WO2025006870A1 - Novel formulations - Google Patents

Novel formulations Download PDF

Info

Publication number
WO2025006870A1
WO2025006870A1 PCT/US2024/035999 US2024035999W WO2025006870A1 WO 2025006870 A1 WO2025006870 A1 WO 2025006870A1 US 2024035999 W US2024035999 W US 2024035999W WO 2025006870 A1 WO2025006870 A1 WO 2025006870A1
Authority
WO
WIPO (PCT)
Prior art keywords
aqueous liquid
liquid pharmaceutical
pharmaceutical formulation
polynucleotide molecule
acid
Prior art date
Application number
PCT/US2024/035999
Other languages
French (fr)
Inventor
Kazuhiro Ito
Jagdeep SHUR
William Garth Rapeport
Radhakrishnan P. Iyer
Susan SOBOLOV
Original Assignee
Rigimmune Inc.
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 PCT/US2024/030546 external-priority patent/WO2024243293A1/en
Application filed by Rigimmune Inc. filed Critical Rigimmune Inc.
Publication of WO2025006870A1 publication Critical patent/WO2025006870A1/en

Links

Classifications

    • 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
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • 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
    • A61K47/02Inorganic compounds
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles

Definitions

  • the invention also relates inter alia to aqueous liquid pharmaceutical formulations for use as a medicament, for example for use in the treatment or prevention of viral infection and disease associated with viral infection, and related methods of treatment.
  • a therapeutic agent is delivered to the target tissues and/or cells of a subject in an amount that allows said therapeutic agent to exert a therapeutic effect. Indeed, insufficient delivery of a therapeutic agent often impedes the ability of an otherwise efficacious therapeutic agent to achieve desired therapeutic outcomes.
  • biologic therapeutics such as immunoglobulins and polynucleotide molecules, in particular genomic DNA (gDNA), complementary DNA (cDNA), mRNA, siRNA, and shRNA, whose macromolecular nature and net ionic charge add additional complexities to targeted delivery.
  • biologics do not undergo passive diffusion across cell membranes.
  • polynucleotide molecules are susceptible to degradation by endonuclease and exonuclease enzymes which can hydrolyse the phosphodiester bonds of the polynucleotide backbone.
  • polynucleotide therapeutics are dependent on delivery technologies that improve the stability of the polynucleotide, facilitate efficient internalisation, and increase target affinity (Kulkarni et al.2021).
  • ASOs chemically-modified antisense oligonucleotides
  • GalNAc N- acetylgalactosamine
  • AAV adeno-associated virus
  • lipid delivery systems including lipid nanoparticles (LNPs) and liposomes (Kulkarni et al.2021).
  • ASOs refer to polynucleotide molecules comprising a number of chemical modifications to the backbone, sugar moiety, or nitrogenous base to enhance affinity to target RNA, improve nuclease resistance and modulate the immunological profile of the polynucleotide (Khvorova and Watts, 2017).
  • GalNAc conjugation facilitates accumulation of relevant polynucleotide therapeutics, particularly in the liver.
  • the GalNAc construct targets the asialoglycoprotein receptor which is predominantly expressed on liver hepatocytes. Subsequent internalisation via clathrin-mediated endocytosis, and endosomal escape by the polynucleotide molecule provides targeted delivery (Springer and Dowdy, 2018).
  • AAV vector systems are comparably simple and may provide efficient delivery of polynucleotide molecule to the nucleus.
  • different AAVs display different cell tropisms, such that the platform technology can be adapted to different target tissues or cells.
  • lipid delivery systems including lipid nanoparticles and liposomes, have increasingly become recognised as the most promising delivery system for polynucleotide molecules.
  • biocompatibility of lipid-based formulations and their respective ease of manufacturing, particularly at a large-scale has made such formulations an attractive avenue for research and development.
  • lipid delivery systems are highly efficient in delivering a polynucleotide molecule into a target cell. Nevertheless, lipid delivery systems are associated with a number of problems.
  • lipid delivery systems such as lipid nanoparticles and liposomes, frequently include a polyethylene glycol (PEG)-based compound which prevents the aggregation, and subsequent immune recognition and elimination, of lipid particles (Jokerst et al.2011). Indeed, PEGylated lipid particles appear to have an increased half-life in the circulation (Huang and Liu, 2011).
  • PEGylated lipid nanoparticles have been reported to severely inhibit endosomal release of polynucleotide molecules (Song et al.
  • Lipid delivery systems comprising polynucleotide molecules must be administered to a subject via a route that is tolerable to the subject, supporting patient compliance, and that ensures the polynucleotide molecule is delivered to the target tissue or cells at a concentration suitable for exerting a therapeutic effect.
  • lipid delivery systems comprising polynucleotide molecules are administered intravenously, for example to facilitate systemic administration, or via local injection to a target tissue or organ, for example via the intradermal, subcutaneous, intra-ocular, intramuscular, intra-myocardial or intra-tumoral route.
  • lipid delivery systems comprising polynucleotide molecules are less commonly administered topically to the lung or nose (Li et al.
  • lipid delivery systems comprising polynucleotide molecules to the respiratory tract, in particular via topical administration to the lung or nose, are associated with a number of well-documented challenges.
  • a lipid delivery system comprising a polynucleotide molecule must be administered intranasally, or via inhalation, in a manner that ensures a therapeutically effective dose is provided in the region of the respiratory tract where the therapeutic effect is desired, e.g. the upper or lower respiratory tract. Moreover, administration should occur within a time frame that supports high patient compliance. Furthermore, regulatory requirements provide that lipid nanoparticle and liposome drug products are subject to additional considerations such as vesicle or particle size, size distribution and morphology (FDA Guidance for Industry entitled “Liposome Drug Products Chemistry, Manufacturing, and Controls; Human Pharmacokinetics and Bioavailability; and Labelling Documentation”, 2018).
  • lipid nanoparticles and liposomes are susceptible to fusion (i.e., combination of smaller lipid particles into larger lipid particles), aggregation, and leakage of the contained polynucleotide molecule, each of which can be detrimental to the stability of the contained polynucleotide molecule.
  • fusion i.e., combination of smaller lipid particles into larger lipid particles
  • leakage of the contained polynucleotide molecule
  • aqueous liquid pharmaceutical formulation comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule.
  • the present invention further provides an aqueous liquid pharmaceutical formulation
  • a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule for use as a medicament, for example for use in the in the treatment or prevention of viral infection and disease associated with viral infection.
  • a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant
  • a polynucleotide molecule for use as a medicament, for example for use in the in the treatment or prevention of viral infection and disease associated with viral infection.
  • such formulations are, for example, suitable for topical administration to the lung or nose, as well as other routes.
  • the formulations of the present invention suitably form a stable colloidal emulsion.
  • Figure 1 shows the effect of apical treatment with vehicle (water alone), a polynucleotide molecule (shRNA 1) in vehicle (water alone), or with a surfactant component in vehicle at three different concentrations (surfactant component 1: 0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80; surfactant component 2: 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80; and surfactant component 3: 0.2% (w/w) oleic acid and 0.15% (w/w) polysorbate 80) on CXCL10 release in the basal chamber from air-liquid interface (ALI) cultured nasal epithelium on days 1, 2 and 3 post-treatment.
  • surfactant component 1 0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80
  • surfactant component 2 0.05% (w/w) oleic acid and 0.045% (w/w) polysorb
  • Figure 2 shows the effect of apical treatment with vehicle (water alone) a polynucleotide molecule (shRNA 1) in vehicle (water alone), or with a surfactant component in vehicle at two different concentrations (0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80; and 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) on CXCL10 release in an apical wash from ALI cultured nasal epithelium on day 1 post-treatment (i.e. prior to virus infection). The effects of these treatments are compared to basolateral treatment with oseltamivir.
  • shRNA 1 polynucleotide molecule
  • Figure 3 shows the effect of apical treatment with vehicle (water alone), a polynucleotide molecule (shRNA 1) in vehicle (water alone), or with a surfactant component in vehicle at two different concentrations (0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80; and 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) on viral load in an apical wash from influenza virus infected ALI cultured nasal epithelium on day 2 post-infection (i.e. day 3 post- 4 treatment).
  • the effects of these treatments are compared to basolateral treatment with oseltamivir.
  • Figure 4 shows the effect of apical treatment with vehicle (water alone), a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) alone in vehicle (water) , a polynucleotide molecule (shRNA 1) in vehicle (water), or a polynucleotide molecule (shRNA 1) with said surfactant component in vehicle (water), on CXCL10 release in an apical wash from ALI cultured nasal epithelium on day 1 post-treatment (i.e. prior to virus infection).
  • the effects of these treatments are compared to basolateral treatment with oseltamivir.
  • Figure 5 shows the effect of apical treatment with vehicle (water alone), a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) alone in vehicle, and a polynucleotide molecule (shRNA 1) in vehicle or a polynucleotide molecule (shRNA 1) with said surfactant component in vehicle, on viral load in an apical wash from influenza virus infected ALI cultured nasal epithelium on day 2 post-infection (i.e. day 3 post-treatment).
  • the effects of these treatments are compared to basolateral treatment with oseltamivir.
  • Figure 6 shows the effect of apical treatment of dsRNA 1 or dsRNA 2 in vehicle (buffer: 0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate), or with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, on CXCL10 release in an apical wash from ALI cultured nasal epithelium on day 1 post-treatment (i.e. prior to virus infection).
  • vehicle buffer: 0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate
  • a surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • Figure 7 shows the effect of apical treatment of dsRNA 1 or dsRNA 2 in vehicle (buffer: 0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate), or with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, on viral load in an apical wash from influenza virus infected ALI cultured nasal epithelium on day 1 post- infection (i.e. day 2 post-treatment).
  • vehicle buffer: 0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate
  • a surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • Figure 8 shows the effect of intranasal treatment with vehicle (water alone), with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle together with further pharmaceutically acceptable excipients (see Formulation Example 1A minus the shRNA 1, hereinafter “surfactant formulation”), and a polynucleotide molecule (shRNA 1) in vehicle, or in the above described surfactant formulation (see Formulation Example 1A, hereinafter “shRNA 1 with surfactant formulation”), on viral load in nasal tissue from influenza (PR8) infected mice on day 1 and day 5 post-infection. The effects of these treatments are compared to oral treatment with oseltamivir phosphate.
  • surfactant formulation a polynucleotide molecule
  • Figure 9 shows the effect of intranasal treatment with vehicle (water alone), with a formulation comprising a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) and further pharmaceutically acceptable excipients in vehicle (see Formulation Example 1A minus the shRNA 1, hereinafter “surfactant formulation”), and a polynucleotide molecule (shRNA 1) in vehicle, or in the above described surfactant formulation (see Formulation 5 Example 1A, hereinafter “shRNA 1 with surfactant formulation”), on neutrophil accumulation in the nasal lavage of influenza (PR8) infected mice on Day 1 and Day 5 post-infection.
  • a surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • surfactant formulation a polynucleotide molecule
  • Figure 11 shows the effect of apical treatment with a GFP-encoding mRNA in a buffer (0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate), or with said GFP- encoding mRNA with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in said buffer, on mRNA exposure and consequently on GFP signal in ALI cultured nasal epithelial cells 24 hrs post-treatment.
  • a buffer 0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate
  • a surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • Figure 12 shows the effect of apical treatment with a ⁇ -galactosidase-encoding ( ⁇ -gal) plasmid in a buffer (0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate), or with said ⁇ -gal plasmid with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in said buffer, on plasmid exposure and consequently on ⁇ - galactosidase enzyme activity in ALI cultured nasal epithelial cells 24 hrs post-treatment.
  • a buffer 0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate
  • a surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • Figures 13 (A-D) show the effect of the presence of a surfactant component of three different concentrations (surfactant component 1: 0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80; surfactant component 2: 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80; and surfactant component 3: 0.2% (w/w) oleic acid and 0.15% (w/w) polysorbate 80) on the polydispersity and particle size of formulations comprising a polynucleotide molecule (shRNA 1) in vehicle (water).
  • shRNA 1 polynucleotide molecule
  • Figure 14 shows the effect of apical treatment with vehicle (buffer alone), a polynucleotide molecule (shRNA 1) in buffer, a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) or shRNA 1 with surfactant component in vehicle, on CXCL10 release in an apical wash from ALI cultured nasal epithelium on day 1 post-treatment (i.e. prior to virus infection).
  • vehicle buffer alone
  • shRNA 1 polynucleotide molecule
  • surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • Figure 15 shows the effect of apical treatment, on day 1 and on day 0 pre-infection, with vehicle (buffer alone), a polynucleotide molecule (shRNA 1) in buffer, a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) or shRNA 1 with surfactant component in vehicle, on viral load in an apical wash from human rhinovirus (HRV16) infected ALI cultured nasal epithelium on day 2 post-infection (i.e. day 3 post-first treatment).
  • vehicle buffer alone
  • shRNA 1 polynucleotide molecule
  • a surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • shRNA 1 with surfactant component in vehicle on viral load in an apical wash from human rhinovirus (HRV16) infected ALI cultured nasal epithelium on day 2 post-infection (i.e.
  • FIG 16 shows the effect of apical treatment, on day 1 and on day 0 pre-infection, with vehicle (buffer alone), a polynucleotide molecule (shRNA 1) in buffer, a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) or shRNA 1 with surfactant component in vehicle, on viral load in an apical wash from respiratory syncytial virus (RSV) A2 infected ALI cultured nasal epithelium on day 3 post-infection (i.e. day 4 post-first treatment).
  • vehicle buffer alone
  • shRNA 1 polynucleotide molecule
  • a surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • shRNA 1 with surfactant component in vehicle
  • Figure 17 shows the effect of apical treatment, on day 1 and on day 0 pre-infection, with vehicle (buffer alone), a polynucleotide molecule (shRNA 1) in buffer, a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) or shRNA 1 with surfactant component in vehicle, on cell integrity, as represented by transepithelial electrical resistance (TEER), in a respiratory syncytial virus (RSV) A2 infected ALI cultured nasal epithelium model, on days 0, 1, 2 and 3 post-infection.
  • vehicle buffer alone
  • shRNA 1 polynucleotide molecule
  • a surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • shRNA 1 with surfactant component in vehicle
  • Figure 18 shows the effect of intranasal treatment with vehicle (saline alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, or a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, on viral load in lung tissue from respiratory syncytial virus (RSV) A2 infected mice on day 4 post-infection.
  • RSV respiratory syncytial virus
  • Figure 19 shows the effect of intranasal treatment with vehicle (saline alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, or a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, on neutrophil accumulation in the nasal lavage of respiratory syncytial virus (RSV) A2 infected mice on day 4 post-infection.
  • RSV respiratory syncytial virus
  • Figure 20 shows the effect of intranasal treatment with vehicle (saline alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, or a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, on body weight loss observed in respiratory syncytial virus (RSV) infected mice during the four days post-infection.
  • vehicle saline alone
  • surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • shRNA 1 polynucleotide molecule
  • Figure 21 shows the effect of subcutaneous treatment with vehicle (saline alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer, or a polynucleotide molecule (shRNA 1) with surfactant component (2 or 20 mg/mL) in phosphate buffer, on CXCL10 release in the serum of mice 24 hrs post-treatment.
  • Figure 22 shows the effect of subcutaneous treatment with vehicle (saline alone; i.e.
  • FIG. 24 shows the effect of subcutaneous treatment with vehicle (saline alone; i.e.
  • FIG. 25 shows the effect of subcutaneous treatment with vehicle (saline alone; i.e.
  • surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • vehicle a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, surfactant component in vehicle in combination with oral oseltamivir treatment, or shRNA 1 with surfactant component in vehicle in combination with oral oseltamivir treatment, on neutrophil accumulation in the nasal tissue of influenza (PR8) infected mice on day 5 post- infection.
  • shRNA 1 polynucleotide molecule
  • Figure 26 shows the effect of subcutaneous treatment with vehicle (saline alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, surfactant component in vehicle in combination with oral oseltamivir treatment, or shRNA 1 with surfactant component in vehicle in combination with oral oseltamivir treatment, on body weight loss observed in influenza (PR8) infected mice during the five days post-infection.
  • Figure 27 shows the effect of intranasal vaccination with vehicle (i.e.
  • H1N1 haemagglutinin
  • rH1N1 HA in vehicle following previous treatment with a polynucleotide molecule (shRNA 1) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or rH1N1 HA in vehicle following previous treatment with the control adjuvant, CPG-ODN, on viral load in the lung tissue of influenza (PR8) infected mice on day 5 post-infection.
  • Figure 28 shows the effect of intranasal vaccination with vehicle (i.e.
  • H1N1 haemagglutinin
  • rH1N1 HA in vehicle following previous treatment with a polynucleotide molecule (shRNA 1) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or rH1N1 HA in vehicle following previous 8 treatment with the control adjuvant, CPG-ODN, on viral load in the nasal tissue of influenza (PR8) infected mice on day 5 post-infection.
  • Figure 29 shows the effect of intranasal vaccination with vehicle (i.e.
  • H1N1 haemagglutinin
  • rH1N1 HA in vehicle following previous treatment with a polynucleotide molecule (shRNA 1) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or rH1N1 HA in vehicle following previous treatment with the control adjuvant, CPG-ODN, on neutrophil accumulation in the lung tissue of influenza (PR8) infected mice on day 5 post-infection.
  • Figure 30 shows the effect of intranasal vaccination with vehicle (i.e.
  • H1N1 haemagglutinin
  • rH1N1 HA in vehicle following previous treatment with a polynucleotide molecule (shRNA 1) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or rH1N1 HA in vehicle following previous treatment with the control adjuvant, CPG-ODN, in vehicle, on neutrophil accumulation in the nasal tissue of influenza (PR8) infected mice on day 5 post-infection.
  • Figure 31 shows the effect of intranasal vaccination with vehicle (i.e.
  • H1N1 haemagglutinin
  • rH1N1 HA in vehicle following previous treatment with a polynucleotide molecule (shRNA 1) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or rH1N1 HA in vehicle following previous treatment with the control adjuvant, CPG-ODN, on body weight loss observed in influenza (PR8) infected mice during the five days post-infection.
  • shRNA 1 polynucleotide molecule
  • CPG-ODN control adjuvant
  • Figure 32 shows the effect of apical treatment with a GFP-encoding mRNA in a buffer (0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate), or with said GFP- encoding mRNA with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in said buffer, on mRNA exposure and consequently on GFP signal in ALI cultured nasal epithelial cells 24 hrs post-treatment.
  • Figures 33 and 34 show the effect of apical treatment of ALI cultured nasal epithelium with H1N1 haemagglutinin (HA) mRNA in vehicle (i.e.
  • Figure 36 shows the effect of apical treatment of ALI cultured nasal epithelium with both a low- shear mixed (i.e. magnetically stirred) and high-shear mixed formulation comprising vehicle 9 (i.e.
  • citrate or phosphate buffer alone surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in citrate or phosphate buffer, or a polynucleotide molecule (shRNA 1) with surfactant component in citrate or phosphate buffer, on viral load in an apical wash from influenza virus (PR8) infected ALI cultured nasal epithelium on day 2 post-infection (i.e. day 3 post-treatment).
  • shRNA 1 polynucleotide molecule
  • Figure 37 shows the effect of apical treatment of ALI cultured corneal epithelium with Cy3 labelled siRNA in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate), or with said Cy3 labelled siRNA with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in said phosphate buffer, on siRNA exposure and consequently on fluorescent signal in ALI cultured corneal epithelial cells 4 hrs post-treatment.
  • phosphate buffer 0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate
  • a surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • Figure 38 shows the effect of apical treatment of ALI cultured corneal epithelium with a GFP- encoding mRNA in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate), or with said GFP-encoding mRNA with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in said phosphate buffer, on mRNA exposure and consequently on GFP signal in ALI cultured corneal epithelial cells 72 hrs post-treatment.
  • phosphate buffer 0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate
  • a surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • Figure 39 shows the effect of apical treatment of surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in buffer, shRNA 1 in buffer, RNA Conjugate 1 in buffer, RNA Conjugate 1 with surfactant component in buffer, RNA Conjugate 2 in buffer, and RNA Conjugate 2 with surfactant component in buffer, on CXCL10 release in an apical wash from ALI cultured nasal epithelium on day 1 post-treatment
  • Figures 40 to 44 show the effect of the presence of a surfactant component, wherein said surfactant component is polysorbate 80 (0.045% (w/w)) + caprylic acid (0.05% (w/w)), polysorbate 80 (0.045% (w/w)) + oleic acid (0.05% (w/w)), Brij 35 (0.045% (w/w)) + oleic acid (0.05% (w/w)), Brij 35 (0.45% (w/w)) + capry
  • Figure 46 shows differential scanning calorimetry thermographs for formulations of the present invention which comprise a surfactant component, in particular polysorbate 80 (0.045% (w/w)) + oleic acid (0.05% (w/w)), including a formulation comprising surfactant component only (top 10 panel), a formulation comprising 2 mg/mL polynucleotide molecule (shRNA 1) with surfactant component (middle panel), and a formulation comprising 20 mg/mL polynucleotide molecule (shRNA 1) with surfactant component (bottom panel).
  • a surfactant component in particular polysorbate 80 (0.045% (w/w)) + oleic acid (0.05% (w/w)
  • Figures 47 to 49 show the effect of apical treatment with media, surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer, shRNA 1 with surfactant component in phosphate buffer, or pleconaril, a known human rhinovirus (HRV) inhibitor, in media, on CXCL10, CXCL8, and CCL5 (respectively, Figures 47 to 49) release in an apical wash from HRV-infected, ALI cultured bronchial epithelium from an asthma donor on day 5 post-virus inoculation.
  • RSV human rhinovirus
  • RNA or DNA molecules to cells upon formulation with specific surfactant components.
  • the present invention is further based on discoveries made by testing the exposure and therefore anti-viral activity of diverse anti-viral polynucleotide molecules, e.g. RNA or DNA molecules, in combination with specific surfactant components.
  • an apically administered formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with one of a number of diverse anti-viral polynucleotide molecules, for example a shRNA molecule, dsRNA 1, dsRNA 2, RNA Conjugate 1, or RNA Conjugate 2, has a potent effect in stimulating the innate immune response, as a result of improved polynucleotide molecule delivery, as determined by the production of CXCL10, a surrogate marker of anti-viral interferon (IFN) signalling, in air-liquid interface (ALI) cultured nasal epithelium (see Biological Examples 1 to 4 and 15, and Figures 1, 2, 4, 6, and 39); (ii) an apically administered formulation comprising a surfactant component comprising a mixture of a fatty acid,
  • a surfactant component comprising a mixture of a
  • the aqueous liquid pharmaceutical formulations of the invention comprise a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant.
  • Fatty Acid refers to a carboxylic acid molecule comprising a carboxylic acid group attached to an aliphatic hydrocarbon “tail”, which is typically between 4 and 24 carbon atoms in length.
  • the aliphatic hydrocarbon “tail” may be between 4 and 22, such as between 4 and 20, such as between 4 and 18, such as between 4 and 16, such as between 4 and 14, such as between 4 and 12, such as between 4 and 10, such as between 4 and 8, such as between 4 and 6, carbon atoms in length.
  • the aliphatic hydrocarbon “tail” may be between 6 and 24, such as between 8 and 24, such as between 10 and 24, such as between 12 and 24, such as between 14 and 24, carbon atoms in length.
  • the aliphatic hydrocarbon “tail” may be between 6 and 22, such as between 6 and 20, such as between 8 and 20, such as between 8 and 18, such as between 10 and 18, carbon atoms in length.
  • the aliphatic hydrocarbon “tail” is between 4 and 6 carbon atoms in length i.e. the fatty acid is a short-chain fatty acid such as butyric acid (4 carbon atoms).
  • the aliphatic hydrocarbon “tail” is between 6 and 12 carbon atoms in length i.e.
  • the fatty acid is a medium-chain fatty acid such as caprylic acid (8 carbon atoms) and capric acid (10 carbon atoms).
  • the aliphatic hydrocarbon “tail” is between 14 and 24 carbon atoms in length i.e. the fatty acid is a long-chain fatty acid such as oleic acid (18 carbon atoms), stearic acid (18 carbon atoms) and arachidic acid (20 carbon atoms).
  • Fatty acids may be sub-categorised based on the length and degree of saturation of the aliphatic hydrocarbon “tail”.
  • fatty acids have a molar mass of from about 100 g/mol to about 400 g/mol, such as from about 100 g/mol to about 350 g/mol, such as from about 120 g/mol to about 350 g/mol, such as from about 140 g/mol to about 350 g/mol, such as from about 140 g/mol to about 300 g/mol.
  • Exemplary fatty acids may typically have a molar mass of from about 150 g/mol to about 400 g/mol, for example from about 200 g/mol to about 350 g/mol e.g.
  • exemplary fatty acids may typically have a molar mass of from about 140 g/mol to about 200 g/mol. They include but are not limited to arachidic acid, arachidonic acid, lauric acid, linoleic acid, linolenic acid, myristic acid, myristoleic acid, oleic acid, palmitic acid, palmitoleic acid, sapienic acid, stearic acid, and vaccenic acid.
  • a further example is caprylic acid.
  • the fatty acid is oleic acid.
  • the fatty acid is caprylic acid.
  • the fatty acid is not linoleic acid.
  • the fatty acid has a hydrophilic-lipophilic balance (HLB) in the range 1 - 4, for example in the range 1 – 3, e.g.1 - 2.5, e.g.1 – 2, e.g.1 - 1.5.
  • HLB hydrophilic-lipophilic balance
  • MHL is the molecular weight of the hydrophilic portion of the molecule
  • MOH is the molecular weight of the lipophilic portion of the molecule.
  • MHL is taken as represented by O with a molecular weight of 16.
  • MOH is the balance of the molecular weight of the molecule.
  • oleic acid has an HLB value of 1 and caprylic acid has an HLB value of 2.2.
  • the fatty acid has a Log P value in the range 2 – 8, such as caprylic acid, oleic acid, lauric acid and palmitic acid.
  • caprylic acid has a Log P value of 3.05 and oleic acid has a Log P value of 7.7.
  • lauric acid has a Log P value of 4.6 and palmitic acid has a Log P value of 7.15.
  • Log P refers to the logarithm of the partition coefficient P of a compound between two immiscible phases, typically octanol and water.
  • the fatty acid has a critical micellar concentration (CMC) in the range 0.001 – 0.01 mM.
  • CMC critical micellar concentration
  • oleic acid has an CMC value of 0.006 mM.
  • CMC refers to the concentration of surfactants in a bulk phase above which micelles start to form spontaneously. Below the CMC, surfactants exist mainly as individual molecules dispersed in the solution.
  • Non-Ionic Surfactant Exemplary non-ionic surfactants may typically have a molar mass of from about 100 g/mol to about 10000 g/mol, in particular from about 100 g/mol to about 2000 g/mol. Exemplary non- ionic surfactants typically comprise one or more polyoxyalkylene moieties e.g. polyoxyethylene and/or polyoxypropylene moieties. Exemplary non-ionic surfactants include polyoxyalkylenes, particularly poloxamers, such as poloxamer 188, poloxamer 407, poloxamer 171, and poloxamer 185.
  • non-ionic surfactants include alkyl ethers of polyethylene glycol, such as those known under the brand names Brij 35 (polyoxyethylene (23) lauryl ether), Brij 52 (polyoxyethylene (20) cetyl ether), Brij 93 (polyoxyethylene (2) oleyl ether), Brij 97 (polyoxyethylene (10) oleyl ether), Brij L4 (polyoxyethylene (4) lauryl ether), Brij 30 (polyoxyethylene (4) lauryl ether), and Brij 78 (polyoxyethylene (20) stearyl ether).
  • Additional exemplary non-ionic surfactants include alkylphenyl ethers of polyethylene glycol, such as that known under the brand name Triton X-100.
  • non-ionic surfactants include fatty acid esters, such as fatty acid esters of polyols. Such fatty acid esters may comprise one or more e.g. one, two or three fatty acid chains e.g. one fatty acid chain. Specific examples include polyoxyethylene sorbitan fatty acid esters. In particular, the non-ionic surfactant is a polyoxyethylene sorbitan fatty acid ester. Suitable polyoxyethylene sorbitan fatty acid esters include polysorbate 80 (e.g. Tween 80), polysorbate 120, polysorbate 85, polysorbate 65, polysorbate 60, polysorbate 40, and polysorbate 20, in particular polysorbate 80.
  • polysorbate 80 e.g. Tween 80
  • the non-ionic surfactant is not polysorbate 60. In an embodiment, the non-ionic surfactant is not polysorbate 85.
  • the non-ionic surfactant has a hydrophilic-lipophilic balance (HLB) value of 10 or more, for example in the range 10 – 20, such as polysorbates and other highly ethoxylated non-ionic surfactants.
  • HLB hydrophilic-lipophilic balance
  • polysorbate 20 has a HLB value of 16.7.
  • polysorbate 40 has a HLB value of 15.6.
  • polysorbate 65 has a HLB value of 10.5.
  • polysorbate 120 has a HLB value of 14.9.
  • polysorbate 80 16 has a HLB value of 15.0.
  • Brij 35 has a HLB value of 16.9.
  • Brij 97 has a HLB value of 12.4.
  • the non-ionic surfactant has a Log P value in the range 1 – 5, such as polysorbate 20.
  • polysorbate 80 has a Log P value of 4.7.
  • the Log P value of the non- ionic surfactant is lower than that of the fatty acid.in the surfactant component.
  • the non-ionic surfactant has a critical micellar concentration (CMC) in the range 0.01 – 0.5 mM, such as polysorbate 20 and Brij 35.
  • CMC critical micellar concentration
  • polysorbate 20 has a CMC value of 0.06 mM.
  • polysorbate 80 has a CMC value of 0.012 mM.
  • Brij 35 has a CMC value of 0.09 mM.
  • the CMC of the non-ionic surfactant is greater than that of the fatty acid.in the surfactant component.
  • the aqueous liquid pharmaceutical formulation comprises a single non-ionic surfactant as part of the surfactant component. Alternatively, it comprises a mixture of e.g. of two (or more) non-ionic surfactants as part of the surfactant component.
  • the surfactant component is selected from the group consisting of mixtures of (a) oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (b) lauric acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (c) linoleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (d) linolenic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (e) palmitic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (f) stearic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (g) oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyalkylene, such as a poloxamer, (h)
  • the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular a polyoxyethylene sorbitan fatty acid ester selected from polysorbate 80 and polysorbate 20.
  • the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof, especially oleic acid, and polysorbate 80.
  • the surfactant component is suitably a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular a polyoxyethylene sorbitan fatty acid ester selected from polysorbate 80 and polysorbate 20.
  • the surfactant component is suitably a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol, such as those known under the brand names Brij 35, Brij 52, Brij 93, Brij 97, Brij L4, Brij 30, and Brij 78.
  • the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof, especially caprylic acid, and Brij 35.
  • the surfactant component is not a mixture of linoleic acid and polysorbate 60.
  • Pharmaceutically acceptable salt forms of fatty acid that may be employed include sodium, potassium, and ammonium salts, and in particular the sodium salt.
  • the fatty acid is used as, i.e.
  • Such a formulation may more suitably be achieved when the non-ionic surfactant is water-miscible, and/or has a HLB value of 10 or more, for example has a HLB in the range 10-20, and/or when present at a concentration provided for 18 in the present invention below.
  • Physical measurements are suitably made at a temperature of 23 o C and a pressure of 1 standard atmosphere.
  • the surfactant component is present in the formulation at a concentration of 1 – 3000 ⁇ g/mL, for example 1 – 2000 ⁇ g/mL, for example 5 – 2000 ⁇ g/mL, for example 5 – 1500 ⁇ g/mL, for example 5 – 1000 ⁇ g/mL, for example 5-500 ⁇ g/mL.
  • the surfactant component is present at a concentration of 50-200 ⁇ g/mL, for example 75-150 ⁇ g/mL, for example 90 – 120 ⁇ g/mL, or about 100 ⁇ g/mL.
  • the fatty acid may be present in the formulation at a concentration of 100 – 1000 ug/mL, for example 200 – 800 ⁇ g/mL, for example 250 – 750 ⁇ g/mL, for example 400 – 600 ⁇ g/mL, or about 500 ⁇ g/mL
  • the non-ionic surfactant may be present in the formulation at a concentration of 100 – 1000 ug/mL, for example 200 – 800 ⁇ g/mL, for example 250 – 750 ⁇ g/mL, for example 300 – 600 ⁇ g/mL, for example 400 – 500 ⁇ g/mL.
  • the surfactant component is present at a concentration of 0.005% (w/w) – 0.02% (w/w), for example 0.0075% (w/w) – 0.015% (w/w), for example 0.009% (w/w) – 0.012% (w/w), or about 0.01% (w/w), wherein the % by weight is with respect to the total weight of the formulation.
  • the surfactant component is present at a concentration of 0.05% (w/w) – 0.2% (w/w), for example 0.075% (w/w) – 0.15% (w/w), for example 0.09% (w/w) – 0.12% (w/w), or about 0.1% (w/w), wherein the % by weight Is with respect to the total weight of the formulation.
  • the fatty acid may be present in the formulation at a concentration of 0.01% (w/w) – 0.1% (w/w), for example 0.02% (w/w) – 0.08% (w/w), for example 0.025% (w/w) – 0.075% (w/w), for example 0.04% (w/w) – 0.06% (w/w), or about 0.05% (w/w), and the non-ionic surfactant may be present in the formulation at a concentration of 0.01% (w/w) – 0.1% (w/w), for example 0.02% (w/w) – 0.08% (w/w), for example 0.025% (w/w) – 0.075% (w/w), for example 0.03% (w/w) – 0.06% (w/w), for example 0.04% (w/w) – 0.05% (w/w), wherein the % by weight is with respect to the total weight of the formulation.
  • the surfactant component forms the oil phase of an oil in water emulsion when dispersed in the aqueous formulation.
  • the surfactant component forms the oil phase of said 20 emulsion in which the fatty acid (such as oleic acid) forms the core (or internal phase) of the particles and the non-ionic surfactant (such as polysorbate 80) stabilises the interface between the oil phase and the aqueous phase.
  • the polynucleotide molecule is present within (i.e.
  • aqueous liquid pharmaceutical formulations of the present invention comprise a polynucleotide molecule.
  • the aqueous liquid pharmaceutical formulation comprises a single polynucleotide molecule.
  • the aqueous liquid formulations of the present invention may comprise more than one, for example two, three, four, five, six, seven, eight, nine, or ten etc. different polynucleotide molecules.
  • polynucleotide molecule refers to a molecule comprising two or more nucleotides. Therefore, a polynucleotide molecule may suitably comprise from two to about 100 nucleotides, such as from two to about 90 nucleotides, such as from two to about 80 nucleotides, such as from two to about 70 nucleotides, such as from two to about 60 nucleotides, or such as from two to about 50 nucleotides.
  • the polynucleotide molecule suitably comprises from about five to about 50 nucleotides, such as from about five to about 40 nucleotides, such as from five to about 30 nucleotides or from about 20 to about 40 nucleotides, such as from 10 to about 30 nucleotides or from about 20 21 to about 35 nucleotides, such as from 10 to about 20 nucleotides or from about 20 to about 30 nucleotides.
  • a polynucleotide molecule may suitably comprise more than about 100 nucleotides.
  • the polynucleotide molecule suitably comprises more than about 100 nucleotides, such as more than about 200 nucleotides, such as more than about 400 nucleotides, such as more than about 500 nucleotides, such as more than about 750 nucleotides, such as more than about 1000 nucleotides, such as more than about 1250 nucleotides, such as more than about 1500 nucleotides, such as more than about 2000 nucleotides, such as more than about 2500 nucleotides, such as more than about 5000 nucleotides.
  • the polynucleotide molecule suitably comprises from about 100 to about 20000, such as from about 100 to about 10000 nucleotides, such as from about 200 to about 8000 nucleotides or from about 500 to about 10000 nucleotides, such as from about 2500 to about 1000 nucleotides or from about 500 to about 7500 nucleotides, such as from about 1000 to about 5000 nucleotides, such as from about 2000 to about 5000 nucleotides, such as from about 2000 nucleotides to about 4000 nucleotides.
  • nucleotide refers to monomeric organic molecules comprising a nitrogenous base, in particular a primary or canonical nitrogenous base such as adenine, cytosine, guanine, thymine, or uracil, a sugar molecule, in particular a pentose sugar, such as ribose or deoxyribose, and a phosphate group or an analogue thereof, such as a thiophosphate group.
  • a nitrogenous base in particular a primary or canonical nitrogenous base such as adenine, cytosine, guanine, thymine, or uracil
  • a sugar molecule in particular a pentose sugar, such as ribose or deoxyribose, and a phosphate group or an analogue thereof, such as a thiophosphate group.
  • Suitable alternative nitrogenous bases include modified purine nitrogenous bases such as 7-methyl guanine, hypoxanthine, and xanthine, modified pyrimidine nitrogenous bases such as 5,6-dihydrouracil, 5-methylcytosine and 5’hydroxymethylcytosine, and artificial or synthetic nitrogenous bases.
  • Suitable nucleotides therefore include ribonucleotides and deoxyribonucleotides.
  • polynucleotide molecules therefore include ribonucleic acid (RNA) molecules, and deoxyribonucleic acids (DNA) molecules.
  • nucleotide may further refer to peptide nucleotides, threose nucleotides, glycol nucleotides, serinol nucleotides or locked nucleotides.
  • polynucleotides molecules therefore include peptide nucleic acid (PNA) molecules, threose nucleic acid (TNA) molecules, glycol nucleic acid (GNA) molecules, serinol nucleic acid (SNA) molecules and locked nucleic acid (LNA) molecules.
  • polynucleotides molecules further include hybrid polynucleotide molecules comprising one or more different types of nucleotide, in particular from those nucleotides described above.
  • a particularly suitable hybrid polynucleotide molecule is a DNA:RNA hybrid polynucleotide molecule i.e., a polynucleotide molecule comprising ribonucleotides and deoxyribonucleotides.
  • the polynucleotide molecule is an RNA molecule or a DNA molecule. 22
  • the polynucleotide molecule may be of any origin, e.g.
  • the polynucleotide molecule may arise from any biological sample and any organ, tissue, cell, or sub-cellular compartment.
  • the polynucleotide molecule may be pre-treated before use, for example isolated, purified and/or modified.
  • the polynucleotide molecule, or any number of individual nucleotides within the polynucleotide molecule may be artificial or synthetic.
  • the polynucleotide molecule may consist of a single strand, i.e. be single-stranded, e.g.
  • the polynucleotide molecule may consist of two strands, i.e. be double- stranded, e.g. of dsRNA.
  • a polynucleotide molecule may consist of three strands i.e. be triple-stranded, e.g. triple-stranded DNA.
  • the single-stranded polynucleotide molecule consists of a sense strand.
  • the single-stranded polynucleotide molecule consists of an antisense strand.
  • the polynucleotide molecule comprises a double-stranded region.
  • a single-stranded polynucleotide molecule may comprise a first region which hybridizes with a second region of the single-stranded polynucleotide molecule to form an intramolecular double-stranded region, e.g., duplex.
  • the polynucleotide molecule comprises an intramolecular structure.
  • the polynucleotide molecule may comprise a helix, a bulge (separation of a double helical tract on one strand), an internal loop (separation of a double helical tract on both strands), a stem-loop or hairpin, a tetraloop (four base pair hairpin), pseudoknot, or junction.
  • the polynucleotide molecule comprises at least one, e.g., one or two, 5’ or 3’ monophosphate(s), and/or at least one, e.g. one or two, 5’ or 3’ diphosphate(s), and/or at least one, e.g. one or two, 5’ or 3’ triphosphate(s), and/or at least one, e.g. one or two, 5’ or 3’ hydroxyl (OH) groups.
  • the 5’ or 3’ monophosphate(s) and/or 5’ or 3’ diphosphate(s) and/or 5’ or 3’ triphosphate(s) and/or 5’ or 3’ hydroxyl (OH) groups are located at the 5’ and/or 3’ terminus or termini of the single-stranded or double-stranded polynucleotide molecule.
  • the polynucleotide molecule is polyinosinic-polycytidylic acid (poly I:C), which is a synthetic dsRNA typically between 100-10000, such as between 200- 8000, for example between 300-6000 base pairs in length (see, for example, dsRNA 2 in the Examples below and US9682096B2, which is incorporated herein by reference).
  • poly I:C polyinosinic-polycytidylic acid
  • the polynucleotide molecule is an RNA molecule.
  • the polynucleotide molecule, which is an RNA molecule is an mRNA molecule, miRNA molecule, shRNA molecule, or siRNA molecule.
  • the polynucleotide molecule is an mRNA molecule.
  • the polynucleotide molecule is a single-stranded RNA molecule which may encode one or more proteins.
  • the mRNA molecule is capable of being translated.
  • the mRNA molecule comprises from about 100 to about 10000 nucleotides, such as from about 200 to about 8000 nucleotides, such as from about 500 to about 7500 nucleotides, such as from about 1000 to about 5000 nucleotides.
  • the mRNA molecule comprises one or more coding regions, which may optionally be stabilised by internal base pairs.
  • the coding regions may further comprise regulatory sequences, exonic splicing enhancers or exonic splicing silencers.
  • the mRNA molecule comprises a 5’ untranslated region (5’ UTR) and/or a 3’ untranslated region (3’ UTR).
  • the mRNA molecule comprises a 3’ tail of adenine nucleotides, known as a polyA tail.
  • the polyA tail comprises 50 or more, such as 100 or more adenine nucleotides.
  • the polynucleotide molecule for example mRNA molecule, comprises a 5’ cap, comprising a terminal 7-methylguanosine residue linked via 5’-5’ triphosphate bond to the first 5’ nucleotide.
  • the mRNA molecule is a circularised mRNA molecule, for example due to a protein-mediated intramolecular interaction between the 5’ cap and polyA tail of an mRNA molecule.
  • the polynucleotide molecule is a miRNA molecule.
  • the polynucleotide molecule is a single-stranded RNA molecule which does not encode a protein and which functions in RNA silencing and post-transcriptional regulation of gene expression.
  • the miRNA molecule comprises from about 20 to about 25 nucleotides. In particular the miRNA molecule comprises 20, 21, 22, 23, 24 or 25 nucleotides.
  • the polynucleotide molecule is an shRNA molecule.
  • the polynucleotide molecule is a single-stranded RNA molecule wherein a first region hybridizes with a second region of the single-stranded RNA molecule to form an intramolecular double- stranded region, e.g., duplex. Said hybridization will create a hairpin structure, including a hairpin loop, within the RNA molecule.
  • the shRNA comprises from about 10 to about 70 nucleotides, for example from about 20 to about 70 nucleotides, for example from about 35 to about 70 nucleotides or from about 25 to about 35 nucleotides.
  • the shRNA molecule comprises 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 nucleotides.
  • the double-stranded region, or duplex comprises 30 base pairs or less, such as 25 base pairs or less, such as 20 base pairs or less, such as 18 base pairs or less, 24 such as 16 base pairs or less, such as 14 base pairs or less, such as 12 base pairs or less, such as 10 base pairs or less, such as eight base pairs or less, such as six base pairs or less, such as four base pairs or less.
  • the double-stranded region, or duplex comprises from about two to about 30 base pairs, such as from about two to about 25 base pairs, such as from about four to about 20 base pairs, such as from about 4 to about 18 base pairs, such as from about six to about 18 base pairs, such as from about eight to about 18 base pairs, such as from about 10 to about 18 base pairs.
  • the double-stranded region comprises one or more mispaired bases, according to Watson-Crick base pairing.
  • the double-stranded region may comprise one to 10 mispaired bases, such as one to eight mispaired bases, such as one to six mispaired bases, in particular one, two, three, four, five, or six mispaired bases.
  • the first region and second region, which hybridize to form an intramolecular double- stranded region, e.g., duplex are each 20 nucleotides or less, for example 19 nucleotides or less, such as 18 nucleotides or less, in length.
  • the first and second region are each between about five and about 20 nucleotides, for example between about five and about 18 nucleotides, such as between about eight and about 18 nucleotides, such as between about 10 and about 18 nucleotides in length.
  • the first region and second region, which hybridize to form an intramolecular double- stranded region, e.g., duplex, are substantially complementary to each other, for example are at least about 80% complementary, in particular at least about 90% complementary, or most preferably 100% complementary.
  • the hairpin structure may comprise a hairpin loop.
  • the length the hairpin loop is zero, or one or more, nucleotides in length.
  • the hairpin loop is two or more, four or more, five or more, eight or more, 10 or more, 15 or more, 20 or more, 40 or more, 50 or more, or 100 or more, nucleotides in length.
  • the hairpin loop is from about one to about 20, such as from about one to about 15, such as from about two to about 10, such as from about four to about 10 nucleotides in length.
  • the shRNA molecule comprises a blunt end.
  • a blunt end refers to a polynucleotide molecule, where at least one strand of the duplex does not overhang, for example a 3’ dinucleotide overhang, such that the 5’ and 3’ strand end together.
  • the shRNA molecule may comprise a 3’ overhang or a 5’ overhang.
  • the shRNA molecule comprises at least one 5’ triphosphate or at least one 5’ diphosphate.
  • the 5’ triphosphate or 5’ diphosphate is located at the 5’ terminus of the shRNA molecule.
  • the shRNA molecule comprises one 5’ triphosphate or one 5’ diphosphate, in particular wherein the 5’ triphosphate or 5’ diphosphate is located at the 5’ terminus.
  • the shRNA molecule which is a single-stranded RNA molecule which forms a hairpin structure comprising an intramolecular double-stranded region and a hairpin loop, comprises a blunt end and a 5’ triphosphate or 5’ diphosphate located at the 5’ terminus, wherein the double-stranded region is between about 10 and about 18 nucleotides in length.
  • the shRNA molecule of this embodiment may comprise one or more sugar-modified nucleotides, which each comprise a 2’ OH modification, and/or one or more backbone- modified nucleotides and/or one or more base-modified nucleotides.
  • such an shRNA molecule is capable of inducing an interferon response in a vertebrate cell.
  • the shRNA molecule which is a single-stranded RNA molecule which forms a hairpin structure comprising an intramolecular double-stranded region and a hairpin loop, comprises, consists of, or consists essentially of a sequence disclosed in WO2019/246450A1, which is incorporated herein by reference.
  • the shRNA molecule which is a single-stranded RNA molecule which forms a hairpin structure comprising an intramolecular double-stranded region and a hairpin loop, comprises, consists of, or consists essentially of SEQ ID NO. 2.
  • the shRNA molecule comprises, consists of, or consists essentially of a variant of SEQ ID NO.2.
  • the shRNA molecule which is a single-stranded RNA molecule which forms a hairpin structure comprising an intramolecular double-stranded region and a hairpin loop, comprises, consists of, or consists essentially of a sequence disclosed in US2023/0159923A1, which is incorporated herein by reference.
  • the polynucleotide molecule is an siRNA molecule.
  • the polynucleotide molecule is a double-stranded RNA molecule which does not encode a protein and which functions in RNA silencing and post-transcriptional regulation of gene expression.
  • the siRNA molecule comprises from about 20 to about 60 nucleotides, for example from about 30 to about 60 nucleotides, such as from about 40 to about 50 nucleotides.
  • the siRNA molecule comprises 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, or 60 nucleotides.
  • the siRNA molecule comprises a double-stranded region, or duplex, comprising 30 base pairs or less, such as 25 base pairs or less, such as 20 base pairs or less.
  • the double-stranded region, or duplex comprises from about 10 to about 30 base pairs, such as from about 15 to about 30 base pairs, such as from about 20 to about 30 base pairs, such as from about 20 to about 25 base pairs or from about 25 base pairs to about 30 base pairs.
  • the double-stranded region comprises one or more mispaired bases, according to Watson-Crick base pairing.
  • the double-stranded region may comprise one to 10 mispaired bases, such as one to eight mispaired bases, such as one to six mispaired bases, in particular one, two, three, four, five, or six mispaired bases.
  • the siRNA molecule comprises one or more, e.g., two, 3’ overhangs.
  • the siRNA molecule may comprise one or more, e.g., two 5’ overhangs.
  • the siRNA molecule may comprise one or more, e.g., one, 3’ overhang and/or one or more, e.g., one, 5’ overhang.
  • the siRNA molecule may comprise one or more blunt ends.
  • the siRNA molecule comprises at least one, e.g., two, 5’ monophosphate(s), or at least one, e.g., two, 5’ diphosphate(s), or at least one, e.g., two, 5’ triphosphate(s).
  • the 5’ monophosphate(s) and/or 5’ diphosphate(s) and/or 5’ triphosphate(s) are located at the 5’ termini of the siRNA molecule.
  • the siRNA molecule comprises at least one, e.g., two, 3’ OH (hydroxyl) groups.
  • the 3’ OH group(s) are located at the 3’ termini of the siRNA molecule.
  • the siRNA molecule which is a double-stranded RNA molecule, comprises the sequence of SEQ ID NO.11 or a variant thereof.
  • one strand of the double-stranded siRNA molecule comprises, consists of, or consists essentially of the sequence of SEQ ID NO.11, or a variant thereof (see Ren et al.2019, which is incorporated herein by reference).
  • the polynucleotide molecule is a guide RNA (gRNA) molecule, which is a polynucleotide molecule comprising crispr RNA (crRNA), which is a nucleotide sequence, typically between 15 and 20 nucleotides in length which is complementary to a host target DNA, and tracr RNA (trRNA) which enables binding to a Cas nuclease.
  • gRNA guide RNA
  • crRNA crispr RNA
  • trRNA tracr RNA
  • the polynucleotide molecule is a DNA molecule.
  • the DNA molecule is a genomic DNA (gDNA) molecule, for example a chromosomal DNA molecule or a mitochondrial DNA molecule, a complementary DNA (cDNA) molecule, or an extra- chromosomal DNA molecule, for example a plasmid DNA molecule.
  • the DNA molecule is a gDNA molecule.
  • the DNA molecule is a cDNA molecule.
  • the DNA molecule is an extra-chromosomal DNA molecule, in particular a plasmid DNA molecule.
  • the DNA molecule is a coding DNA molecule.
  • the DNA molecule comprises one or more coding regions, which may optionally be stabilised by internal base pairs.
  • the DNA molecule may encode one or more protein.
  • the DNA molecule, which is a coding DNA molecule is capable of being transcribed.
  • the DNA molecule, which is a coding DNA molecule further comprises one or more promoter sequences, in particular wherein the one or more promoter sequence flanks the corresponding coding region at the 5’ end.
  • the DNA molecule, which is a coding DNA molecule further comprises a termination sequence, in particular wherein the termination sequence flanks the one or more coding regions at the 3’ end.
  • a coding DNA molecule may further comprise any number of non-coding DNA elements, as described below.
  • the DNA molecule is a non-coding DNA molecule, that is the DNA molecule does not comprise a coding region and consequently does not encode a protein.
  • the non-coding DNA molecule is capable of being transcribed, for example to produce tRNA, miRNA, siRNA or ribosomal RNA.
  • the non-coding DNA molecule may comprise a regulatory sequence that controls gene expression, may comprise a scaffold attachment region, may comprise a centromere, or may comprise a telomere.
  • the non-coding DNA molecule may comprise non-functional elements such as an intron, a pseudogene, intergenic DNA, or transposons.
  • the non-coding DNA molecule may comprise a combination of the above-described elements.
  • the DNA molecule comprises from about 100 to about 20000 nucleotides, such as from about 100 to about 15000 nucleotides, such as from about 500 to about 15000 nucleotides, such as from about 500 to about 10000 nucleotides, such as from about 2500 to about 10000 nucleotides, such as from about 2500 to about 8000 nucleotides.
  • the polynucleotide molecule has the structure of Formula (I): 5’-P z -(N) b N-3’-(E) y (E)-L-(E)(E) y’ -5’-N(N) b’ -3’ 28 wherein 5’-P z -(N) b N-3’ represents the first nucleic acid sequence; 5’-N(N) b’ -3’ represents the second nucleic acid sequence; P at each instance is independently a phosphate or analog thereof; z is 2 or 3; N is, at each instance, any nucleotide or modified nucleotide or analog or derivative there of; b and b’ are independently 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18; 5’-(E) y (E)-L-(E)(E) y’ -3’ represents the connector element wherein E at each occurrence is independently any nucleotide, modified nucleotide, or abasic;
  • the polynucleotide molecule has the structure of Formula (II): 5’-P z -Nu-3’-(E) y (E)-L-(E)(E) y’ -5’-Nu’-3’ wherein 5’-P z -Nu-3’ represents the first nucleic acid sequence; 5’-Nu’-3’ represents the second nucleic acid sequence; P at each instance is independently a phosphate or analog thereof.
  • E at each occurrence is independently any nucleotide, modified nucleotide, or abasic; y and y’ are independently 0-9, provided that y + y’ equals 0-8; L is a non-nucleotide segment having the structure wherein X and X’ are independently O or S; Y and Y’ are independently OR”, SR”, or NRR’; V and V’ are independently O, S, or NRR’; q is 1-20; k is 1-20; t is 1-20; M selected from aliphatic, substituted aliphatic, aryl, substituted aryl, heteroalkyl, heterocyclyl or substituted heterocyclyl; W is any reactive group or conjugation group; and d is 0 or 1.
  • z is 2. Moreover, suitably, when z is 2, at least one P is a phosphate analogue. In particular, suitably, when z is 2, both P are phosphate analogues. Alternatively, 30 in one embodiment, z is 3. Furthermore, suitably, when z is 3, at least one P is a phosphate analogue. In particular, suitably, when z is 3, at least two P are phosphate analogues. For example, suitably, when z is 3, all P are phosphate analogues.
  • b and b’ are independently 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18; preferably 11, 12, 13, 14, 15, 16, 17, or 18; preferably 13, 14, 15, 16, 17, or 18.
  • b is 9 and b’ is less than, equal to, or greater than 9.
  • b is 10 and b’ is less than, equal to, or greater than 10. In one embodiment, b is 11 and b’ is less than, equal to, or greater than 11. In one embodiment, b is 12 and b’ is less than, equal to, or greater than 12. In one embodiment, b is 13 and b’ is less than, equal to, or greater than 13. In one embodiment, b is 14 and b’ is less than, equal to, or greater than 14. In one embodiment, b is 15 and b’ is less than, equal to, or greater than 15. In one embodiment, b is 16 and b’ is less than, equal to, or greater than 16. In one embodiment, b is 17 and b’ is less than, equal to, or greater than 17.
  • b is 18 and b’ is less than, equal to, or greater than 18. In one embodiment, b equals b’. In an alternative embodiment, b does not equal b’. In one embodiment, when b is less than b’, the polynucleotide molecule has a 3’-overhang. In an alternative embodiment, when b is greater than b’, the nucleic acid molecule has a 5′-overhang. In one embodiment, q is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20. For example, q is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15, and in particular q is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • q is 1, 2, 3, 4, or 5, and in particular q is 1, or q is 2, or q is 3 ,or q is 4, or q is 5.
  • k and t are independently 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20, and in particular k and t are independently 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15.
  • k and t are independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, and in particular k and t are independently 1, 2, 3, 4, or 5.
  • k is 1, or k is 2, or k is 3, or k is 4, or k is 5.
  • t is 1, or t is 2, or t is 3, or t is 4, or t is 5.
  • k and t are the same, and L is therefore 31 symmetrical. In an alternative embodiment, k and t are different, and L is therefore asymmetrical.
  • R, R’, and R are independently selected from the group consisting of an alkyl, an amino alkyl, a carboxamido, polyethylene glycol (PEG), aralkyl, hetero-ar-alkyl, hetero-alkyl, substituted or unsubstituted cycloalkyl.
  • R and R” groups may contain functionalities such as amino, hydroxy, azido, or thiol, that can be optionally used for the attachment of which can be used to link to a targeting molecule (Tm), such as a vitamin, peptide, antibody, or protein.
  • Tm targeting molecule
  • the R and R” group can be a peptide group.
  • Peptide groups include a variety of enzymatically cleavable or non-cleavable peptides.
  • the individual amino acids groups of the peptide could be natural or synthetic amino acids.
  • R 1 methyl, isopropyl, t-butyl, - (CH2)n-R 2 , wherein R 2 is selected from aryl, aralkyl, heteroaryl, hetero-aralkyl, alkyl, an amino alkyl, a carboxamido, polyethylene glycol (PEG), hetero-alkyl, substituted or unsubstituted cycloalkyl.
  • the reactive group W may be additionally connected to alkyl, an amino alkyl, a carboxamido, polyethylene glycol (PEG), aralkyl, hetero-ar-alkyl, hetero-alkyl, substituted or unsubstituted cycloalkyl.
  • R and R” groups may contain functionalities such as amino, hydroxy, azido, or thiol, that can be optionally used for the attachment to a targeting molecule (Tm) such as a vitamin, peptide, antibody, or protein.
  • Tm targeting molecule
  • M is selected from aliphatic, substituted aliphatic, aryl, substituted aryl, heteroralkyl, heterocyclyl or substituted heterocyclyl.
  • aliphatic group refers to a non-aromatic moiety that may be saturated (e.g., single bond) or contain one or more units of unsaturation, e.g., double and/or triple bonds.
  • An aliphatic group may be straight chained, branched or cyclic, contain carbon, hydrogen or, optionally, one or more heteroatoms and may be substituted or unsubstituted.
  • aliphatic groups include, for example, polyalkoxyalkyls, such as polyalkylene glycols, polyamines, and polyimines, for example. Such aliphatic groups may be further substituted.
  • aliphatic groups may include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, and substituted or unsubstituted cycloalkyl groups as described herein.
  • alkyl is intended to include both branched and straight chain, substituted or unsubstituted saturated aliphatic hydrocarbon radicals/groups having the specified number of carbons.
  • Preferred alkyl groups comprise about 1 to about 24 carbon atoms (“C 1 -C 24 ”).
  • Such radicals preferably contain from about two to about twenty-four carbon atoms (“C 2 -C 24 ”).
  • Other preferred alkenyl radicals are “lower alkenyl” radicals having two to about ten carbon atoms (“C 2 -C 10 ”) such as ethenyl, allyl, propenyl, butenyl and 4- methylbutenyl.
  • Preferred lower alkenyl radicals include 2 to about 6 carbon atoms (“C 2 -C 6 ”).
  • alkenyl”, and “lower alkenyl”, embrace radicals having “cis” and “trans” orientations, or alternatively, “E” and “Z” orientations.
  • alkynyl refers to linear or branched radicals having at least one carbon-carbon triple bond. Such radicals preferably contain from about two to about twenty-four carbon atoms (“C 2 -C 24 ”). Other preferred alkynyl radicals are “lower alkynyl” radicals having two to about ten carbon atoms such as propargyl, 1-propynyl, 2-propynyl, 1-butyne, 2-butynyl and 1-pentynyl. Preferred lower alkynyl radicals include 2 to about 6 carbon atoms (“C 2 -C 6 ”).
  • cycloalkyl refers to saturated carbocyclic radicals having three to about twelve carbon atoms (“C 3 -C 12 ”).
  • cycloalkyl embraces saturated carbocyclic radicals having three to about twelve carbon atoms. Examples of such radicals include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • alkoxy refers to linear or branched oxy-containing radicals each having alkyl portions of one to about twenty-four carbon atoms or, preferably, one to about twelve carbon atoms.
  • More preferred alkoxy radicals are “lower alkoxy” radicals having one to about ten carbon atoms and more preferably having one to about eight carbon atoms. Examples of such radicals include methoxy, ethoxy, propoxy, butoxy and tert-butoxy.
  • aryl alone or in combination, means an aromatic system containing one, two or three rings wherein such rings may be attached together in a pendent manner or may be fused.
  • aryl embraces aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indane furanyl, quinazolinyl, pyridyl and biphenyl.
  • heterocyclyl refers to saturated, partially unsaturated and unsaturated heteroatom-containing ring-shaped radicals, which can also be called “heterocyclyl”, “heterocycloalkenyl” and “heteroaryl” correspondingly, where the heteroatoms may be selected from nitrogen, sulfur and oxygen.
  • saturated heterocyclyl radicals include saturated 3 to 6-membered heteromonocyclic group containing 1 to 4 nitrogen atoms (e.g., pyrrolidinyl, imidazolidinyl, piperidino, piperazinyl, etc.); saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms (e.g., morpholinyl, etc.); saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms (e.g., thiazolidinyl, etc.).
  • heteroaryl radicals include unsaturated 3 to 6 membered heteromonocyclic group containing 1 to 4 nitrogen atoms, for example, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl (e.g., 4H-1,2,4-triazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3- triazolyl, etc.) tetrazolyl (e.g., 1H-tetrazolyl, 2H-tetrazolyl, etc.), etc.; unsaturated condensed heterocyclyl group containing 1 to 5 nitrogen atoms, for example, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl
  • alkyl or “arylalkyl” refer to aryl-substituted alkyl radicals such as benzyl, diphenylmethyl, triphenylmethyl, phenylethyl, and diphenylethyl.
  • aryloxy refers to aryl radicals attached through an oxygen atom to other radicals.
  • alkylamino denotes amino groups which are substituted with one or two alkyl radicals.
  • Preferred alkylamino radicals have alkyl radicals having about one to about twenty carbon atoms or, preferably, one to about twelve carbon atoms.
  • alkylamino radicals are “lower alkylamino” that have alkyl radicals having one to about ten carbon atoms. Most preferred are alkylamino radicals having lower alkyl radicals having one to about eight carbon atoms. Suitable lower alkylamino may be monosubstituted N-alkylamino or disubstituted N,N-alkylamino, such as N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-diethylamino or the like.
  • halogen or “halo” as used herein, refers to an atom selected from fluorine, chlorine, bromine and iodine.
  • substituted refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent including, but not limited to: halo, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, thiol, alkylthio, arylthio, alkylthioalkyl, arylthioalkyl, alkylsulfonyl, alkylsulfonylalkyl, arylsulfonylalkyl, alkoxy, aryloxy, aralkoxy, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkoxycarbonyl, aryloxycarbonyl, haloalkyl, amino, trifluoromethyl, cyano, nitro, alkylamino, arylamino, alkylaminoalkyl, arylaminoalkyl, aminoalkylamino,
  • the polynucleotide molecule may comprise, consist of, or consist essentially of SEQ ID NO.13 and SEQ ID NO. 14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L, as defined above.
  • the polynucleotide molecule comprises, consists of, or consists essentially of a variant of SEQ ID NO.13 and/or a variant of SEQ ID NO.14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L, wherein a “variant” as used herein refers to a sequence having for example at least about 75% identity, for example at least about 80% identity, for example at least about 85% identity, in particular at least about 90% identity, such as at least about 95%, 98% or 99% identity to the associated reference sequence over their entire lengths.
  • L is selected from the group consisting of: 35 . , .
  • the polynucleotide molecule may comprise, consist of, or consist essentially a variant of SEQ ID NO. 13 and/or a variant of SEQ ID NO. 14 wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L1.
  • the polynucleotide molecule may comprise, consist of, or consist essentially of SEQ ID NO.13 and SEQ ID NO. 14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L2.
  • the 36 example 10 – 1000 ⁇ g/mL for example 10 – 500 ⁇ g/mL, for example 10 – 250 ⁇ g/mL, for example 50 – 150 ⁇ g/mL, such as about 100 ⁇ g/mL.
  • the polynucleotide molecule may be present in the formulation at a concentration of 1 – 10000 ⁇ g/mL, for example 10 – 10000 ⁇ g/mL, for example 100 – 10000 ⁇ g/mL, for example 500 – 5000 ⁇ g/mL, for example 500 – 2500 ⁇ g/mL, such as about 1000 ⁇ g/mL.
  • the polynucleotide molecule may be present in the formulation at a concentration of 1 – 100000 ⁇ g/mL, for example 10 – 100000 ⁇ g/mL, for example 100 – 100000 ⁇ g/mL, for example 1000 – 100000 ⁇ g/mL, for example 5000 – 75000 ⁇ g/mL, for example 5000 – 50000 ⁇ g/mL, such as 5000 – 15000 ⁇ g/mL, 10000 – 20000 ⁇ g/mL, 15000 – 25000 ⁇ g/mL, or 25000 – 50000 ⁇ g/mL.
  • the concentration of polynucleotide molecule in the formulation is highly variable and dependent on the type, sequence, structure, and size of the polynucleotide molecule.
  • the polynucleotide molecule may be present in the formulation at a concentration of 0.000001% (w/w) – 10% (w/w), for example 0.00001% (w/w) – 10% (w/w), for example 0.0001% (w/w) – 10% (w/w), for example 0.0001% (w/w) – 5% (w/w), wherein the % by weight is with respect to the total weight of the formulation.
  • the polynucleotide molecule may be present in the formulation at a concentration of 0.0001% (w/w) – 1% (w/w), for example 0.0001% (w/w) – 0.1% (w/w), for example 0.0001% (w/w) – 0.01% (w/w), for example 0.0001% (w/w) – 0.005% (w/w), for example 0.0001% (w/w) – 0.002% (w/w), such as about 0.001% (w/w), wherein the % by weight is with respect to the total weight of the formulation.
  • the polynucleotide molecule may be present in the formulation at a concentration of 0.0001% (w/w) – 1% (w/w), for example 0.0001% (w/w) – 0.1% (w/w), for example 0.001% (w/w) – 0.1% (w/w), for example 0.001% (w/w) – 0.05% (w/w), for example 0.001% (w/w) – 0.025% (w/w), for example 0.005% (w/w) – 0.015% (w/w), such as about 0.01% (w/w), wherein the % by weight is with respect to the total weight of the formulation.
  • the polynucleotide molecule may be present in the formulation at a concentration of 0.0001% (w/w) – 1% (w/w), for example 0.001% (w/w) – 1% 37 (w/w), for example 0.01% (w/w) – 1% (w/w), for example 0.05% (w/w) – 0.5% (w/w), for example 0.05% (w/w) – 0.25% (w/w), such as about 0.1% (w/w), wherein the % by weight is with respect to the total weight of the formulation.
  • the polynucleotide molecule may be present in the formulation at a concentration of 0.0001% (w/w) – 10% (w/w), for example 0.001% (w/w) – 10% (w/w), for example 0.01% (w/w) – 10% (w/w), for example 0.1% (w/w) – 10% (w/w), for example 0.5% (w/w) – 7.5% (w/w), for example 0.5% (w/w) – 5% (w/w), such as 0.5% (w/w) – 1.5% (w/w), 1% (w/w) – 2% (w/w), 1.5% (w/w) – 2.5% (w/w) or 2.5% (w/w) – 5% (w/w), wherein the % by weight is with respect to the total weight of the formulation.
  • the ratio of the amount of the surfactant component (meaning the total concentration of the surfactants of the surfactant component) to the amount of polynucleotide molecule, for example wherein each is measured in ⁇ g/mL, is between about 100:1 and about 1:1000, for example between about 100:1 and about 1:750, for example between about 75:1 and about 1:750, for example between about 50:1 and about 1:750, for example between about 50:1 and about 1:500, for example between about 50:1 and about 1:250, for example between about 50:1 and about 1:200, for example between about 40:1 and about 1:200, for example between about 40:1 and about 1:150, for example between about 25:1 and about 1:150, for example between about 25:1 and about 1:100.
  • the polynucleotide molecules described above comprise one or more modified nucleotides.
  • the polynucleotide molecule comprises two or more, three or more, four or more, five or more, eight or more, or 10 or more modified nucleotides. Modifications to nucleotides suitably can enhance stability, functionality, and/or specificity, and can minimise the immunostimulatory properties of a polynucleotide molecule.
  • the polynucleotide molecule comprises at least one modified nucleotide which confers reduced immunostimulatory activity, and at least one modified nucleotide which confers increased serum half-life.
  • the same modified nucleotide elicits both effects.
  • the polynucleotide molecule comprises one or more modified nucleotides which confer enhanced resistance to nuclear enzymes.
  • Modified nucleotides include, but are not limited to, sugar-, backbone-, and base-modified nucleotides. Suitable modified nucleotides may comprise any combination of sugar-, backbone-, and base-modifications.
  • Sugar-modified nucleotides include, but are not limited to, nucleotides in which the 2’ OH- group (ribonucleotide) or 2’-H group (deoxyribonucleotide) is replaced by a group selected from the group consisting of H, OR, R, halo, SH, SR, NH 2 , NHR, NR 2 , or ON, wherein R is C 1 - C 6 alkyl, alkenyl, or alkynyl.
  • 2’ OH-group (ribonucleotide) or 2’-H group (deoxyribonucleotide) modifications include alkoxy or aryloxy modifications, e.g.
  • R is H, alkyl e.g. C 1 - 38 C 6 alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar; polyethyleneglycol (PEG) modifications; “locked” nucleotide modifications wherein the 2’ OH-group or 2’ H-group is connected, for example via a methylene bridge, to the 4’ carbon of the same sugar; amine, O-amine and aminoalkoxy, e.g.
  • amine O(CH 2 ) n amine, modifications, wherein amine is NH 2 , alkylamino, dialkylamino, heterocyclyl amino, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, ethylene diamine, or polyamino).
  • Additional examples of 2’ OH-group (ribonucleotide) or 2’-H group (deoxyribonucleotide) modifications include H (for ribonucleotides); halo; amino, for example NH 2 , alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroaryl amino, diheteroaryl amino, or amino acid); NH(CH 2 CH 2 NH) n CH 2 CH 2 -amine modifications wherein amine is NH 2 , alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroaryl amino, diheteroaryl amino; NHC(O)R modifications, wherein R is alkyl e.g.
