US20210322568A1 - Virucidal Nanoparticles And Use Thereof Against Influenza Virus - Google Patents

Virucidal Nanoparticles And Use Thereof Against Influenza Virus Download PDF

Info

Publication number
US20210322568A1
US20210322568A1 US17/273,113 US201917273113A US2021322568A1 US 20210322568 A1 US20210322568 A1 US 20210322568A1 US 201917273113 A US201917273113 A US 201917273113A US 2021322568 A1 US2021322568 A1 US 2021322568A1
Authority
US
United States
Prior art keywords
virucidal
sln
ligands
nanoparticle
nanoparticles
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
US17/273,113
Other languages
English (en)
Inventor
Caroline TAPPAREL VU
Valeria Cagno
Özgün KOCABIYIK
Francesco Stellacci
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.)
Ecole Polytechnique Federale de Lausanne EPFL
Original Assignee
Ecole Polytechnique Federale de Lausanne EPFL
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 Ecole Polytechnique Federale de Lausanne EPFL filed Critical Ecole Polytechnique Federale de Lausanne EPFL
Assigned to ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE (EPFL) reassignment ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE (EPFL) ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: STELLACCI, FRANCESCO, CAGNO, Valeria, TAPPAREL VU, Caroline, KOCABIYIK, OZGUN
Publication of US20210322568A1 publication Critical patent/US20210322568A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/61Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule the organic macromolecular compound being a polysaccharide or a derivative thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6923Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being an inorganic particle, e.g. ceramic particles, silica particles, ferrite or synsorb
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • A61K47/6939Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being a polysaccharide, e.g. starch, chitosan, chitin, cellulose or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6949Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit inclusion complexes, e.g. clathrates, cavitates or fullerenes
    • A61K47/6951Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit inclusion complexes, e.g. clathrates, cavitates or fullerenes using cyclodextrin
    • 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

