetic, Toiletry, and Fragrance Association) International Cosmetic Ingredient Dictionary and Handbook, Tenth Edition (2004); and the “Inactive Ingredient Guide,” U.S. Food and Drug Administration (FDA) Center for Drug Evaluation and Research (CDER) Office of Management, the contents of all of which are hereby incorporated by reference in their entirety.
  • Examples of pharmaceutically acceptable excipients, carriers and diluents useful in the present compositions include distilled water, physiological saline, Ringer's solution, dextrose solution, Hank's solution, and DMSO. These additional inactive components, as well as effective formulations and administration procedures, are well known in the art and are described in standard textbooks, such as Goodman and Gillman’s: The Pharmacological Bases of Therapeutics, 8th Ed., Gilman et al. Eds. Pergamon Press (1990); Remington’s Pharmaceutical Sciences, 18th Ed., Mack Publishing Co., Easton, Pa.
  • compositions may also be contained in artificially created structures such as liposomes, ISCOMS, slow-releasing particles, and other vehicles which increase the half-life of the peptides or polypeptides in serum.
  • liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers, and the like.
  • Liposomes for use with the presently described peptides are formed from standard vesicle-forming lipids which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol.
  • the selection of lipids is generally determined by considerations such as liposome size and stability in the blood.
  • a variety of methods are available for preparing liposomes as reviewed, for example, by Coligan, J. E. et al, Current Protocols in Protein Science, 1999, John Wiley & Sons, Inc., New York, and see also U.S. Pat. Nos. 4,235,871, 4,501,728, 4,837,028, and 5,019,369.
  • the carrier may comprise, in total, from about 0.1% to about 99.99999% by weight of the pharmaceutical compositions presented herein.
  • a method of screening effectiveness of an agent in treating or preventing a Zika virus infection comprising providing a cell comprising a membrane permeabilized Zika M protein, contacting the cell with the agent, and determining effect of the agent on growth of the cell, wherein a substantial effect of the agent on cellular growth is indicative of the agent as being effective for treating or preventing a Zika virus infection, thereby screening effectiveness of an agent in treating or preventing a Zika virus infection.
  • the method is a negative assay.
  • the cell is characterized by growth retardation due to the membrane permeabilized M protein.
  • an agent that alleviates growth retardation is indicative as being effective for treating or preventing a Zika virus infection.
  • the method is a positive assay.
  • the cell is a K + -uptake deficient cell grown in low [K + ] media experience growth, due to the channel formed by the M protein.
  • an agent that induces growth retardation is indicative as being effective for treating or preventing a Zika virus infection.
  • the method comprises performing both the negative assay and the positive assay.
  • Non-limiting examples for growing a bacterial cell applicable for the screening methods provided herein include: Astrahan, P. et al., Acta 1808, 394-8 (2011); Santner, P. et al. Biochemistry 57, 5949-5956 (2016), and Taube, R., Alhadeff, R., Assa, D., Krugliak, M. & Arkin, I. T. PLoS One 9, el05387 (2014).
  • the assay is for determining susceptibility of the virus to develop resistance against the agent.
  • a length of about 1,000 nanometers (nm) refers to a length of 1,000 nm ⁇ 100 nm.
  • the plasmid was subsequently transformed into ultra-competent cells and the positive clones were confirmed by sequencing. Multiple sequence alignment of the proteins was done using Clustal Omega. The visualization of the alignment was conducted by ESPript 3.0.
  • the cryo-EM structures (PDB id- 6CO8, 5IRE) of ZikV-M were taken as template for generating the model of the transmembrane domain of ZikV-M.