  • the polynucleotide molecule in particular an RNA molecule, comprises one or more sugar-modified nucleotides which each comprise a 2’ OH (or 2’ H) modification.
  • the polynucleotide molecule in particular an RNA molecule, comprises one or more sugar-modified nucleotides which each comprise a 2’ OH (or 2’ H) modification selected from the group consisting of 2’-deoxy, 2’-fluoro, 2'-deoxy-2'-fluoro, 2’-O-methyl, 2’-O- methoxyethyl (2’-O-MOE), 2’-O-aminopropyl (2’-O-AP), 2’-O-dimethylaminoethyl (2’-O- DMAOE), 2’-O-dimethylaminopropyl (2’-O-DMAP), 2’-O-dimethylaminoethyloxyethyl (2’-O- DMAEOE), or 2’-O—N-methylacetamido (2’-O-NMA).
  • 2’ OH (or 2’ H) modification selected from the group consisting of 2’-deoxy, 2’-fluoro
  • the polynucleotide molecule in particular an RNA molecule, comprises one or more sugar-modified nucleotides which each comprise a 2’ OH modification selected from the group consisting of 2’-H, and 2’-O-methyl.
  • the polynucleotide molecule in particular an RNA molecule, comprises one or more nucleotides comprising a 2’-O-methyl modification.
  • all of the nucleotides in the polynucleotide molecule, in particular an RNA molecule comprise a 2’-O-methyl modification.
  • some or all of the pyrimidine nucleotides of the polynucleotide molecule, in particular an RNA molecule comprise a 2’ OH modification. In particular, some or all of the pyrimidine nucleotides may comprise a modification selected from the group consisting of 2’- H, and 2’-O-methyl. In one embodiment, some or all of the purine nucleotides of the polynucleotide molecule, in particular an RNA molecule, comprise a 2’ OH modification. In particular, some or all of the purine nucleotides may comprise a modification selected from the group consisting of 2’-H, and 2’-O-methyl.
  • the polynucleotide molecule in particular an RNA molecule, comprises one or more dinucleotides which are susceptible to endonuclease cleavage, wherein the 5’ 39 nucleotide of the dinucleotide comprises a 2’ OH (or 2’ H) modification.
  • the dinucleotide is s 5’-UA-3’, 5’-UG-3’, 5’-CA-3’, 5’-UU-3’, or 5’-CC-3’.
  • Backbone-modified nucleotides include, but are not limited to, nucleotides which include modifications to the phosphate-sugar backbone of polynucleotides molecules, and in particular nucleotides which include modifications to the phosphodiester bonds which link the phosphate group and sugar molecule of adjacent nucleotides.
  • the phosphodiester bonds may be modified to include at least one heteroatom, such as a nitrogen or sulfur heteroatom.
  • the phosphoester group of a nucleotide which links to the sugar molecule of an adjacent nucleotide may be replaced with a phosphothioate group.
  • the polynucleotide molecule in particular an RNA molecule, comprises one or more backbone-modified nucleotides, in particular wherein the backbone modification is replacement of the phosphoester group of a nucleotide with a phosphorothioate group.
  • the phosphorothioate group when the backbone modification is replacement of the phosphoester group of a nucleotide with a phosphorothioate group, the phosphorothioate group is located at the first, second, third, fourth, of fifth internucleotide linkage at the 5’ and/or 3’ end of the polynucleotide molecule, in particular an RNA molecule.
  • the polynucleotide molecule in particular an RNA molecule, comprises one or more backbone-modified nucleotides wherein the backbone modification is the replacement of the ribose (ribonucleotide) or deoxyribose (deoxyribonucleotide) sugar moiety with a pyranose or furanose sugar moiety.
  • Base-modified nucleotides include, but are not limited to, nucleotides comprising a non- naturally occurring base rather than a naturally occurring base (adenine, cytosine, guanine, thymine, or uracil).
  • Suitable non-naturally occurring bases include, but are not limited to, uridine and/or cytidine modified at the 5-position, for example 5-(2-amino)propyl uridine, 5- bromo uridine; adenosine and/or guanosine modified at the 8 position, for example 8-bromo guanosine; deaza nucleotides, for example 7-deaza-adenosine; and O- and N-alkylated nucleotides, for example N6-methyl adenosine. Notably, these modifications may be combined.
  • the polynucleotide molecule in particular an RNA molecule, comprises one or more base-modified nucleotides.
  • the modified nucleotides for example sugar-modified, backbone- modified, or based-modified nucleotides, are located proximal to, for example within three nucleotides, five nucleotides, or 10 nucleotides, of the 5’ and/or 3’ end of the polynucleotide molecule.
  • the polynucleotide is not conjugated to any small molecule e.g. the polynucleotide is not conjugated to any to any organic molecule having a molecular weight in the range 250-1500 e.g.300-1000 g/mol. 40
  • the aqueous liquid pharmaceutical formulations of the invention include water as the solvent.
  • Water includes but not is limited to sterile or purified water, sterile water for injection, RNAse free water, or bacteriostatic water for injection.
  • the aqueous liquid pharmaceutical formulation is substantially free of any solvent or co-solvent other than water.
  • the aqueous liquid pharmaceutical formulation does not comprise an organic solvent or co-solvent, such as inter alia ethanol, acetone, dimethyl sulfoxide (DMSO), dichloromethane (DCM), N-methyl pyrrolidinone (NMP), ⁇ , ⁇ ’- dimethylformamide (DMF), N,N’-dimethylacetamide (DMAC), 1,3-dimethyl-2-imidazolidinone (DMEU), 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, and benzyl benzoate.
  • organic solvent or co-solvent such as inter alia ethanol, acetone, dimethyl sulfoxide (DMSO), dichloromethane (DCM), N-methyl pyrrolidinone (NMP), ⁇ , ⁇ ’- dimethylform
  • the term “substantially free of” means that the formulations comprise less than 2% (w/w), for example less than 1% (w/w), such as less than 0.5% (w/w), wherein the % by weight is with respect to the total weight of the formulation.
  • the formulation does not comprise any solvent or co-solvent other than water.
  • the aqueous liquid pharmaceutical formulations according to the present invention may further comprise pharmaceutically acceptable excipients including, but not limited to, antioxidants, buffers, diluents, emulsifiers, lubricants, preservatives, solvents, stabilizers, suspending agents, thickeners, tonicity adjusting (osmotic) agents, vehicles, wetting agents.
  • Suitable antioxidants include but are not limited to ascorbic acid (vitamin C), glutathione (reduced), lipoic acid, uric acid, carotenes, including ⁇ -carotene and retinol (vitamin A), cc- tocopherol (vitamin E), ubiquinol (coenzyme Q), butylated hydroxyanisole, butylated hydroxytoluene, propyl gallate, tert-butylhydroquinone, monothioglycerol, lutein, selenium, manganese, zeaxanthin, or a combination thereof.
  • the aqueous liquid pharmaceutical formulations of the invention may comprise one or more buffers.
  • Suitable buffers include but are not limited to citrate, borate, formate, glycine, alanine, acetate, aspartate, malate, glyoxylate, gluconate, lactate, glycolate, oxalate, histidine, tartarate and succinate buffer systems.
  • references to a “citrate” buffer will be understood to refer to a mixture of citrate and the corresponding acid as a buffer system in a ratio according to the target pH, that is the pH at which the aqueous liquid pharmaceutical formulation is intended to be buffered.
  • the buffer may comprise sodium citrate dihydrate and citric acid monohydrate.
  • the buffer is based on a weak organic acid, for example the buffer is citrate, acetate, lactate, or formate, in particular citrate.
  • a further suitable buffer is phosphate.
  • the buffer may comprise sodium phosphate and disodium phosphate.
  • Suitable, pharmaceutically acceptable, diluents include but are not limited to isotonic saline (0.9% w/v), isotonic dextrose (5% w/v), isotonic mixtures of saline and dextrose (e.g. saline 41 (0.45 % w/v) and dextrose (2.5 % w/v)), sterile or purified water, sterile water for injection or bacteriostatic water for injection.
  • the diluent is sterile or purified water, sterile water for injection, RNAse free water or bacteriostatic water for injection.
  • the diluent is sterile or purified water.
  • the diluent is isotonic saline (0.9% w/v).
  • Suitable preservatives include, but are not limited to, edetic acid and alkali salts thereof, such as disodium edetate (also known as “disodium EDTA”) or calcium edetate (also known as calcium EDTA), phenol, m-cresol, chlorocresol, benzyl alcohol, propyl paraben, methyl paraben, butyl paraben, chlorobutanol, phenylethyl alcohol, benzalkonium chloride, thimerosal, propylene glycol, sorbic acid, benzoic acid derivatives and combinations thereof.
  • disodium edetate also known as “disodium EDTA”
  • calcium EDTA also known as calcium EDTA
  • Suitable suspending agents include, but are not limited to, acacia (gum), sodium alginate, starch and starch derivatives, xanthan gum, pectin, methylcellulose, hydroxyethylcellulose, sodium carboxymethylcellulose (Avicel RC591), microcrystalline cellulose, hypromellose, hyaluronic acid, and combinations thereof.
  • Particularly suitable suspending agents include, microcrystalline cellulose, sodium carboxymethylcellulose (Avicel RC591), hyaluronic acid, and combinations thereof.
  • the properties of certain suspending agents may further render them as suitable thickening agents and/or wetting agents. Accordingly, suitable thickening agents and/or wetting agents, may include, but are not limited to the suspending agents recited above.
  • suitable thickening agents and/or wetting agents include microcrystalline cellulose, sodium carboxymethylcellulose (Avicel RC591), hyaluronic acid, and combinations thereof.
  • suitable tonicity adjusting (osmotic) agents include, but are not limited to, polyols, such as sugars and sugar alcohols, for example erythritol, glycerol, lactose, maltitol, mannitol, sorbitol, trehalose, and xylitol, and salts, for example sodium acetate, sodium lactate, sodium chloride, potassium chloride, and calcium chloride.
  • a particularly suitable tonicity adjusting (osmotic) agent is glycerol.
  • the aqueous liquid pharmaceutical formulation according to the present invention has an osmolarity from about 50 mOsm to about 750 mOsm, for example from about 100 mOsm to about 600 mOsm, for example from about 100 mOsm to about 500 mOsm.
  • the aqueous liquid pharmaceutical formulation according to the present invention suitably has an osmolarity from about 100 mOsm to about 400 mOsm, for example from about 150 mOsm to about 350 mOsm, for example from about 200 mOsm to about 300 mOsm.
  • the pH of the aqueous liquid pharmaceutical formulation according to the present invention is suitably between about 4.0 and about 9.0, such as between about 4.0 and about 8.0, such as between about 4.0 and about 7.0 or between about 5.0 and about 8.0.
  • the pH is suitably between about 4.0 and about 6.0, such as between about 4.0 and about 5.5.
  • the pH of the aqueous liquid pharmaceutical formulation is about 4.0, about 4.1, 42 about 4.2, about 4.3, about 4.4, about 4.5, about, 4.6, about 4.7, about 4.8, about 4.9 or about 5.0.
  • pH is suitably between about 5.5 and about 8.0, such as between about 6.0 and about 8.0, such as between about 6.5 and about 7.5 or between about 7.0 and about 80.
  • the pH of the aqueous liquid pharmaceutical formulation is about 6.5, about 6.6, about 6.7. about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0.
  • pH of such a pharmaceutical composition may be adjusted by pH adjusting agents including acidifying agents such as hydrochloric acid, tartaric acid, citric acid, succinic acid, phosphoric acid, ascorbic acid, acetic acid, lactic acid, sulphuric acid, formic acid and mixtures thereof, or alkaline buffering agents such as ammonium hydroxide, ethylamine, dipropylamine, triethylamine, alkanediamines, ethanolamines, polyalkylene polyamines, heterocyclic amines, hydroxides of alkali metals, such as sodium and potassium hydroxide, hydroxides of alkali earth metals, such as magnesium and calcium hydroxide, and basic amino acids such as L-arginine, lysine, alanine, leucine, isoleucine, oxylysine and histidine, and mixtures thereof.
  • acidifying agents such as hydrochloric acid, tartaric acid, citric acid, succinic acid, phosphoric acid
  • the aqueous liquid pharmaceutical formulations according to the present invention may suitably have a pH between about 4.0 and about 9.0, such as between about 4.0 and about 8.0. It will be understood by the skilled person that an aqueous liquid pharmaceutical formulation suitable for topical administration to the nose may suitably have a pH between about 4.0 and about 9.0, such as between about 4.0 and about 8.0, such as between about 4.0 and about 7.0, such as between about 4.0 and about 6.0, such as between about 4.0 and about 5.0.
  • a particularly suitable buffer for compositions of this desired pH is citrate.
  • an aqueous liquid pharmaceutical formulation suitable for topical administration to the lung may suitably have a pH between about 5.5 and about 8.0, such as between about 6.0 and about 8.0, for example between about 6.0 and about 7.0 or between about 7.0 and about 8.0.
  • a particularly suitable buffer for compositions of this desired pH is phosphate.
  • the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a protein.
  • the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a cationic lipopeptide, for example it does not comprise a polymyxin such as polymyxin B.
  • the aqueous liquid pharmaceutical formulation of the present invention suitably does not comprise an inorganic nanoparticle, suitable or typical examples of which are known in the art and may include inorganic nanoparticles of metal salts, such as zinc oxide, or may include gold, silver, or silica nanoparticles.
  • the aqueous liquid pharmaceutical formulation of the present invention suitably does not comprise a lipid nanoparticle (LNP) or a liposome.
  • the aqueous liquid pharmaceutical formulation of the present invention suitably does not comprise a solid or partly solid (e.g. part solid and part liquid) nanoparticle. Lipid nanoparticles (LNPs) which 43 have in the prior art been described as delivery systems for nucleic acids (see e.g.
  • Kulkarni et al.2021) typically are particles formed of a core comprising a cationic lipid that can bind the nucleic acid, a neutral lipid (such as cholesterol), a helper lipid which contributes to the structure of the particle and an outer formed of a stabiliser molecule.
  • the helper lipids which can include phospholipids, such as phosphatidylcholine or phosphatidylethanolamine, play a role in enhancing the stability of the LNPs by providing structural integrity and facilitating the formation of the lipid bilayer.
  • the stabiliser often a polyethylene glycol (PEG)-lipid, provides a hydrophilic shield that reduces aggregation and opsonization, thus prolonging circulation time in the bloodstream and enhancing the delivery efficiency to target cells.
  • All of the components in LNPs are required for encapsulation efficiency and facilitate cellular uptake and ensures its stability in biological environments.
  • Liposomes are typically spherical vesicles which contain a lipid bilayer.
  • the particles of the stable colloidal emulsion of the present invention do contain any lipid bilayer.
  • the aqueous liquid pharmaceutical formulation according to the present invention is substantially free of lipid nanoparticle (LNP) and liposome components.
  • Lipid nanoparticles are typically described as nanoparticles which comprise a core of cationic or ionizable lipids designed to encapsulate nucleic acids, such as mRNA or siRNA, forming a complex that can be efficiently delivered to target cells.
  • the core structure is stabilised by neutral lipids like cholesterol, which help to maintain the integrity and fluidity of the lipid bilayer.
  • helper lipids such as phosphatidylcholine or phosphatidylethanolamine are incorporated to enhance the encapsulation efficiency of the nucleic acids and facilitate the fusion of the LNP with cellular membranes, thereby improving uptake by target cells.
  • the outer surface of the LNP is often modified with stabiliser molecules, such as polyethylene glycol (PEG)-lipids, which provide a hydrophilic coating that reduces aggregation and opsonization by the immune system.
  • PEG polyethylene glycol
  • This coating prolongs the circulation time of the LNPs in the bloodstream, increasing the likelihood of reaching the target cells.
  • these components work synergistically to create a delivery system for nucleic acids, capable of protecting the genetic material and ensuring its effective delivery and expression within the target cells. Therefore, for example, suitably, the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a neutral lipid.
  • a neutral lipid typically has no ionizable groups, which therefore leads to no charge (whether positive or negative) at around neutral pH (i.e.
  • the pharmaceutical formulation does not comprise cholesterol, or an analogue thereof.
  • the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a cationic lipid, such as quaternary ammonium lipid.
  • the pharmaceutical formulation does not comprise a cationic lipid, such as N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA) or 1,2-dioleoyl-3-trimethylammonium propane (DOTAP).
  • DOTMA N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DOTAP 1,2-dioleoyl-3-trimethylammonium propane
  • Such a cationic 44 lipid typically has at least one ionizable group leading to a net positive charge at around neutral pH (i.e. pH 7.0 or 7.4).
  • the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a helper lipid, such as dioleoylphosphatidylethanolamine (DOPE) or phosphatidylcholine.
  • DOPE dioleoylphosphatidylethanolamine
  • the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a lipid, other than the fatty acid present therein.
  • the fatty acid is oleic acid
  • the aqueous liquid pharmaceutical formulation does not comprise a lipid, other than oleic acid.
  • the aqueous liquid pharmaceutical formulations (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention may be suitable for oral, inhalational, sub- lingual, buccal, or parenteral, including intravenous, subcutaneous, topical, transdermal, pulmonary, rectal, vaginal, ocular, intranasal, intradermal, transmucosal, intraperitoneal, intramuscular, intracapsular, intraorbital, intracardiac, transtracheal, subcuticular, intraarticular, intraspinal, and intrasternal administration.
  • the aqueous liquid pharmaceutical formulations (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention are suitable for topical administration to the lung or nose.
  • the aqueous liquid pharmaceutical formulations of the present invention are suitable for administration via inhalation, for example suitable for administration topically to the lung via oral inhalation, or for intranasal administration.
  • the aqueous liquid pharmaceutical formulation (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention is administered topically to the lung or nose.
  • the aqueous liquid pharmaceutical formulations (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention are administered via inhalation or are administered intranasally.
  • aqueous liquid pharmaceutical formulations of the invention suitable for topical administration to the lung or nose when administered topically to the lung by oral inhalation or topically to the nose may thereby involve administration to the pharynx.
  • a formulation suitable for topical administration to the lung may comprise different pharmaceutically acceptable excipients to a formulation suitable for topical administration to the nose.
  • a formulation suitable for topical administration to the nose may comprise a suspending and/or wetting and/or thickening agent such as microcrystalline cellulose, sodium carboxymethylcellulose (Avicel RC591), hyaluronic acid, or a combination thereof, whilst a formulation suitable for topical administration to the lung may not.
  • the aqueous liquid pharmaceutical formulations (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention are 45 suitable for subcutaneous administration, and in particular via subcutaneous injection.
  • the aqueous liquid pharmaceutical formulation (or the immunostimulatory, immunogenic, or vaccine composition) of the present invention is administered subcutaneously, for example via subcutaneous injection.
  • a formulations suitable for subcutaneous administration will comprise pharmaceutically acceptable excipients appropriate for that route, and that those pharmaceutically acceptable excipients will be different to those present in a formulation suitable for topical administration to the lung or nose.
  • the aqueous liquid pharmaceutical formulations (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention are suitable for ocular administration.
  • the aqueous liquid pharmaceutical formulations of the present invention may be suitable for intra-ocular administration.
  • the aqueous liquid pharmaceutical formulations of the present invention may be suitable for topical administration to the eye.
  • the aqueous liquid pharmaceutical formulation (or the immunostimulatory, immunogenic, or vaccine composition) of the present invention is administered via the ocular administration route (e.g. via the intra-ocular route or via topical administration to the eye).
  • a formulations suitable for ocular administration will comprise pharmaceutically acceptable excipients appropriate for that route, and that those pharmaceutically acceptable excipients will be different to those present in a formulation suitable for topical administration to the lung or nose or a formulation suitable for subcutaneous administration.
  • the aqueous liquid pharmaceutical formulations of the present invention are suitable for administration to a mammal. More suitably, the aqueous liquid pharmaceutical formulations of the present invention are suitable for administration to a human.
  • the aqueous liquid pharmaceutical formulations of the present invention are administered to a mammal.
  • the aqueous liquid pharmaceutical formulations of the present invention are administered to a human.
  • the aqueous liquid pharmaceutical formulations disclosed herein may be administered to a patient or subject once or more than once a day, for example two times a day, three time a day, four times a day or five times a day. Such treatment may extend for a number of weeks or months.
  • Medical Use Without wishing to be bound by theory, the present inventors intend that the aqueous liquid pharmaceutical formulations of the present invention should (in at least some embodiments) enhance, improve, or make more efficient the delivery, and thereby increase the exposure, of a given polynucleotide molecule to a target cell or tissue.
  • a method for enhancing or improving the delivery of a polynucleotide 46 molecule to a target cell or tissue comprising formulating the polynucleotide molecule in an aqueous liquid pharmaceutical formulation of the present invention and administering said formulation to a target cell or tissue.
  • a “target cell or tissue” refers to a cell or tissue which is targeted for the purpose of administration of a polynucleotide molecule, wherein said cell or tissue may also be, or may not be, the cell or tissue that is targeted for the for the purpose of eliciting a therapeutic effect.
  • the target cell or tissue is a cell or tissue of the lung or nose, and in particular a cell or tissue of the nasal or respiratory epithelium.
  • Polynucleotide molecules can elicit a therapeutic effect, upon interaction of the polynucleotide molecule with molecules, organelles, cells, or tissues. Therefore, in one embodiment there is provided an aqueous liquid pharmaceutical formulation according to the present invention for use as a medicament.
  • the aqueous liquid pharmaceutical formulation according to the present invention is for use as a medicament for administration topically to the lung (e.g., by oral inhalation) or topically to the nose.
  • the aqueous liquid pharmaceutical formulations according to the present invention for use as a medicament are administered topically to the lung (e.g., by oral inhalation) or intranasally.
  • the aqueous liquid pharmaceutical formulation according to the present invention is for use as a medicament for administration subcutaneously (e.g. by subcutaneous injection).
  • the aqueous liquid pharmaceutical formulation according to the present invention may be for use as a medicament for administration ocularly.
  • the aqueous liquid pharmaceutical formulation for use according to the present invention is for use therapeutically i.e., in the treatment of disease.
  • a suitable dose of the aqueous liquid pharmaceutical formulation for use as described herein is a therapeutically or prophylactically effective dose which can be determined by the skilled person.
  • the aqueous liquid pharmaceutical formulation for use may be administered to a patient or subject in an amount such that the dose of the polynucleotide molecule is 0.01 ⁇ g/mL – 100000 ⁇ g/mL, for example 0.1 – 100000 ⁇ g/mL, for example 1 – 100000 ⁇ g/mL, for example 1 – 50000 ⁇ g/mL.
  • the aqueous liquid pharmaceutical formulation for use may be administered to a patient or subject in an amount such that the dose of the polynucleotide molecule is 1 – 10000 ⁇ g/mL, for example 1 – 1000 ⁇ g/mL, for example 1 – 100 ⁇ g/mL, for example 1 – 50 ⁇ g/mL, for example 1 – 20 ⁇ g/mL, such as about 10 ⁇ g/mL.
  • the aqueous liquid pharmaceutical formulation for use may be administered to a patient or subject in an amount such that the dose of the polynucleotide molecule is 1 – 10000 ⁇ g/mL, for example 1 – 1000 ⁇ g/mL, for example 10 – 1000 ⁇ g/mL, for example 10 – 500 ⁇ g/mL, for example 10 – 250 ⁇ g/mL, for example 50 – 150 ⁇ g/mL, such as about 100 ⁇ g/mL.
  • the aqueous liquid 47 pharmaceutical formulation for use may be administered to a patient or subject in an amount such that the dose of the polynucleotide molecule is 1 – 100000 ⁇ g/mL, for example 10 – 100000 ⁇ g/mL, for example 100 – 100000 ⁇ g/mL, for example 1000 – 100000 ⁇ g/mL, for example 5000 – 75000 ⁇ g/mL, for example 5000 – 50000 ⁇ g/mL, such as 5000 – 15000 ⁇ g/mL, 10000 – 20000 ⁇ g/mL, 15000 – 25000 ⁇ g/mL, or 25000 – 50000 ⁇ g/mL..
  • the dose will depend on the administration route, sequence, structure, and size of the polynucleotide molecule and indication to be treated.
  • the aqueous liquid pharmaceutical formulations for use as described herein may be administered to a patient or subject once or more than once a day, for example two times a day, three time a day, four times a day or five times a day. Such treatment may extend for a number of weeks or months.
  • the aqueous liquid pharmaceutical formulations for use according to the present invention are administered to a mammal.
  • the aqueous liquid pharmaceutical formulations of the present invention are administered to a human.
  • a polynucleotide molecule in particular an shRNA molecule, may directly or indirectly (e.g., via an encoded protein or other gene product) interact with molecules or cells of the immune system, resulting in activation of the innate and/or adaptive immune system.
  • immune stimulation or activation may have utility in the treatment of infectious disease or cancer.
  • other gene products include inter alia RNA or cDNA molecules. Therefore, in one embodiment there is provided an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the aqueous liquid pharmaceutical formulation stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response.
  • an aqueous liquid pharmaceutical formulation for use according to the present invention wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response, for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response.
  • the present invention provides a method for the treatment of a disease or condition, which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation as described herein, wherein the 48 polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response.
  • the present invention provides use of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response.
  • Immunostimulatory Compositions As discussed above, a polynucleotide molecule may interact with molecules or cells of the immune system, resulting in stimulation activation of the innate and/or adaptive immune system.
  • an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system, for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system.
  • the present invention provides an aqueous liquid pharmaceutical formulation, or immunostimulatory composition, for use in stimulating or activating the immune system in a subject. Accordingly, the present invention provides a method of stimulating the immune system in a subject, comprising administering to the subject an aqueous liquid formulation, or immunostimulatory composition, as described herein.
  • an aqueous liquid pharmaceutical formulation, or immunostimulatory composition in the manufacture of a medicament for stimulating the immune system in a subject.
  • the aqueous liquid formulation, or immunostimulatory compositions of the present invention is for use in stimulating or activating an anti-viral innate and/or adaptive immune response in a subject.
  • the polynucleotide molecule present in an aqueous liquid pharmaceutical formulation for use in stimulating the immune system in a subject is an e.g., directly immunostimulatory polynucleotide molecule, in particular wherein the polynucleotide molecule is an shRNA molecule.
  • the immunostimulatory polynucleotide may be poly I:C (see, for example, dsRNA 2 in the Examples below and US9682096B2, which is incorporated herein by reference). Therefore, in one embodiment, the present invention provides an immunostimulatory composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic 49 surfactant and (ii) an immunostimulatory polynucleotide molecule.
  • the immunostimulatory polynucleotide molecule is capable of, or for use in, stimulating the immune system in a subject.
  • the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular polysorbate 80.
  • the immunostimulatory polynucleotide molecule is an shRNA molecule.
  • the immune system in a subject is considered stimulated if an immune response, including an innate immune response, antibody response or cell-mediated immune response, is initiated, potentiated or enhanced in response to an antigen or immunogen, in particular an exogenous antigen or immunogen.
  • an immunogen is an immunogenic antigen.
  • the immune response is stimulated by inducing a pro-inflammatory cytokine response.
  • the immune response is stimulated by inducing a pro-inflammatory interleukin response, such as one or more of an IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL- 9, IL-11, IL-17, and IL-18 response, or by inducing a pro-inflammatory interform response, such as one or more of an IFN- ⁇ , IFN- ⁇ , and IFN- ⁇ response, or by inducing a pro- inflammatory chemokine response, or by including a pro-inflammatory tumor necrosis factor response, such as a TNF- ⁇ and/or TNF- ⁇ response, or a combination thereof.
  • a pro-inflammatory interleukin response such as one or more of an IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL- 9, IL-11, IL-17, and IL-18 response
  • a pro-inflammatory interform response such as one or more
  • the immune response is stimulated by inducing an interferon response, such as one or more of an IFN- ⁇ , IFN- ⁇ , and IFN- ⁇ response.
  • an interferon response such as one or more of an IFN- ⁇ , IFN- ⁇ , and IFN- ⁇ response.
  • the immune response is stimulated by activating a pattern recognition receptor (PRR).
  • PRRs can induce pro-inflammatory cytokine response, such as pro-inflammatory interferon responses.
  • the PRR can be any member within a family of PRRs selected from the toll-like receptors (TLRs), such as TLR2, TLR3, TLR4, TLR7, TLR8, or TLR9, C-type lectin receptors (CLRs), NOD-like receptors (NLRs) and RIG-I-like receptors (RLRs), such as RIG-I, MDA5 or LGP2.
  • TLRs toll-like receptors
  • CLRs C-type lectin receptors
  • NLRs NOD-like receptors
  • RIG-I-like receptors RIG-I-like receptors
  • the PRR can be any member within the RLRs, such as RIG-I, MDA5, or LGP2, in particular RIG-I.
  • a polynucleotide molecule may interact, for example indirectly (e.g., via an encoded protein or other gene product), with molecules or cells of the immune system, resulting in the raising of an innate and/or adaptive immune response. Therefore, in one embodiment, there is provided an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the polynucleotide molecule is a polynucleotide molecule which raises an innate and/or adaptive immune response in a subject, for use in the treatment of a disease or condition which is treated by raising an innate and/or adaptive immune response.
  • an aqueous liquid pharmaceutical formulation, immunogenic composition, or vaccine composition for use in raising an immune response in a subject, wherein the immune response is raised against the immunogen or vaccine immunogen encoded by the polynucleotide molecule.
  • the present invention provides a method of raising an immune response in a subject, comprising administering to the subject an aqueous liquid formulation as described herein.
  • use of an aqueous liquid pharmaceutical formulation in the manufacture of a medicament for raising an immune response in a subject is provided.
  • the polynucleotide molecule present in an aqueous liquid pharmaceutical formulation, or vaccine composition, for use in raising an immune response in a subject encodes an immunogen.
  • the aqueous liquid pharmaceutical formulation, or vaccine compositions, for use in raising an immune response in a subject is for use therapeutically, that it is the immune response is raised in order to have curative effect on a disease or condition treated by the raising of an innate and/or adaptive immune response e.g., cancer.
  • the compositions are for use prophylactically, that it is the immune response is raised in order to have a protective or prophylactic effect against a disease or condition treated by the raising of an innate and/or adaptive immune response e.g., an infectious disease.
  • an immunogenic composition which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule which encodes an immunogen.
  • the polynucleotide molecule which encodes an immunogen is capable of, or for use in, raising an immune response against said immunogen in a subject.
  • the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular polysorbate 80.
  • the polynucleotide molecule which encodes an immunogen is an mRNA molecule or DNA molecule.
  • a vaccine composition which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule which encodes a vaccine immunogen.
  • the polynucleotide molecule which encodes a vaccine immunogen is capable of, or for use in, raising an immune response against said vaccine immunogen in a subject.
  • the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in 51 particular polysorbate 80.
  • the polynucleotide molecule which encodes a vaccine immunogen is an mRNA molecule or DNA molecule.
  • a vaccine immunogen is an immunogen, that is an immunogenic antigen, capable of raising a therapeutic or protective or immune response in a subject.
  • an aqueous liquid pharmaceutical formulation for use in raising an immune response in a subject may comprise (i) a polynucleotide molecule which encodes an immunogen, against which the immune response may be raised and (ii) a polynucleotide molecule which stimulates the immune system in a subject. That is, an immunogenic composition or vaccine composition according to the present invention may further comprise an immunostimulatory polynucleotide molecule. Suitably said immunostimulatory polynucleotide molecule is an shRNA molecule.
  • the present invention provides an immunogenic composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule which encodes an immunogen, and (iii) an immunostimulatory polynucleotide molecule.
  • the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular polysorbate 80.
  • the polynucleotide molecule which encodes an immunogen is capable of, or for use in, raising an immune response against said immunogen in a subject.
  • the polynucleotide molecule which encodes an immunogen is an mRNA or DNA molecule.
  • the immunostimulatory polynucleotide molecule is capable of, or for use in, stimulating the immune system in a subject.
  • the immunostimulatory polynucleotide molecule is an shRNA molecule.
  • a vaccine composition which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule which encodes a vaccine immunogen, and (iii) an immunostimulatory polynucleotide molecule.
  • the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular polysorbate 80.
  • the polynucleotide molecule which encodes a vaccine immunogen is capable of, or for use in, raising an immune response against said vaccine immunogen in a subject.
  • the polynucleotide molecule which encodes a vaccine immunogen is an mRNA or DNA molecule.
  • the immunostimulatory polynucleotide molecule is capable of, or for use in, stimulating the immune system in a subject.
  • the immunostimulatory polynucleotide molecule is an shRNA molecule.
  • the immunostimulatory or immunogenic compositions described above do not comprise a protein.
  • said compositions do not comprise a cationic lipopeptide, such 52 as polymyxin B.
  • said compositions do not comprise a neutral lipid.
  • said compositions do not comprise cholesterol, or an analogue thereof.
  • said compositions do not comprise a cationic lipid.
  • said compositions do not comprise a helper lipid, such as dioleoylphosphatidylethanolamine (DOPE) or phosphatidylcholine.
  • DOPE dioleoylphosphatidylethanolamine
  • the immunostimulatory or immunogenic compositions described above do not comprise an inorganic nanoparticle, a lipid nanoparticle (LNP), or a liposome and/or are substantially free of LNP and liposome components.
  • the immunostimulatory or immunogenic compositions described above may further comprise pharmaceutically acceptable excipients, at the relevant amounts, as described above.
  • compositions may be limited as described above in relation to the aqueous liquid pharmaceutical formulations of the invention.
  • Immune Stimulation Indications Suitably, a disease or condition treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response is an infectious disease.
  • the infectious disease is suitably bacterial, fungal, parasitic, or viral in origin.
  • the aqueous liquid pharmaceutical formulation of the present invention is for use in the treatment or prevention of infection by a bacteria, fungus or parasite, or disease associated with infection with such a bacteria, fungus, or parasite.
  • the disease or condition treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response is infection by a bacteria, fungus or parasite or disease associated with infection with such a bacteria, fungus, or parasite
  • the bacteria, fungus or parasite infects the brain, circulatory system, endocrine system, eyes, gastrointestinal tract, genital tract, kidneys, liver, respiratory tract, or skin.
  • the disease associated with infection is a disease of the brain, circulatory system, endocrine system, eyes, gastrointestinal tract, genital tract, kidneys, liver, respiratory tract, or skin.
  • the bacteria, fungus or parasite infects the respiratory tract and the disease associated with infection is a disease of the respiratory tract.
  • the bacteria belongs to the genus Bordetella, Chlamydophila, Corynebacterium, Coxiella, Escherichia, Haemophilus, Klebsiella, Legionella, Moraxella, Mycobacterium, Mycoplasma, Proteus, Pseudomonas, Serratia, Staphylococcus, Streptococcus
  • the aqueous liquid pharmaceutical formulation of the present invention is for use in the treatment or prevention of infection by a virus or disease associated with infection with such a virus.
  • the virus is a DNA virus or an RNA virus.
  • the virus has a single-stranded genome, double-stranded genome, or partially double-stranded genome.
  • the single- stranded genome is a sense (+) genome.
  • the single-stranded genome is an antisense (-) genome.
  • the virus may have a single-stranded RNA genome, double-stranded DNA genome, or double-stranded RNA genome.
  • the virus may have a sense (+) single-stranded RNA genome or an antisense (-) single-stranded RNA genome.
  • the virus is naked i.e., is not enveloped.
  • the virus is enveloped.
  • the virus belongs to the family Adenoviridae, Arenaviridae, Astroviridae, Bornaviridae, Bunyaviridae, Caliciviridae, Circoviridae, Coronaviridae, Filoviridae, Flaviviridae, Hepadnaviridae, Hepeviridae, Herpesviridae, Orthomyxoviridae, Papillomaviridae, Polyomaviridae, Paramyxoviridae, Parvoviridae, Picornaviridae, Poxviridae, Reoviridae, Retroviridae, Rhabdoviridae, or Togaviridae.
  • the virus is SARS-CoV-2 and the disease associated with viral infection is COVID-19.
  • the virus is seasonal coronavirus, for example 229E, NL63, OC43, or HKU1
  • the disease associated with viral infection is the disease associated with seasonal coronavirus, for example 229E, NL63, OC43, or HKU1, infection.
  • the virus is influenza virus and the disease associated with viral infection is influenza.
  • the virus is respiratory syncytial virus (RSV) and the disease associated with viral infection is the disease associated with RSV infection.
  • the virus is human rhinovirus (HRV) and the disease associated with viral infection is the disease associated with HRV infection.
  • the virus is Middle East respiratory syndrome (MERS)-CoV and the disease associated with viral infection is MERS.
  • MERS Middle East respiratory syndrome
  • the virus is an avian influenza virus and the disease associated with viral infection is avian influenza.
  • the virus is Nipah virus and the disease associated with viral infection is the disease associated with Nipah virus infection.
  • the virus is a human parainfluenza virus (HPIV) and the disease associated with viral infection is the disease associated with HPIV infection.
  • the virus is a human metapneumovirus (hMPV) and the disease associated with viral infection is the disease associated with hMPV infection.
  • a disease or condition treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response is cancer.
  • the cancer may be breast cancer, bladder cancer, renal cancer, lung cancer, prostate cancer, bone cancer, brain cancer, cervical cancer, anal cancer, colon cancer, colorectal cancer, gastric cancer, blood cancer, such as leukaemia, lymphoma, or myeloma, liver cancer, skin cancer, ovarian cancer, pancreatic cancer, testicular cancer, thyroid cancer, vaginal cancer, cardiac cancer or sarcoma.
  • a polynucleotide molecule in particular an mRNA or DNA molecule, may be intended to establish expression of one or more proteins (or other gene products) which are encoded by the polynucleotide molecule, in order to supplement endogenous gene expression such that over-expression of the protein (or other gene product) facilitates interference with endogenous cellular process, such as the regulation of gene expression or signal transduction.
  • over-expression of the protein (or other gene product) may be intended to activate an immune response or stimulate the immune system, for example against a tumour over- expressing an antigen, or in response to an immune cell presenting an exogenous antigen.
  • an aqueous liquid pharmaceutical formulation for use according to the present invention wherein the aqueous liquid pharmaceutical formulation increases the endogenous expression of a protein (or other gene product).
  • the aqueous liquid pharmaceutical formulation increases the endogenous expression of a functional protein (or other gene product).
  • the aqueous liquid pharmaceutical formulation may establish the expression of a functional protein (or other gene product), whilst endogenous expression of said protein (or other gene product) produces a non-functional or dysfunctional e.g., truncated or misfolded protein (or other gene product).
  • the term “increases” includes restoring i.e., increasing from zero or a low value to a “normal” value and enhancing i.e., increasing from a “normal” value to a high value, endogenous gene expression.
  • the present invention provides an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein (or other gene product) or increases endogenous expression of a functional protein (or other gene product), is for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein (or other gene product), or by increasing the expression of a functional protein (or other gene product).
  • an aqueous liquid pharmaceutical formulation for use in a method of gene therapy, wherein the polynucleotide molecule, in particular an mRNA or DNA molecule, encodes a therapeutic gene, protein (or other gene product).
  • the polynucleotide molecule in particular an mRNA or DNA molecule, encodes a therapeutic gene, protein (or other gene product).
  • said use in a method of gene therapy is for the treatment of a disease or condition which is treated by increasing the endogenous expression of the therapeutic gene, protein (or other gene product) encoded by said polynucleotide molecule.
  • the present invention provides a method for the treatment of a disease or condition, which is treated by increasing the endogenous expression of a protein (or other gene product), or by increasing the expression of a functional protein (or other gene product), comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein (or other gene product) or increases endogenous expression of a functional protein (or other gene product).
  • a method of gene therapy comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule, in particular an mRNA or DNA molecule, encodes a therapeutic gene, protein (or other gene product).
  • said method of gene therapy is for the treatment of a disease or condition 56 which is treated by increasing the endogenous expression of the therapeutic gene, protein (or other gene product) encoded by said polynucleotide.
  • the present invention also provides use of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein (or other gene product) or increases endogenous expression of a functional protein (or other gene product), in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein (or other gene product), or by increasing the expression of a functional protein (or other gene product).
  • said use in a method of gene therapy is for the treatment of a disease or condition which is treated by increasing the endogenous expression of the therapeutic gene, protein (or other gene product) encoded by said polynucleotide.
  • a disease or condition treated by increasing the endogenous expression of a protein (or other gene product), or by increasing the endogenous expression of a functional protein (or other gene product) is a monogenic or polygenic disease or condition.
  • the disease or condition is a blood disease or condition, such as anaemia, in particular sickle-cell anaemia, haemophilia, in particular haemophilia A or B, severe combined immune deficiency (SCID), thalassemia, or Von Willebrand disease, a hearing disease or condition, such as deafness, a heart disease or condition, such as atherosclerosis, coronary heart disease, Long QT syndrome, or Von-Hippel Lindau syndrome, a metabolic disease or condition, including lysosomal storage diseases and conditions, such as Type I diabetes, Gaucher disease, glycogen storage disease, or obesity, a musculoskeletal disease or condition, such as Duchenne muscular dystrophy, or achondroplasias, a nervous system or brain disease or condition, such as Alzheimer’s disease, amyotrophic lateral sclerosis (ALS), Rett’s syndrome, Fragile X syndrome, Huntington’s disease, or Parkinson’s disease, a digestive or renal disease or condition, such
  • the disease or condition is a respiratory disease or condition, such as asthma, alpha-1 antitrypsin deficiency, chronic obstructive pulmonary disease (COPD), primary ciliary dyskinesia (PCD), pulmonary fibrosis, sarcoidosis or cystic fibrosis.
  • COPD chronic obstructive pulmonary disease
  • PCD primary ciliary dyskinesia
  • pulmonary fibrosis sarcoidosis or cystic fibrosis.
  • the disease or condition is cystic fibrosis and the polynucleotide molecule present in the formulation according to the present invention increases the endogenous expression of the, or increases the endogenous expression of the functional, cystic fibrosis transmembrane conductance regulator (CFTR) protein (see WO2022/204270A1, which is herein incorporated by reference).
  • CFTR cystic fibrosis transmembrane conductance regulator
  • the disease or condition is PCD and the polynucleotide molecule present in the formulation according to the present invention increases the endogenous expression of a, or increases the endogenous expression of a functional, PCD- associated protein, such as dynein axonemal intermediate chain 1 (see WO2022/198099A1 and WO2022/204215A1, which are herein incorporated by reference).
  • a functional, PCD- associated protein such as dynein axonemal intermediate chain 1 (see WO2022/198099A1 and WO2022/204215A1, which are herein incorporated by reference).
  • a polynucleotide molecule in particular an miRNA or siRNA molecule, may be intended to down-regulate, reduce, silence or knock-down expression of an endogenous gene, in particular when said gene, and consequently the encoded protein (or other gene product), is over-expressed and said over-expression is contributing to cellular dysfunction, or when said gene, and consequently the encoded protein (or other gene product) is defective or dysfunctional and therefore contributing to cellular dysfunction.
  • a polynucleotide molecule in particular an miRNA or siRNA molecule, may be intended to down-regulate, reduce, silence or knock-down expression of an endogenous gene in order to reduce endogenous gene expression such that under-expression of the protein (or other gene product) interferes with other endogenous cellular processes such as the regulation of gene expression or signal transduction.
  • under-expression of the protein (or other gene product) may be intended to activate an immune response or stimulate the immune system, for example by removal of “self-antigens”. Therefore, in one embodiment there is provided an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the aqueous liquid pharmaceutical formulation decreases the endogenous expression of a protein (or other gene product).
  • an aqueous liquid pharmaceutical formulation for use according to the present invention wherein the polynucleotide molecule is a polynucleotide molecule which decreases the endogenous expression of a protein (or other gene product), is for use in the treatment of a disease or condition which is treated by decreasing the endogenous expression of a protein (or other gene product).
  • the present invention provides a method for the treatment of a disease or condition, which is treated by decreasing the endogenous expression of a protein 58 (or other gene product), comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which decreases the endogenous expression of a protein (or other gene product).
  • a polynucleotide molecule in particular a DNA molecule, may be intended to modify, for example repair, excise, or insert or exchange bases or stretches of, endogenous nucleic acid or polynucleotide sequences, for example RNA or DNA sequences.
  • the polynucleotide molecule which is intended to modify endogenous nucleic acid or polynucleotide sequences may be a guide RNA (gRNA), which may function as a component of the CRISPR-Cas9 gene editing technology.
  • gRNA guide RNA
  • an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the aqueous liquid pharmaceutical formulation modifies an endogenous nucleic acid sequence, for example an mRNA molecule or the genome.
  • the aqueous liquid pharmaceutical formulation may repair, excise bases or stretches from, insert bases or stretches into, or exchange bases or stretches with, an endogenous nucleic acid sequence, e.g., the genome.
  • an aqueous liquid pharmaceutical formulation for use according to the present invention wherein the polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence, is for use in the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence.
  • a method for the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation as described herein, wherein the 59 polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence.
  • the present invention provides use of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence.
  • a polynucleotide molecule can interact with, interfere with, modify or otherwise influence essentially any cellular process.
  • the aqueous liquid pharmaceutical formulation according to the present invention may be for use as a medicament for administration ocularly, such as by administration topically to the eye.
  • the aqueous liquid pharmaceutical formulation according to the present invention is for use as a medicament for administration ocularly, it is suitably for use in the prevention or treatment of an ocular disease or condition, for example an ocular disease or condition selected from dry eye–Sjogren’s syndrome, Meesmann epithelial corneal dystrophy, herpes simplex keratitis (HSK), mucopolysaccharidosis (MPS), ectrodactyly-ectodermal dysplasia- clefting (EEC) syndrome, ocular hypertension and open angle glaucoma, retinoschisis, choroideremia, achromatopsia, recurrent retinoblastoma.
  • an ocular disease or condition selected from dry eye–Sjogren’s syndrome, Meesmann epithelial corneal dystrophy, herpes simplex keratitis (HSK), mucopolysaccharidosis (MPS), ectrodactyly
  • an ocular disease or condition for example an ocular disease or condition selected from dry eye–Sjogren’s syndrome, Meesmann epithelial corneal dystrophy, herpes simplex keratitis (HSK), mucopolysaccharidosis (MPS), ectrodactyly-ectodermal dysplasia-clefting (EEC) syndrome, ocular hypertension and open angle glaucoma, retinoschisis, choroideremia, achromatopsia, recurrent retinoblastoma, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation according to the present invention.
  • an ocular disease or condition selected from dry eye–Sjogren’s syndrome, Meesmann epithelial corneal dystrophy, herpes simplex keratitis (HSK), mucopolysaccharidosis (MPS), ectrodactyly-ecto
  • an aqueous liquid pharmaceutical formulation according to the present invention in the manufacture of a medicament for use in the treatment of an ocular disease or condition, for example an ocular disease or condition selected from dry eye–Sjogren’s syndrome, Meesmann epithelial corneal dystrophy, herpes simplex keratitis (HSK), mucopolysaccharidosis (MPS), ectrodactyly-ectodermal dysplasia-clefting (EEC) syndrome, ocular hypertension and open angle glaucoma, retinoschisis, choroideremia, achromatopsia, recurrent retinoblastoma.
  • an ocular disease or condition selected from dry eye–Sjogren’s syndrome, Meesmann epithelial corneal dystrophy, herpes simplex keratitis (HSK), mucopolysaccharidosis (MPS), ectrodactyly-ectodermal dysplasia-clefting
  • Formulations of the invention may be produced by mixing the ingredients leading to a colloidal emulsion.
  • an aqueous liquid pharmaceutical formulation of the present invention is produced by a stepwise process as described below. It will be understood that the skilled person would be aware of minor modifications to the process which may nevertheless lead to a process suitable for the production of an aqueous liquid pharmaceutical formulation as described herein. Moreover, the skilled person would be able to determine optimised parameters for the process described below, for example temperature, mixing time, pH, based on the surfactant component, polynucleotide molecule, and concentrations thereof etc.
  • a process for the production or preparation of an aqueous liquid pharmaceutical formulation, or immunostimulatory composition, immunogenic composition, or vaccine composition according to the present invention.
  • the process may comprise, consist of, or consist essentially of one or more of the steps described below.
  • the surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, for example a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, is added at the required concentration to water, for example RNAse free water, and mixed using an overhead stirrer at between 25% and 75% power, for example at between 40% and 50% power, or at about 50% power. Mixing continues for any period of time until a homogenous surfactant component mixture is achieved, such as for between about 1-60 minutes, for example for between about 1-30 minutes, for example for between about 5-20 minutes, such as for about 10 minutes.
  • a homogenous surfactant component mixture is achieved, such as for between about 1-60 minutes, for example for between about 1-30 minutes, for example for between about 5-20 minutes, such as for about 10 minutes.
  • An exemplary overhead stirrer is the IKA EURO-STAR Overhead Lab Mixer.
  • a specific volume of polynucleotide molecule stock solution which typically comprises the polynucleotide molecule at a concentration of about 0.2 – 20% (w/w), wherein the % by weight is with respect to the total weight of the formulation, is dispersed into the homogenous surfactant component mixture described above in order to achieve a desired final concentration of polynucleotide molecule.
  • the formulation is then mixed using an overhead stirrer at between 25% and 75% power, for example at between 40% and 50% power, or at about 50% power, for any period of time until a uniform active surfactant component mixture is produced.
  • Suitably mixing may occur for between about 1-60 minutes, for example for between about 1-30 minutes, for example for between about 5-20 minutes, such as for about 10 minutes.
  • these may be added, either simultaneously, stepwise, or a combination 61 thereof, at the required concentration to water, for example RNAse free water and homogenised until the mixture achieves a desired rheology.
  • homogenisation may occur at an rpm (revolutions per minutes) of 2000-20000, such as 2000-15000, 4000- 12000, or 5000-10000 for between about 1-20 minutes, for example for between about 1-10 minutes, such as for between about 1-5 minutes.
  • the further pharmaceutically acceptable excipient composition will be combined with the active surfactant component mixture described above and mixed using an overhead stirrer at between 25% and 75% power, for example at between 40% and 50% power, or at about 50% power. Mixing will continue for any period of time until a homogenous and uniform aqueous liquid pharmaceutical formulation is produced. Suitably mixing may occur for between about 1-60 minutes, for example for between about 1-30 minutes, for example for between about 5-20 minutes, such as for about 10 minutes.
  • An exemplary homogeniser is the Silverson L5M homogenizer. It will be understood by the skilled person that the further pharmaceutically acceptable excipients included in an aqueous liquid pharmaceutical formulation of the present invention will depend on the surfactant component, polynucleotide molecule, and administration route.
  • an aqueous liquid pharmaceutical formulation suitable for topical administration to the nose may comprise carboxymethyl cellulose, for example at a concentration of about 2% (w/w), hyaluronic acid, for example at a concentration of about 1% (w/w), and glycerol, for example at a concentration of about 2.1% (w/w), wherein the % by weight is with respect to the total weight of the formulation. Therefore, it is these excipients that may be present in the further pharmaceutically acceptable excipient composition described above.
  • the aqueous liquid pharmaceutical formulation will be buffered to a final desired pH, for example using a buffer such as a citrate buffer (e.g.
  • the buffered aqueous liquid pharmaceutical formulation will be mixed, for example using an overhead stirrer as described above, until unform pH adjustment is achieved. For example, mixing may occur for between about 1-60 minutes, for example for between about 1-30 minutes, for example for between about 5-20 minutes, such as for about 10 minutes.
  • each of the steps described above is performed at room temperature (i.e. between about 20 and 30 o C).
  • An aqueous liquid pharmaceutical formulation comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule.
  • a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule.
  • the aqueous liquid pharmaceutical formulation according to clause 2 wherein the fatty acid is selected from caprylic acid and oleic acid.
  • Clause 4. The aqueous liquid pharmaceutical formulation according to clause 3, wherein the fatty acid is caprylic acid.
  • Clause 5. The aqueous liquid pharmaceutical formulation according to clause 3, wherein the fatty acid is oleic acid.
  • Clause 6. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 5, wherein the non-ionic surfactant is selected from the group consisting of polyoxyalkylenes, such as poloxamers, alkyl ethers of polyethylene glycol, alkylphenyl ethers of polyethylene glycol, and fatty acid esters, such as polyoxyethylene sorbitan fatty acid esters.
  • aqueous liquid pharmaceutical formulation according to clause 6 wherein the non-ionic surfactant is selected from alkyl ethers of polyethylene glycol and polyoxyethylene sorbitan fatty acid esters.
  • the non-ionic surfactant is an alkyl ether of polyethylene glycol.
  • the non-ionic surfactant is Brij 35 (polyoxyethylene (23) lauryl ether).
  • Clause 12 The aqueous liquid pharmaceutical formulation according to clause 11, wherein the non-ionic surfactant is polysorbate 80, polysorbate 120, polysorbate 85, polysorbate 65, polysorbate 60, polysorbate 40, or polysorbate 20.
  • Clause 13 The aqueous liquid pharmaceutical formulation according to clause 12, wherein the non-ionic surfactant is polysorbate 80. 63 Clause 14.
  • aqueous liquid pharmaceutical formulation according to clause 1, wherein the surfactant component is selected from the group consisting of mixtures of (a) oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (b) lauric acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (c) linoleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (d) linolenic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (e) palmitic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (f) stearic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (g) oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyalkylene, such as a polox
  • Clause 15 The aqueous liquid pharmaceutical formulation according to clause 14, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester.
  • Clause 16 The aqueous liquid pharmaceutical formulation according to clause 15, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester selected from polysorbate 80 and polysorbate 20.
  • Clause 17 The aqueous liquid pharmaceutical formulation according to clause 16, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and polysorbate 80.
  • Clause 19 The aqueous liquid pharmaceutical formulation according to clause 18, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester selected from polysorbate 80 and polysorbate 20.
  • 64 Clause 20 The aqueous liquid pharmaceutical formulation according to clause 19, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and polysorbate 80.
  • Clause 22. The aqueous liquid pharmaceutical formulation according to clause 21, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol selected from Brij 35 (polyoxyethylene (23) lauryl ether), Brij 52 (polyoxyethylene (20) cetyl ether), Brij 93 (polyoxyethylene (2) oleyl ether), Brij 97 (polyoxyethylene (10) oleyl ether), Brij L4 (polyoxyethylene (4) lauryl ether), Brij 30 (polyoxyethylene (4) lauryl ether), or Brij 78 (polyoxyethylene(20) stearyl ether).
  • Clause 23 The aqueous liquid pharmaceutical formulation according to clause 22, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and Brij 35 (polyoxyethylene (23) lauryl ether).
  • Clause 24 The aqueous liquid pharmaceutical formulation according to clause 14, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol.
  • aqueous liquid pharmaceutical formulation according to clause 24, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol selected from Brij 35 (polyoxyethylene (23) lauryl ether), Brij 52 (polyoxyethylene (20) cetyl ether), Brij 93 (polyoxyethylene (2) oleyl ether), Brij 97 (polyoxyethylene (10) oleyl ether), Brij L4 (polyoxyethylene (4) lauryl ether), Brij 30 (polyoxyethylene (4) lauryl ether), or Brij 78 (polyoxyethylene(20) stearyl ether).
  • the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol selected from Brij 35 (polyoxyethylene (23) lauryl ether), Brij 52 (polyoxyethylene (20) cetyl ether), Brij 93 (polyoxyethylene (2) oleyl
  • aqueous liquid pharmaceutical formulation according to clause 25, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and Brij 35 (polyoxyethylene (23) lauryl ether).
  • the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and Brij 35 (polyoxyethylene (23) lauryl ether).
  • Brij 35 polyoxyethylene (23) lauryl ether.
  • aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 27, wherein the concentration of the surfactant component in the formulation is 0.2 – 30000 ⁇ g/mL, for example 1 – 30000 ⁇ g/mL, for example 1 – 20000 ⁇ g/mL, 5 – 20000 ⁇ g/mL, 5 – 15000 ⁇ g/mL, 5 – 10000 ⁇ g/mL, or 5-5000 ⁇ g/mL.
  • 65 Clause 30.
  • the aqueous liquid pharmaceutical formulation according to clause 29, wherein the concentration of the surfactant component in the formulation is 50-200 ⁇ g/mL, for example 75-150 ⁇ g/mL, for example 90 – 120 ⁇ g/mL, or about 100 ⁇ g/mL. Clause 31.
  • aqueous liquid pharmaceutical formulation according to clause 28, wherein the concentration of the surfactant component in the formulation is 500-2000 ⁇ g/mL, for example 750-1500 ⁇ g/mL, for example 900 – 1200 ⁇ g/mL, or about 1000 ⁇ g/mL. Clause 32.
  • aqueous liquid pharmaceutical formulation according to clause 28, wherein the fatty acid is present in the formulation at a concentration of 10 – 100 ug/mL, for example 20 – 80 ⁇ g/mL, for example 25 – 75 ⁇ g/mL, for example 40 – 60 ⁇ g/mL, or about 50 ⁇ g/mL; and wherein the non-ionic surfactant is present in the formulation at a concentration of 10 – 100 ug/mL, for example 20 – 80 ⁇ g/mL, for example 25 – 75 ⁇ g/mL, for example 30 – 60 ⁇ g/mL, for example 40 – 50 ⁇ g/mL.
  • Clause 33 wherein the fatty acid is present in the formulation at a concentration of 10 – 100 ug/mL, for example 20 – 80 ⁇ g/mL, for example 25 – 75 ⁇ g/mL, for example 40 – 60 ⁇ g/mL, or about 50 ⁇ g/mL; and where
  • aqueous liquid pharmaceutical formulation according to clause 28, wherein the fatty acid is present in the formulation at a concentration of 100 – 1000 ug/mL, for example 200 – 800 ⁇ g/mL, for example 250 – 750 ⁇ g/mL, for example 400 – 600 ⁇ g/mL, or about 500 ⁇ g/mL; and wherein the non-ionic surfactant is present in the formulation at a concentration of 100 – 1000 ug/mL, for example 200 – 800 ⁇ g/mL, for example 250 – 750 ⁇ g/mL, for example 300 – 600 ⁇ g/mL, for example 400 – 500 ⁇ g/mL.
  • Clause 35. The aqueous liquid pharmaceutical formulation according to clause 34, wherein the ratio of the amount of fatty acid or a pharmaceutically acceptable salt thereof to non-ionic surfactant, wherein each is measured in ⁇ g/mL, is between about 3:2 and about 2:3, for example between about 6:5 and about 1:1, e.g. about 10:9 or about 11:10.
  • Clause 36 The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 35, wherein the ratio of the amount of the surfactant component to polynucleotide molecule, wherein each is measured in ⁇ g/mL, is between about 100:1 and about 1:1000, between about 100:1 and about 1:750, between about 75:1 and about 1:750, between about 50:1 and about 1:750, between about 50:1 and about 1:500, between about 50:1 and about 1:250, between about 50:1 and about 1:200, between about 40:1 and about 1:200, between about 40:1 and about 1:150, between about 25:1 and about 1:150, for example between about 25:1 and about 1:100.
  • the ratio of the amount of the surfactant component to polynucleotide molecule, wherein each is measured in ⁇ g/mL is between about 100:1 and about 1:1000, between about 100:1 and about 1:750, between about 75:1 and about 1:750, between about 50:1 and about
  • Clause 38. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 36, wherein the polynucleotide molecule is a double-stranded polynucleotide molecule.
  • Clause 39. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 38, wherein the polynucleotide molecule comprises an intramolecular structure. Clause 40.
  • RNA ribonucleic acid
  • Clause 41 The aqueous liquid pharmaceutical formulation according to clause 40, wherein the RNA molecule is an mRNA, miRNA, shRNA or siRNA molecule.
  • Clause 42 The aqueous liquid pharmaceutical formulation according to clause 41, wherein the RNA molecule is an mRNA molecule.
  • Clause 45. The aqueous liquid pharmaceutical formulation according to clause 44, wherein the miRNA molecule comprises from about 20 to about 25 nucleotides.
  • RNA molecule is an shRNA molecule.
  • shRNA molecule comprises from about 10 to about 70 nucleotides, for example from about 20 to about 70 nucleotides, for example from about 35 to about 70 nucleotides or from about 25 to about 35 nucleotides.
  • shRNA molecule comprises 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 nucleotides.
  • the shRNA molecule comprises a double-stranded region comprising 30 base pairs or less, such as 25 base pairs or less, such as 20 base pairs or less, such as 18 base pairs or less, such as 16 base pairs or less, such as 14 base pairs or less, such as 12 base pairs or less, such as 10 base pairs or less, such as 8 base pairs or less, such as 6 base pairs or less, such as 4 base pairs or less.
  • aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 51, wherein the shRNA molecule comprises a blunt end.
  • Clause 53 The aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 52, wherein the shRNA molecule comprises at least one 5’ triphosphate or at least one 5’ diphosphate, in particular wherein the 5’ triphosphate or 5’ diphosphate is located at the 5’ terminus.
  • Clause 55. The aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 54, wherein the shRNA molecule comprises or consists of SEQ ID NO.1 or a variant of SEQ ID NO.1.
  • Clause 56. The aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 54, wherein the shRNA molecule comprises or consists of SEQ ID NO.2, SEQ ID NO.
  • Clause 59 The aqueous liquid pharmaceutical formulation according to clause 58, wherein the siRNA molecule comprises from about 20 to about 60 nucleotides, for example from about 30 to about 60 nucleotides, such as from about 40 to about 50 nucleotides.
  • Clause 60 The aqueous liquid pharmaceutical formulation according to clause 59, wherein the siRNA molecule comprises 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, or 60 nucleotides.
  • aqueous liquid pharmaceutical formulation according to any one of clauses 58 to 63, wherein the siRNA molecule comprises at least one, e.g. two, 5’ monophosphate(s), or at least one, e.g. two, 5’ diphosphate(s), or at least one, e.g. two, 5’ triphosphate(s).
  • Clause 65 The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 39, wherein the polynucleotide molecule is a deoxyribonucleic acid (DNA) molecule.
  • the aqueous liquid pharmaceutical formulation according to clause 65 wherein the DNA molecule is a genomic DNA (gDNA) molecule, a complementary DNA (cDNA) molecule, or an extra-chromosomal DNA molecule, for example a plasmid DNA molecule.
  • gDNA genomic DNA
  • cDNA complementary DNA
  • an extra-chromosomal DNA molecule for example a plasmid DNA molecule.
  • aqueous liquid pharmaceutical formulation according to any one of clauses 65 to 69, wherein the DNA molecule comprises from about 100 to about 20000 nucleotides, such as from about 100 to about 15000 nucleotides, such as from about 500 to about 15000 nucleotides, such as from about 500 to about 10000 nucleotides, such as from about 2500 to about 10000 nucleotides, such as from about 2500 to about 8000 nucleotides.
  • Clause 71 Clause 71.
  • aqueous liquid pharmaceutical formulation according to clause 1, wherein the polynucleotide molecule has the structure of Formula (I): 5’-P z -(N) b N-3’-(E) y (E)-L-(E)(E) y’ -5’-N(N) b’ -3’ wherein 5’-P z -(N) b N-3’ represents the first nucleic acid sequence; 5’-N(N) b’ -3’ represents the second nucleic acid sequence; P at each instance is independently a phosphate or analog thereof; z is 2 or 3; N is, at each instance, any nucleotide or modified nucleotide or analog or derivative thereof; b and b’ are independently 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18; 5’-(E) y (E)-L-(E)(E) y’ -3’ represents the connector element wherein 69 E at each occurrence is independently any nucleotide, modified nucleo
  • Clause 72 The aqueous liquid pharmaceutical formulation according to clause 71, wherein the polynucleotide molecule has the structure of Formula (II): 5’-P z -Nu-3’-(E) y (E)-L-(E)(E) y’ -5’-Nu’-3’ wherein 5’-P z -Nu-3’ represents the first nucleic acid sequence; 5’-Nu’-3’ represents the second nucleic acid sequence; P at each instance is independently a phosphate or analog thereof.
  • Formula (II) 5’-P z -Nu-3’-(E) y (E)-L-(E)(E) y’ -5’-Nu’-3’ wherein 5’-P z -Nu-3’ represents the first nucleic acid sequence; 5’-Nu’-3’ represents the second nucleic acid sequence; P at each instance is independently a phosphate or analog thereof.
  • E at each occurrence is independently any nucleotide, modified nucleotide, or abasic; 70 y and y’ are independently 0-9, provided that y + y’ equals 0-8; L is a non-nucleotide segment having the structure O X X and X’ are independently O or S; Y and Y’ are independently OR”, SR”, or NRR’; V and V’ are independently O, S, or NRR’; q is 1-20; k is 1-20; t is 1-20; M selected from aliphatic, substituted aliphatic, aryl, substituted aryl, heteroalkyl, heterocyclyl or substituted heterocyclyl; W is any reactive group or conjugation group; and d is 0 or 1.
  • Clause 73 The aqueous liquid pharmaceutical formulation according to clause 1, wherein the polynucleotide molecule comprises, consists of, or consists essentially of SEQ ID NO.13 and SEQ ID NO.14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L1.
  • Clause 74 The aqueous liquid pharmaceutical formulation according to clause 1, wherein the polynucleotide molecule comprises, consists of, or consists essentially of SEQ ID NO.13 and SEQ ID NO.14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L1.
  • Clause 75 The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 74, wherein the polynucleotide molecule comprises one or more sugar-modified nucleotides which each comprise a 2’ OH (or 2’ H) modification. 71 Clause 76.
  • the 2’ OH (or 2’ H) modification is selected from the group consisting of 2'-deoxy, 2’-fluoro, 2'- deoxy- methoxyethyl (2'-O-MOE), 2'-O-amino-propyl (2'-O-AP), 2'- O- , 2'-O-dimethylaminopropyl (2'-O-DMAP), 2'
