WO2023118014A1 - Inhalable compositions comprising a complex of hpbcd and budesonide or ciclesonide for the treatment or prevention of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease - Google Patents

Inhalable compositions comprising a complex of hpbcd and budesonide or ciclesonide for the treatment or prevention of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease Download PDF

Info

Publication number
WO2023118014A1
WO2023118014A1 PCT/EP2022/086808 EP2022086808W WO2023118014A1 WO 2023118014 A1 WO2023118014 A1 WO 2023118014A1 EP 2022086808 W EP2022086808 W EP 2022086808W WO 2023118014 A1 WO2023118014 A1 WO 2023118014A1
Authority
WO
WIPO (PCT)
Prior art keywords
lipopolysaccharide
viral disease
induced inflammation
respiratory viral
respiratory
Prior art date
Application number
PCT/EP2022/086808
Other languages
French (fr)
Inventor
Didier Cataldo
Natacha Rocks
Thomas Marichal
Maurizio PASSANISI
Original Assignee
Aquilon Pharmaceuticals
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
Priority claimed from BE20216002A external-priority patent/BE1029585B1/en
Priority claimed from BE20216105A external-priority patent/BE1030002B1/en
Application filed by Aquilon Pharmaceuticals filed Critical Aquilon Pharmaceuticals
Publication of WO2023118014A1 publication Critical patent/WO2023118014A1/en

Links

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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/40Cyclodextrins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0078Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a nebulizer such as a jet nebulizer, ultrasonic nebulizer, e.g. in the form of aqueous drug solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses

Definitions

  • the present invention relates to an inhalable composition
  • an inhalable composition comprising a complex of hydroxypropyl-beta-cyclodextrin (HPBCD) or derivatives thereof and budesonide or ciclesonide for use in the topical prevention or topical treatment of respiratory viral disease.
  • HPBCD hydroxypropyl-beta-cyclodextrin
  • HPBCD has already been described for the treatment of inflammatory disease.
  • EP1799231 to the University of Minnesota proposes the direct administration of cyclodextrins for the treatment of bronchial inflammatory disease, preferably asthma and chronic obstructive pulmonary disease (COPD).
  • bronchial inflammatory disease preferably asthma and chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • the microparticles have a median mass aerodynamic diameter of 0.1 microns or more and 5 microns or less;
  • the microparticles have a core-shell structure, with the carrier forming the shell and the active pharmaceutical ingredient forming the core;
  • the microparticles have a plurality of golf ball-like surface depressions identifiable by scanning electron microscopy; ⁇ The average maximum depth of the surface depressions is 5 % or more and 30 % or less as compared to the average maximum diameter D of the microparticles; and
  • EP3151836 to the University of Minnesota and Paul Maes discloses the use of HPBCD in conjunction with the corticosteroid budesonide for the treatment and prevention of bronchial inflammatory diseases, preferably for chronic obstructive pulmonary disease, such as chronic bronchitis, obstructive bronchiolitis, emphysema, pulmonary fibrosis, cystic fibrosis and most preferably for tobacco-induced chronic obstructive pulmonary disease and cystic fibrosis disease.
  • chronic obstructive pulmonary disease such as chronic bronchitis, obstructive bronchiolitis, emphysema, pulmonary fibrosis, cystic fibrosis and most preferably for tobacco-induced chronic obstructive pulmonary disease and cystic fibrosis disease.
  • HPBCD respiratory for the treatment of respiratory viral disease is not disclosed.
  • Pari EP1894559 discloses several corticosteroids solubilized through cyclodextrins for the treatment of a long list of human diseases, comprising viral sinusitis.
  • an inhalable composition comprising a complex of HPBCD and budesonide.
  • W02021001349 to Aquilon Pharmaceuticals discloses an inhalable composition comprising one or more cyclodextrins or pharmaceutically acceptable derivatives thereof for use in the prevention or treatment of nasal inflammations, wherein the treatment or prevention of nasal inflammations is a topical treatment.
  • a preferred cyclodextrin is HPBCD.
  • HPBCD is a preferred cyclodextrin.
  • this document does not disclose the use of HPBCD for the treatment of respiratory viral disease.
  • EP0423240B1 to the University of Pennsylvania discloses the use of a beta-cyclodextrin as a blocking agent for the preparation of a medicament for inhibiting infection of cells by a virus.
  • this document does not suggest the use of HPBCD nor its use for the treatment of respiratory viral disease.
  • WO2019067145 to ASDERA LLC discloses cyclodextrin compositions and methods that may be useful in the treatment and/or prevention of respiratory diseases, including, but not limited to the protection from virus or bacterial infections.
  • HPBCD the specific selection of HPBCD for the topical treatment of respiratory viral disease is not disclosed. Baglivo et al., 2020, Natural small molecules as inhibitors of coronavirus lipid dependent attachment to host cells: a possible strategy for reducing SARS-COV-2 infectivity? Acta Biomed 2020; Vol. 91 , N.
  • Combinations comprising cyclodextrins use in the treatment of viral diseases
  • inhalable compositions comprising a pharmaceutically effective amount of HPBCD and budesonide or ciclesonide complex have synergistic effects in respect of respiratory viral disease, in particular by reducing the neutrophilic inflammation in respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease.
  • the inhalable compositions modulate the neutrophilic or macrophage response to viruses.
  • the budesonide or the ciclesonide can be administered by inhalation in very low daily dosages and still be effective against respiratory viral disease, in particular human corona virus or lipopolysaccharide-induced infections or inflammations, in particular lipopolysaccharide-induced infections or inflammations associated with a respiratory viral disease.
  • a first aspect of the present invention is an inhalable composition
  • a complex of HPBCD or a pharmaceutically acceptable derivative thereof and budesonide or ciclesonide for use in the topical prevention or topical treatment of a respiratory viral disease, or lipopolysaccharide-induced infections or inflammations, in particular lipopolysaccharide-induced infections or inflammations associated with a respiratory viral disease, preferably by inhalation.
  • the respiratory virus is selected from the group consisting of adenovirus, human bocavirus, human coronavirus, human metapneumovirus, human parainfluenza virus, influenza, human respiratory syncytial virus, human rhinovirus, pharyngoconjunctival fever, and any other virus causing severe acute respiratory syndrome or combinations thereof.
  • no further active pharmaceutical ingredient is present in the inhalable composition in therapeutically effective amounts for which HPBCD is used to deliver an active pharmaceutical ingredient to other parts of the body than the respiratory system or the bronchial epithelium.
  • no further active pharmaceutical ingredient is present in the inhalable composition in therapeutically effective amounts for which HPBCD is used to increase the solubility of the further active pharmaceutical ingredient in water.
  • the inhalable composition is an aqueous solution, wherein the concentration of HPBCD is from 5 millimolar to 50 millimolar, preferably from 7 millimolar to 40 millimolar, even more preferably from 10 millimolar to 30 millimolar.
  • the inhalable composition comprises HPBCD in an amount from 1 mg/ml to 100 mg/ml, preferably from 1 mg/ml to 100 mg/ml, or from about 5 mg/ml to about 80 mg/ml and more preferably from about 8.00 mg/ml to 60 mg/ml.
  • the inhalable composition comprises budesonide or ciclesonide in an amount from 0,020 mg/ml to 1 mg/ml, or from 0,020 mg/ml to 0,70 mg/ml, preferably from about 0,05 mg/ml to about 0,5 mg/ml and more preferably from about 0,1 mg/ml to 0,25 mg/ml.
  • the viscosity of the inhalable composition is in the range of about 0,01 mPa.s to about 10 mPa.s, preferably in the range of about 0,5 mPa.s to about 5 mPa.s and most preferably in the range from about 0,8 mPa.s to about 3 mPa.s at 20 °C as measured by EP/LISP (https://www.roquette.com/innovation-hub/pharma/product- profile-pages/kleptose-hpb-oral-grade).
  • the composition is a spray-dried powder.
  • the HPBCD is administered per inhalation in the amount of 0.1 mg to 105 mg per day, or in the amount of 0.1 mg to 30 mg per day, preferably in the amount of 0.5 mg to 20 mg per day, even more preferably in the amount of 1 mg to 10 mg per day.
  • the HPBCD is administered in the amount of 1 mg to 105 mg per day.
  • the budesonide or ciclesonide is administered per inhalation in the amount of 0.020 mg to 0,700 mg per day, preferably in the amount of 0.05 mg to 0,5 mg per day, even more preferably in the amount of 0,07 mg to 0,25 per day, even more preferably in an amount of 0,07 mg to 0, 15 mg per day, even more preferably in an amount of 0,07 mg to 0, 10 mg per day.
  • the budesonide or ciclesonide is administered per inhalation in the amount of 0.02 to 1 mg per day.
  • the HPBCD is administered to children aged up to two years per inhalation in the amount of 0.1 mg to 1 ,0 mg per day and wherein the budesonide or the ciclesonide is administered in an amount of 0,05 mg to 0,12 mg per day.
  • the HPBCD is administered to children aged up to two years per inhalation in the amount of 0,025 mg to 0,12 mg per day.
  • the HPBCD is administered to children aged from 2 to 6 years in the amount of 0,05 mg to 0,20 mg per day and wherein the budesonide or the ciclesonide is administered per inhalation in an amount of 0,15 mg to 0,20 mg per day. In another embodiment, the HPBCD is administered to children aged from 2 to 6 years in the amount of 0,15 mg to 0,50 mg per day.
  • the HPBCD is administered to children aged from 6 to 14 years in the amount of 0, 1 mg to 20 mg per day and wherein the budesonide or the ciclesonide is administered per inhalation in an amount of 0,25 mg to 0,5 mg per day.
  • the HPBCD is administered to children aged from 6 to 14 years in the amount of 0,25 mg to 1 mg per day.
  • Another aspect of the invention is a Rayleigh-jet based inhaler comprising the inhalable composition of the invention for use in the topical prevention or topical treatment of a respiratory viral disease.
  • Another aspect of the invention is the use of a Rayleigh-jet based inhaler for delivering the inhalable composition of the invention in the topical prevention or topical treatment of a respiratory viral disease, or lipopolysaccharide-induced infections or inflammations, in particular lipopolysaccharide-induced infections or inflammations associated with a respiratory viral disease.
  • Another aspect is a method of topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced infections or inflammations, in particular lipopolysaccharide-induced infections or inflammations associated with a respiratory viral disease, wherein a composition comprising a complex of HPBCD (HPBCD) or a pharmaceutically acceptable derivative thereof and budesonide or the ciclesonide is administered in a therapeutically effective amount without treatment-limiting side-effects to a subject in need thereof.
  • HPBCD HPBCD
  • HPBCD is an oligosaccharide composed of glucopyranose units.
  • the major unsubstituted cyclodextrins are usually prepared by the enzymatic degradation of starch.
  • HPBCD is a chemically modified cyclodextrin which may have increased water solubility over unmodified cyclodextrins.
  • HPBCD Also comprised are pharmaceutically acceptable derivatives of HPBCD as long as they are structurally and functionally similar to HPBCD. Treatment
  • treatment or “treat” describes any treatment of respiratory viral disease for example by inhalation of an aerosol or a micronized powder in view of reducing the symptoms or causes of respiratory viral disease.
  • a first aspect of the present invention is an inhalable composition
  • the inhalable composition is free of alpha or gamma-cyclodextrin.
  • the inhalable composition comprises one or more further cyclodextrins, for example alpha-cyclodextrin, betacyclodextrins, gamma-cyclodextrin, and particularly 2-hydroxypropyl-gamma-cyclodextrin, sulfobutylether-beta-cyclodextrin, and methyl-beta-cyclodextrin.
  • cyclodextrins for example alpha-cyclodextrin, betacyclodextrins, gamma-cyclodextrin, and particularly 2-hydroxypropyl-gamma-cyclodextrin, sulfobutylether-beta-cyclodextrin, and methyl-beta-cyclodextrin.
  • inhalable refers to a composition that may be administered by inhalation.
  • inhalable means that the composition is capable of being micronized for inhalation purposes to an average particle size of 1 micrometer to 20 micrometers, preferably 1 micrometer to 10 micrometers and even more preferably 1 to 5 micrometers.
  • Suitable devices for micronization for inhalation include dry powder inhalers, metered dose inhaler or nebulizers.
  • composition of the present invention is for topical treatment, particularly by inhalation.
  • topical means that the composition of the invention is applied to the respiratory system, in particular to the nasal mucosa or the bronchial epithelium, for example by intranasal application or through inhalation of an aerosol generated by an aerosolgenerating device.
  • the treatment is a respiratory topical treatment in the sense that no other tissue or part of the body than the respiratory system, the nasal mucosa or the bronchial epithelium are targeted by the HPBCD.
  • the nasal mucosa is not targeted by the composition of the present invention.
  • no other part of the body than the lower respiratory system is targeted by the composition.
  • the primary purpose of the HPBCD of the composition of the invention is the use for the topical prevention or topical treatment of respiratory viral disease or the lipopolysaccharide-induced infections, in particular lipopolysaccharide- induced infections associated with a respiratory viral disease. Consequently, in one embodiment, the primary purpose of the cyclodextrins is not to enhance or improve the delivery or solubility of another active pharmaceutical ingredient.
  • active pharmaceutical ingredient refers to any substance or combination of substances used in a finished pharmaceutical product, intended to furnish pharmacological activity or to otherwise have direct effect in the diagnosis, cure, mitigation, treatment, or prevention of disease, or to have direct effect in restoring, correcting or modifying physiological functions in human beings.
  • active pharmaceutical ingredient refers to a molecule that is intended to be biologically active, for example for the purpose of treating inflammatory, autoimmune, or pulmonary disease, disorder, or condition.
  • active pharmaceutical ingredient refers to substances whose placing on the market requires a marketing authorization, for example as foreseen under directive 2001/83/EC of 6 November 2001 on the Community Code relating to medicinal products for human use.
  • Respiratory viruses such as human respiratory syncytial virus and influenza are well known to infect the lower airway, and both can cause bronchitis, bronchiolitis, and pneumonia.
  • Human rhinovirus has traditionally been considered to be an upper airway pathogen because of its association with common cold symptoms.
  • respiratory viruses including commonly used abbreviations include:
  • composition of the present invention is used for the increase and/or recovery of the diffusing capacity for carbon monoxide (DLCO) in the treatment of respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease, in particular SARS-COVID19.
  • DLCO carbon monoxide
  • composition of the present invention is used for the increase and/or recovery of the Forced expiratory volume in 1 second (FEV1 ) in the treatment of respiratory viral diseases or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease, in particular SARS-COVID19.
  • FEV1 Forced expiratory volume in 1 second
  • composition of the present invention is used for the increase and/or recovery of the Forced vital capacity (FVC) in the treatment of respiratory viral diseases or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide- induced inflammation associated with a respiratory viral disease, in particular SARS- COVID19.
  • FVC Forced vital capacity
  • the composition of the present invention is used for the treatment of lipopolysaccharides-associated inflammations.
  • the LPS- associated inflammations are associated with a respiratory viral disease, in particular SARS-COVID19.
  • LPS Lipopolysaccharide
  • SARS-COVID19 a respiratory viral disease
  • LPS Lipopolysaccharide
  • LPS can cause an acute inflammatory response by triggering the release of a vast number of inflammatory cytokines in various cell types. Therefore, LPS is widely recognized as a potent activator of monocytes/macrophages.
  • the LPS-associated inflammations may be LPS-produced, LPS-caused or LPS-induced inflammations or infections.
  • LPS-induced, LPS-induced and LPS-produced are used interchangeably. This means that the inflammations of the present invention are directly or indirectly produced by, induced by or induced by the LPS.
  • composition of the present inventions decreases the neutrophilic inflammation in the respiratory system of subjects/patients exposed to LPS.
  • composition of the present invention for treating the neutrophilic inflammation in the respiratory system of subjects/patients exposed to LPS or to LPS-associated inflammations or infections.
  • the composition of the present invention is used to reduce the neutrophil recruitment into lungs in particular following LPS exposure.
  • This typically characterized by the expression of the specific inflammatory markers such as CXCR4.
  • CXCR-4 is an alpha-chemokine receptor specific for stromal-derived-factor-1 (SDF-1 also called CXCL12).
  • SDF-1 is a molecule endowed with potent chemotactic activity for lymphocytes.
  • CXCR4 neutrophils are susceptible to release neutrophil extracellular traps (NETs).
  • composition of the present invention for decreasing the NETs numbers in the lungs of subjects/patients exposed to LPS or to LPS-associated inflammations or infections.
  • Steroids are anti-inflammatory compounds which are commonly also referred to as steroids, corticoids, glucocorticoids, or cortisol analogues.
  • corticosteroids include beclomethasone, budesonide, flunisolide, fluticasone, ciclesonide, mometasone, or any compounds comprising the active moiety of any of these corticosteroids, such as salts, derivatives, and prodrugs thereof.
  • corticosteroids selected from the group consisting of fluticasone and budesonide.
  • a preferred corticosteroid is budesonide.
  • Budesonide is also named (R,S)- 1 1 (3,16a, 17,21 -tetrahydroxypregna- 1 ,4-diene-3, 20- dione cyclic 16, 17-acetal with butyraidehyde or 16, 17-(butylidenebis(oxy))- 1 1 ,21 - dihydroxy-, (1 1 -[3,16-a)-pregna-l ,4-diene-3, 20-dione.
  • the chemical formula, molecular weight and CAS number for Budesonide are C25H34O6, MW: 430.5 and 51333-22-3, respectively.
  • Budesonide is a racemate consisting of a mixture of the two diastereomers 22 R and 22 S and is provided commercially as a mixture of the two isomers (22R and 22 S).
  • Commercial formulations of budesonide in solution are provided by AstraZeneca LP (Wilmington, Del.) under the trademark Pulmicort.
  • the molar ratio between steroid, preferably the budesonide or ciclesonide and the HPBCD may vary widely. In some embodiments, the molar ratio is from about 1 :1 to about 1 : 100, preferably from about 1 :1 to 1 :75 and even more preferably from about 1 :1 to about 1 : 50.
  • no further active pharmaceutical ingredient than budesonide or ciclesonide is present in the inhalable composition in therapeutically effective amounts for which HPBCD is used to deliver an active pharmaceutical ingredient to other parts of the body than the respiratory system or the bronchial epithelium.
  • no further active pharmaceutical ingredient than budesonide is present in the inhalable composition in therapeutically effective amounts for which HPBCD is used to increase the solubility of the further active pharmaceutical ingredient in water.
  • the composition is free of flavonoids agents, such as Citrox. In another embodiment, the composition is free of oils.
  • the composition does not comprise any bronchodilators such as formoterol. In other embodiments, the composition further comprises bronchodilators such as formoterol.
  • an effective amount refers to an amount of an active agent as described herein that contributes to achieving one or more desirable clinical outcomes, such as those described in the 'treatment" description above.
  • An appropriate “effective” amount in any individual case may be determined using standard techniques known in the art, such as a dose escalation study.
  • the term “therapeutically effective amount” is meant to refer to an amount of an active agent or combination of agents effective to ameliorate, delay, or prevent the symptoms. Determination of a therapeutically effective amount is well within the capabilities of the skilled person.
  • the present inventors have found that the inhalation of a composition comprising a complex of HPBCD and budesonide or ciclesonide is particularly suitable for the direct prevention or treatment of respiratory viral disease.
  • this is the direct effect of the topical administration of a composition comprising a complex of HPBCD and budesonide or ciclesonide to the respiratory system, in particular the nasal mucosa or the pulmonary epithelium.
  • the composition is only used to treat the lower airways, but not the upper airways.
  • no further active pharmaceutical ingredient than budesonide or ciclesonide is present in the composition of the present invention, wherein HPBCD is primarily used as solubilizing excipient or as a drug delivery system.
  • the composition of the invention is aqueous, which means that it is preferably a liquid composition comprising water as the predominant liquid constituent.
  • the inhalable composition is a composition comprising HPBCD, water and optionally one or more other components suitable for use in pharmaceutical delivery such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, or excipients, and antimicrobial preservatives.
  • these components are present in very low amounts, typically in the range of 0,1 to 5 mg/ml.
  • the viscosity of the inhalable aqueous composition is in the range of about 0,0,1 mPa.s to about 10 mPa.s, preferably in the range of about 0,5 mPa.s to about 5 mPa.s and most preferably in the range from about 0,8 mPa.s to about 3 mPa.s at 20 °C as measured by the Ell or U.S. Pharmacopoeia (USP) 911 - Viscosity (Capillary Viscometer Methods).
  • the composition comprises an effective amount of HPBCD, salt or derivative thereof and a pharmaceutically acceptable carrier.
  • a preferred composition for nebulization comprises HPBCD, NaCI and water.
  • liquids for inhalation When administered in aerosol form, it is further preferred for safety and tolerability reasons that water is the only liquid present in the composition. However, in some cases it may be acceptable even for liquids for inhalation to comprise some amount of other liquids, in particular one or more organic solvents having a relatively low inhalation toxicity such as ethanol, propylene glycol, or glycerol. In one embodiment, these liquids are present in very low amounts, typically in the range of 0,1 to 5 mg/ml.
  • the aqueous composition is suitable for administration as an aerosol.
  • it is designed for inhalation for the prevention or treatment of respiratory viral disease affecting a region of the upper respiratory tract, such as the nasal mucosa or the paranasal sinuses.
  • the aqueous composition is designed for inhalation for the prevention or treatment of respiratory viral disease affecting the lower airways, in particular the bronchial system.
  • the aqueous solution may be a isotonic or hypertonic.
  • a solution is isotonic when its effective osmotic concentration is the same as that of the cytosol inside the cells of the respiratory system.
  • a solution is hypertonic if it has a greater concentration of solutes than the cytosol inside the cells of the respiratory system.
  • the pH of the composition is adjusted to 3.5 to 7.5, preferably from 6,5 to 7 and even more preferably from 4,8 to 5,2.
  • the pH in the range from 4,8 to 5,2 may preferably be obtained by adding a pH buffer.
  • Preferable pH buffers are citric acid/citrate/ascorbic acid or citric acid/citrate/EDTA.
  • the composition may comprise one or more excipients selected from pharmaceutically acceptable organic acids, salts of organic acids, inorganic acids, inorganic salts, bases, sugars, sugar alcohols, stabilizers, antioxidants, surfactants, preservatives, and taste masking agents.
  • the composition of the present invention enables aqueous and dry powder formulations whose properties allow highly efficient and convenient aerosol or dry powder delivery using currently available aerosol generating devices, or dry powder inhalers.