Definitions

  • the invention relates to virucidal nanoparticles comprising a trisaccharide moiety and the use thereof against influenza virus.
  • Influenza viruses are among the most infective viruses. Every year different influenza strains infect a large fraction of both the animal and human population, endangering infants, the elderly and immunocompromised people, all having a risk of hospitalization and death due to influenza-related complications. As a result, seasonal influenza poses remarkable impacts on socio-economy. In fact, respiratory diseases can cost a significant fraction of the total health expenditures in developed and mainly in developing countries. Because influenza mutates so rapidly, the development of a vaccine is still a major challenge. Vaccine development would pose even higher challenges when focused on the occasional pandemics instead of yearly outbreaks. In such case, the development time of a new vaccine, which is on average 6 months, would represent a serious risk. Furthermore, even in the presence of a vaccine, reaching a reasonable vaccination coverage is far from a foregone conclusion. Therefore, the risk of a new pandemic, such as the Spanish-flu, is still present and recognised as one of the top threats to global health.
  • antiviral drugs Naturally, the second line of defence after vaccines, are antiviral drugs.
  • a number of anti-influenza drugs are currently approved: neuraminidase inhibitors such as zanamivir and oseltamivir, ion channel inhibitors such as amantadine, fusion inhibitors such as umifenovir (only in Russia and China) and polymerase inhibitor such as baloxavir marboxil, which was recently approved in US and Japan.
  • neuraminidase inhibitors such as zanamivir and oseltamivir
  • ion channel inhibitors such as amantadine
  • fusion inhibitors such as umifenovir (only in Russia and China)
  • polymerase inhibitor such as baloxavir marboxil
  • the interaction between the viral hemagglutinin (I-TA) and the sialic acid (SA)-bearing glycoproteins on host-cells is the primary step of influenza infection.
  • the binding affinity between the SA and HA is low and compensated for by multivalent binding.
  • SA coated multivalent materials such as polymers, dendrimers and nanoparticles.
  • NPs gold nanoparticles
  • Micromolar material concentrations to inhibit the influenza virus in vitro are still too high and in vivo concentrations will be even higher. Also, most of the entry inhibitors are virustatic; the virus becomes infective again upon dilution of virus-material complex in vitro.
  • An ideal anti-influenza drug should be broad-spectrum, target a highly conserved part of the virus, have an irreversible effect, i.e. be virucidal (in order to avoid loss of efficacy due to the dilution in body fluids) at low concentrations, and obviously be non-toxic.
  • the present invention provides nanoparticles that strongly interact with the HA, irreversibly inhibit the infectivity of influenza virus at low concentrations, and display exceedingly-low toxicity.
  • An aspect of the present invention provides a virucidal nanoparticle comprising a core and a plurality of ligands covalently linked to the core, wherein at least a portion of said ligands comprise a trisaccharide moiety and wherein:
  • Another aspect of the present invention provides a virucidal nanoparticle represented by Formula (I)
  • Another aspect of the present invention provides a virucidal nanoparticle represented by Formula (II)
  • a further aspect of the present invention provides a pharmaceutical composition comprising an effective amount of one or more virucidal nanoparticles of the present invention and at least one pharmaceutically acceptable excipient, carrier and/or diluent.
  • a further aspect of the present invention provides the virucidal nanoparticle of the present invention for use in treating and/or preventing influenza virus infections and/or diseases associated with influenza viruses.
  • a further aspect of the present invention provides a virucidal composition
  • a virucidal composition comprising an effective amount of one or more virucidal nanoparticles of the present invention and optionally at least one suitable aerosol carrier.
  • a further aspect of the present invention provides a method of disinfection and/or sterilization comprising using the virucidal compositions of the present invention, or a virucidal nanoparticle of the present invention.
  • a further aspect of the present invention provides a device comprising the virucidal compositions of the present invention, or one or more virucidal nanoparticles of the present invention and means for applying or dispensing the virucidal compositions or the virucidal nanoparticles.
  • a further aspect of the present invention provides a use of the virucidal nanoparticles of the present invention or the virucidal compositions of the present inventions for sterilization and/or for disinfection.
  • FIG. 1 shows chemical structure of NPs with different ligand composition. Average NP diameter: 2.9 ⁇ 0.9 nm.
  • FIG. 2 shows TEM images of H3N2 virus Vic/11 strain: without NPs (A), the virus after 1 hour incubation with LD 6′SLN NPs (B) and with PEG(5) NPs (C).
  • FIG. 3 shows chemical structure of C15-6′SLN modified ⁇ -CD (A). EC 50 concentrations of the modified CD against different influenza strains (B). Virucidal activity against Neth/09 strain (C) ex vivo experiment on the MucilAir (D). Illustration of modified CD interacting with HA globular head (E).
  • FIG. 4 shows exemplary modified cyclodextrins.
  • Number of 6′SLN or 3′SLN per ⁇ -CD is the average number calculated by 1 H NMR.
  • the representative chemical structures of modified cyclodextrins were constructed based on NMR results.
  • EC 50 represents the half-inhibitory concentrations on MDCK cells at 24 hpi against A/Netherlands/2009 (H1N1) ( FIG. 7 ).
  • N/A not assessable.
  • some of the rearward cyclodextrin sugars, ligands and trisaccharides are not shown.
  • FIG. 5 shows 1 H NMR studies were conducted in order to characterize the modified cyclodextrins shown in FIG. 4 .
  • Average number of 6′SLN or 3′SLN per ⁇ -cyclodextrin was calculated by comparing a distinctive peak from the trisaccharide ( ⁇ ) to the one from ⁇ -cyclodextrin (•). Both peaks correspond to a single hydrogen.
  • FIG. 6 shows dose-response curves demonstrating the antiviral activity of C6-6′ (A), C14-6′ (B), C11-3′ (C) and C1-6′ (D) against A/Netherlands/2009 H1N1. The results are the mean of 2 independent experiments performed in duplicates.
  • FIG. 7 shows dose-response curves demonstrating the antiviral activity of C11-6′ against B/Wisconsin/2010 (A), A/Clinical/2018 H1N1 (B), A/Singapore/2004 H3N2 (C) and B/Clinical/2018 (D). The results are the mean of 2 independent experiments performed in duplicates.
  • FIG. 8 shows dose-response curves demonstrating the antiviral activity of C11-3′ against avian strains A/turkey/Turkey/2005 H5N1 (together with C11-6′ control) (A) and A/Turkey/Italy/977/1999 H7N1 (B).
  • graph A The infection was quantified with both FACS and ICC methods. The results are the mean of 2 independent experiments performed in duplicates.
  • FIG. 9 shows antiviral activity comparison of C11-6′ and P8-6′ in vitro.
  • Panels (a) and (b) show on the left graphs the inhibitory activity of each compound against A/NL/09, superimposed with the results of the cell viability assays. All the compounds show very similar behaviour.
  • On the right graphs in these panels are shown the results of virucidal (i.e. dilution) test. Note that in the figure's axes ffu stands for focus forming units and NT for non-treated.
  • C11-6′ was tested against the following viral strains: A/Singapore/2004 (H3N2), B/Wisconsin/2010, and A/Clinical/2018 (H1N1). Results are the mean and SEM of 2 independent experiments performed in duplicate.
  • FIG. 10 shows virucidal activity of the C14-6′ (A), C6-6′ (B) and C11-3′ (C) against A/Netherhands/2009 (H1N1).
  • the experiments were performed with a compound concentration of 100 ⁇ g/mL.
  • the results are the mean and SEM of 2 independent experiments.
  • FIG. 11 shows ex vivo inhibitory activity comparison of C11-6′ and P8-6′.
  • C11-6′ provided a full protection against clinical pandemic H1N1 09 strain in co-treatment condition, whereas P8-6′ only provided a minor protection in the beginning of the infection (a).
  • FIG. 11( b ) immunofluorescence at 7 days post-infection (co-treatment condition) confirms the protection provided by C11-6′. Red: monoclonal antibody Influenza A, blue: DAPI, green: ⁇ -IV-tubulin (marker of ciliated cells). The thickness of each tissue was demonstrated at the bottom of the corresponding image (b).
  • C11-6′ also showed high efficacy in post-treatment condition (c). Results of (a) and (c) are mean and SEM of 2 to 4 independent experiments performed in duplicate. Images of (b) are representative of 10 images taken for each condition.
  • FIG. 12 shows LDH release from infected tissues. Tissues were infected and treated with C11-6′ (50 ⁇ g) or P8-6′ at the time of infection. Apical washes performed at 96 and 24 hpi were subjected to LDH measurement. Results are the mean and SEM of 2 independent experiments for H1N1 and H1N1 C11-6′ and of a single experiment performed in duplicate for P8-6′.