  • Clause pharmaceutical formulation according to any one of clauses 1 to molecule comprises one or more backbone-modified the backbone modification is replacement of the with a phosphorothioate group.
  • Clause pharmaceutical formulation according to any one of clauses 1 to molecule comprises one or more base-modified
  • Clause pharmaceutical formulation according to any one of clauses 1 to pharmaceutical formulation is in the form of a stable colloidal emulsion.
  • Clause 80. An aqueous liquid pharmaceutical formulation in the form of a stable colloidal emulsion comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule.
  • aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 82, wherein the aqueous liquid pharmaceutical formulation is suitable for oral, inhalational, sub-lingual, buccal, or parenteral, including intravenous, subcutaneous, topical, transdermal, pulmonary, rectal, vaginal, ocular, intranasal, intradermal, transmucosal, intraperitoneal, intramuscular, intracapsular, intraorbital, intracardiac, transtracheal, subcuticular, intraarticular, intraspinal, or intrasternal administration.
  • Clause 84 The aqueous liquid pharmaceutical formulation according to clause 83, wherein the aqueous liquid pharmaceutical formulation is suitable for topical administration to the lung or nose. 72 Clause 85.
  • Clause 90. The aqueous liquid pharmaceutical formulations for use according to clause 87, for administration ocularly, for example intra-ocularly or topically to the eye.
  • Clause 91. The aqueous liquid pharmaceutical formulation for use according to any one of clauses 87 to 90, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response, for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response.
  • an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response.
  • Clause 94 The aqueous liquid pharmaceutical formulation for use, method, or use, according to any one of clauses 91 to 93, wherein the disease or condition is infection by a virus or associated with infection with such a virus. 73 Clause 95.
  • Clause 96. The aqueous liquid pharmaceutical formulation for use, method, or use, according to clause 95, wherein the virus is SARS-CoV-2 and the disease associated with viral infection is COVID-19; or wherein the virus is seasonal coronavirus, for example 229E, NL63, OC43, or HKU1, and the disease associated with viral infection is the disease associated with seasonal coronavirus, for example 229E, NL63, OC43, or HKU1, infection; or wherein the virus is influenza virus and the disease associated with viral infection is influenza; or wherein the virus is respiratory syncytial virus (RSV) and the disease associated with viral infection is the disease associated with RSV infection; or wherein the virus is human rhinovirus (HRV) and the disease associated with viral infection is the disease associated with HRV infection; or wherein the virus is Middle East
  • Clause 97 The aqueous liquid pharmaceutical formulation for use according to any one of clauses 87 to 90, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein or other gene product, or which increases endogenous expression of a functional protein or other gene product, for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein or other gene product, or by increasing the expression of a functional protein or other gene product.
  • Clause 98 Clause 98.
  • a method for the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein or other gene product, or by increasing the expression of a functional protein or other gene product comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the 74 polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein or other gene product, or which increases endogenous expression of a functional protein or other gene product.
  • aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein or other gene product, or which increases endogenous expression of a functional protein or other gene product, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein or other gene product, or by increasing the expression of a functional protein or other gene product.
  • Clause 100 is a polynucleotide molecule which increases endogenous expression of a protein or other gene product, or which increases endogenous expression of a functional protein or other gene product
  • the aqueous liquid pharmaceutical formulation for use according to clause 101 for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of the therapeutic gene, protein or other gene product encoded by the polynucleotide molecule.
  • Clause 103 The aqueous liquid pharmaceutical formulation for use according to any one of clauses to 87 to 90, wherein the polynucleotide molecule is a polynucleotide molecule which decreases the endogenous expression of a protein or other gene product, for use in the treatment of a disease or condition which is treated by decreasing the endogenous expression of protein or other gene product.
  • an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which decreases the endogenous expression of a protein or other gene product, in the manufacture 75 of a medicament for use in the treatment of a disease or condition which is treated by decreasing the endogenous expression of a protein or other gene product.
  • Clause 106 The aqueous liquid pharmaceutical formulation for use according to any one of clauses 87 to 90, wherein the polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence, for use in the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence.
  • Clause 107 A method for the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence.
  • an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence.
  • An immunostimulatory composition which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) an immunostimulatory polynucleotide molecule.
  • An immunogenic composition which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule which encodes an immunogen.
  • a vaccine composition which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule which encodes a vaccine immunogen. 76 Clause 115.
  • An immunogenic composition or vaccine composition according to any one of clauses 113 to 117, wherein the composition does not comprise a lipid nanoparticle (LNP) or a liposome, and/or wherein the composition is substantially free of LNP and liposome components.
  • Clause 120 An immunogenic composition or vaccine composition for use according to clause 119, for use therapeutically i.e.
  • aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a protein.
  • Clause 125 The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a cationic lipopeptide, for example the aqueous liquid pharmaceutical formulation does not comprise polymyxin B.
  • Clause 126 The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise an inorganic nanoparticle. 77 Clause 127.
  • aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a lipid nanoparticle (LNP) or a liposome.
  • LNP lipid nanoparticle
  • Clause 128 The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a neutral lipid, for example the aqueous liquid pharmaceutical formulation does not comprise cholesterol or an analogue thereof.
  • Clause 129. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a cationic lipid.
  • aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers, and (iv) a diluent.
  • An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers selected from citrate, acetate, lactate, formate, and phosphate, and (iv) a diluent selected from isotonic saline (0.9% w/v), isotonic dextrose (5% w/v), isotonic mixtures of saline and dextrose (e.g.
  • An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers selected from citrate and phosphate, and (iv) a diluent selected from isotonic saline (0.9% w/v) and sterile or purified water.
  • An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) citrate, and (iv) isotonic saline (0.9% w/v). Clause 135.
  • An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a 78 pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) citrate, and (iv) sterile or purified water. Clause 136.
  • An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) phosphate, and (iv) isotonic saline (0.9% w/v). Clause 137.
  • An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) phosphate, and (iv) sterile or purified water. Clause 138.
  • An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers, (iv) a diluent, (v) optionally, one or more suspending agents, (vi) optionally, one or more wetting or thickening agents, and (vii) optionally one or more osmotic or tonicity adjusting agents. Clause 139.
  • An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers selected from citrate and phosphate, (iv) a diluent selected from isotonic saline (0.9% w/v) and sterile or purified water, (v) optionally, one or more suspending agents, (vi) optionally, one or more wetting or thickening agents, and (vii) optionally one or more osmotic or tonicity adjusting agents.
  • Clause 140 The aqueous liquid pharmaceutical formulation according to any one of clauses 131 to 139 in the form of a stable colloidal emulsion. Examples Abbreviations used herein are defined below (see Table 1). Any abbreviations not defined are intended to convey their generally accepted meaning.
  • Table 1 Abbreviations ALI Air Liquid Interface BSA Bovine Serum Albumin CPE Cytopathic Effects DMEM Dulbecco’s Modified Eagles Medium GFP Green Fluorescent Protein HAE Human Airway Epithelium 79 hr Hour(s) IFN Interferon L litre MDCK Madin-Darby canine kidney cells MFI Mean Fluorescent Intensity min Minute(s) MOI Multiplicity of Infection MSD Meso Scale Discovery PBS Phosphate Buffered Saline PCR Polymerase Chain Reaction PFU Plaque-Forming Units PR8 A/Puerto Rico/8/1934 (H1N1) TPCK tosyl phenylalanyl chloromethyl ketone The structure of molecules used herein are illustrated below (see Table 2).
  • Table 2 Structures of Polynucleotide Molecules d sRNA 1 5’pppGCAUGCGACCUCUGUUUGA-3’ SEQ ID NO.12 3’-CGUACGCUGGAGACAAACU-5’ dsRNA 2 polyinosinic-polycytidylic acid dsRNA between 200-8000 base Poly I:C pairs in length (CAS: 31852-29-6) shRNA 1 SEQ ID NO.1 shRNA 2 SEQ ID NO.2 RNA 5’-ppGGAUCGAUCGAUCGUU-L1- SEQ ID NO 13 SEQ ID NO 14 M A 80 F F n c T d, f Stable Polysorbate 80 Active Ingredient Colloidal 0.045% 45 ⁇ g
  • the formulation may be administered intranasally via a nasal spray device, for example with a spray volume 100 ⁇ L suitable for 1 or 2 sprays per nostril.
  • the formulation may be suitable 81 for the treatment or prevention of infection by a virus or disease associated with infection with such a virus.
  • Formulation Example 1B Example aqueous liquid pharmaceutical formulation comprising dsRNA 1
  • the following aqueous liquid pharmaceutical formulation at about pH 4.7 may be prepared, Purified Water Diluent q.s. 85.92 mg
  • the formulation may be administered intranasally via a nasal spray device, for example with a spray volume 100 ⁇ L suitable for 1 or 2 sprays per nostril.
  • the formulation may be suitable 82 for the treatment or prevention of infection by a virus or disease associated with infection with such a virus.
  • Example aqueous liquid pharmaceutical formulation comprising dsRNA 2
  • the following aqueous liquid pharmaceutical formulation at about pH 4.7 may be prepared, for example largely according to the protocol described in Biophysical Example 1:
  • the formulation may be administered intranasally via a nasal spray device, for example with a spray volume 100 ⁇ L suitable for 1 or 2 sprays per nostril.
  • the formulation may be suitable 83 for the treatment or prevention of infection by a virus or disease associated with infection with such a virus.
  • Example aqueous liquid pharmaceutical formulation comprising shRNA 1
  • the following aqueous liquid pharmaceutical formulation at about pH 7.2 may be prepared, for example largely according to the protocol described in Biophysical Example 1: Stable shRNA 1 Active Ingredient Colloidal 2.00% 2000 ⁇ g Particle 84 Stable Oleic Acid Active Ingredient Colloidal 0.050% 50 ⁇ g Particle Stable Polysorbate 80 Active Ingredient Colloidal 0.045% 45 ⁇ g Particle Stable Colloidal shRNA 1 Active Ingredient 2.00% Particle Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle 85 Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle Citric Acid, Monohydrate Buffer Solution 0.20% (Promega, E1081) 86 Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle Citric Acid Monoh drate Buffer Solution 020% Stable Colloidal Oleic
  • a e oo a Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle Stable Colloidal RNA Conjugate 2 Active Ingredient 2 mg/mL Particle 90 Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal differentiated at an air-liquid interface (ALI), forming a pseudostratified mucociliary airway epithelium that is composed of ciliated cells, goblet cells, club cells, and/or basal cells with an arrangement closely reflective of an in vivo cellular organization.
  • ALI air-liquid interface
  • HAE-ALI human airway epithelium
  • shRNA 1 with surfactant component 2 and surfactant component 3 induced significantly higher levels of CXCL10 expression compared to shRNA 1 in water.
  • surfactant component 3 (with shRNA 1) which comprises the highest surfactant concentration, only marginally increased induction of CXCL10 relative to surfactant component 2 (with shRNA 1).
  • Biological Example 2 Assessment of CXCL10 production and virus load, upon treatment with shRNA 1, in influenza virus infected ALI cultured human epithelial cells
  • ALI cultured pooled donors human nasal epithelium (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase with MucilAirTM culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions.
  • influenza virus inoculum strain PR8; 100 ⁇ L; diluted in MucilAirTM culture medium to give a final MOI of 0.1
  • Virus inoculum was subsequently removed, and inserts were washed with sterile PBS (with Ca 2+ /Mg 2+ ).
  • the treatment formulations described below were produced largely as described in Biophysical Example 1.
  • ALI cultures were dosed apically with 50 ⁇ L vehicle control (water alone), shRNA 1 in vehicle (2 mg/mL) or shRNA 1 (2 mg/mL) with surfactant component 1 (0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80), or surfactant component 2 (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) each in vehicle, for 30 min (then being removed with a pipette) one day prior to virus inoculation, before subsequent re-application of the treatments 30 min prior to virus inoculation (then being removed with a pipette) and subsequent incubation with the virus inoculum for 60 min.
  • oseltamivir carboxylate (10 ⁇ M) was added to the basolateral chambers of sample wells on the day prior to virus inoculation, and on Day 0.
  • sampling was conducted by adding 300 ⁇ L of culture medium to the 92 apical surface of each for well for 10 min (being stored at -80 o C).
  • virus load was quantified using the 50% Tissue Culture Infectious Dose (TCID 50 ).
  • shRNA 1 with surfactant component 1 which comprises the lowest surfactant concentration
  • shRNA 1 with surfactant component 2 which comprises a comparatively higher surfactant concentration
  • oseltamivir control 10 ⁇ M failed to induce CXCL10 expression as it is a direct anti-viral agent and does not stimulate the innate immune system.
  • high levels of influenza replication were detected in an apical wash following treatment with the vehicle control (mean: 3.5 Log, TCID 50 /mL) at 48 hrs post- inoculation.
  • shRNA 1 Upon comparison of the detected viral load of each treatment with that following treatment with vehicle control (water), shRNA 1 alone failed to demonstrate a significant anti- viral effect, and similarly shRNA 1 with surfactant component 1, which comprises the lowest surfactant concentration, failed to significantly reduce viral load. However, shRNA with surfactant component 2 had a significant anti-viral effect, as demonstrated by a 0.7 Log reduction in viral load.
  • the assay control, oseltamivir (10 ⁇ M) had a similar anti-viral effect (0.7 Log reduction) as predicted (Boda et al.2018).
  • ALI cultures were dosed apically with 50 ⁇ L vehicle control (water alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, shRNA 1 in vehicle or shRNA 1 with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, for 30 min (then being removed with a pipette) one day prior to virus inoculation, before subsequent re-application of the treatments 30 min prior to virus inoculation (then being removed with a pipette) and subsequent incubation with the virus inoculum for 60 min.
  • vehicle control water alone
  • surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • shRNA 1 in vehicle or shRNA 1 with surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in
  • oseltamivir carboxylate (10 ⁇ M) was added to the basolateral chambers of sample wells on the day prior to virus inoculation, and on Day 0.
  • sampling was conducted by adding 300 ⁇ L of culture medium to the apical surface of each for well for 10 min (being stored at -80 o C).
  • Treatment formulations used in this Example were produced largely as described in Biophysical Example 1.
  • shRNA 1 with surfactant component had a very potent anti-viral effect, as represented by a 3.2 Log reduction of viral load relative to water alone (vehicle control).
  • shRNA 1 with surfactant component elicited a more significant reduction in viral load than the assay control, oseltamivir (10 ⁇ M), which led to a 2.2 Log reduction in viral load versus the vehicle control.
  • oseltamivir 10 ⁇ M
  • ALI cultures were dosed apically with 50 ⁇ L of a vehicle control (buffer: 0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate in water), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, dsRNA 1 (10 ⁇ g/mL) or dsRNA 2 (100 ⁇ g/mL) in vehicle or dsRNA 1 (10 ⁇ g/mL) or dsRNA 2 (100 ⁇ g/mL) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle for 30 min (then being removed with a pipette) one day prior to virus inoculation, before subsequent re-application of the treatments for 30 min prior to virus inoculation (then being removed with a pipette) and subsequent incubation with the virus inoculum for 60 min
  • dsRNA 1 failed to induce CXCL10 expression relative to vehicle control.
  • dsRNA 1 with surfactant component induced strong CXCL10 expression.
  • dsRNA 2 alone induced significant CXCL10 expression, however formulation of dsRNA 2 with surfactant component elicited a further increase in the observed induction of CXCL10 expression.
  • high levels of influenza replication were detected in an apical wash following treatment with the vehicle control (buffer).
  • dsRNA 1 alone failed to demonstrate a significant anti-viral effect.
  • surfactant component alone appeared to elicit a mild anti-viral effect, as represented by a 0.7 Log reduction in viral load relative to vehicle control.
  • dsRNA 1 with surfactant component had potent anti-viral activity, as represented by a significant reduction in viral load (1.8 Log reduction versus dsRNA 1.
  • dsRNA 2 alone demonstrated significant anti-viral activity relative to vehicle control.
  • formulation of dsRNA 2 with the surfactant component enhanced the anti-viral activity of dsRNA 2, as 95 represented by a 2.8 Log reduction in viral load versus surfactant component alone and a 1.5 Log reduction in viral load versus dsRNA 2 alone.
  • Treatment formulations of vehicle control (water), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) and further pharmaceutically acceptable excipients in vehicle (see Formulation Example 1A minus the shRNA 1, hereinafter “surfactant formulation”), or shRNA 1 (2 mg/mL) in vehicle, or with surfactant component and further pharmaceutically acceptable excipients in vehicle (see Formulation Example 1A, hereinafter “shRNA 1 with surfactant formulation”) were produced largely as described in Biophysical Example 1 below.
  • the vehicle control or test formulations were administered intranasally (10 ⁇ L each nostril) with a pipette 3 days, 1 day and 1 hr prior to virus infection on Day 0.
  • oseltamivir phosphate was formulated in PBS and administered orally to control mice (10 mg/kg) 1 hr prior to virus infection and once daily on Day 1 post-inoculation. On Day 1 and Day 5 post-infection, the mice were euthanised (intraperitoneal injection of pentobarbitone overdose). Subsequently, the trachea was cannulated and the airway lavaged by flushing out the lungs using 0.5 mL PBS. Immediately after bronchoalveolar lavage, a catheter was inserted into the posterior naris/nares from the opening of the trachea that was used for bronchoalveolar lavage to collect nasal lavage fluid (NLF).
  • NVF nasal lavage fluid
  • mice Immediately after NLF collection, the thoracic cavity of euthanised mice was opened and the right lung lobes from each mouse were removed and homogenised in DMEM (with 1% BSA and 25% sucrose; 10 mL/g lung) for two 20 second periods. The homogenate was transferred into a sterile tube and spun at 4 o C at 2000 rpm for 5 min. The clarified homogenate was then 96 transferred to a chilled cryovial and snap frozen in liquid nitrogen and stored at -80 o C. Following removal of the right lung lobe, the nasal tissue was removed from each animal. The dissection involved firstly removing the mandible and skin over the head, followed by removal of the head.
  • DMEM with 1% BSA and 25% sucrose; 10 mL/g lung
  • the palette and bone protecting the brain and olfactory bulbs was removed to expose the nasal tissue.
  • the extracted nasal tissue was homogenised in DMEM (with 1% BSA and 25% sucrose; 10mL/g tissue) for two 20 second periods.
  • the nasal tissue homogenate was transferred into a sterile tube and spun at 4 o C at 2000 rpm for 5 min before transfer into a cryovial, snap freezing in liquid nitrogen and storage at -80 o C.
  • shRNA 1 alone demonstrated a moderate anti-viral effect, as represented by a 41% reduction in viral load on both Day 1 and Day 5 post-inoculation (versus water alone)
  • formulation of shRNA 1 with the surfactant formulation resulted in a more significant anti-viral effect, as represented by a 60% and 51% reduction in viral load on Day 1 and Day 5 post-inoculation respectively (versus surfactant formulation alone).
  • shRNA 1 with surfactant formulation therefore had a significantly more potent anti-viral effect than the oseltamivir control, which elicited a 38% and 33% reduction in viral load on Day 1 and Day 5 post-inoculation respectively (versus vehicle).
  • the oseltamivir control also resulted in a reduction in neutrophil accumulation, as represented by a 57% and 69% reduction in the neutrophil number on Day 1 and Day 5 post-inoculation respectively, albeit to a lesser extent than shRNA 1 with the surfactant formulation.
  • influenza virus infection led to substantial body weight loss in influenza virus infected mice treated with vehicle.
  • the surfactant formulation alone failed to prevent this influenza virus-induced weight loss.
  • shRNA 1 in vehicle had a moderate protective effect against body weight loss, whilst shRNA 1 with surfactant formulation further prevented influenza virus-induced weight loss.
  • shRNA 1 with surfactant formulation had a similar protective effect to the oseltamivir control.
  • Biological Example 6 Assessment of the exposure of mRNA and plasmid DNA to ALI cultured human nasal epithelium Experimental Methods ALI cultured pooled donors’ human nasal epithelium (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase with MucilAirTM culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions.
  • ALI cultures were dosed apically with 50 ⁇ L of vehicle control, that is 0.5 mg/mL GFP-encoding mRNA (Vetnal) in citrate buffer (0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate), or test formulation, that is 0.5 mg/mL GFP-encoding mRNA (Vetnal) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs. The treatment was then removed, and the apical surface of the ALI cultures were washed with 100 ⁇ L culture.
  • vehicle control that is 0.5 mg/mL GFP-encoding mRNA (Vetnal) in citrate buffer (0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate
  • test formulation that is 0.5 mg/mL GFP-encoding mRNA (V
  • ALI cultured nasal epithelial cells were collected by gentle agitation in sterile PBS prior to fixation with 4% paraformaldehyde in PBS. Samples from two independent inserts were combined and analysed by flow cytometry using a 530/30-nm bandpass (BP) filter on the BD AccuriTM apparatus (Becton Dickinson). The MFI was calculated, and the MFI of non- treated control (autofluorescence) subtracted.
  • BP bandpass
  • ALI cultures were dosed apically with 50 ⁇ L of vehicle control, that is 0.033 ⁇ g/mL pSV- ⁇ -Galactosidase Control Vector (Promega, E1081) ( ⁇ -gal) in citrate buffer (0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate), or test formulation, that is 0.033 ⁇ g/mL pSV- ⁇ -Galactosidase Control Vector (Promega, E1081) ( ⁇ -gal) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs.
  • vehicle control that is 0.033 ⁇ g/mL pSV- ⁇ -Galactosidase Control Vector (Promega, E1081) ( ⁇ -gal) in citrate buffer (0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate
  • ALI cultured nasal epithelial cells from three inserts were lysed together in a reporter lysis buffer according to the manufacturer’s instructions (Promega, #E2000, ⁇ -galactosidase enzyme assay system with Reporter Lysis Buffer). Following 98 overnight incubation with the substrate provided in the kit ⁇ -galactosidase enzyme activity (24 hrs post-treatment) was determined by reading absorbance at 420 nm. Treatment formulations used in this Example were produced largely as described in Biophysical Example 1.
  • formulation of a ⁇ -galactosidase encoding plasmid with surfactant component may similarly increase ALI cultured nasal epithelial cell exposure to said plasmid.
  • human rhinovirus type 16 (HRV16) virus inoculum 100 ⁇ L of 400,000 PFU/mL in MucilAirTM culture medium to give a final MOI of approximately 0.2
  • HRV16 human rhinovirus type 16 virus inoculum
  • 100 ⁇ L of 400,000 PFU/mL in MucilAirTM culture medium 100 ⁇ L was added to the apical surface of the epithelium for 1 hr (34°C/5% CO 2 ).
  • Virus inoculum was subsequently removed, and inserts were washed with 100 ⁇ L of sterile PBS (with Ca 2+ /Mg 2+ ).
  • ALI cultures were dosed apically with 50 ⁇ L vehicle control (citrate buffer: 0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate), shRNA 1 in vehicle (2 mg/mL), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or shRNA 1 (2 mg/mL) with surfactant component for 30 min (then being removed with a pipette almost completely without additional wash) one day prior to virus inoculation, before subsequent re- application of the treatments 30 min prior to virus inoculation (then being removed as described above) and subsequent incubation with virus inoculum for 60 min.
  • vehicle control citrate buffer: 0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate
  • shRNA 1 in vehicle 2 mg/mL
  • surfactant component 0.05% (w/w) oleic acid and 0.045% (
  • human respiratory syncytial virus A2 (RSV A2) virus inoculum (100 ⁇ L of 40,000 PFU/mL in MucilAirTM culture medium to give a final MOI of approximately 0.02) was added to the apical surface of the epithelium for 1 hr (37°C/5% CO 2 ). Virus inoculum was subsequently removed, and inserts were washed with 100 ⁇ L of sterile PBS (with Ca 2+ /Mg 2+ ).
  • ALI cultures were dosed apically with 50 ⁇ L vehicle control (citrate buffer: 0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate), shRNA 1 in vehicle (2 mg/mL), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or shRNA 1 (2 mg/mL) with surfactant component for 30 min (then being removed with a pipette almost completely without additional wash) one day prior to virus inoculation, before subsequent re- application of the treatments 30 min prior to virus inoculation (then being removed as described above) and subsequent incubation with virus inoculum for 60 min.
  • vehicle control citrate buffer: 0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate
  • shRNA 1 in vehicle 2 mg/mL
  • surfactant component 0.05% (w/w) oleic acid and 0.045% (
  • HEp-2 cells were seeded into 24-well plates (Corning, NY, USA) at a density of between 5-10 ⁇ 10 4 cells/well and grown, for 48 hrs prior to infection, in 10% FBS DMEM until they attained 100% confluency.
  • virus infected samples were thawed at room temperature and ten-fold serial dilutions were prepared in serum-free DMEM.
  • the growth medium from HEp-2 cells was aspirated and replaced with 300 ⁇ L of the serially diluted virus collections and left to infect at 37°C/5% CO 2 for 4 hrs.
  • Plaque Assay Overlay (0.3% Avicel RC-591 [FMC Biopolymer UK, Girvan, Scotland]) in MEM, supplemented to a final concentration of 2% FBS), and left for 7 days at 37°C/5% CO 2 .
  • Cells were then fixed with ice-cold methanol for 10 min, prior to methanol removal and subsequent washing of fixed cells (x 2) with sterile PBS.
  • transepithelial electrical resistance was measured to investigate the integrity of tight junction dynamics in ALI cultured pseudostratified epithelium pre- and 3 days post-RSV A2 infection.
  • This measurement of TEER is a surrogate measure for epithelial damage. Specifically, chopstick-electrodes were placed in the apical and basolateral chambers and the TEER was measured using a dedicated 101 Volt/Ohm meter (EVOM2, Epithelial Volt/Ohm Meter for TEER). The measure of TEER is expressed as Ohm/cm 2 .
  • Biological Example 9 Assessment of viral load, virus-induced inflammation and body weight loss, upon treatment with shRNA 1, in a respiratory syncytial virus (RSV) infected mouse model
  • RSV respiratory syncytial virus
  • Non-fasted mice male BALB/C, 20-30 g
  • RSV respiratory syncytial virus
  • the RSV virus (RSV A2 strain; 25 ⁇ L per nostril of 2.5 x10 6 PFU) was administered into each nostril in a drop-wise manner (25 ⁇ L each nostril).
  • Treatment formulations of saline, surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer and shRNA 1 (2 mg/mL) with surfactant component and further pharmaceutically acceptable excipients in vehicle were produced largely as described in Biophysical Examples 1 and 2 below.
  • the vehicle control or test formulations were administered intranasally (25 ⁇ L each nostril) with a pipette both 4 days and 1 day prior to virus infection on Day 0.
  • ribavirin was formulated in PBS and administered intranasally to control mice (12.5 mg/kg) 3 hr prior to virus infection, and twice daily on Days 0 to 4 post-inoculation. On Day 4 post-infection, the mice were euthanised (intraperitoneal injection of pentobarbitone overdose). Subsequently, the trachea was cannulated and the airway lavaged by flushing out 102 the lungs using 0.5 mL PBS. Immediately after bronchoalveolar lavage, a catheter was inserted into the posterior naris/nares from the opening of the trachea that was used for bronchoalveolar lavage to collect nasal lavage fluid (NLF).
  • NVF nasal lavage fluid
  • mice Immediately after NLF collection, the thoracic cavity of euthanised mice was opened and the right lung lobes from each mouse were removed and homogenised in DMEM (with 1% BSA and 25% sucrose; 10 mL/g lung) for two 20 second periods. The homogenate was transferred into a sterile tube and spun at 4 o C at 2000 rpm for 5 min. The clarified homogenate was then transferred to a chilled cryovial and snap frozen in liquid nitrogen and stored at -80 o C. Following removal of the right lung lobe, the nasal tissue was removed from each animal. The dissection involved firstly removing the mandible and skin over the head, followed by removal of the head.
  • DMEM with 1% BSA and 25% sucrose; 10 mL/g lung
  • the extracted nasal tissue was homogenised in DMEM (with 1% BSA and 25% sucrose; 10 mL/g tissue) for two 20 second periods.
  • DMEM with 1% BSA and 25% sucrose; 10 mL/g tissue
  • the nasal tissue homogenate was transferred into a sterile tube and spun at 4 o C at 2000 rpm for 5 min before transfer into a cryovial, snap freezing in liquid nitrogen and storage at -80 o C.
  • plaque assay was conducted.
  • HEp2 cells were grown in 24-well plates prior to infection in DMEM containing 10% (v/v) FBS until they achieved 100% confluency.
  • the supernatant collected from the lung and nasal tissue homogenates were thawed out at room temperature and serial dilutions was prepared in serum-free DMEM.
  • the growth medium from HEp2 cells were aspirated and replaced with 300 ⁇ L of serially diluted lung homogenate (along with stock RSV only positive control) and left to infect at 37°C/5% CO 2 for four hrs.
  • the infectious media was then aspirated and replaced with 500 ⁇ L Plaque Assay Overlay (1% (w/v) methylcellulose in MEM, 2% (v/v) FBS, 1% (w/v) pen/strep, 0.5 ⁇ g/ml amphotericin B), and left for 7 days at 37°C/5% CO 2 .
  • Anti-RSV F-protein antibody [2F7] was diluted to a 1:150 concentration in blocking buffer (5% (w/v) powdered milk (Marvel) in 0.05% (v/v) PBS-Tween 20) and 150 ⁇ L was added to cells for 2 hrs at room temperature with shaking. Cells were washed twice using PBS before 150 ⁇ L of secondary antibody (goat anti-mouse/HRP conjugate) diluted 1:400 in 103 blocking buffer were added to cells for 1 hr at room temperature, with shaking.
  • blocking buffer 5% (w/v) powdered milk (Marvel) in 0.05% (v/v) PBS-Tween 20
  • mice with the surfactant component formulation only failed to demonstrate any significant reduction in neutrophil count.
  • treatment of mice with the shRNA 1 with surfactant component formulation resulted in a significant reduction in the degree of neutrophil accumulation, as represented by a 57% reduction in the neutrophil number on Day 4 post-inoculation relative to treatment with vehicle control (i.e. saline), and by a 55% reduction in the neutrophil number on Day 4 post-inoculation relative to treatment with surfactant component alone.
  • treatment of mice with the shRNA 1 with surfactant component formulation resulted in a reduction in neutrophil accumulation to that observed following treatment with ribavirin.
  • ribavirin treatment resulted in a 61% reduction in the neutrophil number on Day 4 post-inoculation relative to treatment with vehicle control (i.e. saline).
  • vehicle control i.e. saline
  • RSV A2 infection led to substantial body weight loss in mice treated with vehicle control (i.e. saline).
  • treatment of mice with the surfactant component formulation failed to limit or prevent the RSV A2 infection-induced body weight loss.
  • treatment of mice with both the shRNA 1 with surfactant component formulation and with the ribavirin control provided a protective effect against said RSV A2 infection-induced body weight loss. The extent of this protective effect was comparable between the shRNA 1 with surfactant component formulation and the ribavirin control.
  • Non-fasted mice male BALB/C, 20-30 g were injected subcutaneously with saline, surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer, shRNA1 (2 mg/mL) with surfactant component, or shRNA1 (20 mg/mL) with surfactant component at a dose volume of 10 mL/kg.
  • the airway was then lavaged by flushing out the lungs using 0.5 mL PBS. This procedure was repeated until the recovered volume totalled 1.6 mL.
  • the isolated BALF was then centrifuged at 1500 rpm for 10 mins at 4 o C and the supernatant was aliquoted (400 ⁇ L) at -80 o C.
  • the levels of CXCL10 in serum and BALF was determined using mouse CXCL10 kit using MSD multi scanner (Meso Scale Diagnosis). Separately, non-fasted mice (male BALB/C, 20-30 g) were infected intranasally with influenza (strain PR8) under isoflurane (5% in O 2 ) anaesthesia.
  • influenza virus PR8 strain; 10 ⁇ L per nostril of 2 x10 2 PFU
  • the influenza virus was administered into each nostril in a drop-wise manner (10 ⁇ L each nostril).
  • oseltamivir phosphate was formulated in PBS and administered orally 4 hr prior to virus infection to control mice (10 mg/kg), wherein said mice were also treated with subcutaneous injection of surfactant component alone 4 days and 1 day prior to virus infection on Day 0.
  • Futhermore one group of mice was treated both with oral oseltamivir, and with the subcutaneously administered shRNA 1 with surfactant component formulation, as a combination therapy.
  • Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2. 105 On Day 5 post-infection, the mice were euthanised (intraperitoneal injection of pentobarbitone overdose).
  • the trachea was cannulated and the airway lavaged by flushing out the lungs using 0.5 mL PBS.
  • a catheter was inserted into the posterior naris/nares from the opening of the trachea that was used for bronchoalveolar lavage to collect nasal lavage fluid (NLF).
  • PBS (1 mL) was gently perfused into the nasal cavities, and the NLF was collected from the anterior naris/nares.
  • the isolated NLF was centrifuged at 1500 rpm for 10 min at 4 o C and the supernatant was aliquoted (350 ⁇ L) at -80 o C for future cytokine analysis.
  • the cell pellets were re-suspended in 1.6 mL PBS and the NLF cells were analysed for total and differential numbers.
  • DMEM DMEM with 1% BSA and 25% sucrose; 10 mL/g lung
  • the homogenate was transferred into a sterile tube and spun at 4 o C at 2000 rpm for 5 min. The clarified homogenate was then transferred to a chilled cryovial and snap frozen in liquid nitrogen and stored at -80 o C. Following removal of the right lung lobe, the nasal tissue was removed from each animal. The dissection involved firstly removing the mandible and skin over the head, followed by removal of the head. Next, the palette and bone protecting the brain and olfactory bulbs was removed to expose the nasal tissue. The extracted nasal tissue was homogenised in DMEM (with 1% BSA and 25% sucrose; 10mL/g tissue) for two 20 second periods.
  • DMEM with 1% BSA and 25% sucrose; 10mL/g tissue
  • the nasal tissue homogenate was transferred into a sterile tube and spun at 4 o C at 2000 rpm for 5 min before transfer into a cryovial, snap freezing in liquid nitrogen and storage at -80 o C.
  • mice with the surfactant component formulation demonstrated no clear anti-viral effect in either the lung or nasal tissue.
  • treatment of mice with the shRNA 1 with surfactant component formulation had a clear and strong anti-viral effect, as represented by a 29% reduction in viral load in the lung, and by a 45% reduction in viral load in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only.
  • treatment of mice with the control drug oseltamivir also resulted in a reduction in viral load, as represented by a 41% reduction in viral load in the lung, and by a 49% reduction in viral load in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only.
  • the anti-viral effect of the control drug oseltamivir is comparable to that of the shRNA 1 with surfactant component formulation.
  • the combination treatment of oral oseltamivir and subcutaneous shRNA 1 with surfactant component showed an even more significant anti-viral effect, as represented by a 73% reduction in viral load in the lung, and by a 67% reduction in viral load in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only.
  • significant neutrophil accumulation was observed in both the lung (see Figure 24) and nasal tissue (see Figure 25) of influenza virus-infected mice treated with saline only (i.e. non-treatment).
  • mice with the control drug oseltamivir also resulted in a reduction in neutrophil accumulation in both the lung and nasal tissue, as represented by a 60% reduction in neutrophil number in the lung, and by a 70% reduction in the neutrophil number in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only.
  • the combination treatment of oral oseltamivir and subcutaneous shRNA 1 with surfactant component elicited a yet more significant reduction in neutrophil accumulation, as represented by an 80% reduction in 107 neutrophil number in the lung, and by a 74% reduction in the neutrophil number in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only.
  • influenza virus infection led to substantial body weight loss in mice treated with vehicle control (i.e. saline).
  • vehicle control i.e. saline
  • Subcutaneous treatment of mice with the surfactant component formulation failed to limit or prevent the influenza virus infection-induced body weight loss.
  • subcutaneous treatment of mice with both the shRNA 1 with surfactant component formulation and with the oral oseltamivir control provided a protective effect against said influenza virus infection-induced body weight loss.
  • the combination treatment of oral oseltamivir and subcutaneous shRNA 1 with surfactant component entirely prevented any influenza virus infection-induced body weight loss.
  • Biological Example 11 Assessment of the adjuvant effect of shRNA 1 with surfactant component on recombinant H1N1 haemagglutinin (HA) vaccinated and influenza virus infected mice Experimental Methods Non-fasted mice (male BALB/C, 20-30 g) were treated with PBS, shRNA 1 with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer or the control adjuvant and TLR9 agonist, CPG-ODN solution (InvivoGen), intranasally (10 ⁇ L/nostril). Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2.
  • mice were treated, on Day 0, with either vehicle (i.e. PBS) or recombinant haemagglutinin (HA) (1mg/mL in PBS) from the influenza virus PR8 strain (SinoBiologics) under isoflurane to deliver a volume of 10 ⁇ L per nostril. Said treatments were introduced into each nostril in a drop-wise fashion, alternating between the two, until the required volume had been delivered (10 ⁇ g HA/10 ⁇ L). A qualitative assessment for each group was recorded for the level of piloerection generated in each animal following priming dose. Booster doses (adjuvant and HA antigen) in all animals were re-administered on Day 28.
  • vehicle i.e. PBS
  • HA haemagglutinin
  • mice On Day 56 each animal was dosed intranasally (10 ⁇ L) with influenza virus (PR8 strain; 2 x 10 2 PFU). Following infection each animal was weighed on a daily basis to monitor changes in body weight. On Day 61 (i.e.5 days post-infection), terminal blood samples were taken by venipuncture (via the lateral tail vein) and placed into LiHep tubes. Each blood sample was mixed gently and centrifuged (2000 g for 5 min at 4 o C) from which the resulting plasma was extracted, aliquoted and stored at -80 o C. Immediately after the bleeds, mice were euthanised (intraperitoneal injection of pentobarbitone overdose).
  • influenza virus PR8 strain; 2 x 10 2 PFU
  • the trachea was cannulated and the airway lavaged by flushing out the lungs using 0.5 mL PBS.
  • a catheter was inserted into the posterior naris/nares from the opening of the trachea that was used for bronchoalveolar lavage to collect nasal lavage fluid (NLF).
  • PBS (1 mL) was gently 108 perfused into the nasal cavities, and the NLF was collected from the anterior naris/nares.
  • the isolated NLF was centrifuged at 1500 rpm for 10 min at 4 o C and the supernatant was aliquoted (350 ⁇ L) at -80 o C for future cytokine analysis.
  • the cell pellets were re-suspended in 1.6 mL PBS and the NLF cells were analysed for total and differential numbers.
  • DMEM DMEM with 1% BSA and 25% sucrose; 10 mL/g lung
  • the homogenate was transferred into a sterile tube and spun at 4 o C and 2000 rpm for 5 min. The clarified homogenate was then transferred to a chilled cryovial and snap frozen in liquid nitrogen and stored at -80 o C. Following removal of the right lung lobe, the nasal tissue was removed from each animal. The dissection involved firstly removing the mandible and skin over the head, followed by removal of the head. Next, the palette and bone protecting the brain and olfactory bulbs was removed to expose the nasal tissue. The extracted nasal tissue was homogenised in DMEM (with 1% BSA and 25% sucrose; 10mL/g tissue) for two 20 second periods.
  • DMEM with 1% BSA and 25% sucrose; 10mL/g tissue
  • the nasal tissue homogenate was transferred into a sterile tube and spun at 4 o C and 2000 rpm for 5 min before transfer into a cryovial, snap freezing in liquid nitrogen and storage at -80 o C.
  • mice treated with the rH1N1 HA vaccination and the control adjuvant, CPG-ODN similarly experienced a moderate reduction in viral load in both the lung and nasal tissue.
  • mice treated with the rH1N1 HA vaccination and the shRNA 1 with surfactant component formulation demonstrated a considerably greater, and highly statistically significant reduction in viral load, in both the lung and nasal tissue.
  • the anti-viral effect of the rH1N1 HA vaccination 109 following treatment with the shRNA 1 with surfactant component formulation was therefore considerably greater than that observed following treatment of said vaccination with the control adjuvant, CPG-ODN.
  • mice vaccinated with rH1N1 HA alone demonstrated a marked reduction in neutrophil accumulation in both the lung and nasal tissue and, notably, mice vaccinated with rH1N1 HA post-treatment with the control adjuvant, CPG- ODN, showed a similar degree of reduction in neutrophil accumulation. That is, the control adjuvant, CPG-ODN, failed to enhance the reduction in neutrophil accumulation induced by rH1N1 HA vaccination.
  • mice treated with rH1N1 HA vaccination post- treatment with the shRNA 1 with surfactant component formulation demonstrated a further significant reduction in neutrophil accumulation in both the lung and nasal tissue, as represented by a 40% reduction in neutrophil accumulation in the lung, and by a 83% reduction in neutrophil accumulation in the nasal tissue, relative to treatment with vehicle (i.e. PBS) on Day 5 post-inoculation. That is, the shRNA 1 with surfactant component formulation enhanced the rH1N1 HA vaccination associated reduction in neutrophil accumulation.
  • mice with the rH1N1 HA vaccination post-treatment with the shRNA 1 with surfactant component formulation resulted in a more significant protective effect against the influenza virus infection-induced body weight loss, and in fact almost completely mitigated any such body weight loss.
  • Biological Example 12 Assessment of the exposure of mRNA to ALI cultured human nasal epithelium Experimental Methods Following the encouraging results of Biological Example 6 above, the inventors have better characterised the data presented in relation to exposure of GFP-encoding mRNA, which supports the use of the formulations of the present invention in improving the delivery, and thereby increasing exposure, of mRNA molecules.
  • ALI nasal epithelium cultures (Epithelix Sarl) were dosed apically with 50 ⁇ L of vehicle control, that is 0.1 mg/mL H1N1 haemagglutinin (HA)-encoding mRNA (Oz Bioscience) in phosphate buffer, or 0.1 110 mg/mL H1N1 HA-encoding mRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs. The treatment was then removed, and the apical surface of the ALI cultures was washed with 100 ⁇ L culture.
  • vehicle control that is 0.1 mg/mL H1N1 haemagglutinin (HA)-encoding mRNA (Oz Bioscience) in phosphate buffer, or 0.1 110 mg/mL H1N1 HA-encoding mRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/
  • H1N1 HA protein was detected by the standard SDS-PAGE/Western blotting system using anti-H1N1 HA antibody (#ab281949, Abcam). Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2.
  • ALI bronchial epithelium cultures (Epithelix Sarl) were dosed apically with 50 ⁇ L of vehicle control, that is cystic fibrosis transmembrane conductance regulator (CFTR)-encoded mRNA (northern RNA) in phosphate buffer, or 0.1 mg/mL CFTR-encoded mRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs.
  • CFTR cystic fibrosis transmembrane conductance regulator
  • surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • ALI cultured bronchial epithelial cells were collected and suspended in lysis buffer.
  • the expression of CFTR protein was detected by the standard SDS- PAGE/Western blotting system using anti-CFTR antibody (#sc376683, Santa Cruz Biotechnology). Results
  • treatment of ALI cultured nasal epithelial cells with GFP-mRNA in citrate buffer (i.e. vehicle) failed to result in any detectable fluorescent signal relative to background fluorescence. This is indicative of the failure of citrate buffer alone to expose ALI cultured nasal epithelial cells to GFP-encoding mRNA.
  • ALI cultured nasal epithelial cells treated with GFP-encoding mRNA with surfactant component demonstrated a strong and significant fluorescent signal (MFI), indicative of the ability of the surfactant component to increase exposure of GFP-encoding mRNA to cells.
  • MFI fluorescent signal
  • only cells treated with H1N1 haemagglutinin (HA) mRNA with surfactant component expressed the H1N1 HA proteins 48 hrs post- treatment. That is, cells treated with H1N1 HA mRNA in PBS failed to be exposed to, and therefore failed to express the H1N1 HA protein.
  • ALI cultures were dosed apically with 50 ⁇ L vehicle (citrate or phosphate buffer only), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in citrate or phosphate buffer, or shRNA 1 (2 mg/mL) in with surfactant component in citrate or phosphate buffer, for 30 min (then being removed with a pipette) four days and one day prior to virus inoculation, (influenza virus, strain PR8; 100 ⁇ L; diluted in MucilAirTM culture medium to give a final MOI of 0.2) for 60 min on Day 0.
  • vehicle citrate or phosphate buffer only
  • surfactant component 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80
  • shRNA 1 (2 mg/mL) in with surfactant component in citrate or phosphate buffer
  • vehicle alone i.e. citrate or phosphate buffer alone
  • Biological Example 14 Assessment of the exposure of siRNA and mRNA to ALI cultured human corneal epithelium Experimental Methods ALI cultured human corneal epithelium cells (EpiCorneal COR-100, provided by Mattek Corp. (Ashland, MA) were maintained in air-liquid interphase with COR-100-MM maintenance medium, in the sterile 12-well hanging top-plates (HNG-TOP-12), according to the manufacturer’s instructions.
  • HNG-TOP-12 sterile 12-well hanging top-plates
  • ALI cultures were dosed apically with 50 ⁇ L control formulation, i.e.10nM Silencer TM Cy TM 3- labelled negative control No.1 siRNA (Cy3 labelled siRNA, #AM4621, ThermoFisher Scientific, UK) in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate), or test formulation, i.e.10nM Cy3-labelled siRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs at 37 ⁇ C, 5% CO 2 .
  • control formulation i.e.10nM Silencer TM Cy TM 3- labelled negative control No.1 siRNA (Cy3 labelled siRNA, #AM4621, ThermoFisher Scientific, UK) in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and
  • ALI cultured corneal epithelial cells were collected via trypsinization (0.25% in PBS for 15 min) prior to resuspension in PBS (plus 1% bovine serum albumin (BSA) and 2% paraformaldehyde) and immediate analysis by flow cytometry using a 532 nm bandpass (BP) filter on the BD Canto II apparatus (Becton Dickinson). The count was calculated by subtracting the count of non-treated control (autofluorescence) from the treated cells.
  • BSA bovine serum albumin
  • BP 532 nm bandpass
  • ALI cultures were dosed apically with 50 ⁇ L control formulation, i.e. 0.1 mg/mL GFP-encoding mRNA (Vernal) in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate), or test formulation, i.e.0.1 mg/mL GFP-encoding mRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs. The treatment was then removed, and incubated for 72 hrs at 37 o C, 5% CO 2 .
  • control formulation i.e. 0.1 mg/mL GFP-encoding mRNA (Vernal) in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate)
  • test formulation i.e.0.1 mg/mL GFP-en
  • ALI cultured corneal epithelial cells were collected via trypsinization and analysed by flow cytometry using a 488 nm bandpass (BP) filter on the BD Canto IITM apparatus (Becton Dickinson). The cell count was determined, and the cell count of non-treated control (autofluorescence) was subtracted.
  • BP bandpass
  • Biological Example 15 Assessment of CXCL10 production, upon treatment with shRNA 1, RNA Conjugate 1, and RNA Conjugate 2 in ALI cultured human nasal epithelial cells
  • ALI cultured pooled donors human nasal epithelium (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase with MucilAirTM culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions.
  • ALI cultures were dosed apically with 50 ⁇ L vehicle control (citrate buffer alone: 0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in citrate buffer, shRNA 1 (2 mg/mL) with surfactant component in citrate buffer, RNA Conjugate 1 (2 mg/mL) in citrate buffer, RNA Conjugate 1 (2 mg/mL) with surfactant component in citrate buffer, RNA Conjugate 2 (2 mg/mL) in citrate buffer, or RNA Conjugate 2 (2 mg/mL) with surfactant component in citrate buffer, for 30 min (then being removed with a pipette).24 hrs post-treatment, ALI cultures were harvested and CXCL10 concentration was quantified via the method described in Biological Example 1.
  • Biological Example 16 Assessment of viral load and virus-induced inflammation, upon treatment with shRNA 1, in human rhinovirus (HRV) infected, ALI cultured, human bronchial epithelial cells obtained from an asthma donor Experimental Methods ALI cultured human bronchial epithelial cells obtained from an asthma donor (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase (ALI) with MucilAirTM culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions.
  • HRV human rhinovirus
  • ALI human bronchial epithelial cells obtained from an asthma donor
  • ALI air-liquid interphase
  • MucilAirTM culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions.
  • human rhinovirus type 16 (HRV16) virus inoculum 100 ⁇ L of 400,000 PFU/mL in MucilAirTM culture medium to give a final MOI of approximately 0.2
  • HRV16 human rhinovirus type 16 virus inoculum
  • ALI cultures were dosed apically with 50 ⁇ L media, surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate), shRNA 1 (2 mg/mL) with surfactant component in phosphate buffer, or pleconaril, a known HRV inhibitor (10 ⁇ M in media) for 30 min (then being removed with a pipette; almost completely and without additional wash) both two days (Day -2) and one day (Day -1) prior to virus inoculation (on Day 0), before subsequent re-application of the treatments 30 min prior to virus inoculation (then being removed as described above) and subsequent incubation with virus inoculum for 90 min.
  • phosphate buffer 0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate
  • TCID 50 Tissue Culture Infectious Dose
  • treatment with a formulation comprising shRNA 1 with surfactant component as greater effect in reducing pro-inflammatory cytokine release relative to treatment with the known HRV inhibitor, pleconaril.
  • high levels of HRV replication were detected in an apical wash from HRV-infected bronchial epithelial cells treated with media (i.e. vehicle) on day 2 post-inoculation (mean: 4.1 Log, TCID 50 /mL). Treatment of HRV-infected bronchial epithelial cells with surfactant component alone had no notable anti-viral effect.
  • treatment with a formulation comprising shRNA 1 with surfactant component had a clear anti-viral effect, as represented by a 0.9 and 1.6 Log reduction in viral load relative to treatment with media only or surfactant component only, respectively.
  • the known HRV inhibitor, pleconaril had a more significant anti-viral effect, as represented by a 2.3 Log reduction in viral load relative to treatment with media only.
  • treatment with a formulation comprising shRNA 1 with surfactant component has a clear anti-inflammatory and anti-viral effect.
  • Thiis is particularly pertinent as HRV, and in particular HRV16, is a known driver of asthma exacerbations.
  • CXCL10 expression was quantified by an enzyme-linked immunosorbent assay using the MSD multiplex platform and viral load was quantified using TCID 50 values calculated using the Reed-Muench formula. More pertinently, the surfactant component, which itself demonstrated minimal anti-viral activity, was able to potentiate the activity of anti-viral polynucleotide molecules, as represented by increased induction of CXCL10 expression prior to virus infection and decreased viral load in influenza virus infected ALI cultured nasal epithelium.
  • the in vitro anti-viral activity of a formulation of an anti-viral polynucleotide molecule, such as shRNA 1, with a surfactant component has also been demonstrated in a human rhinovirus (HRV16)-infected and a human respiratory syncytial virus (RSV) A2-infected ALI cultured nasal epithelium model (see Biological Examples 7 and 8).
  • HRV16 human rhinovirus
  • RSV respiratory syncytial virus
  • formulations comprising surfactant component were demonstrated to clearly increase the integrity and improve the barrier function of the epithelium, as represented by an increase in transepithelial electrical resistance (TEER), which has a clear protective effect against virus-induced epithelial damage in a human respiratory syncytial virus (RSV) A2- infected ALI cultured nasal epithelium model (see Biological Example 8).
  • TEER transepithelial electrical resistance
  • RSV respiratory syncytial virus
  • formulations comprising surfactant component were improved in their protective against such virus-induced epithelial damage relative to formulations comprising the anti-viral polynucleotide molecule, shRNA 1, which alone was only able to mitigate the loss in TEER/restore the reduced TEER observed following human RSV A2 infection of ALI cultured nasal epithelium.
  • the anti-viral activity of the polynucleotide molecule with surfactant formulation was comparable to, if not slightly superior to, the anti-viral activity of the commercially available anti-viral drug oseltamivir.
  • the polynucleotide molecule, shRNA 1, in vehicle had a protective effect against influenza virus-induced body weight loss in infected mice
  • formulation of said polynucleotide molecule with surfactant formulation improved the protective effect of the polynucleotide molecule such that said polynucleotide molecule prevented influenza virus- induced body weight loss to a similar extent to oseltamivir.
  • the anti-viral activity of the formulation of shRNA 1 with surfactant component resulted in a formulation with in vivo anti-viral activity comparable to the commercially available anti-viral drug ribavirin (see Biological Example 9).
  • the surfactant component alone had no significant protective effect against human RSV A2-induced body weight loss in infected mice
  • formulation of the anti-viral polynucleotide molecule, shRNA 1, in said surfactant component resulted in a significant protective effect, and in fact prevented human RSV A2-induced body weight loss in infected mice to a similar extent to ribavirin.
  • the in vivo anti-viral activity of the formulations disclosed herein was further assessed upon administration via different routes, and in particular following subcutaneous administration.
  • saline i.e. non-treatment
  • surfactant component alone had no significant anti-viral activity
  • the anti-viral activity of a subcutaneously administered formulations comprising an anti-viral polynucleotide molecule, shRNA 1, with surfactant component was highly significant, as represented by a substantial reduction in viral load and simultaneous reduction in neutrophil accumulation in both the lung and nasal tissue of influenza virus (PR8)-infected mice (see Biological Example 10).
  • the anti-viral 118 activity of the formulation comprising shRNA 1 with surfactant component was comparable to the anti-viral effect observed in influenza virus-infected mice orally treated with the commercially available drug oseltamivir, and simultaneously subcutaneously treated with a formulation comprising surfactant component alone. More pertinently, the anti-viral effect observed in influenza virus-infected mice subcutaneously treated with the formulation comprising shRNA 1 with surfactant component in combination with oral treatment with oseltamivir, was more significant that the anti-viral effect observed following treatment with either formulation/drug alone (see Biological Example 10). Moreover, whilst subcutaneous treatment of influenza virus-infected mice with vehicle (i.e.
  • saline alone failed to protect against influenza virus-induced body weight loss in infected mice
  • formulation of the anti-viral polynucleotide molecule, shRNA 1, in said surfactant component resulted in a significant protective effect, and in fact prevented influenza virus-induced body weight loss in infected mice to a similar extent to oral oseltamivir treatment (when co-administered with subcutaneous surfactant component alone) (see Biological Example 10).
  • mice with rH1N1 HA in PBS alone, or following treatment with the control adjuvant, CPG-ODN resulted in moderate protection against influenza virus infection
  • vaccination of mice with rH1N1 HA following treatment with the shRNA 1 with surfactant component formulation resulted in more significant protection against influenza virus infection, as represented by a significant reduction in viral and simultaneous reduction in neutrophil accumulation in both the lung and nasal tissue of infected mice (see Biological Example 11).
  • the protective effect of intranasal rH1N1 HA vaccination was more greatly enhanced by previous treatment with the shRNA 1 with surfactant component formulation of the present invention than by previous treatment with the control adjuvant, CPG-ODN.
  • the present inventors consider that the liquid formulations of the present invention improve delivery of a formulated polynucleotide molecule to target cells, in particular ALI cultured nasal epithelial cells, and consequently enhance downstream activity of said formulated polynucleotide molecule.
  • target cells in particular ALI cultured nasal epithelial cells
  • the present inventors consider that the liquid formulations of the present invention improve delivery of a formulated polynucleotide molecule to target cells, in particular ALI cultured nasal epithelial cells, and consequently enhance downstream activity of said formulated polynucleotide molecule.
  • Support for this hypothesis is derived from the observation that the expression of mRNA or DNA encoded biomarkers was significantly higher upon treatment of cells with the biomarker-encoding mRNA or DNA with surfactant component relative to corresponding treatment with said mRNA or DNA in vehicle (buffer component) only (see Biological Examples 6, 12, and 14).
  • treatment of ALI cultured nasal epithelial cells with shRNA 1 with surfactant component in either citrate or phosphate buffer led to a strong anti-viral effect, as represented by a significant reduction in viral load relative to treatment with vehicle, i.e. citrate or phosphate buffer alone (see Biological Example 13).
  • treatment of ALI cultured nasal epithelial cells with shRNA 1 with surfactant component formulations, in either citrate or phosphate buffer which were produced using either low-shear mixing, i.e. magnetic stirring, or high-shear mixing led to a strong anti-viral effect, as represented by a significant reduction in viral load relative to treatment with vehicle, i.e. citrate or phosphate buffer alone (see Biological Example 13).
  • the present inventors investigated the utility of the liquid formulations of the present invention as a prophylactic treatment for asthma exacerbations, in particular human rhinovirus (HRV) infection-associated asthma exacerbations.
  • HRV human rhinovirus
  • treatment of HRV-infected bronchial epithelial cells from an asthma donor with formulations comprising shRNA 1 with surfactant component effectively inhibited virus-induced release of the pro-inflammatory cytokines CXCL10, CXCL8, and CCL5, by day 5 post-inoculation (see Biological Example 16), and in fact inhibited such release to a greater extent than the known HRV inhibitor pleconaril.
  • liquid pharmaceutical formulations of the invention including a surfactant component comprising a mixture of a fatty acid and a non-ionic surfactant, and a polynucleotide molecule, and specifically in the form of a stable colloidal emulsion, are expected to be useful for improving the delivery and consequently increasing the exposure of said polynucleotide molecule, and consequently enhancing the therapeutic effect of said polynucleotide molecule.
  • Biophysical Examples Biophysical Example 1 Formulations comprising a polynucleotide molecule, shRNA 1, a surfactant component of three different concentrations (1: 0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80; 2: 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80; and 3: 0.2% (w/w) oleic acid and 0.15% (w/w) polysorbate 80) and vehicle (water) were produced by first adding oleic acid and polysorbate 80 at the concentration required to achieve the desired final concentration to RNAse free water in a 0.5 L glass vessel prior to mixing for 10 min using an IKA EURO-STAR Overhead Lab Mixer at about 50% power.
  • shRNA 1 a solution of shRNA 1 (0.2% (w/w)) was dispersed in the surfactant component and mixed as described above to produce a shRNA 1 surfactant composition.
  • carboxymethyl cellulose 2% (w/w)
  • hyaluronic acid were dispersed in purified water and homogenised using a Silverson L5M homogenizer at 8500 rpm for 2 min. This suspension was further combined with glycerol (2.1% (w/w)) by continuous stirring for 10 min.
  • shRNA 1 surfactant composition and the suspension were then combined by continuous mixing for 10 min, prior to buffering with sodium citrate dihydrate (0.28% (w/w)) and citric acid monohydrate (0.20% (w/w)) and mixing for a further 10 min to ensure uniform pH adjustment.
  • a comparator formulation comprising shRNA 1 in vehicle was produced similarly.
  • dynamic light scattering techniques by means of the Zetasizer Nano S apparatus (Malvern Instruments Ltd., UK), were used to measure the translational diffusion of polynucleotide:surfactant component particles due to Brownian motion.
  • shRNA 1 with surfactant component 1 (0.005% (w/w) oleic acid 121 and 0.0045% (w/w) polysorbate 80) in vehicle, which comprises surfactant at the lowest of the tested concentrations, results in a formulation of high polydispersity and comprising particles of a highly variable size.
  • Table 3 The effect of formulation of shRNA 1 with surfactant components of different concentrations on particle size and polydispersity Polydispersity Formulation Z-Average Size (Standard Deviation) (nm) Index shRNA 1 in water 174.57 (25.91) 0.42 (0.05) shRNA 1 with surfactant 124.15 (27.17) 0.61 (0.15) component 1 in water shRNA 1 with surfactant 197.43 (6.55) 0.47 (0.02) component 2 in water shRNA 1 with surfactant 182.63 (4.60) 0.32 (0.03) component 3 in water 122 Biophysical Example 2 Formulations comprising a polynucleotide molecule, shRNA 1, and a variety of surfactant components (see Table 4, below) were produced by firstly adding 5 mL of RNAse free water to the desired concentration of the relevant fatty acid (i.e.
  • RNAse free water by simultaneously, in a separate vessel, adding 5 mL of RNAse free water to the desired concentration of the relevant non-ionic surfactant (i.e. polysorbate 80 or Brij 35).
  • the relevant non-ionic surfactant i.e. polysorbate 80 or Brij 35.
  • Each of these fatty acid and non-ionic surfactant mixtures was manually mixed using a stainless steel spatula.
  • Each of the fatty acid mixture and non-ionic surfactant mixture were then added to 80 mL of RNAse free water in a 0.25 L glass vessel. This solution was mixed for about 10 min using an IKA EURO-STAR Overhead Lab Mixer at about 50% power, until a homogenous mixture was produced.
  • shRNA 1 (0.2% (w/w) was dispersed in the surfactant component mixture solution and mixed as descried above, until entirely unfirm in constitution, to produce a formulations of shRNA in the relevant surfactant component.
  • the pH of the shRNA 1 in surfactant component formulations was then adjusted to the desired value using either 1 M 1 in L5M nual o S on of said kes- y co- dded ent.
  • terial ment As illustrated in Figures 40 to 44 and Table 4 (below), the hydrodynamic diameter of shRNA 1 in each of the tested surfactant component formulations is largely consistent as indicated by the small value of the standard deviation of the Z-average size. This consistent particle size is 123 advantageous in a pharmaceutical product.
  • each of the tested surfactant component formulations results in a relatively low polydispersity, which is advantageous in a pharmaceutical product.
  • the different composition of the surfactant components appeared to influence the average particle size, although each of the tested surfactant components formulations resulted in an average particle size which is suitable for use in a pharmaceutical product.
  • the larger average particle size observed in relation to the surfactant component of polysorbate 80 (4.5% (w/w)) + caprylic acid (5.0% (w/w)) is likely due to use of the surfactant component elements at a higher concentration.
  • the formulations were imaged using cryo-transmission 124 electron microscopy (cryo-TEM), in order to observe the particles formed by the formulations, and in particular by the surfactant component of the formulation.
  • cryo-TEM cryo-transmission 124 electron microscopy
  • a sample of the formulations was plunge frozen onto Lacey Carbon grids (size: 300), which had been made hydrophilic via glow discharge at 15 mA for 25 seconds.
  • the plunge freezing process was performed under controlled conditions: 15 o C and 90% humidity, and utilised a blot time of 2 seconds.
  • the Lacey Carbon grids were plunge frozen into liquid ethane and stored under liquid nitrogen until imaging.
  • Imaging was performed using the FEI Talos L120C G2 Transmission Electron Microscope, where sample Lacey Carbon grids were loaded into the Gatan transfer holder and maintained at temperatures below -170°C.
  • Low dose imagine techniques were used to minimise radiation- associated damage, involving use of a software-controlled beam blanking system to limit electron exposure prior to image capture.
  • the beam was focused on an area of limited interest adjacent to an area of interest, prior to re-tilting to the original position during exposure mode for capture of the image at the area of interest.
  • Images were taken using the BM-Ceta CCD Camera, employed under focus to enhance Figures 45 (B), (E), and (F), show that the polynucleotide molecule (e.g.
  • shRNA 1 is present within (i.e. is a component of) the stable colloidal particles, and for example is present around the core of the particles and at the interface between the core of the particles and the non- ionic surfactant (see the inner dark band of the particles in these Figures).
  • 125 Biophysical Example 4 Formulations comprising a polynucleotide molecule, shRNA 1, were prepared using a surfactant component comprising oleic acid (0.05 % (w/w)) and polysorbate 80 (0.045% (w/w)) according to Biophysical Example 1 and Biophysical Example 2 and using high-shear mixing.
  • the resultant DSC data was processed via subtraction of the formulation buffer baseline from the sample endotherm data, prior to integration using a sigmoidal and polynomial baseline where applicable.
  • An overlay of baseline-subtracted thermograms allows for direct comparison of thermal transitions between samples, revealing variations in thermal properties and in particular in melting temperature (T m ), which is indicative of thermal stability.
  • T m melting temperature
  • the melting temperature (T m ) of shRNA 1 in vehicle (water) has been determined as 30 o C.
  • the T m of shRNA 1 when prepared in the surfactant component formulation is about 67 o C, which is indicative of incorporation of shRNA 1 into a stable colloidal emulsion, and resultant stabilisation of said shRNA.
  • a clear exothermic event is present within the DSC thermogram at between 70 o C and 90 o C, which indicates a stabilising interaction between the non-ionic surfactant (i.e. polysorbate 80) and fatty acid (i.e. oleic acid), which is therefore somewhat responsible for the structural stability of the stable colloidal emulsion, and consequently the stabilisation of any polynucleotide molecule packaged therein.
  • non-ionic surfactant i.e. polysorbate 80
  • fatty acid i.e. oleic acid
  • Figure 46 demonstrates the stability of stable colloidal emulsions formed by formulations comprising surfactant component (polysorbate 80 0.045% (w/w) + oleic acid (0.05% (w/w)) only (top panel), formulations comprising 2 mg/mL polynucleotide molecule (shRNA 1) with surfactant component (middle panel), and formulations comprising 20 mg/mL polynucleotide molecule (shRNA 1) with surfactant component (bottom panel).
  • surfactant component polysorbate 80 0.045% (w/w) + oleic acid (0.05% (w/w)
  • a surfactant component which comprises a mixture of a fatty acid and a non-ionic surfactant
  • a surfactant component which comprises a mixture of a fatty acid and a non-ionic surfactant
  • formulation of a polynucleotide molecule, specifically shRNA 1 with a surfactant component comprising caprylic acid or oleic acid (as the fatty acid) and polysorbate 80 or Brij 35 (as the non-ionic surfactant), in each case produced a formulation in which stable colloidal particles of a consistent particle size are formed, and in which the polydispersity of the formulation is low (see Biophysical Example 2).
  • formulations of the present invention producing stable colloidal emulsions was provided upon visualisation of said formulation.
  • formulations comprising a range of surfactant components (i.e. different fatty acids and non-ionic surfactants), each produced clearly visible, but sparsely distributed, particles, of a consistent size and shape, and which typically ranged between 50 and 300 nm in diameter.
  • surfactant components i.e. different fatty acids and non-ionic surfactants
  • the stable colloidal emulsions formed by the formulations of the invention in particular as a result of the presence of a surfactant component comprising a mixture of a fatty acid and a non-ionic surfactant, have been demonstrated to be highly stable, in particular as a result of specific interaction between said fatty acid and said non-ionic surfactant.
  • the stable colloidal emulsion formulations of the present invention to significantly stabilise a polynucleotide molecule, specifically shRNA 1, packaged therein, as represented by an increase in melting temperature (T m ) (see Biophysical Example 4).
  • liquid pharmaceutical formulations of the invention which include a surfactant component comprising a mixture of a fatty acid and a non-ionic surfactant and a polynucleotide molecule, and specifically in the form of a stable colloidal emulsion, are expected to be useful for efficiently packaging and delivering and said polynucleotide molecule, whilst also maintaining stability of said polynucleotide molecule.
  • the liquid pharmaceutical formulations of the invention are expected to be particularly suitable pharmaceutical products due to the consistent particle size of the stable colloidal particles in said formulations, and the low polydispersity of the formulations.
  • Boda B Benaoudia S, Huang S, Bonfante R, Wiszniewski L, Tseligka ED, Tapparel C, Constant S. Antiviral drug screening by assessing epithelial functions and innate immune responses in human 3D airway epithelium model. Antiviral Res. 2018 Aug; 156:72-79. doi: 10.1016/j. antiviral.2018.06.007