  • the cell pellet was lysed using lysis buffer (40 mM NaCl; 2.7 mM KC1; 10 mM Na2HPO4; 1.8 mM KH 2 PO 4 ; pH 7.4) containing 100 mM PMSF, 100 u/ml Dnase and 0.2 mg/ml lysozyme.
  • the cells were kept at 37 °C for 15 min on a dry bath and then freeze-thawed twice immediately followed by ultrasonication for 5 cycles of 10 seconds with a 10 second gap at 40 watts (Vibra-CellTM Ultrasonic Liquid Processors; Newtown, CT).
  • TBS-T tris-buffered saline containing Tween-20
  • 1% low-fat milk incubated overnight with a primary antibody (Anti-His, New England BioLabs; Ipswich, MA)
  • a primary antibody Anti-His, New England BioLabs; Ipswich, MA
  • a secondary antibody linked to horseradish peroxidase anti-rabbit, Novagen; Temecula, CA
  • Immunoreactive bands were detected using an ECL kit (Biological Industries; Haemek, Israel).
  • a plasmid containing the ZikV-M chimera was transformed into NT326 cells AmalE). An empty pUC-19 plasmid with ampicillin resistance was used as control. Transformed cells were grown on M9 agar plate containing 0.4% maltose as the sole carbon source with 100 pM IPTG for 72 hours at 37 °C.
  • LR1 cells were grown in the same conditions as DH10B cells. After 3 hours of secondary culture, protein expression was induced by adding 100 pM IPTG at 37 °C for 2 hrs. The cells were then washed twice and dissolved in buffer (200 mM Na2HPO4, 0.9% NaCl adjusted to pH 7.6 with 0.1 M citric acid, 0.9% NaCl) to an optical density of 0.25 at 600 nm. 200 pl of dissolved cells were put in 96 well optical black plates (Thermo Scientific, Waltham, MA) with 30 pl buffer (or buffer + Drug/DMSO). 230 pl buffer was used for baseline reduction. Uninduced bacterial culture was used as control.
  • Fluorescent measurements were carried out after adding 70 pl of citric acid (300 mM, 0.9% NaCl) by a liquid handling system (Tecan Mannedorf, Switzerland). The plate reader was set for 390 nm and 466 nm excitation filters paired with a 520 nm emission filter. Emission from wells was read out alternating between two filter pairs for 60 seconds. The proton flow was calculated according to the procedure described previously. All experiments were performed in duplicates or triplicates.
  • Membrane Transporter/Ion Channel Compound Library (HY-L011, MedChem express; Monmouth Junction, NJ) with 372 compounds and Drug Repurposing Compound Library (HY-L035, MedChem Express; Monmouth Junction, NJ) with 2839 compounds were used for the screening.
  • the high throughput screening method was used for the initial screening of compounds by a the Tecan robotic system (Mannedorf, Switzerland).
  • the negative genetic assay was used for the initial screening of compounds.
  • the screening was done in 96 well plates, of which the first and last columns were used for control with 1% DMSO.
  • the bacterial cells without IPTG were taken as negative control while bacteria with 100 pM IPTG were taken as the positive control.
  • NT326 cells that lack a native MBP and consequently are unable to grow on M9 media with maltose as the only carbon source.
  • a plasmid based MBP directs proteins expression to the inter-membrane space and allows the bacteria to grow on M9 maltose.
  • Supplementary figure 2B NT326 cells with MBP-ZikV-M construct were able to grow on M9 media supplemented with 1% maltose after 72 hours at 37 °C. Taken together, the construct results in protein expression and integration in the inner bacterial membrane.
  • the negative assay is based on the fact that elevated levels of protein expression result in growth retardation due to membrane permeabilization by an active viroporin. This assay is well established for the characterization of viroporins in earlier studies. Indeed, ZikV-M when expressed at increasing levels showed commensurate bacterial growth inhibition, as shown in Fig. 1.
  • LB650 cells are K + -uptake deficient bacteria that exhibit retarded growth in low potassium media (/'. ⁇ ?., LB).
  • the ZikV-M construct when transformed into K + -uptake deficient bacteria and upon induction with IPTG, resulted in improved bacterial growth, due most likely to the potassium channel conductivity of ZikV-M. As seen in Fig. 2, the bacterial growth was improved up to 20 pM IPTG. Beyond that, the growth started to slow down due to excess membrane permeabilization, as seen in other viroporins.