  • FIG. 13 shows long co-treatment experiment. Tissues were infected and treated with C11-6′ (50 ⁇ g) at the time of infection. Daily apical washes were collected for the first 5 days, and subsequently at 9, 17 and 23 days, with a wash the previous day in order to evaluate daily virus production. Results are the mean of a single experiment performed in duplicate.
  • FIG. 14 shows ex vivo toxicity.
  • Tissues were treated with different doses of C11-6′ or an equal volume of medium or triton 5% with daily addition.
  • tissues were subjected to: A) MTT assay, B) LDH assay in which the viability of the tissues was evaluated, C) trans epithelial resistance evaluation and D) ELISAs assay to evaluate the release of pro-inflammatory cytokines.
  • LDH and ELISA were performed on collected basal medium. The experiments are the mean and SEM of 2 independent experiments.
  • FIG. 15 shows in vivo antiviral activity of C11-6′.
  • (a to c) Mice were intra-nasally treated with PBS or C11-6′ simultaneously and 48-hours post-infection with A/NL/09. Viral loads were quantified 48- and 96-hours post-infection (a) and morbidity (loss of temperature (b) and weight (c)) of infected mice was monitored daily.
  • (d to 0 Mice were intra-nasally treated with PBS or C11-6′ 6-hours post-infection and daily for 3 days (14 ⁇ g/mouse). Morbidity (d-e) and survival (f) of infected mice were monitored daily. Results are expressed as means ⁇ SEM. Arrows indicate the treatment times.
  • FIG. 16 shows stacked 1 H NMR specta of ⁇ -clycodextrin, 6′SLN- ⁇ -ethylamine and C11-6′.
  • FIG. 17 shows DOSY spectrum of the C11-6′ demonstrating that the resulting compound is free from unbound trisaccharides.
  • FIG. 18 shows the gating strategy performed for FACS.
  • the term “at least one” used in a phrase such as “at least one C atom” can mean “one C atom” or “two C atoms” or more C atoms.
  • virustatic refers to a characterization of antiviral efficacy determined by in vitro testing demonstrating reversible inhibition of the infectivity of a virus following interaction with an antiviral composition. The interaction inhibits infectivity, for example, by binding to the virus or otherwise interfering with the virus' surface ligands.
  • the term “virucidal” refers to a characterization of antiviral efficacy determined by in vitro testing demonstrating irreversible inhibition of the infectivity of a virus following interaction with an antiviral compound or composition.
  • the interaction inhibits infectivity, for example, by binding to the virus or otherwise interfering with the virus' surface ligands.
  • termination of the interaction for example, by dilution
  • biocompatible refers to compatibility with living cells, tissues, organs, or systems, and having no significant risk of injury, toxicity, or rejection by the immune system.
  • nano such as used in “nanoparticle” refers to nanometric size, such as a particle having a nanometric size, and is not intended to convey any specific shape limitation.
  • nanoparticle encompasses nanospheres, nanotubes, nanoboxes, nanoclusters, nanorods and the like.
  • the nanoparticles and/or nanoparticle cores contemplated herein have a generally polyhedral or spherical geometry.
  • influenza refers to sialic acid-seeking, airborne transmissible (human or animal) RNA viruses, such as influenza A virus, influenza B virus, influenza C virus and influenza D virus.
  • Influenza A virus encompasess the following serotypes: H1N1, H2N2, H3N2, H5N1, H7N7, H1N2, H9N2, H7N2, H7N3, H10N7, H7N9, and H6N1.
  • alkyl refers to a straight hydrocarbon chain containing from 1 to 50 carbon atoms, preferably 4 to 30 carbon atoms.
  • Representative examples of alkyl include, but are not limited to methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, . . . .
  • carboxyalkyl refers to a carboxy group appended to the parent molecular moiety through an alkyl group as defined herein.
  • An aspect of the present invention provides a virucidal nanoparticle comprising a core and a plurality of ligands covalently linked to the core, wherein at least a portion of said ligands comprise a trisaccharide moiety and wherein
  • the trisaccharide moiety is exposed on a ligand covalently bound to the outer surface of a nanoparticle (NP) in a manner so that any other ligands do not hinder the interaction between the trisaccharide moiety and the influenza virus.
  • NP nanoparticle
  • the mean diameter of the core ranges from about 1.0 nm to about 200 nm, preferably from 1 nm to 5 nm, most preferably from 1.5 nm to 3 nm.
  • the overall nanoparticle size has a mean particle diameter of from 3 nm to 250 nm, or from 3 nm to 200 nm, preferably from 3 nm to 10 nm, more preferably from 4.5 nm to 6 nm.
  • the core in virucidal nanoparticles of the invention is organic material, preferably a polymer, wherein a polymer is selected from the group comprising polyacrylic acid (PAA), polyvinyl alcohol (PVA), polyethylene glycol (PEG), polylactic acid (PLA), polyglycolide (PGA), polydioxanone (PDO), and poly(lactic-co-glycolic acid).
  • PAA polyacrylic acid
  • PVA polyvinyl alcohol
  • PEG polyethylene glycol
  • PLA polylactic acid
  • PGA polyglycolide
  • PDO polydioxanone
  • poly(lactic-co-glycolic acid) lactic-co-glycolic acid
  • the core in virucidal nanoparticles of the invention is organic material, preferably a dendrimer selected from the group comprising poly(amidoamine) (PAMAM) and bis-MPA.
  • PAMAM poly(amidoamine)
  • bis-MPA bis-MPA
  • the core in virucidal nanoparticles of the invention is organic material, preferably a dendron selected from the group comprising poly(amidoamine) (PAMAM) and bis-MPA.
  • PAMAM poly(amidoamine)
  • bis-MPA bis-MPA
  • the core in virucidal nanoparticles of the invention is organic material which is cyclodextrin.
  • Cyclodextrins are naturally occurring cyclic glucose derivatives consisting of alpha(14)-linked glucopyranoside units. Their cyclic structure creates a truncated cone shape with the primary hydroxyls of the glucose units on the narrow face and the secondary hydroxyls on the wider face. Each face can be readily and independently functionalised.
  • the most commonly used natural CDs have 6, 7, and 8 glucopyranoside units, referred to as alpha, beta and gamma cyclodextrin respectively.
  • the preferred cyclodextrin is beta. Because of the cyclic structure of CDs, they have a cavity capable of forming supramolecular inclusion complexes with guest molecules.
  • CDs are naturally occurring, readily functionalised, have a cavity for guest inclusion and are biocompatible, they have found use in many commercial applications including drug delivery, air fresheners, etc.
  • the difference in reactivity of each face of CDs has been used for the synthesis of a wide range of modified cyclodextrins.
  • the primary face of CDs is more readily modified, with control over the degree and location of substitution being possible.
  • CD derivatives that bear a good leaving group, such as halogenated CDs are important intermediates in CD functionalisation.
  • heptakis-6-iodo-6-deoxy-beta-cyclodextrin was synthetized followed by reaction with mercaptoundecaosulphonate (MUS) to yield a CD functionalised on the primary face with undecanaosulfonate groups. It is then possible to independently modify the secondary face of the cyclodextrin to introduce further solubilising groups, dye molecules, polymers, etc.
  • MUS mercaptoundecaosulphonate
  • the size of ⁇ -CD falls within the preferred nano size for cores of the invention and matches well with the HA globular head ( ⁇ 5 nm).
  • Beta-cyclodextrin has a rigid chemical structure that is believed to contribute to virucidal activity, and can have maximum of 7 trisaccharide-bearing ligands depending from the narrow face, preferably 3 to 4 trisaccharide-bearing ligands, three being the number of sialic acid binding points in the influenza virus HA globular head.
  • the virucidal nanoparticles of the present invention can also be purified single molecules or compounds which are also intended to be encompassed within the scope of the present invention.
  • An embodiment of the present invention provides a virucidal nanoparticle comprising a core and a plurality of ligands covalently linked to the core, wherein at least a portion of said ligands comprise a trisaccharide moiety and wherein
  • some or all of the ligands comprise 3′SLN, some or all of said ligands comprise 6′SLN, and some but not all of said ligands comprise no trisaccharide moiety.
  • a virucidal nanoparticle of the present invention, wherein the core is cyclodextrin, can be represented by Formula (I)
  • Another aspect of the present invention provides a virucidal nanoparticle represented by Formula (II)
  • the polymer in the virucidal nanoparticles of the invention can be selected from both synthetic and natural polymers.