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Emergency Medicine (AREA)
  • Dispersion Chemistry (AREA)
  • Inorganic Chemistry (AREA)
  • Otolaryngology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides inter alia aqueous liquid pharmaceutical formulations comprising a surfactant component and a polynucleotide molecule. The formulations of the present invention are for use as medicaments, for example for use in the treatment or prevention of viral infection and disease associated with viral infection.

Description

Novel Formulations Related Applications This application claims the benefit of U.S. Provisional Application No.63/510,985, filed on June 29, 2023 and International Application No. PCT/US24/30546 filed on May 22, 2024. The entire teachings of the above applications are incorporated herein by reference. Sequence Listing The sequence listing submitted via EFS, in compliance with 37 CFR §1.52(e)(5), is incorporated herein by reference. The sequence listing XML file submitted via EFS contains the file “4329.3002 WO SEQ Listing.xml”, created on June 27, 2024, which is 13,174 bytes in size. Field This invention relates to aqueous liquid pharmaceutical formulations comprising a surfactant component and a polynucleotide molecule. The invention also relates inter alia to aqueous liquid pharmaceutical formulations for use as a medicament, for example for use in the treatment or prevention of viral infection and disease associated with viral infection, and related methods of treatment. Background of the Invention It is essential that a therapeutic agent is delivered to the target tissues and/or cells of a subject in an amount that allows said therapeutic agent to exert a therapeutic effect. Indeed, insufficient delivery of a therapeutic agent often impedes the ability of an otherwise efficacious therapeutic agent to achieve desired therapeutic outcomes. This problem is particularly pronounced for biologic therapeutics, such as immunoglobulins and polynucleotide molecules, in particular genomic DNA (gDNA), complementary DNA (cDNA), mRNA, siRNA, and shRNA, whose macromolecular nature and net ionic charge add additional complexities to targeted delivery. Unlike small molecule therapeutics, biologics do not undergo passive diffusion across cell membranes. Moreover, upon introduction intro a subject, polynucleotide molecules are susceptible to degradation by endonuclease and exonuclease enzymes which can hydrolyse the phosphodiester bonds of the polynucleotide backbone. Accordingly, the clinical value of polynucleotide therapeutics is dependent on delivery technologies that improve the stability of the polynucleotide, facilitate efficient internalisation, and increase target affinity (Kulkarni et al.2021). In view of the challenges associated with delivery of polynucleotide therapeutics, a number of platform delivery technologies have been developed, in particular in the context of gene 1    therapy products. These include chemically-modified antisense oligonucleotides (ASOs), N- acetylgalactosamine (GalNAc) conjugates, adeno-associated virus (AAV) vectors and lipid delivery systems, including lipid nanoparticles (LNPs) and liposomes (Kulkarni et al.2021). ASOs refer to polynucleotide molecules comprising a number of chemical modifications to the backbone, sugar moiety, or nitrogenous base to enhance affinity to target RNA, improve nuclease resistance and modulate the immunological profile of the polynucleotide (Khvorova and Watts, 2017). GalNAc conjugation facilitates accumulation of relevant polynucleotide therapeutics, particularly in the liver. Specifically, the GalNAc construct targets the asialoglycoprotein receptor which is predominantly expressed on liver hepatocytes. Subsequent internalisation via clathrin-mediated endocytosis, and endosomal escape by the polynucleotide molecule provides targeted delivery (Springer and Dowdy, 2018). These chemical modification technologies have significant utility but may complicate manufacturing processes and increase costs. Conversely, AAV vector systems are comparably simple and may provide efficient delivery of polynucleotide molecule to the nucleus. Moreover, different AAVs display different cell tropisms, such that the platform technology can be adapted to different target tissues or cells. However, lipid delivery systems, including lipid nanoparticles and liposomes, have increasingly become recognised as the most promising delivery system for polynucleotide molecules. In particular, the biocompatibility of lipid-based formulations and their respective ease of manufacturing, particularly at a large-scale, has made such formulations an attractive avenue for research and development. Moreover, lipid delivery systems are highly efficient in delivering a polynucleotide molecule into a target cell. Nevertheless, lipid delivery systems are associated with a number of problems. For example, lipid delivery systems, such as lipid nanoparticles and liposomes, frequently include a polyethylene glycol (PEG)-based compound which prevents the aggregation, and subsequent immune recognition and elimination, of lipid particles (Jokerst et al.2011). Indeed, PEGylated lipid particles appear to have an increased half-life in the circulation (Huang and Liu, 2011). However, PEGylated lipid nanoparticles have been reported to severely inhibit endosomal release of polynucleotide molecules (Song et al. 2002), as well as stimulating undesirable immune response, for example the raising of an antibody response (Garay and Labaune, 2011). Alternative polymers to PEG have been investigated, including naturally occurring polymers, such as serum albumin, and zwitterionic polymers, such as poly(carboxybetaine) (Hoang Thi et al. 2020). However, these polymers generally perform less favourably and are found in a number of other common products or pharmaceutical compositions leading to concerns about their immunogenicity. Moreover, the above-described polymers, and alternative technologies 2    such as XTEN peptides, although able to extend lipid particle half-life in vivo, are often large and therefore inhibitory to polynucleotide molecule uptake. Lipid delivery systems comprising polynucleotide molecules must be administered to a subject via a route that is tolerable to the subject, supporting patient compliance, and that ensures the polynucleotide molecule is delivered to the target tissue or cells at a concentration suitable for exerting a therapeutic effect. Typically, lipid delivery systems comprising polynucleotide molecules are administered intravenously, for example to facilitate systemic administration, or via local injection to a target tissue or organ, for example via the intradermal, subcutaneous, intra-ocular, intramuscular, intra-myocardial or intra-tumoral route. Notably, lipid delivery systems comprising polynucleotide molecules are less commonly administered topically to the lung or nose (Li et al. 2023), i.e., via inhalation or intranasal administration, despite the apparent suitability of these administration routes for delivering a polynucleotide therapeutic to the respiratory tract, particularly wherein the polynucleotide therapeutic serves to treat a disease of the respiratory tract, for example a respiratory viral infection or cystic fibrosis. However, administration of lipid delivery systems comprising polynucleotide molecules to the respiratory tract, in particular via topical administration to the lung or nose, are associated with a number of well-documented challenges. Firstly, a lipid delivery system comprising a polynucleotide molecule must be administered intranasally, or via inhalation, in a manner that ensures a therapeutically effective dose is provided in the region of the respiratory tract where the therapeutic effect is desired, e.g. the upper or lower respiratory tract. Moreover, administration should occur within a time frame that supports high patient compliance. Furthermore, regulatory requirements provide that lipid nanoparticle and liposome drug products are subject to additional considerations such as vesicle or particle size, size distribution and morphology (FDA Guidance for Industry entitled “Liposome Drug Products Chemistry, Manufacturing, and Controls; Human Pharmacokinetics and Bioavailability; and Labelling Documentation”, 2018). Moreover, lipid nanoparticles and liposomes are susceptible to fusion (i.e., combination of smaller lipid particles into larger lipid particles), aggregation, and leakage of the contained polynucleotide molecule, each of which can be detrimental to the stability of the contained polynucleotide molecule. In conclusion, there remains a need to develop liquid pharmaceutical formulations, and in particular liquid pharmaceutical formulations, which enable efficient delivery of a polynucleotide molecule to a target tissue or cell(s), and which avoid some of the above- described challenges associated with use of lipid delivery systems, in particular use of lipid delivery systems to deliver polynucleotide molecules via inhalation or intranasally. 3    Summary of the Invention Commonly used excipients are generally regarded as well-understood and pharmacologically inert. However, the present inventors have made the surprising discovery that certain surfactants, which have been used as conventional excipients in pharmaceutical formulations, have specific biophysical and biological activity. The present invention therefore provides an aqueous liquid pharmaceutical formulation comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule. The present invention further provides an aqueous liquid pharmaceutical formulation comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule for use as a medicament, for example for use in the in the treatment or prevention of viral infection and disease associated with viral infection. In embodiments such formulations are, for example, suitable for topical administration to the lung or nose, as well as other routes. In embodiments, the formulations of the present invention suitably form a stable colloidal emulsion. Brief Description of the Figures Figure 1 shows the effect of apical treatment with vehicle (water alone), a polynucleotide molecule (shRNA 1) in vehicle (water alone), or with a surfactant component in vehicle at three different concentrations (surfactant component 1: 0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80; surfactant component 2: 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80; and surfactant component 3: 0.2% (w/w) oleic acid and 0.15% (w/w) polysorbate 80) on CXCL10 release in the basal chamber from air-liquid interface (ALI) cultured nasal epithelium on days 1, 2 and 3 post-treatment. Figure 2 shows the effect of apical treatment with vehicle (water alone) a polynucleotide molecule (shRNA 1) in vehicle (water alone), or with a surfactant component in vehicle at two different concentrations (0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80; and 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) on CXCL10 release in an apical wash from ALI cultured nasal epithelium on day 1 post-treatment (i.e. prior to virus infection). The effects of these treatments are compared to basolateral treatment with oseltamivir. Figure 3 shows the effect of apical treatment with vehicle (water alone), a polynucleotide molecule (shRNA 1) in vehicle (water alone), or with a surfactant component in vehicle at two different concentrations (0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80; and 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) on viral load in an apical wash from influenza virus infected ALI cultured nasal epithelium on day 2 post-infection (i.e. day 3 post- 4    treatment). The effects of these treatments are compared to basolateral treatment with oseltamivir. Figure 4 shows the effect of apical treatment with vehicle (water alone), a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) alone in vehicle (water) , a polynucleotide molecule (shRNA 1) in vehicle (water), or a polynucleotide molecule (shRNA 1) with said surfactant component in vehicle (water), on CXCL10 release in an apical wash from ALI cultured nasal epithelium on day 1 post-treatment (i.e. prior to virus infection). The effects of these treatments are compared to basolateral treatment with oseltamivir. Figure 5 shows the effect of apical treatment with vehicle (water alone), a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) alone in vehicle, and a polynucleotide molecule (shRNA 1) in vehicle or a polynucleotide molecule (shRNA 1) with said surfactant component in vehicle, on viral load in an apical wash from influenza virus infected ALI cultured nasal epithelium on day 2 post-infection (i.e. day 3 post-treatment). The effects of these treatments are compared to basolateral treatment with oseltamivir. Figure 6 shows the effect of apical treatment of dsRNA 1 or dsRNA 2 in vehicle (buffer: 0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate), or with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, on CXCL10 release in an apical wash from ALI cultured nasal epithelium on day 1 post-treatment (i.e. prior to virus infection). Figure 7 shows the effect of apical treatment of dsRNA 1 or dsRNA 2 in vehicle (buffer: 0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate), or with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, on viral load in an apical wash from influenza virus infected ALI cultured nasal epithelium on day 1 post- infection (i.e. day 2 post-treatment). Figure 8 shows the effect of intranasal treatment with vehicle (water alone), with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle together with further pharmaceutically acceptable excipients (see Formulation Example 1A minus the shRNA 1, hereinafter “surfactant formulation”), and a polynucleotide molecule (shRNA 1) in vehicle, or in the above described surfactant formulation (see Formulation Example 1A, hereinafter “shRNA 1 with surfactant formulation”), on viral load in nasal tissue from influenza (PR8) infected mice on day 1 and day 5 post-infection. The effects of these treatments are compared to oral treatment with oseltamivir phosphate. Figure 9 shows the effect of intranasal treatment with vehicle (water alone), with a formulation comprising a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) and further pharmaceutically acceptable excipients in vehicle (see Formulation Example 1A minus the shRNA 1, hereinafter “surfactant formulation”), and a polynucleotide molecule (shRNA 1) in vehicle, or in the above described surfactant formulation (see Formulation 5    Example 1A, hereinafter “shRNA 1 with surfactant formulation”), on neutrophil accumulation in the nasal lavage of influenza (PR8) infected mice on Day 1 and Day 5 post-infection. The effects of these treatments are compared to oral treatment with oseltamivir phosphate. Figure 10 shows the effect of intranasal treatment with vehicle (water alone), with a formulation comprising a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) and further pharmaceutically acceptable excipients (see Formulation Example 1A minus the shRNA 1, hereinafter “surfactant formulation”), and a polynucleotide molecule (shRNA 1) in vehicle, or in the above described surfactant formulation (see Formulation Example 1A, hereinafter “shRNA 1 with surfactant formulation”), on body weight loss observed in influenza (PR8) infected mice during the five days post-infection. The effects of these treatments are compared to oral treatment with oseltamivir phosphate. Figure 11 shows the effect of apical treatment with a GFP-encoding mRNA in a buffer (0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate), or with said GFP- encoding mRNA with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in said buffer, on mRNA exposure and consequently on GFP signal in ALI cultured nasal epithelial cells 24 hrs post-treatment. Figure 12 shows the effect of apical treatment with a β-galactosidase-encoding (β-gal) plasmid in a buffer (0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate), or with said β-gal plasmid with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in said buffer, on plasmid exposure and consequently on β- galactosidase enzyme activity in ALI cultured nasal epithelial cells 24 hrs post-treatment. Figures 13 (A-D) show the effect of the presence of a surfactant component of three different concentrations (surfactant component 1: 0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80; surfactant component 2: 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80; and surfactant component 3: 0.2% (w/w) oleic acid and 0.15% (w/w) polysorbate 80) on the polydispersity and particle size of formulations comprising a polynucleotide molecule (shRNA 1) in vehicle (water). Figure 14 shows the effect of apical treatment with vehicle (buffer alone), a polynucleotide molecule (shRNA 1) in buffer, a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) or shRNA 1 with surfactant component in vehicle, on CXCL10 release in an apical wash from ALI cultured nasal epithelium on day 1 post-treatment (i.e. prior to virus infection). Figure 15 shows the effect of apical treatment, on day 1 and on day 0 pre-infection, with vehicle (buffer alone), a polynucleotide molecule (shRNA 1) in buffer, a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) or shRNA 1 with surfactant component in vehicle, on viral load in an apical wash from human rhinovirus (HRV16) infected ALI cultured nasal epithelium on day 2 post-infection (i.e. day 3 post-first treatment). 6    Figure 16 shows the effect of apical treatment, on day 1 and on day 0 pre-infection, with vehicle (buffer alone), a polynucleotide molecule (shRNA 1) in buffer, a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) or shRNA 1 with surfactant component in vehicle, on viral load in an apical wash from respiratory syncytial virus (RSV) A2 infected ALI cultured nasal epithelium on day 3 post-infection (i.e. day 4 post-first treatment). Figure 17 shows the effect of apical treatment, on day 1 and on day 0 pre-infection, with vehicle (buffer alone), a polynucleotide molecule (shRNA 1) in buffer, a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) or shRNA 1 with surfactant component in vehicle, on cell integrity, as represented by transepithelial electrical resistance (TEER), in a respiratory syncytial virus (RSV) A2 infected ALI cultured nasal epithelium model, on days 0, 1, 2 and 3 post-infection. Figure 18 shows the effect of intranasal treatment with vehicle (saline alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, or a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, on viral load in lung tissue from respiratory syncytial virus (RSV) A2 infected mice on day 4 post-infection. The effects of these treatments are compared to intranasal treatment with ribavirin. Figure 19 shows the effect of intranasal treatment with vehicle (saline alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, or a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, on neutrophil accumulation in the nasal lavage of respiratory syncytial virus (RSV) A2 infected mice on day 4 post-infection. The effects of these treatments are compared to intranasal treatment with ribavirin. Figure 20 shows the effect of intranasal treatment with vehicle (saline alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, or a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, on body weight loss observed in respiratory syncytial virus (RSV) infected mice during the four days post-infection. The effects of these treatments are compared to intranasal treatment with ribavirin. Figure 21 shows the effect of subcutaneous treatment with vehicle (saline alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer, or a polynucleotide molecule (shRNA 1) with surfactant component (2 or 20 mg/mL) in phosphate buffer, on CXCL10 release in the serum of mice 24 hrs post-treatment. Figure 22 shows the effect of subcutaneous treatment with vehicle (saline alone; i.e. non- treatment), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, surfactant component in vehicle in combination with oral oseltamivir treatment, or shRNA 1 with surfactant component in vehicle in combination with oral oseltamivir treatment, on viral load in the lung tissue of influenza (PR8) virus infected mice on day 5 post-infection. 7    Figure 23 shows the effect of subcutaneous treatment with vehicle (saline alone; i.e. non- treatment), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, surfactant component in vehicle in combination with oral oseltamivir treatment, or shRNA 1 with surfactant component in vehicle in combination with oral oseltamivir treatment, on viral load in the nasal tissue of influenza (PR8) virus infected mice on day 5 post-infection. Figure 24 shows the effect of subcutaneous treatment with vehicle (saline alone; i.e. non- treatment), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, surfactant component in vehicle in combination with oral oseltamivir treatment, or shRNA 1 with surfactant component in vehicle in combination with oral oseltamivir treatment, on neutrophil accumulation in the lung tissue of influenza (PR8) infected mice on day 5 post- infection. Figure 25 shows the effect of subcutaneous treatment with vehicle (saline alone; i.e. non- treatment), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, surfactant component in vehicle in combination with oral oseltamivir treatment, or shRNA 1 with surfactant component in vehicle in combination with oral oseltamivir treatment, on neutrophil accumulation in the nasal tissue of influenza (PR8) infected mice on day 5 post- infection. Figure 26 shows the effect of subcutaneous treatment with vehicle (saline alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, a polynucleotide molecule (shRNA 1) with surfactant component in vehicle, surfactant component in vehicle in combination with oral oseltamivir treatment, or shRNA 1 with surfactant component in vehicle in combination with oral oseltamivir treatment, on body weight loss observed in influenza (PR8) infected mice during the five days post-infection. Figure 27 shows the effect of intranasal vaccination with vehicle (i.e. PBS alone), recombinant H1N1 (rH1N1) haemagglutinin (HA) in vehicle, rH1N1 HA in vehicle following previous treatment with a polynucleotide molecule (shRNA 1) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or rH1N1 HA in vehicle following previous treatment with the control adjuvant, CPG-ODN, on viral load in the lung tissue of influenza (PR8) infected mice on day 5 post-infection. Figure 28 shows the effect of intranasal vaccination with vehicle (i.e. PBS alone), recombinant H1N1 (rH1N1) haemagglutinin (HA) in vehicle, rH1N1 HA in vehicle following previous treatment with a polynucleotide molecule (shRNA 1) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or rH1N1 HA in vehicle following previous 8    treatment with the control adjuvant, CPG-ODN, on viral load in the nasal tissue of influenza (PR8) infected mice on day 5 post-infection. Figure 29 shows the effect of intranasal vaccination with vehicle (i.e. PBS alone), recombinant H1N1 (rH1N1) haemagglutinin (HA) in vehicle, rH1N1 HA in vehicle following previous treatment with a polynucleotide molecule (shRNA 1) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or rH1N1 HA in vehicle following previous treatment with the control adjuvant, CPG-ODN, on neutrophil accumulation in the lung tissue of influenza (PR8) infected mice on day 5 post-infection. Figure 30 shows the effect of intranasal vaccination with vehicle (i.e. PBS alone), recombinant H1N1 (rH1N1) haemagglutinin (HA) in vehicle, rH1N1 HA in vehicle following previous treatment with a polynucleotide molecule (shRNA 1) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or rH1N1 HA in vehicle following previous treatment with the control adjuvant, CPG-ODN, in vehicle, on neutrophil accumulation in the nasal tissue of influenza (PR8) infected mice on day 5 post-infection. Figure 31 shows the effect of intranasal vaccination with vehicle (i.e. PBS alone), recombinant H1N1 (rH1N1) haemagglutinin (HA) in vehicle, rH1N1 HA in vehicle following previous treatment with a polynucleotide molecule (shRNA 1) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or rH1N1 HA in vehicle following previous treatment with the control adjuvant, CPG-ODN, on body weight loss observed in influenza (PR8) infected mice during the five days post-infection. Figure 32 shows the effect of apical treatment with a GFP-encoding mRNA in a buffer (0.28% (w/w) sodium citrate dihydrate and 0.20% (w/w) citric acid monohydrate), or with said GFP- encoding mRNA with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in said buffer, on mRNA exposure and consequently on GFP signal in ALI cultured nasal epithelial cells 24 hrs post-treatment. Figures 33 and 34 show the effect of apical treatment of ALI cultured nasal epithelium with H1N1 haemagglutinin (HA) mRNA in vehicle (i.e. PBS) or with said H1N1 HA mRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle (i.e. SF), on expression of the H1N1 HA protein in ALI cultured nasal epithelial cells 48 hrs post-treatment. Figure 35 shows effect of apical treatment of ALI cultured bronchial epithelium with cystic fibrosis transmembrane conductance regulator (CFTR)-encoding mRNA in vehicle (i.e. PBS alone) or with said CFTR-encoding mRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, on expression of the CFTR protein in ALI cultured bronchial epithelial cells 72 hrs post-treatment. Figure 36 shows the effect of apical treatment of ALI cultured nasal epithelium with both a low- shear mixed (i.e. magnetically stirred) and high-shear mixed formulation comprising vehicle 9    (i.e. citrate or phosphate buffer alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in citrate or phosphate buffer, or a polynucleotide molecule (shRNA 1) with surfactant component in citrate or phosphate buffer, on viral load in an apical wash from influenza virus (PR8) infected ALI cultured nasal epithelium on day 2 post-infection (i.e. day 3 post-treatment). Figure 37 shows the effect of apical treatment of ALI cultured corneal epithelium with Cy3 labelled siRNA in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate), or with said Cy3 labelled siRNA with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in said phosphate buffer, on siRNA exposure and consequently on fluorescent signal in ALI cultured corneal epithelial cells 4 hrs post-treatment. Figure 38 shows the effect of apical treatment of ALI cultured corneal epithelium with a GFP- encoding mRNA in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate), or with said GFP-encoding mRNA with a surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in said phosphate buffer, on mRNA exposure and consequently on GFP signal in ALI cultured corneal epithelial cells 72 hrs post-treatment. Figure 39 shows the effect of apical treatment of surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in buffer, shRNA 1 in buffer, RNA Conjugate 1 in buffer, RNA Conjugate 1 with surfactant component in buffer, RNA Conjugate 2 in buffer, and RNA Conjugate 2 with surfactant component in buffer, on CXCL10 release in an apical wash from ALI cultured nasal epithelium on day 1 post-treatment Figures 40 to 44 show the effect of the presence of a surfactant component, wherein said surfactant component is polysorbate 80 (0.045% (w/w)) + caprylic acid (0.05% (w/w)), polysorbate 80 (0.045% (w/w)) + oleic acid (0.05% (w/w)), Brij 35 (0.045% (w/w)) + oleic acid (0.05% (w/w)), Brij 35 (0.45% (w/w)) + caprylic acid (0.5% (w/w)), or polysorbate 80 (4.5% (w/w)) + caprylic acid (5.0% (w/w)), on the polydispersity and particle size of formulations comprising a polynucleotide molecule (shRNA 1) in vehicle (water). Figures 45 (A-F) show the particle structures, and diameter thereof, formed in formulations comprising a surfactant component of the present invention, including when the surfactant components comprises (i) (A to D) oleic acid (as the fatty acid) + polysorbate 80 (as the non- ionic surfactant), (ii) (E) caprylic acid (as the fatty acid) + polysorbate 80 (as the non-ionic surfactant), and (iii) (F) caprylic acid (as the fatty acid) + Brij 35 (as the non-ionic surfactant) as viewed under a transmission electron microscope. Figure 46 shows differential scanning calorimetry thermographs for formulations of the present invention which comprise a surfactant component, in particular polysorbate 80 (0.045% (w/w)) + oleic acid (0.05% (w/w)), including a formulation comprising surfactant component only (top 10    panel), a formulation comprising 2 mg/mL polynucleotide molecule (shRNA 1) with surfactant component (middle panel), and a formulation comprising 20 mg/mL polynucleotide molecule (shRNA 1) with surfactant component (bottom panel). Figures 47 to 49 show the effect of apical treatment with media, surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer, shRNA 1 with surfactant component in phosphate buffer, or pleconaril, a known human rhinovirus (HRV) inhibitor, in media, on CXCL10, CXCL8, and CCL5 (respectively, Figures 47 to 49) release in an apical wash from HRV-infected, ALI cultured bronchial epithelium from an asthma donor on day 5 post-virus inoculation. Figure 50 shows the effect of apical treatment with media, surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer, shRNA 1 with surfactant component in phosphate buffer, or pleconaril, a known human rhinovirus (HRV) inhibitor, in media, on viral load in HRV-infected, ALI cultured bronchial epithelium from an asthma donor on day 2 post-virus inoculation. Figure 51 shows a graphic representation of different types of particles formed by formulations comprising a lipid and a surfactant. Detailed Description of the Invention The present invention is based on discoveries made by testing the exposure of polynucleotide molecules, e.g. RNA or DNA molecules, to cells upon formulation with specific surfactant components. The present invention is further based on discoveries made by testing the exposure and therefore anti-viral activity of diverse anti-viral polynucleotide molecules, e.g. RNA or DNA molecules, in combination with specific surfactant components. In particular, the present invention is based on the surprising discoveries that: (i) an apically administered formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with one of a number of diverse anti-viral polynucleotide molecules, for example a shRNA molecule, dsRNA 1, dsRNA 2, RNA Conjugate 1, or RNA Conjugate 2, has a potent effect in stimulating the innate immune response, as a result of improved polynucleotide molecule delivery, as determined by the production of CXCL10, a surrogate marker of anti-viral interferon (IFN) signalling, in air-liquid interface (ALI) cultured nasal epithelium (see Biological Examples 1 to 4 and 15, and Figures 1, 2, 4, 6, and 39); (ii) an apically administered formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with one of a number of diverse anti-viral polynucleotide molecules, for example a shRNA molecule, dsRNA 1, or dsRNA 2, has a powerful effect in 11    reducing viral load in a model of infection comprising influenza virus infected ALI cultured nasal epithelium (see Biological Examples 2 to 4, and Figures 3, 5, and 7); (iii) an apically administered formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with an anti-viral polynucleotide molecule (shRNA 1), has a potent effect in stimulating the innate immune response, as determined by the production of CXCL10, and in reducing viral load in a model of infection comprising human rhinovirus type 16 (HRV16) infected ALI cultured nasal epithelium (see Biological Example 7 and Figures 14 and 15); (iv) an apically administered formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with an anti-viral polynucleotide molecule (shRNA 1), has a potent effect in reducing viral load, and in improving epithelial cell/barrier integrity, as represented by the transepithelial electrical resistance (TEER), in a model of infection comprising human respiratory syncytial virus (RSV) A2 infected ALI cultured nasal epithelium (see Biological Example 8 and Figures 16 and 17); (v) intranasal administration of a formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with an anti-viral polynucleotide molecule (shRNA 1), has a potent effect in reducing viral load, reducing virus-induced inflammation, and protecting from virus-induced weight loss in an in vivo model comprising influenza (PR8) virus infected mice (see Biological Example 5, and Figures 8 to 10); and (vi) intranasal administration of a formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with an anti-viral polynucleotide molecule (shRNA 1), has a potent effect in reducing viral load, reducing virus-induced inflammation, and protecting from virus-induced weight loss in an in vivo model comprising human respiratory syncytial virus (RSV) A2 infected mice (see Biological Example 9 and Figures 18 to 20); (vii) subcutaneous administration of a formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with an anti-viral polynucleotide molecule (shRNA 1), significantly stimulates the innate immune response, as determined by the production of CXCL10, in non-infected mice, and moreover has a potent effect in reducing viral load, reducing virus-induced inflammation, and protecting from virus-induced weight loss in an in vivo model comprising influenza (PR8) virus infected mice (see Biological Example 10 and Figures 21 to 26); 12    (viii) subcutaneous administration of a formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with an anti-viral polynucleotide molecule (shRNA 1), has a potent adjuvant effect on subcutaneous vaccination with recombinant H1N1 (rH1N1) haemagglutinin (HA), as represented via an enhancement of the capacity of said rH1N1 HA vaccination to reduce viral load, reduce virus-induced inflammation, and protect against virus-induced weight loss in an in vivo model comprising influenza (PR8) virus infected mice (see Biological Example 11 and Figures 27 to 31); (ix) an apically administered formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with a polynucleotide molecule, particularly an siRNA, mRNA or DNA molecule, regulating or expressing a marker protein, effectively increases cell exposure to the polynucleotide molecule, as determined by modulated or increased expression of the marker protein, in a model comprising ALI cultured nasal epithelium or ALI cultured corneal epithelium (see Biological Examples 6, 12, and 14, and Figures 11,12, 32 to 35, 37, and 38); (x) the in vitro activity of formulations comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with an anti-viral polynucleotide molecule (shRNA 1), is not significantly influenced by buffer identity or shear mixing, as represented by reduction in viral load in a model of infection comprising influenza virus infected ALI cultured nasal epithelium (see Biological Example 13 and Figure 36); (xi) an apically administered formulation comprising a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, in combination with an anti-viral polynucleotide molecule (shRNA 1), has a potent effect in reducing human rhinovirus (HRV) induced release of the pro-inflammatory cytokines CXCL10, CXCL8, and CCL5, and in reducing viral load, in a model of infection comprising human rhinovirus type 16 (HRV16) infected ALI cultured bronchial epithelium from an asthma donor (see Biological Example 16, and Figures 47 to 50); (xii) formulation of a polynucleotide molecule, in particular a shRNA molecule, with a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non- ionic surfactant, in particular polysorbate 80, at specific concentrations, results in a formulation wherein the colloidal particle size of the colloidal particles containing the polynucleotide molecule is stabilised, as represented by a low polydispersity and consistent particle size (see Biophysical Example 1, and Figures 13 (A-D)); (xiii) formulation of a polynucleotide molecule, in particular a shRNA molecule, with surfactant components comprising caprylic acid or oleic acid as the fatty acid, and 13    polysorbate 80 or Brij 35 as the non-ionic surfactant, each result in a formulation in which colloidal particles are formed, and in which the particle size of said colloidal particles, which contain the polynucleotide molecule, is stabilised, as represented by a low polydispersity and consistent particle size (see Biophysical Examples 2 and 3, and Figures 40 to 45 (A-F)); and (xiv) formulations comprising a polynucleotide molecule, in particular shRNA 1, and a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non- ionic surfactant, in particular polysorbate 80, produce a stable colloidal emulsion in which colloidal particles are highly stable, and in which the stability of the polynucleotide molecule is increased, as represented by an increased melting temperature (see Biophysical Example 4, and Figure 46). Surfactant Component The aqueous liquid pharmaceutical formulations of the invention comprise a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant. Fatty Acid By way of definition, as used herein a “fatty acid” refers to a carboxylic acid molecule comprising a carboxylic acid group attached to an aliphatic hydrocarbon “tail”, which is typically between 4 and 24 carbon atoms in length. For example, the aliphatic hydrocarbon “tail” may be between 4 and 22, such as between 4 and 20, such as between 4 and 18, such as between 4 and 16, such as between 4 and 14, such as between 4 and 12, such as between 4 and 10, such as between 4 and 8, such as between 4 and 6, carbon atoms in length. Alternatively, the aliphatic hydrocarbon “tail” may be between 6 and 24, such as between 8 and 24, such as between 10 and 24, such as between 12 and 24, such as between 14 and 24, carbon atoms in length. For example, the aliphatic hydrocarbon “tail” may be between 6 and 22, such as between 6 and 20, such as between 8 and 20, such as between 8 and 18, such as between 10 and 18, carbon atoms in length. In an embodiment, the aliphatic hydrocarbon “tail” is between 4 and 6 carbon atoms in length i.e. the fatty acid is a short-chain fatty acid such as butyric acid (4 carbon atoms). Alternatively, the aliphatic hydrocarbon “tail” is between 6 and 12 carbon atoms in length i.e. the fatty acid is a medium-chain fatty acid such as caprylic acid (8 carbon atoms) and capric acid (10 carbon atoms). Alternatively, the aliphatic hydrocarbon “tail” is between 14 and 24 carbon atoms in length i.e. the fatty acid is a long-chain fatty acid such as oleic acid (18 carbon atoms), stearic acid (18 carbon atoms) and arachidic acid (20 carbon atoms). The aliphatic hydrocarbon “tail” may be saturated or unsaturated. If unsaturated, the aliphatic hydrocarbon “tail” may comprise, for example, one, two, three, four, five, six etc. C=C double bonds, in particular one or two, especially one C=C double bond. Fatty acids may be sub-categorised based on the length and degree of saturation of the aliphatic hydrocarbon “tail”. 14    Suitably, fatty acids have a molar mass of from about 100 g/mol to about 400 g/mol, such as from about 100 g/mol to about 350 g/mol, such as from about 120 g/mol to about 350 g/mol, such as from about 140 g/mol to about 350 g/mol, such as from about 140 g/mol to about 300 g/mol. Exemplary fatty acids may typically have a molar mass of from about 150 g/mol to about 400 g/mol, for example from about 200 g/mol to about 350 g/mol e.g. from about 200 g/mol to about 300 g/mol. Further exemplary fatty acids may typically have a molar mass of from about 140 g/mol to about 200 g/mol. They include but are not limited to arachidic acid, arachidonic acid, lauric acid, linoleic acid, linolenic acid, myristic acid, myristoleic acid, oleic acid, palmitic acid, palmitoleic acid, sapienic acid, stearic acid, and vaccenic acid. A further example is caprylic acid. In particular, the fatty acid is oleic acid. Alternatively, the fatty acid is caprylic acid. In one embodiment, the fatty acid is not linoleic acid. Suitably the fatty acid has a hydrophilic-lipophilic balance (HLB) in the range 1 - 4, for example in the range 1 – 3, e.g.1 - 2.5, e.g.1 – 2, e.g.1 - 1.5. As used herein, HLB is defined as 20 * (MHL / MHL+MOH) where MHL is the molecular weight of the hydrophilic portion of the molecule and MOH is the molecular weight of the lipophilic portion of the molecule. For fatty acids, the MHL is taken as represented by O with a molecular weight of 16. MOH is the balance of the molecular weight of the molecule. For example, oleic acid has an HLB value of 1 and caprylic acid has an HLB value of 2.2. Suitably the fatty acid has a Log P value in the range 2 – 8, such as caprylic acid, oleic acid, lauric acid and palmitic acid. For example, caprylic acid has a Log P value of 3.05 and oleic acid has a Log P value of 7.7. For example, lauric acid has a Log P value of 4.6 and palmitic acid has a Log P value of 7.15. As used herein, Log P refers to the logarithm of the partition coefficient P of a compound between two immiscible phases, typically octanol and water. This value is a measure of the compound's lipophilicity, indicating how much the compound prefers a lipid (fat-soluble) environment over a watery (aqueous) environment. A higher log P value suggests that the compound is more lipophilic, meaning it dissolves better in lipids or non-polar solvents than in water. Suitably the fatty acid has a critical micellar concentration (CMC) in the range 0.001 – 0.01 mM. For example, oleic acid has an CMC value of 0.006 mM. As used herein, CMC refers to the concentration of surfactants in a bulk phase above which micelles start to form spontaneously. Below the CMC, surfactants exist mainly as individual molecules dispersed in the solution. When the concentration reaches the CMC, these molecules begin to aggregate into micelles, which are spherical structures where the 15    hydrophobic (water-repelling) tails of the surfactant molecules are shielded from the water by the hydrophilic (water-attracting) heads. It would be understood by the skilled person that the HLB, LogP, and CMC values of fatty acids and non-ionic surfactants (as discussed further below) can be easily determined, or can be easily found, for example by consulting references including the Handbook of Pharmaceutical Excipients, 5th Edition (Rowe, Sheskey, and Owen), 2006. Suitably the aqueous liquid pharmaceutical formulation comprises a single fatty acid as part of the surfactant component. Alternatively, it comprises a mixture of e.g. of two (or more) fatty acids as part of the surfactant component. Non-Ionic Surfactant Exemplary non-ionic surfactants may typically have a molar mass of from about 100 g/mol to about 10000 g/mol, in particular from about 100 g/mol to about 2000 g/mol. Exemplary non- ionic surfactants typically comprise one or more polyoxyalkylene moieties e.g. polyoxyethylene and/or polyoxypropylene moieties. Exemplary non-ionic surfactants include polyoxyalkylenes, particularly poloxamers, such as poloxamer 188, poloxamer 407, poloxamer 171, and poloxamer 185. Further exemplary non-ionic surfactants include alkyl ethers of polyethylene glycol, such as those known under the brand names Brij 35 (polyoxyethylene (23) lauryl ether), Brij 52 (polyoxyethylene (20) cetyl ether), Brij 93 (polyoxyethylene (2) oleyl ether), Brij 97 (polyoxyethylene (10) oleyl ether), Brij L4 (polyoxyethylene (4) lauryl ether), Brij 30 (polyoxyethylene (4) lauryl ether), and Brij 78 (polyoxyethylene (20) stearyl ether). Additional exemplary non-ionic surfactants include alkylphenyl ethers of polyethylene glycol, such as that known under the brand name Triton X-100. Particular exemplary non-ionic surfactants include fatty acid esters, such as fatty acid esters of polyols. Such fatty acid esters may comprise one or more e.g. one, two or three fatty acid chains e.g. one fatty acid chain. Specific examples include polyoxyethylene sorbitan fatty acid esters. In particular, the non-ionic surfactant is a polyoxyethylene sorbitan fatty acid ester. Suitable polyoxyethylene sorbitan fatty acid esters include polysorbate 80 (e.g. Tween 80), polysorbate 120, polysorbate 85, polysorbate 65, polysorbate 60, polysorbate 40, and polysorbate 20, in particular polysorbate 80. In an embodiment, the non-ionic surfactant is not polysorbate 60. In an embodiment, the non-ionic surfactant is not polysorbate 85. Suitably the non-ionic surfactant has a hydrophilic-lipophilic balance (HLB) value of 10 or more, for example in the range 10 – 20, such as polysorbates and other highly ethoxylated non-ionic surfactants. For example, polysorbate 20 has a HLB value of 16.7. For example, polysorbate 40 has a HLB value of 15.6. For example, polysorbate 65 has a HLB value of 10.5. For example, polysorbate 120 has a HLB value of 14.9. For example, polysorbate 80 16    has a HLB value of 15.0. For example, Brij 35 has a HLB value of 16.9. For example, Brij 97 has a HLB value of 12.4. Suitably the non-ionic surfactant has a Log P value in the range 1 – 5, such as polysorbate 20. For example, polysorbate 80 has a Log P value of 4.7. Suitably the Log P value of the non- ionic surfactant is lower than that of the fatty acid.in the surfactant component. Suitably the non-ionic surfactant has a critical micellar concentration (CMC) in the range 0.01 – 0.5 mM, such as polysorbate 20 and Brij 35. For example, polysorbate 20 has a CMC value of 0.06 mM. For example, polysorbate 80 has a CMC value of 0.012 mM. For example, Brij 35 has a CMC value of 0.09 mM. Suitably the CMC of the non-ionic surfactant is greater than that of the fatty acid.in the surfactant component. Suitably the aqueous liquid pharmaceutical formulation comprises a single non-ionic surfactant as part of the surfactant component. Alternatively, it comprises a mixture of e.g. of two (or more) non-ionic surfactants as part of the surfactant component. Other Aspects Suitably, the surfactant component is selected from the group consisting of mixtures of (a) oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (b) lauric acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (c) linoleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (d) linolenic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (e) palmitic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (f) stearic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (g) oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyalkylene, such as a poloxamer, (h) oleic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol, and (i) oleic acid or a pharmaceutically acceptable salt thereof and an alkylphenyl ether of polyethylene glycol. Most suitably the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular a polyoxyethylene sorbitan fatty acid ester selected from polysorbate 80 and polysorbate 20. In particular the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof, especially oleic acid, and polysorbate 80. Alternatively, the surfactant component is suitably a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular a polyoxyethylene sorbitan fatty acid ester selected from polysorbate 80 and polysorbate 20. In particular the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof, especially caprylic acid, and polysorbate 80. 17    Alternatively, the surfactant component is suitably a mixture of oleic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol, such as those known under the brand names Brij 35, Brij 52, Brij 93, Brij 97, Brij L4, Brij 30, and Brij 78. In particular the surfactant component is a mixture of oleic acid, or a pharmaceutically acceptable salt thereof, especially oleic acid, and Brij 35. Alternatively, the surfactant component is suitably a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol, such as those known under the brand names Brij 35, Brij 52, Brij 93, Brij 97, Brij L4, Brij 30, and Brij 78. In particular the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof, especially caprylic acid, and Brij 35. In an embodiment, the surfactant component is not a mixture of linoleic acid and polysorbate 60. Pharmaceutically acceptable salt forms of fatty acid that may be employed include sodium, potassium, and ammonium salts, and in particular the sodium salt. Suitably the fatty acid is used as, i.e. is in the form of, the free acid. The aqueous liquid pharmaceutical formulations of the present invention should suitably form a stable colloidal emulsion (i.e. an oil in water emulsion) such as a stable colloidal nanoemulsion. Typically, the stable colloidal emulsion will comprise stable colloidal particles (i.e. particles comprising the oil phase in the oil in water emulsion) with an average particle size of between about 10 and about 1000 nm such as about 50 and about 1000 nm, such as between about 50 and about 750 nm, such as between about 50 and about 500 nm, for example between about 50 and about 400 nm, for example between about 50 and about 300 nm, for example between about 50 and about 100 nm, or between about 100 and about 300 nm, or about 100 and about 250 nm, or between about 250 and about 500 nm. Thus, for example, in a preferred embodiment the average particle size is between about 100 and about 300 nm, such as between about 100 and 200 nm (see Biophysical Example 1). Thus suitably the particles of the colloidal emulsion are droplets formed of the surfactant component and containing the polynucleotide molecule. The aforesaid particle size means hydrodynamic diameter (Z-average size) which can be measured as described in Biophysical Example 1. Such a formulation is suitably achieved upon use of a surfactant component which is a mixture of a fatty acid, or a pharmaceutically acceptable salt thereof, and a non-ionic surfactant. This mixture forms the oil phase of the stable colloidal emulsion. Such a formulation may more suitably be achieved when the fatty acid is present at a concentration, as provided for in the present invention below, which is at or above (but ideally close to) the critical micellar concentration (CMC) of said fatty acid. Such a formulation may more suitably be achieved when the non-ionic surfactant is water-miscible, and/or has a HLB value of 10 or more, for example has a HLB in the range 10-20, and/or when present at a concentration provided for 18    in the present invention below. Physical measurements are suitably made at a temperature of 23 oC and a pressure of 1 standard atmosphere. Typically, the surfactant component may be present in the formulation at a concentration (meaning the total concentration of the surfactants of the surfactant component) of 0.2 – 30000 µg/mL, for example 1 – 30000 µg/mL, for example 1 – 20000 µg/mL, for example 5 – 20000 µg/mL, for example 5 – 15000 µg/mL, for example 5 – 10000 µg/mL, for example 5-5000 µg/mL. Suitably, the surfactant component is present in the formulation at a concentration of 1 – 3000 µg/mL, for example 1 – 2000 µg/mL, for example 5 – 2000 µg/mL, for example 5 – 1500 µg/mL, for example 5 – 1000 µg/mL, for example 5-500 µg/mL. In one embodiment, the surfactant component is present at a concentration of 50-200 µg/mL, for example 75-150 µg/mL, for example 90 – 120 µg/mL, or about 100 µg/mL. In an alternative embodiment, the surfactant component is present at a concentration of 500-2000 µg/mL, for example 750-1500 µg/mL, for example 900 – 1200 µg/mL, or about 1000 µg/mL. Suitably, the fatty acid may be present in the formulation at a concentration of 0.2 – 30000 µg/mL, for example 1 – 30000 µg/mL, for example 1 – 20000 µg/mL, for example 5 – 10000 µg/mL and the non-ionic surfactant may be present in the formulation at a concentration of 0.2 – 20000 µg/mL, for example 1 – 20000 µg/mL, for example 1 – 15000 µg/mL, for example 5 – 5000 ug/mL. More suitably, the fatty acid may be present in the formulation at a concentration of 10 – 100 ug/mL, for example 20 – 80 µg/mL, for example 25 – 75 µg/mL, for example 40 – 60 µg/mL, or about 50 µg/mL, and the non-ionic surfactant may be present in the formulation at a concentration of 10 – 100 ug/mL, for example 20 – 80 µg/mL, for example 25 – 75 µg/mL, for example 30 – 60 µg/mL, for example 40 – 50 µg/mL. Alternatively, in another suitable embodiment, the fatty acid may be present in the formulation at a concentration of 100 – 1000 ug/mL, for example 200 – 800 µg/mL, for example 250 – 750 µg/mL, for example 400 – 600 µg/mL, or about 500 µg/mL, and the non-ionic surfactant may be present in the formulation at a concentration of 100 – 1000 ug/mL, for example 200 – 800 µg/mL, for example 250 – 750 µg/mL, for example 300 – 600 µg/mL, for example 400 – 500 µg/mL. Typically, the surfactant component may be present in the formulation at a concentration of 0.00002% (w/w) – 3% (w/w), for example 0.0001% (w/w) – 3% (w/w), for example 0.0001% (w/w) – 2% (w/w), for example 0.0005% (w/w) – 2% (w/w), for example 0.0005% (w/w) – 1.5% (w/w), for example 0.0005% (w/w) – 1% (w/w), for example 0.0005% (w/w) – 0.5% (w/w), wherein the % by weight is with respect to the total weight of the formulation. Suitably, the surfactant component is present in the formulation at a concentration of 0.0001% (w/w) – 0.3% (w/w), for example 0.0001% (w/w) – 0.2% (w/w), for example 0.0005% (w/w) – 0.2% (w/w), for example 0.0005% (w/w) – 0.15% (w/w), for example 0.0005% (w/w) – 0.1% (w/w), for example 0.0005% (w/w) – 0.05% (w/w), wherein the % by weight is with respect to the total 19    weight of the formulation. In one embodiment, the surfactant component is present at a concentration of 0.005% (w/w) – 0.02% (w/w), for example 0.0075% (w/w) – 0.015% (w/w), for example 0.009% (w/w) – 0.012% (w/w), or about 0.01% (w/w), wherein the % by weight is with respect to the total weight of the formulation. In one embodiment, the surfactant component is present at a concentration of 0.05% (w/w) – 0.2% (w/w), for example 0.075% (w/w) – 0.15% (w/w), for example 0.09% (w/w) – 0.12% (w/w), or about 0.1% (w/w), wherein the % by weight Is with respect to the total weight of the formulation. Suitably, the fatty acid may be present in the formulation at a concentration of 0.00002% (w/w) – 3% (w/w), for example 0.0001% (w/w) – 3% (w/w), for example 0.0001% (w/w) – 2% (w/w), for example 0.0005% (w/w) – 1% (w/w), and the non-ionic surfactant may be present in the formulation at a concentration of 0.00002% (w/w) – 2% (w/w), for example 0.0001% (w/w) – 2% (w/w), for example 0.0001% (w/w) – 1.5% (w/w), for example 0.0005% (w/w) – 0.5% (w/w), wherein the % by weight is with respect to the total weight of the formulation. More suitably, the fatty acid may be present in the formulation at a concentration of 0.001% (w/w) – 0.01% (w/w), for example 0.002% (w/w) – 0.008% (w/w), for example 0.0025% (w/w) – 0.0075% (w/w), for example 0.004% (w/w) – 0.006% (w/w), or about 0.005% (w/w), and the non-ionic surfactant may be present in the formulation at a concentration of 0.001% (w/w) – 0.01% (w/w), for example 0.002% (w/w) – 0.008% (w/w), for example 0.0025% (w/w) – 0.0075% (w/w), for example 0.003% (w/w) – 0.006% (w/w), for example 0.004% (w/w) – 0.005% (w/w), wherein the % by weight is with respect to the total weight of the formulation. Alternatively, in another suitable embodiment, the fatty acid may be present in the formulation at a concentration of 0.01% (w/w) – 0.1% (w/w), for example 0.02% (w/w) – 0.08% (w/w), for example 0.025% (w/w) – 0.075% (w/w), for example 0.04% (w/w) – 0.06% (w/w), or about 0.05% (w/w), and the non-ionic surfactant may be present in the formulation at a concentration of 0.01% (w/w) – 0.1% (w/w), for example 0.02% (w/w) – 0.08% (w/w), for example 0.025% (w/w) – 0.075% (w/w), for example 0.03% (w/w) – 0.06% (w/w), for example 0.04% (w/w) – 0.05% (w/w), wherein the % by weight is with respect to the total weight of the formulation. Suitably, the ratio of the amount of fatty acid or a pharmaceutically acceptable salt thereof to non-ionic surfactant, for example wherein each is measured in µg/mL, is between about 5:1 and about 1:5, for example between about 5:1 and about 1:2, for example between about 4:1 and about 1:2, for example between about 2:1 and about 1:2. More suitably, the ratio of the amount of fatty acid or a pharmaceutically acceptable salt thereof to non-ionic surfactant, for example wherein each is measured in µg/mL, is between about 3:2 and about 2:3, for example between about 6:5 and about 1:1, e.g. about 10:9 or about 11:10. As described above, and without wishing to be bound by theory, suitably the surfactant component forms the oil phase of an oil in water emulsion when dispersed in the aqueous formulation. For example, suitably the surfactant component forms the oil phase of said 20    emulsion in which the fatty acid (such as oleic acid) forms the core (or internal phase) of the particles and the non-ionic surfactant (such as polysorbate 80) stabilises the interface between the oil phase and the aqueous phase. Suitably the polynucleotide molecule is present within (i.e. is a component of) the stable colloidal particles, and for example is present around the core of the particles, and for example is present around the core and at the interface between the core of the particles and the non-ionic surfactant which stabilises the interface between the oil phase and the aqueous phase. A graphic representation of different types of particles formed by formulations comprising a surfactant component, which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, is shown in Figure 51. In accordance with the description of the above paragraph and, again without wishing to be bound by theory, the present inventors consider that formulations of the present invention suitably form a stable colloidal emulsion (e.g. a stable colloidal or lipid nanoemulsion), as shown in the left most of the three graphics in Figure 51. For images of the particles according to the present invention as viewed under a Transmission Electron Microscope see Figures 45 (A)-(F) discussed further below. The particles of the emulsion (i.e. stable colloidal particles containing polynucleotide) may spontaneously self-assemble when present in the aqueous formulation. Suitably, the emulsion having particles of suitable size may be formed by mixing the ingredients. Mixing may be performed at higher or lower shear depending on the target size of particles. For example, mixing speeds of 100 – 7000 rpm, or preferably of 500 – 3000 rpm, may be employed to yield emulsions with particle size in the range 100 to 300 nM (see Biophysical Examples 1 to 3). Polynucleotide Molecule The aqueous liquid pharmaceutical formulations of the present invention comprise a polynucleotide molecule. Suitably the aqueous liquid pharmaceutical formulation comprises a single polynucleotide molecule. However, it will be understood that the aqueous liquid formulations of the present invention may comprise more than one, for example two, three, four, five, six, seven, eight, nine, or ten etc. different polynucleotide molecules. As used herein, the term “polynucleotide molecule” refers to a molecule comprising two or more nucleotides. Therefore, a polynucleotide molecule may suitably comprise from two to about 100 nucleotides, such as from two to about 90 nucleotides, such as from two to about 80 nucleotides, such as from two to about 70 nucleotides, such as from two to about 60 nucleotides, or such as from two to about 50 nucleotides. In one embodiment, the polynucleotide molecule suitably comprises from about five to about 50 nucleotides, such as from about five to about 40 nucleotides, such as from five to about 30 nucleotides or from about 20 to about 40 nucleotides, such as from 10 to about 30 nucleotides or from about 20 21    to about 35 nucleotides, such as from 10 to about 20 nucleotides or from about 20 to about 30 nucleotides. Alternatively, a polynucleotide molecule may suitably comprise more than about 100 nucleotides. Therefore, in one embodiment, the polynucleotide molecule suitably comprises more than about 100 nucleotides, such as more than about 200 nucleotides, such as more than about 400 nucleotides, such as more than about 500 nucleotides, such as more than about 750 nucleotides, such as more than about 1000 nucleotides, such as more than about 1250 nucleotides, such as more than about 1500 nucleotides, such as more than about 2000 nucleotides, such as more than about 2500 nucleotides, such as more than about 5000 nucleotides. In one embodiment, the polynucleotide molecule suitably comprises from about 100 to about 20000, such as from about 100 to about 10000 nucleotides, such as from about 200 to about 8000 nucleotides or from about 500 to about 10000 nucleotides, such as from about 2500 to about 1000 nucleotides or from about 500 to about 7500 nucleotides, such as from about 1000 to about 5000 nucleotides, such as from about 2000 to about 5000 nucleotides, such as from about 2000 nucleotides to about 4000 nucleotides. As used herein, the term “nucleotide” refers to monomeric organic molecules comprising a nitrogenous base, in particular a primary or canonical nitrogenous base such as adenine, cytosine, guanine, thymine, or uracil, a sugar molecule, in particular a pentose sugar, such as ribose or deoxyribose, and a phosphate group or an analogue thereof, such as a thiophosphate group. Suitable alternative nitrogenous bases include modified purine nitrogenous bases such as 7-methyl guanine, hypoxanthine, and xanthine, modified pyrimidine nitrogenous bases such as 5,6-dihydrouracil, 5-methylcytosine and 5’hydroxymethylcytosine, and artificial or synthetic nitrogenous bases. Suitable nucleotides therefore include ribonucleotides and deoxyribonucleotides. Suitably polynucleotide molecules therefore include ribonucleic acid (RNA) molecules, and deoxyribonucleic acids (DNA) molecules. As used herein, the term “nucleotide” may further refer to peptide nucleotides, threose nucleotides, glycol nucleotides, serinol nucleotides or locked nucleotides. Suitably polynucleotides molecules therefore include peptide nucleic acid (PNA) molecules, threose nucleic acid (TNA) molecules, glycol nucleic acid (GNA) molecules, serinol nucleic acid (SNA) molecules and locked nucleic acid (LNA) molecules. Suitably polynucleotides molecules further include hybrid polynucleotide molecules comprising one or more different types of nucleotide, in particular from those nucleotides described above. A particularly suitable hybrid polynucleotide molecule is a DNA:RNA hybrid polynucleotide molecule i.e., a polynucleotide molecule comprising ribonucleotides and deoxyribonucleotides. In particular, the polynucleotide molecule is an RNA molecule or a DNA molecule. 22    The polynucleotide molecule may be of any origin, e.g. viral, bacterial, archae-bacterial, fungal, ribosomal, eukaryotic or prokaryotic, and may be from any organism. The polynucleotide molecule may arise from any biological sample and any organ, tissue, cell, or sub-cellular compartment. The polynucleotide molecule may be pre-treated before use, for example isolated, purified and/or modified. The polynucleotide molecule, or any number of individual nucleotides within the polynucleotide molecule, may be artificial or synthetic. The polynucleotide molecule may consist of a single strand, i.e. be single-stranded, e.g. of RNA, or alternatively the polynucleotide molecule may consist of two strands, i.e. be double- stranded, e.g. of dsRNA. In an embodiment, a polynucleotide molecule may consist of three strands i.e. be triple-stranded, e.g. triple-stranded DNA. Suitably, the single-stranded polynucleotide molecule consists of a sense strand. Alternatively, the single-stranded polynucleotide molecule consists of an antisense strand. In one embodiment, the polynucleotide molecule comprises a double-stranded region. For example, a double-stranded polynucleotide molecule may form a double-stranded duplex. Alternatively, two single-stranded polynucleotide molecules may hybridize, that is bind non- covalently, to form a double-stranded polynucleotide molecule. Hybridization may occur between two complementary or partially complementary sequences and may occur between polynucleotide molecules of the same, DNA:DNA, or different, DNA:RNA, types. Alternatively, a single-stranded polynucleotide molecule may comprise a first region which hybridizes with a second region of the single-stranded polynucleotide molecule to form an intramolecular double-stranded region, e.g., duplex. In one embodiment, the polynucleotide molecule comprises an intramolecular structure. For example, the polynucleotide molecule may comprise a helix, a bulge (separation of a double helical tract on one strand), an internal loop (separation of a double helical tract on both strands), a stem-loop or hairpin, a tetraloop (four base pair hairpin), pseudoknot, or junction. In one embodiment, the polynucleotide molecule comprises at least one, e.g., one or two, 5’ or 3’ monophosphate(s), and/or at least one, e.g. one or two, 5’ or 3’ diphosphate(s), and/or at least one, e.g. one or two, 5’ or 3’ triphosphate(s), and/or at least one, e.g. one or two, 5’ or 3’ hydroxyl (OH) groups. In particular, the 5’ or 3’ monophosphate(s) and/or 5’ or 3’ diphosphate(s) and/or 5’ or 3’ triphosphate(s) and/or 5’ or 3’ hydroxyl (OH) groups are located at the 5’ and/or 3’ terminus or termini of the single-stranded or double-stranded polynucleotide molecule. For example, in one embodiment the polynucleotide molecule is polyinosinic-polycytidylic acid (poly I:C), which is a synthetic dsRNA typically between 100-10000, such as between 200- 8000, for example between 300-6000 base pairs in length (see, for example, dsRNA 2 in the Examples below and US9682096B2, which is incorporated herein by reference). 23    In one embodiment, the polynucleotide molecule is an RNA molecule. Suitably, the polynucleotide molecule, which is an RNA molecule, is an mRNA molecule, miRNA molecule, shRNA molecule, or siRNA molecule. In one embodiment, the polynucleotide molecule is an mRNA molecule. In particular, the polynucleotide molecule is a single-stranded RNA molecule which may encode one or more proteins. Suitably, the mRNA molecule, is capable of being translated. In one embodiment, the mRNA molecule comprises from about 100 to about 10000 nucleotides, such as from about 200 to about 8000 nucleotides, such as from about 500 to about 7500 nucleotides, such as from about 1000 to about 5000 nucleotides. In one embodiment, the mRNA molecule comprises one or more coding regions, which may optionally be stabilised by internal base pairs. The coding regions may further comprise regulatory sequences, exonic splicing enhancers or exonic splicing silencers. Suitably, the mRNA molecule, comprises a 5’ untranslated region (5’ UTR) and/or a 3’ untranslated region (3’ UTR). Suitably, the mRNA molecule comprises a 3’ tail of adenine nucleotides, known as a polyA tail. Suitably, the polyA tail comprises 50 or more, such as 100 or more adenine nucleotides. Suitably, the polynucleotide molecule, for example mRNA molecule, comprises a 5’ cap, comprising a terminal 7-methylguanosine residue linked via 5’-5’ triphosphate bond to the first 5’ nucleotide. In one embodiment, the mRNA molecule is a circularised mRNA molecule, for example due to a protein-mediated intramolecular interaction between the 5’ cap and polyA tail of an mRNA molecule. In one embodiment, the polynucleotide molecule is a miRNA molecule. In particular, the polynucleotide molecule is a single-stranded RNA molecule which does not encode a protein and which functions in RNA silencing and post-transcriptional regulation of gene expression. In one embodiment, the miRNA molecule comprises from about 20 to about 25 nucleotides. In particular the miRNA molecule comprises 20, 21, 22, 23, 24 or 25 nucleotides. In one embodiment, the polynucleotide molecule is an shRNA molecule. In particular, the polynucleotide molecule is a single-stranded RNA molecule wherein a first region hybridizes with a second region of the single-stranded RNA molecule to form an intramolecular double- stranded region, e.g., duplex. Said hybridization will create a hairpin structure, including a hairpin loop, within the RNA molecule. In one embodiment, the shRNA comprises from about 10 to about 70 nucleotides, for example from about 20 to about 70 nucleotides, for example from about 35 to about 70 nucleotides or from about 25 to about 35 nucleotides. In particular the shRNA molecule comprises 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 nucleotides. In one embodiment, the double-stranded region, or duplex, comprises 30 base pairs or less, such as 25 base pairs or less, such as 20 base pairs or less, such as 18 base pairs or less, 24    such as 16 base pairs or less, such as 14 base pairs or less, such as 12 base pairs or less, such as 10 base pairs or less, such as eight base pairs or less, such as six base pairs or less, such as four base pairs or less. In one embodiment, the double-stranded region, or duplex, comprises from about two to about 30 base pairs, such as from about two to about 25 base pairs, such as from about four to about 20 base pairs, such as from about 4 to about 18 base pairs, such as from about six to about 18 base pairs, such as from about eight to about 18 base pairs, such as from about 10 to about 18 base pairs. In one embodiment, the double-stranded region comprises one or more mispaired bases, according to Watson-Crick base pairing. For example, the double-stranded region may comprise one to 10 mispaired bases, such as one to eight mispaired bases, such as one to six mispaired bases, in particular one, two, three, four, five, or six mispaired bases. Suitably, the first region and second region, which hybridize to form an intramolecular double- stranded region, e.g., duplex, are each 20 nucleotides or less, for example 19 nucleotides or less, such as 18 nucleotides or less, in length. Suitably, the first and second region are each between about five and about 20 nucleotides, for example between about five and about 18 nucleotides, such as between about eight and about 18 nucleotides, such as between about 10 and about 18 nucleotides in length. Suitably, the first region and second region, which hybridize to form an intramolecular double- stranded region, e.g., duplex, are substantially complementary to each other, for example are at least about 80% complementary, in particular at least about 90% complementary, or most preferably 100% complementary. The hairpin structure may comprise a hairpin loop. Suitably, the length the hairpin loop is zero, or one or more, nucleotides in length. For example, the hairpin loop is two or more, four or more, five or more, eight or more, 10 or more, 15 or more, 20 or more, 40 or more, 50 or more, or 100 or more, nucleotides in length. In particular, the hairpin loop is from about one to about 20, such as from about one to about 15, such as from about two to about 10, such as from about four to about 10 nucleotides in length. In one suitable embodiment, the shRNA molecule comprises a blunt end. A blunt end refers to a polynucleotide molecule, where at least one strand of the duplex does not overhang, for example a 3’ dinucleotide overhang, such that the 5’ and 3’ strand end together. Alternatively, the shRNA molecule may comprise a 3’ overhang or a 5’ overhang. In one embodiment, the shRNA molecule comprises at least one 5’ triphosphate or at least one 5’ diphosphate. In particular, the 5’ triphosphate or 5’ diphosphate is located at the 5’ terminus of the shRNA molecule. Suitably, the shRNA molecule comprises one 5’ triphosphate or one 5’ diphosphate, in particular wherein the 5’ triphosphate or 5’ diphosphate is located at the 5’ terminus. 25    In one embodiment, the shRNA molecule, which is a single-stranded RNA molecule which forms a hairpin structure comprising an intramolecular double-stranded region and a hairpin loop, comprises a blunt end and a 5’ triphosphate or 5’ diphosphate located at the 5’ terminus, wherein the double-stranded region is between about 10 and about 18 nucleotides in length. The shRNA molecule of this embodiment may comprise one or more sugar-modified nucleotides, which each comprise a 2’ OH modification, and/or one or more backbone- modified nucleotides and/or one or more base-modified nucleotides. In an embodiment, such an shRNA molecule is capable of inducing an interferon response in a vertebrate cell. In one embodiment, the shRNA molecule, which is a single-stranded RNA molecule which forms a hairpin structure comprising an intramolecular double-stranded region and a hairpin loop, comprises, consists of, or consists essentially of a sequence disclosed in WO2019/246450A1, which is incorporated herein by reference. Specifically, the shRNA molecule may comprise, consist of, or consist essentially of SEQ ID NO.1. Alternatively, the shRNA molecule comprises, consists of, or consists essentially of a variant of SEQ ID NO.1, wherein a “variant” as used herein refers to a sequence having for example at least about 75% identity, for example at least about 80% identity, for example at least about 85% identity, in particular at least about 90% identity, such as at least about 95%, 98% or 99% identity to the associated reference sequence over their entire lengths. Alternatively, in one embodiment, the shRNA molecule, which is a single-stranded RNA molecule which forms a hairpin structure comprising an intramolecular double-stranded region and a hairpin loop, comprises, consists of, or consists essentially of SEQ ID NO. 2. Alternatively, the shRNA molecule comprises, consists of, or consists essentially of a variant of SEQ ID NO.2. In one embodiment, the shRNA molecule, which is a single-stranded RNA molecule which forms a hairpin structure comprising an intramolecular double-stranded region and a hairpin loop, comprises, consists of, or consists essentially of a sequence disclosed in US2023/0159923A1, which is incorporated herein by reference. In particular, the shRNA molecule may comprise, consist of, or consist essentially of SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO.5, SEQ ID NO.6, SEQ ID NO.7, SEQ ID NO.8, SEQ ID NO.9 or SEQ ID NO. 10. Alternatively, the shRNA molecule comprises, consists of, or consists essentially of a variant of SEQ ID NO.3, SEQ ID NO.4, SEQ ID NO.5, SEQ ID NO.6, SEQ ID NO.7, SEQ ID NO.8, SEQ ID NO.9 or SEQ ID NO.10. The molecule shRNA 1 used in the Examples below is an shRNA molecule of SEQ ID NO.1 comprising a blunt end and a 5’ diphosphate at the 5’ terminus. Thus, in an embodiment, the shRNA molecule is an shRNA molecule having the sequence of SEQ ID NO.1 and comprising a blunt end and a 5’ diphosphate at the 5’ terminus. It will be understood that an shRNA molecule of SEQ ID NO.1 may also comprise a 5’ triphosphate at the 5’ terminus. 26    The molecule shRNA 2 is an shRNA molecule of SEQ ID NO.2 comprising a blunt end and a 5’ diphosphate at the 5’ terminus. Thus, in an embodiment, the shRNA molecule is an shRNA molecule having the sequence of SEQ ID NO. 2 and comprising a blunt end and a 5’ diphosphate at the 5’ terminus. It will be understood that an shRNA molecule of SEQ ID NO. 2 may also comprise a 5’ triphosphate at the 5’ terminus. In one embodiment, the polynucleotide molecule is an siRNA molecule. In particular, the polynucleotide molecule is a double-stranded RNA molecule which does not encode a protein and which functions in RNA silencing and post-transcriptional regulation of gene expression. In one embodiment, the siRNA molecule comprises from about 20 to about 60 nucleotides, for example from about 30 to about 60 nucleotides, such as from about 40 to about 50 nucleotides. In particular the siRNA molecule comprises 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, or 60 nucleotides. In one embodiment, the siRNA molecule comprises a double-stranded region, or duplex, comprising 30 base pairs or less, such as 25 base pairs or less, such as 20 base pairs or less. In one embodiment, the double-stranded region, or duplex, comprises from about 10 to about 30 base pairs, such as from about 15 to about 30 base pairs, such as from about 20 to about 30 base pairs, such as from about 20 to about 25 base pairs or from about 25 base pairs to about 30 base pairs. In one embodiment, the double-stranded region comprises one or more mispaired bases, according to Watson-Crick base pairing. For example, the double-stranded region may comprise one to 10 mispaired bases, such as one to eight mispaired bases, such as one to six mispaired bases, in particular one, two, three, four, five, or six mispaired bases. Suitably, the siRNA molecule comprises one or more, e.g., two, 3’ overhangs. Alternatively, the siRNA molecule may comprise one or more, e.g., two 5’ overhangs. Alternatively, the siRNA molecule may comprise one or more, e.g., one, 3’ overhang and/or one or more, e.g., one, 5’ overhang. Alternatively, the siRNA molecule may comprise one or more blunt ends. In one embodiment, the siRNA molecule comprises at least one, e.g., two, 5’ monophosphate(s), or at least one, e.g., two, 5’ diphosphate(s), or at least one, e.g., two, 5’ triphosphate(s). In particular, the 5’ monophosphate(s) and/or 5’ diphosphate(s) and/or 5’ triphosphate(s) are located at the 5’ termini of the siRNA molecule. Suitably, the siRNA molecule comprises at least one, e.g., two, 3’ OH (hydroxyl) groups. In particular, the 3’ OH group(s) are located at the 3’ termini of the siRNA molecule. In one embodiment, the siRNA molecule, which is a double-stranded RNA molecule, comprises the sequence of SEQ ID NO.11 or a variant thereof. In particular, one strand of the double-stranded siRNA molecule comprises, consists of, or consists essentially of the sequence of SEQ ID NO.11, or a variant thereof (see Ren et al.2019, which is incorporated herein by reference). 27    In one embodiment, the polynucleotide molecule is a guide RNA (gRNA) molecule, which is a polynucleotide molecule comprising crispr RNA (crRNA), which is a nucleotide sequence, typically between 15 and 20 nucleotides in length which is complementary to a host target DNA, and tracr RNA (trRNA) which enables binding to a Cas nuclease. Suitably, the gRNA molecule is a component of the CRISPR-Cas9 gene editing technology. In one embodiment, the polynucleotide molecule is a DNA molecule. Suitably the DNA molecule is a genomic DNA (gDNA) molecule, for example a chromosomal DNA molecule or a mitochondrial DNA molecule, a complementary DNA (cDNA) molecule, or an extra- chromosomal DNA molecule, for example a plasmid DNA molecule. In one embodiment, the DNA molecule is a gDNA molecule. In one embodiment, the DNA molecule is a cDNA molecule. In one embodiment, the DNA molecule is an extra-chromosomal DNA molecule, in particular a plasmid DNA molecule. In one embodiment, the DNA molecule is a coding DNA molecule. For example, the DNA molecule comprises one or more coding regions, which may optionally be stabilised by internal base pairs. Accordingly, the DNA molecule may encode one or more protein. Suitably, the DNA molecule, which is a coding DNA molecule, is capable of being transcribed. Suitably, the DNA molecule, which is a coding DNA molecule, further comprises one or more promoter sequences, in particular wherein the one or more promoter sequence flanks the corresponding coding region at the 5’ end. Suitably, the DNA molecule, which is a coding DNA molecule, further comprises a termination sequence, in particular wherein the termination sequence flanks the one or more coding regions at the 3’ end. Suitably, a coding DNA molecule may further comprise any number of non-coding DNA elements, as described below. In one embodiment, the DNA molecule is a non-coding DNA molecule, that is the DNA molecule does not comprise a coding region and consequently does not encode a protein. Suitably, the non-coding DNA molecule is capable of being transcribed, for example to produce tRNA, miRNA, siRNA or ribosomal RNA. Alternatively, the non-coding DNA molecule may comprise a regulatory sequence that controls gene expression, may comprise a scaffold attachment region, may comprise a centromere, or may comprise a telomere. Alternatively, the non-coding DNA molecule may comprise non-functional elements such as an intron, a pseudogene, intergenic DNA, or transposons. Suitably, the non-coding DNA molecule may comprise a combination of the above-described elements. In one embodiment, the DNA molecule comprises from about 100 to about 20000 nucleotides, such as from about 100 to about 15000 nucleotides, such as from about 500 to about 15000 nucleotides, such as from about 500 to about 10000 nucleotides, such as from about 2500 to about 10000 nucleotides, such as from about 2500 to about 8000 nucleotides. In one embodiment, the polynucleotide molecule has the structure of Formula (I): 5’-Pz-(N)bN-3’-(E)y(E)-L-(E)(E)y’-5’-N(N)b’-3’ 28    wherein 5’-Pz-(N)bN-3’ represents the first nucleic acid sequence; 5’-N(N)b’-3’ represents the second nucleic acid sequence; P at each instance is independently a phosphate or analog thereof; z is 2 or 3; N is, at each instance, any nucleotide or modified nucleotide or analog or derivative there of; b and b’ are independently 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18; 5’-(E)y(E)-L-(E)(E)y’-3’ represents the connector element wherein E at each occurrence is independently any nucleotide, modified nucleotide, or abasic; y and y’ are independently 0-9, provided that y + y’ equals 0-8; L is a non-nucleotide segment having the structure