  • a fluorescent-based conductivity assay was used to examine if ZikV-M can facilitate proton flow.
  • LR1 bacteria harbor a pH-sensitive green fluorescent protein in their genome. Subsequently, injection of an acidic solution to the media will result in a detectable fluorescence change if the bacteria express a protein capable of proton transport.
  • Residues L69 lies in helix 3, whereas P72 is positioned outside the transmembrane region of the protein. Subsequently, the channel activities of each of the mutants and wild type proteins was examined in each of the three bacteria-based assay as expounded below.
  • the inventors performed the second assay to corroborate the results from the negative genetic assay.
  • ZikV-M mutants were transformed into K + -uptake deficient bacteria and the corresponding bacteria were grown at low potassium media (5 mM) by inducing the channel (10 pM IPTG).
  • the wild-type protein enables the bacteria to thrive.
  • mutants P40A, G54A, Q59A, and V61Stop reduce the ability of bacteria to grow in the aforementioned conditions. This again confirmed the importance of these amino acids to the channel activity of studied viroporin (Figs. 4 and 9A).
  • the inventors subsequently, performed the assay for WNV and DenV MgM to compare once more their behavior with ZikV-M.
  • Fig. 5A To determine the effect of the mutations on proton conductivity the inventors utilized the third, fluorescent-based assay (Fig. 5A). Wild type ZikV-M and all mutants were grown up to three hours and were then induced by 100 pM IPTG for two hours. Uninduced ZikV- M was taken as control. All mutations exhibited less proton flow than the wild-type protein. Mutants G54A, Q59A, and V61-stop, again exhibited less channel activity comparable to uninduced wild type and were much lower than IPTG induced wild type. The result once more pointed at the crucial role of G54, Q59, and helix 3 in channel activity.
  • Fig. 6A ten compounds were able to increase bacterial growth in the negative assay to varying extents: Floxuridine, Nafamostat, Kasugamycin (hydrochloride hydrate), 5-Azacytidine, AZD-5423, Streptomycin (sulfate), Zosuquidar, Dequalinium chloride, Capreomycin (sulfate), and Pentamidine (isethionate).
  • the above ten compounds were subsequently examined in the positive genetic assay in K + -uptake deficient bacteria. In this assay, ZikV-M enhances the growth of the bacteria and therefore, blockers are expected to reduce growth. Gratifyingly, all of the compounds that scored positively in the first assay were able to decrease bacterial growth, with the exception of Nafromastat and Zosuquidar (Fig. 6B).
  • Vero E6 cells were grown to 80% confluency in 96-well plates.
  • the following drugs 5- Azacytidine, Nafamostat, Floxuridine, Kasugamycin, Desogestrel, Spiramycin, AZD-5423, Dabigatran etexillate, Zosuquidar, Streptomycin, Capreomycin, and Pentamidine were added at concentrations of 1 pM, 3 pM, and 10 pM in DMSO with a total DMSO amount of 01.%.
  • Capreomycin and Pentamidine were shown to increase cell viability in all tested concentrations, yet at relatively low % (-5- 20%).
  • Streptomycin (3 and 10 pM) were shown to increase cell viability by -90% and -60%, respectively.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne des compositions pharmaceutiques comprenant un inhibiteur de la protéine canal Zika M et destinées à traiter ou prévenir la virulence du virus Zika chez un sujet. L'invention concerne en outre une composition pharmaceutique comprenant un inhibiteur de la protéine canal Zika M destinée à prévenir l'entrée cellulaire, la décapsidation et/ou la libération hors d'une cellule du virus Zika.
PCT/IL2023/050105 2022-01-30 2023-01-30 Inhibiteurs de la protéine zika m utilisés en tant qu'agents anti-virus zika WO2023144830A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263304627P 2022-01-30 2022-01-30
US63/304,627 2022-01-30