  • the synthetic polymers are selected from the group comprising, but not limited to, poly(ethylene glycol) (PEG), poly(vinyl alcohol) (PVA), poly(acrylamide) (PAAm), poly(n-butyl acrylate), poly-( ⁇ -esters), (PEG-b-PPO-b-PEG), poly(N-isopropylacrylamide) (pNIPAAM), polylacticglycolic acid (PLGA) and/or combinations thereof.
  • the natural polymers are selected from the group comprising dextran, dextrins, glucose, cellulose and/or combinations thereof.
  • the trisaccharide moiety is preferably 6′SLN, which is specific to human influenza strains. In other embodiments of virucidal nanoparticles of the invention, the trisaccharide moiety is preferably 3′SLN, which is specific to avian influenza strains.
  • the ligands (or ligand compounds) of virucidal nanoparticles of the invention are typically sufficiently long (at least 2, or at least 4 or at least 6 carbon atoms) and hydrophobic.
  • the optionally substituted alkyl-based ligands are selected from the group comprising hexane-, pentane-, octane-, undecane-, hexadecane-based ligands.
  • Substituted alkyl-based ligands, substituted C 4 -C 30 alkyl based ligands, and substituted C 4 -C 30 carboxyalkyls of virucidal nanoparticles of the present invention are substituted with 1, 2, 3, 4 or 5 substituents independently selected from the group comprising alkenyl, alkenylthio, alkenyloxy, alkoxy, alkoxyalkoxy, alkoxyalkoxyalkoxy, alkoxyalkoxyalkyl, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkoxy, alkoxycarbonylalkyl, alkoxysulfonyl, alkyl, alkylamidoalkyl, alkylcarbonyl, alkylcarbonylalkoxy, alkylcarbonylalkyl, alkylcarbonylalkylthio, alkylcarbonyloxy, alkylcarbonylthio, alkylsulfinyl,
  • substituted alkyl-based ligands Preferably substituted alkyl-based ligands, substituted C 4 -C 30 alkyl-based ligands, and substituted C 4 -C 30 carboxyalkyls are substituted with one mercapto group (replacing the corresponding oxygen of the unmodified cyclodextrin).
  • Preferred substituted alkyl-based ligand is alkylamidoalkyl substituted C 4 -C 30 or C 2 to C 28 or C 4 to C 13 alkyl-based ligand.
  • the percentage ratio between the ligands and the ligands comprising a trisaccharides moiety is of 75%:25% to 95%:5%; preferably 88%:12%.
  • plurality of ligands refers to a virucidal nanoparticle core that is coated, partially or completely, by a plurality of ligands of the invention, wherein at least a portion of said ligands comprise a trisaccharides moiety of the invention.
  • the coating can be homogenous, unstructured or structured.
  • the virucidal nanoparticle comprises very high density (HD) of ligands comprising trisaccharide moiety of the invention, for example 25% of total ligands.
  • HD very high density
  • the virucidal nanoparticle comprises about 2 to about 5 ligands of the invention per nm 2 , wherein at least a portion of said ligands comprise a trisaccharide moiety. In other embodiments, the virucidal nanoparticle comprises four ligands of the invention per nm 2 , wherein at least a portion of such ligands comprise a trisaccharide moiety.
  • the plurality of ligands of the invention comprises a mixture of at least two structurally different ligands, such as polyethylene glycol 5 (PEG(5)) and 16-mercaptohexadeconic acid (C15).
  • PEG(5) polyethylene glycol 5
  • C15 16-mercaptohexadeconic acid
  • the mixture can advantageously be organized so that the ligands bearing no trisaccharide moiety provide optimal spacing for the ligands that do bear a trisaccharide moiety and do not hinder the interactions between the trisaccharide moieties and the influenza viral HA.
  • a percentage ratio will exist between ligands not bearing versus those bearing a trisaccharide moiety, ranging from about 75:25 to about 95:5, preferably about 88:12.
  • PEG(5) & 16-mercaptohexadeconic acid (C15) mixed ligand gold nanoparticles (NPs) have been synthesized.
  • PEG(5) enhances water solubility of NPs whereas C15 is the ligand to graft the triscahharide.
  • the ligand choice was based on two reasons: 1) C15 is sufficiently long to target three sialic acid binding points, that are ⁇ 4 nm apart, on the HA, 2) Carbon based rigid ligand enhances the virucidal activity.
  • the ligands are generally present on the surface of the core in an amount that optimizes binding of the trisaccaharide moieties to the influenza hemagglutinin.
  • the core has typically four ligands per nm 2 . In some embodiments, the core has about 2 to about 5 ligands per nm 2.
  • virucidal nanoparticles of the invention Important advantages of the virucidal nanoparticles of the invention is that detailed toxicity analysis did not show any alteration of the tissue structure nor release of pro-inflammatory cytokines. In vivo tests showed that the treatment with the virucidal nanoparticles of the invention significantly improved the health condition of infected mice and reduced the viral load in the lungs, independently of the addition of the drug before or after infection.
  • Another aspect of the invention discloses a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of one or more virucidal nanoparticles of the invention and at least one pharmaceutically acceptable excipient, carrier and/or diluent.
  • pharmaceutically acceptable carrier, excipient and/or diluent means a carrier, excipient or diluent that is useful in preparing a pharmaceutical composition that is generally safe, and possesses acceptable toxicities.
  • Acceptable carriers, excipients or diluents include those that are acceptable for veterinary use as well as human pharmaceutical use.
  • a “pharmaceutically acceptable carrier, excipient and/or diluent” as used in the specification and claims includes both one and more than one such carrier, excipient and/or diluent.
  • the pharmaceutical composition of the present invention further comprises one or more additional active agents, preferably anti-viral agents.
  • virucidal nanoparticles of the invention that are used in the methods of the present invention can be incorporated into a variety of formulations and medicaments for therapeutic administration. More particularly, a virucidal nanoparticle as provided herein can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers, excipients and/or diluents, and can be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, pills, powders, granules, dragees, gels, slurries, ointments, solutions, suppositories, injections, inhalants and aerosols.
  • administration of the virucidal nanoparticles can be achieved in various ways, including oral, buccal, inhalation (pulmonary, nasal), rectal, parenteral, intraperitoneal, intradermal, transdermal, intracranial and/or intratracheal administration.
  • the virucidal nanoparticles can be administered in a local rather than systemic manner, in a depot or sustained release formulation.
  • the virucidal nanoparticles can be formulated with common excipients, diluents or carriers, and compressed into tablets, or formulated as elixirs or solutions for convenient oral administration, or administered by the intramuscular or intravenous routes.
  • the virucidal nanoparticles can be administered transdermally, and can be formulated as sustained release dosage forms and the like.
  • the virucidal nanoparticles can be administered alone, in combination with each other, or they can be used in combination with other known compounds.
  • Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences (Mack Publishing Company (1985) Philadelphia, Pa., 17th ed.), which is incorporated herein by reference.
  • Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences (Mack Publishing Company (1985) Philadelphia, Pa., 17th ed.), which is incorporated herein by reference.
  • Langer, Science (1990) 249:1527-1533 which is incorporated herein by reference.
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semi permeable matrices of solid hydrophobic polymers containing the virucidal nanoparticles of the invention, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and [gamma] ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-( ⁇ )-3-hydroxybutyric acid.
  • the virucidal nanoparticles of the present invention may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • macroemulsions for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • compositions described herein can be manufactured in a manner that is known to those of skill in the art, i.e., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • the following methods and excipients are merely exemplary and are in no way limiting.
  • a virucidal nanoparticle (and optionally another active agent) can be formulated into preparations by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • an aqueous or nonaqueous solvent such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol
  • solubilizers isotonic agents
  • suspending agents emulsifying agents, stabilizers and preservatives.
  • the virucidal nanoparticles of the present invention can be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • pharmaceutical formulations for parenteral administration include aqueous solutions of the virucidal nanoparticles in water-soluble form.