Figure imgf000031_0001
wherein X and X’ are independently O or S; Y and Y’ are independently OR”, SR”, or NRR’; V and V’ are independently O, S, or NRR’; q is 1-20; k is 1-20; t is 1-20; M selected from aliphatic, substituted aliphatic, aryl, substituted aryl, heteroalkyl, heterocyclyl or substituted heterocyclyl; W is any reactive group; and d is 0 or 1. 29    Suitably, the polynucleotide molecule has the structure of Formula (II): 5’-Pz-Nu-3’-(E)y(E)-L-(E)(E)y’-5’-Nu’-3’ wherein 5’-Pz-Nu-3’ represents the first nucleic acid sequence; 5’-Nu’-3’ represents the second nucleic acid sequence; P at each instance is independently a phosphate or analog thereof. z is 0, 1, 2, or 3; 5’-(E)y(E)-L-(E)(E)y’-3’ represents the connector element wherein E at each occurrence is independently any nucleotide, modified nucleotide, or abasic; y and y’ are independently 0-9, provided that y + y’ equals 0-8; L is a non-nucleotide segment having the structure
Figure imgf000032_0001
wherein X and X’ are independently O or S; Y and Y’ are independently OR”, SR”, or NRR’; V and V’ are independently O, S, or NRR’; q is 1-20; k is 1-20; t is 1-20; M selected from aliphatic, substituted aliphatic, aryl, substituted aryl, heteroalkyl, heterocyclyl or substituted heterocyclyl; W is any reactive group or conjugation group; and d is 0 or 1. In one embodiment, z is 2. Moreover, suitably, when z is 2, at least one P is a phosphate analogue. In particular, suitably, when z is 2, both P are phosphate analogues. Alternatively, 30    in one embodiment, z is 3. Furthermore, suitably, when z is 3, at least one P is a phosphate analogue. In particular, suitably, when z is 3, at least two P are phosphate analogues. For example, suitably, when z is 3, all P are phosphate analogues. In one embodiment, the phosphate analogue comprises the structure: X OH–P–Y Z wherein, Y is O or S, or CH-R where R = alkyl, ar-alkyl, heteroaryl, cycloalkylamines (e.g., piperazines), X is O or S, and Z is OH, SH, NHR’, wherein R’ is H, alkyl, aralkyl, and heteroaryl. In one embodiment, b and b’ are independently 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18; preferably 11, 12, 13, 14, 15, 16, 17, or 18; preferably 13, 14, 15, 16, 17, or 18. In one embodiment, b is 9 and b’ is less than, equal to, or greater than 9. In one embodiment, b is 10 and b’ is less than, equal to, or greater than 10. In one embodiment, b is 11 and b’ is less than, equal to, or greater than 11. In one embodiment, b is 12 and b’ is less than, equal to, or greater than 12. In one embodiment, b is 13 and b’ is less than, equal to, or greater than 13. In one embodiment, b is 14 and b’ is less than, equal to, or greater than 14. In one embodiment, b is 15 and b’ is less than, equal to, or greater than 15. In one embodiment, b is 16 and b’ is less than, equal to, or greater than 16. In one embodiment, b is 17 and b’ is less than, equal to, or greater than 17. In one embodiment, b is 18 and b’ is less than, equal to, or greater than 18. In one embodiment, b equals b’. In an alternative embodiment, b does not equal b’. In one embodiment, when b is less than b’, the polynucleotide molecule has a 3’-overhang. In an alternative embodiment, when b is greater than b’, the nucleic acid molecule has a 5′-overhang. In one embodiment, q is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20. For example, q is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15, and in particular q is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10. For example, in one embodiment, q is 1, 2, 3, 4, or 5, and in particular q is 1, or q is 2, or q is 3 ,or q is 4, or q is 5. In one embodiment, k and t are independently 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20, and in particular k and t are independently 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15. For example, in one embodiment, k and t are independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, and in particular k and t are independently 1, 2, 3, 4, or 5. In one embodiment, k is 1, or k is 2, or k is 3, or k is 4, or k is 5. In one embodiment, t is 1, or t is 2, or t is 3, or t is 4, or t is 5. In one embodiment, k and t are the same, and L is therefore 31    symmetrical. In an alternative embodiment, k and t are different, and L is therefore asymmetrical. In one embodiment, R, R’, and R” are independently selected from the group consisting of an alkyl, an amino alkyl, a carboxamido, polyethylene glycol (PEG), aralkyl, hetero-ar-alkyl, hetero-alkyl, substituted or unsubstituted cycloalkyl. In embodiments, R and R” groups may contain functionalities such as amino, hydroxy, azido, or thiol, that can be optionally used for the attachment of which can be used to link to a targeting molecule (Tm), such as a vitamin, peptide, antibody, or protein. In one embodiment, the R and R” group can be a peptide group. Peptide groups include a variety of enzymatically cleavable or non-cleavable peptides. The individual amino acids groups of the peptide could be natural or synthetic amino acids. In one embodiment, R and R” can be -CH2-O-CO-R1, where R1 = methyl, isopropyl, t-butyl, - (CH2)n-R2, wherein R2 is selected from aryl, aralkyl, heteroaryl, hetero-aralkyl, alkyl, an amino alkyl, a carboxamido, polyethylene glycol (PEG), hetero-alkyl, substituted or unsubstituted cycloalkyl. In one embodiment, the reactive group W may be additionally connected to alkyl, an amino alkyl, a carboxamido, polyethylene glycol (PEG), aralkyl, hetero-ar-alkyl, hetero-alkyl, substituted or unsubstituted cycloalkyl. In certain embodiments, R and R” groups may contain functionalities such as amino, hydroxy, azido, or thiol, that can be optionally used for the attachment to a targeting molecule (Tm) such as a vitamin, peptide, antibody, or protein. In one embodiment, M is selected from aliphatic, substituted aliphatic, aryl, substituted aryl, heteroralkyl, heterocyclyl or substituted heterocyclyl. The term “aliphatic group” or “aliphatic” refers to a non-aromatic moiety that may be saturated (e.g., single bond) or contain one or more units of unsaturation, e.g., double and/or triple bonds. An aliphatic group may be straight chained, branched or cyclic, contain carbon, hydrogen or, optionally, one or more heteroatoms and may be substituted or unsubstituted. In addition to aliphatic hydrocarbon groups, aliphatic groups include, for example, polyalkoxyalkyls, such as polyalkylene glycols, polyamines, and polyimines, for example. Such aliphatic groups may be further substituted. It is understood that aliphatic groups may include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, and substituted or unsubstituted cycloalkyl groups as described herein. The term “alkyl” is intended to include both branched and straight chain, substituted or unsubstituted saturated aliphatic hydrocarbon radicals/groups having the specified number of carbons. Preferred alkyl groups comprise about 1 to about 24 carbon atoms (“C1-C24”). Other preferred alkyl groups comprise at about 1 to about 8 carbon atoms (“C1-C8”) such as about 1 to about 6 carbon atoms (“C1-C6”), or such as about 1 to about 3 carbon atoms (“C1- 32    C3”). Examples of C1-C6 alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, n-pentyl, neopentyl and n-hexyl radicals. The term “alkenyl” refers to linear or branched radicals having at least one carbon-carbon double bond. Such radicals preferably contain from about two to about twenty-four carbon atoms (“C2-C24”). Other preferred alkenyl radicals are “lower alkenyl” radicals having two to about ten carbon atoms (“C2-C10”) such as ethenyl, allyl, propenyl, butenyl and 4- methylbutenyl. Preferred lower alkenyl radicals include 2 to about 6 carbon atoms (“C2-C6”). The terms “alkenyl”, and “lower alkenyl”, embrace radicals having “cis” and “trans” orientations, or alternatively, “E” and “Z” orientations. The term “alkynyl” refers to linear or branched radicals having at least one carbon-carbon triple bond. Such radicals preferably contain from about two to about twenty-four carbon atoms (“C2-C24”). Other preferred alkynyl radicals are “lower alkynyl” radicals having two to about ten carbon atoms such as propargyl, 1-propynyl, 2-propynyl, 1-butyne, 2-butynyl and 1-pentynyl. Preferred lower alkynyl radicals include 2 to about 6 carbon atoms (“C2-C6”). The term “cycloalkyl” refers to saturated carbocyclic radicals having three to about twelve carbon atoms (“C3-C12”). The term "cycloalkyl" embraces saturated carbocyclic radicals having three to about twelve carbon atoms. Examples of such radicals include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The term “alkoxy” refers to linear or branched oxy-containing radicals each having alkyl portions of one to about twenty-four carbon atoms or, preferably, one to about twelve carbon atoms. More preferred alkoxy radicals are “lower alkoxy” radicals having one to about ten carbon atoms and more preferably having one to about eight carbon atoms. Examples of such radicals include methoxy, ethoxy, propoxy, butoxy and tert-butoxy. The term “aryl”, alone or in combination, means an aromatic system containing one, two or three rings wherein such rings may be attached together in a pendent manner or may be fused. The term “aryl” embraces aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indane furanyl, quinazolinyl, pyridyl and biphenyl. The terms “heterocyclyl”, “heterocycle” “heterocyclic” or “heterocyclo” refer to saturated, partially unsaturated and unsaturated heteroatom-containing ring-shaped radicals, which can also be called “heterocyclyl”, “heterocycloalkenyl” and “heteroaryl” correspondingly, where the heteroatoms may be selected from nitrogen, sulfur and oxygen. Examples of saturated heterocyclyl radicals include saturated 3 to 6-membered heteromonocyclic group containing 1 to 4 nitrogen atoms (e.g., pyrrolidinyl, imidazolidinyl, piperidino, piperazinyl, etc.); saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms (e.g., morpholinyl, etc.); saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms (e.g., thiazolidinyl, etc.). Examples of partially unsaturated heterocyclyl radicals include dihydrothiophene, dihydropyran, 33    dihydrofuran and dihydrothiazole. Heterocyclyl radicals may include a pentavalent nitrogen, such as in tetrazolium and pyridinium radicals. The term “heterocycle” also embraces radicals where heterocyclyl radicals are fused with aryl or cycloalkyl radicals. Examples of such fused bicyclic radicals include benzofuran, benzothiophene, and the like. The term “heteroaryl” refers to unsaturated aromatic heterocyclyl radicals. Examples of heteroaryl radicals include unsaturated 3 to 6 membered heteromonocyclic group containing 1 to 4 nitrogen atoms, for example, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl (e.g., 4H-1,2,4-triazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3- triazolyl, etc.) tetrazolyl (e.g., 1H-tetrazolyl, 2H-tetrazolyl, etc.), etc.; unsaturated condensed heterocyclyl group containing 1 to 5 nitrogen atoms, for example, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl, tetrazolopyridazinyl (e.g., tetrazolo[1,5-b]pyridazinyl, etc.), etc.; unsaturated 3 to 6-membered heteromonocyclic group containing an oxygen atom, for example, pyranyl, furyl, etc.; unsaturated 3 to 6- membered heteromonocyclic group containing a sulfur atom, for example, thienyl, etc.; unsaturated 3- to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, for example, oxazolyl, isoxazolyl, oxadiazolyl (e.g., 1,2,4-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,5-oxadiazolyl, etc.) etc.; unsaturated condensed heterocyclyl group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms (e.g., benzoxazolyl, benzoxadiazolyl, etc.); unsaturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, for example, thiazolyl, thiadiazolyl (e.g., 1,2,4- thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, etc.) etc.; unsaturated condensed heterocyclyl group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms (e.g., benzothiazolyl, benzothiadiazolyl, etc.) and the like. The terms “aralkyl” or “arylalkyl” refer to aryl-substituted alkyl radicals such as benzyl, diphenylmethyl, triphenylmethyl, phenylethyl, and diphenylethyl. The term “aryloxy” refers to aryl radicals attached through an oxygen atom to other radicals. The term “alkylamino” denotes amino groups which are substituted with one or two alkyl radicals. Preferred alkylamino radicals have alkyl radicals having about one to about twenty carbon atoms or, preferably, one to about twelve carbon atoms. More preferred alkylamino radicals are “lower alkylamino” that have alkyl radicals having one to about ten carbon atoms. Most preferred are alkylamino radicals having lower alkyl radicals having one to about eight carbon atoms. Suitable lower alkylamino may be monosubstituted N-alkylamino or disubstituted N,N-alkylamino, such as N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-diethylamino or the like. The terms “halogen” or “halo” as used herein, refers to an atom selected from fluorine, chlorine, bromine and iodine. 34    The term ”substituted” refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent including, but not limited to: halo, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, thiol, alkylthio, arylthio, alkylthioalkyl, arylthioalkyl, alkylsulfonyl, alkylsulfonylalkyl, arylsulfonylalkyl, alkoxy, aryloxy, aralkoxy, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkoxycarbonyl, aryloxycarbonyl, haloalkyl, amino, trifluoromethyl, cyano, nitro, alkylamino, arylamino, alkylaminoalkyl, arylaminoalkyl, aminoalkylamino, hydroxy, alkoxyalkyl, carboxyalkyl, alkoxycarbonylalkyl, aminocarbonylalkyl, acyl, aralkoxycarbonyl, carboxylic acid, sulfonic acid, sulfonyl, phosphonic acid, aryl, heteroaryl, heterocyclic, and aliphatic. It is understood that the substituent may be further substituted. In one embodiment, the polynucleotide molecule may comprise, consist of, or consist essentially of SEQ ID NO.13 and SEQ ID NO. 14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L, as defined above. Alternatively, the polynucleotide molecule comprises, consists of, or consists essentially of a variant of SEQ ID NO.13 and/or a variant of SEQ ID NO.14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L, wherein a “variant” as used herein refers to a sequence having for example at least about 75% identity, for example at least about 80% identity, for example at least about 85% identity, in particular at least about 90% identity, such as at least about 95%, 98% or 99% identity to the associated reference sequence over their entire lengths. Suitably, L is selected from the group consisting of:
Figure imgf000037_0001
35   
Figure imgf000038_0001
. , . Alternatively, the polynucleotide molecule may comprise, consist of, or consist essentially a variant of SEQ ID NO. 13 and/or a variant of SEQ ID NO. 14 wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L1. In a further embodiment, the polynucleotide molecule may comprise, consist of, or consist essentially of SEQ ID NO.13 and SEQ ID NO. 14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L2. Alternatively, the 36   
Figure imgf000039_0001
example 10 – 1000 µg/mL, for example 10 – 500 µg/mL, for example 10 – 250 µg/mL, for example 50 – 150 µg/mL, such as about 100 µg/mL. In an alternative embodiment, the polynucleotide molecule may be present in the formulation at a concentration of 1 – 10000 µg/mL, for example 10 – 10000 µg/mL, for example 100 – 10000 µg/mL, for example 500 – 5000 µg/mL, for example 500 – 2500 µg/mL, such as about 1000 µg/mL. Alternatively, the polynucleotide molecule may be present in the formulation at a concentration of 1 – 100000 µg/mL, for example 10 – 100000 µg/mL, for example 100 – 100000 µg/mL, for example 1000 – 100000 µg/mL, for example 5000 – 75000 µg/mL, for example 5000 – 50000 µg/mL, such as 5000 – 15000 µg/mL, 10000 – 20000 µg/mL, 15000 – 25000 µg/mL, or 25000 – 50000 µg/mL. Significantly, as would be appreciated by the skilled person, the concentration of polynucleotide molecule in the formulation is highly variable and dependent on the type, sequence, structure, and size of the polynucleotide molecule. Suitably, the polynucleotide molecule may be present in the formulation at a concentration of 0.000001% (w/w) – 10% (w/w), for example 0.00001% (w/w) – 10% (w/w), for example 0.0001% (w/w) – 10% (w/w), for example 0.0001% (w/w) – 5% (w/w), wherein the % by weight is with respect to the total weight of the formulation. In one embodiment, the polynucleotide molecule may be present in the formulation at a concentration of 0.0001% (w/w) – 1% (w/w), for example 0.0001% (w/w) – 0.1% (w/w), for example 0.0001% (w/w) – 0.01% (w/w), for example 0.0001% (w/w) – 0.005% (w/w), for example 0.0001% (w/w) – 0.002% (w/w), such as about 0.001% (w/w), wherein the % by weight is with respect to the total weight of the formulation. In an alternative embodiment, the polynucleotide molecule may be present in the formulation at a concentration of 0.0001% (w/w) – 1% (w/w), for example 0.0001% (w/w) – 0.1% (w/w), for example 0.001% (w/w) – 0.1% (w/w), for example 0.001% (w/w) – 0.05% (w/w), for example 0.001% (w/w) – 0.025% (w/w), for example 0.005% (w/w) – 0.015% (w/w), such as about 0.01% (w/w), wherein the % by weight is with respect to the total weight of the formulation. In an alternative embodiment, the polynucleotide molecule may be present in the formulation at a concentration of 0.0001% (w/w) – 1% (w/w), for example 0.001% (w/w) – 1% 37    (w/w), for example 0.01% (w/w) – 1% (w/w), for example 0.05% (w/w) – 0.5% (w/w), for example 0.05% (w/w) – 0.25% (w/w), such as about 0.1% (w/w), wherein the % by weight is with respect to the total weight of the formulation. Alternatively, the polynucleotide molecule may be present in the formulation at a concentration of 0.0001% (w/w) – 10% (w/w), for example 0.001% (w/w) – 10% (w/w), for example 0.01% (w/w) – 10% (w/w), for example 0.1% (w/w) – 10% (w/w), for example 0.5% (w/w) – 7.5% (w/w), for example 0.5% (w/w) – 5% (w/w), such as 0.5% (w/w) – 1.5% (w/w), 1% (w/w) – 2% (w/w), 1.5% (w/w) – 2.5% (w/w) or 2.5% (w/w) – 5% (w/w), wherein the % by weight is with respect to the total weight of the formulation. Suitably, the ratio of the amount of the surfactant component (meaning the total concentration of the surfactants of the surfactant component) to the amount of polynucleotide molecule, for example wherein each is measured in µg/mL, is between about 100:1 and about 1:1000, for example between about 100:1 and about 1:750, for example between about 75:1 and about 1:750, for example between about 50:1 and about 1:750, for example between about 50:1 and about 1:500, for example between about 50:1 and about 1:250, for example between about 50:1 and about 1:200, for example between about 40:1 and about 1:200, for example between about 40:1 and about 1:150, for example between about 25:1 and about 1:150, for example between about 25:1 and about 1:100. Nucleic Acid Modifications In one embodiment, the polynucleotide molecules described above comprise one or more modified nucleotides. For example, the polynucleotide molecule comprises two or more, three or more, four or more, five or more, eight or more, or 10 or more modified nucleotides. Modifications to nucleotides suitably can enhance stability, functionality, and/or specificity, and can minimise the immunostimulatory properties of a polynucleotide molecule. Suitably, the polynucleotide molecule comprises at least one modified nucleotide which confers reduced immunostimulatory activity, and at least one modified nucleotide which confers increased serum half-life. Suitably, the same modified nucleotide elicits both effects. Suitably, the polynucleotide molecule comprises one or more modified nucleotides which confer enhanced resistance to nuclear enzymes. Modified nucleotides include, but are not limited to, sugar-, backbone-, and base-modified nucleotides. Suitable modified nucleotides may comprise any combination of sugar-, backbone-, and base-modifications. Sugar-modified nucleotides include, but are not limited to, nucleotides in which the 2’ OH- group (ribonucleotide) or 2’-H group (deoxyribonucleotide) is replaced by a group selected from the group consisting of H, OR, R, halo, SH, SR, NH2, NHR, NR2, or ON, wherein R is C1- C6 alkyl, alkenyl, or alkynyl. Further examples of 2’ OH-group (ribonucleotide) or 2’-H group (deoxyribonucleotide) modifications include alkoxy or aryloxy modifications, e.g. OR, wherein R is H, alkyl e.g. C1- 38    C6 alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar; polyethyleneglycol (PEG) modifications; “locked” nucleotide modifications wherein the 2’ OH-group or 2’ H-group is connected, for example via a methylene bridge, to the 4’ carbon of the same sugar; amine, O-amine and aminoalkoxy, e.g. O(CH2)namine, modifications, wherein amine is NH2, alkylamino, dialkylamino, heterocyclyl amino, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, ethylene diamine, or polyamino). Additional examples of 2’ OH-group (ribonucleotide) or 2’-H group (deoxyribonucleotide) modifications include H (for ribonucleotides); halo; amino, for example NH2, alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroaryl amino, diheteroaryl amino, or amino acid); NH(CH2CH2NH)nCH2CH2-amine modifications wherein amine is NH2, alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroaryl amino, diheteroaryl amino; NHC(O)R modifications, wherein R is alkyl e.g. C1-C6 alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar; cyano; mercapto; alkyl-thio-alkyl; thioalkoxy; and alkyl e.g. C1-C6 alkyl, cycloalkyl, aryl, alkenyl and alkynyl, which may be optionally substituted. In one embodiment, the polynucleotide molecule, in particular an RNA molecule, comprises one or more sugar-modified nucleotides which each comprise a 2’ OH (or 2’ H) modification. In one embodiment, the polynucleotide molecule, in particular an RNA molecule, comprises one or more sugar-modified nucleotides which each comprise a 2’ OH (or 2’ H) modification selected from the group consisting of 2’-deoxy, 2’-fluoro, 2'-deoxy-2'-fluoro, 2’-O-methyl, 2’-O- methoxyethyl (2’-O-MOE), 2’-O-aminopropyl (2’-O-AP), 2’-O-dimethylaminoethyl (2’-O- DMAOE), 2’-O-dimethylaminopropyl (2’-O-DMAP), 2’-O-dimethylaminoethyloxyethyl (2’-O- DMAEOE), or 2’-O—N-methylacetamido (2’-O-NMA). In one embodiment, the polynucleotide molecule, in particular an RNA molecule, comprises one or more sugar-modified nucleotides which each comprise a 2’ OH modification selected from the group consisting of 2’-H, and 2’-O-methyl. Suitably, the polynucleotide molecule, in particular an RNA molecule, comprises one or more nucleotides comprising a 2’-O-methyl modification. In one embodiment, all of the nucleotides in the polynucleotide molecule, in particular an RNA molecule, comprise a 2’-O-methyl modification. In one embodiment, some or all of the pyrimidine nucleotides of the polynucleotide molecule, in particular an RNA molecule, comprise a 2’ OH modification. In particular, some or all of the pyrimidine nucleotides may comprise a modification selected from the group consisting of 2’- H, and 2’-O-methyl. In one embodiment, some or all of the purine nucleotides of the polynucleotide molecule, in particular an RNA molecule, comprise a 2’ OH modification. In particular, some or all of the purine nucleotides may comprise a modification selected from the group consisting of 2’-H, and 2’-O-methyl. In one embodiment, the polynucleotide molecule, in particular an RNA molecule, comprises one or more dinucleotides which are susceptible to endonuclease cleavage, wherein the 5’ 39    nucleotide of the dinucleotide comprises a 2’ OH (or 2’ H) modification. Suitably the dinucleotide is s 5’-UA-3’, 5’-UG-3’, 5’-CA-3’, 5’-UU-3’, or 5’-CC-3’. Backbone-modified nucleotides include, but are not limited to, nucleotides which include modifications to the phosphate-sugar backbone of polynucleotides molecules, and in particular nucleotides which include modifications to the phosphodiester bonds which link the phosphate group and sugar molecule of adjacent nucleotides. For example, the phosphodiester bonds may be modified to include at least one heteroatom, such as a nitrogen or sulfur heteroatom. In particular, the phosphoester group of a nucleotide which links to the sugar molecule of an adjacent nucleotide may be replaced with a phosphothioate group. In one embodiment, the polynucleotide molecule, in particular an RNA molecule, comprises one or more backbone-modified nucleotides, in particular wherein the backbone modification is replacement of the phosphoester group of a nucleotide with a phosphorothioate group. In one embodiment, when the backbone modification is replacement of the phosphoester group of a nucleotide with a phosphorothioate group, the phosphorothioate group is located at the first, second, third, fourth, of fifth internucleotide linkage at the 5’ and/or 3’ end of the polynucleotide molecule, in particular an RNA molecule. In one embodiment, the polynucleotide molecule, in particular an RNA molecule, comprises one or more backbone-modified nucleotides wherein the backbone modification is the replacement of the ribose (ribonucleotide) or deoxyribose (deoxyribonucleotide) sugar moiety with a pyranose or furanose sugar moiety. Base-modified nucleotides include, but are not limited to, nucleotides comprising a non- naturally occurring base rather than a naturally occurring base (adenine, cytosine, guanine, thymine, or uracil). Suitable non-naturally occurring bases include, but are not limited to, uridine and/or cytidine modified at the 5-position, for example 5-(2-amino)propyl uridine, 5- bromo uridine; adenosine and/or guanosine modified at the 8 position, for example 8-bromo guanosine; deaza nucleotides, for example 7-deaza-adenosine; and O- and N-alkylated nucleotides, for example N6-methyl adenosine. Notably, these modifications may be combined. In one embodiment, the polynucleotide molecule, in particular an RNA molecule, comprises one or more base-modified nucleotides. In one embodiment, the modified nucleotides, for example sugar-modified, backbone- modified, or based-modified nucleotides, are located proximal to, for example within three nucleotides, five nucleotides, or 10 nucleotides, of the 5’ and/or 3’ end of the polynucleotide molecule. In an embodiment the polynucleotide is not conjugated to any small molecule e.g. the polynucleotide is not conjugated to any to any organic molecule having a molecular weight in the range 250-1500 e.g.300-1000 g/mol. 40    Pharmaceutical Formulation The aqueous liquid pharmaceutical formulations of the invention include water as the solvent. Water includes but not is limited to sterile or purified water, sterile water for injection, RNAse free water, or bacteriostatic water for injection. Suitably, the aqueous liquid pharmaceutical formulation is substantially free of any solvent or co-solvent other than water. In particular, the aqueous liquid pharmaceutical formulation does not comprise an organic solvent or co-solvent, such as inter alia ethanol, acetone, dimethyl sulfoxide (DMSO), dichloromethane (DCM), N-methyl pyrrolidinone (NMP), Ν,Ν’- dimethylformamide (DMF), N,N’-dimethylacetamide (DMAC), 1,3-dimethyl-2-imidazolidinone (DMEU), 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, and benzyl benzoate. As used herein, the term “substantially free of” means that the formulations comprise less than 2% (w/w), for example less than 1% (w/w), such as less than 0.5% (w/w), wherein the % by weight is with respect to the total weight of the formulation. Preferably, the formulation does not comprise any solvent or co-solvent other than water. The aqueous liquid pharmaceutical formulations according to the present invention may further comprise pharmaceutically acceptable excipients including, but not limited to, antioxidants, buffers, diluents, emulsifiers, lubricants, preservatives, solvents, stabilizers, suspending agents, thickeners, tonicity adjusting (osmotic) agents, vehicles, wetting agents. Suitable antioxidants include but are not limited to ascorbic acid (vitamin C), glutathione (reduced), lipoic acid, uric acid, carotenes, including β-carotene and retinol (vitamin A), cc- tocopherol (vitamin E), ubiquinol (coenzyme Q), butylated hydroxyanisole, butylated hydroxytoluene, propyl gallate, tert-butylhydroquinone, monothioglycerol, lutein, selenium, manganese, zeaxanthin, or a combination thereof. The aqueous liquid pharmaceutical formulations of the invention may comprise one or more buffers. Suitable buffers include but are not limited to citrate, borate, formate, glycine, alanine, acetate, aspartate, malate, glyoxylate, gluconate, lactate, glycolate, oxalate, histidine, tartarate and succinate buffer systems. As used herein, references to a “citrate” buffer will be understood to refer to a mixture of citrate and the corresponding acid as a buffer system in a ratio according to the target pH, that is the pH at which the aqueous liquid pharmaceutical formulation is intended to be buffered. For example, the buffer may comprise sodium citrate dihydrate and citric acid monohydrate. In particular, the buffer is based on a weak organic acid, for example the buffer is citrate, acetate, lactate, or formate, in particular citrate. A further suitable buffer is phosphate. For example, the buffer may comprise sodium phosphate and disodium phosphate. Suitable, pharmaceutically acceptable, diluents include but are not limited to isotonic saline (0.9% w/v), isotonic dextrose (5% w/v), isotonic mixtures of saline and dextrose (e.g. saline 41    (0.45 % w/v) and dextrose (2.5 % w/v)), sterile or purified water, sterile water for injection or bacteriostatic water for injection. In particular, the diluent is sterile or purified water, sterile water for injection, RNAse free water or bacteriostatic water for injection. For example, in one embodiment, the diluent is sterile or purified water. In an alternative embodiment, the diluent is isotonic saline (0.9% w/v). Suitable preservatives include, but are not limited to, edetic acid and alkali salts thereof, such as disodium edetate (also known as “disodium EDTA”) or calcium edetate (also known as calcium EDTA), phenol, m-cresol, chlorocresol, benzyl alcohol, propyl paraben, methyl paraben, butyl paraben, chlorobutanol, phenylethyl alcohol, benzalkonium chloride, thimerosal, propylene glycol, sorbic acid, benzoic acid derivatives and combinations thereof. Suitable suspending agents include, but are not limited to, acacia (gum), sodium alginate, starch and starch derivatives, xanthan gum, pectin, methylcellulose, hydroxyethylcellulose, sodium carboxymethylcellulose (Avicel RC591), microcrystalline cellulose, hypromellose, hyaluronic acid, and combinations thereof. Particularly suitable suspending agents include, microcrystalline cellulose, sodium carboxymethylcellulose (Avicel RC591), hyaluronic acid, and combinations thereof. The properties of certain suspending agents may further render them as suitable thickening agents and/or wetting agents. Accordingly, suitable thickening agents and/or wetting agents, may include, but are not limited to the suspending agents recited above. In particular, suitable thickening agents and/or wetting agents include microcrystalline cellulose, sodium carboxymethylcellulose (Avicel RC591), hyaluronic acid, and combinations thereof. Suitable tonicity adjusting (osmotic) agents include, but are not limited to, polyols, such as sugars and sugar alcohols, for example erythritol, glycerol, lactose, maltitol, mannitol, sorbitol, trehalose, and xylitol, and salts, for example sodium acetate, sodium lactate, sodium chloride, potassium chloride, and calcium chloride. A particularly suitable tonicity adjusting (osmotic) agent is glycerol. Suitably, the aqueous liquid pharmaceutical formulation according to the present invention has an osmolarity from about 50 mOsm to about 750 mOsm, for example from about 100 mOsm to about 600 mOsm, for example from about 100 mOsm to about 500 mOsm. In particular, the aqueous liquid pharmaceutical formulation according to the present invention suitably has an osmolarity from about 100 mOsm to about 400 mOsm, for example from about 150 mOsm to about 350 mOsm, for example from about 200 mOsm to about 300 mOsm. The pH of the aqueous liquid pharmaceutical formulation according to the present invention is suitably between about 4.0 and about 9.0, such as between about 4.0 and about 8.0, such as between about 4.0 and about 7.0 or between about 5.0 and about 8.0. In particular, the pH is suitably between about 4.0 and about 6.0, such as between about 4.0 and about 5.5. For example, the pH of the aqueous liquid pharmaceutical formulation is about 4.0, about 4.1, 42    about 4.2, about 4.3, about 4.4, about 4.5, about, 4.6, about 4.7, about 4.8, about 4.9 or about 5.0. Alternatively, pH is suitably between about 5.5 and about 8.0, such as between about 6.0 and about 8.0, such as between about 6.5 and about 7.5 or between about 7.0 and about 80. For example, the pH of the aqueous liquid pharmaceutical formulation is about 6.5, about 6.6, about 6.7. about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0. The pH of such a pharmaceutical composition may be adjusted by pH adjusting agents including acidifying agents such as hydrochloric acid, tartaric acid, citric acid, succinic acid, phosphoric acid, ascorbic acid, acetic acid, lactic acid, sulphuric acid, formic acid and mixtures thereof, or alkaline buffering agents such as ammonium hydroxide, ethylamine, dipropylamine, triethylamine, alkanediamines, ethanolamines, polyalkylene polyamines, heterocyclic amines, hydroxides of alkali metals, such as sodium and potassium hydroxide, hydroxides of alkali earth metals, such as magnesium and calcium hydroxide, and basic amino acids such as L-arginine, lysine, alanine, leucine, isoleucine, oxylysine and histidine, and mixtures thereof. The aqueous liquid pharmaceutical formulations according to the present invention may suitably have a pH between about 4.0 and about 9.0, such as between about 4.0 and about 8.0. It will be understood by the skilled person that an aqueous liquid pharmaceutical formulation suitable for topical administration to the nose may suitably have a pH between about 4.0 and about 9.0, such as between about 4.0 and about 8.0, such as between about 4.0 and about 7.0, such as between about 4.0 and about 6.0, such as between about 4.0 and about 5.0. A particularly suitable buffer for compositions of this desired pH is citrate. The skilled person would further understand that an aqueous liquid pharmaceutical formulation suitable for topical administration to the lung may suitably have a pH between about 5.5 and about 8.0, such as between about 6.0 and about 8.0, for example between about 6.0 and about 7.0 or between about 7.0 and about 8.0. A particularly suitable buffer for compositions of this desired pH is phosphate. Suitably, the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a protein. Suitably the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a cationic lipopeptide, for example it does not comprise a polymyxin such as polymyxin B. Moreover, the aqueous liquid pharmaceutical formulation of the present invention suitably does not comprise an inorganic nanoparticle, suitable or typical examples of which are known in the art and may include inorganic nanoparticles of metal salts, such as zinc oxide, or may include gold, silver, or silica nanoparticles. Furthermore, the aqueous liquid pharmaceutical formulation of the present invention suitably does not comprise a lipid nanoparticle (LNP) or a liposome. The aqueous liquid pharmaceutical formulation of the present invention suitably does not comprise a solid or partly solid (e.g. part solid and part liquid) nanoparticle. Lipid nanoparticles (LNPs) which 43    have in the prior art been described as delivery systems for nucleic acids (see e.g. Kulkarni et al.2021) typically are particles formed of a core comprising a cationic lipid that can bind the nucleic acid, a neutral lipid (such as cholesterol), a helper lipid which contributes to the structure of the particle and an outer formed of a stabiliser molecule. The helper lipids, which can include phospholipids, such as phosphatidylcholine or phosphatidylethanolamine, play a role in enhancing the stability of the LNPs by providing structural integrity and facilitating the formation of the lipid bilayer. The stabiliser, often a polyethylene glycol (PEG)-lipid, provides a hydrophilic shield that reduces aggregation and opsonization, thus prolonging circulation time in the bloodstream and enhancing the delivery efficiency to target cells. All of the components in LNPs are required for encapsulation efficiency and facilitate cellular uptake and ensures its stability in biological environments. Liposomes are typically spherical vesicles which contain a lipid bilayer. Suitably the particles of the stable colloidal emulsion of the present invention do contain any lipid bilayer. Moreover, suitably the aqueous liquid pharmaceutical formulation according to the present invention is substantially free of lipid nanoparticle (LNP) and liposome components. Lipid nanoparticles (LNPs) are typically described as nanoparticles which comprise a core of cationic or ionizable lipids designed to encapsulate nucleic acids, such as mRNA or siRNA, forming a complex that can be efficiently delivered to target cells. The core structure is stabilised by neutral lipids like cholesterol, which help to maintain the integrity and fluidity of the lipid bilayer. Additionally, helper lipids such as phosphatidylcholine or phosphatidylethanolamine are incorporated to enhance the encapsulation efficiency of the nucleic acids and facilitate the fusion of the LNP with cellular membranes, thereby improving uptake by target cells. The outer surface of the LNP is often modified with stabiliser molecules, such as polyethylene glycol (PEG)-lipids, which provide a hydrophilic coating that reduces aggregation and opsonization by the immune system. This coating prolongs the circulation time of the LNPs in the bloodstream, increasing the likelihood of reaching the target cells. Together, these components work synergistically to create a delivery system for nucleic acids, capable of protecting the genetic material and ensuring its effective delivery and expression within the target cells. Therefore, for example, suitably, the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a neutral lipid. Such a neutral lipid typically has no ionizable groups, which therefore leads to no charge (whether positive or negative) at around neutral pH (i.e. pH 7.0-7.4). In particular, the pharmaceutical formulation does not comprise cholesterol, or an analogue thereof. Suitably, the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a cationic lipid, such as quaternary ammonium lipid. In particular, the pharmaceutical formulation does not comprise a cationic lipid, such as N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA) or 1,2-dioleoyl-3-trimethylammonium propane (DOTAP). Such a cationic 44    lipid typically has at least one ionizable group leading to a net positive charge at around neutral pH (i.e. pH 7.0 or 7.4). Suitably, the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a helper lipid, such as dioleoylphosphatidylethanolamine (DOPE) or phosphatidylcholine. Furthermore, for example, suitably, the aqueous liquid pharmaceutical formulation according to the present invention does not comprise a lipid, other than the fatty acid present therein. In one embodiment in which the fatty acid is oleic acid, suitably the aqueous liquid pharmaceutical formulation does not comprise a lipid, other than oleic acid. The aqueous liquid pharmaceutical formulations (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention may be suitable for oral, inhalational, sub- lingual, buccal, or parenteral, including intravenous, subcutaneous, topical, transdermal, pulmonary, rectal, vaginal, ocular, intranasal, intradermal, transmucosal, intraperitoneal, intramuscular, intracapsular, intraorbital, intracardiac, transtracheal, subcuticular, intraarticular, intraspinal, and intrasternal administration. In one embodiment, the aqueous liquid pharmaceutical formulations (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention are suitable for topical administration to the lung or nose. Accordingly, the aqueous liquid pharmaceutical formulations of the present invention are suitable for administration via inhalation, for example suitable for administration topically to the lung via oral inhalation, or for intranasal administration. In one embodiment, the aqueous liquid pharmaceutical formulation (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention is administered topically to the lung or nose. Accordingly, in one embodiment, the aqueous liquid pharmaceutical formulations (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention are administered via inhalation or are administered intranasally. It should be noted that the aqueous liquid pharmaceutical formulations of the invention suitable for topical administration to the lung or nose, when administered topically to the lung by oral inhalation or topically to the nose may thereby involve administration to the pharynx. It will be understood that a formulation suitable for topical administration to the lung may comprise different pharmaceutically acceptable excipients to a formulation suitable for topical administration to the nose. By way of example, a formulation suitable for topical administration to the nose may comprise a suspending and/or wetting and/or thickening agent such as microcrystalline cellulose, sodium carboxymethylcellulose (Avicel RC591), hyaluronic acid, or a combination thereof, whilst a formulation suitable for topical administration to the lung may not. In an alternative embodiment, the aqueous liquid pharmaceutical formulations (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention are 45    suitable for subcutaneous administration, and in particular via subcutaneous injection. In one embodiment, the aqueous liquid pharmaceutical formulation (or the immunostimulatory, immunogenic, or vaccine composition) of the present invention is administered subcutaneously, for example via subcutaneous injection. It will be understood that a formulations suitable for subcutaneous administration will comprise pharmaceutically acceptable excipients appropriate for that route, and that those pharmaceutically acceptable excipients will be different to those present in a formulation suitable for topical administration to the lung or nose. In an alternative embodiment, the aqueous liquid pharmaceutical formulations (or the immunostimulatory, immunogenic, or vaccine compositions) of the present invention are suitable for ocular administration. For example, the aqueous liquid pharmaceutical formulations of the present invention may be suitable for intra-ocular administration. Alternatively, the aqueous liquid pharmaceutical formulations of the present invention may be suitable for topical administration to the eye. Accordingly, in one embodiment the aqueous liquid pharmaceutical formulation (or the immunostimulatory, immunogenic, or vaccine composition) of the present invention is administered via the ocular administration route (e.g. via the intra-ocular route or via topical administration to the eye). It will be understood that a formulations suitable for ocular administration will comprise pharmaceutically acceptable excipients appropriate for that route, and that those pharmaceutically acceptable excipients will be different to those present in a formulation suitable for topical administration to the lung or nose or a formulation suitable for subcutaneous administration. Suitably, the aqueous liquid pharmaceutical formulations of the present invention are suitable for administration to a mammal. More suitably, the aqueous liquid pharmaceutical formulations of the present invention are suitable for administration to a human. In one embodiment, the aqueous liquid pharmaceutical formulations of the present invention are administered to a mammal. In particular, the aqueous liquid pharmaceutical formulations of the present invention are administered to a human. Suitably, the aqueous liquid pharmaceutical formulations disclosed herein may be administered to a patient or subject once or more than once a day, for example two times a day, three time a day, four times a day or five times a day. Such treatment may extend for a number of weeks or months. Medical Use Without wishing to be bound by theory, the present inventors intend that the aqueous liquid pharmaceutical formulations of the present invention should (in at least some embodiments) enhance, improve, or make more efficient the delivery, and thereby increase the exposure, of a given polynucleotide molecule to a target cell or tissue. Accordingly, in one embodiment, there is provided a method for enhancing or improving the delivery of a polynucleotide 46    molecule to a target cell or tissue, comprising formulating the polynucleotide molecule in an aqueous liquid pharmaceutical formulation of the present invention and administering said formulation to a target cell or tissue. As used herein, a “target cell or tissue” refers to a cell or tissue which is targeted for the purpose of administration of a polynucleotide molecule, wherein said cell or tissue may also be, or may not be, the cell or tissue that is targeted for the for the purpose of eliciting a therapeutic effect. Suitably, the target cell or tissue is a cell or tissue of the lung or nose, and in particular a cell or tissue of the nasal or respiratory epithelium. Polynucleotide molecules can elicit a therapeutic effect, upon interaction of the polynucleotide molecule with molecules, organelles, cells, or tissues. Therefore, in one embodiment there is provided an aqueous liquid pharmaceutical formulation according to the present invention for use as a medicament. Suitably, the aqueous liquid pharmaceutical formulation according to the present invention is for use as a medicament for administration topically to the lung (e.g., by oral inhalation) or topically to the nose. Suitably, the aqueous liquid pharmaceutical formulations according to the present invention for use as a medicament are administered topically to the lung (e.g., by oral inhalation) or intranasally. Alternatively, suitably, the aqueous liquid pharmaceutical formulation according to the present invention is for use as a medicament for administration subcutaneously (e.g. by subcutaneous injection). Alternatively, the aqueous liquid pharmaceutical formulation according to the present invention may be for use as a medicament for administration ocularly. In one embodiment, the aqueous liquid pharmaceutical formulation for use according to the present invention is for use therapeutically i.e., in the treatment of disease. Alternatively, the aqueous liquid pharmaceutical formulation for use prophylactically i.e., in the prevention of disease. A suitable dose of the aqueous liquid pharmaceutical formulation for use as described herein is a therapeutically or prophylactically effective dose which can be determined by the skilled person. By way of example, the aqueous liquid pharmaceutical formulation for use may be administered to a patient or subject in an amount such that the dose of the polynucleotide molecule is 0.01 µg/mL – 100000 µg/mL, for example 0.1 – 100000 µg/mL, for example 1 – 100000 µg/mL, for example 1 – 50000 µg/mL. In one embodiment, the aqueous liquid pharmaceutical formulation for use may be administered to a patient or subject in an amount such that the dose of the polynucleotide molecule is 1 – 10000 µg/mL, for example 1 – 1000 µg/mL, for example 1 – 100 µg/mL, for example 1 – 50 µg/mL, for example 1 – 20 µg/mL, such as about 10 µg/mL. In an alternative embodiment, the aqueous liquid pharmaceutical formulation for use may be administered to a patient or subject in an amount such that the dose of the polynucleotide molecule is 1 – 10000 µg/mL, for example 1 – 1000 µg/mL, for example 10 – 1000 µg/mL, for example 10 – 500 µg/mL, for example 10 – 250 µg/mL, for example 50 – 150 µg/mL, such as about 100 µg/mL. Alternatively, the aqueous liquid 47    pharmaceutical formulation for use may be administered to a patient or subject in an amount such that the dose of the polynucleotide molecule is 1 – 100000 µg/mL, for example 10 – 100000 µg/mL, for example 100 – 100000 µg/mL, for example 1000 – 100000 µg/mL, for example 5000 – 75000 µg/mL, for example 5000 – 50000 µg/mL, such as 5000 – 15000 µg/mL, 10000 – 20000 µg/mL, 15000 – 25000 µg/mL, or 25000 – 50000 µg/mL.. Significantly, as would be appreciated by the skilled person, the dose will depend on the administration route, sequence, structure, and size of the polynucleotide molecule and indication to be treated. Suitably, the aqueous liquid pharmaceutical formulations for use as described herein may be administered to a patient or subject once or more than once a day, for example two times a day, three time a day, four times a day or five times a day. Such treatment may extend for a number of weeks or months. Suitably, the aqueous liquid pharmaceutical formulations for use according to the present invention are administered to a mammal. In particular, the aqueous liquid pharmaceutical formulations of the present invention are administered to a human. Immune Stimulation For example, a polynucleotide molecule, in particular an shRNA molecule, may directly or indirectly (e.g., via an encoded protein or other gene product) interact with molecules or cells of the immune system, resulting in activation of the innate and/or adaptive immune system. By way of example, such immune stimulation or activation may have utility in the treatment of infectious disease or cancer. By way of definition, other gene products include inter alia RNA or cDNA molecules. Therefore, in one embodiment there is provided an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the aqueous liquid pharmaceutical formulation stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response. Accordingly, in one embodiment, an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response, for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response. In an alternative embodiment, the present invention provides a method for the treatment of a disease or condition, which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation as described herein, wherein the 48    polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response. Moreover, the present invention provides use of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response. Immunostimulatory Compositions As discussed above, a polynucleotide molecule may interact with molecules or cells of the immune system, resulting in stimulation activation of the innate and/or adaptive immune system. Therefore, in one embodiment, there is provided an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system, for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system. Furthermore, the present invention provides an aqueous liquid pharmaceutical formulation, or immunostimulatory composition, for use in stimulating or activating the immune system in a subject. Accordingly, the present invention provides a method of stimulating the immune system in a subject, comprising administering to the subject an aqueous liquid formulation, or immunostimulatory composition, as described herein. Furthermore, there is provided use of an aqueous liquid pharmaceutical formulation, or immunostimulatory composition, in the manufacture of a medicament for stimulating the immune system in a subject. Suitably, the aqueous liquid formulation, or immunostimulatory compositions of the present invention is for use in stimulating or activating an anti-viral innate and/or adaptive immune response in a subject. Suitably, the polynucleotide molecule present in an aqueous liquid pharmaceutical formulation for use in stimulating the immune system in a subject is an e.g., directly immunostimulatory polynucleotide molecule, in particular wherein the polynucleotide molecule is an shRNA molecule. Alternatively, the immunostimulatory polynucleotide may be poly I:C (see, for example, dsRNA 2 in the Examples below and US9682096B2, which is incorporated herein by reference). Therefore, in one embodiment, the present invention provides an immunostimulatory composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic 49    surfactant and (ii) an immunostimulatory polynucleotide molecule. The immunostimulatory polynucleotide molecule is capable of, or for use in, stimulating the immune system in a subject. Suitably, the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular polysorbate 80. Suitably, the immunostimulatory polynucleotide molecule is an shRNA molecule. By way of definition, the immune system in a subject is considered stimulated if an immune response, including an innate immune response, antibody response or cell-mediated immune response, is initiated, potentiated or enhanced in response to an antigen or immunogen, in particular an exogenous antigen or immunogen. By way of definition, an immunogen is an immunogenic antigen. Suitably, the immune response is stimulated by inducing a pro-inflammatory cytokine response. For example, the immune response is stimulated by inducing a pro-inflammatory interleukin response, such as one or more of an IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL- 9, IL-11, IL-17, and IL-18 response, or by inducing a pro-inflammatory interform response, such as one or more of an IFN-α, IFN-β, and IFN-γ response, or by inducing a pro- inflammatory chemokine response, or by including a pro-inflammatory tumor necrosis factor response, such as a TNF-α and/or TNF-β response, or a combination thereof. In particular, the immune response is stimulated by inducing an interferon response, such as one or more of an IFN-α, IFN-β, and IFN-γ response. Suitably, the immune response is stimulated by activating a pattern recognition receptor (PRR). PRRs can induce pro-inflammatory cytokine response, such as pro-inflammatory interferon responses. For example, the PRR can be any member within a family of PRRs selected from the toll-like receptors (TLRs), such as TLR2, TLR3, TLR4, TLR7, TLR8, or TLR9, C-type lectin receptors (CLRs), NOD-like receptors (NLRs) and RIG-I-like receptors (RLRs), such as RIG-I, MDA5 or LGP2. Suitably, the PRR can be any member within the RLRs, such as RIG-I, MDA5, or LGP2, in particular RIG-I. Immunogenic Compositions As discussed above, a polynucleotide molecule may interact, for example indirectly (e.g., via an encoded protein or other gene product), with molecules or cells of the immune system, resulting in the raising of an innate and/or adaptive immune response. Therefore, in one embodiment, there is provided an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the polynucleotide molecule is a polynucleotide molecule which raises an innate and/or adaptive immune response in a subject, for use in the treatment of a disease or condition which is treated by raising an innate and/or adaptive immune response. 50    Furthermore, there is provided an aqueous liquid pharmaceutical formulation, immunogenic composition, or vaccine composition, for use in raising an immune response in a subject, wherein the immune response is raised against the immunogen or vaccine immunogen encoded by the polynucleotide molecule. Accordingly, the present invention provides a method of raising an immune response in a subject, comprising administering to the subject an aqueous liquid formulation as described herein. Moreover, there is provided use of an aqueous liquid pharmaceutical formulation in the manufacture of a medicament for raising an immune response in a subject. Suitably, the polynucleotide molecule present in an aqueous liquid pharmaceutical formulation, or vaccine composition, for use in raising an immune response in a subject encodes an immunogen. Endogenous expression of the immunogen or vaccine immunogen leads to the raising of an immune response against said immunogen in a subject. In particular, said polynucleotide molecule is an mRNA molecule or DNA molecule. In one embodiment, the aqueous liquid pharmaceutical formulation, or vaccine compositions, for use in raising an immune response in a subject is for use therapeutically, that it is the immune response is raised in order to have curative effect on a disease or condition treated by the raising of an innate and/or adaptive immune response e.g., cancer. Alternatively, the compositions are for use prophylactically, that it is the immune response is raised in order to have a protective or prophylactic effect against a disease or condition treated by the raising of an innate and/or adaptive immune response e.g., an infectious disease. Therefore, in one embodiment, the present invention provides an immunogenic composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule which encodes an immunogen. The polynucleotide molecule which encodes an immunogen is capable of, or for use in, raising an immune response against said immunogen in a subject. Suitably, the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular polysorbate 80. Suitably, the polynucleotide molecule which encodes an immunogen is an mRNA molecule or DNA molecule. Furthermore, there is provided a vaccine composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule which encodes a vaccine immunogen. The polynucleotide molecule which encodes a vaccine immunogen is capable of, or for use in, raising an immune response against said vaccine immunogen in a subject. Suitably, the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in 51    particular polysorbate 80. Suitably, the polynucleotide molecule which encodes a vaccine immunogen is an mRNA molecule or DNA molecule. By way of definition, a vaccine immunogen is an immunogen, that is an immunogenic antigen, capable of raising a therapeutic or protective or immune response in a subject. Significantly, an aqueous liquid pharmaceutical formulation for use in raising an immune response in a subject may comprise (i) a polynucleotide molecule which encodes an immunogen, against which the immune response may be raised and (ii) a polynucleotide molecule which stimulates the immune system in a subject. That is, an immunogenic composition or vaccine composition according to the present invention may further comprise an immunostimulatory polynucleotide molecule. Suitably said immunostimulatory polynucleotide molecule is an shRNA molecule. Therefore, in one embodiment, the present invention provides an immunogenic composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule which encodes an immunogen, and (iii) an immunostimulatory polynucleotide molecule. Suitably, the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular polysorbate 80. The polynucleotide molecule which encodes an immunogen is capable of, or for use in, raising an immune response against said immunogen in a subject. Suitably, the polynucleotide molecule which encodes an immunogen is an mRNA or DNA molecule. The immunostimulatory polynucleotide molecule is capable of, or for use in, stimulating the immune system in a subject. Suitably, the immunostimulatory polynucleotide molecule is an shRNA molecule. Furthermore, there is provided a vaccine composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule which encodes a vaccine immunogen, and (iii) an immunostimulatory polynucleotide molecule. Suitably, the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, in particular polysorbate 80. The polynucleotide molecule which encodes a vaccine immunogen is capable of, or for use in, raising an immune response against said vaccine immunogen in a subject. Suitably, the polynucleotide molecule which encodes a vaccine immunogen is an mRNA or DNA molecule. The immunostimulatory polynucleotide molecule is capable of, or for use in, stimulating the immune system in a subject. Suitably, the immunostimulatory polynucleotide molecule is an shRNA molecule. Suitably, the immunostimulatory or immunogenic compositions described above do not comprise a protein. Suitably said compositions do not comprise a cationic lipopeptide, such 52    as polymyxin B. Suitably, said compositions do not comprise a neutral lipid. In particular, said compositions do not comprise cholesterol, or an analogue thereof. Suitably, said compositions do not comprise a cationic lipid. Suitably said compositions do not comprise a helper lipid, such as dioleoylphosphatidylethanolamine (DOPE) or phosphatidylcholine. Furthermore, the immunostimulatory or immunogenic compositions described above do not comprise an inorganic nanoparticle, a lipid nanoparticle (LNP), or a liposome and/or are substantially free of LNP and liposome components. Suitably, the immunostimulatory or immunogenic compositions described above may further comprise pharmaceutically acceptable excipients, at the relevant amounts, as described above. Indeed, said compositions may be limited as described above in relation to the aqueous liquid pharmaceutical formulations of the invention. Immune Stimulation Indications Suitably, a disease or condition treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response is an infectious disease. The infectious disease is suitably bacterial, fungal, parasitic, or viral in origin. In one embodiment, the aqueous liquid pharmaceutical formulation of the present invention is for use in the treatment or prevention of infection by a bacteria, fungus or parasite, or disease associated with infection with such a bacteria, fungus, or parasite. That is the disease or condition treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response is infection by a bacteria, fungus or parasite or disease associated with infection with such a bacteria, fungus, or parasite Suitably, the bacteria, fungus or parasite infects the brain, circulatory system, endocrine system, eyes, gastrointestinal tract, genital tract, kidneys, liver, respiratory tract, or skin. Accordingly, the disease associated with infection is a disease of the brain, circulatory system, endocrine system, eyes, gastrointestinal tract, genital tract, kidneys, liver, respiratory tract, or skin. In particular, the bacteria, fungus or parasite infects the respiratory tract and the disease associated with infection is a disease of the respiratory tract. For example, the bacteria belongs to the genus Bordetella, Chlamydophila, Corynebacterium, Coxiella, Escherichia, Haemophilus, Klebsiella, Legionella, Moraxella, Mycobacterium, Mycoplasma, Proteus, Pseudomonas, Serratia, Staphylococcus, Streptococcus In one embodiment, the aqueous liquid pharmaceutical formulation of the present invention is for use in the treatment or prevention of infection by a virus or disease associated with infection with such a virus. That is the disease or condition treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response is infection by a virus or disease associated with infection with such a virus. 53    Suitably, the virus is a DNA virus or an RNA virus. Suitably, the virus has a single-stranded genome, double-stranded genome, or partially double-stranded genome. Sutiably, the single- stranded genome is a sense (+) genome. Alternatively, the single-stranded genome is an antisense (-) genome. In particular, the virus may have a single-stranded RNA genome, double-stranded DNA genome, or double-stranded RNA genome. For example, the virus may have a sense (+) single-stranded RNA genome or an antisense (-) single-stranded RNA genome. Suitably the virus is naked i.e., is not enveloped. Alternatively, the virus is enveloped. Suitably, the virus belongs to the family Adenoviridae, Arenaviridae, Astroviridae, Bornaviridae, Bunyaviridae, Caliciviridae, Circoviridae, Coronaviridae, Filoviridae, Flaviviridae, Hepadnaviridae, Hepeviridae, Herpesviridae, Orthomyxoviridae, Papillomaviridae, Polyomaviridae, Paramyxoviridae, Parvoviridae, Picornaviridae, Poxviridae, Reoviridae, Retroviridae, Rhabdoviridae, or Togaviridae. In particular, the virus belongs to the family Adenoviridae, Coronaviridae, Herpesviridae, Orthomyxoviridae, Paramyxoviridae, or Picornaviridae. Suitably, the virus infects the brain, circulatory system, endocrine system, eyes, gastrointestinal tract, genital tract, kidneys, liver, respiratory tract, or skin. Accordingly, the disease associated with viral infection is a disease of the brain, circulatory system, endocrine system, eyes, gastrointestinal tract, genital tract, kidneys, liver, respiratory tract, or skin. In particular, the virus infects the respiratory tract and the disease associated with viral infection is a disease of the respiratory tract. For example, the virus is SARS-CoV-2 and the disease associated with viral infection is COVID-19. For example, the virus is seasonal coronavirus, for example 229E, NL63, OC43, or HKU1, and the disease associated with viral infection is the disease associated with seasonal coronavirus, for example 229E, NL63, OC43, or HKU1, infection. For example, the virus is influenza virus and the disease associated with viral infection is influenza. For example, the virus is respiratory syncytial virus (RSV) and the disease associated with viral infection is the disease associated with RSV infection. For example, the virus is human rhinovirus (HRV) and the disease associated with viral infection is the disease associated with HRV infection. For example, the virus is Middle East respiratory syndrome (MERS)-CoV and the disease associated with viral infection is MERS. For example, wherein the virus is an avian influenza virus and the disease associated with viral infection is avian influenza. For example, the virus is Nipah virus and the disease associated with viral infection is the disease associated with Nipah virus infection. For example, the virus is a human parainfluenza virus (HPIV) and the disease associated with viral infection is the disease associated with HPIV infection. For example, the virus is a human metapneumovirus (hMPV) and the disease associated with viral infection is the disease associated with hMPV infection. 54    In an alternative embodiment, the virus infects the respiratory tract, but the disease associated with viral infection is a systemic disease, or is a disease of the brain, circulatory system, endocrine system, eyes, gastrointestinal tract, genital tract, kidneys, liver, skin, or other organ or organ system. For example, the virus is Ebola virus and the disease associated with viral infection is Ebola or Ebola virus disease (EVD). For example, the virus is Lassa virus and the disease associate with viral infection is Lassa fever. As used herein, “influenza virus” includes influenza A virus, influenza B virus, influenza C virus and influenza D virus, for example influenza A virus or influenza B virus. Alternatively, a disease or condition treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response is cancer. For example, the cancer may be breast cancer, bladder cancer, renal cancer, lung cancer, prostate cancer, bone cancer, brain cancer, cervical cancer, anal cancer, colon cancer, colorectal cancer, gastric cancer, blood cancer, such as leukaemia, lymphoma, or myeloma, liver cancer, skin cancer, ovarian cancer, pancreatic cancer, testicular cancer, thyroid cancer, vaginal cancer, cardiac cancer or sarcoma. Increasing Gene Expression and Gene Therapy Alternatively, a polynucleotide molecule, in particular an mRNA or DNA molecule, may be intended to establish expression of one or more proteins (or other gene products) which are encoded by the polynucleotide molecule, in particular wherein endogenous expression of said protein (or other gene product) is defective or too low, e.g. silent, leading to insufficient expression of, or expression of a defective or dysfunctional variant of, a protein (or other gene product), wherein the insufficient expression of, or expression of a defective or dysfunctional variant of, a protein (or other gene product) is contributing to cellular dysfunction, and consequently disease. Notably, a polynucleotide molecule, in particular an mRNA or DNA molecule, may be intended to establish expression of one or more proteins (or other gene products) which are encoded by the polynucleotide molecule, in order to supplement endogenous gene expression such that over-expression of the protein (or other gene product) facilitates interference with endogenous cellular process, such as the regulation of gene expression or signal transduction. Moreover, over-expression of the protein (or other gene product) may be intended to activate an immune response or stimulate the immune system, for example against a tumour over- expressing an antigen, or in response to an immune cell presenting an exogenous antigen. Therefore, in one embodiment there is provided an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the aqueous liquid pharmaceutical formulation increases the endogenous expression of a protein (or other gene product). 55    In an alternative embodiment there is provided an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the aqueous liquid pharmaceutical formulation increases the endogenous expression of a functional protein (or other gene product). For example, the aqueous liquid pharmaceutical formulation may establish the expression of a functional protein (or other gene product), whilst endogenous expression of said protein (or other gene product) produces a non-functional or dysfunctional e.g., truncated or misfolded protein (or other gene product). As used herein, the term “increases” includes restoring i.e., increasing from zero or a low value to a “normal” value and enhancing i.e., increasing from a “normal” value to a high value, endogenous gene expression. Accordingly, in one embodiment the present invention provides an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein (or other gene product) or increases endogenous expression of a functional protein (or other gene product), is for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein (or other gene product), or by increasing the expression of a functional protein (or other gene product). Therefore, in one embodiment, there is provided an aqueous liquid pharmaceutical formulation for use in a method of gene therapy, wherein the polynucleotide molecule, in particular an mRNA or DNA molecule, encodes a therapeutic gene, protein (or other gene product). Suitably said use in a method of gene therapy is for the treatment of a disease or condition which is treated by increasing the endogenous expression of the therapeutic gene, protein (or other gene product) encoded by said polynucleotide molecule. In an alternative embodiment, the present invention provides a method for the treatment of a disease or condition, which is treated by increasing the endogenous expression of a protein (or other gene product), or by increasing the expression of a functional protein (or other gene product), comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein (or other gene product) or increases endogenous expression of a functional protein (or other gene product). Furthermore, there is provided a method of gene therapy, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule, in particular an mRNA or DNA molecule, encodes a therapeutic gene, protein (or other gene product). Suitably said method of gene therapy is for the treatment of a disease or condition 56    which is treated by increasing the endogenous expression of the therapeutic gene, protein (or other gene product) encoded by said polynucleotide. The present invention also provides use of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein (or other gene product) or increases endogenous expression of a functional protein (or other gene product), in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein (or other gene product), or by increasing the expression of a functional protein (or other gene product). Furthermore, there is provided use of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule, in particular an mRNA or DNA molecule, encodes a therapeutic gene, protein (or other gene product), in the manufacture of a medicament for use in a method of gene therapy. Suitably said use in a method of gene therapy is for the treatment of a disease or condition which is treated by increasing the endogenous expression of the therapeutic gene, protein (or other gene product) encoded by said polynucleotide. Suitably, a disease or condition treated by increasing the endogenous expression of a protein (or other gene product), or by increasing the endogenous expression of a functional protein (or other gene product) is a monogenic or polygenic disease or condition. For example, the disease or condition is a blood disease or condition, such as anaemia, in particular sickle-cell anaemia, haemophilia, in particular haemophilia A or B, severe combined immune deficiency (SCID), thalassemia, or Von Willebrand disease, a hearing disease or condition, such as deafness, a heart disease or condition, such as atherosclerosis, coronary heart disease, Long QT syndrome, or Von-Hippel Lindau syndrome, a metabolic disease or condition, including lysosomal storage diseases and conditions, such as Type I diabetes, Gaucher disease, glycogen storage disease, or obesity, a musculoskeletal disease or condition, such as Duchenne muscular dystrophy, or achondroplasias, a nervous system or brain disease or condition, such as Alzheimer’s disease, amyotrophic lateral sclerosis (ALS), Rett’s syndrome, Fragile X syndrome, Huntington’s disease, or Parkinson’s disease, a digestive or renal disease or condition, such as polycystic kidney disease, a respiratory disease or condition, such as asthma, alpha-1 antitrypsin deficiency, chronic obstructive pulmonary disease (COPD), primary ciliary dyskinesia (PCD), pulmonary fibrosis, sarcoidosis, or cystic fibrosis, a skin disease or condition, such as albinism, male pattern baldness, or alopecia. In particular, the disease or condition is a respiratory disease or condition, such as asthma, alpha-1 antitrypsin deficiency, chronic obstructive pulmonary disease (COPD), primary ciliary dyskinesia (PCD), pulmonary fibrosis, sarcoidosis or cystic fibrosis. 