  • composition of the invention for the topical prevention or topical treatment of respiratory viral disease by an aerosol or a micronized powder.
  • Aerosolization in the sense of the present invention, means any spraying process that produces droplets.
  • the size of the droplets may vary. Typically, the size of the droplets is between 1 and 100 microns, preferably, between 1 ,5 and 10 microns and even more preferably between 2 and 7 microns.
  • Micronization in the sense of the present invention, means any process to decrease the particle size of dry powders to an inhalable size, preferably between 1 and 100 microns, preferably, between 1 ,5 and 10 microns and even more preferably between 2 and 7 microns.
  • Dry powder inhaler in the sense of the present invention, means any device suitable for micronization of inhalable dry powder compositions.
  • composition of the present invention may have very little side effects even when administered even at very high dosages, such as daily delivered doses up to 180 mg/kg.
  • composition of the present invention may be an aqueous composition or a powder.
  • the amount or content of the cyclodextrin in the composition is selected to ensure a sufficiently low viscosity at ambient temperature in view of inhalation.
  • the dynamic viscosity which may also be influenced by the choice and quantity of the further excipients, also has a clear influence on the particle size distribution of the aerosol formed by nebulization and on the efficiency of nebulization.
  • the viscosity may be adjusted to a range of about 0.8 to about 3 mPas at ambient temperature.
  • the dynamic viscosity of the composition is in the range of about 0.8 to about 2.5 mPas or even about 0.9 to about 1 .3 mPas at ambient temperature.
  • the HPBCD concentration in the composition according to the present invention is in the range from about 1 millimolar to about millimolar, preferably from 5 millimolar to about 50 millimolar, and more preferably from about 7 millimolar to about 40 millimolar.
  • Other preferred concentrations range from about 10 millimolar to about 30 millimolar and from about 15 millimolar to about 25 millimolar.
  • the HPBCD is administered per inhalation in the amount of 0.1 mg to 30 mg per day, preferably in the amount of 0.5 mg to 20 mg per day, even more preferably in the amount of 1 mg to 10 mg per day.
  • the composition preferably is a spray-dried powder.
  • the composition is in the form of an inhalable powder, preferably in the form of a spray-dried powder.
  • the inhalable composition is in the form of spherical microparticles, wherein a.
  • the microparticles have a median mass aerodynamic diameter of 0.1 microns or more and 5 microns or less; b.
  • the microparticles have a core-shell structure, with the carrier forming the shell and the active pharmaceutical ingredient forming the core; c.
  • the microparticles have a plurality of golf ball-like surface depressions identifiable by scanning electron microscopy; d.
  • the average maximum depth d of the surface depressions (1 ) is 5 % or more and 30 % or less as compared to the average maximum diameter D of the microparticles; and e. 50 surface area% or more as compared to the total surface of the microparticles are depressed.
  • the inhalable composition is an aqueous solution and wherein the concentration of HPBCD is from 5 millimolar to 50 millimolar, preferably from 7 millimolar to 40 millimolar, even more preferably from 10 millimolar to 30 millimolar.
  • the inhalable composition comprises HPBCD in an amount from 1 mg/ml to 300 mg/ml, preferably from 1 mg/ml to 200 mg/ml, even more preferably from 1 mg/ml to 100 mg/ml.
  • the inhalable composition comprises from about 5 mg/ml to about 80 mg/ml and more preferably from about from 5 mg/ml to about 100 mg/ml, preferably from about 8.00 mg/ml to 60 mg/ml.
  • the HPBCD is administered per inhalation in the amount of 0.1 mg to 30 mg per day. In another embodiment, the HPBCD is administered per inhalation in the amount of 0.5 mg to 20 mg per day. In another embodiment, the HPBCD is administered per inhalation in the amount of 1 mg to 10 mg per day. In another embodiment, the HPBCD is administered to children aged up to two years per inhalation in the amount of 0.1 mg to 0.5 mg per day. In another embodiment, the HPBCD is administered to children aged from two to 6 years per inhalation in the amount of 0.5 mg to 1 mg per day. In another embodiment, the HPBCD is administered to children aged from 6 years to 14 years per inhalation in the amount of 1 mg to 2 mg per day.
  • the inhalable composition comprises budesonide or ciclesonide in an amount from 0,020 mg/ml to 0,70 mg/ml, preferably from about 0,05 mg/ml to about 0,5 mg/ml and more preferably from about 0,1 mg/ml to 0,25 mg/ml.
  • the inhalable composition comprises budesonide or ciclesonide in an amount from 0,02 mg/ml to 2 mg/ml, preferably from 0,06 to 1 ,2 mg/ml and more preferably from 0,1 mg/ml to 0,8 mg/ml.
  • the daily dosage of the steroid compound typically ranges from 1 microgram to 500 micrograms, preferably from about 30 micrograms to about 200 micrograms per day. In some embodiments, the daily dose of the steroid compound is from 50 micrograms to 150 micrograms and even more preferably from 75 micrograms to 125 micrograms. In another embodiment, the budesonide orciclesonide is administered per inhalation in the amount of 0.020 mg to 0,700 mg per day, preferably in the amount of 0.05 mg to 0,5 mg per day, even more preferably in the amount of 0,07 mg to 0,25 per day.
  • the HPBCD is administered to children aged up to two years per inhalation in the amount of 0.1 mg to 1 ,0 mg per day and wherein the budesonide or the ciclesonide is administered in an amount of 0,05 mg to 0,12 mg per day.
  • the HPBCD is administered to children aged from 2 to 6 years in the amount of 0,05 mg to 0,20 mg per day and wherein the budesonide or the ciclesonide is administered per inhalation in an amount of 0,15 mg to 0,20 mg per day.
  • the HPBCD is administered to children aged from 6 to 14 years in the amount of 0,1 mg to 20 mg per day and wherein the budesonide or the ciclesonide is administered per inhalation in an amount of 0,25 mg to 0,5 mg per day.
  • compositions of the present inventions are preferably administered through Rayleigh-jet based inhalers.
  • Rayleigh-jet based inhalers for aqueous solutions are commercially available from Medspray. Such nozzle units are based on plain orifice nozzles, creating Rayleigh jets. A 2 micron hole creates a jet, breaking up into mono-disperse 4 micron droplet trains. The diameter of the droplets is twice the size of the orifice. The hole size can be engineered to meet specific requirements of devices. Spray nozzle chips are typically made with technologies generally seen in computer chips.
  • Figure 1 shows that the DLCO and the functional respiratory tests of Example 9 of the patients who received at least one dose, FCV and FEV1 have been improved in patients using AQ001S, while they have been not at all improved in patients using the placebo (figures 1 to 6).
  • COVID-19 suffering patients do usually not recover to a normal DLCO, see for example Pulmonary function and COVID-19; www.sciencedirect.com; Current Opinion in Physiology 2021 , 21 :29-35; htps://doi.Org/10.1016/j.cophys.2021.03.0051. Therefore, the increase of DLCO is of particular importance.
  • Figure 2 shows the FEV1 of Example 9 of the patients who received at least one dose.
  • Figure 3 shows the FVC of Example 9 of the patients who received at least one dose.
  • Figure 4 shows the DLCO of Example 9 of the patients who finished the study.
  • Figure 5 shows the FEV1 of Example 9 of the patients who finished the study.
  • Figure 6 shows the FVC of Example 9 of the patients who finished the study.
  • Figure 7 shows the experimental outline of Example 10.
  • Naive C57bl/6 mice were exposed to low LPS dose (100 ng) intranasally alone or with excipient or distinct AQS doses (2,5 pg, 12,5 pg or 25 pg).
  • Excipient and AQS have been administered together with LPS and also eight hours after LPS administration intranasally. 24 hours after LPS exposure, early innate immune response has been evaluated by flow cytometry (FACS) and Neutrophil Extracellular Traps (NETs) release has been measured by evaluating free extracellular DNA release using a Quant-it picogreen assay.
  • FACS flow cytometry
  • NETs Neutrophil Extracellular Traps
  • Figure 8 shows the lung neutrophil numbers following LPS exposure and AQS treatment.
  • Naive mice were exposed to LPS with distinct doses of AQS or the excipient alone administered with LPS and 8 hours following LPS exposure.
  • P values were estimated with a one-way ANOVA with Tukey post hoc test.
  • Figure 9 shows the release of extracellular DNA in bronchoalveolar lavage fluids (BALF) following LPS exposure and AQS treatment.
  • BALF bronchoalveolar lavage fluids
  • Figure 10 highlights the presence of CXCR4 on the Neutrophils population in bronchoalveolar lavage fluids (BALF) following LPS exposure and AQS treatment.
  • BALF bronchoalveolar lavage fluids
  • Naive mice were exposed to LPS alone, LPS and excipient or LPS and AQS doses (2,5 pg, 12,5 pg or 25 pg) administered with LPS and 8 hours following LPS exposure.
  • CXCR4 staining level in BALF has been evaluated 24 hours after LPS exposure using Flow Cytometry.
  • P values were estimated with a oneway ANOVA with Tukey post hoc test. Examples
  • Example 2 Evaluation of pharmaceutical compositions from Example 1 or example 7 on the pathophysiology related to viral infection e.g. such as Covid-19 using an experimental in vivo rodent model through a longitudinal study
  • the longitudinal study consists in a follow up of non-invasively measured indicators before and after viral infection of rodent animals; the study is spread over 16 days.
  • the non- invasive measured parameters are described in the paragraph “measured indicators”.
  • blood is collected for analysis of the serological response. Five groups of sex-, age, and weight-matched rodents are compared:
  • MK-4482 Infected rodents treated with Monlupiravir under the code name MK-4482, which is a positive reference against SARS-CoV-2 virus.
  • the MK-4482 is administered per os.
  • the size of each experimental group is 10.
  • a virus stock is prepared, titrating 106 TCID50/ml of a SARS-CoV-2 strain.
  • An inoculum consisting of 100 pl of the stock-virus is inoculated to each rodent through the deposition of 50 pl in each nostril.
  • the inoculation is performed under brief general anesthesia using isoflurane.
  • the awakening of the rodents occurs after 90 seconds maximum.
  • TCID50 describes the virus quantity which is necessary for the destruction of 50% of the infected cellular colonies.
  • a single inhalation administration of pharmaceutical compositions is tested, preferably through intranasal inhalation for a better reproducibility and better pulmonary deposition.
  • the first inhalation administration is performed 1 h post virus inoculation.
  • Successive administrations of test items are performed at 24 h intervals for 14 consecutive days.
  • Respiratory function values by whole body plethysmography, before inoculation (d-1 ) and 3, 5, 7, and 14 days after inoculation.
  • plethysmography describes the volume change measurements in different areas of the body.
  • Lung viral genomic load (RT-qPCR) on day 14 post inoculation.
  • Plasma anti-SARS-CoV-2 neutralization titer on day 14 post inoculation.
  • Example 3 Evaluation of pharmaceutical compositions from Example 1 on the pathophysiology related to viral infection e.g. such as Covid-19 using an experimental in vivo rodent model through a sequential study:
  • the sequential study consists of a sequential follow-up of the induced COVID-19 disease, with sacrifice of a subgroup of animals every 48 hours until the 10th day after inoculation (5 timepoints).
  • the invasive indicators followed are described in the paragraph “sequential measured indicators” and they are measured at each timepoint.
  • the size of each experimental group is 6.
  • a virus stock is prepared, titrating 106 TCID50/ml of a SARS-CoV-2 strain.
  • An inoculum consisting of 100 pl of the stock-virus is inoculated to each rodent through the deposition of 50 pl in each nostril.
  • the inoculation is performed under brief general anesthesia using isoflurane. The awakening of the rodent occurs after 90 seconds maximum.
  • a single inhalation administration of pharmaceutical compositions is tested, preferably through intranasal inhalation for a better reproducibility and better pulmonary deposition.
  • the first inhalation administration is performed 1 h post virus inoculation.
  • Successive administrations of test items are performed at 24 h intervals for 10 consecutive days.
  • Respiratory function values by whole-body plethysmography, before inoculation (d-1) and 2, 4, 6, 8 and 10 days after viral inoculation.
  • RT-qPCR Lung viral genomic load
  • TID50 Lung infectious viral load at each timepoint (i.e., 2, 4, 6, 8 and 10 days after viral inoculation).
  • SARS-CoV-2 virus detection by immunohistochemistry at each timepoint i.e., 2, 4, 6, 8 and 10 days after viral inoculation.
  • lung cytokine/chemokine expression for example IL-1 , IL-2, IL-4, IL-6, IL-10, IL-12p35, TGFb, TNF, IFN et IP10, by RT-qPCR (ELISA or qPCR) at each timepoint (i.e. , 2, 4, 6, 8 and 10 days after viral inoculation).
  • Example 4 Evaluation of pharmaceutical compositions from Example 1 or example 1.2 on the pathophysiology related to viral infection e.g. HRSV using an experimental in vivo non-rodent model e.g. Ferret-model:
  • Virus preparation Wild-type HRSV subgroup A strain was passaged exclusively in HEp-2 cells.
  • Inhalation Per each day of the experiment, a single inhalation administration of pharmaceutical compositions is tested, preferably through intranasal inhalation for a better reproducibility and better pulmonary deposition. The first inhalation administration is performed 1 h post virus inoculation. Successive administrations of test items are performed at 24 h intervals for 21 consecutive days.
  • RT-PCR virus isolation and reverse transcription-PCR
  • Samples from right lungs and nasal turbinates as well as from the trachea and bronchus are processed before viral load assessment by virus isolation and quantitative PCR (qPCR). Infectious virus titers in tissue are expressed as Iog10 TCID50 per gram tissue, and infectious virus titer in nose washes and swabs are expressed as log 10 TCID50/mL.
  • Formalin-fixed tissue sections will be routinely processed, paraffin embedded and sectioned at 3-4 pm, and stained with hematoxylin and eosin for histopathological examination by light microscopy.
  • additional serial slides are sectioned simultaneously and incubated for 1 h with a goat anti-HRSV-peroxidase (PO).
  • PO goat anti-HRSV-peroxidase
  • Example s Evaluation of pharmaceutical compositions from Example 1 or example 7 on the pathophysiology related to Influenza A infection using an experimental in vivo mice model:
  • a variation of the example 2 consists to infect rodents, preferably mice, with Influenza A virus strain A/PR8/34 (H1 N1 ).
  • the virus stock suspension (108 PFU/ml) has been diluted and 50 pl of solution (corresponding to 5 PFU) have been administered to isoflurane - anesthetized mice (Nat lmmunol.2019 November; 20 (11 ): 1444-1455. doi:10.1038/s41590-019-0496-9).
  • the term PFU means Plaque Forming Units and it describes the number of virus particles capable of forming plaques per unit volume.
  • ⁇ Inflammatory cell subtypes macrophages (alveolar, interstitial), polynuclear neutrophil, lymphocytes, NK cells, Dendritic cells, granulocytes, NET production at each timepoint (i.e., 24 hours, 2, 4, 6, 8 and 10 days after viral inoculation).
  • DNA nets i.e., 24 hours, 2, 4, 6, 8 and 10 days after viral inoculation.
  • Example 6 Evaluation of the efficacy / pharmacodynamics (PD) of pharmaceutical compositions from Example 1 on the symptoms related to Covid-19 infection through a phase 2a clinical trial:
  • the objectives of the clinical test are to assess the efficacy/pharmacodynamics and the safety of liquid drug products composed of HP-beta-CD and Budesonide in the treatment of COVID-19 symptoms in hospitalized patients, during and after their hospitalization.
  • composition The pharmaceutical composition tested in this clinical trial is composed of 17,5 mg/ml HP-beta-CD and 0,125 mg/ml Budesonide.
  • the liquid composition also comprises antioxidants i.e. EDTA, Citric Acid monohydrate and Trisodium Citrate Dihydrate as a pH adjuster in the range of 4-7.
  • the osmolarity of the solution is adjusted with adequate amounts of Sodium Chloride.
  • Tow dose strengths of this pharmaceutical solution will be tested in the clinical trial.
  • the placebo is Saline e.g. parenteral NaCI 0.9% isotonic solution.
  • the pharmaceutical composition and the placebo are delivered to the patients using inhaler devices e.g. PulmoSprayTM SP and PulmoSprayTM ST softmist inhaler when patient is hospitalized or with PARI BOY® Classic inhalation system when atient is discharged from hospital.
  • PulmoSprayTM SP is a softmist inhaler device to be used in combination with the Respi Lever DriveTM RP004 when the patient is hospitalized but not under a mechanical ventilation therapy.
  • PulmoSprayTM ST is a softmist inhaler device to be used without Respi Lever DriveTM RP004 when the patient is hospitalized and under a mechanical ventilation therapy.
  • the efficacy is assessed by :
  • COVID-19 clinical progression scale e.g. a composite scale allowing the analysis of reproducible, non-invasive and widely used well-being metrics describing the clinical course of the disease.
  • the endpoint will be the change from baseline to Day 28 ⁇ 2 , and over the dosing period.
  • Clinical Progression Scale reflects patient trajectory and resource use over the course of clinical illness (https://doi.org/10.1016/S1473- 3099(20)30483-7).
  • mMRC Modified Medical Research Council
  • mMRC Modified Medical Research Council
  • the term (mMRC) dyspnea scale quantifies disability attributable to breathlessness, and is useful for characterizing baseline dyspnea in patients with respiratory diseases.
  • Usefulness of the Medical Research Council (MRC) dyspnoea scale as a measure of disability in patients with chronic obstructive pulmonary disease. (Thorax 1999; 54:581-586); https://pubmed.ncbi.nlm.nih.gov/10377201/).
  • CT Scan pulmonary computer-assisted tomography
  • the PD is assessed by :
  • AE adverse events
  • SAE serious adverse events
  • the exploratory endpoints are:
  • VAS Visual analog scale
  • ACQ-5 Asthma Control Questionnaire -5
  • Naive mice will be exposed to low doses of LPS and treated by intratracheal instillation of AQ001S (different doses). Acute and chronic inflammation parameters will be assessed 24 hours and 7 days after LPS stimulation, respectively.
  • ⁇ NETs release in lungs (analyzed by western blot on lung protein extracts on citrullinated histone 3, or by confocal microscopy on tissue sections) (after 24h)
  • Inflammation and tissue repair will be evaluated by bi-photon microscopy (to identify collagen deposition), histological examination (7 days).
  • pCT will help highlight various radiological opacities and disease-specific patterns in lungs with high resolution, both in vivo and ex vivo. These could be further used by RDX to determine and validate new biomarkers for the non-invasive diagnosis of different inflammatory conditions in the lung by means of their Al proprietary processing tools. To obtain high resolution images, respiratory motion should be minimized. To this aim, different strategies will be tested and assessed on pCT for pulmonary gating, i.e. image acquisition synchronized to the animal respiratory cycle.
  • fluorodeoxyglucose (18F-FDG) and radiolabeled neutrophils as radiotracers in pPET experiments will provide in vivo quantification data over time related to lung inflammation and neutrophil recruitment since both radiolabeled moleculeswill accumulate in inflammatory sites with different kinetics.
  • Histology parameters will be correlated with imaging data collected throughout the model, with a particular interest in the timing of inflammation development and its response to AQ001 S treatment.
  • NETs markers myeloperoxidase, neutrophil elastase, peptidyl- arginine deaminase 4, mTOR and MEK/ERK
  • NETs markers myeloperoxidase, neutrophil elastase, peptidyl- arginine deaminase 4, mTOR and MEK/ERK
  • Example 9 inhalable aqueous composition AQ001S
  • the inhalable aqueous composition AQ001 S had the following composition:
  • composition AQ001 S was clinically tested in a Phase Ila -Proof of claim randomized double-blind (at hospital), placebo-controlled, parallel, multiple dose clinical trial in adults admitted to hospital with a confirmed positive SARS-CoV-2 RT-PCR test
  • AQ001 S Two regimens of AQ001 S were tested and compared to a placebo inhalation solution.
  • 0.4mL AQ001 S were administered by a PulmoSprayTM softmist inhaler.
  • 2mL AQ001 S 0,125mg/ml have been administered using PARI BOY Classic Inhalation System at home, i.e. once patients were discharged from hospital.
  • Subjects adult patients (99) admitted to hospital with a positive SARS-CoV-2 test.
  • Duration of subject study participation 36 ⁇ 2 days from screening visit with treatments administered for a duration of 28 ( ⁇ 2) days.
  • the FEV1 is the amount of the exhaled air during the 1st second of the FVC maneuver. It decreases in case of diseases that obstruct the airway, such as asthma or emphysema.
  • the forced vital capacity (FVC) A spirometry manoeuvre begins with the patient inhaling as deeply as he or she can. Then the patient exhales as long and as forcefully as possible; the amount exhaled in this manner is the FVC.
  • DLCO carbon monoxide
  • AEs Adverse Events
  • SAEs serious adverse events
  • Figures 1 to 6 show that the DLCO and the functional respiratory tests, FCV and FEV1 have been improved in patients using AQ001 S, while they have been improved in patients using the placebo.
  • Example 10 AQ001S Pharmacology in rodent models of acute respiratory inflammation induced by LPS (focus on neutrophils
  • composition AQ001 S has been tested in Pharmacodynamic (PD) study in rodents to demonstrate its pharmacological performance on acute and chronic models mimicking inflammatory processes induced by respiratory infections.
  • PD Pharmacodynamic
  • mice Female wild-type C57BL/6 mice (6 to 8 weeks old) purchased from Janvier laboratories were used. Mice were housed under specific pathogen free (SPF) conditions and maintained in a 12h light-dark cycle with food and water ad libitum. The animals were allocated to 6 experimental groups, with 4 mice per group:
  • mice were injected i.n. with PBS, LPS, LPS+ HP-b-CD, LPS+AQ2,5, LPS+AQ12,5, LPS+AQ25 in a total volume of 50 pL.
  • mice were injected i.n. with vehicle, AQ2,5, AQ12,5, AQ25 in a total volume of 50 pL.
  • mice were sacrificed.
  • dsDNA double-stranded DNA
  • Example 10 The results of Example 10 are shown in Figures 7 to 10.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Communicable Diseases (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Pulmonology (AREA)
  • Engineering & Computer Science (AREA)
  • Inorganic Chemistry (AREA)
  • Dispersion Chemistry (AREA)
  • Otolaryngology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