  • suspensions of the virucidal nanoparticles can be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions can contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension can also contain suitable stabilizers or agents that increase the solubility of the virucidal nanoparticles to allow for the preparation of highly concentrated solutions.
  • the amount of a virucidal nanoparticle of the invention that can be combined with a carrier material to produce a single dosage form will vary depending upon the viral disease treated, the mammalian species, and the particular mode of administration. It will be also understood, that the specific dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed; the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion; other drugs that have previously been administered; and the severity of the particular viral disease undergoing therapy, as is well understood by those of skill in the area.
  • Further aspect of the invention provides a method of treating and/or preventing influenza virus infections and/or diseases associated with influenza viruses, comprising administering to a subject in need thereof, a therapeutically effective amount of one or more virucidal nanoparticles of the invention.
  • Another aspect of the invention provides the virucidal nanoparticles of the invention for use in treating and/or preventing influenza virus infections and/or diseases associated with influenza viruses.
  • the terms “subject” or “patient” are well-recognized in the art, and, are used interchangeably herein to refer to a mammal, including dog, cat, rat, mouse, monkey, cow, horse, goat, sheep, pig, camel, and, most preferably, a human. Other animals, such as a chicken, are also encompassed by these terms.
  • the terms “subject” or “patient” refer to a human and animals, such as dog, cat, rat, mouse, monkey, cow, horse, goat, sheep, pig, camel, chicken.
  • the subject is a subject in need of treatment or a subject being infected by an influenza virus.
  • a subject can be an animal infected by avian influenza, such as a chicken.
  • the subject can be a healthy subject or a subject who has already undergone treatment.
  • the term does not denote a particular age or sex. Thus, adult, children and newborn subjects, whether male or female, are intended to be covered.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already being infected by an influenza virus, as well as those in which the influenza viral infection is to be prevented. Hence, the mammal, preferably human, to be treated herein may have been diagnosed as being infected by an influenza virus, or may be predisposed or susceptible to be infected by an influenza virus. Treatment includes ameliorating at least one symptom of, curing and/or preventing the development of a disease or condition due to influenza viral infection. Preventing is meant attenuating or reducing the ability of an influenza virus to cause infection or disease, for example by affecting a post-entry viral event.
  • “Mammal” for purposes of treatment refers to any animal classified as a mammal, including humans, domestic and farm animals or pet animals, such as dogs, horses, cats, cows, monkeys etc. Preferably, the mammal is human.
  • terapéuticaally effective amount refers to an amount of a virucidal nanoparticle of the invention effective to alter an influenza virus, and to render it inert, in a recipient subject, and/or if its presence results in a detectable change in the physiology of a recipient subject, for example ameliorates at least one symptom associated with a viral infection, prevents or reduces the rate transmission of at least one viral agent.
  • a virucidal composition comprising an effective amount of one or more virucidal nanoparticles of the invention and optionally at least one suitable carrier or aerosol carrier.
  • “An effective amount” refers to the amount sufficient for irreversibly inhibiting influenza viruses; i.e. sufficient for obtaining virucidal effect.
  • the suitable carrier is selected from the group comprising stabilisers, fragrance, colorants, emulsifiers, thickeners, wetting agents, or mixtures thereof.
  • the virucidal composition can be in the form of a liquid, a gel, a foam, a spray or an emulsion.
  • the virucidal composition can be an air freshener, a sterilizing solution or a disinfecting solution.
  • a device comprising the virucidal composition of the invention or one or more virucidal nanoparticles of the invention and means for applying and/or dispensing the virucidal nanoparticles of the invention.
  • the means comprise a dispenser, a spray applicator or a solid support soaked with the virucidal nanoparticles of the invention.
  • the support is a woven or non-woven fabric, a textile, a paper towel, cotton wool, an absorbent polymer sheet, or a sponge.
  • Another aspect of the invention provides a method of disinfection and/or sterilization using the virucidal nanoparticles of the invention or the virucidal composition of the invention or the pharmaceutical composition of the invention.
  • the method of disinfection and/or sterilization comprises the steps of (i) providing at least one virucidal nanoparticle of the invention or a virucidal composition of the invention, or pharmaceutical composition of the invention, (ii) contacting an influenza virus-contaminated surface or a surface suspected to be contaminated by influenza virus with the at least one virucidal nanoparticle of the invention or a virucidal composition of the invention or pharmaceutical composition of the invention for a time sufficient to obtain virucidal effect.
  • the influenza virus-contaminated surface is human or animal skin.
  • the influenza virus-contaminated surface is a non-living surface, such as medical equipments, clothing, masks, furnitures, rooms, etc.
  • Another aspect of the invention provides a use of a virucidal nanoparticle of the invention or a virucidal composition of the invention or a pharmaceutical composition of the invention for sterilization and/or for disinfection.
  • sterilization and disinfection is for influenza virus-contaminated surfaces or surfaces suspected to be contaminated by influenza viruses.
  • the surfaces are human or animal skin.
  • the surfaces are non-living surfaces, such as medical equipments, clothing, masks, furnitures, rooms, etc.
  • the virucidal composition of the invention or the pharmaceutical composition of the invention is used as virucidal hand disinfectant for frequent use.
  • the virucidal composition of the invention or the pharmaceutical composition of the invention is applied by spraying.
  • the virucidal composition of the invention of the pharmaceutical composition of the invention is applied on a protective mask.
  • NPs at three different 6-sialyl-N-acetyllactoseamine (6′SLN) densities were synthetized in order to demonstrate the optimum 6′SLN density ( FIG. 1 ).
  • Control experiments with PEG(5) and 3-sialyl-N-acetyllactoseamine (3′SLN)—coated gold NPs were conducted as well.
  • LD-6′SLN NPs have the lowest half inhibitory concentration (EC 50 ) against all the viruses tested. NPs, in general, inhibited influenza A subtype better than B subtype.
  • EC 50 and CC 50 of different NPs against Influenza B/Yamagata EC 50 EC 50 Influenza EC 50 NPs 6′SLN CC 50 Strain ( ⁇ g/mL) (nM) (nM) ( ⁇ g/ml) LD 6′SLN NPs B/Yamagata 13.7 67 1206 >500 (18 6′SLN/NP) HD 6′SLN NPs B/Yamagata 17.25 85 3230 >500 (38 6′SLN/NP) LD(-) 6′SLN B/Yamagata 53.3 260 2080 >500 NPs (8 6′SLN/NP) LD 3′SLN NPs B/Yamagata >100 >100 >1000 >500 (18 3′SLN/NP) PEG5 NPs B/Yamagata N/A N/A N/A >500
  • Virucidal assays were then conducted, to determine whether the mechanism of inhibition is irreversible.
  • the NPs were incubated with the virus at corresponding IC99 concentration for a certain amount of time. Serial dilutions were then conducted of the inoculum and the residual infectivity of the virus was measured.
  • Virucidal activity of LD 6′SLN NPs against Neth/09 and Sing/04 strains was tested by increasing the virus concentration ten times relative to the dose-response experiments. The titer of Neth/09 strain was reduced by 2 logs whereas Sing/04 was reduced by 1.5 logs. 1-2 logs reduction in virus titer indicates that the virus is irreversibly inhibited.
  • the virus-NP interaction were demonstrated also with electron microscopy (TEM). H3N2 Vic/11 virus was incubated with LD 6′SLN NPs for 1 hour. After preparation of TEM grids, methyl tungsten staining was conducted. Majority of viruses were fully covered with LD 6′SLN NPs ( FIG. 2B ). The control experiments were conducted with PEG(5) NPs in which the NPs were all around but not attached to the viral envelop ( FIG. 2C ).
  • Influenza pandemics usually appear when animal influenza strains mix with human influenza strains. Therefore, the next goal is to irreversibly inhibit the avian flu strains with NPs of the invention.
  • Preliminary research was conducted with an egg-adapted virus strain, CAL/09.
  • Neth/09 and CAL/09 are two very similar human H1N1 strains.
  • LD-6′SLN NPs have a strong activity on the Neth/09 strain.
  • CAL/09 strain which is replicated using chicken eggs, binds LD 3′SLN NPs with higher affinity (Table 4). This result indicate that LD-3′SLN NPs inhibit avian influenza strains.