57    By way of example, the disease or condition is cystic fibrosis and the polynucleotide molecule present in the formulation according to the present invention increases the endogenous expression of the, or increases the endogenous expression of the functional, cystic fibrosis transmembrane conductance regulator (CFTR) protein (see WO2022/204270A1, which is herein incorporated by reference). Alternatively, by way of example, the disease or condition is PCD and the polynucleotide molecule present in the formulation according to the present invention increases the endogenous expression of a, or increases the endogenous expression of a functional, PCD- associated protein, such as dynein axonemal intermediate chain 1 (see WO2022/198099A1 and WO2022/204215A1, which are herein incorporated by reference). Decreasing or Silencing Gene Expression Alternatively, a polynucleotide molecule, in particular an miRNA or siRNA molecule, may be intended to down-regulate, reduce, silence or knock-down expression of an endogenous gene, in particular when said gene, and consequently the encoded protein (or other gene product), is over-expressed and said over-expression is contributing to cellular dysfunction, or when said gene, and consequently the encoded protein (or other gene product) is defective or dysfunctional and therefore contributing to cellular dysfunction. Notably, a polynucleotide molecule, in particular an miRNA or siRNA molecule, may be intended to down-regulate, reduce, silence or knock-down expression of an endogenous gene in order to reduce endogenous gene expression such that under-expression of the protein (or other gene product) interferes with other endogenous cellular processes such as the regulation of gene expression or signal transduction. Moreover, under-expression of the protein (or other gene product) may be intended to activate an immune response or stimulate the immune system, for example by removal of “self-antigens”. Therefore, in one embodiment there is provided an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the aqueous liquid pharmaceutical formulation decreases the endogenous expression of a protein (or other gene product). As used herein, the term “decreases” includes restoring i.e. decreasing from a high value to a “normal” value and impairing i.e., decreasing from a “normal” value to a low value or zero e.g. silencing, endogenous gene expression. Accordingly, in one embodiment, an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the polynucleotide molecule is a polynucleotide molecule which decreases the endogenous expression of a protein (or other gene product), is for use in the treatment of a disease or condition which is treated by decreasing the endogenous expression of a protein (or other gene product). In an alternative embodiment, the present invention provides a method for the treatment of a disease or condition, which is treated by decreasing the endogenous expression of a protein 58    (or other gene product), comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which decreases the endogenous expression of a protein (or other gene product). Moreover, the present invention provides use of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which decreases the endogenous expression of a protein (or other gene product), in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by decreasing the endogenous expression of a protein (or other gene product). In one embodiment, a disease or condition treated by decreasing the endogenous expression of a protein (or other gene product) is an infectious disease. The infectious disease is suitably bacterial, fungal, parasitic, or viral in origin. In particular, the infectious disease is infection by a virus or disease associated with infection with such a virus. Suitably, virus infects the respiratory tract and the disease associated with viral infection is a disease of the respiratory tract. Modifying Endogenous Nucleic Acid Sequences Alternatively, a polynucleotide molecule, in particular a DNA molecule, may be intended to modify, for example repair, excise, or insert or exchange bases or stretches of, endogenous nucleic acid or polynucleotide sequences, for example RNA or DNA sequences. For example, the polynucleotide molecule which is intended to modify endogenous nucleic acid or polynucleotide sequences may be a guide RNA (gRNA), which may function as a component of the CRISPR-Cas9 gene editing technology. Therefore, in one embodiment, there is provided an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the aqueous liquid pharmaceutical formulation modifies an endogenous nucleic acid sequence, for example an mRNA molecule or the genome. For example, the aqueous liquid pharmaceutical formulation may repair, excise bases or stretches from, insert bases or stretches into, or exchange bases or stretches with, an endogenous nucleic acid sequence, e.g., the genome. Therefore, in one embodiment, an aqueous liquid pharmaceutical formulation for use according to the present invention, wherein the polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence, is for use in the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence. Alternatively, in one embodiment, there is provided a method for the treatment of a disease or condition, which is treated by modifying an endogenous nucleic acid sequence, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation as described herein, wherein the 59    polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence. Furthermore, the present invention provides use of an aqueous liquid pharmaceutical formulation as described herein, wherein the polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence. By way of these disparate mechanisms, a polynucleotide molecule can interact with, interfere with, modify or otherwise influence essentially any cellular process. Ocular Indications Suitably, the aqueous liquid pharmaceutical formulation according to the present invention may be for use as a medicament for administration ocularly, such as by administration topically to the eye. When the aqueous liquid pharmaceutical formulation according to the present invention is for use as a medicament for administration ocularly, it is suitably for use in the prevention or treatment of an ocular disease or condition, for example an ocular disease or condition selected from dry eye–Sjogren’s syndrome, Meesmann epithelial corneal dystrophy, herpes simplex keratitis (HSK), mucopolysaccharidosis (MPS), ectrodactyly-ectodermal dysplasia- clefting (EEC) syndrome, ocular hypertension and open angle glaucoma, retinoschisis, choroideremia, achromatopsia, recurrent retinoblastoma. Also provided is a method for the prevention or treatment of an ocular disease or condition, for example an ocular disease or condition selected from dry eye–Sjogren’s syndrome, Meesmann epithelial corneal dystrophy, herpes simplex keratitis (HSK), mucopolysaccharidosis (MPS), ectrodactyly-ectodermal dysplasia-clefting (EEC) syndrome, ocular hypertension and open angle glaucoma, retinoschisis, choroideremia, achromatopsia, recurrent retinoblastoma, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation according to the present invention. Similarly, there is provided the use of an aqueous liquid pharmaceutical formulation according to the present invention in the manufacture of a medicament for use in the treatment of an ocular disease or condition, for example an ocular disease or condition selected from dry eye–Sjogren’s syndrome, Meesmann epithelial corneal dystrophy, herpes simplex keratitis (HSK), mucopolysaccharidosis (MPS), ectrodactyly-ectodermal dysplasia-clefting (EEC) syndrome, ocular hypertension and open angle glaucoma, retinoschisis, choroideremia, achromatopsia, recurrent retinoblastoma. 60    Production Methods Formulations of the invention may be produced by mixing the ingredients leading to a colloidal emulsion. Suitably, an aqueous liquid pharmaceutical formulation of the present invention is produced by a stepwise process as described below. It will be understood that the skilled person would be aware of minor modifications to the process which may nevertheless lead to a process suitable for the production of an aqueous liquid pharmaceutical formulation as described herein. Moreover, the skilled person would be able to determine optimised parameters for the process described below, for example temperature, mixing time, pH, based on the surfactant component, polynucleotide molecule, and concentrations thereof etc. Therefore, in one embodiment, there is provided a process for the production or preparation of an aqueous liquid pharmaceutical formulation, or immunostimulatory composition, immunogenic composition, or vaccine composition according to the present invention. Suitably, the process may comprise, consist of, or consist essentially of one or more of the steps described below. Suitably, firstly, the surfactant component, which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, for example a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, is added at the required concentration to water, for example RNAse free water, and mixed using an overhead stirrer at between 25% and 75% power, for example at between 40% and 50% power, or at about 50% power. Mixing continues for any period of time until a homogenous surfactant component mixture is achieved, such as for between about 1-60 minutes, for example for between about 1-30 minutes, for example for between about 5-20 minutes, such as for about 10 minutes. An exemplary overhead stirrer is the IKA EURO-STAR Overhead Lab Mixer. Secondly, a specific volume of polynucleotide molecule stock solution, which typically comprises the polynucleotide molecule at a concentration of about 0.2 – 20% (w/w), wherein the % by weight is with respect to the total weight of the formulation, is dispersed into the homogenous surfactant component mixture described above in order to achieve a desired final concentration of polynucleotide molecule. The formulation is then mixed using an overhead stirrer at between 25% and 75% power, for example at between 40% and 50% power, or at about 50% power, for any period of time until a uniform active surfactant component mixture is produced. Suitably mixing may occur for between about 1-60 minutes, for example for between about 1-30 minutes, for example for between about 5-20 minutes, such as for about 10 minutes. In a variant process, if it is intended that the formulation contains further pharmaceutically acceptable excipients, these may be added, either simultaneously, stepwise, or a combination 61    thereof, at the required concentration to water, for example RNAse free water and homogenised until the mixture achieves a desired rheology. For example, homogenisation may occur at an rpm (revolutions per minutes) of 2000-20000, such as 2000-15000, 4000- 12000, or 5000-10000 for between about 1-20 minutes, for example for between about 1-10 minutes, such as for between about 1-5 minutes. Next, the further pharmaceutically acceptable excipient composition will be combined with the active surfactant component mixture described above and mixed using an overhead stirrer at between 25% and 75% power, for example at between 40% and 50% power, or at about 50% power. Mixing will continue for any period of time until a homogenous and uniform aqueous liquid pharmaceutical formulation is produced. Suitably mixing may occur for between about 1-60 minutes, for example for between about 1-30 minutes, for example for between about 5-20 minutes, such as for about 10 minutes. An exemplary homogeniser is the Silverson L5M homogenizer. It will be understood by the skilled person that the further pharmaceutically acceptable excipients included in an aqueous liquid pharmaceutical formulation of the present invention will depend on the surfactant component, polynucleotide molecule, and administration route. By way of example, an aqueous liquid pharmaceutical formulation suitable for topical administration to the nose may comprise carboxymethyl cellulose, for example at a concentration of about 2% (w/w), hyaluronic acid, for example at a concentration of about 1% (w/w), and glycerol, for example at a concentration of about 2.1% (w/w), wherein the % by weight is with respect to the total weight of the formulation. Therefore, it is these excipients that may be present in the further pharmaceutically acceptable excipient composition described above. Finally, the aqueous liquid pharmaceutical formulation will be buffered to a final desired pH, for example using a buffer such as a citrate buffer (e.g. comprising 0.2% (w/w) citric acid and 0.28% (w/w) sodium citrate, wherein the % by weight is with respect to the total weight of the formulation) which would be suitable for achieving a pH of between about 4.0 and 6.0, such as about 5.0). The buffered aqueous liquid pharmaceutical formulation will be mixed, for example using an overhead stirrer as described above, until unform pH adjustment is achieved. For example, mixing may occur for between about 1-60 minutes, for example for between about 1-30 minutes, for example for between about 5-20 minutes, such as for about 10 minutes. Suitably, each of the steps described above is performed at room temperature (i.e. between about 20 and 30 oC). 62    The invention is further defined by the following clauses: Clause 1. An aqueous liquid pharmaceutical formulation comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule. Clause 2. The aqueous liquid pharmaceutical formulation according to clause 1, wherein the fatty acid is selected from the group consisting of arachidic acid, arachidonic acid, caprylic acid, lauric acid, linoleic acid, linolenic acid, myristic acid, myristoleic acid, oleic acid, palmitic acid, palmitoleic acid, sapienic acid, stearic acid, and vaccenic acid. Clause 3. The aqueous liquid pharmaceutical formulation according to clause 2, wherein the fatty acid is selected from caprylic acid and oleic acid. Clause 4. The aqueous liquid pharmaceutical formulation according to clause 3, wherein the fatty acid is caprylic acid. Clause 5. The aqueous liquid pharmaceutical formulation according to clause 3, wherein the fatty acid is oleic acid. Clause 6. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 5, wherein the non-ionic surfactant is selected from the group consisting of polyoxyalkylenes, such as poloxamers, alkyl ethers of polyethylene glycol, alkylphenyl ethers of polyethylene glycol, and fatty acid esters, such as polyoxyethylene sorbitan fatty acid esters. Clause 7. The aqueous liquid pharmaceutical formulation according to clause 6, wherein the non-ionic surfactant is selected from alkyl ethers of polyethylene glycol and polyoxyethylene sorbitan fatty acid esters. Clause 8. The aqueous liquid pharmaceutical formulation according to clause 7, wherein the non-ionic surfactant is an alkyl ether of polyethylene glycol. Clause 9. The aqueous liquid pharmaceutical formulation according to clause 8, wherein the non-ionic surfactant is Brij 35 (polyoxyethylene (23) lauryl ether), Brij 52 (polyoxyethylene (20) cetyl ether), Brij 93 (polyoxyethylene (2) oleyl ether), Brij 97 (polyoxyethylene (10) oleyl ether), Brij L4 (polyoxyethylene (4) lauryl ether), Brij 30 (polyoxyethylene (4) lauryl ether), or Brij 78 (polyoxyethylene(20) stearyl ether). Clause 10. The aqueous liquid pharmaceutical formulation according to clause 9, wherein the non-ionic surfactant is Brij 35 (polyoxyethylene (23) lauryl ether). Clause 11. The aqueous liquid pharmaceutical formulation according to clause 7, wherein the non-ionic surfactant is a polyoxyethylene sorbitan fatty acid ester. Clause 12. The aqueous liquid pharmaceutical formulation according to clause 11, wherein the non-ionic surfactant is polysorbate 80, polysorbate 120, polysorbate 85, polysorbate 65, polysorbate 60, polysorbate 40, or polysorbate 20. Clause 13. The aqueous liquid pharmaceutical formulation according to clause 12, wherein the non-ionic surfactant is polysorbate 80. 63    Clause 14. The aqueous liquid pharmaceutical formulation according to clause 1, wherein the surfactant component is selected from the group consisting of mixtures of (a) oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (b) lauric acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (c) linoleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (d) linolenic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (e) palmitic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (f) stearic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (g) oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyalkylene, such as a poloxamer, (h) oleic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol, (i) oleic acid or a pharmaceutically acceptable salt thereof and an alkylphenyl ether of polyethylene glycol, (j) caprylic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (k) caprylic acid or a pharmaceutically acceptable salt thereof and a polyoxyalkylene, such as a poloxamer, (l) caprylic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol, and (m) caprylic acid or a pharmaceutically acceptable salt thereof and an alkylphenyl ether of polyethylene glycol. Clause 15. The aqueous liquid pharmaceutical formulation according to clause 14, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester. Clause 16. The aqueous liquid pharmaceutical formulation according to clause 15, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester selected from polysorbate 80 and polysorbate 20. Clause 17. The aqueous liquid pharmaceutical formulation according to clause 16, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and polysorbate 80. Clause 18. The aqueous liquid pharmaceutical formulation according to clause 14, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester. Clause 19. The aqueous liquid pharmaceutical formulation according to clause 18, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester selected from polysorbate 80 and polysorbate 20. 64    Clause 20. The aqueous liquid pharmaceutical formulation according to clause 19, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and polysorbate 80. Clause 21. The aqueous liquid pharmaceutical formulation according to clause 14, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol. Clause 22. The aqueous liquid pharmaceutical formulation according to clause 21, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol selected from Brij 35 (polyoxyethylene (23) lauryl ether), Brij 52 (polyoxyethylene (20) cetyl ether), Brij 93 (polyoxyethylene (2) oleyl ether), Brij 97 (polyoxyethylene (10) oleyl ether), Brij L4 (polyoxyethylene (4) lauryl ether), Brij 30 (polyoxyethylene (4) lauryl ether), or Brij 78 (polyoxyethylene(20) stearyl ether). Clause 23. The aqueous liquid pharmaceutical formulation according to clause 22, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and Brij 35 (polyoxyethylene (23) lauryl ether). Clause 24. The aqueous liquid pharmaceutical formulation according to clause 14, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol. Clause 25. The aqueous liquid pharmaceutical formulation according to clause 24, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol selected from Brij 35 (polyoxyethylene (23) lauryl ether), Brij 52 (polyoxyethylene (20) cetyl ether), Brij 93 (polyoxyethylene (2) oleyl ether), Brij 97 (polyoxyethylene (10) oleyl ether), Brij L4 (polyoxyethylene (4) lauryl ether), Brij 30 (polyoxyethylene (4) lauryl ether), or Brij 78 (polyoxyethylene(20) stearyl ether). Clause 26. The aqueous liquid pharmaceutical formulation according to clause 25, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and Brij 35 (polyoxyethylene (23) lauryl ether). Clause 27. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 26, wherein the fatty acid is in the form of the free acid. Clause 28. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 27, wherein the concentration of the surfactant component in the formulation is 0.2 – 30000 µg/mL, for example 1 – 30000 µg/mL, for example 1 – 20000 µg/mL, 5 – 20000 µg/mL, 5 – 15000 µg/mL, 5 – 10000 µg/mL, or 5-5000 µg/mL. Clause 29. The aqueous liquid pharmaceutical formulation according to clause 28, wherein the concentration of the surfactant component in the formulation is 1 – 3000 µg/mL, for example 1 – 2000 µg/mL, 5 – 2000 µg/mL, 5 – 1500 µg/mL, 5 – 1000 µg/mL, or 5-500 µg/mL. 65    Clause 30. The aqueous liquid pharmaceutical formulation according to clause 29, wherein the concentration of the surfactant component in the formulation is 50-200 µg/mL, for example 75-150 µg/mL, for example 90 – 120 µg/mL, or about 100 µg/mL. Clause 31. The aqueous liquid pharmaceutical formulation according to clause 28, wherein the concentration of the surfactant component in the formulation is 500-2000 µg/mL, for example 750-1500 µg/mL, for example 900 – 1200 µg/mL, or about 1000 µg/mL. Clause 32. The aqueous liquid pharmaceutical formulation according to clause 28, wherein the fatty acid is present in the formulation at a concentration of 10 – 100 ug/mL, for example 20 – 80 µg/mL, for example 25 – 75 µg/mL, for example 40 – 60 µg/mL, or about 50 µg/mL; and wherein the non-ionic surfactant is present in the formulation at a concentration of 10 – 100 ug/mL, for example 20 – 80 µg/mL, for example 25 – 75 µg/mL, for example 30 – 60 µg/mL, for example 40 – 50 µg/mL. Clause 33. The aqueous liquid pharmaceutical formulation according to clause 28, wherein the fatty acid is present in the formulation at a concentration of 100 – 1000 ug/mL, for example 200 – 800 µg/mL, for example 250 – 750 µg/mL, for example 400 – 600 µg/mL, or about 500 µg/mL; and wherein the non-ionic surfactant is present in the formulation at a concentration of 100 – 1000 ug/mL, for example 200 – 800 µg/mL, for example 250 – 750 µg/mL, for example 300 – 600 µg/mL, for example 400 – 500 µg/mL. Clause 34. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 33, wherein the ratio of the amount of fatty acid or a pharmaceutically acceptable salt thereof to non-ionic surfactant, wherein each is measured in µg/mL, is between about 5:1 and about 1:5, between about 5:1 and about 1:2, between about 4:1 and about 1:2, or between about 2:1 and about 1:2. Clause 35. The aqueous liquid pharmaceutical formulation according to clause 34, wherein the ratio of the amount of fatty acid or a pharmaceutically acceptable salt thereof to non-ionic surfactant, wherein each is measured in µg/mL, is between about 3:2 and about 2:3, for example between about 6:5 and about 1:1, e.g. about 10:9 or about 11:10. Clause 36. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 35, wherein the ratio of the amount of the surfactant component to polynucleotide molecule, wherein each is measured in µg/mL, is between about 100:1 and about 1:1000, between about 100:1 and about 1:750, between about 75:1 and about 1:750, between about 50:1 and about 1:750, between about 50:1 and about 1:500, between about 50:1 and about 1:250, between about 50:1 and about 1:200, between about 40:1 and about 1:200, between about 40:1 and about 1:150, between about 25:1 and about 1:150, for example between about 25:1 and about 1:100. 66    Clause 37. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 36, wherein the polynucleotide molecule is a single-stranded polynucleotide molecule. Clause 38. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 36, wherein the polynucleotide molecule is a double-stranded polynucleotide molecule. Clause 39. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 38, wherein the polynucleotide molecule comprises an intramolecular structure. Clause 40. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 39, wherein the polynucleotide molecule is a ribonucleic acid (RNA) molecule. Clause 41. The aqueous liquid pharmaceutical formulation according to clause 40, wherein the RNA molecule is an mRNA, miRNA, shRNA or siRNA molecule. Clause 42. The aqueous liquid pharmaceutical formulation according to clause 41, wherein the RNA molecule is an mRNA molecule. Clause 43. The aqueous liquid pharmaceutical formulation according to clause 42, wherein the mRNA molecule comprises from about 100 to about 10000 nucleotides, such as from about 200 to about 8000 nucleotides, such as from about 500 to about 7500 nucleotides, such as from about 1000 to about 5000 nucleotides. Clause 44. The aqueous liquid pharmaceutical formulation according to clause 41, wherein the RNA molecule is an miRNA molecule. Clause 45. The aqueous liquid pharmaceutical formulation according to clause 44, wherein the miRNA molecule comprises from about 20 to about 25 nucleotides. Clause 46. The aqueous liquid pharmaceutical formulation according to clause 45, wherein the miRNA molecule comprises 20, 21, 22, 23, 24 or 25 nucleotides. Clause 47. The aqueous liquid pharmaceutical formulation according to clause 41, wherein the RNA molecule is an shRNA molecule. Clause 48. The aqueous liquid pharmaceutical formulation according to clause 47, wherein the shRNA molecule comprises from about 10 to about 70 nucleotides, for example from about 20 to about 70 nucleotides, for example from about 35 to about 70 nucleotides or from about 25 to about 35 nucleotides. Clause 49. The aqueous liquid pharmaceutical formulation according to clause 48, wherein the shRNA molecule comprises 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 nucleotides. Clause 50. The aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 49, wherein the shRNA molecule comprises a double-stranded region comprising 30 base pairs or less, such as 25 base pairs or less, such as 20 base pairs or less, such as 18 base pairs or less, such as 16 base pairs or less, such as 14 base pairs or less, such as 12 base pairs or less, such as 10 base pairs or less, such as 8 base pairs or less, such as 6 base pairs or less, such as 4 base pairs or less. 67    Clause 51. The aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 50, wherein the shRNA molecule comprises a double-stranded region comprising one or more mispaired bases. Clause 52. The aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 51, wherein the shRNA molecule comprises a blunt end. Clause 53. The aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 52, wherein the shRNA molecule comprises at least one 5’ triphosphate or at least one 5’ diphosphate, in particular wherein the 5’ triphosphate or 5’ diphosphate is located at the 5’ terminus. Clause 54. The aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 53, wherein the shRNA molecule comprises (i) a blunt end, (ii) a 5’ triphosphate or 5’ diphosphate moiety located at the 5’ terminus, and (iii) a double-stranded region between about 10 and about 18 nucleotides in length. Clause 55. The aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 54, wherein the shRNA molecule comprises or consists of SEQ ID NO.1 or a variant of SEQ ID NO.1. Clause 56. The aqueous liquid pharmaceutical formulation according to any one of clauses 47 to 54, wherein the shRNA molecule comprises or consists of SEQ ID NO.2, SEQ ID NO. 3, SEQ ID NO.4, SEQ ID NO.5, SEQ ID NO.6, SEQ ID NO.7, SEQ ID NO.8, SEQ ID NO. 9 or SEQ ID NO. 10, or a variant of SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 4, SEQ ID NO.5, SEQ ID NO.6, SEQ ID NO.7, SEQ ID NO.8, SEQ ID NO.9 or SEQ ID NO.10. Clause 57. The aqueous liquid formulations according to any one of clauses 54 to 56, wherein the shRNA molecule is capable of inducing an interferon response in a vertebrate cell. Clause 58. The aqueous liquid pharmaceutical formulation according to clause 41, wherein the RNA molecule is an siRNA molecule. Clause 59. The aqueous liquid pharmaceutical formulation according to clause 58, wherein the siRNA molecule comprises from about 20 to about 60 nucleotides, for example from about 30 to about 60 nucleotides, such as from about 40 to about 50 nucleotides. Clause 60. The aqueous liquid pharmaceutical formulation according to clause 59, wherein the siRNA molecule comprises 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, or 60 nucleotides. Clause 61. The aqueous liquid pharmaceutical formulation according to any one of clauses 58 to 60, wherein the siRNA molecule comprises a double-stranded region comprising 30 base pairs or less, such as 25 base pairs or less, such as 20 base pairs or less. 68    Clause 62. The aqueous liquid pharmaceutical formulation according to any one of clauses 58 to 61, wherein the siRNA molecule comprises a double-stranded region comprising one or more mispaired bases. Clause 63. The aqueous liquid pharmaceutical formulation according to any one of clauses 58 to 62, wherein the siRNA molecule comprises one or more, e.g. two, 3’ overhangs. Clause 64. The aqueous liquid pharmaceutical formulation according to any one of clauses 58 to 63, wherein the siRNA molecule comprises at least one, e.g. two, 5’ monophosphate(s), or at least one, e.g. two, 5’ diphosphate(s), or at least one, e.g. two, 5’ triphosphate(s). Clause 65. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 39, wherein the polynucleotide molecule is a deoxyribonucleic acid (DNA) molecule. Clause 66. The aqueous liquid pharmaceutical formulation according to clause 65, wherein the DNA molecule is a genomic DNA (gDNA) molecule, a complementary DNA (cDNA) molecule, or an extra-chromosomal DNA molecule, for example a plasmid DNA molecule. Clause 67. The aqueous liquid pharmaceutical formulation according to clause 66, wherein the DNA molecule is a gDNA molecule. Clause 68. The aqueous liquid pharmaceutical formulation according to clause 66, wherein the DNA molecule is a cDNA molecule. Clause 69. The aqueous liquid pharmaceutical formulation according to clause 66, wherein the DNA molecule is an extra-chromosomal DNA molecule, for example a plasmid DNA molecule. Clause 70. The aqueous liquid pharmaceutical formulation according to any one of clauses 65 to 69, wherein the DNA molecule comprises from about 100 to about 20000 nucleotides, such as from about 100 to about 15000 nucleotides, such as from about 500 to about 15000 nucleotides, such as from about 500 to about 10000 nucleotides, such as from about 2500 to about 10000 nucleotides, such as from about 2500 to about 8000 nucleotides. Clause 71. The aqueous liquid pharmaceutical formulation according to clause 1, wherein the polynucleotide molecule has the structure of Formula (I): 5’-Pz-(N)bN-3’-(E)y(E)-L-(E)(E)y’-5’-N(N)b’-3’ wherein 5’-Pz-(N)bN-3’ represents the first nucleic acid sequence; 5’-N(N)b’-3’ represents the second nucleic acid sequence; P at each instance is independently a phosphate or analog thereof; z is 2 or 3; N is, at each instance, any nucleotide or modified nucleotide or analog or derivative thereof; b and b’ are independently 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18; 5’-(E)y(E)-L-(E)(E)y’-3’ represents the connector element wherein 69    E at each occurrence is independently any nucleotide, modified nucleotide, or abasic; y and y’ are independently 0-9, provided that y + y’ equals 0-8; L is a non-nucleotide segment having the structure O X P V Y k W d q M t V' x' O P Y' wherein X and X’ are independently O or S; Y and Y’ are independently OR”, SR”, or NRR’; V and V’ are independently O, S, or NRR’; q is 1-20; k is 1-20; t is 1-20; M selected from aliphatic, substituted aliphatic, aryl, substituted aryl, heteroalkyl, heterocyclyl or substituted heterocyclyl; W is any reactive group; and d is 0 or 1. Clause 72. The aqueous liquid pharmaceutical formulation according to clause 71, wherein the polynucleotide molecule has the structure of Formula (II): 5’-Pz-Nu-3’-(E)y(E)-L-(E)(E)y’-5’-Nu’-3’ wherein 5’-Pz-Nu-3’ represents the first nucleic acid sequence; 5’-Nu’-3’ represents the second nucleic acid sequence; P at each instance is independently a phosphate or analog thereof. z is 0, 1, 2, or 3; 5’-(E)y(E)-L-(E)(E)y’-3’ represents the connector element wherein E at each occurrence is independently any nucleotide, modified nucleotide, or abasic; 70    y and y’ are independently 0-9, provided that y + y’ equals 0-8; L is a non-nucleotide segment having the structure O X
Figure imgf000073_0001
X and X’ are independently O or S; Y and Y’ are independently OR”, SR”, or NRR’; V and V’ are independently O, S, or NRR’; q is 1-20; k is 1-20; t is 1-20; M selected from aliphatic, substituted aliphatic, aryl, substituted aryl, heteroalkyl, heterocyclyl or substituted heterocyclyl; W is any reactive group or conjugation group; and d is 0 or 1. Clause 73. The aqueous liquid pharmaceutical formulation according to clause 1, wherein the polynucleotide molecule comprises, consists of, or consists essentially of SEQ ID NO.13 and SEQ ID NO.14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L1. Clause 74. The aqueous liquid pharmaceutical formulation according to clause 1, wherein the polynucleotide molecule comprises, consists of, or consists essentially of SEQ ID NO.13 and SEQ ID NO.14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L2. Clause 75. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 74, wherein the polynucleotide molecule comprises one or more sugar-modified nucleotides which each comprise a 2’ OH (or 2’ H) modification. 71    Clause 76. The aqueous liquid pharmaceutical formulation according to clause 75, wherein the 2’ OH (or 2’ H) modification is selected from the group consisting of 2'-deoxy, 2’-fluoro, 2'- deoxy- methoxyethyl (2'-O-MOE), 2'-O-amino-propyl (2'-O-AP), 2'- O- , 2'-O-dimethylaminopropyl (2'-O-DMAP), 2'-O- DMAEOE), and 2'-O-N-methylacetamido (2'-O-NMA). Clause pharmaceutical formulation according to any one of clauses 1 to molecule comprises one or more backbone-modified the backbone modification is replacement of the with a phosphorothioate group. Clause pharmaceutical formulation according to any one of clauses 1 to molecule comprises one or more base-modified Clause pharmaceutical formulation according to any one of clauses 1 to
Figure imgf000074_0001
pharmaceutical formulation is in the form of a stable colloidal emulsion. Clause 80. An aqueous liquid pharmaceutical formulation in the form of a stable colloidal emulsion comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule. Clause 81. The aqueous liquid pharmaceutical formulation according to clause 79 or clause 80, wherein the average particle size of the stable colloidal particles is between about 10 and about 1000 nm, such as between about 50 and about 1000 nm, such as between about 50 and about 750 nm, such as between about 50 and about 500 nm, for example between about 50 and about 400 nm, for example between about 50 and about 300 nm Clause 82. The aqueous liquid pharmaceutical formulation according to clause 81, wherein the average particle size of the stable colloidal particles is between about 100 and about 300 nm. Clause 83. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 82, wherein the aqueous liquid pharmaceutical formulation is suitable for oral, inhalational, sub-lingual, buccal, or parenteral, including intravenous, subcutaneous, topical, transdermal, pulmonary, rectal, vaginal, ocular, intranasal, intradermal, transmucosal, intraperitoneal, intramuscular, intracapsular, intraorbital, intracardiac, transtracheal, subcuticular, intraarticular, intraspinal, or intrasternal administration. Clause 84. The aqueous liquid pharmaceutical formulation according to clause 83, wherein the aqueous liquid pharmaceutical formulation is suitable for topical administration to the lung or nose. 72    Clause 85. The aqueous liquid pharmaceutical formulation according to clause 83, wherein the aqueous liquid pharmaceutical formulation is suitable for subcutaneous administration, e.g. subcutaneous injection. Clause 86. The aqueous liquid pharmaceutical formulation according to clause 83, wherein the aqueous liquid pharmaceutical formulation is suitable for ocular administration, e.g. intra- ocular administration or topical administration to the eye. Clause 87. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86 for use as a medicament. Clause 88. The aqueous liquid pharmaceutical formulations for use according to clause 87, for administration topically to the lung (e.g. by oral inhalation) or topically to the nose. Clause 89. The aqueous liquid pharmaceutical formulations for use according to clause 87, for administration subcutaneously. Clause 90. The aqueous liquid pharmaceutical formulations for use according to clause 87, for administration ocularly, for example intra-ocularly or topically to the eye. Clause 91. The aqueous liquid pharmaceutical formulation for use according to any one of clauses 87 to 90, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response, for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response. Clause 92. A method for the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response. Clause 93. Use of an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response. Clause 94. The aqueous liquid pharmaceutical formulation for use, method, or use, according to any one of clauses 91 to 93, wherein the disease or condition is infection by a virus or associated with infection with such a virus. 73    Clause 95. The aqueous liquid pharmaceutical formulation for use, method, or use, according to clause 94, wherein the virus infects the respiratory tract and the disease associated with infection is a disease of the respiratory tract. Clause 96. The aqueous liquid pharmaceutical formulation for use, method, or use, according to clause 95, wherein the virus is SARS-CoV-2 and the disease associated with viral infection is COVID-19; or wherein the virus is seasonal coronavirus, for example 229E, NL63, OC43, or HKU1, and the disease associated with viral infection is the disease associated with seasonal coronavirus, for example 229E, NL63, OC43, or HKU1, infection; or wherein the virus is influenza virus and the disease associated with viral infection is influenza; or wherein the virus is respiratory syncytial virus (RSV) and the disease associated with viral infection is the disease associated with RSV infection; or wherein the virus is human rhinovirus (HRV) and the disease associated with viral infection is the disease associated with HRV infection; or wherein the virus is Middle East respiratory syndrome (MERS)-CoV and the disease associated with viral infection is MERS; or wherein the virus is an avian influenza virus and the disease associated with viral infection is avian influenza; or wherein the virus is Nipah virus and the disease associated with viral infection is the disease associated with Nipah virus infection; or wherein the virus is a human parainfluenza virus (HPIV) and the disease associated with viral infection is the disease associated with HPIV infection; or wherein the virus is a human metapneumovirus (hMPV) and the disease associated with viral infection is the disease associated with hMPV infection. Clause 97. The aqueous liquid pharmaceutical formulation for use according to any one of clauses 87 to 90, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein or other gene product, or which increases endogenous expression of a functional protein or other gene product, for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein or other gene product, or by increasing the expression of a functional protein or other gene product. Clause 98. A method for the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein or other gene product, or by increasing the expression of a functional protein or other gene product, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the 74    polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein or other gene product, or which increases endogenous expression of a functional protein or other gene product. Clause 99. Use of an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein or other gene product, or which increases endogenous expression of a functional protein or other gene product, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein or other gene product, or by increasing the expression of a functional protein or other gene product. Clause 100. The aqueous liquid pharmaceutical formulation for use, method, or use, according to any one of clauses 97 to 99, wherein the disease or condition is a respiratory disease or condition, such as asthma, alpha-1 antitrypsin deficiency, chronic obstructive pulmonary disease (COPD), primary ciliary dyskinesia (PCD), pulmonary fibrosis, sarcoidosis or cystic fibrosis. Clause 101. The aqueous liquid pharmaceutical formulation for use according to any one of clauses 87 to 90, for use in a method of gene therapy, wherein the polynucleotide molecule, in particular an mRNA or DNA molecule, encodes a therapeutic gene, protein or other gene product. Clause 102. The aqueous liquid pharmaceutical formulation for use according to clause 101, for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of the therapeutic gene, protein or other gene product encoded by the polynucleotide molecule. Clause 103. The aqueous liquid pharmaceutical formulation for use according to any one of clauses to 87 to 90, wherein the polynucleotide molecule is a polynucleotide molecule which decreases the endogenous expression of a protein or other gene product, for use in the treatment of a disease or condition which is treated by decreasing the endogenous expression of protein or other gene product. Clause 104. A method for the treatment of a disease or condition which is treated by decreasing the endogenous expression of a protein or other gene product, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which decreases the endogenous expression of a protein or other gene product. Clause 105. Use of an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which decreases the endogenous expression of a protein or other gene product, in the manufacture 75    of a medicament for use in the treatment of a disease or condition which is treated by decreasing the endogenous expression of a protein or other gene product. Clause 106. The aqueous liquid pharmaceutical formulation for use according to any one of clauses 87 to 90, wherein the polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence, for use in the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence. Clause 107. A method for the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence. Clause 108. Use of an aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the polynucleotide molecule is a polynucleotide molecule which modifies an endogenous nucleic acid sequence, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by modifying an endogenous nucleic acid sequence. Clause 109. An immunostimulatory composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) an immunostimulatory polynucleotide molecule. Clause 110. An immunostimulatory composition according to clause 109, wherein the immunostimulatory polynucleotide molecule is an shRNA molecule. Clause 111. An immunostimulatory composition according to clause 109 or clause 110, for use in stimulating or activating the immune system in a subject. Clause 112. An immunostimulatory composition according to clause 111, for use in stimulating or activating an anti-viral innate and/or adaptive immune response in a subject. Clause 113. An immunogenic composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule which encodes an immunogen. Clause 114. A vaccine composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule which encodes a vaccine immunogen. 76    Clause 115. An immunogenic composition or vaccine composition according to clause 113 or clause 114, wherein the polynucleotide molecule which encodes an immunogen or vaccine immunogen is an mRNA molecule or DNA molecule. Clause 116. An immunogenic composition or vaccine composition according to any one of clauses 113 to 115, further comprising an immunostimulatory polynucleotide molecule. Clause 117. An immunogenic composition or vaccine composition according to clause 116, wherein the immunostimulatory polynucleotide molecule is an shRNA molecule. Clause 118. An immunogenic composition or vaccine composition according to any one of clauses 113 to 117, wherein the composition does not comprise a lipid nanoparticle (LNP) or a liposome, and/or wherein the composition is substantially free of LNP and liposome components. Clause 119. An immunogenic composition or vaccine composition according to any one of clauses 113 to 118, for use in raising an immune response in a subject, wherein the immune response is raised against the immunogen or vaccine immunogen encoded by the polynucleotide molecule. Clause 120. An immunogenic composition or vaccine composition for use according to clause 119, for use therapeutically i.e. for use in raising an immune response to have a curative effect on a disease or condition treated by the raising of an innate and/or adaptive immune response. Clause 121. An immunogenic composition or vaccine composition for use according to clause 120 where the disease or condition is cancer. Clause 122. An immunogenic composition or vaccine composition for use according to clause 119, for use prophylactically i.e. for use in raising an immune response to have a protective effect against a disease or condition treated by the raising of an innate and/or adaptive immune response. Clause 123. An immunogenic composition or vaccine composition for use according to clause 122 where the disease or condition is an infectious disease. Clause 124. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a protein. Clause 125. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a cationic lipopeptide, for example the aqueous liquid pharmaceutical formulation does not comprise polymyxin B. Clause 126. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise an inorganic nanoparticle. 77    Clause 127. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a lipid nanoparticle (LNP) or a liposome. Clause 128. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a neutral lipid, for example the aqueous liquid pharmaceutical formulation does not comprise cholesterol or an analogue thereof. Clause 129. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a cationic lipid. Clause 130. The aqueous liquid pharmaceutical formulation according to any one of clauses 1 to 86, wherein the aqueous liquid pharmaceutical formulation does not comprise a helper lipid, for example the aqueous liquid pharmaceutical formulation does not comprise dioleoylphosphatidylethanolamine (DOPE) or phosphatidylcholine. Clause 131. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers, and (iv) a diluent. Clause 132. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers selected from citrate, acetate, lactate, formate, and phosphate, and (iv) a diluent selected from isotonic saline (0.9% w/v), isotonic dextrose (5% w/v), isotonic mixtures of saline and dextrose (e.g. saline (0.45 % w/v) and dextrose (2.5 % w/v)), sterile or purified water, sterile water for injection or bacteriostatic water for injection. Clause 133. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers selected from citrate and phosphate, and (iv) a diluent selected from isotonic saline (0.9% w/v) and sterile or purified water. Clause 134. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) citrate, and (iv) isotonic saline (0.9% w/v). Clause 135. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a 78    pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) citrate, and (iv) sterile or purified water. Clause 136. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) phosphate, and (iv) isotonic saline (0.9% w/v). Clause 137. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) phosphate, and (iv) sterile or purified water. Clause 138. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers, (iv) a diluent, (v) optionally, one or more suspending agents, (vi) optionally, one or more wetting or thickening agents, and (vii) optionally one or more osmotic or tonicity adjusting agents. Clause 139. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers selected from citrate and phosphate, (iv) a diluent selected from isotonic saline (0.9% w/v) and sterile or purified water, (v) optionally, one or more suspending agents, (vi) optionally, one or more wetting or thickening agents, and (vii) optionally one or more osmotic or tonicity adjusting agents. Clause 140: The aqueous liquid pharmaceutical formulation according to any one of clauses 131 to 139 in the form of a stable colloidal emulsion. Examples Abbreviations used herein are defined below (see Table 1). Any abbreviations not defined are intended to convey their generally accepted meaning. Table 1: Abbreviations ALI Air Liquid Interface BSA Bovine Serum Albumin CPE Cytopathic Effects DMEM Dulbecco’s Modified Eagles Medium GFP Green Fluorescent Protein HAE Human Airway Epithelium 79    hr Hour(s) IFN Interferon L litre MDCK Madin-Darby canine kidney cells MFI Mean Fluorescent Intensity min Minute(s) MOI Multiplicity of Infection MSD Meso Scale Discovery PBS Phosphate Buffered Saline PCR Polymerase Chain Reaction PFU Plaque-Forming Units PR8 A/Puerto Rico/8/1934 (H1N1) TPCK tosyl phenylalanyl chloromethyl ketone The structure of molecules used herein are illustrated below (see Table 2). Table 2: Structures of Polynucleotide Molecules dsRNA 1 5’pppGCAUGCGACCUCUGUUUGA-3’ SEQ ID NO.12 3’-CGUACGCUGGAGACAAACU-5’ dsRNA 2 polyinosinic-polycytidylic acid dsRNA between 200-8000 base Poly I:C pairs in length (CAS: 31852-29-6) shRNA 1 SEQ ID NO.1 shRNA 2 SEQ ID NO.2 RNA 5’-ppGGAUCGAUCGAUCGUU-L1- SEQ ID NO 13 SEQ ID NO 14 M A
Figure imgf000082_0001
80    F F n c T d, f
Figure imgf000083_0001
Stable Polysorbate 80 Active Ingredient Colloidal 0.045% 45 µg
Figure imgf000083_0002
The formulation may be administered intranasally via a nasal spray device, for example with a spray volume 100 µL suitable for 1 or 2 sprays per nostril. The formulation may be suitable 81    for the treatment or prevention of infection by a virus or disease associated with infection with such a virus. Formulation Example 1B: Example aqueous liquid pharmaceutical formulation comprising dsRNA 1 The following aqueous liquid pharmaceutical formulation at about pH 4.7 may be prepared,
Figure imgf000084_0001
Purified Water Diluent q.s. 85.92 mg The formulation may be administered intranasally via a nasal spray device, for example with a spray volume 100 µL suitable for 1 or 2 sprays per nostril. The formulation may be suitable 82    for the treatment or prevention of infection by a virus or disease associated with infection with such a virus. Formulation Example 1C: Example aqueous liquid pharmaceutical formulation comprising dsRNA 2 The following aqueous liquid pharmaceutical formulation at about pH 4.7 may be prepared, for example largely according to the protocol described in Biophysical Example 1:
Figure imgf000085_0001
The formulation may be administered intranasally via a nasal spray device, for example with a spray volume 100 µL suitable for 1 or 2 sprays per nostril. The formulation may be suitable 83    for the treatment or prevention of infection by a virus or disease associated with infection with such a virus. Formulation Example 1D: Example aqueous liquid pharmaceutical formulation comprising shRNA 1 The following aqueous liquid pharmaceutical formulation at about pH 7.2 may be prepared, for example largely according to the protocol described in Biophysical Example 1:
Figure imgf000086_0001
Stable shRNA 1 Active Ingredient Colloidal 2.00% 2000 µg Particle 84    Stable Oleic Acid Active Ingredient Colloidal 0.050% 50 µg Particle Stable Polysorbate 80 Active Ingredient Colloidal 0.045% 45 µg Particle
Figure imgf000087_0001
Stable Colloidal shRNA 1 Active Ingredient 2.00% Particle Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle
Figure imgf000087_0002
85   
Figure imgf000088_0001
Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle Citric Acid, Monohydrate Buffer Solution 0.20%
Figure imgf000088_0002
(Promega, E1081) 86    Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle Citric Acid Monoh drate Buffer Solution 020%
Figure imgf000089_0001
Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle Sodium Phosphate Buffer Solution 0.06%
Figure imgf000089_0002
87   
Figure imgf000090_0001
Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle Sodium Phosphate Buffer Solution 0.06%
Figure imgf000090_0002
Scientific, UK) 88    Stable Colloidal Oleic Acid Active Ingredient 0.050% (w/w) Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% (w/w) Particle
Figure imgf000091_0001
(Vetnal) Particle Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle Sodium Phos hate Buffer Solution 006%
Figure imgf000091_0002
. , may be administered ocularly. 89   
Figure imgf000092_0001
a e oo a Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal Polysorbate 80 Active Ingredient 0.045% Particle
Figure imgf000092_0002
Stable Colloidal RNA Conjugate 2 Active Ingredient 2 mg/mL Particle 90    Stable Colloidal Oleic Acid Active Ingredient 0.050% Particle Stable Colloidal
Figure imgf000093_0001
differentiated at an air-liquid interface (ALI), forming a pseudostratified mucociliary airway epithelium that is composed of ciliated cells, goblet cells, club cells, and/or basal cells with an arrangement closely reflective of an in vivo cellular organization. This in vitro model of human airway epithelium (HAE) cultured at an ALI (HAE-ALI) closely recapitulates important characteristics of the infected and/or immune stimulated upper and lower airways in vivo. Accordingly, the HAE-ALI culture has been used to study respiratory virus-host cell interactions and immune responses, for example in the context of influenza virus infection. ALI cultured pooled donors’ human nasal epithelium (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase with MucilAir™ culture media in Costar
Figure imgf000093_0002
with a pipette). On Days 1, 2 and 3 post-treatment, sampling was collected from basal 91   
Figure imgf000094_0001
to shRNA 1 alone. Conversely, shRNA 1 with surfactant component 2 and surfactant component 3 induced significantly higher levels of CXCL10 expression compared to shRNA 1 in water. Notably, surfactant component 3 (with shRNA 1), which comprises the highest surfactant concentration, only marginally increased induction of CXCL10 relative to surfactant component 2 (with shRNA 1). Biological Example 2: Assessment of CXCL10 production and virus load, upon treatment with shRNA 1, in influenza virus infected ALI cultured human epithelial cells Experimental Methods ALI cultured pooled donors’ human nasal epithelium (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase with MucilAir™ culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions. On Day 0, influenza virus inoculum (strain PR8; 100 µL; diluted in MucilAir™ culture medium to give a final MOI of 0.1) was added to the apical surface of the epithelium for 1 hr (34°C/5% CO2). Virus inoculum was subsequently removed, and inserts were washed with sterile PBS (with Ca2+/Mg2+). The treatment formulations described below were produced largely as described in Biophysical Example 1. ALI cultures were dosed apically with 50 µL vehicle control (water alone), shRNA 1 in vehicle (2 mg/mL) or shRNA 1 (2 mg/mL) with surfactant component 1 (0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80), or surfactant component 2 (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) each in vehicle, for 30 min (then being removed with a pipette) one day prior to virus inoculation, before subsequent re-application of the treatments 30 min prior to virus inoculation (then being removed with a pipette) and subsequent incubation with the virus inoculum for 60 min. Simultaneously, oseltamivir carboxylate (10 µM) was added to the basolateral chambers of sample wells on the day prior to virus inoculation, and on Day 0. On Day 0, and Day 2, sampling was conducted by adding 300 µL of culture medium to the 92    apical surface of each for well for 10 min (being stored at -80oC). On Day 0 (day 1 post- treatment), CXCL10 concentration, prior to virus infection, was quantified by the method described in Biological Example 1 (n=3, mean ± SEM). On Day 2 post-virus inoculation (day 3 post-treatment), virus load was quantified using the 50% Tissue Culture Infectious Dose (TCID50). Briefly, the apical wash was defrosted and serial dilutions of the apical wash samples with medium containing 0.1 μg/ml TPCK Trypsin, were applied to plates lined with MDCK cells (80% confluency), prior to incubation at 35oC with 5% CO2 for 2-3 days, and in particular until the CPE induced by the vehicle control became visible. TCID50 was calculated using the Reed-Muench formula (Bullen et al. 2022) (n=3, mean ± range). Specifically, the first dilution with a CPE positivity % ≥ 50 (“Dilution More”) and the first dilution with CPE positivity % < 50% (“Dilution Less”) were identified and the proportionate distance (PD) was calculated using the formula: PD = (% of CPE positive wells in Dilution More – 50%) / (% of CPE positive wells in Dilution More - % of CPE positive wells in Dilution Less) Log TCID50 = Log Dilution More + PD Results As shown in Figure 2, shRNA 1 alone failed to induce CXCL10, which is a marker of anti-viral IFN production, expression. However, shRNA 1 with surfactant component 1, which comprises the lowest surfactant concentration, enhanced CXCL10 induction relative to vehicle control (water) or shRNA 1 alone. shRNA 1 with surfactant component 2, which comprises a comparatively higher surfactant concentration, induced a further significant increase in CXCL10 expression, relative to both shRNA 1 alone and shRNA 1 with surfactant component 1. Notably, the oseltamivir control (10 µM) failed to induce CXCL10 expression as it is a direct anti-viral agent and does not stimulate the innate immune system. As demonstrated in Figure 3, high levels of influenza replication were detected in an apical wash following treatment with the vehicle control (mean: 3.5 Log, TCID50/mL) at 48 hrs post- inoculation. Upon comparison of the detected viral load of each treatment with that following treatment with vehicle control (water), shRNA 1 alone failed to demonstrate a significant anti- viral effect, and similarly shRNA 1 with surfactant component 1, which comprises the lowest surfactant concentration, failed to significantly reduce viral load. However, shRNA with surfactant component 2 had a significant anti-viral effect, as demonstrated by a 0.7 Log reduction in viral load. The assay control, oseltamivir (10 µM), had a similar anti-viral effect (0.7 Log reduction) as predicted (Boda et al.2018). 93    Biological Example 3: Assessment of CXCL10 production and virus load, upon treatment with shRNA 1 or surfactants alone, in influenza virus infected ALI cultured human epithelial cells Experimental Methods ALI cultured human nasal epithelium was maintained and inoculated with influenza virus (PR8 strain) as described in Biological Example 2. ALI cultures were dosed apically with 50 µL vehicle control (water alone), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, shRNA 1 in vehicle or shRNA 1 with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, for 30 min (then being removed with a pipette) one day prior to virus inoculation, before subsequent re-application of the treatments 30 min prior to virus inoculation (then being removed with a pipette) and subsequent incubation with the virus inoculum for 60 min. Simultaneously, oseltamivir carboxylate (10 µM) was added to the basolateral chambers of sample wells on the day prior to virus inoculation, and on Day 0. On Day 2 (48 hrs post-inoculation), sampling was conducted by adding 300 µL of culture medium to the apical surface of each for well for 10 min (being stored at -80oC). Virus load (n=3, mean ± range) on Day 2 post infection (day 3 post-treatment) and CXCL10 levels on Day 0 (day 1 post-treatment), prior to virus infection, (n=3, mean ± SEM) were calculated as described in Biological Example 2 above. Treatment formulations used in this Example were produced largely as described in Biophysical Example 1. Results As illustrated in Figure 4, vehicle control (water), shRNA 1 alone, and, notably, the surfactant component alone each failed to induce CXCL10 expression on Day 1 post-treatment. Conversely, shRNA 1 with surfactant component significantly induced CXCL10 expression. As discussed previously, oseltamivir control (10 µM) failed to induce CXCL10 expression as it is a direct anti-viral agent. As demonstrated in Figure 5, high levels of influenza replication were detected in an apical wash following treatment with the vehicle control (mean: about 3.4 Log, TCID50/mL) at 48 hrs post-inoculation. shRNA 1 alone, and, notably, the surfactant component alone, each failed to reduce viral load relative to the vehicle control. However, shRNA 1 with surfactant component had a very potent anti-viral effect, as represented by a 3.2 Log reduction of viral load relative to water alone (vehicle control). Significantly, shRNA 1 with surfactant component elicited a more significant reduction in viral load than the assay control, oseltamivir (10 µM), which led to a 2.2 Log reduction in viral load versus the vehicle control. 94    Biological Example 4: Assessment of CXCL10 production and virus load, upon treatment with dsRNA 1 or dsRNA 2, in influenza virus infected ALI cultured human epithelial cells Experimental Methods ALI cultured human nasal epithelium was maintained and inoculated with influenza virus (PR8 strain) as described in Biological Example 2. ALI cultures were dosed apically with 50 µL of a vehicle control (buffer: 0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate in water), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle, dsRNA 1 (10 µg/mL) or dsRNA 2 (100 µg/mL) in vehicle or dsRNA 1 (10 µg/mL) or dsRNA 2 (100 µg/mL) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in vehicle for 30 min (then being removed with a pipette) one day prior to virus inoculation, before subsequent re-application of the treatments for 30 min prior to virus inoculation (then being removed with a pipette) and subsequent incubation with the virus inoculum for 60 min. On Day 0 (24 hrs post-treatment) and Day 1 (24 hrs post-virus inoculation and 48 hrs post-treatment), sampling was conducted by adding 300 µL of culture medium to the apical surface of each for well for 10 min (being stored at -80oC). Virus load (n=3, mean ± range) and CXCL10 levels (n=3, mean ± SEM) were calculated as described in Biological Example 2 above. Upon comparison of the detected viral load of each treatment with that following treatment with vehicle control Treatment formulations used in this Example were produced largely as described in Biophysical Example 1. Results As illustrated in Figure 6, surfactant component alone failed to induce CXCL10 expression in the apical wash relative to vehicle control (buffer). Moreover, dsRNA 1 failed to induce CXCL10 expression relative to vehicle control. However, dsRNA 1 with surfactant component induced strong CXCL10 expression. dsRNA 2 alone induced significant CXCL10 expression, however formulation of dsRNA 2 with surfactant component elicited a further increase in the observed induction of CXCL10 expression. As demonstrated in Figure 7, high levels of influenza replication were detected in an apical wash following treatment with the vehicle control (buffer). dsRNA 1 alone failed to demonstrate a significant anti-viral effect. Notably, surfactant component alone appeared to elicit a mild anti-viral effect, as represented by a 0.7 Log reduction in viral load relative to vehicle control. dsRNA 1 with surfactant component had potent anti-viral activity, as represented by a significant reduction in viral load (1.8 Log reduction versus dsRNA 1. dsRNA 2 alone demonstrated significant anti-viral activity relative to vehicle control. However, formulation of dsRNA 2 with the surfactant component enhanced the anti-viral activity of dsRNA 2, as 95    represented by a 2.8 Log reduction in viral load versus surfactant component alone and a 1.5 Log reduction in viral load versus dsRNA 2 alone. Biological Example 5: Assessment of viral load, virus-induced inflammation and body weight loss, upon treatment with shRNA 1, in an influenza virus infected mouse model Experimental Methods Non-fasted mice (male BALB/C, 20-30 g) were infected intranasally with influenza (strain PR8) or virus diluent (DMEM, 2%F BS, 12.5% sucrose) under isoflurane (5% in O2) anaesthesia. The influenza virus (PR8 strain; 10uL per nostril of 2 x102 PFU) was administered into each nostril in a drop-wise manner (10 µl each nostril). Following infection, each mouse was weighed daily (n=5, mean ± SEM). Treatment formulations of vehicle control (water), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) and further pharmaceutically acceptable excipients in vehicle (see Formulation Example 1A minus the shRNA 1, hereinafter “surfactant formulation”), or shRNA 1 (2 mg/mL) in vehicle, or with surfactant component and further pharmaceutically acceptable excipients in vehicle (see Formulation Example 1A, hereinafter “shRNA 1 with surfactant formulation”) were produced largely as described in Biophysical Example 1 below. The vehicle control or test formulations were administered intranasally (10 µL each nostril) with a pipette 3 days, 1 day and 1 hr prior to virus infection on Day 0. By way of a drug control, oseltamivir phosphate was formulated in PBS and administered orally to control mice (10 mg/kg) 1 hr prior to virus infection and once daily on Day 1 post-inoculation. On Day 1 and Day 5 post-infection, the mice were euthanised (intraperitoneal injection of pentobarbitone overdose). Subsequently, the trachea was cannulated and the airway lavaged by flushing out the lungs using 0.5 mL PBS. Immediately after bronchoalveolar lavage, a catheter was inserted into the posterior naris/nares from the opening of the trachea that was used for bronchoalveolar lavage to collect nasal lavage fluid (NLF). PBS (1 mL) was gently perfused into the nasal cavities, and the NLF was collected from the anterior naris/nares. The isolated NLF was centrifuged at 1500 rpm for 10 min at 4oC and the supernatant was aliquoted (350 µL) at -80oC for future cytokine analysis. The cell pellets were re-suspended in 1.6 mL PBS and the NLF cells were analysed for total and differential numbers. Total and differential cell counts of the NLF fluid samples were measured using a XT-2000iV analyser (Sysmex). Results are expressed as cells/mL e.g., neutrophils/mL (total and differential) (n=5, mean ± SEM). Immediately after NLF collection, the thoracic cavity of euthanised mice was opened and the right lung lobes from each mouse were removed and homogenised in DMEM (with 1% BSA and 25% sucrose; 10 mL/g lung) for two 20 second periods. The homogenate was transferred into a sterile tube and spun at 4oC at 2000 rpm for 5 min. The clarified homogenate was then 96    transferred to a chilled cryovial and snap frozen in liquid nitrogen and stored at -80oC. Following removal of the right lung lobe, the nasal tissue was removed from each animal. The dissection involved firstly removing the mandible and skin over the head, followed by removal of the head. Next, the palette and bone protecting the brain and olfactory bulbs was removed to expose the nasal tissue. The extracted nasal tissue was homogenised in DMEM (with 1% BSA and 25% sucrose; 10mL/g tissue) for two 20 second periods. As for the lung homogenate, the nasal tissue homogenate was transferred into a sterile tube and spun at 4oC at 2000 rpm for 5 min before transfer into a cryovial, snap freezing in liquid nitrogen and storage at -80oC. Serial dilutions of the supernatant collected from the lung and nasal tissue homogenates, with medium containing 0.1 μg/ml TPCK Trypsin, were applied to plates lined with MDCK cells (80% confluency), prior to incubation of the inoculated cells at 37oC for 1 hr. Next, the inoculum was removed from the well and the cells were washed twice with PBS before applying an overlay of 1% methylcellulose agar media (with the growth medium and 0.1 μg/ml TPCK trypsin) to each well. Once the agar medium was set, the plates were incubated at 37oC with 5% CO2 for 3 days, after which the resulting plaques are counted. A second count was performed following removal of the agar overlay and staining of the cells with crystal violet (n=5, mean ± SEM, PFU/group). Results High levels of influenza virus replication were detected in the nasal tissue of mice on Day 1 post-inoculation following treatment with vehicle control (water alone), whilst only a marginal reduction in viral load was observed on Day 5 post-inoculation. As illustrated in Figure 8, the surfactant formulation alone had no anti-viral effect. However, whilst shRNA 1 alone demonstrated a moderate anti-viral effect, as represented by a 41% reduction in viral load on both Day 1 and Day 5 post-inoculation (versus water alone), formulation of shRNA 1 with the surfactant formulation resulted in a more significant anti-viral effect, as represented by a 60% and 51% reduction in viral load on Day 1 and Day 5 post-inoculation respectively (versus surfactant formulation alone). shRNA 1 with surfactant formulation therefore had a significantly more potent anti-viral effect than the oseltamivir control, which elicited a 38% and 33% reduction in viral load on Day 1 and Day 5 post-inoculation respectively (versus vehicle). Moreover, as illustrated in Figure 9, significant neutrophil accumulation was observed in the nose of influenza virus infected mice on Day 1 post-inoculation following treatment with vehicle, although the neutrophil count was moderately reduced by Day 5 post-inoculation. shRNA 1 in water elicited a moderate reduction in neutrophil accumulation, as represented by a 41% and 67% reduction in the neutrophil number on Day 1 and Day 5 post-inoculation respectively (versus water alone). However, formulation of shRNA 1 with the surfactant formulation resulted in a more significant reduction in neutrophil accumulation, as represented by an 85% and 82% reduction in the neutrophil number on Day 1 and Day 5 post-inoculation 97    respectively (versus surfactant formulation alone which elicited no reduction in neutrophil accumulation relative to vehicle). The oseltamivir control also resulted in a reduction in neutrophil accumulation, as represented by a 57% and 69% reduction in the neutrophil number on Day 1 and Day 5 post-inoculation respectively, albeit to a lesser extent than shRNA 1 with the surfactant formulation. Finally, influenza virus infection led to substantial body weight loss in influenza virus infected mice treated with vehicle. As illustrated in Figure 10, the surfactant formulation alone failed to prevent this influenza virus-induced weight loss. However, shRNA 1 in vehicle had a moderate protective effect against body weight loss, whilst shRNA 1 with surfactant formulation further prevented influenza virus-induced weight loss. Indeed, shRNA 1 with surfactant formulation had a similar protective effect to the oseltamivir control. Biological Example 6: Assessment of the exposure of mRNA and plasmid DNA to ALI cultured human nasal epithelium Experimental Methods ALI cultured pooled donors’ human nasal epithelium (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase with MucilAir™ culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions. ALI cultures were dosed apically with 50 µL of vehicle control, that is 0.5 mg/mL GFP-encoding mRNA (Vetnal) in citrate buffer (0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate), or test formulation, that is 0.5 mg/mL GFP-encoding mRNA (Vetnal) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs. The treatment was then removed, and the apical surface of the ALI cultures were washed with 100 µL culture. Following overnight incubation (24 hrs post- treatment), ALI cultured nasal epithelial cells were collected by gentle agitation in sterile PBS prior to fixation with 4% paraformaldehyde in PBS. Samples from two independent inserts were combined and analysed by flow cytometry using a 530/30-nm bandpass (BP) filter on the BD Accuri™ apparatus (Becton Dickinson). The MFI was calculated, and the MFI of non- treated control (autofluorescence) subtracted. Alternatively, ALI cultures were dosed apically with 50 µL of vehicle control, that is 0.033 µg/mL pSV-β-Galactosidase Control Vector (Promega, E1081) (β-gal) in citrate buffer (0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate), or test formulation, that is 0.033 µg/mL pSV-β-Galactosidase Control Vector (Promega, E1081) (β-gal) with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs. The treatment was then removed, and the apical surface of the ALI cultures were washed with 100 µL culture. ALI cultured nasal epithelial cells from three inserts were lysed together in a reporter lysis buffer according to the manufacturer’s instructions (Promega, #E2000, β-galactosidase enzyme assay system with Reporter Lysis Buffer). Following 98    overnight incubation with the substrate provided in the kit β-galactosidase enzyme activity (24 hrs post-treatment) was determined by reading absorbance at 420 nm. Treatment formulations used in this Example were produced largely as described in Biophysical Example 1. Results As illustrated in Figure 11, treatment of ALI cultured nasal epithelial cells with formulation of GFP-encoding mRNA with surfactant component led to a significant MFI (in particular versus background autofluorescence). Moreover, the MFI observed upon treatment with GFP- encoding mRNA with surfactant component was significantly higher than that observed upon treatment with GFP-encoding mRNA alone. These data suggest that formulation of GFP- encoding mRNA with surfactant component facilitates a greater degree of epithelial cell exposure to said mRNA, resulting in greater GFP expression and consequently higher levels of fluorescence. Moreover, as illustrated in Figure 12, it appears that formulation of a β-galactosidase encoding plasmid with surfactant component may similarly increase ALI cultured nasal epithelial cell exposure to said plasmid. These data provide encouragement that the formulations of the present invention could be suitable for improving the delivery, and thereby increasing exposure, of a diverse array of polynucleotide molecules, including DNA. Biological Example 7: Assessment of viral load and virus-induced inflammation, upon treatment with shRNA 1, in human rhinovirus infected ALI cultured human epithelial cells Experimental Methods ALI cultured pooled donors’ human nasal epithelium (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase with MucilAir™ culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions. On Day 0 of the study, human rhinovirus type 16 (HRV16) virus inoculum (100 µL of 400,000 PFU/mL in MucilAir™ culture medium to give a final MOI of approximately 0.2) was added to the apical surface of the epithelium for 1 hr (34°C/5% CO2). Virus inoculum was subsequently removed, and inserts were washed with 100 µL of sterile PBS (with Ca2+/Mg2+). ALI cultures were dosed apically with 50 µL vehicle control (citrate buffer: 0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate), shRNA 1 in vehicle (2 mg/mL), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or shRNA 1 (2 mg/mL) with surfactant component for 30 min (then being removed with a pipette almost completely without additional wash) one day prior to virus inoculation, before subsequent re- application of the treatments 30 min prior to virus inoculation (then being removed as described above) and subsequent incubation with virus inoculum for 60 min. On Day 0 (before 99    virus inoculation) and Day 2, sampling was conducted by adding 300 µL of culture medium to the apical surface of each for well for 10 min (being stored at -80oC). On Day 0 (day 1 post- the first treatment), CXCL10 concentration, prior to virus infection, was quantified by the method described in Biological Example 1 (n=3, mean ± SEM). On Day 2 post virus inoculation (day 3 post-the first treatment), virus load was quantified using the 50% Tissue Culture Infectious Dose (TCID50). Briefly, the apical wash was defrosted and serial dilutions of the apical wash samples with 1% FBS DMEM medium containing 15mM MgCl2, were applied to plates lined with Hela cells (ATCC®, Manassas, #CCL-2, 80% confluency), prior to incubation at 34oC with 5% CO2 for 5 days, and in particular until the CPE induced by the vehicle control became visible. TCID50 was calculated using the Reed-Muench formula (Bullen et al.2022) (n=3, mean ± range). Specifically, the first dilution with a CPE positivity % ≥ 50 (“Dilution More”) and the first dilution with CPE positivity % < 50% (“Dilution Less”) were identified and the proportionate distance (PD) was calculated using the formula: PD = (% of CPE positive wells in Dilution More – 50%) / (% of CPE positive wells in Dilution More - % of CPE positive wells in Dilution Less) Log TCID50 = Log Dilution More + PD Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2. Results As illustrated in Figure 14, shRNA 1 alone (i.e. in vehicle) marginally induced CXCL10 expression, which is a marker of anti-viral IFN production, expression. However, shRNA 1 with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), induced a considerably more significant increase in CXCL10 expression, relative to vehicle control. Moreover, as demonstrated in Figure 15, high levels of HRV16 replication were detected in an apical wash following treatment with the vehicle control (mean: 3.1 Log, TCID50/mL) at 48 hrs post-inoculation. shRNA 1 alone (i.e. in vehicle) failed to demonstrate a significant anti- viral effect. However, whilst treatment with surfactant component alone had a moderate anti- viral effect, shRNA 1 with surfactant component had a specific, and strong anti-viral effect, as demonstrated by a 2.8 Log reduction in viral load relative to vehicle control. Biological Example 8: Assessment of viral load and cell integrity, upon treatment with shRNA 1, in human respiratory syncytial virus (RSV) infected ALI cultured human epithelial cells Experimental Methods ALI cultured pooled donors’ human nasal epithelium (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase with MucilAir™ culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions. On Day 0 100    of the study, human respiratory syncytial virus A2 (RSV A2) virus inoculum (100 µL of 40,000 PFU/mL in MucilAir™ culture medium to give a final MOI of approximately 0.02) was added to the apical surface of the epithelium for 1 hr (37°C/5% CO2). Virus inoculum was subsequently removed, and inserts were washed with 100 µL of sterile PBS (with Ca2+/Mg2+). ALI cultures were dosed apically with 50 µL vehicle control (citrate buffer: 0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate), shRNA 1 in vehicle (2 mg/mL), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80), or shRNA 1 (2 mg/mL) with surfactant component for 30 min (then being removed with a pipette almost completely without additional wash) one day prior to virus inoculation, before subsequent re- application of the treatments 30 min prior to virus inoculation (then being removed as described above) and subsequent incubation with virus inoculum for 60 min. On Day 0 (before virus inoculation) and Day 3, sampling was conducted by adding 300 µL of culture medium to the apical surface of each well for 10 min. All apical wash was collected in sterile Eppendorf tube containing 100µL of 50% sucrose solution (being stored at -80oC). Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2. On Day 3 post virus inoculation (day 4 post-the first treatment), virus load was quantified via a plaque assay. Briefly, HEp-2 cells were seeded into 24-well plates (Corning, NY, USA) at a density of between 5-10 × 104 cells/well and grown, for 48 hrs prior to infection, in 10% FBS DMEM until they attained 100% confluency. Next, virus infected samples were thawed at room temperature and ten-fold serial dilutions were prepared in serum-free DMEM. The growth medium from HEp-2 cells was aspirated and replaced with 300 µL of the serially diluted virus collections and left to infect at 37°C/5% CO2 for 4 hrs. Post-infection, the virus media was aspirated and replaced with 1 mL of Plaque Assay Overlay (0.3% Avicel RC-591 [FMC Biopolymer UK, Girvan, Scotland]) in MEM, supplemented to a final concentration of 2% FBS), and left for 7 days at 37°C/5% CO2. Cells were then fixed with ice-cold methanol for 10 min, prior to methanol removal and subsequent washing of fixed cells (x 2) with sterile PBS. Cells were then stained with 200 µL 0.1% crystal violet solution (in distilled water) for 1 hr. Crystal violet solution was removed, and cells were rinsed with water before plaques were counted and viral load enumerated (n=3, mean ± range). In addition to the above-described plaque assay, transepithelial electrical resistance (TEER) was measured to investigate the integrity of tight junction dynamics in ALI cultured pseudostratified epithelium pre- and 3 days post-RSV A2 infection. This measurement of TEER is a surrogate measure for epithelial damage. Specifically, chopstick-electrodes were placed in the apical and basolateral chambers and the TEER was measured using a dedicated 101    Volt/Ohm meter (EVOM2, Epithelial Volt/Ohm Meter for TEER). The measure of TEER is expressed as Ohm/cm2. Results As shown in Figure 16, high levels of human RSV A2 replication was detected in an apical wash following treatment with the vehicle control (mean: 6.4 Log, PFU/mL) at 72 hrs post- inoculation. shRNA 1 alone (i.e. in vehicle) failed to demonstrate a significant anti-viral effect. However, upon comparison of the detected viral load of with that following treatment with vehicle (buffer), surfactant component alone had a moderate anti-viral effect, whilst shRNA 1 with surfactant component had a significant anti-viral effect, as demonstrated by a 1.6 Log reduction in viral load. Furthermore, as demonstrated in Figure 17, TEER was reduced following human RSV A2 infection following treatment with vehicle control (buffer). As noted above, this is indicative of virus induced epithelial damage. Treatment of cells with shRNA 1 alone (i.e. in vehicle) post- infection was able to mitigate the loss in TEER and/or restore the reduced TEER. However, treatment with surfactant component alone, clearly increased the TEER (measured 72 hrs post-infection) relative to that measured pre-infection, which is suggestive of a protective effect and increased epithelial cell integrity in the presence of surfactant component. This protective effect is even more significant following treatment with shRNA 1 in surfactant component, as evidence by a further increased TEER. Biological Example 9: Assessment of viral load, virus-induced inflammation and body weight loss, upon treatment with shRNA 1, in a respiratory syncytial virus (RSV) infected mouse model Experimental Methods Non-fasted mice (male BALB/C, 20-30 g) were inoculated intranasally with RSV (5 x 106 PFU/mouse) under isoflurane (5% in O2) anaesthesia. The RSV virus (RSV A2 strain; 25 µL per nostril of 2.5 x106 PFU) was administered into each nostril in a drop-wise manner (25 µL each nostril). Following infection, each mouse was weighed daily (n=5, mean ± SEM). Treatment formulations of saline, surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer and shRNA 1 (2 mg/mL) with surfactant component and further pharmaceutically acceptable excipients in vehicle (see Formulation Example 1A) were produced largely as described in Biophysical Examples 1 and 2 below. The vehicle control or test formulations were administered intranasally (25 µL each nostril) with a pipette both 4 days and 1 day prior to virus infection on Day 0. By way of a drug control, ribavirin was formulated in PBS and administered intranasally to control mice (12.5 mg/kg) 3 hr prior to virus infection, and twice daily on Days 0 to 4 post-inoculation. On Day 4 post-infection, the mice were euthanised (intraperitoneal injection of pentobarbitone overdose). Subsequently, the trachea was cannulated and the airway lavaged by flushing out 102    the lungs using 0.5 mL PBS. Immediately after bronchoalveolar lavage, a catheter was inserted into the posterior naris/nares from the opening of the trachea that was used for bronchoalveolar lavage to collect nasal lavage fluid (NLF). PBS (1 mL) was gently perfused into the nasal cavities, and the NLF was collected from the anterior naris/nares. The isolated NLF was centrifuged at 1500 rpm for 10 min at 4oC and the supernatant was aliquoted (350 µL) at -80oC for future cytokine analysis. The cell pellets were re-suspended in 1.6 mL PBS and the NLF cells were analysed for total and differential numbers. Total and differential cell counts of the NLF fluid samples were measured using a XT-2000iV analyser (Sysmex). Results are expressed as cells/mL e.g. neutrophils/mL (total and differential) (n=5, mean ± SEM). Immediately after NLF collection, the thoracic cavity of euthanised mice was opened and the right lung lobes from each mouse were removed and homogenised in DMEM (with 1% BSA and 25% sucrose; 10 mL/g lung) for two 20 second periods. The homogenate was transferred into a sterile tube and spun at 4oC at 2000 rpm for 5 min. The clarified homogenate was then transferred to a chilled cryovial and snap frozen in liquid nitrogen and stored at -80oC. Following removal of the right lung lobe, the nasal tissue was removed from each animal. The dissection involved firstly removing the mandible and skin over the head, followed by removal of the head. Next, the palette and bone protecting the brain and olfactory bulbs was removed to expose the nasal tissue. The extracted nasal tissue was homogenised in DMEM (with 1% BSA and 25% sucrose; 10 mL/g tissue) for two 20 second periods. As for the lung homogenate, the nasal tissue homogenate was transferred into a sterile tube and spun at 4oC at 2000 rpm for 5 min before transfer into a cryovial, snap freezing in liquid nitrogen and storage at -80oC. To determine the viral load, plaque assay was conducted. HEp2 cells were grown in 24-well plates prior to infection in DMEM containing 10% (v/v) FBS until they achieved 100% confluency. The supernatant collected from the lung and nasal tissue homogenates were thawed out at room temperature and serial dilutions was prepared in serum-free DMEM. The growth medium from HEp2 cells were aspirated and replaced with 300 µL of serially diluted lung homogenate (along with stock RSV only positive control) and left to infect at 37°C/5% CO2 for four hrs. The infectious media was then aspirated and replaced with 500 µL Plaque Assay Overlay (1% (w/v) methylcellulose in MEM, 2% (v/v) FBS, 1% (w/v) pen/strep, 0.5 µg/ml amphotericin B), and left for 7 days at 37°C/5% CO2. Next, cells were fixed with ice-cold methanol for 10 mins after which they were washed twice with sterile PBS. Anti-RSV F-protein antibody [2F7] was diluted to a 1:150 concentration in blocking buffer (5% (w/v) powdered milk (Marvel) in 0.05% (v/v) PBS-Tween 20) and 150 µL was added to cells for 2 hrs at room temperature with shaking. Cells were washed twice using PBS before 150 µL of secondary antibody (goat anti-mouse/HRP conjugate) diluted 1:400 in 103    blocking buffer were added to cells for 1 hr at room temperature, with shaking. The secondary antibody solution was removed, and cells was washed twice with PBS.150 µL of the metal- enhanced development substrate, DAB, prepared in ultra-pure water was applied to the cells until plaques are visible. Plaques were counted by eye and confirmed using light microscopy, allowing the calculation of plaque-forming units per mL (n=5, mean ± SEM, PFU/group). Results As shown in Figure 18, high levels of RSV A2 were detected in the lung tissue of mice on Day 4 post-inoculation following treatment with vehicle control (i.e. saline). Notably, treatment of mice with the surfactant component formulation only had no anti-viral effect in the lung. However, treatment of mice with the shRNA 1 with surfactant component formulation resulted in a significant anti-viral effect, as represented by a 54% reduction in viral load on Day 4 post- inoculation, relative to treatment with surfactant component alone. Moreover, the anti-viral effect of the shRNA 1 with surfactant component formulation was comparable to that of ribavirin which, as expected, had a strong anti-viral effect, as represented by a 66% reduction in viral load on Day 4 post-inoculation, relative to treatment with vehicle control (i.e. saline). Furthermore, as demonstrated in Figure 19, significant neutrophil accumulation was observed in the lung of RSV A2-infected mice on Day 4 post-inoculation following treatment with vehicle control (i.e. saline). Treatment of mice with the surfactant component formulation only failed to demonstrate any significant reduction in neutrophil count. However, treatment of mice with the shRNA 1 with surfactant component formulation resulted in a significant reduction in the degree of neutrophil accumulation, as represented by a 57% reduction in the neutrophil number on Day 4 post-inoculation relative to treatment with vehicle control (i.e. saline), and by a 55% reduction in the neutrophil number on Day 4 post-inoculation relative to treatment with surfactant component alone. Notably, treatment of mice with the shRNA 1 with surfactant component formulation resulted in a reduction in neutrophil accumulation to that observed following treatment with ribavirin. Specifically, ribavirin treatment resulted in a 61% reduction in the neutrophil number on Day 4 post-inoculation relative to treatment with vehicle control (i.e. saline). Finally, as shown in Figure 20, RSV A2 infection led to substantial body weight loss in mice treated with vehicle control (i.e. saline). Treatment of mice with the surfactant component formulation failed to limit or prevent the RSV A2 infection-induced body weight loss. On the contrary, treatment of mice with both the shRNA 1 with surfactant component formulation and with the ribavirin control, provided a protective effect against said RSV A2 infection-induced body weight loss. The extent of this protective effect was comparable between the shRNA 1 with surfactant component formulation and the ribavirin control. 104    Biological Example 10: Assessment of CXCL10 release in non-infected mice, and assessment of viral load, virus-induced inflammation and body weight loss, in an influenza virus infected mouse model, upon subcutaneous treatment with shRNA 1 Experimental Methods Non-fasted mice (male BALB/C, 20-30 g) were injected subcutaneously with saline, surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer, shRNA1 (2 mg/mL) with surfactant component, or shRNA1 (20 mg/mL) with surfactant component at a dose volume of 10 mL/kg. 24 hrs after dosing (n=4) blood samples were collected from the lateral tail vein of infected mice and placed into a serum tube. Each serum sample was kept at room temperature for 45 mins to allow coagulation, prior to centrifugation at 2000 g for 15 min at 4oC. The resulting supernatant was extracted, aliquoted (2 x 50 µL) and stored at -80oC for shipping. Immediately after blood collection each mouse was euthanized via intraperitoneal pentobarbitone overdose. The trachea was then isolated by a midline incision in the neck and separation of the muscle layers. A small incision was made into the trachea and a plastic cannula was inserted and secured in place with a suture. The airway was then lavaged by flushing out the lungs using 0.5 mL PBS. This procedure was repeated until the recovered volume totalled 1.6 mL. The isolated BALF was then centrifuged at 1500 rpm for 10 mins at 4oC and the supernatant was aliquoted (400 µL) at -80oC. The levels of CXCL10 in serum and BALF was determined using mouse CXCL10 kit using MSD multi scanner (Meso Scale Diagnosis). Separately, non-fasted mice (male BALB/C, 20-30 g) were infected intranasally with influenza (strain PR8) under isoflurane (5% in O2) anaesthesia. The influenza virus (PR8 strain; 10 µL per nostril of 2 x102 PFU) was administered into each nostril in a drop-wise manner (10 µL each nostril). Following infection, each mouse was weighed daily (n=5, mean ± SEM). Treatment formulations of saline (i.e. non-treatment), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer, or shRNA 1 (2 mg/mL) with surfactant component in phosphate buffer, were administered subcutaneously at a dose volume of 10 mL/kg both 4 days and 1 day prior to virus infection on Day 0. By way of a drug control, oseltamivir phosphate was formulated in PBS and administered orally 4 hr prior to virus infection to control mice (10 mg/kg), wherein said mice were also treated with subcutaneous injection of surfactant component alone 4 days and 1 day prior to virus infection on Day 0. Futhermore, one group of mice was treated both with oral oseltamivir, and with the subcutaneously administered shRNA 1 with surfactant component formulation, as a combination therapy. Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2. 105    On Day 5 post-infection, the mice were euthanised (intraperitoneal injection of pentobarbitone overdose). Subsequently, the trachea was cannulated and the airway lavaged by flushing out the lungs using 0.5 mL PBS. Immediately after bronchoalveolar lavage, a catheter was inserted into the posterior naris/nares from the opening of the trachea that was used for bronchoalveolar lavage to collect nasal lavage fluid (NLF). PBS (1 mL) was gently perfused into the nasal cavities, and the NLF was collected from the anterior naris/nares. The isolated NLF was centrifuged at 1500 rpm for 10 min at 4oC and the supernatant was aliquoted (350 µL) at -80oC for future cytokine analysis. The cell pellets were re-suspended in 1.6 mL PBS and the NLF cells were analysed for total and differential numbers. Total and differential cell counts of the NLF fluid samples were measured using a XT-2000iV analyser (Sysmex). Results are expressed as cells/mL e.g. neutrophils/mL (total and differential) (n=5, mean ± SEM). Immediately after NLF collection, the thoracic cavity of euthanised mice was opened and the right lung lobes from each mouse were removed and homogenised in DMEM (with 1% BSA and 25% sucrose; 10 mL/g lung) for two 20 second periods. The homogenate was transferred into a sterile tube and spun at 4oC at 2000 rpm for 5 min. The clarified homogenate was then transferred to a chilled cryovial and snap frozen in liquid nitrogen and stored at -80oC. Following removal of the right lung lobe, the nasal tissue was removed from each animal. The dissection involved firstly removing the mandible and skin over the head, followed by removal of the head. Next, the palette and bone protecting the brain and olfactory bulbs was removed to expose the nasal tissue. The extracted nasal tissue was homogenised in DMEM (with 1% BSA and 25% sucrose; 10mL/g tissue) for two 20 second periods. As for the lung homogenate, the nasal tissue homogenate was transferred into a sterile tube and spun at 4oC at 2000 rpm for 5 min before transfer into a cryovial, snap freezing in liquid nitrogen and storage at -80oC. Serial dilutions of the supernatant collected from the lung and nasal tissue homogenates, with medium containing 0.1 μg/mL TPCK Trypsin, were applied to plates lined with MDCK cells (80% confluency), prior to incubation of the inoculated cells at 37oC for 1 hr. Next, the inoculum was removed from the well and the cells were washed twice with PBS before applying an overlay of 1% methylcellulose agar media (with the growth medium and 0.1 μg/mL TPCK trypsin) to each well. Once the agar medium was set, the plates were incubated at 37oC with 5% CO2 for 3 days, after which the resulting plaques are counted. A second count was performed following removal of the agar overlay and staining of the cells with crystal violet (n=5, mean ± SEM, PFU/group). Results As shown in Figure 21, subcutaneous injection of saline in treated mice failed to induce notable CXCL10 expression. Subcutaneous treatment of mice with surfactant component alone induced CXCL10 expression significantly relative to treatment with saline. However, 106    subcutaneous injection of the shRNA 1 with surfactant component formulation resulted in a considerably more significant increase in CXCL10 expression, relative to treatment with either saline or surfactant component alone. It should be noted that there was no significant between the level of induction of CXCL10 expression between the 2 mg/mL and 20 mg/mL shRNA 1 doses. The data shown is from 24 hrs post-injection. In the influenza virus-infected mouse model, high levels of influenza virus replication were detected in both the lung (see Figure 22) and nasal tissue (see Figure 23) of non-treated mice (i.e. saline injection only). Moreover, as illustrated in Figures 22 and 23, treatment of mice with the surfactant component formulation demonstrated no clear anti-viral effect in either the lung or nasal tissue. On the contrary, treatment of mice with the shRNA 1 with surfactant component formulation had a clear and strong anti-viral effect, as represented by a 29% reduction in viral load in the lung, and by a 45% reduction in viral load in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only. Treatment of mice with the control drug oseltamivir also resulted in a reduction in viral load, as represented by a 41% reduction in viral load in the lung, and by a 49% reduction in viral load in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only. It should be noted that the anti-viral effect of the control drug oseltamivir is comparable to that of the shRNA 1 with surfactant component formulation. Finally, the combination treatment of oral oseltamivir and subcutaneous shRNA 1 with surfactant component showed an even more significant anti-viral effect, as represented by a 73% reduction in viral load in the lung, and by a 67% reduction in viral load in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only. Furthermore, significant neutrophil accumulation was observed in both the lung (see Figure 24) and nasal tissue (see Figure 25) of influenza virus-infected mice treated with saline only (i.e. non-treatment). Moreover, as illustrated in Figures 24 and 25, treatment of mice with the surfactant component formulation alone failed to reduce neutrophil accumulation in both the lung and nasal tissue. However, treatment of mice with the shRNA 1 with surfactant component formulation resulted in a significant reduction in neutrophil accumulation in both the lung and nasal tissue, as represented by a 40% reduction in neutrophil number in the lung, and by a 56% reduction in the neutrophil number in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only. Treatment of mice with the control drug oseltamivir also resulted in a reduction in neutrophil accumulation in both the lung and nasal tissue, as represented by a 60% reduction in neutrophil number in the lung, and by a 70% reduction in the neutrophil number in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only. More significantly, the combination treatment of oral oseltamivir and subcutaneous shRNA 1 with surfactant component elicited a yet more significant reduction in neutrophil accumulation, as represented by an 80% reduction in 107    neutrophil number in the lung, and by a 74% reduction in the neutrophil number in the nasal tissue, on Day 5 post-inoculation, relative to treatment with surfactant component only. Finally, as shown in Figure 26, influenza virus infection led to substantial body weight loss in mice treated with vehicle control (i.e. saline). Subcutaneous treatment of mice with the surfactant component formulation failed to limit or prevent the influenza virus infection-induced body weight loss. However, subcutaneous treatment of mice with both the shRNA 1 with surfactant component formulation and with the oral oseltamivir control, provided a protective effect against said influenza virus infection-induced body weight loss. Moreover, the combination treatment of oral oseltamivir and subcutaneous shRNA 1 with surfactant component entirely prevented any influenza virus infection-induced body weight loss. Biological Example 11: Assessment of the adjuvant effect of shRNA 1 with surfactant component on recombinant H1N1 haemagglutinin (HA) vaccinated and influenza virus infected mice Experimental Methods Non-fasted mice (male BALB/C, 20-30 g) were treated with PBS, shRNA 1 with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer or the control adjuvant and TLR9 agonist, CPG-ODN solution (InvivoGen), intranasally (10 µL/nostril). Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2. Following adjuvant dosing, mice were treated, on Day 0, with either vehicle (i.e. PBS) or recombinant haemagglutinin (HA) (1mg/mL in PBS) from the influenza virus PR8 strain (SinoBiologics) under isoflurane to deliver a volume of 10 µL per nostril. Said treatments were introduced into each nostril in a drop-wise fashion, alternating between the two, until the required volume had been delivered (10 µg HA/10µL). A qualitative assessment for each group was recorded for the level of piloerection generated in each animal following priming dose. Booster doses (adjuvant and HA antigen) in all animals were re-administered on Day 28. On Day 56 each animal was dosed intranasally (10 µL) with influenza virus (PR8 strain; 2 x 102 PFU). Following infection each animal was weighed on a daily basis to monitor changes in body weight. On Day 61 (i.e.5 days post-infection), terminal blood samples were taken by venipuncture (via the lateral tail vein) and placed into LiHep tubes. Each blood sample was mixed gently and centrifuged (2000 g for 5 min at 4oC) from which the resulting plasma was extracted, aliquoted and stored at -80oC. Immediately after the bleeds, mice were euthanised (intraperitoneal injection of pentobarbitone overdose). Subsequently, the trachea was cannulated and the airway lavaged by flushing out the lungs using 0.5 mL PBS. Immediately after bronchoalveolar lavage, a catheter was inserted into the posterior naris/nares from the opening of the trachea that was used for bronchoalveolar lavage to collect nasal lavage fluid (NLF). PBS (1 mL) was gently 108    perfused into the nasal cavities, and the NLF was collected from the anterior naris/nares. The isolated NLF was centrifuged at 1500 rpm for 10 min at 4oC and the supernatant was aliquoted (350 µL) at -80oC for future cytokine analysis. The cell pellets were re-suspended in 1.6 mL PBS and the NLF cells were analysed for total and differential numbers. Total and differential cell counts of the NLF fluid samples were measured using a XT-2000iV analyser (Sysmex). Results are expressed as cells/mL e.g. neutrophils/mL (total and differential) (n=6, mean ± SEM). Immediately after NLF collection, the thoracic cavity of euthanised mice was opened and the right lung lobes from each mouse were removed and homogenised in DMEM (with 1% BSA and 25% sucrose; 10 mL/g lung) for two 20 second periods. The homogenate was transferred into a sterile tube and spun at 4oC and 2000 rpm for 5 min. The clarified homogenate was then transferred to a chilled cryovial and snap frozen in liquid nitrogen and stored at -80oC. Following removal of the right lung lobe, the nasal tissue was removed from each animal. The dissection involved firstly removing the mandible and skin over the head, followed by removal of the head. Next, the palette and bone protecting the brain and olfactory bulbs was removed to expose the nasal tissue. The extracted nasal tissue was homogenised in DMEM (with 1% BSA and 25% sucrose; 10mL/g tissue) for two 20 second periods. As for the lung homogenate, the nasal tissue homogenate was transferred into a sterile tube and spun at 4oC and 2000 rpm for 5 min before transfer into a cryovial, snap freezing in liquid nitrogen and storage at -80oC. Serial dilutions of the supernatant collected from the lung and nasal tissue homogenates, with medium containing 0.1 μg/ml TPCK Trypsin, were applied to plates lined with MDCK cells (80% confluency), prior to incubation of the inoculated cells at 37oC for 1 hr. Next, the inoculum was removed from the well and the cells were washed twice with PBS before applying an overlay of 1% methylcellulose agar media (with the growth medium and 0.1 μg/ml TPCK trypsin) to each well. Once the agar medium was set, the plates were incubated at 37oC with 5% CO2 for 3 days, after which the resulting plaques are counted. A second count was performed following removal of the agar overlay and staining of the cells with crystal violet (n=5, mean ± SEM, PFU/group). Results Mice treated with the recombinant H1N1 (rH1N1) haemagglutinin (HA) vaccination alone (i.e. in PBS) demonstrated a moderate, but not statistically significant, reduction in viral load in both the lung (see Figure 27) and nasal tissue (see Figure 28). Mice treated with the rH1N1 HA vaccination and the control adjuvant, CPG-ODN, similarly experienced a moderate reduction in viral load in both the lung and nasal tissue. However, and most pertinently, mice treated with the rH1N1 HA vaccination and the shRNA 1 with surfactant component formulation demonstrated a considerably greater, and highly statistically significant reduction in viral load, in both the lung and nasal tissue. The anti-viral effect of the rH1N1 HA vaccination 109    following treatment with the shRNA 1 with surfactant component formulation was therefore considerably greater than that observed following treatment of said vaccination with the control adjuvant, CPG-ODN. Furthermore, significant neutrophil accumulation was observed in both the lung (see Figure 29) and nasal tissue (see Figure 30) of influenza virus-infected mice treated with vehicle (i.e. PBS) on Day 5 post-inoculation. Mice vaccinated with rH1N1 HA alone (i.e. in PBS) demonstrated a marked reduction in neutrophil accumulation in both the lung and nasal tissue and, notably, mice vaccinated with rH1N1 HA post-treatment with the control adjuvant, CPG- ODN, showed a similar degree of reduction in neutrophil accumulation. That is, the control adjuvant, CPG-ODN, failed to enhance the reduction in neutrophil accumulation induced by rH1N1 HA vaccination. On the contrary, mice treated with rH1N1 HA vaccination post- treatment with the shRNA 1 with surfactant component formulation demonstrated a further significant reduction in neutrophil accumulation in both the lung and nasal tissue, as represented by a 40% reduction in neutrophil accumulation in the lung, and by a 83% reduction in neutrophil accumulation in the nasal tissue, relative to treatment with vehicle (i.e. PBS) on Day 5 post-inoculation. That is, the shRNA 1 with surfactant component formulation enhanced the rH1N1 HA vaccination associated reduction in neutrophil accumulation. Finally, as shown in Figure 31, influenza virus infection led to substantial body weight loss in mice treated with vehicle (i.e. PBS). Intranasal vaccination of rH1N1 HA mitigated the influenza infection-induced body weight loss to a moderate extent. The control adjuvant, CPG- ODN, failed to enhance the protective effects of the rH1N1 HA vaccination, and in fact resulted in mice suffering from a greater loss in body weight than those treated with the rH1N1 HA vaccination alone (i.e. in PBS). On the contrary, treatment of mice with the rH1N1 HA vaccination post-treatment with the shRNA 1 with surfactant component formulation resulted in a more significant protective effect against the influenza virus infection-induced body weight loss, and in fact almost completely mitigated any such body weight loss. Biological Example 12: Assessment of the exposure of mRNA to ALI cultured human nasal epithelium Experimental Methods Following the encouraging results of Biological Example 6 above, the inventors have better characterised the data presented in relation to exposure of GFP-encoding mRNA, which supports the use of the formulations of the present invention in improving the delivery, and thereby increasing exposure, of mRNA molecules. Independently, the inventors conducted a further experiment in which ALI nasal epithelium cultures (Epithelix Sarl) were dosed apically with 50 µL of vehicle control, that is 0.1 mg/mL H1N1 haemagglutinin (HA)-encoding mRNA (Oz Bioscience) in phosphate buffer, or 0.1 110    mg/mL H1N1 HA-encoding mRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs. The treatment was then removed, and the apical surface of the ALI cultures was washed with 100 µL culture. Following 48 hrs incubation, ALI cultured nasal epithelial cells were collected and suspended in lysis buffer. The expression of H1N1 HA protein was detected by the standard SDS-PAGE/Western blotting system using anti-H1N1 HA antibody (#ab281949, Abcam). Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2. Separately, the inventors conducted a further experiment in which ALI bronchial epithelium cultures (Epithelix Sarl) were dosed apically with 50 µL of vehicle control, that is cystic fibrosis transmembrane conductance regulator (CFTR)-encoded mRNA (northern RNA) in phosphate buffer, or 0.1 mg/mL CFTR-encoded mRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs. The treatment was then removed, and the apical surface of the ALI cultures were washed with 100 µL culture. Following 72 hrs incubation, ALI cultured bronchial epithelial cells were collected and suspended in lysis buffer. The expression of CFTR protein was detected by the standard SDS- PAGE/Western blotting system using anti-CFTR antibody (#sc376683, Santa Cruz Biotechnology). Results As illustrated in Figure 32, treatment of ALI cultured nasal epithelial cells with GFP-mRNA in citrate buffer (i.e. vehicle) failed to result in any detectable fluorescent signal relative to background fluorescence. This is indicative of the failure of citrate buffer alone to expose ALI cultured nasal epithelial cells to GFP-encoding mRNA. On the contrary, ALI cultured nasal epithelial cells treated with GFP-encoding mRNA with surfactant component demonstrated a strong and significant fluorescent signal (MFI), indicative of the ability of the surfactant component to increase exposure of GFP-encoding mRNA to cells. Moreover, as demonstrated by Figures 33 and 34, only cells treated with H1N1 haemagglutinin (HA) mRNA with surfactant component expressed the H1N1 HA proteins 48 hrs post- treatment. That is, cells treated with H1N1 HA mRNA in PBS failed to be exposed to, and therefore failed to express the H1N1 HA protein. On the contrary, cells treated with H1N1 HA mRNA with surfactant component were able to express large quantities of H1N1 HA by the 48 hr timepoint. This is indicative of the surfactant component facilitating exposure of cells to the H1N1 HA mRNA. Finally, as illustrated in Figure 35, the cystic fibrosis transmembrane conductance regulator (CFTR) protein was constitutively expressed in bronchial epithelial cells. Transfection of said cells with CFTR mRNA with PBS failed to elicit any increase in expression of the CFTR protein 72 hrs post-treatment. On the contrary, treatment of cells with CFTR-mRNA with surfactant 111    component resulted in a significant increase in CFTR protein expression 72 hrs post- treatment, indicative of the surfactant component facilitating exposure of cells to the CFTR mRNA. Biological Example 13: Assessment of the impact of buffer identity and shear mixing on the effect of treatment of influenza virus infected ALI cultured human epithelial cells with formulations comprising shRNA 1 with surfactant component Experimental Methods ALI cultured pooled donors’ human nasal epithelium (purchased from Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase with MucilAir™ culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions. ALI cultures were dosed apically with 50 µL vehicle (citrate or phosphate buffer only), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in citrate or phosphate buffer, or shRNA 1 (2 mg/mL) in with surfactant component in citrate or phosphate buffer, for 30 min (then being removed with a pipette) four days and one day prior to virus inoculation, (influenza virus, strain PR8; 100 µL; diluted in MucilAir™ culture medium to give a final MOI of 0.2) for 60 min on Day 0. On Day 2 (48 hrs post-inoculation), sampling was conducted by adding 300 µL of culture medium to the apical surface of each for well for 10 min (being stored at -80oC). Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2 below. However, the above-described formulations were each prepared in duplicate, with one formulation produced using low-shear mixing (i.e. magnetic stirring) and one formulation produced using high-shear mixing (i.e. using a Silverson L5M homogenizer at 5000 rpm, for 5 min). Virus load (n=3, mean ± range) on Day 2 post infection (day 3 post-treatment) and CXCL10 levels on Day 0 (day 1 post-treatment), prior to virus infection, (n=3, mean ± SEM) were calculated as described in Biological Example 2 above. Results As illustrated in Figure 36, high levels of influenza virus replication were observed on Day 2 post-infection following treatment of ALI cultured nasal epithelial cells with vehicle alone (i.e. citrate or phosphate buffer alone). Treatment of cells with surfactant component in buffer (i.e. either citrate or phosphate buffer) alone resulted in a moderate anti-viral effect. However, treatment of cells with shRNA 1 with surfactant component in either citrate or phosphate buffer led to a strong anti-viral effect. Notably, there was no discernible difference between the performance of formulations produced using citrate buffer and the formulations produced using phosphate buffer. Moreover, these trends were observed irrespective of whether the formulations were produced via low-shear mixing (i.e. magnetic stirring) or via high-shear 112    mixing. In summary, neither buffer identity nor shear mixing appear to significantly influence the performance of the formulations of the present invention. Additional data (not shown) also demonstrate that formulations prepared using citrate buffer or phosphate buffer and/or produced via low-shear or high-shear mixing, perform similarly in CXCL10 induction assays, which further supports the theory that these factors do not influence the biological performance of the formulations of the present invention. Biological Example 14: Assessment of the exposure of siRNA and mRNA to ALI cultured human corneal epithelium Experimental Methods ALI cultured human corneal epithelium cells (EpiCorneal COR-100, provided by Mattek Corp. (Ashland, MA) were maintained in air-liquid interphase with COR-100-MM maintenance medium, in the sterile 12-well hanging top-plates (HNG-TOP-12), according to the manufacturer’s instructions. ALI cultures were dosed apically with 50 µL control formulation, i.e.10nM SilencerTM CyTM3- labelled negative control No.1 siRNA (Cy3 labelled siRNA, #AM4621, ThermoFisher Scientific, UK) in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate), or test formulation, i.e.10nM Cy3-labelled siRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs at 37̊ C, 5% CO2. The treatment was then removed, and the apical surface of the ALI cultures were washed with 100 µL phosphate buffered saline (PBS). Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2. ALI cultured corneal epithelial cells were collected via trypsinization (0.25% in PBS for 15 min) prior to resuspension in PBS (plus 1% bovine serum albumin (BSA) and 2% paraformaldehyde) and immediate analysis by flow cytometry using a 532 nm bandpass (BP) filter on the BD Canto II apparatus (Becton Dickinson). The count was calculated by subtracting the count of non-treated control (autofluorescence) from the treated cells. In a parallel experiment, ALI cultures were dosed apically with 50 µL control formulation, i.e. 0.1 mg/mL GFP-encoding mRNA (Vernal) in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate), or test formulation, i.e.0.1 mg/mL GFP-encoding mRNA with surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in the aforementioned buffer, for 4 hrs. The treatment was then removed, and incubated for 72 hrs at 37oC, 5% CO2. Following 72 hrs incubation, ALI cultured corneal epithelial cells were collected via trypsinization and analysed by flow cytometry using a 488 nm bandpass (BP) filter on the BD Canto II™ apparatus (Becton Dickinson). The cell count was determined, and the cell count of non-treated control (autofluorescence) was subtracted. 113    Results As illustrated in Figure 37, treatment of ALI cultured corneal epithelial cells with a formulation comprising Cy3-labelled siRNA with surfactant component led to a significant increase in the number of Cy3 positive cells (in particular versus background autofluorescence), whilst treatment of ALI cultured corneal epithelial cells with Cy3-labelled siRNA formulated in phosphate buffer alone did not show any positive fluorescent signals relative to background autofluorescence. These data suggest that formulation of Cy3-labelled siRNA with surfactant component facilitates a greater degree of corneal epithelial cell exposure to said siRNA, resulting in greater Cy3 expression and consequently higher levels of fluorescence. As illustrated in Figure 38, treatment of ALI cultured corneal epithelial cells with a formulation comprising GFP-encoding mRNA with surfactant component led to a significant increase in the number of GFP-positive cells (in particular versus background autofluorescence, whilst treatment of ALI cultured corneal epithelial cells with GFP-encoding mRNA formulated in phosphate buffer alone did not show any positive fluorescent signals relative to background autofluorescence. These data suggest that formulation of GFP-encoding mRNA with surfactant component facilitates a greater degree of corneal epithelial cell exposure to said mRNA, resulting in greater GFP expression and consequently higher levels of fluorescence. Biological Example 15: Assessment of CXCL10 production, upon treatment with shRNA 1, RNA Conjugate 1, and RNA Conjugate 2 in ALI cultured human nasal epithelial cells Experimental Methods ALI cultured pooled donors’ human nasal epithelium (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase with MucilAir™ culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions. ALI cultures were dosed apically with 50 µL vehicle control (citrate buffer alone: 0.20% (w/w) citric acid monohydrate and 0.28% (w/w) sodium citrate dihydrate), surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in citrate buffer, shRNA 1 (2 mg/mL) with surfactant component in citrate buffer, RNA Conjugate 1 (2 mg/mL) in citrate buffer, RNA Conjugate 1 (2 mg/mL) with surfactant component in citrate buffer, RNA Conjugate 2 (2 mg/mL) in citrate buffer, or RNA Conjugate 2 (2 mg/mL) with surfactant component in citrate buffer, for 30 min (then being removed with a pipette).24 hrs post-treatment, ALI cultures were harvested and CXCL10 concentration was quantified via the method described in Biological Example 1. Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2. Results As demonstrated by Figure 39, each of the treatment formulations induced at least moderate CXCL10 expression. However, more pertinently, whilst each of RNA Conjugate 1 and RNA 114    Conjugate 2 in buffer alone induced CXCL10 expression to some extent, formulation of each of said RNA Conjugates in surfactant component resulted in a significant enhancement of their ability to induce CXCL10 expression (i.e. increased CXCL10 expression relative to the appropriate RNA Conjugate in buffer alone). Biological Example 16: Assessment of viral load and virus-induced inflammation, upon treatment with shRNA 1, in human rhinovirus (HRV) infected, ALI cultured, human bronchial epithelial cells obtained from an asthma donor Experimental Methods ALI cultured human bronchial epithelial cells obtained from an asthma donor (provided by Epithelix Sàrl (Geneva, Switzerland)) were maintained in air-liquid interphase (ALI) with MucilAir™ culture media in Costar Transwell inserts (Corning, NY, USA) according to the manufacturer’s instructions. On Day 0 of the study, human rhinovirus type 16 (HRV16) virus inoculum (100 µL of 400,000 PFU/mL in MucilAir™ culture medium to give a final MOI of approximately 0.2) was added to the apical surface of the epithelium for 1.5 hrs (33°C/5% CO2). Virus inoculum was subsequently removed, and inserts were washed with 100 µL sterile PBS (with Ca2+/Mg2+). Subsequently, ALI cultures were dosed apically with 50 µL media, surfactant component (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) in phosphate buffer (0.06% (w/w) sodium dihydrogen phosphate and 0.08% (w/w) disodium hydrogen phosphate), shRNA 1 (2 mg/mL) with surfactant component in phosphate buffer, or pleconaril, a known HRV inhibitor (10µM in media) for 30 min (then being removed with a pipette; almost completely and without additional wash) both two days (Day -2) and one day (Day -1) prior to virus inoculation (on Day 0), before subsequent re-application of the treatments 30 min prior to virus inoculation (then being removed as described above) and subsequent incubation with virus inoculum for 90 min. Treatment formulations used in this Example were produced largely as described in Biophysical Examples 1 and 2. On Day 0 (prior to virus inoculation), and Days 1, 2 and 5, sampling was conducted via addition of 300 µL of culture medium to the apical surface of each for well for 10 min (being stored at - 80oC). CXCL10, CXCL8 and CCL5 concentration was quantified via an enzyme-linked immunosorbent assay using the MSD multiplex platform (U-PLEX Human IP-10 Assay, catalogue #K151UFK-2; U-PLEX Human IL-8 Assay, Cat# K151TYK; U-PLEX Human RANTES Assay, Cat# K151A2K, n=3, mean ± SEM). On Day 2 post-virus inoculation (day 3 post-the first treatment), virus load was quantified using the 50% Tissue Culture Infectious Dose (TCID50). Briefly, the apical wash was defrosted and serial dilutions of the apical wash samples with 1% FBS DMEM medium containing 15 mM MgCl2, were applied to plates lined with HeLa cells (ATCC®, Manassas, #CCL-2, 80% 115    confluency), prior to incubation at 34oC with 5% CO2 for 5 days, and in particular until the CPE induced by the vehicle control became visible. TCID50 was calculated using the Reed-Muench formula (n=3, mean ± range). Specifically, the first dilution with a CPE positivity % ≥ 50 (“Dilution More”) and the first dilution with CPE positivity % < 50% (“Dilution Less”) were identified and the proportionate distance (PD) was calculated using the formula: PD = (% of CPE positive wells in Dilution More – 50%) / (% of CPE positive wells in Dilution More - % of CPE positive wells in Dilution Less) Log TCID50 = Log Dilution More + PD Results As illustrated in Figures 47 to 49, human rhinovirus (HRV) infection of the bronchial epithelium from an asthma donor induced significant release of the pro-inflammatory cytokines CXCL10 (Figure 47), CXCL8 (Figure 48), and CCL5 (Figure 49). Treatment of the bronchial epithelium with surfactant component only had a moderate effect in reducing virus-induced release of these pro-inflammatory cytokines by day 5 post-inoculation. Similarly, the known HRV inhibitor, pleconaril, had a moderate effect in reducing virus-induced release of CXCL10, CXCL8, and CCL5, by day 5 post-inoculation. However, treatment of the bronchial epithelium with shRNA 1 with surfactant component had a significant effect in reducing virus-induced release of these pro-inflammatory cytokines by day 5 post-inoculation, and in fact said treatment almost completely inhibited the virus-induced release of CXCL10, CXCL8, and CCL5. Notably, treatment with a formulation comprising shRNA 1 with surfactant component as greater effect in reducing pro-inflammatory cytokine release relative to treatment with the known HRV inhibitor, pleconaril. Moreover, as illustrated in Figure 50, high levels of HRV replication were detected in an apical wash from HRV-infected bronchial epithelial cells treated with media (i.e. vehicle) on day 2 post-inoculation (mean: 4.1 Log, TCID50/mL). Treatment of HRV-infected bronchial epithelial cells with surfactant component alone had no notable anti-viral effect. However, treatment with a formulation comprising shRNA 1 with surfactant component had a clear anti-viral effect, as represented by a 0.9 and 1.6 Log reduction in viral load relative to treatment with media only or surfactant component only, respectively. The known HRV inhibitor, pleconaril, had a more significant anti-viral effect, as represented by a 2.3 Log reduction in viral load relative to treatment with media only. In summary, in the bronchial epithelium of an asthma patient, infected by HRV, treatment with a formulation comprising shRNA 1 with surfactant component, has a clear anti-inflammatory and anti-viral effect. Thiis is particularly pertinent as HRV, and in particular HRV16, is a known driver of asthma exacerbations. Therefore these data suggests that formulations comprising 116    shRNA 1 with surfactant component can effectively be used prophylactically against and/or treat asthma exacerbations, including HRV-related asthma exacerbations. Summary of the Biological Data The in vitro anti-viral activity of certain polynucleotide molecules has been demonstrated via induction of CXCL10, which is a marker of anti-viral interferon signalling, expression in ALI cultured nasal epithelium prior to virus infection, and reduction of influenza virus viral load in infected ALI cultured nasal epithelium. In these assay systems, CXCL10 expression was quantified by an enzyme-linked immunosorbent assay using the MSD multiplex platform and viral load was quantified using TCID50 values calculated using the Reed-Muench formula. More pertinently, the surfactant component, which itself demonstrated minimal anti-viral activity, was able to potentiate the activity of anti-viral polynucleotide molecules, as represented by increased induction of CXCL10 expression prior to virus infection and decreased viral load in influenza virus infected ALI cultured nasal epithelium. Notably, these effects were observed for five different anti-viral polynucleotide molecules of significantly different sequence and structure, shRNA 1, dsRNA 1, dsRNA 2, RNA Conjugate 1, and RNA Conjugate 2, and using three different surfactant concentrations (see Biological Examples 2 and 15). Furthermore, in some cases, formulation of an anti-viral polynucleotide molecule, such as shRNA 1 with a surfactant component resulted in a formulation with in vitro anti-viral activity comparable to, or improved relative to, the commercially available anti-viral drug oseltamivir, in an influenza virus-infected ALI cultured nasal epithelium model (see Biological Examples 3 and 4). Significantly, the in vitro anti-viral activity of a formulation of an anti-viral polynucleotide molecule, such as shRNA 1, with a surfactant component, and the potentiation of the anti-viral activity of said anti-viral polynucleotide molecule by formulation with said surfactant component, has also been demonstrated in a human rhinovirus (HRV16)-infected and a human respiratory syncytial virus (RSV) A2-infected ALI cultured nasal epithelium model (see Biological Examples 7 and 8). In addition, formulations comprising surfactant component were demonstrated to clearly increase the integrity and improve the barrier function of the epithelium, as represented by an increase in transepithelial electrical resistance (TEER), which has a clear protective effect against virus-induced epithelial damage in a human respiratory syncytial virus (RSV) A2- infected ALI cultured nasal epithelium model (see Biological Example 8). In fact, formulations comprising surfactant component were improved in their protective against such virus-induced epithelial damage relative to formulations comprising the anti-viral polynucleotide molecule, shRNA 1, which alone was only able to mitigate the loss in TEER/restore the reduced TEER observed following human RSV A2 infection of ALI cultured nasal epithelium. 117    The in vitro anti-viral activity of the formulations disclosed herein was subsequently confirmed in an in vivo mouse model of influenza infection. In particular, whilst both vehicle (water alone) and the surfactant formulation alone had no significant anti-viral activity, the anti-viral activity of an intranasally administered polynucleotide molecule, in particular shRNA 1, was potentiated by the surfactant component to a significant extent, as represented by a significant reduction in viral load and simultaneous reduction in neutrophil accumulation. The anti-viral activity of the polynucleotide molecule was more potent upon formulation with the surfactant formulation in comparison to formulation in vehicle (water). Moreover, the anti-viral activity of the polynucleotide molecule with surfactant formulation was comparable to, if not slightly superior to, the anti-viral activity of the commercially available anti-viral drug oseltamivir. Furthermore, although the polynucleotide molecule, shRNA 1, in vehicle had a protective effect against influenza virus-induced body weight loss in infected mice, formulation of said polynucleotide molecule with surfactant formulation improved the protective effect of the polynucleotide molecule such that said polynucleotide molecule prevented influenza virus- induced body weight loss to a similar extent to oseltamivir. The in vivo anti-viral activity of the formulations disclosed herein was further confirmed in an in vivo mouse model of human respiratory syncytial virus (RSV) A2 infection. In particular, whilst both vehicle (saline alone) and the surfactant component alone had no significant anti- viral activity, the anti-viral activity of an intranasally administered polynucleotide molecule, shRNA 1 with surfactant component was highly significant, as represented by a substantial reduction in viral load and simultaneous reduction in neutrophil accumulation (see Biological Example 9). In fact, the anti-viral activity of the formulation of shRNA 1 with surfactant component resulted in a formulation with in vivo anti-viral activity comparable to the commercially available anti-viral drug ribavirin (see Biological Example 9). Furthermore, although the surfactant component alone had no significant protective effect against human RSV A2-induced body weight loss in infected mice, formulation of the anti-viral polynucleotide molecule, shRNA 1, in said surfactant component resulted in a significant protective effect, and in fact prevented human RSV A2-induced body weight loss in infected mice to a similar extent to ribavirin. The in vivo anti-viral activity of the formulations disclosed herein was further assessed upon administration via different routes, and in particular following subcutaneous administration. Notably, whilst subcutaneous administration of saline (i.e. non-treatment) or surfactant component alone had no significant anti-viral activity, the anti-viral activity of a subcutaneously administered formulations comprising an anti-viral polynucleotide molecule, shRNA 1, with surfactant component was highly significant, as represented by a substantial reduction in viral load and simultaneous reduction in neutrophil accumulation in both the lung and nasal tissue of influenza virus (PR8)-infected mice (see Biological Example 10). Notably, the anti-viral 118    activity of the formulation comprising shRNA 1 with surfactant component was comparable to the anti-viral effect observed in influenza virus-infected mice orally treated with the commercially available drug oseltamivir, and simultaneously subcutaneously treated with a formulation comprising surfactant component alone. More pertinently, the anti-viral effect observed in influenza virus-infected mice subcutaneously treated with the formulation comprising shRNA 1 with surfactant component in combination with oral treatment with oseltamivir, was more significant that the anti-viral effect observed following treatment with either formulation/drug alone (see Biological Example 10). Moreover, whilst subcutaneous treatment of influenza virus-infected mice with vehicle (i.e. saline alone) or surfactant component alone failed to protect against influenza virus-induced body weight loss in infected mice, formulation of the anti-viral polynucleotide molecule, shRNA 1, in said surfactant component resulted in a significant protective effect, and in fact prevented influenza virus-induced body weight loss in infected mice to a similar extent to oral oseltamivir treatment (when co-administered with subcutaneous surfactant component alone) (see Biological Example 10). In addition, subcutaneous treatment of influenza virus-infected mice with a formulation comprising shRNA 1 with surfactant component, in combination with oral treatment of said mice with the commercially available drug oseltamivir, resulted in a further protective effect against influenza virus-induced body weight loss in infected mice and in fact entirely prevented any such weight loss. The pro-inflammatory effect of the formulations disclosed herein in vivo was further assessed in the context of intranasal vaccination of influenza virus infected mice with recombinant H1N1 (rH1N1) haemagglutinin (HA). Significantly, vaccination of mice with rH1N1 HA in PBS alone, or following treatment with the control adjuvant, CPG-ODN, resulted in moderate protection against influenza virus infection, whilst vaccination of mice with rH1N1 HA following treatment with the shRNA 1 with surfactant component formulation resulted in more significant protection against influenza virus infection, as represented by a significant reduction in viral and simultaneous reduction in neutrophil accumulation in both the lung and nasal tissue of infected mice (see Biological Example 11). Notably, the protective effect of intranasal rH1N1 HA vaccination was more greatly enhanced by previous treatment with the shRNA 1 with surfactant component formulation of the present invention than by previous treatment with the control adjuvant, CPG-ODN. Moreover, whilst intranasal vaccination of influenza-virus infected mice with rH1N1 HA in PBS resulted in moderate protection against influenza virus-induced body weight loss in infected mice (relative to treatment with vehicle, i.e. PBS alone), formulation of said rH1N1 HA in the shRNA 1 with surfactant component formulation resulted in a more significant protective effect against said influenza virus-induced body weight loss in infected mice, and in fact almost completely mitigated any such weight loss (see Biological Example 11). Moreover, the 119    protective effect of the rH1N1 HA vaccination upon formulation in the shRNA 1 with surfactant component formulation was far more significant than that observed upon formulation of the vaccination in the control adjuvant, CPG-ODN. Without wishing to be bound by theory, the present inventors consider that the liquid formulations of the present invention improve delivery of a formulated polynucleotide molecule to target cells, in particular ALI cultured nasal epithelial cells, and consequently enhance downstream activity of said formulated polynucleotide molecule. Support for this hypothesis is derived from the observation that the expression of mRNA or DNA encoded biomarkers was significantly higher upon treatment of cells with the biomarker-encoding mRNA or DNA with surfactant component relative to corresponding treatment with said mRNA or DNA in vehicle (buffer component) only (see Biological Examples 6, 12, and 14). Furthermore, it has been observed that the exposure of cells to specific siRNA molecules is significantly increased upon formulation of said siRNA molecule in surfactant component formulations relative to formulation in vehicle (phosphate buffer component) only (see Biological Example 14). Significantly, this effect has been observed in different epithelium types, including the bronchial (see Biological Example 12), nasal (see Biological Examples 6 and 12), and corneal (see Biological Example 14) epithelia. In addition, the impact of buffer identity and shear mixing on the in vitro activity of the formulations disclosed herein was assessed. Importantly, treatment of ALI cultured nasal epithelial cells with shRNA 1 with surfactant component in either citrate or phosphate buffer led to a strong anti-viral effect, as represented by a significant reduction in viral load relative to treatment with vehicle, i.e. citrate or phosphate buffer alone (see Biological Example 13). Similarly, treatment of ALI cultured nasal epithelial cells with shRNA 1 with surfactant component formulations, in either citrate or phosphate buffer, which were produced using either low-shear mixing, i.e. magnetic stirring, or high-shear mixing led to a strong anti-viral effect, as represented by a significant reduction in viral load relative to treatment with vehicle, i.e. citrate or phosphate buffer alone (see Biological Example 13). Finally, the present inventors investigated the utility of the liquid formulations of the present invention as a prophylactic treatment for asthma exacerbations, in particular human rhinovirus (HRV) infection-associated asthma exacerbations. In particular, treatment of HRV-infected bronchial epithelial cells from an asthma donor with formulations comprising shRNA 1 with surfactant component effectively inhibited virus-induced release of the pro-inflammatory cytokines CXCL10, CXCL8, and CCL5, by day 5 post-inoculation (see Biological Example 16), and in fact inhibited such release to a greater extent than the known HRV inhibitor pleconaril. Moreover, treatment of HRV-infected bronchial epithelial cells from an asthma donor with formulations comprising shRNA 1 with surfactant component resulted in a reduction in HRV viral load, relative to treatment with media alone (see Biological Example 16). 120    These results show that liquid pharmaceutical formulations of the invention including a surfactant component comprising a mixture of a fatty acid and a non-ionic surfactant, and a polynucleotide molecule, and specifically in the form of a stable colloidal emulsion, are expected to be useful for improving the delivery and consequently increasing the exposure of said polynucleotide molecule, and consequently enhancing the therapeutic effect of said polynucleotide molecule. Biophysical Examples Biophysical Example 1 Formulations comprising a polynucleotide molecule, shRNA 1, a surfactant component of three different concentrations (1: 0.005% (w/w) oleic acid and 0.0045% (w/w) polysorbate 80; 2: 0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80; and 3: 0.2% (w/w) oleic acid and 0.15% (w/w) polysorbate 80) and vehicle (water) were produced by first adding oleic acid and polysorbate 80 at the concentration required to achieve the desired final concentration to RNAse free water in a 0.5 L glass vessel prior to mixing for 10 min using an IKA EURO-STAR Overhead Lab Mixer at about 50% power. Next, a solution of shRNA 1 (0.2% (w/w)) was dispersed in the surfactant component and mixed as described above to produce a shRNA 1 surfactant composition. In a separate 1 L glass vessel, carboxymethyl cellulose (2% (w/w)) and hyaluronic acid were dispersed in purified water and homogenised using a Silverson L5M homogenizer at 8500 rpm for 2 min. This suspension was further combined with glycerol (2.1% (w/w)) by continuous stirring for 10 min. The shRNA 1 surfactant composition and the suspension were then combined by continuous mixing for 10 min, prior to buffering with sodium citrate dihydrate (0.28% (w/w)) and citric acid monohydrate (0.20% (w/w)) and mixing for a further 10 min to ensure uniform pH adjustment. A comparator formulation comprising shRNA 1 in vehicle was produced similarly. Subsequently, dynamic light scattering techniques, by means of the Zetasizer Nano S apparatus (Malvern Instruments Ltd., UK), were used to measure the translational diffusion of polynucleotide:surfactant component particles due to Brownian motion. The size of said particles can be measured relative to the viscosity of the solution according to the Stokes- Einstein equation: ^^ ^^ ^^ ൌ 3 ^^ ^^ ^^ wherein k is the Boltzmann constant, T is the absolute temperature, η is the viscosity co- efficient of the solvent, a is the particle size, and D is the diffusion co-efficient (n=3). Results As illustrated in Figures 13 (A-D) and Table 3 (below), the hydrodynamic diameter of shRNA 1 in vehicle is highly variable, as indicated by the large standard deviation of the Z-average size. Similarly, formulation of shRNA 1 with surfactant component 1 (0.005% (w/w) oleic acid 121    and 0.0045% (w/w) polysorbate 80) in vehicle, which comprises surfactant at the lowest of the tested concentrations, results in a formulation of high polydispersity and comprising particles of a highly variable size. Conversely, formulation of shRNA 1 with surfactant component 2 (0.05% (w/w) oleic acid and 0.045% (w/w) polysorbate 80) or surfactant component 3 (0.2% (w/w) oleic acid and 0.15% (w/w) polysorbate 80) in vehicle, resulted in formulations of low polydispersity, relative to a formulation comprising shRNA 1 with vehicle alone or with surfactant component 1 and vehicle, and with a consistent particle size, as indicated by small values of the standard deviation of the Z-average size. This data suggests that formulation of a polynucleotide molecule, in particular shRNA 1, with surfactant component 2 or surfactant component 3 and vehicle, results in a formulation with low polydispersity and consistent particle size, which is advantageous in a pharmaceutical product. It should be noted that each plot of Figure 13 (A-D) comprises three repeat measurements (n=3) performed with each formulation. Table 3: The effect of formulation of shRNA 1 with surfactant components of different concentrations on particle size and polydispersity Polydispersity Formulation Z-Average Size (Standard Deviation) (nm) Index shRNA 1 in water 174.57 (25.91) 0.42 (0.05) shRNA 1 with surfactant 124.15 (27.17) 0.61 (0.15) component 1 in water shRNA 1 with surfactant 197.43 (6.55) 0.47 (0.02) component 2 in water shRNA 1 with surfactant 182.63 (4.60) 0.32 (0.03) component 3 in water 122    Biophysical Example 2 Formulations comprising a polynucleotide molecule, shRNA 1, and a variety of surfactant components (see Table 4, below) were produced by firstly adding 5 mL of RNAse free water to the desired concentration of the relevant fatty acid (i.e. caprylic acid or oleic acid) and by simultaneously, in a separate vessel, adding 5 mL of RNAse free water to the desired concentration of the relevant non-ionic surfactant (i.e. polysorbate 80 or Brij 35). Each of these fatty acid and non-ionic surfactant mixtures was manually mixed using a stainless steel spatula. Each of the fatty acid mixture and non-ionic surfactant mixture were then added to 80 mL of RNAse free water in a 0.25 L glass vessel. This solution was mixed for about 10 min using an IKA EURO-STAR Overhead Lab Mixer at about 50% power, until a homogenous mixture was produced. Next, a solution of shRNA 1 (0.2% (w/w)) was dispersed in the surfactant component mixture solution and mixed as descried above, until entirely unfirm in constitution, to produce a formulations of shRNA in the relevant surfactant component. The pH of the shRNA 1 in surfactant component formulations was then adjusted to the desired value using either 1 M 1 in L5M nual o S on of said kes- y co- dded ent. terial ment
Figure imgf000125_0001
As illustrated in Figures 40 to 44 and Table 4 (below), the hydrodynamic diameter of shRNA 1 in each of the tested surfactant component formulations is largely consistent as indicated by the small value of the standard deviation of the Z-average size. This consistent particle size is 123    advantageous in a pharmaceutical product. Moreover, each of the tested surfactant component formulations results in a relatively low polydispersity, which is advantageous in a pharmaceutical product. It should be noted that the different composition of the surfactant components appeared to influence the average particle size, although each of the tested surfactant components formulations resulted in an average particle size which is suitable for use in a pharmaceutical product. It should be noted that the larger average particle size observed in relation to the surfactant component of polysorbate 80 (4.5% (w/w)) + caprylic acid (5.0% (w/w)) is likely due to use of the surfactant component elements at a higher concentration. Moreover, whilst it appears that polysorbate 80 as non-ionic surfactant results in particles of a lower average size, formulations comprising Brij 35 as the non-ionic surfactant also comprised particles of a suitable size and with a low polydispersity. It should be noted that each of Figures 40 to 44 comprise three repeat measurements (n=3) performed with each surfactant component formulation. Table 4: The effect of formulation of shRNA 1 with surfactant components comprising different fatty acids and non-ionic surfactants on particle size and polydispersity Z-Average Size Surfactant Component (Standard Deviation) Polydispersity Index (nm) polysorbate 80 (0.045% (w/w)) + caprylic 80.56 (2.73) 0.36 (0.06) acid (0.05% (w/w)) polysorbate 80 (0.045% (w/w)) + oleic 135.80 (2.69) 0.23 (0.03) acid (0.05% (w/w)) Brij 35 (0.045% (w/w)) + oleic acid 123.50 (9.62) 0.32 (0.01) (0.05% (w/w)) Brij 35 (0.45% (w/w)) + caprylic acid 243.10 (4.48) 0.11 (0.04) (0.5% (w/w)) polysorbate 80 (4.5% (w/w)) + caprylic 372.67 (10.27) 0.29 (0.01) acid (5.0% (w/w)) Biophysical Example 3 Formulations comprising a polynucleotide molecule, shRNA 1, and a variety of surfactant components (see Figures 45 (A-F) were produced as described in Biophysical Example 1 and Biophysical Example 2. Subsequently, the formulations were imaged using cryo-transmission 124    electron microscopy (cryo-TEM), in order to observe the particles formed by the formulations, and in particular by the surfactant component of the formulation. Specifically, a sample of the formulations was plunge frozen onto Lacey Carbon grids (size: 300), which had been made hydrophilic via glow discharge at 15 mA for 25 seconds. The plunge freezing process was performed under controlled conditions: 15oC and 90% humidity, and utilised a blot time of 2 seconds. Subsequently, the Lacey Carbon grids were plunge frozen into liquid ethane and stored under liquid nitrogen until imaging. Imaging was performed using the FEI Talos L120C G2 Transmission Electron Microscope, where sample Lacey Carbon grids were loaded into the Gatan transfer holder and maintained at temperatures below -170°C. Low dose imagine techniques were used to minimise radiation- associated damage, involving use of a software-controlled beam blanking system to limit electron exposure prior to image capture. In particular, the beam was focused on an area of limited interest adjacent to an area of interest, prior to re-tilting to the original position during exposure mode for capture of the image at the area of interest. Images were taken using the BM-Ceta CCD Camera, employed under focus to enhance
Figure imgf000127_0001
Figures 45 (B), (E), and (F), show that the polynucleotide molecule (e.g. shRNA 1) is present within (i.e. is a component of) the stable colloidal particles, and for example is present around the core of the particles and at the interface between the core of the particles and the non- ionic surfactant (see the inner dark band of the particles in these Figures). 125    Biophysical Example 4 Formulations comprising a polynucleotide molecule, shRNA 1, were prepared using a surfactant component comprising oleic acid (0.05 % (w/w)) and polysorbate 80 (0.045% (w/w)) according to Biophysical Example 1 and Biophysical Example 2 and using high-shear mixing. Said formulations were then analysed using differential scanning calorimetry (DSC) techniques in order to evaluate the thermal properties of the formulations Specifically, samples of the relevant formulation were diluted with the surfactant component, to a final concentration of 1 mg/mL shRNA 1, prior to analysis with a TA Instruments Nano Differential Scanning Calorimeter. Both sample formulations, and formulation buffers, were analysed between 25oC and 95oC, at a scanning rate of 1oC per min and using a pre-run equilibration at 25oC for 10 min, under a pressure of 3 atmospheres. The resultant DSC data was processed via subtraction of the formulation buffer baseline from the sample endotherm data, prior to integration using a sigmoidal and polynomial baseline where applicable. An overlay of baseline-subtracted thermograms allows for direct comparison of thermal transitions between samples, revealing variations in thermal properties and in particular in melting temperature (Tm), which is indicative of thermal stability. Results The melting temperature (Tm) of shRNA 1 in vehicle (water) has been determined as 30oC. As determined from Figure 46, the Tm of shRNA 1 when prepared in the surfactant component formulation is about 67oC, which is indicative of incorporation of shRNA 1 into a stable colloidal emulsion, and resultant stabilisation of said shRNA. Moreover, as illustrated in Figure 46, a clear exothermic event is present within the DSC thermogram at between 70oC and 90oC, which indicates a stabilising interaction between the non-ionic surfactant (i.e. polysorbate 80) and fatty acid (i.e. oleic acid), which is therefore somewhat responsible for the structural stability of the stable colloidal emulsion, and consequently the stabilisation of any polynucleotide molecule packaged therein. It should be noted that Figure 46 demonstrates the stability of stable colloidal emulsions formed by formulations comprising surfactant component (polysorbate 80 0.045% (w/w) + oleic acid (0.05% (w/w)) only (top panel), formulations comprising 2 mg/mL polynucleotide molecule (shRNA 1) with surfactant component (middle panel), and formulations comprising 20 mg/mL polynucleotide molecule (shRNA 1) with surfactant component (bottom panel). Summary of the Biophysical Data The formulation of a polynucleotide molecule, specifically shRNA 1, with a surfactant component comprising a mixture of a fatty acid, in particular oleic acid, and a non-ionic surfactant, in particular polysorbate 80, at specific concentrations, has been demonstrated to result in a formulation, and specifically a stable colloidal emulsion, in which the colloidal particle size is highly consistent and in which the polydispersity of the formulation is low. Each 126    of these factors is highly advantageous in a pharmaceutical product (see Biophysical Example 1). Moreover, to further investigate the suitability of a surfactant component, which comprises a mixture of a fatty acid and a non-ionic surfactant, for producing stable colloidal emulsion formulations which will function well as pharmaceutical products, the present inventors tested alternative fatty acid and non-ionic surfactant combinations. In particular, formulation of a polynucleotide molecule, specifically shRNA 1, with a surfactant component comprising caprylic acid or oleic acid (as the fatty acid) and polysorbate 80 or Brij 35 (as the non-ionic surfactant), in each case produced a formulation in which stable colloidal particles of a consistent particle size are formed, and in which the polydispersity of the formulation is low (see Biophysical Example 2). Further support for the formulations of the present invention producing stable colloidal emulsions was provided upon visualisation of said formulation. Specifically, formulations comprising a range of surfactant components (i.e. different fatty acids and non-ionic surfactants), each produced clearly visible, but sparsely distributed, particles, of a consistent size and shape, and which typically ranged between 50 and 300 nm in diameter. Without wishing to be bound by theory, the present inventors that particles of this size are likely to be optimal for efficient polynucleotide packaging, and consequently delivery (see Biophysical Example 3). Finally, the stable colloidal emulsions formed by the formulations of the invention, in particular as a result of the presence of a surfactant component comprising a mixture of a fatty acid and a non-ionic surfactant, have been demonstrated to be highly stable, in particular as a result of specific interaction between said fatty acid and said non-ionic surfactant. Moreover, the stable colloidal emulsion formulations of the present invention to significantly stabilise a polynucleotide molecule, specifically shRNA 1, packaged therein, as represented by an increase in melting temperature (Tm) (see Biophysical Example 4). These results show that liquid pharmaceutical formulations of the invention, which include a surfactant component comprising a mixture of a fatty acid and a non-ionic surfactant and a polynucleotide molecule, and specifically in the form of a stable colloidal emulsion, are expected to be useful for efficiently packaging and delivering and said polynucleotide molecule, whilst also maintaining stability of said polynucleotide molecule. In addition, the liquid pharmaceutical formulations of the invention are expected to be particularly suitable pharmaceutical products due to the consistent particle size of the stable colloidal particles in said formulations, and the low polydispersity of the formulations. General statements Throughout the specification and the claims which follow, unless the context requires otherwise, the word ‘comprise’, and variations such as ‘comprises’ and ‘comprising’, will be 127    understood to imply the inclusion of a stated integer, step, group of integers or group of steps but not to the exclusion of any other integer, step, group of integers or group of steps. All patents, patent applications and references mentioned throughout the specification of the present invention are herein incorporated in their entirety by reference. The invention embraces all combinations of preferred and more preferred groups and suitable and more suitable groups and embodiments of groups recited above. Sequence Listing SEQ ID NO.1: GGAUCGAUCGAUCGUUCGCGAUCGAUCGAUCC SEQ ID NO.2: GGACGUACGUUUCGACGUACGUCC SEQ ID NO.3: GGCGCGGGUUCGCCCGCGCC SEQ ID NO.4: GGCGCCGGGUUCGCCCGGCGCC SEQ ID NO.5: GGCGACGUUUCGACGUCGCC SEQ ID NO.6: GGCGUACGUUUCGACGUACGCC SEQ ID NO.7: GGAUCGAUCGAUCGGAACCGAUCGAUCGAUCC SEQ ID NO.8: GGAUCGAUCGAUCGCUUGCGAUCGAUCGAUCC SEQ ID NO.9: GGAUCGAUCGAUCGUUUUUCGAUCGAUCGAUCC SEQ ID NO.10: GGAUCGAUCGAUCGACAAUGCCGAUCGAUCGAUCC SEQ ID NO.11: GCCCAGACCACC SEQ ID NO.12: GCAUGCGACCUCUGUUUGA SEQ ID NO.13: GGAUCGAUCGAUCGUU SEQ ID NO.14: CGCGAUCGAUCGAUCC 128    References Boda B, Benaoudia S, Huang S, Bonfante R, Wiszniewski L, Tseligka ED, Tapparel C, Constant S. Antiviral drug screening by assessing epithelial functions and innate immune responses in human 3D airway epithelium model. Antiviral Res. 2018 Aug;156:72-79. doi: 10.1016/j.antiviral.2018.06.007 Bullen CK, Davis SL, Looney MM. Quantification of Infectious SARS-CoV-2 by the 50% Tissue Culture Infectious Dose Endpoint Dilution Assay. Methods Mol Biol.2022;2452:131-146. doi: 10.1007/978-1-0716-2111-0_9. PMID: 35554905 Garay RP, Labaune JP. Immunogenicity of Polyethylene Glycol (PEG). The Open Conference Proceedings Journal.2011; 2:104-107. doi: 10.2174/2210289201102010104 Handbook of Pharmaceutical Excipients, 5th Edition (Rowe, Sheskey, and Owen), 2006 Hoang Thi TT, Pilkington EH, Nguyen DH, Lee JS, Park KD, Truong NP. The Importance of Poly(ethylene glycol) Alternatives for Overcoming PEG Immunogenicity in Drug Delivery and Bioconjugation. Polymers (Basel).2020 Feb 2;12(2):298. doi: 10.3390/polym12020298 Huang L, Liu Y. In vivo delivery of RNAi with lipid-based nanoparticles. Annu Rev Biomed Eng.2011 Aug 15;13:507-30. doi: 10.1146/annurev-bioeng-071910-124709 Jokerst JV, Lobovkina T, Zare RN, Gambhir SS. Nanoparticle PEGylation for imaging and therapy. Nanomedicine (Lond).2011 Jun;6(4):715-28. doi: 10.2217/nnm.11.19 Khvorova A, Watts JK. The chemical evolution of oligonucleotide therapies of clinical utility. Nat Biotechnol.2017 Mar;35(3):238-248. doi: 10.1038/nbt.3765 Kulkarni JA, Witzigmann D, Thomson SB, Chen S, Leavitt BR, Cullis PR, van der Meel R. The current landscape of nucleic acid therapeutics. Nat Nanotechnol.2021 Jun;16(6):630-643. doi: 10.1038/s41565-021-00898-0 Li B, Manan RS, Liang SQ, Gordon A, Jiang A, Varley A, Gao G, Langer R, Xue W, Anderson D. Combinatorial design of nanoparticles for pulmonary mRNA delivery and genome editing. Nat Biotechnol.2023 Mar 30. doi: 10.1038/s41587-023-01679-x Ren X, Linehan MM, Iwasaki A, Pyle AM. RIG-I Selectively Discriminates against 5'- Monophosphate RNA. Cell Rep. 2019 Feb 19;26(8):2019-2027 doi: 10.1016/j.celrep.2019.01.107 Springer AD, Dowdy SF. GalNAc-siRNA Conjugates: Leading the Way for Delivery of RNAi Therapeutics. Nucleic Acid Ther.2018 Jun;28(3):109-118. doi: 10.1089/nat.2018.0736 Song LY, Ahkong QF, Rong Q, Wang Z, Ansell S, Hope MJ, Mui B. Characterization of the inhibitory effect of PEG-lipid conjugates on the intracellular delivery of plasmid and antisense DNA mediated by cationic lipid liposomes. Biochim Biophys Acta.2002 Jan 2;1558(1):1-13. doi: 10.1016/s0005-2736(01)00399-6 129    References
Boda B, Benaoudia S, Huang S, Bonfante R, Wiszniewski L, Tseligka ED, Tapparel C, Constant S. Antiviral drug screening by assessing epithelial functions and innate immune responses in human 3D airway epithelium model. Antiviral Res. 2018 Aug; 156:72-79. doi: 10.1016/j. antiviral.2018.06.007
Bullen CK, Davis SL, Looney MM. Quantification of Infectious SARS-CoV-2 by the 50% Tissue Culture Infectious Dose Endpoint Dilution Assay. Methods Mol Biol. 2022;2452:131-146. doi: 10.1007/978-1-0716-2111-0_9. PMID: 35554905
Garay RP, Labaune JP. Immunogenicity of Polyethylene Glycol (PEG). The Open Conference Proceedings Journal. 2011 ; 2:104-107. doi: 10.2174/2210289201102010104
Handbook of Pharmaceutical Excipients, 5th Edition (Rowe, Sheskey, and Owen), 2006 Hoang Thi TT, Pilkington EH, Nguyen DH, Lee JS, Park KD, Truong NP. The Importance of Poly(ethylene glycol) Alternatives for Overcoming PEG Immunogenicity in Drug Delivery and Bioconjugation. Polymers (Basel). 2020 Feb 2;12(2):298. doi: 10.3390/polym12020298 Huang L, Liu Y. In vivo delivery of RNAi with lipid-based nanoparticles. Annu Rev Biomed Eng. 2011 Aug 15;13:507-30. doi: 10.1146/annurev-bioeng-071910-124709
Jokerst JV, Lobovkina T, Zare RN, Gambhir SS. Nanoparticle PEGylation for imaging and therapy. Nanomedicine (Lond). 2011 Jun;6(4):715-28. doi: 10.2217/nnm.11.19
Khvorova A, Watts JK. The chemical evolution of oligonucleotide therapies of clinical utility. Nat Biotechnol. 2017 Mar;35(3):238-248. doi: 10.1038/nbt.3765
Kulkarni JA, Witzigmann D, Thomson SB, Chen S, Leavitt BR, Cullis PR, van der Meel R. The current landscape of nucleic acid therapeutics. Nat Nanotechnol. 2021 Jun;16(6):630-643. doi: 10.1038/S41565-021 -00898-0
Li B, Manan RS, Liang SQ, Gordon A, Jiang A, Varley A, Gao G, Langer R, Xue W, Anderson D. Combinatorial design of nanoparticles for pulmonary mRNA delivery and genome editing. Nat Biotechnol. 2023 Mar 30. doi: 10.1038/s41587-023-01679-x
Ren X, Linehan MM, Iwasaki A, Pyle AM. RIG-I Selectively Discriminates against 5'- Monophosphate RNA. Cell Rep. 2019 Feb 19;26(8):2019-2027 doi: 10.1016/j. celrep.2019.01.107
Springer AD, Dowdy SF. GalNAc-siRNA Conjugates: Leading the Way for Delivery of RNAi Therapeutics. Nucleic Acid Ther. 2018 Jun;28(3):109-118. doi: 10.1089/nat.2018.0736
Song LY, Ahkong QF, Rong Q, Wang Z, Ansell S, Hope MJ, Mui B. Characterization of the inhibitory effect of PEG-lipid conjugates on the intracellular delivery of plasmid and antisense DNA mediated by cationic lipid liposomes. Biochim Biophys Acta. 2002 Jan 2; 1558(1):1-13. doi: 10.1016/s0005-2736(01 )00399-6