An inhalable composition comprising a complex of hydroxypropyl-beta-cyclodextrin (HPBCD) or a pharmaceutically acceptable derivative thereof and budesonide or ciclesonide for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide- induced inflammation associated with a respiratory viral disease, wherein the respiratory virus is selected from the group consisting of adenovirus, human bocavirus, human coronavirus, human metapneumovirus, human parainfluenza virus, human respiratory syncytial virus, human rhinovirus, pharyngoconjunctival fever, and any other virus causing severe acute respiratory syndrome or combinations thereof.

Description

INHALABLE COMPOSITIONS COMPRISING A COMPLEX OF HPBCD AND BUDESONIDE OR CICLESONIDE FOR THE TREATMENT OR PREVENTION OF A RESPIRATORY VIRAL DISEASE OR LIPOPOLYSACCHARIDE-INDUCED INFLAMMATION, IN PARTICULAR LIPOPOLYSACCHARIDE-INDUCED INFLAMMATION ASSOCIATED WITH A RESPIRATORY VIRAL DISEASE
HELD OF THE INVENTION
The present invention relates to an inhalable composition comprising a complex of hydroxypropyl-beta-cyclodextrin (HPBCD) or derivatives thereof and budesonide or ciclesonide for use in the topical prevention or topical treatment of respiratory viral disease.
In recent years, the number of respiratory viral diseases has substantially increased through multiple factors, particularly through the recent Corona virus epidemic.
HPBCD has already been described for the treatment of inflammatory disease.
Cyclodextrins for treating inflammatory disease
EP1799231 to the University of Liege proposes the direct administration of cyclodextrins for the treatment of bronchial inflammatory disease, preferably asthma and chronic obstructive pulmonary disease (COPD). However, the use of HPBCD for the treatment of respiratory viral disease is not disclosed.
International patent application WO2021048322A1 to Aquilon Pharmaceuticals discloses spherical microparticles for use in the treatment and prevention of respiratory disease comprising one or more carriers, preferably HPBCD and one or more active pharmaceutical ingredients, preferably budesonide, wherein
■ The microparticles have a median mass aerodynamic diameter of 0.1 microns or more and 5 microns or less;
■ The microparticles have a core-shell structure, with the carrier forming the shell and the active pharmaceutical ingredient forming the core;
■ The microparticles have a plurality of golf ball-like surface depressions identifiable by scanning electron microscopy; ■ The average maximum depth of the surface depressions is 5 % or more and 30 % or less as compared to the average maximum diameter D of the microparticles; and
■ 50 surface area percent or more as compared to the total surface of the microparticles are depressed.
EP3151836 to the University of Liege and Paul Maes discloses the use of HPBCD in conjunction with the corticosteroid budesonide for the treatment and prevention of bronchial inflammatory diseases, preferably for chronic obstructive pulmonary disease, such as chronic bronchitis, obstructive bronchiolitis, emphysema, pulmonary fibrosis, cystic fibrosis and most preferably for tobacco-induced chronic obstructive pulmonary disease and cystic fibrosis disease. However, the use of HPBCD respiratory for the treatment of respiratory viral disease is not disclosed.
Pari, EP1894559, discloses several corticosteroids solubilized through cyclodextrins for the treatment of a long list of human diseases, comprising viral sinusitis. However, there is no credible disclosure on the synergistic antiviral effect of an inhalable composition comprising a complex of HPBCD and budesonide.
Cyclodextrins for direct use in the treatment of viral diseases
W02021001349 to Aquilon Pharmaceuticals discloses an inhalable composition comprising one or more cyclodextrins or pharmaceutically acceptable derivatives thereof for use in the prevention or treatment of nasal inflammations, wherein the treatment or prevention of nasal inflammations is a topical treatment. A preferred cyclodextrin is HPBCD. However, this document does not disclose the use of HPBCD for the treatment of respiratory viral disease.
EP0423240B1 to the University of Pennsylvania discloses the use of a beta-cyclodextrin as a blocking agent for the preparation of a medicament for inhibiting infection of cells by a virus. However, this document does not suggest the use of HPBCD nor its use for the treatment of respiratory viral disease.
WO2019067145 to ASDERA LLC discloses cyclodextrin compositions and methods that may be useful in the treatment and/or prevention of respiratory diseases, including, but not limited to the protection from virus or bacterial infections. However, the specific selection of HPBCD for the topical treatment of respiratory viral disease is not disclosed. Baglivo et al., 2020, Natural small molecules as inhibitors of coronavirus lipid dependent attachment to host cells: a possible strategy for reducing SARS-COV-2 infectivity? Acta Biomed 2020; Vol. 91 , N. 1 : 161-164, DOI: 10.23750/abm.v91 i1.9402, discloses the use of methyl-[3-cyclodextrin as a molecular inhibitor of virus lipid-dependent attachment. However, this document does not disclose the use of HPBCD for use in the treatment of respiratory viral disease.
Combinations comprising cyclodextrins use in the treatment of viral diseases
Carrouel etc., COVID-19: A Recommendation to Examine the Effect of Mouthrinses with [3-Cyclodextrin Combined with Citrox in Preventing Infection and Progression, J. Clin. Med. 2020, 9, 1126; doi:10.3390/jcm9041126, discloses the use of a mouth rinses and/or with local nasal applications that contain [3-cyclodextrins combined with flavonoids agents, such as Citrox, could provide valuable adjunctive treatment to reduce the viral load of saliva and nasopharyngeal microbiota, including potential SARS-CoV-2 carriage. However, this does not disclose the direct use of HPBCD.
Kusakabe et al., Intranasal HPBCD-adjuvanted influenza vaccine protects against sub- heterologous virus infection, http://dx.doi.Org/10.1016/j.vaccine.2016.04.001 , discloses the intranasal use of HP-beta-CD as an adjuvant inducing significantly lower antigen-specific IgE responses than that induced by aluminum salt adjuvant. Kusakabe further suggests that HPBCD may be a potent mucosal adjuvant for seasonal and pandemic influenza vaccine. This document further suggests the use of therapeutic oral biofilm rinses and/or nasal applications in preventing viral transmission via the oropharyngeal route. However, this document does not disclose the direct use of HPBCD as an active pharmaceutical ingredient.
There still is a need for an efficient treatment of respiratory viral disease in particular of the corona virus that produce less side effects even when administered over long periods of time.
SHORT DESCRIPTION OF THE INVENTION
The inventors have surprisingly found that inhalable compositions comprising a pharmaceutically effective amount of HPBCD and budesonide or ciclesonide complex have synergistic effects in respect of respiratory viral disease, in particular by reducing the neutrophilic inflammation in respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease. In another embodiment, the inhalable compositions modulate the neutrophilic or macrophage response to viruses. Consequently, through the complexation with the HPBCD, the budesonide or the ciclesonide can be administered by inhalation in very low daily dosages and still be effective against respiratory viral disease, in particular human corona virus or lipopolysaccharide-induced infections or inflammations, in particular lipopolysaccharide-induced infections or inflammations associated with a respiratory viral disease.
Accordingly, a first aspect of the present invention is an inhalable composition comprising a complex of HPBCD or a pharmaceutically acceptable derivative thereof and budesonide or ciclesonide for use in the topical prevention or topical treatment of a respiratory viral disease, or lipopolysaccharide-induced infections or inflammations, in particular lipopolysaccharide-induced infections or inflammations associated with a respiratory viral disease, preferably by inhalation.
In one embodiment, the respiratory virus is selected from the group consisting of adenovirus, human bocavirus, human coronavirus, human metapneumovirus, human parainfluenza virus, influenza, human respiratory syncytial virus, human rhinovirus, pharyngoconjunctival fever, and any other virus causing severe acute respiratory syndrome or combinations thereof.
In one embodiment, no further active pharmaceutical ingredient is present in the inhalable composition in therapeutically effective amounts for which HPBCD is used to deliver an active pharmaceutical ingredient to other parts of the body than the respiratory system or the bronchial epithelium.
In one embodiment, no further active pharmaceutical ingredient is present in the inhalable composition in therapeutically effective amounts for which HPBCD is used to increase the solubility of the further active pharmaceutical ingredient in water.
In one embodiment, the inhalable composition is an aqueous solution, wherein the concentration of HPBCD is from 5 millimolar to 50 millimolar, preferably from 7 millimolar to 40 millimolar, even more preferably from 10 millimolar to 30 millimolar.
In another embodiment, the inhalable composition comprises HPBCD in an amount from 1 mg/ml to 100 mg/ml, preferably from 1 mg/ml to 100 mg/ml, or from about 5 mg/ml to about 80 mg/ml and more preferably from about 8.00 mg/ml to 60 mg/ml. In another embodiment, the inhalable composition comprises budesonide or ciclesonide in an amount from 0,020 mg/ml to 1 mg/ml, or from 0,020 mg/ml to 0,70 mg/ml, preferably from about 0,05 mg/ml to about 0,5 mg/ml and more preferably from about 0,1 mg/ml to 0,25 mg/ml.
In another embodiment, the viscosity of the inhalable composition is in the range of about 0,01 mPa.s to about 10 mPa.s, preferably in the range of about 0,5 mPa.s to about 5 mPa.s and most preferably in the range from about 0,8 mPa.s to about 3 mPa.s at 20 °C as measured by EP/LISP (https://www.roquette.com/innovation-hub/pharma/product- profile-pages/kleptose-hpb-oral-grade).
In another embodiment, the composition is a spray-dried powder.
In another embodiment, the HPBCD is administered per inhalation in the amount of 0.1 mg to 105 mg per day, or in the amount of 0.1 mg to 30 mg per day, preferably in the amount of 0.5 mg to 20 mg per day, even more preferably in the amount of 1 mg to 10 mg per day.
In another embodiment, the HPBCD is administered in the amount of 1 mg to 105 mg per day.
In another embodiment, the budesonide or ciclesonide is administered per inhalation in the amount of 0.020 mg to 0,700 mg per day, preferably in the amount of 0.05 mg to 0,5 mg per day, even more preferably in the amount of 0,07 mg to 0,25 per day, even more preferably in an amount of 0,07 mg to 0, 15 mg per day, even more preferably in an amount of 0,07 mg to 0, 10 mg per day.
In another embodiment, the budesonide or ciclesonide is administered per inhalation in the amount of 0.02 to 1 mg per day.
In another embodiment, the HPBCD is administered to children aged up to two years per inhalation in the amount of 0.1 mg to 1 ,0 mg per day and wherein the budesonide or the ciclesonide is administered in an amount of 0,05 mg to 0,12 mg per day.
In another embodiment, the HPBCD is administered to children aged up to two years per inhalation in the amount of 0,025 mg to 0,12 mg per day.
In another embodiment, the HPBCD is administered to children aged from 2 to 6 years in the amount of 0,05 mg to 0,20 mg per day and wherein the budesonide or the ciclesonide is administered per inhalation in an amount of 0,15 mg to 0,20 mg per day. In another embodiment, the HPBCD is administered to children aged from 2 to 6 years in the amount of 0,15 mg to 0,50 mg per day.
In another embodiment, the HPBCD is administered to children aged from 6 to 14 years in the amount of 0, 1 mg to 20 mg per day and wherein the budesonide or the ciclesonide is administered per inhalation in an amount of 0,25 mg to 0,5 mg per day.
In another embodiment, the HPBCD is administered to children aged from 6 to 14 years in the amount of 0,25 mg to 1 mg per day.
Another aspect of the invention is a Rayleigh-jet based inhaler comprising the inhalable composition of the invention for use in the topical prevention or topical treatment of a respiratory viral disease.
Another aspect of the invention is the use of a Rayleigh-jet based inhaler for delivering the inhalable composition of the invention in the topical prevention or topical treatment of a respiratory viral disease, or lipopolysaccharide-induced infections or inflammations, in particular lipopolysaccharide-induced infections or inflammations associated with a respiratory viral disease.
Another aspect is a method of topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced infections or inflammations, in particular lipopolysaccharide-induced infections or inflammations associated with a respiratory viral disease, wherein a composition comprising a complex of HPBCD (HPBCD) or a pharmaceutically acceptable derivative thereof and budesonide or the ciclesonide is administered in a therapeutically effective amount without treatment-limiting side-effects to a subject in need thereof.
DETAILED DESCRIPTION OF THE INVENTION
HPBCD
HPBCD is an oligosaccharide composed of glucopyranose units. The major unsubstituted cyclodextrins are usually prepared by the enzymatic degradation of starch. HPBCD is a chemically modified cyclodextrin which may have increased water solubility over unmodified cyclodextrins.
Also comprised are pharmaceutically acceptable derivatives of HPBCD as long as they are structurally and functionally similar to HPBCD. Treatment
The term “treatment” or “treat” describes any treatment of respiratory viral disease for example by inhalation of an aerosol or a micronized powder in view of reducing the symptoms or causes of respiratory viral disease.
Inhalable composition
A first aspect of the present invention is an inhalable composition comprising a complex of HPBCD (HPBCD) or a pharmaceutically acceptable derivative thereof and budesonide ciclesonide for use in the topical prevention or topical treatment of a respiratory viral disease, preferably by inhalation. In some embodiments, the inhalable composition is free of alpha or gamma-cyclodextrin. In other embodiments, the inhalable composition comprises one or more further cyclodextrins, for example alpha-cyclodextrin, betacyclodextrins, gamma-cyclodextrin, and particularly 2-hydroxypropyl-gamma-cyclodextrin, sulfobutylether-beta-cyclodextrin, and methyl-beta-cyclodextrin.
The term “inhalable” as used herein refers to a composition that may be administered by inhalation. In particular, the term “inhalable” means that the composition is capable of being micronized for inhalation purposes to an average particle size of 1 micrometer to 20 micrometers, preferably 1 micrometer to 10 micrometers and even more preferably 1 to 5 micrometers. Suitable devices for micronization for inhalation include dry powder inhalers, metered dose inhaler or nebulizers.
Topical treatment
The composition of the present invention is for topical treatment, particularly by inhalation. The term “topical” means that the composition of the invention is applied to the respiratory system, in particular to the nasal mucosa or the bronchial epithelium, for example by intranasal application or through inhalation of an aerosol generated by an aerosolgenerating device. In some embodiments the treatment is a respiratory topical treatment in the sense that no other tissue or part of the body than the respiratory system, the nasal mucosa or the bronchial epithelium are targeted by the HPBCD.
In another embodiment, the nasal mucosa is not targeted by the composition of the present invention.
In another embodiment, no other part of the body than the lower respiratory system is targeted by the composition. Direct application of HPBCD
The term “directly” means that the primary purpose of the HPBCD of the composition of the invention is the use for the topical prevention or topical treatment of respiratory viral disease or the lipopolysaccharide-induced infections, in particular lipopolysaccharide- induced infections associated with a respiratory viral disease. Consequently, in one embodiment, the primary purpose of the cyclodextrins is not to enhance or improve the delivery or solubility of another active pharmaceutical ingredient.
Active pharmaceutical ingredient