  • CD The size of CD (d ⁇ 1.5 nm) is comparable to gold (metal) NPs of the invention ( ⁇ 3 nm) and matches well with the HA globular head ( ⁇ 5 nm).
  • cyclodextrin Similar to gold (metal) NPs, cyclodextrin has a rigid chemical structure, which contributes to the virucidal activity together with the ligand.
  • beta-cyclodextrin can have maximum of 7 trisaccharides (and more preferably 3 or 4 trisaccharides), three being the exact number of sialic acid binding points in the HA globular head ( FIG. 3A ). Therefore, ⁇ -CD was modified with C15-6′SLN in a very similar way to gold nanoparticles.
  • ⁇ -cyclodextrin ( ⁇ -CD) was modified with different ligands, with and without trisaccharides, in order to investigate the relationship between the chemical structure and the antiviral activity.
  • exemplary modified cyclodextrins are shown in FIG. 4 . They bear a comparable number of trisaccharides (see FIG. 5 and Table 5), determined with 1 H Nuclear Magnetic Resonance Spectroscopy (NMR)).
  • NMR Nuclear Magnetic Resonance Spectroscopy
  • Dose-response assays against influenza A/Netherlands/2009 (H1N1) strain (A/NL/09) were conducted to compare the inhibitory activity of these NPs ( FIG. 6 ). The infection was quantified with immunocytochemical assays, 24 hours post-infection (hpi).
  • ⁇ -CDs bearing a sufficiently long, hydrophobic ligand and 6′SLN end-group showed strong inhibitory activity against infection of cells by the influenza A/NL/09, having EC 50 values in the nanomolar range.
  • the ⁇ -CD with a shorter ligand, C1-6′SLN poorly inhibited the infection.
  • Introducing a sufficiently long ligand clearly enhanced the end-group flexibility; hence the inhibitory concentrations decreased.
  • the EC 50 was comparable (yet slightly higher) when the hydrophobic ligand was replaced with a hydrophilic PEG8 ligand (PEG8-6′SLN).
  • C11-6′SLN the nanoparticle that showed the best inhibitory activity against A/NL/09, displayed strong antiviral activity against human influenza strains from both the A and the B types (Table 6 and FIG. 7 ). Importantly, it inhibited very recent A (H1N1) and B clinical strains (from the 2017/2018 influenza season), isolated from patients in the University Hospital of Geneva and passaged only once in cells. C11-6′SLN did not show any antiviral activity against HSV-2, an HSPG-binding virus, indicating specificity of the compound for sialic acid dependent viruses.
  • 6′SLN is known to be specific to human influenza strains whereas 3′SLN is preferred for avian influenza strains as a primary attachment point.
  • C11-3′SLN was synthesized and tested ( FIG. 4 ) against avian influenza strains.
  • C11-3′SLN successfully inhibited two avian strains, H5N1 and H7N1, at 4.1 and 8.8 ⁇ g/ml concentrations respectively (see Table 6). De facto, these results confirm the strategy adopted against human strains.
  • H5N1 has a significant potential to cause the next influenza pandemic.
  • virucidal assays were conducted to determine the mechanism of inhibition, i.e., virucidal (irreversible) or virustatic (reversible).
  • the synthesis of similar nanoparticles sharing the ⁇ -cylodextrin core and the 6′SLN moiety but different ligands highlights a structural feature conferring virucidal action ( FIGS. 9 and 10 ). It was hypothesized that one of the key components of irreversible viral inhibition is that the binding moiety (here 6′SLN) is borne by a hydrophobic ligand. To test this hypothesis, C11-6′SLN and P8-6′SLN were compared.
  • C11-6′SLN has an irreversible inhibitory effect on the virus while the effect of P8-6′SLN is reversible. It is worth mentioning that both nanoparticles are non-toxic to cells ( FIGS. 9 a and 9 b ). Virucidal activity of C11-6′SLN against other influenza strains was further investigated confirming its irreversible activity independently of the strain ( FIG. 9 c ).
  • C11-6′SLN completely prevented virus replication throughout the entire course of the experiment, while P8-6′SLN slightly reduced viral replication the first two days post-infection (dpi) but not thereafter ( FIG. 11 a ).
  • mice were administered with C11-6′SLN (25 ⁇ g/mouse) and A/NL/09 (100 infectious particles/mouse) simultaneously via the intranasal route.
  • the body temperature and the weight of the mice were measured on a daily basis in order to estimate the impact of C11-6′SLN administration on the infected animals' physiological condition.
  • half of the mice were randomly euthanized and the rest of the mice retreated with C11-6′SLN.
  • the second group of mice was euthanized at 4 dpi.
  • Viral titers were quantified from broncho-alveolar lavages (BAL) ( FIG. 15 a ). Significant decrease of viral titers was observed at day 2 and 4 post-infection in treated mice ( FIG. 15 a ). The antiviral activity of the C11-6′SLN also significantly diminished morbidity with a significant preservation of both weight and body temperature compared to untreated mice ( FIGS. 15 b and 15 c ). Collectively, these results demonstrate the capacity of the C11-6′SLN nanoparticles to prevent in vivo infection and spread of the virus in the lungs.
  • C11-6′SLN The in vivo therapeutic potential of C11-6′SLN was also assessed via post-treatment condition. Mice were infected with A/NL/09 (100 infectious particles/mouse) and treated with C11-6′SLN 6 hpi (14 ⁇ g/mouse) daily for the next three days with the same dose of nanoparticle ( FIGS. 15 d to f ). Weight and body temperature of the mice were measured each day. Although C11-6′SLN was less potent in the post-treatment condition, it still delayed the progress of the infection. The treated mice displayed a reduction of the morbidity signs ( FIGS. 15 d and 15 e ) and better clinical scores. These improvements in the infected-animal's physiological states correlated with prolonged survival ( FIG. 15 f ).
  • Neu5Ac ⁇ (2,6)-Gal ⁇ (1-4)-GlcNAc- ⁇ -ethylamine and Neu5Ac ⁇ (2,3)-Gal ⁇ (1-4)-GlcNAc- ⁇ -ethylamine were purchased from TCI chemicals.
  • Heptakis-(6-deoxy-6-mercapto)-beta-cyclo dextrin and carboxymethyl-beta-cyclodextrin sodium salt were purchased from Cyclodextrin-Shop.
  • 11-dodecenoic acid was purchased from abcr GmbH.
  • 14-pentadecenoic acid was purchased from Larodan AB.
  • Maleimide-PEG 8 -CH 2 CH 2 COOH was purchased from PurePEG. All the other chemicals and solvents were purchased from Sigma-Aldrich.
  • Cyclodextrin derivatives to target influenza virus were synthesized in three steps.
  • the first step was the conjugation of the ligands onto the cyclodextrin.
  • the second step was N-hydroxysuccinimide (NHS) activation of the —COOH end group of the ligands.
  • the third step was SLN grafting onto the ligands.
  • Heptakis-(6-deoxy-6-mercapto)-beta-cyclodextrin was stirred with 0.28 mmol of bi-functional molecules (ligands) bearing allyl and carboxylic acid end-groups (such as 6-heptenoic acid, 11-dodecenoic acid or 14-pentadecenoic acid) in 5 mL of DMSO, in the presence of a UV lamp (250 W), overnight.
  • the resulting heptakis-(6-deoxy-6-alkanoic acidthion)-beta-cyclodextrin derivatives were precipitated from DCM-diethyl ether mixture by centrifugation and dried under vacuum.
  • cyclodextrin derivatives obtained in step 1 were stirred with 1.12 mmol N-hydroxysuccinimide (NHS), 0.56 mmol ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC-hcl) and 0.02 mmol 4-dimethylaminopyridine (DMAP) in 5 mL of DMSO, overnight.
  • NHS-activated cyclodextrin derivatives were firstly precipitated from ice-cold water, and then washed with acetonitrile and diethyl ether. The products were dried under vacuum.
  • step 1 was skipped and NHS activation reaction was directly conducted.
  • Carboxymethyl-beta-cyclodextrin sodium salt directly (0.04 mmol) was activated with 1.12 mmol NHS 0.56 mmol EDC-hcl and 0.02 mmol DMAP was added. The activation reaction was conducted overnight. The resulting cyclodextrin derivative was first precipitated from a DCM-diethyl ether mixture and further washed three times. The product was dried under vacuum. 6′SLN grafting was conducted in the same way as step 3.
  • NHS Activation of C15- ⁇ -CD The resulting material was activated using 100 mg NHS, 75 mg of EDC and 2.5 mg of DMAP in DMF, overnight. NHS-activated C15- ⁇ -CD was precipitated from ice-cold water and further washed three times. The last precipitation was done in acetonitrile.
  • 6′SLN grafting onto C15- ⁇ -CD 5 mg C15- ⁇ -CD and 5 mg amine functionalized 6′SLN and 3 mg TEA were stirred overnight in DMSO. The resulting material cleaned using amicon filters.
  • PEG(5)-C15 mixed ligand NPs A ligand exchange reaction was performed with 1.5 mg of 16-mercaptohexacanoic acid (HS—C15-COOH) and 20 mg of PEG(5) NPs, in DMF, overnight. The NPs were precipitated from DMF-diethyl ether mixture and further washed three times.
  • NPs 15 mg were activated using 10 mg N-hydroxysuccinimide ester (NHS), 2 mg of ethylcarbodiimide hydrochloride (EDC) and 0.1 mg of 4-(dimethylamino)-pyridin (DMAP) in DMF, overnight. The resulting NPs were precipitated from DMF-diethyl ether mixture and further washed three times.
  • NHS N-hydroxysuccinimide ester
  • EDC ethylcarbodiimide hydrochloride