Claims

Claims 1. An aqueous liquid pharmaceutical formulation, in the form of a stable colloidal emulsion, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule.
2. The aqueous liquid pharmaceutical formulation according to claim 1, wherein the fatty acid is selected from the group consisting of arachidic acid, arachidonic acid, caprylic acid, lauric acid, linoleic acid, linolenic acid, myristic acid, myristoleic acid, oleic acid, palmitic acid, palmitoleic acid, sapienic acid, stearic acid, and vaccenic acid.
3. The aqueous liquid pharmaceutical formulation according to claim 2, wherein the fatty acid is selected from caprylic acid and oleic acid.
4. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 3, wherein the non-ionic surfactant is selected from the group consisting of polyoxyalkylenes, such as poloxamers, alkyl ethers of polyethylene glycol, alkylphenyl ethers of polyethylene glycol, and fatty acid esters, such as polyoxyethylene sorbitan fatty acid esters.
5. The aqueous liquid pharmaceutical formulation according to claim 4, wherein the non- ionic surfactant is selected from alkyl ethers of polyethylene glycol and polyoxyethylene sorbitan fatty acid esters.
6. The aqueous liquid pharmaceutical formulation according to claim 1, wherein the surfactant component is selected from the group consisting of mixtures of (a) oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (b) lauric acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (c) linoleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (d) linolenic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (e) palmitic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (f) stearic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (g) oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyalkylene, such as a poloxamer, (h) oleic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol, (i) oleic acid or a pharmaceutically acceptable salt thereof and an alkylphenyl ether of polyethylene glycol, (j) caprylic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester, (k) caprylic acid or a pharmaceutically acceptable salt thereof and a polyoxyalkylene, such as a poloxamer, (l) caprylic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol, and (m) caprylic acid or a pharmaceutically acceptable salt thereof and an alkylphenyl ether of polyethylene glycol. 130   
7. The aqueous liquid pharmaceutical formulation according to claim 6, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester.
8. The aqueous liquid pharmaceutical formulation according to claim 7, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and polysorbate 80.
9. The aqueous liquid pharmaceutical formulation according to claim 6, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and a polyoxyethylene sorbitan fatty acid ester.
10. The aqueous liquid pharmaceutical formulation according to claim 9, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and polysorbate 80.
11. The aqueous liquid pharmaceutical formulation according to claim 6, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol.
12. The aqueous liquid pharmaceutical formulation according to claim 11, wherein the surfactant component is a mixture of oleic acid or a pharmaceutically acceptable salt thereof and Brij 35 (polyoxyethylene (23) lauryl ether).
13. The aqueous liquid pharmaceutical formulation according to claim 6, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and an alkyl ether of polyethylene glycol.
14. The aqueous liquid pharmaceutical formulation according to claim 13, wherein the surfactant component is a mixture of caprylic acid or a pharmaceutically acceptable salt thereof and Brij 35 (polyoxyethylene (23) lauryl ether).
15. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 14, wherein the fatty acid is in the form of the free acid.
16. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 15, wherein the concentration of the surfactant component in the formulation is 0.2 – 30000 µg/mL, for example 1 – 30000 µg/mL, for example 1 – 20000 µg/mL, 5 – 20000 µg/mL, 5 – 15000 µg/mL, 5 – 10000 µg/mL, or 5-5000 µg/mL.
17. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 16 wherein the ratio of the amount of fatty acid or a pharmaceutically acceptable salt thereof to non-ionic surfactant, wherein each is measured in µg/mL, is between about 5:1 and about 1:5, between about 5:1 and about 1:2, between about 4:1 and about 1:2, or between about 2:1 and about 1:2.
18. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 17, wherein the polynucleotide molecule is a ribonucleic acid (RNA) molecule. 131   
19. The aqueous liquid pharmaceutical formulation according to claim 18, wherein the RNA molecule is an mRNA, miRNA, shRNA or siRNA molecule.
20. The aqueous liquid pharmaceutical formulation according to claim 19, wherein the RNA molecule is an shRNA molecule.
21. The aqueous liquid pharmaceutical formulation according to claim 20, wherein the shRNA molecule comprises (i) a blunt end, (ii) a 5’ triphosphate or 5’ diphosphate moiety located at the 5’ terminus, and (iii) a double-stranded region between about 10 and about 18 nucleotides in length.
22. The aqueous liquid pharmaceutical formulation according to claim 20 or claim 21, wherein the shRNA molecule comprises or consists of SEQ ID NO.1 or a variant of SEQ ID NO.1.
23. The aqueous liquid pharmaceutical formulation according to claim 20 or claim 21, wherein the shRNA molecule comprises or consists of SEQ ID NO.2, SEQ ID NO.3, SEQ ID NO.4, SEQ ID NO.5, SEQ ID NO.6, SEQ ID NO.7, SEQ ID NO.8, SEQ ID NO.9 or SEQ ID NO.10, or a variant of SEQ ID NO.2, SEQ ID NO.3, SEQ ID NO.4, SEQ ID NO.5, SEQ ID NO.6, SEQ ID NO.7, SEQ ID NO.8, SEQ ID NO.9 or SEQ ID NO.10.
24. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 17, wherein the polynucleotide molecule is a deoxyribonucleic acid (DNA) molecule.
25. The aqueous liquid pharmaceutical formulation according to claim 24, wherein the DNA molecule is a genomic DNA (gDNA) molecule, a complementary DNA (cDNA) molecule, or an extra-chromosomal DNA molecule, for example a plasmid DNA molecule.
26. The aqueous liquid pharmaceutical formulation according to claim 1, wherein the polynucleotide molecule has the structure of Formula (I): 5’-Pz-(N)bN-3’-(E)y(E)-L-(E)(E)y’-5’-N(N)b’-3’ wherein 5’-Pz-(N)bN-3’ represents the first nucleic acid sequence; 5’-N(N)b’-3’ represents the second nucleic acid sequence; P at each instance is independently a phosphate or analog thereof; z is 2 or 3; N is, at each instance, any nucleotide or modified nucleotide or analog or derivative thereof; b and b’ are independently 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18; 5’-(E)y(E)-L-(E)(E)y’-3’ represents the connector element wherein E at each occurrence is independently any nucleotide, modified nucleotide, or abasic; y and y’ are independently 0-9, provided that y + y’ equals 0-8; L is a non-nucleotide segment having the structure 132    wherein X and X’ are independently O or S; Y and Y’ are independently OR”, SR”, or NRR’; V and V’ are independently O, S, or NRR’; q is 1-20; k is 1-20; t is 1-20; M selected from aliphatic, substituted aliphatic, aryl, substituted aryl, heteroalkyl, heterocyclyl or substituted heterocyclyl; W is any reactive group; and d is 0 or 1.
27. The aqueous liquid pharmaceutical formulation according to claim 1, wherein the polynucleotide molecule comprises, consists of, or consists essentially of SEQ ID NO.13 and SEQ ID NO.14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L1.
28. The aqueous liquid pharmaceutical formulation according to claim 1, wherein the polynucleotide molecule comprises, consists of, or consists essentially of SEQ ID NO.13 and SEQ ID NO.14, wherein the 3’ end of SEQ ID NO.13 is connected to the 5’ end of SEQ ID NO.14 via the non-nucleotide moiety, L2.
29. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 28, wherein the polynucleotide molecule comprises one or more sugar-modified nucleotides which each comprise a 2’ OH (or 2’ H) modification.
30. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 29, wherein the polynucleotide molecule comprises one or more backbone-modified nucleotides, 133    in particular wherein the backbone modification is replacement of the phosphoester group of a nucleotide with a phosphorothioate group.
31. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 30, wherein the polynucleotide molecule comprises one or more base-modified nucleotides.
32. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 31, wherein the average particle size of the stable colloidal particles is between about 10 and about 1000 nm, such as between about 50 and about 1000 nm, such as between about 50 and about 750 nm, such as between about 50 and about 500 nm, for example between about 50 and about 400 nm, for example between about 50 and about 300 nm.
33. The aqueous liquid pharmaceutical formulation according to claim 32, wherein the average particle size of the stable colloidal particles is between about 100 and about 300 nm.
34. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 33, wherein the aqueous liquid pharmaceutical formulation is suitable for topical administration to the lung or nose.
35. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 33, wherein the aqueous liquid pharmaceutical formulation is suitable for subcutaneous administration, e.g. subcutaneous injection.
36. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 33, wherein the aqueous liquid pharmaceutical formulation is suitable for ocular administration, e.g. intra-ocular administration or topical administration to the eye.
37. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 36, wherein the aqueous liquid pharmaceutical formulation does not comprise a protein.
38. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 37, wherein the aqueous liquid pharmaceutical formulation does not comprise a cationic lipopeptide, for example the aqueous liquid pharmaceutical formulation does not comprise polymyxin B.
39. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 38, wherein the aqueous liquid pharmaceutical formulation does not comprise an inorganic nanoparticle.
40. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 39, wherein the aqueous liquid pharmaceutical formulation does not comprise a lipid nanoparticle (LNP) or a liposome.
41. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 40, wherein the aqueous liquid pharmaceutical formulation does not comprise a neutral lipid, for example the aqueous liquid pharmaceutical formulation does not comprise cholesterol or an analogue thereof. 134   
42. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 41, wherein the aqueous liquid pharmaceutical formulation does not comprise a cationic lipid.
43. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 42, wherein the aqueous liquid pharmaceutical formulation does not comprise a helper lipid, for example the aqueous liquid pharmaceutical formulation does not comprise dioleoylphosphatidylethanolamine (DOPE) or phosphatidylcholine.
44. The aqueous liquid pharmaceutical formulation according to any one of claims 1 to 43 for use as a medicament.
45. The aqueous liquid pharmaceutical formulation for use according to claim 44, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response, for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response.
46. A method for the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation according to any one of claims 1 to 43, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response.
47. Use of an aqueous liquid pharmaceutical formulation according to any one of claims 1 to 43, wherein the polynucleotide molecule is a polynucleotide molecule which stimulates or activates the innate and/or adaptive immune system and/or raises an innate and/or adaptive immune response, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by stimulation or activation of the innate and/or adaptive immune system and/or the raising of an innate and/or adaptive immune response.
48. The aqueous liquid pharmaceutical formulation for use, method, or use, according to any one of claims 45 to 47, wherein the disease or condition is infection by a virus or associated with infection with such a virus.
49. The aqueous liquid pharmaceutical formulation for use, method, or use, according to claim 48, wherein the virus is SARS-CoV-2 and the disease associated with viral infection is COVID-19; or wherein the virus is seasonal coronavirus, for example 229E, NL63, OC43, or HKU1, and the disease associated with viral infection is the disease associated with seasonal coronavirus, for example 229E, NL63, OC43, or HKU1, infection; or 135    wherein the virus is influenza virus and the disease associated with viral infection is influenza; or wherein the virus is respiratory syncytial virus (RSV) and the disease associated with viral infection is the disease associated with RSV infection; or wherein the virus is human rhinovirus (HRV) and the disease associated with viral infection is the disease associated with HRV infection; or wherein the virus is Middle East respiratory syndrome (MERS)-CoV and the disease associated with viral infection is MERS; or wherein the virus is an avian influenza virus and the disease associated with viral infection is avian influenza; or wherein the virus is Nipah virus and the disease associated with viral infection is the disease associated with Nipah virus infection; or wherein the virus is a human parainfluenza virus (HPIV) and the disease associated with viral infection is the disease associated with HPIV infection; or wherein the virus is a human metapneumovirus (hMPV) and the disease associated with viral infection is the disease associated with hMPV infection.
50. The aqueous liquid pharmaceutical formulation for use according to claim 44, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein or other gene product, or which increases endogenous expression of a functional protein or other gene product, for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein or other gene product, or by increasing the expression of a functional protein or other gene product.
51. A method for the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein or other gene product, or by increasing the expression of a functional protein or other gene product, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an aqueous liquid pharmaceutical formulation according to any one of claims 1 to 43, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein or other gene product, or which increases endogenous expression of a functional protein or other gene product.
52. Use of an aqueous liquid pharmaceutical formulation according to any one of claims 1 to 43, wherein the polynucleotide molecule is a polynucleotide molecule which increases endogenous expression of a protein or other gene product, or which increases endogenous expression of a functional protein or other gene product, in the manufacture of a medicament for use in the treatment of a disease or condition which is treated by increasing the endogenous expression of a protein or other gene product, or by increasing the expression of a functional protein or other gene product. 136   
53. The aqueous liquid pharmaceutical formulation for use, method, or use, according to any one of claims 50 to 52, wherein the disease or condition is a respiratory disease or condition, such as asthma, alpha-1 antitrypsin deficiency, chronic obstructive pulmonary disease (COPD), primary ciliary dyskinesia (PCD), pulmonary fibrosis, sarcoidosis or cystic fibrosis.
54. An immunostimulatory composition, which is an aqueous liquid formulation, in the form of a stable colloidal emulsion, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) an immunostimulatory polynucleotide molecule.
55. An immunostimulatory composition according to claim 54, for use in stimulating or activating an anti-viral innate and/or adaptive immune response in a subject.
56. An immunogenic composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule which encodes an immunogen.
57. A vaccine composition, which is an aqueous liquid formulation, comprising (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant and (ii) a polynucleotide molecule which encodes a vaccine immunogen.
58. An immunogenic composition or vaccine composition according to claim 56 or claim 57, further comprising an immunostimulatory polynucleotide molecule.
59. An immunogenic composition or vaccine composition according to any one of claims 56 to 58, for use in raising an immune response in a subject, wherein the immune response is raised against the immunogen or vaccine immunogen encoded by the polynucleotide molecule.
60. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers, and (iv) a diluent.
61. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers selected from citrate and phosphate, and (iv) a diluent selected from isotonic saline (0.9% w/v) and sterile or purified water.
62. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or 137    more buffers, (iv) a diluent, (v) optionally, one or more suspending agents, (vi) optionally, one or more wetting or thickening agents, and (vii) optionally one or more osmotic or tonicity adjusting agents.
63. An aqueous liquid pharmaceutical formulation consisting of, or consisting essentially of: (i) a surfactant component which is a mixture of a fatty acid or a pharmaceutically acceptable salt thereof and a non-ionic surfactant, (ii) a polynucleotide molecule, (iii) one or more buffers selected from citrate and phosphate, (iv) a diluent selected from isotonic saline (0.9% w/v) and sterile or purified water, (v) optionally, one or more suspending agents, (vi) optionally, one or more wetting or thickening agents, and (vii) optionally one or more osmotic or tonicity adjusting agents. 138   
PCT/US2024/035999 2023-06-29 2024-06-28 Novel formulations WO2025006870A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202363510985P 2023-06-29 2023-06-29
US63/510,985 2023-06-29
USPCT/US2024/030546 2024-05-22
PCT/US2024/030546 WO2024243293A1 (en) 2023-05-24 2024-05-22 Connector modified synthetic rig-i agonists and methods of using the same

Publications (1)

Publication Number Publication Date
WO2025006870A1 true WO2025006870A1 (en) 2025-01-02

Family

ID=93939889

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/035999 WO2025006870A1 (en) 2023-06-29 2024-06-28 Novel formulations

Country Status (1)

Country Link
WO (1) WO2025006870A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6267985B1 (en) * 1999-06-30 2001-07-31 Lipocine Inc. Clear oil-containing pharmaceutical compositions
WO2016057693A1 (en) * 2014-10-10 2016-04-14 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhalation delivery of conjugated oligonucleotide
US11141377B2 (en) * 2017-06-15 2021-10-12 Infectious Disease Research Institute Nanostructured lipid carriers and stable emulsions and uses thereof
WO2023286076A1 (en) * 2021-07-13 2023-01-19 Gennova Biopharmaceuticals Ltd. Rna adsorbed onto lipid nano-emulsion particles and its formulations.
WO2023118896A1 (en) * 2021-12-23 2023-06-29 Subintro Limited Novel antiviral compositions comprising oleic acid

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6267985B1 (en) * 1999-06-30 2001-07-31 Lipocine Inc. Clear oil-containing pharmaceutical compositions
WO2016057693A1 (en) * 2014-10-10 2016-04-14 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhalation delivery of conjugated oligonucleotide
US11141377B2 (en) * 2017-06-15 2021-10-12 Infectious Disease Research Institute Nanostructured lipid carriers and stable emulsions and uses thereof
WO2023286076A1 (en) * 2021-07-13 2023-01-19 Gennova Biopharmaceuticals Ltd. Rna adsorbed onto lipid nano-emulsion particles and its formulations.
WO2023118896A1 (en) * 2021-12-23 2023-06-29 Subintro Limited Novel antiviral compositions comprising oleic acid

Similar Documents

Publication Publication Date Title
JP6921144B2 (en) RNA interference-mediated inhibition of hepatitis B virus (HBV) gene expression using small molecule interfering nucleic acid (siNA)
ES2992653T3 (en) Methods of preparing lipid nanoparticles
US20220195433A1 (en) Rna interference mediated inhibition of gene expression using short interfering nucleic acids (sina)
DK2494993T3 (en) Amino acid lipids and uses thereof
US9080186B2 (en) Compositions comprising cationic amphiphiles and colipids for delivering therapeutic molecules
US20070218122A1 (en) siRNA silencing of influenza virus gene expression
US9320798B2 (en) Lipid delivery formulations
Zhang et al. Airway epithelial cell-specific delivery of lipid nanoparticles loading siRNA for asthma treatment
AU2012268619B2 (en) RNA interference mediated inhibition of isocitrate dehydrogenase (IDH1) gene expression
TW201701887A (en) RNA interference therapeutics against Ebola virus
CN117550985A (en) Lipid compound and application thereof
WO2025006870A1 (en) Novel formulations
US20220056444A1 (en) Process to inhibit or eliminate eosinophilic diseases of the airway and related conditions
WO2012021383A2 (en) RNA INTERFERENCE MEDIATED INHIBITION OF MITOGEN-ACTIVATED PROTEIN KINASE 1 (MAPK1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP3284822A1 (en) Therapeutic agent for eye disease
EP4271364A1 (en) Micellar nanoparticles and uses thereof
US20230129386A1 (en) Treatment of covid-19 with reverse micelle system comprising unmodified oligonucleotides
US20240209035A1 (en) Polynucleotides capable of enhanced protein expression and uses thereof
WO2024175707A1 (en) A synthetic oligonucleotide for treating nidovirales infections
HK1259870A1 (en) Rna interference mediated inhibition of hepatitis b virus (hbv) gene expression using short interfering nucleic acid (sina)

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: 24832995

Country of ref document: EP

Kind code of ref document: A1