The term "active pharmaceutical ingredient" or “API” refers to any substance or combination of substances used in a finished pharmaceutical product, intended to furnish pharmacological activity or to otherwise have direct effect in the diagnosis, cure, mitigation, treatment, or prevention of disease, or to have direct effect in restoring, correcting or modifying physiological functions in human beings. Preferably, the term “active pharmaceutical ingredient" refers to a molecule that is intended to be biologically active, for example for the purpose of treating inflammatory, autoimmune, or pulmonary disease, disorder, or condition. Even more preferably, the term “active pharmaceutical ingredient" refers to substances whose placing on the market requires a marketing authorization, for example as foreseen under directive 2001/83/EC of 6 November 2001 on the Community Code relating to medicinal products for human use.
Respiratory viruses
Respiratory viruses such as human respiratory syncytial virus and influenza are well known to infect the lower airway, and both can cause bronchitis, bronchiolitis, and pneumonia. Human rhinovirus has traditionally been considered to be an upper airway pathogen because of its association with common cold symptoms.
Examples of respiratory viruses including commonly used abbreviations include:
Figure imgf000009_0001
Figure imgf000010_0001
Diffusing capacity for carbon monoxide - DLCO
In another embodiment, the composition of the present invention is used for the increase and/or recovery of the diffusing capacity for carbon monoxide (DLCO) in the treatment of respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease, in particular SARS-COVID19.
Forced expiratory volume in 1 second - FEV1
In another embodiment, the composition of the present invention is used for the increase and/or recovery of the Forced expiratory volume in 1 second (FEV1 ) in the treatment of respiratory viral diseases or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease, in particular SARS-COVID19.
Forced vital capacity - FVC
In another embodiment, the composition of the present invention is used for the increase and/or recovery of the Forced vital capacity (FVC) in the treatment of respiratory viral diseases or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide- induced inflammation associated with a respiratory viral disease, in particular SARS- COVID19.
Lipopolysaccharides - LPS
In another embodiment, the composition of the present invention is used for the treatment of lipopolysaccharides-associated inflammations. In a preferred embodiment, the LPS- associated inflammations are associated with a respiratory viral disease, in particular SARS-COVID19. Lipopolysaccharide (LPS) is the major component of Gram-negative bacteria cell walls. LPS can cause an acute inflammatory response by triggering the release of a vast number of inflammatory cytokines in various cell types. Therefore, LPS is widely recognized as a potent activator of monocytes/macrophages.
The LPS-associated inflammations may be LPS-produced, LPS-caused or LPS-induced inflammations or infections. The terms LPS-induced, LPS-induced and LPS-produced are used interchangeably. This means that the inflammations of the present invention are directly or indirectly produced by, induced by or induced by the LPS.
Neutrophils
In one embodiment, the composition of the present inventions decreases the neutrophilic inflammation in the respiratory system of subjects/patients exposed to LPS.
Accordingly, another aspect of the invention is the use of the composition of the present invention for treating the neutrophilic inflammation in the respiratory system of subjects/patients exposed to LPS or to LPS-associated inflammations or infections.
Neutrophil extracellular traps - NETs
In another embodiment, the composition of the present invention is used to reduce the neutrophil recruitment into lungs in particular following LPS exposure. This typically characterized by the expression of the specific inflammatory markers such as CXCR4. CXCR-4 is an alpha-chemokine receptor specific for stromal-derived-factor-1 (SDF-1 also called CXCL12). SDF-1 is a molecule endowed with potent chemotactic activity for lymphocytes. CXCR4 neutrophils are susceptible to release neutrophil extracellular traps (NETs).
Accordingly, another aspect of the invention is the use of the composition of the present invention for decreasing the NETs numbers in the lungs of subjects/patients exposed to LPS or to LPS-associated inflammations or infections.
Steroids
Steroids are anti-inflammatory compounds which are commonly also referred to as steroids, corticoids, glucocorticoids, or cortisol analogues. Examples of such corticosteroids include beclomethasone, budesonide, flunisolide, fluticasone, ciclesonide, mometasone, or any compounds comprising the active moiety of any of these corticosteroids, such as salts, derivatives, and prodrugs thereof. In particular corticosteroids selected from the group consisting of fluticasone and budesonide. A preferred corticosteroid is budesonide.
Budesonide, is also named (R,S)- 1 1 (3,16a, 17,21 -tetrahydroxypregna- 1 ,4-diene-3, 20- dione cyclic 16, 17-acetal with butyraidehyde or 16, 17-(butylidenebis(oxy))- 1 1 ,21 - dihydroxy-, (1 1 -[3,16-a)-pregna-l ,4-diene-3, 20-dione. The chemical formula, molecular weight and CAS number for Budesonide are C25H34O6, MW: 430.5 and 51333-22-3, respectively. Budesonide is a racemate consisting of a mixture of the two diastereomers 22 R and 22 S and is provided commercially as a mixture of the two isomers (22R and 22 S). Commercial formulations of budesonide in solution are provided by AstraZeneca LP (Wilmington, Del.) under the trademark Pulmicort.
The molar ratio between steroid, preferably the budesonide or ciclesonide and the HPBCD may vary widely. In some embodiments, the molar ratio is from about 1 :1 to about 1 : 100, preferably from about 1 :1 to 1 :75 and even more preferably from about 1 :1 to about 1 : 50.
In one embodiment, no further active pharmaceutical ingredient than budesonide or ciclesonide is present in the inhalable composition in therapeutically effective amounts for which HPBCD is used to deliver an active pharmaceutical ingredient to other parts of the body than the respiratory system or the bronchial epithelium.
In one embodiment, no further active pharmaceutical ingredient than budesonide is present in the inhalable composition in therapeutically effective amounts for which HPBCD is used to increase the solubility of the further active pharmaceutical ingredient in water.
Further disclaimed substances
In one embodiment, the composition is free of flavonoids agents, such as Citrox. In another embodiment, the composition is free of oils.
Bronchodilators
In some embodiments, the composition does not comprise any bronchodilators such as formoterol. In other embodiments, the composition further comprises bronchodilators such as formoterol.
Effective amount
The terms "effective amount" or "therapeutically effective amount," as used herein, refer to an amount of an active agent as described herein that contributes to achieving one or more desirable clinical outcomes, such as those described in the 'treatment" description above. An appropriate "effective" amount in any individual case may be determined using standard techniques known in the art, such as a dose escalation study. In some embodiments, as used herein, the term "therapeutically effective amount" is meant to refer to an amount of an active agent or combination of agents effective to ameliorate, delay, or prevent the symptoms. Determination of a therapeutically effective amount is well within the capabilities of the skilled person.
Effect
The present inventors have found that the inhalation of a composition comprising a complex of HPBCD and budesonide or ciclesonide is particularly suitable for the direct prevention or treatment of respiratory viral disease. In particular, the present inventors have found that this is the direct effect of the topical administration of a composition comprising a complex of HPBCD and budesonide or ciclesonide to the respiratory system, in particular the nasal mucosa or the pulmonary epithelium. In some embodiments, the composition is only used to treat the lower airways, but not the upper airways.
Whilst other compounds may be present, there is no need for administering further active pharmaceutical ingredients and in particular for a further anti-inflammatory active pharmaceutical ingredient to obtain a positive effect in respect of the respiratory viral disease.
In one embodiment, no further active pharmaceutical ingredient than budesonide or ciclesonide is present in the composition of the present invention, wherein HPBCD is primarily used as solubilizing excipient or as a drug delivery system.
Inhalable aqueous composition
In one embodiment, the composition of the invention is aqueous, which means that it is preferably a liquid composition comprising water as the predominant liquid constituent. Accordingly, in some embodiments, the inhalable composition is a composition comprising HPBCD, water and optionally one or more other components suitable for use in pharmaceutical delivery such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, or excipients, and antimicrobial preservatives. Typically, these components are present in very low amounts, typically in the range of 0,1 to 5 mg/ml.
In another embodiment, the viscosity of the inhalable aqueous composition is in the range of about 0,0,1 mPa.s to about 10 mPa.s, preferably in the range of about 0,5 mPa.s to about 5 mPa.s and most preferably in the range from about 0,8 mPa.s to about 3 mPa.s at 20 °C as measured by the Ell or U.S. Pharmacopoeia (USP) 911 - Viscosity (Capillary Viscometer Methods).
In another embodiment, the composition comprises an effective amount of HPBCD, salt or derivative thereof and a pharmaceutically acceptable carrier. A preferred composition for nebulization comprises HPBCD, NaCI and water.
When administered in aerosol form, it is further preferred for safety and tolerability reasons that water is the only liquid present in the composition. However, in some cases it may be acceptable even for liquids for inhalation to comprise some amount of other liquids, in particular one or more organic solvents having a relatively low inhalation toxicity such as ethanol, propylene glycol, or glycerol. In one embodiment, these liquids are present in very low amounts, typically in the range of 0,1 to 5 mg/ml.
The aqueous composition is suitable for administration as an aerosol. In particular, it is designed for inhalation for the prevention or treatment of respiratory viral disease affecting a region of the upper respiratory tract, such as the nasal mucosa or the paranasal sinuses. In another embodiment, the aqueous composition is designed for inhalation for the prevention or treatment of respiratory viral disease affecting the lower airways, in particular the bronchial system.
The aqueous solution may be a isotonic or hypertonic. A solution is isotonic when its effective osmotic concentration is the same as that of the cytosol inside the cells of the respiratory system. A solution is hypertonic if it has a greater concentration of solutes than the cytosol inside the cells of the respiratory system.
It is further preferred that the pH of the composition is adjusted to 3.5 to 7.5, preferably from 6,5 to 7 and even more preferably from 4,8 to 5,2. The pH in the range from 4,8 to 5,2 may preferably be obtained by adding a pH buffer. Preferable pH buffers are citric acid/citrate/ascorbic acid or citric acid/citrate/EDTA.
In order to adjust the pH, surface tension, viscosity, osmolality, stability, taste and other properties of the composition, one or more further excipients may be used. For example, the composition may comprise one or more excipients selected from pharmaceutically acceptable organic acids, salts of organic acids, inorganic acids, inorganic salts, bases, sugars, sugar alcohols, stabilizers, antioxidants, surfactants, preservatives, and taste masking agents. The composition of the present invention enables aqueous and dry powder formulations whose properties allow highly efficient and convenient aerosol or dry powder delivery using currently available aerosol generating devices, or dry powder inhalers.
Accordingly, a further object of the invention is the use of the composition of the invention for the topical prevention or topical treatment of respiratory viral disease by an aerosol or a micronized powder.
Aerosolization, in the sense of the present invention, means any spraying process that produces droplets. The size of the droplets may vary. Typically, the size of the droplets is between 1 and 100 microns, preferably, between 1 ,5 and 10 microns and even more preferably between 2 and 7 microns.
Micronization, in the sense of the present invention, means any process to decrease the particle size of dry powders to an inhalable size, preferably between 1 and 100 microns, preferably, between 1 ,5 and 10 microns and even more preferably between 2 and 7 microns.
Dry powder inhaler, in the sense of the present invention, means any device suitable for micronization of inhalable dry powder compositions.
Finally, the composition of the present invention may have very little side effects even when administered even at very high dosages, such as daily delivered doses up to 180 mg/kg.
Following examples represent embodiments of the present inventions and are in no ways meant to impact the scope of the present invention.
Amount of HPBCD in the composition
The composition of the present invention may be an aqueous composition or a powder.
In the case of an aqueous solution, the amount or content of the cyclodextrin in the composition is selected to ensure a sufficiently low viscosity at ambient temperature in view of inhalation. The dynamic viscosity, which may also be influenced by the choice and quantity of the further excipients, also has a clear influence on the particle size distribution of the aerosol formed by nebulization and on the efficiency of nebulization. In one embodiment, the viscosity may be adjusted to a range of about 0.8 to about 3 mPas at ambient temperature. In another embodiment, the dynamic viscosity of the composition is in the range of about 0.8 to about 2.5 mPas or even about 0.9 to about 1 .3 mPas at ambient temperature. Typically, the HPBCD concentration in the composition according to the present invention is in the range from about 1 millimolar to about millimolar, preferably from 5 millimolar to about 50 millimolar, and more preferably from about 7 millimolar to about 40 millimolar. Other preferred concentrations range from about 10 millimolar to about 30 millimolar and from about 15 millimolar to about 25 millimolar.
In another embodiment, the HPBCD is administered per inhalation in the amount of 0.1 mg to 30 mg per day, preferably in the amount of 0.5 mg to 20 mg per day, even more preferably in the amount of 1 mg to 10 mg per day.
Inhalable powder
In the case of a powder, the composition preferably is a spray-dried powder.
In another embodiment, the composition is in the form of an inhalable powder, preferably in the form of a spray-dried powder.
In another embodiment, the inhalable composition is in the form of spherical microparticles, wherein a. The microparticles have a median mass aerodynamic diameter of 0.1 microns or more and 5 microns or less; b. The microparticles have a core-shell structure, with the carrier forming the shell and the active pharmaceutical ingredient forming the core; c. The microparticles have a plurality of golf ball-like surface depressions identifiable by scanning electron microscopy; d. The average maximum depth d of the surface depressions (1 ) is 5 % or more and 30 % or less as compared to the average maximum diameter D of the microparticles; and e. 50 surface area% or more as compared to the total surface of the microparticles are depressed.
An exemplary process of manufacturing such spherical microparticles by spraydrying as well as suitable compositions are described in example 1 , in particular in tables 1 and 2 of International patent application WO2021048322A1 to Aquilon Pharmaceuticals. Concentration and daily dosage of cyclodextrin
In one embodiment, the inhalable composition is an aqueous solution and wherein the concentration of HPBCD is from 5 millimolar to 50 millimolar, preferably from 7 millimolar to 40 millimolar, even more preferably from 10 millimolar to 30 millimolar.
In another embodiment, the inhalable composition comprises HPBCD in an amount from 1 mg/ml to 300 mg/ml, preferably from 1 mg/ml to 200 mg/ml, even more preferably from 1 mg/ml to 100 mg/ml.
In another embodiment, the inhalable composition, comprises from about 5 mg/ml to about 80 mg/ml and more preferably from about from 5 mg/ml to about 100 mg/ml, preferably from about 8.00 mg/ml to 60 mg/ml.
In one embodiment the HPBCD is administered per inhalation in the amount of 0.1 mg to 30 mg per day. In another embodiment, the HPBCD is administered per inhalation in the amount of 0.5 mg to 20 mg per day. In another embodiment, the HPBCD is administered per inhalation in the amount of 1 mg to 10 mg per day. In another embodiment, the HPBCD is administered to children aged up to two years per inhalation in the amount of 0.1 mg to 0.5 mg per day. In another embodiment, the HPBCD is administered to children aged from two to 6 years per inhalation in the amount of 0.5 mg to 1 mg per day. In another embodiment, the HPBCD is administered to children aged from 6 years to 14 years per inhalation in the amount of 1 mg to 2 mg per day.