  • DMAP 4-(dimethylamino)-pyridin
  • 6′SLN grafting onto NPs 5 mg of NHS-activate NPs in 1 mL of DMSO were mixed with 1.7 mg of amine functionalized 6′SLN in 3 mL of 0.1 M phosphate buffer (pH: 7.5). After 5 hours reaction, 6′SLN grafted NPs were cleaned with amicon filters.
  • DMEM Glutamax medium was purchased from Thermo Fischer Scientific. Tween 20® for washing buffer and 3,3′-diaminobenzidine (DAB) tablets were purchased from Sigma Aldrich.
  • Primary antibody Influenza A monoclonal antibody
  • Secondary antibody Anti-mouse IgG, HRP-linked antibody
  • the CellTiter 96® AQ ueous One Solution Cell Proliferation Assay that contains a tetrazolium compound [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt; MTS] and an electron coupling reagent (phenazine ethosulfate; PES) were purchased from Promega.
  • MDCK Mesarby Canine Kidney Cells
  • ATCC American Type Culture Collection, Rockville, Md.
  • the cells were cultured in Dulbecco's modified Eagle's medium with glucose supplement (DMEM+ GlutaMAXTM) containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin.
  • MDCK cell lines was grown in humidified atmosphere with CO 2 (5%) at 37° C.
  • H1N1 Neth09 and B Yamagata were a kind gift from Prof M. Schmolke (University of Geneva).
  • Avian strain NIBRG-23 prepared by reverse genetics using A/turkey/Turkey/1/2005 H5N1 surface proteins and A/PR/8/34 (H1N1) backbone was obtained from National Institute for Biological Standards and Controls, Potters Bar, United Kingdom and was grown further in 10 days old embryonated chicken eggs followed by virus purification and characterization. Clinical samples were provided from the Geneva University Hospital from anonymized patients. All influenza strains were propagated and titrated by ICC on MDCK cells in presence of TPCK-treated trypsin (0.2 mg/ml)
  • Cell viability was measured by the MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assay. Confluent cell cultures seeded in 96-well plates were incubated with different concentrations of nanoparticles or ligand in triplicate under the same experimental conditions described for the antiviral assays. Cell viability was determined by the CellTiter 96 Proliferation Assay Kit (Promega, Madison, Wis., USA) according to the manufacturer's instructions. Absorbance was measured using a Microplate Reader (Model 680, BIORAD) at 490 nm.
  • the effect on cell viability at different concentrations of nanoparticles or cyclodextrins was expressed as a percentage, by comparing the absorbance of treated cells with the one of cells incubated with culture medium alone.
  • the 50% cytotoxic concentrations (CC 50 ) and 95% confidence intervals (CIs) were determined using Prism software (Graph-Pad Software, San Diego, Calif.).
  • MDCK cells were pre-plated 24 h in advance in 96-well plates. Increasing concentrations of materials were incubated with the influenza virus (MOI: 0.02 or 0.01 for H5N1 and 0.1 for other viruses) at 37° C. for one hour and then the mixtures were added to cells. Following the virus adsorption (1 h at 37° C.), the virus inoculum was removed, the cells were washed and fresh medium was added. After 24 h of incubation at 37° C., the infection was analyzed with immunocytochemical (ICC) assay. The cells were fixed and permeabilized with methanol. Then the primary antibody (1:100 dilution) was added and incubated for 1 hour at 37° C.
  • influenza virus MOI: 0.02 or 0.01 for H5N1 and 0.1 for other viruses
  • the cells were washed with wash buffer (DPBS+Tween 0.05%) three times; then secondary antibody (1:750 dilution) was added. After 1 hour the cells were washed and the DAB solution was added. Infected cells were counted and percentages of infection were calculated comparing the number of infected cells in treated and untreated conditions.
  • MDCK cells were pre-plated 24 h advanced in 24-well plates (75,000 cells/well). Increasing concentrations of materials were incubated with the influenza virus (MOI: 0.04) at 37° C. for one hour and then the mixtures were added to cells. Following the virus adsorption (1 h at 37° C.), the virus inoculum was removed, the cells were washed and fresh medium was added. After 5 h of incubation at 37° C., the infection was analyzed with flow cytometry.
  • influenza virus MOI: 0.04
  • H7N1 infectivity was evaluated through Luciferase activity.
  • MDCK cells were seeded at 5 ⁇ 10 4 on 96-wells plates. After 24 h, the medium was replaced by serum-free medium. Increasing concentrations of C11-3′ were incubated with 100 pfu of H7N1 A/Turkey/Italy/977/1999 encoding the NanoLuciferase. The mixture was incubated 1 h at 37° C. before addition to the cells for another 1 h incubation at 37° C. (100 ⁇ L per well). Cells were washed and medium replaced by serum-free medium with 1 ⁇ g/mL TPCK-Trypsin.
  • Viruses (focus forming unit (ffu):10 5 /mL) and the test materials (EC 99 concentration, Table 7) were incubated for 1 hour at 37° C. Serial dilutions of the virus-material complexes together with the non-treated controls were conducted and transferred onto the cells. After 1 hour, the mixture was removed and fresh medium was added. The next day, viral titers were evaluated with ICC assay. For the ICC assay, the same procedure described above was followed.
  • Co-treatment No. 1 H1N1 Neth/09 strain (pfu: 10 4 -10 5 ) and CD-C15-6′SLN (400 ug/mL) were simultaneously added to MucilAir (reconstruction of human airway epithelia). After 4 hours, the supernatant was removed and fresh medium was added. The viral titer of the supernatant was followed every 24 hours with qPCR.
  • Co-treatment No. 2 Human airway epithelia reconstructed in vitro, MucilAir tissues, (Epithelix Sàrl, Geneva, Switzerland) were cultured at the air-liquid interface from a mixture of nasal polyp epithelial cells originating from healthy donors. Influenza H1N1 pdm 2009 clinical strain (1e4 rna copies/tissue) and C11-6′ (50 ⁇ g/tissue) were transferred onto tissues without pre-incubation, together with non-treated control. After 4 hours of incubation time, the tissues were washed twice. On a daily basis, the basal medium was changed. To conduct daily qPCR measurements, 200 uL of medium was added onto tissues and then collected 20 minutes later. RNA extracted with EZNA viral extraction kit (Omega Biotek) was quantified by using qPCR with the QuantiTect kit (#204443; Qiagen, Hilden, Germany) in a StepOne ABI Thermocycler.
  • Influenza infected cells were detected by direct with Influenza A antibody (Light Diagnostic) and beta tubulin primary rabbit antibody (Abcam) was used as a marker of ciliated cells.
  • the Alexa 488-goat anti-rabbit Ab and the Alexa 594-goat anti-mouse Ab were used as the secondary Ab and nuclei were stained with DAPI. Images were acquired with Zeiss LSM 700 Meta confocal microscope and processed by Imaris.
  • MTT solution was diluted in MucilAir medium (1 mg/ml) and 300 ⁇ l were added basally in a 24 well plate. After 4 hours incubation at 37° C. the tissues were transferred in new plates and lysed with 1 ml of DMSO. The supernatant was read at 570 nm. Percentages of viability were calculated comparing treated and untreated tissues.
  • Interleukin-6 (IL-6), CXC motif chemokine 10 (CXCL10 or IP-10), CC motif chemokine5 (CCL5 or RANTES), interleukin-8 (IL-8 or CXCL-8) and interferon lambda (IL-29/IL-28B) were measured in the basal medium by ELISA (R&D DY206-05, DY266-05, DY278-05, DY208-05 and DY1598B-05) following everyday treatment with different concentrations of CD.
  • Pre-treatment Four groups of five BALB/c mice were treated at day 0 with 50 ⁇ l of PBS or C11-6′ (25 ⁇ g in 50 ⁇ l) and immediately inoculated with A/NL/09 (10 2 ffu). On a daily basis, body temperature and weight of the mice were measured. Two days post-infection, 2 groups of mice were sacrificed (one group pbs and the other C11-6′ treated). Lung homogenate and nasal mucosa and bronchoalveolar lavage were collected to quantify the viral titer through qPCR measurements. The C11-6′ treated group was retreated with the same amount of the nanoparticle. Two days after, all the remaining mice were sacrificed, lung homogenate and nasal mucosa were collected. After the tissue disruption, the RNA was extracted with Trizol and quantified by using qPCR while BAL were subjected to plaque assay. Two independent experiments were performed.
  • mice Post-treatment: 2 groups of ten BALB/c mice were infected with A/NL/09 (10 2 ffu) and treated at 6 hours post infection and subsequently daily for 7 days. Body temperature and weight of the mice were measured daily. Humane endpoint was used during survival study: mice were euthanized via cervical dislocation when body weights were reduced to 75% of the starting weights. In addition, animals that reached moribund state (unresponsive and unaware of stimuli) were also euthanized.
  • a fix virus concentration (ffu: 10 3 ) was incubated with varying dose of nanoparticles, for 1 hour at 37° C. The mixture then transferred onto cells. After one 1 hour, the mixture was removed and cells were washed. Next day, the infection was analyzed with immunocytochemical (ICC) assay.
  • ICC immunocytochemical