Publications (1)

Publication Number Publication Date
WO2023144830A1 true WO2023144830A1 (fr) 2023-08-03

Family

ID=85278273

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2023/050105 WO2023144830A1 (fr) 2022-01-30 2023-01-30 Inhibiteurs de la protéine zika m utilisés en tant qu'agents anti-virus zika

Country Status (1)

Country Link
WO (1) WO2023144830A1 (fr)

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5019369A (en) 1984-10-22 1991-05-28 Vestar, Inc. Method of targeting tumors in humans
WO2003092616A2 (fr) 2002-05-01 2003-11-13 The Cleveland Clinic Foundation Compositions therapeutiques constituees de pentamidine et methodes d'utilisation de ces dernieres pour traiter le cancer
WO2007008490A2 (fr) 2005-07-06 2007-01-18 Kanisa Pharmaceuticals, Inc. Zosuquidar, daunorubicine et cytarabine pour le traitement du cancer
WO2007130743A2 (fr) 2006-05-01 2007-11-15 Array Biopharma, Inc. Dérivés de capréomycine, et leur utilisation en tant qu'agents antibactériens
CN102532232A (zh) 2010-12-27 2012-07-04 上海信谊药厂有限公司 去氧孕烯的制备方法
WO2014007326A1 (fr) 2012-07-04 2014-01-09 味の素株式会社 Procédé de préparation de mésylate de nafamostat
CN106854179A (zh) 2016-12-26 2017-06-16 华润双鹤药业股份有限公司 地喹氯铵及其类似物的制备方法
US20180015153A1 (en) * 2016-07-16 2018-01-18 Florida State University Research Foundation, Inc. Compounds and methods for treatment and prevention of flavivirus infection

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US5019369A (en) 1984-10-22 1991-05-28 Vestar, Inc. Method of targeting tumors in humans
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
WO2003092616A2 (fr) 2002-05-01 2003-11-13 The Cleveland Clinic Foundation Compositions therapeutiques constituees de pentamidine et methodes d'utilisation de ces dernieres pour traiter le cancer
WO2007008490A2 (fr) 2005-07-06 2007-01-18 Kanisa Pharmaceuticals, Inc. Zosuquidar, daunorubicine et cytarabine pour le traitement du cancer
WO2007130743A2 (fr) 2006-05-01 2007-11-15 Array Biopharma, Inc. Dérivés de capréomycine, et leur utilisation en tant qu'agents antibactériens
CN102532232A (zh) 2010-12-27 2012-07-04 上海信谊药厂有限公司 去氧孕烯的制备方法
WO2014007326A1 (fr) 2012-07-04 2014-01-09 味の素株式会社 Procédé de préparation de mésylate de nafamostat
US20180015153A1 (en) * 2016-07-16 2018-01-18 Florida State University Research Foundation, Inc. Compounds and methods for treatment and prevention of flavivirus infection
CN106854179A (zh) 2016-12-26 2017-06-16 华润双鹤药业股份有限公司 地喹氯铵及其类似物的制备方法