Concentration and daily dosage of corticosteriod
In another embodiment, the inhalable composition comprises budesonide or ciclesonide in an amount from 0,020 mg/ml to 0,70 mg/ml, preferably from about 0,05 mg/ml to about 0,5 mg/ml and more preferably from about 0,1 mg/ml to 0,25 mg/ml.
In another embodiment, the inhalable composition comprises budesonide or ciclesonide in an amount from 0,02 mg/ml to 2 mg/ml, preferably from 0,06 to 1 ,2 mg/ml and more preferably from 0,1 mg/ml to 0,8 mg/ml.
If the composition also comprises a steroid compound, the daily dosage of the steroid compound typically ranges from 1 microgram to 500 micrograms, preferably from about 30 micrograms to about 200 micrograms per day. In some embodiments, the daily dose of the steroid compound is from 50 micrograms to 150 micrograms and even more preferably from 75 micrograms to 125 micrograms. In another embodiment, the budesonide orciclesonide is administered per inhalation in the amount of 0.020 mg to 0,700 mg per day, preferably in the amount of 0.05 mg to 0,5 mg per day, even more preferably in the amount of 0,07 mg to 0,25 per day.
In another embodiment, the HPBCD is administered to children aged up to two years per inhalation in the amount of 0.1 mg to 1 ,0 mg per day and wherein the budesonide or the ciclesonide is administered in an amount of 0,05 mg to 0,12 mg per day.
In another embodiment, the HPBCD is administered to children aged from 2 to 6 years in the amount of 0,05 mg to 0,20 mg per day and wherein the budesonide or the ciclesonide is administered per inhalation in an amount of 0,15 mg to 0,20 mg per day.
In another embodiment, the HPBCD is administered to children aged from 6 to 14 years in the amount of 0,1 mg to 20 mg per day and wherein the budesonide or the ciclesonide is administered per inhalation in an amount of 0,25 mg to 0,5 mg per day.
Rayleigh-jet based inhaler
The inhalable compositions of the present inventions are preferably administered through Rayleigh-jet based inhalers.
In one embodiment, Rayleigh-jet based inhalers for aqueous solutions are commercially available from Medspray. Such nozzle units are based on plain orifice nozzles, creating Rayleigh jets. A 2 micron hole creates a jet, breaking up into mono-disperse 4 micron droplet trains. The diameter of the droplets is twice the size of the orifice. The hole size can be engineered to meet specific requirements of devices. Spray nozzle chips are typically made with technologies generally seen in computer chips.
Short description of the drawings
Figure 1 shows that the DLCO and the functional respiratory tests of Example 9 of the patients who received at least one dose, FCV and FEV1 have been improved in patients using AQ001S, while they have been not at all improved in patients using the placebo (figures 1 to 6). COVID-19 suffering patients do usually not recover to a normal DLCO, see for example Pulmonary function and COVID-19; www.sciencedirect.com; Current Opinion in Physiology 2021 , 21 :29-35; htps://doi.Org/10.1016/j.cophys.2021.03.0051. Therefore, the increase of DLCO is of particular importance.
Figure 2 shows the FEV1 of Example 9 of the patients who received at least one dose.
Figure 3 shows the FVC of Example 9 of the patients who received at least one dose. Figure 4 shows the DLCO of Example 9 of the patients who finished the study.
Figure 5 shows the FEV1 of Example 9 of the patients who finished the study.
Figure 6 shows the FVC of Example 9 of the patients who finished the study.
Figure 7 shows the experimental outline of Example 10. Naive C57bl/6 mice were exposed to low LPS dose (100 ng) intranasally alone or with excipient or distinct AQS doses (2,5 pg, 12,5 pg or 25 pg). Excipient and AQS have been administered together with LPS and also eight hours after LPS administration intranasally. 24 hours after LPS exposure, early innate immune response has been evaluated by flow cytometry (FACS) and Neutrophil Extracellular Traps (NETs) release has been measured by evaluating free extracellular DNA release using a Quant-it picogreen assay.
Figure 8 shows the lung neutrophil numbers following LPS exposure and AQS treatment. Naive mice were exposed to LPS with distinct doses of AQS or the excipient alone administered with LPS and 8 hours following LPS exposure. Lung neutrophil numbers have been evaluated 24 hours after LPS exposure by flow cytometry. P<0,05=*; P<0,01=**; P<0,001=***; P<0, 0001 =****. P values were estimated with a one-way ANOVA with Tukey post hoc test.
Figure 9 shows the release of extracellular DNA in bronchoalveolar lavage fluids (BALF) following LPS exposure and AQS treatment. Naive mice were exposed to LPS alone, LPS and excipient or LPS and 2,5pg AQS administered with LPS and 8 hours following LPS exposure. Release of extracellular DNA in BALF has been evaluated 24 hours after LPS exposure using Quant-it Picogreen assay. P<0,05=*; P<0,01=**; P<0,001 =***; P<0,0001 =****. P values were estimated with a one-way ANOVA with T ukey post hoc test.
Figure 10 highlights the presence of CXCR4 on the Neutrophils population in bronchoalveolar lavage fluids (BALF) following LPS exposure and AQS treatment. Naive mice were exposed to LPS alone, LPS and excipient or LPS and AQS doses (2,5 pg, 12,5 pg or 25 pg) administered with LPS and 8 hours following LPS exposure. CXCR4 staining level in BALF has been evaluated 24 hours after LPS exposure using Flow Cytometry. P<0,05=*; P<0,01 =**; P<0,001 =***; P<0, 0001 =****. P values were estimated with a oneway ANOVA with Tukey post hoc test. Examples
Following examples are given for illustration purposes. However, they are not meant to limit the invention in any way.
Example 1.1 - AQ001S
Figure imgf000020_0001
Example 1.2 - Ciclesonide based AQ003S
Figure imgf000020_0002
Figure imgf000021_0001
Example 2: Evaluation of pharmaceutical compositions from Example 1 or example 7 on the pathophysiology related to viral infection e.g. such as Covid-19 using an experimental in vivo rodent model through a longitudinal study
2.1. Study design:
The longitudinal study consists in a follow up of non-invasively measured indicators before and after viral infection of rodent animals; the study is spread over 16 days. The non- invasive measured parameters are described in the paragraph “measured indicators”. When rodents are sacrificed, blood is collected for analysis of the serological response. Five groups of sex-, age, and weight-matched rodents are compared:
(1) Uninfected rodents only treated with the buffer solution B (see description 2),
(2) Infected rodents treated with buffer solution B (see description 2),
(3) Infected rodents treated with hydroxypropyl-beta-cyclodextrin (HP-beta-CD)-containing aqueous solution of ( see description 2), selected from the compositions 1 to 4 of the Example 1 ,
(4) Infected rodents treated with HP-beta-CD-containing aqueous solution which results from the adequate mixing of solution A and solution B (see description 2 ) selected from the compositions 5 to 12 of the Example 1 (HP-beta-CD with Budesonide in solution),
(5) Infected rodents treated with Monlupiravir under the code name MK-4482, which is a positive reference against SARS-CoV-2 virus. The MK-4482 is administered per os.
Add a summary of protocol on non-rodents and HRSV The size of each experimental group is 10.
The size of each experimental group is 10.
Inoculation:
A virus stock is prepared, titrating 106 TCID50/ml of a SARS-CoV-2 strain. An inoculum consisting of 100 pl of the stock-virus is inoculated to each rodent through the deposition of 50 pl in each nostril. The inoculation is performed under brief general anesthesia using isoflurane. The awakening of the rodents occurs after 90 seconds maximum. The term TCID50 describes the virus quantity which is necessary for the destruction of 50% of the infected cellular colonies.
Inhalation:
Per each day of the experiment, a single inhalation administration of pharmaceutical compositions is tested, preferably through intranasal inhalation for a better reproducibility and better pulmonary deposition. The first inhalation administration is performed 1 h post virus inoculation. Successive administrations of test items are performed at 24 h intervals for 14 consecutive days.
2.2. Measured indicators:
Body weight twice a day, over 16 days (2 days before viral inoculation, 14 days after inoculation).
Clinical score, once a day over 16 days (2 days before viral inoculation, 14 days after inoculation). The observed parameters are summarized in the following table 1 according to the table mentioned in the publication: Sci Rep. 2019; 9: 5919. Published online 2019 Apr 11 . doi: 10.1038/s41598-019^2414^t
Table 1 : Score sheet:
Table 1.1. Observation:
Figure imgf000022_0001
Figure imgf000023_0001
Table 1.2. Examination:
Figure imgf000024_0001
Figure imgf000025_0001
Table 1.3: Evaluation A
Figure imgf000025_0002
Table 1.4: Evaluation B
Figure imgf000025_0003
Table 1.5: Evaluation B
Figure imgf000025_0004
Table 1.6: Evaluation C
Figure imgf000026_0001
Table 1 : Clinical scores (Sci Rep. 2019; 9: 5919. Published online 2019 Apr 11. doi: 10.1038/S41598-019-42414-4)
Respiratory function values, by whole body plethysmography, before inoculation (d-1 ) and 3, 5, 7, and 14 days after inoculation. The term “plethysmography” describes the volume change measurements in different areas of the body.
Description of lung, kidney, heart and brain histological alteration on Hematoxylin&Eosin- stained section on day 14 post inoculation.
SARS-CoV-2 virus detection by immunohistochemistry on day 14 post inoculation.
Lung viral genomic load (RT-qPCR) on day 14 post inoculation.
Plasma anti-SARS-CoV-2 neutralization titer, on day 14 post inoculation.
Example 3. Evaluation of pharmaceutical compositions from Example 1 on the pathophysiology related to viral infection e.g. such as Covid-19 using an experimental in vivo rodent model through a sequential study:
3.1. Study design:
The sequential study consists of a sequential follow-up of the induced COVID-19 disease, with sacrifice of a subgroup of animals every 48 hours until the 10th day after inoculation (5 timepoints). The invasive indicators followed are described in the paragraph “sequential measured indicators” and they are measured at each timepoint.
Five groups of sex-, age, and weight-matched rodents are compared:
(1) Uninfected rodents only treated with the buffer solution B (see description 2),
(2) Infected rodents treated with buffer solution B (see description 2),
(3) Infected rodents treated with HP-beta-CD-containing aqueous solution of ( see description 2), selected from the compositions 1 to 4 of the Example 1 ,
(4) Infected rodents treated with HP-beta-CD-containing aqueous solution which results from the adequate mixing of solution A and solution B (see description 2 ) selected from the compositions 5 to 8 of the Example 1 (HP-beta-CD with Budesonide in solution), (5) Infected rodents treated with Monlupiravir under the code name MK-4482, which is a positive reference against SARS-CoV-2 virus. The MK-4482 is administered per os.
The size of each experimental group is 6.
Inoculation:
A virus stock is prepared, titrating 106 TCID50/ml of a SARS-CoV-2 strain. An inoculum consisting of 100 pl of the stock-virus is inoculated to each rodent through the deposition of 50 pl in each nostril. The inoculation is performed under brief general anesthesia using isoflurane. The awakening of the rodent occurs after 90 seconds maximum.
Inhalation:
Per each day of the experiment, a single inhalation administration of pharmaceutical compositions is tested, preferably through intranasal inhalation for a better reproducibility and better pulmonary deposition. The first inhalation administration is performed 1 h post virus inoculation. Successive administrations of test items are performed at 24 h intervals for 10 consecutive days.
3.2. Sequential measured indicators:
Body weight, twice a day, over 12 days (2 days before and 10 days after viral inoculation)
Clinical score, once a day, over 12 days (2 days before viral inoculation, 10 days after inoculation). The observed parameters are summarized in the above table 1 according to the table mentioned in the publication : Sci Rep. 2019; 9: 5919. Published online 2019 Apr 11 . doi: 10.1038/S41598-019-424144.
Respiratory function values, by whole-body plethysmography, before inoculation (d-1) and 2, 4, 6, 8 and 10 days after viral inoculation.
Lung viral genomic load (RT-qPCR) at each timepoint (i.e. , 2, 4, 6, 8 and 10 days after viral inoculation).
Lung infectious viral load (TCID50) at each timepoint (i.e., 2, 4, 6, 8 and 10 days after viral inoculation).
Lung weights and description of lung histological alterations on Hematoxylin&Eosin- stained sections at each timepoint (i.e., 2, 4, 6, 8 and 10 days after viral inoculation).
SARS-CoV-2 virus detection by immunohistochemistry at each timepoint (i.e., 2, 4, 6, 8 and 10 days after viral inoculation). Follow-up of lung cytokine/chemokine expression, for example IL-1 , IL-2, IL-4, IL-6, IL-10, IL-12p35, TGFb, TNF, IFN et IP10, by RT-qPCR (ELISA or qPCR) at each timepoint (i.e. , 2, 4, 6, 8 and 10 days after viral inoculation).
Example 4 : Evaluation of pharmaceutical compositions from Example 1 or example 1.2 on the pathophysiology related to viral infection e.g. HRSV using an experimental in vivo non-rodent model e.g. Ferret-model:
4.1. Study design:
Virus preparation: Wild-type HRSV subgroup A strain was passaged exclusively in HEp-2 cells.
Ferrets were randomly assigned to treatment groups of 5 animals.
Five groups of sex-, age, and weight-matched ferrets are compared:
(1 ) Uninfected ferrets only treated with the buffer solution B (see description 2), ,
(2) Infected ferrets treated with buffer solution B (see description 2),
(3) Infected ferrets treated with HP-[3-CD-containing aqueous solution selected from the compositions 1 to 4 of the Example 1 (dose 1 ),
(4) Infected ferrets treated with HP-[3-CD-containing aqueous solution which results from the adequate mixing of solution A and solution B (see description 2), selected from the compositions 5 to 8 of the Example 1 (HP-beta-CD with Budesonide in solution) (dose 2),
(5) Infected ferrets treated with an antiviral drug, which is a positive reference against antiviral.
Inoculation:
On day 0, all animals were infected with 105 TCID50 low-passage wild-type HRSV subgroup A by intra-nasal inoculation with a volume of 0.3 mL. Throat and nose swabs are collected daily in a 3-mL virus transport medium, and blood samples are collected -3, -2, 0, 2, 4, 6, 14, and 21 days post injection. Animals are euthanized by exsanguination at 4, 7, or 21 days post injection.
Inhalation: Per each day of the experiment, a single inhalation administration of pharmaceutical compositions is tested, preferably through intranasal inhalation for a better reproducibility and better pulmonary deposition. The first inhalation administration is performed 1 h post virus inoculation. Successive administrations of test items are performed at 24 h intervals for 21 consecutive days.
4.2. Measured indicators:
After collection, nose washes, nose swabs, and throat swabs are processed and infectious virus titers and concentrations of viral RNA are measured by virus isolation and reverse transcription-PCR (RT-PCR).
Samples from right lungs and nasal turbinates as well as from the trachea and bronchus are processed before viral load assessment by virus isolation and quantitative PCR (qPCR). Infectious virus titers in tissue are expressed as Iog10 TCID50 per gram tissue, and infectious virus titer in nose washes and swabs are expressed as log 10 TCID50/mL.
Formalin-fixed tissue sections will be routinely processed, paraffin embedded and sectioned at 3-4 pm, and stained with hematoxylin and eosin for histopathological examination by light microscopy. For immunohistochemistry, additional serial slides are sectioned simultaneously and incubated for 1 h with a goat anti-HRSV-peroxidase (PO).
Measurement of DNA Nets produced by neutophils
Example s: Evaluation of pharmaceutical compositions from Example 1 or example 7 on the pathophysiology related to Influenza A infection using an experimental in vivo mice model:
A variation of the example 2 consists to infect rodents, preferably mice, with Influenza A virus strain A/PR8/34 (H1 N1 ). The virus stock suspension (108 PFU/ml) has been diluted and 50 pl of solution (corresponding to 5 PFU) have been administered to isoflurane - anesthetized mice (Nat lmmunol.2019 November; 20 (11 ): 1444-1455. doi:10.1038/s41590-019-0496-9). The term PFU means Plaque Forming Units and it describes the number of virus particles capable of forming plaques per unit volume. It is a proxy measurement rather than a measurement of the absolute quantity of particles: viral particles that are defective or which fail to infect their target cell will not produce a plaque and thus will not be counted. Another variation over the example 2 will be the timing of sacrifice: some animals being already sacrificed 24 hours post inoculation. Other time points of sacrifice: 2, 4, 6, 8 and 10 days after viral inoculation.
Measured indicators:
■ Body weight, twice a day, over 12 days (2 days before and 10 days after viral inoculation)
■ Clinical score, once a day, over 12 days (2 days before and 10 days after viral inoculation). The observed parameters are summarized in the above table 1 according to the table mentioned in the publication : Sci Rep. 2019; 9: 5919. Published online 2019 Apr 11 . doi: 10.1038/s41598-019-42414-4.
■ Lung weights and description of lung histological alterations on Hematoxylin&Eosin-stained sections at each timepoint (i.e. , 24 hours, 2, 4, 6, 8 and 10 days after viral inoculation).
■ Inflammatory cell subtypes: macrophages (alveolar, interstitial), polynuclear neutrophil, lymphocytes, NK cells, Dendritic cells, granulocytes, NET production at each timepoint (i.e., 24 hours, 2, 4, 6, 8 and 10 days after viral inoculation). DNA nets
Example 6: Evaluation of the efficacy / pharmacodynamics (PD) of pharmaceutical compositions from Example 1 on the symptoms related to Covid-19 infection through a phase 2a clinical trial:
6.1. Purpose:
The objectives of the clinical test are to assess the efficacy/pharmacodynamics and the safety of liquid drug products composed of HP-beta-CD and Budesonide in the treatment of COVID-19 symptoms in hospitalized patients, during and after their hospitalization.
6.2. Methodology:
- Phase 2a, randomized, double-blind, placebo-controlled, parallel, multiple centre, multiple dose clinical study in adults admitted to hospital with a confirmed positive SARS-CoV-2 RT-PCR test.
- Size of the study: A total of 99 adult patients will be enrolled in the trial. - Two (2) doses of pharmaceutical compositions (cf. below) are tested in the clinical study and compared to a placebo inhalation solution.
- Treatments will be given for 28±2 days.
- Two planned interim of the safety and efficacy data will be performed following the first 30 patients and 60 patients.
6.3. Composition: The pharmaceutical composition tested in this clinical trial is composed of 17,5 mg/ml HP-beta-CD and 0,125 mg/ml Budesonide. The liquid composition also comprises antioxidants i.e. EDTA, Citric Acid monohydrate and Trisodium Citrate Dihydrate as a pH adjuster in the range of 4-7. The osmolarity of the solution is adjusted with adequate amounts of Sodium Chloride. Tow dose strengths of this pharmaceutical solution will be tested in the clinical trial. The placebo is Saline e.g. parenteral NaCI 0.9% isotonic solution. The pharmaceutical composition and the placebo are delivered to the patients using inhaler devices e.g. PulmoSprayTM SP and PulmoSprayTM ST softmist inhaler when patient is hospitalized or with PARI BOY® Classic inhalation system when atient is discharged from hospital.
PulmoSprayTM SP is a softmist inhaler device to be used in combination with the Respi Lever DriveTM RP004 when the patient is hospitalized but not under a mechanical ventilation therapy.
PulmoSprayTM ST is a softmist inhaler device to be used without Respi Lever DriveTM RP004 when the patient is hospitalized and under a mechanical ventilation therapy.
6.4.. Measured indicators:
For the efficacy/PD endpoint, the efficacy is assessed by :
COVID-19 clinical progression scale, e.g. a composite scale allowing the analysis of reproducible, non-invasive and widely used well-being metrics describing the clinical course of the disease. The endpoint will be the change from baseline to Day 28 ±2 , and over the dosing period. The term Clinical Progression Scale reflects patient trajectory and resource use over the course of clinical illness (https://doi.org/10.1016/S1473- 3099(20)30483-7).
COVID-19 clinical endpoints from baseline to Day 28 ±2 , and at each visit over the dosing periodchange in Modified Medical Research Council (mMRC) Dyspnea Scale from baseline to Day 28±2, and at each visit over the dosing period. The term (mMRC) dyspnea scale quantifies disability attributable to breathlessness, and is useful for characterizing baseline dyspnea in patients with respiratory diseases. (J C Bestall, E A Paul, R Garrod, R Garnham, P W Jones JAW. Usefulness of the Medical Research Council (MRC) dyspnoea scale as a measure of disability in patients with chronic obstructive pulmonary disease. (Thorax 1999; 54:581-586); https://pubmed.ncbi.nlm.nih.gov/10377201/).
Changes in the pulmonary function assessments from baseline to Day 28±2, and at each visit over the dosing period.
Changes in the diffusion capacity for carbon monoxide from baseline to Day 28±2, and at each visit over the dosing period.
Changes in the pulmonary computer-assisted tomography (CT Scan) between baseline and Day 28±2.
For the efficacy/PD endpoint, the PD is assessed by :
■ Changes in the systemic inflammatory and cardiovascular biomarkers from baseline to Day 28±2, and at each visit over the dosing period.
■ Changes in Immunoglobulin rates from baseline to Day 28±2, and at each visit over the dosing period.
■ The primary endpoints for safety are adverse events (AE) and serious adverse events (SAE), general tolerability, laboratory parameters for hematology, biochemistry and urinalysis, local tolerability, respiratory rate and oxygen saturation.
The exploratory endpoints are:
■ Results of 6-minute walking test (6MWT) at Day 28±2
■ Changes in nocturnal oximetry from baseline to Day 28±2
■ Changes in Saint-Georges’ questionnaire from baseline to Day 28±2
■ Changes in Visual analog scale chest pain from baseline to Day 28±2, and at each visit over the dosing period. The term Visual analog scale (VAS) defines a psychometric response scale which can be used in questionnaires. It is a measurement instrument for subjective characteristics or attitudes that cannot be directly measured. When responding to a VAS item, respondents specify their level of agreement to a statement by indicating a position along a continuous line between two endpoints. ■ Changes in Asthma Control Questionnaire -5 (ACQ-5) quality of life questionnaire from baseline to Day 28±2, and at each visit over the dosing period
■ Measurement of the levels of circulating exosomes in blood from baseline and to Day 28±2, and at each visit over the dosing period.
Example 7
AQ001S Pharmacology in rodent models of acute respiratory inflammation induced by LPS (focus on neutrophil and macrophage cell types)
Naive mice will be exposed to low doses of LPS and treated by intratracheal instillation of AQ001S (different doses). Acute and chronic inflammation parameters will be assessed 24 hours and 7 days after LPS stimulation, respectively.
The following direct PD markers will be used as readouts to assess drug efficacy in this experimental model:
■ Neutrophil numbers and phenotype in lung tissues (analyzed by flow cytometry) (after 24h)
■ NETs release in lungs (analyzed by western blot on lung protein extracts on citrullinated histone 3, or by confocal microscopy on tissue sections) (after 24h)
■ Presence of extracellular DNA or MPO-DNA complexes in BAL (after 24h)
■ Pro-inflammatory cytokine production in total lung supernatant (after 24h)
■ Monocyte, alveolar macrophage and interstitial macrophage numbers and phenotype in lung tissues (analyzed by flow cytometry) (after 24h)
■ Monocyte, alveolar macrophage and interstitial macrophage numbers, phenotype and regulatory functions in lung tissues (analyzed by flow cytometry) (after 7 days)
Ex vivo assessment of anti-inflammatory or pro-inflammatory cytokine secretion by the distinct type of macrophages isolated from lungs of LPS-stimulated mice treated or not with AQ001S.
Inflammation and tissue repair will be evaluated by bi-photon microscopy (to identify collagen deposition), histological examination (7 days).
Investigations will be performed to study acute lung lesions (emphysema, lung inflammation,... ) and their evolution in response to AQ001 S treatment (real-time in vivo data combined with histology data). More precisely, the following imaging outcomes are expected: pCT will help highlight various radiological opacities and disease-specific patterns in lungs with high resolution, both in vivo and ex vivo. These could be further used by RDX to determine and validate new biomarkers for the non-invasive diagnosis of different inflammatory conditions in the lung by means of their Al proprietary processing tools. To obtain high resolution images, respiratory motion should be minimized. To this aim, different strategies will be tested and assessed on pCT for pulmonary gating, i.e. image acquisition synchronized to the animal respiratory cycle.
The use of fluorodeoxyglucose (18F-FDG) and radiolabeled neutrophils as radiotracers in pPET experiments will provide in vivo quantification data over time related to lung inflammation and neutrophil recruitment since both radiolabeled moleculeswill accumulate in inflammatory sites with different kinetics.
Histology parameters will be correlated with imaging data collected throughout the model, with a particular interest in the timing of inflammation development and its response to AQ001 S treatment.
AQ001M solutions might also be tested in these experiments to provide bridging data between the 2 budesonide formulations.
Example 8
AQ001 S Pharmacology in in vitro culture models or in samples recovered from in vivo experiments
The following parameters will be investigated on in vitro models:
■ Neutrophil apoptosis and ROS production (flow cytometry),
■ Expression of NETs markers (myeloperoxidase, neutrophil elastase, peptidyl- arginine deaminase 4, mTOR and MEK/ERK) (qRT-PCR).
■ Dynamic and kinetic analysis of NETs formation and release (real-time fluorescence microscopy)
■ Dynamic pneumocyte (and/or neutrophil) membrane composition and biophysical property modifications (real-time fluorescence confocal or spinning-disk microscopy)
■ Subcellular modifications (high-resolution fluorescence microscopy, electron microscopy) ■ AQ001M solutions might also be tested in these experiments to provide bridging data between the 2 budesonide formulations.
Example 9 - inhalable aqueous composition AQ001S The inhalable aqueous composition AQ001 S had the following composition:
Figure imgf000035_0001
Clinical performance:
The composition AQ001 S was clinically tested in a Phase Ila -Proof of claim randomized double-blind (at hospital), placebo-controlled, parallel, multiple dose clinical trial in adults admitted to hospital with a confirmed positive SARS-CoV-2 RT-PCR test
Study design:
Two regimens of AQ001 S were tested and compared to a placebo inhalation solution. In a hospital setting, 0.4mL AQ001 S were administered by a PulmoSprayTM softmist inhaler. 2mL AQ001 S 0,125mg/ml have been administered using PARI BOY Classic Inhalation System at home, i.e. once patients were discharged from hospital.
Investigational Medicinal Products:
■ Budesonide inhalation solution (AQ001 S) - corticosteroid Placebo inhalation solution (isotonic saline solution 0,9%)
Route of administration: inhalation (nebulization).
Subjects: adult patients (99) admitted to hospital with a positive SARS-CoV-2 test.
In hospital setting (use of PulmoSpray): 0.4mL AQ001 S (0,125mg/ml) have been administered by PulmoSprayTM softmist inhaler. The total Budesonide dose is 0,125x0,4 = 0,05 mg.
■ QID patients received 0,05 mg x 4 = 0,2 mg Budesonide/day
■ BID patients received 0,05 mg x 2 = 0, 1 mg Budesonide/day
■ QID Placebo patients received 0 mg Budesonide/day.
At home (after hospital discharge, using the PARI BOY nebulizer):
■ BID patients received 4,0 ml/day AQ001 S (2 ml morning & 2 ml evening) +4,0 mUday placebo (2 ml mid-morning & 2 ml mid-afternoon).
■ QID patients received 8,0 ml/day AQ001 S
■ QID Placebo patients received 8,0 ml/day Placebo inhalation solution.
The study aimed at:
■ Assessing the safety of AQ001S in the management of acute COVID-19 symptoms.
■ Assessing the efficacy of AQ001 S in the management of acute COVID-19 symptoms.
■ Assessing the effect of AQ001 S on pharmacodynam ic (PD) parameters related to COVID-19.
Duration of subject study participation: 36 ±2 days from screening visit with treatments administered for a duration of 28 (±2) days.
Efficacy parameters:
The efficacy analysis of the clinical study were based on the following parameters:
■ Forced Expiratory Volume in 1 second (FEV1 ): The FEV1 is the amount of the exhaled air during the 1st second of the FVC maneuver. It decreases in case of diseases that obstruct the airway, such as asthma or emphysema. ■ The forced vital capacity (FVC): A spirometry manoeuvre begins with the patient inhaling as deeply as he or she can. Then the patient exhales as long and as forcefully as possible; the amount exhaled in this manner is the FVC.
■ Diffusing capacity for carbon monoxide (DLCO): It is a measure of the ability to transfer gases from the alveoli to the red blood cells through the alveolar epithelium and capillary endothelium.
Safety parameters:
The safety was evaluated collecting the following information on:
■ Adverse Events (AEs) and serious adverse events (SAEs).
■ General tolerability: vital signs, ECG, physical examination.
■ Laboratory parameters: hematology, biochemistry, urinalysis.
■ Safety and tolerability: respiratory rate, 02 saturation.
■ Local tolerability as possible specific AE, related to AQ001 S administration:
- Increased bronchial irritability
- Paradoxical bronchospasm
- Oropharyngeal examination
Efficacy results:
Figures 1 to 6 show that the DLCO and the functional respiratory tests, FCV and FEV1 have been improved in patients using AQ001 S, while they have been improved in patients using the placebo.
The Figures show that the increase of DLCO is of particular importance, as it is well known that COVID-19 suffering patients do not recover a normal DLCO [Pulmonary function and COVID-19; www.sciencedirect.com Current Opinion in Physiology 2021 , 21 :29-35; https://doi.Org/10.1016/j.cophys.2021 .03.005],
For the DLCO there is no value in visit 2. Two Reasons forthat: DLCO involves the patient blowing into a device. The CHU had forbidden this parameter because it did not want caregivers to be contaminated with COVID in the event that saliva particles were released by the patient. The second reason: some patients were unable to take the measurement because of their condition due to covid (difficulty breathing)
Safety assessment: 22 AEs were reported among those 16 AEs were not related to the study drug, 1 AE was possibly related to the study drug, 5 were classified as SAEs
Figure imgf000038_0001
According to the principal investigator, rhabdomyolysis could be explained by fall of the patient, renal insufficiency and statin treatment.
Example 10: AQ001S Pharmacology in rodent models of acute respiratory inflammation induced by LPS (focus on neutrophils
The composition AQ001 S has been tested in Pharmacodynamic (PD) study in rodents to demonstrate its pharmacological performance on acute and chronic models mimicking inflammatory processes induced by respiratory infections.
A robust acute model of neutrophil recruitment into lungs in mice following intranasal administration of low doses of LPS has been used. Following the model, recruited neutrophils have acquired a particular pro-inflammatory phenotype when entering the lung, characterized by their high expression of the specific markers and notably CXCR4. Indeed, CXCR4 neutrophils are susceptible to release neutrophil extracellular traps (NETs).
Material and methods:
Animals: Female wild-type C57BL/6 mice (6 to 8 weeks old) purchased from Janvier laboratories were used. Mice were housed under specific pathogen free (SPF) conditions and maintained in a 12h light-dark cycle with food and water ad libitum. The animals were allocated to 6 experimental groups, with 4 mice per group:
■ PBS,
■ LPS, ■ LPS+HP-beta-CD,
■ LPS+AQ1 S at different doses:
- 2,5 pg (AQ2,5),
- 12,5 pg (AQ12,5)
- 25 pg (AQ25) Route and protocol of administration: PBS (50 pL), LPS (100 ng), vehicle and AQ001S were administered through the intranasal (i.n.) route in isoflurane-anesthetized mice. At TOh, mice were injected i.n. with PBS, LPS, LPS+ HP-b-CD, LPS+AQ2,5, LPS+AQ12,5, LPS+AQ25 in a total volume of 50 pL. At T8h, mice were injected i.n. with vehicle, AQ2,5, AQ12,5, AQ25 in a total volume of 50 pL. At T24h, mice were sacrificed. After 24 hours, levels of double-stranded DNA (dsDNA) were measured in the broncho-alveolar lavage fluids (BALF - as a proxy for NETs) and lungs were digested to perform flow cytometry phenotyping of lung myeloid cells (Figure 7).
The results of Example 10 are shown in Figures 7 to 10.