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Virology (AREA)
  • Nanotechnology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Ceramic Engineering (AREA)
  • Inorganic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
US17/273,113 2018-09-04 2019-09-03 Virucidal Nanoparticles And Use Thereof Against Influenza Virus Abandoned US20210322568A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP18192559 2018-09-04
EP18192559.5 2018-09-04
PCT/EP2019/073459 WO2020048976A1 (en) 2018-09-04 2019-09-03 Virucidal nanoparticles and use thereof against influenza virus

Publications (1)

Publication Number Publication Date
US20210322568A1 true US20210322568A1 (en) 2021-10-21

Family

ID=63678383

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/273,113 Abandoned US20210322568A1 (en) 2018-09-04 2019-09-03 Virucidal Nanoparticles And Use Thereof Against Influenza Virus

Country Status (9)

Country Link
US (1) US20210322568A1 (pt)
EP (1) EP3846853A1 (pt)
JP (1) JP2021535150A (pt)
KR (1) KR20210056379A (pt)
CN (1) CN112770780B (pt)
BR (1) BR112021004081A2 (pt)
CA (1) CA3110766A1 (pt)
IL (1) IL280936A (pt)
WO (1) WO2020048976A1 (pt)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024086510A1 (en) * 2022-10-17 2024-04-25 Ecole Polytechnique Federale De Lausanne (Epfl) Virucidal compounds and use thereof

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4125935A1 (en) 2020-03-30 2023-02-08 Ecole Polytechnique Federale De Lausanne (Epfl) Virucidal compositions and use thereof
CN113984747B (zh) * 2021-10-21 2024-04-05 南京理工大学 一种金纳米阵列表面修饰唾液酸的方法

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200166506A1 (en) * 2017-04-28 2020-05-28 National University Corporation Tokyo Medical And Dental University Modified nanoparticle, dispersion containing modified nanoparticle, set for resistive pulse sensing, set and reagent for detecting virus or bacterium, and method for detecting virus or bacterium

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
KR20050099536A (ko) * 2003-02-06 2005-10-13 트리펩 아베 글리코실화된 특이성 교환체
CN101511385A (zh) * 2006-09-11 2009-08-19 诺华有限公司 不使用蛋制备流感病毒疫苗
JP5360497B2 (ja) * 2007-06-28 2013-12-04 国立大学法人静岡大学 新規なn結合型人工シアロ糖鎖含有ポリマーおよびその製造方法
JP2009001805A (ja) * 2008-07-04 2009-01-08 Okura Ind Co Ltd ポリアミド系電子写真用部材
AU2008362921B2 (en) * 2008-10-13 2012-07-26 Semiotik Llc Multiligand constructs
FR2967677B1 (fr) * 2010-11-18 2014-05-16 Centre Nat Rech Scient Derives de polysaccharides comprenant un motif alcene et reaction de couplage par chimie thio-clic
RU2752620C2 (ru) * 2015-05-27 2021-07-29 Нортвестерн Юниверсити Модифицированные углеводами частицы и порошкообразные композиции для модуляции иммунного ответа
US20190275073A1 (en) * 2016-07-22 2019-09-12 Ecole polytechnique fédérale de Lausanne (EPFL) Virucidal compounds and uses thereof
WO2018034522A1 (ko) * 2016-08-18 2018-02-22 주식회사 고암바이오알앤디수 코어와 그의 표면에 결합된 시알산, 시알릴락토스 또는 이들의 유도체를 포함하는 결합체 및 그의 용도

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200166506A1 (en) * 2017-04-28 2020-05-28 National University Corporation Tokyo Medical And Dental University Modified nanoparticle, dispersion containing modified nanoparticle, set for resistive pulse sensing, set and reagent for detecting virus or bacterium, and method for detecting virus or bacterium

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Zhang et al. Clustering of PK-trisaccharides on amphiphilic cyclodextrin reveals unprecedented affinity for the Shiga-like toxin Stx2, Chem. Commun., 2017, 53, 10528-10531 (Year: 2017) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024086510A1 (en) * 2022-10-17 2024-04-25 Ecole Polytechnique Federale De Lausanne (Epfl) Virucidal compounds and use thereof

Also Published As

Publication number Publication date
EP3846853A1 (en) 2021-07-14
CN112770780B (zh) 2024-05-17
WO2020048976A1 (en) 2020-03-12
JP2021535150A (ja) 2021-12-16
BR112021004081A2 (pt) 2021-05-25
CN112770780A (zh) 2021-05-07
IL280936A (en) 2021-04-29
KR20210056379A (ko) 2021-05-18
CA3110766A1 (en) 2020-03-12

Similar Documents

Publication Publication Date Title
US20210322568A1 (en) Virucidal Nanoparticles And Use Thereof Against Influenza Virus
US10881673B2 (en) Conjugate including core and sialic acid or derivative thereof bound to surface of core and use thereof
KR102007278B1 (ko) 바이러스 진단 및 치료를 위한 인지능 물질 및 이의 제조방법
IL261345A (en) Preparations and methods for protection against pathogens and irritants carried in the air
US20210137966A1 (en) Virucidal compounds and uses thereof
WO2021207060A1 (en) Methods of treatment of coronavirus-induced inflammation conditions
US20170360815A1 (en) Compositions and methods for protecting against airborne pathogens and irritants
US20230099027A1 (en) Virucidal compositions and use thereof
WO2021236626A1 (en) Mucoretentive antiviral technologies
WO2023213310A1 (zh) 调节细胞膜跨膜运输和流动性的碳链物质、制备及应用
JP7497868B2 (ja) アンジオテンシン変換酵素2の発現抑制剤、および、アンジオテンシン変換酵素2を受容体とするウイルスに対する抗ウイルス剤
US20230201249A1 (en) Compositions incorporating sulfated polysaccharides for inhibiting sars-cov-2
KR20220037373A (ko) 시알산 유도체가 결합된 필라멘트성 파지, 및 이의 바이러스 감염증 예방 또는 치료 용도
Kocabiyik Virucidal nanomaterials against influenza
High et al. Article type: Communication
CA3192515A1 (en) Viral inhibitors
JP5633717B2 (ja) 抗ウイルス剤

Legal Events

Date Code Title Description
AS Assignment

Owner name: ECOLE POLYTECHNIQUE FEDERALE DE LAUSANNE (EPFL), SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TAPPAREL VU, CAROLINE;CAGNO, VALERIA;KOCABIYIK, OZGUN;AND OTHERS;SIGNING DATES FROM 20210303 TO 20210317;REEL/FRAME:055638/0705

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

STCB Information on status: application discontinuation

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