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
"CTFA (Cosmetic, Toiletry, and Fragrance Association) International Cosmetic Ingredient Dictionary and Handbook", 2004
"GenBank", Database accession no. YP_009227197.1
"Goodman and Gillman's: The Pharmacological Bases of Therapeutics", 1990, MACK PUBLISHING CO.
"Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
"The Merck Index", 2001, MERCK & CO., INC.
ARIAS-ARIAS JORGE L. ET AL: "In Vitro Inhibition of Zika Virus Replication with Amantadine and Rimantadine Hydrochlorides", MICROBIOLOGY RESEARCH, vol. 12, no. 3, 15 September 2021 (2021-09-15), pages 727 - 738, XP093043304, DOI: 10.3390/microbiolres12030052 *
ASTRAHAN, P. ET AL., ACTA, vol. 1808, 2011, pages 394 - 8
BROWN EMMA ET AL: "Inhibitors of the Small Membrane (M) Protein Viroporin Prevent Zika Virus Infection", BIORXIV, 12 March 2021 (2021-03-12), XP093043311, Retrieved from the Internet <URL:https://www.biorxiv.org/content/10.1101/2021.03.11.435022v1.full.pdf> [retrieved on 20230501], DOI: 10.1101/2021.03.11.435022 *
BROWN EMMA TABITHA: "Zika virus M protein as a viroporin drug target", 1 August 2020 (2020-08-01), XP093043306, Retrieved from the Internet <URL:https://etheses.whiterose.ac.uk/28649/1/Thesis%20with%20corrections%2007Jan21C.pdf> [retrieved on 20230501] *
CAS, no. 167354-41-8
COLIGAN, J. E. ET AL.: "Current Protocols in Protein Science", 1999, JOHN WILEY & SONS
SANTNER, P. ET AL., BIOCHEMISTRY, vol. 57, 2018, pages 5949 - 5956
TAUBE, R.ALHADEFF, R.ASSA, D.KRUGLIAK, M.ARKIN, I. T., PLOS ONE, vol. 9, 2014, pages e105387
TOMAR PST ET AL: "Zika M-A Potential Viroporin: Mutational Study and Drug Repurposing", BIOMEDICINES, vol. 10, no. 3, 10 March 2022 (2022-03-10), pages 641, XP093043416, DOI: 10.3390/biomedicines10030641 *
WANG LILI ET AL: "Development of Small-Molecule Inhibitors Against Zika Virus Infection", FRONTIERS IN MICROBIOLOGY, vol. 10, no. 2725, 6 December 2019 (2019-12-06), Lausanne, XP093043307, ISSN: 1664-302X, DOI: 10.3389/fmicb.2019.02725 *

Similar Documents

Publication Publication Date Title
US20200368343A1 (en) Zika virus mrna vaccines
AU2007293239B2 (en) Method of treating peripheral nerve disorders
TW580387B (en) Pharmaceutical combination comprising tipranavir and ritonavir
US7488800B2 (en) Apoptosis-inducing polypeptides
US11364292B2 (en) CHIKV RNA vaccines
CN112023045A (zh) 抑制masp-2及其下游补体活化效应的物质在制备冠状病毒所致疾病药物中的应用
Lundstrom et al. COVID-19 signalome: Potential therapeutic interventions
WO2023144830A1 (fr) Inhibiteurs de la protéine zika m utilisés en tant qu&#39;agents anti-virus zika
KR20230035524A (ko) Covid-19 치료제로서 e 단백질 채널 차단제 및 orf3 억제제
US20220211810A1 (en) Method of Treatment
US20230190769A1 (en) E protein channel blockers and orf3 inhibitors as anti-covid-19 agents
US20060094679A1 (en) Method of preventing or treating liver disease
US20230330089A1 (en) Covid prevention and treatment using an integrin inhibitor
WO2024009307A1 (fr) Bloqueurs/inhibiteurs de viroporines en tant qu&#39;agents anti-flavivirus
JP4149713B2 (ja) 感染症治療剤
WO2022113069A1 (fr) Bloqueurs de canal de la protéine e et inhibiteurs de l&#39;orf3 utilisés en tant qu&#39;agents anti-covid-19
JP2009533478A (ja) 黄色ブドウ球菌感染症の治療用薬剤の製造のための活性成分としてtrapタンパク質自体の使用
TWI737972B (zh) 心肌再灌流的方法及套组與減緩或減少心肌再灌流損傷的方法
US20230355711A1 (en) Vimentin and fragments or derivatives thereof for coronavirus treatment and prevention
JP2007153837A (ja) C型肝炎ウイルス粒子形成・細胞外放出を抑制する方法
CN115770292A (zh) 新型冠状病毒SARS-CoV-2的协同免疫及其用途
RU2265439C2 (ru) Лечение инфекции вируса иммунодефицита человека с лекарственной устойчивостью
KR20020073494A (ko) 약물내성 인체 면역결핍 바이러스 감염의 치료
KR20090121428A (ko) 씨형간염 바이러스 중합효소의 작용을 저해하는 화합물

Legal Events

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

Ref document number: 23705684

Country of ref document: EP

Kind code of ref document: A1