Claims

1. An inhalable composition comprising a complex of hydroxypropyl-beta- cyclodextrin (HPBCD) or a pharmaceutically acceptable derivative thereof and budesonide or ciclesonide for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease.
2. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of claim 1 , wherein the respiratory virus is selected from the group consisting of adenovirus, human bocavirus, human coronavirus, human metapneumovirus, human parainfluenza virus, human respiratory syncytial virus, human rhinovirus, pharyngoconjunctival fever, and any other virus causing severe acute respiratory syndrome or combinations thereof.
3. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of any one of the preceding claims, wherein a. no further active pharmaceutical ingredient is present in therapeutically effective amounts for which HPBCD is used to deliver an active pharmaceutical ingredient to other parts of the body than the respiratory system or the bronchial epithelium. b. no further active pharmaceutical ingredient is present in therapeutically effective amounts for which HPBCD is used to increase the solubility of the further active pharmaceutical ingredient in water.
4. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of any one of the preceding claims, wherein the inhalable composition comprises HPBCD in an amount from 1 mg/ml to 100 mg/ml, preferably from about 5 mg/ml to about 80 mg/ml and more preferably from about 8.00 mg/ml to 60 mg/ml. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of any one of the preceding claims, wherein the inhalable composition comprises budesonide or ciclesonide in an amount from 0,20 mg/ml to 1 mg/ml, preferably from 0,020 mg/ml to 0,70 mg/ml, preferably from about 0,05 mg/ml to about 0,5 mg/ml and more preferably from about 0,1 mg/ml to 0,25 mg/ml.. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of any one of the preceding claims, wherein the viscosity of the inhalable composition is in the range of about 0,01 mPa.s to about 10 mPa.s, preferably in the range of about 0,5 mPa.s to about 5 mPa.s and most preferably in the range from about 0,8 mPa.s to about 3 mPa.s at 20 °C as measured according to USP. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of any one of the preceding claims, wherein composition is a spray-dried powder. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of any one of the preceding claims, wherein the HPBCD is administered per inhalation in the amount of 0.1 mg to 105 mg per day, or from 0.1 mg to 30 mg per day, preferably in the amount of 0.5 mg to 20 mg per day, even more preferably in the amount of 1 mg to 10 mg per day. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of any one of the preceding claims, wherein the budesonide or the ciclesonide is administered per inhalation in the amount of 0.020 mg to 1 mg per day, or of 0.020 mg to 0,700 mg per day, preferably in the amount of 0.05 mg to 0,5 mg per day, even more preferably in the amount of 0,07 mg to 0,25 per day. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of any one of the preceding claims, wherein the inhalable composition is administered to children aged up to two years per inhalation, wherein the HPBCD is administered in the amount of 0.1 mg to 50 mg per day, or of 0.1 mg to 1 ,0 mg per day and wherein the budesonide or the ciclesonide is administered in an amount of 0,05 mg to 0,12 mg per day. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of any one of the preceding claims, wherein the inhalable composition is administered to children aged from 2 to 6 years per inhalation, wherein the HPBCD is administered in the amount of 0,05 mg to 50 mg per day, or of 0,05 mg to 0,20 mg per day and wherein the budesonide or the ciclesonide is administered per inhalation in an amount of 0,15 mg to 0,50 mg per day, or of 0,15 mg to 0,20 mg per day. The inhalable composition for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease of any one of the preceding claims, wherein the inhalable composition is administered to children aged from 6 to 14 years per inhalation, wherein the HPBCD is administered in the amount of 0,1 mg to 50 mg per day, or of 0,1 mg to 20 mg per day and wherein the budesonide or the ciclesonide is administered per inhalation in an amount of 0,25 mg to 1 mg per day, or of 0,25 mg to 0,5 mg per day. A Rayleigh-jet based inhaler comprising the inhalable composition of any one of the preceding claims for use in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease. Use of a Rayleigh-jet based inhaler for delivering the inhalable composition of any one of claims 1 to 12 in the topical prevention or topical treatment of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease.
PCT/EP2022/086808 2021-12-20 2022-12-19 Inhalable compositions comprising a complex of hpbcd and budesonide or ciclesonide for the treatment or prevention of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease WO2023118014A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
BE20216002A BE1029585B1 (en) 2021-12-20 2021-12-20 INHALABLE COMPOSITIONS COMPRISING A COMPLEX OF HPBCD AND BUDESONIDE OR CICLESONIDE FOR THE TREATMENT OR PREVENTION OF A RESPIRATORY VIRAL DISEASE
BEBE2021/6002 2021-12-20
BE20216105A BE1030002B1 (en) 2021-12-29 2021-12-29 INHABLE COMPOSITIONS COMPRISING A COMPLEX OF HPBCD AND BUDESONIDE OR CICLESONIDE FOR THE TREATMENT OR PREVENTION OF A VIRAL RESPIRATORY DISEASE
BEBE2021/6105 2021-12-29

Publications (1)

Publication Number Publication Date
WO2023118014A1 true WO2023118014A1 (en) 2023-06-29

Family

ID=84923307

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/086808 WO2023118014A1 (en) 2021-12-20 2022-12-19 Inhalable compositions comprising a complex of hpbcd and budesonide or ciclesonide for the treatment or prevention of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease

Country Status (1)

Country Link
WO (1) WO2023118014A1 (en)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0423240B1 (en) 1988-07-07 1995-11-08 The Trustees Of The University Of Pennsylvania Modulation of virus-host cell interactions using cyclic oligosaccharides
EP1799231A1 (en) 2004-10-10 2007-06-27 Université de Liège Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases
EP1894559A1 (en) 2006-09-01 2008-03-05 PARI Pharma GmbH Means to solubilise steroids for inhalation
EP3151836A2 (en) 2014-03-28 2017-04-12 Université de Liège Composition comprising cyclodextrin and budesonide derivatives for the treatment and prophylaxis of pulmonary inflammations
WO2019067145A1 (en) 2017-08-28 2019-04-04 Asdera Llc Use of cyclodextrins in diseases and disorders involving phospholipid dysregulation
WO2021001349A1 (en) 2019-07-01 2021-01-07 Aquilon Pharmaceuticals Inhalable composition comprising cyclodextrin for use in the treatment of nasal inflammations
WO2021048322A1 (en) 2019-09-10 2021-03-18 Aquilon Pharmaceuticals Golf ball-like microparticles for use in the treatment and prevention of pulmonary diseases
US20210346613A1 (en) * 2020-05-05 2021-11-11 Mahesh Kumar KHAITAN Controlled delivery device for treating coronavirus infections and methods thereof

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0423240B1 (en) 1988-07-07 1995-11-08 The Trustees Of The University Of Pennsylvania Modulation of virus-host cell interactions using cyclic oligosaccharides
EP1799231A1 (en) 2004-10-10 2007-06-27 Université de Liège Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases
EP1894559A1 (en) 2006-09-01 2008-03-05 PARI Pharma GmbH Means to solubilise steroids for inhalation
EP3151836A2 (en) 2014-03-28 2017-04-12 Université de Liège Composition comprising cyclodextrin and budesonide derivatives for the treatment and prophylaxis of pulmonary inflammations
WO2019067145A1 (en) 2017-08-28 2019-04-04 Asdera Llc Use of cyclodextrins in diseases and disorders involving phospholipid dysregulation
WO2021001349A1 (en) 2019-07-01 2021-01-07 Aquilon Pharmaceuticals Inhalable composition comprising cyclodextrin for use in the treatment of nasal inflammations
WO2021048322A1 (en) 2019-09-10 2021-03-18 Aquilon Pharmaceuticals Golf ball-like microparticles for use in the treatment and prevention of pulmonary diseases
US20210346613A1 (en) * 2020-05-05 2021-11-11 Mahesh Kumar KHAITAN Controlled delivery device for treating coronavirus infections and methods thereof

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
CARROUEL: "COVID-19: A Recommendation to Examine the Effect of Mouthrinses with β-Cyclodextrin Combined with Citrox in Preventing Infection and Progression", J. CLIN. MED., vol. 9, 2020, pages 1126
CURRENT OPINION IN PHYSIOLOGY, vol. 21, 2021, pages 29 - 35, Retrieved from the Internet <URL:https://doi.ora/10.1016/i.cophys.2021.03.005>
FLORENCE CARROUEL ET AL: "COVID-19: A Recommendation to Examine the Effect of Mouthrinses with b-Cyclodextrin Combined with Citrox in Preventing Infection and Progression", JOURNAL OF CLINICAL MEDICINE, vol. 9, no. 4, 15 April 2020 (2020-04-15), pages 1126, XP055759526, DOI: 10.3390/jcm9041126 *
J C BESTALLE A PAULR GARRODR GARNHAMP W JONES JAW: "Usefulness of the Medical Research Council (MRC) dyspnoea scale as a measure of disability in patients with chronic obstructive pulmonary disease", THORAX, vol. 54, 1999, pages 581 - 586, Retrieved from the Internet <URL:https://pubmed.ncbi.nlm.nih.gov/10377201>
NAT IMMUNOL, vol. 20, no. 11, November 2019 (2019-11-01), pages 1444 - 1455
SCI REP, vol. 9, 11 April 2019 (2019-04-11), pages 5919

Similar Documents

Publication Publication Date Title
JP5247265B2 (en) Use of mometasone furoate for the treatment of airway channels and lung diseases
Newman et al. Lung deposition of fenoterol and flunisolide delivered using a novel device for inhaled medicines: comparison of RESPIMAT with conventional metered-dose inhalers with and without spacer devices
EP3332767B1 (en) Liquid inhalation formulation comprising rpl554
ES2340511T3 (en) USE OF CYCLODEXTRINE FOR THE TREATMENT AND PREVENTION OF BRONCHIAL INFLAMMATORY DISEASES.
Madney et al. Nebulizers and spacers for aerosol delivery through adult nasal cannula at low oxygen flow rate: an in-vitro study
KR20010042531A (en) Pulmonary and Nasal Delivery of Raloxifene
JP2023526547A (en) Certain drug systems, methods, and uses for reducing viral replication in airway mucosa
JP7359519B2 (en) Drugs for the prevention or treatment of rhinovirus infections
WO2021168173A1 (en) Methods and compositions for treating viral respiratory infections
EP3892275A1 (en) Aerosolization of hcq or its metabolites for the treatment of lung infections
WO2023118014A1 (en) Inhalable compositions comprising a complex of hpbcd and budesonide or ciclesonide for the treatment or prevention of a respiratory viral disease or lipopolysaccharide-induced inflammation, in particular lipopolysaccharide-induced inflammation associated with a respiratory viral disease
Terzano et al. Beclomethasone dipropionate versus budesonide inhalation suspension in children with mild to moderate persistent asthma
EP4232073A1 (en) New dosage regimen for inhaled vasoactive intestinal polypeptide
CN116916889A (en) Inhalation-type imatinib for lung high-pressure field
WO2010009288A1 (en) Compositions and uses of antiviral active pharmaceutical agents
Gümrü et al. Oral candidiasis as a local adverse effect of inhaled corticosteroids: what the dental practitioner should know
US20220168297A1 (en) Methods and compositions for treating chronic obstructive pulmonary disease, asthma, pneumonia, bronchitis, cystic fibrosis, pulmonary edema, interstitial lung disease, sarcoidosis, idiopathic pulmonary fibrosis, acute respiratory distress syndrome, and pulmonary arterial hypertension
KR20180030399A (en) Composition for nebulizers
US20240009227A1 (en) Use of a heparin composition in the treatment of viral lung diseases, acute and/or chronic lung diseases by soft mist inhaler or vibration mesh technology nebulizer through inhalation route
WO2021211923A1 (en) Compositions and methods for treating disease
Cabrera et al. Influence of mesh nebulizer characteristics on aerosol delivery in non-human primates
JP2023525662A (en) Medicines and nasal sprays containing trehalose or derivatives of trehalose
ES2942710T3 (en) Composition comprising cyclodextrin and budesonide derivatives for use in the treatment and prophylaxis of pulmonary inflammations
EP4274552A1 (en) Genistein treatment of inflammatory and immunological disorders
CN116744934A (en) Inhaled statins for the treatment of viral respiratory diseases

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: 22840643

Country of ref document: EP

Kind code of ref document: A1